Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 500
Filtrar
1.
Am J Hum Genet ; 2024 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-39053459

RESUMO

Mendelian randomization (MR) provides valuable assessments of the causal effect of exposure on outcome, yet the application of conventional MR methods for mapping risk genes encounters new challenges. One of the issues is the limited availability of expression quantitative trait loci (eQTLs) as instrumental variables (IVs), hampering the estimation of sparse causal effects. Additionally, the often context- or tissue-specific eQTL effects challenge the MR assumption of consistent IV effects across eQTL and GWAS data. To address these challenges, we propose a multi-context multivariable integrative MR framework, mintMR, for mapping expression and molecular traits as joint exposures. It models the effects of molecular exposures across multiple tissues in each gene region, while simultaneously estimating across multiple gene regions. It uses eQTLs with consistent effects across more than one tissue type as IVs, improving IV consistency. A major innovation of mintMR involves employing multi-view learning methods to collectively model latent indicators of disease relevance across multiple tissues, molecular traits, and gene regions. The multi-view learning captures the major patterns of disease relevance and uses these patterns to update the estimated tissue relevance probabilities. The proposed mintMR iterates between performing a multi-tissue MR for each gene region and joint learning the disease-relevant tissue probabilities across gene regions, improving the estimation of sparse effects across genes. We apply mintMR to evaluate the causal effects of gene expression and DNA methylation for 35 complex traits using multi-tissue QTLs as IVs. The proposed mintMR controls genome-wide inflation and offers insights into disease mechanisms.

2.
Brief Bioinform ; 25(4)2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-38920342

RESUMO

Effective molecular representation learning is very important for Artificial Intelligence-driven Drug Design because it affects the accuracy and efficiency of molecular property prediction and other molecular modeling relevant tasks. However, previous molecular representation learning studies often suffer from limitations, such as over-reliance on a single molecular representation, failure to fully capture both local and global information in molecular structure, and ineffective integration of multiscale features from different molecular representations. These limitations restrict the complete and accurate representation of molecular structure and properties, ultimately impacting the accuracy of predicting molecular properties. To this end, we propose a novel multi-view molecular representation learning method called MvMRL, which can incorporate feature information from multiple molecular representations and capture both local and global information from different views well, thus improving molecular property prediction. Specifically, MvMRL consists of four parts: a multiscale CNN-SE Simplified Molecular Input Line Entry System (SMILES) learning component and a multiscale Graph Neural Network encoder to extract local feature information and global feature information from the SMILES view and the molecular graph view, respectively; a Multi-Layer Perceptron network to capture complex non-linear relationship features from the molecular fingerprint view; and a dual cross-attention component to fuse feature information on the multi-views deeply for predicting molecular properties. We evaluate the performance of MvMRL on 11 benchmark datasets, and experimental results show that MvMRL outperforms state-of-the-art methods, indicating its rationality and effectiveness in molecular property prediction. The source code of MvMRL was released in https://github.com/jedison-github/MvMRL.


Assuntos
Redes Neurais de Computação , Algoritmos , Aprendizado de Máquina , Modelos Moleculares , Desenho de Fármacos , Software , Estrutura Molecular , Inteligência Artificial
3.
Brief Bioinform ; 25(2)2024 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-38517692

RESUMO

Graph learning models have received increasing attention in the computational analysis of single-cell RNA sequencing (scRNA-seq) data. Compared with conventional deep neural networks, graph neural networks and language models have exhibited superior performance by extracting graph-structured data from raw gene count matrices. Established deep neural network-based clustering approaches generally focus on temporal expression patterns while ignoring inherent interactions at gene-level as well as cell-level, which could be regarded as spatial dynamics in single-cell data. Both gene-gene and cell-cell interactions are able to boost the performance of cell type detection, under the framework of multi-view modeling. In this study, spatiotemporal embedding and cell graphs are extracted to capture spatial dynamics at the molecular level. In order to enhance the accuracy of cell type detection, this study proposes the scHybridBERT architecture to conduct multi-view modeling of scRNA-seq data using extracted spatiotemporal patterns. In this scHybridBERT method, graph learning models are employed to deal with cell graphs and the Performer model employs spatiotemporal embeddings. Experimental outcomes about benchmark scRNA-seq datasets indicate that the proposed scHybridBERT method is able to enhance the accuracy of single-cell clustering tasks by integrating spatiotemporal embeddings and cell graphs.


Assuntos
Benchmarking , Regulação da Expressão Gênica , Comunicação Celular , Análise por Conglomerados , Aprendizagem
4.
Brief Bioinform ; 25(3)2024 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-38605642

RESUMO

MicroRNAs (miRNAs) synergize with various biomolecules in human cells resulting in diverse functions in regulating a wide range of biological processes. Predicting potential disease-associated miRNAs as valuable biomarkers contributes to the treatment of human diseases. However, few previous methods take a holistic perspective and only concentrate on isolated miRNA and disease objects, thereby ignoring that human cells are responsible for multiple relationships. In this work, we first constructed a multi-view graph based on the relationships between miRNAs and various biomolecules, and then utilized graph attention neural network to learn the graph topology features of miRNAs and diseases for each view. Next, we added an attention mechanism again, and developed a multi-scale feature fusion module, aiming to determine the optimal fusion results for the multi-view topology features of miRNAs and diseases. In addition, the prior attribute knowledge of miRNAs and diseases was simultaneously added to achieve better prediction results and solve the cold start problem. Finally, the learned miRNA and disease representations were then concatenated and fed into a multi-layer perceptron for end-to-end training and predicting potential miRNA-disease associations. To assess the efficacy of our model (called MUSCLE), we performed 5- and 10-fold cross-validation (CV), which got average the Area under ROC curves of 0.966${\pm }$0.0102 and 0.973${\pm }$0.0135, respectively, outperforming most current state-of-the-art models. We then examined the impact of crucial parameters on prediction performance and performed ablation experiments on the feature combination and model architecture. Furthermore, the case studies about colon cancer, lung cancer and breast cancer also fully demonstrate the good inductive capability of MUSCLE. Our data and code are free available at a public GitHub repository: https://github.com/zht-code/MUSCLE.git.


Assuntos
Neoplasias do Colo , Neoplasias Pulmonares , MicroRNAs , Humanos , Músculos , Aprendizagem , MicroRNAs/genética , Algoritmos , Biologia Computacional
5.
Brief Bioinform ; 25(4)2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-38801701

RESUMO

Spatially resolved transcriptomics data are being used in a revolutionary way to decipher the spatial pattern of gene expression and the spatial architecture of cell types. Much work has been done to exploit the genomic spatial architectures of cells. Such work is based on the common assumption that gene expression profiles of spatially adjacent spots are more similar than those of more distant spots. However, related work might not consider the nonlocal spatial co-expression dependency, which can better characterize the tissue architectures. Therefore, we propose MuCoST, a Multi-view graph Contrastive learning framework for deciphering complex Spatially resolved Transcriptomic architectures with dual scale structural dependency. To achieve this, we employ spot dependency augmentation by fusing gene expression correlation and spatial location proximity, thereby enabling MuCoST to model both nonlocal spatial co-expression dependency and spatially adjacent dependency. We benchmark MuCoST on four datasets, and we compare it with other state-of-the-art spatial domain identification methods. We demonstrate that MuCoST achieves the highest accuracy on spatial domain identification from various datasets. In particular, MuCoST accurately deciphers subtle biological textures and elaborates the variation of spatially functional patterns.


Assuntos
Perfilação da Expressão Gênica , Transcriptoma , Perfilação da Expressão Gênica/métodos , Humanos , Algoritmos , Aprendizado de Máquina , Biologia Computacional/métodos
6.
Brief Bioinform ; 25(4)2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-38975896

RESUMO

Mechanisms of protein-DNA interactions are involved in a wide range of biological activities and processes. Accurately identifying binding sites between proteins and DNA is crucial for analyzing genetic material, exploring protein functions, and designing novel drugs. In recent years, several computational methods have been proposed as alternatives to time-consuming and expensive traditional experiments. However, accurately predicting protein-DNA binding sites still remains a challenge. Existing computational methods often rely on handcrafted features and a single-model architecture, leaving room for improvement. We propose a novel computational method, called EGPDI, based on multi-view graph embedding fusion. This approach involves the integration of Equivariant Graph Neural Networks (EGNN) and Graph Convolutional Networks II (GCNII), independently configured to profoundly mine the global and local node embedding representations. An advanced gated multi-head attention mechanism is subsequently employed to capture the attention weights of the dual embedding representations, thereby facilitating the integration of node features. Besides, extra node features from protein language models are introduced to provide more structural information. To our knowledge, this is the first time that multi-view graph embedding fusion has been applied to the task of protein-DNA binding site prediction. The results of five-fold cross-validation and independent testing demonstrate that EGPDI outperforms state-of-the-art methods. Further comparative experiments and case studies also verify the superiority and generalization ability of EGPDI.


Assuntos
Biologia Computacional , Proteínas de Ligação a DNA , DNA , Redes Neurais de Computação , Sítios de Ligação , DNA/metabolismo , DNA/química , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/química , Biologia Computacional/métodos , Algoritmos , Ligação Proteica
7.
Proc Natl Acad Sci U S A ; 120(46): e2309240120, 2023 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-37943836

RESUMO

A bottleneck in high-throughput nanomaterials discovery is the pace at which new materials can be structurally characterized. Although current machine learning (ML) methods show promise for the automated processing of electron diffraction patterns (DPs), they fail in high-throughput experiments where DPs are collected from crystals with random orientations. Inspired by the human decision-making process, a framework for automated crystal system classification from DPs with arbitrary orientations was developed. A convolutional neural network was trained using evidential deep learning, and the predictive uncertainties were quantified and leveraged to fuse multiview predictions. Using vector map representations of DPs, the framework achieves a testing accuracy of 0.94 in the examples considered, is robust to noise, and retains remarkable accuracy using experimental data. This work highlights the ability of ML to be used to accelerate experimental high-throughput materials data analytics.

8.
Proc Natl Acad Sci U S A ; 120(32): e2303647120, 2023 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-37523521

RESUMO

Multimodal single-cell technologies profile multiple modalities for each cell simultaneously, enabling a more thorough characterization of cell populations. Existing dimension-reduction methods for multimodal data capture the "union of information," producing a lower-dimensional embedding that combines the information across modalities. While these tools are useful, we focus on a fundamentally different task of separating and quantifying the information among cells that is shared between the two modalities as well as unique to only one modality. Hence, we develop Tilted Canonical Correlation Analysis (Tilted-CCA), a method that decomposes a paired multimodal dataset into three lower-dimensional embeddings-one embedding captures the "intersection of information," representing the geometric relations among the cells that is common to both modalities, while the remaining two embeddings capture the "distinct information for a modality," representing the modality-specific geometric relations. We analyze single-cell multimodal datasets sequencing RNA along surface antibodies (i.e., CITE-seq) as well as RNA alongside chromatin accessibility (i.e., 10x) for blood cells and developing neurons via Tilted-CCA. These analyses show that Tilted-CCA enables meaningful visualization and quantification of the cross-modal information. Finally, Tilted-CCA's framework allows us to perform two specific downstream analyses. First, for single-cell datasets that simultaneously profile transcriptome and surface antibody markers, we show that Tilted-CCA helps design the target antibody panel to complement the transcriptome best. Second, for developmental single-cell datasets that simultaneously profile transcriptome and chromatin accessibility, we show that Tilted-CCA helps identify development-informative genes and distinguish between transient versus terminal cell types.


Assuntos
Algoritmos , Análise de Correlação Canônica , Transcriptoma , Análise de Célula Única/métodos
9.
Development ; 149(4)2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-35072204

RESUMO

Understanding how development is coordinated in multiple tissues and gives rise to fully functional organs or whole organisms necessitates microscopy tools. Over the last decade numerous advances have been made in live-imaging, enabling high resolution imaging of whole organisms at cellular resolution. Yet, these advances mainly rely on mounting the specimen in agarose or aqueous solutions, precluding imaging of organisms whose oxygen uptake depends on ventilation. Here, we implemented a multi-view multi-scale microscopy strategy based on confocal spinning disk microscopy, called Multi-View confocal microScopy (MuViScopy). MuViScopy enables live-imaging of multiple organs with cellular resolution using sample rotation and confocal imaging without the need of sample embedding. We illustrate the capacity of MuViScopy by live-imaging Drosophila melanogaster pupal development throughout metamorphosis, highlighting how internal organs are formed and multiple organ development is coordinated. We foresee that MuViScopy will open the path to better understand developmental processes at the whole organism scale in living systems that require gas exchange by ventilation.


Assuntos
Drosophila melanogaster/anatomia & histologia , Microscopia Confocal/métodos , Animais , Metamorfose Biológica , Pupa/anatomia & histologia , Imagem com Lapso de Tempo
10.
Brief Bioinform ; 24(2)2023 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-36790856

RESUMO

Potential miRNA-disease associations (MDA) play an important role in the discovery of complex human disease etiology. Therefore, MDA prediction is an attractive research topic in the field of biomedical machine learning. Recently, several models have been proposed for this task, but their performance limited by over-reliance on relevant network information with noisy graph structure connections. However, the application of self-supervised graph structure learning to MDA tasks remains unexplored. Our study is the first to use multi-view self-supervised contrastive learning (MSGCL) for MDA prediction. Specifically, we generated a learner view without association labels of miRNAs and diseases as input, and utilized the known association network to generate an anchor view that provides guiding signals for the learner view. The graph structure was optimized by designing a contrastive loss to maximize the consistency between the anchor and learner views. Our model is similar to a pre-trained model that continuously optimizes upstream tasks for high-quality association graph topology, thereby enhancing the latent representation of association predictions. The experimental results show that our proposed method outperforms state-of-the-art methods by 2.79$\%$ and 3.20$\%$ in area under the receiver operating characteristic curve (AUC) and area under the precision/recall curve (AUPR), respectively.


Assuntos
Aprendizado de Máquina , MicroRNAs , Humanos , Área Sob a Curva , MicroRNAs/genética , Curva ROC
11.
Brief Bioinform ; 24(2)2023 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-36847701

RESUMO

Emerging studies have shown that circular RNAs (circRNAs) are involved in a variety of biological processes and play a key role in disease diagnosing, treating and inferring. Although many methods, including traditional machine learning and deep learning, have been developed to predict associations between circRNAs and diseases, the biological function of circRNAs has not been fully exploited. Some methods have explored disease-related circRNAs based on different views, but how to efficiently use the multi-view data about circRNA is still not well studied. Therefore, we propose a computational model to predict potential circRNA-disease associations based on collaborative learning with circRNA multi-view functional annotations. First, we extract circRNA multi-view functional annotations and build circRNA association networks, respectively, to enable effective network fusion. Then, a collaborative deep learning framework for multi-view information is designed to get circRNA multi-source information features, which can make full use of the internal relationship among circRNA multi-view information. We build a network consisting of circRNAs and diseases by their functional similarity and extract the consistency description information of circRNAs and diseases. Last, we predict potential associations between circRNAs and diseases based on graph auto encoder. Our computational model has better performance in predicting candidate disease-related circRNAs than the existing ones. Furthermore, it shows the high practicability of the method that we use several common diseases as case studies to find some unknown circRNAs related to them. The experiments show that CLCDA can efficiently predict disease-related circRNAs and are helpful for the diagnosis and treatment of human disease.


Assuntos
Aprendizado Profundo , Práticas Interdisciplinares , Humanos , RNA Circular/genética , Aprendizado de Máquina , Biologia Computacional/métodos
12.
Brief Bioinform ; 24(5)2023 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-37539822

RESUMO

Cancer heterogeneity has posed great challenges in exploring precise therapeutic strategies for cancer treatment. The identification of cancer subtypes aims to detect patients with distinct molecular profiles and thus could provide new clues on effective clinical therapies. While great efforts have been made, it remains challenging to develop powerful computational methods that can efficiently integrate multi-omics datasets for the task. In this paper, we propose a novel self-supervised learning model called Deep Multi-view Contrastive Learning (DMCL) for cancer subtype identification. Specifically, by incorporating the reconstruction loss, contrastive loss and clustering loss into a unified framework, our model simultaneously encodes the sample discriminative information into the extracted feature representations and well preserves the sample cluster structures in the embedded space. Moreover, DMCL is an end-to-end framework where the cancer subtypes could be directly obtained from the model outputs. We compare DMCL with eight alternatives ranging from classic cancer subtype identification methods to recently developed state-of-the-art systems on 10 widely used cancer multi-omics datasets as well as an integrated dataset, and the experimental results validate the superior performance of our method. We further conduct a case study on liver cancer and the analysis results indicate that different subtypes might have different responses to the selected chemotherapeutic drugs.


Assuntos
Neoplasias Hepáticas , Humanos , Neoplasias Hepáticas/genética , Análise por Conglomerados , Multiômica
13.
Brief Bioinform ; 24(4)2023 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-37427977

RESUMO

Studies have shown that the mechanism of action of many drugs is related to miRNA. In-depth research on the relationship between miRNA and drugs can provide theoretical foundations and practical approaches for various areas, such as drug target discovery, drug repositioning and biomarker research. Traditional biological experiments to test miRNA-drug susceptibility are costly and time-consuming. Thus, sequence- or topology-based deep learning methods are recognized in this field for their efficiency and accuracy. However, these methods have limitations in dealing with sparse topologies and higher-order information of miRNA (drug) feature. In this work, we propose GCFMCL, a model for multi-view contrastive learning based on graph collaborative filtering. To the best of our knowledge, this is the first attempt that incorporates contrastive learning strategy into the graph collaborative filtering framework to predict the sensitivity relationships between miRNA and drug. The proposed multi-view contrastive learning method is divided into topological contrastive objective and feature contrastive objective: (1) For the homogeneous neighbors of the topological graph, we propose a novel topological contrastive learning method via constructing the contrastive target through the topological neighborhood information of nodes. (2) The proposed model obtains feature contrastive targets from high-order feature information according to the correlation of node features, and mines potential neighborhood relationships in the feature space. The proposed multi-view comparative learning effectively alleviates the impact of heterogeneous node noise and graph data sparsity in graph collaborative filtering, and significantly enhances the performance of the model. Our study employs a dataset derived from the NoncoRNA and ncDR databases, encompassing 2049 experimentally validated miRNA-drug sensitivity associations. Five-fold cross-validation shows that the Area Under the Curve (AUC), Area Under the Precision-Recall Curve (AUPR) and F1-score (F1) of GCFMCL reach 95.28%, 95.66% and 89.77%, which outperforms the state-of-the-art (SOTA) method by the margin of 2.73%, 3.42% and 4.96%, respectively. Our code and data can be accessed at https://github.com/kkkayle/GCFMCL.


Assuntos
Sistemas de Liberação de Medicamentos , MicroRNAs , Área Sob a Curva , Bases de Dados Factuais , Descoberta de Drogas , MicroRNAs/genética
14.
Brief Bioinform ; 25(1)2023 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-38058188

RESUMO

Biclustering is a useful method for simultaneously grouping samples and features and has been applied across various biomedical data types. However, most existing biclustering methods lack the ability to integratively analyze multi-modal data such as multi-omics data such as genome, transcriptome and epigenome. Moreover, the potential of leveraging biological knowledge represented by graphs, which has been demonstrated to be beneficial in various statistical tasks such as variable selection and prediction, remains largely untapped in the context of biclustering. To address both, we propose a novel Bayesian biclustering method called Bayesian graph-guided biclustering (BGB). Specifically, we introduce a new hierarchical sparsity-inducing prior to effectively incorporate biological graph information and establish a unified framework to model multi-view data. We develop an efficient Markov chain Monte Carlo algorithm to conduct posterior sampling and inference. Extensive simulations and real data analysis show that BGB outperforms other popular biclustering methods. Notably, BGB is robust in terms of utilizing biological knowledge and has the capability to reveal biologically meaningful information from heterogeneous multi-modal data.


Assuntos
Algoritmos , Multiômica , Teorema de Bayes , Análise por Conglomerados , Transcriptoma
15.
Brief Bioinform ; 24(1)2023 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-36445207

RESUMO

Driven by multi-omics data, some multi-view clustering algorithms have been successfully applied to cancer subtypes prediction, aiming to identify subtypes with biometric differences in the same cancer, thereby improving the clinical prognosis of patients and designing personalized treatment plan. Due to the fact that the number of patients in omics data is much smaller than the number of genes, multi-view spectral clustering based on similarity learning has been widely developed. However, these algorithms still suffer some problems, such as over-reliance on the quality of pre-defined similarity matrices for clustering results, inability to reasonably handle noise and redundant information in high-dimensional omics data, ignoring complementary information between omics data, etc. This paper proposes multi-view spectral clustering with latent representation learning (MSCLRL) method to alleviate the above problems. First, MSCLRL generates a corresponding low-dimensional latent representation for each omics data, which can effectively retain the unique information of each omics and improve the robustness and accuracy of the similarity matrix. Second, the obtained latent representations are assigned appropriate weights by MSCLRL, and global similarity learning is performed to generate an integrated similarity matrix. Third, the integrated similarity matrix is used to feed back and update the low-dimensional representation of each omics. Finally, the final integrated similarity matrix is used for clustering. In 10 benchmark multi-omics datasets and 2 separate cancer case studies, the experiments confirmed that the proposed method obtained statistically and biologically meaningful cancer subtypes.


Assuntos
Multiômica , Neoplasias , Humanos , Algoritmos , Neoplasias/genética , Análise por Conglomerados
16.
Brief Bioinform ; 24(1)2023 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-36527428

RESUMO

Understanding the mechanisms of candidate drugs play an important role in drug discovery. The activating/inhibiting mechanisms between drugs and targets are major types of mechanisms of drugs. Owing to the complexity of drug-target (DT) mechanisms and data scarcity, modelling this problem based on deep learning methods to accurately predict DT activating/inhibiting mechanisms remains a considerable challenge. Here, by considering network pharmacology, we propose a multi-view deep learning model, DrugAI, which combines four modules, i.e. a graph neural network for drugs, a convolutional neural network for targets, a network embedding module for drugs and targets and a deep neural network for predicting activating/inhibiting mechanisms between drugs and targets. Computational experiments show that DrugAI performs better than state-of-the-art methods and has good robustness and generalization. To demonstrate the reliability of the predictive results of DrugAI, bioassay experiments are conducted to validate two drugs (notopterol and alpha-asarone) predicted to activate TRPV1. Moreover, external validation bears out 61 pairs of mechanism relationships between natural products and their targets predicted by DrugAI based on independent literatures and PubChem bioassays. DrugAI, for the first time, provides a powerful multi-view deep learning framework for robust prediction of DT activating/inhibiting mechanisms.


Assuntos
Aprendizado Profundo , Algoritmos , Reprodutibilidade dos Testes , Redes Neurais de Computação , Descoberta de Drogas
17.
Brief Bioinform ; 24(1)2023 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-36511221

RESUMO

Cumulative studies have shown that many long non-coding RNAs (lncRNAs) are crucial in a number of diseases. Predicting potential lncRNA-disease associations (LDAs) can facilitate disease prevention, diagnosis and treatment. Therefore, it is vital to develop practical computational methods for LDA prediction. In this study, we propose a novel predictor named capsule network (CapsNet)-LDA for LDA prediction. CapsNet-LDA first uses a stacked autoencoder for acquiring the informative low-dimensional representations of the lncRNA-disease pairs under multiple views, then the attention mechanism is leveraged to implement an adaptive allocation of importance weights to them, and they are subsequently processed using a CapsNet-based architecture for predicting LDAs. Different from the conventional convolutional neural networks (CNNs) that have some restrictions with the usage of scalar neurons and pooling operations. the CapsNets use vector neurons instead of scalar neurons that have better robustness for the complex combination of features and they use dynamic routing processes for updating parameters. CapsNet-LDA is superior to other five state-of-the-art models on four benchmark datasets, four perturbed datasets and an independent test set in the comparison experiments, demonstrating that CapsNet-LDA has excellent performance and robustness against perturbation, as well as good generalization ability. The ablation studies verify the effectiveness of some modules of CapsNet-LDA. Moreover, the ability of multi-view data to improve performance is proven. Case studies further indicate that CapsNet-LDA can accurately predict novel LDAs for specific diseases.


Assuntos
RNA Longo não Codificante , RNA Longo não Codificante/genética , Redes Neurais de Computação
18.
Brief Bioinform ; 24(2)2023 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-36907654

RESUMO

In recent years, many experiments have proved that microRNAs (miRNAs) play a variety of important regulatory roles in cells, and their abnormal expression can lead to the emergence of specific diseases. Therefore, it is greatly valuable to do research on the association between miRNAs and diseases, which can effectively help prevent and treat miRNA-related diseases. At present, effective computational methods still need to be developed to better identify potential miRNA-disease associations. Inspired by graph convolutional networks, in this study, we propose a new method based on Attention aware Multi-view similarity networks and Hypergraph learning for MiRNA-Disease Associations identification (AMHMDA). First, we construct multiple similarity networks for miRNAs and diseases, and exploit the graph convolutional networks fusion attention mechanism to obtain the important information from different views. Then, in order to obtain high-quality links and richer nodes information, we introduce a kind of virtual nodes called hypernodes to construct heterogeneous hypergraph of miRNAs and diseases. Finally, we employ the attention mechanism to fuse the outputs of graph convolutional networks, predicting miRNA-disease associations. To verify the effectiveness of this method, we carry out a series of experiments on the Human MicroRNA Disease Database (HMDD v3.2). The experimental results show that AMHMDA has good performance compared with other methods. In addition, the case study results also fully demonstrate the reliable predictive performance of AMHMDA.


Assuntos
MicroRNAs , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Predisposição Genética para Doença , Algoritmos , Biologia Computacional/métodos , Bases de Dados Genéticas
19.
Brief Bioinform ; 24(5)2023 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-37466210

RESUMO

MOTIVATION: Recent advances in spatial transcriptomics technologies have enabled gene expression profiles while preserving spatial context. Accurately identifying spatial domains is crucial for downstream analysis and it requires the effective integration of gene expression profiles and spatial information. While increasingly computational methods have been developed for spatial domain detection, most of them cannot adaptively learn the complex relationship between gene expression and spatial information, leading to sub-optimal performance. RESULTS: To overcome these challenges, we propose a novel deep learning method named Spatial-MGCN for identifying spatial domains, which is a Multi-view Graph Convolutional Network (GCN) with attention mechanism. We first construct two neighbor graphs using gene expression profiles and spatial information, respectively. Then, a multi-view GCN encoder is designed to extract unique embeddings from both the feature and spatial graphs, as well as their shared embeddings by combining both graphs. Finally, a zero-inflated negative binomial decoder is used to reconstruct the original expression matrix by capturing the global probability distribution of gene expression profiles. Moreover, Spatial-MGCN incorporates a spatial regularization constraint into the features learning to preserve spatial neighbor information in an end-to-end manner. The experimental results show that Spatial-MGCN outperforms state-of-the-art methods consistently in several tasks, including spatial clustering and trajectory inference.


Assuntos
Oftalmopatias Hereditárias , Doenças Genéticas Ligadas ao Cromossomo X , Humanos , Perfilação da Expressão Gênica
20.
Brief Bioinform ; 24(6)2023 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-37742052

RESUMO

Drug-drug interaction (DDI) prediction can discover potential risks of drug combinations in advance by detecting drug pairs that are likely to interact with each other, sparking an increasing demand for computational methods of DDI prediction. However, existing computational DDI methods mostly rely on the single-view paradigm, failing to handle the complex features and intricate patterns of DDIs due to the limited expressiveness of the single view. To this end, we propose a Hierarchical Triple-view Contrastive Learning framework for Drug-Drug Interaction prediction (HTCL-DDI), leveraging the molecular, structural and semantic views to model the complicated information involved in DDI prediction. To aggregate the intra-molecular compositional and structural information, we present a dual attention-aware network in the molecular view. Based on the molecular view, to further capture inter-molecular information, we utilize the one-hop neighboring information and high-order semantic relations in the structural view and semantic view, respectively. Then, we introduce contrastive learning to enhance drug representation learning from multifaceted aspects and improve the robustness of HTCL-DDI. Finally, we conduct extensive experiments on three real-world datasets. All the experimental results show the significant improvement of HTCL-DDI over the state-of-the-art methods, which also demonstrates that HTCL-DDI opens new avenues for ensuring medication safety and identifying synergistic drug combinations.


Assuntos
Aprendizado Profundo , Interações Medicamentosas , Semântica
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa