Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 179(3): 750-771.e22, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31626773

ABSTRACT

Tissue-specific regulatory regions harbor substantial genetic risk for disease. Because brain development is a critical epoch for neuropsychiatric disease susceptibility, we characterized the genetic control of the transcriptome in 201 mid-gestational human brains, identifying 7,962 expression quantitative trait loci (eQTL) and 4,635 spliceQTL (sQTL), including several thousand prenatal-specific regulatory regions. We show that significant genetic liability for neuropsychiatric disease lies within prenatal eQTL and sQTL. Integration of eQTL and sQTL with genome-wide association studies (GWAS) via transcriptome-wide association identified dozens of novel candidate risk genes, highlighting shared and stage-specific mechanisms in schizophrenia (SCZ). Gene network analysis revealed that SCZ and autism spectrum disorder (ASD) affect distinct developmental gene co-expression modules. Yet, in each disorder, common and rare genetic variation converges within modules, which in ASD implicates superficial cortical neurons. More broadly, these data, available as a web browser and our analyses, demonstrate the genetic mechanisms by which developmental events have a widespread influence on adult anatomical and behavioral phenotypes.


Subject(s)
Autism Spectrum Disorder/genetics , Quantitative Trait Loci/genetics , Schizophrenia/genetics , Transcriptome/genetics , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/pathology , Brain/growth & development , Brain/metabolism , Female , Fetus/metabolism , Gene Expression Regulation, Developmental , Genetic Predisposition to Disease , Genome-Wide Association Study , Gestational Age , Humans , Male , Neurons/metabolism , Polymorphism, Single Nucleotide/genetics , RNA Splicing/genetics , Schizophrenia/metabolism , Schizophrenia/pathology
2.
Cell ; 178(4): 850-866.e26, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31398340

ABSTRACT

We performed a comprehensive assessment of rare inherited variation in autism spectrum disorder (ASD) by analyzing whole-genome sequences of 2,308 individuals from families with multiple affected children. We implicate 69 genes in ASD risk, including 24 passing genome-wide Bonferroni correction and 16 new ASD risk genes, most supported by rare inherited variants, a substantial extension of previous findings. Biological pathways enriched for genes harboring inherited variants represent cytoskeletal organization and ion transport, which are distinct from pathways implicated in previous studies. Nevertheless, the de novo and inherited genes contribute to a common protein-protein interaction network. We also identified structural variants (SVs) affecting non-coding regions, implicating recurrent deletions in the promoters of DLG2 and NR3C2. Loss of nr3c2 function in zebrafish disrupts sleep and social function, overlapping with human ASD-related phenotypes. These data support the utility of studying multiplex families in ASD and are available through the Hartwell Autism Research and Technology portal.


Subject(s)
Autism Spectrum Disorder/genetics , Genetic Predisposition to Disease/genetics , Pedigree , Protein Interaction Maps/genetics , Animals , Child , Databases, Genetic , Disease Models, Animal , Female , Gene Deletion , Guanylate Kinases/genetics , Humans , Inheritance Patterns/genetics , Machine Learning , Male , Nuclear Family , Promoter Regions, Genetic/genetics , Receptors, Mineralocorticoid/genetics , Risk Factors , Tumor Suppressor Proteins/genetics , Whole Genome Sequencing , Zebrafish/genetics
3.
Cell ; 177(6): 1600-1618.e17, 2019 05 30.
Article in English | MEDLINE | ID: mdl-31150625

ABSTRACT

Autism spectrum disorder (ASD) manifests as alterations in complex human behaviors including social communication and stereotypies. In addition to genetic risks, the gut microbiome differs between typically developing (TD) and ASD individuals, though it remains unclear whether the microbiome contributes to symptoms. We transplanted gut microbiota from human donors with ASD or TD controls into germ-free mice and reveal that colonization with ASD microbiota is sufficient to induce hallmark autistic behaviors. The brains of mice colonized with ASD microbiota display alternative splicing of ASD-relevant genes. Microbiome and metabolome profiles of mice harboring human microbiota predict that specific bacterial taxa and their metabolites modulate ASD behaviors. Indeed, treatment of an ASD mouse model with candidate microbial metabolites improves behavioral abnormalities and modulates neuronal excitability in the brain. We propose that the gut microbiota regulates behaviors in mice via production of neuroactive metabolites, suggesting that gut-brain connections contribute to the pathophysiology of ASD.


Subject(s)
Autism Spectrum Disorder/microbiology , Behavioral Symptoms/microbiology , Gastrointestinal Microbiome/physiology , Animals , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/physiopathology , Bacteria , Behavior, Animal/physiology , Brain/metabolism , Disease Models, Animal , Humans , Mice , Microbiota , Risk Factors
6.
Nature ; 611(7936): 532-539, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36323788

ABSTRACT

Neuropsychiatric disorders classically lack defining brain pathologies, but recent work has demonstrated dysregulation at the molecular level, characterized by transcriptomic and epigenetic alterations1-3. In autism spectrum disorder (ASD), this molecular pathology involves the upregulation of microglial, astrocyte and neural-immune genes, the downregulation of synaptic genes, and attenuation of gene-expression gradients in cortex1,2,4-6. However, whether these changes are limited to cortical association regions or are more widespread remains unknown. To address this issue, we performed RNA-sequencing analysis of 725 brain samples spanning 11 cortical areas from 112 post-mortem samples from individuals with ASD and neurotypical controls. We find widespread transcriptomic changes across the cortex in ASD, exhibiting an anterior-to-posterior gradient, with the greatest differences in primary visual cortex, coincident with an attenuation of the typical transcriptomic differences between cortical regions. Single-nucleus RNA-sequencing and methylation profiling demonstrate that this robust molecular signature reflects changes in cell-type-specific gene expression, particularly affecting excitatory neurons and glia. Both rare and common ASD-associated genetic variation converge within a downregulated co-expression module involving synaptic signalling, and common variation alone is enriched within a module of upregulated protein chaperone genes. These results highlight widespread molecular changes across the cerebral cortex in ASD, extending beyond association cortex to broadly involve primary sensory regions.


Subject(s)
Autism Spectrum Disorder , Cerebral Cortex , Genetic Variation , Transcriptome , Humans , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/pathology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Neurons/metabolism , RNA/analysis , RNA/genetics , Transcriptome/genetics , Autopsy , Sequence Analysis, RNA , Primary Visual Cortex/metabolism , Neuroglia/metabolism
7.
Proc Natl Acad Sci U S A ; 120(10): e2206758120, 2023 03 07.
Article in English | MEDLINE | ID: mdl-36862688

ABSTRACT

Autism spectrum disorder (ASD) is a highly heterogeneous disorder, yet transcriptomic profiling of bulk brain tissue has identified substantial convergence among dysregulated genes and pathways in ASD. However, this approach lacks cell-specific resolution. We performed comprehensive transcriptomic analyses on bulk tissue and laser-capture microdissected (LCM) neurons from 59 postmortem human brains (27 ASD and 32 controls) in the superior temporal gyrus (STG) of individuals ranging from 2 to 73 years of age. In bulk tissue, synaptic signaling, heat shock protein-related pathways, and RNA splicing were significantly altered in ASD. There was age-dependent dysregulation of genes involved in gamma aminobutyric acid (GABA) (GAD1 and GAD2) and glutamate (SLC38A1) signaling pathways. In LCM neurons, AP-1-mediated neuroinflammation and insulin/IGF-1 signaling pathways were upregulated in ASD, while mitochondrial function, ribosome, and spliceosome components were downregulated. GABA synthesizing enzymes GAD1 and GAD2 were both downregulated in ASD neurons. Mechanistic modeling suggested a direct link between inflammation and ASD in neurons, and prioritized inflammation-associated genes for future study. Alterations in small nucleolar RNAs (snoRNAs) associated with splicing events suggested interplay between snoRNA dysregulation and splicing disruption in neurons of individuals with ASD. Our findings supported the fundamental hypothesis of altered neuronal communication in ASD, demonstrated that inflammation was elevated at least in part in ASD neurons, and may reveal windows of opportunity for biotherapeutics to target the trajectory of gene expression and clinical manifestation of ASD throughout the human lifespan.


Subject(s)
Autism Spectrum Disorder , Transcriptome , Humans , Neuroinflammatory Diseases , Autism Spectrum Disorder/genetics , Inflammation/genetics , Neurons , Glutamic Acid
8.
Bioinformatics ; 39(1)2023 01 01.
Article in English | MEDLINE | ID: mdl-36495218

ABSTRACT

SUMMARY: With the continued deluge of results from genome-wide association and functional genomic studies, it has become increasingly imperative to quickly combine and visualize different layers of genetic and genomic data within a given locus to facilitate exploratory and integrative data analyses. While several tools have been developed to visualize locus-level genetic results, the limited speed, scalability and flexibility of current approaches remain a significant bottleneck. Here, we present a Julia package for high-performance genetics and genomics-related data visualization that enables fast, simultaneous plotting of hundreds of association results along with multiple relevant genomic annotations. Leveraging the powerful plotting and layout utilities from Makie.jl facilitates the customization and extensibility of every component of a plot, enabling generation of publication-ready figures. AVAILABILITY AND IMPLEMENTATION: The GeneticsMakie.jl package is open source and distributed under the MIT license via GitHub (https://github.com/mmkim1210/GeneticsMakie.jl). The GitHub repository contains installation instructions as well as examples and documentation for built-in functions. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Genome-Wide Association Study , Software , Genome-Wide Association Study/methods , Genomics/methods , Genome , Data Analysis
9.
Nature ; 560(7718): E30, 2018 08.
Article in English | MEDLINE | ID: mdl-29995847

ABSTRACT

Change history: In this Letter, the labels for splicing events A3SS and A5SS were swapped in column D of Supplementary Table 3a and b. This has been corrected online.

10.
J Neurosci ; 42(8): 1587-1603, 2022 02 23.
Article in English | MEDLINE | ID: mdl-34987109

ABSTRACT

Astrocytes are critical for the development and function of synapses. There are notable species differences between human astrocytes and commonly used animal models. Yet, it is unclear whether astrocytic genes involved in synaptic function are stable or exhibit dynamic changes associated with disease states and age in humans, which is a barrier in understanding human astrocyte biology and its potential involvement in neurologic diseases. To better understand the properties of human astrocytes, we acutely purified astrocytes from the cerebral cortices of over 40 humans across various ages, sexes, and disease states. We performed RNA sequencing to generate transcriptomic profiles of these astrocytes and identified genes associated with these biological variables. We found that human astrocytes in tumor-surrounding regions downregulate genes involved in synaptic function and sensing of signals in the microenvironment, suggesting involvement of peritumor astrocytes in tumor-associated neural circuit dysfunction. In aging, we also found downregulation of synaptic regulators and upregulation of markers of cytokine signaling, while in maturation we identified changes in ionic transport with implications for calcium signaling. In addition, we identified subtle sexual dimorphism in human cortical astrocytes, which has implications for observed sex differences across many neurologic disorders. Overall, genes involved in synaptic function exhibit dynamic changes in the peritumor microenvironment and aging. These data provide powerful new insights into human astrocyte biology in several biologically relevant states that will aid in generating novel testable hypotheses about homeostatic and reactive astrocytes in humans.SIGNIFICANCE STATEMENT Astrocytes are an abundant class of cells playing integral roles at synapses. Astrocyte dysfunction is implicated in a variety of human neurologic diseases. Yet our knowledge of astrocytes is largely based on mouse studies. Direct knowledge of human astrocyte biology remains limited. Here, we present transcriptomic profiles of human cortical astrocytes, and we identified molecular differences associated with age, sex, and disease state. We found that peritumor and aging astrocytes downregulate genes involved in astrocyte-synapse interactions. These data provide necessary insight into human astrocyte biology that will improve our understanding of human disease.


Subject(s)
Astrocytes , Transcriptome , Aging/pathology , Animals , Astrocytes/physiology , Female , Humans , Male , Mice , Synapses/physiology , Tumor Microenvironment
11.
Am J Hum Genet ; 106(1): 71-91, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31901249

ABSTRACT

Gene-environment interactions (GxE) can be fundamental in applications ranging from functional genomics to precision medicine and is a conjectured source of substantial heritability. However, unbiased methods to profile GxE genome-wide are nascent and, as we show, cannot accommodate general environment variables, modest sample sizes, heterogeneous noise, and binary traits. To address this gap, we propose a simple, unifying mixed model for gene-environment interaction (GxEMM). In simulations and theory, we show that GxEMM can dramatically improve estimates and eliminate false positives when the assumptions of existing methods fail. We apply GxEMM to a range of human and model organism datasets and find broad evidence of context-specific genetic effects, including GxSex, GxAdversity, and GxDisease interactions across thousands of clinical and molecular phenotypes. Overall, GxEMM is broadly applicable for testing and quantifying polygenic interactions, which can be useful for explaining heritability and invaluable for determining biologically relevant environments.


Subject(s)
Gene-Environment Interaction , Genetic Markers , Mental Disorders/genetics , Mental Disorders/pathology , Models, Genetic , Multifactorial Inheritance/genetics , Adult , Animals , Computer Simulation , Female , Genome-Wide Association Study , Humans , Male , Middle Aged , Phenomics , Phenotype , Rats
12.
Hum Brain Mapp ; 44(2): 535-548, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36177528

ABSTRACT

Sex differences in white matter microstructure have been robustly demonstrated in the adult brain using both conventional and advanced diffusion-weighted magnetic resonance imaging approaches. However, sex differences in white matter microstructure prior to adulthood remain poorly understood; previous developmental work focused on conventional microstructure metrics and yielded mixed results. Here, we rigorously characterized sex differences in white matter microstructure among over 6000 children from the Adolescent Brain Cognitive Development study who were between 9 and 10 years old. Microstructure was quantified using both the conventional model-diffusion tensor imaging (DTI)-and an advanced model, restriction spectrum imaging (RSI). DTI metrics included fractional anisotropy (FA) and mean, axial, and radial diffusivity (MD, AD, RD). RSI metrics included normalized isotropic, directional, and total intracellular diffusion (N0, ND, NT). We found significant and replicable sex differences in DTI or RSI microstructure metrics in every white matter region examined across the brain. Sex differences in FA were regionally specific. Across white matter regions, boys exhibited greater MD, AD, and RD than girls, on average. Girls displayed increased N0, ND, and NT compared to boys, on average, suggesting greater cell and neurite density in girls. Together, these robust and replicable findings provide an important foundation for understanding sex differences in health and disease.


Subject(s)
White Matter , Adult , Adolescent , Humans , Child , Male , Female , White Matter/pathology , Diffusion Tensor Imaging/methods , Sex Characteristics , Brain/pathology , Diffusion Magnetic Resonance Imaging , Anisotropy
13.
J Neurosci ; 41(48): 9971-9987, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34607967

ABSTRACT

Human epidemiological studies implicate exposure to infection during gestation in the etiology of neurodevelopmental disorders. Animal models of maternal immune activation (MIA) have identified the maternal immune response as the critical link between maternal infection and aberrant offspring brain and behavior development. Here we evaluate neurodevelopment of male rhesus monkeys (Macaca mulatta) born to MIA-treated dams (n = 14) injected with a modified form of the viral mimic polyinosinic:polycytidylic acid at the end of the first trimester. Control dams received saline injections at the same gestational time points (n = 10) or were untreated (n = 4). MIA-treated dams exhibited a strong immune response as indexed by transient increases in sickness behavior, temperature, and inflammatory cytokines. Although offspring born to control or MIA-treated dams did not differ on measures of physical growth and early developmental milestones, the MIA-treated animals exhibited subtle changes in cognitive development and deviated from species-typical brain growth trajectories. Longitudinal MRI revealed significant gray matter volume reductions in the prefrontal and frontal cortices of MIA-treated offspring at 6 months that persisted through the final time point at 45 months along with smaller frontal white matter volumes in MIA-treated animals at 36 and 45 months. These findings provide the first evidence of early postnatal changes in brain development in MIA-exposed nonhuman primates and establish a translationally relevant model system to explore the neurodevelopmental trajectory of risk associated with prenatal immune challenge from birth through late adolescence.SIGNIFICANCE STATEMENT Women exposed to infection during pregnancy have an increased risk of giving birth to a child who will later be diagnosed with a neurodevelopmental disorder. Preclinical maternal immune activation (MIA) models have demonstrated that the effects of maternal infection on fetal brain development are mediated by maternal immune response. Since the majority of MIA models are conducted in rodents, the nonhuman primate provides a unique system to evaluate the MIA hypothesis in a species closely related to humans. Here we report the first longitudinal study conducted in a nonhuman primate MIA model. MIA-exposed offspring demonstrate subtle changes in cognitive development paired with marked reductions in frontal gray and white matter, further supporting the association between prenatal immune challenge and alterations in offspring neurodevelopment.


Subject(s)
Brain/pathology , Disease Models, Animal , Neurodevelopmental Disorders/etiology , Pregnancy Complications, Infectious , Prenatal Exposure Delayed Effects/pathology , Animals , Female , Interferon Inducers/toxicity , Macaca mulatta , Male , Neurodevelopmental Disorders/pathology , Neurogenesis/physiology , Poly I-C/toxicity , Pregnancy , Pregnancy Complications, Infectious/chemically induced , Prenatal Exposure Delayed Effects/chemically induced
14.
Nature ; 538(7626): 523-527, 2016 10 27.
Article in English | MEDLINE | ID: mdl-27760116

ABSTRACT

Three-dimensional physical interactions within chromosomes dynamically regulate gene expression in a tissue-specific manner. However, the 3D organization of chromosomes during human brain development and its role in regulating gene networks dysregulated in neurodevelopmental disorders, such as autism or schizophrenia, are unknown. Here we generate high-resolution 3D maps of chromatin contacts during human corticogenesis, permitting large-scale annotation of previously uncharacterized regulatory relationships relevant to the evolution of human cognition and disease. Our analyses identify hundreds of genes that physically interact with enhancers gained on the human lineage, many of which are under purifying selection and associated with human cognitive function. We integrate chromatin contacts with non-coding variants identified in schizophrenia genome-wide association studies (GWAS), highlighting multiple candidate schizophrenia risk genes and pathways, including transcription factors involved in neurogenesis, and cholinergic signalling molecules, several of which are supported by independent expression quantitative trait loci and gene expression analyses. Genome editing in human neural progenitors suggests that one of these distal schizophrenia GWAS loci regulates FOXG1 expression, supporting its potential role as a schizophrenia risk gene. This work provides a framework for understanding the effect of non-coding regulatory elements on human brain development and the evolution of cognition, and highlights novel mechanisms underlying neuropsychiatric disorders.


Subject(s)
Brain/embryology , Brain/metabolism , Chromatin/chemistry , Chromatin/genetics , Chromosomes, Human/chemistry , Chromosomes, Human/genetics , Gene Expression Regulation, Developmental , Nucleic Acid Conformation , Chromatin/metabolism , Chromosomes, Human/metabolism , Cognition , Enhancer Elements, Genetic/genetics , Epigenesis, Genetic , Forkhead Transcription Factors/genetics , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Humans , Nerve Tissue Proteins/genetics , Neural Stem Cells/metabolism , Neurogenesis , Organ Specificity , Polymorphism, Single Nucleotide/genetics , Promoter Regions, Genetic/genetics , Reproducibility of Results , Schizophrenia/genetics , Schizophrenia/pathology
15.
Nature ; 540(7633): 423-427, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27919067

ABSTRACT

Autism spectrum disorder (ASD) involves substantial genetic contributions. These contributions are profoundly heterogeneous but may converge on common pathways that are not yet well understood. Here, through post-mortem genome-wide transcriptome analysis of the largest cohort of samples analysed so far, to our knowledge, we interrogate the noncoding transcriptome, alternative splicing, and upstream molecular regulators to broaden our understanding of molecular convergence in ASD. Our analysis reveals ASD-associated dysregulation of primate-specific long noncoding RNAs (lncRNAs), downregulation of the alternative splicing of activity-dependent neuron-specific exons, and attenuation of normal differences in gene expression between the frontal and temporal lobes. Our data suggest that SOX5, a transcription factor involved in neuron fate specification, contributes to this reduction in regional differences. We further demonstrate that a genetically defined subtype of ASD, chromosome 15q11.2-13.1 duplication syndrome (dup15q), shares the core transcriptomic signature observed in idiopathic ASD. Co-expression network analysis reveals that individuals with ASD show age-related changes in the trajectory of microglial and synaptic function over the first two decades, and suggests that genetic risk for ASD may influence changes in regional cortical gene expression. Our findings illustrate how diverse genetic perturbations can lead to phenotypic convergence at multiple biological levels in a complex neuropsychiatric disorder.


Subject(s)
Alternative Splicing/genetics , Autism Spectrum Disorder/genetics , Gene Expression Profiling , Gene Expression Regulation , Genome, Human/genetics , RNA, Long Noncoding/genetics , Animals , Autopsy , Case-Control Studies , Chromosome Aberrations , Chromosomes, Human, Pair 15/genetics , Exons/genetics , Frontal Lobe/metabolism , Humans , Intellectual Disability/genetics , Neurons/metabolism , Primates/genetics , SOXD Transcription Factors/metabolism , Species Specificity , Temporal Lobe/metabolism , Transcriptome/genetics
16.
Nat Rev Genet ; 16(8): 441-58, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26149713

ABSTRACT

Genetic and genomic approaches have implicated hundreds of genetic loci in neurodevelopmental disorders and neurodegeneration, but mechanistic understanding continues to lag behind the pace of gene discovery. Understanding the role of specific genetic variants in the brain involves dissecting a functional hierarchy that encompasses molecular pathways, diverse cell types, neural circuits and, ultimately, cognition and behaviour. With a focus on transcriptomics, this Review discusses how high-throughput molecular, integrative and network approaches inform disease biology by placing human genetics in a molecular systems and neurobiological context. We provide a framework for interpreting network biology studies and leveraging big genomics data sets in neurobiology.


Subject(s)
Brain/metabolism , Developmental Disabilities/genetics , Gene Expression Profiling/methods , Gene Regulatory Networks/genetics , High-Throughput Screening Assays/methods , Models, Neurological , Neurobiology/methods , Neurodegenerative Diseases/genetics , Brain/cytology , High-Throughput Screening Assays/trends , Humans , Neurobiology/trends , Systems Biology/methods , Systems Biology/trends
17.
J Neurosci ; 39(42): 8193-8199, 2019 10 16.
Article in English | MEDLINE | ID: mdl-31619487

ABSTRACT

Many cellular and physiological processes are coordinated by regulatory networks that produce a remarkable complexity of transcript isoforms. In the mammalian nervous system, alternative pre-mRNA splicing generates functionally distinct isoforms that play key roles in normal physiology, supporting development, plasticity, complex behaviors, and cognition. Neuronal splicing programs controlled by RNA-binding proteins, are influenced by chromatin modifications and can exhibit neuronal subtype specificity. As highlighted in recent publications, aberrant alternative splicing is a major contributor to disease phenotypes. Therefore, understanding the underlying mechanisms of alternative splicing regulation and identifying functional splicing isoforms with critical phenotypic roles are expected to provide a comprehensive resource for therapeutic development, as illuminated by recent successful interventions of spinal muscular atrophy. Here, we discuss the latest progress in the study of the emerging complexity of alternative splicing mechanisms in neurons, and how these findings inform new therapies to correct and control splicing defects.


Subject(s)
Alternative Splicing/physiology , Autism Spectrum Disorder/therapy , Muscular Atrophy, Spinal/therapy , Neurons/metabolism , Animals , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Humans , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , Protein Isoforms/metabolism , RNA Splicing
19.
J Child Psychol Psychiatry ; 60(5): 585-598, 2019 05.
Article in English | MEDLINE | ID: mdl-30362171

ABSTRACT

BACKGROUND: Williams syndrome (WS) is a neurodevelopmental disorder that has been attributed to heterozygous deletions in chromosome 7q11.23 and exhibits a variety of physical, cognitive, and behavioral features. However, the genetic basis of this phenotypic variability is unclear. In this study, we identified genetic clues underlying these complex phenotypes. METHODS: Neurobehavioral function was assessed in WS patients and healthy controls. Total RNA was extracted from peripheral blood and subjected to microarray analysis, RNA-sequencing, and qRT-PCR. Weighted gene co-expression network analysis was performed to identify specific alterations related to intermediate disease phenotypes. To functionally interpret each WS-related module, gene ontology and disease-related gene enrichment were examined. We also investigated the micro (mi)RNA expression profiles and miRNA co-expression networks to better explain the regulation of the transcriptome in WS. RESULTS: Our analysis identified four significant co-expression modules related to intermediate WS phenotypes. Notably, the three upregulated WS-related modules were composed exclusively of genes located outside the 7q11.23 region. They were significantly enriched in genes related to B-cell activation, RNA processing, and RNA transport. BCL11A, which is known for its association with speech disorders and intellectual disabilities, was identified as one of the hub genes in the top WS-related module. Finally, these key upregulated mRNA co-expression modules appear to be inversely correlated with a specific downregulated WS-related miRNA co-expression module. CONCLUSIONS: Dysregulation of the mRNA/miRNA network involving genes outside of the 7q11.23 region is likely related to the complex phenotypes observed in WS patients.


Subject(s)
Autism Spectrum Disorder/genetics , Gene Expression Profiling , Gene Expression/genetics , Williams Syndrome/genetics , Child , Chromosomes, Human, Pair 7/genetics , Humans , MicroRNAs/genetics , RNA, Messenger/genetics
20.
Proc Natl Acad Sci U S A ; 108(43): E962-70, 2011 Oct 25.
Article in English | MEDLINE | ID: mdl-21969553

ABSTRACT

DTNBP1 (dystrobrevin binding protein 1) is a leading candidate susceptibility gene in schizophrenia and is associated with working memory capacity in normal subjects. In schizophrenia, the encoded protein dystrobrevin-binding protein 1 (dysbindin-1) is often reduced in excitatory cortical limbic synapses. We found that reduced dysbindin-1 in mice yielded deficits in auditory-evoked response adaptation, prepulse inhibition of startle, and evoked γ-activity, similar to patterns in schizophrenia. In contrast to the role of dysbindin-1 in glutamatergic transmission, γ-band abnormalities in schizophrenia are most often attributed to disrupted inhibition and reductions in parvalbumin-positive interneuron (PV cell) activity. To determine the mechanism underlying electrophysiological deficits related to reduced dysbindin-1 and the potential role of PV cells, we examined PV cell immunoreactivity and measured changes in net circuit activity using voltage-sensitive dye imaging. The dominant circuit impact of reduced dysbindin-1 was impaired inhibition, and PV cell immunoreactivity was reduced. Thus, this model provides a link between a validated candidate gene and an auditory endophenotypes. Furthermore, these data implicate reduced fast-phasic inhibition as a common underlying mechanism of schizophrenia-associated intermediate phenotypes.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Evoked Potentials, Auditory/physiology , Limbic System/metabolism , Schizophrenia/genetics , Synapses/metabolism , Animals , Dysbindin , Dystrophin-Associated Proteins , Electrophysiology , Evoked Potentials, Auditory/genetics , Female , Genotype , Immunohistochemistry , Male , Mice , Mice, Mutant Strains , Parvalbumins
SELECTION OF CITATIONS
SEARCH DETAIL