1
|
Yang B, Liu Y, Wu J, Bai F, Zheng M, Zheng J. GENNDTI: Drug-Target Interaction Prediction Using Graph Neural Network Enhanced by Router Nodes. IEEE J Biomed Health Inform 2024; 28:7588-7598. [PMID: 40030413 DOI: 10.1109/jbhi.2024.3402529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Identifying drug-target interactions (DTI) is crucial in drug discovery and repurposing, and in silico techniques for DTI predictions are becoming increasingly important for reducing time and cost. Most interaction-based DTI models rely on the guilt-by-association principle that "similar drugs can interact with similar targets". However, such methods utilize precomputed similarity matrices and cannot dynamically discover intricate correlations. Meanwhile, some methods enrich DTI networks by incorporating additional networks like DDI and PPI networks, enriching biological signals to enhance DTI prediction. While these approaches have achieved promising performance in DTI prediction, such coarse-grained association data do not explain the specific biological mechanisms underlying DTIs. In this work, we propose GENNDTI, which constructs biologically meaningful routers to represent and integrate the salient properties of drugs and targets. Similar drugs or targets connect to more same router nodes, capturing property sharing. In addition, heterogeneous encoders are designed to distinguish different types of interactions, modeling both real and constructed interactions. This strategy enriches graph topology and enhances prediction efficiency as well. We evaluate the proposed method on benchmark datasets, demonstrating comparative performance over existing methods. We specifically analyze router nodes to validate their efficacy in improving predictions and providing biological explanations.
Collapse
|
2
|
Dao NA, Le MH, Dang XT. Label Transfer for Drug Disease Association in Three Meta-Paths. Evol Bioinform Online 2024; 20:11769343241272414. [PMID: 39279816 PMCID: PMC11401013 DOI: 10.1177/11769343241272414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 07/15/2024] [Indexed: 09/18/2024] Open
Abstract
The identification of potential interactions and relationships between diseases and drugs is significant in public health care and drug discovery. As we all know, experimenting to determine the drug-disease interactions is very expensive in both time and money. However, there are still many drug-disease associations that are still undiscovered and potential. Therefore, the development of computational methods to explore the relationship between drugs and diseases is very important and essential. Many computational methods for predicting drug-disease associations have been developed based on known interactions to learn potential interactions of unknown drug-disease pairs. In this paper, we propose 3 new main groups of meta-paths based on the heterogeneous biological network of drug-protein-disease objects. For each meta-path, we design a machine learning model, then an integrated learning method is formed by these models. We evaluated our approach on 3 standard datasets which are DrugBank, OMIM, and Gottlieb's dataset. Experimental results demonstrate that the proposed method is better than some recent methods such as EMP-SVD, LRSSL, MBiRW, MPG-DDA, SCMFDD,. . . in some measures such as AUC, AUPR, and F1-score.
Collapse
|
3
|
Peng L, Liu X, Chen M, Liao W, Mao J, Zhou L. MGNDTI: A Drug-Target Interaction Prediction Framework Based on Multimodal Representation Learning and the Gating Mechanism. J Chem Inf Model 2024; 64:6684-6698. [PMID: 39137398 DOI: 10.1021/acs.jcim.4c00957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
Drug-Target Interaction (DTI) prediction facilitates acceleration of drug discovery and promotes drug repositioning. Most existing deep learning-based DTI prediction methods can better extract discriminative features for drugs and proteins, but they rarely consider multimodal features of drugs. Moreover, learning the interaction representations between drugs and targets needs further exploration. Here, we proposed a simple M ulti-modal G ating N etwork for DTI prediction, MGNDTI, based on multimodal representation learning and the gating mechanism. MGNDTI first learns the sequence representations of drugs and targets using different retentive networks. Next, it extracts molecular graph features of drugs through a graph convolutional network. Subsequently, it devises a multimodal gating network to obtain the joint representations of drugs and targets. Finally, it builds a fully connected network for computing the interaction probability. MGNDTI was benchmarked against seven state-of-the-art DTI prediction models (CPI-GNN, TransformerCPI, MolTrans, BACPI, CPGL, GIFDTI, and FOTF-CPI) using four data sets (i.e., Human, C. elegans, BioSNAP, and BindingDB) under four different experimental settings. Through evaluation with AUROC, AUPRC, accuracy, F1 score, and MCC, MGNDTI significantly outperformed the above seven methods. MGNDTI is a powerful tool for DTI prediction, showcasing its superior robustness and generalization ability on diverse data sets and different experimental settings. It is freely available at https://github.com/plhhnu/MGNDTI.
Collapse
Affiliation(s)
- Lihong Peng
- College of Life Science and Chemistry, Hunan University of Technology, Zhuzhou, Hunan 412007, China
| | - Xin Liu
- College of Life Science and Chemistry, Hunan University of Technology, Zhuzhou, Hunan 412007, China
| | - Min Chen
- School of Computer Science and Engineering, Hunan Institute of Technology, Hengyang, Hunan 421002, China
| | - Wen Liao
- School of Computer Science, Hunan University of Technology, Zhuzhou, Hunan 412007, China
| | - Jiale Mao
- School of Computer Science, Hunan University of Technology, Zhuzhou, Hunan 412007, China
| | - Liqian Zhou
- College of Life Science and Chemistry, Hunan University of Technology, Zhuzhou, Hunan 412007, China
| |
Collapse
|
4
|
Li Y, Liang W, Peng L, Zhang D, Yang C, Li KC. Predicting Drug-Target Interactions Via Dual-Stream Graph Neural Network. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2024; 21:948-958. [PMID: 36074878 DOI: 10.1109/tcbb.2022.3204188] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Drug target interaction prediction is a crucial stage in drug discovery. However, brute-force search over a compound database is financially infeasible. We have witnessed the increasing measured drug-target interactions records in recent years, and the rich drug/protein-related information allows the usage of graph machine learning. Despite the advances in deep learning-enabled drug-target interaction, there are still open challenges: (1) rich and complex relationship between drugs and proteins can be explored; (2) the intermediate node is not calibrated in the heterogeneous graph. To tackle with above issues, this paper proposed a framework named DSG-DTI. Specifically, DSG-DTI has the heterogeneous graph autoencoder and heterogeneous attention network-based Matrix Completion. Our framework ensures that the known types of nodes (e.g., drug, target, side effects, diseases) are precisely embedded into high-dimensional space with our pretraining skills. Also, the attention-based heterogeneous graph-based matrix completion achieves highly competitive results via effective long-range dependencies extraction. We verify our model on two public benchmarks. The result of two publicly available benchmark application programs show that the proposed scheme effectively predicts drug-target interactions and can generalize to newly registered drugs and targets with slight performance degradation, outperforming the best accuracy compared with other baselines.
Collapse
|
5
|
Zhang Z, Zhao L, Wang J, Wang C. A Hierarchical Graph Neural Network Framework for Predicting Protein-Protein Interaction Modulators With Functional Group Information and Hypergraph Structure. IEEE J Biomed Health Inform 2024; 28:4295-4305. [PMID: 38564358 DOI: 10.1109/jbhi.2024.3384238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Accurate prediction of small molecule modulators targeting protein-protein interactions (PPIMs) remains a significant challenge in drug discovery. Existing machine learning-based models rely on manual feature engineering, which is tedious and task-specific. Recently, deep learning models based on graph neural networks have made remarkable progress in molecular representation learning. However, many graph-based approaches ignore molecular hierarchical structure modeling guided by domain knowledge. In chemistry, the functional groups of a molecule determine its interaction with specific targets. Therefore, we propose a hierarchical graph neural network framework (called HiGPPIM) for predicting PPIMs by integrating atom-level and functional group-level features of molecules. HiGPPIM constructs atom-level and functional group-level graphs based on chemical knowledge and learns graph representations using graph attention networks. Furthermore, a hypergraph attention network is designed in HiGPPIM to aggregate and transform two-level graph information. We evaluate the performance of HiGPPIM on eight PPI families and two prediction tasks, namely PPIM identification and potency prediction. Experimental results demonstrate that HiGPPIM achieves state-of-the-art performance on both tasks and that using functional group information to guide PPIM prediction is effective.
Collapse
|
6
|
Zhang Q, Zuo L, Ren Y, Wang S, Wang W, Ma L, Zhang J, Xia B. FMCA-DTI: a fragment-oriented method based on a multihead cross attention mechanism to improve drug-target interaction prediction. Bioinformatics 2024; 40:btae347. [PMID: 38810106 PMCID: PMC11256963 DOI: 10.1093/bioinformatics/btae347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 04/23/2024] [Accepted: 05/28/2024] [Indexed: 05/31/2024] Open
Abstract
MOTIVATION Identifying drug-target interactions (DTI) is crucial in drug discovery. Fragments are less complex and can accurately characterize local features, which is important in DTI prediction. Recently, deep learning (DL)-based methods predict DTI more efficiently. However, two challenges remain in existing DL-based methods: (i) some methods directly encode drugs and proteins into integers, ignoring the substructure representation; (ii) some methods learn the features of the drugs and proteins separately instead of considering their interactions. RESULTS In this article, we propose a fragment-oriented method based on a multihead cross attention mechanism for predicting DTI, named FMCA-DTI. FMCA-DTI obtains multiple types of fragments of drugs and proteins by branch chain mining and category fragment mining. Importantly, FMCA-DTI utilizes the shared-weight-based multihead cross attention mechanism to learn the complex interaction features between different fragments. Experiments on three benchmark datasets show that FMCA-DTI achieves significantly improved performance by comparing it with four state-of-the-art baselines. AVAILABILITY AND IMPLEMENTATION The code for this workflow is available at: https://github.com/jacky102022/FMCA-DTI.
Collapse
Affiliation(s)
- Qi Zhang
- College of Mathematics and Computer Science, Yan'an University, Yan'an 716000, China
| | - Le Zuo
- College of Mathematics and Computer Science, Yan'an University, Yan'an 716000, China
| | - Ying Ren
- College of Mathematics and Computer Science, Yan'an University, Yan'an 716000, China
| | - Siyuan Wang
- College of Mathematics and Computer Science, Yan'an University, Yan'an 716000, China
| | - Wenfa Wang
- College of Mathematics and Computer Science, Yan'an University, Yan'an 716000, China
| | - Lerong Ma
- College of Mathematics and Computer Science, Yan'an University, Yan'an 716000, China
| | - Jing Zhang
- Medical College of Yan'an University, Yan'an University, Yan'an 716000, China
- Medical Research and Experimental Center, The Second Affiliated Hospital of Xi'an Medical University, Xi'an 710021, China
| | - Bisheng Xia
- College of Mathematics and Computer Science, Yan'an University, Yan'an 716000, China
| |
Collapse
|
7
|
Mushtaq A, Asif R, Humayun WA, Naseer MM. Novel isatin-triazole based thiosemicarbazones as potential anticancer agents: synthesis, DFT and molecular docking studies. RSC Adv 2024; 14:14051-14067. [PMID: 38686286 PMCID: PMC11057040 DOI: 10.1039/d4ra01937g] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 04/23/2024] [Indexed: 05/02/2024] Open
Abstract
Thiosemicarbazones of isatin have been found to exhibit versatile bioactivities. In this study, two distinct types of isatin-triazole hybrids 3a and 3b were accessed via copper-catalyzed azide-alkyne cycloaddition reaction (CuAAC), together with their mono and bis-thiosemicarbazone derivatives 4a-h and 5a-h. In addition to the characterization by physical, spectral and analytical data, a DFT study was carried out to obtain the optimized geometries of all thiosemicarbazones. The global reactivity values showed that among the synthesized derivatives, 4c, 4g and 5c having nitro substituents are the most soft compounds, with compound 5c having the highest electronegativity and electrophilicity index values among the synthesized series, thus possessing strong binding ability with biomolecules. Molecular docking studies were performed to explore the inhibitory ability of the selected compounds against the active sites of the anticancer protein of phosphoinositide 3-kinase (PI3K). Among the synthesized derivatives, 4-nitro substituted bisthiosemicarbazone 5c showed the highest binding energy of -10.3 kcal mol-1. These findings demonstrated that compound 5c could be used as a favored anticancer scaffold via the mechanism of inhibition against the PI3K signaling pathways.
Collapse
Affiliation(s)
- Alia Mushtaq
- Department of Chemistry, Quaid-i-Azam University Islamabad 45320 Pakistan
| | - Rabbia Asif
- Department of Chemistry, Quaid-i-Azam University Islamabad 45320 Pakistan
| | - Waqar Ahmed Humayun
- Department of Medical Oncology & Radiotherapy, King Edward Medical University Lahore 54000 Pakistan
| | | |
Collapse
|
8
|
Gu X, Liu J, Yu Y, Xiao P, Ding Y. MFD-GDrug: multimodal feature fusion-based deep learning for GPCR-drug interaction prediction. Methods 2024; 223:75-82. [PMID: 38286333 DOI: 10.1016/j.ymeth.2024.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 01/14/2024] [Accepted: 01/26/2024] [Indexed: 01/31/2024] Open
Abstract
The accurate identification of drug-protein interactions (DPIs) is crucial in drug development, especially concerning G protein-coupled receptors (GPCRs), which are vital targets in drug discovery. However, experimental validation of GPCR-drug pairings is costly, prompting the need for accurate predictive methods. To address this, we propose MFD-GDrug, a multimodal deep learning model. Leveraging the ESM pretrained model, we extract protein features and employ a CNN for protein feature representation. For drugs, we integrated multimodal features of drug molecular structures, including three-dimensional features derived from Mol2vec and the topological information of drug graph structures extracted through Graph Convolutional Neural Networks (GCN). By combining structural characterizations and pretrained embeddings, our model effectively captures GPCR-drug interactions. Our tests on leading GPCR-drug interaction datasets show that MFD-GDrug outperforms other methods, demonstrating superior predictive accuracy.
Collapse
Affiliation(s)
- Xingyue Gu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Junkai Liu
- School of Electronic and Information Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Yue Yu
- School of Computer and Artificial Intelligence, Zhengzhou University, Zhengzhou 450001, China
| | - Pengfeng Xiao
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Yijie Ding
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou, Zhejiang 324003, China; Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu 611730, China.
| |
Collapse
|
9
|
Wang Q, Gu J, Zhang J, Liu M, Jin X, Xie M. A Heterogeneous Cross Contrastive Learning Method for Drug-Target Interaction Prediction. LECTURE NOTES IN COMPUTER SCIENCE 2024:183-194. [DOI: 10.1007/978-981-97-5689-6_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
10
|
Wu K, Karapetyan E, Schloss J, Vadgama J, Wu Y. Advancements in small molecule drug design: A structural perspective. Drug Discov Today 2023; 28:103730. [PMID: 37536390 PMCID: PMC10543554 DOI: 10.1016/j.drudis.2023.103730] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 07/19/2023] [Accepted: 07/27/2023] [Indexed: 08/05/2023]
Abstract
In this review, we outline recent advancements in small molecule drug design from a structural perspective. We compare protein structure prediction methods and explore the role of the ligand binding pocket in structure-based drug design. We examine various structural features used to optimize drug candidates, including functional groups, stereochemistry, and molecular weight. Computational tools such as molecular docking and virtual screening are discussed for predicting and optimizing drug candidate structures. We present examples of drug candidates designed based on their molecular structure and discuss future directions in the field. By effectively integrating structural information with other valuable data sources, we can improve the drug discovery process, leading to the identification of novel therapeutics with improved efficacy, specificity, and safety profiles.
Collapse
Affiliation(s)
- Ke Wu
- Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA
| | - Eduard Karapetyan
- Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA
| | - John Schloss
- Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA; School of Pharmacy, American University of Health Sciences, Signal Hill, CA 90755, USA
| | - Jaydutt Vadgama
- Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA; School of Pharmacy, American University of Health Sciences, Signal Hill, CA 90755, USA.
| | - Yong Wu
- Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA.
| |
Collapse
|
11
|
Huang Y, Huang HY, Chen Y, Lin YCD, Yao L, Lin T, Leng J, Chang Y, Zhang Y, Zhu Z, Ma K, Cheng YN, Lee TY, Huang HD. A Robust Drug-Target Interaction Prediction Framework with Capsule Network and Transfer Learning. Int J Mol Sci 2023; 24:14061. [PMID: 37762364 PMCID: PMC10531393 DOI: 10.3390/ijms241814061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/27/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023] Open
Abstract
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.
Collapse
Affiliation(s)
- Yixian Huang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (Y.H.); (Y.C.); (J.L.)
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (L.Y.); (Y.C.)
| | - Hsi-Yuan Huang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (Y.H.); (Y.C.); (J.L.)
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (L.Y.); (Y.C.)
| | - Yigang Chen
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (Y.H.); (Y.C.); (J.L.)
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (L.Y.); (Y.C.)
| | - Yang-Chi-Dung Lin
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (Y.H.); (Y.C.); (J.L.)
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (L.Y.); (Y.C.)
| | - Lantian Yao
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (L.Y.); (Y.C.)
| | - Tianxiu Lin
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (Y.H.); (Y.C.); (J.L.)
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (L.Y.); (Y.C.)
| | - Junlin Leng
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (Y.H.); (Y.C.); (J.L.)
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (L.Y.); (Y.C.)
| | - Yuan Chang
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (L.Y.); (Y.C.)
| | - Yuntian Zhang
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (L.Y.); (Y.C.)
| | - Zihao Zhu
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (Y.H.); (Y.C.); (J.L.)
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (L.Y.); (Y.C.)
| | - Kun Ma
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (Y.H.); (Y.C.); (J.L.)
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (L.Y.); (Y.C.)
| | - Yeong-Nan Cheng
- Institute of Bioinformatics and Systems Biology, Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan; (Y.-N.C.)
| | - Tzong-Yi Lee
- Institute of Bioinformatics and Systems Biology, Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan; (Y.-N.C.)
| | - Hsien-Da Huang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (Y.H.); (Y.C.); (J.L.)
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (L.Y.); (Y.C.)
| |
Collapse
|
12
|
Malik MS, Faazil S, Alsharif MA, Sajid Jamal QM, Al-Fahemi JH, Banerjee A, Chattopadhyay A, Pal SK, Kamal A, Ahmed SA. Antibacterial Properties and Computational Insights of Potent Novel Linezolid-Based Oxazolidinones. Pharmaceuticals (Basel) 2023; 16:516. [PMID: 37111273 PMCID: PMC10143092 DOI: 10.3390/ph16040516] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/23/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023] Open
Abstract
The mounting evidence of bacterial resistance against commonly prescribed antibiotics warrants the development of new antibacterial drugs on an urgent basis. Linezolid, an oxazolidinone antibiotic, is a lead molecule in designing new oxazolidinones as antibacterial agents. In this study, we report the antibacterial potential of the novel oxazolidinone-sulphonamide/amide conjugates that were recently reported by our research group. The antibacterial assays showed that, from the series, oxazolidinones 2 and 3a exhibited excellent potency (MIC of 1.17 μg/mL) against B. subtilis and P. aeruginosa strains, along with good antibiofilm activity. Docking studies revealed higher binding affinities of oxazolidinones 2 and 3a compared to linezolid, which were further validated by molecular dynamics simulations. In addition to this, other computational studies, one-descriptor (log P) analysis, ADME-T and drug likeness studies demonstrated the potential of these novel linezolid-based oxazolidinones to be taken forward for further studies.
Collapse
Affiliation(s)
- M. Shaheer Malik
- Department of Chemistry, Faculty of Applied Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (M.A.A.)
| | - Shaikh Faazil
- Department of Chemistry, Poona College of Arts, Science and Commerce, Pune 411001, India
- Department of Medicinal Chemistry and Pharmacology, CSIR—Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Meshari A. Alsharif
- Department of Chemistry, Faculty of Applied Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (M.A.A.)
| | - Qazi Mohammad Sajid Jamal
- Department of Health Informatics, College of Public Health and Health Informatics, Qassim University, Al Bukayriyah 52741, Saudi Arabia;
| | - Jabir H. Al-Fahemi
- Department of Chemistry, Faculty of Applied Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (M.A.A.)
| | - Amrita Banerjee
- Department of Physics, Jadavpur University, 188, Raja S.C. Mallick Rd., Kolkata 700032, India;
- Technical Research Centre, S. N. Bose National Centre for Basic Sciences, Block JD, Sector III, Salt Lake, Kolkata 700106, India
| | - Arpita Chattopadhyay
- Department of Basic Science and Humanities, Techno International New Town, Block—DG 1/1, Action Area 1, New Town, Rajarhat, Kolkata 700156, India;
| | - Samir Kumar Pal
- Department of Chemical and Biological Sciences, S. N. Bose National Centre for Basic Sciences, Block JD, Sector 3, Salt Lake, Kolkata 700106, India;
| | - Ahmed Kamal
- Department of Medicinal Chemistry and Pharmacology, CSIR—Indian Institute of Chemical Technology, Hyderabad 500007, India
- Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad 500078, India
| | - Saleh A. Ahmed
- Department of Chemistry, Faculty of Applied Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (M.A.A.)
- Chemistry Department, Faculty of Science, Assiut University, Assiut 71516, Egypt
| |
Collapse
|
13
|
Jia ZJ, Lan XW, Lu K, Meng X, Jing WJ, Jia SR, Zhao K, Dai YJ. Synthesis, molecular docking, and binding Gibbs free energy calculation of β-nitrostyrene derivatives: Potential inhibitors of SARS-CoV-2 3CL protease. J Mol Struct 2023; 1284:135409. [PMID: 36993878 PMCID: PMC10033154 DOI: 10.1016/j.molstruc.2023.135409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/10/2023] [Accepted: 03/21/2023] [Indexed: 03/24/2023]
Abstract
The outbreak of novel coronavirus disease 2019 (COVID-19), caused by the novel coronavirus (SARS-CoV-2), has had a significant impact on human health and the economic development. SARS-CoV-2 3CL protease (3CLpro) is highly conserved and plays a key role in mediating the transcription of virus replication. It is an ideal target for the design and screening of anti-coronavirus drugs. In this work, seven β-nitrostyrene derivatives were synthesized by Henry reaction and β-dehydration reaction, and their inhibitory effects on SARS-CoV-2 3CL protease were identified by enzyme activity inhibition assay in vitro. Among them, 4-nitro-β-nitrostyrene (compound a) showed the lowest IC50 values of 0.7297 μM. To investigate the key groups that determine the activity of β-nitrostyrene derivatives and their interaction mode with the receptor, the molecular docking using the CDOCKER protocol in Discovery Studio 2016 was performed. The results showed that the hydrogen bonds between β-NO2 and receptor GLY-143 and the π-π stacking between the aryl ring of the ligand and the imidazole ring of receptor HIS-41 significantly contributed to the ligand activity. Furthermore, the ligand-receptor absolute binding Gibbs free energies were calculated using the Binding Affinity Tool (BAT.py) to verify its correlation with the activity of β-nitrostyrene 3CLpro inhibitors as a scoring function. The higher correlation(r2=0.6) indicates that the absolute binding Gibbs free energy based on molecular dynamics can be used to predict the activity of new β-nitrostyrene 3CLpro inhibitors. These results provide valuable insights for the functional group-based design, structure optimization and the discovery of high accuracy activity prediction means of anti-COVID-19 lead compounds.
Collapse
Affiliation(s)
- Ze-Jun Jia
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, PR China
| | - Xiao-Wei Lan
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, PR China
| | - Kui Lu
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, PR China
| | - Xuan Meng
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, PR China
| | - Wen-Jie Jing
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, PR China
| | - Shi-Ru Jia
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, PR China
| | - Kai Zhao
- Hebei Kaisheng Medical Technology Co. LTD, No.319 of Xiangjiang Road, High-tech Zone, Shijiazhuang 050000, PR China
- Jiangxi Oushi Pharmaceutical Co. LTD, 1115 Saiwei Dadao, Yushui District, Xinyu 338004, PR China
| | - Yu-Jie Dai
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, PR China
| |
Collapse
|
14
|
MOZART, a QSAR Multi-Target Web-Based Tool to Predict Multiple Drug-Enzyme Interactions. Molecules 2023; 28:molecules28031182. [PMID: 36770857 PMCID: PMC9921108 DOI: 10.3390/molecules28031182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/09/2023] [Accepted: 01/16/2023] [Indexed: 01/27/2023] Open
Abstract
Developing models able to predict interactions between drugs and enzymes is a primary goal in computational biology since these models may be used for predicting both new active drugs and the interactions between known drugs on untested targets. With the compilation of a large dataset of drug-enzyme pairs (62,524), we recognized a unique opportunity to attempt to build a novel multi-target machine learning (MTML) quantitative structure-activity relationship (QSAR) model for probing interactions among different drugs and enzyme targets. To this end, this paper presents an MTML-QSAR model based on using the features of topological drugs together with the artificial neural network (ANN) multi-layer perceptron (MLP). Validation of the final best model found was carried out by internal cross-validation statistics and other relevant diagnostic statistical parameters. The overall accuracy of the derived model was found to be higher than 96%. Finally, to maximize the diffusion of this model, a public and accessible tool has been developed to allow users to perform their own predictions. The developed web-based tool is public accessible and can be downloaded as free open-source software.
Collapse
|
15
|
Ahn S, Lee SE, Kim MH. Random-forest model for drug-target interaction prediction via Kullbeck-Leibler divergence. J Cheminform 2022; 14:67. [PMID: 36192818 PMCID: PMC9531514 DOI: 10.1186/s13321-022-00644-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 09/11/2022] [Indexed: 12/04/2022] Open
Abstract
Virtual screening has significantly improved the success rate of early stage drug discovery. Recent virtual screening methods have improved owing to advances in machine learning and chemical information. Among these advances, the creative extraction of drug features is important for predicting drug–target interaction (DTI), which is a large-scale virtual screening of known drugs. Herein, we report Kullbeck–Leibler divergence (KLD) as a DTI feature and the feature-driven classification model applicable to DTI prediction. For the purpose, E3FP three-dimensional (3D) molecular fingerprints of drugs as a molecular representation allow the computation of 3D similarities between ligands within each target (Q–Q matrix) to identify the uniqueness of pharmacological targets and those between a query and a ligand (Q–L vector) in DTIs. The 3D similarity matrices are transformed into probability density functions via kernel density estimation as a nonparametric estimation. Each density model can exploit the characteristics of each pharmacological target and measure the quasi-distance between the ligands. Furthermore, we developed a random forest model from the KLD feature vectors to successfully predict DTIs for representative 17 targets (mean accuracy: 0.882, out-of-bag score estimate: 0.876, ROC AUC: 0.990). The method is applicable for 2D chemical similarity.
Collapse
Affiliation(s)
- Sangjin Ahn
- Gachon Institute of Pharmaceutical Science and Department of Pharmacy, College of Pharmacy, Gachon University, 191 Hambakmoeiro, Yeonsu-gu, Incheon, Republic of Korea.,Department of Artificial Intelligence, Ajou University, Suwon, 16499, Republic of Korea
| | - Si Eun Lee
- Gachon Institute of Pharmaceutical Science and Department of Pharmacy, College of Pharmacy, Gachon University, 191 Hambakmoeiro, Yeonsu-gu, Incheon, Republic of Korea
| | - Mi-Hyun Kim
- Gachon Institute of Pharmaceutical Science and Department of Pharmacy, College of Pharmacy, Gachon University, 191 Hambakmoeiro, Yeonsu-gu, Incheon, Republic of Korea.
| |
Collapse
|
16
|
Yeh SJ, Chen BS. Systems Medicine Design based on Systems Biology Approaches and Deep Neural Network for Gastric Cancer. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:3019-3031. [PMID: 34232888 DOI: 10.1109/tcbb.2021.3095369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Gastric cancer (GC) is the third leading cause of cancer death in the world. It is associated with the stimulation of microenvironment, aberrant epigenetic modification, and chronic inflammation. However, few researches discuss the GC molecular progression mechanisms from the perspective of the system level. In this study, we proposed a systems medicine design procedure to identify essential biomarkers and find corresponding drugs for GC. At first, we did big database mining to construct candidate protein-protein interaction network (PPIN) and candidate gene regulation network (GRN). Second, by leveraging the next-generation sequencing (NGS) data, we performed system modeling and applied system identification and model selection to obtain real genome-wide genetic and epigenetic networks (GWGENs). To make the real GWGENs easy to analyze, the principal network projection method was used to extract the core signaling pathways denoted by KEGG pathways. Subsequently, based on the identified biomarkers, we trained a deep neural network of drug-target interaction (DeepDTI) with supervised learning and filtered our candidate drugs considering drug regulation ability and drug sensitivity. With the proposed systematic strategy, we not only shed the light on the progression of GC but also suggested potential multiple-molecule drugs efficiently.
Collapse
|
17
|
Lian M, Wang X, Du W. Integrated multi-similarity fusion and heterogeneous graph inference for drug-target interaction prediction. Neurocomputing 2022. [DOI: 10.1016/j.neucom.2022.04.104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
18
|
Margulis E, Slavutsky Y, Lang T, Behrens M, Benjamini Y, Niv MY. BitterMatch: recommendation systems for matching molecules with bitter taste receptors. J Cheminform 2022; 14:45. [PMID: 35799226 PMCID: PMC9261901 DOI: 10.1186/s13321-022-00612-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 05/14/2022] [Indexed: 11/10/2022] Open
Abstract
Bitterness is an aversive cue elicited by thousands of chemically diverse compounds. Bitter taste may prevent consumption of foods and jeopardize drug compliance. The G protein-coupled receptors for bitter taste, TAS2Rs, have species-dependent number of subtypes and varying expression levels in extraoral tissues. Molecular recognition by TAS2R subtypes is physiologically important, and presents a challenging case study for ligand-receptor matchmaking. Inspired by hybrid recommendation systems, we developed a new set of similarity features, and created the BitterMatch algorithm that predicts associations of ligands to receptors with ~ 80% precision at ~ 50% recall. Associations for several compounds were tested in-vitro, resulting in 80% precision and 42% recall. The encouraging performance was achieved by including receptor properties and integrating experimentally determined ligand-receptor associations with chemical ligand-to-ligand similarities. BitterMatch can predict off-targets for bitter drugs, identify novel ligands and guide flavor design. The novel features capture information regarding the molecules and their receptors, which could inform various chemoinformatic tasks. Inclusion of neighbor-informed similarities improves as experimental data mounts, and provides a generalizable framework for molecule-biotarget matching.
Collapse
Affiliation(s)
- Eitan Margulis
- The Institute of Biochemistry, Food Science and Nutrition, The Robert H Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Yuli Slavutsky
- Department of Statistics and Data Science, Faculty of Social Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Tatjana Lang
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
| | - Maik Behrens
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
| | - Yuval Benjamini
- Department of Statistics and Data Science, Faculty of Social Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Masha Y Niv
- The Institute of Biochemistry, Food Science and Nutrition, The Robert H Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel.
| |
Collapse
|
19
|
Zheng J, Xiao X, Qiu WR. DTI-BERT: Identifying Drug-Target Interactions in Cellular Networking Based on BERT and Deep Learning Method. Front Genet 2022; 13:859188. [PMID: 35754843 PMCID: PMC9213727 DOI: 10.3389/fgene.2022.859188] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/25/2022] [Indexed: 11/20/2022] Open
Abstract
Drug–target interactions (DTIs) are regarded as an essential part of genomic drug discovery, and computational prediction of DTIs can accelerate to find the lead drug for the target, which can make up for the lack of time-consuming and expensive wet-lab techniques. Currently, many computational methods predict DTIs based on sequential composition or physicochemical properties of drug and target, but further efforts are needed to improve them. In this article, we proposed a new sequence-based method for accurately identifying DTIs. For target protein, we explore using pre-trained Bidirectional Encoder Representations from Transformers (BERT) to extract sequence features, which can provide unique and valuable pattern information. For drug molecules, Discrete Wavelet Transform (DWT) is employed to generate information from drug molecular fingerprints. Then we concatenate the feature vectors of the DTIs, and input them into a feature extraction module consisting of a batch-norm layer, rectified linear activation layer and linear layer, called BRL block and a Convolutional Neural Networks module to extract DTIs features further. Subsequently, a BRL block is used as the prediction engine. After optimizing the model based on contrastive loss and cross-entropy loss, it gave prediction accuracies of the target families of G Protein-coupled receptors, ion channels, enzymes, and nuclear receptors up to 90.1, 94.7, 94.9, and 89%, which indicated that the proposed method can outperform the existing predictors. To make it as convenient as possible for researchers, the web server for the new predictor is freely accessible at: https://bioinfo.jcu.edu.cn/dtibert or http://121.36.221.79/dtibert/. The proposed method may also be a potential option for other DITs.
Collapse
Affiliation(s)
- Jie Zheng
- Computer Department, Jing-De-Zhen Ceramic Institute, Jing-De-Zhen, China
| | - Xuan Xiao
- Computer Department, Jing-De-Zhen Ceramic Institute, Jing-De-Zhen, China
| | - Wang-Ren Qiu
- Computer Department, Jing-De-Zhen Ceramic Institute, Jing-De-Zhen, China
| |
Collapse
|
20
|
Ahmed F, Lee JW, Samantasinghar A, Kim YS, Kim KH, Kang IS, Memon FH, Lim JH, Choi KH. SperoPredictor: An Integrated Machine Learning and Molecular Docking-Based Drug Repurposing Framework With Use Case of COVID-19. Front Public Health 2022; 10:902123. [PMID: 35784208 PMCID: PMC9244710 DOI: 10.3389/fpubh.2022.902123] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/02/2022] [Indexed: 12/13/2022] Open
Abstract
The global spread of the SARS coronavirus 2 (SARS-CoV-2), its manifestation in human hosts as a contagious disease, and its variants have induced a pandemic resulting in the deaths of over 6,000,000 people. Extensive efforts have been devoted to drug research to cure and refrain the spread of COVID-19, but only one drug has received FDA approval yet. Traditional drug discovery is inefficient, costly, and unable to react to pandemic threats. Drug repurposing represents an effective strategy for drug discovery and reduces the time and cost compared to de novo drug discovery. In this study, a generic drug repurposing framework (SperoPredictor) has been developed which systematically integrates the various types of drugs and disease data and takes the advantage of machine learning (Random Forest, Tree Ensemble, and Gradient Boosted Trees) to repurpose potential drug candidates against any disease of interest. Drug and disease data for FDA-approved drugs (n = 2,865), containing four drug features and three disease features, were collected from chemical and biological databases and integrated with the form of drug-disease association tables. The resulting dataset was split into 70% for training, 15% for testing, and the remaining 15% for validation. The testing and validation accuracies of the models were 99.3% for Random Forest and 99.03% for Tree Ensemble. In practice, SperoPredictor identified 25 potential drug candidates against 6 human host-target proteomes identified from a systematic review of journals. Literature-based validation indicated 12 of 25 predicted drugs (48%) have been already used for COVID-19 followed by molecular docking and re-docking which indicated 4 of 13 drugs (30%) as potential candidates against COVID-19 to be pre-clinically and clinically validated. Finally, SperoPredictor results illustrated the ability of the platform to be rapidly deployed to repurpose the drugs as a rapid response to emergent situations (like COVID-19 and other pandemics).
Collapse
Affiliation(s)
- Faheem Ahmed
- Department of Mechatronics Engineering, Jeju National University, Jeju, South Korea
| | - Jae Wook Lee
- Department of Mechatronics Engineering, Jeju National University, Jeju, South Korea
- BioSpero, Inc., Jeju, South Korea
| | | | | | - Kyung Hwan Kim
- Department of Mechatronics Engineering, Jeju National University, Jeju, South Korea
| | - In Suk Kang
- Department of Mechatronics Engineering, Jeju National University, Jeju, South Korea
| | - Fida Hussain Memon
- Department of Mechatronics Engineering, Jeju National University, Jeju, South Korea
| | - Jong Hwan Lim
- Department of Mechatronics Engineering, Jeju National University, Jeju, South Korea
| | - Kyung Hyun Choi
- Department of Mechatronics Engineering, Jeju National University, Jeju, South Korea
- BioSpero, Inc., Jeju, South Korea
| |
Collapse
|
21
|
Rout RK, Umer S, Sheikh S, Sindhwani S, Pati S. EightyDVec: a method for protein sequence similarity analysis using physicochemical properties of amino acids. COMPUTER METHODS IN BIOMECHANICS AND BIOMEDICAL ENGINEERING: IMAGING & VISUALIZATION 2022. [DOI: 10.1080/21681163.2021.1956369] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Ranjeet Kumar Rout
- Computer Science & Engineering, National Institute of Technology Srinagar, Hazratbal, India
| | - Saiyed Umer
- Computer Science & Engineering, Aliah University, West Bengal, India
| | - Sabha Sheikh
- Computer Science & Engineering, National Institute of Technology Srinagar, Hazratbal, India
| | - Sanchit Sindhwani
- , DR. B. R. Ambedkar National Institute of Technology, Jalandhar, Punjab, India
| | - Smitarani Pati
- , DR. B. R. Ambedkar National Institute of Technology, Jalandhar, Punjab, India
| |
Collapse
|
22
|
Alakus TB, Turkoglu I. A Comparative Study of Amino Acid Encoding Methods for Predicting Drug-Target Interactions in COVID-19 Disease. MODELING, CONTROL AND DRUG DEVELOPMENT FOR COVID-19 OUTBREAK PREVENTION 2022:619-643. [DOI: 10.1007/978-3-030-72834-2_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
|
23
|
Jin Y, Lu J, Shi R, Yang Y. EmbedDTI: Enhancing the Molecular Representations via Sequence Embedding and Graph Convolutional Network for the Prediction of Drug-Target Interaction. Biomolecules 2021; 11:biom11121783. [PMID: 34944427 PMCID: PMC8698792 DOI: 10.3390/biom11121783] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/20/2021] [Accepted: 11/24/2021] [Indexed: 01/09/2023] Open
Abstract
The identification of drug-target interaction (DTI) plays a key role in drug discovery and development. Benefitting from large-scale drug databases and verified DTI relationships, a lot of machine-learning methods have been developed to predict DTIs. However, due to the difficulty in extracting useful information from molecules, the performance of these methods is limited by the representation of drugs and target proteins. This study proposes a new model called EmbedDTI to enhance the representation of both drugs and target proteins, and improve the performance of DTI prediction. For protein sequences, we leverage language modeling for pretraining the feature embeddings of amino acids and feed them to a convolutional neural network model for further representation learning. For drugs, we build two levels of graphs to represent compound structural information, namely the atom graph and substructure graph, and adopt graph convolutional network with an attention module to learn the embedding vectors for the graphs. We compare EmbedDTI with the existing DTI predictors on two benchmark datasets. The experimental results show that EmbedDTI outperforms the state-of-the-art models, and the attention module can identify the components crucial for DTIs in compounds.
Collapse
Affiliation(s)
- Yuan Jin
- Center for Brain-Like Computing and Machine Intelligence, Department of Computer Science and Engineering, Shanghai Jiao Tong University, 800 Dong Chuan Rd., Shanghai 200240, China; (Y.J.); (R.S.)
| | - Jiarui Lu
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dong Chuan Rd., Shanghai 200240, China;
| | - Runhan Shi
- Center for Brain-Like Computing and Machine Intelligence, Department of Computer Science and Engineering, Shanghai Jiao Tong University, 800 Dong Chuan Rd., Shanghai 200240, China; (Y.J.); (R.S.)
| | - Yang Yang
- Center for Brain-Like Computing and Machine Intelligence, Department of Computer Science and Engineering, Shanghai Jiao Tong University, 800 Dong Chuan Rd., Shanghai 200240, China; (Y.J.); (R.S.)
- Key Laboratory of Shanghai Education Commission for Intelligent Interaction and Cognitive Engineering, Shanghai Jiao Tong University, 800 Dong Chuan Rd., Shanghai 200240, China
- Correspondence:
| |
Collapse
|
24
|
Sorkhi AG, Abbasi Z, Mobarakeh MI, Pirgazi J. Drug-target interaction prediction using unifying of graph regularized nuclear norm with bilinear factorization. BMC Bioinformatics 2021; 22:555. [PMID: 34789169 PMCID: PMC8597250 DOI: 10.1186/s12859-021-04464-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 10/29/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Wet-lab experiments for identification of interactions between drugs and target proteins are time-consuming, costly and labor-intensive. The use of computational prediction of drug-target interactions (DTIs), which is one of the significant points in drug discovery, has been considered by many researchers in recent years. It also reduces the search space of interactions by proposing potential interaction candidates. RESULTS In this paper, a new approach based on unifying matrix factorization and nuclear norm minimization is proposed to find a low-rank interaction. In this combined method, to solve the low-rank matrix approximation, the terms in the DTI problem are used in such a way that the nuclear norm regularized problem is optimized by a bilinear factorization based on Rank-Restricted Soft Singular Value Decomposition (RRSSVD). In the proposed method, adjacencies between drugs and targets are encoded by graphs. Drug-target interaction, drug-drug similarity, target-target, and combination of similarities have also been used as input. CONCLUSIONS The proposed method is evaluated on four benchmark datasets known as Enzymes (E), Ion channels (ICs), G protein-coupled receptors (GPCRs) and nuclear receptors (NRs) based on AUC, AUPR, and time measure. The results show an improvement in the performance of the proposed method compared to the state-of-the-art techniques.
Collapse
Affiliation(s)
- Ali Ghanbari Sorkhi
- Faculty of Electrical and Computer Engineering, University of Science and Technology of Mazandaran, P.O. Box, 48518-78195 Behshahr, Iran
| | - Zahra Abbasi
- School of Medicine, Faculty of Medical Biotechnology, Shahroud University of Medical Sciences, Shahroud, Iran
| | | | - Jamshid Pirgazi
- Faculty of Electrical and Computer Engineering, University of Science and Technology of Mazandaran, P.O. Box, 48518-78195 Behshahr, Iran
| |
Collapse
|
25
|
Hu S, Xia D, Su B, Chen P, Wang B, Li J. A Convolutional Neural Network System to Discriminate Drug-Target Interactions. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2021; 18:1315-1324. [PMID: 31514149 DOI: 10.1109/tcbb.2019.2940187] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Biological targets are most commonly proteins such as enzymes, ion channels, and receptors. They are anything within a living organism to bind with some other entities (like an endogenous ligand or a drug), resulting in change in their behaviors or functions. Exploring potential drug-target interactions (DTIs) are crucial for drug discovery and effective drug development. Computational methods were widely applied in drug-target interactions, since experimental methods are extremely time-consuming and resource-intensive. In this paper, we proposed a novel deep learning-based prediction system, with a new negative instance generation, to identify DTIs. As a result, our method achieved an accuracy of 0.9800 on our created dataset. Another dataset derived from DrugBank was used to further assess the generalization of the model, which yielded a good performance with accuracy of 0.8814 and AUC value of 0.9527 on the dataset. The outcome of our experimental results indicated that the proposed method, involving the credible negative generation, can be employed to discriminate the interactions between drugs and targets. Website: http://www.dlearningapp.com/web/DrugCNN.htm.
Collapse
|
26
|
Predicting Drug-Target Interactions Based on the Ensemble Models of Multiple Feature Pairs. Int J Mol Sci 2021; 22:ijms22126598. [PMID: 34202954 PMCID: PMC8234024 DOI: 10.3390/ijms22126598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/09/2021] [Accepted: 06/16/2021] [Indexed: 11/30/2022] Open
Abstract
Backgroud: The prediction of drug–target interactions (DTIs) is of great significance in drug development. It is time-consuming and expensive in traditional experimental methods. Machine learning can reduce the cost of prediction and is limited by the characteristics of imbalanced datasets and problems of essential feature selection. Methods: The prediction method based on the Ensemble model of Multiple Feature Pairs (Ensemble-MFP) is introduced. Firstly, three negative sets are generated according to the Euclidean distance of three feature pairs. Then, the negative samples of the validation set/test set are randomly selected from the union set of the three negative sets in the validation set/test set. At the same time, the ensemble model with weight is optimized and applied to the test set. Results: The area under the receiver operating characteristic curve (area under ROC, AUC) in three out of four sub-datasets in gold standard datasets was more than 94.0% in the prediction of new drugs. The effectiveness of the proposed method is also shown with the comparison of state-of-the-art methods and demonstration of predicted drug–target pairs. Conclusion: The Ensemble-MFP can weigh the existing feature pairs and has a good prediction effect for general prediction on new drugs.
Collapse
|
27
|
Yuan T, Werman JM, Sampson NS. The pursuit of mechanism of action: uncovering drug complexity in TB drug discovery. RSC Chem Biol 2021; 2:423-440. [PMID: 33928253 PMCID: PMC8081351 DOI: 10.1039/d0cb00226g] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 12/23/2020] [Indexed: 12/21/2022] Open
Abstract
Whole cell-based phenotypic screens have become the primary mode of hit generation in tuberculosis (TB) drug discovery during the last two decades. Different drug screening models have been developed to mirror the complexity of TB disease in the laboratory. As these culture conditions are becoming more and more sophisticated, unraveling the drug target and the identification of the mechanism of action (MOA) of compounds of interest have additionally become more challenging. A good understanding of MOA is essential for the successful delivery of drug candidates for TB treatment due to the high level of complexity in the interactions between Mycobacterium tuberculosis (Mtb) and the TB drug used to treat the disease. There is no single "standard" protocol to follow and no single approach that is sufficient to fully investigate how a drug restrains Mtb. However, with the recent advancements in -omics technologies, there are multiple strategies that have been developed generally in the field of drug discovery that have been adapted to comprehensively characterize the MOAs of TB drugs in the laboratory. These approaches have led to the successful development of preclinical TB drug candidates, and to a better understanding of the pathogenesis of Mtb infection. In this review, we describe a plethora of efforts based upon genetic, metabolomic, biochemical, and computational approaches to investigate TB drug MOAs. We assess these different platforms for their strengths and limitations in TB drug MOA elucidation in the context of Mtb pathogenesis. With an emphasis on the essentiality of MOA identification, we outline the unmet needs in delivering TB drug candidates and provide direction for further TB drug discovery.
Collapse
Affiliation(s)
- Tianao Yuan
- Department of Chemistry, Stony Brook UniversityStony BrookNY 11794-3400USA+1-631-632-5738+1-631-632-7952
| | - Joshua M. Werman
- Department of Chemistry, Stony Brook UniversityStony BrookNY 11794-3400USA+1-631-632-5738+1-631-632-7952
| | - Nicole S. Sampson
- Department of Chemistry, Stony Brook UniversityStony BrookNY 11794-3400USA+1-631-632-5738+1-631-632-7952
| |
Collapse
|
28
|
Mahmud SMH, Chen W, Liu Y, Awal MA, Ahmed K, Rahman MH, Moni MA. PreDTIs: prediction of drug-target interactions based on multiple feature information using gradient boosting framework with data balancing and feature selection techniques. Brief Bioinform 2021; 22:6168499. [PMID: 33709119 PMCID: PMC7989622 DOI: 10.1093/bib/bbab046] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 01/25/2021] [Accepted: 01/29/2021] [Indexed: 12/13/2022] Open
Abstract
Discovering drug–target (protein) interactions (DTIs) is of great significance for researching and developing novel drugs, having a tremendous advantage to pharmaceutical industries and patients. However, the prediction of DTIs using wet-lab experimental methods is generally expensive and time-consuming. Therefore, different machine learning-based methods have been developed for this purpose, but there are still substantial unknown interactions needed to discover. Furthermore, data imbalance and feature dimensionality problems are a critical challenge in drug-target datasets, which can decrease the classifier performances that have not been significantly addressed yet. This paper proposed a novel drug–target interaction prediction method called PreDTIs. First, the feature vectors of the protein sequence are extracted by the pseudo-position-specific scoring matrix (PsePSSM), dipeptide composition (DC) and pseudo amino acid composition (PseAAC); and the drug is encoded with MACCS substructure fingerings. Besides, we propose a FastUS algorithm to handle the class imbalance problem and also develop a MoIFS algorithm to remove the irrelevant and redundant features for getting the best optimal features. Finally, balanced and optimal features are provided to the LightGBM Classifier to identify DTIs, and the 5-fold CV validation test method was applied to evaluate the prediction ability of the proposed method. Prediction results indicate that the proposed model PreDTIs is significantly superior to other existing methods in predicting DTIs, and our model could be used to discover new drugs for unknown disorders or infections, such as for the coronavirus disease 2019 using existing drugs compounds and severe acute respiratory syndrome coronavirus 2 protein sequences.
Collapse
Affiliation(s)
- S M Hasan Mahmud
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Wenyu Chen
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Yongsheng Liu
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Md Abdul Awal
- Electronics and Communication Engineering Discipline, Khulna University, Khulna 9208, Bangladesh
| | - Kawsar Ahmed
- Department of Information and Communication Technology, Mawlana Bhashani Science and Technology University, Santosh, Tangail-1902, Bangladesh
| | - Md Habibur Rahman
- Department of Computer Science and Engineering, Islamic University, Kushtia-7003, Bangladesh
| | - Mohammad Ali Moni
- UNSW Digital Health, WHO Center for eHealth, School of Public Health and Community Medicine, Faculty of Medicine, The University of New South Wales, Sydney, Australia
| |
Collapse
|
29
|
Zhou M, Zheng C, Xu R. Combining phenome-driven drug-target interaction prediction with patients' electronic health records-based clinical corroboration toward drug discovery. Bioinformatics 2021; 36:i436-i444. [PMID: 32657406 PMCID: PMC7355254 DOI: 10.1093/bioinformatics/btaa451] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Motivation Predicting drug–target interactions (DTIs) using human phenotypic data have the potential in eliminating the translational gap between animal experiments and clinical outcomes in humans. One challenge in human phenome-driven DTI predictions is integrating and modeling diverse drug and disease phenotypic relationships. Leveraging large amounts of clinical observed phenotypes of drugs and diseases and electronic health records (EHRs) of 72 million patients, we developed a novel integrated computational drug discovery approach by seamlessly combining DTI prediction and clinical corroboration. Results We developed a network-based DTI prediction system (TargetPredict) by modeling 855 904 phenotypic and genetic relationships among 1430 drugs, 4251 side effects, 1059 diseases and 17 860 genes. We systematically evaluated TargetPredict in de novo cross-validation and compared it to a state-of-the-art phenome-driven DTI prediction approach. We applied TargetPredict in identifying novel repositioned candidate drugs for Alzheimer’s disease (AD), a disease affecting over 5.8 million people in the United States. We evaluated the clinical efficiency of top repositioned drug candidates using EHRs of over 72 million patients. The area under the receiver operating characteristic (ROC) curve was 0.97 in the de novo cross-validation when evaluated using 910 drugs. TargetPredict outperformed a state-of-the-art phenome-driven DTI prediction system as measured by precision–recall curves [measured by average precision (MAP): 0.28 versus 0.23, P-value < 0.0001]. The EHR-based case–control studies identified that the prescriptions top-ranked repositioned drugs are significantly associated with lower odds of AD diagnosis. For example, we showed that the prescription of liraglutide, a type 2 diabetes drug, is significantly associated with decreased risk of AD diagnosis [adjusted odds ratios (AORs): 0.76; 95% confidence intervals (CI) (0.70, 0.82), P-value < 0.0001]. In summary, our integrated approach that seamlessly combines computational DTI prediction and large-scale patients’ EHRs-based clinical corroboration has high potential in rapidly identifying novel drug targets and drug candidates for complex diseases. Availability and implementation nlp.case.edu/public/data/TargetPredict.
Collapse
Affiliation(s)
- Mengshi Zhou
- Center for Artificial Intelligence in Drug Discovery, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA.,Department of Population and Quantitative Health Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Chunlei Zheng
- Center for Artificial Intelligence in Drug Discovery, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Rong Xu
- Center for Artificial Intelligence in Drug Discovery, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
30
|
Recent Advances in Predicting Protein S-Nitrosylation Sites. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5542224. [PMID: 33628788 PMCID: PMC7892234 DOI: 10.1155/2021/5542224] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 01/24/2021] [Accepted: 01/25/2021] [Indexed: 01/09/2023]
Abstract
Protein S-nitrosylation (SNO) is a process of covalent modification of nitric oxide (NO) and its derivatives and cysteine residues. SNO plays an essential role in reversible posttranslational modifications of proteins. The accurate prediction of SNO sites is crucial in revealing a certain biological mechanism of NO regulation and related drug development. Identification of the sites of SNO in proteins is currently a very hot topic. In this review, we briefly summarize recent advances in computationally identifying SNO sites. The challenges and future perspectives for identifying SNO sites are also discussed. We anticipate that this review will provide insights into research on SNO site prediction.
Collapse
|
31
|
Qiu W, Lv Z, Hong Y, Jia J, Xiao X. BOW-GBDT: A GBDT Classifier Combining With Artificial Neural Network for Identifying GPCR-Drug Interaction Based on Wordbook Learning From Sequences. Front Cell Dev Biol 2021; 8:623858. [PMID: 33598456 PMCID: PMC7882597 DOI: 10.3389/fcell.2020.623858] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/15/2020] [Indexed: 12/28/2022] Open
Abstract
Background: As a class of membrane protein receptors, G protein-coupled receptors (GPCRs) are very important for cells to complete normal life function and have been proven to be a major drug target for widespread clinical application. Hence, it is of great significance to find GPCR targets that interact with drugs in the process of drug development. However, identifying the interaction of the GPCR–drug pairs by experimental methods is very expensive and time-consuming on a large scale. As more and more database about GPCR–drug pairs are opened, it is viable to develop machine learning models to accurately predict whether there is an interaction existing in a GPCR–drug pair. Methods: In this paper, the proposed model aims to improve the accuracy of predicting the interactions of GPCR–drug pairs. For GPCRs, the work extracts protein sequence features based on a novel bag-of-words (BOW) model improved with weighted Silhouette Coefficient and has been confirmed that it can extract more pattern information and limit the dimension of feature. For drug molecules, discrete wavelet transform (DWT) is used to extract features from the original molecular fingerprints. Subsequently, the above-mentioned two types of features are contacted, and SMOTE algorithm is selected to balance the training dataset. Then, artificial neural network is used to extract features further. Finally, a gradient boosting decision tree (GBDT) model is trained with the selected features. In this paper, the proposed model is named as BOW-GBDT. Results: D92M and Check390 are selected for testing BOW-GBDT. D92M is used for a cross-validation dataset which contains 635 interactive GPCR–drug pairs and 1,225 non-interactive pairs. Check390 is used for an independent test dataset which consists of 130 interactive GPCR–drug pairs and 260 non-interactive GPCR–drug pairs, and each element in Check390 cannot be found in D92M. According to the results, the proposed model has a better performance in generation ability compared with the existing machine learning models. Conclusion: The proposed predictor improves the accuracy of the interactions of GPCR–drug pairs. In order to facilitate more researchers to use the BOW-GBDT, the predictor has been settled into a brand-new server, which is available at http://www.jci-bioinfo.cn/bowgbdt.
Collapse
Affiliation(s)
- Wangren Qiu
- School of Information Engineering, Jingdezhen Ceramic Institute, Jingdezhen, China
| | - Zhe Lv
- School of Information Engineering, Jingdezhen Ceramic Institute, Jingdezhen, China
| | - Yaoqiu Hong
- School of Information Engineering, Jingdezhen University, Jingdezhen, China
| | - Jianhua Jia
- School of Information Engineering, Jingdezhen Ceramic Institute, Jingdezhen, China
| | - Xuan Xiao
- School of Information Engineering, Jingdezhen Ceramic Institute, Jingdezhen, China
| |
Collapse
|
32
|
Wang C, Kurgan L. Survey of Similarity-Based Prediction of Drug-Protein Interactions. Curr Med Chem 2021; 27:5856-5886. [PMID: 31393241 DOI: 10.2174/0929867326666190808154841] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 04/16/2018] [Accepted: 10/23/2018] [Indexed: 12/20/2022]
Abstract
Therapeutic activity of a significant majority of drugs is determined by their interactions with proteins. Databases of drug-protein interactions (DPIs) primarily focus on the therapeutic protein targets while the knowledge of the off-targets is fragmented and partial. One way to bridge this knowledge gap is to employ computational methods to predict protein targets for a given drug molecule, or interacting drugs for given protein targets. We survey a comprehensive set of 35 methods that were published in high-impact venues and that predict DPIs based on similarity between drugs and similarity between protein targets. We analyze the internal databases of known PDIs that these methods utilize to compute similarities, and investigate how they are linked to the 12 publicly available source databases. We discuss contents, impact and relationships between these internal and source databases, and well as the timeline of their releases and publications. The 35 predictors exploit and often combine three types of similarities that consider drug structures, drug profiles, and target sequences. We review the predictive architectures of these methods, their impact, and we explain how their internal DPIs databases are linked to the source databases. We also include a detailed timeline of the development of these predictors and discuss the underlying limitations of the current resources and predictive tools. Finally, we provide several recommendations concerning the future development of the related databases and methods.
Collapse
Affiliation(s)
- Chen Wang
- Department of Computer Science, Virginia Commonwealth University, Richmond, VA 23284, United States
| | - Lukasz Kurgan
- Department of Computer Science, Virginia Commonwealth University, Richmond, VA 23284, United States
| |
Collapse
|
33
|
Peng J, Lu G, Shang X. A Survey of Network Representation Learning Methods for Link Prediction in Biological Network. Curr Pharm Des 2021; 26:3076-3084. [PMID: 31951161 DOI: 10.2174/1381612826666200116145057] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 01/09/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Networks are powerful resources for describing complex systems. Link prediction is an important issue in network analysis and has important practical application value. Network representation learning has proven to be useful for network analysis, especially for link prediction tasks. OBJECTIVE To review the application of network representation learning on link prediction in a biological network, we summarize recent methods for link prediction in a biological network and discuss the application and significance of network representation learning in link prediction task. METHOD & RESULTS We first introduce the widely used link prediction algorithms, then briefly introduce the development of network representation learning methods, focusing on a few widely used methods, and their application in biological network link prediction. Existing studies demonstrate that using network representation learning to predict links in biological networks can achieve better performance. In the end, some possible future directions have been discussed.
Collapse
Affiliation(s)
- Jiajie Peng
- School of Computer Science, Northwestern Polytechnical University, Xi'an, China
| | - Guilin Lu
- School of Computer Science, Northwestern Polytechnical University, Xi'an, China
| | - Xuequn Shang
- School of Computer Science, Northwestern Polytechnical University, Xi'an, China
| |
Collapse
|
34
|
Karasev D, Sobolev B, Lagunin A, Filimonov D, Poroikov V. Prediction of Protein-ligand Interaction Based on Sequence Similarity and Ligand Structural Features. Int J Mol Sci 2020; 21:ijms21218152. [PMID: 33142754 PMCID: PMC7663273 DOI: 10.3390/ijms21218152] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 01/09/2023] Open
Abstract
Computationally predicting the interaction of proteins and ligands presents three main directions: the search of new target proteins for ligands, the search of new ligands for targets, and predicting the interaction of new proteins and new ligands. We proposed an approach providing the fuzzy classification of protein sequences based on the ligand structural features to analyze the latter most complicated case. We tested our approach on five protein groups, which represented promised targets for drug-like ligands and differed in functional peculiarities. The training sets were built with the original procedure overcoming the data ambiguity. Our study showed the effective prediction of new targets for ligands with an average accuracy of 0.96. The prediction of new ligands for targets displayed the average accuracy 0.95; accuracy estimates were close to our previous results, comparable in accuracy to those of other methods or exceeded them. Using the fuzzy coefficients reflecting the target-to-ligand specificity, we provided predicting interactions for new proteins and new ligands; the obtained accuracy values from 0.89 to 0.99 were acceptable for such a sophisticated task. The protein kinase family case demonstrated the ability to account for subtle features of proteins and ligands required for the specificity of protein–ligand interaction.
Collapse
Affiliation(s)
- Dmitry Karasev
- Department of Bioinformatics, Institute of Biomedical Chemistry, Moscow 119121, Russia; (B.S.); (A.L.); (D.F.); (V.P.)
- Correspondence:
| | - Boris Sobolev
- Department of Bioinformatics, Institute of Biomedical Chemistry, Moscow 119121, Russia; (B.S.); (A.L.); (D.F.); (V.P.)
| | - Alexey Lagunin
- Department of Bioinformatics, Institute of Biomedical Chemistry, Moscow 119121, Russia; (B.S.); (A.L.); (D.F.); (V.P.)
- Department of Bioinformatics, Russian National Research Medical University, Moscow 117997, Russia
| | - Dmitry Filimonov
- Department of Bioinformatics, Institute of Biomedical Chemistry, Moscow 119121, Russia; (B.S.); (A.L.); (D.F.); (V.P.)
| | - Vladimir Poroikov
- Department of Bioinformatics, Institute of Biomedical Chemistry, Moscow 119121, Russia; (B.S.); (A.L.); (D.F.); (V.P.)
| |
Collapse
|
35
|
Hasan Mahmud SM, Chen W, Jahan H, Dai B, Din SU, Dzisoo AM. DeepACTION: A deep learning-based method for predicting novel drug-target interactions. Anal Biochem 2020; 610:113978. [PMID: 33035462 DOI: 10.1016/j.ab.2020.113978] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 09/23/2020] [Accepted: 09/25/2020] [Indexed: 12/13/2022]
Abstract
Drug-target interactions (DTIs) play a key role in drug development and discovery processes. Wet lab prediction of DTIs is time-consuming, expensive, and tedious. Fortunately, computational approaches can identify new interactions (drug-target pairs) and accelerate the process of drug repurposing. However, a vast number of interactions remain undiscovered; therefore, we proposed a deep learning-based method (deepACTION) for predicting potential or unknown DTIs. Here, each drug chemical structure and protein sequence are transformed according to structural and sequence information using different descriptors to represent their features correctly. There have been some challenges, such as the high dimensionality and class imbalance of data during the prediction process. To address these problems, we developed the MMIB technique to balance the majority and minority instances in the dataset and utilized a LASSO model to handle the high dimensionality of the data. In addition, we trained the convolutional neural network algorithm with balanced and reduced features for accurate prediction of DTIs. In this study, the AUC is considered a primary evaluation metric for comparing the performance of the deep ACTION model with that of existing methods by a 5-fold cross-validation test. Our experiential dataset obtained from the DrugBank database and our deepACTION model achieved an AUC of 0.9836 for this dataset. The experimental results ensured that the model can predict significant numbers of new DTIs and provide complete information to motivate scientists to develop drugs.
Collapse
Affiliation(s)
- S M Hasan Mahmud
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Wenyu Chen
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu, 611731, China.
| | - Hosney Jahan
- College of Computer Science, Sichuan University, Chengdu, 610065, China
| | - Bo Dai
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Salah Ud Din
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Anthony Mackitz Dzisoo
- Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| |
Collapse
|
36
|
Martynowycz MW, Gonen T. Ligand Incorporation into Protein Microcrystals for MicroED by On-Grid Soaking. Structure 2020; 29:88-95.e2. [PMID: 33007196 DOI: 10.1016/j.str.2020.09.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/29/2020] [Accepted: 09/15/2020] [Indexed: 11/17/2022]
Abstract
A high throughout method for soaking ligands into protein microcrystals on TEM grids is presented. Every crystal on the grid is soaked simultaneously using only standard cryoelectron microscopy vitrification equipment. The method is demonstrated using proteinase K microcrystals soaked with the 5-amino-2,4,6-triodoisophthalic acid (I3C) magic triangle. A soaked microcrystal is milled to a thickness of approximately 200 nm using a focused ion beam, and MicroED data are collected. A high-resolution structure of the protein with four ligands at high occupancy is determined. Both the number of ligands bound and their occupancy is higher using on-grid soaking of microcrystals compared with much larger crystals treated similarly and investigated by X-ray crystallography. These results indicate that on-grid soaking ligands into microcrystals results in efficient uptake of ligands into protein microcrystals.
Collapse
Affiliation(s)
- Michael W Martynowycz
- Department of Biological Chemistry, University of California Los Angeles, 615 Charles E Young Drive South, Los Angeles, CA 90095, USA; Department of Physiology, University of California Los Angeles, 615 Charles E Young Drive South, Los Angeles, CA 90095, USA; Howard Hughes Medical Institute, University of California Los Angeles, Los Angeles CA90095, USA
| | - Tamir Gonen
- Department of Biological Chemistry, University of California Los Angeles, 615 Charles E Young Drive South, Los Angeles, CA 90095, USA; Department of Physiology, University of California Los Angeles, 615 Charles E Young Drive South, Los Angeles, CA 90095, USA; Howard Hughes Medical Institute, University of California Los Angeles, Los Angeles CA90095, USA.
| |
Collapse
|
37
|
Wang C, Wang W, Lu K, Zhang J, Chen P, Wang B. Predicting Drug-Target Interactions with Electrotopological State Fingerprints and Amphiphilic Pseudo Amino Acid Composition. Int J Mol Sci 2020; 21:ijms21165694. [PMID: 32784497 PMCID: PMC7570185 DOI: 10.3390/ijms21165694] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 08/05/2020] [Accepted: 08/06/2020] [Indexed: 12/13/2022] Open
Abstract
The task of drug-target interaction (DTI) prediction plays important roles in drug development. The experimental methods in DTIs are time-consuming, expensive and challenging. To solve these problems, machine learning-based methods are introduced, which are restricted by effective feature extraction and negative sampling. In this work, features with electrotopological state (E-state) fingerprints for drugs and amphiphilic pseudo amino acid composition (APAAC) for target proteins are tested. E-state fingerprints are extracted based on both molecular electronic and topological features with the same metric. APAAC is an extension of amino acid composition (AAC), which is calculated based on hydrophilic and hydrophobic characters to construct sequence order information. Using the combination of these feature pairs, the prediction model is established by support vector machines. In order to enhance the effectiveness of features, a distance-based negative sampling is proposed to obtain reliable negative samples. It is shown that the prediction results of area under curve for Receiver Operating Characteristic (AUC) are above 98.5% for all the three datasets in this work. The comparison of state-of-the-art methods demonstrates the effectiveness and efficiency of proposed method, which will be helpful for further drug development.
Collapse
Affiliation(s)
- Cheng Wang
- Department of Computer Science & Technology, Tongji University, Shanghai 201804, China;
| | - Wenyan Wang
- School of Electrical & Information Engineering, Anhui University of Technology, Ma’anshan 243002, China; (W.W.); (K.L.)
- Key Laboratory of Power Electronics and Motion Control Anhui Education Department, Ma’anshan 243032, China
| | - Kun Lu
- School of Electrical & Information Engineering, Anhui University of Technology, Ma’anshan 243002, China; (W.W.); (K.L.)
| | - Jun Zhang
- Institutes of Physical Science and Information Technology & School of Internet, Anhui University, Hefei 230601, China;
| | - Peng Chen
- Institutes of Physical Science and Information Technology & School of Internet, Anhui University, Hefei 230601, China;
- Correspondence: (P.C.); (B.W.)
| | - Bing Wang
- Department of Computer Science & Technology, Tongji University, Shanghai 201804, China;
- School of Electrical & Information Engineering, Anhui University of Technology, Ma’anshan 243002, China; (W.W.); (K.L.)
- Key Laboratory of Power Electronics and Motion Control Anhui Education Department, Ma’anshan 243032, China
- Correspondence: (P.C.); (B.W.)
| |
Collapse
|
38
|
Sachdev K, Gupta MK. A hybrid ensemble-based technique for predicting drug-target interactions. Chem Biol Drug Des 2020; 96:1447-1455. [PMID: 32638508 DOI: 10.1111/cbdd.13753] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/14/2020] [Accepted: 06/14/2020] [Indexed: 11/30/2022]
Abstract
Drug-target interaction is the intercommunication between chemical drugs and target proteins to produce any kind of change in the human body. The laboratory experiments conducted to recognize these potential intercommunications are costly and tedious. Various computational methods have been established recently, including the chemogenomic approach for identifying drug-target intercommunication, that integrates the chemical data of the drugs and the genomic properties of the proteins for identifying the interactions between them. This paper proposes a novel technique based on hybrid ensemble to predict drug target interactions. Hybrid ensembles introduce diversity in classification that helps to improve the performance of prediction. The proposed method has been evaluated by comparing the technique with state-of-the-art methods on two different databases under three cross-validation settings. The comparison clearly shows that the technique produced significant improvement in the interaction prediction.
Collapse
Affiliation(s)
- Kanica Sachdev
- School of Computer Science and Engineering, Shri Mata Vaishno Devi University, Jammu and Kashmir, India
| | - Manoj Kumar Gupta
- School of Computer Science and Engineering, Shri Mata Vaishno Devi University, Jammu and Kashmir, India
| |
Collapse
|
39
|
Gong J, Chen Y, Pu F, Sun P, He F, Zhang L, Li Y, Ma Z, Wang H. Understanding Membrane Protein Drug Targets in Computational Perspective. Curr Drug Targets 2020; 20:551-564. [PMID: 30516106 DOI: 10.2174/1389450120666181204164721] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 09/03/2018] [Accepted: 09/04/2018] [Indexed: 01/16/2023]
Abstract
Membrane proteins play crucial physiological roles in vivo and are the major category of drug targets for pharmaceuticals. The research on membrane protein is a significant part in the drug discovery. The biological process is a cycled network, and the membrane protein is a vital hub in the network since most drugs achieve the therapeutic effect via interacting with the membrane protein. In this review, typical membrane protein targets are described, including GPCRs, transporters and ion channels. Also, we conclude network servers and databases that are referring to the drug, drug-target information and their relevant data. Furthermore, we chiefly introduce the development and practice of modern medicines, particularly demonstrating a series of state-of-the-art computational models for the prediction of drug-target interaction containing network-based approach and machine-learningbased approach as well as showing current achievements. Finally, we discuss the prospective orientation of drug repurposing and drug discovery as well as propose some improved framework in bioactivity data, created or improved predicted approaches, alternative understanding approaches of drugs bioactivity and their biological processes.
Collapse
Affiliation(s)
- Jianting Gong
- School of Information Science and Technology, Northeast Normal University, Changchun, China.,Institution of Computational Biology, Northeast Normal University, Changchun, China
| | - Yongbing Chen
- School of Information Science and Technology, Northeast Normal University, Changchun, China.,Institution of Computational Biology, Northeast Normal University, Changchun, China
| | - Feng Pu
- School of Information Science and Technology, Northeast Normal University, Changchun, China.,Institution of Computational Biology, Northeast Normal University, Changchun, China
| | - Pingping Sun
- School of Information Science and Technology, Northeast Normal University, Changchun, China.,Institution of Computational Biology, Northeast Normal University, Changchun, China
| | - Fei He
- School of Information Science and Technology, Northeast Normal University, Changchun, China.,Institution of Computational Biology, Northeast Normal University, Changchun, China
| | - Li Zhang
- School of Computer Science and Engineering, Changchun University of Technology, Changchun, China
| | - Yanwen Li
- School of Information Science and Technology, Northeast Normal University, Changchun, China.,Institution of Computational Biology, Northeast Normal University, Changchun, China
| | - Zhiqiang Ma
- School of Information Science and Technology, Northeast Normal University, Changchun, China.,Institution of Computational Biology, Northeast Normal University, Changchun, China
| | - Han Wang
- School of Information Science and Technology, Northeast Normal University, Changchun, China.,Institution of Computational Biology, Northeast Normal University, Changchun, China
| |
Collapse
|
40
|
Wang L, You ZH, Li LP, Yan X, Zhang W, Song KJ, Song CD. Identification of potential drug-targets by combining evolutionary information extracted from frequency profiles and molecular topological structures. Chem Biol Drug Des 2020; 96:758-767. [PMID: 31393672 DOI: 10.1111/cbdd.13599] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/29/2019] [Accepted: 08/03/2019] [Indexed: 01/09/2023]
Abstract
Identifying interactions among drug compounds and target proteins is the basis of drug research and plays a crucial role in drug discovery. However, determining drug-target interactions (DTIs) and potential protein-compound interactions by biological experiment-based method alone is a very complicated, expensive, and time-consuming process. Hence, there is an intense motivation to design in silico prediction methods to overcome these obstacles. In this work, we designed a novel in silico strategy to predict proteome-scale DTIs based on the assumption that DTI pairs can be expressed through the evolutionary information derived from frequency profiles and drugs' structural properties. To achieve this, drug molecules are encoded into the substructure fingerprints to represent certain fragments; target proteins are first converted into position-specific scoring matrix (PSSM) and then encoded as 2-dimensional principal component analysis (2DPCA) descriptors. In the prediction phase, the feature weighted rotation forest (RF) classifier is used to estimate whether drug and target interact with each other on four benchmark datasets, including Enzymes, Ion Channels, GPCRs, and Nuclear Receptors. The prediction accuracy of cross-validation on the four datasets is 95.40%, 88.82%, 85.67%, and 82.22%, respectively. In order to have a clearer assessment of the proposed approach, we compared it with the discrete cosine transform (DCT) descriptor model, support vector machine (SVM) classifier model, and existing excellent approaches, including DBSI, NetCBP, KBMF2K, SIMCOMP, and RFDT. The excellent results of the experiment indicated that the proposed approach can effectively improve the DTI prediction accuracy and can be used as a practical tool for the research and design of new drugs.
Collapse
Affiliation(s)
- Lei Wang
- College of Information Science and Engineering, Zaozhuang University, Zaozhuang, China.,Xinjiang Technical Institutes of Physics and Chemistry, Chinese Academy of Science, Urumqi, China
| | - Zhu-Hong You
- Xinjiang Technical Institutes of Physics and Chemistry, Chinese Academy of Science, Urumqi, China
| | - Li-Ping Li
- Xinjiang Technical Institutes of Physics and Chemistry, Chinese Academy of Science, Urumqi, China
| | - Xin Yan
- School of Foreign Languages, Zaozhuang University, Zaozhuang, China
| | - Wei Zhang
- College of Information Science and Engineering, Zaozhuang University, Zaozhuang, China
| | - Ke-Jian Song
- School of information engineering, JiangXi University of Science and Technology, Ganzhou, China
| | - Chuan-Dong Song
- College of Information Science and Engineering, Zaozhuang University, Zaozhuang, China
| |
Collapse
|
41
|
Che J, Chen L, Guo ZH, Wang S, Aorigele. Drug Target Group Prediction with Multiple Drug Networks. Comb Chem High Throughput Screen 2020; 23:274-284. [DOI: 10.2174/1386207322666190702103927] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 03/11/2019] [Accepted: 04/15/2019] [Indexed: 02/07/2023]
Abstract
Background:
Identification of drug-target interaction is essential in drug discovery. It is
beneficial to predict unexpected therapeutic or adverse side effects of drugs. To date, several
computational methods have been proposed to predict drug-target interactions because they are
prompt and low-cost compared with traditional wet experiments.
Methods:
In this study, we investigated this problem in a different way. According to KEGG,
drugs were classified into several groups based on their target proteins. A multi-label classification
model was presented to assign drugs into correct target groups. To make full use of the known drug
properties, five networks were constructed, each of which represented drug associations in one
property. A powerful network embedding method, Mashup, was adopted to extract drug features
from above-mentioned networks, based on which several machine learning algorithms, including
RAndom k-labELsets (RAKEL) algorithm, Label Powerset (LP) algorithm and Support Vector
Machine (SVM), were used to build the classification model.
Results and Conclusion:
Tenfold cross-validation yielded the accuracy of 0.839, exact match of
0.816 and hamming loss of 0.037, indicating good performance of the model. The contribution of
each network was also analyzed. Furthermore, the network model with multiple networks was
found to be superior to the one with a single network and classic model, indicating the superiority
of the proposed model.
Collapse
Affiliation(s)
- Jingang Che
- College of Information Engineering, Shanghai Maritime University, Shanghai 201306, China
| | - Lei Chen
- College of Information Engineering, Shanghai Maritime University, Shanghai 201306, China
| | - Zi-Han Guo
- College of Information Engineering, Shanghai Maritime University, Shanghai 201306, China
| | - Shuaiqun Wang
- College of Information Engineering, Shanghai Maritime University, Shanghai 201306, China
| | - Aorigele
- Faculty of Engineering, University of Toyama, Toyama, Japan
| |
Collapse
|
42
|
Zeng X, Zhu S, Hou Y, Zhang P, Li L, Li J, Huang LF, Lewis SJ, Nussinov R, Cheng F. Network-based prediction of drug-target interactions using an arbitrary-order proximity embedded deep forest. Bioinformatics 2020; 36:2805-2812. [PMID: 31971579 PMCID: PMC7203727 DOI: 10.1093/bioinformatics/btaa010] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 12/24/2019] [Accepted: 01/17/2020] [Indexed: 11/14/2022] Open
Abstract
MOTIVATION Systematic identification of molecular targets among known drugs plays an essential role in drug repurposing and understanding of their unexpected side effects. Computational approaches for prediction of drug-target interactions (DTIs) are highly desired in comparison to traditional experimental assays. Furthermore, recent advances of multiomics technologies and systems biology approaches have generated large-scale heterogeneous, biological networks, which offer unexpected opportunities for network-based identification of new molecular targets among known drugs. RESULTS In this study, we present a network-based computational framework, termed AOPEDF, an arbitrary-order proximity embedded deep forest approach, for prediction of DTIs. AOPEDF learns a low-dimensional vector representation of features that preserve arbitrary-order proximity from a highly integrated, heterogeneous biological network connecting drugs, targets (proteins) and diseases. In total, we construct a heterogeneous network by uniquely integrating 15 networks covering chemical, genomic, phenotypic and network profiles among drugs, proteins/targets and diseases. Then, we build a cascade deep forest classifier to infer new DTIs. Via systematic performance evaluation, AOPEDF achieves high accuracy in identifying molecular targets among known drugs on two external validation sets collected from DrugCentral [area under the receiver operating characteristic curve (AUROC) = 0.868] and ChEMBL (AUROC = 0.768) databases, outperforming several state-of-the-art methods. In a case study, we showcase that multiple molecular targets predicted by AOPEDF are associated with mechanism-of-action of substance abuse disorder for several marketed drugs (such as aripiprazole, risperidone and haloperidol). AVAILABILITY AND IMPLEMENTATION Source code and data can be downloaded from https://github.com/ChengF-Lab/AOPEDF.
Collapse
Affiliation(s)
- Xiangxiang Zeng
- Department of Computer Science, College of Information Science and Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Siyi Zhu
- Department of Computer Science, Xiamen University, Xiamen 361005, China
| | - Yuan Hou
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Pengyue Zhang
- Department of Biomedical Informatics, Ohio State University, Columbus, OH 43210, USA
| | - Lang Li
- Department of Biomedical Informatics, Ohio State University, Columbus, OH 43210, USA
| | - Jing Li
- Department of Computer and Data Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - L Frank Huang
- Division of Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children’s Hospital Medical Center
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Stephen J Lewis
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Ruth Nussinov
- Computational Structural Biology Section, Basic Science Program, Frederick National Laboratory for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
43
|
Kaushik AC, Mehmood A, Dai X, Wei DQ. A comparative chemogenic analysis for predicting Drug-Target Pair via Machine Learning Approaches. Sci Rep 2020; 10:6870. [PMID: 32322011 PMCID: PMC7176722 DOI: 10.1038/s41598-020-63842-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 04/04/2020] [Indexed: 12/26/2022] Open
Abstract
A computational technique for predicting the DTIs has now turned out to be an indispensable job during the process of drug finding. It tapers the exploration room for interactions by propounding possible interaction contenders for authentication through experiments of wet-lab which are known for their expensiveness and time consumption. Chemogenomics, an emerging research area focused on the systematic examination of the biological impact of a broad series of minute molecular-weighting ligands on a broad raiment of macromolecular target spots. Additionally, with the advancement in time, the complexity of the algorithms is increasing which may result in the entry of big data technologies like Spark in this field soon. In the presented work, we intend to offer an inclusive idea and realistic evaluation of the computational Drug Target Interaction projection approaches, to perform as a guide and reference for researchers who are carrying out work in a similar direction. Precisely, we first explain the data utilized in computational Drug Target Interaction prediction attempts like this. We then sort and explain the best and most modern techniques for the prediction of DTIs. Then, a realistic assessment is executed to show the projection performance of several illustrative approaches in various situations. Ultimately, we underline possible opportunities for additional improvement of Drug Target Interaction projection enactment and also linked study objectives.
Collapse
Affiliation(s)
- Aman Chandra Kaushik
- Wuxi School of Medicine, Jiangnan University, Wuxi, China.
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China.
| | - Aamir Mehmood
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Xiaofeng Dai
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Dong-Qing Wei
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China.
| |
Collapse
|
44
|
Wang P, Huang X, Qiu W, Xiao X. Identifying GPCR-drug interaction based on wordbook learning from sequences. BMC Bioinformatics 2020; 21:150. [PMID: 32312232 PMCID: PMC7171867 DOI: 10.1186/s12859-020-3488-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 04/13/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND G protein-coupled receptors (GPCRs) mediate a variety of important physiological functions, are closely related to many diseases, and constitute the most important target family of modern drugs. Therefore, the research of GPCR analysis and GPCR ligand screening is the hotspot of new drug development. Accurately identifying the GPCR-drug interaction is one of the key steps for designing GPCR-targeted drugs. However, it is prohibitively expensive to experimentally ascertain the interaction of GPCR-drug pairs on a large scale. Therefore, it is of great significance to predict the interaction of GPCR-drug pairs directly from the molecular sequences. With the accumulation of known GPCR-drug interaction data, it is feasible to develop sequence-based machine learning models for query GPCR-drug pairs. RESULTS In this paper, a new sequence-based method is proposed to identify GPCR-drug interactions. For GPCRs, we use a novel bag-of-words (BoW) model to extract sequence features, which can extract more pattern information from low-order to high-order and limit the feature space dimension. For drug molecules, we use discrete Fourier transform (DFT) to extract higher-order pattern information from the original molecular fingerprints. The feature vectors of two kinds of molecules are concatenated and input into a simple prediction engine distance-weighted K-nearest-neighbor (DWKNN). This basic method is easy to be enhanced through ensemble learning. Through testing on recently constructed GPCR-drug interaction datasets, it is found that the proposed methods are better than the existing sequence-based machine learning methods in generalization ability, even an unconventional method in which the prediction performance was further improved by post-processing procedure (PPP). CONCLUSIONS The proposed methods are effective for GPCR-drug interaction prediction, and may also be potential methods for other target-drug interaction prediction, or protein-protein interaction prediction. In addition, the new proposed feature extraction method for GPCR sequences is the modified version of the traditional BoW model and may be useful to solve problems of protein classification or attribute prediction. The source code of the proposed methods is freely available for academic research at https://github.com/wp3751/GPCR-Drug-Interaction.
Collapse
Affiliation(s)
- Pu Wang
- Computer School, Hubei University of Arts and Science, Xiangyang, 441053 China
| | - Xiaotong Huang
- Computer School, Hubei University of Arts and Science, Xiangyang, 441053 China
| | - Wangren Qiu
- Computer Department, Jingdezhen Ceramic Institute, Jingdezhen, 333403 China
| | - Xuan Xiao
- Computer Department, Jingdezhen Ceramic Institute, Jingdezhen, 333403 China
| |
Collapse
|
45
|
Rayhan F, Ahmed S, Mousavian Z, Farid DM, Shatabda S. FRnet-DTI: Deep convolutional neural network for drug-target interaction prediction. Heliyon 2020; 6:e03444. [PMID: 32154410 PMCID: PMC7052404 DOI: 10.1016/j.heliyon.2020.e03444] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 06/16/2019] [Accepted: 02/14/2020] [Indexed: 01/09/2023] Open
Abstract
The task of drug-target interaction prediction holds significant importance in pharmacology and therapeutic drug design. In this paper, we present FRnet-DTI, an auto-encoder based feature manipulation and a convolutional neural network based classifier for drug target interaction prediction. Two convolutional neural networks are proposed: FRnet-Encode and FRnet-Predict. Here, one model is used for feature manipulation and the other one for classification. Using the first method FRnet-Encode, we generate 4096 features for each of the instances in each of the datasets and use the second method, FRnet-Predict, to identify interaction probability employing those features. We have tested our method on four gold standard datasets extensively used by other researchers. Experimental results shows that our method significantly improves over the state-of-the-art method on three out of four drug-target interaction gold standard datasets on both area under curve for Receiver Operating Characteristic (auROC) and area under Precision Recall curve (auPR) metric. We also introduce twenty new potential drug-target pairs for interaction based on high prediction scores. The source codes and implementation details of our methods are available from https://github.com/farshidrayhanuiu/FRnet-DTI/ and also readily available to use as an web application from http://farshidrayhan.pythonanywhere.com/FRnet-DTI/.
Collapse
Affiliation(s)
- Farshid Rayhan
- Department of Computer Science and Engineering, United International University, Plot 2, United City, Madani Avenue, Satarkul, Badda, Dhaka-1212, Bangladesh
| | - Sajid Ahmed
- Department of Computer Science and Engineering, United International University, Plot 2, United City, Madani Avenue, Satarkul, Badda, Dhaka-1212, Bangladesh
| | - Zaynab Mousavian
- School of Mathematics, Statistics, and Computer Science, College of Science, University of Tehran, Tehran, Iran
| | - Dewan Md Farid
- Department of Computer Science and Engineering, United International University, Plot 2, United City, Madani Avenue, Satarkul, Badda, Dhaka-1212, Bangladesh
| | - Swakkhar Shatabda
- Department of Computer Science and Engineering, United International University, Plot 2, United City, Madani Avenue, Satarkul, Badda, Dhaka-1212, Bangladesh
| |
Collapse
|
46
|
Redkar S, Mondal S, Joseph A, Hareesha KS. A Machine Learning Approach for Drug-target Interaction Prediction using Wrapper Feature Selection and Class Balancing. Mol Inform 2020; 39:e1900062. [PMID: 32003548 DOI: 10.1002/minf.201900062] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 01/28/2020] [Indexed: 01/19/2023]
Abstract
Drug-Target interaction (DTI) plays a crucial role in drug discovery, drug repositioning and understanding the drug side effects which helps to identify new therapeutic profiles for various diseases. However, the exponential growth in the genomic and drugs data makes it difficult to identify the new associations between drugs and targets. Therefore, we use computational methods as it helps in accelerating the DTI identification process. Usually, available data driven sources consisting of known DTI is used to train the classifier to predict the new DTIs. Such datasets often face the problem of class imbalance. Therefore, in this study we address two challenges faced by such datasets, i. e., class imbalance and high dimensionality to develop a predictive model for DTI prediction. The study is carried out on four protein classes namely Enzyme, Ion Channel, G Protein-Coupled Receptor (GPCR) and Nuclear Receptor. We encoded the target protein sequence using the dipeptide composition and drug with a molecular descriptor. A machine learning approach is employed to predict the DTI using wrapper feature selection and synthetic minority oversampling technique (SMOTE). The ensemble approach achieved at the best an accuracy of 95.9 %, 93.4 %, 90.8 % and 90.6 % and 96.3 %, 92.8 %, 90.1 %, and 90.2 % of precision on Enzyme, Ion Channel, GPCR and Nuclear Receptor datasets, respectively, when evaluated excluding SMOTE samples with 10-fold cross validation. Furthermore, our method could predict new drug-target interactions not contained in training dataset. Selected features using wrapper feature selection may be important to understand the DTI for the protein categories under this study. Based on our evaluation, the proposed method can be used for understanding and identifying new drug-target interactions. We provide the readers with a standalone package available at https://github.com/shwetagithub1/predDTI which will be able to provide the DTI predictions to user for new query DTI pairs.
Collapse
Affiliation(s)
- Shweta Redkar
- Department of Computer Applications, Manipal Institute of Technology, Manipal Academy of Higher Education, 576104, Manipal, Karnataka, India
| | - Sukanta Mondal
- Department of Biological Sciences, Birla Institute of Technology and Science-Pilani, K.K.Birla Goa Campus, 403726, Zuarinagar, Goa, -India
| | - Alex Joseph
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, 576104, Manipal, Karnataka, India
| | - K S Hareesha
- Department of Computer Applications, Manipal Institute of Technology, Manipal Academy of Higher Education, 576104, Manipal, Karnataka, India
| |
Collapse
|
47
|
Bagherian M, Sabeti E, Wang K, Sartor MA, Nikolovska-Coleska Z, Najarian K. Machine learning approaches and databases for prediction of drug-target interaction: a survey paper. Brief Bioinform 2020; 22:247-269. [PMID: 31950972 PMCID: PMC7820849 DOI: 10.1093/bib/bbz157] [Citation(s) in RCA: 201] [Impact Index Per Article: 40.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 11/01/2019] [Accepted: 11/07/2019] [Indexed: 12/12/2022] Open
Abstract
The task of predicting the interactions between drugs and targets plays a key role in the process of drug discovery. There is a need to develop novel and efficient prediction approaches in order to avoid costly and laborious yet not-always-deterministic experiments to determine drug–target interactions (DTIs) by experiments alone. These approaches should be capable of identifying the potential DTIs in a timely manner. In this article, we describe the data required for the task of DTI prediction followed by a comprehensive catalog consisting of machine learning methods and databases, which have been proposed and utilized to predict DTIs. The advantages and disadvantages of each set of methods are also briefly discussed. Lastly, the challenges one may face in prediction of DTI using machine learning approaches are highlighted and we conclude by shedding some lights on important future research directions.
Collapse
Affiliation(s)
- Maryam Bagherian
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Elyas Sabeti
- Michigan Institute for Data Science, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kai Wang
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Maureen A Sartor
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | | | - Kayvan Najarian
- Department of Electrical Engineering and Computer Science, College of Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
48
|
Mongia A, Majumdar A. Drug-target interaction prediction using Multi Graph Regularized Nuclear Norm Minimization. PLoS One 2020; 15:e0226484. [PMID: 31945078 PMCID: PMC6964976 DOI: 10.1371/journal.pone.0226484] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 11/27/2019] [Indexed: 01/09/2023] Open
Abstract
The identification of potential interactions between drugs and target proteins is crucial in pharmaceutical sciences. The experimental validation of interactions in genomic drug discovery is laborious and expensive; hence, there is a need for efficient and accurate in-silico techniques which can predict potential drug-target interactions to narrow down the search space for experimental verification. In this work, we propose a new framework, namely, Multi-Graph Regularized Nuclear Norm Minimization, which predicts the interactions between drugs and target proteins from three inputs: known drug-target interaction network, similarities over drugs and those over targets. The proposed method focuses on finding a low-rank interaction matrix that is structured by the proximities of drugs and targets encoded by graphs. Previous works on Drug Target Interaction (DTI) prediction have shown that incorporating drug and target similarities helps in learning the data manifold better by preserving the local geometries of the original data. But, there is no clear consensus on which kind and what combination of similarities would best assist the prediction task. Hence, we propose to use various multiple drug-drug similarities and target-target similarities as multiple graph Laplacian (over drugs/targets) regularization terms to capture the proximities exhaustively. Extensive cross-validation experiments on four benchmark datasets using standard evaluation metrics (AUPR and AUC) show that the proposed algorithm improves the predictive performance and outperforms recent state-of-the-art computational methods by a large margin. Software is publicly available at https://github.com/aanchalMongia/MGRNNMforDTI.
Collapse
Affiliation(s)
- Aanchal Mongia
- Dept. of Computer Science and Engineering, IIIT-Delhi, Delhi, India
| | - Angshul Majumdar
- Dept. of Electronics and Communications Engineering, IIIT-Delhi, Delhi, India
| |
Collapse
|
49
|
Zhang YF, Wang X, Kaushik AC, Chu Y, Shan X, Zhao MZ, Xu Q, Wei DQ. SPVec: A Word2vec-Inspired Feature Representation Method for Drug-Target Interaction Prediction. Front Chem 2020; 7:895. [PMID: 31998687 PMCID: PMC6967417 DOI: 10.3389/fchem.2019.00895] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 12/12/2019] [Indexed: 11/13/2022] Open
Abstract
Drug discovery is an academical and commercial process of global importance. Accurate identification of drug-target interactions (DTIs) can significantly facilitate the drug discovery process. Compared to the costly, labor-intensive and time-consuming experimental methods, machine learning (ML) plays an ever-increasingly important role in effective, efficient and high-throughput identification of DTIs. However, upstream feature extraction methods require tremendous human resources and expert insights, which limits the application of ML approaches. Inspired by the unsupervised representation learning methods like Word2vec, we here proposed SPVec, a novel way to automatically represent raw data such as SMILES strings and protein sequences into continuous, information-rich and lower-dimensional vectors, so as to avoid the sparseness and bit collisions from the cumbersomely manually extracted features. Visualization of SPVec nicely illustrated that the similar compounds or proteins occupy similar vector space, which indicated that SPVec not only encodes compound substructures or protein sequences efficiently, but also implicitly reveals some important biophysical and biochemical patterns. Compared with manually-designed features like MACCS fingerprints and amino acid composition (AAC), SPVec showed better performance with several state-of-art machine learning classifiers such as Gradient Boosting Decision Tree, Random Forest and Deep Neural Network on BindingDB. The performance and robustness of SPVec were also confirmed on independent test sets obtained from DrugBank database. Also, based on the whole DrugBank dataset, we predicted the possibilities of all unlabeled DTIs, where two of the top five predicted novel DTIs were supported by external evidences. These results indicated that SPVec can provide an effective and efficient way to discover reliable DTIs, which would be beneficial for drug reprofiling.
Collapse
Affiliation(s)
- Yu-Fang Zhang
- State Key Laboratory of Microbial Metabolism, and SJTU-Yale Joint Center for Biostatistics and Data Science, School of Life Sciences and Biotechnology, and Joint Laboratory of International Cooperation in Metabolic and Developmental Sciences, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Xiangeng Wang
- State Key Laboratory of Microbial Metabolism, and SJTU-Yale Joint Center for Biostatistics and Data Science, School of Life Sciences and Biotechnology, and Joint Laboratory of International Cooperation in Metabolic and Developmental Sciences, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Aman Chandra Kaushik
- State Key Laboratory of Microbial Metabolism, and SJTU-Yale Joint Center for Biostatistics and Data Science, School of Life Sciences and Biotechnology, and Joint Laboratory of International Cooperation in Metabolic and Developmental Sciences, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China.,Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yanyi Chu
- State Key Laboratory of Microbial Metabolism, and SJTU-Yale Joint Center for Biostatistics and Data Science, School of Life Sciences and Biotechnology, and Joint Laboratory of International Cooperation in Metabolic and Developmental Sciences, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoqi Shan
- State Key Laboratory of Microbial Metabolism, and SJTU-Yale Joint Center for Biostatistics and Data Science, School of Life Sciences and Biotechnology, and Joint Laboratory of International Cooperation in Metabolic and Developmental Sciences, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Ming-Zhu Zhao
- Instrumental Analysis Center, Shanghai Jiao Tong University, Shanghai, China
| | - Qin Xu
- State Key Laboratory of Microbial Metabolism, and SJTU-Yale Joint Center for Biostatistics and Data Science, School of Life Sciences and Biotechnology, and Joint Laboratory of International Cooperation in Metabolic and Developmental Sciences, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Dong-Qing Wei
- State Key Laboratory of Microbial Metabolism, and SJTU-Yale Joint Center for Biostatistics and Data Science, School of Life Sciences and Biotechnology, and Joint Laboratory of International Cooperation in Metabolic and Developmental Sciences, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China.,Peng Cheng Laboratory, Shenzhen, China
| |
Collapse
|
50
|
Hu S, Zhang C, Chen P, Gu P, Zhang J, Wang B. Predicting drug-target interactions from drug structure and protein sequence using novel convolutional neural networks. BMC Bioinformatics 2019; 20:689. [PMID: 31874614 PMCID: PMC6929541 DOI: 10.1186/s12859-019-3263-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background Accurate identification of potential interactions between drugs and protein targets is a critical step to accelerate drug discovery. Despite many relative experimental researches have been done in the past decades, detecting drug-target interactions (DTIs) remains to be extremely resource-intensive and time-consuming. Therefore, many computational approaches have been developed for predicting drug-target associations on a large scale. Results In this paper, we proposed an deep learning-based method to predict DTIs only using the information of drug structures and protein sequences. The final results showed that our method can achieve good performance with the accuracies up to 92.0%, 90.0%, 92.0% and 90.7% for the target families of enzymes, ion channels, GPCRs and nuclear receptors of our created dataset, respectively. Another dataset derived from DrugBank was used to further assess the generalization of the model, which yielded an accuracy of 0.9015 and an AUC value of 0.9557. Conclusion It was elucidated that our model shows improved performance in comparison with other state-of-the-art computational methods on the common benchmark datasets. Experimental results demonstrated that our model successfully extracted more nuanced yet useful features, and therefore can be used as a practical tool to discover new drugs. Availability http://deeplearner.ahu.edu.cn/web/CnnDTI.htm.
Collapse
Affiliation(s)
- ShanShan Hu
- School of Computer Science and Technology, Anhui University, Jiulong Road, Hefei, 230601, China
| | - Chenglin Zhang
- Institutes of Physical Science and Information Technology, Anhui University, Jiulong Road, Hefei, 230601, China
| | - Peng Chen
- School of Computer Science and Technology, Anhui University, Jiulong Road, Hefei, 230601, China. .,Institutes of Physical Science and Information Technology, Anhui University, Jiulong Road, Hefei, 230601, China. .,Cadre's Ward (South District), The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China.
| | - Pengying Gu
- Cadre's Ward (South District), The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China.
| | - Jun Zhang
- School of Electrical and Information Engineering, Anhui University, Hefei, 230601, China
| | - Bing Wang
- School of Electrical and Information Engineering, Anhui University of Technology, Ma'anshan, 243032, China
| |
Collapse
|