Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Brief Bioinform ; 25(2)2024 Jan 22.
Article in English | MEDLINE | ID: mdl-38517693

ABSTRACT

Numerous investigations increasingly indicate the significance of microRNA (miRNA) in human diseases. Hence, unearthing associations between miRNA and diseases can contribute to precise diagnosis and efficacious remediation of medical conditions. The detection of miRNA-disease linkages via computational techniques utilizing biological information has emerged as a cost-effective and highly efficient approach. Here, we introduced a computational framework named ReHoGCNES, designed for prospective miRNA-disease association prediction (ReHoGCNES-MDA). This method constructs homogenous graph convolutional network with regular graph structure (ReHoGCN) encompassing disease similarity network, miRNA similarity network and known MDA network and then was tested on four experimental tasks. A random edge sampler strategy was utilized to expedite processes and diminish training complexity. Experimental results demonstrate that the proposed ReHoGCNES-MDA method outperforms both homogenous graph convolutional network and heterogeneous graph convolutional network with non-regular graph structure in all four tasks, which implicitly reveals steadily degree distribution of a graph does play an important role in enhancement of model performance. Besides, ReHoGCNES-MDA is superior to several machine learning algorithms and state-of-the-art methods on the MDA prediction. Furthermore, three case studies were conducted to further demonstrate the predictive ability of ReHoGCNES. Consequently, 93.3% (breast neoplasms), 90% (prostate neoplasms) and 93.3% (prostate neoplasms) of the top 30 forecasted miRNAs were validated by public databases. Hence, ReHoGCNES-MDA might serve as a dependable and beneficial model for predicting possible MDAs.


Subject(s)
MicroRNAs , Prostatic Neoplasms , Humans , Male , Algorithms , Computational Biology/methods , Databases, Genetic , MicroRNAs/genetics , Prospective Studies , Prostatic Neoplasms/genetics , Female
2.
Brief Bioinform ; 25(5)2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39175132

ABSTRACT

Numerous studies have demonstrated that microRNAs (miRNAs) are critically important for the prediction, diagnosis, and characterization of diseases. However, identifying miRNA-disease associations through traditional biological experiments is both costly and time-consuming. To further explore these associations, we proposed a model based on hybrid high-order moments combined with element-level attention mechanisms (HHOMR). This model innovatively fused hybrid higher-order statistical information along with structural and community information. Specifically, we first constructed a heterogeneous graph based on existing associations between miRNAs and diseases. HHOMR employs a structural fusion layer to capture structure-level embeddings and leverages a hybrid high-order moments encoder layer to enhance features. Element-level attention mechanisms are then used to adaptively integrate the features of these hybrid moments. Finally, a multi-layer perceptron is utilized to calculate the association scores between miRNAs and diseases. Through five-fold cross-validation on HMDD v2.0, we achieved a mean AUC of 93.28%. Compared with four state-of-the-art models, HHOMR exhibited superior performance. Additionally, case studies on three diseases-esophageal neoplasms, lymphoma, and prostate neoplasms-were conducted. Among the top 50 miRNAs with high disease association scores, 46, 47, and 45 associated with these diseases were confirmed by the dbDEMC and miR2Disease databases, respectively. Our results demonstrate that HHOMR not only outperforms existing models but also shows significant potential in predicting miRNA-disease associations.


Subject(s)
MicroRNAs , MicroRNAs/genetics , Humans , Computational Biology/methods , Genetic Predisposition to Disease , Algorithms , Prostatic Neoplasms/genetics , Models, Genetic
3.
Brief Bioinform ; 25(3)2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38605642

ABSTRACT

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.


Subject(s)
Colonic Neoplasms , Lung Neoplasms , MicroRNAs , Humans , Muscles , Learning , MicroRNAs/genetics , Algorithms , Computational Biology
4.
Brief Bioinform ; 24(2)2023 03 19.
Article in English | MEDLINE | ID: mdl-36790856

ABSTRACT

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.


Subject(s)
Machine Learning , MicroRNAs , Humans , Area Under Curve , MicroRNAs/genetics , ROC Curve
5.
Brief Bioinform ; 24(1)2023 01 19.
Article in English | MEDLINE | ID: mdl-36526276

ABSTRACT

Increasing studies have proved that microRNAs (miRNAs) are critical biomarkers in the development of human complex diseases. Identifying disease-related miRNAs is beneficial to disease prevention, diagnosis and remedy. Based on the assumption that similar miRNAs tend to associate with similar diseases, various computational methods have been developed to predict novel miRNA-disease associations (MDAs). However, selecting proper features for similarity calculation is a challenging task because of data deficiencies in biomedical science. In this study, we propose a deep learning-based computational method named MAGCN to predict potential MDAs without using any similarity measurements. Our method predicts novel MDAs based on known lncRNA-miRNA interactions via graph convolution networks with multichannel attention mechanism and convolutional neural network combiner. Extensive experiments show that the average area under the receiver operating characteristic values obtained by our method under 2-fold, 5-fold and 10-fold cross-validations are 0.8994, 0.9032 and 0.9044, respectively. When compared with five state-of-the-art methods, MAGCN shows improvement in terms of prediction accuracy. In addition, we conduct case studies on three diseases to discover their related miRNAs, and find that all the top 50 predictions for all the three diseases have been supported by established databases. The comprehensive results demonstrate that our method is a reliable tool in detecting new disease-related miRNAs.


Subject(s)
MicroRNAs , RNA, Long Noncoding , Humans , Algorithms , Computational Biology/methods , Databases, Genetic , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Deep Learning
6.
Brief Bioinform ; 24(4)2023 07 20.
Article in English | MEDLINE | ID: mdl-37287133

ABSTRACT

MicroRNAs (miRNAs) are a family of non-coding RNA molecules with vital roles in regulating gene expression. Although researchers have recognized the importance of miRNAs in the development of human diseases, it is very resource-consuming to use experimental methods for identifying which dysregulated miRNA is associated with a specific disease. To reduce the cost of human effort, a growing body of studies has leveraged computational methods for predicting the potential miRNA-disease associations. However, the extant computational methods usually ignore the crucial mediating role of genes and suffer from the data sparsity problem. To address this limitation, we introduce the multi-task learning technique and develop a new model called MTLMDA (Multi-Task Learning model for predicting potential MicroRNA-Disease Associations). Different from existing models that only learn from the miRNA-disease network, our MTLMDA model exploits both miRNA-disease and gene-disease networks for improving the identification of miRNA-disease associations. To evaluate model performance, we compare our model with competitive baselines on a real-world dataset of experimentally supported miRNA-disease associations. Empirical results show that our model performs best using various performance metrics. We also examine the effectiveness of model components via ablation study and further showcase the predictive power of our model for six types of common cancers. The data and source code are available from https://github.com/qwslle/MTLMDA.


Subject(s)
MicroRNAs , Neoplasms , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Algorithms , Computational Biology/methods , Neoplasms/genetics , Software
7.
Methods ; 221: 73-81, 2024 01.
Article in English | MEDLINE | ID: mdl-38123109

ABSTRACT

Research indicates that miRNAs present in herbal medicines are crucial for identifying disease markers, advancing gene therapy, facilitating drug delivery, and so on. These miRNAs maintain stability in the extracellular environment, making them viable tools for disease diagnosis. They can withstand the digestive processes in the gastrointestinal tract, positioning them as potential carriers for specific oral drug delivery. By engineering plants to generate effective, non-toxic miRNA interference sequences, it's possible to broaden their applicability, including the treatment of diseases such as hepatitis C. Consequently, delving into the miRNA-disease associations (MDAs) within herbal medicines holds immense promise for diagnosing and addressing miRNA-related diseases. In our research, we propose the SGAE-MDA model, which harnesses the strengths of a graph autoencoder (GAE) combined with a semi-supervised approach to uncover potential MDAs in herbal medicines more effectively. Leveraging the GAE framework, the SGAE-MDA model exactly integrates the inherent feature vectors of miRNAs and disease nodes with the regulatory data in the miRNA-disease network. Additionally, the proposed semi-supervised learning approach randomly hides the partial structure of the miRNA-disease network, subsequently reconstructing them within the GAE framework. This technique effectively minimizes network noise interference. Through comparison against other leading deep learning models, the results consistently highlighted the superior performance of the proposed SGAE-MDA model. Our code and dataset can be available at: https://github.com/22n9n23/SGAE-MDA.


Subject(s)
MicroRNAs , MicroRNAs/genetics , Algorithms , Computational Biology/methods , Supervised Machine Learning , Plant Extracts
8.
Methods ; 229: 71-81, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38909974

ABSTRACT

Identifying miRNA-disease associations (MDAs) is crucial for improving the diagnosis and treatment of various diseases. However, biological experiments can be time-consuming and expensive. To overcome these challenges, computational approaches have been developed, with Graph Convolutional Network (GCN) showing promising results in MDA prediction. The success of GCN-based methods relies on learning a meaningful spatial operator to extract effective node feature representations. To enhance the inference of MDAs, we propose a novel method called PGCNMDA, which employs graph convolutional networks with a learning graph spatial operator from paths. This approach enables the generation of meaningful spatial convolutions from paths in GCN, leading to improved prediction performance. On HMDD v2.0, PGCNMDA obtains a mean AUC of 0.9229 and an AUPRC of 0.9206 under 5-fold cross-validation (5-CV), and a mean AUC of 0.9235 and an AUPRC of 0.9212 under 10-fold cross-validation (10-CV), respectively. Additionally, the AUC of PGCNMDA also reaches 0.9238 under global leave-one-out cross-validation (GLOOCV). On HMDD v3.2, PGCNMDA obtains a mean AUC of 0.9413 and an AUPRC of 0.9417 under 5-CV, and a mean AUC of 0.9419 and an AUPRC of 0.9425 under 10-CV, respectively. Furthermore, the AUC of PGCNMDA also reaches 0.9415 under GLOOCV. The results show that PGCNMDA is superior to other compared methods. In addition, the case studies on pancreatic neoplasms, thyroid neoplasms and leukemia show that 50, 50 and 48 of the top 50 predicted miRNAs linked to these diseases are confirmed, respectively. It further validates the effectiveness and feasibility of PGCNMDA in practical applications.


Subject(s)
MicroRNAs , Humans , MicroRNAs/genetics , Computational Biology/methods , Neural Networks, Computer , Genetic Predisposition to Disease , Area Under Curve , Pancreatic Neoplasms/genetics , Algorithms
9.
BMC Bioinformatics ; 25(1): 139, 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38553698

ABSTRACT

BACKGROUND: MicroRNA (miRNA) has been shown to play a key role in the occurrence and progression of diseases, making uncovering miRNA-disease associations vital for disease prevention and therapy. However, traditional laboratory methods for detecting these associations are slow, strenuous, expensive, and uncertain. Although numerous advanced algorithms have emerged, it is still a challenge to develop more effective methods to explore underlying miRNA-disease associations. RESULTS: In the study, we designed a novel approach on the basis of deep autoencoder and combined feature representation (DAE-CFR) to predict possible miRNA-disease associations. We began by creating integrated similarity matrices of miRNAs and diseases, performing a logistic function transformation, balancing positive and negative samples with k-means clustering, and constructing training samples. Then, deep autoencoder was used to extract low-dimensional feature from two kinds of feature representations for miRNAs and diseases, namely, original association information-based and similarity information-based. Next, we combined the resulting features for each miRNA-disease pair and used a logistic regression (LR) classifier to infer all unknown miRNA-disease interactions. Under five and tenfold cross-validation (CV) frameworks, DAE-CFR not only outperformed six popular algorithms and nine classifiers, but also demonstrated superior performance on an additional dataset. Furthermore, case studies on three diseases (myocardial infarction, hypertension and stroke) confirmed the validity of DAE-CFR in practice. CONCLUSIONS: DAE-CFR achieved outstanding performance in predicting miRNA-disease associations and can provide evidence to inform biological experiments and clinical therapy.


Subject(s)
MicroRNAs , Humans , MicroRNAs/genetics , Computational Biology/methods , Algorithms , Genetic Predisposition to Disease
10.
Brief Bioinform ; 23(1)2022 01 17.
Article in English | MEDLINE | ID: mdl-34875683

ABSTRACT

MiRNAs are a class of small non-coding RNA molecules that play an important role in many biological processes, and determining miRNA-disease associations can benefit drug development and clinical diagnosis. Although great efforts have been made to develop miRNA-disease association prediction methods, few attention has been paid to in-depth classification of miRNA-disease associations, e.g. up/down-regulation of miRNAs in diseases. In this paper, we regard known miRNA-disease associations as a signed bipartite network, which has miRNA nodes, disease nodes and two types of edges representing up/down-regulation of miRNAs in diseases, and propose a signed graph neural network method (SGNNMD) for predicting deregulation types of miRNA-disease associations. SGNNMD extracts subgraphs around miRNA-disease pairs from the signed bipartite network and learns structural features of subgraphs via a labeling algorithm and a neural network, and then combines them with biological features (i.e. miRNA-miRNA functional similarity and disease-disease semantic similarity) to build the prediction model. In the computational experiments, SGNNMD achieves highly competitive performance when compared with several baselines, including the signed graph link prediction methods, multi-relation prediction methods and one existing deregulation type prediction method. Moreover, SGNNMD has good inductive capability and can generalize to miRNAs/diseases unseen during the training.


Subject(s)
MicroRNAs , Neural Networks, Computer , Algorithms , Computational Biology/methods , Down-Regulation , MicroRNAs/genetics
11.
Brief Bioinform ; 23(5)2022 09 20.
Article in English | MEDLINE | ID: mdl-35849099

ABSTRACT

Increasing biomedical evidence has proved that the dysregulation of miRNAs is associated with human complex diseases. Identification of disease-related miRNAs is of great importance for disease prevention, diagnosis and remedy. To reduce the time and cost of biomedical experiments, there is a strong incentive to develop efficient computational methods to infer potential miRNA-disease associations. Although many computational approaches have been proposed to address this issue, the prediction accuracy needs to be further improved. In this study, we present a computational framework MKGAT to predict possible associations between miRNAs and diseases through graph attention networks (GATs) using dual Laplacian regularized least squares. We use GATs to learn embeddings of miRNAs and diseases on each layer from initial input features of known miRNA-disease associations, intra-miRNA similarities and intra-disease similarities. We then calculate kernel matrices of miRNAs and diseases based on Gaussian interaction profile (GIP) with the learned embeddings. We further fuse the kernel matrices of each layer and initial similarities with attention mechanism. Dual Laplacian regularized least squares are finally applied for new miRNA-disease association predictions with the fused miRNA and disease kernels. Compared with six state-of-the-art methods by 5-fold cross-validations, our method MKGAT receives the highest AUROC value of 0.9627 and AUPR value of 0.7372. We use MKGAT to predict related miRNAs for three cancers and discover that all the top 50 predicted results in the three diseases are confirmed by existing databases. The excellent performance indicates that MKGAT would be a useful computational tool for revealing disease-related miRNAs.


Subject(s)
MicroRNAs , Neoplasms , Algorithms , Computational Biology/methods , Databases, Factual , Humans , Least-Squares Analysis , MicroRNAs/genetics , Neoplasms/genetics
12.
Brief Bioinform ; 23(5)2022 09 20.
Article in English | MEDLINE | ID: mdl-36037084

ABSTRACT

Increasing evidence has suggested that microRNAs (miRNAs) are important biomarkers of various diseases. Numerous graph neural network (GNN) models have been proposed for predicting miRNA-disease associations. However, the existing GNN-based methods have over-smoothing issue-the learned feature embeddings of miRNA nodes and disease nodes are indistinguishable when stacking multiple GNN layers. This issue makes the performance of the methods sensitive to the number of layers, and significantly hurts the performance when more layers are employed. In this study, we resolve this issue by a novel self-feature-based graph autoencoder model, shortened as SFGAE. The key novelty of SFGAE is to construct miRNA-self embeddings and disease-self embeddings, and let them be independent of graph interactions between two types of nodes. The novel self-feature embeddings enrich the information of typical aggregated feature embeddings, which aggregate the information from direct neighbors and hence heavily rely on graph interactions. SFGAE adopts a graph encoder with attention mechanism to concatenate aggregated feature embeddings and self-feature embeddings, and adopts a bilinear decoder to predict links. Our experiments show that SFGAE achieves state-of-the-art performance. In particular, SFGAE improves the average AUC upon recent GAEMDA [1] on the benchmark datasets HMDD v2.0 and HMDD v3.2, and consistently performs better when less (e.g. 10%) training samples are used. Furthermore, SFGAE effectively overcomes the over-smoothing issue and performs stably well on deeper models (e.g. eight layers). Finally, we carry out case studies on three human diseases, colon neoplasms, esophageal neoplasms and kidney neoplasms, and perform a survival analysis using kidney neoplasm as an example. The results suggest that SFGAE is a reliable tool for predicting potential miRNA-disease associations.


Subject(s)
Colonic Neoplasms , MicroRNAs , Algorithms , Colonic Neoplasms/genetics , Computational Biology/methods , Humans , MicroRNAs/genetics , Neural Networks, Computer
13.
Brief Bioinform ; 23(6)2022 11 19.
Article in English | MEDLINE | ID: mdl-36168938

ABSTRACT

More and more evidence indicates that the dysregulations of microRNAs (miRNAs) lead to diseases through various kinds of underlying mechanisms. Identifying the multiple types of disease-related miRNAs plays an important role in studying the molecular mechanism of miRNAs in diseases. Moreover, compared with traditional biological experiments, computational models are time-saving and cost-minimized. However, most tensor-based computational models still face three main challenges: (i) easy to fall into bad local minima; (ii) preservation of high-order relations; (iii) false-negative samples. To this end, we propose a novel tensor completion framework integrating self-paced learning, hypergraph regularization and adaptive weight tensor into nonnegative tensor factorization, called SPLDHyperAWNTF, for the discovery of potential multiple types of miRNA-disease associations. We first combine self-paced learning with nonnegative tensor factorization to effectively alleviate the model from falling into bad local minima. Then, hypergraphs for miRNAs and diseases are constructed, and hypergraph regularization is used to preserve the high-order complex relations of these hypergraphs. Finally, we innovatively introduce adaptive weight tensor, which can effectively alleviate the impact of false-negative samples on the prediction performance. The average results of 5-fold and 10-fold cross-validation on four datasets show that SPLDHyperAWNTF can achieve better prediction performance than baseline models in terms of Top-1 precision, Top-1 recall and Top-1 F1. Furthermore, we implement case studies to further evaluate the accuracy of SPLDHyperAWNTF. As a result, 98 (MDAv2.0) and 98 (MDAv2.0-2) of top-100 are confirmed by HMDDv3.2 dataset. Moreover, the results of enrichment analysis illustrate that unconfirmed potential associations have biological significance.


Subject(s)
MicroRNAs , Humans , MicroRNAs/genetics , Computational Biology/methods , Algorithms , Genetic Predisposition to Disease
14.
Brief Bioinform ; 23(5)2022 09 20.
Article in English | MEDLINE | ID: mdl-35901464

ABSTRACT

MOTIVATION: The associations between biomarkers and human diseases play a key role in understanding complex pathology and developing targeted therapies. Wet lab experiments for biomarker discovery are costly, laborious and time-consuming. Computational prediction methods can be used to greatly expedite the identification of candidate biomarkers. RESULTS: Here, we present a novel computational model named GTGenie for predicting the biomarker-disease associations based on graph and text features. In GTGenie, a graph attention network is utilized to characterize diverse similarities of biomarkers and diseases from heterogeneous information resources. Meanwhile, a pretrained BERT-based model is applied to learn the text-based representation of biomarker-disease relation from biomedical literature. The captured graph and text features are then integrated in a bimodal fusion network to model the hybrid entity representation. Finally, inductive matrix completion is adopted to infer the missing entries for reconstructing relation matrix, with which the unknown biomarker-disease associations are predicted. Experimental results on HMDD, HMDAD and LncRNADisease data sets showed that GTGenie can obtain competitive prediction performance with other state-of-the-art methods. AVAILABILITY: The source code of GTGenie and the test data are available at: https://github.com/Wolverinerine/GTGenie.


Subject(s)
Computational Biology , Software , Computational Biology/methods , Humans
15.
Methods ; 212: 1-9, 2023 04.
Article in English | MEDLINE | ID: mdl-36813017

ABSTRACT

MicroRNA(miRNA) is a class of short non-coding RNAs with a length of about 22 nucleotides, which participates in various biological processes of cells. A number of studies have shown that miRNAs are closely related to the occurrence of cancer and various human diseases. Therefore, studying miRNA-disease associations is helpful to understand the pathogenesis of diseases as well as the prevention, diagnosis, treatment and prognosis of diseases. Traditional biological experimental methods for studying miRNA-disease associations have disadvantages such as expensive equipment, time-consuming and labor-intensive. With the rapid development of bioinformatics, more and more researchers are committed to developing effective computational methods to predict miRNA-disease associations in roder to reduce the time and money cost of experiments. In this study, we proposed a neural network-based deep matrix factorization method named NNDMF to predict miRNA-disease associations. To address the problem that traditional matrix factorization methods can only extract linear features, NNDMF used neural network to perform deep matrix factorization to extract nonlinear features, which makes up for the shortcomings of traditional matrix factorization methods. We compared NNDMF with four previous classical prediction models (IMCMDA, GRMDA, SACMDA and ICFMDA) in global LOOCV and local LOOCV, respectively. The AUCs achieved by NNDMF in two cross-validation methods were 0.9340 and 0.8763, respectively. Furthermore, we conducted case studies on three important human diseases (lymphoma, colorectal cancer and lung cancer) to validate the effectiveness of NNDMF. In conclusion, NNDMF could effectively predict the potential miRNA-disease associations.


Subject(s)
Lung Neoplasms , MicroRNAs , Humans , MicroRNAs/genetics , Genetic Predisposition to Disease , Algorithms , Neural Networks, Computer , Computational Biology/methods
16.
BMC Bioinformatics ; 24(1): 229, 2023 Jun 02.
Article in English | MEDLINE | ID: mdl-37268893

ABSTRACT

BACKGROUND: Clinical studies have shown that miRNAs are closely related to human health. The study of potential associations between miRNAs and diseases will contribute to a profound understanding of the mechanism of disease development, as well as human disease prevention and treatment. MiRNA-disease associations predicted by computational methods are the best complement to biological experiments. RESULTS: In this research, a federated computational model KATZNCP was proposed on the basis of the KATZ algorithm and network consistency projection to infer the potential miRNA-disease associations. In KATZNCP, a heterogeneous network was initially constructed by integrating the known miRNA-disease association, integrated miRNA similarities, and integrated disease similarities; then, the KATZ algorithm was implemented in the heterogeneous network to obtain the estimated miRNA-disease prediction scores. Finally, the precise scores were obtained by the network consistency projection method as the final prediction results. KATZNCP achieved the reliable predictive performance in leave-one-out cross-validation (LOOCV) with an AUC value of 0.9325, which was better than the state-of-the-art comparable algorithms. Furthermore, case studies of lung neoplasms and esophageal neoplasms demonstrated the excellent predictive performance of KATZNCP. CONCLUSION: A new computational model KATZNCP was proposed for predicting potential miRNA-drug associations based on KATZ and network consistency projections, which can effectively predict the potential miRNA-disease interactions. Therefore, KATZNCP can be used to provide guidance for future experiments.


Subject(s)
Esophageal Neoplasms , Lung Neoplasms , MicroRNAs , Humans , MicroRNAs/genetics , Algorithms , Lung Neoplasms/genetics , Computational Biology/methods , Genetic Predisposition to Disease
17.
Brief Bioinform ; 22(6)2021 11 05.
Article in English | MEDLINE | ID: mdl-34009265

ABSTRACT

Accurate identification of the miRNA-disease associations (MDAs) helps to understand the etiology and mechanisms of various diseases. However, the experimental methods are costly and time-consuming. Thus, it is urgent to develop computational methods towards the prediction of MDAs. Based on the graph theory, the MDA prediction is regarded as a node classification task in the present study. To solve this task, we propose a novel method MDA-GCNFTG, which predicts MDAs based on Graph Convolutional Networks (GCNs) via graph sampling through the Feature and Topology Graph to improve the training efficiency and accuracy. This method models both the potential connections of feature space and the structural relationships of MDA data. The nodes of the graphs are represented by the disease semantic similarity, miRNA functional similarity and Gaussian interaction profile kernel similarity. Moreover, we considered six tasks simultaneously on the MDA prediction problem at the first time, which ensure that under both balanced and unbalanced sample distribution, MDA-GCNFTG can predict not only new MDAs but also new diseases without known related miRNAs and new miRNAs without known related diseases. The results of 5-fold cross-validation show that the MDA-GCNFTG method has achieved satisfactory performance on all six tasks and is significantly superior to the classic machine learning methods and the state-of-the-art MDA prediction methods. Moreover, the effectiveness of GCNs via the graph sampling strategy and the feature and topology graph in MDA-GCNFTG has also been demonstrated. More importantly, case studies for two diseases and three miRNAs are conducted and achieved satisfactory performance.


Subject(s)
Biomarkers , Computational Biology/methods , Disease Susceptibility , Gene Expression Regulation , MicroRNAs/genetics , Software , Algorithms , Databases, Genetic , Humans , Reproducibility of Results , Workflow
18.
Brief Bioinform ; 22(5)2021 09 02.
Article in English | MEDLINE | ID: mdl-33778850

ABSTRACT

MOTIVATION: There is growing evidence showing that the dysregulations of miRNAs cause diseases through various kinds of the underlying mechanism. Thus, predicting the multiple-category associations between microRNAs (miRNAs) and diseases plays an important role in investigating the roles of miRNAs in diseases. Moreover, in contrast with traditional biological experiments which are time-consuming and expensive, computational approaches for the prediction of multicategory miRNA-disease associations are time-saving and cost-effective that are highly desired for us. RESULTS: We present a novel data-driven end-to-end learning-based method of neural multiple-category miRNA-disease association prediction (NMCMDA) for predicting multiple-category miRNA-disease associations. The NMCMDA has two main components: (i) encoder operates directly on the miRNA-disease heterogeneous network and leverages Graph Neural Network to learn miRNA and disease latent representations, respectively. (ii) Decoder yields miRNA-disease association scores with the learned latent representations as input. Various kinds of encoders and decoders are proposed for NMCMDA. Finally, the NMCMDA with the encoder of Relational Graph Convolutional Network and the neural multirelational decoder (NMR-RGCN) achieves the best prediction performance. We compared the NMCMDA with other baselines on three experimental datasets. The experimental results show that the NMR-RGCN is significantly superior to the state-of-the-art method TDRC in terms of Top-1 precision, Top-1 Recall, and Top-1 F1. Additionally, case studies are provided for two high-risk human diseases (namely, breast cancer and lung cancer) and we also provide the prediction and validation of top-10 miRNA-disease-category associations based on all known data of HMDD v3.2, which further validate the effectiveness and feasibility of the proposed method.


Subject(s)
Breast Neoplasms/genetics , Computational Biology/methods , Genetic Predisposition to Disease/genetics , Lung Neoplasms/genetics , Machine Learning , MicroRNAs/genetics , Neural Networks, Computer , Data Accuracy , Databases, Genetic , Feasibility Studies , Female , Humans
19.
Brief Bioinform ; 22(4)2021 07 20.
Article in English | MEDLINE | ID: mdl-34293850

ABSTRACT

Emerging evidence indicates that the abnormal expression of miRNAs involves in the evolution and progression of various human complex diseases. Identifying disease-related miRNAs as new biomarkers can promote the development of disease pathology and clinical medicine. However, designing biological experiments to validate disease-related miRNAs is usually time-consuming and expensive. Therefore, it is urgent to design effective computational methods for predicting potential miRNA-disease associations. Inspired by the great progress of graph neural networks in link prediction, we propose a novel graph auto-encoder model, named GAEMDA, to identify the potential miRNA-disease associations in an end-to-end manner. More specifically, the GAEMDA model applies a graph neural networks-based encoder, which contains aggregator function and multi-layer perceptron for aggregating nodes' neighborhood information, to generate the low-dimensional embeddings of miRNA and disease nodes and realize the effective fusion of heterogeneous information. Then, the embeddings of miRNA and disease nodes are fed into a bilinear decoder to identify the potential links between miRNA and disease nodes. The experimental results indicate that GAEMDA achieves the average area under the curve of $93.56\pm 0.44\%$ under 5-fold cross-validation. Besides, we further carried out case studies on colon neoplasms, esophageal neoplasms and kidney neoplasms. As a result, 48 of the top 50 predicted miRNAs associated with these diseases are confirmed by the database of differentially expressed miRNAs in human cancers and microRNA deregulation in human disease database, respectively. The satisfactory prediction performance suggests that GAEMDA model could serve as a reliable tool to guide the following researches on the regulatory role of miRNAs. Besides, the source codes are available at https://github.com/chimianbuhetang/GAEMDA.


Subject(s)
Databases, Genetic , Gene Expression Regulation, Neoplastic , MicroRNAs , Models, Genetic , Neoplasms , Neural Networks, Computer , RNA, Neoplasm , Software , Humans , MicroRNAs/biosynthesis , MicroRNAs/genetics , Neoplasms/genetics , Neoplasms/metabolism , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics
20.
Brief Bioinform ; 22(6)2021 11 05.
Article in English | MEDLINE | ID: mdl-33963829

ABSTRACT

MOTIVATION: In recent years, a growing number of studies have proved that microRNAs (miRNAs) play significant roles in the development of human complex diseases. Discovering the associations between miRNAs and diseases has become an important part of the discovery and treatment of disease. Since uncovering associations via traditional experimental methods is complicated and time-consuming, many computational methods have been proposed to identify the potential associations. However, there are still challenges in accurately determining potential associations between miRNA and disease by using multisource data. RESULTS: In this study, we develop a Multi-view Multichannel Attention Graph Convolutional Network (MMGCN) to predict potential miRNA-disease associations. Different from simple multisource information integration, MMGCN employs GCN encoder to obtain the features of miRNA and disease in different similarity views, respectively. Moreover, our MMGCN can enhance the learned latent representations for association prediction by utilizing multichannel attention, which adaptively learns the importance of different features. Empirical results on two datasets demonstrate that MMGCN model can achieve superior performance compared with nine state-of-the-art methods on most of the metrics. Furthermore, we prove the effectiveness of multichannel attention mechanism and the validity of multisource data in miRNA and disease association prediction. Case studies also indicate the ability of the method for discovering new associations.


Subject(s)
Algorithms , Biomarkers , Computational Biology/methods , Disease Susceptibility , MicroRNAs/genetics , Neural Networks, Computer , Databases, Genetic , Humans , ROC Curve , Web Browser
SELECTION OF CITATIONS
SEARCH DETAIL