Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Brief Bioinform ; 24(1)2023 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-36545805

RESUMO

Discovering the relationships between long non-coding RNAs (lncRNAs) and diseases is significant in the treatment, diagnosis and prevention of diseases. However, current identified lncRNA-disease associations are not enough because of the expensive and heavy workload of wet laboratory experiments. Therefore, it is greatly important to develop an efficient computational method for predicting potential lncRNA-disease associations. Previous methods showed that combining the prediction results of the lncRNA-disease associations predicted by different classification methods via Learning to Rank (LTR) algorithm can be effective for predicting potential lncRNA-disease associations. However, when the classification results are incorrect, the ranking results will inevitably be affected. We propose the GraLTR-LDA predictor based on biological knowledge graphs and ranking framework for predicting potential lncRNA-disease associations. Firstly, homogeneous graph and heterogeneous graph are constructed by integrating multi-source biological information. Then, GraLTR-LDA integrates graph auto-encoder and attention mechanism to extract embedded features from the constructed graphs. Finally, GraLTR-LDA incorporates the embedded features into the LTR via feature crossing statistical strategies to predict priority order of diseases associated with query lncRNAs. Experimental results demonstrate that GraLTR-LDA outperforms the other state-of-the-art predictors and can effectively detect potential lncRNA-disease associations. Availability and implementation: Datasets and source codes are available at http://bliulab.net/GraLTR-LDA.


Assuntos
Neoplasias , RNA Longo não Codificante , Humanos , RNA Longo não Codificante/genética , Biologia Computacional/métodos , Algoritmos , Software
2.
Brief Bioinform ; 24(1)2023 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-36592062

RESUMO

Recent studies have revealed that long noncoding RNAs (lncRNAs) are closely linked to several human diseases, providing new opportunities for their use in detection and therapy. Many graph propagation and similarity fusion approaches can be used for predicting potential lncRNA-disease associations. However, existing similarity fusion approaches suffer from noise and self-similarity loss in the fusion process. To address these problems, a new prediction approach, termed SSMF-BLNP, based on organically combining selective similarity matrix fusion (SSMF) and bidirectional linear neighborhood label propagation (BLNP), is proposed in this paper to predict lncRNA-disease associations. In SSMF, self-similarity networks of lncRNAs and diseases are obtained by selective preprocessing and nonlinear iterative fusion. The fusion process assigns weights to each initial similarity network and introduces a unit matrix that can reduce noise and compensate for the loss of self-similarity. In BLNP, the initial lncRNA-disease associations are employed in both lncRNA and disease directions as label information for linear neighborhood label propagation. The propagation was then performed on the self-similarity network obtained from SSMF to derive the scoring matrix for predicting the relationships between lncRNAs and diseases. Experimental results showed that SSMF-BLNP performed better than seven other state of-the-art approaches. Furthermore, a case study demonstrated up to 100% and 80% accuracy in 10 lncRNAs associated with hepatocellular carcinoma and 10 lncRNAs associated with renal cell carcinoma, respectively. The source code and datasets used in this paper are available at: https://github.com/RuiBingo/SSMF-BLNP.


Assuntos
RNA Longo não Codificante , Humanos , Algoritmos , Biologia Computacional/métodos , RNA Longo não Codificante/genética , Software , Carcinoma Hepatocelular/genética , Carcinoma de Células Renais/genética , Neoplasias Hepáticas/genética , Neoplasias Renais/genética
3.
Brief Bioinform ; 24(1)2023 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-36528809

RESUMO

MOTIVATION: Exploring the potential long noncoding RNA (lncRNA)-disease associations (LDAs) plays a critical role for understanding disease etiology and pathogenesis. Given the high cost of biological experiments, developing a computational method is a practical necessity to effectively accelerate experimental screening process of candidate LDAs. However, under the high sparsity of LDA dataset, many computational models hardly exploit enough knowledge to learn comprehensive patterns of node representations. Moreover, although the metapath-based GNN has been recently introduced into LDA prediction, it discards intermediate nodes along the meta-path and results in information loss. RESULTS: This paper presents a new multi-view contrastive heterogeneous graph attention network (GAT) for lncRNA-disease association prediction, MCHNLDA for brevity. Specifically, MCHNLDA firstly leverages rich biological data sources of lncRNA, gene and disease to construct two-view graphs, feature structural graph of feature schema view and lncRNA-gene-disease heterogeneous graph of network topology view. Then, we design a cross-contrastive learning task to collaboratively guide graph embeddings of the two views without relying on any labels. In this way, we can pull closer the nodes of similar features and network topology, and push other nodes away. Furthermore, we propose a heterogeneous contextual GAT, where long short-term memory network is incorporated into attention mechanism to effectively capture sequential structure information along the meta-path. Extensive experimental comparisons against several state-of-the-art methods show the effectiveness of proposed framework.The code and data of proposed framework is freely available at https://github.com/zhaoxs686/MCHNLDA.


Assuntos
RNA Longo não Codificante , RNA Longo não Codificante/genética , Aprendizagem
4.
BMC Bioinformatics ; 25(1): 46, 2024 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-38287236

RESUMO

BACKGROUND: Many biological studies have shown that lncRNAs regulate the expression of epigenetically related genes. The study of lncRNAs has helped to deepen our understanding of the pathogenesis of complex diseases at the molecular level. Due to the large number of lncRNAs and the complex and time-consuming nature of biological experiments, applying computer techniques to predict potential lncRNA-disease associations is very effective. To explore information between complex network structures, existing methods rely mainly on lncRNA and disease information. Metapaths have been applied to network models as an effective method for exploring information in heterogeneous graphs. However, existing methods are dominated by lncRNAs or disease nodes and tend to ignore the paths provided by intermediate nodes. METHODS: We propose a deep learning model based on hierarchical graphical attention networks to predict unknown lncRNA-disease associations using multiple types of metapaths to extract features. We have named this model the MMHGAN. First, the model constructs a lncRNA-disease-miRNA heterogeneous graph based on known associations and two homogeneous graphs of lncRNAs and diseases. Second, for homogeneous graphs, the features of neighboring nodes are aggregated using a multihead attention mechanism. Third, for the heterogeneous graph, metapaths of different intermediate nodes are selected to construct subgraphs, and the importance of different types of metapaths is calculated and aggregated to obtain the final embedded features. Finally, the features are reconstructed using a fully connected layer to obtain the prediction results. RESULTS: We used a fivefold cross-validation method and obtained an average AUC value of 96.07% and an average AUPR value of 93.23%. Additionally, ablation experiments demonstrated the role of homogeneous graphs and different intermediate node path weights. In addition, we studied lung cancer, esophageal carcinoma, and breast cancer. Among the 15 lncRNAs associated with these diseases, 15, 12, and 14 lncRNAs were validated by the lncRNA Disease Database and the Lnc2Cancer Database, respectively. CONCLUSION: We compared the MMHGAN model with six existing models with better performance, and the case study demonstrated that the model was effective in predicting the correlation between potential lncRNAs and diseases.


Assuntos
Neoplasias da Mama , Neoplasias Pulmonares , MicroRNAs , RNA Longo não Codificante , Humanos , Feminino , RNA Longo não Codificante/genética , Biologia Computacional/métodos , MicroRNAs/genética , Algoritmos
5.
BMC Bioinformatics ; 25(1): 187, 2024 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-38741200

RESUMO

MOTIVATION: Long non-coding RNAs (lncRNAs) are a class of molecules involved in important biological processes. Extensive efforts have been provided to get deeper understanding of disease mechanisms at the lncRNA level, guiding towards the detection of biomarkers for disease diagnosis, treatment, prognosis and prevention. Unfortunately, due to costs and time complexity, the number of possible disease-related lncRNAs verified by traditional biological experiments is very limited. Computational approaches for the prediction of disease-lncRNA associations allow to identify the most promising candidates to be verified in laboratory, reducing costs and time consuming. RESULTS: We propose novel approaches for the prediction of lncRNA-disease associations, all sharing the idea of exploring associations among lncRNAs, other intermediate molecules (e.g., miRNAs) and diseases, suitably represented by tripartite graphs. Indeed, while only a few lncRNA-disease associations are still known, plenty of interactions between lncRNAs and other molecules, as well as associations of the latters with diseases, are available. A first approach presented here, NGH, relies on neighborhood analysis performed on a tripartite graph, built upon lncRNAs, miRNAs and diseases. A second approach (CF) relies on collaborative filtering; a third approach (NGH-CF) is obtained boosting NGH by collaborative filtering. The proposed approaches have been validated on both synthetic and real data, and compared against other methods from the literature. It results that neighborhood analysis allows to outperform competitors, and when it is combined with collaborative filtering the prediction accuracy further improves, scoring a value of AUC equal to 0966. AVAILABILITY: Source code and sample datasets are available at: https://github.com/marybonomo/LDAsPredictionApproaches.git.


Assuntos
Biologia Computacional , RNA Longo não Codificante , RNA Longo não Codificante/genética , Humanos , Biologia Computacional/métodos , Algoritmos , MicroRNAs/genética , MicroRNAs/metabolismo , Predisposição Genética para Doença/genética
6.
BMC Genomics ; 25(1): 73, 2024 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-38233788

RESUMO

BACKGROUND: Long noncoding RNAs (lncRNAs) are integral to a plethora of critical cellular biological processes, including the regulation of gene expression, cell differentiation, and the development of tumors and cancers. Predicting the relationships between lncRNAs and diseases can contribute to a better understanding of the pathogenic mechanisms of disease and provide strong support for the development of advanced treatment methods. RESULTS: Therefore, we present an innovative Node-Adaptive Graph Transformer model for predicting unknown LncRNA-Disease Associations, named NAGTLDA. First, we utilize the node-adaptive feature smoothing (NAFS) method to learn the local feature information of nodes and encode the structural information of the fusion similarity network of diseases and lncRNAs using Structural Deep Network Embedding (SDNE). Next, the Transformer module is used to capture potential association information between the network nodes. Finally, we employ a Transformer module with two multi-headed attention layers for learning global-level embedding fusion. Network structure coding is added as the structural inductive bias of the network to compensate for the missing message-passing mechanism in Transformer. NAGTLDA achieved an average AUC of 0.9531 and AUPR of 0.9537 significantly higher than state-of-the-art methods in 5-fold cross validation. We perform case studies on 4 diseases; 55 out of 60 associations between lncRNAs and diseases have been validated in the literatures. The results demonstrate the enormous potential of the graph Transformer structure to incorporate graph structural information for uncovering lncRNA-disease unknown correlations. CONCLUSIONS: Our proposed NAGTLDA model can serve as a highly efficient computational method for predicting biological information associations.


Assuntos
Neoplasias , RNA Longo não Codificante , Humanos , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Biologia Computacional/métodos , Neoplasias/genética , Algoritmos
7.
Brief Bioinform ; 23(1)2022 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-34486019

RESUMO

Long noncoding RNAs (lncRNAs) play important roles in various biological regulatory processes, and are closely related to the occurrence and development of diseases. Identifying lncRNA-disease associations is valuable for revealing the molecular mechanism of diseases and exploring treatment strategies. Thus, it is necessary to computationally predict lncRNA-disease associations as a complementary method for biological experiments. In this study, we proposed a novel prediction method GCRFLDA based on the graph convolutional matrix completion. GCRFLDA first constructed a graph using the available lncRNA-disease association information. Then, it constructed an encoder consisting of conditional random field and attention mechanism to learn efficient embeddings of nodes, and a decoder layer to score lncRNA-disease associations. In GCRFLDA, the Gaussian interaction profile kernels similarity and cosine similarity were fused as side information of lncRNA and disease nodes. Experimental results on four benchmark datasets show that GCRFLDA is superior to other existing methods. Moreover, we conducted case studies on four diseases and observed that 70 of 80 predicted associated lncRNAs were confirmed by the literature.


Assuntos
RNA Longo não Codificante , Algoritmos , Biologia Computacional/métodos , RNA Longo não Codificante/genética , Projetos de Pesquisa
8.
Brief Bioinform ; 23(5)2022 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-35901464

RESUMO

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.


Assuntos
Biologia Computacional , Software , Biologia Computacional/métodos , Humanos
9.
BMC Bioinformatics ; 23(1): 5, 2022 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-34983367

RESUMO

BACKGROUND: More and more evidence showed that long non-coding RNAs (lncRNAs) play important roles in the development and progression of human sophisticated diseases. Therefore, predicting human lncRNA-disease associations is a challenging and urgently task in bioinformatics to research of human sophisticated diseases. RESULTS: In the work, a global network-based computational framework called as LRWRHLDA were proposed which is a universal network-based method. Firstly, four isomorphic networks include lncRNA similarity network, disease similarity network, gene similarity network and miRNA similarity network were constructed. And then, six heterogeneous networks include known lncRNA-disease, lncRNA-gene, lncRNA-miRNA, disease-gene, disease-miRNA, and gene-miRNA associations network were applied to design a multi-layer network. Finally, the Laplace normalized random walk with restart algorithm in this global network is suggested to predict the relationship between lncRNAs and diseases. CONCLUSIONS: The ten-fold cross validation is used to evaluate the performance of LRWRHLDA. As a result, LRWRHLDA achieves an AUC of 0.98402, which is higher than other compared methods. Furthermore, LRWRHLDA can predict isolated disease-related lnRNA (isolated lnRNA related disease). The results for colorectal cancer, lung adenocarcinoma, stomach cancer and breast cancer have been verified by other researches. The case studies indicated that our method is effective.


Assuntos
MicroRNAs , Neoplasias/genética , RNA Longo não Codificante , Algoritmos , Biologia Computacional , Redes Reguladoras de Genes , Humanos , MicroRNAs/genética , RNA Longo não Codificante/genética
10.
BMC Bioinformatics ; 23(1): 189, 2022 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-35590258

RESUMO

BACKGROUND: Many long non-coding RNAs (lncRNAs) have key roles in different human biologic processes and are closely linked to numerous human diseases, according to cumulative evidence. Predicting potential lncRNA-disease associations can help to detect disease biomarkers and perform disease analysis and prevention. Establishing effective computational methods for lncRNA-disease association prediction is critical. RESULTS: In this paper, we propose a novel model named MAGCNSE to predict underlying lncRNA-disease associations. We first obtain multiple feature matrices from the multi-view similarity graphs of lncRNAs and diseases utilizing graph convolutional network. Then, the weights are adaptively assigned to different feature matrices of lncRNAs and diseases using the attention mechanism. Next, the final representations of lncRNAs and diseases is acquired by further extracting features from the multi-channel feature matrices of lncRNAs and diseases using convolutional neural network. Finally, we employ a stacking ensemble classifier, consisting of multiple traditional machine learning classifiers, to make the final prediction. The results of ablation studies in both representation learning methods and classification methods demonstrate the validity of each module. Furthermore, we compare the overall performance of MAGCNSE with that of six other state-of-the-art models, the results show that it outperforms the other methods. Moreover, we verify the effectiveness of using multi-view data of lncRNAs and diseases. Case studies further reveal the outstanding ability of MAGCNSE in the identification of potential lncRNA-disease associations. CONCLUSIONS: The experimental results indicate that MAGCNSE is a useful approach for predicting potential lncRNA-disease associations.


Assuntos
RNA Longo não Codificante , Biologia Computacional/métodos , Humanos , Aprendizado de Máquina , Redes Neurais de Computação , RNA Longo não Codificante/genética
11.
BMC Bioinformatics ; 22(Suppl 3): 241, 2021 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-33980147

RESUMO

BACKGROUND: In the development of science and technology, there are increasing evidences that there are some associations between lncRNAs and human diseases. Therefore, finding these associations between them will have a huge impact on our treatment and prevention of some diseases. However, the process of finding the associations between them is very difficult and requires a lot of time and effort. Therefore, it is particularly important to find some good methods for predicting lncRNA-disease associations (LDAs). RESULTS: In this paper, we propose a method based on dual sparse collaborative matrix factorization (DSCMF) to predict LDAs. The DSCMF method is improved on the traditional collaborative matrix factorization method. To increase the sparsity, the L2,1-norm is added in our method. At the same time, Gaussian interaction profile kernel is added to our method, which increase the network similarity between lncRNA and disease. Finally, the AUC value obtained by the experiment is used to evaluate the quality of our method, and the AUC value is obtained by the ten-fold cross-validation method. CONCLUSIONS: The AUC value obtained by the DSCMF method is 0.8523. At the end of the paper, simulation experiment is carried out, and the experimental results of prostate cancer, breast cancer, ovarian cancer and colorectal cancer are analyzed in detail. The DSCMF method is expected to bring some help to lncRNA-disease associations research. The code can access the https://github.com/Ming-0113/DSCMF website.


Assuntos
Neoplasias da Mama , Neoplasias da Próstata , RNA Longo não Codificante , Algoritmos , Simulação por Computador , Humanos , Masculino , Neoplasias da Próstata/genética , RNA Longo não Codificante/genética
12.
Methods ; 173: 32-43, 2020 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-31226302

RESUMO

Influx evidences show that red long non-coding RNAs (lncRNAs) play important roles in various critical biological processes, and they afffect the development and progression of various human diseases. Therefore, it is necessary to precisely identify the lncRNA-disease associations. The identification precision can be improved by developing data integrative models. However, current models mainly need to project heterogeneous data onto the homologous networks, and then merge these networks into a composite one for integrative prediction. We recognize that this projection overrides the individual structure of the heterogeneous data, and the combination is impacted by noisy networks. As a result, the performance is compromised. Given that, we introduce a weighted matrix factorization model on multi-relational data to predict LncRNA-disease associations (WMFLDA). WMFLDA firstly uses a heterogeneous network to capture the inter(intra)-associations between different types of nodes (including genes, lncRNAs, and Disease Ontology terms). Then, it presets weights to these inter-association and intra-association matrices of the network, and cooperatively decomposes these matrices into low-rank ones to explore the underlying relationships between nodes. Next, it jointly optimizes the low-rank matrices and the weights. After that, WMFLDA approximates the lncRNA-disease association matrix using the optimized matrices and weights, and thus to achieve the prediction. WMFLDA obtains a much better performance than related data integrative solutions across different experiment settings and evaluation metrics. It can not only respect the intrinsic structures of individual data sources, but can also fuse them with selection.


Assuntos
Biologia Computacional/métodos , Predisposição Genética para Doença , RNA Longo não Codificante/genética , Algoritmos , Progressão da Doença , Humanos
13.
Genomics ; 112(6): 4777-4787, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33348478

RESUMO

An increasing number of research shows that long non-coding RNA plays a key role in many important biological processes. However, the number of disease-related lncRNAs found by researchers remains relatively small, and experimental identification is time consuming and labor intensive. In this study, we propose a novel method, namely HAUBRW, to predict undiscovered lncRNA-disease associations. First, the hybrid algorithm, which combines the heat spread algorithm and the probability diffusion algorithm, redistributes the resources. Second, unbalanced bi-random walk, is used to infer undiscovered lncRNA disease associations. Seven advanced models, i.e. BRWLDA, DSCMF, RWRlncD, IDLDA, KATZ, Ping's, and Yang's were compared with our method, and simulation results show that the AUC of our method is more perfect than the other models. In addition, case studies have shown that HAUBRW can effectively predict candidate lncRNAs for breast, osteosarcoma and cervical cancer. Therefore, our approach may be a good choice in future biomedical research.


Assuntos
Algoritmos , Biologia Computacional , Predisposição Genética para Doença , RNA Longo não Codificante/genética , Simulação por Computador , Estudos de Associação Genética , Humanos
14.
BMC Bioinformatics ; 21(1): 339, 2020 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-32736513

RESUMO

BACKGROUND: It has been widely accepted that long non-coding RNAs (lncRNAs) play important roles in the development and progression of human diseases. Many association prediction models have been proposed for predicting lncRNA functions and identifying potential lncRNA-disease associations. Nevertheless, among them, little effort has been attempted to measure lncRNA functional similarity, which is an essential part of association prediction models. RESULTS: In this study, we presented an lncRNA functional similarity calculation model, IDSSIM for short, based on an improved disease semantic similarity method, highlight of which is the introduction of information content contribution factor into the semantic value calculation to take into account both the hierarchical structures of disease directed acyclic graphs and the disease specificities. IDSSIM and three state-of-the-art models, i.e., LNCSIM1, LNCSIM2, and ILNCSIM, were evaluated by applying their disease semantic similarity matrices and the lncRNA functional similarity matrices, as well as corresponding matrices of human lncRNA-disease associations coming from either lncRNADisease database or MNDR database, into an association prediction method WKNKN for lncRNA-disease association prediction. In addition, case studies of breast cancer and adenocarcinoma were also performed to validate the effectiveness of IDSSIM. CONCLUSIONS: Results demonstrated that in terms of ROC curves and AUC values, IDSSIM is superior to compared models, and can improve accuracy of disease semantic similarity effectively, leading to increase the association prediction ability of the IDSSIM-WKNKN model; in terms of case studies, most of potential disease-associated lncRNAs predicted by IDSSIM can be confirmed by databases and literatures, implying that IDSSIM can serve as a promising tool for predicting lncRNA functions, identifying potential lncRNA-disease associations, and pre-screening candidate lncRNAs to perform biological experiments. The IDSSIM code, all experimental data and prediction results are available online at https://github.com/CDMB-lab/IDSSIM .


Assuntos
Algoritmos , Biologia Computacional/métodos , Doença/genética , Modelos Genéticos , RNA Longo não Codificante/genética , Semântica , Adenocarcinoma/genética , Área Sob a Curva , Neoplasias da Mama/genética , Bases de Dados Genéticas , Feminino , Humanos , Curva ROC
15.
BMC Bioinformatics ; 20(1): 396, 2019 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-31315558

RESUMO

BACKGROUND: Since the number of known lncRNA-disease associations verified by biological experiments is quite limited, it has been a challenging task to uncover human disease-related lncRNAs in recent years. Moreover, considering the fact that biological experiments are very expensive and time-consuming, it is important to develop efficient computational models to discover potential lncRNA-disease associations. RESULTS: In this manuscript, a novel Collaborative Filtering model called CFNBC for inferring potential lncRNA-disease associations is proposed based on Naïve Bayesian Classifier. In CFNBC, an original lncRNA-miRNA-disease tripartite network is constructed first by integrating known miRNA-lncRNA associations, miRNA-disease associations and lncRNA-disease associations, and then, an updated lncRNA-miRNA-disease tripartite network is further constructed through applying the item-based collaborative filtering algorithm on the original tripartite network. Finally, based on the updated tripartite network, a novel approach based on the Naïve Bayesian Classifier is proposed to predict potential associations between lncRNAs and diseases. The novelty of CFNBC lies in the construction of the updated lncRNA-miRNA-disease tripartite network and the introduction of the item-based collaborative filtering algorithm and Naïve Bayesian Classifier, which guarantee that CFNBC can be applied to predict potential lncRNA-disease associations efficiently without entirely relying on known miRNA-disease associations. Simulation results show that CFNBC can achieve a reliable AUC of 0.8576 in the Leave-One-Out Cross Validation (LOOCV), which is considerably better than previous state-of-the-art results. Moreover, case studies of glioma, colorectal cancer and gastric cancer demonstrate the excellent prediction performance of CFNBC as well. CONCLUSIONS: According to simulation results, due to the satisfactory prediction performance, CFNBC may be an excellent addition to biomedical researches in the future.


Assuntos
Doença/genética , RNA Longo não Codificante/metabolismo , Algoritmos , Teorema de Bayes , Neoplasias Colorretais/genética , Simulação por Computador , Glioma/genética , Humanos , MicroRNAs/metabolismo , Neoplasias Gástricas/genética
16.
Mol Genet Genomics ; 294(6): 1477-1486, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31250107

RESUMO

Long noncoding RNAs play a significant role in the occurrence of diseases. Thus, studying the relationship prediction between lncRNAs and disease is becoming more popular. Researchers hope to determine effective treatments by revealing the occurrence and development of diseases at the molecular level. However, the traditional biological experimental way to verify the association between lncRNAs and disease is very time-consuming and expensive. Therefore, we developed a method called LLCLPLDA to predict potential lncRNA-disease associations. First, locality-constrained linear coding (LLC) is leveraged to project the features of lncRNAs and diseases to local-constraint features, and then, a label propagation (LP) strategy is used to mix up the initial association matrix and the obtained features of lncRNAs and diseases. To demonstrate the performance of our method, we compared LLCLPLDA with five methods in the leave-one-out cross-validation and fivefold cross-validation scheme, and the experimental results show that the proposed method outperforms the other five methods. Additionally, we conducted case studies on three diseases: cervical cancer, gliomas, and breast cancer. The top five predicted lncRNAs for cervical cancer and gliomas were verified, and four of the five lncRNAs for breast cancer were also confirmed.


Assuntos
Algoritmos , Doença/genética , RNA Longo não Codificante/metabolismo , Neoplasias da Mama/genética , Feminino , Glioma/genética , Humanos , Modelos Genéticos , Neoplasias do Colo do Útero/genética
17.
Math Biosci Eng ; 21(4): 4814-4834, 2024 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-38872515

RESUMO

Long non-coding RNA (lncRNA) is considered to be a crucial regulator involved in various human biological processes, including the regulation of tumor immune checkpoint proteins. It has great potential as both a cancer biomolecular biomarker and therapeutic target. Nevertheless, conventional biological experimental techniques are both resource-intensive and laborious, making it essential to develop an accurate and efficient computational method to facilitate the discovery of potential links between lncRNAs and diseases. In this study, we proposed HRGCNLDA, a computational approach utilizing hierarchical refinement of graph convolutional neural networks for forecasting lncRNA-disease potential associations. This approach effectively addresses the over-smoothing problem that arises from stacking multiple layers of graph convolutional neural networks. Specifically, HRGCNLDA enhances the layer representation during message propagation and node updates, thereby amplifying the contribution of hidden layers that resemble the ego layer while reducing discrepancies. The results of the experiments showed that HRGCNLDA achieved the highest AUC-ROC (area under the receiver operating characteristic curve, AUC for short) and AUC-PR (area under the precision versus recall curve, AUPR for short) values compared to other methods. Finally, to further demonstrate the reliability and efficacy of our approach, we performed case studies on the case of three prevalent human diseases, namely, breast cancer, lung cancer and gastric cancer.


Assuntos
Algoritmos , Área Sob a Curva , Biologia Computacional , Redes Neurais de Computação , RNA Longo não Codificante , Curva ROC , RNA Longo não Codificante/genética , Humanos , Biologia Computacional/métodos , Neoplasias/genética , Neoplasias Pulmonares/genética , Neoplasias da Mama/genética , Biomarcadores Tumorais/genética , Feminino , Previsões
18.
Sci Rep ; 14(1): 5185, 2024 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-38431702

RESUMO

LncRNAs are non-coding RNAs with a length of more than 200 nucleotides. More and more evidence shows that lncRNAs are inextricably linked with diseases. To make up for the shortcomings of traditional methods, researchers began to collect relevant biological data in the database and used bioinformatics prediction tools to predict the associations between lncRNAs and diseases, which greatly improved the efficiency of the study. To improve the prediction accuracy of current methods, we propose a new lncRNA-disease associations prediction method with attention mechanism, called ResGCN-A. Firstly, we integrated lncRNA functional similarity, lncRNA Gaussian interaction profile kernel similarity, disease semantic similarity, and disease Gaussian interaction profile kernel similarity to obtain lncRNA comprehensive similarity and disease comprehensive similarity. Secondly, the residual graph convolutional network was used to extract the local features of lncRNAs and diseases. Thirdly, the new attention mechanism was used to assign the weight of the above features to further obtain the potential features of lncRNAs and diseases. Finally, the training set required by the Extra-Trees classifier was obtained by concatenating potential features, and the potential associations between lncRNAs and diseases were obtained by the trained Extra-Trees classifier. ResGCN-A combines the residual graph convolutional network with the attention mechanism to realize the local and global features fusion of lncRNA and diseases, which is beneficial to obtain more accurate features and improve the prediction accuracy. In the experiment, ResGCN-A was compared with five other methods through 5-fold cross-validation. The results show that the AUC value and AUPR value obtained by ResGCN-A are 0.9916 and 0.9951, which are superior to the other five methods. In addition, case studies and robustness evaluation have shown that ResGCN-A is an effective method for predicting lncRNA-disease associations. The source code for ResGCN-A will be available at https://github.com/Wangxiuxiun/ResGCN-A .


Assuntos
RNA Longo não Codificante , RNA Longo não Codificante/genética , Algoritmos , Software , Biologia Computacional/métodos , Bases de Dados Factuais
19.
Interdiscip Sci ; 15(3): 439-451, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37308797

RESUMO

Numerous scientific evidences have revealed that long non-coding RNAs (lncRNAs) are involved in the progression of human complex diseases and biological life activities. Therefore, identifying novel and potential disease-related lncRNAs is helpful to diagnosis, prognosis and therapy of many human complex diseases. Since traditional laboratory experiments are cost and time-consuming, a great quantity of computer algorithms have been proposed for predicting the relationships between lncRNAs and diseases. However, there are still much room for the improvement. In this paper, we introduce an accurate framework named LDAEXC to infer LncRNA-Disease Associations with deep autoencoder and XGBoost Classifier. LDAEXC utilizes different similarity views of lncRNAs and human diseases to construct features for each data sources. Then, the reduced features are obtained by feeding the constructed feature vectors into a deep autoencoder, and at last an XGBoost classifier is leveraged to calculate the latent lncRNA-disease-associated scores using reduced features. The fivefold cross-validation experiments on four datasets showed that LDAEXC reached AUC scores of 0.9676 ± 0.0043, 0.9449 ± 0.022, 0.9375 ± 0.0331 and 0.9556 ± 0.0134, respectively, significantly higher than other advanced similar computer methods. Extensive experiment results and case studies of two complex diseases (colon and breast cancers) further indicated the practicability and excellent prediction performance of LDAEXC in inferring unknown lncRNA-disease associations. TLDAEXC utilizes disease semantic similarity, lncRNA expression similarity, and Gaussian interaction profile kernel similarity of lncRNAs and diseases for feature construction. The constructed features are fed to a deep autoencoder to extract reduced features, and an XGBoost classifier is used to predict the lncRNA-disease associations based on the reduced features. The fivefold and tenfold cross-validation experiments on a benchmark dataset showed that LDAEXC could achieve AUC scores of 0.9676 and 0.9682, respectively, significantly higher than other state-of-the-art similar methods.


Assuntos
Neoplasias da Mama , RNA Longo não Codificante , Humanos , Feminino , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Algoritmos , Neoplasias da Mama/genética , Biologia Computacional/métodos
20.
Front Genet ; 14: 1084482, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37274787

RESUMO

Identification of long non-coding RNAs (lncRNAs) associated with common diseases is crucial for patient self-diagnosis and monitoring of health conditions using artificial intelligence (AI) technology at home. LncRNAs have gained significant attention due to their crucial roles in the pathogenesis of complex human diseases and identifying their associations with diseases can aid in developing diagnostic biomarkers at the molecular level. Computational methods for predicting lncRNA-disease associations (LDAs) have become necessary due to the time-consuming and labor-intensive nature of wet biological experiments in hospitals, enabling patients to access LDAs through their AI terminal devices at any time. Here, we have developed a predictive tool, LDAGRL, for identifying potential LDAs using a bridge heterogeneous information network (BHnet) constructed via Structural Deep Network Embedding (SDNE). The BHnet consists of three types of molecules as bridge nodes to implicitly link the lncRNA with disease nodes and the SDNE is used to learn high-quality node representations and make LDA predictions in a unified graph space. To assess the feasibility and performance of LDAGRL, extensive experiments, including 5-fold cross-validation, comparison with state-of-the-art methods, comparison on different classifiers and comparison of different node feature combinations, were conducted, and the results showed that LDAGRL achieved satisfactory prediction performance, indicating its potential as an effective LDAs prediction tool for family medicine and primary care.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA