Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Science ; 384(6698): eadh0829, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38781368

ABSTRACT

Neuropsychiatric genome-wide association studies (GWASs), including those for autism spectrum disorder and schizophrenia, show strong enrichment for regulatory elements in the developing brain. However, prioritizing risk genes and mechanisms is challenging without a unified regulatory atlas. Across 672 diverse developing human brains, we identified 15,752 genes harboring gene, isoform, and/or splicing quantitative trait loci, mapping 3739 to cellular contexts. Gene expression heritability drops during development, likely reflecting both increasing cellular heterogeneity and the intrinsic properties of neuronal maturation. Isoform-level regulation, particularly in the second trimester, mediated the largest proportion of GWAS heritability. Through colocalization, we prioritized mechanisms for about 60% of GWAS loci across five disorders, exceeding adult brain findings. Finally, we contextualized results within gene and isoform coexpression networks, revealing the comprehensive landscape of transcriptome regulation in development and disease.


Subject(s)
Autism Spectrum Disorder , Brain , Genome-Wide Association Study , Protein Isoforms , Quantitative Trait Loci , Schizophrenia , Humans , Brain/metabolism , Brain/growth & development , Brain/embryology , Schizophrenia/genetics , Autism Spectrum Disorder/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Transcriptome , RNA Splicing , Gene Expression Regulation, Developmental , Alternative Splicing , Atlases as Topic , Gene Regulatory Networks
2.
Science ; 384(6698): eadh0559, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38781390

ABSTRACT

Nucleotide changes in gene regulatory elements are important determinants of neuronal development and diseases. Using massively parallel reporter assays in primary human cells from mid-gestation cortex and cerebral organoids, we interrogated the cis-regulatory activity of 102,767 open chromatin regions, including thousands of sequences with cell type-specific accessibility and variants associated with brain gene regulation. In primary cells, we identified 46,802 active enhancer sequences and 164 variants that alter enhancer activity. Activity was comparable in organoids and primary cells, suggesting that organoids provide an adequate model for the developing cortex. Using deep learning we decoded the sequence basis and upstream regulators of enhancer activity. This work establishes a comprehensive catalog of functional gene regulatory elements and variants in human neuronal development.


Subject(s)
Cerebral Cortex , Enhancer Elements, Genetic , Organoids , Humans , Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Cerebral Cortex/growth & development , Organoids/metabolism , Deep Learning , Chromatin/metabolism , Chromatin/genetics , Gene Expression Regulation, Developmental , Neurogenesis/genetics , Regulatory Sequences, Nucleic Acid , Neurons/metabolism
3.
Science ; 380(6643): eabm1696, 2023 04 28.
Article in English | MEDLINE | ID: mdl-37104607

ABSTRACT

Human accelerated regions (HARs) are conserved genomic loci that evolved at an accelerated rate in the human lineage and may underlie human-specific traits. We generated HARs and chimpanzee accelerated regions with an automated pipeline and an alignment of 241 mammalian genomes. Combining deep learning with chromatin capture experiments in human and chimpanzee neural progenitor cells, we discovered a significant enrichment of HARs in topologically associating domains containing human-specific genomic variants that change three-dimensional (3D) genome organization. Differential gene expression between humans and chimpanzees at these loci suggests rewiring of regulatory interactions between HARs and neurodevelopmental genes. Thus, comparative genomics together with models of 3D genome folding revealed enhancer hijacking as an explanation for the rapid evolution of HARs.


Subject(s)
Genetic Loci , Neurogenesis , Animals , Humans , Chromatin/genetics , Genome, Human , Genomics , Pan troglodytes/genetics , Neurogenesis/genetics , Deep Learning
4.
medRxiv ; 2023 Mar 06.
Article in English | MEDLINE | ID: mdl-36945630

ABSTRACT

Genomic regulatory elements active in the developing human brain are notably enriched in genetic risk for neuropsychiatric disorders, including autism spectrum disorder (ASD), schizophrenia, and bipolar disorder. However, prioritizing the specific risk genes and candidate molecular mechanisms underlying these genetic enrichments has been hindered by the lack of a single unified large-scale gene regulatory atlas of human brain development. Here, we uniformly process and systematically characterize gene, isoform, and splicing quantitative trait loci (xQTLs) in 672 fetal brain samples from unique subjects across multiple ancestral populations. We identify 15,752 genes harboring a significant xQTL and map 3,739 eQTLs to a specific cellular context. We observe a striking drop in gene expression and splicing heritability as the human brain develops. Isoform-level regulation, particularly in the second trimester, mediates the greatest proportion of heritability across multiple psychiatric GWAS, compared with eQTLs. Via colocalization and TWAS, we prioritize biological mechanisms for ~60% of GWAS loci across five neuropsychiatric disorders, nearly two-fold that observed in the adult brain. Finally, we build a comprehensive set of developmentally regulated gene and isoform co-expression networks capturing unique genetic enrichments across disorders. Together, this work provides a comprehensive view of genetic regulation across human brain development as well as the stage-and cell type-informed mechanistic underpinnings of neuropsychiatric disorders.

5.
bioRxiv ; 2023 Feb 16.
Article in English | MEDLINE | ID: mdl-36824845

ABSTRACT

Nucleotide changes in gene regulatory elements are important determinants of neuronal development and disease. Using massively parallel reporter assays in primary human cells from mid-gestation cortex and cerebral organoids, we interrogated the cis-regulatory activity of 102,767 sequences, including differentially accessible cell-type specific regions in the developing cortex and single-nucleotide variants associated with psychiatric disorders. In primary cells, we identified 46,802 active enhancer sequences and 164 disorder-associated variants that significantly alter enhancer activity. Activity was comparable in organoids and primary cells, suggesting that organoids provide an adequate model for the developing cortex. Using deep learning, we decoded the sequence basis and upstream regulators of enhancer activity. This work establishes a comprehensive catalog of functional gene regulatory elements and variants in human neuronal development.

6.
bioRxiv ; 2023 Jan 07.
Article in English | MEDLINE | ID: mdl-36711864

ABSTRACT

Chronic inflammation and tissue fibrosis are common stress responses that worsen organ function, yet the molecular mechanisms governing their crosstalk are poorly understood. In diseased organs, stress-induced changes in gene expression fuel maladaptive cell state transitions and pathological interaction between diverse cellular compartments. Although chronic fibroblast activation worsens dysfunction of lung, liver, kidney, and heart, and exacerbates many cancers, the stress-sensing mechanisms initiating the transcriptional activation of fibroblasts are not well understood. Here, we show that conditional deletion of the transcription co-activator Brd4 in Cx3cr1-positive myeloid cells ameliorates heart failure and is associated with a dramatic reduction in fibroblast activation. Analysis of single-cell chromatin accessibility and BRD4 occupancy in vivo in Cx3cr1-positive cells identified a large enhancer proximal to Interleukin-1 beta (Il1b), and a series of CRISPR deletions revealed the precise stress-dependent regulatory element that controlled expression of Il1b in disease. Secreted IL1B functioned non-cell autonomously to activate a p65/RELA-dependent enhancer near the transcription factor MEOX1, resulting in a profibrotic response in human cardiac fibroblasts. In vivo, antibody-mediated IL1B neutralization prevented stress-induced expression of MEOX1, inhibited fibroblast activation, and improved cardiac function in heart failure. The elucidation of BRD4-dependent crosstalk between a specific immune cell subset and fibroblasts through IL1B provides new therapeutic strategies for heart disease and other disorders of chronic inflammation and maladaptive tissue remodeling.

8.
Bioinform Adv ; 2(1): vbac051, 2022.
Article in English | MEDLINE | ID: mdl-35967929

ABSTRACT

Motivation: Unsupervised clustering of single-cell transcriptomics is a powerful method for identifying cell populations. Static visualization techniques for single-cell clustering only display results for a single resolution parameter. Analysts will often evaluate more than one resolution parameter but then only report one. Results: We developed Cell Layers, an interactive Sankey tool for the quantitative investigation of gene expression, co-expression, biological processes and cluster integrity across clustering resolutions. Cell Layers enhances the interpretability of single-cell clustering by linking molecular data and cluster evaluation metrics, providing novel insight into cell populations. Availability and implementation: https://github.com/apblair/CellLayers.

9.
Bioinformatics ; 38(9): 2621-2623, 2022 04 28.
Article in English | MEDLINE | ID: mdl-35274675

ABSTRACT

SUMMARY: CellWalkR is an R package that integrates single-cell open chromatin data with cell type labels and bulk epigenetic data to identify cell type-specific regulatory regions. A Graphics Processing Unit (GPU) implementation and downsampling strategies enable thousands of cells to be processed in seconds. CellWalkR's user-friendly interface provides interactive analysis and visualization of cell labels and regulatory region mappings. AVAILABILITY AND IMPLEMENTATION: CellWalkR is freely available as an R package under a GNU GPL-2.0 License and can be accessed from https://github.com/PFPrzytycki/CellWalkR with an accompanying vignette. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Regulatory Sequences, Nucleic Acid , Software , Data Interpretation, Statistical
10.
Nature ; 598(7879): 205-213, 2021 10.
Article in English | MEDLINE | ID: mdl-34616060

ABSTRACT

During mammalian development, differences in chromatin state coincide with cellular differentiation and reflect changes in the gene regulatory landscape1. In the developing brain, cell fate specification and topographic identity are important for defining cell identity2 and confer selective vulnerabilities to neurodevelopmental disorders3. Here, to identify cell-type-specific chromatin accessibility patterns in the developing human brain, we used a single-cell assay for transposase accessibility by sequencing (scATAC-seq) in primary tissue samples from the human forebrain. We applied unbiased analyses to identify genomic loci that undergo extensive cell-type- and brain-region-specific changes in accessibility during neurogenesis, and an integrative analysis to predict cell-type-specific candidate regulatory elements. We found that cerebral organoids recapitulate most putative cell-type-specific enhancer accessibility patterns but lack many cell-type-specific open chromatin regions that are found in vivo. Systematic comparison of chromatin accessibility across brain regions revealed unexpected diversity among neural progenitor cells in the cerebral cortex and implicated retinoic acid signalling in the specification of neuronal lineage identity in the prefrontal cortex. Together, our results reveal the important contribution of chromatin state to the emerging patterns of cell type diversity and cell fate specification and provide a blueprint for evaluating the fidelity and robustness of cerebral organoids as a model for cortical development.


Subject(s)
Brain/cytology , Epigenomics , Neurogenesis , Single-Cell Analysis , Atlases as Topic , Brain/growth & development , Brain/metabolism , Chromatin/chemistry , Chromatin/genetics , Chromatin/metabolism , Disease Susceptibility , Enhancer Elements, Genetic , Humans , Neurons/cytology , Neurons/metabolism , Organoids/cytology , Tretinoin/metabolism
11.
Nature ; 595(7867): 438-443, 2021 07.
Article in English | MEDLINE | ID: mdl-34163071

ABSTRACT

In diseased organs, stress-activated signalling cascades alter chromatin, thereby triggering maladaptive cell state transitions. Fibroblast activation is a common stress response in tissues that worsens lung, liver, kidney and heart disease, yet its mechanistic basis remains unclear1,2. Pharmacological inhibition of bromodomain and extra-terminal domain (BET) proteins alleviates cardiac dysfunction3-7, providing a tool to interrogate and modulate cardiac cell states as a potential therapeutic approach. Here we use single-cell epigenomic analyses of hearts dynamically exposed to BET inhibitors to reveal a reversible transcriptional switch that underlies the activation of fibroblasts. Resident cardiac fibroblasts demonstrated robust toggling between the quiescent and activated state in a manner directly correlating with BET inhibitor exposure and cardiac function. Single-cell chromatin accessibility revealed previously undescribed DNA elements, the accessibility of which dynamically correlated with cardiac performance. Among the most dynamic elements was an enhancer that regulated the transcription factor MEOX1, which was specifically expressed in activated fibroblasts, occupied putative regulatory elements of a broad fibrotic gene program and was required for TGFß-induced fibroblast activation. Selective CRISPR inhibition of the single most dynamic cis-element within the enhancer blocked TGFß-induced Meox1 activation. We identify MEOX1 as a central regulator of fibroblast activation associated with cardiac dysfunction and demonstrate its upregulation after activation of human lung, liver and kidney fibroblasts. The plasticity and specificity of BET-dependent regulation of MEOX1 in tissue fibroblasts provide previously unknown trans- and cis-targets for treating fibrotic disease.


Subject(s)
Enhancer Elements, Genetic , Fibroblasts/cytology , Heart Diseases/genetics , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Animals , Chromatin/metabolism , Epigenomics , Gene Expression Regulation , Humans , Mice , Proteins/antagonists & inhibitors , Single-Cell Analysis , Transcriptome , Transforming Growth Factor beta/metabolism
12.
Genome Biol ; 22(1): 61, 2021 02 14.
Article in English | MEDLINE | ID: mdl-33583425

ABSTRACT

Single-cell and bulk genomics assays have complementary strengths and weaknesses, and alone neither strategy can fully capture regulatory elements across the diversity of cells in complex tissues. We present CellWalker, a method that integrates single-cell open chromatin (scATAC-seq) data with gene expression (RNA-seq) and other data types using a network model that simultaneously improves cell labeling in noisy scATAC-seq and annotates cell type-specific regulatory elements in bulk data. We demonstrate CellWalker's robustness to sparse annotations and noise using simulations and combined RNA-seq and ATAC-seq in individual cells. We then apply CellWalker to the developing brain. We identify cells transitioning between transcriptional states, resolve regulatory elements to cell types, and observe that autism and other neurological traits can be mapped to specific cell types through their regulatory elements.


Subject(s)
Computational Biology/methods , Gene Expression Regulation , Regulatory Sequences, Nucleic Acid , Single-Cell Analysis , Software , Chromatin Immunoprecipitation Sequencing/methods , Genetic Loci , Molecular Sequence Annotation , Organ Specificity/genetics , Reproducibility of Results , Single-Cell Analysis/methods
13.
Cell Syst ; 10(2): 193-203.e4, 2020 02 26.
Article in English | MEDLINE | ID: mdl-32078798

ABSTRACT

Identifying cancer-relevant mutations in noncoding regions is challenging due to the large numbers of such mutations, their low levels of recurrence, and difficulties in interpreting their functional impact. To uncover genes that are dysregulated due to somatic mutations in cis, we build upon the concept of differential allele-specific expression (ASE) and introduce methods to identify genes within an individual's cancer whose ASE differs from what is found in matched normal tissue. When applied to breast cancer tumor samples, our methods detect the known allele-specific effects of copy number variation and nonsense-mediated decay. Further, genes that are found to recurrently exhibit differential ASE across samples are cancer relevant. Genes with cis mutations are enriched for differential ASE, and we find 147 potentially functional noncoding mutations cis to genes that exhibit significant differential ASE. We conclude that differential ASE is a promising means for discovering gene dysregulation due to cis noncoding mutations.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Profiling/methods , Whole Genome Sequencing/methods , Alleles , Female , Humans , Mutation
14.
Genome Med ; 10(1): 35, 2018 05 10.
Article in English | MEDLINE | ID: mdl-29747675

ABSTRACT

Due to an error introduced during copyediting of this article [1], reference [8] incorrectly reads.

15.
Genome Med ; 9(1): 79, 2017 08 25.
Article in English | MEDLINE | ID: mdl-28841835

ABSTRACT

A major aim of cancer genomics is to pinpoint which somatically mutated genes are involved in tumor initiation and progression. We introduce a new framework for uncovering cancer genes, differential mutation analysis, which compares the mutational profiles of genes across cancer genomes with their natural germline variation across healthy individuals. We present DiffMut, a fast and simple approach for differential mutational analysis, and demonstrate that it is more effective in discovering cancer genes than considerably more sophisticated approaches. We conclude that germline variation across healthy human genomes provides a powerful means for characterizing somatic mutation frequency and identifying cancer driver genes. DiffMut is available at https://github.com/Singh-Lab/Differential-Mutation-Analysis .


Subject(s)
DNA Mutational Analysis/methods , Genes, Neoplasm , Germ-Line Mutation , Mutation , Neoplasms/genetics , Exome , Female , Genetic Predisposition to Disease , Genomics/methods , Humans , Male , Polymorphism, Genetic , Software
16.
Bioinformatics ; 26(1): 141-2, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19887508

ABSTRACT

SUMMARY: SimBoolNet is an open source Cytoscape plugin that simulates the dynamics of signaling transduction using Boolean networks. Given a user-specified level of stimulation to signal receptors, SimBoolNet simulates the response of downstream molecules and visualizes with animation and records the dynamic changes of the network. It can be used to generate hypotheses and facilitate experimental studies about causal relations and crosstalk among cellular signaling pathways. AVAILABILITY: SimBoolNet package (with manual) is freely available at http://www.ncbi.nlm.nih.gov/CBBresearch/Przytycka/SimBoolNet


Subject(s)
Algorithms , Computer Simulation , Models, Biological , Models, Statistical , Proteome/metabolism , Signal Transduction/physiology , Software
17.
BMC Syst Biol ; 3: 88, 2009 Sep 04.
Article in English | MEDLINE | ID: mdl-19732446

ABSTRACT

BACKGROUND: Cellular response to external stimuli requires propagation of corresponding signals through molecular signaling pathways. However, signaling pathways are not isolated information highways, but rather interact in a number of ways forming sophisticated signaling networks. Since defects in signaling pathways are associated with many serious diseases, understanding of the crosstalk between them is fundamental for designing molecularly targeted therapy. Unfortunately, we still lack technology that would allow high throughput detailed measurement of activity of individual signaling molecules and their interactions. This necessitates developing methods to prioritize selection of the molecules such that measuring their activity would be most informative for understanding the crosstalk. Furthermore, absence of the reaction coefficients necessary for detailed modeling of signal propagation raises the question whether simple parameter-free models could provide useful information about such pathways. RESULTS: We study the combined signaling network of three major pro-survival signaling pathways: Epidermal Growth Factor Receptor (EGFR), Insulin-like Growth Factor-1 Receptor (IGF-1R), and Insulin Receptor (IR). Our study involves static analysis and dynamic modeling of this network, as well as an experimental verification of the model by measuring the response of selected signaling molecules to differential stimulation of EGF, IGF and insulin receptors. We introduced two novel measures of the importance of a node in the context of such crosstalk. Based on these measures several molecules, namely Erk1/2, Akt1, Jnk, p70S6K, were selected for monitoring in the network simulation and for experimental studies. Our simulation method relies on the Boolean network model combined with stochastic propagation of the signal. Most (although not all) trends suggested by the simulations have been confirmed by experiments. CONCLUSION: The simple model implemented in this paper provides a valuable first step in modeling signaling networks. However, to obtain a fully predictive model, a more detailed knowledge regarding parameters of individual interactions might be necessary.


Subject(s)
Cell Survival , Epidermal Growth Factor/metabolism , Insulin/metabolism , Models, Biological , Ovarian Neoplasms/physiopathology , Signal Transduction , Somatomedins/metabolism , Cell Line , Computer Simulation , Female , Humans , Ovarian Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...