Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
Add more filters

Publication year range
1.
Cell ; 152(1-2): 262-75, 2013 Jan 17.
Article in English | MEDLINE | ID: mdl-23332760

ABSTRACT

22q11.2 microdeletions result in specific cognitive deficits and schizophrenia. Analysis of Df(16)A(+/-) mice, which model this microdeletion, revealed abnormalities in the formation of neuronal dendrites and spines, as well as altered brain microRNAs. Here, we show a drastic reduction of miR-185, which resides within the 22q11.2 locus, to levels more than expected by a hemizygous deletion, and we demonstrate that this reduction alters dendritic and spine development. miR-185 represses, through an evolutionarily conserved target site, a previously unknown inhibitor of these processes that resides in the Golgi apparatus and shows higher prenatal brain expression. Sustained derepression of this inhibitor after birth represents the most robust transcriptional disturbance in the brains of Df(16)A(+/-) mice and results in structural alterations in the hippocampus. Reduction of miR-185 also has milder age- and region-specific effects on the expression of some Golgi-related genes. Our findings illuminate the contribution of microRNAs in psychiatric disorders and cognitive dysfunction.


Subject(s)
Brain/metabolism , Embryo, Mammalian/metabolism , MicroRNAs/metabolism , Amino Acid Sequence , Animals , Brain/embryology , Chromosome Deletion , Chromosomes, Human, Pair 22/genetics , Disease Models, Animal , Golgi Apparatus/metabolism , Hippocampus/metabolism , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , MicroRNAs/genetics , Molecular Sequence Data , Neuronal Plasticity , Neurons/metabolism , Proteins/chemistry , Proteins/genetics , Proteins/metabolism , RNA-Binding Proteins
2.
Am J Hum Genet ; 105(3): 493-508, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31447100

ABSTRACT

Histones mediate dynamic packaging of nuclear DNA in chromatin, a process that is precisely controlled to guarantee efficient compaction of the genome and proper chromosomal segregation during cell division and to accomplish DNA replication, transcription, and repair. Due to the important structural and regulatory roles played by histones, it is not surprising that histone functional dysregulation or aberrant levels of histones can have severe consequences for multiple cellular processes and ultimately might affect development or contribute to cell transformation. Recently, germline frameshift mutations involving the C-terminal tail of HIST1H1E, which is a widely expressed member of the linker histone family and facilitates higher-order chromatin folding, have been causally linked to an as-yet poorly defined syndrome that includes intellectual disability. We report that these mutations result in stable proteins that reside in the nucleus, bind to chromatin, disrupt proper compaction of DNA, and are associated with a specific methylation pattern. Cells expressing these mutant proteins have a dramatically reduced proliferation rate and competence, hardly enter into the S phase, and undergo accelerated senescence. Remarkably, clinical assessment of a relatively large cohort of subjects sharing these mutations revealed a premature aging phenotype as a previously unrecognized feature of the disorder. Our findings identify a direct link between aberrant chromatin remodeling, cellular senescence, and accelerated aging.


Subject(s)
Cellular Senescence/physiology , Histones/physiology , Aneuploidy , Cell Nucleolus/metabolism , Child , Chromatin/metabolism , DNA Methylation , Female , Histones/chemistry , Humans , Infant , Male , Middle Aged
3.
Proc Natl Acad Sci U S A ; 114(30): E6127-E6136, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28696314

ABSTRACT

Identification of protective loss-of-function (LoF) mutations holds great promise for devising novel therapeutic interventions, although it faces challenges due to the scarcity of protective LoF alleles in the human genome. Exploiting the detailed mechanistic characterization of animal models of validated disease mutations offers an alternative. Here, we provide insights into protective-variant biology based on our characterization of a model of the 22q11.2 deletion, a strong genetic risk factor for schizophrenia (SCZ). Postnatal brain up-regulation of Mirta22/Emc10, an inhibitor of neuronal maturation, represents the major transcriptional effect of the 22q11.2-associated microRNA dysregulation. Here, we demonstrate that mice in which the Df(16)A deficiency is combined with a LoF Mirta22 allele show rescue of key SCZ-related deficits, namely prepulse inhibition decrease, working memory impairment, and social memory deficits, as well as synaptic and structural plasticity abnormalities in the prefrontal cortex. Additional analysis of homozygous Mirta22 knockout mice, in which no alteration is observed in the above-mentioned SCZ-related phenotypes, highlights the deleterious effects of Mirta22 up-regulation. Our results support a causal link between dysregulation of a miRNA target and SCZ-related deficits and provide key insights into beneficial LoF mutations and potential new treatments.


Subject(s)
Loss of Function Mutation , Membrane Proteins/genetics , Schizophrenia/genetics , Animals , Female , Genetic Predisposition to Disease , Male , Membrane Proteins/metabolism , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Neuronal Plasticity/genetics , Phenotype , Schizophrenia/physiopathology
4.
S Afr J Psychiatr ; 26: 1393, 2020.
Article in English | MEDLINE | ID: mdl-32391182

ABSTRACT

BACKGROUND: Schizophrenia is a heterogeneous disorder with strong genetic vulnerability. Family history of schizophrenia has been considered in genetic studies under several models. De novo genetic events seem to play a larger role in sporadic cases. AIM: This study used the familial-sporadic distinction with the aim of identifying a more homogeneous phenotype to delineate the genetic and clinical complexity of schizophrenia. SETTING: The study was conducted at Weskoppies Hospital, Pretoria, South Africa. METHODS: The study included 384 participants with schizophrenia or schizoaffective disorder from the Afrikaner founder population in South Africa who are considered comparable to Caucasian patients from the United States. A comprehensive data capturing sheet was completed. RESULTS: When schizophrenia and schizoaffective disorder diagnoses were considered jointly, we found no significant differences between the sporadic and the familial groups for age at disease onset, season of birth, comorbid diagnoses, clinical symptomatology, history of suicide or marital status. When the diagnoses were examined separately, however, the sporadic schizoaffective disorder, bipolar type, was found to have a significantly lower age at onset (mean 20.6 vs. 25.3 years). CONCLUSION: The sporadic schizoaffective disorder, bipolar type, forms a more homogeneous subgroup for genetic studies.

6.
Nature ; 471(7339): 499-503, 2011 Mar 24.
Article in English | MEDLINE | ID: mdl-21346763

ABSTRACT

Rare copy number variants (CNVs) have a prominent role in the aetiology of schizophrenia and other neuropsychiatric disorders. Substantial risk for schizophrenia is conferred by large (>500-kilobase) CNVs at several loci, including microdeletions at 1q21.1 (ref. 2), 3q29 (ref. 3), 15q13.3 (ref. 2) and 22q11.2 (ref. 4) and microduplication at 16p11.2 (ref. 5). However, these CNVs collectively account for a small fraction (2-4%) of cases, and the relevant genes and neurobiological mechanisms are not well understood. Here we performed a large two-stage genome-wide scan of rare CNVs and report the significant association of copy number gains at chromosome 7q36.3 with schizophrenia. Microduplications with variable breakpoints occurred within a 362-kilobase region and were detected in 29 of 8,290 (0.35%) patients versus 2 of 7,431 (0.03%) controls in the combined sample. All duplications overlapped or were located within 89 kilobases upstream of the vasoactive intestinal peptide receptor gene VIPR2. VIPR2 transcription and cyclic-AMP signalling were significantly increased in cultured lymphocytes from patients with microduplications of 7q36.3. These findings implicate altered vasoactive intestinal peptide signalling in the pathogenesis of schizophrenia and indicate the VPAC2 receptor as a potential target for the development of new antipsychotic drugs.


Subject(s)
DNA Copy Number Variations/genetics , Genes, Duplicate/genetics , Genetic Predisposition to Disease/genetics , Receptors, Vasoactive Intestinal Peptide, Type II/genetics , Schizophrenia/genetics , Cell Line , Chromosomes, Human, Pair 7/genetics , Cohort Studies , Cyclic AMP/metabolism , Female , Gene Dosage/genetics , Genome-Wide Association Study , Humans , Inheritance Patterns/genetics , Male , Pedigree , Receptors, Vasoactive Intestinal Peptide, Type II/metabolism , Reproducibility of Results , Schizophrenia/metabolism , Signal Transduction , Transcription, Genetic/genetics
7.
Proc Natl Acad Sci U S A ; 111(1): 343-8, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24344280

ABSTRACT

We used a family-based cluster detection approach designed to localize significant rare disease-risk variants clusters within a region of interest to systematically search for schizophrenia (SCZ) susceptibility genes within 49 genomic loci previously implicated by de novo copy number variants. Using two independent whole-exome sequencing family datasets and a follow-up autism spectrum disorder (ASD) case/control whole-exome sequencing dataset, we identified variants in one gene, Fanconi-associated nuclease 1 (FAN1), as being associated with both SCZ and ASD. FAN1 is located in a region on chromosome 15q13.3 implicated by a recurrent copy number variant, which predisposes to an array of psychiatric and neurodevelopmental phenotypes. In both SCZ and ASD datasets, rare nonsynonymous risk variants cluster significantly in affected individuals within a 20-kb window that spans several key functional domains of the gene. Our finding suggests that FAN1 is a key driver in the 15q13.3 locus for the associated psychiatric and neurodevelopmental phenotypes. FAN1 encodes a DNA repair enzyme, thus implicating abnormalities in DNA repair in the susceptibility to SCZ or ASD.


Subject(s)
Autistic Disorder/genetics , Chromosomes, Human, Pair 15 , Exodeoxyribonucleases/genetics , Genetic Predisposition to Disease , Schizophrenia/genetics , Amino Acid Sequence , Cluster Analysis , Computer Simulation , DNA Repair , Endodeoxyribonucleases , Exome , Female , Genetic Markers , Genetic Variation , Humans , Male , Molecular Sequence Data , Multifunctional Enzymes , Nucleotides/genetics , Phenotype , Risk , Sequence Homology, Amino Acid , South Africa , United States
8.
Nature ; 464(7289): 763-7, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20360742

ABSTRACT

Abnormalities in functional connectivity between brain areas have been postulated as an important pathophysiological mechanism underlying schizophrenia. In particular, macroscopic measurements of brain activity in patients suggest that functional connectivity between the frontal and temporal lobes may be altered. However, it remains unclear whether such dysconnectivity relates to the aetiology of the illness, and how it is manifested in the activity of neural circuits. Because schizophrenia has a strong genetic component, animal models of genetic risk factors are likely to aid our understanding of the pathogenesis and pathophysiology of the disease. Here we study Df(16)A(+/-) mice, which model a microdeletion on human chromosome 22 (22q11.2) that constitutes one of the largest known genetic risk factors for schizophrenia. To examine functional connectivity in these mice, we measured the synchronization of neural activity between the hippocampus and the prefrontal cortex during the performance of a task requiring working memory, which is one of the cognitive functions disrupted in the disease. In wild-type mice, hippocampal-prefrontal synchrony increased during working memory performance, consistent with previous reports in rats. Df(16)A(+/-) mice, which are impaired in the acquisition of the task, showed drastically reduced synchrony, measured both by phase-locking of prefrontal cells to hippocampal theta oscillations and by coherence of prefrontal and hippocampal local field potentials. Furthermore, the magnitude of hippocampal-prefrontal coherence at the onset of training could be used to predict the time it took the Df(16)A(+/-) mice to learn the task and increased more slowly during task acquisition. These data suggest how the deficits in functional connectivity observed in patients with schizophrenia may be realized at the single-neuron level. Our findings further suggest that impaired long-range synchrony of neural activity is one consequence of the 22q11.2 deletion and may be a fundamental component of the pathophysiology underlying schizophrenia.


Subject(s)
Chromosomes, Mammalian/genetics , Disease Models, Animal , Hippocampus/physiopathology , Prefrontal Cortex/physiopathology , Schizophrenia/genetics , Schizophrenia/physiopathology , Action Potentials/physiology , Alleles , Animals , Behavior, Animal/physiology , Chromosomes, Human, Pair 22/genetics , Female , Genetic Predisposition to Disease/genetics , Humans , Male , Memory/physiology , Mice , Mice, Inbred C57BL , Models, Genetic , Models, Neurological
9.
Neurobiol Dis ; 77: 228-37, 2015 May.
Article in English | MEDLINE | ID: mdl-25771167

ABSTRACT

Variation in gene expression is an important mechanism underlying susceptibility to complex disease and traits. Single nucleotide polymorphisms (SNPs) account for a substantial portion of the total detected genetic variation in gene expression but how exactly variants acting in trans modulate gene expression and disease susceptibility remains largely unknown. The BDNF Val66Met SNP has been associated with a number of psychiatric disorders such as depression, anxiety disorders, schizophrenia and related traits. Using global microRNA expression profiling in hippocampus of humanized BDNF Val66Met knock-in mice we showed that this variant results in dysregulation of at least one microRNA, which in turn affects downstream target genes. Specifically, we show that reduced levels of miR-146b (mir146b), lead to increased Per1 and Npas4 mRNA levels and increased Irak1 protein levels in vitro and are associated with similar changes in the hippocampus of hBDNF(Met/Met) mice. Our findings highlight trans effects of common variants on microRNA-mediated gene expression as an integral part of the genetic architecture of complex disorders and traits.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Gene Expression Regulation/genetics , Methionine/genetics , MicroRNAs/metabolism , Valine/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Hippocampus/metabolism , Humans , In Vitro Techniques , Interleukin-1 Receptor-Associated Kinases/metabolism , Mice , Mice, Transgenic , Molecular Sequence Data , Period Circadian Proteins/metabolism
10.
Nat Rev Neurosci ; 11(6): 402-16, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20485365

ABSTRACT

Recent studies are beginning to paint a clear and consistent picture of the impairments in psychological and cognitive competencies that are associated with microdeletions in chromosome 22q11.2. These studies have highlighted a strong link between this genetic lesion and schizophrenia. Parallel studies in humans and animal models are starting to uncover the complex genetic and neural substrates altered by the microdeletion. In addition to offering a deeper understanding of the effects of this genetic lesion, these findings may guide analysis of other copy-number variants associated with cognitive dysfunction and psychiatric disorders.


Subject(s)
Brain Diseases/etiology , Chromosome Aberrations , Chromosome Deletion , Chromosomes, Human, Pair 22 , Schizophrenia , Animals , Brain Diseases/genetics , Humans , Schizophrenia/complications , Schizophrenia/genetics , Schizophrenia/pathology
11.
Nat Genet ; 38(5): 556-60, 2006 May.
Article in English | MEDLINE | ID: mdl-16582909

ABSTRACT

The genome-wide distribution of linkage disequilibrium (LD) determines the strategy for selecting markers for association studies, but it varies between populations. We assayed LD in large samples (200 individuals) from each of 11 well-described population isolates and an outbred European-derived sample, using SNP markers spaced across chromosome 22. Most isolates show substantially higher levels of LD than the outbred sample and many fewer regions of very low LD (termed 'holes'). Young isolates known to have had relatively few founders show particularly extensive LD with very few holes; these populations offer substantial advantages for genome-wide association mapping.


Subject(s)
Genetics, Population , Genome, Human , Linkage Disequilibrium , Chromosomes, Human, Pair 22 , Humans , Polymorphism, Single Nucleotide
12.
J Neurosci ; 33(37): 14825-39, 2013 Sep 11.
Article in English | MEDLINE | ID: mdl-24027283

ABSTRACT

We used a mouse model of the schizophrenia-predisposing 22q11.2 microdeletion to evaluate how this genetic lesion affects cortical neural circuits at the synaptic, cellular, and molecular levels. Guided by cognitive deficits, we demonstrated that mutant mice display robust deficits in high-frequency synaptic transmission and short-term plasticity (synaptic depression and potentiation), as well as alterations in long-term plasticity and dendritic spine stability. Apart from previously reported reduction in dendritic complexity of layer 5 pyramidal neurons, altered synaptic plasticity occurs in the context of relatively circumscribed and often subtle cytoarchitectural changes in neuronal density and inhibitory neuron numbers. We confirmed the pronounced DiGeorge critical region 8 (Dgcr8)-dependent deficits in primary micro-RNA processing and identified additional changes in gene expression and RNA splicing that may underlie the effects of this mutation. Reduction in Dgcr8 levels appears to be a major driver of altered short-term synaptic plasticity in prefrontal cortex and working memory but not of long-term plasticity and cytoarchitecture. Our findings inform the cortical synaptic and neuronal mechanisms of working memory impairment in the context of psychiatric disorders. They also provide insight into the link between micro-RNA dysregulation and genetic liability to schizophrenia and cognitive dysfunction.


Subject(s)
DiGeorge Syndrome/pathology , Long-Term Potentiation/genetics , Long-Term Synaptic Depression/genetics , Neurons/physiology , Prefrontal Cortex/pathology , Animals , Cognition Disorders/etiology , Cognition Disorders/genetics , Dendritic Spines/pathology , Dendritic Spines/ultrastructure , DiGeorge Syndrome/complications , DiGeorge Syndrome/genetics , Disease Models, Animal , Gene Expression Regulation/genetics , Gene Regulatory Networks/genetics , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Neurons/pathology , Phosphopyruvate Hydratase/metabolism , Proteins/genetics , RNA-Binding Proteins , Recognition, Psychology/physiology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
13.
Annu Rev Med ; 63: 63-80, 2012.
Article in English | MEDLINE | ID: mdl-22034867

ABSTRACT

Although a genetic component of schizophrenia has been acknowledged for a long time, the underlying architecture of the genetic risk remains a contentious issue. Early linkage and candidate association studies led to largely inconclusive results. More recently, the availability of powerful technologies, samples of sufficient sizes, and genome-wide panels of genetic markers facilitated systematic and agnostic scans throughout the genome for either common or rare disease risk variants of small or large effect size, respectively. Although the former had limited success, the role of rare genetic events, such as copy-number variants (CNVs) or rare point mutations, has become increasingly important in gene discovery for schizophrenia. Importantly, recent research building upon earlier findings of de novo recurrent CNVs at the 22q11.2 locus, has highlighted a de novo mutational paradigm as a major component of the genetic architecture of schizophrenia. Recent progress is bringing us closer to earlier intervention and new therapeutic targets.


Subject(s)
Genetic Predisposition to Disease/epidemiology , Genetic Predisposition to Disease/genetics , Genetics/trends , Schizophrenia/epidemiology , Schizophrenia/genetics , Humans , Risk Factors
14.
Proc Natl Acad Sci U S A ; 108(11): 4447-52, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21368174

ABSTRACT

Individuals with 22q11.2 microdeletions have cognitive and behavioral impairments and the highest known genetic risk for developing schizophrenia. One gene disrupted by the 22q11.2 microdeletion is DGCR8, a component of the "microprocessor" complex that is essential for microRNA production, resulting in abnormal processing of specific brain miRNAs and working memory deficits. Here, we determine the effect of Dgcr8 deficiency on the structure and function of cortical circuits by assessing their laminar organization, as well as the neuronal morphology, and intrinsic and synaptic properties of layer 5 pyramidal neurons in the prefrontal cortex of Dgcr8(+/-) mutant mice. We found that heterozygous Dgcr8 mutant mice have slightly fewer cortical layer 2/4 neurons and that the basal dendrites of layer 5 pyramidal neurons have slightly smaller spines. In addition to the modest structural changes, field potential and whole-cell electrophysiological recordings performed in layer 5 of the prefrontal cortex revealed greater short-term synaptic depression during brief stimulation trains applied at 50 Hz to superficial cortical layers. This finding was accompanied by a decrease in the initial phase of synaptic potentiation. Our results identify altered short-term plasticity as a neural substrate underlying the cognitive dysfunction and the increased risk for schizophrenia associated with the 22q11.2 microdeletions.


Subject(s)
Gene Deletion , Neuronal Plasticity/physiology , Prefrontal Cortex/physiopathology , Proteins/metabolism , Animals , CA1 Region, Hippocampal/physiopathology , CA3 Region, Hippocampal/physiopathology , Chromosome Deletion , Chromosomes, Human, Pair 22/genetics , Dendritic Spines/metabolism , Dendritic Spines/pathology , Excitatory Postsynaptic Potentials/physiology , Mice , Prefrontal Cortex/pathology , RNA-Binding Proteins , Synapses/metabolism , Time Factors
15.
Proc Natl Acad Sci U S A ; 108(49): E1349-58, 2011 Dec 06.
Article in English | MEDLINE | ID: mdl-22049344

ABSTRACT

Carefully designed animal models of genetic risk factors are likely to aid our understanding of the pathogenesis of schizophrenia. Here, we study a mouse strain with a truncating lesion in the endogenous Disc1 ortholog designed to model the effects of a schizophrenia-predisposing mutation and offer a detailed account of the consequences that this mutation has on the development and function of a hippocampal circuit. We uncover widespread and cumulative cytoarchitectural alterations in the dentate gyrus during neonatal and adult neurogenesis, which include errors in axonal targeting and are accompanied by changes in short-term plasticity at the mossy fiber/CA3 circuit. We also provide evidence that cAMP levels are elevated as a result of the Disc1 mutation, leading to altered axonal targeting and dendritic growth. The identified structural alterations are, for the most part, not consistent with the growth-promoting and premature maturation effects inferred from previous RNAi-based Disc1 knockdown. Our results provide support to the notion that modest disturbances of neuronal connectivity and accompanying deficits in short-term synaptic dynamics is a general feature of schizophrenia-predisposing mutations.


Subject(s)
Axons/metabolism , Hippocampus/metabolism , Nerve Tissue Proteins/metabolism , Neuronal Plasticity , Action Potentials , Animals , Animals, Newborn , Cell Proliferation , Cells, Cultured , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Dendrites/metabolism , Dendrites/physiology , Dentate Gyrus/cytology , Dentate Gyrus/growth & development , Dentate Gyrus/metabolism , Hippocampus/cytology , Hippocampus/growth & development , Immunohistochemistry , Long-Term Potentiation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mossy Fibers, Hippocampal/metabolism , Nerve Tissue Proteins/genetics , Neurogenesis , Neurons/cytology , Neurons/metabolism , Neurons/physiology , Patch-Clamp Techniques
16.
Nat Genet ; 36(2): 131-7, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14745448

ABSTRACT

AKT-GSK3beta signaling is a target of lithium and as such has been implicated in the pathogenesis of mood disorders. Here, we provide evidence that this signaling pathway also has a role in schizophrenia. Specifically, we present convergent evidence for a decrease in AKT1 protein levels and levels of phosphorylation of GSK3beta at Ser9 in the peripheral lymphocytes and brains of individuals with schizophrenia; a significant association between schizophrenia and an AKT1 haplotype associated with lower AKT1 protein levels; and a greater sensitivity to the sensorimotor gating-disruptive effect of amphetamine, conferred by AKT1 deficiency. Our findings support the proposal that alterations in AKT1-GSK3beta signaling contribute to schizophrenia pathogenesis and identify AKT1 as a potential schizophrenia susceptibility gene. Consistent with this proposal, we also show that haloperidol induces a stepwise increase in regulatory phosphorylation of AKT1 in the brains of treated mice that could compensate for an impaired function of this signaling pathway in schizophrenia.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins , Schizophrenia/enzymology , Signal Transduction/physiology , Antipsychotic Agents/pharmacology , Glycogen Synthase Kinase 3 beta , Haloperidol/pharmacology , Haplotypes , Humans , Phosphorylation/drug effects , Phosphotransferases/drug effects , Phosphotransferases/metabolism , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt , Schizophrenia/genetics , Schizophrenia/metabolism , Serine/metabolism
17.
Nat Genet ; 36(7): 725-31, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15184899

ABSTRACT

Using a relatively dense genetic map of 72 single-nucleotide polymorphisms (SNPs) distributed across the entire 1.5-Mb locus on chromosome 22q11 associated with susceptibilit to schizophrenia, we previously identified two subregions that were consistently associated with the disease. In the distal subregion, we detected an association signal with five neighboring SNPs distributed over a haplotypic block of 80 kb encompassing six known genes. One of these five SNPs, rs175174, had the strongest association of all 72 SNPs that we tested. Here we show that rs175174 regulates the level of the fully functional transcript by modulating the retention of intron 4 of the gene ZDHHC8, which encodes a putative transmembrane palmitoyltransferase. Zdhhc8-knockout mice had a sexually dimorphic deficit in prepulse inhibition, a gene dosage-dependent decrease in exploratory activity in a new environment and a decreased sensitivity to the locomotor stimulatory effects of the psychomimetic drug dizocilpine (MK801). SNP rs175174 shows differences in transmission distortion between sexes in individuals with schizophrenia. Our results indicate that there is an unexpected connection between impaired palmitate modification of neuronal proteins and the psychiatric phenotypes associated with microdeletions of chromosome 22q11.


Subject(s)
Acyltransferases/genetics , Genetic Predisposition to Disease , Membrane Proteins/genetics , Schizophrenia/genetics , Acyltransferases/chemistry , Alternative Splicing , Amino Acid Sequence , Animals , Base Sequence , DNA , Dizocilpine Maleate/administration & dosage , Excitatory Amino Acid Antagonists/administration & dosage , Exons , Membrane Proteins/chemistry , Mice , Mice, Knockout , Molecular Sequence Data , Sequence Homology, Amino Acid
18.
Neurobiol Dis ; 46(2): 291-301, 2012 May.
Article in English | MEDLINE | ID: mdl-22406400

ABSTRACT

MicroRNAs (miRNA), a class of non-coding RNAs, are emerging as important modulators of neuronal development, structure and function. A connection has been established between abnormalities in miRNA expression and miRNA-mediated gene regulation and psychiatric and neurodevelopmental disorders as well as cognitive dysfunction. Establishment of this connection has been driven by progress in elucidating the genetic etiology of these phenotypes and has provided a context to interpret additional supporting evidence accumulating from parallel expression profiling studies in brains and peripheral blood of patients. Here we review relevant evidence that supports this connection and explore possible mechanisms that underlie the contribution of individual miRNAs and miRNA-related pathways to the pathogenesis and pathophysiology of these complex clinical phenotypes. The existing evidence provides useful hypotheses for further investigation as well as important clues for identifying novel therapeutic targets.


Subject(s)
Cognition Disorders/genetics , Gene Expression Regulation , Mental Disorders/genetics , MicroRNAs/physiology , Animals , Brain/pathology , Brain/physiology , Cognition Disorders/pathology , Cognition Disorders/physiopathology , Humans , Mental Disorders/pathology , Mental Disorders/physiopathology , Neuronal Plasticity/physiology , Signal Transduction/physiology
19.
Int J Neuropsychopharmacol ; 15(9): 1331-42, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22074909

ABSTRACT

Catechol-O-methyltransferase (COMT) is an important enzyme in the metabolism of dopamine and disturbance in dopamine function is proposed to be central to the pathogenesis of schizophrenia. Clinical epidemiological studies have indicated cannabis use to confer a 2-fold increase in risk for subsequent onset of psychosis, with adolescent-onset use conveying even higher risk. There is evidence that a high activity COMT polymorphism moderates the effects of adolescent exposure to cannabis on risk for adult psychosis. In this paper we compared the effect of chronic adolescent exposure to the cannabinoid WIN 55212 on sensorimotor gating, behaviours related to the negative symptoms of schizophrenia, anxiety- and stress-related behaviours, as well as ex-vivo brain dopamine and serotonin levels, in COMT KO vs. wild-type (WT) mice. Additionally, we examined the effect of pretreatment with the COMT inhibitor tolcapone on acute effects of this cannabinoid on sensorimotor gating in C57BL/6 mice. COMT KO mice were shown to be more vulnerable than WT to the disruptive effects of adolescent cannabinoid treatment on prepulse inhibition (PPI). Acute pharmacological inhibition of COMT in C57BL/6 mice also modified acute cannabinoid effects on startle reactivity, as well as PPI, indicating that chronic and acute loss of COMT can produce dissociable effects on the behavioural effects of cannabinoids. COMT KO mice also demonstrated differential effects of adolescent cannabinoid administration on sociability and anxiety-related behaviour, both confirming and extending earlier reports of COMT×cannabinoid effects on the expression of schizophrenia-related endophenotypes.


Subject(s)
Cannabinoids/pharmacology , Catechol O-Methyltransferase Inhibitors , Catechol O-Methyltransferase/genetics , Schizophrenia/genetics , Schizophrenic Psychology , Alleles , Animals , Anxiety/psychology , Benzophenones/pharmacology , Benzoxazines/pharmacology , Biogenic Monoamines/metabolism , Cannabinoid Receptor Agonists/pharmacology , Chromatography, High Pressure Liquid , Cyclohexanols/pharmacology , Enzyme Inhibitors/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Morpholines/pharmacology , Naphthalenes/pharmacology , Nitrophenols/pharmacology , Pain Measurement/drug effects , Phenotype , Reflex, Startle/drug effects , Reflex, Startle/genetics , Schizophrenia/enzymology , Social Behavior , Swimming/psychology , Tolcapone
20.
Mol Cell Neurosci ; 47(4): 293-305, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21635953

ABSTRACT

22q11.2 chromosomal deletions are recurrent copy number mutations that increase the risk of schizophrenia around thirty-fold. Deletion of the orthologous chromosomal region in mice offers an opportunity to characterize changes to neuronal structure and function that may account for the development of this disease. The hippocampus has been implicated in schizophrenia pathogenesis, is reduced in volume in 22q11.2 deletion carriers and displays altered neuronal structure in a mouse model of the mutation (Df(16)A(+/-) mice). Here we investigate hippocampal CA1 physiology, hippocampal-dependent spatial memory and novelty-induced hippocampal activation in Df(16)A(+/-) mice. We found normal spatial reference memory (as assayed by the Morris water maze test) as well as modest but potentially important deficits in physiology. In particular, a reduction in the level of inhibition of CA1 pyramidal neurons was observed, implying a decrease in interneuron activity. Additionally, deficits in LTP were observed using certain induction protocols. Induction of c-Fos expression by exploration of a novel environment suggested a relative sparing of CA1 and dentate gyrus function but showed a robust decrease in the number of activated CA3 pyramidal neurons in Df(16)A(+/-) mice. Overall, experiments performed in this 22q11.2 deletion model demonstrated deficits of various degrees across different regions of the hippocampus, which together may contribute to the increased risk of developing schizophrenia.


Subject(s)
Chromosome Deletion , Hippocampus/physiology , Models, Animal , Action Potentials/physiology , Animals , Chromosomes, Human, Pair 22 , Humans , Interneurons/metabolism , Maze Learning/physiology , Memory/physiology , Mice , Mice, Inbred C57BL , Neuronal Plasticity/physiology , Neurons/cytology , Neurons/physiology , Patch-Clamp Techniques , Proto-Oncogene Proteins c-fos/metabolism , Risk Factors , Schizophrenia/genetics
SELECTION OF CITATIONS
SEARCH DETAIL