Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Brief Funct Genomics ; 2024 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-38944027

RESUMO

Acute myeloid leukemia (AML) is a type of blood cancer with diverse genetic variations and DNA methylation alterations. By studying the interaction of gene mutations, expression, and DNA methylation, we aimed to gain valuable insights into the processes that lead to block differentiation in AML. We analyzed TCGA-LAML data (173 samples) with RNA sequencing and DNA methylation arrays, comparing FLT3 mutant (48) and wild-type (125) cases. We conducted differential gene expression analysis using cBioPortal, identified DNA methylation differences with ChAMP tool, and correlated them with gene expression changes. Gene set enrichment analysis (g:Profiler) revealed significant biological processes and pathways. ShinyGo and GeneCards were used to find potential transcription factors and their binding sites among significant genes. We found significant differentially expressed genes (DEGs) negatively correlated with their most significant methylation probes (Pearson correlation coefficient of -0.49, P-value <0.001) between FLT3 mutant and wild-type groups. Moreover, our exploration of 450 k CpG sites uncovered a global hypo-methylated status in 168 DEGs. Notably, these methylation changes were enriched in the promoter regions of Homebox superfamily gene, which are crucial in transcriptional-regulating pathways in blood cancer. Furthermore, in FLT3 mutant AML patient samples, we observed overexpress of WT1, a transcription factor known to bind homeobox gene family. This finding suggests a potential mechanism by which WT1 recruits TET2 to demethylate specific genomic regions. Integrating gene expression and DNA methylation analyses shed light on the impact of FLT3 mutations on cancer cell development and differentiation, supporting a two-hit model in AML. This research advances understanding of AML and fosters targeted therapeutic strategy development.

2.
Hum Genet ; 143(1): 35-47, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38095720

RESUMO

Complex multi-omics effects drive the clustering of cardiometabolic risk factors, underscoring the imperative to comprehend how individual and combined omics shape phenotypic variation. Our study partitions phenotypic variance in metabolic syndrome (MetS), blood glucose (GLU), triglycerides (TG), high-density lipoprotein cholesterol (HDL-C), and blood pressure through genome, transcriptome, metabolome, and exposome (i.e., lifestyle exposome) analyses. Our analysis included a cohort of 62,822 unrelated individuals with white British ancestry, sourced from the UK biobank. We employed linear mixed models to partition phenotypic variance using the restricted maximum likelihood (REML) method, implemented in MTG2 (v2.22). We initiated the analysis by individually modeling omics, followed by subsequent integration of pairwise omics in a joint model that also accounted for the covariance and interaction between omics layers. Finally, we estimated the correlations of various omics effects between the phenotypes using bivariate REML. Significant proportions of the MetS variance were attributed to distinct data sources: genome (9.47%), transcriptome (4.24%), metabolome (14.34%), and exposome (3.77%). The phenotypic variances explained by the genome, transcriptome, metabolome, and exposome ranged from 3.28% for GLU to 25.35% for HDL-C, 0% for GLU to 19.34% for HDL-C, 4.29% for systolic blood pressure (SBP) to 35.75% for TG, and 0.89% for GLU to 10.17% for HDL-C, respectively. Significant correlations were found between genomic and transcriptomic effects for TG and HDL-C. Furthermore, significant interaction effects between omics data were detected for both MetS and its components. Interestingly, significant correlation of omics effect between the phenotypes was found. This study underscores omics' roles, interaction effects, and random-effects covariance in unveiling phenotypic variation in multi-omics domains.


Assuntos
Síndrome Metabólica , Humanos , Síndrome Metabólica/genética , Multiômica , Fenótipo , Triglicerídeos/genética , HDL-Colesterol
3.
PLoS Comput Biol ; 19(10): e1011308, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37812646

RESUMO

Non-coding RNAs (ncRNAs) act as important modulators of gene expression and they have been confirmed to play critical roles in the physiology and development of malignant tumors. Understanding the synergism of multiple ncRNAs in competing endogenous RNA (ceRNA) regulation can provide important insights into the mechanisms of malignant tumors caused by ncRNA regulation. In this work, we present a framework, SCOM, for identifying ncRNA synergistic competition. We systematically construct the landscape of ncRNA synergistic competition across 31 malignant tumors, and reveal that malignant tumors tend to share hub ncRNAs rather than the ncRNA interactions involved in the synergistic competition. In addition, the synergistic competition ncRNAs (i.e. ncRNAs involved in the synergistic competition) are likely to be involved in drug resistance, contribute to distinguishing molecular subtypes of malignant tumors, and participate in immune regulation. Furthermore, SCOM can help to infer ncRNA synergistic competition across malignant tumors and uncover potential diagnostic and prognostic biomarkers of malignant tumors. Altogether, the SCOM framework (https://github.com/zhangjunpeng411/SCOM/) and the resulting web-based database SCOMdb (https://comblab.cn/SCOMdb/) serve as a useful resource for exploring ncRNA regulation and to accelerate the identification of carcinogenic biomarkers.


Assuntos
Carcinógenos , Neoplasias , Humanos , RNA não Traduzido/genética , Neoplasias/genética , Carcinogênese/genética , Biomarcadores
4.
Artigo em Inglês | MEDLINE | ID: mdl-37018092

RESUMO

Instrumental variable (IV) is a powerful approach to inferring the causal effect of a treatment on an outcome of interest from observational data even when there exist latent confounders between the treatment and the outcome. However, existing IV methods require that an IV is selected and justified with domain knowledge. An invalid IV may lead to biased estimates. Hence, discovering a valid IV is critical to the applications of IV methods. In this article, we study and design a data-driven algorithm to discover valid IVs from data under mild assumptions. We develop the theory based on partial ancestral graphs (PAGs) to support the search for a set of candidate ancestral IVs (AIVs), and for each possible AIV, the identification of its conditioning set. Based on the theory, we propose a data-driven algorithm to discover a pair of IVs from data. The experiments on synthetic and real-world datasets show that the developed IV discovery algorithm estimates accurate estimates of causal effects in comparison with the state-of-the-art IV-based causal effect estimators.

5.
Wiley Interdiscip Rev RNA ; 13(2): e1686, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34342388

RESUMO

Inferring competing endogenous RNA (ceRNA) or microRNA (miRNA) sponge modules is a challenging and meaningful task for revealing ceRNA regulation mechanism at the module level. Modules in this context refer to groups of miRNA sponges which have mutual competitions and act as functional units for achieving biological processes. The recent development of computational methods based on heterogeneous data provides a novel way to discern the competitive effects of miRNA sponges on human complex diseases. This article aims to provide a comprehensive perspective of miRNA sponge module discovery methods. We first review the publicly available databases of cancer-related miRNA sponges, as the miRNA sponges involved in human cancers contribute to the discovery of cancer-associated modules. Then we review the existing computational methods for inferring miRNA sponge modules. Furthermore, we conduct an assessment on the performance of the module discovery methods with the pan-cancer dataset, and the comparison study indicates that it is useful to infer biologically meaningful miRNA sponge modules by directly mapping heterogeneous data to the competitive modules. Finally, we discuss the future directions and associated challenges in developing in silico methods to infer miRNA sponge modules. This article is categorized under: RNA Interactions with Proteins and Other Molecules > Small Molecule-RNA Interactions Regulatory RNAs/RNAi/Riboswitches > Regulatory RNAs.


Assuntos
MicroRNAs , Neoplasias , Redes Reguladoras de Genes , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Neoplasias/genética , RNA Mensageiro/metabolismo
6.
Bioinform Adv ; 2(1): vbac063, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36699386

RESUMO

Summary: MicroRNA (miRNA) sponges influence the capability of miRNA-mediated gene silencing by competing for shared miRNA response elements and play significant roles in many physiological and pathological processes. It has been proved that computational or dry-lab approaches are useful to guide wet-lab experiments for uncovering miRNA sponge regulation. However, all of the existing tools only allow the analysis of miRNA sponge regulation regarding a group of samples, rather than the miRNA sponge regulation unique to individual samples. Furthermore, most existing tools do not allow parallel computing for the fast identification of miRNA sponge regulation. Here, we present an enhanced version of our R/Bioconductor package, miRspongeR 2.0. Compared with the original version introduced in 2019, this package extends the resolution of miRNA sponge regulation from the multi-sample level to the single-sample level. Moreover, it supports the identification of miRNA sponge networks using parallel computing, and the construction of sample-sample correlation networks. It also provides more computational methods to infer miRNA sponge regulation and expands the ground truth for validation. With these new features, we anticipate that miRspongeR 2.0 will further accelerate the research on miRNA sponges with higher resolution and more utilities. Availability and implementation: http://bioconductor.org/packages/miRspongeR/. Supplementary information: Supplementary data are available at Bioinformatics Advances online.

7.
BMC Bioinformatics ; 22(1): 578, 2021 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-34856921

RESUMO

BACKGROUND: Existing computational methods for studying miRNA regulation are mostly based on bulk miRNA and mRNA expression data. However, bulk data only allows the analysis of miRNA regulation regarding a group of cells, rather than the miRNA regulation unique to individual cells. Recent advance in single-cell miRNA-mRNA co-sequencing technology has opened a way for investigating miRNA regulation at single-cell level. However, as currently single-cell miRNA-mRNA co-sequencing data is just emerging and only available at small-scale, there is a strong need of novel methods to exploit existing single-cell data for the study of cell-specific miRNA regulation. RESULTS: In this work, we propose a new method, CSmiR (Cell-Specific miRNA regulation) to combine single-cell miRNA-mRNA co-sequencing data and putative miRNA-mRNA binding information to identify miRNA regulatory networks at the resolution of individual cells. We apply CSmiR to the miRNA-mRNA co-sequencing data in 19 K562 single-cells to identify cell-specific miRNA-mRNA regulatory networks for understanding miRNA regulation in each K562 single-cell. By analyzing the obtained cell-specific miRNA-mRNA regulatory networks, we observe that the miRNA regulation in each K562 single-cell is unique. Moreover, we conduct detailed analysis on the cell-specific miRNA regulation associated with the miR-17/92 family as a case study. The comparison results indicate that CSmiR is effective in predicting cell-specific miRNA targets. Finally, through exploring cell-cell similarity matrix characterized by cell-specific miRNA regulation, CSmiR provides a novel strategy for clustering single-cells and helps to understand cell-cell crosstalk. CONCLUSIONS: To the best of our knowledge, CSmiR is the first method to explore miRNA regulation at a single-cell resolution level, and we believe that it can be a useful method to enhance the understanding of cell-specific miRNA regulation.


Assuntos
MicroRNAs , Análise por Conglomerados , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , MicroRNAs/genética , RNA Mensageiro/genética
8.
Theranostics ; 11(11): 5553-5568, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33859763

RESUMO

Identifying the genes responsible for driving cancer is of critical importance for directing treatment. Accordingly, multiple computational tools have been developed to facilitate this task. Due to the different methods employed by these tools, different data considered by the tools, and the rapidly evolving nature of the field, the selection of an appropriate tool for cancer driver discovery is not straightforward. This survey seeks to provide a comprehensive review of the different computational methods for discovering cancer drivers. We categorise the methods into three groups; methods for single driver identification, methods for driver module identification, and methods for identifying personalised cancer drivers. In addition to providing a "one-stop" reference of these methods, by evaluating and comparing their performance, we also provide readers the information about the different capabilities of the methods in identifying biologically significant cancer drivers. The biologically relevant information identified by these tools can be seen through the enrichment of discovered cancer drivers in GO biological processes and KEGG pathways and through our identification of a small cancer-driver cohort that is capable of stratifying patient survival.


Assuntos
Biologia Computacional/métodos , Neoplasias/genética , Humanos , Transdução de Sinais/genética
9.
IEEE/ACM Trans Comput Biol Bioinform ; 18(6): 2841-2847, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33909569

RESUMO

The classification of clinical samples based on gene expression data is an important part of precision medicine. In this manuscript, we show how transforming gene expression data into a set of personalized (sample-specific) networks can allow us to harness existing graph-based methods to improve classifier performance. Existing approaches to personalized gene networks have the limitation that they depend on other samples in the data and must get re-computed whenever a new sample is introduced. Here, we propose a novel method, called Personalized Annotation-based Networks (PAN), that avoids this limitation by using curated annotation databases to transform gene expression data into a graph. Unlike competing methods, PANs are calculated for each sample independent of the population, making it a more efficient way to obtain single-sample networks. Using three breast cancer datasets as a case study, we show that PAN classifiers not only predict cancer relapse better than gene features alone, but also outperform PPI (protein-protein interactions) and population-level graph-based classifiers. This work demonstrates the practical advantages of graph-based classification for high-dimensional genomic data, while offering a new approach to making sample-specific networks. Supplementary information: PAN and the baselines are implemented in Python. Source code and data are available at https://github.com/thinng/PAN.


Assuntos
Neoplasias da Mama , Genômica/métodos , Anotação de Sequência Molecular/métodos , Recidiva Local de Neoplasia , Medicina de Precisão/métodos , Algoritmos , Neoplasias da Mama/diagnóstico , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Bases de Dados Genéticas , Feminino , Humanos , Recidiva Local de Neoplasia/diagnóstico , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/metabolismo , Recidiva Local de Neoplasia/patologia , Mapas de Interação de Proteínas/genética , Software , Transcriptoma/genética
10.
RNA Biol ; 18(12): 2308-2320, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-33822666

RESUMO

In molecular biology, microRNA (miRNA) sponges are RNA transcripts which compete with other RNA transcripts for binding with miRNAs. Research has shown that miRNA sponges have a fundamental impact on tissue development and disease progression. Generally, to achieve a specific biological function, miRNA sponges tend to form modules or communities in a biological system. Until now, however, there is still a lack of tools to aid researchers to infer and analyse miRNA sponge modules from heterogeneous data. To fill this gap, we develop an R/Bioconductor package, miRSM, for facilitating the procedure of inferring and analysing miRNA sponge modules. miRSM provides a collection of 50 co-expression analysis methods to identify gene co-expression modules (which are candidate miRNA sponge modules), four module discovery methods to infer miRNA sponge modules and seven modular analysis methods for investigating miRNA sponge modules. miRSM will enable researchers to quickly apply new datasets to infer and analyse miRNA sponge modules, and will consequently accelerate the research on miRNA sponges.


Assuntos
Regulação da Expressão Gênica , Redes Reguladoras de Genes , MicroRNAs/genética , RNA Mensageiro/genética , Software , Ligação Competitiva , Humanos , MicroRNAs/metabolismo , RNA Mensageiro/metabolismo
11.
Bioinformatics ; 37(8): 1140-1147, 2021 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-33119053

RESUMO

SUMMARY: The development of new drugs is costly, time consuming and often accompanied with safety issues. Drug repurposing can avoid the expensive and lengthy process of drug development by finding new uses for already approved drugs. In order to repurpose drugs effectively, it is useful to know which proteins are targeted by which drugs. Computational models that estimate the interaction strength of new drug-target pairs have the potential to expedite drug repurposing. Several models have been proposed for this task. However, these models represent the drugs as strings, which is not a natural way to represent molecules. We propose a new model called GraphDTA that represents drugs as graphs and uses graph neural networks to predict drug-target affinity. We show that graph neural networks not only predict drug-target affinity better than non-deep learning models, but also outperform competing deep learning methods. Our results confirm that deep learning models are appropriate for drug-target binding affinity prediction, and that representing drugs as graphs can lead to further improvements. AVAILABILITY OF IMPLEMENTATION: The proposed models are implemented in Python. Related data, pre-trained models and source code are publicly available at https://github.com/thinng/GraphDTA. All scripts and data needed to reproduce the post hoc statistical analysis are available from https://doi.org/10.5281/zenodo.3603523. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Redes Neurais de Computação , Preparações Farmacêuticas , Reposicionamento de Medicamentos , Proteínas , Software
12.
Bioinformatics ; 37(6): 807-814, 2021 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-33070184

RESUMO

MOTIVATION: microRNAs (miRNAs) are important gene regulators and they are involved in many biological processes, including cancer progression. Therefore, correctly identifying miRNA-mRNA interactions is a crucial task. To this end, a huge number of computational methods has been developed, but they mainly use the data at one snapshot and ignore the dynamics of a biological process. The recent development of single cell data and the booming of the exploration of cell trajectories using 'pseudotime' concept have inspired us to develop a pseudotime-based method to infer the miRNA-mRNA relationships characterizing a biological process by taking into account the temporal aspect of the process. RESULTS: We have developed a novel approach, called pseudotime causality, to find the causal relationships between miRNAs and mRNAs during a biological process. We have applied the proposed method to both single cell and bulk sequencing datasets for Epithelia to Mesenchymal Transition, a key process in cancer metastasis. The evaluation results show that our method significantly outperforms existing methods in finding miRNA-mRNA interactions in both single cell and bulk data. The results suggest that utilizing the pseudotemporal information from the data helps reveal the gene regulation in a biological process much better than using the static information. AVAILABILITY AND IMPLEMENTATION: R scripts and datasets can be found at https://github.com/AndresMCB/PTC. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Fenômenos Biológicos , MicroRNAs , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Redes Reguladoras de Genes , MicroRNAs/genética , RNA Mensageiro/genética
13.
PLoS Comput Biol ; 16(4): e1007851, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32324747

RESUMO

Until now, existing methods for identifying lncRNA related miRNA sponge modules mainly rely on lncRNA related miRNA sponge interaction networks, which may not provide a full picture of miRNA sponging activities in biological conditions. Hence there is a strong need of new computational methods to identify lncRNA related miRNA sponge modules. In this work, we propose a framework, LMSM, to identify LncRNA related MiRNA Sponge Modules from heterogeneous data. To understand the miRNA sponging activities in biological conditions, LMSM uses gene expression data to evaluate the influence of the shared miRNAs on the clustered sponge lncRNAs and mRNAs. We have applied LMSM to the human breast cancer (BRCA) dataset from The Cancer Genome Atlas (TCGA). As a result, we have found that the majority of LMSM modules are significantly implicated in BRCA and most of them are BRCA subtype-specific. Most of the mediating miRNAs act as crosslinks across different LMSM modules, and all of LMSM modules are statistically significant. Multi-label classification analysis shows that the performance of LMSM modules is significantly higher than baseline's performance, indicating the biological meanings of LMSM modules in classifying BRCA subtypes. The consistent results suggest that LMSM is robust in identifying lncRNA related miRNA sponge modules. Moreover, LMSM can be used to predict miRNA targets. Finally, LMSM outperforms a graph clustering-based strategy in identifying BRCA-related modules. Altogether, our study shows that LMSM is a promising method to investigate modular regulatory mechanism of sponge lncRNAs from heterogeneous data.


Assuntos
Neoplasias da Mama , Biologia Computacional/métodos , MicroRNAs/genética , RNA Longo não Codificante/genética , Algoritmos , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Análise por Conglomerados , Bases de Dados Genéticas , Feminino , Perfilação da Expressão Gênica , Humanos , MicroRNAs/análise , MicroRNAs/metabolismo , RNA Longo não Codificante/análise , RNA Longo não Codificante/metabolismo
14.
BMC Bioinformatics ; 21(1): 32, 2020 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-31996128

RESUMO

After publication of this supplement article [1], it was brought to our attention that the Fig. 3 was incorrect. The correct Fig. 3 is as below.

15.
IEEE Trans Pattern Anal Mach Intell ; 42(9): 2240-2256, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-30946660

RESUMO

Causal feature selection has attracted much attention in recent years, as the causal features selected imply the causal mechanism related to the class attribute, leading to more reliable prediction models built using them. Currently there is a need of developing multi-source feature selection methods, since in many applications data for studying the same problem has been collected from various sources, such as multiple gene expression datasets obtained from different experiments for studying the causes of the same disease. However, the state-of-the-art causal feature selection methods generally tackle a single dataset, and a direct application of the methods to multiple datasets will result in unreliable results as the datasets may have different distributions. To address the challenges, by utilizing the concept of causal invariance in causal inference, we first formulate the problem of causal feature selection with multiple datasets as a search problem for an invariant set across the datasets, then give the upper and lower bounds of the invariant set, and finally we propose a new Multi-source Causal Feature Selection algorithm, MCFS. Using synthetic and real world datasets and 16 feature selection methods, the extensive experiments have validated the effectiveness of MCFS.

16.
BMC Bioinformatics ; 20(Suppl 23): 613, 2019 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-31881825

RESUMO

BACKGROUND: Studying multiple microRNAs (miRNAs) synergism in gene regulation could help to understand the regulatory mechanisms of complicated human diseases caused by miRNAs. Several existing methods have been presented to infer miRNA synergism. Most of the current methods assume that miRNAs with shared targets at the sequence level are working synergistically. However, it is unclear if miRNAs with shared targets are working in concert to regulate the targets or they individually regulate the targets at different time points or different biological processes. A standard method to test the synergistic activities is to knock-down multiple miRNAs at the same time and measure the changes in the target genes. However, this approach may not be practical as we would have too many sets of miRNAs to test. RESULTS: n this paper, we present a novel framework called miRsyn for inferring miRNA synergism by using a causal inference method that mimics the multiple-intervention experiments, e.g. knocking-down multiple miRNAs, with observational data. Our results show that several miRNA-miRNA pairs that have shared targets at the sequence level are not working synergistically at the expression level. Moreover, the identified miRNA synergistic network is small-world and biologically meaningful, and a number of miRNA synergistic modules are significantly enriched in breast cancer. Our further analyses also reveal that most of synergistic miRNA-miRNA pairs show the same expression patterns. The comparison results indicate that the proposed multiple-intervention causal inference method performs better than the single-intervention causal inference method in identifying miRNA synergistic network. CONCLUSIONS: Taken together, the results imply that miRsyn is a promising framework for identifying miRNA synergism, and it could enhance the understanding of miRNA synergism in breast cancer.


Assuntos
Algoritmos , MicroRNAs/genética , Neoplasias da Mama/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Humanos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
17.
BMC Bioinformatics ; 20(1): 235, 2019 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-31077152

RESUMO

BACKGROUND: A microRNA (miRNA) sponge is an RNA molecule with multiple tandem miRNA response elements that can sequester miRNAs from their target mRNAs. Despite growing appreciation of the importance of miRNA sponges, our knowledge of their complex functions remains limited. Moreover, there is still a lack of miRNA sponge research tools that help researchers to quickly compare their proposed methods with other methods, apply existing methods to new datasets, or select appropriate methods for assisting in subsequent experimental design. RESULTS: To fill the gap, we present an R/Bioconductor package, miRspongeR, for simplifying the procedure of identifying and analyzing miRNA sponge interaction networks and modules. It provides seven popular methods and an integrative method to identify miRNA sponge interactions. Moreover, it supports the validation of miRNA sponge interactions and the identification of miRNA sponge modules, as well as functional enrichment and survival analysis of miRNA sponge modules. CONCLUSIONS: This package enables researchers to quickly evaluate their new methods, apply existing methods to new datasets, and consequently speed up miRNA sponge research.


Assuntos
Redes Reguladoras de Genes/genética , MicroRNAs/genética , Humanos
18.
Brief Bioinform ; 20(4): 1403-1419, 2019 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-29401217

RESUMO

It is known that noncoding RNAs (ncRNAs) cover ∼98% of the transcriptome, but do not encode proteins. Among ncRNAs, long noncoding RNAs (lncRNAs) are a large and diverse class of RNA molecules, and are thought to be a gold mine of potential oncogenes, anti-oncogenes and new biomarkers. Although only a minority of lncRNAs is functionally characterized, it is clear that they are important regulators to modulate gene expression and involve in many biological functions. To reveal the functions and regulatory mechanisms of lncRNAs, it is vital to understand how lncRNAs regulate their target genes for implementing specific biological functions. In this article, we review the computational methods for inferring lncRNA-mRNA interactions and the third-party databases of storing lncRNA-mRNA regulatory relationships. We have found that the existing methods are based on statistical correlations between the gene expression levels of lncRNAs and mRNAs, and may not reveal gene regulatory relationships which are causal relationships. Moreover, these methods do not consider the modularity of lncRNA-mRNA regulatory networks, and thus, the networks identified are not module-specific. To address the above two issues, we propose a novel method, MSLCRN, to infer and analyze module-specific lncRNA-mRNA causal regulatory networks. We have applied it into glioblastoma multiforme, lung squamous cell carcinoma, ovarian cancer and prostate cancer, respectively. The experimental results show that MSLCRN, as an expression-based method, could be a useful complementary method to study lncRNA regulations.


Assuntos
Redes Reguladoras de Genes , Neoplasias/genética , RNA Longo não Codificante/genética , RNA Mensageiro/genética , Neoplasias Encefálicas/genética , Carcinoma de Células Escamosas/genética , Causalidade , Biologia Computacional/métodos , Bases de Dados de Ácidos Nucleicos/estatística & dados numéricos , Feminino , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Humanos , Neoplasias Pulmonares/genética , Masculino , Análise de Sequência com Séries de Oligonucleotídeos/estatística & dados numéricos , Neoplasias Ovarianas/genética , Neoplasias da Próstata/genética
19.
IEEE/ACM Trans Comput Biol Bioinform ; 16(5): 1483-1495, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-27429444

RESUMO

Discovering causal relationships from observational data is a crucial problem and it has applications in many research areas. The PC algorithm is the state-of-the-art constraint based method for causal discovery. However, runtime of the PC algorithm, in the worst-case, is exponential to the number of nodes (variables), and thus it is inefficient when being applied to high dimensional data, e.g., gene expression datasets. On another note, the advancement of computer hardware in the last decade has resulted in the widespread availability of multi-core personal computers. There is a significant motivation for designing a parallelized PC algorithm that is suitable for personal computers and does not require end users' parallel computing knowledge beyond their competency in using the PC algorithm. In this paper, we develop parallel-PC, a fast and memory efficient PC algorithm using the parallel computing technique. We apply our method to a range of synthetic and real-world high dimensional datasets. Experimental results on a dataset from the DREAM 5 challenge show that the original PC algorithm could not produce any results after running more than 24 hours; meanwhile, our parallel-PC algorithm managed to finish within around 12 hours with a 4-core CPU computer, and less than six hours with a 8-core CPU computer. Furthermore, we integrate parallel-PC into a causal inference method for inferring miRNA-mRNA regulatory relationships. The experimental results show that parallel-PC helps improve both the efficiency and accuracy of the causal inference algorithm.


Assuntos
Algoritmos , Biologia Computacional/métodos , Mineração de Dados/métodos , Teorema de Bayes , Bases de Dados Genéticas , Perfilação da Expressão Gênica , Aprendizado de Máquina , Modelos Genéticos , Transcriptoma
20.
Bioinformatics ; 34(24): 4232-4240, 2018 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-29955818

RESUMO

Motivation: MicroRNAs (miRNAs) are small non-coding RNAs with the length of ∼22 nucleotides. miRNAs are involved in many biological processes including cancers. Recent studies show that long non-coding RNAs (lncRNAs) are emerging as miRNA sponges, playing important roles in cancer physiology and development. Despite accumulating appreciation of the importance of lncRNAs, the study of their complex functions is still in its preliminary stage. Based on the hypothesis of competing endogenous RNAs (ceRNAs), several computational methods have been proposed for investigating the competitive relationships between lncRNAs and miRNA target messenger RNAs (mRNAs). However, when the mRNAs are released from the control of miRNAs, it remains largely unknown as to how the sponge lncRNAs influence the expression levels of the endogenous miRNA targets. Results: We propose a novel method to construct lncRNA related miRNA sponge regulatory networks (LncmiRSRNs) by integrating matched lncRNA and mRNA expression profiles with clinical information and putative miRNA-target interactions. Using the method, we have constructed the LncmiRSRNs for four human cancers (glioblastoma multiforme, lung cancer, ovarian cancer and prostate cancer). Based on the networks, we discover that after being released from miRNA control, the target mRNAs are normally up-regulated by the sponge lncRNAs, and only a fraction of sponge lncRNA-mRNA regulatory relationships and hub lncRNAs are shared by the four cancers. Moreover, most sponge lncRNA-mRNA regulatory relationships show a rewired mode between different cancers, and a minority of sponge lncRNA-mRNA regulatory relationships conserved (appearing) in different cancers may act as a common pivot across cancers. Besides, differential and conserved hub lncRNAs may act as potential cancer drivers to influence the cancerous state in cancers. Functional enrichment and survival analysis indicate that the identified differential and conserved LncmiRSRN network modules work as functional units in biological processes, and can distinguish metastasis risks of cancers. Our analysis demonstrates the potential of integrating expression profiles, clinical information and miRNA-target interactions for investigating lncRNA regulatory mechanism. Availability and implementation: LncmiRSRN is freely available (https://github.com/zhangjunpeng411/LncmiRSRN). Supplementary information: Supplementary data are available at Bioinformatics online.


Assuntos
Redes Reguladoras de Genes , MicroRNAs , RNA Longo não Codificante , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , RNA Mensageiro/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...