Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
2.
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.

3.
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
4.
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
5.
Nat Biotechnol ; 35(2): 154-163, 2017 02.
Article in English | MEDLINE | ID: mdl-28112759

ABSTRACT

Considerable progress has been made in converting human pluripotent stem cells (hPSCs) into functional neurons. However, the protracted timing of human neuron specification and functional maturation remains a key challenge that hampers the routine application of hPSC-derived lineages in disease modeling and regenerative medicine. Using a combinatorial small-molecule screen, we previously identified conditions to rapidly differentiate hPSCs into peripheral sensory neurons. Here we generalize the approach to central nervous system (CNS) fates by developing a small-molecule approach for accelerated induction of early-born cortical neurons. Combinatorial application of six pathway inhibitors induces post-mitotic cortical neurons with functional electrophysiological properties by day 16 of differentiation, in the absence of glial cell co-culture. The resulting neurons, transplanted at 8 d of differentiation into the postnatal mouse cortex, are functional and establish long-distance projections, as shown using iDISCO whole-brain imaging. Accelerated differentiation into cortical neuron fates should facilitate hPSC-based strategies for disease modeling and cell therapy in CNS disorders.


Subject(s)
Cell Differentiation/physiology , Central Nervous System Agents/administration & dosage , Neurons/cytology , Neurons/physiology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/physiology , Batch Cell Culture Techniques/methods , Cell Differentiation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Humans , Neurogenesis/drug effects , Neurogenesis/physiology , Neurons/drug effects , Pluripotent Stem Cells/drug effects
6.
Neuron ; 89(5): 940-7, 2016 Mar 02.
Article in English | MEDLINE | ID: mdl-26938441

ABSTRACT

We analyze de novo synonymous mutations identified in autism spectrum disorders (ASDs) and schizophrenia (SCZ) with potential impact on regulatory elements using data from whole-exome sequencing (WESs) studies. Focusing on five types of genetic regulatory functions, we found that de novo near-splice site synonymous mutations changing exonic splicing regulators and those within frontal cortex-derived DNase I hypersensitivity sites are significantly enriched in ASD and SCZ, respectively. These results remained significant, albeit less so, after incorporating two additional ASD datasets. Among the genes identified, several are hit by multiple functional de novo mutations, with RAB2A and SETD1A showing the highest statistical significance in ASD and SCZ, respectively. The estimated contribution of these synonymous mutations to disease liability is comparable to de novo protein-truncating mutations. These findings expand the repertoire of functional de novo mutations to include "functional" synonymous ones and strengthen the role of rare variants in neuropsychiatric disease risk.


Subject(s)
Autistic Disorder/genetics , Genetic Predisposition to Disease/genetics , Histone-Lysine N-Methyltransferase/genetics , Mutation/genetics , Regulatory Elements, Transcriptional/genetics , Schizophrenia/genetics , rab2 GTP-Binding Protein/genetics , Databases, Factual/statistics & numerical data , Exome , Female , Genetic Association Studies , Humans , Machine Learning , Male , MicroRNAs/genetics , Nerve Tissue Proteins/genetics , Sequence Analysis, DNA
7.
Chem Biol ; 22(6): 793-802, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-26091172

ABSTRACT

Processing of microRNA primary transcripts (pri-miRNAs) is highly regulated and defects in the processing machinery play a key role in many human diseases. In 22q11.2 deletion syndrome (22q11.2DS), heterozygous deletion of DiGeorge critical region gene 8 (DGCR8) causes a processing deficiency, which contributes to abnormal brain development. The DGCR8 protein is the RNA-binding partner of Drosha RNase, both essential for processing canonical pri-miRNAs. To identify an agent that can compensate reduced DGCR8 expression, we screened for metalloporphyrins that can mimic the natural DGCR8 heme cofactor. We found that Co(III) protoporphyrin IX (PPIX) stably binds DGCR8 and activates it for pri-miRNA processing in vitro and in HeLa cells. Importantly, treating cultured Dgcr8(+/-) mouse neurons with Co(III)PPIX can compensate the pri-miRNA processing defects. Co(III)PPIX is effective at concentrations as low as 0.2 µM and is not degraded by heme degradation enzymes, making it useful as a research tool and a potential therapeutic.


Subject(s)
Cobalt/chemistry , Coordination Complexes/chemistry , MicroRNAs/metabolism , Protoporphyrins/chemistry , RNA-Binding Proteins/metabolism , Animals , Cells, Cultured , Coordination Complexes/metabolism , Coordination Complexes/toxicity , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Protein Binding , RNA Processing, Post-Transcriptional/drug effects , RNA-Binding Proteins/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification
8.
Neuron ; 86(3): 680-95, 2015 May 06.
Article in English | MEDLINE | ID: mdl-25913858

ABSTRACT

22q11.2 deletion carriers show specific cognitive deficits, and ∼30% of them develop schizophrenia. One of the disrupted genes is ZDHHC8, which encodes for a palmitoyltransferase. We show that Zdhhc8-deficient mice have reduced palmitoylation of proteins that regulate axonal growth and branching. Analysis of axonal projections of pyramidal neurons from both Zdhhc8-deficient and Df(16)A(+/-) mice, which model the 22q11.2 deletion, revealed deficits in axonal growth and terminal arborization, which can be prevented by reintroduction of active ZDHHC8 protein. Impaired terminal arborization is accompanied by a reduction in the strength of synaptic connections and altered functional connectivity and working memory. The effect of ZDHHC8 is mediated in part via Cdc42-dependent modulation of Akt/Gsk3ß signaling at the tip of the axon and can be reversed by pharmacologically decreasing Gsk3ß activity during postnatal brain development. Our findings provide valuable mechanistic insights into the cognitive and psychiatric symptoms associated with a schizophrenia-predisposing mutation.


Subject(s)
Alzheimer Disease/pathology , Axons/pathology , Brain/pathology , DiGeorge Syndrome/pathology , Acyltransferases/deficiency , Acyltransferases/genetics , Age Factors , Alzheimer Disease/genetics , Animals , Animals, Newborn , Brain/embryology , Brain/metabolism , Channelrhodopsins , DiGeorge Syndrome/genetics , Disease Models, Animal , Embryo, Mammalian , Excitatory Postsynaptic Potentials/genetics , Gene Expression Regulation, Developmental/genetics , Green Fluorescent Proteins , Humans , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Transgenic , Neural Pathways/embryology , Neural Pathways/growth & development , Neural Pathways/physiology , Neurons/pathology , Neurons/ultrastructure , Phosphopyruvate Hydratase/metabolism , Signal Transduction/genetics , Synapsins/metabolism
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.
Neuron ; 82(4): 773-80, 2014 May 21.
Article in English | MEDLINE | ID: mdl-24853937

ABSTRACT

Loss-of-function (LOF) (i.e., nonsense, splice site, and frameshift) variants that lead to disruption of gene function are likely to contribute to the etiology of neuropsychiatric disorders. Here, we perform a systematic investigation of the role of both de novo and inherited LOF variants in schizophrenia using exome sequencing data from 231 case and 34 control trios. We identify two de novo LOF variants in the SETD1A gene, which encodes a subunit of histone methyltransferase, a finding unlikely to have occurred by chance, and provide evidence for a more general role of chromatin regulators in schizophrenia risk. Transmission pattern analyses reveal that LOF variants are more likely to be transmitted to affected individuals than controls. This is especially true for private LOF variants in genes intolerant to functional genetic variation. These findings highlight the contribution of LOF mutations to the genetic architecture of schizophrenia and provide important insights into disease pathogenesis.


Subject(s)
Genetic Predisposition to Disease/genetics , Genetic Variation/genetics , Histone-Lysine N-Methyltransferase/genetics , Schizophrenia/genetics , Chromatin Assembly and Disassembly , Cohort Studies , Female , Gene Frequency , Gene Regulatory Networks , Genome, Human , Genotype , Humans , Likelihood Functions , Male , Risk Factors , Sequence Analysis, DNA
11.
Neuropsychopharmacology ; 39(4): 934-43, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24141571

ABSTRACT

We previously reported linkage of schizophrenia and schizoaffective disorder to 13q32-34 in the European descent Afrikaner population from South Africa. The nature of genetic variation underlying linkage peaks in psychiatric disorders remains largely unknown and both rare and common variants may be contributing. Here, we examine the contribution of common variants located under the 13q32-34 linkage region. We used densely spaced SNPs to fine map the linkage peak region using both a discovery sample of 415 families and a meta-analysis incorporating two additional replication family samples. In a second phase of the study, we use one family-based data set with 237 families and independent case-control data sets for fine mapping of the common variant association signal using HapMap SNPs. We report a significant association with a genetic variant (rs9583277) within the gene encoding for the myosin heavy-chain Myr 8 (MYO16), which has been implicated in neuronal phosphoinositide 3-kinase signaling. Follow-up analysis of HapMap variation within MYO16 in a second set of Afrikaner families and additional case-control data sets of European descent highlighted a region across introns 2-6 as the most likely region to harbor common MYO16 risk variants. Expression analysis revealed a significant increase in the level of MYO16 expression in the brains of schizophrenia patients. Our results suggest that common variation within MYO16 may contribute to the genetic liability to schizophrenia.


Subject(s)
Brain/pathology , Chromosomes, Human, Pair 13/genetics , Genetic Predisposition to Disease/genetics , Myosin Heavy Chains/genetics , Schizophrenia/genetics , Schizophrenia/pathology , Chromosome Mapping , Female , Genetic Linkage , Genetic Variation/genetics , Genotype , Humans , Male , South Africa , White People
12.
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
13.
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
14.
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
15.
Neurobiol Aging ; 34(4): 1309.e9-10, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23036583

ABSTRACT

A hexanucleotide repeat expansion in C9ORF72 was recently found to cause some cases of frontotemporal lobar degeneration, frontotemporal dementia (FTD)-amyotrophic lateral sclerosis, and amyotrophic lateral sclerosis. Patients with frontotemporal lobar degeneration with the C9ORF72 repeat expansion are more likely than those without to present with psychosis. In this study, we screened DNA samples from 192 unrelated subjects with schizophrenia for the C9ORF72 repeat expansion. None of the subjects with schizophrenia had the pathogenic expansion. C9ORF72 repeat expansions either do not cause schizophrenia, or do so rarely (less than 1% of cases).


Subject(s)
DNA Repeat Expansion/genetics , Genetic Predisposition to Disease/epidemiology , Genetic Predisposition to Disease/genetics , Polymorphism, Single Nucleotide/genetics , Proteins/genetics , Schizophrenia/epidemiology , Schizophrenia/genetics , C9orf72 Protein , Causality , Genetic Markers/genetics , Humans , Incidence , Risk Factors , United States/epidemiology
16.
Nat Neurosci ; 15(12): 1723-8, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23143521

ABSTRACT

Despite the successful identification of several relevant genomic loci, the underlying molecular mechanisms of schizophrenia remain largely unclear. We developed a computational approach (NETBAG+) that allows an integrated analysis of diverse disease-related genetic data using a unified statistical framework. The application of this approach to schizophrenia-associated genetic variations, obtained using unbiased whole-genome methods, allowed us to identify several cohesive gene networks related to axon guidance, neuronal cell mobility, synaptic function and chromosomal remodeling. The genes forming the networks are highly expressed in the brain, with higher brain expression during prenatal development. The identified networks are functionally related to genes previously implicated in schizophrenia, autism and intellectual disability. A comparative analysis of copy number variants associated with autism and schizophrenia suggests that although the molecular networks implicated in these distinct disorders may be related, the mutations associated with each disease are likely to lead, at least on average, to different functional consequences.


Subject(s)
Gene Regulatory Networks/genetics , Genetic Variation/genetics , Multigene Family/genetics , Phenotype , Schizophrenia/genetics , Autistic Disorder/diagnosis , Autistic Disorder/epidemiology , Autistic Disorder/genetics , Cluster Analysis , Gene Expression Regulation, Developmental , Genetic Predisposition to Disease/epidemiology , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Humans , Intellectual Disability/diagnosis , Intellectual Disability/epidemiology , Intellectual Disability/genetics , Protein Interaction Domains and Motifs/genetics , Schizophrenia/diagnosis , Schizophrenia/epidemiology
17.
Nat Genet ; 44(12): 1365-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23042115

ABSTRACT

To evaluate evidence for de novo etiologies in schizophrenia, we sequenced at high coverage the exomes of families recruited from two populations with distinct demographic structures and history. We sequenced a total of 795 exomes from 231 parent-proband trios enriched for sporadic schizophrenia cases, as well as 34 unaffected trios. We observed in cases an excess of de novo nonsynonymous single-nucleotide variants as well as a higher prevalence of gene-disruptive de novo mutations relative to controls. We found four genes (LAMA2, DPYD, TRRAP and VPS39) affected by recurrent de novo events within or across the two populations, which is unlikely to have occurred by chance. We show that de novo mutations affect genes with diverse functions and developmental profiles, but we also find a substantial contribution of mutations in genes with higher expression in early fetal life. Our results help define the genomic and neural architecture of schizophrenia.


Subject(s)
Mutation , Neurons/physiology , Schizophrenia/genetics , Adaptor Proteins, Signal Transducing/genetics , Adult , Autophagy-Related Proteins , Base Sequence , Brain/growth & development , Cohort Studies , Dihydrouracil Dehydrogenase (NADP)/genetics , Exome/genetics , Female , Gene Expression Regulation, Developmental/genetics , Genetic Predisposition to Disease , Humans , Intracellular Signaling Peptides and Proteins/genetics , Laminin/genetics , Male , Molecular Sequence Data , Nuclear Proteins/genetics , Polymorphism, Single Nucleotide , Sequence Analysis, DNA , South Africa , United States , Vesicular Transport Proteins/genetics , Young Adult
19.
Neuropsychopharmacology ; 37(7): 1773-83, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22434221

ABSTRACT

Cannabis use confers a two-fold increase in risk for psychosis, with adolescent use conferring an even greater risk. A high-low activity polymorphism in catechol-O-methyltransferase (COMT), a gene encoding the COMT enzyme involved in dopamine clearance in the brain, may interact with adolescent cannabis exposure to increase risk for schizophrenia. The impact of such an interaction on central neurotransmitter pathways implicated in schizophrenia is unknown. Male mice with knockout of the COMT gene were treated chronically with delta-9-tetrahydrocannabinol (THC) during adolescence (postnatal day 32-52). We measured the size and density of GABAergic cells and the protein expression of cannabinoid receptor 1 (CB1R) in the prefrontal cortex (PFC) and hippocampus (HPC) in knockout mice relative to heterozygous mutants and wild-type controls. Size and density of dopaminergic neurons was also assessed in the ventral tegmental area (VTA) across the genotypes. COMT genotype × THC treatment interactions were observed for: (1) dopaminergic cell size in the VTA, (2) CB1R protein expression in the HPC, and (3) parvalbumin (PV) cell size in the PFC. No effects of adolescent THC treatment were observed for PV and dopaminergic cell density across the COMT genotypes. COMT genotype modulates the effects of chronic THC administration during adolescence on indices of neurotransmitter function in the brain. These findings illuminate how COMT deletion and adolescent cannabis use can interact to modulate the function of neurotransmitters systems implicated in schizophrenia.


Subject(s)
Brain/drug effects , Cannabinoid Receptor Modulators/metabolism , Catechol O-Methyltransferase/genetics , Dopaminergic Neurons/drug effects , Dronabinol/pharmacology , Endocannabinoids , GABAergic Neurons/drug effects , Animals , Brain/metabolism , Catechol O-Methyltransferase/metabolism , Cell Count , Dopamine/metabolism , Male , Mice , Mice, Knockout , Tyrosine 3-Monooxygenase/metabolism , gamma-Aminobutyric Acid/metabolism
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...