Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 113
Filter
1.
Cell ; 182(3): 754-769.e18, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32610082

ABSTRACT

To discover regulatory elements driving the specificity of gene expression in different cell types and regions of the developing human brain, we generated an atlas of open chromatin from nine dissected regions of the mid-gestation human telencephalon, as well as microdissected upper and deep layers of the prefrontal cortex. We identified a subset of open chromatin regions (OCRs), termed predicted regulatory elements (pREs), that are likely to function as developmental brain enhancers. pREs showed temporal, regional, and laminar differences in chromatin accessibility and were correlated with gene expression differences across regions and gestational ages. We identified two functional de novo variants in a pRE for autism risk gene SLC6A1, and using CRISPRa, demonstrated that this pRE regulates SCL6A1. Additionally, mouse transgenic experiments validated enhancer activity for pREs proximal to FEZF2 and BCL11A. Thus, this atlas serves as a resource for decoding neurodevelopmental gene regulation in health and disease.


Subject(s)
Chromatin/genetics , Chromatin/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental/genetics , Prefrontal Cortex/embryology , Telencephalon/embryology , Animals , Autistic Disorder/genetics , Cell Line , Chromatin Immunoprecipitation Sequencing , Euchromatin/genetics , GABA Plasma Membrane Transport Proteins/genetics , Gene Ontology , Genetic Predisposition to Disease , Gestational Age , Humans , Mice , Mice, Transgenic , Nucleotide Motifs , Point Mutation , Prefrontal Cortex/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Spatio-Temporal Analysis , Telencephalon/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
2.
Cell ; 180(3): 568-584.e23, 2020 02 06.
Article in English | MEDLINE | ID: mdl-31981491

ABSTRACT

We present the largest exome sequencing study of autism spectrum disorder (ASD) to date (n = 35,584 total samples, 11,986 with ASD). Using an enhanced analytical framework to integrate de novo and case-control rare variation, we identify 102 risk genes at a false discovery rate of 0.1 or less. Of these genes, 49 show higher frequencies of disruptive de novo variants in individuals ascertained to have severe neurodevelopmental delay, whereas 53 show higher frequencies in individuals ascertained to have ASD; comparing ASD cases with mutations in these groups reveals phenotypic differences. Expressed early in brain development, most risk genes have roles in regulation of gene expression or neuronal communication (i.e., mutations effect neurodevelopmental and neurophysiological changes), and 13 fall within loci recurrently hit by copy number variants. In cells from the human cortex, expression of risk genes is enriched in excitatory and inhibitory neuronal lineages, consistent with multiple paths to an excitatory-inhibitory imbalance underlying ASD.


Subject(s)
Autistic Disorder/genetics , Cerebral Cortex/growth & development , Exome Sequencing/methods , Gene Expression Regulation, Developmental , Neurobiology/methods , Case-Control Studies , Cell Lineage , Cohort Studies , Exome , Female , Gene Frequency , Genetic Predisposition to Disease , Humans , Male , Mutation, Missense , Neurons/metabolism , Phenotype , Sex Factors , Single-Cell Analysis/methods
3.
Cell ; 174(3): 505-520, 2018 07 26.
Article in English | MEDLINE | ID: mdl-30053424

ABSTRACT

Although gene discovery in neuropsychiatric disorders, including autism spectrum disorder, intellectual disability, epilepsy, schizophrenia, and Tourette disorder, has accelerated, resulting in a large number of molecular clues, it has proven difficult to generate specific hypotheses without the corresponding datasets at the protein complex and functional pathway level. Here, we describe one path forward-an initiative aimed at mapping the physical and genetic interaction networks of these conditions and then using these maps to connect the genomic data to neurobiology and, ultimately, the clinic. These efforts will include a team of geneticists, structural biologists, neurobiologists, systems biologists, and clinicians, leveraging a wide array of experimental approaches and creating a collaborative infrastructure necessary for long-term investigation. This initiative will ultimately intersect with parallel studies that focus on other diseases, as there is a significant overlap with genes implicated in cancer, infectious disease, and congenital heart defects.


Subject(s)
Chromosome Mapping/methods , Neurodevelopmental Disorders/genetics , Systems Biology/methods , Gene Regulatory Networks/genetics , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study/methods , Genomics/methods , Humans , Neurobiology/methods , Neuropsychiatry
4.
Cell ; 157(1): 201-14, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-24679536

ABSTRACT

Recent findings in a range of scientific disciplines are challenging the conventional wisdom regarding the etiology, classification, and treatment of psychiatric disorders. This Review focuses on the current state of the psychiatric diagnostic nosology and recent progress in three areas: genomics, neuroimaging, and therapeutics development. The accelerating pace of novel and unexpected findings is transforming the understanding of mental illness and represents a hopeful sign that the approaches and models that have sustained the field for the past 40 years are yielding to a flood of new data and presaging the emergence of a new and more powerful scientific paradigm.


Subject(s)
Mental Disorders/diagnosis , Mental Disorders/therapy , Neurosciences/methods , Animals , Humans , Mental Disorders/classification , Mental Disorders/genetics , Neuroimaging
5.
Cell ; 155(5): 997-1007, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-24267886

ABSTRACT

Autism spectrum disorder (ASD) is a complex developmental syndrome of unknown etiology. Recent studies employing exome- and genome-wide sequencing have identified nine high-confidence ASD (hcASD) genes. Working from the hypothesis that ASD-associated mutations in these biologically pleiotropic genes will disrupt intersecting developmental processes to contribute to a common phenotype, we have attempted to identify time periods, brain regions, and cell types in which these genes converge. We have constructed coexpression networks based on the hcASD "seed" genes, leveraging a rich expression data set encompassing multiple human brain regions across human development and into adulthood. By assessing enrichment of an independent set of probable ASD (pASD) genes, derived from the same sequencing studies, we demonstrate a key point of convergence in midfetal layer 5/6 cortical projection neurons. This approach informs when, where, and in what cell types mutations in these specific genes may be productively studied to clarify ASD pathophysiology.


Subject(s)
Brain/metabolism , Child Development Disorders, Pervasive/genetics , Child Development Disorders, Pervasive/physiopathology , Animals , Brain/embryology , Brain/growth & development , Brain/pathology , Child Development Disorders, Pervasive/pathology , Exome , Female , Fetus/metabolism , Fetus/pathology , Gene Expression Profiling , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Male , Mice , Mutation , Neurons/metabolism , Prefrontal Cortex/metabolism , Sequence Analysis, DNA
6.
Nat Rev Neurosci ; 23(6): 323-341, 2022 06.
Article in English | MEDLINE | ID: mdl-35440779

ABSTRACT

More than a hundred genes have been identified that, when disrupted, impart large risk for autism spectrum disorder (ASD). Current knowledge about the encoded proteins - although incomplete - points to a very wide range of developmentally dynamic and diverse biological processes. Moreover, the core symptoms of ASD involve distinctly human characteristics, presenting challenges to interpreting evolutionarily distant model systems. Indeed, despite a decade of striking progress in gene discovery, an actionable understanding of pathobiology remains elusive. Increasingly, convergent neuroscience approaches have been recognized as an important complement to traditional uses of genetics to illuminate the biology of human disorders. These methods seek to identify intersection among molecular-level, cellular-level and circuit-level functions across multiple risk genes and have highlighted developing excitatory neurons in the human mid-gestational prefrontal cortex as an important pathobiological nexus in ASD. In addition, neurogenesis, chromatin modification and synaptic function have emerged as key potential mediators of genetic vulnerability. The continued expansion of foundational 'omics' data sets, the application of higher-throughput model systems and incorporating developmental trajectories and sex differences into future analyses will refine and extend these results. Ultimately, a systems-level understanding of ASD genetic risk holds promise for clarifying pathobiology and advancing therapeutics.


Subject(s)
Autism Spectrum Disorder , Neurosciences , Autism Spectrum Disorder/genetics , Female , Genomics , Humans , Male , Neurogenesis , Neurons
7.
Development ; 150(14)2023 07 15.
Article in English | MEDLINE | ID: mdl-37366052

ABSTRACT

Gene ontology analyses of high-confidence autism spectrum disorder (ASD) risk genes highlight chromatin regulation and synaptic function as major contributors to pathobiology. Our recent functional work in vivo has additionally implicated tubulin biology and cellular proliferation. As many chromatin regulators, including the ASD risk genes ADNP and CHD3, are known to directly regulate both tubulins and histones, we studied the five chromatin regulators most strongly associated with ASD (ADNP, CHD8, CHD2, POGZ and KMT5B) specifically with respect to tubulin biology. We observe that all five localize to microtubules of the mitotic spindle in vitro in human cells and in vivo in Xenopus. Investigation of CHD2 provides evidence that mutations present in individuals with ASD cause a range of microtubule-related phenotypes, including disrupted localization of the protein at mitotic spindles, cell cycle stalling, DNA damage and cell death. Lastly, we observe that ASD genetic risk is significantly enriched among tubulin-associated proteins, suggesting broader relevance. Together, these results provide additional evidence that the role of tubulin biology and cellular proliferation in ASD warrants further investigation and highlight the pitfalls of relying solely on annotated gene functions in the search for pathological mechanisms.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Humans , Autistic Disorder/genetics , Autistic Disorder/complications , Autistic Disorder/metabolism , Chromatin/metabolism , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/pathology , Tubulin/metabolism , Histones/metabolism , Microtubules/metabolism , Spindle Apparatus/metabolism
8.
Development ; 147(21)2020 06 22.
Article in English | MEDLINE | ID: mdl-32467234

ABSTRACT

DYRK1A [dual specificity tyrosine-(Y)-phosphorylation-regulated kinase 1 A] is a high-confidence autism risk gene that encodes a conserved kinase. In addition to autism, individuals with putative loss-of-function variants in DYRK1A exhibit microcephaly, intellectual disability, developmental delay and/or congenital anomalies of the kidney and urinary tract. DYRK1A is also located within the critical region for Down syndrome; therefore, understanding the role of DYRK1A in brain development is crucial for understanding the pathobiology of multiple developmental disorders. To characterize the function of this gene, we used the diploid frog Xenopus tropicalis We discover that Dyrk1a is expressed in ciliated tissues, localizes to ciliary axonemes and basal bodies, and is required for ciliogenesis. We also demonstrate that Dyrk1a localizes to mitotic spindles and that its inhibition leads to decreased forebrain size, abnormal cell cycle progression and cell death during brain development. These findings provide hypotheses about potential mechanisms of pathobiology and underscore the utility of X. tropicalis as a model system for understanding neurodevelopmental disorders.


Subject(s)
Brain/anatomy & histology , Cilia/metabolism , Embryo, Nonmammalian/anatomy & histology , Neurodevelopmental Disorders/genetics , Organogenesis/genetics , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Xenopus Proteins/genetics , Xenopus/embryology , Xenopus/genetics , Animals , Brain/embryology , Cell Cycle/genetics , Cell Survival , Gene Expression Regulation, Developmental , Genetic Predisposition to Disease , Organ Size , Phenotype , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Risk Factors , Spindle Apparatus/metabolism , Telencephalon/anatomy & histology , Xenopus Proteins/metabolism
9.
Nature ; 520(7545): 51-6, 2015 Apr 02.
Article in English | MEDLINE | ID: mdl-25807484

ABSTRACT

Autism is a multifactorial neurodevelopmental disorder affecting more males than females; consequently, under a multifactorial genetic hypothesis, females are affected only when they cross a higher biological threshold. We hypothesize that deleterious variants at conserved residues are enriched in severely affected patients arising from female-enriched multiplex families with severe disease, enhancing the detection of key autism genes in modest numbers of cases. Here we show the use of this strategy by identifying missense and dosage sequence variants in the gene encoding the adhesive junction-associated δ-catenin protein (CTNND2) in female-enriched multiplex families and demonstrating their loss-of-function effect by functional analyses in zebrafish embryos and cultured hippocampal neurons from wild-type and Ctnnd2 null mouse embryos. Finally, through gene expression and network analyses, we highlight a critical role for CTNND2 in neuronal development and an intimate connection to chromatin biology. Our data contribute to the understanding of the genetic architecture of autism and suggest that genetic analyses of phenotypic extremes, such as female-enriched multiplex families, are of innate value in multifactorial disorders.


Subject(s)
Autistic Disorder/genetics , Autistic Disorder/metabolism , Brain/metabolism , Catenins/deficiency , Catenins/genetics , Animals , Brain/embryology , Catenins/metabolism , Cells, Cultured , Chromatin/genetics , Chromatin/metabolism , DNA Copy Number Variations/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Exome/genetics , Female , Gene Expression , Gene Expression Regulation, Developmental , Hippocampus/pathology , Humans , Male , Mice , Models, Genetic , Multifactorial Inheritance/genetics , Mutation, Missense , Nerve Net , Neurons/cytology , Neurons/metabolism , Sex Characteristics , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism , Delta Catenin
10.
Nature ; 515(7526): 216-21, 2014 Nov 13.
Article in English | MEDLINE | ID: mdl-25363768

ABSTRACT

Whole exome sequencing has proven to be a powerful tool for understanding the genetic architecture of human disease. Here we apply it to more than 2,500 simplex families, each having a child with an autistic spectrum disorder. By comparing affected to unaffected siblings, we show that 13% of de novo missense mutations and 43% of de novo likely gene-disrupting (LGD) mutations contribute to 12% and 9% of diagnoses, respectively. Including copy number variants, coding de novo mutations contribute to about 30% of all simplex and 45% of female diagnoses. Almost all LGD mutations occur opposite wild-type alleles. LGD targets in affected females significantly overlap the targets in males of lower intelligence quotient (IQ), but neither overlaps significantly with targets in males of higher IQ. We estimate that LGD mutation in about 400 genes can contribute to the joint class of affected females and males of lower IQ, with an overlapping and similar number of genes vulnerable to contributory missense mutation. LGD targets in the joint class overlap with published targets for intellectual disability and schizophrenia, and are enriched for chromatin modifiers, FMRP-associated genes and embryonically expressed genes. Most of the significance for the latter comes from affected females.


Subject(s)
Child Development Disorders, Pervasive/genetics , Genetic Predisposition to Disease/genetics , Mutation/genetics , Open Reading Frames/genetics , Child , Cluster Analysis , Exome/genetics , Female , Genes , Humans , Intelligence Tests , Male , Reproducibility of Results
12.
Nature ; 485(7397): 237-41, 2012 Apr 04.
Article in English | MEDLINE | ID: mdl-22495306

ABSTRACT

Multiple studies have confirmed the contribution of rare de novo copy number variations to the risk for autism spectrum disorders. But whereas de novo single nucleotide variants have been identified in affected individuals, their contribution to risk has yet to be clarified. Specifically, the frequency and distribution of these mutations have not been well characterized in matched unaffected controls, and such data are vital to the interpretation of de novo coding mutations observed in probands. Here we show, using whole-exome sequencing of 928 individuals, including 200 phenotypically discordant sibling pairs, that highly disruptive (nonsense and splice-site) de novo mutations in brain-expressed genes are associated with autism spectrum disorders and carry large effects. On the basis of mutation rates in unaffected individuals, we demonstrate that multiple independent de novo single nucleotide variants in the same gene among unrelated probands reliably identifies risk alleles, providing a clear path forward for gene discovery. Among a total of 279 identified de novo coding mutations, there is a single instance in probands, and none in siblings, in which two independent nonsense variants disrupt the same gene, SCN2A (sodium channel, voltage-gated, type II, α subunit), a result that is highly unlikely by chance.


Subject(s)
Autistic Disorder/genetics , Exome/genetics , Exons/genetics , Genetic Predisposition to Disease/genetics , Mutation/genetics , Nerve Tissue Proteins/genetics , Sodium Channels/genetics , Alleles , Codon, Nonsense/genetics , Genetic Heterogeneity , Humans , NAV1.2 Voltage-Gated Sodium Channel , RNA Splice Sites/genetics , Siblings
13.
PLoS Genet ; 11(1): e1004852, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25621974

ABSTRACT

Contactins and Contactin-Associated Proteins, and Contactin-Associated Protein-Like 2 (CNTNAP2) in particular, have been widely cited as autism risk genes based on findings from homozygosity mapping, molecular cytogenetics, copy number variation analyses, and both common and rare single nucleotide association studies. However, data specifically with regard to the contribution of heterozygous single nucleotide variants (SNVs) have been inconsistent. In an effort to clarify the role of rare point mutations in CNTNAP2 and related gene families, we have conducted targeted next-generation sequencing and evaluated existing sequence data in cohorts totaling 2704 cases and 2747 controls. We find no evidence for statistically significant association of rare heterozygous mutations in any of the CNTN or CNTNAP genes, including CNTNAP2, placing marked limits on the scale of their plausible contribution to risk.


Subject(s)
Autistic Disorder/genetics , Contactins/genetics , Genetic Association Studies , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Autistic Disorder/pathology , Codon, Nonsense , DNA Copy Number Variations , Genetic Predisposition to Disease , Humans , Point Mutation , Polymorphism, Single Nucleotide , Sequence Analysis, DNA , Sequence Deletion
14.
Am J Hum Genet ; 93(1): 103-9, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-23830515

ABSTRACT

Intellectual disability (ID), often attributed to autosomal-recessive mutations, occurs in 40% of autism spectrum disorders (ASDs). For this reason, we conducted a genome-wide analysis of runs of homozygosity (ROH) in simplex ASD-affected families consisting of a proband diagnosed with ASD and at least one unaffected sibling. In these families, probands with an IQ ≤ 70 show more ROH than their unaffected siblings, whereas probands with an IQ > 70 do not show this excess. Although ASD is far more common in males than in females, the proportion of females increases with decreasing IQ. Our data do support an association between ROH burden and autism diagnosis in girls; however, we are not able to show that this effect is independent of low IQ. We have also discovered several autism candidate genes on the basis of finding (1) a single gene that is within an ROH interval and that is recurrent in autism or (2) a gene that is within an autism ROH block and that harbors a homozygous, rare deleterious variant upon analysis of exome-sequencing data. In summary, our data suggest a distinct genetic architecture for participants with autism and co-occurring intellectual disability and that this architecture could involve a role for recessively inherited loci for this autism subgroup.


Subject(s)
Child Development Disorders, Pervasive/genetics , Genetic Association Studies/methods , Intellectual Disability/genetics , Child , Chromosomes, Human/genetics , Female , Genetic Diseases, Inborn/genetics , Genetic Predisposition to Disease/genetics , Genetics, Population/methods , Homozygote , Humans , Intelligence Tests , Male , Pedigree , Phenotype , Sex Factors
15.
Circ Res ; 115(10): 884-896, 2014 Oct 24.
Article in English | MEDLINE | ID: mdl-25205790

ABSTRACT

RATIONALE: Congenital heart disease (CHD) is among the most common birth defects. Most cases are of unknown pathogenesis. OBJECTIVE: To determine the contribution of de novo copy number variants (CNVs) in the pathogenesis of sporadic CHD. METHODS AND RESULTS: We studied 538 CHD trios using genome-wide dense single nucleotide polymorphism arrays and whole exome sequencing. Results were experimentally validated using digital droplet polymerase chain reaction. We compared validated CNVs in CHD cases with CNVs in 1301 healthy control trios. The 2 complementary high-resolution technologies identified 63 validated de novo CNVs in 51 CHD cases. A significant increase in CNV burden was observed when comparing CHD trios with healthy trios, using either single nucleotide polymorphism array (P=7×10(-5); odds ratio, 4.6) or whole exome sequencing data (P=6×10(-4); odds ratio, 3.5) and remained after removing 16% of de novo CNV loci previously reported as pathogenic (P=0.02; odds ratio, 2.7). We observed recurrent de novo CNVs on 15q11.2 encompassing CYFIP1, NIPA1, and NIPA2 and single de novo CNVs encompassing DUSP1, JUN, JUP, MED15, MED9, PTPRE SREBF1, TOP2A, and ZEB2, genes that interact with established CHD proteins NKX2-5 and GATA4. Integrating de novo variants in whole exome sequencing and CNV data suggests that ETS1 is the pathogenic gene altered by 11q24.2-q25 deletions in Jacobsen syndrome and that CTBP2 is the pathogenic gene in 10q subtelomeric deletions. CONCLUSIONS: We demonstrate a significantly increased frequency of rare de novo CNVs in CHD patients compared with healthy controls and suggest several novel genetic loci for CHD.


Subject(s)
DNA Copy Number Variations/genetics , Exome/genetics , Gene Frequency/genetics , Heart Defects, Congenital/genetics , Polymorphism, Single Nucleotide/genetics , Case-Control Studies , Cohort Studies , Gene Regulatory Networks/genetics , Heart Defects, Congenital/diagnosis , Humans , Molecular Sequence Data
16.
Nature ; 467(7312): 207-10, 2010 Sep 09.
Article in English | MEDLINE | ID: mdl-20729831

ABSTRACT

The development of the human cerebral cortex is an orchestrated process involving the generation of neural progenitors in the periventricular germinal zones, cell proliferation characterized by symmetric and asymmetric mitoses, followed by migration of post-mitotic neurons to their final destinations in six highly ordered, functionally specialized layers. An understanding of the molecular mechanisms guiding these intricate processes is in its infancy, substantially driven by the discovery of rare mutations that cause malformations of cortical development. Mapping of disease loci in putative Mendelian forms of malformations of cortical development has been hindered by marked locus heterogeneity, small kindred sizes and diagnostic classifications that may not reflect molecular pathogenesis. Here we demonstrate the use of whole-exome sequencing to overcome these obstacles by identifying recessive mutations in WD repeat domain 62 (WDR62) as the cause of a wide spectrum of severe cerebral cortical malformations including microcephaly, pachygyria with cortical thickening as well as hypoplasia of the corpus callosum. Some patients with mutations in WDR62 had evidence of additional abnormalities including lissencephaly, schizencephaly, polymicrogyria and, in one instance, cerebellar hypoplasia, all traits traditionally regarded as distinct entities. In mice and humans, WDR62 transcripts and protein are enriched in neural progenitors within the ventricular and subventricular zones. Expression of WDR62 in the neocortex is transient, spanning the period of embryonic neurogenesis. Unlike other known microcephaly genes, WDR62 does not apparently associate with centrosomes and is predominantly nuclear in localization. These findings unify previously disparate aspects of cerebral cortical development and highlight the use of whole-exome sequencing to identify disease loci in settings in which traditional methods have proved challenging.


Subject(s)
Brain Diseases/genetics , Brain/abnormalities , DNA Mutational Analysis/methods , Nerve Tissue Proteins/genetics , Animals , Base Sequence , Brain/growth & development , Brain/pathology , Brain Diseases/pathology , Cell Cycle Proteins , Female , Genes, Recessive , Humans , Male , Mice , Microcephaly/genetics , Microcephaly/pathology , Molecular Sequence Data , Mutation , Nerve Tissue Proteins/metabolism , Pedigree
17.
PLoS Genet ; 9(8): e1003671, 2013.
Article in English | MEDLINE | ID: mdl-23966865

ABSTRACT

De novo mutations affect risk for many diseases and disorders, especially those with early-onset. An example is autism spectrum disorders (ASD). Four recent whole-exome sequencing (WES) studies of ASD families revealed a handful of novel risk genes, based on independent de novo loss-of-function (LoF) mutations falling in the same gene, and found that de novo LoF mutations occurred at a twofold higher rate than expected by chance. However successful these studies were, they used only a small fraction of the data, excluding other types of de novo mutations and inherited rare variants. Moreover, such analyses cannot readily incorporate data from case-control studies. An important research challenge in gene discovery, therefore, is to develop statistical methods that accommodate a broader class of rare variation. We develop methods that can incorporate WES data regarding de novo mutations, inherited variants present, and variants identified within cases and controls. TADA, for Transmission And De novo Association, integrates these data by a gene-based likelihood model involving parameters for allele frequencies and gene-specific penetrances. Inference is based on a Hierarchical Bayes strategy that borrows information across all genes to infer parameters that would be difficult to estimate for individual genes. In addition to theoretical development we validated TADA using realistic simulations mimicking rare, large-effect mutations affecting risk for ASD and show it has dramatically better power than other common methods of analysis. Thus TADA's integration of various kinds of WES data can be a highly effective means of identifying novel risk genes. Indeed, application of TADA to WES data from subjects with ASD and their families, as well as from a study of ASD subjects and controls, revealed several novel and promising ASD candidate genes with strong statistical support.


Subject(s)
Child Development Disorders, Pervasive/genetics , Genetic Variation , Models, Theoretical , Mutation , Bayes Theorem , Case-Control Studies , Exome , Genetic Predisposition to Disease , Humans , Likelihood Functions , Risk Factors , Sequence Analysis, DNA
18.
Am J Hum Genet ; 91(1): 38-55, 2012 Jul 13.
Article in English | MEDLINE | ID: mdl-22726847

ABSTRACT

Copy-number variants (CNVs) are a major contributor to the pathophysiology of autism spectrum disorders (ASDs), but the functional impact of CNVs remains largely unexplored. Because brain tissue is not available from most samples, we interrogated gene expression in lymphoblasts from 244 families with discordant siblings in the Simons Simplex Collection in order to identify potentially pathogenic variation. Our results reveal that the overall frequency of significantly misexpressed genes (which we refer to here as outliers) identified in probands and unaffected siblings does not differ. However, in probands, but not their unaffected siblings, the group of outlier genes is significantly enriched in neural-related pathways, including neuropeptide signaling, synaptogenesis, and cell adhesion. We demonstrate that outlier genes cluster within the most pathogenic CNVs (rare de novo CNVs) and can be used for the prioritization of rare CNVs of potentially unknown significance. Several nonrecurrent CNVs with significant gene-expression alterations are identified (these include deletions in chromosomal regions 3q27, 3p13, and 3p26 and duplications at 2p15), suggesting that these are potential candidate ASD loci. In addition, we identify distinct expression changes in 16p11.2 microdeletions, 16p11.2 microduplications, and 7q11.23 duplications, and we show that specific genes within the 16p CNV interval correlate with differences in head circumference, an ASD-relevant phenotype. This study provides evidence that pathogenic structural variants have a functional impact via transcriptome alterations in ASDs at a genome-wide level and demonstrates the utility of integrating gene expression with mutation data for the prioritization of genes disrupted by potentially pathogenic mutations.


Subject(s)
Child Development Disorders, Pervasive/genetics , DNA Copy Number Variations , Child , Child, Preschool , Chromosomes, Human, Pair 16 , Gene Expression Profiling , Humans , Mutation
19.
Nature ; 459(7246): 528-33, 2009 May 28.
Article in English | MEDLINE | ID: mdl-19404256

ABSTRACT

Autism spectrum disorders (ASDs) represent a group of childhood neurodevelopmental and neuropsychiatric disorders characterized by deficits in verbal communication, impairment of social interaction, and restricted and repetitive patterns of interests and behaviour. To identify common genetic risk factors underlying ASDs, here we present the results of genome-wide association studies on a cohort of 780 families (3,101 subjects) with affected children, and a second cohort of 1,204 affected subjects and 6,491 control subjects, all of whom were of European ancestry. Six single nucleotide polymorphisms between cadherin 10 (CDH10) and cadherin 9 (CDH9)-two genes encoding neuronal cell-adhesion molecules-revealed strong association signals, with the most significant SNP being rs4307059 (P = 3.4 x 10(-8), odds ratio = 1.19). These signals were replicated in two independent cohorts, with combined P values ranging from 7.4 x 10(-8) to 2.1 x 10(-10). Our results implicate neuronal cell-adhesion molecules in the pathogenesis of ASDs, and represent, to our knowledge, the first demonstration of genome-wide significant association of common variants with susceptibility to ASDs.


Subject(s)
Autistic Disorder/genetics , Chromosomes, Human, Pair 5/genetics , Genetic Predisposition to Disease/genetics , Genetic Variation/genetics , Brain/metabolism , Cadherins/genetics , Case-Control Studies , Cell Adhesion/genetics , Cell Adhesion Molecules, Neuronal/genetics , Cohort Studies , Genetic Markers/genetics , Genome-Wide Association Study , Genotype , Humans , Polymorphism, Single Nucleotide/genetics , Reproducibility of Results
20.
Eur Child Adolesc Psychiatry ; 24(2): 141-51, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24771252

ABSTRACT

Tourette syndrome (TS) is a neuropsychiatric disorder characterized by recurrent motor and vocal tics, often accompanied by obsessive-compulsive disorder and/or attention-deficit/hyperactivity disorder. While the evidence for a genetic contribution is strong, its exact nature has yet to be clarified fully. There is now mounting evidence that the genetic risks for TS include both common and rare variants and may involve complex multigenic inheritance or, in rare cases, a single major gene. Based on recent progress in many other common disorders with apparently similar genetic architectures, it is clear that large patient cohorts and open-access repositories will be essential to further advance the field. To that end, the large multicenter Tourette International Collaborative Genetics (TIC Genetics) study was established. The goal of the TIC Genetics study is to undertake a comprehensive gene discovery effort, focusing both on familial genetic variants with large effects within multiply affected pedigrees and on de novo mutations ascertained through the analysis of apparently simplex parent-child trios with non-familial tics. The clinical data and biomaterials (DNA, transformed cell lines, RNA) are part of a sharing repository located within the National Institute for Mental Health Center for Collaborative Genomics Research on Mental Disorders, USA, and will be made available to the broad scientific community. This resource will ultimately facilitate better understanding of the pathophysiology of TS and related disorders and the development of novel therapies. Here, we describe the objectives and methods of the TIC Genetics study as a reference for future studies from our group and to facilitate collaboration between genetics consortia in the field of TS.


Subject(s)
Attention Deficit Disorder with Hyperactivity/complications , Obsessive-Compulsive Disorder/complications , Tic Disorders/complications , Tics/complications , Tourette Syndrome/genetics , Attention Deficit Disorder with Hyperactivity/psychology , Child , Cooperative Behavior , Female , Gene-Environment Interaction , Genetic Linkage , Genetic Predisposition to Disease , Genomics , Humans , Obsessive-Compulsive Disorder/psychology , Pedigree , Tic Disorders/psychology , Tics/psychology , Tourette Syndrome/psychology
SELECTION OF CITATIONS
SEARCH DETAIL