Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
1.
Annu Rev Pharmacol Toxicol ; 64: 507-526, 2024 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-37722721

RESUMO

Recent advances in chemical, molecular, and genetic approaches have provided us with an unprecedented capacity to identify drug-target interactions across the whole proteome and genome. Meanwhile, rapid developments of single-cell and spatial omics technologies are revolutionizing our understanding of the molecular architecture of biological systems. However, a significant gap remains in how we align our understanding of drug actions, traditionally based on molecular affinities, with the in vivo cellular and spatial tissue heterogeneity revealed by these newer techniques. Here, we review state-of-the-art methods for profiling drug-target interactions and emerging multiomics tools to delineate the tissue heterogeneity at single-cell resolution. Highlighting the recent technical advances enabling high-resolution, multiplexable in situ small-molecule drug imaging (clearing-assisted tissue click chemistry, or CATCH), we foresee the integration of single-cell and spatial omics platforms, data, and concepts into the future framework of defining and understanding in vivo drug-target interactions and mechanisms of actions.


Assuntos
Sistemas de Liberação de Medicamentos , Proteoma , Humanos , Tecnologia
2.
Methods ; 224: 79-92, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38430967

RESUMO

The identification of drug-target interactions (DTI) is a valuable step in the drug discovery and repositioning process. However, traditional laboratory experiments are time-consuming and expensive. Computational methods have streamlined research to determine DTIs. The application of deep learning methods has significantly improved the prediction performance for DTIs. Modern deep learning methods can leverage multiple sources of information, including sequence data that contains biological structural information, and interaction data. While useful, these methods cannot be effectively applied to each type of information individually (e.g., chemical structure and interaction network) and do not take into account the specificity of DTI data such as low- or zero-interaction biological entities. To overcome these limitations, we propose a method called MFA-DTI (Multi-feature Fusion Adopted framework for DTI). MFA-DTI consists of three modules: an interaction graph learning module that processes the interaction network to generate interaction vectors, a chemical structure learning module that extracts features from the chemical structure, and a fusion module that combines these features for the final prediction. To validate the performance of MFA-DTI, we conducted experiments on six public datasets under different settings. The results indicate that the proposed method is highly effective in various settings and outperforms state-of-the-art methods.


Assuntos
Descoberta de Drogas , Laboratórios , Interações Medicamentosas
3.
BMC Bioinformatics ; 25(1): 10, 2024 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-38177981

RESUMO

Examining potential drug-target interactions (DTIs) is a pivotal component of drug discovery and repurposing. Recently, there has been a significant rise in the use of computational techniques to predict DTIs. Nevertheless, previous investigations have predominantly concentrated on assessing either the connections between nodes or the consistency of the network's topological structure in isolation. Such one-sided approaches could severely hinder the accuracy of DTI predictions. In this study, we propose a novel method called TTGCN, which combines heterogeneous graph convolutional neural networks (GCN) and graph attention networks (GAT) to address the task of DTI prediction. TTGCN employs a two-tiered feature learning strategy, utilizing GAT and residual GCN (R-GCN) to extract drug and target embeddings from the diverse network, respectively. These drug and target embeddings are then fused through a mean-pooling layer. Finally, we employ an inductive matrix completion technique to forecast DTIs while preserving the network's node connectivity and topological structure. Our approach demonstrates superior performance in terms of area under the curve and area under the precision-recall curve in experimental comparisons, highlighting its significant advantages in predicting DTIs. Furthermore, case studies provide additional evidence of its ability to identify potential DTIs.


Assuntos
Descoberta de Drogas , Aprendizagem , Interações Medicamentosas , Redes Neurais de Computação
4.
Brief Bioinform ; 23(2)2022 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-35043158

RESUMO

Drug-target interactions (DTIs) prediction research presents important significance for promoting the development of modern medicine and pharmacology. Traditional biochemical experiments for DTIs prediction confront the challenges including long time period, high cost and high failure rate, and finally leading to a low-drug productivity. Chemogenomic-based computational methods can realize high-throughput prediction. In this study, we develop a deep collaborative filtering prediction model with multiembeddings, named DCFME (deep collaborative filtering prediction model with multiembeddings), which can jointly utilize multiple feature information from multiembeddings. Two different representation learning algorithms are first employed to extract heterogeneous network features. DCFME uses the generated low-dimensional dense vectors as input, and then simulates the drug-target relationship from the perspective of both couplings and heterogeneity. In addition, the model employs focal loss that concentrates the loss on sparse and hard samples in the training process. Comparative experiments with five baseline methods show that DCFME achieves more significant performance improvement on sparse datasets. Moreover, the model has better robustness and generalization capacity under several harder prediction scenarios.


Assuntos
Algoritmos , Desenvolvimento de Medicamentos , Desenvolvimento de Medicamentos/métodos
5.
Brief Bioinform ; 23(3)2022 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-35262678

RESUMO

Accurate prediction of drug-target interactions (DTIs) can reduce the cost and time of drug repositioning and drug discovery. Many current methods integrate information from multiple data sources of drug and target to improve DTIs prediction accuracy. However, these methods do not consider the complex relationship between different data sources. In this study, we propose a novel computational framework, called MccDTI, to predict the potential DTIs by multiview network embedding, which can integrate the heterogenous information of drug and target. MccDTI learns high-quality low-dimensional representations of drug and target by preserving the consistent and complementary information between multiview networks. Then MccDTI adopts matrix completion scheme for DTIs prediction based on drug and target representations. Experimental results on two datasets show that the prediction accuracy of MccDTI outperforms four state-of-the-art methods for DTIs prediction. Moreover, literature verification for DTIs prediction shows that MccDTI can predict the reliable potential DTIs. These results indicate that MccDTI can provide a powerful tool to predict new DTIs and accelerate drug discovery. The code and data are available at: https://github.com/ShangCS/MccDTI.


Assuntos
Desenvolvimento de Medicamentos , Reposicionamento de Medicamentos , Descoberta de Drogas , Interações Medicamentosas
6.
Brief Bioinform ; 23(2)2022 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-35134117

RESUMO

Targeted drugs have been applied to the treatment of cancer on a large scale, and some patients have certain therapeutic effects. It is a time-consuming task to detect drug-target interactions (DTIs) through biochemical experiments. At present, machine learning (ML) has been widely applied in large-scale drug screening. However, there are few methods for multiple information fusion. We propose a multiple kernel-based triple collaborative matrix factorization (MK-TCMF) method to predict DTIs. The multiple kernel matrices (contain chemical, biological and clinical information) are integrated via multi-kernel learning (MKL) algorithm. And the original adjacency matrix of DTIs could be decomposed into three matrices, including the latent feature matrix of the drug space, latent feature matrix of the target space and the bi-projection matrix (used to join the two feature spaces). To obtain better prediction performance, MKL algorithm can regulate the weight of each kernel matrix according to the prediction error. The weights of drug side-effects and target sequence are the highest. Compared with other computational methods, our model has better performance on four test data sets.


Assuntos
Algoritmos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Interações Medicamentosas , Humanos , Aprendizado de Máquina
7.
Int J Mol Sci ; 25(10)2024 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-38791165

RESUMO

Studying drug-target interactions (DTIs) is the foundational and crucial phase in drug discovery. Biochemical experiments, while being the most reliable method for determining drug-target affinity (DTA), are time-consuming and costly, making it challenging to meet the current demands for swift and efficient drug development. Consequently, computational DTA prediction methods have emerged as indispensable tools for this research. In this article, we propose a novel deep learning algorithm named GRA-DTA, for DTA prediction. Specifically, we introduce Bidirectional Gated Recurrent Unit (BiGRU) combined with a soft attention mechanism to learn target representations. We employ Graph Sample and Aggregate (GraphSAGE) to learn drug representation, especially to distinguish the different features of drug and target representations and their dimensional contributions. We merge drug and target representations by an attention neural network (ANN) to learn drug-target pair representations, which are fed into fully connected layers to yield predictive DTA. The experimental results showed that GRA-DTA achieved mean squared error of 0.142 and 0.225 and concordance index reached 0.897 and 0.890 on the benchmark datasets KIBA and Davis, respectively, surpassing the most state-of-the-art DTA prediction algorithms.


Assuntos
Algoritmos , Aprendizado Profundo , Redes Neurais de Computação , Descoberta de Drogas/métodos , Humanos , Preparações Farmacêuticas/química
8.
BMC Bioinformatics ; 24(1): 38, 2023 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-36737694

RESUMO

BACKGROUND: The experimental verification of a drug discovery process is expensive and time-consuming. Therefore, efficiently and effectively identifying drug-target interactions (DTIs) has been the focus of research. At present, many machine learning algorithms are used for predicting DTIs. The key idea is to train the classifier using an existing DTI to predict a new or unknown DTI. However, there are various challenges, such as class imbalance and the parameter optimization of many classifiers, that need to be solved before an optimal DTI model is developed. METHODS: In this study, we propose a framework called SSELM-neg for DTI prediction, in which we use a screening approach to choose high-quality negative samples and a spherical search approach to optimize the parameters of the extreme learning machine. RESULTS: The results demonstrated that the proposed technique outperformed other state-of-the-art methods in 10-fold cross-validation experiments in terms of the area under the receiver operating characteristic curve (0.986, 0.993, 0.988, and 0.969) and AUPR (0.982, 0.991, 0.982, and 0.946) for the enzyme dataset, G-protein coupled receptor dataset, ion channel dataset, and nuclear receptor dataset, respectively. CONCLUSION: The screening approach produced high-quality negative samples with the same number of positive samples, which solved the class imbalance problem. We optimized an extreme learning machine using a spherical search approach to identify DTIs. Therefore, our models performed better than other state-of-the-art methods.


Assuntos
Desenvolvimento de Medicamentos , Descoberta de Drogas , Descoberta de Drogas/métodos , Aprendizado de Máquina , Algoritmos , Interações Medicamentosas
9.
BMC Bioinformatics ; 24(1): 375, 2023 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-37789278

RESUMO

BACKGROUND: Identifying drug-target interactions (DTIs) plays a key role in drug development. Traditional wet experiments to identify DTIs are costly and time consuming. Effective computational methods to predict DTIs are useful to speed up the process of drug discovery. A variety of non-negativity matrix factorization based methods are proposed to predict DTIs, but most of them overlooked the sparsity of feature matrices and the convergence of adopted matrix factorization algorithms, therefore their performances can be further improved. RESULTS: In order to predict DTIs more accurately, we propose a novel method iPALM-DLMF. iPALM-DLMF models DTIs prediction as a problem of non-negative matrix factorization with graph dual regularization terms and [Formula: see text] norm regularization terms. The graph dual regularization terms are used to integrate the information from the drug similarity matrix and the target similarity matrix, and [Formula: see text] norm regularization terms are used to ensure the sparsity of the feature matrices obtained by non-negative matrix factorization. To solve the model, iPALM-DLMF adopts non-negative double singular value decomposition to initialize the nonnegative matrix factorization, and an inertial Proximal Alternating Linearized Minimization iterating process, which has been proved to converge to a KKT point, to obtain the final result of the matrix factorization. Extensive experimental results show that iPALM-DLMF has better performance than other state-of-the-art methods. In case studies, in 50 highest-scoring proteins targeted by the drug gabapentin predicted by iPALM-DLMF, 46 have been validated, and in 50 highest-scoring drugs targeting prostaglandin-endoperoxide synthase 2 predicted by iPALM-DLMF, 47 have been validated.


Assuntos
Algoritmos , Descoberta de Drogas , Interações Medicamentosas , Descoberta de Drogas/métodos , Proteínas/química
10.
Brief Bioinform ; 22(2): 1679-1693, 2021 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-32065227

RESUMO

Complex biological systems are traditionally modelled as graphs of interconnected biological entities. These graphs, i.e. biological knowledge graphs, are then processed using graph exploratory approaches to perform different types of analytical and predictive tasks. Despite the high predictive accuracy of these approaches, they have limited scalability due to their dependency on time-consuming path exploratory procedures. In recent years, owing to the rapid advances of computational technologies, new approaches for modelling graphs and mining them with high accuracy and scalability have emerged. These approaches, i.e. knowledge graph embedding (KGE) models, operate by learning low-rank vector representations of graph nodes and edges that preserve the graph's inherent structure. These approaches were used to analyse knowledge graphs from different domains where they showed superior performance and accuracy compared to previous graph exploratory approaches. In this work, we study this class of models in the context of biological knowledge graphs and their different applications. We then show how KGE models can be a natural fit for representing complex biological knowledge modelled as graphs. We also discuss their predictive and analytical capabilities in different biology applications. In this regard, we present two example case studies that demonstrate the capabilities of KGE models: prediction of drug-target interactions and polypharmacy side effects. Finally, we analyse different practical considerations for KGEs, and we discuss possible opportunities and challenges related to adopting them for modelling biological systems.


Assuntos
Biologia Computacional/métodos , Redes Neurais de Computação , Algoritmos , Interações Medicamentosas , Humanos , Aprendizado de Máquina
11.
Methods ; 206: 101-107, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36058415

RESUMO

Determining the interaction of drug and target plays a key role in the process of drug development and discovery. The calculation methods can predict new interactions and speed up the process of drug development. In recent studies, the network-based approaches have been proposed to predict drug-target interactions. However, these methods cannot fully utilize the node information from heterogeneous networks. Therefore, we propose a method based on heterogeneous graph convolutional neural network for drug-target interaction prediction, GCHN-DTI (Predicting drug-target interactions by graph convolution on heterogeneous net-works), to predict potential DTIs. GCHN-DTI integrates network information from drug-target interactions, drug-drug interactions, drug-similarities, target-target interactions, and target-similarities. Then, the graph convolution operation is used in the heterogeneous network to obtain the node embedding of the drugs and the targets. Furthermore, we incorporate an attention mechanism between graph convolutional layers to combine node embedding from each layer. Finally, the drug-target interaction score is predicted based on the node embedding of the drugs and the targets. Our model uses fewer network types and achieves higher prediction performance. In addition, the prediction performance of the model will be significantly improved on the dataset with a higher proportion of positive samples. The experimental evaluations show that GCHN-DTI outperforms several state-of-the-art prediction methods.


Assuntos
Desenvolvimento de Medicamentos , Redes Neurais de Computação , Desenvolvimento de Medicamentos/métodos , Interações Medicamentosas
12.
Methods ; 198: 19-31, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34737033

RESUMO

Computational prediction of drug-target interactions (DTIs) is of particular importance in the process of drug repositioning because of its efficiency in selecting potential candidates for DTIs. A variety of computational methods for predicting DTIs have been proposed over the past decade. Our interest is which methods or techniques are the most advantageous for increasing prediction accuracy. This article provides a comprehensive overview of network-based, machine learning, and integrated DTI prediction methods. The network-based methods handle a DTI network along with drug and target similarities in a matrix form and apply graph-theoretic algorithms to identify new DTIs. Machine learning methods use known DTIs and the features of drugs and target proteins as training data to build a predictive model. Integrated methods combine these two techniques. We assessed the prediction performance of the selected state-of-the-art methods using two different benchmark datasets. Our experimental results demonstrate that the integrated methods outperform the others in general. Some previous methods showed low accuracy on predicting interactions of unknown drugs which do not exist in the training dataset. Combining similarity matrices from multiple features by data fusion was not beneficial in increasing prediction accuracy. Finally, we analyzed future directions for further improvements in DTI predictions.


Assuntos
Algoritmos , Aprendizado de Máquina , Interações Medicamentosas , Reposicionamento de Medicamentos , Proteínas/metabolismo
13.
Mol Divers ; 27(3): 1333-1343, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35871213

RESUMO

Drug-target interaction is crucial in the discovery of new drugs. Computational methods can be used to identify new drug-target interactions at low costs and with reasonable accuracy. Recent studies pay more attention to machine-learning methods, ranging from matrix factorization to deep learning, in the DTI prediction. Since the interaction matrix is often extremely sparse, DTI prediction performance is significantly decreased with matrix factorization-based methods. Therefore, some matrix factorization methods utilize side information to address both the sparsity issue of the interaction matrix and the cold-start issue. By combining matrix factorization and autoencoders, we propose a hybrid DTI prediction model that simultaneously learn the hidden factors of drugs and targets from their side information and interaction matrix. The proposed method is composed of two steps: the pre-processing of the interaction matrix, and the hybrid model. We leverage the similarity matrices of both drugs and targets to address the sparsity problem of the interaction matrix. The comparison of our approach against other algorithms on the same reference datasets has shown good results regarding area under receiver operating characteristic curve and the area under precision-recall curve. More specifically, experimental results achieve high accuracy on golden standard datasets (e.g., Nuclear Receptors, GPCRs, Ion Channels, and Enzymes) when performed with five repetitions of tenfold cross-validation. Display graphical of the hybrid model of Matrix Factorization with Denoising Autoencoders with the help side information of drugs and targets for Prediction of Drug-Target Interactions.


Assuntos
Algoritmos , Aprendizado de Máquina , Interações Medicamentosas , Projetos de Pesquisa , Curva ROC
14.
Int J Mol Sci ; 24(18)2023 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-37762364

RESUMO

Drug-target interactions (DTIs) are considered a crucial component of drug design and drug discovery. To date, many computational methods were developed for drug-target interactions, but they are insufficiently informative for accurately predicting DTIs due to the lack of experimentally verified negative datasets, inaccurate molecular feature representation, and ineffective DTI classifiers. Therefore, we address the limitations of randomly selecting negative DTI data from unknown drug-target pairs by establishing two experimentally validated datasets and propose a capsule network-based framework called CapBM-DTI to capture hierarchical relationships of drugs and targets, which adopts pre-trained bidirectional encoder representations from transformers (BERT) for contextual sequence feature extraction from target proteins through transfer learning and the message-passing neural network (MPNN) for the 2-D graph feature extraction of compounds to accurately and robustly identify drug-target interactions. We compared the performance of CapBM-DTI with state-of-the-art methods using four experimentally validated DTI datasets of different sizes, including human (Homo sapiens) and worm (Caenorhabditis elegans) species datasets, as well as three subsets (new compounds, new proteins, and new pairs). Our results demonstrate that the proposed model achieved robust performance and powerful generalization ability in all experiments. The case study on treating COVID-19 demonstrates the applicability of the model in virtual screening.

15.
Molecules ; 28(18)2023 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-37764321

RESUMO

The prediction of drug-target interaction (DTI) is crucial to drug discovery. Although the interactions between the drug and target can be accurately verified by traditional biochemical experiments, the determination of DTI through biochemical experiments is a time-consuming, laborious, and expensive process. Therefore, we propose a learning-based framework named BG-DTI for drug-target interaction prediction. Our model combines two main approaches based on biological features and heterogeneous networks to identify interactions between drugs and targets. First, we extract original features from the sequence to encode each drug and target. Later, we further consider the relationships among various biological entities by constructing drug-drug similarity networks and target-target similarity networks. Furthermore, a graph convolutional network and a graph attention network in the graph representation learning module help us learn the features representation of drugs and targets. After obtaining the features from graph representation learning modules, these features are combined into fusion descriptors for drug-target pairs. Finally, we send the fusion descriptors and labels to a random forest classifier for predicting DTI. The evaluation results show that BG-DTI achieves an average AUC of 0.938 and an average AUPR of 0.930, which is better than those of five existing state-of-the-art methods. We believe that BG-DTI can facilitate the development of drug discovery or drug repurposing.


Assuntos
Descoberta de Drogas , Aprendizagem , Reposicionamento de Medicamentos , Sistemas de Liberação de Medicamentos , Algoritmo Florestas Aleatórias
16.
BMC Bioinformatics ; 23(1): 245, 2022 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-35729494

RESUMO

BACKGROUND: Drug-target interactions (DTIs) are critical for drug repurposing and elucidation of drug mechanisms, and are manually curated by large databases, such as ChEMBL, BindingDB, DrugBank and DrugTargetCommons. However, the number of curated articles likely constitutes only a fraction of all the articles that contain experimentally determined DTIs. Finding such articles and extracting the experimental information is a challenging task, and there is a pressing need for systematic approaches to assist the curation of DTIs. To this end, we applied Bidirectional Encoder Representations from Transformers (BERT) to identify such articles. Because DTI data intimately depends on the type of assays used to generate it, we also aimed to incorporate functions to predict the assay format. RESULTS: Our novel method identified 0.6 million articles (along with drug and protein information) which are not previously included in public DTI databases. Using 10-fold cross-validation, we obtained ~ 99% accuracy for identifying articles containing quantitative drug-target profiles. The F1 micro for the prediction of assay format is 88%, which leaves room for improvement in future studies. CONCLUSION: The BERT model in this study is robust and the proposed pipeline can be used to identify previously overlooked articles containing quantitative DTIs. Overall, our method provides a significant advancement in machine-assisted DTI extraction and curation. We expect it to be a useful addition to drug mechanism discovery and repurposing.


Assuntos
Reposicionamento de Medicamentos , Proteínas , Bases de Dados Factuais , Interações Medicamentosas , Proteínas/metabolismo , PubMed
17.
Brief Bioinform ; 21(1): 211-220, 2020 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-30566623

RESUMO

Knowledge of the full target space of drugs (or drug-like compounds) provides important insights into the potential therapeutic use of the agents to modulate or avoid their various on- and off-targets in drug discovery and precision medicine. However, there is a lack of consolidated databases and associated data exploration tools that allow for systematic profiling of drug target-binding potencies of both approved and investigational agents using a network-centric approach. We recently initiated a community-driven platform, Drug Target Commons (DTC), which is an open-data crowdsourcing platform designed to improve the management, reproducibility and extended use of compound-target bioactivity data for drug discovery and repurposing, as well as target identification applications. In this work, we demonstrate an integrated use of the rich bioactivity data from DTC and related drug databases using Drug Target Profiler (DTP), an open-source software and web tool for interactive exploration of drug-target interaction networks. DTP was designed for network-centric modeling of mode-of-action of multi-targeting anticancer compounds, especially for precision oncology applications. DTP enables users to construct an interaction network based on integrated bioactivity data across selected chemical compounds and their protein targets, further customizable using various visualization and filtering options, as well as cross-links to several drug and protein databases to provide comprehensive information of the network nodes and interactions. We demonstrate here the operation of the DTP tool and its unique features by several use cases related to both drug discovery and drug repurposing applications, using examples of anticancer drugs with shared target profiles. DTP is freely accessible at http://drugtargetprofiler.fimm.fi/.

18.
Molecules ; 27(16)2022 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-36014371

RESUMO

Nowadays, drug-target interactions (DTIs) prediction is a fundamental part of drug repositioning. However, on the one hand, drug-target interactions prediction models usually consider drugs or targets information, which ignore prior knowledge between drugs and targets. On the other hand, models incorporating priori knowledge cannot make interactions prediction for under-studied drugs and targets. Hence, this article proposes a novel dual-network integrated logistic matrix factorization DTIs prediction scheme (Ro-DNILMF) via a knowledge graph embedding approach. This model adds prior knowledge as input data into the prediction model and inherits the advantages of the DNILMF model, which can predict under-studied drug-target interactions. Firstly, a knowledge graph embedding model based on relational rotation (RotatE) is trained to construct the interaction adjacency matrix and integrate prior knowledge. Secondly, a dual-network integrated logistic matrix factorization prediction model (DNILMF) is used to predict new drugs and targets. Finally, several experiments conducted on the public datasets are used to demonstrate that the proposed method outperforms the single base-line model and some mainstream methods on efficiency.


Assuntos
Reposicionamento de Medicamentos , Reconhecimento Automatizado de Padrão , Algoritmos , Sistemas de Liberação de Medicamentos , Interações Medicamentosas , Modelos Logísticos
19.
BMC Bioinformatics ; 22(1): 567, 2021 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-34836495

RESUMO

BACKGROUND: Experimental verification of a drug discovery process is expensive and time-consuming. Therefore, recently, the demand to more efficiently and effectively identify drug-target interactions (DTIs) has intensified. RESULTS: We treat the prediction of DTIs as a ranking problem and propose a neural network architecture, NeuRank, to address it. Also, we assume that similar drug compounds are likely to interact with similar target proteins. Thus, in our model, we add drug and target similarities, which are very effective at improving the prediction of DTIs. Then, we develop NeuRank from a point-wise to a pair-wise, and further to list-wise model. CONCLUSION: Finally, results from extensive experiments on five public data sets (DrugBank, Enzymes, Ion Channels, G-Protein-Coupled Receptors, and Nuclear Receptors) show that, in identifying DTIs, our models achieve better performance than other state-of-the-art methods.


Assuntos
Desenvolvimento de Medicamentos , Preparações Farmacêuticas , Descoberta de Drogas , Redes Neurais de Computação , Receptores Citoplasmáticos e Nucleares
20.
BMC Bioinformatics ; 22(1): 497, 2021 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-34649499

RESUMO

BACKGROUND: The study of drug-target interactions (DTIs) affinity plays an important role in safety assessment and pharmacology. Currently, quantitative structure-activity relationship (QSAR) and molecular docking (MD) are most common methods in research of DTIs affinity. However, they often built for a specific target or several targets, and most QSAR and MD methods were based either on structure of drug molecules or on structure of receptors with low accuracy and small scope of application. How to construct quantitative prediction models with high accuracy and wide applicability remains a challenge. To this end, this paper screened molecular descriptors based on molecular vibrations and took molecule-target as a whole system to construct prediction models with high accuracy-wide applicability based on dissociation constant (Kd) and concentration for 50% of maximal effect (EC50), and to provide reference for quantifying affinity of DTIs. RESULTS: After comprehensive comparison, the results showed that RF models are optimal models to analyze and predict DTIs affinity with coefficients of determination (R2) are all greater than 0.94. Compared to the quantitative models reported in literatures, the RF models developed in this paper have higher accuracy and wide applicability. In addition, E-state molecular descriptors associated with molecular vibrations and normalized Moreau-Broto autocorrelation (G3), Moran autocorrelation (G4), transition-distribution (G7) protein descriptors are of higher importance in the quantification of DTIs. CONCLUSION: Through screening molecular descriptors based on molecular vibrations and taking molecule-target as whole system, we obtained optimal models based on RF with more accurate-widely applicable, which indicated that selection of molecular descriptors associated with molecular vibrations and the use of molecular-target as whole system are reliable methods for improving performance of models. It can provide reference for quantifying affinity of DTIs.


Assuntos
Preparações Farmacêuticas , Vibração , Ligantes , Simulação de Acoplamento Molecular , Relação Quantitativa Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa