51
|
Shoombuatong W, Homdee N, Schaduangrat N, Chumnanpuen P. Leveraging a meta-learning approach to advance the accuracy of Na v blocking peptides prediction. Sci Rep 2024; 14:4463. [PMID: 38396246 PMCID: PMC10891130 DOI: 10.1038/s41598-024-55160-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: 12/28/2023] [Accepted: 02/21/2024] [Indexed: 02/25/2024] Open
Abstract
The voltage-gated sodium (Nav) channel is a crucial molecular component responsible for initiating and propagating action potentials. While the α subunit, forming the channel pore, plays a central role in this function, the complete physiological function of Nav channels relies on crucial interactions between the α subunit and auxiliary proteins, known as protein-protein interactions (PPI). Nav blocking peptides (NaBPs) have been recognized as a promising and alternative therapeutic agent for pain and itch. Although traditional experimental methods can precisely determine the effect and activity of NaBPs, they remain time-consuming and costly. Hence, machine learning (ML)-based methods that are capable of accurately contributing in silico prediction of NaBPs are highly desirable. In this study, we develop an innovative meta-learning-based NaBP prediction method (MetaNaBP). MetaNaBP generates new feature representations by employing a wide range of sequence-based feature descriptors that cover multiple perspectives, in combination with powerful ML algorithms. Then, these feature representations were optimized to identify informative features using a two-step feature selection method. Finally, the selected informative features were applied to develop the final meta-predictor. To the best of our knowledge, MetaNaBP is the first meta-predictor for NaBP prediction. Experimental results demonstrated that MetaNaBP achieved an accuracy of 0.948 and a Matthews correlation coefficient of 0.898 over the independent test dataset, which were 5.79% and 11.76% higher than the existing method. In addition, the discriminative power of our feature representations surpassed that of conventional feature descriptors over both the training and independent test datasets. We anticipate that MetaNaBP will be exploited for the large-scale prediction and analysis of NaBPs to narrow down the potential NaBPs.
Collapse
Affiliation(s)
- Watshara Shoombuatong
- Center for Research Innovation and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand.
| | - Nutta Homdee
- Center for Research Innovation and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand
| | - Nalini Schaduangrat
- Center for Research Innovation and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand
| | - Pramote Chumnanpuen
- Department of Zoology, Faculty of Science, Kasetsart University, Bangkok, 10900, Thailand
- Omics Center for Agriculture, Bioresources, Food, and Health, Kasetsart University (OmiKU), Bangkok, 10900, Thailand
| |
Collapse
|
52
|
Charoenkwan P, Chumnanpuen P, Schaduangrat N, Shoombuatong W. Accelerating the identification of the allergenic potential of plant proteins using a stacked ensemble-learning framework. J Biomol Struct Dyn 2024:1-13. [PMID: 38385478 DOI: 10.1080/07391102.2024.2318482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/08/2024] [Indexed: 02/23/2024]
Abstract
Plant-allergenic proteins (PAPs) have the potential to induce allergic reactions in certain individuals. While these proteins are generally innocuous for the majority of people, they can elicit an immune response in those with particular sensitivities. Thus, screening and prioritizing the allergenic potential of plant proteins is indispensable for the development of diagnostic tools, therapeutic interventions or medications to treat allergic reactions. However, investigating the allergenic potential of plant proteins based on experimental methods is costly and labour-intensive. Therefore, we develop StackPAP, a three-layer stacking ensemble framework for accurate large-scale identification of PAPs. In StackPAP, at the first layer, we conducted a comprehensive analysis of an extensive set of feature descriptors. Subsequently, we selected and fused five potential sequence-based feature descriptors, including amphiphilic pseudo-amino acid composition, dipeptide deviation from expected mean, amino acid composition, pseudo amino acid composition and dipeptide composition. Additionally, we applied an efficient genetic algorithm (GA-SAR) to determine informative feature sets. In the second layer, 12 powerful machine learning (ML) methods, in combination with all the informative feature sets, were employed to construct a pool of base classifiers. Finally, 13 potential base classifiers were selected using the GA-SAR method and combined to develop the final meta-classifier. Our experimental results revealed the promising prediction performance of StackPAP, with an accuracy, Matthew's correlation coefficient and AUC of 0.984, 0.969 and 0.993, respectively, as judged by the independent test dataset. In conclusion, both cross-validation and independent test results indicated the superior performance of StackPAP compared with several ML-based classifiers. To accelerate the identification of the allergenicity of plant proteins, we developed a user-friendly web server for StackPAP (https://pmlabqsar.pythonanywhere.com/StackPAP). We anticipate that StackPAP will be an efficient and useful tool for rapidly screening PAPs from a vast number of plant proteins.
Collapse
Affiliation(s)
- Phasit Charoenkwan
- Modern Management and Information Technology, College of Arts, Media and Technology, Chiang Mai University, Thailand
| | - Pramote Chumnanpuen
- Department of Zoology, Faculty of Science, Kasetsart University, Bangkok, Thailand
- Omics Center for Agriculture, Bioresources, Food, and Health, Kasetsart University (OmiKU), Bangkok, Thailand
| | - Nalini Schaduangrat
- Center for Research Innovation and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Watshara Shoombuatong
- Center for Research Innovation and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| |
Collapse
|
53
|
Karakaya O, Kilimci ZH. An efficient consolidation of word embedding and deep learning techniques for classifying anticancer peptides: FastText+BiLSTM. PeerJ Comput Sci 2024; 10:e1831. [PMID: 38435607 PMCID: PMC10909209 DOI: 10.7717/peerj-cs.1831] [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: 09/19/2023] [Accepted: 12/31/2023] [Indexed: 03/05/2024]
Abstract
Anticancer peptides (ACPs) are a group of peptides that exhibit antineoplastic properties. The utilization of ACPs in cancer prevention can present a viable substitute for conventional cancer therapeutics, as they possess a higher degree of selectivity and safety. Recent scientific advancements generate an interest in peptide-based therapies which offer the advantage of efficiently treating intended cells without negatively impacting normal cells. However, as the number of peptide sequences continues to increase rapidly, developing a reliable and precise prediction model becomes a challenging task. In this work, our motivation is to advance an efficient model for categorizing anticancer peptides employing the consolidation of word embedding and deep learning models. First, Word2Vec, GloVe, FastText, One-Hot-Encoding approaches are evaluated as embedding techniques for the purpose of extracting peptide sequences. Then, the output of embedding models are fed into deep learning approaches CNN, LSTM, BiLSTM. To demonstrate the contribution of proposed framework, extensive experiments are carried on widely-used datasets in the literature, ACPs250 and independent. Experiment results show the usage of proposed model enhances classification accuracy when compared to the state-of-the-art studies. The proposed combination, FastText+BiLSTM, exhibits 92.50% of accuracy for ACPs250 dataset, and 96.15% of accuracy for the Independent dataset, thence determining new state-of-the-art.
Collapse
Affiliation(s)
- Onur Karakaya
- Research and Development Inc., Turkcell Technology, İstanbul, Turkey
| | - Zeynep Hilal Kilimci
- Department of Information Systems Engineering, Kocaeli University, Kocaeli, Turkey
| |
Collapse
|
54
|
Karim T, Shaon MSH, Sultan MF, Hasan MZ, Kafy AA. ANNprob-ACPs: A novel anticancer peptide identifier based on probabilistic feature fusion approach. Comput Biol Med 2024; 169:107915. [PMID: 38171261 DOI: 10.1016/j.compbiomed.2023.107915] [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: 08/20/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/05/2024]
Abstract
Anticancer Peptides (ACPs) offer significant potential as cancer treatment drugs in this modern era. Quickly identifying active compounds from protein sequences is crucial for healthcare and cancer treatment. In this paper ANNprob-ACPs, a novel and effective model for detecting ACPs has been implemented based on nine feature encoding techniques, including AAC, CC, W2V, DPC, PAAC, QSO, CTDC, CTDT, and CKSAAGP. After analyzing the performance of several machine learning models, the six best models were selected based on their overall performances in every evaluation metric. The probability scores of each model were subsequently aggregated and used as input of our meta- model, called ANNprob-ACPs. Our model outperformed all others and its potential to lead to phenomenal identification of ACPs. The results of this study showed notable improvement in 10-fold cross-validation and independent test, with accuracy of 93.72% and 90.62%, respectively. Our proposed model, ANNprob-ACPs outperformed existing approaches in terms of accuracy and effectiveness in discovering ACPs. By using SHAP, this study obtained the physicochemical properties of QSO, and compositional properties of DPC, AAC, and PAAC are more impactful for our model's performances, which have a major impact on a drug's interactions and future discoveries. Consequently, this model is crucial for the future and has a high probability of detecting ACPs more frequently. We developed a web server of ANNprob-ACPs, which is accessible at ANNprob-ACPs webserver.
Collapse
Affiliation(s)
- Tasmin Karim
- Department of Computer Science & Engineering, Daffodil International University, Daffodil Smart City, Birulia, Dhaka, 1216, Bangladesh; Health Informatics Research Lab, Department of Computer Science and Engineering, Daffodil International University, Daffodil Smart City, Birulia, Dhaka, 1216, Bangladesh.
| | - Md Shazzad Hossain Shaon
- Department of Computer Science & Engineering, Daffodil International University, Daffodil Smart City, Birulia, Dhaka, 1216, Bangladesh; Health Informatics Research Lab, Department of Computer Science and Engineering, Daffodil International University, Daffodil Smart City, Birulia, Dhaka, 1216, Bangladesh.
| | - Md Fahim Sultan
- Department of Computer Science & Engineering, Daffodil International University, Daffodil Smart City, Birulia, Dhaka, 1216, Bangladesh; Health Informatics Research Lab, Department of Computer Science and Engineering, Daffodil International University, Daffodil Smart City, Birulia, Dhaka, 1216, Bangladesh.
| | - Md Zahid Hasan
- Department of Computer Science & Engineering, Daffodil International University, Daffodil Smart City, Birulia, Dhaka, 1216, Bangladesh; Health Informatics Research Lab, Department of Computer Science and Engineering, Daffodil International University, Daffodil Smart City, Birulia, Dhaka, 1216, Bangladesh.
| | - Abdulla-Al Kafy
- Department of Urban & Regional Planning, Rajshahi University of Engineering & Technology (RUET), Rajshahi, 6204, Bangladesh.
| |
Collapse
|
55
|
Zuo W, Kwok HF. Design of Bioengineered Peptides/Proteases as Anti-cancer Reagents with Integrated Omics and Machine Learning Approaches. Methods Mol Biol 2024; 2747:295-309. [PMID: 38038948 DOI: 10.1007/978-1-0716-3589-6_22] [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] [Indexed: 12/02/2023]
Abstract
Cancer is a heterogeneous disorder of uncontrolled growth of cells, which has proven to be a major burden worldwide. Many treatment options are available for cancer therapy, yet side effects and drug resistance remain major hurdles. Therefore, it is necessary to develop novel drugs for cancer therapy. Anti-cancer peptides (ACPs) are attractive candidates with remarkable potency, low toxicity, and high specificity advantages. However, traditional experimental identification of ACPs is time-consuming and expensive. Integrated omics combined with machine learning (ML) is considered a new powerful and cost-effective strategy to discover ACPs from natural products. In this chapter, we describe in detail experimental procedures for collecting both transcriptomic and proteomic data from venoms, followed by descriptive approaches to ML prediction.
Collapse
Affiliation(s)
- Weimin Zuo
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR, China
- School of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR, China
| | - Hang Fai Kwok
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR, China.
- School of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR, China.
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Avenida de Universidade, Taipa, Macau SAR, China.
| |
Collapse
|
56
|
Li C, Jin K. Chemical Strategies towards the Development of Effective Anticancer Peptides. Curr Med Chem 2024; 31:1839-1873. [PMID: 37170992 DOI: 10.2174/0929867330666230426111157] [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: 11/01/2022] [Revised: 01/28/2023] [Accepted: 02/24/2023] [Indexed: 05/13/2023]
Abstract
Cancer is increasingly recognized as one of the primary causes of death and has become a multifaceted global health issue. Modern medical science has made significant advancements in the diagnosis and therapy of cancer over the past decade. The detrimental side effects, lack of efficacy, and multidrug resistance of conventional cancer therapies have created an urgent need for novel anticancer therapeutics or treatments with low cytotoxicity and drug resistance. The pharmaceutical groups have recognized the crucial role that peptide therapeutic agents can play in addressing unsatisfied healthcare demands and how these become great supplements or even preferable alternatives to biological therapies and small molecules. Anticancer peptides, as a vibrant therapeutic strategy against various cancer cells, have demonstrated incredible anticancer potential due to high specificity and selectivity, low toxicity, and the ability to target the surface of traditional "undruggable" proteins. This review will provide the research progression of anticancer peptides, mainly focusing on the discovery and modifications along with the optimization and application of these peptides in clinical practice.
Collapse
Affiliation(s)
- Cuicui Li
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Medicinal Chemistry, School of Pharmacy, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Kang Jin
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Medicinal Chemistry, School of Pharmacy, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| |
Collapse
|
57
|
La Paglia L, Vazzana M, Mauro M, Urso A, Arizza V, Vizzini A. Bioactive Molecules from the Innate Immunity of Ascidians and Innovative Methods of Drug Discovery: A Computational Approach Based on Artificial Intelligence. Mar Drugs 2023; 22:6. [PMID: 38276644 PMCID: PMC10817596 DOI: 10.3390/md22010006] [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: 11/20/2023] [Revised: 12/12/2023] [Accepted: 12/17/2023] [Indexed: 01/27/2024] Open
Abstract
The study of bioactive molecules of marine origin has created an important bridge between biological knowledge and its applications in biotechnology and biomedicine. Current studies in different research fields, such as biomedicine, aim to discover marine molecules characterized by biological activities that can be used to produce potential drugs for human use. In recent decades, increasing attention has been paid to a particular group of marine invertebrates, the Ascidians, as they are a source of bioactive products. We describe omics data and computational methods relevant to identifying the mechanisms and processes of innate immunity underlying the biosynthesis of bioactive molecules, focusing on innovative computational approaches based on Artificial Intelligence. Since there is increasing attention on finding new solutions for a sustainable supply of bioactive compounds, we propose that a possible improvement in the biodiscovery pipeline might also come from the study and utilization of marine invertebrates' innate immunity.
Collapse
Affiliation(s)
- Laura La Paglia
- Istituto di Calcolo e Reti ad Alte Prestazioni–Consiglio Nazionale delle Ricerche, Via Ugo La Malfa 153, 90146 Palermo, Italy; (L.L.P.); (A.U.)
| | - Mirella Vazzana
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche–Università di Palermo, Via Archirafi 18, 90100 Palermo, Italy; (M.V.); (M.M.); (V.A.)
| | - Manuela Mauro
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche–Università di Palermo, Via Archirafi 18, 90100 Palermo, Italy; (M.V.); (M.M.); (V.A.)
| | - Alfonso Urso
- Istituto di Calcolo e Reti ad Alte Prestazioni–Consiglio Nazionale delle Ricerche, Via Ugo La Malfa 153, 90146 Palermo, Italy; (L.L.P.); (A.U.)
| | - Vincenzo Arizza
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche–Università di Palermo, Via Archirafi 18, 90100 Palermo, Italy; (M.V.); (M.M.); (V.A.)
| | - Aiti Vizzini
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche–Università di Palermo, Via Archirafi 18, 90100 Palermo, Italy; (M.V.); (M.M.); (V.A.)
| |
Collapse
|
58
|
Menotti L, Vannini A. Oncolytic Viruses in the Era of Omics, Computational Technologies, and Modeling: Thesis, Antithesis, and Synthesis. Int J Mol Sci 2023; 24:17378. [PMID: 38139207 PMCID: PMC10743452 DOI: 10.3390/ijms242417378] [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: 11/09/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
Oncolytic viruses (OVs) are the frontier therapy for refractory cancers, especially in integration with immunomodulation strategies. In cancer immunovirotherapy, the many available "omics" and systems biology technologies generate at a fast pace a challenging huge amount of data, where apparently clashing information mirrors the complexity of individual clinical situations and OV used. In this review, we present and discuss how currently big data analysis, on one hand and, on the other, simulation, modeling, and computational technologies, provide invaluable support to interpret and integrate "omic" information and drive novel synthetic biology and personalized OV engineering approaches for effective immunovirotherapy. Altogether, these tools, possibly aided in the future by artificial intelligence as well, will allow for the blending of the information into OV recombinants able to achieve tumor clearance in a patient-tailored way. Various endeavors to the envisioned "synthesis" of turning OVs into personalized theranostic agents are presented.
Collapse
Affiliation(s)
- Laura Menotti
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy;
| | | |
Collapse
|
59
|
Ding Y, Zhou H, Zou Q, Yuan L. Identification of drug-side effect association via correntropy-loss based matrix factorization with neural tangent kernel. Methods 2023; 219:73-81. [PMID: 37783242 DOI: 10.1016/j.ymeth.2023.09.008] [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: 07/26/2023] [Revised: 09/18/2023] [Accepted: 09/20/2023] [Indexed: 10/04/2023] Open
Abstract
Adverse drug reactions include side effects, allergic reactions, and secondary infections. Severe adverse reactions can cause cancer, deformity, or mutation. The monitoring of drug side effects is an important support for post marketing safety supervision of drugs, and an important basis for revising drug instructions. Its purpose is to timely detect and control drug safety risks. Traditional methods are time-consuming. To accelerate the discovery of side effects, we propose a machine learning based method, called correntropy-loss based matrix factorization with neural tangent kernel (CLMF-NTK), to solve the prediction of drug side effects. Our method and other computational methods are tested on three benchmark datasets, and the results show that our method achieves the best predictive performance.
Collapse
Affiliation(s)
- Yijie Ding
- Key Laboratory of Computational Science and Application of Hainan Province, Hainan Normal University, Haikou 571158, China; Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou 324000, China; School of Electronic and Information Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Hongmei Zhou
- Beidahuang Industry Group General Hospital, Harbin 150001, China
| | - Quan Zou
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou 324000, China.
| | - Lei Yuan
- Department of Hepatobiliary Surgery, Quzhou People's Hospital, 100# Minjiang Main Road, Quzhou 324000, China.
| |
Collapse
|
60
|
Zou X, Ren L, Cai P, Zhang Y, Ding H, Deng K, Yu X, Lin H, Huang C. Accurately identifying hemagglutinin using sequence information and machine learning methods. Front Med (Lausanne) 2023; 10:1281880. [PMID: 38020152 PMCID: PMC10644030 DOI: 10.3389/fmed.2023.1281880] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Hemagglutinin (HA) is responsible for facilitating viral entry and infection by promoting the fusion between the host membrane and the virus. Given its significance in the process of influenza virus infestation, HA has garnered attention as a target for influenza drug and vaccine development. Thus, accurately identifying HA is crucial for the development of targeted vaccine drugs. However, the identification of HA using in-silico methods is still lacking. This study aims to design a computational model to identify HA. Methods In this study, a benchmark dataset comprising 106 HA and 106 non-HA sequences were obtained from UniProt. Various sequence-based features were used to formulate samples. By perform feature optimization and inputting them four kinds of machine learning methods, we constructed an integrated classifier model using the stacking algorithm. Results and discussion The model achieved an accuracy of 95.85% and with an area under the receiver operating characteristic (ROC) curve of 0.9863 in the 5-fold cross-validation. In the independent test, the model exhibited an accuracy of 93.18% and with an area under the ROC curve of 0.9793. The code can be found from https://github.com/Zouxidan/HA_predict.git. The proposed model has excellent prediction performance. The model will provide convenience for biochemical scholars for the study of HA.
Collapse
Affiliation(s)
- Xidan Zou
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu, China
| | - Liping Ren
- School of Healthcare Technology, Chengdu Neusoft University, Chengdu, China
| | - Peiling Cai
- School of Basic Medical Sciences, Chengdu University, Chengdu, China
| | - Yang Zhang
- Innovative Institute of Chinese Medicine and Pharmacy, Academy for Interdiscipline, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hui Ding
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu, China
| | - Kejun Deng
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaolong Yu
- School of Materials Science and Engineering, Hainan University, Haikou, China
| | - Hao Lin
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu, China
| | - Chengbing Huang
- School of Computer Science and Technology, Aba Teachers University, Aba, China
| |
Collapse
|
61
|
Sun M, Hu H, Pang W, Zhou Y. ACP-BC: A Model for Accurate Identification of Anticancer Peptides Based on Fusion Features of Bidirectional Long Short-Term Memory and Chemically Derived Information. Int J Mol Sci 2023; 24:15447. [PMID: 37895128 PMCID: PMC10607064 DOI: 10.3390/ijms242015447] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 09/10/2023] [Accepted: 10/20/2023] [Indexed: 10/29/2023] Open
Abstract
Anticancer peptides (ACPs) have been proven to possess potent anticancer activities. Although computational methods have emerged for rapid ACPs identification, their accuracy still needs improvement. In this study, we propose a model called ACP-BC, a three-channel end-to-end model that utilizes various combinations of data augmentation techniques. In the first channel, features are extracted from the raw sequence using a bidirectional long short-term memory network. In the second channel, the entire sequence is converted into a chemical molecular formula, which is further simplified using Simplified Molecular Input Line Entry System notation to obtain deep abstract features through a bidirectional encoder representation transformer (BERT). In the third channel, we manually selected four effective features according to dipeptide composition, binary profile feature, k-mer sparse matrix, and pseudo amino acid composition. Notably, the application of chemical BERT in predicting ACPs is novel and successfully integrated into our model. To validate the performance of our model, we selected two benchmark datasets, ACPs740 and ACPs240. ACP-BC achieved prediction accuracy with 87% and 90% on these two datasets, respectively, representing improvements of 1.3% and 7% compared to existing state-of-the-art methods on these datasets. Therefore, systematic comparative experiments have shown that the ACP-BC can effectively identify anticancer peptides.
Collapse
Affiliation(s)
- Mingwei Sun
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun 130012, China; (M.S.); (H.H.)
| | - Haoyuan Hu
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun 130012, China; (M.S.); (H.H.)
| | - Wei Pang
- School of Mathematical and Computer Sciences, Heriot-Watt University, Edinburgh EH14 4AS, UK;
| | - You Zhou
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun 130012, China; (M.S.); (H.H.)
- College of Software, Jilin University, Changchun 130012, China
| |
Collapse
|
62
|
Basith S, Pham NT, Song M, Lee G, Manavalan B. ADP-Fuse: A novel two-layer machine learning predictor to identify antidiabetic peptides and diabetes types using multiview information. Comput Biol Med 2023; 165:107386. [PMID: 37619323 DOI: 10.1016/j.compbiomed.2023.107386] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/03/2023] [Accepted: 08/14/2023] [Indexed: 08/26/2023]
Abstract
Diabetes mellitus has become a major public health concern associated with high mortality and reduced life expectancy and can cause blindness, heart attacks, kidney failure, lower limb amputations, and strokes. A new generation of antidiabetic peptides (ADPs) that act on β-cells or T-cells to regulate insulin production is being developed to alleviate the effects of diabetes. However, the lack of effective peptide-mining tools has hampered the discovery of these promising drugs. Hence, novel computational tools need to be developed urgently. In this study, we present ADP-Fuse, a novel two-layer prediction framework capable of accurately identifying ADPs or non-ADPs and categorizing them into type 1 and type 2 ADPs. First, we comprehensively evaluated 22 peptide sequence-derived features coupled with eight notable machine learning algorithms. Subsequently, the most suitable feature descriptors and classifiers for both layers were identified. The output of these single-feature models, embedded with multiview information, was trained with an appropriate classifier to provide the final prediction. Comprehensive cross-validation and independent tests substantiate that ADP-Fuse surpasses single-feature models and the feature fusion approach for the prediction of ADPs and their types. In addition, the SHapley Additive exPlanation method was used to elucidate the contributions of individual features to the prediction of ADPs and their types. Finally, a user-friendly web server for ADP-Fuse was developed and made publicly accessible (https://balalab-skku.org/ADP-Fuse), enabling the swift screening and identification of novel ADPs and their types. This framework is expected to contribute significantly to antidiabetic peptide identification.
Collapse
Affiliation(s)
- Shaherin Basith
- Department of Physiology, Ajou University School of Medicine, Suwon, 16499, Republic of Korea
| | - Nhat Truong Pham
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Minkyung Song
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea; Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Gwang Lee
- Department of Physiology, Ajou University School of Medicine, Suwon, 16499, Republic of Korea; Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea.
| | - Balachandran Manavalan
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
63
|
Sui J, Chen J, Chen Y, Iwamori N, Sun J. Identification of plant vacuole proteins by using graph neural network and contact maps. BMC Bioinformatics 2023; 24:357. [PMID: 37740195 PMCID: PMC10517492 DOI: 10.1186/s12859-023-05475-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: 05/13/2023] [Accepted: 09/12/2023] [Indexed: 09/24/2023] Open
Abstract
Plant vacuoles are essential organelles in the growth and development of plants, and accurate identification of their proteins is crucial for understanding their biological properties. In this study, we developed a novel model called GraphIdn for the identification of plant vacuole proteins. The model uses SeqVec, a deep representation learning model, to initialize the amino acid sequence. We utilized the AlphaFold2 algorithm to obtain the structural information of corresponding plant vacuole proteins, and then fed the calculated contact maps into a graph convolutional neural network. GraphIdn achieved accuracy values of 88.51% and 89.93% in independent testing and fivefold cross-validation, respectively, outperforming previous state-of-the-art predictors. As far as we know, this is the first model to use predicted protein topology structure graphs to identify plant vacuole proteins. Furthermore, we assessed the effectiveness and generalization capability of our GraphIdn model by applying it to identify and locate peroxisomal proteins, which yielded promising outcomes. The source code and datasets can be accessed at https://github.com/SJNNNN/GraphIdn .
Collapse
Affiliation(s)
- Jianan Sui
- School of Information Science and Engineering, University of Jinan, Jinan, China
| | - Jiazi Chen
- Laboratory of Zoology, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka-Shi, Fukuoka, Japan
| | - Yuehui Chen
- School of Artificial Intelligence Institute and Information Science and Engineering, University of Jinan, Jinan, China.
| | - Naoki Iwamori
- Laboratory of Zoology, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka-Shi, Fukuoka, Japan
| | - Jin Sun
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu, 611731, China
| |
Collapse
|
64
|
Cui Z, Wang SG, He Y, Chen ZH, Zhang QH. DeepTPpred: A Deep Learning Approach With Matrix Factorization for Predicting Therapeutic Peptides by Integrating Length Information. IEEE J Biomed Health Inform 2023; 27:4611-4622. [PMID: 37368803 DOI: 10.1109/jbhi.2023.3290014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
The abuse of traditional antibiotics has led to increased resistance of bacteria and viruses. Efficient therapeutic peptide prediction is critical for peptide drug discovery. However, most of the existing methods only make effective predictions for one class of therapeutic peptides. It is worth noting that currently no predictive method considers sequence length information as a distinct feature of therapeutic peptides. In this article, a novel deep learning approach with matrix factorization for predicting therapeutic peptides (DeepTPpred) by integrating length information are proposed. The matrix factorization layer can learn the potential features of the encoded sequence through the mechanism of first compression and then restoration. And the length features of the sequence of therapeutic peptides are embedded with encoded amino acid sequences. To automatically learn therapeutic peptide predictions, these latent features are input into the neural networks with self-attention mechanism. On eight therapeutic peptide datasets, DeepTPpred achieved excellent prediction results. Based on these datasets, we first integrated eight datasets to obtain a full therapeutic peptide integration dataset. Then, we obtained two functional integration datasets based on the functional similarity of the peptides. Finally, we also conduct experiments on the latest versions of the ACP and CPP datasets. Overall, the experimental results show that our work is effective for the identification of therapeutic peptides.
Collapse
|
65
|
Liu N, Zhang Z, Wu Y, Wang Y, Liang Y. CRBSP:Prediction of CircRNA-RBP Binding Sites Based on Multimodal Intermediate Fusion. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:2898-2906. [PMID: 37130249 DOI: 10.1109/tcbb.2023.3272400] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Circular RNA (CircRNA) is widely expressed and has physiological and pathological significance, regulating post-transcriptional processes via its protein-binding activity. However, whereas much work has been done on linear RNA and RNA binding protein (RBP), little is known about the binding sites of CircRNA. The current report is on the development of a medium-term multimodal data fusion strategy, CRBSP, to predict CircRNA-RBP binding sites. CRBSP represents the CircRNA trinucleotide semantic, location, composition and frequency information as the corresponding coding methods of Word to vector (Word2vec), Position-specific trinucleotide propensity (PSTNP), Pseudo trinucleotide composition (PseTNC) and Trinucleotide nucleotide composition (TNC), respectively. CNN (Convolution Neural Networks) was used to extract global information and BiLSTM (bidirectional Long- and Short-Term Memory network) encoder and LSTM (Long- and Short-Term Memory network) decoder for local sequence information. Enhancement of the contributions of key features by the self-attention mechanism was followed by mid-term fusion of the four enhanced features. Logistic Regression (LR) classifier showed that CRBSP gives a mean AUC value of 0.9362 through 5-fold Cross Validation of all 37 datasets, a performance which is superior to five current state-of-the-art models. Similar evaluation of linear RNA-RBP binding sites gave an AUC value of 0.7615 which is also higher than other prediction methods, demonstrating the robustness of CRBSP.
Collapse
|
66
|
Chen S, Liao Y, Zhao J, Bin Y, Zheng C. PACVP: Prediction of Anti-Coronavirus Peptides Using a Stacking Learning Strategy With Effective Feature Representation. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:3106-3116. [PMID: 37022025 DOI: 10.1109/tcbb.2023.3238370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Due to the global outbreak of COVID-19 and its variants, antiviral peptides with anti-coronavirus activity (ACVPs) represent a promising new drug candidate for the treatment of coronavirus infection. At present, several computational tools have been developed to identify ACVPs, but the overall prediction performance is still not enough to meet the actual therapeutic application. In this study, we constructed an efficient and reliable prediction model PACVP (Prediction of Anti-CoronaVirus Peptides) for identifying ACVPs based on effective feature representation and a two-layer stacking learning framework. In the first layer, we use nine feature encoding methods with different feature representation angles to characterize the rich sequence information and fuse them into a feature matrix. Secondly, data normalization and unbalanced data processing are carried out. Next, 12 baseline models are constructed by combining three feature selection methods and four machine learning classification algorithms. In the second layer, we input the optimal probability features into the logistic regression algorithm (LR) to train the final model PACVP. The experiments show that PACVP achieves favorable prediction performance on independent test dataset, with ACC of 0.9208 and AUC of 0.9465. We hope that PACVP will become a useful method for identifying, annotating and characterizing novel ACVPs.
Collapse
|
67
|
Fan R, Ding Y, Zou Q, Yuan L. Multi-view local hyperplane nearest neighbor model based on independence criterion for identifying vesicular transport proteins. Int J Biol Macromol 2023; 247:125774. [PMID: 37437677 DOI: 10.1016/j.ijbiomac.2023.125774] [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: 05/11/2023] [Revised: 06/30/2023] [Accepted: 07/07/2023] [Indexed: 07/14/2023]
Abstract
Vesicular transport proteins participate in various biological processes and play a significant role in the movement of substances within cells. These proteins are associated with numerous human diseases, making their identification particularly important. In this study, we developed a novel strategy for accurately identifying vesicular transport proteins. We developed a novel multi-view classifier called graph-regularized k-local hyperplane distance nearest neighbor model (HSIC-GHKNN), which combines the Hilbert-Schmidt independence criterion (HSIC)-based multi-view learning method with a local hyperplane distance nearest-neighbor classifier. We first extracted protein evolution information using two feature extraction methods, pseudo-position-specific scoring matrix (PsePSSM) and AATP, and addressed dataset imbalance using the Edited Nearest Neighbors (ENN) algorithm. Subsequently, we employed a local hyperplane distance nearest-neighbor classifier for each view identification and added an HSIC term to maintain independence between views. We then assessed the performance of our identification strategy and analyzed the PsePSSM and AATP feature sets to determine the influencing factors of the classification results. The experimental results demonstrate that the accurate and Matthew correlation coefficients of our strategy on the independent test set are 85.8 % and 0.548, respectively. Our approach outperformed existing methods in most evaluation metrics. In addition, the proposed multi-view classification model can easily be applied to similar identification tasks.
Collapse
Affiliation(s)
- Rui Fan
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu 610054, China; Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou, Zhejiang 324000, China
| | - Yijie Ding
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou, Zhejiang 324000, China.
| | - Quan Zou
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu 610054, China; Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou, Zhejiang 324000, China.
| | - Lei Yuan
- Department of Hepatobiliary Surgery, Quzhou People's Hospital, Quzhou, Zhejiang 324000, China.
| |
Collapse
|
68
|
Shoombuatong W, Schaduangrat N, Nikom J. Empirical comparison and analysis of machine learning-based approaches for druggable protein identification. EXCLI JOURNAL 2023; 22:915-927. [PMID: 37780939 PMCID: PMC10539545 DOI: 10.17179/excli2023-6410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 08/15/2023] [Indexed: 10/03/2023]
Abstract
Efficiently and precisely identifying drug targets is crucial for developing and discovering potential medications. While conventional experimental approaches can accurately pinpoint these targets, they suffer from time constraints and are not easily adaptable to high-throughput processes. On the other hand, computational approaches, particularly those utilizing machine learning (ML), offer an efficient means to accelerate the prediction of druggable proteins based solely on their primary sequences. Recently, several state-of-the-art computational methods have been developed for predicting and analyzing druggable proteins. These computational methods showed high diversity in terms of benchmark datasets, feature extraction schemes, ML algorithms, evaluation strategies and webserver/software usability. Thus, our objective is to reexamine these computational approaches and conduct a comprehensive assessment of their strengths and weaknesses across multiple aspects. In this study, we deliver the first comprehensive survey regarding the state-of-the-art computational approaches for in silico prediction of druggable proteins. First, we provided information regarding the existing benchmark datasets and the types of ML methods employed. Second, we investigated the effectiveness of these computational methods in druggable protein identification for each benchmark dataset. Third, we summarized the important features used in this field and the existing webserver/software. Finally, we addressed the present constraints of the existing methods and offer valuable guidance to the scientific community in designing and developing novel prediction models. We anticipate that this comprehensive review will provide crucial information for the development of more accurate and efficient druggable protein predictors.
Collapse
Affiliation(s)
- Watshara Shoombuatong
- Center for Research Innovation and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand, 10700
| | - Nalini Schaduangrat
- Center for Research Innovation and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand, 10700
| | - Jaru Nikom
- Research Methodology and Data Analytics Program, Faculty of Science & Technology, Prince of Songkla University, Pattani, Thailand, 94000
| |
Collapse
|
69
|
Charoenkwan P, Schaduangrat N, Shoombuatong W. StackTTCA: a stacking ensemble learning-based framework for accurate and high-throughput identification of tumor T cell antigens. BMC Bioinformatics 2023; 24:301. [PMID: 37507654 PMCID: PMC10386778 DOI: 10.1186/s12859-023-05421-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND The identification of tumor T cell antigens (TTCAs) is crucial for providing insights into their functional mechanisms and utilizing their potential in anticancer vaccines development. In this context, TTCAs are highly promising. Meanwhile, experimental technologies for discovering and characterizing new TTCAs are expensive and time-consuming. Although many machine learning (ML)-based models have been proposed for identifying new TTCAs, there is still a need to develop a robust model that can achieve higher rates of accuracy and precision. RESULTS In this study, we propose a new stacking ensemble learning-based framework, termed StackTTCA, for accurate and large-scale identification of TTCAs. Firstly, we constructed 156 different baseline models by using 12 different feature encoding schemes and 13 popular ML algorithms. Secondly, these baseline models were trained and employed to create a new probabilistic feature vector. Finally, the optimal probabilistic feature vector was determined based the feature selection strategy and then used for the construction of our stacked model. Comparative benchmarking experiments indicated that StackTTCA clearly outperformed several ML classifiers and the existing methods in terms of the independent test, with an accuracy of 0.932 and Matthew's correlation coefficient of 0.866. CONCLUSIONS In summary, the proposed stacking ensemble learning-based framework of StackTTCA could help to precisely and rapidly identify true TTCAs for follow-up experimental verification. In addition, we developed an online web server ( http://2pmlab.camt.cmu.ac.th/StackTTCA ) to maximize user convenience for high-throughput screening of novel TTCAs.
Collapse
Affiliation(s)
- Phasit Charoenkwan
- Modern Management and Information Technology, College of Arts, Media and Technology, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nalini Schaduangrat
- Center for Research Innovation and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand
| | - Watshara Shoombuatong
- Center for Research Innovation and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand.
| |
Collapse
|
70
|
Xu J, Li F, Li C, Guo X, Landersdorfer C, Shen HH, Peleg AY, Li J, Imoto S, Yao J, Akutsu T, Song J. iAMPCN: a deep-learning approach for identifying antimicrobial peptides and their functional activities. Brief Bioinform 2023; 24:bbad240. [PMID: 37369638 PMCID: PMC10359087 DOI: 10.1093/bib/bbad240] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 05/30/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Antimicrobial peptides (AMPs) are short peptides that play crucial roles in diverse biological processes and have various functional activities against target organisms. Due to the abuse of chemical antibiotics and microbial pathogens' increasing resistance to antibiotics, AMPs have the potential to be alternatives to antibiotics. As such, the identification of AMPs has become a widely discussed topic. A variety of computational approaches have been developed to identify AMPs based on machine learning algorithms. However, most of them are not capable of predicting the functional activities of AMPs, and those predictors that can specify activities only focus on a few of them. In this study, we first surveyed 10 predictors that can identify AMPs and their functional activities in terms of the features they employed and the algorithms they utilized. Then, we constructed comprehensive AMP datasets and proposed a new deep learning-based framework, iAMPCN (identification of AMPs based on CNNs), to identify AMPs and their related 22 functional activities. Our experiments demonstrate that iAMPCN significantly improved the prediction performance of AMPs and their corresponding functional activities based on four types of sequence features. Benchmarking experiments on the independent test datasets showed that iAMPCN outperformed a number of state-of-the-art approaches for predicting AMPs and their functional activities. Furthermore, we analyzed the amino acid preferences of different AMP activities and evaluated the model on datasets of varying sequence redundancy thresholds. To facilitate the community-wide identification of AMPs and their corresponding functional types, we have made the source codes of iAMPCN publicly available at https://github.com/joy50706/iAMPCN/tree/master. We anticipate that iAMPCN can be explored as a valuable tool for identifying potential AMPs with specific functional activities for further experimental validation.
Collapse
Affiliation(s)
- Jing Xu
- Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
- Monash Data Futures Institute, Monash University, Melbourne, VIC 3800, Australia
| | - Fuyi Li
- Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
- College of Information Engineering, Northwest A&F University, Shaanxi 712100, China
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3800, Australia
| | - Chen Li
- Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
- Monash Data Futures Institute, Monash University, Melbourne, VIC 3800, Australia
| | - Xudong Guo
- College of Information Engineering, Northwest A&F University, Shaanxi 712100, China
| | - Cornelia Landersdorfer
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3800, Australia
| | - Hsin-Hui Shen
- Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
- Department of Materials Science and Engineering, Faculty of Engineering, Monash University, Clayton, VIC, 3800, Australia
| | - Anton Y Peleg
- Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
- Department of Infectious Diseases, Alfred Hospital, Alfred Health, Melbourne, Victoria, Australia
| | - Jian Li
- Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Melbourne, VIC 3800, Australia
| | - Seiya Imoto
- Division of Health Medical Intelligence, Human Genome Center, Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
- Collaborative Research Institute for Innovative Microbiology, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | | | - Tatsuya Akutsu
- Bioinformatics Center, Institute for Chemical Research, Kyoto University, Uji 611-0011, Japan
| | - Jiangning Song
- Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
- Monash Data Futures Institute, Monash University, Melbourne, VIC 3800, Australia
- Bioinformatics Center, Institute for Chemical Research, Kyoto University, Uji 611-0011, Japan
| |
Collapse
|
71
|
Deng Y, Ma S, Li J, Zheng B, Lv Z. Using the Random Forest for Identifying Key Physicochemical Properties of Amino Acids to Discriminate Anticancer and Non-Anticancer Peptides. Int J Mol Sci 2023; 24:10854. [PMID: 37446031 PMCID: PMC10341712 DOI: 10.3390/ijms241310854] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/17/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Anticancer peptides (ACPs) represent a promising new therapeutic approach in cancer treatment. They can target cancer cells without affecting healthy tissues or altering normal physiological functions. Machine learning algorithms have increasingly been utilized for predicting peptide sequences with potential ACP effects. This study analyzed four benchmark datasets based on a well-established random forest (RF) algorithm. The peptide sequences were converted into 566 physicochemical features extracted from the amino acid index (AAindex) library, which were then subjected to feature selection using four methods: light gradient-boosting machine (LGBM), analysis of variance (ANOVA), chi-squared test (Chi2), and mutual information (MI). Presenting and merging the identified features using Venn diagrams, 19 key amino acid physicochemical properties were identified that can be used to predict the likelihood of a peptide sequence functioning as an ACP. The results were quantified by performance evaluation metrics to determine the accuracy of predictions. This study aims to enhance the efficiency of designing peptide sequences for cancer treatment.
Collapse
Affiliation(s)
- Yiting Deng
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China; (Y.D.); (S.M.); (B.Z.)
| | - Shuhan Ma
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China; (Y.D.); (S.M.); (B.Z.)
| | - Jiayu Li
- College of Life Science, Sichuan University, Chengdu 610065, China;
| | - Bowen Zheng
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China; (Y.D.); (S.M.); (B.Z.)
| | - Zhibin Lv
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China; (Y.D.); (S.M.); (B.Z.)
| |
Collapse
|
72
|
Wang Z, Deng Z, Zhang W, Lou Q, Choi KS, Wei Z, Wang L, Wu J. MMSMAPlus: a multi-view multi-scale multi-attention embedding model for protein function prediction. Brief Bioinform 2023:7187109. [PMID: 37258453 DOI: 10.1093/bib/bbad201] [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: 12/29/2022] [Revised: 04/16/2023] [Accepted: 05/08/2023] [Indexed: 06/02/2023] Open
Abstract
Protein is the most important component in organisms and plays an indispensable role in life activities. In recent years, a large number of intelligent methods have been proposed to predict protein function. These methods obtain different types of protein information, including sequence, structure and interaction network. Among them, protein sequences have gained significant attention where methods are investigated to extract the information from different views of features. However, how to fully exploit the views for effective protein sequence analysis remains a challenge. In this regard, we propose a multi-view, multi-scale and multi-attention deep neural model (MMSMA) for protein function prediction. First, MMSMA extracts multi-view features from protein sequences, including one-hot encoding features, evolutionary information features, deep semantic features and overlapping property features based on physiochemistry. Second, a specific multi-scale multi-attention deep network model (MSMA) is built for each view to realize the deep feature learning and preliminary classification. In MSMA, both multi-scale local patterns and long-range dependence from protein sequences can be captured. Third, a multi-view adaptive decision mechanism is developed to make a comprehensive decision based on the classification results of all the views. To further improve the prediction performance, an extended version of MMSMA, MMSMAPlus, is proposed to integrate homology-based protein prediction under the framework of multi-view deep neural model. Experimental results show that the MMSMAPlus has promising performance and is significantly superior to the state-of-the-art methods. The source code can be found at https://github.com/wzy-2020/MMSMAPlus.
Collapse
Affiliation(s)
- Zhongyu Wang
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, China
| | - Zhaohong Deng
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, China
| | - Wei Zhang
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, China
| | - Qiongdan Lou
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, China
| | | | - Zhisheng Wei
- National Key Laboratory of Food Science and Resource Mining, Jiangnan University, Wuxi, China
| | - Lei Wang
- National Key Laboratory of Food Science and Resource Mining, Jiangnan University, Wuxi, China
| | - Jing Wu
- National Key Laboratory of Food Science and Resource Mining, Jiangnan University, Wuxi, China
| |
Collapse
|
73
|
Chen R, Li F, Guo X, Bi Y, Li C, Pan S, Coin LJM, Song J. ATTIC is an integrated approach for predicting A-to-I RNA editing sites in three species. Brief Bioinform 2023; 24:bbad170. [PMID: 37150785 PMCID: PMC10565902 DOI: 10.1093/bib/bbad170] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 05/09/2023] Open
Abstract
A-to-I editing is the most prevalent RNA editing event, which refers to the change of adenosine (A) bases to inosine (I) bases in double-stranded RNAs. Several studies have revealed that A-to-I editing can regulate cellular processes and is associated with various human diseases. Therefore, accurate identification of A-to-I editing sites is crucial for understanding RNA-level (i.e. transcriptional) modifications and their potential roles in molecular functions. To date, various computational approaches for A-to-I editing site identification have been developed; however, their performance is still unsatisfactory and needs further improvement. In this study, we developed a novel stacked-ensemble learning model, ATTIC (A-To-I ediTing predICtor), to accurately identify A-to-I editing sites across three species, including Homo sapiens, Mus musculus and Drosophila melanogaster. We first comprehensively evaluated 37 RNA sequence-derived features combined with 14 popular machine learning algorithms. Then, we selected the optimal base models to build a series of stacked ensemble models. The final ATTIC framework was developed based on the optimal models improved by the feature selection strategy for specific species. Extensive cross-validation and independent tests illustrate that ATTIC outperforms state-of-the-art tools for predicting A-to-I editing sites. We also developed a web server for ATTIC, which is publicly available at http://web.unimelb-bioinfortools.cloud.edu.au/ATTIC/. We anticipate that ATTIC can be utilized as a useful tool to accelerate the identification of A-to-I RNA editing events and help characterize their roles in post-transcriptional regulation.
Collapse
Affiliation(s)
- Ruyi Chen
- College of Information Engineering, Northwest A&F University, Shaanxi 712100, China
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, VIC 3000, Australia
| | - Fuyi Li
- College of Information Engineering, Northwest A&F University, Shaanxi 712100, China
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, VIC 3000, Australia
| | - Xudong Guo
- College of Information Engineering, Northwest A&F University, Shaanxi 712100, China
| | - Yue Bi
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, VIC 3800, Australia
| | - Chen Li
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, VIC 3800, Australia
| | - Shirui Pan
- School of Information and Communication Technology, Griffith University, QLD 4222, Australia
| | - Lachlan J M Coin
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, VIC 3000, Australia
| | - Jiangning Song
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, VIC 3800, Australia
- Monash Data Futures Institute, Monash University, VIC 3800, Australia
| |
Collapse
|
74
|
Xia WT, Qiu WR, Yu WK, Xu ZC, Zhang SH. Identifying TME signatures for cervical cancer prognosis based on GEO and TCGA databases. Heliyon 2023; 9:e15096. [PMID: 37095983 PMCID: PMC10121839 DOI: 10.1016/j.heliyon.2023.e15096] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 03/24/2023] [Accepted: 03/27/2023] [Indexed: 04/26/2023] Open
Abstract
The mortality rate from cervical cancer (CESC), a malignant tumor that affects women, has increased significantly globally in recent years. The discovery of biomarkers points to a direction for the diagnosis of cervical cancer with the advancement of bioinformatics technology. The goal of this study was to look for potential biomarkers for the diagnosis and prognosis of CESC using the GEO and TCGA databases. Because of the high dimension and small sample size of the omic data, or the use of biomarkers generated from a single omic data, the diagnosis of cervical cancer may be inaccurate and unreliable. The purpose of this study was to search the GEO and TCGA databases for potential biomarkers for the diagnosis and prognosis of CESC. We begin by downloading CESC (GSE30760) DNA methylation data from GEO, then perform differential analysis on the downloaded methylation data and screen out the differential genes. Then, using estimation algorithms, we score immune cells and stromal cells in the tumor microenvironment and perform survival analysis on the gene expression profile data and the most recent clinical data of CESC from TCGA. Then, using the 'limma' package and Venn plot in R language to perform differential analysis of genes and screen out overlapping genes, these overlapping genes were then subjected to GO and KEGG functional enrichment analysis. The differential genes screened by the GEO methylation data and the differential genes screened by the TCGA gene expression data were intersected to screen out the common differential genes. A protein-protein interaction (PPI) network of gene expression data was then created in order to discover important genes. The PPI network's key genes were crossed with previously identified common differential genes to further validate them. The Kaplan-Meier curve was then used to determine the prognostic importance of the key genes. Survival analysis has shown that CD3E and CD80 are important for the identification of cervical cancer and can be considered as potential biomarkers for cervical cancer.
Collapse
Affiliation(s)
- Wen-Tao Xia
- School of Information Engineering, Jingdezhen Ceramic University, Jingdezhen, China
| | - Wang-Ren Qiu
- School of Information Engineering, Jingdezhen Ceramic University, Jingdezhen, China
- Corresponding author.
| | - Wang-Ke Yu
- School of Information Engineering, Jingdezhen Ceramic University, Jingdezhen, China
| | - Zhao-Chun Xu
- School of Information Engineering, Jingdezhen Ceramic University, Jingdezhen, China
| | - Shou-Hua Zhang
- Department of General Surgery, Jiangxi Provincial Children's Hospital, Nanchang, China
| |
Collapse
|
75
|
Deng H, Ding M, Wang Y, Li W, Liu G, Tang Y. ACP-MLC: A two-level prediction engine for identification of anticancer peptides and multi-label classification of their functional types. Comput Biol Med 2023; 158:106844. [PMID: 37058760 DOI: 10.1016/j.compbiomed.2023.106844] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 03/09/2023] [Accepted: 03/30/2023] [Indexed: 04/07/2023]
Abstract
Anticancer peptides (ACPs), a series of short bioactive peptides, are promising candidates in fighting against cancer due to their high activity, low toxicity, and not likely cause drug resistance. The accurate identification of ACPs and classification of their functional types is of great importance for investigating their mechanisms of action and developing peptide-based anticancer therapies. Here, we provided a computational tool, called ACP-MLC, to address binary classification and multi-label classification of ACPs for a given peptide sequence. Briefly, ACP-MLC is a two-level prediction engine, in which the 1st-level model predicts whether a query sequence is an ACP or not by random forest algorithm, and the 2nd-level model predicts which tissue types the sequence might target by the binary relevance algorithm. Development and evaluation by high-quality datasets, our ACP-MLC yielded an area under the receiver operating characteristic curve (AUC) of 0.888 on the independent test set for the 1st-level prediction, and obtained 0.157 hamming loss, 0.577 subset accuracy, 0.802 F1-scoremacro, and 0.826 F1-scoremicro on the independent test set for the 2nd-level prediction. A systematic comparison demonstrated that ACP-MLC outperformed existing binary classifiers and other multi-label learning classifiers for ACP prediction. Finally, we interpreted the important features of ACP-MLC by the SHAP method. User-friendly software and the datasets are available at https://github.com/Nicole-DH/ACP-MLC. We believe that the ACP-MLC would be a powerful tool in ACP discovery.
Collapse
|
76
|
Li Y, Ma D, Chen D, Chen Y. ACP-GBDT: An improved anticancer peptide identification method with gradient boosting decision tree. Front Genet 2023; 14:1165765. [PMID: 37065496 PMCID: PMC10090421 DOI: 10.3389/fgene.2023.1165765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 03/09/2023] [Indexed: 03/31/2023] Open
Abstract
Cancer is one of the most dangerous diseases in the world, killing millions of people every year. Drugs composed of anticancer peptides have been used to treat cancer with low side effects in recent years. Therefore, identifying anticancer peptides has become a focus of research. In this study, an improved anticancer peptide predictor named ACP-GBDT, based on gradient boosting decision tree (GBDT) and sequence information, is proposed. To encode the peptide sequences included in the anticancer peptide dataset, ACP-GBDT uses a merged-feature composed of AAIndex and SVMProt-188D. A GBDT is adopted to train the prediction model in ACP-GBDT. Independent testing and ten-fold cross-validation show that ACP-GBDT can effectively distinguish anticancer peptides from non-anticancer ones. The comparison results of the benchmark dataset show that ACP-GBDT is simpler and more effective than other existing anticancer peptide prediction methods.
Collapse
Affiliation(s)
- Yanjuan Li
- College of Electrical and Information Engineering, Quzhou University, Quzhou, China
| | - Di Ma
- College of Computer, Hangzhou Dianzi University, Hangzhou, China
| | - Dong Chen
- College of Electrical and Information Engineering, Quzhou University, Quzhou, China
- *Correspondence: Dong Chen, ; Yu Chen,
| | - Yu Chen
- College of Information and Computer Engineering, Northeast Forestry University, Harbin, China
- *Correspondence: Dong Chen, ; Yu Chen,
| |
Collapse
|
77
|
Zhou W, Liu Y, Li Y, Kong S, Wang W, Ding B, Han J, Mou C, Gao X, Liu J. TriNet: A tri-fusion neural network for the prediction of anticancer and antimicrobial peptides. PATTERNS (NEW YORK, N.Y.) 2023; 4:100702. [PMID: 36960450 PMCID: PMC10028424 DOI: 10.1016/j.patter.2023.100702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 12/20/2022] [Accepted: 02/03/2023] [Indexed: 03/04/2023]
Abstract
The accurate identification of anticancer peptides (ACPs) and antimicrobial peptides (AMPs) remains a computational challenge. We propose a tri-fusion neural network termed TriNet for the accurate prediction of both ACPs and AMPs. The framework first defines three kinds of features to capture the peptide information contained in serial fingerprints, sequence evolutions, and physicochemical properties, which are then fed into three parallel modules: a convolutional neural network module enhanced by channel attention, a bidirectional long short-term memory module, and an encoder module for training and final classification. To achieve a better training effect, TriNet is trained via a training approach using iterative interactions between the samples in the training and validation datasets. TriNet is tested on multiple challenging ACP and AMP datasets and exhibits significant improvements over various state-of-the-art methods. The web server and source code of TriNet are respectively available at http://liulab.top/TriNet/server and https://github.com/wanyunzh/TriNet.
Collapse
Affiliation(s)
- Wanyun Zhou
- SDU-ANU Joint Science College, Shandong University (Weihai), Weihai 264209, China
| | - Yufei Liu
- SDU-ANU Joint Science College, Shandong University (Weihai), Weihai 264209, China
| | - Yingxin Li
- School of Mechanical, Electrical & Information Engineering, Shandong University (Weihai), Weihai 264209, China
| | - Siqi Kong
- SDU-ANU Joint Science College, Shandong University (Weihai), Weihai 264209, China
| | - Weilin Wang
- SDU-ANU Joint Science College, Shandong University (Weihai), Weihai 264209, China
| | - Boyun Ding
- SDU-ANU Joint Science College, Shandong University (Weihai), Weihai 264209, China
| | - Jiyun Han
- School of Mathematics and Statistics, Shandong University (Weihai), Weihai 264209, China
| | - Chaozhou Mou
- School of Mathematics and Statistics, Shandong University (Weihai), Weihai 264209, China
| | - Xin Gao
- Computational Bioscience Research Center (CBRC), Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955, Saudi Arabia
| | - Juntao Liu
- School of Mathematics and Statistics, Shandong University (Weihai), Weihai 264209, China
| |
Collapse
|
78
|
Charoenkwan P, Chumnanpuen P, Schaduangrat N, Oh C, Manavalan B, Shoombuatong W. PSRQSP: An effective approach for the interpretable prediction of quorum sensing peptide using propensity score representation learning. Comput Biol Med 2023; 158:106784. [PMID: 36989748 DOI: 10.1016/j.compbiomed.2023.106784] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 02/07/2023] [Accepted: 03/10/2023] [Indexed: 03/14/2023]
Abstract
Quorum sensing peptides (QSPs) are microbial signaling molecules involved in several cellular processes, such as cellular communication, virulence expression, bioluminescence, and swarming, in various bacterial species. Understanding QSPs is essential for identifying novel drug targets for controlling bacterial populations and pathogenicity. In this study, we present a novel computational approach (PSRQSP) for improving the prediction and analysis of QSPs. In PSRQSP, we develop a novel propensity score representation learning (PSR) scheme. Specifically, we utilized the PSR approach to extract and learn a comprehensive set of estimated propensities of 20 amino acids, 400 dipeptides, and 400 g-gap dipeptides from a pool of scoring card method-based models. Finally, to maximize the utility of the propensity scores, we explored a set of optimal propensity scores and combined them to construct a final meta-predictor. Our experimental results showed that combining multiview propensity scores was more beneficial for identifying QSPs than the conventional feature descriptors. Moreover, extensive benchmarking experiments based on the independent test were sufficient to demonstrate the predictive capability and effectiveness of PSRQSP by outperforming the conventional ML-based and existing methods, with an accuracy of 94.44% and AUC of 0.967. PSR-derived propensity scores were employed to determine the crucial physicochemical properties for a better understanding of the functional mechanisms of QSPs. Finally, we constructed an easy-to-use web server for the PSRQSP (http://pmlabstack.pythonanywhere.com/PSRQSP). PSRQSP is anticipated to be an efficient computational tool for accelerating the data-driven discovery of potential QSPs for drug discovery and development.
Collapse
Affiliation(s)
- Phasit Charoenkwan
- Modern Management and Information Technology, College of Arts, Media and Technology, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Pramote Chumnanpuen
- Department of Zoology, Faculty of Science, Kasetsart University, Bangkok, 10900, Thailand; Omics Center for Agriculture, Bioresources, Food, and Health, Kasetsart University (OmiKU), Bangkok, 10900, Thailand
| | - Nalini Schaduangrat
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand
| | - Changmin Oh
- Computational Biology and Bioinformatics Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, 16419, Gyeonggi-do, Republic of Korea
| | - Balachandran Manavalan
- Computational Biology and Bioinformatics Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, 16419, Gyeonggi-do, Republic of Korea.
| | - Watshara Shoombuatong
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand.
| |
Collapse
|
79
|
Charoenkwan P, Schaduangrat N, Pham NT, Manavalan B, Shoombuatong W. Pretoria: An effective computational approach for accurate and high-throughput identification of CD8+ t-cell epitopes of eukaryotic pathogens. Int J Biol Macromol 2023; 238:124228. [PMID: 36996953 DOI: 10.1016/j.ijbiomac.2023.124228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/11/2023] [Accepted: 03/25/2023] [Indexed: 03/31/2023]
Abstract
T-cells recognize antigenic epitopes present on major histocompatibility complex (MHC) molecules, triggering an adaptive immune response in the host. T-cell epitope (TCE) identification is challenging because of the extensive number of undetermined proteins found in eukaryotic pathogens, as well as MHC polymorphisms. In addition, conventional experimental approaches for TCE identification are time-consuming and expensive. Thus, computational approaches that can accurately and rapidly identify CD8+ T-cell epitopes (TCEs) of eukaryotic pathogens based solely on sequence information may facilitate the discovery of novel CD8+ TCEs in a cost-effective manner. Here, Pretoria (Predictor of CD8+ TCEs of eukaryotic pathogens) is proposed as the first stack-based approach for accurate and large-scale identification of CD8+ TCEs of eukaryotic pathogens. In particular, Pretoria enabled the extraction and exploration of crucial information embedded in CD8+ TCEs by employing a comprehensive set of 12 well-known feature descriptors extracted from multiple groups, including physicochemical properties, composition-transition-distribution, pseudo-amino acid composition, and amino acid composition. These feature descriptors were then utilized to construct a pool of 144 different machine learning (ML)-based classifiers based on 12 popular ML algorithms. Finally, the feature selection method was used to effectively determine the important ML classifiers for the construction of our stacked model. The experimental results indicated that Pretoria is an accurate and effective computational approach for CD8+ TCE prediction; it was superior to several conventional ML classifiers and the existing method in terms of the independent test, with an accuracy of 0.866, MCC of 0.732, and AUC of 0.921. Additionally, to maximize user convenience for high-throughput identification of CD8+ TCEs of eukaryotic pathogens, a user-friendly web server of Pretoria (http://pmlabstack.pythonanywhere.com/Pretoria) was developed and made freely available.
Collapse
Affiliation(s)
- Phasit Charoenkwan
- Modern Management and Information Technology, College of Arts, Media and Technology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nalini Schaduangrat
- Center for Research Innovation and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand
| | - Nhat Truong Pham
- Computational Biology and Bioinformatics Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419, Gyeonggi-do, Republic of Korea
| | - Balachandran Manavalan
- Computational Biology and Bioinformatics Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419, Gyeonggi-do, Republic of Korea.
| | - Watshara Shoombuatong
- Center for Research Innovation and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand.
| |
Collapse
|
80
|
Lerksuthirat T, On‐yam P, Chitphuk S, Stitchantrakul W, Newburg DS, Morrow AL, Hongeng S, Chiangjong W, Chutipongtanate S. ALA-A2 Is a Novel Anticancer Peptide Inspired by Alpha-Lactalbumin: A Discovery from a Computational Peptide Library, In Silico Anticancer Peptide Screening and In Vitro Experimental Validation. GLOBAL CHALLENGES (HOBOKEN, NJ) 2023; 7:2200213. [PMID: 36910465 PMCID: PMC10000267 DOI: 10.1002/gch2.202200213] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Indexed: 06/18/2023]
Abstract
Anticancer peptides (ACPs) are rising as a new strategy for cancer therapy. However, traditional laboratory screening to find and identify novel ACPs from hundreds to thousands of peptides is costly and time consuming. Here, a sequential procedure is applied to identify candidate ACPs from a computer-generated peptide library inspired by alpha-lactalbumin, a milk protein with known anticancer properties. A total of 2688 distinct peptides, 5-25 amino acids in length, are generated from alpha-lactalbumin. In silico ACP screening using the physicochemical and structural filters and three machine learning models lead to the top candidate peptides ALA-A1 and ALA-A2. In vitro screening against five human cancer cell lines supports ALA-A2 as the positive hit. ALA-A2 selectively kills A549 lung cancer cells in a dose-dependent manner, with no hemolytic side effects, and acts as a cell penetrating peptide without membranolytic effects. Sequential window acquisition of all theorical fragment ions-proteomics and functional validation reveal that ALA-A2 induces autophagy to mediate lung cancer cell death. This approach to identify ALA-A2 is time and cost-effective. Further investigations are warranted to elucidate the exact intracellular targets of ALA-A2. Moreover, these findings support the use of larger computational peptide libraries built upon multiple proteins to further advance ACP research and development.
Collapse
Affiliation(s)
- Tassanee Lerksuthirat
- Research CenterFaculty of Medicine Ramathibodi HospitalMahidol UniversityBangkok10400Thailand
| | - Pasinee On‐yam
- Pediatric Translational Research UnitDepartment of PediatricsFaculty of Medicine Ramathibodi HospitalMahidol UniversityBangkok10400Thailand
- Faculty of Medicine Ramathibodi HospitalMahidol UniversityBangkok10400Thailand
| | - Sermsiri Chitphuk
- Research CenterFaculty of Medicine Ramathibodi HospitalMahidol UniversityBangkok10400Thailand
| | - Wasana Stitchantrakul
- Research CenterFaculty of Medicine Ramathibodi HospitalMahidol UniversityBangkok10400Thailand
| | - David S. Newburg
- Division of EpidemiologyDepartment of Environmental and Public Health SciencesUniversity of Cincinnati College of MedicineCincinnatiOH45267USA
| | - Ardythe L. Morrow
- Division of EpidemiologyDepartment of Environmental and Public Health SciencesUniversity of Cincinnati College of MedicineCincinnatiOH45267USA
- Division of Infectious DiseasesDepartment of PediatricsCincinnati Children's Hospital Medical CenterUniversity of Cincinnati College of MedicineCincinnatiOH45267USA
| | - Suradej Hongeng
- Division of Hematology and OncologyDepartment of PediatricsFaculty of Medicine Ramathibodi HospitalMahidol UniversityBangkok10400Thailand
| | - Wararat Chiangjong
- Pediatric Translational Research UnitDepartment of PediatricsFaculty of Medicine Ramathibodi HospitalMahidol UniversityBangkok10400Thailand
| | - Somchai Chutipongtanate
- Pediatric Translational Research UnitDepartment of PediatricsFaculty of Medicine Ramathibodi HospitalMahidol UniversityBangkok10400Thailand
- Division of EpidemiologyDepartment of Environmental and Public Health SciencesUniversity of Cincinnati College of MedicineCincinnatiOH45267USA
| |
Collapse
|
81
|
Wang C, Zou Q, Ju Y, Shi H. Enhancer-FRL: Improved and Robust Identification of Enhancers and Their Activities Using Feature Representation Learning. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:967-975. [PMID: 36063523 DOI: 10.1109/tcbb.2022.3204365] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Enhancers are crucial for precise regulation of gene expression, while enhancer identification and strength prediction are challenging because of their free distribution and tremendous number of similar fractions in the genome. Although several bioinformatics tools have been developed, shortfalls in these models remain, and their performances need further improvement. In the present study, a two-layer predictor called Enhancer-FRL was proposed for identifying enhancers (enhancers or nonenhancers) and their activities (strong and weak). More specifically, to build an efficient model, the feature representation learning scheme was applied to generate a 50D probabilistic vector based on 10 feature encodings and five machine learning algorithms. Subsequently, the multiview probabilistic features were integrated to construct the final prediction model. Compared with the single feature-based model, Enhancer-FRL showed significant performance improvement and model robustness. Performance assessment on the independent test dataset indicated that the proposed model outperformed state-of-the-art available toolkits. The webserver Enhancer-FRL is freely accessible at http://lab.malab.cn/∼wangchao/softwares/Enhancer-FRL/, The code and datasets can be downloaded at the webserver page or at the Github https://github.com/wangchao-malab/Enhancer-FRL/.
Collapse
|
82
|
Yan K, Lv H, Wen J, Guo Y, Xu Y, Liu B. PreTP-Stack: Prediction of Therapeutic Peptides Based on the Stacked Ensemble Learing. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:1337-1344. [PMID: 35700248 DOI: 10.1109/tcbb.2022.3183018] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Therapeutic peptide prediction is critical for drug development and therapeutic therapy. Researchers have developed several computational methods to identify different therapeutic peptide types. However, most computational methods focus on identifying the specific type of therapeutic peptides and fail to accurately predict all types of therapeutic peptides. Moreover, it is still challenging to utilize different properties features to predict the therapeutic peptides. In this study, a novel stacking framework PreTP-Stack is proposed for predicting different types of therapeutic peptide. PreTP-Stack is constructed based on ten different features and four predictors (Random Forest, Linear Discriminant Analysis, XGBoost and Support Vector Machine). Then the proposed method constructs an auto-weighted multi-view learning model as a final meta-classifier to enhance the performance of the basic models. Experimental results showed that the proposed method achieved better or highly comparable performance with the state-of-the-art methods for predicting eight types of therapeutic peptides A user-friendly web-server predictor is available at http://bliulab.net/PreTP-Stack.
Collapse
|
83
|
Zhang YF, Wang YH, Gu ZF, Pan XR, Li J, Ding H, Zhang Y, Deng KJ. Bitter-RF: A random forest machine model for recognizing bitter peptides. Front Med (Lausanne) 2023; 10:1052923. [PMID: 36778738 PMCID: PMC9909039 DOI: 10.3389/fmed.2023.1052923] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 01/05/2023] [Indexed: 01/27/2023] Open
Abstract
Introduction Bitter peptides are short peptides with potential medical applications. The huge potential behind its bitter taste remains to be tapped. To better explore the value of bitter peptides in practice, we need a more effective classification method for identifying bitter peptides. Methods In this study, we developed a Random forest (RF)-based model, called Bitter-RF, using sequence information of the bitter peptide. Bitter-RF covers more comprehensive and extensive information by integrating 10 features extracted from the bitter peptides and achieves better results than the latest generation model on independent validation set. Results The proposed model can improve the accurate classification of bitter peptides (AUROC = 0.98 on independent set test) and enrich the practical application of RF method in protein classification tasks which has not been used to build a prediction model for bitter peptides. Discussion We hope the Bitter-RF could provide more conveniences to scholars for bitter peptide research.
Collapse
Affiliation(s)
- Yu-Fei Zhang
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu, China
| | - Yu-Hao Wang
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu, China
| | - Zhi-Feng Gu
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu, China
| | - Xian-Run Pan
- Innovative Institute of Chinese Medicine and Pharmacy, Academy for Interdiscipline, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jian Li
- School of Basic Medical Sciences, Chengdu University, Chengdu, China
| | - Hui Ding
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu, China
| | - Yang Zhang
- Innovative Institute of Chinese Medicine and Pharmacy, Academy for Interdiscipline, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ke-Jun Deng
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
84
|
Ghaly G, Tallima H, Dabbish E, Badr ElDin N, Abd El-Rahman MK, Ibrahim MAA, Shoeib T. Anti-Cancer Peptides: Status and Future Prospects. Molecules 2023; 28:molecules28031148. [PMID: 36770815 PMCID: PMC9920184 DOI: 10.3390/molecules28031148] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/26/2022] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
The dramatic rise in cancer incidence, alongside treatment deficiencies, has elevated cancer to the second-leading cause of death globally. The increasing morbidity and mortality of this disease can be traced back to a number of causes, including treatment-related side effects, drug resistance, inadequate curative treatment and tumor relapse. Recently, anti-cancer bioactive peptides (ACPs) have emerged as a potential therapeutic choice within the pharmaceutical arsenal due to their high penetration, specificity and fewer side effects. In this contribution, we present a general overview of the literature concerning the conformational structures, modes of action and membrane interaction mechanisms of ACPs, as well as provide recent examples of their successful employment as targeting ligands in cancer treatment. The use of ACPs as a diagnostic tool is summarized, and their advantages in these applications are highlighted. This review expounds on the main approaches for peptide synthesis along with their reconstruction and modification needed to enhance their therapeutic effect. Computational approaches that could predict therapeutic efficacy and suggest ACP candidates for experimental studies are discussed. Future research prospects in this rapidly expanding area are also offered.
Collapse
Affiliation(s)
- Gehane Ghaly
- Department of Chemistry, The American University in Cairo, New Cairo 11835, Egypt
| | - Hatem Tallima
- Department of Chemistry, The American University in Cairo, New Cairo 11835, Egypt
| | - Eslam Dabbish
- Department of Chemistry, The American University in Cairo, New Cairo 11835, Egypt
| | - Norhan Badr ElDin
- Analytical Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr-El Aini Street, Cairo 11562, Egypt
| | - Mohamed K. Abd El-Rahman
- Analytical Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr-El Aini Street, Cairo 11562, Egypt
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, MA 02138, USA
| | - Mahmoud A. A. Ibrahim
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia 61519, Egypt
- School of Health Sciences, University of Kwa-Zulu-Natal, Westville, Durban 4000, South Africa
| | - Tamer Shoeib
- Department of Chemistry, The American University in Cairo, New Cairo 11835, Egypt
- Correspondence:
| |
Collapse
|
85
|
Wang H, Sun J, Liu M, Zheng CH, Xia J, Cheng N. frDSM: An Ensemble Predictor With Effective Feature Representation for Deleterious Synonymous Mutation in Human Genome. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:371-377. [PMID: 35420988 DOI: 10.1109/tcbb.2022.3167468] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
With the discovery of causality between synonymous mutations and diseases, it has become increasingly important to identify deleterious synonymous mutations for better understanding of their functional mechanisms. Although several machine learning methods have been proposed to solve the task, an effective feature representation method that can make use of the inner difference and relevance between deleterious and benign synonymous mutations is still challenging considering the vast number of synonymous mutations in human genome. In this work, we developed a robust and accurate predictor called frDSM for deleterious synonymous mutation prediction using logistic regression. More specifically, we introduced an effective feature representation learning method which exploits multiple feature descriptors from different perspectives including functional scores obtained from previously computational methods, evolutionary conservation, splicing and sequence feature descriptors, and these features descriptors were input into the 76 XGBoost classifiers to obtain the predictive probabilities values. These probabilities were concatenated to generate the 76-dimension new feature vector, and feature selection method was used to remove redundant and irrelevant features. Experimental results show that frDSM enables robust and accurate prediction than the competing prediction methods with 31 optimal features, which demonstrated the effectiveness of the feature representation learning method. frDSM is freely available at http://frdsm.xialab.info.
Collapse
|
86
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [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
|
87
|
Wang N, Zhang J, Liu B. iDRBP-EL: Identifying DNA- and RNA- Binding Proteins Based on Hierarchical Ensemble Learning. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:432-441. [PMID: 34932484 DOI: 10.1109/tcbb.2021.3136905] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Identification of DNA-binding proteins (DBPs) and RNA-binding proteins (RBPs) from the primary sequences is essential for further exploring protein-nucleic acid interactions. Previous studies have shown that machine-learning-based methods can efficiently identify DBPs or RBPs. However, the information used in these methods is slightly unitary, and most of them only can predict DBPs or RBPs. In this study, we proposed a computational predictor iDRBP-EL to identify DNA- and RNA- binding proteins, and introduced hierarchical ensemble learning to integrate three level information. The method can integrate the information of different features, machine learning algorithms and data into one multi-label model. The ablation experiment showed that the fusion of different information can improve the prediction performance and overcome the cross-prediction problem. Experimental results on the independent datasets showed that iDRBP-EL outperformed all the other competing methods. Moreover, we established a user-friendly webserver iDRBP-EL (http://bliulab.net/iDRBP-EL), which can predict both DBPs and RBPs only based on protein sequences.
Collapse
|
88
|
Liang Y, Ma X. iACP-GE: accurate identification of anticancer peptides by using gradient boosting decision tree and extra tree. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2023; 34:1-19. [PMID: 36562289 DOI: 10.1080/1062936x.2022.2160011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 12/12/2022] [Indexed: 06/17/2023]
Abstract
Cancer is one of the main diseases threatening human life, accounting for millions of deaths around the world each year. Traditional physical and chemical methods for cancer treatment are extremely time-consuming, lab-intensive, expensive, inefficient and difficult to be applied in a high-throughput way. Hence, it is an urgent task to develop automated computational methods to enable fast and accurate identification of anticancer peptides (ACPs). In this paper, we develop a novel model named iACP-GE to identify ACPs. Multi-features are extracted by using binary encoding, enhanced grouped amino acid composition and BLOSUM62 encoding based on the N5C5 sequence, as well as detrended forward moving-average auto-cross correlation analysis based on physicochemical properties of 20 natural amino acids. Thus, 835 features are obtained for each sample, in order to avoid information redundancy, gradient boosting decision tree was adopted as the feature selection strategy. Then, the optimal feature subset is input to the extra tree classifier. The accuracies of ACP740 and ACP240 datasets with the 5-fold cross-validation were 90.54% and 91.25%, respectively. Experimental results indicate that iACP-GE significantly outperforms several existing models on ACP740 and ACP240 datasets and can be used as an effective tool for the identification of ACPs. The datasets and source codes for iACP-GE are available at https://github.com/yunyunliang88/iACP-GE.
Collapse
Affiliation(s)
- Y Liang
- School of Science, Xi'an Polytechnic University, Xi'an, P. R. China
| | - X Ma
- School of Science, Xi'an Polytechnic University, Xi'an, P. R. China
| |
Collapse
|
89
|
ACPred-BMF: bidirectional LSTM with multiple feature representations for explainable anticancer peptide prediction. Sci Rep 2022; 12:21915. [PMID: 36535969 PMCID: PMC9763336 DOI: 10.1038/s41598-022-24404-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 11/15/2022] [Indexed: 12/24/2022] Open
Abstract
Cancer has become a major factor threatening human life and health. Under the circumstance that traditional treatment methods such as chemotherapy and radiotherapy are not highly specific and often cause severe side effects and toxicity, new treatment methods are urgently needed. Anticancer peptide drugs have low toxicity, stronger efficacy and specificity, and have emerged as a new type of cancer treatment drugs. However, experimental identification of anticancer peptides is time-consuming and expensive, and difficult to perform in a high-throughput manner. Computational identification of anticancer peptides can make up for the shortcomings of experimental identification. In this study, a deep learning-based predictor named ACPred-BMF is proposed for the prediction of anticancer peptides. This method uses the quantitative and qualitative properties of amino acids, binary profile feature to numerical representation for the peptide sequences. The Bidirectional LSTM network architecture is used in the model, and the attention mechanism is also considered. To alleviate the black-box problem of deep learning model prediction, we visualized the automatically extracted features and used the Shapley additive explanations algorithm to determine the importance of features to further understand the anticancer peptide mechanism. The results show that our method is one of the state-of-the-art anticancer peptide predictors. A web server as the implementation of ACPred-BMF that can be accessed via: http://mialab.ruc.edu.cn/ACPredBMFServer/ .
Collapse
|
90
|
Ao C, Jiao S, Wang Y, Yu L, Zou Q. Biological Sequence Classification: A Review on Data and General Methods. RESEARCH (WASHINGTON, D.C.) 2022; 2022:0011. [PMID: 39285948 PMCID: PMC11404319 DOI: 10.34133/research.0011] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 10/25/2022] [Indexed: 09/19/2024]
Abstract
With the rapid development of biotechnology, the number of biological sequences has grown exponentially. The continuous expansion of biological sequence data promotes the application of machine learning in biological sequences to construct predictive models for mining biological sequence information. There are many branches of biological sequence classification research. In this review, we mainly focus on the function and modification classification of biological sequences based on machine learning. Sequence-based prediction and analysis are the basic tasks to understand the biological functions of DNA, RNA, proteins, and peptides. However, there are hundreds of classification models developed for biological sequences, and the quite varied specific methods seem dizzying at first glance. Here, we aim to establish a long-term support website (http://lab.malab.cn/~acy/BioseqData/home.html), which provides readers with detailed information on the classification method and download links to relevant datasets. We briefly introduce the steps to build an effective model framework for biological sequence data. In addition, a brief introduction to single-cell sequencing data analysis methods and applications in biology is also included. Finally, we discuss the current challenges and future perspectives of biological sequence classification research.
Collapse
Affiliation(s)
- Chunyan Ao
- School of Computer Science and Technology, Xidian University, Xi'an, China
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou, China
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, China
| | - Shihu Jiao
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou, China
| | - Yansu Wang
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, China
| | - Liang Yu
- School of Computer Science and Technology, Xidian University, Xi'an, China
| | - Quan Zou
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou, China
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
91
|
Accurate Prediction of Anti-hypertensive Peptides Based on Convolutional Neural Network and Gated Recurrent unit. Interdiscip Sci 2022; 14:879-894. [PMID: 35474167 DOI: 10.1007/s12539-022-00521-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 03/30/2022] [Accepted: 04/06/2022] [Indexed: 12/30/2022]
Abstract
Hypertension (HT) is a general disease, and also one of the most ordinary and major causes of cardiovascular disease. Some diseases are caused by high blood pressure, including impairment of heart and kidney function, cerebral hemorrhage and myocardial infarction. Due to the limitations of laboratory methods, bioactive peptides for the treatment of HT need a long time to be identified. Therefore, it is of great immediate significance for the identification of anti-hypertensive peptides (AHTPs). With the prevalence of machine learning, it is suggested to use it as a supplementary method for AHTPs classification. Therefore, we develop a new model to identify AHTPs based on multiple features and deep learning. And the deep model is constructed by combining a convolutional neural network (CNN) and a gated recurrent unit (GRU). The unique convolution structure is used to reduce the feature dimension and running time. The data processed by CNN is input into the recurrent structure GRU, and important information is filtered out through the reset gate and update gate. Finally, the output layer adopts Sigmoid activation function. Firstly, we use Kmer, the deviation between the dipeptide frequency and the expected mean (DDE), encoding based on grouped weight (EBGW), enhanced grouped amino acid composition (EGAAC) and dipeptide binary profile and frequency (DBPF) to extract features. For Kmer, DDE, EBGW and EGAAC, it is widely used in the field of protein research. DBPF is a new feature representation method designed by us. It corresponds dipeptides to binary numbers, and finally obtains a binary coding file and a frequency file. Then these features are spliced together and input into our proposed model for prediction and analysis. After a tenfold cross-validation test, this model has a better competitive advantage than the previous methods, and the accuracy is 96.23% and 99.10%, respectively. From the results, compared with the previous methods, it has been greatly improved. It shows that the combination of convolution calculation and recurrent structure has a positive impact on the classification of AHTPs. The results show that this method is a feasible, efficient and competitive sequence analysis tool for AHTPs. Meanwhile, we design a friendly online prediction tool and it is freely accessible at http://ahtps.zhanglab.site/ .
Collapse
|
92
|
Gu X, Ding Y, Xiao P, He T. A GHKNN model based on the physicochemical property extraction method to identify SNARE proteins. Front Genet 2022; 13:935717. [PMID: 36506312 PMCID: PMC9727185 DOI: 10.3389/fgene.2022.935717] [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: 08/16/2022] [Accepted: 11/02/2022] [Indexed: 11/24/2022] Open
Abstract
There is a great deal of importance to SNARE proteins, and their absence from function can lead to a variety of diseases. The SNARE protein is known as a membrane fusion protein, and it is crucial for mediating vesicle fusion. The identification of SNARE proteins must therefore be conducted with an accurate method. Through extensive experiments, we have developed a model based on graph-regularized k-local hyperplane distance nearest neighbor model (GHKNN) binary classification. In this, the model uses the physicochemical property extraction method to extract protein sequence features and the SMOTE method to upsample protein sequence features. The combination achieves the most accurate performance for identifying all protein sequences. Finally, we compare the model based on GHKNN binary classification with other classifiers and measure them using four different metrics: SN, SP, ACC, and MCC. In experiments, the model performs significantly better than other classifiers.
Collapse
Affiliation(s)
- Xingyue Gu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Yijie Ding
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou, Zhejiang, China
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, China
| | - Pengfeng Xiao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Tao He
- Beidahuang Industry Group General Hospital, Harbin, China
| |
Collapse
|
93
|
Yan J, Cai J, Zhang B, Wang Y, Wong DF, Siu SWI. Recent Progress in the Discovery and Design of Antimicrobial Peptides Using Traditional Machine Learning and Deep Learning. Antibiotics (Basel) 2022; 11:1451. [PMID: 36290108 PMCID: PMC9598685 DOI: 10.3390/antibiotics11101451] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/11/2022] [Accepted: 10/13/2022] [Indexed: 11/16/2022] Open
Abstract
Antimicrobial resistance has become a critical global health problem due to the abuse of conventional antibiotics and the rise of multi-drug-resistant microbes. Antimicrobial peptides (AMPs) are a group of natural peptides that show promise as next-generation antibiotics due to their low toxicity to the host, broad spectrum of biological activity, including antibacterial, antifungal, antiviral, and anti-parasitic activities, and great therapeutic potential, such as anticancer, anti-inflammatory, etc. Most importantly, AMPs kill bacteria by damaging cell membranes using multiple mechanisms of action rather than targeting a single molecule or pathway, making it difficult for bacterial drug resistance to develop. However, experimental approaches used to discover and design new AMPs are very expensive and time-consuming. In recent years, there has been considerable interest in using in silico methods, including traditional machine learning (ML) and deep learning (DL) approaches, to drug discovery. While there are a few papers summarizing computational AMP prediction methods, none of them focused on DL methods. In this review, we aim to survey the latest AMP prediction methods achieved by DL approaches. First, the biology background of AMP is introduced, then various feature encoding methods used to represent the features of peptide sequences are presented. We explain the most popular DL techniques and highlight the recent works based on them to classify AMPs and design novel peptide sequences. Finally, we discuss the limitations and challenges of AMP prediction.
Collapse
Affiliation(s)
- Jielu Yan
- PAMI Research Group, Department of Computer and Information Science, University of Macau, Taipa, Macau, China
| | - Jianxiu Cai
- Faculty of Applied Sciences, Macao Polytechnic University, Macau, China
- Institute of Science and Environment, University of Saint Joseph, Estr. Marginal da Ilha Verde, Macau, China
| | - Bob Zhang
- PAMI Research Group, Department of Computer and Information Science, University of Macau, Taipa, Macau, China
| | - Yapeng Wang
- Faculty of Applied Sciences, Macao Polytechnic University, Macau, China
| | - Derek F. Wong
- NLP2CT Lab, Department of Computer and Information Science, University of Macau, Taipa, Macau, China
| | - Shirley W. I. Siu
- Institute of Science and Environment, University of Saint Joseph, Estr. Marginal da Ilha Verde, Macau, China
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Pulau Pinang 11800, Malaysia
| |
Collapse
|
94
|
ACP-ADA: A Boosting Method with Data Augmentation for Improved Prediction of Anticancer Peptides. Int J Mol Sci 2022; 23:ijms232012194. [PMID: 36293050 PMCID: PMC9603247 DOI: 10.3390/ijms232012194] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/08/2022] [Accepted: 10/11/2022] [Indexed: 11/30/2022] Open
Abstract
Cancer is the second-leading cause of death worldwide, and therapeutic peptides that target and destroy cancer cells have received a great deal of interest in recent years. Traditional wet experiments are expensive and inefficient for identifying novel anticancer peptides; therefore, the development of an effective computational approach is essential to recognize ACP candidates before experimental methods are used. In this study, we proposed an Ada-boosting algorithm with the base learner random forest called ACP-ADA, which integrates binary profile feature, amino acid index, and amino acid composition with a 210-dimensional feature space vector to represent the peptides. Training samples in the feature space were augmented to increase the sample size and further improve the performance of the model in the case of insufficient samples. Furthermore, we used five-fold cross-validation to find model parameters, and the cross-validation results showed that ACP-ADA outperforms existing methods for this feature combination with data augmentation in terms of performance metrics. Specifically, ACP-ADA recorded an average accuracy of 86.4% and a Mathew’s correlation coefficient of 74.01% for dataset ACP740 and 90.83% and 81.65% for dataset ACP240; consequently, it can be a very useful tool in drug development and biomedical research.
Collapse
|
95
|
Xu D, Liu B, Wang J, Zhang Z. Bibliometric analysis of artificial intelligence for biotechnology and applied microbiology: Exploring research hotspots and frontiers. Front Bioeng Biotechnol 2022; 10:998298. [PMID: 36277390 PMCID: PMC9585160 DOI: 10.3389/fbioe.2022.998298] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/23/2022] [Indexed: 11/13/2022] Open
Abstract
Background: In the biotechnology and applied microbiology sectors, artificial intelligence (AI) has been extensively used in disease diagnostics, drug research and development, functional genomics, biomarker recognition, and medical imaging diagnostics. In our study, from 2000 to 2021, science publications focusing on AI in biotechnology were reviewed, and quantitative, qualitative, and modeling analyses were performed. Methods: On 6 May 2022, the Web of Science Core Collection (WoSCC) was screened for AI applications in biotechnology and applied microbiology; 3,529 studies were identified between 2000 and 2022, and analyzed. The following information was collected: publication, country or region, references, knowledgebase, institution, keywords, journal name, and research hotspots, and examined using VOSviewer and CiteSpace V bibliometric platforms. Results: We showed that 128 countries published articles related to AI in biotechnology and applied microbiology; the United States had the most publications. In addition, 584 global institutions contributed to publications, with the Chinese Academy of Science publishing the most. Reference clusters from studies were categorized into ten headings: deep learning, prediction, support vector machines (SVM), object detection, feature representation, synthetic biology, amyloid, human microRNA precursors, systems biology, and single cell RNA-Sequencing. Research frontier keywords were represented by microRNA (2012–2020) and protein-protein interactions (PPIs) (2012–2020). Conclusion: We systematically, objectively, and comprehensively analyzed AI-related biotechnology and applied microbiology literature, and additionally, identified current hot spots and future trends in this area. Our review provides researchers with a comprehensive overview of the dynamic evolution of AI in biotechnology and applied microbiology and identifies future key research areas.
Collapse
Affiliation(s)
- Dongyu Xu
- Department of Computer, School of Intelligent Medicine, China Medical University, Shenyang, Liaoning, China
| | - Bing Liu
- Department of Bone Oncology, The People’s Hospital of Liaoning Province, Shenyang, Liaoning, China
| | - Jian Wang
- Department of Pathogenic Biology, School of Basic Medicine, China Medical University, Shenyang, Liaoning, China
| | - Zhichang Zhang
- Department of Computer, School of Intelligent Medicine, China Medical University, Shenyang, Liaoning, China
- *Correspondence: Zhichang Zhang,
| |
Collapse
|
96
|
Rodrigues CHM, Garg A, Keizer D, Pires DEV, Ascher DB. CSM-peptides: A computational approach to rapid identification of therapeutic peptides. Protein Sci 2022; 31:e4442. [PMID: 36173168 PMCID: PMC9518225 DOI: 10.1002/pro.4442] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/25/2022]
Abstract
Peptides are attractive alternatives for the development of new therapeutic strategies due to their versatility and low complexity of synthesis. Increasing interest in these molecules has led to the creation of large collections of experimentally characterized therapeutic peptides, which greatly contributes to development of data-driven computational approaches. Here we propose CSM-peptides, a novel machine learning method for rapid identification of eight different types of therapeutic peptides: anti-angiogenic, anti-bacterial, anti-cancer, anti-inflammatory, anti-viral, cell-penetrating, quorum sensing, and surface binding. Our method has shown to outperform existing approaches, achieving an AUC of up to 0.92 on independent blind tests, and consistent performance on cross-validation. We anticipate CSM-peptides to be of great value in helping screening large libraries to identify novel peptides with therapeutic potential and have made it freely available as a user-friendly web server and Application Programming Interface at https://biosig.lab.uq.edu.au/csm_peptides.
Collapse
Affiliation(s)
- Carlos H. M. Rodrigues
- Structural Biology and Bioinformatics, Department of BiochemistryUniversity of MelbourneMelbourneVictoriaAustralia
- Systems and Computational Biology, Bio21 Institute, University of MelbourneMelbourneVictoriaAustralia
- Computational Biology and Clinical InformaticsBaker Heart and Diabetes InstituteMelbourneVictoriaAustralia
- School of Chemistry and Molecular BiosciencesUniversity of QueenslandSt LuciaQueenslandAustralia
| | - Anjali Garg
- Structural Biology and Bioinformatics, Department of BiochemistryUniversity of MelbourneMelbourneVictoriaAustralia
- Systems and Computational Biology, Bio21 Institute, University of MelbourneMelbourneVictoriaAustralia
| | - David Keizer
- Structural Biology and Bioinformatics, Department of BiochemistryUniversity of MelbourneMelbourneVictoriaAustralia
- Systems and Computational Biology, Bio21 Institute, University of MelbourneMelbourneVictoriaAustralia
| | - Douglas E. V. Pires
- Systems and Computational Biology, Bio21 Institute, University of MelbourneMelbourneVictoriaAustralia
- Computational Biology and Clinical InformaticsBaker Heart and Diabetes InstituteMelbourneVictoriaAustralia
- School of Computing and Information SystemsUniversity of MelbourneMelbourneVictoriaAustralia
| | - David B. Ascher
- Structural Biology and Bioinformatics, Department of BiochemistryUniversity of MelbourneMelbourneVictoriaAustralia
- Systems and Computational Biology, Bio21 Institute, University of MelbourneMelbourneVictoriaAustralia
- Computational Biology and Clinical InformaticsBaker Heart and Diabetes InstituteMelbourneVictoriaAustralia
- School of Chemistry and Molecular BiosciencesUniversity of QueenslandSt LuciaQueenslandAustralia
| |
Collapse
|
97
|
Bahadory S, Sadraei J, Zibaei M, Pirestani M, Dalimi A. In vitro anti-gastrointestinal cancer activity of Toxocara canis-derived peptide: Analyzing the expression level of factors related to cell proliferation and tumor growth. Front Pharmacol 2022; 13:878724. [PMID: 36204226 PMCID: PMC9530354 DOI: 10.3389/fphar.2022.878724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 08/01/2022] [Indexed: 11/23/2022] Open
Abstract
Background: Recently, a hypothesis about the negative relationship between cancers and parasites has been proposed and investigated; some parasitic worms and their products can affect the cancer cell proliferation. Due to the potential anti-cancer effect of helminthic parasites, in the present study, the excretory–secretory protein of Toxocara canis (T. canis) parasite was used to evaluate the possible anti-cancer properties and their effect on gastrointestinal and liver cancer cell proliferation-related genes in laboratory conditions. Methods and materials: The selected synthesized peptide fraction from the T. canis excretory–secretory Troponin protein peptide (ES TPP) was exposed at 32, 64, 128, and 256 μg/ml concentrations to three gastrointestinal cancer cell lines AGS, HT-29, and Caco 2, as well as HDF cells as normal cell lines. We used the MTT assay to evaluate cellular changes and cell viability (CV). Variations in gene (Bcl-2, APAF1, ZEB1, VEGF, cyclin-D1, and caspase-3) expression were analyzed by real-time RT-PCR. Results: After 24 h of exposure to pept1ides and cell lines, a decrease in CV was observed at a concentration of 64 μg/ml and compared to the control group. Then, after 48 h, a significant decrease in the CV of Caco 2 cells was observed at a concentration of 32 μg/ml; in the other cancer cell lines, concentrations above 32 μg/ml were effective. The peptide was able to significantly alter the expression of the studied genes at a concentration of 100 μg/ml. Conclusion: Although the studied peptide at high concentrations could have a statistically significant effect on cancer cells, it is still far from the standard drug and can be optimized and promising in future studies.
Collapse
Affiliation(s)
- Saeed Bahadory
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Javid Sadraei
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- *Correspondence: Javid Sadraei,
| | - Mohammad Zibaei
- Department of Parasitology and Mycology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
- Evidence-Based Phytotherapy and Complementary Medicine Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Majid Pirestani
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Abdolhossein Dalimi
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
98
|
Liu J, Li M, Chen X. AntiMF: A deep learning framework for predicting anticancer peptides based on multi-view feature extraction. Methods 2022; 207:38-43. [PMID: 36100141 DOI: 10.1016/j.ymeth.2022.07.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/20/2022] [Accepted: 07/26/2022] [Indexed: 01/10/2023] Open
Abstract
In recent years, anticancer peptides have emerged as a new viable option in cancer therapy, with the ability to overcome the considerable side effects and poor outcomes of standard cancer therapies. Accurate anticancer peptide identification can facilitate its finding and speed up its application in treating cancer. However, many recent approaches are based on machine learning, which not only restricts the representation ability of the models but also requires a complex hand-crafted feature extraction process. Here, we propose AntiMF, a deep learning model that utilizes multi-view mechanism based on different feature extraction models. Comparative results show that our model has a better performance than the state-of-the-art methods in the prediction of anticancer peptides. By using an ensemble learning framework to extract representation, AntiMF can capture the different dimensional information, which can make model representation more complete. Moreover, we visualize what AntiMF learns on one of its ensemble models to intuitively show the effectivity of our model, indicating that AntiMF has the great potential ability to be an effective and useful model to identify anticancer peptides accurately.
Collapse
Affiliation(s)
- Jingjing Liu
- Eye Hospital, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Minghao Li
- Beidahuang Industry Group General Hospital, Harbin 150001, China
| | - Xin Chen
- Eye Hospital, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China; Department of Neurosurgical Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China.
| |
Collapse
|
99
|
Zhu L, Ye C, Hu X, Yang S, Zhu C. ACP-check: An anticancer peptide prediction model based on bidirectional long short-term memory and multi-features fusion strategy. Comput Biol Med 2022; 148:105868. [PMID: 35868046 DOI: 10.1016/j.compbiomed.2022.105868] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 06/14/2022] [Accepted: 07/09/2022] [Indexed: 11/16/2022]
Abstract
The anticancer peptide is an emerging anticancer drug that has become an effective alternative to chemotherapy and targeted therapy due to fewer side effects and resistance. The traditional biological experimental method for identifying anticancer peptides is a time-consuming and complicated process that hinders large-scale, rapid, and effective identification. In this paper, we propose a model based on a bidirectional long short-term memory network and multi-features fusion, called ACP-check, which employs a bidirectional long short-term memory network to extract time-dependent information features from peptide sequences, and combines them with amino acid sequence features including binary profile feature, dipeptide composition, the composition of k-spaced amino acid group pairs, amino acid composition, and sequence-order-coupling number. To verify the performance of the model, six benchmark datasets are selected, including ACPred-Fuse, ACPred-FL, ACP240, ACP740, main and alternate datasets of AntiCP2.0. In terms of Matthews correlation coefficients, ACP-check obtains 0.37, 0.82, 0.80, 0.75, 0.56, and 0.86 on six datasets respectively, which is an improvement by 2%-86% than existing state-of-the-art anticancer peptides prediction methods. Furthermore, ACP-check achieves prediction accuracy with 0.91, 0.91, 0.90, 0.87, 0.78, and 0.93 respectively, which increases range from 1%-49%. Overall, the comparison experiment shows that ACP-check can accurately identify anticancer peptides by sequence-level information. The code and data are available at http://www.cczubio.top/ACP-check/.
Collapse
Affiliation(s)
- Lun Zhu
- School of Computer Science and Artificial Intelligence Aliyun School of Big Data School of Software, Changzhou University, Changzhou, 213164, China
| | - Chenyang Ye
- School of Computer Science and Artificial Intelligence Aliyun School of Big Data School of Software, Changzhou University, Changzhou, 213164, China
| | - Xuemei Hu
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun, 130012, China
| | - Sen Yang
- School of Computer Science and Artificial Intelligence Aliyun School of Big Data School of Software, Changzhou University, Changzhou, 213164, China; Changzhou No.2 People's Hospital, the Affiliated Hospital of Nanjing Medical University, Changzhou, 213164, China.
| | - Chenyang Zhu
- School of Computer Science and Artificial Intelligence Aliyun School of Big Data School of Software, Changzhou University, Changzhou, 213164, China
| |
Collapse
|
100
|
Pu Y, Li J, Tang J, Guo F. DeepFusionDTA: Drug-Target Binding Affinity Prediction With Information Fusion and Hybrid Deep-Learning Ensemble Model. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:2760-2769. [PMID: 34379594 DOI: 10.1109/tcbb.2021.3103966] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Identification of drug-target interaction (DTI) is the most important issue in the broad field of drug discovery. Using purely biological experiments to verify drug-target binding profiles takes lots of time and effort, so computational technologies for this task obviously have great benefits in reducing the drug search space. Most of computational methods to predict DTI are proposed to solve a binary classification problem, which ignore the influence of binding strength. Therefore, drug-target binding affinity prediction is still a challenging issue. Currently, lots of studies only extract sequence information that lacks feature-rich representation, but we consider more spatial features in order to merge various data in drug and target spaces. In this study, we propose a two-stage deep neural network ensemble model for detecting drug-target binding affinity, called DeepFusionDTA, via various information analysis modules. First stage is to utilize sequence and structure information to generate fusion feature map of candidate protein and drug pair through various analysis modules based deep learning. Second stage is to apply bagging-based ensemble learning strategy for regression prediction, and we obtain outstanding results by combining the advantages of various algorithms in efficient feature abstraction and regression calculation. Importantly, we evaluate our novel method, DeepFusionDTA, which delivers 1.5 percent CI increase on KIBA dataset and 1.0 percent increase on Davis dataset, by comparing with existing prediction tools, DeepDTA. Furthermore, the ideas we have offered can be applied to in-silico screening of the interaction space, to provide novel DTIs which can be experimentally pursued. The codes and data are available from https://github.com/guofei-tju/DeepFusionDTA.
Collapse
|