Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters

Publication year range
1.
Cell ; 180(5): 915-927.e16, 2020 03 05.
Article in English | MEDLINE | ID: mdl-32084333

ABSTRACT

The dichotomous model of "drivers" and "passengers" in cancer posits that only a few mutations in a tumor strongly affect its progression, with the remaining ones being inconsequential. Here, we leveraged the comprehensive variant dataset from the ICGC/TCGA Pan-Cancer Analysis of Whole Genomes (PCAWG) project to demonstrate that-in addition to the dichotomy of high- and low-impact variants-there is a third group of medium-impact putative passengers. Moreover, we also found that molecular impact correlates with subclonal architecture (i.e., early versus late mutations), and different signatures encode for mutations with divergent impact. Furthermore, we adapted an additive-effects model from complex-trait studies to show that the aggregated effect of putative passengers, including undetected weak drivers, provides significant additional power (∼12% additive variance) for predicting cancerous phenotypes, beyond PCAWG-identified driver mutations. Finally, this framework allowed us to estimate the frequency of potential weak-driver mutations in PCAWG samples lacking any well-characterized driver alterations.


Subject(s)
Genome, Human/genetics , Genomics/methods , Mutation/genetics , Neoplasms/genetics , DNA Mutational Analysis/methods , Disease Progression , Humans , Neoplasms/pathology , Whole Genome Sequencing
2.
Cell ; 177(2): 463-477.e15, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30951672

ABSTRACT

To develop a map of cell-cell communication mediated by extracellular RNA (exRNA), the NIH Extracellular RNA Communication Consortium created the exRNA Atlas resource (https://exrna-atlas.org). The Atlas version 4P1 hosts 5,309 exRNA-seq and exRNA qPCR profiles from 19 studies and a suite of analysis and visualization tools. To analyze variation between profiles, we apply computational deconvolution. The analysis leads to a model with six exRNA cargo types (CT1, CT2, CT3A, CT3B, CT3C, CT4), each detectable in multiple biofluids (serum, plasma, CSF, saliva, urine). Five of the cargo types associate with known vesicular and non-vesicular (lipoprotein and ribonucleoprotein) exRNA carriers. To validate utility of this model, we re-analyze an exercise response study by deconvolution to identify physiologically relevant response pathways that were not detected previously. To enable wide application of this model, as part of the exRNA Atlas resource, we provide tools for deconvolution and analysis of user-provided case-control studies.


Subject(s)
Cell Communication/physiology , RNA/metabolism , Adult , Body Fluids/chemistry , Cell-Free Nucleic Acids/metabolism , Circulating MicroRNA/metabolism , Extracellular Vesicles/metabolism , Female , Humans , Male , Reproducibility of Results , Sequence Analysis, RNA/methods , Software
3.
Bioinformatics ; 40(Suppl 2): ii111-ii119, 2024 09 01.
Article in English | MEDLINE | ID: mdl-39230702

ABSTRACT

MOTIVATION: Spatial transcriptomics technologies, which generate a spatial map of gene activity, can deepen the understanding of tissue architecture and its molecular underpinnings in health and disease. However, the high cost makes these technologies difficult to use in practice. Histological images co-registered with targeted tissues are more affordable and routinely generated in many research and clinical studies. Hence, predicting spatial gene expression from the morphological clues embedded in tissue histological images provides a scalable alternative approach to decoding tissue complexity. RESULTS: Here, we present a graph neural network based framework to predict the spatial expression of highly expressed genes from tissue histological images. Extensive experiments on two separate breast cancer data cohorts demonstrate that our method improves the prediction performance compared to the state-of-the-art, and that our model can be used to better delineate spatial domains of biological interest. AVAILABILITY AND IMPLEMENTATION: https://github.com/song0309/asGNN/.


Subject(s)
Breast Neoplasms , Neural Networks, Computer , Humans , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , Gene Expression Profiling/methods , Transcriptome
4.
Bioinformatics ; 39(1)2023 01 01.
Article in English | MEDLINE | ID: mdl-36477833

ABSTRACT

MOTIVATION: While many quantum computing (QC) methods promise theoretical advantages over classical counterparts, quantum hardware remains limited. Exploiting near-term QC in computer-aided drug design (CADD) thus requires judicious partitioning between classical and quantum calculations. RESULTS: We present HypaCADD, a hybrid classical-quantum workflow for finding ligands binding to proteins, while accounting for genetic mutations. We explicitly identify modules of our drug-design workflow currently amenable to replacement by QC: non-intuitively, we identify the mutation-impact predictor as the best candidate. HypaCADD thus combines classical docking and molecular dynamics with quantum machine learning (QML) to infer the impact of mutations. We present a case study with the coronavirus (SARS-CoV-2) protease and associated mutants. We map a classical machine-learning module onto QC, using a neural network constructed from qubit-rotation gates. We have implemented this in simulation and on two commercial quantum computers. We find that the QML models can perform on par with, if not better than, classical baselines. In summary, HypaCADD offers a successful strategy for leveraging QC for CADD. AVAILABILITY AND IMPLEMENTATION: Jupyter Notebooks with Python code are freely available for academic use on GitHub: https://www.github.com/hypahub/hypacadd_notebook. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
COVID-19 , Software , Humans , Workflow , Computing Methodologies , Quantum Theory , SARS-CoV-2 , Drug Design , Molecular Dynamics Simulation
5.
Mod Pathol ; 34(7): 1261-1270, 2021 07.
Article in English | MEDLINE | ID: mdl-33536573

ABSTRACT

Ki67, a nuclear proliferation-related protein, is heavily used in anatomic pathology but has not become a companion diagnostic or a standard-of-care biomarker due to analytic variability in both assay protocols and interpretation. The International Ki67 Working Group in breast cancer has published and has ongoing efforts in the standardization of the interpretation of Ki67, but they have not yet assessed technical issues of assay production representing multiple sources of variation, including antibody clones, antibody formats, staining platforms, and operators. The goal of this work is to address these issues with a new standardization tool. We have developed a cell line microarray system in which mixes of human Karpas 299 or Jurkat cells (Ki67+) with Sf9 (Spodoptera frugiperda) (Ki67-) cells are present in incremental standardized ratios. To validate the tool, six different antibodies, including both ready-to-use and concentrate formats from six vendors, were used to measure Ki67 proliferation indices using IHC protocols for manual (bench-top) and automated platforms. The assays were performed by three different laboratories at Yale and analyzed using two image analysis software packages, including QuPath and Visiopharm. Results showed statistically significant differences in Ki67 reactivity between each antibody clone. However, subsets of Ki67 assays using three clones performed in three different labs show no significant differences. This work shows the need for analytic standardization of the Ki67 assay and provides a new tool to do so. We show here how a cell line standardization system can be used to normalize the staining variability in proliferation indices between different antibody clones in a triple negative breast cancer cohort. We believe that this cell line standardization array has the potential to improve reproducibility among Ki67 assays and laboratories, which is critical for establishing Ki67 as a standard-of-care assay.


Subject(s)
Biomarkers, Tumor/analysis , Immunohistochemistry/standards , Ki-67 Antigen/analysis , Mitotic Index/standards , Triple Negative Breast Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Female , Humans , Middle Aged
7.
J Theor Biol ; 420: 304-317, 2017 05 07.
Article in English | MEDLINE | ID: mdl-27866978

ABSTRACT

Coregulation of the expression of groups of genes has been extensively demonstrated empirically in bacterial and eukaryotic systems. Such coregulation can arise through the use of shared regulatory motifs, which allow the coordinated expression of modules (and module groups) of functionally related genes across the genome. Coregulation can also arise through the physical association of multi-gene complexes through chromosomal looping, which are then transcribed together. We present a general formalism for modeling coregulation rules in the framework of Random Boolean Networks (RBN), and develop specific models for transcription factor networks with modular structure (including module groups, and multi-input modules (MIM) with autoregulation) and multi-gene complexes (including hierarchical differentiation between multi-gene complex members). We develop a mean-field approach to analyse the dynamical stability of large networks incorporating coregulation, and show that autoregulated MIM and hierarchical gene-complex models can achieve greater stability than networks without coregulation whose rules have matching activation frequency. We provide further analysis of the stability of small networks of both kinds through simulations. We also characterize several general properties of the transients and attractors in the hierarchical coregulation model, and show using simulations that the steady-state distribution factorizes hierarchically as a Bayesian network in a Markov Jump Process analogue of the RBN model.


Subject(s)
Gene Expression Regulation/genetics , Gene Regulatory Networks/genetics , Models, Genetic , Bayes Theorem , Computer Simulation , Markov Chains
8.
J R Soc Interface ; 21(212): 20230647, 2024 03.
Article in English | MEDLINE | ID: mdl-38503341

ABSTRACT

Cultural processes of change bear many resemblances to biological evolution. The underlying units of non-biological evolution have, however, remained elusive, especially in the domain of music. Here, we introduce a general framework to jointly identify underlying units and their associated evolutionary processes. We model musical styles and principles of organization in dimensions such as harmony and form as following an evolutionary process. Furthermore, we propose that such processes can be identified by extracting latent evolutionary signatures from musical corpora, analogously to identifying mutational signatures in genomics. These signatures provide a latent embedding for each song or musical piece. We develop a deep generative architecture for our model, which can be viewed as a type of variational autoencoder with an evolutionary prior constraining the latent space; specifically, the embeddings for each song are tied together via an energy-based prior, which encourages songs close in evolutionary space to share similar representations. As illustration, we analyse songs from the McGill Billboard dataset. We find frequent chord transitions and formal repetition schemes and identify latent evolutionary signatures related to these features. Finally, we show that the latent evolutionary representations learned by our model outperform non-evolutionary representations in such tasks as period and genre prediction.


Subject(s)
Cultural Evolution , Music , Genomics
9.
Clin Cancer Res ; 30(16): 3520-3532, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38837895

ABSTRACT

PURPOSE: We aim to improve the prediction of response or resistance to immunotherapies in patients with melanoma. This goal is based on the hypothesis that current gene signatures predicting immunotherapy outcomes show only modest accuracy due to the lack of spatial information about cellular functions and molecular processes within tumors and their microenvironment. EXPERIMENTAL DESIGN: We collected gene expression data spatially from three cellular compartments defined by CD68+ macrophages, CD45+ leukocytes, and S100B+ tumor cells in 55 immunotherapy-treated melanoma specimens using Digital Spatial Profiling-Whole Transcriptome Atlas. We developed a computational pipeline to discover compartment-specific gene signatures and determine if adding spatial information can improve patient stratification. RESULTS: We achieved robust performance of compartment-specific signatures in predicting the outcome of immune checkpoint inhibitors in the discovery cohort. Of the three signatures, the S100B signature showed the best performance in the validation cohort (N = 45). We also compared our compartment-specific signatures with published bulk signatures and found the S100B tumor spatial signature outperformed previous signatures. Within the eight-gene S100B signature, five genes (PSMB8, TAX1BP3, NOTCH3, LCP2, and NQO1) with positive coefficients predict the response, and three genes (KMT2C, OVCA2, and MGRN1) with negative coefficients predict the resistance to treatment. CONCLUSIONS: We conclude that the spatially defined compartment signatures utilize tumor and tumor microenvironment-specific information, leading to more accurate prediction of treatment outcome, and thus merit prospective clinical assessment.


Subject(s)
Biomarkers, Tumor , Immunotherapy , Melanoma , Transcriptome , Tumor Microenvironment , Humans , Melanoma/genetics , Melanoma/therapy , Melanoma/immunology , Melanoma/pathology , Immunotherapy/methods , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Biomarkers, Tumor/genetics , Gene Expression Profiling , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic , S100 Calcium Binding Protein beta Subunit/genetics , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, CD/genetics , Female , Male , Prognosis , Macrophages/immunology , Macrophages/metabolism , CD68 Molecule
10.
bioRxiv ; 2024 Mar 30.
Article in English | MEDLINE | ID: mdl-38562822

ABSTRACT

Single-cell genomics is a powerful tool for studying heterogeneous tissues such as the brain. Yet, little is understood about how genetic variants influence cell-level gene expression. Addressing this, we uniformly processed single-nuclei, multi-omics datasets into a resource comprising >2.8M nuclei from the prefrontal cortex across 388 individuals. For 28 cell types, we assessed population-level variation in expression and chromatin across gene families and drug targets. We identified >550K cell-type-specific regulatory elements and >1.4M single-cell expression-quantitative-trait loci, which we used to build cell-type regulatory and cell-to-cell communication networks. These networks manifest cellular changes in aging and neuropsychiatric disorders. We further constructed an integrative model accurately imputing single-cell expression and simulating perturbations; the model prioritized ~250 disease-risk genes and drug targets with associated cell types.

11.
Science ; 384(6698): eadi5199, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38781369

ABSTRACT

Single-cell genomics is a powerful tool for studying heterogeneous tissues such as the brain. Yet little is understood about how genetic variants influence cell-level gene expression. Addressing this, we uniformly processed single-nuclei, multiomics datasets into a resource comprising >2.8 million nuclei from the prefrontal cortex across 388 individuals. For 28 cell types, we assessed population-level variation in expression and chromatin across gene families and drug targets. We identified >550,000 cell type-specific regulatory elements and >1.4 million single-cell expression quantitative trait loci, which we used to build cell-type regulatory and cell-to-cell communication networks. These networks manifest cellular changes in aging and neuropsychiatric disorders. We further constructed an integrative model accurately imputing single-cell expression and simulating perturbations; the model prioritized ~250 disease-risk genes and drug targets with associated cell types.


Subject(s)
Brain , Gene Regulatory Networks , Mental Disorders , Single-Cell Analysis , Humans , Aging/genetics , Brain/metabolism , Cell Communication/genetics , Chromatin/metabolism , Chromatin/genetics , Genomics , Mental Disorders/genetics , Prefrontal Cortex/metabolism , Prefrontal Cortex/physiology , Quantitative Trait Loci
12.
Sci Rep ; 13(1): 8470, 2023 05 25.
Article in English | MEDLINE | ID: mdl-37231011

ABSTRACT

For the COVID-19 pandemic, viral transmission has been documented in many historical and geographical contexts. Nevertheless, few studies have explicitly modeled the spatiotemporal flow based on genetic sequences, to develop mitigation strategies. Additionally, thousands of SARS-CoV-2 genomes have been sequenced with associated records, potentially providing a rich source for such spatiotemporal analysis, an unprecedented amount during a single outbreak. Here, in a case study of seven states, we model the first wave of the outbreak by determining regional connectivity from phylogenetic sequence information (i.e. "genetic connectivity"), in addition to traditional epidemiologic and demographic parameters. Our study shows nearly all of the initial outbreak can be traced to a few lineages, rather than disconnected outbreaks, indicative of a mostly continuous initial viral flow. While the geographic distance from hotspots is initially important in the modeling, genetic connectivity becomes increasingly significant later in the first wave. Moreover, our model predicts that isolated local strategies (e.g. relying on herd immunity) can negatively impact neighboring regions, suggesting more efficient mitigation is possible with unified, cross-border interventions. Finally, our results suggest that a few targeted interventions based on connectivity can have an effect similar to that of an overall lockdown. They also suggest that while successful lockdowns are very effective in mitigating an outbreak, less disciplined lockdowns quickly decrease in effectiveness. Our study provides a framework for combining phylodynamic and computational methods to identify targeted interventions.


Subject(s)
COVID-19 , Humans , COVID-19/epidemiology , COVID-19/prevention & control , SARS-CoV-2/genetics , Pandemics/prevention & control , Phylogeny , Communicable Disease Control/methods , Disease Outbreaks
13.
Cell Rep Methods ; 1(5): 100068, 2021 09 27.
Article in English | MEDLINE | ID: mdl-35474672

ABSTRACT

Advances in single-cell RNA sequencing have allowed for the identification of cellular subtypes on the basis of quantification of the number of transcripts in each cell. However, cells might also differ in the spatial distribution of molecules, including RNAs. Here, we present DypFISH, an approach to quantitatively investigate the subcellular localization of RNA and protein. We introduce a range of analytical techniques to interrogate single-molecule RNA fluorescence in situ hybridization (smFISH) data in combination with protein immunolabeling. DypFISH is suited to study patterns of clustering of molecules, the association of mRNA-protein subcellular localization with microtubule organizing center orientation, and interdependence of mRNA-protein spatial distributions. We showcase how our analytical tools can achieve biological insights by utilizing cell micropatterning to constrain cellular architecture, which leads to reduction in subcellular mRNA distribution variation, allowing for the characterization of their localization patterns. Furthermore, we show that our method can be applied to physiological systems such as skeletal muscle fibers.


Subject(s)
Muscle Fibers, Skeletal , RNA , RNA/genetics , In Situ Hybridization, Fluorescence/methods , RNA, Messenger/genetics , Muscle Fibers, Skeletal/metabolism , Protein Transport
14.
Nat Commun ; 11(1): 732, 2020 02 05.
Article in English | MEDLINE | ID: mdl-32024824

ABSTRACT

Tumors accumulate thousands of mutations, and sequencing them has given rise to methods for finding cancer drivers via mutational recurrence. However, these methods require large cohorts and underperform for low recurrence. Recently, ultra-deep sequencing has enabled accurate measurement of VAFs (variant-allele frequencies) for mutations, allowing the determination of evolutionary trajectories. Here, based solely on the VAF spectrum for an individual sample, we report on a method that identifies drivers and quantifies tumor growth. Drivers introduce perturbations into the spectrum, and our method uses the frequency of hitchhiking mutations preceding a driver to measure this. As validation, we use simulation models and 993 tumors from the Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium with previously identified drivers. Then we apply our method to an ultra-deep sequenced acute myeloid leukemia (AML) tumor and identify known cancer genes and additional driver candidates. In summary, our framework presents opportunities for personalized driver diagnosis using sequencing data from a single individual.


Subject(s)
Genes, Tumor Suppressor , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Models, Genetic , Mutation , Algorithms , Gene Frequency , High-Throughput Nucleotide Sequencing , Humans , Mutation Rate , Mutation, Missense , Neoplasms/genetics , Neoplasms/pathology , Oncogenes , Precision Medicine , Stochastic Processes
15.
Nat Commun ; 11(1): 3696, 2020 07 29.
Article in English | MEDLINE | ID: mdl-32728046

ABSTRACT

ENCODE comprises thousands of functional genomics datasets, and the encyclopedia covers hundreds of cell types, providing a universal annotation for genome interpretation. However, for particular applications, it may be advantageous to use a customized annotation. Here, we develop such a custom annotation by leveraging advanced assays, such as eCLIP, Hi-C, and whole-genome STARR-seq on a number of data-rich ENCODE cell types. A key aspect of this annotation is comprehensive and experimentally derived networks of both transcription factors and RNA-binding proteins (TFs and RBPs). Cancer, a disease of system-wide dysregulation, is an ideal application for such a network-based annotation. Specifically, for cancer-associated cell types, we put regulators into hierarchies and measure their network change (rewiring) during oncogenesis. We also extensively survey TF-RBP crosstalk, highlighting how SUB1, a previously uncharacterized RBP, drives aberrant tumor expression and amplifies the effect of MYC, a well-known oncogenic TF. Furthermore, we show how our annotation allows us to place oncogenic transformations in the context of a broad cell space; here, many normal-to-tumor transitions move towards a stem-like state, while oncogene knockdowns show an opposing trend. Finally, we organize the resource into a coherent workflow to prioritize key elements and variants, in addition to regulators. We showcase the application of this prioritization to somatic burdening, cancer differential expression and GWAS. Targeted validations of the prioritized regulators, elements and variants using siRNA knockdowns, CRISPR-based editing, and luciferase assays demonstrate the value of the ENCODE resource.


Subject(s)
Databases, Genetic , Genomics , Neoplasms/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Gene Regulatory Networks , Humans , Mutation/genetics , Reproducibility of Results , Transcription Factors/metabolism
16.
Cell Syst ; 8(4): 352-357.e3, 2019 04 24.
Article in English | MEDLINE | ID: mdl-30956140

ABSTRACT

Small RNA sequencing has been widely adopted to study the diversity of extracellular RNAs (exRNAs) in biofluids; however, the analysis of exRNA samples can be challenging: they are vulnerable to contamination and artifacts from different isolation techniques, present in lower concentrations than cellular RNA, and occasionally of exogenous origin. To address these challenges, we present exceRpt, the exRNA-processing toolkit of the NIH Extracellular RNA Communication Consortium (ERCC). exceRpt is structured as a cascade of filters and quantifications prioritized based on one's confidence in a given set of annotated RNAs. It generates quality control reports and abundance estimates for RNA biotypes. It is also capable of characterizing mappings to exogenous genomes, which, in turn, can be used to generate phylogenetic trees. exceRpt has been used to uniformly process all ∼3,500 exRNA-seq datasets in the public exRNA Atlas and is available from genboree.org and github.gersteinlab.org/exceRpt.


Subject(s)
Cell-Free Nucleic Acids/chemistry , RNA-Seq/methods , Software , Animals , Cell-Free Nucleic Acids/genetics , Cell-Free Nucleic Acids/metabolism , Humans , Mice , RNA-Seq/standards
17.
IEEE Trans Pattern Anal Mach Intell ; 30(6): 970-84, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18421104

ABSTRACT

Face recognition algorithms perform very unreliably when the pose of the probe face is different from the gallery face: typical feature vectors vary more with pose than with identity. We propose a generative model that creates a one-to-many mapping from an idealized "identity" space to the observed data space. In identity space, the representation for each individual does not vary with pose. We model the measured feature vector as being generated by a pose-contingent linear transformation of the identity variable in the presence of Gaussian noise. We term this model "tied" factor analysis. The choice of linear transformation (factors) depends on the pose, but the loadings are constant (tied) for a given individual. We use the EM algorithm to estimate the linear transformations and the noise parameters from training data. We propose a probabilistic distance metric which allows a full posterior over possible matches to be established. We introduce a novel feature extraction process and investigate recognition performance using the FERET, XM2VTS and PIE databases. Recognition performance compares favourably to contemporary approaches.


Subject(s)
Artificial Intelligence , Biometry/methods , Face/anatomy & histology , Facial Expression , Image Interpretation, Computer-Assisted/methods , Pattern Recognition, Automated/methods , Subtraction Technique , Algorithms , Factor Analysis, Statistical , Humans , Image Enhancement/methods , Likelihood Functions , Reproducibility of Results , Sensitivity and Specificity
18.
Science ; 362(6420)2018 12 14.
Article in English | MEDLINE | ID: mdl-30545856

ABSTRACT

Most genetic risk for psychiatric disease lies in regulatory regions, implicating pathogenic dysregulation of gene expression and splicing. However, comprehensive assessments of transcriptomic organization in diseased brains are limited. In this work, we integrated genotypes and RNA sequencing in brain samples from 1695 individuals with autism spectrum disorder (ASD), schizophrenia, and bipolar disorder, as well as controls. More than 25% of the transcriptome exhibits differential splicing or expression, with isoform-level changes capturing the largest disease effects and genetic enrichments. Coexpression networks isolate disease-specific neuronal alterations, as well as microglial, astrocyte, and interferon-response modules defining previously unidentified neural-immune mechanisms. We integrated genetic and genomic data to perform a transcriptome-wide association study, prioritizing disease loci likely mediated by cis effects on brain expression. This transcriptome-wide characterization of the molecular pathology across three major psychiatric disorders provides a comprehensive resource for mechanistic insight and therapeutic development.


Subject(s)
Autism Spectrum Disorder/genetics , Bipolar Disorder/genetics , Genetic Predisposition to Disease , RNA Splicing , Schizophrenia/genetics , Brain/metabolism , Humans , Protein Isoforms/genetics , Sequence Analysis, RNA , Transcriptome
19.
Science ; 362(6420)2018 12 14.
Article in English | MEDLINE | ID: mdl-30545857

ABSTRACT

Despite progress in defining genetic risk for psychiatric disorders, their molecular mechanisms remain elusive. Addressing this, the PsychENCODE Consortium has generated a comprehensive online resource for the adult brain across 1866 individuals. The PsychENCODE resource contains ~79,000 brain-active enhancers, sets of Hi-C linkages, and topologically associating domains; single-cell expression profiles for many cell types; expression quantitative-trait loci (QTLs); and further QTLs associated with chromatin, splicing, and cell-type proportions. Integration shows that varying cell-type proportions largely account for the cross-population variation in expression (with >88% reconstruction accuracy). It also allows building of a gene regulatory network, linking genome-wide association study variants to genes (e.g., 321 for schizophrenia). We embed this network into an interpretable deep-learning model, which improves disease prediction by ~6-fold versus polygenic risk scores and identifies key genes and pathways in psychiatric disorders.


Subject(s)
Brain/metabolism , Gene Expression Regulation , Mental Disorders/genetics , Datasets as Topic , Deep Learning , Enhancer Elements, Genetic , Epigenesis, Genetic , Epigenomics , Gene Regulatory Networks , Genome-Wide Association Study , Humans , Quantitative Trait Loci , Single-Cell Analysis , Transcriptome
20.
PLoS One ; 7(11): e48701, 2012.
Article in English | MEDLINE | ID: mdl-23144935

ABSTRACT

BACKGROUND: Although rapid diagnostic tests (RDTs) have practical advantages over light microscopy (LM) and good sensitivity in severe falciparum malaria in Africa, their utility where severe non-falciparum malaria occurs is unknown. LM, RDTs and polymerase chain reaction (PCR)-based methods have limitations, and thus conventional comparative malaria diagnostic studies employ imperfect gold standards. We assessed whether, using Bayesian latent class models (LCMs) which do not require a reference method, RDTs could safely direct initial anti-infective therapy in severe ill children from an area of hyperendemic transmission of both Plasmodium falciparum and P. vivax. METHODS AND FINDINGS: We studied 797 Papua New Guinean children hospitalized with well-characterized severe illness for whom LM, RDT and nested PCR (nPCR) results were available. For any severe malaria, the estimated prevalence was 47.5% with RDTs exhibiting similar sensitivity and negative predictive value (NPV) to nPCR (≥96.0%). LM was the least sensitive test (87.4%) and had the lowest NPV (89.7%), but had the highest specificity (99.1%) and positive predictive value (98.9%). For severe falciparum malaria (prevalence 42.9%), the findings were similar. For non-falciparum severe malaria (prevalence 6.9%), no test had the WHO-recommended sensitivity and specificity of >95% and >90%, respectively. RDTs were the least sensitive (69.6%) and had the lowest NPV (96.7%). CONCLUSIONS: RDTs appear a valuable point-of-care test that is at least equivalent to LM in diagnosing severe falciparum malaria in this epidemiologic situation. None of the tests had the required sensitivity/specificity for severe non-falciparum malaria but the number of false-negative RDTs in this group was small.


Subject(s)
Immunologic Tests , Malaria/diagnosis , Plasmodium falciparum/immunology , Plasmodium vivax/immunology , Antigens, Protozoan/immunology , Bayes Theorem , Child, Preschool , Female , Humans , Infant , Male , Markov Chains , Monte Carlo Method , Papua New Guinea , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL