Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
2.
Am J Hum Genet ; 109(10): 1789-1813, 2022 10 06.
Article in English | MEDLINE | ID: mdl-36152629

ABSTRACT

Chromosome 16p11.2 reciprocal genomic disorder, resulting from recurrent copy-number variants (CNVs), involves intellectual disability, autism spectrum disorder (ASD), and schizophrenia, but the responsible mechanisms are not known. To systemically dissect molecular effects, we performed transcriptome profiling of 350 libraries from six tissues (cortex, cerebellum, striatum, liver, brown fat, and white fat) in mouse models harboring CNVs of the syntenic 7qF3 region, as well as cellular, transcriptional, and single-cell analyses in 54 isogenic neural stem cell, induced neuron, and cerebral organoid models of CRISPR-engineered 16p11.2 CNVs. Transcriptome-wide differentially expressed genes were largely tissue-, cell-type-, and dosage-specific, although more effects were shared between deletion and duplication and across tissue than expected by chance. The broadest effects were observed in the cerebellum (2,163 differentially expressed genes), and the greatest enrichments were associated with synaptic pathways in mouse cerebellum and human induced neurons. Pathway and co-expression analyses identified energy and RNA metabolism as shared processes and enrichment for ASD-associated, loss-of-function constraint, and fragile X messenger ribonucleoprotein target gene sets. Intriguingly, reciprocal 16p11.2 dosage changes resulted in consistent decrements in neurite and electrophysiological features, and single-cell profiling of organoids showed reciprocal alterations to the proportions of excitatory and inhibitory GABAergic neurons. Changes both in neuronal ratios and in gene expression in our organoid analyses point most directly to calretinin GABAergic inhibitory neurons and the excitatory/inhibitory balance as targets of disruption that might contribute to changes in neurodevelopmental and cognitive function in 16p11.2 carriers. Collectively, our data indicate the genomic disorder involves disruption of multiple contributing biological processes and that this disruption has relative impacts that are context specific.


Subject(s)
Autism Spectrum Disorder , Chromosome Disorders , Intellectual Disability , Animals , Autism Spectrum Disorder/genetics , Calbindin 2/genetics , Cerebral Cortex , Chromosome Deletion , Chromosome Disorders/genetics , Chromosomes, Human, Pair 16/genetics , DNA Copy Number Variations , Genomics , Humans , Intellectual Disability/genetics , Mice , Neurons , RNA
3.
J Genet Genomics ; 49(7): 654-665, 2022 07.
Article in English | MEDLINE | ID: mdl-34896608

ABSTRACT

Familial dysautonomia (FD), a hereditary sensory and autonomic neuropathy, is caused by a mutation in the Elongator complex protein 1 (ELP1) gene that leads to a tissue-specific reduction of ELP1 protein. Our work to generate a phenotypic mouse model for FD headed to the discovery that homozygous deletion of the mouse Elp1 gene leads to embryonic lethality prior to mid-gestation. Given that FD is caused by a reduction, not loss, of ELP1, we generated two new mouse models by introducing different copy numbers of the human FD ELP1 transgene into the Elp1 knockout mouse (Elp1-/-) and observed that human ELP1 expression rescues embryonic development in a dose-dependent manner. We then conducted a comprehensive transcriptome analysis in mouse embryos to identify genes and pathways whose expression correlates with the amount of ELP1. We found that ELP1 is essential for the expression of genes responsible for nervous system development. Further, gene length analysis of the differentially expressed genes showed that the loss of Elp1 mainly impacts the expression of long genes and that by gradually restoring Elongator, their expression is progressively rescued. Finally, through evaluation of co-expression modules, we identified gene sets with unique expression patterns that depended on ELP1 expression.


Subject(s)
Carrier Proteins , Dysautonomia, Familial , Animals , Carrier Proteins/genetics , Disease Models, Animal , Dysautonomia, Familial/genetics , Dysautonomia, Familial/metabolism , Gene Expression , Homozygote , Humans , Mice , Sequence Deletion
4.
Redox Biol ; 48: 102091, 2021 12.
Article in English | MEDLINE | ID: mdl-34417156

ABSTRACT

Ventilatory support, such as supplemental oxygen, used to save premature infants impairs the growth of the pulmonary microvasculature and distal alveoli, leading to bronchopulmonary dysplasia (BPD). Although lung cellular composition changes with exposure to hyperoxia in neonatal mice, most human BPD survivors are weaned off oxygen within the first weeks to months of life, yet they may have persistent lung injury and pulmonary dysfunction as adults. We hypothesized that early-life hyperoxia alters the cellular landscape in later life and predicts long-term lung injury. Using single-cell RNA sequencing, we mapped lung cell subpopulations at postnatal day (pnd)7 and pnd60 in mice exposed to hyperoxia (95% O2) for 3 days as neonates. We interrogated over 10,000 cells and identified a total of 45 clusters within 32 cell states. Neonatal hyperoxia caused persistent compositional changes in later life (pnd60) in all five type II cell states with unique signatures and function. Premature infants requiring mechanical ventilation with different durations also showed similar alterations in these unique signatures of type II cell states. Pathologically, neonatal hyperoxic exposure caused alveolar simplification in adult mice. We conclude that neonatal hyperoxia alters the lung cellular landscape in later life, uncovering neonatal programing of adult lung dysfunction.


Subject(s)
Bronchopulmonary Dysplasia , Hyperoxia , Adult , Animals , Animals, Newborn , Bronchopulmonary Dysplasia/genetics , Humans , Infant, Newborn , Lung , Mice , Pulmonary Alveoli , Transcriptome
5.
Nat Commun ; 12(1): 3332, 2021 06 07.
Article in English | MEDLINE | ID: mdl-34099697

ABSTRACT

Pre-mRNA splicing is a key controller of human gene expression. Disturbances in splicing due to mutation lead to dysregulated protein expression and contribute to a substantial fraction of human disease. Several classes of splicing modulator compounds (SMCs) have been recently identified and establish that pre-mRNA splicing represents a target for therapy. We describe herein the identification of BPN-15477, a SMC that restores correct splicing of ELP1 exon 20. Using transcriptome sequencing from treated fibroblast cells and a machine learning approach, we identify BPN-15477 responsive sequence signatures. We then leverage this model to discover 155 human disease genes harboring ClinVar mutations predicted to alter pre-mRNA splicing as targets for BPN-15477. Splicing assays confirm successful correction of splicing defects caused by mutations in CFTR, LIPA, MLH1 and MAPT. Subsequent validations in two disease-relevant cellular models demonstrate that BPN-15477 increases functional protein, confirming the clinical potential of our predictions.


Subject(s)
Deep Learning , Gene Targeting/methods , RNA Splicing , Animals , Computational Biology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Exons , HEK293 Cells , Humans , Mice , Mice, Transgenic , MutL Protein Homolog 1/genetics , Mutation , Phenethylamines/administration & dosage , Pyridazines/administration & dosage , Sterol Esterase/genetics , Transcriptome , tau Proteins/genetics
6.
R I Med J (2013) ; 104(2): 54-59, 2021 Mar 01.
Article in English | MEDLINE | ID: mdl-33648321

ABSTRACT

We provide a program update on the COBRE Center for the Computational Biology of Human Disease (CBHD) at Brown University and affiliated hospitals. High throughput data from multiple 'omics-level' technologies are fundamental factors in identifying and treating human disease. The acquisition of these data is now straightforward, but the efficient and creative interpretation of these data remains a serious impediment to progress for faculty at all levels in both the basic and translational aspects of biomedical science. The CBHD COBRE seeks to build close collaboration between laboratory scientists working with model systems and data scientists working with computational and bioinformatics tools that can accelerate human disease research implementation. We describe the accomplishments of junior faculty Project Leaders (9) and Pilots Project leaders (8) and the objectives of the CBHD COBRE's core facility: The Computational Biology Core (CBC). To extend the CBHD COBRE's reach in the future, we encourage one and all to visit the CBHD COBRE and bring your data sets and questions. Only by engaging with new people and challenges can the program grow to serve the broader biomedical research community in the State of Rhode Island.


Subject(s)
Biomedical Research , Computational Biology , Computational Biology/education , Humans , Rhode Island , Universities
7.
Front Genet ; 12: 765985, 2021.
Article in English | MEDLINE | ID: mdl-35719905

ABSTRACT

Preeclampsia is a hypertensive disorder of pregnancy, which complicates up to 15% of US deliveries. It is an idiopathic disorder associated with several different phenotypes. We sought to determine if the genetic architecture of preeclampsia can be described by clusters of patients with variants in genes in shared protein interaction networks. We performed a case-control study using whole exome sequencing on early onset preeclamptic mothers with severe clinical features and control mothers with uncomplicated pregnancies between 2016 and 2020. A total of 143 patients were enrolled, 61 women with early onset preeclampsia with severe features based on ACOG criteria, and 82 control women at term, matched for race and ethnicity. A network analysis and visualization tool, Proteinarium, was used to confirm there are clusters of patients with shared gene networks associated with severe preeclampsia. The majority of the sequenced patients appear in two significant clusters. We identified one case dominant and one control dominant cluster. Thirteen genes were unique to the case dominated cluster. Among these genes, LAMB2, PTK2, RAC1, QSOX1, FN1, and VCAM1 have known associations with the pathogenic mechanisms of preeclampsia. Using bioinformatic analysis, we were able to identify subsets of patients with shared protein interaction networks, thus confirming our hypothesis about the genetic architecture of preeclampsia.

8.
Database (Oxford) ; 20202020 11 28.
Article in English | MEDLINE | ID: mdl-33247936

ABSTRACT

Advances in tumor genome sequencing created an urgent need for bioinformatics tools to support the interpretation of the clinical significance of the variants detected. VarStack is a web tool which is a base to retrieve somatic variant data relating to cancer from existing databases. VarStack incorporates data from several publicly available databases and presents them with an easy-to-navigate user interface. It currently supports data from the Catalogue of Somatic Mutations in Cancer, gnomAD, cBioPortal, ClinVar, OncoKB, CiViC and UCSC Genome Browser. It retrieves the data from these databases and returns them back to the user in a fraction of the time it would take to manually navigate each site independently. Users submit a variant with a gene symbol, peptide change and coding sequence change. They may select a variety of tumor-specific studies in cBioPortal to search through in addition to their original query. The results from the databases are presented in tabs. Users can export the results as an Excel file. VarStack also has the batch search feature in which the user can submit a list of variants and download an Excel file with the data from the databases. With the batch search and data download options, users can easily incorporate VarStack into their workflow or tools. VarStack saves time by providing somatic variant information to the user from multiple databases in an easy-to-export and interpretable format. VarStack is freely available under https://varstack.brown.edu.


Subject(s)
Neoplasms , User-Computer Interface , Computational Biology , Databases, Genetic , Humans , Information Storage and Retrieval , Internet , Neoplasms/genetics , Software
9.
Mol Autism ; 11(1): 45, 2020 06 05.
Article in English | MEDLINE | ID: mdl-32503625

ABSTRACT

BACKGROUND: MBD5, encoding the methyl-CpG-binding domain 5 protein, has been proposed as a necessary and sufficient driver of the 2q23.1 microdeletion syndrome. De novo missense and protein-truncating variants from exome sequencing studies have directly implicated MBD5 in the etiology of autism spectrum disorder (ASD) and related neurodevelopmental disorders (NDDs). However, little is known concerning the specific function(s) of MBD5. METHODS: To gain insight into the complex interactions associated with alteration of MBD5 in individuals with ASD and related NDDs, we explored the transcriptional landscape of MBD5 haploinsufficiency across multiple mouse brain regions of a heterozygous hypomorphic Mbd5+/GT mouse model, and compared these results to CRISPR-mediated mutations of MBD5 in human iPSC-derived neuronal models. RESULTS: Gene expression analyses across three brain regions from Mbd5+/GT mice showed subtle transcriptional changes, with cortex displaying the most widespread changes following Mbd5 reduction, indicating context-dependent effects. Comparison with MBD5 reduction in human neuronal cells reinforced the context-dependence of gene expression changes due to MBD5 deficiency. Gene co-expression network analyses revealed gene clusters that were associated with reduced MBD5 expression and enriched for terms related to ciliary function. LIMITATIONS: These analyses included a limited number of mouse brain regions and neuronal models, and the effects of the gene knockdown are subtle. As such, these results will not reflect the full extent of MBD5 disruption across human brain regions during early neurodevelopment in ASD, or capture the diverse spectrum of cell-type-specific changes associated with MBD5 alterations. CONCLUSIONS: Our study points to modest and context-dependent transcriptional consequences of Mbd5 disruption in the brain. It also suggests a possible link between MBD5 and perturbations in ciliary function, which is an established pathogenic mechanism in developmental disorders and syndromes.


Subject(s)
Brain/metabolism , Methyl-CpG-Binding Protein 2/genetics , Mutation , Neurons/metabolism , Transcription, Genetic , Animals , Autism Spectrum Disorder/etiology , Autism Spectrum Disorder/metabolism , CRISPR-Cas Systems , Cell Differentiation/genetics , Cell Line , Disease Models, Animal , Gene Expression Regulation, Developmental , Gene Targeting , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mice , Mice, Transgenic , Neurons/cytology
10.
Cell ; 172(5): 897-909.e21, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29474918

ABSTRACT

X-linked Dystonia-Parkinsonism (XDP) is a Mendelian neurodegenerative disease that is endemic to the Philippines and is associated with a founder haplotype. We integrated multiple genome and transcriptome assembly technologies to narrow the causal mutation to the TAF1 locus, which included a SINE-VNTR-Alu (SVA) retrotransposition into intron 32 of the gene. Transcriptome analyses identified decreased expression of the canonical cTAF1 transcript among XDP probands, and de novo assembly across multiple pluripotent stem-cell-derived neuronal lineages discovered aberrant TAF1 transcription that involved alternative splicing and intron retention (IR) in proximity to the SVA that was anti-correlated with overall TAF1 expression. CRISPR/Cas9 excision of the SVA rescued this XDP-specific transcriptional signature and normalized TAF1 expression in probands. These data suggest an SVA-mediated aberrant transcriptional mechanism associated with XDP and may provide a roadmap for layered technologies and integrated assembly-based analyses for other unsolved Mendelian disorders.


Subject(s)
Dystonic Disorders/genetics , Genetic Diseases, X-Linked/genetics , Genome, Human , Transcriptome/genetics , Alternative Splicing/genetics , Alu Elements/genetics , Base Sequence , CRISPR-Cas Systems/genetics , Cohort Studies , Family , Female , Genetic Loci , Haplotypes/genetics , High-Throughput Nucleotide Sequencing , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Introns/genetics , Male , Minisatellite Repeats/genetics , Models, Genetic , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Neural Stem Cells/metabolism , Neurons/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Short Interspersed Nucleotide Elements , TATA-Binding Protein Associated Factors/genetics , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/genetics , Transcription Factor TFIID/metabolism
11.
Am J Med Genet A ; 173(9): 2478-2484, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28691782

ABSTRACT

Genetic alterations of ARID1B have been recently recognized as one of the most common mendelian causes of intellectual disability and are associated with both syndromic and non-syndromic phenotypes. The ARID1B protein, a subunit of the chromatin remodeling complex SWI/SNF-A, is involved in the regulation of transcription and multiple downstream cellular processes. We report here the clinical, genetic, and proteomic phenotypes of an individual with a unique apparent de novo mutation of ARID1B due to an intragenic duplication. His neurodevelopmental phenotype includes a severe speech/language disorder with full scale IQ scores 78-98 and scattered academic skill levels, expanding the phenotypic spectrum of ARID1B mutations. Haploinsufficiency of ARID1B was determined both by RNA sequencing and quantitative RT-PCR. Fluorescence in situ hybridization analysis supported an intragenic localization of the ARID1B copy number gain. Principal component analysis revealed marked differentiation of the subject's lymphoblast proteome from that of controls. Of 3426 proteins quantified, 1014 were significantly up- or down-regulated compared to controls (q < 0.01). Pathway analysis revealed highly significant enrichment for canonical pathways of EIF2 and EIF4 signaling, protein ubiquitination, tRNA charging and chromosomal replication, among others. Network analyses revealed down-regulation of: (1) intracellular components involved in organization of membranes, organelles, and vesicles; (2) aspects of cell cycle control, signal transduction, and nuclear protein export; (3) ubiquitination and proteosomal function; and (4) aspects of mRNA synthesis/splicing. Further studies are needed to determine the detailed molecular and cellular mechanisms by which constitutional haploinsufficiency of ARID1B causes syndromic and non-syndromic developmental disabilities.


Subject(s)
DNA-Binding Proteins/genetics , Developmental Disabilities/genetics , Hand Deformities, Congenital/genetics , Intellectual Disability/genetics , Transcription Factors/genetics , Abnormalities, Multiple , Adolescent , Developmental Disabilities/physiopathology , Face/physiopathology , Gene Duplication/genetics , Hand Deformities, Congenital/physiopathology , Haploinsufficiency/genetics , Humans , In Situ Hybridization, Fluorescence , Intellectual Disability/physiopathology , Male , Mutation , Proteomics
12.
Genome Biol ; 18(1): 36, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28260531

ABSTRACT

BACKGROUND: Structural variation (SV) influences genome organization and contributes to human disease. However, the complete mutational spectrum of SV has not been routinely captured in disease association studies. RESULTS: We sequenced 689 participants with autism spectrum disorder (ASD) and other developmental abnormalities to construct a genome-wide map of large SV. Using long-insert jumping libraries at 105X mean physical coverage and linked-read whole-genome sequencing from 10X Genomics, we document seven major SV classes at ~5 kb SV resolution. Our results encompass 11,735 distinct large SV sites, 38.1% of which are novel and 16.8% of which are balanced or complex. We characterize 16 recurrent subclasses of complex SV (cxSV), revealing that: (1) cxSV are larger and rarer than canonical SV; (2) each genome harbors 14 large cxSV on average; (3) 84.4% of large cxSVs involve inversion; and (4) most large cxSV (93.8%) have not been delineated in previous studies. Rare SVs are more likely to disrupt coding and regulatory non-coding loci, particularly when truncating constrained and disease-associated genes. We also identify multiple cases of catastrophic chromosomal rearrangements known as chromoanagenesis, including somatic chromoanasynthesis, and extreme balanced germline chromothripsis events involving up to 65 breakpoints and 60.6 Mb across four chromosomes, further defining rare categories of extreme cxSV. CONCLUSIONS: These data provide a foundational map of large SV in the morbid human genome and demonstrate a previously underappreciated abundance and diversity of cxSV that should be considered in genomic studies of human disease.


Subject(s)
Chromosome Aberrations , Chromosome Inversion , Chromothripsis , Genome, Human , Genomics , Autism Spectrum Disorder/genetics , Gene Order , Gene Rearrangement , Genetic Predisposition to Disease , Genomics/methods , High-Throughput Nucleotide Sequencing , Humans , Mutation
13.
Nat Neurosci ; 19(3): 517-22, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26829649

ABSTRACT

Recurrent, reciprocal genomic disorders resulting from non-allelic homologous recombination (NAHR) between near-identical segmental duplications (SDs) are a major cause of human disease, often producing phenotypically distinct syndromes. The genomic architecture of flanking SDs presents a challenge for modeling these syndromes; however, the capability to efficiently generate reciprocal copy number variants (CNVs) that mimic NAHR would represent a valuable modeling tool. We describe here a CRISPR/Cas9 genome engineering method, single-guide CRISPR/Cas targeting of repetitive elements (SCORE), to model reciprocal genomic disorders and demonstrate its capabilities by generating reciprocal CNVs of 16p11.2 and 15q13.3, including alteration of one copy-equivalent of the SDs that mediate NAHR in vivo. The method is reproducible, and RNA sequencing reliably clusters transcriptional signatures from human subjects with in vivo CNVs and their corresponding in vitro models. This new approach will provide broad applicability for the study of genomic disorders and, with further development, may also permit efficient correction of these defects.


Subject(s)
Autistic Disorder/genetics , CRISPR-Cas Systems/genetics , Chromosome Disorders/genetics , DNA Copy Number Variations/genetics , Genetic Engineering/methods , Intellectual Disability/genetics , Segmental Duplications, Genomic/genetics , Seizures/genetics , Sequence Deletion/genetics , Chromosome Deletion , Chromosomes, Human, Pair 15/genetics , Chromosomes, Human, Pair 16/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Humans
14.
Hum Mol Genet ; 24(9): 2442-57, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25574027

ABSTRACT

The CAG repeat expansion in the Huntington's disease gene HTT extends a polyglutamine tract in mutant huntingtin that enhances its ability to facilitate polycomb repressive complex 2 (PRC2). To gain insight into this dominant gain of function, we mapped histone modifications genome-wide across an isogenic panel of mouse embryonic stem cell (ESC) and neuronal progenitor cell (NPC) lines, comparing the effects of Htt null and different size Htt CAG mutations. We found that Htt is required in ESC for the proper deposition of histone H3K27me3 at a subset of 'bivalent' loci but in NPC it is needed at 'bivalent' loci for both the proper maintenance and the appropriate removal of this mark. In contrast, Htt CAG size, though changing histone H3K27me3, is prominently associated with altered histone H3K4me3 at 'active' loci. The sets of ESC and NPC genes with altered histone marks delineated by the lack of huntingtin or the presence of mutant huntingtin, though distinct, are enriched in similar pathways with apoptosis specifically highlighted for the CAG mutation. Thus, the manner by which huntingtin function facilitates PRC2 may afford mutant huntingtin with multiple opportunities to impinge upon the broader machinery that orchestrates developmentally appropriate chromatin status.


Subject(s)
Chromatin/genetics , Chromatin/metabolism , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Trinucleotide Repeat Expansion , Alleles , Animals , Chromatin Assembly and Disassembly , Chromatin Immunoprecipitation , Cluster Analysis , Embryonic Stem Cells/metabolism , Gene Deletion , Gene Expression Regulation , Genome-Wide Association Study , Genotype , High-Throughput Nucleotide Sequencing , Histones/metabolism , Huntingtin Protein , Mice , Mice, Transgenic , Nerve Tissue Proteins/chemistry , Neural Stem Cells/metabolism , Nuclear Proteins/chemistry , Polycomb Repressive Complex 2/genetics
15.
Proc Natl Acad Sci U S A ; 111(42): E4468-77, 2014 Oct 21.
Article in English | MEDLINE | ID: mdl-25294932

ABSTRACT

Truncating mutations of chromodomain helicase DNA-binding protein 8 (CHD8), and of many other genes with diverse functions, are strong-effect risk factors for autism spectrum disorder (ASD), suggesting multiple mechanisms of pathogenesis. We explored the transcriptional networks that CHD8 regulates in neural progenitor cells (NPCs) by reducing its expression and then integrating transcriptome sequencing (RNA sequencing) with genome-wide CHD8 binding (ChIP sequencing). Suppressing CHD8 to levels comparable with the loss of a single allele caused altered expression of 1,756 genes, 64.9% of which were up-regulated. CHD8 showed widespread binding to chromatin, with 7,324 replicated sites that marked 5,658 genes. Integration of these data suggests that a limited array of direct regulatory effects of CHD8 produced a much larger network of secondary expression changes. Genes indirectly down-regulated (i.e., without CHD8-binding sites) reflect pathways involved in brain development, including synapse formation, neuron differentiation, cell adhesion, and axon guidance, whereas CHD8-bound genes are strongly associated with chromatin modification and transcriptional regulation. Genes associated with ASD were strongly enriched among indirectly down-regulated loci (P < 10(-8)) and CHD8-bound genes (P = 0.0043), which align with previously identified coexpression modules during fetal development. We also find an intriguing enrichment of cancer-related gene sets among CHD8-bound genes (P < 10(-10)). In vivo suppression of chd8 in zebrafish produced macrocephaly comparable to that of humans with inactivating mutations. These data indicate that heterozygous disruption of CHD8 precipitates a network of gene-expression changes involved in neurodevelopmental pathways in which many ASD-associated genes may converge on shared mechanisms of pathogenesis.


Subject(s)
Child Development Disorders, Pervasive/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Gene Expression Regulation, Developmental , Neural Stem Cells/physiology , Transcription Factors/genetics , Transcription Factors/physiology , Zebrafish Proteins/physiology , Animals , Axons/metabolism , Binding Sites , Child Development Disorders, Pervasive/metabolism , Chromatin/metabolism , DNA Helicases/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Genome , Heterozygote , Humans , Megalencephaly/metabolism , Mutation , Neoplasms/metabolism , Neurons/metabolism , Protein Binding , Risk Factors , Sequence Analysis, RNA , Software , Zebrafish , Zebrafish Proteins/genetics
16.
Cell Stem Cell ; 15(1): 27-30, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24996167

ABSTRACT

Genome editing has attracted wide interest for the generation of cellular models of disease using human pluripotent stem cells and other cell types. CRISPR-Cas systems and TALENs can target desired genomic sites with high efficiency in human cells, but recent publications have led to concern about the extent to which these tools may cause off-target mutagenic effects that could potentially confound disease-modeling studies. Using CRISPR-Cas9 and TALEN targeted human pluripotent stem cell clones, we performed whole-genome sequencing at high coverage in order to assess the degree of mutagenesis across the entire genome. In both types of clones, we found that off-target mutations attributable to the nucleases were very rare. From this analysis, we suggest that, although some cell types may be at risk for off-target mutations, the incidence of such effects in human pluripotent stem cells may be sufficiently low and thus not a significant concern for disease modeling and other applications.


Subject(s)
CRISPR-Cas Systems/genetics , Endonucleases/metabolism , Pluripotent Stem Cells/physiology , Sequence Analysis, DNA/methods , Adaptor Proteins, Vesicular Transport/genetics , Base Sequence , Clone Cells , Endonucleases/genetics , Genetic Engineering , Genome/genetics , Humans , Incidence , Molecular Sequence Data , Mutation/genetics , Organ Specificity
17.
Am J Hum Genet ; 94(6): 870-83, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24906019

ABSTRACT

Reciprocal copy-number variation (CNV) of a 593 kb region of 16p11.2 is a common genetic cause of autism spectrum disorder (ASD), yet it is not completely penetrant and can manifest in a wide array of phenotypes. To explore its molecular consequences, we performed RNA sequencing of cerebral cortex from mouse models with CNV of the syntenic 7qF3 region and lymphoblast lines from 34 members of 7 multiplex ASD-affected families harboring the 16p11.2 CNV. Expression of all genes in the CNV region correlated well with their DNA copy number, with no evidence of dosage compensation. We observed effects on gene expression outside the CNV region, including apparent positional effects in cis and in trans at genomic segments with evidence of physical interaction in Hi-C chromosome conformation data. One of the most significant positional effects was telomeric to the 16p11.2 CNV and includes the previously described "distal" 16p11.2 microdeletion. Overall, 16p11.2 CNV was associated with altered expression of genes and networks that converge on multiple hypotheses of ASD pathogenesis, including synaptic function (e.g., NRXN1, NRXN3), chromatin modification (e.g., CHD8, EHMT1, MECP2), transcriptional regulation (e.g., TCF4, SATB2), and intellectual disability (e.g., FMR1, CEP290). However, there were differences between tissues and species, with the strongest effects being consistently within the CNV region itself. Our analyses suggest that through a combination of indirect regulatory effects and direct effects on nuclear architecture, alteration of 16p11.2 genes disrupts expression networks that involve other genes and pathways known to contribute to ASD, suggesting an overlap in mechanisms of pathogenesis.


Subject(s)
Autistic Disorder/genetics , Chromosome Deletion , Chromosome Duplication , Chromosomes, Human, Pair 16/genetics , Animals , Cerebral Cortex/pathology , Child , DNA Copy Number Variations , Female , Genome-Wide Association Study , Genotype , Humans , Intellectual Disability/genetics , Male , Mice , Pedigree , Phenotype , Sequence Analysis, RNA , Transcription, Genetic
18.
Ecol Appl ; 22(5): 1497-511, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22908709

ABSTRACT

As population modeling is increasingly called upon to guide policy and management, it is important that we understand not only the central tendencies of our study systems, but the consequences of their variation in space and time as well. The invasive plant Alliaria petiolata (garlic mustard) is actively managed in the United States and is the focus of a developing biological control program. Two weevils (Coleoptera: Curculionidae: Ceutorhynchus) that reduce fecundity (C. alliariae) and rosette survival plus fecundity (C. scrobicollis) are under consideration for release pending host specificity testing. We used a demographic modeling approach to (1) quantify variability in A. petiolata growth and vital rates and (2) assess the potential for single- or multiple-agent biocontrol to suppress growth of 12 A. petiolata populations in Illinois and Michigan studied over three plant generations. We used perturbation analyses and simulation models with stochastic environments to estimate stochastic growth rates (lambda(S)) and predict the probability of successful management using either a single biocontrol agent or two agent species together. Not all populations exhibited invasive dynamics. Estimates of lambda(S) ranged from 0.78 to 2.21 across sites, while annual, deterministic growth (lambda) varied up to sevenfold within individual sites. Given our knowledge of the biocontrol agents, this analysis suggests that C. scrobicollis alone may control A. petiolata at up to 63% of our study sites where lambda >1, with the combination of both agents predicted to succeed at 88% of sites. Across sites and years, the elasticity rankings were dependent on lambda. Reductions of rosette survival, fecundity, or germination of new seeds are predicted to cause the greatest reduction of lambda in growing populations. In declining populations, transitions affecting seed bank survival have the greatest effect on lambda. This contrasts with past analyses that varied parameters individually in an otherwise constant matrix, which may yield unrealistic predictions by decoupling natural parameter covariances. Overall, comparisons of stochastic and deterministic growth rates illustrate how analyses of individual populations or years could misguide management or fail to characterize complex traits such as invasiveness that emerge as attributes of populations rather than species.


Subject(s)
Brassicaceae/physiology , Introduced Species , Models, Biological , Stochastic Processes , Linear Models , Population Growth , Time Factors
19.
Front Genet ; 3: 29, 2012.
Article in English | MEDLINE | ID: mdl-22403584

ABSTRACT

Most biological traits are regulated by a complex interplay between genetic and environmental factors. By intercrossing divergent lines, it is possible to identify individual and interacting QTL involved in the genetic architecture of these traits. When the loci have been mapped, alternative strategies are needed for fine-mapping and studying the individual and interactive effects of the QTL in detail. We have previously identified, replicated, and fine mapped a four-locus QTL network that determines nearly half of the eightfold difference in body weight at 56 days of age between two divergently selected chicken lines. Here, we describe, to our knowledge, the first generation of a three-locus QTL introgression line in chickens. Recurrent marker-assisted backcrossing was used to simultaneously transfer QTL alleles from the low-weight selected line into the high-weight selected line. Three generations of backcrossing and one generation of intercrossing resulted in an introgression line where all three introgressed QTL and several unlinked and linked control-loci were segregating at nearly expected allele frequencies. We show how intensive selection can be applied using artificial insemination to rapidly generate a multi-locus introgression line and provide recommendations for future breeding of introgression lines. This confirmed introgression line will facilitate later detailed studies of the effects of genetic interactions on complex traits in this population, including growth, and body-composition traits.

20.
Mol Ecol ; 20(11): 2341-52, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21521393

ABSTRACT

Disaccord between the supply and demand of energy (carbon, C) and certain material elements (e.g. phosphorus, P) across trophic levels is common in most ecosystems and impacts the strength of trophic interactions and ecosystem functions such as productivity and nutrient recycling. Yet, we know little about mechanisms operating at the lower levels of biological organization that drive such higher-level ecological processes. Such information should help refine theories integrating biological processes at multiple levels of organization. Understanding the expression and functions of genes that underlie (to a large degree) physiological adjustments made by organisms to stoichiometric imbalances at trophic interfaces is a first step in this enterprise. Here, we investigate adjustments in gene expression to varying supply and demand of phosphorus relative to other dietary components in the keystone limnetic herbivore, Daphnia pulex. Daphniids were fed an algal diet of either LoC-HiP (molar C:P ∼100) or HiC-LoP (molar C:P ∼900) for 5 days, resulting in significant growth reductions under HiC-LoP conditions. Microarrays measured the transcriptional regulation of 8217 annotated protein-coding genes under contrasting dietary conditions and revealed 1818 differentially expressed (DE) genes; 19% are genes unique to the Daphnia lineage. We mapped DE genes onto a global chart of metabolic pathways to obtain a systems-level perspective on the responses to stoichiometric imbalances. Daphnia differentially regulated pathways were involved in sequestering limiting elements, and in dealing with the products of metabolic adjustments that may be triggered by nutrient stress in primary producers. Functional genomics at trophic interfaces illuminate the complexity of processes underlying stoichiometric constraints on energy and nutrient fluxes in ecosystems.


Subject(s)
Daphnia/genetics , Food Chain , Genomics/methods , Animals , Carbon/metabolism , Daphnia/growth & development , Gene Expression Profiling , Gene Expression Regulation , Metabolic Networks and Pathways/genetics , Oligonucleotide Array Sequence Analysis , Phosphorus/metabolism , Scenedesmus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL