Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Brief Bioinform ; 24(1)2023 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-36458445

RESUMEN

Deciphering 3D genome conformation is important for understanding gene regulation and cellular function at a spatial level. The recent advances of single cell Hi-C technologies have enabled the profiling of the 3D architecture of DNA within individual cell, which allows us to study the cell-to-cell variability of 3D chromatin organization. Computational approaches are in urgent need to comprehensively analyze the sparse and heterogeneous single cell Hi-C data. Here, we proposed scDEC-Hi-C, a new framework for single cell Hi-C analysis with deep generative neural networks. scDEC-Hi-C outperforms existing methods in terms of single cell Hi-C data clustering and imputation. Moreover, the generative power of scDEC-Hi-C could help unveil the differences of chromatin architecture across cell types. We expect that scDEC-Hi-C could shed light on deepening our understanding of the complex mechanism underlying the formation of chromatin contacts.


Asunto(s)
Cromatina , Cromosomas , Cromatina/genética , Genoma , ADN , Análisis por Conglomerados
2.
Nucleic Acids Res ; 51(D1): D159-D166, 2023 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-36215037

RESUMEN

Elucidating the role of 3D architecture of DNA in gene regulation is crucial for understanding cell differentiation, tissue homeostasis and disease development. Among various chromatin conformation capture methods, HiChIP has received increasing attention for its significant improvement over other methods in profiling of regulatory (e.g. H3K27ac) and structural (e.g. cohesin) interactions. To facilitate the studies of 3D regulatory interactions, we developed a HiChIP interactions database, HiChIPdb (http://health.tsinghua.edu.cn/hichipdb/). The current version of HiChIPdb contains ∼262M annotated HiChIP interactions from 200 high-throughput HiChIP samples across 108 cell types. The functionalities of HiChIPdb include: (i) standardized categorization of HiChIP interactions in a hierarchical structure based on organ, tissue and cell line and (ii) comprehensive annotations of HiChIP interactions with regulatory genes and GWAS Catalog SNPs. To the best of our knowledge, HiChIPdb is the first comprehensive database that utilizes a unified pipeline to map the functional interactions across diverse cell types and tissues in different resolutions. We believe this database has the potential to advance cutting-edge research in regulatory mechanisms in development and disease by removing the barrier in data aggregation, preprocessing, and analysis.


Asunto(s)
Cromatina , ADN , Línea Celular , Cromatina/genética , Regulación de la Expresión Génica , Análisis de Secuencia de ADN/métodos , Bases de Datos Genéticas
3.
Bioinformatics ; 38(11): 2996-3003, 2022 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-35394015

RESUMEN

MOTIVATION: Single-cell technologies play a crucial role in revolutionizing biological research over the past decade, which strengthens our understanding in cell differentiation, development and regulation from a single-cell level perspective. Single-cell RNA sequencing (scRNA-seq) is one of the most common single cell technologies, which enables probing transcriptional states in thousands of cells in one experiment. Identification of cell types from scRNA-seq measurements is a fundamental and crucial question to answer. Most previous studies directly take gene expression as input while ignoring the comprehensive gene-gene interactions. RESULTS: We propose scGraph, an automatic cell identification algorithm leveraging gene interaction relationships to enhance the performance of the cell-type identification. scGraph is based on a graph neural network to aggregate the information of interacting genes. In a series of experiments, we demonstrate that scGraph is accurate and outperforms eight comparison methods in the task of cell-type identification. Moreover, scGraph automatically learns the gene interaction relationships from biological data and the pathway enrichment analysis shows consistent findings with previous analysis, providing insights on the analysis of regulatory mechanism. AVAILABILITY AND IMPLEMENTATION: scGraph is freely available at https://github.com/QijinYin/scGraph and https://figshare.com/articles/software/scGraph/17157743. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Perfilación de la Expresión Génica , Análisis de la Célula Individual , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Perfilación de la Expresión Génica/métodos , Programas Informáticos , Redes Neurales de la Computación
4.
Nucleic Acids Res ; 49(D1): D221-D228, 2021 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-33045745

RESUMEN

Gene regulatory elements, including promoters, enhancers, silencers, etc., control transcriptional programs in a spatiotemporal manner. Though these elements are known to be able to induce either positive or negative transcriptional control, the community has been mostly studying enhancers which amplify transcription initiation, with less emphasis given to silencers which repress gene expression. To facilitate the study of silencers and the investigation of their potential roles in transcriptional control, we developed SilencerDB (http://health.tsinghua.edu.cn/silencerdb/), a comprehensive database of silencers by manually curating silencers from 2300 published articles. The current version, SilencerDB 1.0, contains (1) 33 060 validated silencers from experimental methods, and (ii) 5 045 547 predicted silencers from state-of-the-art machine learning methods. The functionality of SilencerDB includes (a) standardized categorization of silencers in a tree-structured class hierarchy based on species, organ, tissue and cell line and (b) comprehensive annotations of silencers with the nearest gene and potential regulatory genes. SilencerDB, to the best of our knowledge, is the first comprehensive database at this scale dedicated to silencers, with reliable annotations and user-friendly interactive database features. We believe this database has the potential to enable advanced understanding of silencers in regulatory mechanisms and to empower researchers to devise diverse applications of silencers in disease development.


Asunto(s)
Bases de Datos de Ácidos Nucleicos , Aprendizaje Automático , Elementos Silenciadores Transcripcionales , Transcripción Genética , Interfaz Usuario-Computador , Animales , Búfalos/genética , Línea Celular , Pollos/genética , Drosophila melanogaster/genética , Humanos , Internet , Ratones , Anotación de Secuencia Molecular , Especificidad de Órganos , Ratas , Sus scrofa/genética
5.
Bioinformatics ; 36(2): 496-503, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31318408

RESUMEN

MOTIVATION: Interactions among cis-regulatory elements such as enhancers and promoters are main driving forces shaping context-specific chromatin structure and gene expression. Although there have been computational methods for predicting gene expression from genomic and epigenomic information, most of them neglect long-range enhancer-promoter interactions, due to the difficulty in precisely linking regulatory enhancers to target genes. Recently, HiChIP, a novel high-throughput experimental approach, has generated comprehensive data on high-resolution interactions between promoters and distal enhancers. Moreover, plenty of studies suggest that deep learning achieves state-of-the-art performance in epigenomic signal prediction, and thus promoting the understanding of regulatory elements. In consideration of these two factors, we integrate proximal promoter sequences and HiChIP distal enhancer-promoter interactions to accurately predict gene expression. RESULTS: We propose DeepExpression, a densely connected convolutional neural network, to predict gene expression using both promoter sequences and enhancer-promoter interactions. We demonstrate that our model consistently outperforms baseline methods, not only in the classification of binary gene expression status but also in regression of continuous gene expression levels, in both cross-validation experiments and cross-cell line predictions. We show that the sequential promoter information is more informative than the experimental enhancer information; meanwhile, the enhancer-promoter interactions within ±100 kbp around the TSS of a gene are most beneficial. We finally visualize motifs in both promoter and enhancer regions and show the match of identified sequence signatures with known motifs. We expect to see a wide spectrum of applications using HiChIP data in deciphering the mechanism of gene regulation. AVAILABILITY AND IMPLEMENTATION: DeepExpression is freely available at https://github.com/wanwenzeng/DeepExpression. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Redes Neurales de la Computación , Secuencias Reguladoras de Ácidos Nucleicos , Elementos de Facilitación Genéticos , Epigenómica , Genómica , Regiones Promotoras Genéticas
6.
Proc Natl Acad Sci U S A ; 115(30): 7723-7728, 2018 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-29987051

RESUMEN

When different types of functional genomics data are generated on single cells from different samples of cells from the same heterogeneous population, the clustering of cells in the different samples should be coupled. We formulate this "coupled clustering" problem as an optimization problem and propose the method of coupled nonnegative matrix factorizations (coupled NMF) for its solution. The method is illustrated by the integrative analysis of single-cell RNA-sequencing (RNA-seq) and single-cell ATAC-sequencing (ATAC-seq) data.


Asunto(s)
Bases de Datos Genéticas , Modelos Genéticos , Análisis de Secuencia de ARN/métodos , Animales , Humanos
7.
Methods ; 145: 41-50, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29874547

RESUMEN

Genome-wide association studies (GWAS) have successfully discovered a number of disease-associated genetic variants in the past decade, providing an unprecedented opportunity for deciphering genetic basis of human inherited diseases. However, it is still a challenging task to extract biological knowledge from the GWAS data, due to such issues as missing heritability and weak interpretability. Indeed, the fact that the majority of discovered loci fall into noncoding regions without clear links to genes has been preventing the characterization of their functions and appealing for a sophisticated approach to bridge genetic and genomic studies. Towards this problem, network-based prioritization of candidate genes, which performs integrated analysis of gene networks with GWAS data, has emerged as a promising direction and attracted much attention. However, most existing methods overlook the sparse and noisy properties of gene networks and thus may lead to suboptimal performance. Motivated by this understanding, we proposed a novel method called REGENT for integrating multiple gene networks with GWAS data to prioritize candidate genes for complex diseases. We leveraged a technique called the network representation learning to embed a gene network into a compact and robust feature space, and then designed a hierarchical statistical model to integrate features of multiple gene networks with GWAS data for the effective inference of genes associated with a disease of interest. We applied our method to six complex diseases and demonstrated the superior performance of REGENT over existing approaches in recovering known disease-associated genes. We further conducted a pathway analysis and showed that the ability of REGENT to discover disease-associated pathways. We expect to see applications of our method to a broad spectrum of diseases for post-GWAS analysis. REGENT is freely available at https://github.com/wmmthu/REGENT.


Asunto(s)
Redes Reguladoras de Genes , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo/métodos , Aprendizaje Automático , Polimorfismo de Nucleótido Simple , Programas Informáticos , Humanos
8.
BMC Genomics ; 19(Suppl 2): 84, 2018 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-29764360

RESUMEN

BACKGROUND: Precise identification of three-dimensional genome organization, especially enhancer-promoter interactions (EPIs), is important to deciphering gene regulation, cell differentiation and disease mechanisms. Currently, it is a challenging task to distinguish true interactions from other nearby non-interacting ones since the power of traditional experimental methods is limited due to low resolution or low throughput. RESULTS: We propose a novel computational framework EP2vec to assay three-dimensional genomic interactions. We first extract sequence embedding features, defined as fixed-length vector representations learned from variable-length sequences using an unsupervised deep learning method in natural language processing. Then, we train a classifier to predict EPIs using the learned representations in supervised way. Experimental results demonstrate that EP2vec obtains F1 scores ranging from 0.841~ 0.933 on different datasets, which outperforms existing methods. We prove the robustness of sequence embedding features by carrying out sensitivity analysis. Besides, we identify motifs that represent cell line-specific information through analysis of the learned sequence embedding features by adopting attention mechanism. Last, we show that even superior performance with F1 scores 0.889~ 0.940 can be achieved by combining sequence embedding features and experimental features. CONCLUSIONS: EP2vec sheds light on feature extraction for DNA sequences of arbitrary lengths and provides a powerful approach for EPIs identification.


Asunto(s)
Biología Computacional/métodos , Regiones Promotoras Genéticas , Secuencias Reguladoras de Ácidos Nucleicos , Línea Celular Tumoral , Bases de Datos Genéticas , Células HeLa , Células Endoteliales de la Vena Umbilical Humana , Humanos , Células K562 , Procesamiento de Lenguaje Natural , Análisis de Secuencia de ADN , Aprendizaje Automático no Supervisado
9.
Bioinformatics ; 33(14): i92-i101, 2017 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-28881969

RESUMEN

MOTIVATION: Experimental techniques for measuring chromatin accessibility are expensive and time consuming, appealing for the development of computational approaches to predict open chromatin regions from DNA sequences. Along this direction, existing methods fall into two classes: one based on handcrafted k -mer features and the other based on convolutional neural networks. Although both categories have shown good performance in specific applications thus far, there still lacks a comprehensive framework to integrate useful k -mer co-occurrence information with recent advances in deep learning. RESULTS: We fill this gap by addressing the problem of chromatin accessibility prediction with a convolutional Long Short-Term Memory (LSTM) network with k -mer embedding. We first split DNA sequences into k -mers and pre-train k -mer embedding vectors based on the co-occurrence matrix of k -mers by using an unsupervised representation learning approach. We then construct a supervised deep learning architecture comprised of an embedding layer, three convolutional layers and a Bidirectional LSTM (BLSTM) layer for feature learning and classification. We demonstrate that our method gains high-quality fixed-length features from variable-length sequences and consistently outperforms baseline methods. We show that k -mer embedding can effectively enhance model performance by exploring different embedding strategies. We also prove the efficacy of both the convolution and the BLSTM layers by comparing two variations of the network architecture. We confirm the robustness of our model to hyper-parameters by performing sensitivity analysis. We hope our method can eventually reinforce our understanding of employing deep learning in genomic studies and shed light on research regarding mechanisms of chromatin accessibility. AVAILABILITY AND IMPLEMENTATION: The source code can be downloaded from https://github.com/minxueric/ismb2017_lstm . CONTACT: tingchen@tsinghua.edu.cn or ruijiang@tsinghua.edu.cn. SUPPLEMENTARY INFORMATION: Supplementary materials are available at Bioinformatics online.


Asunto(s)
Cromatina/metabolismo , Biología Computacional/métodos , Redes Neurales de la Computación , Programas Informáticos , Línea Celular , Humanos
10.
BMC Bioinformatics ; 18(Suppl 13): 478, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-29219068

RESUMEN

BACKGROUND: With the rapid development of deep sequencing techniques in the recent years, enhancers have been systematically identified in such projects as FANTOM and ENCODE, forming genome-wide landscapes in a series of human cell lines. Nevertheless, experimental approaches are still costly and time consuming for large scale identification of enhancers across a variety of tissues under different disease status, making computational identification of enhancers indispensable. RESULTS: To facilitate the identification of enhancers, we propose a computational framework, named DeepEnhancer, to distinguish enhancers from background genomic sequences. Our method purely relies on DNA sequences to predict enhancers in an end-to-end manner by using a deep convolutional neural network (CNN). We train our deep learning model on permissive enhancers and then adopt a transfer learning strategy to fine-tune the model on enhancers specific to a cell line. Results demonstrate the effectiveness and efficiency of our method in the classification of enhancers against random sequences, exhibiting advantages of deep learning over traditional sequence-based classifiers. We then construct a variety of neural networks with different architectures and show the usefulness of such techniques as max-pooling and batch normalization in our method. To gain the interpretability of our approach, we further visualize convolutional kernels as sequence logos and successfully identify similar motifs in the JASPAR database. CONCLUSIONS: DeepEnhancer enables the identification of novel enhancers using only DNA sequences via a highly accurate deep learning model. The proposed computational framework can also be applied to similar problems, thereby prompting the use of machine learning methods in life sciences.


Asunto(s)
Algoritmos , ADN/química , Elementos de Facilitación Genéticos , Modelos Genéticos , Redes Neurales de la Computación , Biología Computacional , ADN/genética , Bases de Datos Factuales , Genoma Humano , Genómica , Humanos , Aprendizaje Automático
11.
bioRxiv ; 2024 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-37502861

RESUMEN

The inherent similarities between natural language and biological sequences have given rise to great interest in adapting the transformer-based large language models (LLMs) underlying recent breakthroughs in natural language processing (references), for applications in genomics. However, current LLMs for genomics suffer from several limitations such as the inability to include chromatin interactions in the training data, and the inability to make prediction in new cellular contexts not represented in the training data. To mitigate these problems, we propose EpiGePT, a transformer-based pretrained language model for predicting context-specific epigenomic signals and chromatin contacts. By taking the context-specific activities of transcription factors (TFs) and 3D genome interactions into consideration, EpiGePT offers wider applicability and deeper biological insights than models trained on DNA sequence only. In a series of experiments, EpiGePT demonstrates superior performance in a diverse set of epigenomic signals prediction tasks when compared to existing methods. In particular, our model enables cross-cell-type prediction of long-range interactions and offers insight on the functional impact of genetic variants under different cellular contexts. These new capabilities will enhance the usefulness of LLM in the study of gene regulatory mechanisms. We provide free online prediction service of EpiGePT through http://health.tsinghua.edu.cn/epigept/.

12.
Bioinform Adv ; 3(1): vbad001, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36845200

RESUMEN

Summary: The transformer-based language models, including vanilla transformer, BERT and GPT-3, have achieved revolutionary breakthroughs in the field of natural language processing (NLP). Since there are inherent similarities between various biological sequences and natural languages, the remarkable interpretability and adaptability of these models have prompted a new wave of their application in bioinformatics research. To provide a timely and comprehensive review, we introduce key developments of transformer-based language models by describing the detailed structure of transformers and summarize their contribution to a wide range of bioinformatics research from basic sequence analysis to drug discovery. While transformer-based applications in bioinformatics are diverse and multifaceted, we identify and discuss the common challenges, including heterogeneity of training data, computational expense and model interpretability, and opportunities in the context of bioinformatics research. We hope that the broader community of NLP researchers, bioinformaticians and biologists will be brought together to foster future research and development in transformer-based language models, and inspire novel bioinformatics applications that are unattainable by traditional methods. Supplementary information: Supplementary data are available at Bioinformatics Advances online.

13.
Database (Oxford) ; 20192019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30788500

RESUMEN

Genome-wide association studies have successfully identified thousands of genomic loci potentially associated with hundreds of complex traits in the past decade. Nevertheless, the fact that more than 90% of such disease-associated variants lie in non-coding DNA with unknown functional implications has been appealing for advanced analysis of plenty of genetic variants. Toward this goal, recent studies focusing on individual non-coding variants have revealed that complex diseases are often the consequences of erroneous interactions between enhancers and their target genes. However, such enhancer-disease associations are dispersed in a variety of independent studies, and thus far it is still difficult to carry out comprehensive downstream analysis with these experimentally supported enhancer-disease associations. To fill in this gap, we collected experimentally supported associations between complex diseases and enhancers and then developed a manually curated database called EnDisease (http://bioinfo.au.tsinghua.edu.cn/endisease/). Concretely, EnDisease documents 535 associations between 133 diseases and 454 enhancers, extracted from 199 articles. Moreover, after annotating these enhancers using 649 human and 115 mouse DNase-seq experiments, we find that cancer-related enhancers tend to be open across a large number of cell types. This database provides a user-friendly interface for browsing and searching, and it also allows users to download data freely. EnDisease has the potential to become a helpful and important resource for researchers who aim to understand the molecular mechanisms of enhancers involved in complex diseases.


Asunto(s)
Curaduría de Datos , Bases de Datos de Ácidos Nucleicos , Enfermedad/genética , Elementos de Facilitación Genéticos , Estudio de Asociación del Genoma Completo , Animales , Cromatina/metabolismo , Humanos , Anotación de Secuencia Molecular , Interfaz Usuario-Computador
14.
Nat Commun ; 10(1): 4613, 2019 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-31601804

RESUMEN

Characterizing and interpreting heterogeneous mixtures at the cellular level is a critical problem in genomics. Single-cell assays offer an opportunity to resolve cellular level heterogeneity, e.g., scRNA-seq enables single-cell expression profiling, and scATAC-seq identifies active regulatory elements. Furthermore, while scHi-C can measure the chromatin contacts (i.e., loops) between active regulatory elements to target genes in single cells, bulk HiChIP can measure such contacts in a higher resolution. In this work, we introduce DC3 (De-Convolution and Coupled-Clustering) as a method for the joint analysis of various bulk and single-cell data such as HiChIP, RNA-seq and ATAC-seq from the same heterogeneous cell population. DC3 can simultaneously identify distinct subpopulations, assign single cells to the subpopulations (i.e., clustering) and de-convolve the bulk data into subpopulation-specific data. The subpopulation-specific profiles of gene expression, chromatin accessibility and enhancer-promoter contact obtained by DC3 provide a comprehensive characterization of the gene regulatory system in each subpopulation.


Asunto(s)
Algoritmos , Análisis por Conglomerados , Perfilación de la Expresión Génica/estadística & datos numéricos , Genómica/estadística & datos numéricos , Análisis de la Célula Individual/estadística & datos numéricos , Animales , Línea Celular , Cromatina , Inmunoprecipitación de Cromatina/estadística & datos numéricos , Simulación por Computador , Perfilación de la Expresión Génica/métodos , Redes Reguladoras de Genes , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/estadística & datos numéricos , Humanos , Ratones , Regiones Promotoras Genéticas , Análisis de la Célula Individual/métodos
15.
BMC Syst Biol ; 11(Suppl 4): 76, 2017 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-28950906

RESUMEN

BACKGROUND: The human phenome has been widely used with a variety of genomic data sources in the inference of disease genes. However, most existing methods thus far derive phenotype similarity based on the analysis of biomedical databases by using the traditional term frequency-inverse document frequency (TF-IDF) formulation. This framework, though intuitive, not only ignores semantic relationships between words but also tends to produce high-dimensional vectors, and hence lacks the ability to precisely capture intrinsic semantic characteristics of biomedical documents. To overcome these limitations, we propose a framework called mimvec to analyze the human phenome by making use of the state-of-the-art deep learning technique in natural language processing. RESULTS: We converted 24,061 records in the Online Mendelian Inheritance in Man (OMIM) database to low-dimensional vectors using our method. We demonstrated that the vector presentation not only effectively enabled classification of phenotype records against gene ones, but also succeeded in discriminating diseases of different inheritance styles and different mechanisms. We further derived pairwise phenotype similarities between 7988 human inherited diseases using their vector presentations. With a joint analysis of this phenome with multiple genomic data, we showed that phenotype overlap indeed implied genotype overlap. We finally used the derived phenotype similarities with genomic data to prioritize candidate genes and demonstrated advantages of this method over existing ones. CONCLUSIONS: Our method is capable of not only capturing semantic relationships between words in biomedical records but also alleviating the dimensional disaster accompanying the traditional TF-IDF framework. With the approaching of precision medicine, there will be abundant electronic records of medicine and health awaiting for deep analysis, and we expect to see a wide spectrum of applications borrowing the idea of our method in the near future.


Asunto(s)
Biología Computacional/métodos , Aprendizaje Automático , Fenotipo , Bases de Datos Genéticas , Enfermedad/genética , Humanos , Patrón de Herencia , Procesamiento de Lenguaje Natural
16.
Artículo en Inglés | MEDLINE | ID: mdl-26989155

RESUMEN

The recent advancement of the next generation sequencing technology has enabled the fast and low-cost detection of all genetic variants spreading across the entire human genome, making the application of whole-genome sequencing a tendency in the study of disease-causing genetic variants. Nevertheless, there still lacks a repository that collects predictions of functionally damaging effects of human genetic variants, though it has been well recognized that such predictions play a central role in the analysis of whole-genome sequencing data. To fill this gap, we developed a database named dbWGFP (a database and web server of human whole-genome single nucleotide variants and their functional predictions) that contains functional predictions and annotations of nearly 8.58 billion possible human whole-genome single nucleotide variants. Specifically, this database integrates 48 functional predictions calculated by 17 popular computational methods and 44 valuable annotations obtained from various data sources. Standalone software, user-friendly query services and free downloads of this database are available at http://bioinfo.au.tsinghua.edu.cn/dbwgfp. dbWGFP provides a valuable resource for the analysis of whole-genome sequencing, exome sequencing and SNP array data, thereby complementing existing data sources and computational resources in deciphering genetic bases of human inherited diseases.


Asunto(s)
Bases de Datos Genéticas , Genoma Humano , Internet , Polimorfismo de Nucleótido Simple/genética , Biología Computacional , Humanos , Motor de Búsqueda , Programas Informáticos , Estadísticas no Paramétricas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA