1
|
Hu X, Sun H, Shan L, Ma C, Quan H, Zhang Y, Zhang J, Fan Z, Tang Y, Deng L. Unraveling Disease-Associated PIWI-Interacting RNAs with a Contrastive Learning Methods. J Chem Inf Model 2025; 65:4687-4697. [PMID: 40263714 DOI: 10.1021/acs.jcim.5c00173] [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: 04/24/2025]
Abstract
PIWI-interacting RNAs (piRNAs) are a class of small, non-coding RNAs predominantly expressed in the germ cells of animals and play a crucial role in maintaining genomic integrity, mediating transposon suppression, and ensuring gene stability. Beyond their functions in reproductive cells, piRNAs also play roles in various human diseases, including cancer, suggesting their potential as significant biomarkers critical for disease diagnosis and treatment. Wet-lab methods to identify piRNA-disease associations require substantial resources and are often hit-or-miss. With advancements in computational technologies, an increasing number of researchers are employing computational methods to efficiently predict potential piRNA-disease associations. The sparsity of data in piRNA-disease association studies significantly limits model performance improvement. In this study, we propose a novel computational model, iPiDA_CL, to predict potential piRNA-disease associations through contrastive learning methods, which do not require negative samples. The model represents piRNA-disease association pairs as a bipartite graph and computes the initial embeddings of piRNAs and diseases using Gaussian kernel similarity, with features updated via LightGCN. Based on the siamese network framework, iPiDA_CL constructs online and target networks and employs data augmentation in the target network to build a contrastive learning objective that optimizes model parameters without introducing negative samples. Finally, cross-prediction methods are used to calculate specific piRNA-disease association scores. A series of experimental results demonstrate that iPiDA_CL surpasses state-of-the-art methods in both performance and computational efficiency. The application of iPiDA_CL to the miRNA-disease association dataset underscores its versatility across various ncRNA-disease association task. Furthermore, a case study highlights iPiDA_CL as an efficient and promising tool for predicting piRNA-disease associations.
Collapse
Affiliation(s)
- Xiaowen Hu
- School of Computer Science and Engineering, Center South University, Changsha 410083, China
| | - Hao Sun
- School of Computer Science and Engineering, Center South University, Changsha 410083, China
| | - Linchao Shan
- School of Computer Science and Engineering, Center South University, Changsha 410083, China
| | - Chenxi Ma
- School of Computer Science and Engineering, Center South University, Changsha 410083, China
| | - Hanming Quan
- School of Computer Science and Engineering, Center South University, Changsha 410083, China
| | - Yuanpeng Zhang
- School of software, Xinjiang University, Urumqi 830049, China
| | - Jiaxuan Zhang
- Department of Electrical and Computer Engineering, University of California, San Diego, California 92161, United States
| | - Ziyu Fan
- School of Computer Science and Engineering, Center South University, Changsha 410083, China
| | - Yongjun Tang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha 410083, China
| | - Lei Deng
- School of Computer Science and Engineering, Center South University, Changsha 410083, China
| |
Collapse
|
2
|
Yu D, Yang X, Shang Y, Yuan S, Liu Y, Liu Y. Drug-target interaction prediction based on metapaths and simplified neighbor aggregation. Methods 2025; 240:154-164. [PMID: 40288620 DOI: 10.1016/j.ymeth.2025.04.012] [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: 01/25/2025] [Revised: 04/02/2025] [Accepted: 04/20/2025] [Indexed: 04/29/2025] Open
Abstract
Drug-target interaction (DTI) prediction is critical in drug repositioning and discovery. In current metapath-based prediction methods, attention mechanisms are often used to differentiate the importance of various neighbors, enhancing the model's expressiveness. However, in biological networks with small-scale imbalanced data, attention mechanisms are prone to interference from noise and missing data, leading to instability in weight learning, reduced efficiency, and an increased risk of overfitting. To address these issues, we propose the use of average aggregation to mitigate noise, simplify model complexity, and improve stability. Specifically, we introduce a simplified mean aggregation method for DTI prediction. This approach uses average aggregation, effectively reducing noise interference, lowering model complexity, and preventing overfitting, making it especially suitable for current biological networks. Extensive testing on three heterogeneous biological datasets shows that SNADTI outperforms 12 leading methods across two evaluation metrics, significantly reducing training time and validating its effectiveness in DTI prediction. Complexity analysis reveals that our method offers a substantial computational speed advantage over other methods on the same dataset, highlighting its enhanced efficiency. Experimental results demonstrate that SNADTI excels in prediction accuracy, stability, and reproducibility, confirming its practicality and effectiveness in DTI prediction.
Collapse
Affiliation(s)
- Di Yu
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, 410082, Hunan, China
| | - Xinyu Yang
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, 410082, Hunan, China
| | - Yifan Shang
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, 410082, Hunan, China; Department of Biomedical Engineering, The Chinese University of Hong Kong, 999077, Hong Kong, China.
| | - Sisi Yuan
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, 28223, NC, USA
| | - Yuansheng Liu
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, 410082, Hunan, China
| | - Yiping Liu
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, 410082, Hunan, China
| |
Collapse
|
3
|
Zuo Y, Wu X, Ge F, Yan H, Fei S, Liang J, Deng Z. Research progress on Drug-Target Interactions in the last five years. Anal Biochem 2025; 697:115691. [PMID: 39455038 DOI: 10.1016/j.ab.2024.115691] [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: 08/20/2024] [Revised: 10/06/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024]
Abstract
The identification of Drug-Target Interaction (DTI) is an important step in drug discovery and drug repositioning, and has high application value in multiple fields such as drug discovery, drug repositioning, and repurposing. However, the high cost of experimental validation limits its identification. In contrast, computation-based approaches are both economical and efficient. This review first synthesizes existing chemical genomic approaches, provides a comprehensive summary of prevalent databases for predicting DTIs, and categorizes the feature encodings from recent years. This is followed by an overview and brief description of the methods currently in use for predicting DTIs. The strengths and weaknesses of newly proposed prediction methods in the last five years (2020-2024), including those based on network representation learning and graph neural networks, are then discussed in detail, evaluating the performance of the different methods on a wide range of datasets. Finally, this review explores potential directions for future DTI research, emphasizing how to improve prediction accuracy and efficiency by combining big data and emerging computing technologies.
Collapse
Affiliation(s)
- Yun Zuo
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, 214000, China.
| | - Xubin Wu
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, 214000, China
| | - Fei Ge
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, 214000, China
| | - Hongjin Yan
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, 214000, China
| | - Sirui Fei
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, 214000, China
| | - Jingwen Liang
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, 214000, China
| | - Zhaohong Deng
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, 214000, China.
| |
Collapse
|
4
|
He C, Zhao Z, Wang X, Zheng H, Duan L, Zuo J. Exploring drug-target interaction prediction on cold-start scenarios via meta-learning-based graph transformer. Methods 2025; 234:10-20. [PMID: 39550022 DOI: 10.1016/j.ymeth.2024.11.010] [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: 05/30/2024] [Revised: 11/07/2024] [Accepted: 11/12/2024] [Indexed: 11/18/2024] Open
Abstract
Predicting drug-target interaction (DTI) is of great importance for drug discovery and development. With the rapid development of biological and chemical technologies, computational methods for DTI prediction are becoming a promising approach. However, there are few solutions to the cold-start problem in DTI prediction scenarios, as these methods rely on existing interaction information to support their modeling. Consequently, they are unable to effectively predict DTIs for new drugs or targets with limited interaction data in the existing work. To this end, we propose a graph transformer method based on meta-learning named MGDTI (short for Meta-learning-based Graph Transformer for Drug-Target Interaction prediction) to fill this gap. Technically, we employ drug-drug similarity and target-target similarity as additional information to mitigate the scarcity of interactions. Besides, we trained MGDTI via meta-learning to be adaptive to cold-start tasks. Moreover, we employed graph transformer to prevent over-smoothing by capturing long-range dependencies. Extensive results on the benchmark dataset demonstrate that MGDTI is effective on DTI prediction under cold-start scenarios.
Collapse
Affiliation(s)
- Chengxin He
- School of Computer Science, Sichuan University, Chengdu 610065, China; College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Zhenjiang Zhao
- School of Computer Science, Sichuan University, Chengdu 610065, China
| | - Xinye Wang
- School of Computer Science, Sichuan University, Chengdu 610065, China
| | - Huiru Zheng
- School of Computing, Ulster University, Belfast BT15 1ED, Northern Ireland, UK
| | - Lei Duan
- School of Computer Science, Sichuan University, Chengdu 610065, China
| | - Jie Zuo
- School of Computer Science, Sichuan University, Chengdu 610065, China.
| |
Collapse
|
5
|
Sun L, Yin Z, Lu L. ISLRWR: A network diffusion algorithm for drug-target interactions prediction. PLoS One 2025; 20:e0302281. [PMID: 39883675 PMCID: PMC11781719 DOI: 10.1371/journal.pone.0302281] [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: 12/15/2023] [Accepted: 04/01/2024] [Indexed: 02/01/2025] Open
Abstract
Machine learning techniques and computer-aided methods are now widely used in the pre-discovery tasks of drug discovery, effectively improving the efficiency of drug development and reducing the workload and cost. In this study, we used multi-source heterogeneous network information to build a network model, learn the network topology through multiple network diffusion algorithms, and obtain compressed low-dimensional feature vectors for predicting drug-target interactions (DTIs). We applied the metropolis-hasting random walk (MHRW) algorithm to improve the performance of the random walk with restart (RWR) algorithm, forming the basis by which the self-loop probability of the current node is removed. Additionally, the propagation efficiency of the MHRW was improved using the improved metropolis-hasting random walk (IMRWR) algorithm, facilitating network deep sampling. Finally, we proposed a correction of the transfer probability of the entire network after increasing the self-loop rate of isolated nodes to form the ISLRWR algorithm. Notably, the ISLRWR algorithm improved the area under the receiver operating characteristic curve (AUROC) by 7.53 and 5.72%, and the area under the precision-recall curve (AUPRC) by 5.95 and 4.19% compared to the RWR and MHRW algorithms, respectively, in predicting DTIs performance. Moreover, after excluding the interference of homologous proteins (popular drugs or targets may lead to inflated prediction results), the ISLRWR algorithm still showed a significant performance improvement.
Collapse
Affiliation(s)
- Lu Sun
- School of Mathematics, Physics and Statistics, Institute for Frontier Medical Technology, Center of Intelligent Computing and Applied Statistics, Shanghai University of Engineering Science, Shanghai, China
| | - Zhixiang Yin
- School of Mathematics, Physics and Statistics, Institute for Frontier Medical Technology, Center of Intelligent Computing and Applied Statistics, Shanghai University of Engineering Science, Shanghai, China
| | - Lin Lu
- Shanghai Xinhao Information Technology Co., Ltd., Shanghai, China
| |
Collapse
|
6
|
Zhang B, Quan L, Zhang Z, Cao L, Chen Q, Peng L, Wang J, Jiang Y, Nie L, Li G, Wu T, Lyu Q. MVCL-DTI: Predicting Drug-Target Interactions Using a Multiview Contrastive Learning Model on a Heterogeneous Graph. J Chem Inf Model 2025; 65:1009-1026. [PMID: 39812134 DOI: 10.1021/acs.jcim.4c02073] [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: 01/16/2025]
Abstract
Accurate prediction of drug-target interactions (DTIs) is pivotal for accelerating the processes of drug discovery and drug repurposing. MVCL-DTI, a novel model leveraging heterogeneous graphs for predicting DTIs, tackles the challenge of synthesizing information from varied biological subnetworks. It integrates neighbor view, meta-path view, and diffusion view to capture semantic features and employs an attention-based contrastive learning approach, along with a multiview attention-weighted fusion module, to effectively integrate and adaptively weight the information from the different views. Tested under various conditions on benchmark data sets, including varying positive-to-negative sample ratios, conducting hard negative sampling experiments, and masking known DTIs with different ratios, as well as redundant DTIs with various similarity metrics, MVCL-DTI exhibits strong robust generalization. The model is then employed to predict novel DTIs, with a particular focus on COVID-19-related drugs, highlighting its practical applicability. Ultimately, through features visualization and computational properties analysis, we've pinpointed critical elements, including Gene Ontology and substituent nodes, along with a proper initialization strategy, underscoring their vital role in DTI prediction tasks.
Collapse
Affiliation(s)
- Bei Zhang
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
- China Mobile (Suzhou) Software Technology Company Limited, Suzhou 215163, China
| | - Lijun Quan
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
- Collaborative Innovation Center of Novel Software Technology and Industrialization, Jiangsu 210000, China
| | - Zhijun Zhang
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
| | - Lexin Cao
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
| | - Qiufeng Chen
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
| | - Liangchen Peng
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
| | - Junkai Wang
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
| | - Yelu Jiang
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
| | - Liangpeng Nie
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
| | - Geng Li
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
| | - Tingfang Wu
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
- Collaborative Innovation Center of Novel Software Technology and Industrialization, Jiangsu 210000, China
| | - Qiang Lyu
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
- Collaborative Innovation Center of Novel Software Technology and Industrialization, Jiangsu 210000, China
| |
Collapse
|
7
|
Zhao PC, Wei XX, Wang Q, Wang QH, Li JN, Shang J, Lu C, Shi JY. Single-step retrosynthesis prediction via multitask graph representation learning. Nat Commun 2025; 16:814. [PMID: 39827189 PMCID: PMC11742932 DOI: 10.1038/s41467-025-56062-y] [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: 08/10/2023] [Accepted: 01/08/2025] [Indexed: 01/22/2025] Open
Abstract
Inferring appropriate synthesis reaction (i.e., retrosynthesis) routes for newly designed molecules is vital. Recently, computational methods have produced promising single-step retrosynthesis predictions. However, template-based methods are limited by the known synthesis templates; template-free methods are weakly interpretable; and semi template-based methods are deficient with regard to utilizing the associations between chemical entities. To address these issues, this paper leverages the intra-associations between synthons, the inter-associations between synthons and leaving groups (LGs), and the intra-associations between LGs. It develops a multitask graph representation learning model for single-step retrosynthesis prediction (Retro-MTGR) to solve reaction centre deduction and LG identification simultaneously. A comparison with 16 state-of-the-art methods first demonstrates the superiority of Retro-MTGR. Then, its robustness and scalability and the contributions of its crucial components are validated. More importantly, it can determine whether a bond can be a reaction centre and what LGs are appropriate for a given synthon, respectively. The answers reflect underlying chemical synthesis rules, especially opposite electrical properties between chemical entities (e.g., reaction sites, synthons, and LGs). Finally, case studies demonstrate that the retrosynthesis routes inferred by Retro-MTGR are promising for single-step synthesis reactions. The code and data of this study are freely available at https://doi.org/10.5281/zenodo.14346324 .
Collapse
Affiliation(s)
- Peng-Cheng Zhao
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Xue-Xin Wei
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Qiong Wang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Qi-Hao Wang
- School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, China
| | - Jia-Ning Li
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Jie Shang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine China Academy of Chinese Medical Sciences, Beijing, China.
| | - Jian-Yu Shi
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.
| |
Collapse
|
8
|
Jia Y, Dong H, Li L, Wang F, Juan L, Wang Y, Guo H, Zhao T. xQTLatlas: a comprehensive resource for human cellular-resolution multi-omics genetic regulatory landscape. Nucleic Acids Res 2025; 53:D1270-D1277. [PMID: 39351883 PMCID: PMC11701524 DOI: 10.1093/nar/gkae837] [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: 07/22/2024] [Revised: 08/26/2024] [Accepted: 09/13/2024] [Indexed: 10/03/2024] Open
Abstract
Understanding how genetic variants influence molecular phenotypes in different cellular contexts is crucial for elucidating the molecular and cellular mechanisms behind complex traits, which in turn has spurred significant advances in research into molecular quantitative trait locus (xQTL) at the cellular level. With the rapid proliferation of data, there is a critical need for a comprehensive and accessible platform to integrate this information. To meet this need, we developed xQTLatlas (http://www.hitxqtl.org.cn/), a database that provides a multi-omics genetic regulatory landscape at cellular resolution. xQTLatlas compiles xQTL summary statistics from 151 cell types and 339 cell states across 55 human tissues. It organizes these data into 20 xQTL types, based on four distinct discovery strategies, and spans 13 molecular phenotypes. Each entry in xQTLatlas is meticulously annotated with comprehensive metadata, including the origin of the tissue, cell type, cell state and the QTL discovery strategies utilized. Additionally, xQTLatlas features multiscale data exploration tools and a suite of interactive visualizations, facilitating in-depth analysis of cell-level xQTL. xQTLatlas provides a valuable resource for deepening our understanding of the impact of functional variants on molecular phenotypes in different cellular environments, thereby facilitating extensive research efforts.
Collapse
Affiliation(s)
- Yuran Jia
- Faculty of Computing, Harbin Institute of Technology, Harbin 150001, China
| | - Hongchao Dong
- Faculty of Computing, Harbin Institute of Technology, Harbin 150001, China
| | - Linhao Li
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China
| | - Fang Wang
- Faculty of Computing, Harbin Institute of Technology, Harbin 150001, China
| | - Liran Juan
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Yadong Wang
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Harbin 450000, China
| | - Hongzhe Guo
- Faculty of Computing, Harbin Institute of Technology, Harbin 150001, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Harbin 450000, China
| | - Tianyi Zhao
- Faculty of Computing, Harbin Institute of Technology, Harbin 150001, China
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Harbin 450000, China
| |
Collapse
|
9
|
Xie Y, Wang X, Wang P, Bi X. A pseudo-label supervised graph fusion attention network for drug–target interaction prediction. EXPERT SYSTEMS WITH APPLICATIONS 2025; 259:125264. [DOI: 10.1016/j.eswa.2024.125264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
10
|
Ning Q, Wang Y, Zhao Y, Sun J, Jiang L, Wang K, Yin M. DMHGNN: Double multi-view heterogeneous graph neural network framework for drug-target interaction prediction. Artif Intell Med 2025; 159:103023. [PMID: 39579417 DOI: 10.1016/j.artmed.2024.103023] [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: 10/09/2024] [Accepted: 11/15/2024] [Indexed: 11/25/2024]
Abstract
Accurate identification of drug-target interactions (DTIs) plays a crucial role in drug discovery. Compared with traditional experimental methods that are labor-intensive and time-consuming, computational methods for drug-target interactions prediction are more popular in recent years. Conventional computational methods almost simply view heterogeneous network constructed by the drug-related and protein-related dataset instead of comprehensively exploring drug-protein pair (DPP) information. To address this limitation, we proposed a Double Multi-view Heterogeneous Graph Neural Network framework for drug-target interaction prediction (DMHGNN). In DMHGNN, one multi-view heterogeneous graph neural network is based on meta-paths and denoising autoencoder for protein-, drug-related heterogeneous network learning, and another multi-view heterogeneous graph neural network is based on multi-channel graph convolutional network for drug-protein pair similarity network learning. First, a meta-path-based graph encoder with the attention mechanism is used for substructure learning of complex relationships from heterogeneous network constructed by proteins, drugs, side-effects and diseases, obtaining key information that is easy to be ignored in global learning of heterogeneous networks, and multi-source neighbouring features for drugs and proteins are learned from heterogeneous network via denoising auto-encoder model. Then, multi-view graphs of drug-protein pairs (DPPs) including the topology graph, semantics graph and collaborative graph with shared weights are constructed, and the multi-channel graph convolutional network (GCN) is utilized to learn the deep representation of DPPs. Finally, a multi-layer fully connection network is trained to predict drug-target interactions. Experiments have demonstrated its effectiveness and better performance than state-of-the-art methods.
Collapse
Affiliation(s)
- Qiao Ning
- The School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi 214122, Jiangsu, China; Information Science and Technology, Dalian Maritime University, Dalian 116026, Liaoning, China; Neusoft Education Technology Group, Dalian 116026, Liaoning, China; Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun 130015, Jilin, China.
| | - Yue Wang
- Information Science and Technology, Dalian Maritime University, Dalian 116026, Liaoning, China
| | - Yaomiao Zhao
- Information Science and Technology, Dalian Maritime University, Dalian 116026, Liaoning, China
| | - Jiahao Sun
- Computer Science and Technology, the Northeast Normal University, Changchun 999078, Jilin, China
| | - Lu Jiang
- Information Science and Technology, Dalian Maritime University, Dalian 116026, Liaoning, China; Computer Science and Technology, the Northeast Normal University, Changchun 999078, Jilin, China.
| | - Kaidi Wang
- Computer Science and Technology, the Northeast Normal University, Changchun 999078, Jilin, China
| | - Minghao Yin
- Computer Science and Technology, the Northeast Normal University, Changchun 999078, Jilin, China.
| |
Collapse
|
11
|
Dong Y, Zhang Y, Qian Y, Zhao Y, Yang Z, Feng X. ASGCL: Adaptive Sparse Mapping-based graph contrastive learning network for cancer drug response prediction. PLoS Comput Biol 2025; 21:e1012748. [PMID: 39883719 PMCID: PMC11781687 DOI: 10.1371/journal.pcbi.1012748] [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: 03/21/2024] [Accepted: 12/23/2024] [Indexed: 02/01/2025] Open
Abstract
Personalized cancer drug treatment is emerging as a frontier issue in modern medical research. Considering the genomic differences among cancer patients, determining the most effective drug treatment plan is a complex and crucial task. In response to these challenges, this study introduces the Adaptive Sparse Graph Contrastive Learning Network (ASGCL), an innovative approach to unraveling latent interactions in the complex context of cancer cell lines and drugs. The core of ASGCL is the GraphMorpher module, an innovative component that enhances the input graph structure via strategic node attribute masking and topological pruning. By contrasting the augmented graph with the original input, the model delineates distinct positive and negative sample sets at both node and graph levels. This dual-level contrastive approach significantly amplifies the model's discriminatory prowess in identifying nuanced drug responses. Leveraging a synergistic combination of supervised and contrastive loss, ASGCL accomplishes end-to-end learning of feature representations, substantially outperforming existing methodologies. Comprehensive ablation studies underscore the efficacy of each component, corroborating the model's robustness. Experimental evaluations further illuminate ASGCL's proficiency in predicting drug responses, offering a potent tool for guiding clinical decision-making in cancer therapy.
Collapse
Affiliation(s)
- Yunyun Dong
- School of Software, Taiyuan University of Technology, Taiyuan, China
- Institute of Big Data Science and Industry, Shanxi University, Taiyuan, China
| | - Yuanrong Zhang
- School of Software, Taiyuan University of Technology, Taiyuan, China
| | - Yuhua Qian
- Institute of Big Data Science and Industry, Shanxi University, Taiyuan, China
- School of Computer and Information Technology, Shanxi University, Taiyuan, China
| | - Yiming Zhao
- School of Software, Taiyuan University of Technology, Taiyuan, China
| | - Ziting Yang
- School of Software, Taiyuan University of Technology, Taiyuan, China
| | - Xiufang Feng
- School of Software, Taiyuan University of Technology, Taiyuan, China
| |
Collapse
|
12
|
Ai N, Yuan H, Liang Y, Lu S, Ouyang D, Lai QH, Lai LL. Multi-View Multiattention Graph Learning With Stack Deep Matrix Factorization for circRNA-Drug Sensitivity Association Identification. IEEE J Biomed Health Inform 2024; 28:7670-7682. [PMID: 39186430 DOI: 10.1109/jbhi.2024.3431693] [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: 08/28/2024]
Abstract
Identifying circular RNA (circRNA)-drug sensitivity association (CDsA) is crucial for advancing drug development. As conducting traditional wet experiments for determining CDsA is costly and inefficient, calculation methods have already proven to be a valid approach to cope with this problem. However, there exists limited research addressing the prediction of the CDsA prediction problem, and certain discrepancies persist, particularly concerning false-negative associations. As a consequence, we present a multi-view framework, called MAGSDMF, for identifying latent CDsA. Firstly, MAGSDMF applies ultiple ttention mechanisms and raph learning methods to dynamically extract features and strengthen the features of inside and across multi-similarity networks of circRNA and drug. Secondly, the tack eep atrix Factorization (SDMF) is devised to directly extract features from CDsAs. We consider multi-similarity networks with the original CDsAs as multi-view information. Thirdly, MAGSDMF utilizes a multi-attention channel mechanism to integrate these features for the purpose of reconstructing CDsA. Finally, MAGSDMF performs another DMF based on the reconstruction to identify the latent CDsAs. Simultaneously, contrastive learning (CL) is implemented to enhance the generalization capability of MAGSDMF and oversee the learning process of the underlying links prediction task. In comparative experiments, MAGSDMF achieves superior performance on two datasets with AUC values of 0.9743 and 0.9739 based on 5-fold cross-validation. Moreover, in case studies, the achievements further validate the identification reliability of MAGSDMF.
Collapse
|
13
|
Shi W, Yang H, Xie L, Yin XX, Zhang Y. A review of machine learning-based methods for predicting drug-target interactions. Health Inf Sci Syst 2024; 12:30. [PMID: 38617016 PMCID: PMC11014838 DOI: 10.1007/s13755-024-00287-6] [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: 09/11/2023] [Accepted: 03/04/2024] [Indexed: 04/16/2024] Open
Abstract
The prediction of drug-target interactions (DTI) is a crucial preliminary stage in drug discovery and development, given the substantial risk of failure and the prolonged validation period associated with in vitro and in vivo experiments. In the contemporary landscape, various machine learning-based methods have emerged as indispensable tools for DTI prediction. This paper begins by placing emphasis on the data representation employed by these methods, delineating five representations for drugs and four for proteins. The methods are then categorized into traditional machine learning-based approaches and deep learning-based ones, with a discussion of representative approaches in each category and the introduction of a novel taxonomy for deep neural network models in DTI prediction. Additionally, we present a synthesis of commonly used datasets and evaluation metrics to facilitate practical implementation. In conclusion, we address current challenges and outline potential future directions in this research field.
Collapse
Affiliation(s)
- Wen Shi
- Cyberspace Institute of Advanced Technology, Guangzhou University, Guangzhou, 510006 China
- School of Computer Science and Technology, Zhejiang Normal University, Jinhua, 321004 China
| | - Hong Yang
- Cyberspace Institute of Advanced Technology, Guangzhou University, Guangzhou, 510006 China
| | - Linhai Xie
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing, 102206 China
| | - Xiao-Xia Yin
- Cyberspace Institute of Advanced Technology, Guangzhou University, Guangzhou, 510006 China
| | - Yanchun Zhang
- School of Computer Science and Technology, Zhejiang Normal University, Jinhua, 321004 China
- Department of New Networks, Peng Cheng Laboratory, Shenzhen, 518000 China
| |
Collapse
|
14
|
Zhang M, Hong Y, Shen L, Xu S, Xu Y, Zhang X, Liu J, Liu X. A heterogeneous graph neural network with automatic discovery of effective metapaths for drug–target interaction prediction. FUTURE GENERATION COMPUTER SYSTEMS 2024; 160:283-294. [DOI: 10.1016/j.future.2024.05.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
15
|
Xu B, Chen J, Wang Y, Fu Q, Lu Y. Reinforced Metapath Optimization in Heterogeneous Information Networks for Drug-Target Interaction Prediction. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2024; 21:2315-2329. [PMID: 39316496 DOI: 10.1109/tcbb.2024.3467135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
Graph neural networks offer an effective avenue for predicting drug-target interactions. In this domain, researchers have found that constructing heterogeneous information networks based on metapaths using diverse biological datasets enhances prediction performance. However, the performance of such methods is closely tied to the selection of metapaths and the compatibility between metapath subgraphs and graph neural networks. Most existing approaches still rely on fixed strategies for selecting metapaths and often fail to fully exploit node information along the metapaths, limiting the improvement in model performance. This paper introduces a novel method for predicting drug-target interactions by optimizing metapaths in heterogeneous information networks. On one hand, the method formulates the metapath optimization problem as a Markov decision process, using the enhancement of downstream network performance as a reward signal. Through iterative training of a reinforcement learning agent, a high-quality set of metapaths is learned. On the other hand, to fully leverage node information along the metapaths, the paper constructs subgraphs based on nodes along the metapaths. Different depths of subgraphs are processed using different graph convolutional neural network. The proposed method is validated using standard heterogeneous biological benchmark datasets. Experimental results on standard datasets show significant advantages over traditional methods.
Collapse
|
16
|
Lu Q, Zhou Z, Wang Q. Multi-layer graph attention neural networks for accurate drug-target interaction mapping. Sci Rep 2024; 14:26119. [PMID: 39478027 PMCID: PMC11525987 DOI: 10.1038/s41598-024-75742-1] [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: 02/02/2024] [Accepted: 10/08/2024] [Indexed: 11/02/2024] Open
Abstract
In the crucial process of drug discovery and repurposing, precise prediction of drug-target interactions (DTIs) is paramount. This study introduces a novel DTI prediction approach-Multi-Layer Graph Attention Neural Network (MLGANN), through a groundbreaking computational framework that effectively harnesses multi-source information to enhance prediction accuracy. MLGANN not only strides forward in constructing a multi-layer DTI network by capturing both direct interactions between drugs and targets as well as their multi-level information but also amalgamates Graph Convolutional Networks (GCN) with a self-attention mechanism to comprehensively integrate diverse data sources. This method exhibited significant performance surpassing existing approaches in comparative experiments, underscoring its immense potential in elevating the efficiency and accuracy of DTI predictions. More importantly, this study accentuates the significance of considering multi-source data information and network heterogeneity in the drug discovery process, offering new perspectives and tools for future pharmaceutical research.
Collapse
Affiliation(s)
- Qianwen Lu
- SDU-ANU Joint Science College, Shandong University, Weihai, 264209, Shandong, China
| | - Zhiheng Zhou
- Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing, 100190, China
- School of Mathematical Sciences, University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Qi Wang
- College of Science, China Agricultural University, Beijing, 100083, China.
| |
Collapse
|
17
|
Ren Z, Zeng X, Lao Y, Zheng H, You Z, Xiang H, Zou Q. A spatial hierarchical network learning framework for drug repositioning allowing interpretation from macro to micro scale. Commun Biol 2024; 7:1413. [PMID: 39478146 PMCID: PMC11525566 DOI: 10.1038/s42003-024-07107-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 10/21/2024] [Indexed: 11/02/2024] Open
Abstract
Biomedical network learning offers fresh prospects for expediting drug repositioning. However, traditional network architectures struggle to quantify the relationship between micro-scale drug spatial structures and corresponding macro-scale biomedical networks, limiting their ability to capture key pharmacological properties and complex biomedical information crucial for drug screening and therapeutic discovery. Moreover, challenges such as difficulty in capturing long-range dependencies hinder current network-based approaches. To address these limitations, we introduce the Spatial Hierarchical Network, modeling molecular 3D structures and biological associations into a unified network. We propose an end-to-end framework, SpHN-VDA, integrating spatial hierarchical information through triple attention mechanisms to enhance machine understanding of molecular functionality and improve the accuracy of virus-drug association identification. SpHN-VDA outperforms leading models across three datasets, particularly excelling in out-of-distribution and cold-start scenarios. It also exhibits enhanced robustness against data perturbation, ranging from 20% to 40%. It accurately identifies critical motifs for binding sites, even without protein residue annotations. Leveraging reliability of SpHN-VDA, we have identified 25 potential candidate drugs through gene expression analysis and CMap. Molecular docking experiments with the SARS-CoV-2 spike protein further corroborate the predictions. This research highlights the broad potential of SpHN-VDA to enhance drug repositioning and identify effective treatments for various diseases.
Collapse
Affiliation(s)
- Zhonghao Ren
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China
| | - Xiangxiang Zeng
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China
| | - Yizhen Lao
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China
| | - Heping Zheng
- College of Biology, Department of Molecular Medicine, Hunan University, Changsha, China
| | - Zhuhong You
- School of Computer Science, Northwestern Polytechnical University, Xi'an, China
| | - Hongxin Xiang
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China
| | - Quan Zou
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
18
|
Zhang P, Lin P, Li D, Wang W, Qi X, Li J, Xiong J. MGACL: Prediction Drug-Protein Interaction Based on Meta-Graph Association-Aware Contrastive Learning. Biomolecules 2024; 14:1267. [PMID: 39456200 PMCID: PMC11505808 DOI: 10.3390/biom14101267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 09/20/2024] [Accepted: 10/02/2024] [Indexed: 10/28/2024] Open
Abstract
The identification of drug-target interaction (DTI) is crucial for drug discovery. However, how to reduce the graph neural network's false positives due to its bias and negative transfer in the original bipartite graph remains to be clarified. Considering that the impact of heterogeneous auxiliary information on DTI varies depending on the drug and target, we established an adaptive enhanced personalized meta-knowledge transfer network named Meta Graph Association-Aware Contrastive Learning (MGACL), which can transfer personalized heterogeneous auxiliary information from different nodes and reduce data bias. Meanwhile, we propose a novel DTI association-aware contrastive learning strategy that aligns high-frequency drug representations with learned auxiliary graph representations to prevent negative transfer. Our study improves the DTI prediction performance by about 3%, evaluated by analyzing the area under the curve (AUC) and area under the precision-recall curve (AUPRC) compared with existing methods, which is more conducive to accurately identifying drug targets for the development of new drugs.
Collapse
Affiliation(s)
- Pinglu Zhang
- Faculty of Information Science and Engineering, Ocean University of China, Qingdao 266003, China; (P.Z.); (W.W.)
| | - Peng Lin
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (P.L.); (D.L.); (X.Q.)
| | - Dehai Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (P.L.); (D.L.); (X.Q.)
| | - Wanchun Wang
- Faculty of Information Science and Engineering, Ocean University of China, Qingdao 266003, China; (P.Z.); (W.W.)
| | - Xin Qi
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (P.L.); (D.L.); (X.Q.)
| | - Jing Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (P.L.); (D.L.); (X.Q.)
| | - Jianshe Xiong
- Faculty of Information Science and Engineering, Ocean University of China, Qingdao 266003, China; (P.Z.); (W.W.)
| |
Collapse
|
19
|
Hu X, Zhang P, Zhang J, Deng L. DeepFusionCDR: Employing Multi-Omics Integration and Molecule-Specific Transformers for Enhanced Prediction of Cancer Drug Responses. IEEE J Biomed Health Inform 2024; 28:6248-6258. [PMID: 38935469 DOI: 10.1109/jbhi.2024.3417014] [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: 06/29/2024]
Abstract
Deep learning approaches have demonstrated remarkable potential in predicting cancer drug responses (CDRs), using cell line and drug features. However, existing methods predominantly rely on single-omics data of cell lines, potentially overlooking the complex biological mechanisms governing cell line responses. This paper introduces DeepFusionCDR, a novel approach employing unsupervised contrastive learning to amalgamate multi-omics features, including mutation, transcriptome, methylome, and copy number variation data, from cell lines. Furthermore, we incorporate molecular SMILES-specific transformers to derive drug features from their chemical structures. The unified multi-omics and drug signatures are combined, and a multi-layer perceptron (MLP) is applied to predict IC50 values for cell line-drug pairs. Moreover, this MLP can discern whether a cell line is resistant or sensitive to a particular drug. We assessed DeepFusionCDR's performance on the GDSC dataset and juxtaposed it against cutting-edge methods, demonstrating its superior performance in regression and classification tasks. We also conducted ablation studies and case analyses to exhibit the effectiveness and versatility of our proposed approach. Our results underscore the potential of DeepFusionCDR to enhance CDR predictions by harnessing the power of multi-omics fusion and molecular-specific transformers. The prediction of DeepFusionCDR on TCGA patient data and case study highlight the practical application scenarios of DeepFusionCDR in real-world environments.
Collapse
|
20
|
Qiao G, Wang G, Li Y. Causal enhanced drug-target interaction prediction based on graph generation and multi-source information fusion. Bioinformatics 2024; 40:btae570. [PMID: 39312682 PMCID: PMC11639159 DOI: 10.1093/bioinformatics/btae570] [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: 05/18/2024] [Revised: 08/17/2024] [Accepted: 09/20/2024] [Indexed: 09/25/2024] Open
Abstract
MOTIVATION The prediction of drug-target interaction is a vital task in the biomedical field, aiding in the discovery of potential molecular targets of drugs and the development of targeted therapy methods with higher efficacy and fewer side effects. Although there are various methods for drug-target interaction (DTI) prediction based on heterogeneous information networks, these methods face challenges in capturing the fundamental interaction between drugs and targets and ensuring the interpretability of the model. Moreover, they need to construct meta-paths artificially or a lot of feature engineering (prior knowledge), and graph generation can fuse information more flexibly without meta-path selection. RESULTS We propose a causal enhanced method for drug-target interaction (CE-DTI) prediction that integrates graph generation and multi-source information fusion. First, we represent drugs and targets by modeling the fusion of their multi-source information through automatic graph generation. Once drugs and targets are combined, a network of drug-target pairs is constructed, transforming the prediction of drug-target interactions into a node classification problem. Specifically, the influence of surrounding nodes on the central node is separated into two groups: causal and non-causal variable nodes. Causal variable nodes significantly impact the central node's classification, while non-causal variable nodes do not. Causal invariance is then used to enhance the contrastive learning of the drug-target pairs network. Our method demonstrates excellent performance compared with other competitive benchmark methods across multiple datasets. At the same time, the experimental results also show that the causal enhancement strategy can explore the potential causal effects between DTPs, and discover new potential targets. Additionally, case studies demonstrate that this method can identify potential drug targets. AVAILABILITY AND IMPLEMENTATION The source code of AdaDR is available at: https://github.com/catly/CE-DTI.
Collapse
Affiliation(s)
- Guanyu Qiao
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Guohua Wang
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China
- College of Computer and Control Engineering, Northeast Forestry University, Harbin 150040, China
| | - Yang Li
- College of Computer and Control Engineering, Northeast Forestry University, Harbin 150040, China
| |
Collapse
|
21
|
Tian Z, Yu Y, Ni F, Zou Q. Drug-target interaction prediction with collaborative contrastive learning and adaptive self-paced sampling strategy. BMC Biol 2024; 22:216. [PMID: 39334132 PMCID: PMC11437672 DOI: 10.1186/s12915-024-02012-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Drug-target interaction (DTI) prediction plays a pivotal role in drug discovery and drug repositioning, enabling the identification of potential drug candidates. However, most previous approaches often do not fully utilize the complementary relationships among multiple biological networks, which limits their ability to learn more consistent representations. Additionally, the selection strategy of negative samples significantly affects the performance of contrastive learning methods. RESULTS In this study, we propose CCL-ASPS, a novel deep learning model that incorporates Collaborative Contrastive Learning (CCL) and Adaptive Self-Paced Sampling strategy (ASPS) for drug-target interaction prediction. CCL-ASPS leverages multiple networks to learn the fused embeddings of drugs and targets, ensuring their consistent representations from individual networks. Furthermore, ASPS dynamically selects more informative negative sample pairs for contrastive learning. Experiment results on the established dataset demonstrate that CCL-ASPS achieves significant improvements compared to current state-of-the-art methods. Moreover, ablation experiments confirm the contributions of the proposed CCL and ASPS strategies. CONCLUSIONS By integrating Collaborative Contrastive Learning and Adaptive Self-Paced Sampling, the proposed CCL-ASPS effectively addresses the limitations of previous methods. This study demonstrates that CCL-ASPS achieves notable improvements in DTI predictive performance compared to current state-of-the-art approaches. The case study and cold start experiments further illustrate the capability of CCL-ASPS to effectively predict previously unknown DTI, potentially facilitating the identification of new drug-target interactions.
Collapse
Affiliation(s)
- Zhen Tian
- School of Computer and Artificial Intelligence, Zhengzhou University, Zhengzhou, 450001, Henan, China
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou, China
| | - Yue Yu
- School of Computer and Artificial Intelligence, Zhengzhou University, Zhengzhou, 450001, Henan, China
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou, China
| | - Fengming Ni
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, 130021, China.
| | - Quan Zou
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou, China
| |
Collapse
|
22
|
Wang S, Liu Y, Zhang Y, Zhang K, Song X, Zhang Y, Pang S. CHL-DTI: A Novel High-Low Order Information Convergence Framework for Effective Drug-Target Interaction Prediction. Interdiscip Sci 2024; 16:568-578. [PMID: 38483753 DOI: 10.1007/s12539-024-00608-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 01/05/2024] [Accepted: 01/07/2024] [Indexed: 09/19/2024]
Abstract
Recognizing drug-target interactions (DTI) stands as a pivotal element in the expansive field of drug discovery. Traditional biological wet experiments, although valuable, are time-consuming and costly as methods. Recently, computational methods grounded in network learning have demonstrated great advantages by effective topological feature extraction and attracted extensive research attention. However, most existing network-based learning methods only consider the low-order binary correlation between individual drug and target, neglecting the potential higher-order correlation information derived from multiple drugs and targets. High-order information, as an essential component, exhibits complementarity with low-order information. Hence, the incorporation of higher-order associations between drugs and targets, while adequately integrating them with the existing lower-order information, could potentially yield substantial breakthroughs in predicting drug-target interactions. We propose a novel dual channels network-based learning model CHL-DTI that converges high-order information from hypergraphs and low-order information from ordinary graph for drug-target interaction prediction. The convergence of high-low order information in CHL-DTI is manifested in two key aspects. First, during the feature extraction stage, the model integrates both high-level semantic information and low-level topological information by combining hypergraphs and ordinary graph. Second, CHL-DTI fully fuse the innovative introduced drug-protein pairs (DPP) hypergraph network structure with ordinary topological network structure information. Extensive experimentation conducted on three public datasets showcases the superior performance of CHL-DTI in DTI prediction tasks when compared to SOTA methods. The source code of CHL-DTI is available at https://github.com/UPCLyy/CHL-DTI .
Collapse
Affiliation(s)
- Shudong Wang
- College of Computer Science and Technology, China University of Petroleum (East China), Qingdao, 266580, China
| | - Yingye Liu
- College of Computer Science and Technology, China University of Petroleum (East China), Qingdao, 266580, China
| | - Yuanyuan Zhang
- College of Information and Control Engineering, Qingdao University of Technology, Qingdao, 266520, China.
| | - Kuijie Zhang
- College of Computer Science and Technology, China University of Petroleum (East China), Qingdao, 266580, China
| | - Xuanmo Song
- College of Computer Science and Technology, China University of Petroleum (East China), Qingdao, 266580, China
| | - Yu Zhang
- College of Computer Science and Technology, China University of Petroleum (East China), Qingdao, 266580, China
| | - Shanchen Pang
- College of Computer Science and Technology, China University of Petroleum (East China), Qingdao, 266580, China
| |
Collapse
|
23
|
Li N, Yang Z, Wang J, Lin H. Drug-target interaction prediction using knowledge graph embedding. iScience 2024; 27:109393. [PMID: 38952679 PMCID: PMC11215290 DOI: 10.1016/j.isci.2024.109393] [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: 09/11/2023] [Revised: 01/16/2024] [Accepted: 02/28/2024] [Indexed: 07/03/2024] Open
Abstract
The prediction of drug-target interactions (DTIs) is a critical phase in the sustainable drug development process, especially when the research focus is to capitalize on the repositioning of existing drugs. Computational approaches to predicting DTIs can provide important insights into drug mechanisms of action. However, current methods for predicting DTIs based on the structural information of the knowledge graph may suffer from the sparseness and incompleteness of the knowledge graph and neglect the latent type information of the knowledge graph. In this paper, we propose TTModel, a knowledge graph embedding model for DTI prediction. By exploiting biomedical text and type information, TTModel can learn latent text semantics and type information to improve the performance of representation learning. Comprehensive experiments on two public datasets demonstrate that our model outperforms the state-of-the-art methods significantly on the task of DTI prediction.
Collapse
Affiliation(s)
- Nan Li
- College of Computer Science and Technology, Dalian University of Technology, Dalian, China
| | - Zhihao Yang
- College of Computer Science and Technology, Dalian University of Technology, Dalian, China
| | - Jian Wang
- College of Computer Science and Technology, Dalian University of Technology, Dalian, China
| | - Hongfei Lin
- College of Computer Science and Technology, Dalian University of Technology, Dalian, China
| |
Collapse
|
24
|
Bian J, Lu H, Dong G, Wang G. Hierarchical multimodal self-attention-based graph neural network for DTI prediction. Brief Bioinform 2024; 25:bbae293. [PMID: 38920341 PMCID: PMC11200190 DOI: 10.1093/bib/bbae293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 05/17/2024] [Accepted: 06/06/2024] [Indexed: 06/27/2024] Open
Abstract
Drug-target interactions (DTIs) are a key part of drug development process and their accurate and efficient prediction can significantly boost development efficiency and reduce development time. Recent years have witnessed the rapid advancement of deep learning, resulting in an abundance of deep learning-based models for DTI prediction. However, most of these models used a single representation of drugs and proteins, making it difficult to comprehensively represent their characteristics. Multimodal data fusion can effectively compensate for the limitations of single-modal data. However, existing multimodal models for DTI prediction do not take into account both intra- and inter-modal interactions simultaneously, resulting in limited presentation capabilities of fused features and a reduction in DTI prediction accuracy. A hierarchical multimodal self-attention-based graph neural network for DTI prediction, called HMSA-DTI, is proposed to address multimodal feature fusion. Our proposed HMSA-DTI takes drug SMILES, drug molecular graphs, protein sequences and protein 2-mer sequences as inputs, and utilizes a hierarchical multimodal self-attention mechanism to achieve deep fusion of multimodal features of drugs and proteins, enabling the capture of intra- and inter-modal interactions between drugs and proteins. It is demonstrated that our proposed HMSA-DTI has significant advantages over other baseline methods on multiple evaluation metrics across five benchmark datasets.
Collapse
Affiliation(s)
- Jilong Bian
- College of Computer and Control Engineering, Northeast Forestry University, No. 26 Hexing Road, Xiangfang District, Harbin, Heilongjiang 150040, China
| | - Hao Lu
- College of Computer and Control Engineering, Northeast Forestry University, No. 26 Hexing Road, Xiangfang District, Harbin, Heilongjiang 150040, China
| | - Guanghui Dong
- College of Computer and Control Engineering, Northeast Forestry University, No. 26 Hexing Road, Xiangfang District, Harbin, Heilongjiang 150040, China
| | - Guohua Wang
- College of Computer and Control Engineering, Northeast Forestry University, No. 26 Hexing Road, Xiangfang District, Harbin, Heilongjiang 150040, China
| |
Collapse
|
25
|
Ozalp MK, Vignaux PA, Puhl AC, Lane TR, Urbina F, Ekins S. Sequential Contrastive and Deep Learning Models to Identify Selective Butyrylcholinesterase Inhibitors. J Chem Inf Model 2024; 64:3161-3172. [PMID: 38532612 PMCID: PMC11331448 DOI: 10.1021/acs.jcim.4c00397] [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] [Indexed: 03/28/2024]
Abstract
Butyrylcholinesterase (BChE) is a target of interest in late-stage Alzheimer's Disease (AD) where selective BChE inhibitors (BIs) may offer symptomatic treatment without the harsh side effects of acetylcholinesterase (AChE) inhibitors. In this study, we explore multiple machine learning strategies to identify BIs in silico, optimizing for precision over all other metrics. We compare state-of-the-art supervised contrastive learning (CL) with deep learning (DL) and Random Forest (RF) machine learning, across single and sequential modeling configurations, to identify the best models for BChE selectivity. We used these models to virtually screen a vendor library of 5 million compounds for BIs and tested 20 of these compounds in vitro. Seven of the 20 compounds displayed selectivity for BChE over AChE, reflecting a hit rate of 35% for our model predictions, suggesting a highly efficient strategy for modeling selective inhibition.
Collapse
Affiliation(s)
| | | | - Ana C. Puhl
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC 27606, USA
| | - Thomas R. Lane
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC 27606, USA
| | - Fabio Urbina
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC 27606, USA
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC 27606, USA
| |
Collapse
|
26
|
Chen S, Li M, Semenov I. MFA-DTI: Drug-target interaction prediction based on multi-feature fusion adopted framework. Methods 2024; 224:79-92. [PMID: 38430967 DOI: 10.1016/j.ymeth.2024.02.008] [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: 12/31/2023] [Revised: 02/16/2024] [Accepted: 02/23/2024] [Indexed: 03/05/2024] Open
Abstract
The identification of drug-target interactions (DTI) is a valuable step in the drug discovery and repositioning process. However, traditional laboratory experiments are time-consuming and expensive. Computational methods have streamlined research to determine DTIs. The application of deep learning methods has significantly improved the prediction performance for DTIs. Modern deep learning methods can leverage multiple sources of information, including sequence data that contains biological structural information, and interaction data. While useful, these methods cannot be effectively applied to each type of information individually (e.g., chemical structure and interaction network) and do not take into account the specificity of DTI data such as low- or zero-interaction biological entities. To overcome these limitations, we propose a method called MFA-DTI (Multi-feature Fusion Adopted framework for DTI). MFA-DTI consists of three modules: an interaction graph learning module that processes the interaction network to generate interaction vectors, a chemical structure learning module that extracts features from the chemical structure, and a fusion module that combines these features for the final prediction. To validate the performance of MFA-DTI, we conducted experiments on six public datasets under different settings. The results indicate that the proposed method is highly effective in various settings and outperforms state-of-the-art methods.
Collapse
Affiliation(s)
- Siqi Chen
- School of Information Science and Engineering, Chongqing Jiaotong University, Chongqing, 400074, China.
| | - Minghui Li
- Beidahuang Industry Group General Hospital, Harbin, 150006, China
| | - Ivan Semenov
- College of Intelligence and Computing, Tianjin University, Tianjin, 300072, China
| |
Collapse
|
27
|
Ouyang D, Liang Y, Wang J, Li L, Ai N, Feng J, Lu S, Liao S, Liu X, Xie S. HGCLAMIR: Hypergraph contrastive learning with attention mechanism and integrated multi-view representation for predicting miRNA-disease associations. PLoS Comput Biol 2024; 20:e1011927. [PMID: 38652712 PMCID: PMC11037542 DOI: 10.1371/journal.pcbi.1011927] [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: 09/20/2023] [Accepted: 02/19/2024] [Indexed: 04/25/2024] Open
Abstract
Existing studies have shown that the abnormal expression of microRNAs (miRNAs) usually leads to the occurrence and development of human diseases. Identifying disease-related miRNAs contributes to studying the pathogenesis of diseases at the molecular level. As traditional biological experiments are time-consuming and expensive, computational methods have been used as an effective complement to infer the potential associations between miRNAs and diseases. However, most of the existing computational methods still face three main challenges: (i) learning of high-order relations; (ii) insufficient representation learning ability; (iii) importance learning and integration of multi-view embedding representation. To this end, we developed a HyperGraph Contrastive Learning with view-aware Attention Mechanism and Integrated multi-view Representation (HGCLAMIR) model to discover potential miRNA-disease associations. First, hypergraph convolutional network (HGCN) was utilized to capture high-order complex relations from hypergraphs related to miRNAs and diseases. Then, we combined HGCN with contrastive learning to improve and enhance the embedded representation learning ability of HGCN. Moreover, we introduced view-aware attention mechanism to adaptively weight the embedded representations of different views, thereby obtaining the importance of multi-view latent representations. Next, we innovatively proposed integrated representation learning to integrate the embedded representation information of multiple views for obtaining more reasonable embedding information. Finally, the integrated representation information was fed into a neural network-based matrix completion method to perform miRNA-disease association prediction. Experimental results on the cross-validation set and independent test set indicated that HGCLAMIR can achieve better prediction performance than other baseline models. Furthermore, the results of case studies and enrichment analysis further demonstrated the accuracy of HGCLAMIR and unconfirmed potential associations had biological significance.
Collapse
Affiliation(s)
- Dong Ouyang
- Peng Cheng Laboratory, Shenzhen, China
- School of Biomedical Engineering, Guangdong Medical University, Dongguan, China
| | - Yong Liang
- Peng Cheng Laboratory, Shenzhen, China
- Pazhou Laboratory (Huangpu), Guangzhou, China
| | - Jinfeng Wang
- College of Mathematics and Informatics, South China Agricultural University, Guangzhou, China
| | - Le Li
- School of Computer Science and Engineering, Faculty of Innovation Engineering, Macau University of Science and Technology, Macau, China
| | - Ning Ai
- School of Computer Science and Engineering, Faculty of Innovation Engineering, Macau University of Science and Technology, Macau, China
| | - Junning Feng
- School of Computer Science and Engineering, Faculty of Innovation Engineering, Macau University of Science and Technology, Macau, China
| | - Shanghui Lu
- School of Computer Science and Engineering, Faculty of Innovation Engineering, Macau University of Science and Technology, Macau, China
| | - Shuilin Liao
- School of Computer Science and Engineering, Faculty of Innovation Engineering, Macau University of Science and Technology, Macau, China
| | - Xiaoying Liu
- Computer Engineering Technical College, Guangdong Polytechnic of Science and Technology, Zhuhai, China
| | - Shengli Xie
- Guangdong-HongKong-Macao Joint Laboratory for Smart Discrete Manufacturing, Guangzhou, China
| |
Collapse
|
28
|
E Z, Qiao G, Wang G, Li Y. GSL-DTI: Graph structure learning network for Drug-Target interaction prediction. Methods 2024; 223:136-145. [PMID: 38360082 DOI: 10.1016/j.ymeth.2024.01.018] [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: 07/22/2023] [Revised: 12/23/2023] [Accepted: 01/29/2024] [Indexed: 02/17/2024] Open
Abstract
MOTIVATION Drug-target interaction prediction is an important area of research to predict whether there is an interaction between a drug molecule and its target protein. It plays a critical role in drug discovery and development by facilitating the identification of potential drug candidates and expediting the overall process. Given the time-consuming, expensive, and high-risk nature of traditional drug discovery methods, the prediction of drug-target interactions has become an indispensable tool. Using machine learning and deep learning to tackle this class of problems has become a mainstream approach, and graph-based models have recently received much attention in this field. However, many current graph-based Drug-Target Interaction (DTI) prediction methods rely on manually defined rules to construct the Drug-Protein Pair (DPP) network during the DPP representation learning process. However, these methods fail to capture the true underlying relationships between drug molecules and target proteins. RESULTS We propose GSL-DTI, an automatic graph structure learning model used for predicting drug-target interactions (DTIs). Initially, we integrate large-scale heterogeneous networks using a graph convolution network based on meta-paths, effectively learning the representations of drugs and target proteins. Subsequently, we construct drug-protein pairs based on these representations. In contrast to previous studies that construct DPP networks based on manual rules, our method introduces an automatic graph structure learning approach. This approach utilizes a filter gate on the affinity scores of DPPs and relies on the classification loss of downstream tasks to guide the learning of the underlying DPP network structure. Based on the learned DPP network, we transform the prediction of drug-target interactions into a node classification problem. The comprehensive experiments conducted on three public datasets have shown the superiority of GSL-DTI in the tasks of DTI prediction. Additionally, GSL-DTI provides a fresh perspective for advancing research in graph structure learning for DTI prediction.
Collapse
Affiliation(s)
- Zixuan E
- College of Computer and Control Engineering, Northeast Forestry University,Harbin 150006, China
| | - Guanyu Qiao
- College of Computer and Control Engineering, Northeast Forestry University,Harbin 150006, China
| | - Guohua Wang
- College of Computer and Control Engineering, Northeast Forestry University,Harbin 150006, China.
| | - Yang Li
- College of Computer and Control Engineering, Northeast Forestry University,Harbin 150006, China.
| |
Collapse
|
29
|
Dehghan A, Abbasi K, Razzaghi P, Banadkuki H, Gharaghani S. CCL-DTI: contributing the contrastive loss in drug-target interaction prediction. BMC Bioinformatics 2024; 25:48. [PMID: 38291364 PMCID: PMC11264960 DOI: 10.1186/s12859-024-05671-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 01/22/2024] [Indexed: 02/01/2024] Open
Abstract
BACKGROUND The Drug-Target Interaction (DTI) prediction uses a drug molecule and a protein sequence as inputs to predict the binding affinity value. In recent years, deep learning-based models have gotten more attention. These methods have two modules: the feature extraction module and the task prediction module. In most deep learning-based approaches, a simple task prediction loss (i.e., categorical cross entropy for the classification task and mean squared error for the regression task) is used to learn the model. In machine learning, contrastive-based loss functions are developed to learn more discriminative feature space. In a deep learning-based model, extracting more discriminative feature space leads to performance improvement for the task prediction module. RESULTS In this paper, we have used multimodal knowledge as input and proposed an attention-based fusion technique to combine this knowledge. Also, we investigate how utilizing contrastive loss function along the task prediction loss could help the approach to learn a more powerful model. Four contrastive loss functions are considered: (1) max-margin contrastive loss function, (2) triplet loss function, (3) Multi-class N-pair Loss Objective, and (4) NT-Xent loss function. The proposed model is evaluated using four well-known datasets: Wang et al. dataset, Luo's dataset, Davis, and KIBA datasets. CONCLUSIONS Accordingly, after reviewing the state-of-the-art methods, we developed a multimodal feature extraction network by combining protein sequences and drug molecules, along with protein-protein interaction networks and drug-drug interaction networks. The results show it performs significantly better than the comparable state-of-the-art approaches.
Collapse
Affiliation(s)
- Alireza Dehghan
- Department of Bioinformatics, Kish International Campus, University of Tehran, Kish, 1417614411, Iran
| | - Karim Abbasi
- Laboratory of System Biology, Bioinformatics and Artificial Intelligence in Medicine (LBB&AI), Faculty of Mathematics and Computer Science, Kharazmi University, Tehran, 1417614411, Iran
| | - Parvin Razzaghi
- Department of Computer Science and Information Technology, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, 4513766731, Iran.
| | - Hossein Banadkuki
- Laboratory of Bioinformatics and Drug Design (LBD), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, 1417614411, Iran
| | - Sajjad Gharaghani
- Laboratory of Bioinformatics and Drug Design (LBD), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, 1417614411, Iran.
| |
Collapse
|
30
|
Veleiro U, de la Fuente J, Serrano G, Pizurica M, Casals M, Pineda-Lucena A, Vicent S, Ochoa I, Gevaert O, Hernaez M. GeNNius: an ultrafast drug-target interaction inference method based on graph neural networks. Bioinformatics 2024; 40:btad774. [PMID: 38134424 PMCID: PMC10766589 DOI: 10.1093/bioinformatics/btad774] [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: 06/19/2023] [Revised: 11/20/2023] [Accepted: 12/21/2023] [Indexed: 12/24/2023] Open
Abstract
MOTIVATION Drug-target interaction (DTI) prediction is a relevant but challenging task in the drug repurposing field. In-silico approaches have drawn particular attention as they can reduce associated costs and time commitment of traditional methodologies. Yet, current state-of-the-art methods present several limitations: existing DTI prediction approaches are computationally expensive, thereby hindering the ability to use large networks and exploit available datasets and, the generalization to unseen datasets of DTI prediction methods remains unexplored, which could potentially improve the development processes of DTI inferring approaches in terms of accuracy and robustness. RESULTS In this work, we introduce GeNNius (Graph Embedding Neural Network Interaction Uncovering System), a Graph Neural Network (GNN)-based method that outperforms state-of-the-art models in terms of both accuracy and time efficiency across a variety of datasets. We also demonstrated its prediction power to uncover new interactions by evaluating not previously known DTIs for each dataset. We further assessed the generalization capability of GeNNius by training and testing it on different datasets, showing that this framework can potentially improve the DTI prediction task by training on large datasets and testing on smaller ones. Finally, we investigated qualitatively the embeddings generated by GeNNius, revealing that the GNN encoder maintains biological information after the graph convolutions while diffusing this information through nodes, eventually distinguishing protein families in the node embedding space. AVAILABILITY AND IMPLEMENTATION GeNNius code is available at https://github.com/ubioinformat/GeNNius.
Collapse
Affiliation(s)
- Uxía Veleiro
- CIMA University of Navarra, IdiSNA, 31008 Pamplona, Spain
| | - Jesús de la Fuente
- TECNUN, University of Navarra, 20016 San Sebastian, Spain
- Center for Data Science, New York University, New York, NY 10012, United States
| | - Guillermo Serrano
- CIMA University of Navarra, IdiSNA, 31008 Pamplona, Spain
- TECNUN, University of Navarra, 20016 San Sebastian, Spain
| | - Marija Pizurica
- Stanford Center for Biomedical Informatics Research, Department of Medicine and Department Biomedical Data Science, Stanford University, Stanford, CA 94305, United States
- Internet Technology and Data Science LAB (IDLab), Ghent University, Gent 9052, Belgium
| | - Mikel Casals
- TECNUN, University of Navarra, 20016 San Sebastian, Spain
| | | | - Silve Vicent
- CIMA University of Navarra, IdiSNA, 31008 Pamplona, Spain
| | - Idoia Ochoa
- TECNUN, University of Navarra, 20016 San Sebastian, Spain
- Instituto de Ciencia de los Datos e Inteligencia Artificial (DATAI), University of Navarra, 31008 Pamplona, Spain
| | - Olivier Gevaert
- Stanford Center for Biomedical Informatics Research, Department of Medicine and Department Biomedical Data Science, Stanford University, Stanford, CA 94305, United States
| | - Mikel Hernaez
- CIMA University of Navarra, IdiSNA, 31008 Pamplona, Spain
- Instituto de Ciencia de los Datos e Inteligencia Artificial (DATAI), University of Navarra, 31008 Pamplona, Spain
| |
Collapse
|
31
|
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]
|
32
|
Zhao Z, He C, Qu Y, Zheng H, Duan L, Zuo J. MGDTI: Graph Transformer with Meta-Learning for Drug-Target Interaction Prediction. 2023 IEEE INTERNATIONAL CONFERENCE ON BIOINFORMATICS AND BIOMEDICINE (BIBM) 2023:801-806. [DOI: 10.1109/bibm58861.2023.10385671] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Affiliation(s)
- Zhenjiang Zhao
- Sichuan University,School of Computer Science,Chengdu,China
| | - Chengxin He
- Sichuan University,School of Computer Science,Chengdu,China
| | - Yuening Qu
- Sichuan University,School of Computer Science,Chengdu,China
| | - Huiru Zheng
- Ulster University,School of Computing,Northern Ireland,United Kingdom
| | - Lei Duan
- Sichuan University,School of Computer Science,Chengdu,China
| | - Jie Zuo
- Sichuan University,School of Computer Science,Chengdu,China
| |
Collapse
|
33
|
Sun J, Xu M, Ru J, James-Bott A, Xiong D, Wang X, Cribbs AP. Small molecule-mediated targeting of microRNAs for drug discovery: Experiments, computational techniques, and disease implications. Eur J Med Chem 2023; 257:115500. [PMID: 37262996 PMCID: PMC11554572 DOI: 10.1016/j.ejmech.2023.115500] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/05/2023] [Accepted: 05/15/2023] [Indexed: 06/03/2023]
Abstract
Small molecules have been providing medical breakthroughs for human diseases for more than a century. Recently, identifying small molecule inhibitors that target microRNAs (miRNAs) has gained importance, despite the challenges posed by labour-intensive screening experiments and the significant efforts required for medicinal chemistry optimization. Numerous experimentally-verified cases have demonstrated the potential of miRNA-targeted small molecule inhibitors for disease treatment. This new approach is grounded in their posttranscriptional regulation of the expression of disease-associated genes. Reversing dysregulated gene expression using this mechanism may help control dysfunctional pathways. Furthermore, the ongoing improvement of algorithms has allowed for the integration of computational strategies built on top of laboratory-based data, facilitating a more precise and rational design and discovery of lead compounds. To complement the use of extensive pharmacogenomics data in prioritising potential drugs, our previous work introduced a computational approach based on only molecular sequences. Moreover, various computational tools for predicting molecular interactions in biological networks using similarity-based inference techniques have been accumulated in established studies. However, there are a limited number of comprehensive reviews covering both computational and experimental drug discovery processes. In this review, we outline a cohesive overview of both biological and computational applications in miRNA-targeted drug discovery, along with their disease implications and clinical significance. Finally, utilizing drug-target interaction (DTIs) data from DrugBank, we showcase the effectiveness of deep learning for obtaining the physicochemical characterization of DTIs.
Collapse
Affiliation(s)
- Jianfeng Sun
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, OX3 7LD, UK.
| | - Miaoer Xu
- Department of Biology, Emory University, Atlanta, GA, 30322, USA
| | - Jinlong Ru
- Chair of Prevention of Microbial Diseases, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, 85354, Germany
| | - Anna James-Bott
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, OX3 7LD, UK
| | - Dapeng Xiong
- Department of Computational Biology, Cornell University, Ithaca, NY, 14853, USA; Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Xia Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China.
| | - Adam P Cribbs
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, OX3 7LD, UK.
| |
Collapse
|
34
|
Yao K, Wang X, Li W, Zhu H, Jiang Y, Li Y, Tian T, Yang Z, Liu Q, Liu Q. Semi-supervised heterogeneous graph contrastive learning for drug-target interaction prediction. Comput Biol Med 2023; 163:107199. [PMID: 37421738 DOI: 10.1016/j.compbiomed.2023.107199] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 04/15/2023] [Accepted: 06/19/2023] [Indexed: 07/10/2023]
Abstract
Identification of drug-target interactions (DTIs) is an important step in drug discovery and drug repositioning. In recent years, graph-based methods have attracted great attention and show advantages on predicting potential DTIs. However, these methods face the problem that the known DTIs are very limited and expensive to obtain, which decreases the generalization ability of the methods. Self-supervised contrastive learning is independent of labeled DTIs, which can mitigate the impact of the problem. Therefore, we propose a framework SHGCL-DTI for predicting DTIs, which supplements the classical semi-supervised DTI prediction task with an auxiliary graph contrastive learning module. Specifically, we generate representations for the nodes through the neighbor view and meta-path view, and define positive and negative pairs to maximize the similarity between positive pairs from different views. Subsequently, SHGCL-DTI reconstructs the original heterogeneous network to predict the potential DTIs. The experiments on the public dataset show that SHGCL-DTI has significant improvement in different scenarios, compared with existing state-of-the-art methods. We also demonstrate that the contrastive learning module improves the prediction performance and generalization ability of SHGCL-DTI through ablation study. In addition, we have found several novel predicted DTIs supported by the biological literature. The data and source code are available at: https://github.com/TOJSSE-iData/SHGCL-DTI.
Collapse
Affiliation(s)
- Kainan Yao
- School of Software Engineering, Tongji University, 4800 Caoan Road, Jiading District, Shanghai, 201804, China
| | - Xiaowen Wang
- School of Software Engineering, Tongji University, 4800 Caoan Road, Jiading District, Shanghai, 201804, China
| | - Wannian Li
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department of Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Yangpu District, Shanghai, 200092, China.
| | - Hongming Zhu
- School of Software Engineering, Tongji University, 4800 Caoan Road, Jiading District, Shanghai, 201804, China
| | - Yizhi Jiang
- School of Software Engineering, Tongji University, 4800 Caoan Road, Jiading District, Shanghai, 201804, China
| | - Yulong Li
- School of Software Engineering, Tongji University, 4800 Caoan Road, Jiading District, Shanghai, 201804, China
| | - Tongxuan Tian
- School of Software Engineering, Tongji University, 4800 Caoan Road, Jiading District, Shanghai, 201804, China
| | - Zhaoyi Yang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, No. 96, JinZhai Road Baohe District, Hefei, 230001, Anhui, China.
| | - Qi Liu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department of Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Yangpu District, Shanghai, 200092, China.
| | - Qin Liu
- School of Software Engineering, Tongji University, 4800 Caoan Road, Jiading District, Shanghai, 201804, China.
| |
Collapse
|
35
|
Teng S, Yin C, Wang Y, Chen X, Yan Z, Cui L, Wei L. MolFPG: Multi-level fingerprint-based Graph Transformer for accurate and robust drug toxicity prediction. Comput Biol Med 2023; 164:106904. [PMID: 37453376 DOI: 10.1016/j.compbiomed.2023.106904] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/20/2023] [Accepted: 04/10/2023] [Indexed: 07/18/2023]
Abstract
Drug toxicity prediction is essential to drug development, which can help screen compounds with potential toxicity and reduce the cost and risk of animal experiments and clinical trials. However, traditional handcrafted feature-based and molecular-graph-based approaches are insufficient for molecular representation learning. To address the problem, we developed an innovative molecular fingerprint Graph Transformer framework (MolFPG) with a global-aware module for interpretable toxicity prediction. Our approach encodes compounds using multiple molecular fingerprinting techniques and integrates Graph Transformer-based molecular representation for feature learning and toxic prediction. Experimental results show that our proposed approach has high accuracy and reliability in predicting drug toxicity. In addition, we explored the relationship between drug features and toxicity through an interpretive analysis approach, which improved the interpretability of the approach. Our results highlight the potential of Graph Transformers and multi-level fingerprints for accelerating the drug discovery process by reliably, effectively alarming drug safety. We believe that our study will provide vital support and reference for further development in the field of drug development and toxicity assessment.
Collapse
Affiliation(s)
- Saisai Teng
- School of Software, Shandong University, Jinan, China; Joint SDU-NTU Centre for Artificial Intelligence Research (C-FAIR), Shandong University, Jinan, China
| | - Chenglin Yin
- School of Software, Shandong University, Jinan, China; Joint SDU-NTU Centre for Artificial Intelligence Research (C-FAIR), Shandong University, Jinan, China
| | - Yu Wang
- School of Software, Shandong University, Jinan, China; Joint SDU-NTU Centre for Artificial Intelligence Research (C-FAIR), Shandong University, Jinan, China
| | | | - Zhongmin Yan
- School of Software, Shandong University, Jinan, China; Joint SDU-NTU Centre for Artificial Intelligence Research (C-FAIR), Shandong University, Jinan, China.
| | - Lizhen Cui
- School of Software, Shandong University, Jinan, China; Joint SDU-NTU Centre for Artificial Intelligence Research (C-FAIR), Shandong University, Jinan, China.
| | - Leyi Wei
- School of Software, Shandong University, Jinan, China; Joint SDU-NTU Centre for Artificial Intelligence Research (C-FAIR), Shandong University, Jinan, China.
| |
Collapse
|
36
|
Sinha K, Ghosh N, Sil PC. A Review on the Recent Applications of Deep Learning in Predictive Drug Toxicological Studies. Chem Res Toxicol 2023; 36:1174-1205. [PMID: 37561655 DOI: 10.1021/acs.chemrestox.2c00375] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
Drug toxicity prediction is an important step in ensuring patient safety during drug design studies. While traditional preclinical studies have historically relied on animal models to evaluate toxicity, recent advances in deep-learning approaches have shown great promise in advancing drug safety science and reducing animal use in preclinical studies. However, deep-learning-based approaches also face challenges in handling large biological data sets, model interpretability, and regulatory acceptance. In this review, we provide an overview of recent developments in deep-learning-based approaches for predicting drug toxicity, highlighting their potential advantages over traditional methods and the need to address their limitations. Deep-learning models have demonstrated excellent performance in predicting toxicity outcomes from various data sources such as chemical structures, genomic data, and high-throughput screening assays. The potential of deep learning for automated feature engineering is also discussed. This review emphasizes the need to address ethical concerns related to the use of deep learning in drug toxicity studies, including the reduction of animal use and ensuring regulatory acceptance. Furthermore, emerging applications of deep learning in drug toxicity prediction, such as predicting drug-drug interactions and toxicity in rare subpopulations, are highlighted. The integration of deep-learning-based approaches with traditional methods is discussed as a way to develop more reliable and efficient predictive models for drug safety assessment, paving the way for safer and more effective drug discovery and development. Overall, this review highlights the critical role of deep learning in predictive toxicology and drug safety evaluation, emphasizing the need for continued research and development in this rapidly evolving field. By addressing the limitations of traditional methods, leveraging the potential of deep learning for automated feature engineering, and addressing ethical concerns, deep-learning-based approaches have the potential to revolutionize drug toxicity prediction and improve patient safety in drug discovery and development.
Collapse
Affiliation(s)
- Krishnendu Sinha
- Department of Zoology, Jhargram Raj College, Jhargram 721507, West Bengal, India
| | - Nabanita Ghosh
- Department of Zoology, Maulana Azad College, Kolkata 700013, West Bengal, India
| | - Parames C Sil
- Division of Molecular Medicine, Bose Institute, Kolkata 700054, West Bengal, India
| |
Collapse
|
37
|
Lin X, Dai L, Zhou Y, Yu ZG, Zhang W, Shi JY, Cao DS, Zeng L, Chen H, Song B, Yu PS, Zeng X. Comprehensive evaluation of deep and graph learning on drug-drug interactions prediction. Brief Bioinform 2023:bbad235. [PMID: 37401373 DOI: 10.1093/bib/bbad235] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/30/2023] [Accepted: 06/05/2023] [Indexed: 07/05/2023] Open
Abstract
Recent advances and achievements of artificial intelligence (AI) as well as deep and graph learning models have established their usefulness in biomedical applications, especially in drug-drug interactions (DDIs). DDIs refer to a change in the effect of one drug to the presence of another drug in the human body, which plays an essential role in drug discovery and clinical research. DDIs prediction through traditional clinical trials and experiments is an expensive and time-consuming process. To correctly apply the advanced AI and deep learning, the developer and user meet various challenges such as the availability and encoding of data resources, and the design of computational methods. This review summarizes chemical structure based, network based, natural language processing based and hybrid methods, providing an updated and accessible guide to the broad researchers and development community with different domain knowledge. We introduce widely used molecular representation and describe the theoretical frameworks of graph neural network models for representing molecular structures. We present the advantages and disadvantages of deep and graph learning methods by performing comparative experiments. We discuss the potential technical challenges and highlight future directions of deep and graph learning models for accelerating DDIs prediction.
Collapse
Affiliation(s)
- Xuan Lin
- College of Computer Science, Xiangtan University, Xiangtan, China
| | - Lichang Dai
- College of Computer Science, Xiangtan University, Xiangtan, China
| | - Yafang Zhou
- College of Computer Science, Xiangtan University, Xiangtan, China
| | - Zu-Guo Yu
- Key Laboratory of Intelligent Computing and Information Processing of Ministry of Education, Xiangtan University, Xiangtan, China
| | - Wen Zhang
- College of Informatics, Huazhong Agricultural University, China
| | - Jian-Yu Shi
- Northwestern Polytechnical University, Xian, China
| | - Dong-Sheng Cao
- Xiangya School of Pharmaceutical Sciences, Central South University, China
| | - Li Zeng
- AIDD department of Yuyao Biotech, Shanghai, China
| | - Haowen Chen
- College of Computer Science and Electronic Engineering, Hunan University, 410013 Changsha, P. R. China
| | - Bosheng Song
- College of Information Science and Engineering, Hunan University, Changsha, China
| | - Philip S Yu
- University of Illinois at Chicago and also holds the Wexler Chair in Information Technology
| | - Xiangxiang Zeng
- College of Information Science and Engineering, Hunan University, Changsha, China
| |
Collapse
|
38
|
Zhao Y, Zeng H, Zheng H, Wu J, Kong W, Dai G. A bidirectional interaction-based hybrid network architecture for EEG cognitive recognition. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2023; 238:107593. [PMID: 37209578 DOI: 10.1016/j.cmpb.2023.107593] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/04/2023] [Accepted: 05/08/2023] [Indexed: 05/22/2023]
Abstract
BACKGROUND AND OBJECTIVE Extracting cognitive representation and computational representation information simultaneously from electroencephalography (EEG) data and constructing corresponding information interaction models can effectively improve the recognition capability of brain cognitive status. However, due to the huge gap in the interaction between the two types of information, existing studies have yet to consider the advantages of the interaction of both. METHODS This paper introduces a novel architecture named the bidirectional interaction-based hybrid network (BIHN) for EEG cognitive recognition. BIHN consists of two networks: a cognitive-based network named CogN (e.g., graph convolution network, GCN; capsule network, CapsNet) and a computing-based network named ComN (e.g., EEGNet). CogN is responsible for extracting cognitive representation features from EEG data, while ComN is responsible for extracting computational representation features. Additionally, a bidirectional distillation-based coadaptation (BDC) algorithm is proposed to facilitate information interaction between CogN and ComN to realize the coadaptation of the two networks through bidirectional closed-loop feedback. RESULTS Cross-subject cognitive recognition experiments were performed on the Fatigue-Awake EEG dataset (FAAD, 2-class classification) and SEED dataset (3-class classification), and hybrid network pairs of GCN + EEGNet and CapsNet + EEGNet were verified. The proposed method achieved average accuracies of 78.76% (GCN + EEGNet) and 77.58% (CapsNet + EEGNet) on FAAD and 55.38% (GCN + EEGNet) and 55.10% (CapsNet + EEGNet) on SEED, outperforming the hybrid networks without the bidirectional interaction strategy. CONCLUSIONS Experimental results show that BIHN can achieve superior performance on two EEG datasets and enhance the ability of both CogN and ComN in EEG processing as well as cognitive recognition. We also validated its effectiveness with different hybrid network pairs. The proposed method could greatly promote the development of brain-computer collaborative intelligence.
Collapse
Affiliation(s)
- Yue Zhao
- School of Computer Science and Technology, Hangzhou Dianzi University, Hangzhou 310018, China
| | - Hong Zeng
- School of Computer Science and Technology, Hangzhou Dianzi University, Hangzhou 310018, China; Key Laboratory of Brain Machine Collaborative Intelligence of Zhejiang Province, Hangzhou 310018, China.
| | - Haohao Zheng
- School of Computer Science and Technology, Hangzhou Dianzi University, Hangzhou 310018, China
| | - Jing Wu
- School of Computer Science and Technology, Hangzhou Dianzi University, Hangzhou 310018, China
| | - Wanzeng Kong
- School of Computer Science and Technology, Hangzhou Dianzi University, Hangzhou 310018, China; Key Laboratory of Brain Machine Collaborative Intelligence of Zhejiang Province, Hangzhou 310018, China
| | - Guojun Dai
- School of Computer Science and Technology, Hangzhou Dianzi University, Hangzhou 310018, China; Key Laboratory of Brain Machine Collaborative Intelligence of Zhejiang Province, Hangzhou 310018, China.
| |
Collapse
|
39
|
Zulfiqar H, Ahmed Z, Kissanga Grace-Mercure B, Hassan F, Zhang ZY, Liu F. Computational prediction of promotors in Agrobacterium tumefaciens strain C58 by using the machine learning technique. Front Microbiol 2023; 14:1170785. [PMID: 37125199 PMCID: PMC10133480 DOI: 10.3389/fmicb.2023.1170785] [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: 02/21/2023] [Accepted: 03/17/2023] [Indexed: 05/02/2023] Open
Abstract
Promotors are those genomic regions on the upstream of genes, which are bound by RNA polymerase for starting gene transcription. Because it is the most critical element of gene expression, the recognition of promoters is crucial to understand the regulation of gene expression. This study aimed to develop a machine learning-based model to predict promotors in Agrobacterium tumefaciens (A. tumefaciens) strain C58. In the model, promotor sequences were encoded by three different kinds of feature descriptors, namely, accumulated nucleotide frequency, k-mer nucleotide composition, and binary encodings. The obtained features were optimized by using correlation and the mRMR-based algorithm. These optimized features were inputted into a random forest (RF) classifier to discriminate promotor sequences from non-promotor sequences in A. tumefaciens strain C58. The examination of 10-fold cross-validation showed that the proposed model could yield an overall accuracy of 0.837. This model will provide help for the study of promoters in A. tumefaciens C58 strain.
Collapse
Affiliation(s)
- Hasan Zulfiqar
- Yangtze Delta Region Institute (Huzhou), University of Electronic Science and Technology of China, Huzhou, China
- School of Life Science and Technology and Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu, China
| | - Zahoor Ahmed
- Yangtze Delta Region Institute (Huzhou), University of Electronic Science and Technology of China, Huzhou, China
| | - Bakanina Kissanga Grace-Mercure
- School of Life Science and Technology and Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu, China
| | - Farwa Hassan
- School of Life Science and Technology and Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu, China
| | - Zhao-Yue Zhang
- School of Life Science and Technology and Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu, China
| | - Fen Liu
- Department of Radiation Oncology, Peking University Cancer Hospital (Inner Mongolia Campus), Affiliated Cancer Hospital of Inner Mongolia Medical University, Inner Mongolia Cancer Hospital, Hohhot, China
| |
Collapse
|
40
|
Tian Z, Yu Y, Fang H, Xie W, Guo M. Predicting microbe-drug associations with structure-enhanced contrastive learning and self-paced negative sampling strategy. Brief Bioinform 2023; 24:7009077. [PMID: 36715986 DOI: 10.1093/bib/bbac634] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/19/2022] [Accepted: 12/29/2022] [Indexed: 01/31/2023] Open
Abstract
MOTIVATION Predicting the associations between human microbes and drugs (MDAs) is one critical step in drug development and precision medicine areas. Since discovering these associations through wet experiments is time-consuming and labor-intensive, computational methods have already been an effective way to tackle this problem. Recently, graph contrastive learning (GCL) approaches have shown great advantages in learning the embeddings of nodes from heterogeneous biological graphs (HBGs). However, most GCL-based approaches don't fully capture the rich structure information in HBGs. Besides, fewer MDA prediction methods could screen out the most informative negative samples for effectively training the classifier. Therefore, it still needs to improve the accuracy of MDA predictions. RESULTS In this study, we propose a novel approach that employs the Structure-enhanced Contrastive learning and Self-paced negative sampling strategy for Microbe-Drug Association predictions (SCSMDA). Firstly, SCSMDA constructs the similarity networks of microbes and drugs, as well as their different meta-path-induced networks. Then SCSMDA employs the representations of microbes and drugs learned from meta-path-induced networks to enhance their embeddings learned from the similarity networks by the contrastive learning strategy. After that, we adopt the self-paced negative sampling strategy to select the most informative negative samples to train the MLP classifier. Lastly, SCSMDA predicts the potential microbe-drug associations with the trained MLP classifier. The embeddings of microbes and drugs learning from the similarity networks are enhanced with the contrastive learning strategy, which could obtain their discriminative representations. Extensive results on three public datasets indicate that SCSMDA significantly outperforms other baseline methods on the MDA prediction task. Case studies for two common drugs could further demonstrate the effectiveness of SCSMDA in finding novel MDA associations. AVAILABILITY The source code is publicly available on GitHub https://github.com/Yue-Yuu/SCSMDA-master.
Collapse
Affiliation(s)
- Zhen Tian
- School of Computer and Artificial Intelligence, Zhengzhou University, Zhengzhou 450000, China
| | - Yue Yu
- School of Computer and Artificial Intelligence, Zhengzhou University, Zhengzhou 450000, China
| | - Haichuan Fang
- School of Computer and Artificial Intelligence, Zhengzhou University, Zhengzhou 450000, China
| | - Weixin Xie
- Institute of Intelligent System and Bioinformatics, College of Intelligent Systems Science and Engineering, Harbin Engineering University, Harbin, 150000, China
| | - Maozu Guo
- School of Electrical and Information Engineering, Beijing University of Civil Engineering and Architecture, 100044, Beijing, China
| |
Collapse
|
41
|
Li M, Cai X, Xu S, Ji H. Metapath-aggregated heterogeneous graph neural network for drug-target interaction prediction. Brief Bioinform 2023; 24:6966534. [PMID: 36592060 DOI: 10.1093/bib/bbac578] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 11/03/2022] [Accepted: 11/26/2022] [Indexed: 01/03/2023] Open
Abstract
Drug-target interaction (DTI) prediction is an essential step in drug repositioning. A few graph neural network (GNN)-based methods have been proposed for DTI prediction using heterogeneous biological data. However, existing GNN-based methods only aggregate information from directly connected nodes restricted in a drug-related or a target-related network and are incapable of capturing high-order dependencies in the biological heterogeneous graph. In this paper, we propose a metapath-aggregated heterogeneous graph neural network (MHGNN) to capture complex structures and rich semantics in the biological heterogeneous graph for DTI prediction. Specifically, MHGNN enhances heterogeneous graph structure learning and high-order semantics learning by modeling high-order relations via metapaths. Additionally, MHGNN enriches high-order correlations between drug-target pairs (DTPs) by constructing a DTP correlation graph with DTPs as nodes. We conduct extensive experiments on three biological heterogeneous datasets. MHGNN favorably surpasses 17 state-of-the-art methods over 6 evaluation metrics, which verifies its efficacy for DTI prediction. The code is available at https://github.com/Zora-LM/MHGNN-DTI.
Collapse
Affiliation(s)
- Mei Li
- Tianjin Key Laboratory of Network and Data Security Technology, China.,College of Computer Science, Nankai University, 300350, Tianjin, China
| | - Xiangrui Cai
- Tianjin Key Laboratory of Network and Data Security Technology, China.,College of Computer Science, Nankai University, 300350, Tianjin, China
| | - Sihan Xu
- Tianjin Key Laboratory of Network and Data Security Technology, China.,College of Cyber Science, Nankai University, 300350, Tianjin, China
| | - Hua Ji
- Tianjin Key Laboratory of Network and Data Security Technology, China.,College of Computer Science, Nankai University, 300350, Tianjin, China
| |
Collapse
|
42
|
Su W, Deng S, Gu Z, Yang K, Ding H, Chen H, Zhang Z. Prediction of apoptosis protein subcellular location based on amphiphilic pseudo amino acid composition. Front Genet 2023; 14:1157021. [PMID: 36926588 PMCID: PMC10011625 DOI: 10.3389/fgene.2023.1157021] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 02/20/2023] [Indexed: 03/08/2023] Open
Abstract
Introduction: Apoptosis proteins play an important role in the process of cell apoptosis, which makes the rate of cell proliferation and death reach a relative balance. The function of apoptosis protein is closely related to its subcellular location, it is of great significance to study the subcellular locations of apoptosis proteins. Many efforts in bioinformatics research have been aimed at predicting their subcellular location. However, the subcellular localization of apoptotic proteins needs to be carefully studied. Methods: In this paper, based on amphiphilic pseudo amino acid composition and support vector machine algorithm, a new method was proposed for the prediction of apoptosis proteins\x{2019} subcellular location. Results and Discussion: The method achieved good performance on three data sets. The Jackknife test accuracy of the three data sets reached 90.5%, 93.9% and 84.0%, respectively. Compared with previous methods, the prediction accuracies of APACC_SVM were improved.
Collapse
Affiliation(s)
- Wenxia Su
- College of Science, Inner Mongolia Agriculture University, Hohhot, China
| | - Shuyi Deng
- School of Life Science and Technology, Center for Information Biology, University of Electronic Science and Technology of China, Chengdu, China
| | - Zhifeng Gu
- School of Life Science and Technology, Center for Information Biology, University of Electronic Science and Technology of China, Chengdu, China
| | - Keli Yang
- Nonlinear Research Institute, Baoji University of Arts and Sciences, Baoji, China
| | - Hui Ding
- School of Life Science and Technology, Center for Information Biology, University of Electronic Science and Technology of China, Chengdu, China
| | - Hui Chen
- School of Healthcare Technology, Chengdu Neusoft University, Chengdu, China
| | - Zhaoyue Zhang
- School of Life Science and Technology, Center for Information Biology, University of Electronic Science and Technology of China, Chengdu, China.,School of Healthcare Technology, Chengdu Neusoft University, Chengdu, China
| |
Collapse
|
43
|
Lin S, Chen W, Chen G, Zhou S, Wei DQ, Xiong Y. MDDI-SCL: predicting multi-type drug-drug interactions via supervised contrastive learning. J Cheminform 2022; 14:81. [DOI: 10.1186/s13321-022-00659-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 11/05/2022] [Indexed: 11/16/2022] Open
Abstract
AbstractThe joint use of multiple drugs may cause unintended drug-drug interactions (DDIs) and result in adverse consequence to the patients. Accurate identification of DDI types can not only provide hints to avoid these accidental events, but also elaborate the underlying mechanisms by how DDIs occur. Several computational methods have been proposed for multi-type DDI prediction, but room remains for improvement in prediction performance. In this study, we propose a supervised contrastive learning based method, MDDI-SCL, implemented by three-level loss functions, to predict multi-type DDIs. MDDI-SCL is mainly composed of three modules: drug feature encoder and mean squared error loss module, drug latent feature fusion and supervised contrastive loss module, multi-type DDI prediction and classification loss module. The drug feature encoder and mean squared error loss module uses self-attention mechanism and autoencoder to learn drug-level latent features. The drug latent feature fusion and supervised contrastive loss module uses multi-scale feature fusion to learn drug pair-level latent features. The prediction and classification loss module predicts DDI types of each drug pair. We evaluate MDDI-SCL on three different tasks of two datasets. Experimental results demonstrate that MDDI-SCL achieves better or comparable performance as the state-of-the-art methods. Furthermore, the effectiveness of supervised contrastive learning is validated by ablation experiment, and the feasibility of MDDI-SCL is supported by case studies. The source codes are available at https://github.com/ShenggengLin/MDDI-SCL.
Collapse
|