Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
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 ; 172(1-2): 289-304.e18, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29307494

ABSTRACT

Non-coding regions comprise most of the human genome and harbor a significant fraction of risk alleles for neuropsychiatric diseases, yet their functions remain poorly defined. We created a high-resolution map of non-coding elements involved in human cortical neurogenesis by contrasting chromatin accessibility and gene expression in the germinal zone and cortical plate of the developing cerebral cortex. We link distal regulatory elements (DREs) to their cognate gene(s) together with chromatin interaction data and show that target genes of human-gained enhancers (HGEs) regulate cortical neurogenesis and are enriched in outer radial glia, a cell type linked to human cortical evolution. We experimentally validate the regulatory effects of predicted enhancers for FGFR2 and EOMES. We observe that common genetic variants associated with educational attainment, risk for neuropsychiatric disease, and intracranial volume are enriched within regulatory elements involved in cortical neurogenesis, demonstrating the importance of this early developmental process for adult human cognitive function.


Subject(s)
Cerebral Cortex/metabolism , Chromatin Assembly and Disassembly , Gene Expression Regulation, Developmental , Neurogenesis , Neurons/metabolism , Cell Line , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Chromatin/genetics , Chromatin/metabolism , Enhancer Elements, Genetic , Female , Humans , Male , Neurons/cytology , Polymorphism, Genetic , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
5.
Nature ; 587(7833): 281-284, 2020 11.
Article in English | MEDLINE | ID: mdl-33087932

ABSTRACT

Angelman syndrome (AS) is a severe neurodevelopmental disorder caused by a mutation or deletion of the maternally inherited UBE3A allele. In neurons, the paternally inherited UBE3A allele is silenced in cis by a long non-coding RNA called UBE3A-ATS. Here, as part of a systematic screen, we found that Cas9 can be used to activate ('unsilence') paternal Ube3a in cultured mouse and human neurons when targeted to Snord115 genes, which are small nucleolar RNAs that are clustered in the 3' region of Ube3a-ATS. A short Cas9 variant and guide RNA that target about 75 Snord115 genes were packaged into an adeno-associated virus and administered to a mouse model of AS during the embryonic and early postnatal stages, when the therapeutic benefit of restoring Ube3a is predicted to be greatest1,2. This early treatment unsilenced paternal Ube3a throughout the brain for at least 17 months and rescued anatomical and behavioural phenotypes in AS mice. Genomic integration of the adeno-associated virus vector into Cas9 target sites caused premature termination of Ube3a-ATS at the vector-derived polyA cassette, or when integrated in the reverse orientation, by transcriptional collision with the vector-derived Cas9 transcript. Our study shows that targeted genomic integration of a gene therapy vector can restore the function of paternally inherited UBE3A throughout life, providing a path towards a disease-modifying treatment for a syndromic neurodevelopmental disorder.


Subject(s)
Angelman Syndrome/genetics , Angelman Syndrome/therapy , CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems/genetics , Gene Editing , Genetic Therapy/methods , RNA, Long Noncoding/genetics , Ubiquitin-Protein Ligases/genetics , Animals , CRISPR-Associated Protein 9/genetics , Dependovirus/genetics , Disease Models, Animal , Female , Gene Silencing , Genetic Vectors/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Nervous System/metabolism , Paternal Inheritance/genetics , Phenotype , RNA, Guide, Kinetoplastida/genetics
6.
Hum Mol Genet ; 32(3): 402-416, 2023 01 13.
Article in English | MEDLINE | ID: mdl-35994039

ABSTRACT

Genomic imprinting results in gene expression bias caused by parental chromosome of origin and occurs in genes with important roles during human brain development. However, the cell-type and temporal specificity of imprinting during human neurogenesis is generally unknown. By detecting within-donor allelic biases in chromatin accessibility and gene expression that are unrelated to cross-donor genotype, we inferred imprinting in both primary human neural progenitor cells and their differentiated neuronal progeny from up to 85 donors. We identified 43/20 putatively imprinted regulatory elements (IREs) in neurons/progenitors, and 133/79 putatively imprinted genes in neurons/progenitors. Although 10 IREs and 42 genes were shared between neurons and progenitors, most putative imprinting was only detected within specific cell types. In addition to well-known imprinted genes and their promoters, we inferred novel putative IREs and imprinted genes. Consistent with both DNA methylation-based and H3K27me3-based regulation of imprinted expression, some putative IREs also overlapped with differentially methylated or histone-marked regions. Finally, we identified a progenitor-specific putatively imprinted gene overlapping with copy number variation that is associated with uniparental disomy-like phenotypes. Our results can therefore be useful in interpreting the function of variants identified in future parent-of-origin association studies.


Subject(s)
DNA Copy Number Variations , DNA Methylation , Humans , DNA Methylation/genetics , Genomic Imprinting/genetics , Uniparental Disomy , Cell Differentiation/genetics
7.
Proc Natl Acad Sci U S A ; 119(40): e2200638119, 2022 10 04.
Article in English | MEDLINE | ID: mdl-36161899

ABSTRACT

Alterations in brain size and organization represent some of the most distinctive changes in the emergence of our species. Yet, there is limited understanding of how genetic factors contributed to altered neuroanatomy during human evolution. Here, we analyze neuroimaging and genetic data from up to 30,000 people in the UK Biobank and integrate with genomic annotations for different aspects of human evolution, including those based on ancient DNA and comparative genomics. We show that previously reported signals of recent polygenic selection for cortical anatomy are not replicable in a more ancestrally homogeneous sample. We then investigate relationships between evolutionary annotations and common genetic variants shaping cortical surface area and white-matter connectivity for each hemisphere. Our analyses identify single-nucleotide polymorphism heritability enrichment in human-gained regulatory elements that are active in early brain development, affecting surface areas of several parts of the cortex, including left-hemispheric speech-associated regions. We also detect heritability depletion in genomic regions with Neanderthal ancestry for connectivity of the uncinate fasciculus; this is a white-matter tract involved in memory, language, and socioemotional processing with relevance to neuropsychiatric disorders. Finally, we show that common genetic loci associated with left-hemispheric pars triangularis surface area overlap with a human-gained enhancer and affect regulation of ZIC4, a gene implicated in neurogenesis. This work demonstrates how genomic investigations of present-day neuroanatomical variation can help shed light on the complexities of our evolutionary past.


Subject(s)
Biological Evolution , Brain , Genomics , Neuroimaging , Polymorphism, Single Nucleotide , Brain/growth & development , Brain/ultrastructure , DNA, Ancient , Genomics/methods , Humans , Neuroimaging/methods
8.
Am J Hum Genet ; 108(9): 1647-1668, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34416157

ABSTRACT

Interpretation of the function of non-coding risk loci for neuropsychiatric disorders and brain-relevant traits via gene expression and alternative splicing quantitative trait locus (e/sQTL) analyses is generally performed in bulk post-mortem adult tissue. However, genetic risk loci are enriched in regulatory elements active during neocortical differentiation, and regulatory effects of risk variants may be masked by heterogeneity in bulk tissue. Here, we map e/sQTLs, and allele-specific expression in cultured cells representing two major developmental stages, primary human neural progenitors (n = 85) and their sorted neuronal progeny (n = 74), identifying numerous loci not detected in either bulk developing cortical wall or adult cortex. Using colocalization and genetic imputation via transcriptome-wide association, we uncover cell-type-specific regulatory mechanisms underlying risk for brain-relevant traits that are active during neocortical differentiation. Specifically, we identified a progenitor-specific eQTL for CENPW co-localized with common variant associations for cortical surface area and educational attainment.


Subject(s)
Chromosomal Proteins, Non-Histone/genetics , Gene Expression Regulation, Developmental , Neocortex/metabolism , Neurogenesis/genetics , Neurons/metabolism , Quantitative Trait Loci , Alleles , Alzheimer Disease/diagnosis , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Cell Differentiation , Chromatin/chemistry , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Chromosome Mapping , Educational Status , Female , Fetus , Genetic Predisposition to Disease , Genome, Human , Genome-Wide Association Study , Humans , Male , Neocortex/cytology , Neocortex/growth & development , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurons/cytology , Neuroticism , Parkinson Disease/diagnosis , Parkinson Disease/genetics , Parkinson Disease/metabolism , Primary Cell Culture , Prognosis , Schizophrenia/diagnosis , Schizophrenia/genetics , Schizophrenia/metabolism , Transcriptome
9.
PLoS Genet ; 17(4): e1009455, 2021 04.
Article in English | MEDLINE | ID: mdl-33872308

ABSTRACT

Expression quantitative trait loci (eQTL) studies are used to understand the regulatory function of non-coding genome-wide association study (GWAS) risk loci, but colocalization alone does not demonstrate a causal relationship of gene expression affecting a trait. Evidence for mediation, that perturbation of gene expression in a given tissue or developmental context will induce a change in the downstream GWAS trait, can be provided by two-sample Mendelian Randomization (MR). Here, we introduce a new statistical method, MRLocus, for Bayesian estimation of the gene-to-trait effect from eQTL and GWAS summary data for loci with evidence of allelic heterogeneity, that is, containing multiple causal variants. MRLocus makes use of a colocalization step applied to each nearly-LD-independent eQTL, followed by an MR analysis step across eQTLs. Additionally, our method involves estimation of the extent of allelic heterogeneity through a dispersion parameter, indicating variable mediation effects from each individual eQTL on the downstream trait. Our method is evaluated against other state-of-the-art methods for estimation of the gene-to-trait mediation effect, using an existing simulation framework. In simulation, MRLocus often has the highest accuracy among competing methods, and in each case provides more accurate estimation of uncertainty as assessed through interval coverage. MRLocus is then applied to five candidate causal genes for mediation of particular GWAS traits, where gene-to-trait effects are concordant with those previously reported. We find that MRLocus's estimation of the causal effect across eQTLs within a locus provides useful information for determining how perturbation of gene expression or individual regulatory elements will affect downstream traits. The MRLocus method is implemented as an R package available at https://mikelove.github.io/mrlocus.


Subject(s)
Gene Expression Profiling/statistics & numerical data , Genetic Predisposition to Disease , Genome-Wide Association Study/statistics & numerical data , Quantitative Trait Loci/genetics , Computer Simulation , Gene Expression Regulation/genetics , Humans , Linkage Disequilibrium , Mendelian Randomization Analysis , Models, Genetic , Transcriptome/genetics
10.
Neuroimage ; 265: 119773, 2023 01.
Article in English | MEDLINE | ID: mdl-36442731

ABSTRACT

The expansion of the cerebral cortex is one of the most distinctive changes in the evolution of the human brain. Cortical expansion and related increases in cortical folding may have contributed to emergence of our capacities for high-order cognitive abilities. Molecular analysis of humans, archaic hominins, and non-human primates has allowed identification of chromosomal regions showing evolutionary changes at different points of our phylogenetic history. In this study, we assessed the contributions of genomic annotations spanning 30 million years to human sulcal morphology measured via MRI in more than 18,000 participants from the UK Biobank. We found that variation within brain-expressed human gained enhancers, regulatory genetic elements that emerged since our last common ancestor with Old World monkeys, explained more trait heritability than expected for the left and right calloso-marginal posterior fissures and the right central sulcus. Intriguingly, these are sulci that have been previously linked to the evolution of locomotion in primates and later on bipedalism in our hominin ancestors.


Subject(s)
Brain , Cerebral Cortex , Animals , Humans , Phylogeny , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/anatomy & histology , Brain/anatomy & histology , Primates , Magnetic Resonance Imaging , Genetic Variation , Enhancer Elements, Genetic/genetics
11.
Hum Brain Mapp ; 43(1): 329-340, 2022 01.
Article in English | MEDLINE | ID: mdl-33098356

ABSTRACT

Human brain structure traits have been hypothesized to be broad endophenotypes for neuropsychiatric disorders, implying that brain structure traits are comparatively "closer to the underlying biology." Genome-wide association studies from large sample sizes allow for the comparison of common variant genetic architectures between traits to test the evidence supporting this claim. Endophenotypes, compared to neuropsychiatric disorders, are hypothesized to have less polygenicity, with greater effect size of each susceptible SNP, requiring smaller sample sizes to discover them. Here, we compare polygenicity and discoverability of brain structure traits, neuropsychiatric disorders, and other traits (91 in total) to directly test this hypothesis. We found reduced polygenicity (FDR = 0.01) and increased discoverability (FDR = 3.68 × 10-9 ) of cortical brain structure traits, as compared to aggregated estimates of multiple neuropsychiatric disorders. We predict that ~8 M individuals will be required to explain the full heritability of cortical surface area by genome-wide significant SNPs, whereas sample sizes over 20 M will be required to explain the full heritability of depression. In conclusion, our findings are consistent with brain structure satisfying the higher power criterion of endophenotypes.


Subject(s)
Cerebral Cortex , Endophenotypes , Genetic Predisposition to Disease , Inheritance Patterns , Mental Disorders , Nervous System Diseases , Neuroimaging , Brain Cortical Thickness , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/pathology , Genome-Wide Association Study , Humans , Mental Disorders/diagnostic imaging , Mental Disorders/genetics , Mental Disorders/pathology , Multifactorial Inheritance , Nervous System Diseases/diagnostic imaging , Nervous System Diseases/genetics , Nervous System Diseases/pathology , Polymorphism, Single Nucleotide
12.
Hum Brain Mapp ; 43(1): 292-299, 2022 01.
Article in English | MEDLINE | ID: mdl-33300665

ABSTRACT

Here we review the motivation for creating the enhancing neuroimaging genetics through meta-analysis (ENIGMA) Consortium and the genetic analyses undertaken by the consortium so far. We discuss the methodological challenges, findings, and future directions of the genetics working group. A major goal of the working group is tackling the reproducibility crisis affecting "candidate gene" and genome-wide association analyses in neuroimaging. To address this, we developed harmonized analytic methods, and support their use in coordinated analyses across sites worldwide, which also makes it possible to understand heterogeneity in results across sites. These efforts have resulted in the identification of hundreds of common genomic loci robustly associated with brain structure. We have found both pleiotropic and specific genetic effects associated with brain structures, as well as genetic correlations with psychiatric and neurological diseases.


Subject(s)
Brain , Genetics , Genome-Wide Association Study , Mental Disorders , Meta-Analysis as Topic , Nervous System Diseases , Neuroimaging , Brain/anatomy & histology , Brain/diagnostic imaging , Humans , Mental Disorders/diagnostic imaging , Mental Disorders/genetics , Mental Disorders/pathology , Multicenter Studies as Topic , Nervous System Diseases/diagnostic imaging , Nervous System Diseases/genetics , Nervous System Diseases/pathology
13.
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
14.
Cereb Cortex ; 31(4): 1873-1887, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33290510

ABSTRACT

Structural brain changes along the lineage leading to modern Homo sapiens contributed to our distinctive cognitive and social abilities. However, the evolutionarily relevant molecular variants impacting key aspects of neuroanatomy are largely unknown. Here, we integrate evolutionary annotations of the genome at diverse timescales with common variant associations from large-scale neuroimaging genetic screens. We find that alleles with evidence of recent positive polygenic selection over the past 2000-3000 years are associated with increased surface area (SA) of the entire cortex, as well as specific regions, including those involved in spoken language and visual processing. Therefore, polygenic selective pressures impact the structure of specific cortical areas even over relatively recent timescales. Moreover, common sequence variation within human gained enhancers active in the prenatal cortex is associated with postnatal global SA. We show that such variation modulates the function of a regulatory element of the developmentally relevant transcription factor HEY2 in human neural progenitor cells and is associated with structural changes in the inferior frontal cortex. These results indicate that non-coding genomic regions active during prenatal cortical development are involved in the evolution of human brain structure and identify novel regulatory elements and genes impacting modern human brain structure.


Subject(s)
Biological Evolution , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/physiology , Genetic Variation/genetics , Genome-Wide Association Study/methods , Genetic Testing/methods , Humans , Magnetic Resonance Imaging/trends , Multifactorial Inheritance/genetics , Organ Size/genetics , Quantitative Trait Loci/genetics
15.
BMC Bioinformatics ; 22(1): 260, 2021 May 22.
Article in English | MEDLINE | ID: mdl-34022787

ABSTRACT

BACKGROUND: Recent advances in tissue clearing techniques, combined with high-speed image acquisition through light sheet microscopy, enable rapid three-dimensional (3D) imaging of biological specimens, such as whole mouse brains, in a matter of hours. Quantitative analysis of such 3D images can help us understand how changes in brain structure lead to differences in behavior or cognition, but distinguishing densely packed features of interest, such as nuclei, from background can be challenging. Recent deep learning-based nuclear segmentation algorithms show great promise for automated segmentation, but require large numbers of accurate manually labeled nuclei as training data. RESULTS: We present Segmentor, an open-source tool for reliable, efficient, and user-friendly manual annotation and refinement of objects (e.g., nuclei) within 3D light sheet microscopy images. Segmentor employs a hybrid 2D-3D approach for visualizing and segmenting objects and contains features for automatic region splitting, designed specifically for streamlining the process of 3D segmentation of nuclei. We show that editing simultaneously in 2D and 3D using Segmentor significantly decreases time spent on manual annotations without affecting accuracy as compared to editing the same set of images with only 2D capabilities. CONCLUSIONS: Segmentor is a tool for increased efficiency of manual annotation and refinement of 3D objects that can be used to train deep learning segmentation algorithms, and is available at https://www.nucleininja.org/ and https://github.com/RENCI/Segmentor .


Subject(s)
Image Processing, Computer-Assisted , Microscopy , Algorithms , Animals , Brain , Imaging, Three-Dimensional , Mice
16.
Nature ; 520(7546): 224-9, 2015 Apr 09.
Article in English | MEDLINE | ID: mdl-25607358

ABSTRACT

The highly complex structure of the human brain is strongly shaped by genetic influences. Subcortical brain regions form circuits with cortical areas to coordinate movement, learning, memory and motivation, and altered circuits can lead to abnormal behaviour and disease. To investigate how common genetic variants affect the structure of these brain regions, here we conduct genome-wide association studies of the volumes of seven subcortical regions and the intracranial volume derived from magnetic resonance images of 30,717 individuals from 50 cohorts. We identify five novel genetic variants influencing the volumes of the putamen and caudate nucleus. We also find stronger evidence for three loci with previously established influences on hippocampal volume and intracranial volume. These variants show specific volumetric effects on brain structures rather than global effects across structures. The strongest effects were found for the putamen, where a novel intergenic locus with replicable influence on volume (rs945270; P = 1.08 × 10(-33); 0.52% variance explained) showed evidence of altering the expression of the KTN1 gene in both brain and blood tissue. Variants influencing putamen volume clustered near developmental genes that regulate apoptosis, axon guidance and vesicle transport. Identification of these genetic variants provides insight into the causes of variability in human brain development, and may help to determine mechanisms of neuropsychiatric dysfunction.


Subject(s)
Brain/anatomy & histology , Genetic Variation/genetics , Genome-Wide Association Study , Adolescent , Adult , Aged , Aged, 80 and over , Aging/genetics , Apoptosis/genetics , Caudate Nucleus/anatomy & histology , Child , Female , Gene Expression Regulation, Developmental/genetics , Genetic Loci/genetics , Hippocampus/anatomy & histology , Humans , Magnetic Resonance Imaging , Male , Membrane Proteins/genetics , Middle Aged , Organ Size/genetics , Putamen/anatomy & histology , Sex Characteristics , Skull/anatomy & histology , Young Adult
17.
Psychiatry Clin Neurosci ; 73(7): 357-369, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30864184

ABSTRACT

Imaging genetics aims to identify genetic variants associated with the structure and function of the human brain. Recently, collaborative consortia have been successful in this goal, identifying and replicating common genetic variants influencing gross human brain structure as measured through magnetic resonance imaging. In this review, we contextualize imaging genetic associations as one important link in understanding the causal chain from genetic variant to increased risk for neuropsychiatric disorders. We provide examples in other fields of how identifying genetic variant associations to disease and multiple phenotypes along the causal chain has revealed a mechanistic understanding of disease risk, with implications for how imaging genetics can be similarly applied. We discuss current findings in the imaging genetics research domain, including that common genetic variants can have a slightly larger effect on brain structure than on risk for disorders like schizophrenia, indicating a somewhat simpler genetic architecture. Also, gross brain structure measurements share a genetic basis with some, but not all, neuropsychiatric disorders, invalidating the previously held belief that they are broad endophenotypes, yet pinpointing brain regions likely involved in the pathology of specific disorders. Finally, we suggest that in order to build a more detailed mechanistic understanding of the effects of genetic variants on the brain, future directions in imaging genetics research will require observations of cellular and synaptic structure in specific brain regions beyond the resolution of magnetic resonance imaging. We expect that integrating genetic associations at biological levels from synapse to sulcus will reveal specific causal pathways impacting risk for neuropsychiatric disorders.


Subject(s)
Brain Diseases , Genetics , Genome-Wide Association Study , Mental Disorders , Neuroimaging , Brain Diseases/diagnostic imaging , Brain Diseases/genetics , Brain Diseases/pathology , Brain Diseases/physiopathology , Humans , Mental Disorders/diagnostic imaging , Mental Disorders/genetics , Mental Disorders/pathology , Mental Disorders/physiopathology
18.
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
19.
Nucleic Acids Res ; 43(10): e65, 2015 May 26.
Article in English | MEDLINE | ID: mdl-25765640

ABSTRACT

Isogenic pluripotent stem cells are critical tools for studying human neurological diseases by allowing one to study the effects of a mutation in a fixed genetic background. Of particular interest are the spectrum of autism disorders, some of which are monogenic such as Timothy syndrome (TS); others are multigenic such as the microdeletion and microduplication syndromes of the 16p11.2 chromosomal locus. Here, we report engineered human embryonic stem cell (hESC) lines for modeling these two disorders using locus-specific endonucleases to increase the efficiency of homology-directed repair (HDR). We developed a system to: (1) computationally identify unique transcription activator-like effector nuclease (TALEN) binding sites in the genome using a new software program, TALENSeek, (2) assemble the TALEN genes by combining golden gate cloning with modified constructs from the FLASH protocol, and (3) test the TALEN pairs in an amplification-based HDR assay that is more sensitive than the typical non-homologous end joining assay. We applied these methods to identify, construct, and test TALENs that were used with HDR donors in hESCs to generate an isogenic TS cell line in a scarless manner and to model the 16p11.2 copy number disorder without modifying genomic loci with high sequence similarity.


Subject(s)
Cell Engineering , Child Development Disorders, Pervasive/genetics , Embryonic Stem Cells , Models, Genetic , Autistic Disorder , Binding Sites , Cell Line , Deoxyribonucleases/metabolism , Gene Targeting , Genome, Human , Humans , Long QT Syndrome/genetics , Recombinational DNA Repair , Software , Syndactyly/genetics
20.
Hum Brain Mapp ; 37(5): 1788-800, 2016 May.
Article in English | MEDLINE | ID: mdl-26890892

ABSTRACT

Genome-wide association screens aim to identify common genetic variants contributing to the phenotypic variability of complex traits, such as human height or brain morphology. The identified genetic variants are mostly within noncoding genomic regions and the biology of the genotype-phenotype association typically remains unclear. In this article, we propose a complementary targeted strategy to reveal the genetic underpinnings of variability in subcortical brain volumes, by specifically selecting genomic loci that are experimentally validated forebrain enhancers, active in early embryonic development. We hypothesized that genetic variation within these enhancers may affect the development and ultimately the structure of subcortical brain regions in adults. We tested whether variants in forebrain enhancer regions showed an overall enrichment of association with volumetric variation in subcortical structures of >13,000 healthy adults. We observed significant enrichment of genomic loci that affect the volume of the hippocampus within forebrain enhancers (empirical P = 0.0015), a finding which robustly passed the adjusted threshold for testing of multiple brain phenotypes (cutoff of P < 0.0083 at an alpha of 0.05). In analyses of individual single nucleotide polymorphisms (SNPs), we identified an association upstream of the ID2 gene with rs7588305 and variation in hippocampal volume. This SNP-based association survived multiple-testing correction for the number of SNPs analyzed but not for the number of subcortical structures. Targeting known regulatory regions offers a way to understand the underlying biology that connects genotypes to phenotypes, particularly in the context of neuroimaging genetics. This biology-driven approach generates testable hypotheses regarding the functional biology of identified associations. Hum Brain Mapp 37:1788-1800, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
B7 Antigens/genetics , Brain/anatomy & histology , Brain/growth & development , Enhancer Elements, Genetic/genetics , Polymorphism, Single Nucleotide/genetics , Brain/diagnostic imaging , Genome-Wide Association Study , Humans , Neuroimaging , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL