Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Methods ; 203: 125-138, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35436514

RESUMO

N6-methyladenosine (m6A) is the most abundant eukaryotic modification internal mRNA, which plays the crucial roles in the occurrence and development of cancer. However, current knowledge about m6A-mediated functional circuit and key genes targeted by m6A methylation in cancer is mostly elusive. Thus, here we proposed a novel network-based approach (called m6Acancer-Net) to identify m6A-mediated driver genes and their associated network in specific type of cancer, such as acute myeloid leukemia. m6A-mediated cancer driver genes are defined as genes mediated by m6A methylation, significantly mutated, and functionally interacted in cancer. m6Acancer-Net identified the m6A-mediated cancer driver genes by combining gene functional interaction network with RNA methylation, gene expression and mutation information. A cancer-specific gene-site heterogeneous network was firstly constructed by connecting the m6A site co-methylation network with the functional interaction pruned gene co-expression network generated from large scale gene expression profile of specific cancer. Then, the functional m6A-mediated genes were identified by selecting the m6A regulators as seed genes to perform the random walk with restart algorithm on the gene-site heterogeneous network. Finally, m6A-mediated cancer driver gene subnetworks were constructed by performing the heat diffusion of mutation frequency for functional m6A-mediated genes in protein-protein interaction networks. The experimental results of m6Acancer-Net on the acute myeloid leukemia (AML) and glioblastoma multiforme (GBM) data from TCGA project show that the m6A-mediated caner driver genes identified by m6Acancer-Net are targeted by m6A regulators, and mediate significant cancer-related pathways. They play crucial roles in development and prognostic stratification of cancer. Moreover, 15 m6A-mediated cancer driver genes identified in AML are validated by literatures to mediate AML progress, and 14 m6A-mediated cancer driver genes identified in GBM are validated by literatures to participate in development of GBM. m6Acancer-Net is reliable to identify the functionally significant m6A-mediated driver genes in specific cancer, and it can effectively facilitate the understanding of regulatory and therapeutic mechanism of cancer driver genes in epitranscriptome layer.


Assuntos
Redes Reguladoras de Genes , Glioblastoma , Algoritmos , Glioblastoma/genética , Humanos , Mutação , Mapas de Interação de Proteínas/genética
2.
Methods ; 203: 167-178, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35314342

RESUMO

N6-methyladenosine (m6A) is the most abundant form of mRNA modification and plays an important role in regulating gene expression. However, the mechanisms of m6A regulated gene expression in cell or condition specific, are still poorly understood. Even though, some methods are able to predict m6A regulated expression (m6A-reg-exp) genes in specific context, they don't introduce the m6A reader binding information, while this information can help to predict m6A-reg-exp genes and more clearly to explain the mechanisms of m6A-mediated gene expression process. Thus, by integrating m6A sites and reader binding information, we proposed a novel method (called m6Aexpress-Reader) to predict m6A-reg-exp genes from limited MeRIP-seq data in specific context. m6Aexpress-Reader adopts the reader binding signal strength to weight the posterior distribution of the estimated regulatory coefficients for enhancing the prediction power. By using m6Aexpress-Reader, we found the complex characteristic of m6A on gene expression regulation and the distinct regulated pattern of m6A-reg-exp genes with different reader binding. m6A readers, YTHDF2 or IGF2BP1/3 all play an important role in various cancers and the key cancer pathways. In addition, m6Aexpress-Reader reveals the distinct m6A regulated mode of reader targeted genes in cancer. m6Aexpress-Reader could be a useful tool for studying the m6A regulation on reader target genes in specific context and it can be freely accessible at: https://github.com/NWPU-903PR/m6AexpressReader.


Assuntos
Neoplasias , Proteínas de Ligação a RNA , Adenosina/genética , Adenosina/metabolismo , Regulação da Expressão Gênica , Humanos , Neoplasias/genética , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo
3.
Nucleic Acids Res ; 49(20): e116, 2021 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-34417605

RESUMO

N6-methyladenosine (m6A) is the most abundant form of mRNA modification and controls many aspects of RNA metabolism including gene expression. However, the mechanisms by which m6A regulates cell- and condition-specific gene expression are still poorly understood, partly due to a lack of tools capable of identifying m6A sites that regulate gene expression under different conditions. Here we develop m6A-express, the first algorithm for predicting condition-specific m6A regulation of gene expression (m6A-reg-exp) from limited methylated RNA immunoprecipitation sequencing (MeRIP-seq) data. Comprehensive evaluations of m6A-express using simulated and real data demonstrated its high prediction specificity and sensitivity. When only a few MeRIP-seq samples may be available for the cellular or treatment conditions, m6A-express is particularly more robust than the log-linear model. Using m6A-express, we reported that m6A writers, METTL3 and METTL14, competitively regulate the transcriptional processes by mediating m6A-reg-exp of different genes in Hela cells. In contrast, METTL3 induces different m6A-reg-exp of a distinct group of genes in HepG2 cells to regulate protein functions and stress-related processes. We further uncovered unique m6A-reg-exp patterns in human brain and intestine tissues, which are enriched in organ-specific processes. This study demonstrates the effectiveness of m6A-express in predicting condition-specific m6A-reg-exp and highlights the complex, condition-specific nature of m6A-regulation of gene expression.


Assuntos
Adenosina/análogos & derivados , Processamento Pós-Transcricional do RNA , Análise de Sequência de RNA/métodos , Adenosina/metabolismo , Encéfalo/metabolismo , Células HeLa , Células Hep G2 , Humanos , Mucosa Intestinal/metabolismo
4.
Bioinformatics ; 37(22): 4277-4279, 2021 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-33974000

RESUMO

MOTIVATION: N 6-methyladenosine (m6A) is the most abundant mammalian mRNA methylation with versatile functions. To date, although a number of bioinformatics tools have been developed for location discovery of m6A modification, functional understanding is still quite limited. As the focus of RNA epigenetics gradually shifts from site discovery to functional studies, there is an urgent need for user-friendly tools to identify and explore the functional relevance of context-specific m6A methylation to gain insights into the epitranscriptome layer of gene expression regulation. RESULTS: We introduced here Funm6AViewer, a novel platform to identify, prioritize and visualize the functional gene interaction networks mediated by dynamic m6A RNA methylation unveiled from a case control study. By taking the differential RNA methylation data and differential gene expression data, both of which can be inferred from the widely used MeRIP-seq data, as the inputs, Funm6AViewer enables a series of analysis, including: (i) examining the distribution of differential m6A sites, (ii) prioritizing the genes mediated by dynamic m6A methylation and (iii) characterizing functionally the gene regulatory networks mediated by condition-specific m6A RNA methylation. Funm6AViewer should effectively facilitate the understanding of the epitranscriptome circuitry mediated by this reversible RNA modification. AVAILABILITY AND IMPLEMENTATION: Funm6AViewer is available both as a convenient web server (https://www.xjtlu.edu.cn/biologicalsciences/funm6aviewer) with graphical interface and as an independent R package (https://github.com/NWPU-903PR/Funm6AViewer) for local usage. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Epigênese Genética , RNA , Animais , Metilação , Estudos de Casos e Controles , RNA/metabolismo , Redes Reguladoras de Genes , Adenosina/metabolismo , Mamíferos/genética
5.
Bioinformatics ; 35(14): i90-i98, 2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-31510685

RESUMO

MOTIVATION: As the most abundant mammalian mRNA methylation, N6-methyladenosine (m6A) exists in >25% of human mRNAs and is involved in regulating many different aspects of mRNA metabolism, stem cell differentiation and diseases like cancer. However, our current knowledge about dynamic changes of m6A levels and how the change of m6A levels for a specific gene can play a role in certain biological processes like stem cell differentiation and diseases like cancer is largely elusive. RESULTS: To address this, we propose in this paper FunDMDeep-m6A a novel pipeline for identifying context-specific (e.g. disease versus normal, differentiated cells versus stem cells or gene knockdown cells versus wild-type cells) m6A-mediated functional genes. FunDMDeep-m6A includes, at the first step, DMDeep-m6A a novel method based on a deep learning model and a statistical test for identifying differential m6A methylation (DmM) sites from MeRIP-Seq data at a single-base resolution. FunDMDeep-m6A then identifies and prioritizes functional DmM genes (FDmMGenes) by combing the DmM genes (DmMGenes) with differential expression analysis using a network-based method. This proposed network method includes a novel m6A-signaling bridge (MSB) score to quantify the functional significance of DmMGenes by assessing functional interaction of DmMGenes with their signaling pathways using a heat diffusion process in protein-protein interaction (PPI) networks. The test results on 4 context-specific MeRIP-Seq datasets showed that FunDMDeep-m6A can identify more context-specific and functionally significant FDmMGenes than m6A-Driver. The functional enrichment analysis of these genes revealed that m6A targets key genes of many important context-related biological processes including embryonic development, stem cell differentiation, transcription, translation, cell death, cell proliferation and cancer-related pathways. These results demonstrate the power of FunDMDeep-m6A for elucidating m6A regulatory functions and its roles in biological processes and diseases. AVAILABILITY AND IMPLEMENTATION: The R-package for DMDeep-m6A is freely available from https://github.com/NWPU-903PR/DMDeepm6A1.0. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Neoplasias , Mapas de Interação de Proteínas , RNA , Animais , Humanos , Metilação , Neoplasias/genética , RNA Mensageiro , Software
6.
PLoS Comput Biol ; 15(1): e1006663, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30601803

RESUMO

N6-methyladenosine (m6A) is the most abundant methylation, existing in >25% of human mRNAs. Exciting recent discoveries indicate the close involvement of m6A in regulating many different aspects of mRNA metabolism and diseases like cancer. However, our current knowledge about how m6A levels are controlled and whether and how regulation of m6A levels of a specific gene can play a role in cancer and other diseases is mostly elusive. We propose in this paper a computational scheme for predicting m6A-regulated genes and m6A-associated disease, which includes Deep-m6A, the first model for detecting condition-specific m6A sites from MeRIP-Seq data with a single base resolution using deep learning and Hot-m6A, a new network-based pipeline that prioritizes functional significant m6A genes and its associated diseases using the Protein-Protein Interaction (PPI) and gene-disease heterogeneous networks. We applied Deep-m6A and this pipeline to 75 MeRIP-seq human samples, which produced a compact set of 709 functionally significant m6A-regulated genes and nine functionally enriched subnetworks. The functional enrichment analysis of these genes and networks reveal that m6A targets key genes of many critical biological processes including transcription, cell organization and transport, and cell proliferation and cancer-related pathways such as Wnt pathway. The m6A-associated disease analysis prioritized five significantly associated diseases including leukemia and renal cell carcinoma. These results demonstrate the power of our proposed computational scheme and provide new leads for understanding m6A regulatory functions and its roles in diseases.


Assuntos
Adenosina/análogos & derivados , Biologia Computacional/métodos , Marcadores Genéticos/genética , Neoplasias/genética , Software , Adenosina/genética , Algoritmos , Aprendizado Profundo , Humanos , Neoplasias/metabolismo , Mapas de Interação de Proteínas/genética
7.
Int J Mol Sci ; 21(15)2020 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-32718000

RESUMO

Long non-coding RNAs (lncRNAs) play crucial roles in diverse biological processes and human complex diseases. Distinguishing lncRNAs from protein-coding transcripts is a fundamental step for analyzing the lncRNA functional mechanism. However, the experimental identification of lncRNAs is expensive and time-consuming. In this study, we presented an alignment-free multimodal deep learning framework (namely lncRNA_Mdeep) to distinguish lncRNAs from protein-coding transcripts. LncRNA_Mdeep incorporated three different input modalities, then a multimodal deep learning framework was built for learning the high-level abstract representations and predicting the probability whether a transcript was lncRNA or not. LncRNA_Mdeep achieved 98.73% prediction accuracy in a 10-fold cross-validation test on humans. Compared with other eight state-of-the-art methods, lncRNA_Mdeep showed 93.12% prediction accuracy independent test on humans, which was 0.94%~15.41% higher than that of other eight methods. In addition, the results on 11 cross-species datasets showed that lncRNA_Mdeep was a powerful predictor for predicting lncRNAs.


Assuntos
Bases de Dados de Ácidos Nucleicos , Aprendizado Profundo , RNA Longo não Codificante/genética , Software , Animais , Humanos , Camundongos
8.
BMC Bioinformatics ; 20(1): 87, 2019 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-30782113

RESUMO

BACKGROUND: Long non-coding RNAs play an important role in human complex diseases. Identification of lncRNA-disease associations will gain insight into disease-related lncRNAs and benefit disease diagnoses and treatment. However, using experiments to explore the lncRNA-disease associations is expensive and time consuming. RESULTS: In this study, we developed a novel method to identify potential lncRNA-disease associations by Integrating Diverse Heterogeneous Information sources with positive pointwise Mutual Information and Random Walk with restart algorithm (namely IDHI-MIRW). IDHI-MIRW first constructs multiple lncRNA similarity networks and disease similarity networks from diverse lncRNA-related and disease-related datasets, then implements the random walk with restart algorithm on these similarity networks for extracting the topological similarities which are fused with positive pointwise mutual information to build a large-scale lncRNA-disease heterogeneous network. Finally, IDHI-MIRW implemented random walk with restart algorithm on the lncRNA-disease heterogeneous network to infer potential lncRNA-disease associations. CONCLUSIONS: Compared with other state-of-the-art methods, IDHI-MIRW achieves the best prediction performance. In case studies of breast cancer, stomach cancer, and colorectal cancer, 36/45 (80%) novel lncRNA-disease associations predicted by IDHI-MIRW are supported by recent literatures. Furthermore, we found lncRNA LINC01816 is associated with the survival of colorectal cancer patients. IDHI-MIRW is freely available at https://github.com/NWPU-903PR/IDHI-MIRW .


Assuntos
Algoritmos , Biologia Computacional/métodos , Predisposição Genética para Doença , RNA Longo não Codificante/genética , Neoplasias Colorretais/genética , Estudos de Associação Genética , Humanos , Análise de Sequência de RNA
9.
PLoS Comput Biol ; 12(12): e1005287, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28027310

RESUMO

As the most prevalent mammalian mRNA epigenetic modification, N6-methyladenosine (m6A) has been shown to possess important post-transcriptional regulatory functions. However, the regulatory mechanisms and functional circuits of m6A are still largely elusive. To help unveil the regulatory circuitry mediated by mRNA m6A methylation, we develop here m6A-Driver, an algorithm for predicting m6A-driven genes and associated networks, whose functional interactions are likely to be actively modulated by m6A methylation under a specific condition. Specifically, m6A-Driver integrates the PPI network and the predicted differential m6A methylation sites from methylated RNA immunoprecipitation sequencing (MeRIP-Seq) data using a Random Walk with Restart (RWR) algorithm and then builds a consensus m6A-driven network of m6A-driven genes. To evaluate the performance, we applied m6A-Driver to build the context-specific m6A-driven networks for 4 known m6A (de)methylases, i.e., FTO, METTL3, METTL14 and WTAP. Our results suggest that m6A-Driver can robustly and efficiently identify m6A-driven genes that are functionally more enriched and associated with higher degree of differential expression than differential m6A methylated genes. Pathway analysis of the constructed context-specific m6A-driven gene networks further revealed the regulatory circuitry underlying the dynamic interplays between the methyltransferases and demethylase at the epitranscriptomic layer of gene regulation.


Assuntos
Adenosina/análogos & derivados , Metilação de DNA/genética , Redes Reguladoras de Genes/genética , Modelos Genéticos , RNA/genética , Análise de Sequência de RNA/métodos , Adenosina/genética , Algoritmos , Simulação por Computador
10.
Comput Struct Biotechnol J ; 19: 3015-3026, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34136099

RESUMO

RNA modifications, in particular N 6-methyladenosine (m6A), participate in every stages of RNA metabolism and play diverse roles in essential biological processes and disease pathogenesis. Thanks to the advances in sequencing technology, tens of thousands of RNA modification sites can be identified in a typical high-throughput experiment; however, it remains a major challenge to decipher the functional relevance of these sites, such as, affecting alternative splicing, regulation circuit in essential biological processes or association to diseases. As the focus of RNA epigenetics gradually shifts from site discovery to functional studies, we review here recent progress in functional annotation and prediction of RNA modification sites from a bioinformatics perspective. The review covers naïve annotation with associated biological events, e.g., single nucleotide polymorphism (SNP), RNA binding protein (RBP) and alternative splicing, prediction of key sites and their regulatory functions, inference of disease association, and mining the diagnosis and prognosis value of RNA modification regulators. We further discussed the limitations of existing approaches and some future perspectives.

11.
Comput Struct Biotechnol J ; 18: 1587-1604, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32670500

RESUMO

Post-transcriptional RNA modification occurs on all types of RNA and plays a vital role in regulating every aspect of RNA function. Thanks to the development of high-throughput sequencing technologies, transcriptome-wide profiling of RNA modifications has been made possible. With the accumulation of a large number of high-throughput datasets, bioinformatics approaches have become increasing critical for unraveling the epitranscriptome. We review here the recent progress in bioinformatics approaches for deciphering the epitranscriptomes, including epitranscriptome data analysis techniques, RNA modification databases, disease-association inference, general functional annotation, and studies on RNA modification site prediction. We also discuss the limitations of existing approaches and offer some future perspectives.

12.
Front Genet ; 10: 266, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31001320

RESUMO

Recent studies have revealed that the RNA N 6-methyladenosine (m6A) modification plays a critical role in a variety of biological processes and associated with multiple diseases including cancers. Till this day, transcriptome-wide m6A RNA methylation sites have been identified by high-throughput sequencing technique combined with computational methods, and the information is publicly available in a few bioinformatics databases; however, the association between individual m6A sites and various diseases are still largely unknown. There are yet computational approaches developed for investigating potential association between individual m6A sites and diseases, which represents a major challenge in the epitranscriptome analysis. Thus, to infer the disease-related m6A sites, we implemented a novel multi-layer heterogeneous network-based approach, which incorporates the associations among diseases, genes and m6A RNA methylation sites from gene expression, RNA methylation and disease similarities data with the Random Walk with Restart (RWR) algorithm. To evaluate the performance of the proposed approach, a ten-fold cross validation is performed, in which our approach achieved a reasonable good performance (overall AUC: 0.827, average AUC 0.867), higher than a hypergeometric test-based approach (overall AUC: 0.7333 and average AUC: 0.723) and a random predictor (overall AUC: 0.550 and average AUC: 0.486). Additionally, we show that a number of predicted cancer-associated m6A sites are supported by existing literatures, suggesting that the proposed approach can effectively uncover the underlying epitranscriptome circuits of disease mechanisms. An online database DRUM, which stands for disease-associated ribonucleic acid methylation, was built to support the query of disease-associated RNA m6A methylation sites, and is freely available at: www.xjtlu.edu.cn/biologicalsciences/drum.

13.
Mol Biosyst ; 12(2): 520-31, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26675534

RESUMO

The identification of potential drug-target interaction pairs is very important, which is useful not only for providing greater understanding of protein function, but also for enhancing drug research, especially for drug function repositioning. Recently, numerous machine learning-based algorithms (e.g. kernel-based, matrix factorization-based and network-based inference methods) have been developed for predicting drug-target interactions. All these methods implicitly utilize the assumption that similar drugs tend to target similar proteins and yield better results for predicting interactions between drugs and target proteins. To further improve the accuracy of prediction, a new method of network-based label propagation with mutual interaction information derived from heterogeneous networks, namely LPMIHN, is proposed to infer the potential drug-target interactions. LPMIHN separately performs label propagation on drug and target similarity networks, but the initial label information of the target (or drug) network comes from the drug (or target) label network and the known drug-target interaction bipartite network. The independent label propagation on each similarity network explores the cluster structure in its network, and the label information from the other network is used to capture mutual interactions (bicluster structures) between the nodes in each pair of the similarity networks. As compared to other recent state-of-the-art methods on the four popular benchmark datasets of binary drug-target interactions and two quantitative kinase bioactivity datasets, LPMIHN achieves the best results in terms of AUC and AUPR. In addition, many of the promising drug-target pairs predicted from LPMIHN are also confirmed on the latest publicly available drug-target databases such as ChEMBL, KEGG, SuperTarget and Drugbank. These results demonstrate the effectiveness of our LPMIHN method, indicating that LPMIHN has a great potential for predicting drug-target interactions.


Assuntos
Descoberta de Drogas/métodos , Proteínas/química , Algoritmos , Inteligência Artificial , Biologia Computacional , Simulação por Computador , Bases de Dados Factuais , Humanos
14.
Mol Biosyst ; 10(6): 1400-8, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24695957

RESUMO

The identification of disease genes is very important not only to provide greater understanding of gene function and cellular mechanisms which drive human disease, but also to enhance human disease diagnosis and treatment. Recently, high-throughput techniques have been applied to detect dozens or even hundreds of candidate genes. However, experimental approaches to validate the many candidates are usually time-consuming, tedious and expensive, and sometimes lack reproducibility. Therefore, numerous theoretical and computational methods (e.g. network-based approaches) have been developed to prioritize candidate disease genes. Many network-based approaches implicitly utilize the observation that genes causing the same or similar diseases tend to correlate with each other in gene-protein relationship networks. Of these network approaches, the random walk with restart algorithm (RWR) is considered to be a state-of-the-art approach. To further improve the performance of RWR, we propose a novel method named ESFSC to identify disease-related genes, by enlarging the seed set according to the centrality of disease genes in a network and fusing information of the protein-protein interaction (PPI) network topological similarity and the gene expression correlation. The ESFSC algorithm restarts at all of the nodes in the seed set consisting of the known disease genes and their k-nearest neighbor nodes, then walks in the global network separately guided by the similarity transition matrix constructed with PPI network topological similarity properties and the correlational transition matrix constructed with the gene expression profiles. As a result, all the genes in the network are ranked by weighted fusing the above results of the RWR guided by two types of transition matrices. Comprehensive simulation results of the 10 diseases with 97 known disease genes collected from the Online Mendelian Inheritance in Man (OMIM) database show that ESFSC outperforms existing methods for prioritizing candidate disease genes. The top prediction results of Alzheimer's disease are consistent with previous literature reports.


Assuntos
Biologia Computacional/métodos , Doença/genética , Estudos de Associação Genética/métodos , Algoritmos , Bases de Dados Genéticas , Doença/etiologia , Predisposição Genética para Doença , Humanos , Modelos Genéticos , Mapas de Interação de Proteínas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA