Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Methods ; 229: 71-81, 2024 Jun 22.
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.

2.
Article in English | MEDLINE | ID: mdl-38767995

ABSTRACT

The arduous and costly journey of drug discovery is increasingly intersecting with computational approaches, which promise to accelerate the analysis of bioassays and biomedical literature. The critical role of microRNAs (miRNAs) in disease progression has been underscored in recent studies, elevating them as potential therapeutic targets. This emphasizes the need for the development of sophisticated computational models that can effectively identify promising drug targets, such as miRNAs. Herein, we present a novel method, termed Duplex Link Prediction (DLP), rooted in subspace segmentation, to pinpoint potential miRNA targets. Our approach initiates with the application of the Network Enhancement (NE) algorithm to refine the similarity metric between miRNAs. Thereafter, we construct two matrices by pre-loading the association matrix from both the drug and miRNA perspectives, employing the K Nearest Neighbors (KNN) technique. The DLSR algorithm is then applied to predict potential associations. The final predicted association scores are ascertained through the weighted mean of the two matrices. Our empirical findings suggest that the DLP algorithm outperforms current methodologies in the realm of identifying potential miRNA drug targets. Case study validations further reinforce the real-world applicability and effectiveness of our proposed method. The code of DLP is freely available at https://github.com/kaizheng-academic/DLP.

3.
Methods ; 226: 21-27, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38608849

ABSTRACT

Knowledge graph intent graph attention mechanism Predicting drug-target interactions (DTIs) plays a crucial role in drug discovery and drug development. Considering the high cost and risk of biological experiments, developing computational approaches to explore the interactions between drugs and targets can effectively reduce the time and cost of drug development. Recently, many methods have made significant progress in predicting DTIs. However, existing approaches still suffer from the high sparsity of DTI datasets and the cold start problem. In this paper, we develop a new model to predict drug-target interactions via a knowledge graph and intent graph named DTKGIN. Our method can effectively capture biological environment information for targets and drugs by mining their associated relations in the knowledge graph and considering drug-target interactions at a fine-grained level in the intent graph. DTKGIN learns the representation of drugs and targets from the knowledge graph and the intent graph. Then the probabilities of interactions between drugs and targets are obtained through the inner product of the representation of drugs and targets. Experimental results show that our proposed method outperforms other state-of-the-art methods in 10-fold cross-validation, especially in cold-start experimental settings. Furthermore, the case studies demonstrate the effectiveness of DTKGIN in predicting potential drug-target interactions. The code is available on GitHub: https://github.com/Royluoyi123/DTKGIN.


Subject(s)
Drug Discovery , Drug Discovery/methods , Humans , Algorithms , Computational Biology/methods , Drug Development/methods
4.
Article in English | MEDLINE | ID: mdl-38358864

ABSTRACT

Many studies have proven that microRNAs (miRNAs) can participate in a wide range of biological processes and can be considered as potential noninvasive biomarkers for disease diagnosis and prognosis. Therefore, many computational methods have been developed to identifying miRNA-disease associations, ultimately enhancing the efficiency of disease diagnosis and treatment. In this study, we also introduced a new computational method called PMDAGS, which predicts miRNA-disease associations by utilizing graph nonlinear diffusion convolution network and similarities. PMDAGS first calculates miRNA similarity and disease similarity based on miRNA-target interactions, disease-gene associations and known miRNA-disease associations, respectively. Next, we construct the initial feature of each miRNA (disease) by concatenating its final similarity vector with its known association vector. Based on the known miRNA-disease association network and the initial feature vector of each node, we further apply nonlinear diffusion graph convolution network model to extract the feature embedding vectors. Finally, we concatenate the feature embedding vectors of miRNA and disease and input them into a multi-layer perceptron to identify potential miRNA-disease associations. We conduct 5-fold cross validation (5CV), 10-fold cross validation (10CV), and global leave-one-out cross validation (GLOOCV) on HMDD v2.0 and HMDD v3.2. PMDAGS achieves AUCs of 0.9222, 0.9228, and 0.9221 under 5CV, 10CV and GLOOCV on HMDD v2.0, respectively. In addition, PMDAGS also achieves AUC values of 0.9366, 0.9377, and 0.9376 under 5CV, 10CV and GLOOCV on HMDD v3.2, respectively. According to the experimental results, we can conclude that PMDAGS outperforms other compared methods and can effectively predict miRNA-disease associations.


Subject(s)
Computational Biology , MicroRNAs , MicroRNAs/genetics , Humans , Computational Biology/methods , Algorithms , Neural Networks, Computer , Genetic Predisposition to Disease/genetics
5.
Front Genet ; 14: 1271311, 2023.
Article in English | MEDLINE | ID: mdl-37795241

ABSTRACT

Background: Drug repositioning is considered a promising drug development strategy with the goal of discovering new uses for existing drugs. Compared with the experimental screening for drug discovery, computational drug repositioning offers lower cost and higher efficiency and, hence, has become a hot issue in bioinformatics. However, there are sparse samples, multi-source information, and even some noises, which makes it difficult to accurately identify potential drug-associated indications. Methods: In this article, we propose a new scheme with improved tensor robust principal component analysis (ITRPCA) in multi-source data to predict promising drug-disease associations. First, we use a weighted k-nearest neighbor (WKNN) approach to increase the overall density of the drug-disease association matrix that will assist in prediction. Second, a drug tensor with five frontal slices and a disease tensor with two frontal slices are constructed using multi-similarity matrices and an updated association matrix. The two target tensors naturally integrate multiple sources of data from the drug-side aspect and the disease-side aspect, respectively. Third, ITRPCA is employed to isolate the low-rank tensor and noise information in the tensor. In this step, an additional range constraint is incorporated to ensure that all the predicted entry values of a low-rank tensor are within the specific interval. Finally, we focus on identifying promising drug indications by analyzing drug-disease association pairs derived from the low-rank drug and low-rank disease tensors. Results: We evaluate the effectiveness of the ITRPCA method by comparing it with five prominent existing drug repositioning methods. This evaluation is carried out using 10-fold cross-validation and independent testing experiments. Our numerical results show that ITRPCA not only yields higher prediction accuracy but also exhibits remarkable computational efficiency. Furthermore, case studies demonstrate the practical effectiveness of our method.

6.
IEEE/ACM Trans Comput Biol Bioinform ; 20(6): 3715-3724, 2023.
Article in English | MEDLINE | ID: mdl-37708020

ABSTRACT

Identifying the function of therapeutic peptides is an important issue in the development of novel drugs. To reduce the time and labor costs required to identify therapeutic peptides, computational methods are increasingly required. However, most of the existing peptide therapeutic function prediction models are used for predicting a single therapeutic function, ignoring the fact that a bioactive peptide might simultaneously consist of multi-activities. Furthermore, in the few existing multi-label classification models, the feature extraction procedures are still rough. We propose a multi-label framework, called SCN-MLTPP, with a stacked capsule network for predicting the therapeutic properties of peptides. Instead of using peptide sequence vectors alone, SCN-MLTPP extracts different view representation vectors from the therapeutic peptides and learns the contributions of different views to the properties of therapeutic peptides based on the dynamic routing mechanism. Benchmarking results show that as compared with existing multi-label predictors, SCN-MLTPP achieves better and more robust performance for different peptides. In addition, some visual analyses and case studies also demonstrate the model can reliably capture features from multi-view data and predict different peptides.


Subject(s)
Benchmarking , Peptides , Peptides/pharmacology
7.
IEEE/ACM Trans Comput Biol Bioinform ; 20(5): 2712-2723, 2023.
Article in English | MEDLINE | ID: mdl-34110998

ABSTRACT

The Anatomical Therapeutic Chemical (ATC) classification system, designated by the World Health Organization Collaborating Center (WHOCC), has been widely used in drug screening, repositioning, and similarity research. The ATC classification system assigns different codes to drugs according to the organ or system on which they act and/or their therapeutic and chemical characteristics. Correctly identifying the potential ATC codes for drugs can accelerate drug development and reduce the cost of experiments. Several classifiers have been proposed in this regard. However, they lack of ability to learn basic features from sparsely known drug-ATC code associations. Therefore, there is an urgent need for novel computational methods to precisely predict potential drug-ATC code associations in multiple levels of the ATC classification system based on known associations between drugs and ATC codes. In this paper, we provide a novel end-to-end model, so-called RNPredATC, to predict potential drug-ATC code associations in five ATC classification levels. RNPredATC can extract dense feature vectors from sparsely known drug-ATC code associations and reduce the impact from the degradation problem by a novel deep residual learning. We extensively compare our method with some state-of-the-art methods, including NetPredATC, SPACE, and some multi-label-based methods. Our experimental results show that RNPredATC achieves better performances in five-fold and ten-fold cross validations. Furthermore, the visualization analysis of hidden layers and case studies of predicted associations at the fifth ATC classification level confirm that RNPredATC can effectively identify the potential ATC codes of drugs.

8.
Article in English | MEDLINE | ID: mdl-35471889

ABSTRACT

The identification of drug-target relations (DTRs) is substantial in drug development. A large number of methods treat DTRs as drug-target interactions (DTIs), a binary classification problem. The main drawback of these methods are the lack of reliable negative samples and the absence of many important aspects of DTR, including their dose dependence and quantitative affinities. With increasing number of publications of drug-protein binding affinity data recently, DTRs prediction can be viewed as a regression problem of drug-target affinities (DTAs) which reflects how tightly the drug binds to the target and can present more detailed and specific information than DTIs. The growth of affinity data enables the use of deep learning architectures, which have been shown to be among the state-of-the-art methods in binding affinity prediction. Although relatively effective, due to the black-box nature of deep learning, these models are less biologically interpretable. In this study, we proposed a deep learning-based model, named AttentionDTA, which uses attention mechanism to predict DTAs. Different from the models using 3D structures of drug-target complexes or graph representation of drugs and proteins, the novelty of our work is to use attention mechanism to focus on key subsequences which are important in drug and protein sequences when predicting its affinity. We use two separate one-dimensional Convolution Neural Networks (1D-CNNs) to extract the semantic information of drug's SMILES string and protein's amino acid sequence. Furthermore, a two-side multi-head attention mechanism is developed and embedded to our model to explore the relationship between drug features and protein features. We evaluate our model on three established DTA benchmark datasets, Davis, Metz, and KIBA. AttentionDTA outperforms the state-of-the-art deep learning methods under different evaluation metrics. The results show that the attention-based model can effectively extract protein features related to drug information and drug features related to protein information to better predict drug target affinities. It is worth mentioning that we test our model on IC50 dataset, which provides the binding sites between drugs and proteins, to evaluate the ability of our model to locate binding sites. Finally, we visualize the attention weight to demonstrate the biological significance of the model. The source code of AttentionDTA can be downloaded from https://github.com/zhaoqichang/AttentionDTA_TCBB.


Subject(s)
Deep Learning , Drug Development , Binding Sites , Amino Acid Sequence , Benchmarking
9.
IEEE/ACM Trans Comput Biol Bioinform ; 20(3): 1943-1952, 2023.
Article in English | MEDLINE | ID: mdl-36445997

ABSTRACT

Drug discovery and drug repurposing often rely on the successful prediction of drug-target interactions (DTIs). Recent advances have shown great promise in applying deep learning to drug-target interaction prediction. One challenge in building deep learning-based models is to adequately represent drugs and proteins that encompass the fundamental local chemical environments and long-distance information among amino acids of proteins (or atoms of drugs). Another challenge is to efficiently model the intermolecular interactions between drugs and proteins, which plays vital roles in the DTIs. To this end, we propose a novel model, GIFDTI, which consists of three key components: the sequence feature extractor (CNNFormer), the global molecular feature extractor (GF), and the intermolecular interaction modeling module (IIF). Specifically, CNNFormer incorporates CNN and Transformer to capture the local patterns and encode the long-distance relationship among tokens (atoms or amino acids) in a sequence. Then, GF and IIF extract the global molecular features and the intermolecular interaction features, respectively. We evaluate GIFDTI on six realistic evaluation strategies and the results show it improves DTI prediction performance compared to state-of-the-art methods. Moreover, case studies confirm that our model can be a useful tool to accurately yield low-cost DTIs. The codes of GIFDTI are available at https://github.com/zhaoqichang/GIFDTI.


Subject(s)
Drug Development , Proteins , Proteins/chemistry , Drug Development/methods , Drug Discovery/methods , Drug Repositioning , Amino Acids
10.
Brief Bioinform ; 24(1)2023 Jan 19.
Article in English | MEDLINE | ID: mdl-36572658

ABSTRACT

Emerging evidence has proved that circular RNAs (circRNAs) are implicated in pathogenic processes. They are regarded as promising biomarkers for diagnosis due to covalently closed loop structures. As opposed to traditional experiments, computational approaches can identify circRNA-disease associations at a lower cost. Aggregating multi-source pathogenesis data helps to alleviate data sparsity and infer potential associations at the system level. The majority of computational approaches construct a homologous network using multi-source data, but they lose the heterogeneity of the data. Effective methods that use the features of multi-source data are considered as a matter of urgency. In this paper, we propose a model (CDHGNN) based on edge-weighted graph attention and heterogeneous graph neural networks for potential circRNA-disease association prediction. The circRNA network, micro RNA network, disease network and heterogeneous network are constructed based on multi-source data. To reflect association probabilities between nodes, an edge-weighted graph attention network model is designed for node features. To assign attention weights to different types of edges and learn contextual meta-path, CDHGNN infers potential circRNA-disease association based on heterogeneous neural networks. CDHGNN outperforms state-of-the-art algorithms in terms of accuracy. Edge-weighted graph attention networks and heterogeneous graph networks have both improved performance significantly. Furthermore, case studies suggest that CDHGNN is capable of identifying specific molecular associations and investigating biomolecular regulatory relationships in pathogenesis. The code of CDHGNN is freely available at https://github.com/BioinformaticsCSU/CDHGNN.


Subject(s)
MicroRNAs , RNA, Circular , RNA, Circular/genetics , Neural Networks, Computer , MicroRNAs/genetics , Algorithms , Computational Biology/methods
11.
Front Med (Lausanne) ; 9: 1015278, 2022.
Article in English | MEDLINE | ID: mdl-36465928

ABSTRACT

Increasing evidence has proved that miRNA plays a significant role in biological progress. In order to understand the etiology and mechanisms of various diseases, it is necessary to identify the essential miRNAs. However, it is time-consuming and expensive to identify essential miRNAs by using traditional biological experiments. It is critical to develop computational methods to predict potential essential miRNAs. In this study, we provided a new computational method (called PMMS) to identify essential miRNAs by using multi-head self-attention and sequences. First, PMMS computes the statistic and structure features and extracts the static feature by concatenating them. Second, PMMS extracts the deep learning original feature (BiLSTM-based feature) by using bi-directional long short-term memory (BiLSTM) and pre-miRNA sequences. In addition, we further obtained the multi-head self-attention feature (MS-based feature) based on BiLSTM-based feature and multi-head self-attention mechanism. By considering the importance of the subsequence of pre-miRNA to the static feature of miRNA, we obtained the deep learning final feature (WA-based feature) based on the weighted attention mechanism. Finally, we concatenated WA-based feature and static feature as an input to the multilayer perceptron) model to predict essential miRNAs. We conducted five-fold cross-validation to evaluate the prediction performance of PMMS. The areas under the ROC curves (AUC), the F1-score, and accuracy (ACC) are used as performance metrics. From the experimental results, PMMS obtained best prediction performances (AUC: 0.9556, F1-score: 0.9030, and ACC: 0.9097). It also outperformed other compared methods. The experimental results also illustrated that PMMS is an effective method to identify essential miRNA.

12.
J Chem Inf Model ; 62(22): 5830-5840, 2022 11 28.
Article in English | MEDLINE | ID: mdl-36245217

ABSTRACT

Pathogens producing ß-lactamase pose a great challenge to antibiotic-resistant infection treatment; thus, it is urgent to discover novel ß-lactamase inhibitors for drug development. Conventional high-throughput screening is very costly, and structure-based virtual screening is limited with mechanisms. In this study, we construct a novel multichannel deep neural network (DeepBLI) for ß-lactamase inhibitor screening, pretrained with a label reversal KIBA data set and fine-tuned on ß-lactamase-inhibitor pairs from BindingDB. First, the pairs of encoders (Conv and Att) fuse the information spatially and sequentially for both enzymes and inhibitors. Then, a co-attention module creates the connection between the inhibitor and enzyme embeddings. Finally, multichannel outputs fuse with an element-wise product and then are fed into 3-layer fully connected networks to predict interactions. Comparing the state-of-the-art methods, DeepBLI yields an AUROC of 0.9240 and an AUPRC of 0.9715, which indicates that it can identify new ß-lactamase-inhibitor interactions. To demonstrate its prediction ability, an application of DeepBLI is described to screen potential inhibitor compounds for metallo-ß-lactamase AIM-1 and repurpose rottlerin for four classes of ß-lactamase targets, showing the possibility of being a broad-spectrum inhibitor. DeepBLI provides an effective way for antibacterial drug development, contributing to antibiotic-resistant therapeutics.


Subject(s)
beta-Lactamase Inhibitors , beta-Lactamases , beta-Lactamase Inhibitors/pharmacology , Anti-Bacterial Agents/pharmacology
13.
Materials (Basel) ; 15(17)2022 Sep 01.
Article in English | MEDLINE | ID: mdl-36079462

ABSTRACT

The behavior and the mechanism of fatigue crack propagation in CrCoNi medium-entropy alloys (MEAs) with heterogeneous microstructures were investigated in this paper. After cold-rolling and recrystallization annealing at different temperatures and times, five sets of heterostructured specimens were acquired with different recrystallization levels. Then, the structure characterizations of these five sets of specimens were carried out by nanoindentation testing and electron back-scatter diffraction (EBSD) mapping. Finally, the fatigue crack propagation tests were conducted on single edge crack specimens of these different heterogeneous microstructures. The experimental results indicate that the crack propagation rates of specimens with partial recrystallization microstructures are higher than those with complete recrystallization microstructures, and the effect on fatigue crack thresholds of these specimens is the opposite. The fatigue cracks grow along the slip planes or twin boundaries in recrystallization grains (RGs), which induced crack deflections and the roughness-induced crack closure effect. For this reason, the area percentage of recrystallization and the grain size of RGs have a great effect on the value of the fatigue crack growth threshold.

14.
Materials (Basel) ; 15(14)2022 Jul 07.
Article in English | MEDLINE | ID: mdl-35888222

ABSTRACT

The fracture behavior of the Cu/Sn-3.0Ag-0.5Sn (SAC305)/Cu solder joint was investigated by conducting tensile tests with in situ X-ray micro-computed tomography (µ-CT) observation, and finite element (FE) simulation. The tensile fracture process of solder joints with a real internal defect structure was simulated and compared with the experimental results in terms of defect distribution and fracture path. Additionally, the stress distribution around the defects during the tensile process was calculated. The experimental results reveal that the pores near the intermetallic compound (IMC) layers and the flaky cracks inside the solder significantly affected the crack path. The aggregation degree of the spherical pores and the angle between the crack surface and the loading direction controlled the initiation position and propagation path of the cracks. The fracture morphology indicates that the fracture of the IMC layer was brittle, while the solder fracture exhibited ductile tearing. There are significant differences in the fracture morphology under tensile and shear loading.

15.
Plant Mol Biol ; 110(3): 219-234, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35759052

ABSTRACT

KEY MESSAGE: Identification of infection process and defense response during M. oryzae infecting Acuce. Magnaporthe oryzae is a destructive rice pathogen. Recent studies have focused on the initial infectious stage, with a few studies conducted to elucidate the characteristics of the late infectious stages. This study aims to decipher the characteristics at different stages (biotrophic, biotrophy-necrotrophy switch (BNS), and necrotrophic) between the interaction of two M. oryzae-rice combinations and investigate the resistance mechanisms of rice to M. oryzae using cytological and molecular methods. The biotrophic phase of M. oryzae-LTH compatible interaction was found to be longer than that of M. oryzae-Acuce incompatible interaction. We also found that jasmonic acid (JA) signaling plays an important role in defense by regulating antimicrobial compound accumulation in infected Acuce via a synergistic interaction of JA-salicylic acid (SA) and JA-ethylene (ET). In infected LTH, JA-ET/JA-SA showed antagonistic interaction. Ibuprofen (IBU) is a JA inhibitor. Despite the above findings, we found that exogenous JA-Ile and IBU significantly alleviated blast symptoms in infected LTH at 36 hpi (biotrophic) and 72 hpi (BNS), indicating these two-time points may be critical for managing blast disease in the compatible interaction. Conversely, IBU significantly increased blast symptoms on the infected Acuce at 36 hpi, confirming that the JA signal plays a central role in the defense response in infected Acuce. According to transcriptional analysis, the number of genes enriched in the plant hormone signal pathway was significantly higher than in other pathways. Our findings suggested that JA-mediated defense mechanism is essential in regulating Acuce resistance, particularly during the biotrophic and BNS phases.


Subject(s)
Magnaporthe , Oryza , Ascomycota , Cyclopentanes , Ethylenes/metabolism , Ibuprofen/metabolism , Magnaporthe/metabolism , Oryza/metabolism , Oxylipins , Plant Diseases/microbiology , Plant Growth Regulators/metabolism , Salicylic Acid/metabolism
16.
Brief Bioinform ; 23(3)2022 05 13.
Article in English | MEDLINE | ID: mdl-35289352

ABSTRACT

Determining drug indications is a critical part of the drug development process. However, traditional drug discovery is expensive and time-consuming. Drug repositioning aims to find potential indications for existing drugs, which is considered as an important alternative to the traditional drug discovery. In this article, we propose a multi-view learning with matrix completion (MLMC) method to predict the potential associations between drugs and diseases. Specifically, MLMC first learns the comprehensive similarity matrices from five drug similarity matrices and two disease similarity matrices based on the multi-view learning (ML) with Laplacian graph regularization, and updates the drug-disease association matrix simultaneously. Then, we introduce matrix completion (MC) to add some positive entries in original association matrix based on low-rank structure, and re-execute the multi-view learning algorithm for association prediction. At last, the prediction results of the above two operations are integrated as the final output. Evaluated by 10-fold cross-validation and de novo tests, MLMC achieves higher prediction accuracy than the current state-of-the-art methods. Moreover, case studies confirm the ability of our method in novel drug-disease association discovery. The codes of MLMC are available at https://github.com/BioinformaticsCSU/MLMC. Contact: jxwang@mail.csu.edu.cn.


Subject(s)
Computational Biology , Drug Repositioning , Algorithms , Computational Biology/methods , Drug Discovery , Drug Repositioning/methods
17.
Bioinformatics ; 38(8): 2226-2234, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35150255

ABSTRACT

MOTIVATION: Many studies have shown that microRNAs (miRNAs) play a key role in human diseases. Meanwhile, traditional experimental methods for miRNA-disease association identification are extremely costly, time-consuming and challenging. Therefore, many computational methods have been developed to predict potential associations between miRNAs and diseases. However, those methods mainly predict the existence of miRNA-disease associations, and they cannot predict the deep-level miRNA-disease association types. RESULTS: In this study, we propose a new end-to-end deep learning method (called PDMDA) to predict deep-level miRNA-disease associations with graph neural networks (GNNs) and miRNA sequence features. Based on the sequence and structural features of miRNAs, PDMDA extracts the miRNA feature representations by a fully connected network (FCN). The disease feature representations are extracted from the disease-gene network and gene-gene interaction network by GNN model. Finally, a multilayer with three fully connected layers and a softmax layer is designed to predict the final miRNA-disease association scores based on the concatenated feature representations of miRNAs and diseases. Note that PDMDA does not take the miRNA-disease association matrix as input to compute the Gaussian interaction profile similarity. We conduct three experiments based on six association type samples (including circulations, epigenetics, target, genetics, known association of which their types are unknown and unknown association samples). We conduct fivefold cross-validation validation to assess the prediction performance of PDMDA. The area under the receiver operating characteristic curve scores is used as metric. The experiment results show that PDMDA can accurately predict the deep-level miRNA-disease associations. AVAILABILITY AND IMPLEMENTATION: Data and source codes are available at https://github.com/27167199/PDMDA.


Subject(s)
MicroRNAs , Humans , MicroRNAs/genetics , Algorithms , Computational Biology/methods , Neural Networks, Computer , Software
18.
Article in English | MEDLINE | ID: mdl-32310779

ABSTRACT

A drug-drug interaction (DDI) is defined as an association between two drugs where the pharmacological effects of a drug are influenced by another drug. Positive DDIs can usually improve the therapeutic effects of patients, but negative DDIs cause the major cause of adverse drug reactions and even result in the drug withdrawal from the market and the patient death. Therefore, identifying DDIs has become a key component of the drug development and disease treatment. In this study, we propose a novel method to predict DDIs based on the integrated similarity and semi-supervised learning (DDI-IS-SL). DDI-IS-SL integrates the drug chemical, biological and phenotype data to calculate the feature similarity of drugs with the cosine similarity method. The Gaussian Interaction Profile kernel similarity of drugs is also calculated based on known DDIs. A semi-supervised learning method (the Regularized Least Squares classifier) is used to calculate the interaction possibility scores of drug-drug pairs. In terms of the 5-fold cross validation, 10-fold cross validation and de novo drug validation, DDI-IS-SL can achieve the better prediction performance than other comparative methods. In addition, the average computation time of DDI-IS-SL is shorter than that of other comparative methods. Finally, case studies further demonstrate the performance of DDI-IS-SL in practical applications.


Subject(s)
Pharmaceutical Preparations , Supervised Machine Learning , Algorithms , Drug Interactions , Humans , Least-Squares Analysis
19.
Plant Mol Biol ; 108(1-2): 15-30, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34622380

ABSTRACT

KEY MESSAGE: MoSDT1, a rice blast fungus transcription factor, is as an inducer to activate defense response through mainly mediating phosphorylated proteins in rice. Pathogen effector proteins play a dual role in infecting the host or triggering a defense response. Our previous research found a Magnaporthe oryzae effector, MoSDT1, which could activate the rice defense response when it was overexpressed in rice. However, we still know little about the mechanisms on how MoSDT1 in vivo or in vitro influences the resistance ability of rice. Our results showed that decreased ROS and increased lignin contents appeared along with significant upregulation of defense-related genes, raffinose synthesis gene, and phenylalanine ammonialyase gene. Moreover, we revealed that the contents of lignin were increased, which was in accordance with the upregulation of its precursor phenylalanine gene despite the fact that the glutamate-/thiamine-responsive genes were inhibited in MoSDT1 transgenic rice, and these indicated that MoSDT1 triggered the defense system of rice in vivo. Interestingly, in vitro studies, we further found that MoSDT1 induced the defense system by ROS synthesis, callose deposition, PR gene expression and SA/JA synthesis/signal genes using the purified prokaryotic expression system in rice plants. In addition, this defense response was confirmed to be activated by the zinc finger domain of MoSDT1 via prokaryotic expression of MoSDT1 truncated mutants in rice plants. To elucidate the regulative effects of MoSDT1 on protein phosphorylation in rice, phosphoproteome analysis was performed in both MoSDT1-transgenic and wild type  rice. We found that MoSDT1 specifically up-regulated the expression levels of a few phosphorylated proteins, which were involved in multiple functions, such as biotic/abiotic stress and growth. In addition, the motifs in these specific proteins ranked the top among the top-five conserved motifs in the MoSDT1-transgenic rice. MoSDT1 played a crucial role in enhancing rice resistance by modulating several genes and signaling pathways.


Subject(s)
Ascomycota , Disease Resistance , Fungal Proteins/metabolism , Oryza/microbiology , Phosphoproteins/metabolism , Plant Diseases/microbiology , Plant Proteins/metabolism , Transcription Factors/metabolism , Gene Expression Regulation, Plant , Hydrogen Peroxide/metabolism , Lignin/metabolism , Oryza/metabolism , Phosphorylation , Plant Diseases/immunology , Plant Leaves/metabolism , Plant Leaves/microbiology , Plant Roots/metabolism , Plant Roots/microbiology , Plants, Genetically Modified , Reactive Oxygen Species/metabolism
20.
Exp Ther Med ; 22(4): 1145, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34504590

ABSTRACT

Amyloidosis, a systemic disease characterized by the deposition of misfolded protein, is difficult to rapidly diagnose due to its wide range of symptoms. The present study reported on a case of primary amyloidosis (AL) with involvement of the gastrointestinal tract, mesentery and omentum in a 66-year-old male presenting with recurrent diarrhoea and abdominal distension. Oesophagogastroduodenoscopy and enteroscopy revealed multiple gastric ulcers and multiple protuberant lesions in the colon. Laparotomy indicated multiple nodules in the mesentery of the small intestine. Contrast-enhanced CT revealed dilation of the small bowel with pneumatosis intestinalis and positive Congo red staining of gastric mucosa and mesentery biopsy specimens confirmed amyloid deposition. Therefore, the patient was diagnosed with AL. In this case, the clinical manifestation of mesentery amyloidosis was multiple nodules and extensive peritoneal adhesions, which, to the best of our knowledge, has not been reported by any previous study.

SELECTION OF CITATIONS
SEARCH DETAIL
...