Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
BMC Bioinformatics ; 25(1): 48, 2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38291364

ABSTRACT

BACKGROUND: The Drug-Target Interaction (DTI) prediction uses a drug molecule and a protein sequence as inputs to predict the binding affinity value. In recent years, deep learning-based models have gotten more attention. These methods have two modules: the feature extraction module and the task prediction module. In most deep learning-based approaches, a simple task prediction loss (i.e., categorical cross entropy for the classification task and mean squared error for the regression task) is used to learn the model. In machine learning, contrastive-based loss functions are developed to learn more discriminative feature space. In a deep learning-based model, extracting more discriminative feature space leads to performance improvement for the task prediction module. RESULTS: In this paper, we have used multimodal knowledge as input and proposed an attention-based fusion technique to combine this knowledge. Also, we investigate how utilizing contrastive loss function along the task prediction loss could help the approach to learn a more powerful model. Four contrastive loss functions are considered: (1) max-margin contrastive loss function, (2) triplet loss function, (3) Multi-class N-pair Loss Objective, and (4) NT-Xent loss function. The proposed model is evaluated using four well-known datasets: Wang et al. dataset, Luo's dataset, Davis, and KIBA datasets. CONCLUSIONS: Accordingly, after reviewing the state-of-the-art methods, we developed a multimodal feature extraction network by combining protein sequences and drug molecules, along with protein-protein interaction networks and drug-drug interaction networks. The results show it performs significantly better than the comparable state-of-the-art approaches.


Subject(s)
Knowledge , Machine Learning , Amino Acid Sequence , Drug Interactions , Entropy
2.
J Chem Inf Model ; 64(7): 2577-2585, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38514966

ABSTRACT

Drug synergy prediction plays a vital role in cancer treatment. Because experimental approaches are labor-intensive and expensive, computational-based approaches get more attention. There are two types of computational methods for drug synergy prediction: feature-based and similarity-based. In feature-based methods, the main focus is to extract more discriminative features from drug pairs and cell lines to pass to the task predictor. In similarity-based methods, the similarities among all drugs and cell lines are utilized as features and fed into the task predictor. In this work, a novel approach, called CFSSynergy, that combines these two viewpoints is proposed. First, a discriminative representation is extracted for paired drugs and cell lines as input. We have utilized transformer-based architecture for drugs. For cell lines, we have created a similarity matrix between proteins using the Node2Vec algorithm. Then, the new cell line representation is computed by multiplying the protein-protein similarity matrix and the initial cell line representation. Next, we compute the similarity between unique drugs and unique cells using the learned representation for paired drugs and cell lines. Then, we compute a new representation for paired drugs and cell lines based on the similarity-based features and the learned features. Finally, these features are fed to XGBoost as a task predictor. Two well-known data sets were used to evaluate the performance of our proposed method: DrugCombDB and OncologyScreen. The CFSSynergy approach consistently outperformed existing methods in comparative evaluations. This substantiates the efficacy of our approach in capturing complex synergistic interactions between drugs and cell lines, setting it apart from conventional similarity-based or feature-based methods.


Subject(s)
Algorithms , Computational Biology , Computational Biology/methods , Cell Line
3.
Mol Divers ; 27(1): 249-261, 2023 Feb.
Article in English | MEDLINE | ID: mdl-35438428

ABSTRACT

Caspases (cysteine-aspartic proteases) play critical roles in inflammation and the programming of cell death in the form of necroptosis, apoptosis, and pyroptosis. The name of these enzymes has been chosen in accordance with their cysteine protease activity. They act as cysteines in nucleophilically active sites to attack and cleave target proteins in the aspartic acid and amino acid C-terminal. Based on the substrate's structure and the specificity, the physiological activity of caspases is divided. However, in apoptosis, the division of caspases into initiating caspases (caspase 2, 8, 9, and 10) and executive caspases (caspase 3, 6, and 7) is essential. The present study aimed to perform Proteochemometrics Modeling to generalize the data on caspases, which could predict ligand and protein interactions. In this study, we employed protein and ligand descriptors. Moreover, protein descriptors were computed using the Protr R package, while PADEL-Descriptor was employed for the computation of ligand descriptors. In addition, NCA (Neighborhood Component Analyses) was used for descriptor selection, and SVR, decision tree, and ensemble methods were utilized for the proteochemometrics modeling. This study shows that the ensemble model demonstrates superior performance compared with other models in terms of R2, Q2, and RMSE criteria.


Subject(s)
Apoptosis , Caspases , Caspases/chemistry , Caspases/metabolism , Ligands , Protein Isoforms , Catalytic Domain
4.
Bioinformatics ; 36(17): 4633-4642, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32462178

ABSTRACT

MOTIVATION: An essential part of drug discovery is the accurate prediction of the binding affinity of new compound-protein pairs. Most of the standard computational methods assume that compounds or proteins of the test data are observed during the training phase. However, in real-world situations, the test and training data are sampled from different domains with different distributions. To cope with this challenge, we propose a deep learning-based approach that consists of three steps. In the first step, the training encoder network learns a novel representation of compounds and proteins. To this end, we combine convolutional layers and long-short-term memory layers so that the occurrence patterns of local substructures through a protein and a compound sequence are learned. Also, to encode the interaction strength of the protein and compound substructures, we propose a two-sided attention mechanism. In the second phase, to deal with the different distributions of the training and test domains, a feature encoder network is learned for the test domain by utilizing an adversarial domain adaptation approach. In the third phase, the learned test encoder network is applied to new compound-protein pairs to predict their binding affinity. RESULTS: To evaluate the proposed approach, we applied it to KIBA, Davis and BindingDB datasets. The results show that the proposed method learns a more reliable model for the test domain in more challenging situations. AVAILABILITY AND IMPLEMENTATION: https://github.com/LBBSoft/DeepCDA.


Subject(s)
Neural Networks, Computer , Proteins , Drug Discovery
5.
J Biomol Struct Dyn ; : 1-10, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-38084744

ABSTRACT

Virtual screening has emerged as a valuable computational tool for predicting compound-protein interactions, offering a cost-effective and rapid approach to identifying potential candidate drug molecules. Current machine learning-based methods rely on molecular structures and their relationship in the network. The former utilizes information such as amino acid sequences and chemical structures, while the latter leverages interaction network data, such as protein-protein interactions, drug-disease interactions, and protein-disease interactions. However, there has been limited exploration of integrating molecular information with interaction networks. This study presents DeepCompoundNet, a deep learning-based model that integrates protein features, drug properties, and diverse interaction data to predict chemical-protein interactions. DeepCompoundNet outperforms state-of-the-art methods for compound-protein interaction prediction, as demonstrated through performance evaluations. Our findings highlight the complementary nature of multiple interaction data, extending beyond amino acid sequence homology and chemical structure similarity. Moreover, our model's analysis confirms that DeepCompoundNet gets higher performance in predicting interactions between proteins and chemicals not observed in the training samples.Communicated by Ramaswamy H. Sarma.

6.
Curr Med Chem ; 28(11): 2100-2113, 2021.
Article in English | MEDLINE | ID: mdl-32895036

ABSTRACT

Drug-target Interactions (DTIs) prediction plays a central role in drug discovery. Computational methods in DTIs prediction have gained more attention because carrying out in vitro and in vivo experiments on a large scale is costly and time-consuming. Machine learning methods, especially deep learning, are widely applied to DTIs prediction. In this study, the main goal is to provide a comprehensive overview of deep learning-based DTIs prediction approaches. Here, we investigate the existing approaches from multiple perspectives. We explore these approaches to find out which deep network architectures are utilized to extract features from drug compound and protein sequences. Also, the advantages and limitations of each architecture are analyzed and compared. Moreover, we explore the process of how to combine descriptors for drug and protein features. Likewise, a list of datasets that are commonly used in DTIs prediction is investigated. Finally, current challenges are discussed and a short future outlook of deep learning in DTI prediction is given.


Subject(s)
Deep Learning , Pharmaceutical Preparations , Amino Acid Sequence , Drug Development , Humans , Proteins
SELECTION OF CITATIONS
SEARCH DETAIL