Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Bioinformatics ; 40(5)2024 May 02.
Article in English | MEDLINE | ID: mdl-38648052

ABSTRACT

MOTIVATION: Accurate inference of potential drug-protein interactions (DPIs) aids in understanding drug mechanisms and developing novel treatments. Existing deep learning models, however, struggle with accurate node representation in DPI prediction, limiting their performance. RESULTS: We propose a new computational framework that integrates global and local features of nodes in the drug-protein bipartite graph for efficient DPI inference. Initially, we employ pre-trained models to acquire fundamental knowledge of drugs and proteins and to determine their initial features. Subsequently, the MinHash and HyperLogLog algorithms are utilized to estimate the similarity and set cardinality between drug and protein subgraphs, serving as their local features. Then, an energy-constrained diffusion mechanism is integrated into the transformer architecture, capturing interdependencies between nodes in the drug-protein bipartite graph and extracting their global features. Finally, we fuse the local and global features of nodes and employ multilayer perceptrons to predict the likelihood of potential DPIs. A comprehensive and precise node representation guarantees efficient prediction of unknown DPIs by the model. Various experiments validate the accuracy and reliability of our model, with molecular docking results revealing its capability to identify potential DPIs not present in existing databases. This approach is expected to offer valuable insights for furthering drug repurposing and personalized medicine research. AVAILABILITY AND IMPLEMENTATION: Our code and data are accessible at: https://github.com/ZZCrazy00/DPI.


Subject(s)
Algorithms , Molecular Docking Simulation , Proteins , Proteins/chemistry , Proteins/metabolism , Pharmaceutical Preparations/chemistry , Pharmaceutical Preparations/metabolism , Computational Biology/methods , Deep Learning
2.
Brief Bioinform ; 25(2)2024 Jan 22.
Article in English | MEDLINE | ID: mdl-38446739

ABSTRACT

Antimicrobial peptides (AMPs), short peptides with diverse functions, effectively target and combat various organisms. The widespread misuse of chemical antibiotics has led to increasing microbial resistance. Due to their low drug resistance and toxicity, AMPs are considered promising substitutes for traditional antibiotics. While existing deep learning technology enhances AMP generation, it also presents certain challenges. Firstly, AMP generation overlooks the complex interdependencies among amino acids. Secondly, current models fail to integrate crucial tasks like screening, attribute prediction and iterative optimization. Consequently, we develop a integrated deep learning framework, Diff-AMP, that automates AMP generation, identification, attribute prediction and iterative optimization. We innovatively integrate kinetic diffusion and attention mechanisms into the reinforcement learning framework for efficient AMP generation. Additionally, our prediction module incorporates pre-training and transfer learning strategies for precise AMP identification and screening. We employ a convolutional neural network for multi-attribute prediction and a reinforcement learning-based iterative optimization strategy to produce diverse AMPs. This framework automates molecule generation, screening, attribute prediction and optimization, thereby advancing AMP research. We have also deployed Diff-AMP on a web server, with code, data and server details available in the Data Availability section.


Subject(s)
Amino Acids , Antimicrobial Peptides , Anti-Bacterial Agents , Diffusion , Kinetics
3.
Comput Biol Med ; 171: 108104, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38335821

ABSTRACT

Drug-food interactions (DFIs) crucially impact patient safety and drug efficacy by modifying absorption, distribution, metabolism, and excretion. The application of deep learning for predicting DFIs is promising, yet the development of computational models remains in its early stages. This is mainly due to the complexity of food compounds, challenging dataset developers in acquiring comprehensive ingredient data, often resulting in incomplete or vague food component descriptions. DFI-MS tackles this issue by employing an accurate feature representation method alongside a refined computational model. It innovatively achieves a more precise characterization of food features, a previously daunting task in DFI research. This is accomplished through modules designed for perturbation interactions, feature alignment and domain separation, and inference feedback. These modules extract essential information from features, using a perturbation module and a feature interaction encoder to establish robust representations. The feature alignment and domain separation modules are particularly effective in managing data with diverse frequencies and characteristics. DFI-MS stands out as the first in its field to combine data augmentation, feature alignment, domain separation, and contrastive learning. The flexibility of the inference feedback module allows its application in various downstream tasks. Demonstrating exceptional performance across multiple datasets, DFI-MS represents a significant advancement in food presentations technology. Our code and data are available at https://github.com/kkkayle/DFI-MS.


Subject(s)
Food-Drug Interactions , Food , Humans , Supervised Machine Learning
4.
Brief Bioinform ; 24(4)2023 07 20.
Article in English | MEDLINE | ID: mdl-37427977

ABSTRACT

Studies have shown that the mechanism of action of many drugs is related to miRNA. In-depth research on the relationship between miRNA and drugs can provide theoretical foundations and practical approaches for various areas, such as drug target discovery, drug repositioning and biomarker research. Traditional biological experiments to test miRNA-drug susceptibility are costly and time-consuming. Thus, sequence- or topology-based deep learning methods are recognized in this field for their efficiency and accuracy. However, these methods have limitations in dealing with sparse topologies and higher-order information of miRNA (drug) feature. In this work, we propose GCFMCL, a model for multi-view contrastive learning based on graph collaborative filtering. To the best of our knowledge, this is the first attempt that incorporates contrastive learning strategy into the graph collaborative filtering framework to predict the sensitivity relationships between miRNA and drug. The proposed multi-view contrastive learning method is divided into topological contrastive objective and feature contrastive objective: (1) For the homogeneous neighbors of the topological graph, we propose a novel topological contrastive learning method via constructing the contrastive target through the topological neighborhood information of nodes. (2) The proposed model obtains feature contrastive targets from high-order feature information according to the correlation of node features, and mines potential neighborhood relationships in the feature space. The proposed multi-view comparative learning effectively alleviates the impact of heterogeneous node noise and graph data sparsity in graph collaborative filtering, and significantly enhances the performance of the model. Our study employs a dataset derived from the NoncoRNA and ncDR databases, encompassing 2049 experimentally validated miRNA-drug sensitivity associations. Five-fold cross-validation shows that the Area Under the Curve (AUC), Area Under the Precision-Recall Curve (AUPR) and F1-score (F1) of GCFMCL reach 95.28%, 95.66% and 89.77%, which outperforms the state-of-the-art (SOTA) method by the margin of 2.73%, 3.42% and 4.96%, respectively. Our code and data can be accessed at https://github.com/kkkayle/GCFMCL.


Subject(s)
Drug Delivery Systems , MicroRNAs , Area Under Curve , Databases, Factual , Drug Discovery , MicroRNAs/genetics
5.
Comput Biol Med ; 163: 107143, 2023 09.
Article in English | MEDLINE | ID: mdl-37339574

ABSTRACT

Non-coding RNA (ncRNA) is a functional RNA molecule that plays a key role in various fundamental biological processes, such as gene regulation. Therefore, studying the connection between ncRNA and proteins holds significant importance in exploring the function of ncRNA. Although many efficient and accurate methods have been developed by modern biological scientists, accurate predictions still pose a major challenge for various issues. In our approach, we utilize a multi-head attention mechanism to merge residual connections, allowing for the automatic learning of ncRNA and protein sequence features. Specifically, the proposed method projects node features into multiple spaces based on multi-head attention mechanism, thereby obtaining different feature interaction patterns in these spaces. By stacking interaction layers, higher-order interaction modes can be derived, while still preserving the initial feature information through the residual connection. This strategy effectively leverages the sequence information of ncRNA and protein, enabling the capture of hidden high-order features. The final experimental results demonstrate the effectiveness of our method, with AUC values of 97.4%, 98.5%, and 94.8% achieved on the NPInter v2.0, RPI807, and RPI488 datasets, respectively. These impressive results solidify our method as a powerful tool for exploring the connection between ncRNAs and proteins. We have uploaded the implementation code on GitHub: https://github.com/ZZCrazy00/MHAM-NPI.


Subject(s)
Proteins , RNA, Untranslated , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Proteins/metabolism
6.
Comput Biol Med ; 157: 106783, 2023 05.
Article in English | MEDLINE | ID: mdl-36958237

ABSTRACT

Noncoding RNA (ncRNA) is a functional RNA derived from DNA transcription, and most transcribed genes are transcribed into ncRNA. ncRNA is not directly involved in the translation of proteins, but it can participate in gene expression in cells and affect protein synthesis, thus playing an important role in biological processes such as growth, proliferation, metabolism, and information transmission. Therefore, understanding the interaction between ncRNA and protein is the basis for studying ncRNA regulation of protein-related biological activities. However, it is very expensive and time-consuming to verify ncRNA-protein interaction through biological experiments, and prediction methods based on machine learning have been developed rapidly. Recently, the graph neural network model (GNN) stands out for its excellent performance, but lacks a general framework for predicting ncRNA-protein interactions. We propose a GNN-based framework to predict ncRNA-protein interactions, which can utilize topological structure information to complete prediction tasks faster and more accurately. Meanwhile, for some smaller datasets, many ncRNA nodes lack neighbor information, resulting in lower prediction accuracy. For some larger datasets, the long-tail distribution causes the prediction of the tail nodes (sparse nodes linking few neighbors) to be affected. Therefore, we propose a new sampling method named HeadTailTransfer to mitigate these effects. Experimental results illustrate the effectiveness of this method. Especially for task-specific prediction on the RPI369 dataset in the Graphsage-based neural network framework, the AUC and ACC values increased from 56.8% and 52.2% to 80.2% and 71.8%, respectively. Our data and codes are available: https://github.com/kkkayle/HeadTailTransfer.


Subject(s)
Neural Networks, Computer , RNA, Untranslated , RNA, Untranslated/genetics , RNA, Untranslated/chemistry , RNA, Untranslated/metabolism , Machine Learning , Protein Binding , Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...