1
|
Liu J, Geng A, Cui F, Xu J, Meng Y, Wei L, Zhang Q, Zou Q, Zhang Z. NeuroCL: A deep learning approach for identifying neuropeptides based on contrastive learning. Anal Biochem 2025; 705:115920. [PMID: 40466845 DOI: 10.1016/j.ab.2025.115920] [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: 03/16/2025] [Revised: 05/13/2025] [Accepted: 06/01/2025] [Indexed: 06/16/2025]
Abstract
Neuropeptides (NPs), a unique class of neuronal signaling molecules, involved in neurotransmission, endocrine regulation, immune response, mood, and appetite control. The identification of neuropeptides provides critical scientific insights for early diagnosis, targeted therapy, and personalized medicine of related diseases. Previous models struggle to capture complex relationships among features and inter-sample connections. In this study, we introduce NeuroCL, a deep learning model harnessing contrastive learning and a cross-attention mechanism to efficiently identify NPs through multifaceted attribute representation. Experimental outcomes demonstrate that NeuroCL effectively captures data nuances, achieving an impressive accuracy of 93.8 % and a Matthews correlation coefficient (MCC) of 87.8 % on an independent test set. Contrastive learning enhances class distinction and coherence, while cross-attention mechanisms integrate pre-trained large models with manually encoded features, synergistically boosting their capabilities and strengthening feature connections. Our model surpasses current state-of-the-art predictors in NPs identification. Visualization via uniform manifold approximation and projection (UMAP) reveals that NeuroCL distinctly segregates positive NPs from negative ones. To facilitate the accessibility and application of our model, we have established a web-based platform available at http://www.bioai-lab.com/NeuroCL.
Collapse
Affiliation(s)
- Jian Liu
- School of Computer Science and Technology, Hainan University, Haikou, 570228, China
| | - Aoyun Geng
- School of Computer Science and Technology, Hainan University, Haikou, 570228, China
| | - Feifei Cui
- School of Computer Science and Technology, Hainan University, Haikou, 570228, China
| | - Junlin Xu
- School of Computer Science and Technology, Wuhan University of Science and Technology, Wuhan, 430081, Hubei, China
| | - Yajie Meng
- School of Computer Science and Artificial Intelligence, Wuhan Textile University, Wuhan, 430200, Hubei, China
| | - Leyi Wei
- Centre for Artificial Intelligence Driven Drug Discovery, Faculty of Applied Science, Macao Polytechnic University, Macao Special Administrative Region of China; School of Informatics, Xiamen University, Xiamen, China
| | - Qingchen Zhang
- School of Computer Science and Technology, Hainan University, Haikou, 570228, China
| | - Quan Zou
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Zilong Zhang
- School of Computer Science and Technology, Hainan University, Haikou, 570228, China.
| |
Collapse
|
2
|
Zhang H, Liu S, Su W, Xie X, Yu J, Dao F, Yang M, Lyu H, Lin H. NeuroScale: evolutional scale-based protein language models enable prediction of neuropeptides. BMC Biol 2025; 23:142. [PMID: 40437538 PMCID: PMC12121104 DOI: 10.1186/s12915-025-02243-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Accepted: 05/12/2025] [Indexed: 06/01/2025] Open
Abstract
BACKGROUND Neuropeptides (NPs) are critical signaling molecules involved in various physiological and behavioral processes, including development, metabolism, and memory. They function within both the nervous and endocrine systems and have emerged as promising therapeutic targets for a range of diseases. Despite their significance, the accurate identification of NPs remains a challenge, necessitating the development of more effective computational approaches. RESULTS In this study, we introduce NeuroScale, a multi-channel neural network model leveraging evolutionary scale modeling (ESM) for the precise prediction of NPs. By integrating the GoogLeNet framework, NeuroScale effectively captures multi-scale NP features, enabling robust and accurate classification. Extensive benchmarking demonstrates its superior performance, consistently achieving an area under the receiver operating characteristic curve (AUC) exceeding 0.97. Additionally, we systematically analyzed the impact of protein sequence similarity thresholds and multi-scale sequence lengths on model performance, further validating NeuroScale's robustness and generalizability. CONCLUSIONS NeuroScale represents a significant advancement in neuropeptide prediction, offering both high accuracy and adaptability to diverse sequence characteristics. Its ability to generalize across different sequence similarity thresholds and lengths underscores its potential as a reliable tool for neuropeptide discovery and peptide-based drug development. By providing a scalable and efficient deep learning framework, NeuroScale paves the way for future research in neuropeptide function, disease mechanisms, and therapeutic applications.
Collapse
Affiliation(s)
- Hongqi Zhang
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, People's Republic of China
| | - Shanghua Liu
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, People's Republic of China
| | - Wei Su
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, People's Republic of China
| | - Xueqin Xie
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, People's Republic of China
| | - Junwen Yu
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, People's Republic of China
| | - Fuying Dao
- School of Biological Sciences, Nanyang Technological University, Singapore, 639798, Singapore
| | - Mi Yang
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, People's Republic of China.
| | - Hao Lyu
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, People's Republic of China.
| | - Hao Lin
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, People's Republic of China.
| |
Collapse
|
3
|
Li J, Xiong S, Shi H, Cui F, Zhang Z, Wei L. NeuroPred-AIMP: Multimodal Deep Learning for Neuropeptide Prediction via Protein Language Modeling and Temporal Convolutional Networks. J Chem Inf Model 2025; 65:4740-4750. [PMID: 40258183 DOI: 10.1021/acs.jcim.5c00444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2025]
Abstract
Neuropeptides are key signaling molecules that regulate fundamental physiological processes ranging from metabolism to cognitive function. However, accurate identification is a huge challenge due to sequence heterogeneity, obscured functional motifs and limited experimentally validated data. Accurate identification of neuropeptides is critical for advancing neurological disease therapeutics and peptide-based drug design. Existing neuropeptide identification methods rely on manual features combined with traditional machine learning methods, which are difficult to capture the deep patterns of sequences. To address these limitations, we propose NeuroPred-AIMP (adaptive integrated multimodal predictor), an interpretable model that synergizes global semantic representation of the protein language model (ESM) and the multiscale structural features of the temporal convolutional network (TCN). The model introduced the adaptive features fusion mechanism of residual enhancement to dynamically recalibrate feature contributions, to achieve robust integration of evolutionary and local sequence information. The experimental results demonstrated that the proposed model showed excellent comprehensive performance on the independence test set, with an accuracy of 92.3% and the AUROC of 0.974. Simultaneously, the model showed good balance in the ability to identify positive and negative samples, with a sensitivity of 92.6% and a specificity of 92.1%, with a difference of less than 0.5%. The result fully confirms the effectiveness of the multimodal features strategy in the task of neuropeptide recognition.
Collapse
Affiliation(s)
- Jinjin Li
- Faculty of Applied Sciences, Macao Polytechnic University, R. de Luís Gonzaga Gomes, Macao 999078, China
| | - Shuwen Xiong
- Faculty of Applied Sciences, Macao Polytechnic University, R. de Luís Gonzaga Gomes, Macao 999078, China
| | - Hua Shi
- School of Optoelectronic and Communication Engineering, Xiamen University of Technology, Xiamen 361024, China
| | - Feifei Cui
- School of Computer Science and Technology, Hainan University, Haikou 570228, China
| | - Zilong Zhang
- School of Computer Science and Technology, Hainan University, Haikou 570228, China
| | - Leyi Wei
- Faculty of Applied Sciences, Macao Polytechnic University, R. de Luís Gonzaga Gomes, Macao 999078, China
- School of Software, Shandong University, Jinan 250101, China
| |
Collapse
|
4
|
Akbar S, Raza A, Awan HH, Zou Q, Alghamdi W, Saeed A. pNPs-CapsNet: Predicting Neuropeptides Using Protein Language Models and FastText Encoding-Based Weighted Multi-View Feature Integration with Deep Capsule Neural Network. ACS OMEGA 2025; 10:12403-12416. [PMID: 40191328 PMCID: PMC11966582 DOI: 10.1021/acsomega.4c11449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/04/2025] [Accepted: 03/07/2025] [Indexed: 04/09/2025]
Abstract
Neuropeptides (NPs) are critical signaling molecules that are essential in numerous physiological processes and possess significant therapeutic potential. Computational prediction of NPs has emerged as a promising alternative to traditional experimental methods, often labor-intensive, time-consuming, and expensive. Recent advancements in computational peptide models provide a cost-effective approach to identifying NPs, characterized by high selectivity toward target cells and minimal side effects. In this study, we propose a novel deep capsule neural network-based computational model, namely pNPs-CapsNet, to predict NPs and non-NPs accurately. Input samples are numerically encoded using pretrained protein language models, including ESM, ProtBERT-BFD, and ProtT5, to extract attention mechanism-based contextual and semantic features. A differential evolution-based weighted feature integration method is utilized to construct a multiview vector. Additionally, a two-tier feature selection strategy, comprising MRMD and SHAP analysis, is developed to identify and select optimal features. Finally, the novel capsule neural network (CapsNet) is trained using the selected optimal feature set. The proposed pNPs-CapsNet model achieved a remarkable predictive accuracy of 98.10% and an AUC of 0.98. To validate the generalization capability of the pNPs-CapsNet model, independent samples reported an accuracy of 95.21% and an AUC of 0.96. The pNPs-CapsNet model outperforms existing state-of-the-art models, demonstrating 4% and 2.5% improved predictive accuracy for training and independent data sets, respectively. The demonstrated efficacy and consistency of pNPs-CapsNet underline its potential as a valuable and robust tool for advancing drug discovery and academic research.
Collapse
Affiliation(s)
- Shahid Akbar
- Institute
of Fundamental and Frontier Sciences, University
of Electronic Science and Technology of China, Chengdu 610054, China
- Department
of Computer Science, Abdul Wali Khan University
Mardan, Mardan 23200, Khyber Pakhtunkhwa, Pakistan
| | - Ali Raza
- Department
of Computer Science, Bahria University, Islamabad 44220, Pakistan
| | - Hamid Hussain Awan
- Department
of Computer Science, Rawalpindi Women University, Rawalpindi 46300, Punjab, Pakistan
| | - 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 324000, PR China
| | - Wajdi Alghamdi
- Department
of Information Technology, Faculty of Computing and Information Technology, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Aamir Saeed
- Department
of Computer Science and IT, University of
Engineering and Technology, Jalozai Campus, Peshawar 25000, Pakistan
| |
Collapse
|
5
|
Asim MN, Asif T, Mehmood F, Dengel A. Peptide classification landscape: An in-depth systematic literature review on peptide types, databases, datasets, predictors architectures and performance. Comput Biol Med 2025; 188:109821. [PMID: 39987697 DOI: 10.1016/j.compbiomed.2025.109821] [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: 09/28/2024] [Revised: 02/03/2025] [Accepted: 02/05/2025] [Indexed: 02/25/2025]
Abstract
Peptides are gaining significant attention in diverse fields such as the pharmaceutical market has seen a steady rise in peptide-based therapeutics over the past six decades. Peptides have been utilized in the development of distinct applications including inhibitors of SARS-COV-2 and treatments for conditions like cancer and diabetes. Distinct types of peptides possess unique characteristics, and development of peptide-specific applications require the discrimination of one peptide type from others. To the best of our knowledge, approximately 230 Artificial Intelligence (AI) driven applications have been developed for 22 distinct types of peptides, yet there remains significant room for development of new predictors. A Comprehensive review addresses the critical gap by providing a consolidated platform for the development of AI-driven peptide classification applications. This paper offers several key contributions, including presenting the biological foundations of 22 unique peptide types and categorizes them into four main classes: Regulatory, Therapeutic, Nutritional, and Delivery Peptides. It offers an in-depth overview of 47 databases that have been used to develop peptide classification benchmark datasets. It summarizes details of 288 benchmark datasets that are used in development of diverse types AI-driven peptide classification applications. It provides a detailed summary of 197 sequence representation learning methods and 94 classifiers that have been used to develop 230 distinct AI-driven peptide classification applications. Across 22 distinct types peptide classification tasks related to 288 benchmark datasets, it demonstrates performance values of 230 AI-driven peptide classification applications. It summarizes experimental settings and various evaluation measures that have been employed to assess the performance of AI-driven peptide classification applications. The primary focus of this manuscript is to consolidate scattered information into a single comprehensive platform. This resource will greatly assist researchers who are interested in developing new AI-driven peptide classification applications.
Collapse
Affiliation(s)
- Muhammad Nabeel Asim
- German Research Center for Artificial Intelligence, Kaiserslautern, 67663, Germany; Intelligentx GmbH (intelligentx.com), Kaiserslautern, Germany.
| | - Tayyaba Asif
- Department of Computer Science, Rhineland-Palatinate Technical University of Kaiserslautern-Landau, Kaiserslautern, 67663, Germany
| | - Faiza Mehmood
- Department of Computer Science, Rhineland-Palatinate Technical University of Kaiserslautern-Landau, Kaiserslautern, 67663, Germany; Institute of Data Sciences, University of Engineering and Technology, Lahore, Pakistan
| | - Andreas Dengel
- German Research Center for Artificial Intelligence, Kaiserslautern, 67663, Germany; Department of Computer Science, Rhineland-Palatinate Technical University of Kaiserslautern-Landau, Kaiserslautern, 67663, Germany; Intelligentx GmbH (intelligentx.com), Kaiserslautern, Germany
| |
Collapse
|
6
|
Rahmani R, Kalankesh LR, Ferdousi R. Computational approaches for identifying neuropeptides: A comprehensive review. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102409. [PMID: 40171446 PMCID: PMC11960512 DOI: 10.1016/j.omtn.2024.102409] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
Neuropeptides (NPs) are key signaling molecules that interact with G protein-coupled receptors, influencing neuronal activities and developmental pathways, as well as the endocrine and immune systems. They are significant in disease contexts, offering potential therapeutic targets for conditions such as anxiety, neurological disorders, cardiovascular health, and diabetes. Understanding and detecting NPs is crucial because of their complex functions in health and disease. Historically, identifying NPs via wet lab techniques has been time-consuming and costly. However, integrating computational methods has shown the potential to improve efficiency, accuracy, and cost-effectiveness. Computational techniques, such as artificial intelligence and machine learning, have been extensively researched in recent years for the identification of NP. This review explores the application of machine learning (ML) techniques in predicting various aspects of NPs, including their sequences, cleavage sites, and precursors. Additionally, it provides insights into databases containing NP metadata and specialized tools used in this domain.
Collapse
Affiliation(s)
- Roya Rahmani
- Student Research Committee, Tabriz University of Medical Science, Tabriz, Iran
- Department of Health Information Technology, School of Management and Medical Informatics, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila R. Kalankesh
- Department of Health Information Technology, School of Management and Medical Informatics, Tabriz University of Medical Sciences, Tabriz, Iran
- Tabriz University of Medical Sciences, Research Center of Psychiatry and Behavioral Sciences Tabriz, East Azerbaijan, Iran
| | - Reza Ferdousi
- Department of Health Information Technology, School of Management and Medical Informatics, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
7
|
Phan LT, Rakkiyappan R, Manavalan B. REMED-T2D: A robust ensemble learning model for early detection of type 2 diabetes using healthcare dataset. Comput Biol Med 2025; 187:109771. [PMID: 39914204 DOI: 10.1016/j.compbiomed.2025.109771] [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: 10/30/2024] [Revised: 12/31/2024] [Accepted: 01/29/2025] [Indexed: 02/21/2025]
Abstract
Early diagnosis and timely treatment of diabetes are critical for effective disease management and the prevention of complications. Undiagnosed diabetes can lead to an increased risk of several health issues. Although numerous machine learning (ML) models have been designed to detect diabetes, many exhibit unsatisfactory performance, are not publicly available, and lack validation on external datasets. To address these limitations, we have developed REMED-T2D, an advanced ensemble ML approach that enhances predictive accuracy and robustness through the integration of diverse ML algorithms. Our approach involves a rigorous data preprocessing process and systematic evaluation of 20 different algorithms, encompassing both conventional ML and deep learning for diabetes prediction. Firstly, we applied an under-sampling approach to an imbalanced Pima Indian Diabetes dataset and generated five balanced datasets. Using these datasets, we investigated various computational strategies to select the optimal model for accurate diabetes classification. Our results demonstrate that REMED-T2D outperformed state-of-the-art methods on the training dataset, with notable improvements in ACC (1.40-4.60%) and MCC (3.50-9.80%). Extensive external validations revealed that the model trained on a five-feature subset achieved ACC of 92.61 % and 92.26 % on the RTML1 and Pabna datasets, respectively. Moreover, a model based on a seven-feature subset improved ACC by 2.80 % and MCC by 13.27 % on the RTML2 dataset. These results suggest the potential of REMED-T2D to predict diabetes in Asian females. Notably, this is the first study to conduct such a comprehensive analysis using the Pima dataset, incorporating a diverse set of ML algorithms. Furthermore, we have developed a publicly accessible web server (https://balalab-skku.org/REMED-T2D/) to facilitate self-monitoring and timely medical interventions. We believe REMED-T2D will assist healthcare professionals in detecting diabetes earlier and implementing preventive measures, ultimately improving health outcomes for those at risk.
Collapse
Affiliation(s)
- Le Thi Phan
- Computational Biology and Bioinformatics Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, 16149, Gyeonggi-do, Republic of Korea
| | - Rajan Rakkiyappan
- Department of Mathematics, Bharathiar University, Coimbatore, 641046, Tamil Nadu, India
| | - Balachandran Manavalan
- Computational Biology and Bioinformatics Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, 16149, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
8
|
Li H, Meng J, Wang Z, Luan Y. misORFPred: A Novel Method to Mine Translatable sORFs in Plant Pri-miRNAs Using Enhanced Scalable k-mer and Dynamic Ensemble Voting Strategy. Interdiscip Sci 2025; 17:114-133. [PMID: 39397199 DOI: 10.1007/s12539-024-00661-8] [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: 12/27/2023] [Revised: 09/18/2024] [Accepted: 09/22/2024] [Indexed: 10/15/2024]
Abstract
The primary microRNAs (pri-miRNAs) have been observed to contain translatable small open reading frames (sORFs) that can encode peptides as an independent element. Relevant studies have proven that those of sORFs are of significance in regulating the expression of biological traits. The existing methods for predicting the coding potential of sORFs frequently overlook this data or categorize them as negative samples, impeding the identification of additional translatable sORFs in pri-miRNAs. In light of this, a novel method named misORFPred has been proposed. Specifically, an enhanced scalable k-mer (ESKmer) that simultaneously integrates the composition information within a sequence and distance information between sequences is designed to extract the nucleotide sequence features. After feature selection, the optimal features and several machine learning classifiers are combined to construct the ensemble model, where a newly devised dynamic ensemble voting strategy (DEVS) is proposed to dynamically adjust the weights of base classifiers and adaptively select the optimal base classifiers for each unlabeled sample. Cross-validation results suggest that ESKmer and DEVS are essential for this classification task and could boost model performance. Independent testing results indicate that misORFPred outperforms the state-of-the-art methods. Furthermore, we execute misORFPerd on the genomes of various plant species and perform a thorough analysis of the predicted outcomes. Taken together, misORFPred is a powerful tool for identifying the translatable sORFs in plant pri-miRNAs and can provide highly trusted candidates for subsequent biological experiments.
Collapse
Affiliation(s)
- Haibin Li
- School of Computer Science and Technology, Dalian University of Technology, Dalian, 116024, China
| | - Jun Meng
- School of Computer Science and Technology, Dalian University of Technology, Dalian, 116024, China.
| | - Zhaowei Wang
- School of Computer Science and Technology, Dalian University of Technology, Dalian, 116024, China
| | - Yushi Luan
- School of Bioengineering, Dalian University of Technology, Dalian, 116024, China.
| |
Collapse
|
9
|
Liang Y, Cao M, Zhang S. NeuroPred-ResSE: Predicting neuropeptides by integrating residual block and squeeze-excitation attention mechanism. Anal Biochem 2024; 695:115648. [PMID: 39154878 DOI: 10.1016/j.ab.2024.115648] [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: 06/10/2024] [Revised: 07/31/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
Neuropeptides play crucial roles in regulating neurological function acting as signaling molecules, which provide new opportunity for developing drugs for the treatment of neurological diseases. Therefore, it is very necessary to develop a rapid and accurate prediction model for neuropeptides. Although a few prediction tools have been developed, there is room for improvement in prediction accuracy by using deep learning approach. In this paper, we establish the NeuroPred-ResSE model based on residual block and squeeze-excitation attention mechanism. Firstly, we extract multi-features by using one-hot coding based on the NT5CT5 sequence, dipeptide deviation from expected mean and natural vector. Then, we integrate residual block and squeeze-excitation attention mechanism, which can capture and identify the most relevant attribute features. Finally, the accuracies of the training set and test set are 97.16 % and 96.60 % based on the 5-fold cross-validation and independent test, respectively, and other evaluation metrics have also obtained satisfactory results. The experimental results show that the performance of the NeuroPred-ResSE model outperforms those of existing state-of-the-art models, and our model is an effective, intelligent and robust prediction tool. The datasets and source codes are available at https://github.com/yunyunliang88/NeuroPred-ResSE.
Collapse
Affiliation(s)
- Yunyun Liang
- School of Science, Xi'an Polytechnic University, Xi'an, 710048, PR China.
| | - Mengyi Cao
- School of Science, Xi'an Polytechnic University, Xi'an, 710048, PR China
| | - Shengli Zhang
- School of Mathematics and Statistics, Xidian University, Xi'an, 710071, PR China
| |
Collapse
|
10
|
Lu Q, Xu J, Zhang R, Liu H, Wang M, Liu X, Yue Z, Gao Y. RiceSNP-ABST: a deep learning approach to identify abiotic stress-associated single nucleotide polymorphisms in rice. Brief Bioinform 2024; 26:bbae702. [PMID: 39757606 PMCID: PMC11962596 DOI: 10.1093/bib/bbae702] [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: 09/02/2024] [Revised: 11/16/2024] [Accepted: 12/23/2024] [Indexed: 01/07/2025] Open
Abstract
Given the adverse effects faced by rice due to abiotic stresses, the precise and rapid identification of single nucleotide polymorphisms (SNPs) associated with abiotic stress traits (ABST-SNPs) in rice is crucial for developing resistant rice varieties. The scarcity of high-quality data related to abiotic stress in rice has hindered the development of computational models and constrained research efforts aimed at rice improvement and breeding. Genome-wide association studies provide a better statistical power to consider ABST-SNPs in rice. Meanwhile, deep learning methods have shown their capability in predicting disease- or phenotype-associated loci, but have primarily focused on human species. Therefore, developing predictive models for identifying ABST-SNPs in rice is both urgent and valuable. In this paper, a model called RiceSNP-ABST is proposed for predicting ABST-SNPs in rice. Firstly, six training datasets were generated using a novel strategy for negative sample construction. Secondly, four feature encoding methods were proposed based on DNA sequence fragments, followed by feature selection. Finally, convolutional neural networks with residual connections were used to determine whether the sequences contained rice ABST-SNPs. RiceSNP-ABST outperformed traditional machine learning and state-of-the-art methods on the benchmark dataset and demonstrated consistent generalization on an independent dataset and cross-species datasets. Notably, multi-granularity causal structure learning was employed to elucidate the relationships among DNA structural features, aiming to identify key genetic variants more effectively. The web-based tool for the RiceSNP-ABST can be accessed at http://rice-snp-abst.aielab.cc.
Collapse
Affiliation(s)
- Quan Lu
- School of Information and Artificial Intelligence, Anhui Provincial Engineering Research Center for Beidou Precision Agriculture Information, Anhui Agricultural University, 130, Changjiang West Road, Hefei, Anhui Province 230036, China
| | - Jiajun Xu
- School of Information and Artificial Intelligence, Anhui Provincial Engineering Research Center for Beidou Precision Agriculture Information, Anhui Agricultural University, 130, Changjiang West Road, Hefei, Anhui Province 230036, China
| | - Renyi Zhang
- School of Information and Artificial Intelligence, Anhui Provincial Engineering Research Center for Beidou Precision Agriculture Information, Anhui Agricultural University, 130, Changjiang West Road, Hefei, Anhui Province 230036, China
| | - Hangcheng Liu
- School of Information and Artificial Intelligence, Anhui Provincial Engineering Research Center for Beidou Precision Agriculture Information, Anhui Agricultural University, 130, Changjiang West Road, Hefei, Anhui Province 230036, China
| | - Meng Wang
- School of Information and Artificial Intelligence, Anhui Provincial Engineering Research Center for Beidou Precision Agriculture Information, Anhui Agricultural University, 130, Changjiang West Road, Hefei, Anhui Province 230036, China
| | - Xiaoshuang Liu
- Research Center for Biological Breeding Technology, Advance Academy, Anhui Agricultural University, 130, Changjiang West Road, Hefei, Anhui Province 230036, China
| | - Zhenyu Yue
- School of Information and Artificial Intelligence, Anhui Provincial Engineering Research Center for Beidou Precision Agriculture Information, Anhui Agricultural University, 130, Changjiang West Road, Hefei, Anhui Province 230036, China
- Research Center for Biological Breeding Technology, Advance Academy, Anhui Agricultural University, 130, Changjiang West Road, Hefei, Anhui Province 230036, China
| | - Yujia Gao
- School of Information and Artificial Intelligence, Anhui Provincial Engineering Research Center for Beidou Precision Agriculture Information, Anhui Agricultural University, 130, Changjiang West Road, Hefei, Anhui Province 230036, China
- Research Center for Biological Breeding Technology, Advance Academy, Anhui Agricultural University, 130, Changjiang West Road, Hefei, Anhui Province 230036, China
| |
Collapse
|
11
|
Wen J, Ding Z, Wei Z, Xia H, Zhang Y, Zhu X. NeuroPpred-SHE: An interpretable neuropeptides prediction model based on selected features from hand-crafted features and embeddings of T5 model. Comput Biol Med 2024; 181:109048. [PMID: 39182368 DOI: 10.1016/j.compbiomed.2024.109048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 08/13/2024] [Accepted: 08/18/2024] [Indexed: 08/27/2024]
Abstract
Neuropeptides are the most ubiquitous neurotransmitters in the immune system, regulating various biological processes. Neuropeptides play a significant role for the discovery of new drugs and targets for nervous system disorders. Traditional experimental methods for identifying neuropeptides are time-consuming and costly. Although several computational methods have been developed to predict the neuropeptides, the accuracy is still not satisfactory due to the representability of the extracted features. In this work, we propose an efficient and interpretable model, NeuroPpred-SHE, for predicting neuropeptides by selecting the optimal feature subset from both hand-crafted features and embeddings of a protein language model. Specially, we first employed a pre-trained T5 protein language model to extract embedding features and twelve other encoding methods to extract hand-crafted features from peptide sequences, respectively. Secondly, we fused both embedding features and hand-crafted features to enhance the feature representability. Thirdly, we utilized random forest (RF), Max-Relevance and Min-Redundancy (mRMR) and eXtreme Gradient Boosting (XGBoost) methods to select the optimal feature subset from the fused features. Finally, we employed five machine learning methods (GBDT, XGBoost, SVM, MLP, and LightGBM) to build the models. Our results show that the model based on GBDT achieves the best performance. Furthermore, our final model was compared with other state-of-the-art methods on an independent test set, the results indicate that our model achieves an AUROC of 97.8 % which is higher than all the other state-of-the-art predictors. Our model is available at: https://github.com/wenjean/NeuroPpred-SHE.
Collapse
Affiliation(s)
- Jian Wen
- School of Information and Artificial Intelligence, Anhui Agricultural University, Hefei, 230036, China
| | - Zhijie Ding
- School of Information and Artificial Intelligence, Anhui Agricultural University, Hefei, 230036, China
| | - Zhuoyu Wei
- School of Information and Artificial Intelligence, Anhui Agricultural University, Hefei, 230036, China
| | - Hongwei Xia
- School of Information and Artificial Intelligence, Anhui Agricultural University, Hefei, 230036, China
| | - Yong Zhang
- School of Information and Artificial Intelligence, Anhui Agricultural University, Hefei, 230036, China.
| | - Xiaolei Zhu
- School of Information and Artificial Intelligence, Anhui Agricultural University, Hefei, 230036, China.
| |
Collapse
|
12
|
Kurata H, Harun-Or-Roshid M, Tsukiyama S, Maeda K. PredIL13: Stacking a variety of machine and deep learning methods with ESM-2 language model for identifying IL13-inducing peptides. PLoS One 2024; 19:e0309078. [PMID: 39172871 PMCID: PMC11340954 DOI: 10.1371/journal.pone.0309078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 08/05/2024] [Indexed: 08/24/2024] Open
Abstract
Interleukin (IL)-13 has emerged as one of the recently identified cytokine. Since IL-13 causes the severity of COVID-19 and alters crucial biological processes, it is urgent to explore novel molecules or peptides capable of including IL-13. Computational prediction has received attention as a complementary method to in-vivo and in-vitro experimental identification of IL-13 inducing peptides, because experimental identification is time-consuming, laborious, and expensive. A few computational tools have been presented, including the IL13Pred and iIL13Pred. To increase prediction capability, we have developed PredIL13, a cutting-edge ensemble learning method with the latest ESM-2 protein language model. This method stacked the probability scores outputted by 168 single-feature machine/deep learning models, and then trained a logistic regression-based meta-classifier with the stacked probability score vectors. The key technology was to implement ESM-2 and to select the optimal single-feature models according to their absolute weight coefficient for logistic regression (AWCLR), an indicator of the importance of each single-feature model. Especially, the sequential deletion of single-feature models based on the iterative AWCLR ranking (SDIWC) method constructed the meta-classifier consisting of the top 16 single-feature models, named PredIL13, while considering the model's accuracy. The PredIL13 greatly outperformed the-state-of-the-art predictors, thus is an invaluable tool for accelerating the detection of IL13-inducing peptide within the human genome.
Collapse
Affiliation(s)
- Hiroyuki Kurata
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, Kawazu, Iizuka, Fukuoka, Japan
| | - Md. Harun-Or-Roshid
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, Kawazu, Iizuka, Fukuoka, Japan
| | - Sho Tsukiyama
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, Kawazu, Iizuka, Fukuoka, Japan
| | - Kazuhiro Maeda
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, Kawazu, Iizuka, Fukuoka, Japan
| |
Collapse
|
13
|
Kurata H, Harun-Or-Roshid M, Mehedi Hasan M, Tsukiyama S, Maeda K, Manavalan B. MLm5C: A high-precision human RNA 5-methylcytosine sites predictor based on a combination of hybrid machine learning models. Methods 2024; 227:37-47. [PMID: 38729455 DOI: 10.1016/j.ymeth.2024.05.004] [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: 09/22/2023] [Revised: 04/22/2024] [Accepted: 05/06/2024] [Indexed: 05/12/2024] Open
Abstract
RNA modification serves as a pivotal component in numerous biological processes. Among the prevalent modifications, 5-methylcytosine (m5C) significantly influences mRNA export, translation efficiency and cell differentiation and are also associated with human diseases, including Alzheimer's disease, autoimmune disease, cancer, and cardiovascular diseases. Identification of m5C is critically responsible for understanding the RNA modification mechanisms and the epigenetic regulation of associated diseases. However, the large-scale experimental identification of m5C present significant challenges due to labor intensity and time requirements. Several computational tools, using machine learning, have been developed to supplement experimental methods, but identifying these sites lack accuracy and efficiency. In this study, we introduce a new predictor, MLm5C, for precise prediction of m5C sites using sequence data. Briefly, we evaluated eleven RNA sequence-derived features with four basic machine learning algorithms to generate baseline models. From these 44 models, we ranked them based on their performance and subsequently stacked the Top 20 baseline models as the best model, named MLm5C. The MLm5C outperformed the-state-of-the-art predictors. Notably, the optimization of the sequence length surrounding the modification sites significantly improved the prediction performance. MLm5C is an invaluable tool in accelerating the detection of m5C sites within the human genome, thereby facilitating in the characterization of their roles in post-transcriptional regulation.
Collapse
Affiliation(s)
- Hiroyuki Kurata
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, 680-4 Kawazu, Iizuka, Fukuoka 820-8502, Japan.
| | - Md Harun-Or-Roshid
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, 680-4 Kawazu, Iizuka, Fukuoka 820-8502, Japan
| | - Md Mehedi Hasan
- Division of Biotetecnology and Molecular Medicine, Department of Pathobiological Science, School of Veterinary Medicine, Lousiana State University, Baton Rouge, LA 70803, USA
| | - Sho Tsukiyama
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, 680-4 Kawazu, Iizuka, Fukuoka 820-8502, Japan
| | - Kazuhiro Maeda
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, 680-4 Kawazu, Iizuka, Fukuoka 820-8502, Japan
| | - Balachandran Manavalan
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
14
|
Basith S, Pham NT, Manavalan B, Lee G. SEP-AlgPro: An efficient allergen prediction tool utilizing traditional machine learning and deep learning techniques with protein language model features. Int J Biol Macromol 2024; 273:133085. [PMID: 38871100 DOI: 10.1016/j.ijbiomac.2024.133085] [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: 11/24/2023] [Revised: 05/20/2024] [Accepted: 06/09/2024] [Indexed: 06/15/2024]
Abstract
Allergy is a hypersensitive condition in which individuals develop objective symptoms when exposed to harmless substances at a dose that would cause no harm to a "normal" person. Most current computational methods for allergen identification rely on homology or conventional machine learning using limited set of feature descriptors or validation on specific datasets, making them inefficient and inaccurate. Here, we propose SEP-AlgPro for the accurate identification of allergen protein from sequence information. We analyzed 10 conventional protein-based features and 14 different features derived from protein language models to gauge their effectiveness in differentiating allergens from non-allergens using 15 different classifiers. However, the final optimized model employs top 10 feature descriptors with top seven machine learning classifiers. Results show that the features derived from protein language models exhibit superior discriminative capabilities compared to traditional feature sets. This enabled us to select the most discriminatory baseline models, whose predicted outputs were aggregated and used as input to a deep neural network for the final allergen prediction. Extensive case studies showed that SEP-AlgPro outperforms state-of-the-art predictors in accurately identifying allergens. A user-friendly web server was developed and made freely available at https://balalab-skku.org/SEP-AlgPro/, making it a powerful tool for identifying potential allergens.
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
| | - Balachandran Manavalan
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, 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.
| |
Collapse
|
15
|
Li H, Jiang L, Yang K, Shang S, Li M, Lv Z. iNP_ESM: Neuropeptide Identification Based on Evolutionary Scale Modeling and Unified Representation Embedding Features. Int J Mol Sci 2024; 25:7049. [PMID: 39000158 PMCID: PMC11240975 DOI: 10.3390/ijms25137049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 06/17/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
Neuropeptides are biomolecules with crucial physiological functions. Accurate identification of neuropeptides is essential for understanding nervous system regulatory mechanisms. However, traditional analysis methods are expensive and laborious, and the development of effective machine learning models continues to be a subject of current research. Hence, in this research, we constructed an SVM-based machine learning neuropeptide predictor, iNP_ESM, by integrating protein language models Evolutionary Scale Modeling (ESM) and Unified Representation (UniRep) for the first time. Our model utilized feature fusion and feature selection strategies to improve prediction accuracy during optimization. In addition, we validated the effectiveness of the optimization strategy with UMAP (Uniform Manifold Approximation and Projection) visualization. iNP_ESM outperforms existing models on a variety of machine learning evaluation metrics, with an accuracy of up to 0.937 in cross-validation and 0.928 in independent testing, demonstrating optimal neuropeptide recognition capabilities. We anticipate improved neuropeptide data in the future, and we believe that the iNP_ESM model will have broader applications in the research and clinical treatment of neurological diseases.
Collapse
Affiliation(s)
- Honghao Li
- College of Biomedical Engineering, Sichuan University, Chengdu 610041, China
| | - Liangzhen Jiang
- College of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
- Country Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, Chengdu 610106, China
| | - Kaixiang Yang
- College of Software Engineering, Sichuan University, Chengdu 610041, China
| | - Shulin Shang
- College of Biomedical Engineering, Sichuan University, Chengdu 610041, China
| | - Mingxin Li
- College of Biomedical Engineering, Sichuan University, Chengdu 610041, China
| | - Zhibin Lv
- College of Biomedical Engineering, Sichuan University, Chengdu 610041, China
| |
Collapse
|
16
|
Singh V, Singh SK, Sharma R. A novel framework based on explainable AI and genetic algorithms for designing neurological medicines. Sci Rep 2024; 14:12807. [PMID: 38834718 DOI: 10.1038/s41598-024-63561-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 05/30/2024] [Indexed: 06/06/2024] Open
Abstract
The advent of the fourth industrial revolution, characterized by artificial intelligence (AI) as its central component, has resulted in the mechanization of numerous previously labor-intensive activities. The use of in silico tools has become prevalent in the design of biopharmaceuticals. Upon conducting a comprehensive analysis of the genomes of many organisms, it has been discovered that their tissues can generate specific peptides that confer protection against certain diseases. This study aims to identify a selected group of neuropeptides (NPs) possessing favorable characteristics that render them ideal for production as neurological biopharmaceuticals. Until now, the construction of NP classifiers has been the primary focus, neglecting to optimize these characteristics. Therefore, in this study, the task of creating ideal NPs has been formulated as a multi-objective optimization problem. The proposed framework, NPpred, comprises two distinct components: NSGA-NeuroPred and BERT-NeuroPred. The former employs the NSGA-II algorithm to explore and change a population of NPs, while the latter is an interpretable deep learning-based model. The utilization of explainable AI and motifs has led to the proposal of two novel operators, namely p-crossover and p-mutation. An online application has been deployed at https://neuropred.anvil.app for designing an ideal collection of synthesizable NPs from protein sequences.
Collapse
Affiliation(s)
- Vishakha Singh
- Department of Computer Science and Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, Uttar Pradesh, India.
| | - Sanjay Kumar Singh
- Department of Computer Science and Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, Uttar Pradesh, India.
| | - Ritesh Sharma
- Department of ICT, Manipal Institute of Technology, Manipal, 576104, Uttar Pradesh, India
| |
Collapse
|
17
|
Liao YH, Chen SZ, Bin YN, Zhao JP, Feng XL, Zheng CH. UsIL-6: An unbalanced learning strategy for identifying IL-6 inducing peptides by undersampling technique. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 250:108176. [PMID: 38677081 DOI: 10.1016/j.cmpb.2024.108176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 03/26/2024] [Accepted: 04/11/2024] [Indexed: 04/29/2024]
Abstract
BACKGROUND AND OBJECTIVE Interleukin-6 (IL-6) is the critical factor of early warning, monitoring, and prognosis in the inflammatory storm of COVID-19 cases. IL-6 inducing peptides, which can induce cytokine IL-6 production, are very important for the development of diagnosis and immunotherapy. Although the existing methods have some success in predicting IL-6 inducing peptides, there is still room for improvement in the performance of these models in practical application. METHODS In this study, we proposed UsIL-6, a high-performance bioinformatics tool for identifying IL-6 inducing peptides. First, we extracted five groups of physicochemical properties and sequence structural information from IL-6 inducing peptide sequences, and obtained a 636-dimensional feature vector, we also employed NearMiss3 undersampling method and normalization method StandardScaler to process the data. Then, a 40-dimensional optimal feature vector was obtained by Boruta feature selection method. Finally, we combined this feature vector with extreme randomization tree classifier to build the final model UsIL-6. RESULTS The AUC value of UsIL-6 on the independent test dataset was 0.87, and the BACC value was 0.808, which indicated that UsIL-6 had better performance than the existing methods in IL-6 inducing peptide recognition. CONCLUSIONS The performance comparison on independent test dataset confirmed that UsIL-6 could achieve the highest performance, best robustness, and most excellent generalization ability. We hope that UsIL-6 will become a valuable method to identify, annotate and characterize new IL-6 inducing peptides.
Collapse
Affiliation(s)
- Yan-Hong Liao
- School of Mathematics and System Science, Xinjiang University, Urumqi, Xinjiang 830017, China
| | - Shou-Zhi Chen
- School of Mathematics and System Science, Xinjiang University, Urumqi, Xinjiang 830017, China
| | - Yan-Nan Bin
- School of Computer Science and Technology, Anhui University, Hefei, Anhui 230601, China
| | - Jian-Ping Zhao
- School of Mathematics and System Science, Xinjiang University, Urumqi, Xinjiang 830017, China.
| | - Xin-Long Feng
- School of Mathematics and System Science, Xinjiang University, Urumqi, Xinjiang 830017, China.
| | - Chun-Hou Zheng
- School of Mathematics and System Science, Xinjiang University, Urumqi, Xinjiang 830017, China; School of Computer Science and Technology, Anhui University, Hefei, Anhui 230601, China
| |
Collapse
|
18
|
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
|
19
|
Wang M, Wang L, Xu W, Chu Z, Wang H, Lu J, Xue Z, Wang Y. NeuroPep 2.0: An Updated Database Dedicated to Neuropeptide and Its Receptor Annotations. J Mol Biol 2024; 436:168416. [PMID: 38143020 DOI: 10.1016/j.jmb.2023.168416] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023]
Abstract
Neuropeptides not only work through nervous system but some of them also work peripherally to regulate numerous physiological processes. They are important in regulation of numerous physiological processes including growth, reproduction, social behavior, inflammation, fluid homeostasis, cardiovascular function, and energy homeostasis. The various roles of neuropeptides make them promising candidates for prospective therapeutics of different diseases. Currently, NeuroPep has been updated to version 2.0, it now holds 11,417 unique neuropeptide entries, which is nearly double of the first version of NeuroPep. When available, we collected information about the receptor for each neuropeptide entry and predicted the 3D structures of those neuropeptides without known experimental structure using AlphaFold2 or APPTEST according to the peptide sequence length. In addition, DeepNeuropePred and NeuroPred-PLM, two neuropeptide prediction tools developed by us recently, were also integrated into NeuroPep 2.0 to help to facilitate the identification of new neuropeptides. NeuroPep 2.0 is freely accessible at https://isyslab.info/NeuroPepV2/.
Collapse
Affiliation(s)
- Mingxia Wang
- Institute of Medical Artificial Intelligence, Binzhou Medical University, Yantai, Shandong 264003, China
| | - Lei Wang
- School of Software Engineering, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Wei Xu
- Institute of Medical Artificial Intelligence, Binzhou Medical University, Yantai, Shandong 264003, China
| | - Ziqiang Chu
- School of Software Engineering, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Hengzhi Wang
- School of Software Engineering, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Jingxiang Lu
- Institute of Medical Artificial Intelligence, Binzhou Medical University, Yantai, Shandong 264003, China
| | - Zhidong Xue
- Institute of Medical Artificial Intelligence, Binzhou Medical University, Yantai, Shandong 264003, China; School of Software Engineering, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Yan Wang
- Institute of Medical Artificial Intelligence, Binzhou Medical University, Yantai, Shandong 264003, China; School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China.
| |
Collapse
|
20
|
Harun-Or-Roshid M, Maeda K, Phan LT, Manavalan B, Kurata H. Stack-DHUpred: Advancing the accuracy of dihydrouridine modification sites detection via stacking approach. Comput Biol Med 2024; 169:107848. [PMID: 38145601 DOI: 10.1016/j.compbiomed.2023.107848] [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: 08/26/2023] [Revised: 11/14/2023] [Accepted: 12/11/2023] [Indexed: 12/27/2023]
Abstract
Dihydrouridine (DHU, D) is one of the most abundant post-transcriptional uridine modifications found in tRNA, mRNA, and snoRNA, closely associated with disease pathogenesis and various biological processes in eukaryotes. Identifying D sites is important for understanding the modification mechanisms and/or epigenetic regulation. However, biological experiments for detecting D sites are time-consuming and expensive. Given these challenges, computational methods have been developed for accurately identifying the D sites in genome-wide datasets. However, existing methods have some limitations, and their prediction performance needs to be improved. In this work, we have developed a new computational predictor for accurately identifying D sites called Stack-DHUpred. Briefly, we trained 66 baseline models or single-feature models by connecting six machine learning classifiers with eleven different feature encoding methods and stacked different baseline models to build stacked ensemble learning models. Subsequently, the optimal combination of the baseline models was identified for the construction of the final stacked model. Remarkably, the Stack-DHUpred outperformed the existing predictors on our new independent dataset, indicating that the stacking approach significantly improved the prediction performance. We have made Stack-DHUpred available to the public through a web server (http://kurata35.bio.kyutech.ac.jp/Stack-DHUpred) and a standalone program (https://github.com/kuratahiroyuki/Stack-DHUpred). We believe that Stack-DHUpred will be a valuable tool for accelerating the discovery of D modifications and understanding their role in post-transcriptional regulation.
Collapse
Affiliation(s)
- Md Harun-Or-Roshid
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, 680-4 Kawazu, Iizuka, Fukuoka 820-8502, Japan
| | - Kazuhiro Maeda
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, 680-4 Kawazu, Iizuka, Fukuoka 820-8502, Japan
| | - Le Thi Phan
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Balachandran Manavalan
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Hiroyuki Kurata
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, 680-4 Kawazu, Iizuka, Fukuoka 820-8502, Japan.
| |
Collapse
|
21
|
Toussaint PA, Leiser F, Thiebes S, Schlesner M, Brors B, Sunyaev A. Explainable artificial intelligence for omics data: a systematic mapping study. Brief Bioinform 2023; 25:bbad453. [PMID: 38113073 PMCID: PMC10729786 DOI: 10.1093/bib/bbad453] [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: 12/23/2022] [Revised: 07/28/2023] [Accepted: 11/08/2023] [Indexed: 12/21/2023] Open
Abstract
Researchers increasingly turn to explainable artificial intelligence (XAI) to analyze omics data and gain insights into the underlying biological processes. Yet, given the interdisciplinary nature of the field, many findings have only been shared in their respective research community. An overview of XAI for omics data is needed to highlight promising approaches and help detect common issues. Toward this end, we conducted a systematic mapping study. To identify relevant literature, we queried Scopus, PubMed, Web of Science, BioRxiv, MedRxiv and arXiv. Based on keywording, we developed a coding scheme with 10 facets regarding the studies' AI methods, explainability methods and omics data. Our mapping study resulted in 405 included papers published between 2010 and 2023. The inspected papers analyze DNA-based (mostly genomic), transcriptomic, proteomic or metabolomic data by means of neural networks, tree-based methods, statistical methods and further AI methods. The preferred post-hoc explainability methods are feature relevance (n = 166) and visual explanation (n = 52), while papers using interpretable approaches often resort to the use of transparent models (n = 83) or architecture modifications (n = 72). With many research gaps still apparent for XAI for omics data, we deduced eight research directions and discuss their potential for the field. We also provide exemplary research questions for each direction. Many problems with the adoption of XAI for omics data in clinical practice are yet to be resolved. This systematic mapping study outlines extant research on the topic and provides research directions for researchers and practitioners.
Collapse
Affiliation(s)
- Philipp A Toussaint
- Department of Economics and Management, Karlsruhe Institute of Technology, Karlsruhe, Germany
- HIDSS4Health – Helmholtz Information and Data Science School for Health, Karlsruhe, Heidelberg, Germany
| | - Florian Leiser
- Department of Economics and Management, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Scott Thiebes
- Department of Economics and Management, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Matthias Schlesner
- Biomedical Informatics, Data Mining and Data Analytics, Faculty of Applied Computer Science and Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Benedikt Brors
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Translational Oncology, National Center for Tumor Diseases, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ali Sunyaev
- Department of Economics and Management, Karlsruhe Institute of Technology, Karlsruhe, Germany
| |
Collapse
|
22
|
Zhang X, Guo H, Zhang F, Wang X, Wu K, Qiu S, Liu B, Wang Y, Hu Y, Li J. HNetGO: protein function prediction via heterogeneous network transformer. Brief Bioinform 2023; 24:bbab556. [PMID: 37861172 PMCID: PMC10588005 DOI: 10.1093/bib/bbab556] [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: 08/05/2021] [Revised: 11/18/2021] [Accepted: 12/04/2021] [Indexed: 10/21/2023] Open
Abstract
Protein function annotation is one of the most important research topics for revealing the essence of life at molecular level in the post-genome era. Current research shows that integrating multisource data can effectively improve the performance of protein function prediction models. However, the heavy reliance on complex feature engineering and model integration methods limits the development of existing methods. Besides, models based on deep learning only use labeled data in a certain dataset to extract sequence features, thus ignoring a large amount of existing unlabeled sequence data. Here, we propose an end-to-end protein function annotation model named HNetGO, which innovatively uses heterogeneous network to integrate protein sequence similarity and protein-protein interaction network information and combines the pretraining model to extract the semantic features of the protein sequence. In addition, we design an attention-based graph neural network model, which can effectively extract node-level features from heterogeneous networks and predict protein function by measuring the similarity between protein nodes and gene ontology term nodes. Comparative experiments on the human dataset show that HNetGO achieves state-of-the-art performance on cellular component and molecular function branches.
Collapse
Affiliation(s)
- Xiaoshuai Zhang
- School of Computer Science and Technology, Harbin Institute of Technology (Shenzhen), Shenzhen, Guangdong 518055, China
| | - Huannan Guo
- General Hospital of Heilongjiang Province Land Reclamation Bureau, Harbin 150086, China
| | - Fan Zhang
- Center NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Xuan Wang
- School of Computer Science and Technology, Harbin Institute of Technology (Shenzhen), Shenzhen, Guangdong 518055, China
| | - Kaitao Wu
- School of Computer Science and Technology, Harbin Institute of Technology (Shenzhen), Shenzhen, Guangdong 518055, China
| | - Shizheng Qiu
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Bo Liu
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Yadong Wang
- School of Computer Science and Technology, Harbin Institute of Technology (Shenzhen), Shenzhen, Guangdong 518055, China
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Yang Hu
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Junyi Li
- School of Computer Science and Technology, Harbin Institute of Technology (Shenzhen), Shenzhen, Guangdong 518055, China
| |
Collapse
|
23
|
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
|
24
|
Liu D, Lin Z, Jia C. NeuroCNN_GNB: an ensemble model to predict neuropeptides based on a convolution neural network and Gaussian naive Bayes. Front Genet 2023; 14:1226905. [PMID: 37576553 PMCID: PMC10414792 DOI: 10.3389/fgene.2023.1226905] [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: 05/22/2023] [Accepted: 06/30/2023] [Indexed: 08/15/2023] Open
Abstract
Neuropeptides contain more chemical information than other classical neurotransmitters and have multiple receptor recognition sites. These characteristics allow neuropeptides to have a correspondingly higher selectivity for nerve receptors and fewer side effects. Traditional experimental methods, such as mass spectrometry and liquid chromatography technology, still need the support of a complete neuropeptide precursor database and the basic characteristics of neuropeptides. Incomplete neuropeptide precursor and information databases will lead to false-positives or reduce the sensitivity of recognition. In recent years, studies have proven that machine learning methods can rapidly and effectively predict neuropeptides. In this work, we have made a systematic attempt to create an ensemble tool based on four convolution neural network models. These baseline models were separately trained on one-hot encoding, AAIndex, G-gap dipeptide encoding and word2vec and integrated using Gaussian Naive Bayes (NB) to construct our predictor designated NeuroCNN_GNB. Both 5-fold cross-validation tests using benchmark datasets and independent tests showed that NeuroCNN_GNB outperformed other state-of-the-art methods. Furthermore, this novel framework provides essential interpretations that aid the understanding of model success by leveraging the powerful Shapley Additive exPlanation (SHAP) algorithm, thereby highlighting the most important features relevant for predicting neuropeptides.
Collapse
Affiliation(s)
- Di Liu
- Information Science and Technology College, Dalian Maritime University, Dalian, China
| | - Zhengkui Lin
- Information Science and Technology College, Dalian Maritime University, Dalian, China
| | - Cangzhi Jia
- School of Science, Dalian Maritime University, Dalian, China
| |
Collapse
|
25
|
Zulfiqar H, Guo Z, Grace-Mercure BK, Zhang ZY, Gao H, Lin H, Wu Y. Empirical comparison and recent advances of computational prediction of hormone binding proteins using machine learning methods. Comput Struct Biotechnol J 2023; 21:2253-2261. [PMID: 37035551 PMCID: PMC10073991 DOI: 10.1016/j.csbj.2023.03.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 03/15/2023] [Accepted: 03/16/2023] [Indexed: 03/19/2023] Open
Abstract
Hormone binding proteins (HBPs) belong to the group of soluble carrier proteins. These proteins selectively and non-covalently interact with hormones and promote growth hormone signaling in human and other animals. The HBPs are useful in many medical and commercial fields. Thus, the identification of HBPs is very important because it can help to discover more details about hormone binding proteins. Meanwhile, the experimental methods are time-consuming and expensive for hormone binding proteins recognition. Computational prediction methods have played significant roles in the correct recognition of hormone binding proteins with the use of sequence information and ML algorithms. In this review, we compared and assessed the implementation of ML-based tools in recognition of HBPs in a unique way. We hope that this study will give enough awareness and knowledge for research on HBPs.
Collapse
Affiliation(s)
- Hasan Zulfiqar
- Yangtze Delta Region Institute (Huzhou), University of Electronic Science and Technology of China, Huzhou, Zhejiang 313001, China
- School of Life Science and Technology and Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China
- School of Computer Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Zhiling Guo
- Beidahuang Industry Group General Hospital, Harbin, China
| | - Bakanina Kissanga Grace-Mercure
- School of Life Science and Technology and Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Zhao-Yue Zhang
- School of Life Science and Technology and Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Hui Gao
- School of Computer Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Hao Lin
- Yangtze Delta Region Institute (Huzhou), University of Electronic Science and Technology of China, Huzhou, Zhejiang 313001, China
- School of Life Science and Technology and Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Yun Wu
- College of Computer and Information Engineering, Xiamen University of Technology, Xiamen 361024, China
| |
Collapse
|
26
|
Liu Y, Wang S, Li X, Liu Y, Zhu X. NeuroPpred-SVM: A New Model for Predicting Neuropeptides Based on Embeddings of BERT. J Proteome Res 2023; 22:718-728. [PMID: 36749151 DOI: 10.1021/acs.jproteome.2c00363] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Neuropeptides play pivotal roles in different physiological processes and are related to different kinds of diseases. Identification of neuropeptides is of great benefit for studying the mechanism of these physiological processes and the treatment of neurological disorders. Several state-of-the-art neuropeptide predictors have been developed by using a two-layer stacking ensemble algorithm. Although the two-layer stacking ensemble algorithm can improve the feature representability, these models are complex, which are not as efficient as the models based on one classifier. In this study, we proposed a new model, NeuroPpred-SVM, to predict neuropeptides based on the embeddings of Bidirectional Encoder Representations from Transformers and other sequential features by using a support vector machine (SVM). The experimental results indicate that our model achieved a cross-validation area under the receiver operating characteristic (AUROC) curve of 0.969 on the training data set and an AUROC of 0.966 on the independent test set. By comparing our model with the other four state-of-the-art models including NeuroPIpred, PredNeuroP, NeuroPpred-Fuse, and NeuroPpred-FRL on the independent test set, our model achieved the highest AUROC, Matthews correlation coefficient, accuracy, and specificity, which indicate that our model outperforms the existing models. We believed that NeuroPpred-SVM could be a useful tool for identifying neuropeptides with high accuracy and low cost. The data sets and Python code are available at https://github.com/liuyf-a/NeuroPpred-SVM.
Collapse
Affiliation(s)
- Yufeng Liu
- School of Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Shuyu Wang
- School of Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Xiang Li
- School of Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Yinbo Liu
- School of Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Xiaolei Zhu
- School of Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| |
Collapse
|
27
|
Li C, Hua Y, Pan D, Qi L, Xiao C, Xiong Y, Lu W, Dang Y, Gao X, Zhao Y. A rapid selection strategy for umami peptide screening based on machine learning and molecular docking. Food Chem 2023; 404:134562. [DOI: 10.1016/j.foodchem.2022.134562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 10/02/2022] [Accepted: 10/07/2022] [Indexed: 11/22/2022]
|
28
|
Phetsanthad A, Vu NQ, Yu Q, Buchberger AR, Chen Z, Keller C, Li L. Recent advances in mass spectrometry analysis of neuropeptides. MASS SPECTROMETRY REVIEWS 2023; 42:706-750. [PMID: 34558119 PMCID: PMC9067165 DOI: 10.1002/mas.21734] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 08/22/2021] [Accepted: 08/28/2021] [Indexed: 05/08/2023]
Abstract
Due to their involvement in numerous biochemical pathways, neuropeptides have been the focus of many recent research studies. Unfortunately, classic analytical methods, such as western blots and enzyme-linked immunosorbent assays, are extremely limited in terms of global investigations, leading researchers to search for more advanced techniques capable of probing the entire neuropeptidome of an organism. With recent technological advances, mass spectrometry (MS) has provided methodology to gain global knowledge of a neuropeptidome on a spatial, temporal, and quantitative level. This review will cover key considerations for the analysis of neuropeptides by MS, including sample preparation strategies, instrumental advances for identification, structural characterization, and imaging; insightful functional studies; and newly developed absolute and relative quantitation strategies. While many discoveries have been made with MS, the methodology is still in its infancy. Many of the current challenges and areas that need development will also be highlighted in this review.
Collapse
Affiliation(s)
- Ashley Phetsanthad
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, WI 53706, USA
| | - Nhu Q. Vu
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, WI 53706, USA
| | - Qing Yu
- School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, WI 53705, USA
| | - Amanda R. Buchberger
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, WI 53706, USA
| | - Zhengwei Chen
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, WI 53706, USA
| | - Caitlin Keller
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, WI 53706, USA
| | - Lingjun Li
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, WI 53706, USA
- School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, WI 53705, USA
| |
Collapse
|
29
|
Shi H, Wu C, Bai T, Chen J, Li Y, Wu H. Identify essential genes based on clustering based synthetic minority oversampling technique. Comput Biol Med 2023; 153:106523. [PMID: 36652869 DOI: 10.1016/j.compbiomed.2022.106523] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/13/2022] [Accepted: 12/31/2022] [Indexed: 01/03/2023]
Abstract
Prediction of essential genes in a life organism is one of the central tasks in synthetic biology. Computational predictors are desired because experimental data is often unavailable. Recently, some sequence-based predictors have been constructed to identify essential genes. However, their predictive performance should be further improved. One key problem is how to effectively extract the sequence-based features, which are able to discriminate the essential genes. Another problem is the imbalanced training set. The amount of essential genes in human cell lines is lower than that of non-essential genes. Therefore, predictors trained with such imbalanced training set tend to identify an unseen sequence as a non-essential gene. Here, a new over-sampling strategy was proposed called Clustering based Synthetic Minority Oversampling Technique (CSMOTE) to overcome the imbalanced data issue. Combining CSMOTE with the Z curve, the global features, and Support Vector Machines, a new protocol called iEsGene-CSMOTE was proposed to identify essential genes. The rigorous jackknife cross validation results indicated that iEsGene-CSMOTE is better than the other competing methods. The proposed method outperformed λ-interval Z curve by 35.48% and 11.25% in terms of Sn and BACC, respectively.
Collapse
Affiliation(s)
- Hua Shi
- School of Opto-electronic and Communication Engineering, Xiamen University of Technology, Xiamen, China.
| | - Chenjin Wu
- School of Opto-electronic and Communication Engineering, Xiamen University of Technology, Xiamen, China.
| | - Tao Bai
- School of Computer Science and Technology, Beijing Institute of Technology, Beijing, 100081, China; School of Mathematics & Computer Science, Yanan University, Shanxi, 716000, China.
| | - Jiahai Chen
- Xiamen Sankuai Online Technology Co., Ltd, Xiamen, China.
| | - Yan Li
- School of Opto-electronic and Communication Engineering, Xiamen University of Technology, Xiamen, China.
| | - Hao Wu
- School of Computer Science and Technology, Beijing Institute of Technology, Beijing, 100081, China.
| |
Collapse
|
30
|
Reliability of non-contact tongue diagnosis for Sjögren's syndrome using machine learning method. Sci Rep 2023; 13:1334. [PMID: 36693892 PMCID: PMC9872069 DOI: 10.1038/s41598-023-27764-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 01/06/2023] [Indexed: 01/25/2023] Open
Abstract
Sjögren's syndrome (SS) is an autoimmune disease characterized by dry mouth. The cause of SS is unknown, and its diverse symptoms make diagnosis difficult. The Saxon test, an intraoral examination, is used as the primary diagnostic method for SS, however, the risk of salivary infection is problematic. Therefore, we investigate the possibility of diagnosing SS by non-contact and imaging observation of the tongue surface. In this study, we obtained tongue photographs of 60 patients at the Tsurumi University School of Dentistry outpatient clinic to clarify the relationship between the features of the tongue and SS. We divided the tongue into four regions, and the color of each region was transformed into CIE1976L*a*b* space and statistically analyzed. To clarify experimentally the possibility of SS diagnosis using tongue color, we employed three machine-learning models: logistic regression, support vector machine, and random forest. In addition, we constructed diagnostic prediction models based on the Bagging and Stacking methods combined with three machine-learning models for comparative evaluation. This analysis used dimensionality compression by principal component analysis to eliminate redundancy in tongue color information. We found a significant difference between the a* value of the rear part of the tongue and the b* value of the middle part of the tongue in SS and non-SS patients. In addition to the principal component scores of tongue color, the support vector machine was trained using age, and achieved high accuracy (71.3%) and specificity (78.1%). The results indicate that the prediction of SS diagnosis by tongue color reaches a level comparable to machine learning models trained using the Saxon test. This is the first study using machine learning to predict SS diagnosis by non-contact tongue observation. Our proposed method can potentially support early SS detection simply and conveniently, eliminating the risk of infection at diagnosis, and it should be validated and optimized in clinical practice.
Collapse
|
31
|
Yu H, Luo X. IPPF-FE: an integrated peptide and protein function prediction framework based on fused features and ensemble models. Brief Bioinform 2023; 24:6834141. [PMID: 36403184 DOI: 10.1093/bib/bbac476] [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: 08/05/2022] [Revised: 09/23/2022] [Accepted: 10/05/2022] [Indexed: 11/21/2022] Open
Abstract
The prediction of peptide and protein function is important for research and industrial applications, and many machine learning methods have been developed for this purpose. The existing models have encountered many challenges, including the lack of effective and comprehensive features and the limited applicability of each model. Here, we introduce an Integrated Peptide and Protein function prediction Framework based on Fused features and Ensemble models (IPPF-FE), which can accurately capture the relationship between features and labels. The results indicated that IPPF-FE outperformed existing state-of-the-art (SOTA) models on more than 8 different categories of peptide and protein tasks. In addition, t-distributed Stochastic Neighbour Embedding demonstrated the advantages of IPPF-FE. We anticipate that our method will become a versatile tool for peptide and protein prediction tasks and shed light on the future development of related models. The model is open source and available in the GitHub repository https://github.com/Luo-SynBioLab/IPPF-FE.
Collapse
Affiliation(s)
- Han Yu
- Center for Synthetic Biochemistry, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Xiaozhou Luo
- Shenzhen Key Laboratory for the Intelligent Microbial Manufacturing of Medicines, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.,Center for Synthetic Biochemistry, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
32
|
Bupi N, Sangaraju VK, Phan LT, Lal A, Vo TTB, Ho PT, Qureshi MA, Tabassum M, Lee S, Manavalan B. An Effective Integrated Machine Learning Framework for Identifying Severity of Tomato Yellow Leaf Curl Virus and Their Experimental Validation. RESEARCH (WASHINGTON, D.C.) 2023; 6:0016. [PMID: 36930763 PMCID: PMC10013792 DOI: 10.34133/research.0016] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 11/07/2022] [Indexed: 01/13/2023]
Abstract
Tomato yellow leaf curl virus (TYLCV) dispersed across different countries, specifically to subtropical regions, associated with more severe symptoms. Since TYLCV was first isolated in 1931, it has been a menace to tomato industrial production worldwide over the past century. Three groups were newly isolated from TYLCV-resistant tomatoes in 2022; however, their functions are unknown. The development of machine learning (ML)-based models using characterized sequences and evaluating blind predictions is one of the major challenges in interdisciplinary research. The purpose of this study was to develop an integrated computational framework for the accurate identification of symptoms (mild or severe) based on TYLCV sequences (isolated in Korea). For the development of the framework, we first extracted 11 different feature encodings and hybrid features from the training data and then explored 8 different classifiers and developed their respective prediction models by using randomized 10-fold cross-validation. Subsequently, we carried out a systematic evaluation of these 96 developed models and selected the top 90 models, whose predicted class labels were combined and considered as reduced features. On the basis of these features, a multilayer perceptron was applied and developed the final prediction model (IML-TYLCVs). We conducted blind prediction on 3 groups using IML-TYLCVs, and the results indicated that 2 groups were severe and 1 group was mild. Furthermore, we confirmed the prediction with virus-challenging experiments of tomato plant phenotypes using infectious clones from 3 groups. Plant virologists and plant breeding professionals can access the user-friendly online IML-TYLCVs web server at https://balalab-skku.org/IML-TYLCVs, which can guide them in developing new protection strategies for newly emerging viruses.
Collapse
Affiliation(s)
- Nattanong Bupi
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419, Gyeonggi-do, Republic of Korea
| | - Vinoth Kumar Sangaraju
- Computational Biology and Bioinformatics Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419, Gyeonggi-do, Republic of Korea
| | - Le Thi Phan
- Computational Biology and Bioinformatics Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419, Gyeonggi-do, Republic of Korea
| | - Aamir Lal
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419, Gyeonggi-do, Republic of Korea
| | - Thuy Thi Bich Vo
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419, Gyeonggi-do, Republic of Korea
| | - Phuong Thi Ho
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419, Gyeonggi-do, Republic of Korea
| | - Muhammad Amir Qureshi
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419, Gyeonggi-do, Republic of Korea
| | - Marjia Tabassum
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419, Gyeonggi-do, Republic of Korea
| | - Sukchan Lee
- 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
| |
Collapse
|
33
|
Lim H, No KT. Prediction of polyreactive and nonspecific single-chain fragment variables through structural biochemical features and protein language-based descriptors. BMC Bioinformatics 2022; 23:520. [PMID: 36471239 PMCID: PMC9720949 DOI: 10.1186/s12859-022-05010-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 10/26/2022] [Indexed: 12/09/2022] Open
Abstract
BACKGROUND Monoclonal antibodies (mAbs) have been used as therapeutic agents, which must overcome many developability issues after the discovery from in vitro display libraries. Especially, polyreactive mAbs can strongly bind to a specific target and weakly bind to off-target proteins, which leads to poor antibody pharmacokinetics in clinical development. Although early assessment of polyreactive mAbs is important in the early discovery stage, experimental assessments are usually time-consuming and expensive. Therefore, computational approaches for predicting the polyreactivity of single-chain fragment variables (scFvs) in the early discovery stage would be promising for reducing experimental efforts. RESULTS Here, we made prediction models for the polyreactivity of scFvs with the known polyreactive antibody features and natural language model descriptors. We predicted 19,426 protein structures of scFvs with trRosetta to calculate the polyreactive antibody features and investigated the classifying performance of each factor for polyreactivity. In the known polyreactive features, the net charge of the CDR2 loop, the tryptophan and glycine residues in CDR-H3, and the lengths of the CDR1 and CDR2 loops, importantly contributed to the performance of the models. Additionally, the hydrodynamic features, such as partial specific volume, gyration radius, and isoelectric points of CDR loops and scFvs, were newly added to improve model performance. Finally, we made the prediction model with a robust performance ([Formula: see text]) with an ensemble learning of the top 3 best models. CONCLUSION The prediction models for polyreactivity would help assess polyreactive scFvs in the early discovery stage and our approaches would be promising to develop machine learning models with quantitative data from high throughput assays for antibody screening.
Collapse
Affiliation(s)
- Hocheol Lim
- The Interdisciplinary Graduate Program in Integrative Biotechnology and Translational Medicine, Yonsei University, Incheon, 21983, Republic of Korea
- Bioinformatics and Molecular Design Research Center (BMDRC), Incheon, 21983, Republic of Korea
| | - Kyoung Tai No
- The Interdisciplinary Graduate Program in Integrative Biotechnology and Translational Medicine, Yonsei University, Incheon, 21983, Republic of Korea.
- Bioinformatics and Molecular Design Research Center (BMDRC), Incheon, 21983, Republic of Korea.
- Baobab AiBIO Co., Ltd., Incheon, 21983, Republic of Korea.
| |
Collapse
|
34
|
Chen S, Li Q, Zhao J, Bin Y, Zheng C. NeuroPred-CLQ: incorporating deep temporal convolutional networks and multi-head attention mechanism to predict neuropeptides. Brief Bioinform 2022; 23:6672901. [DOI: 10.1093/bib/bbac319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/27/2022] [Accepted: 07/14/2022] [Indexed: 11/13/2022] Open
Abstract
Abstract
Neuropeptides (NPs) are a particular class of informative substances in the immune system and physiological regulation. They play a crucial role in regulating physiological functions in various biological growth and developmental stages. In addition, NPs are crucial for developing new drugs for the treatment of neurological diseases. With the development of molecular biology techniques, some data-driven tools have emerged to predict NPs. However, it is necessary to improve the predictive performance of these tools for NPs. In this study, we developed a deep learning model (NeuroPred-CLQ) based on the temporal convolutional network (TCN) and multi-head attention mechanism to identify NPs effectively and translate the internal relationships of peptide sequences into numerical features by the Word2vec algorithm. The experimental results show that NeuroPred-CLQ learns data information effectively, achieving 93.6% accuracy and 98.8% AUC on the independent test set. The model has better performance in identifying NPs than the state-of-the-art predictors. Visualization of features using t-distribution random neighbor embedding shows that the NeuroPred-CLQ can clearly distinguish the positive NPs from the negative ones. We believe the NeuroPred-CLQ can facilitate drug development and clinical trial studies to treat neurological disorders.
Collapse
Affiliation(s)
- Shouzhi Chen
- School of Mathematics and System Science, Xinjiang University , Urumqi, China
| | - Qing Li
- School of Mathematics and System Science, Xinjiang University , Urumqi, China
| | - Jianping Zhao
- School of Mathematics and System Science, Xinjiang University , Urumqi, China
| | - Yannan Bin
- School of Computer Science and Technology, Anhui University , Hefei, China
| | - Chunhou Zheng
- School of Mathematics and System Science, Xinjiang University , Urumqi, China
- School of Computer Science and Technology, Anhui University , Hefei, China
| |
Collapse
|
35
|
Ning Q, Li J. DLF-Sul: a multi-module deep learning framework for prediction of S-sulfinylation sites in proteins. Brief Bioinform 2022; 23:6658856. [PMID: 35945138 DOI: 10.1093/bib/bbac323] [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: 04/11/2022] [Revised: 07/16/2022] [Accepted: 07/18/2022] [Indexed: 11/14/2022] Open
Abstract
Protein S-sulfinylation is an important posttranslational modification that regulates a variety of cell and protein functions. This modification has been linked to signal transduction, redox homeostasis and neuronal transmission in studies. Therefore, identification of S-sulfinylation sites is crucial to understanding its structure and function, which is critical in cell biology and human diseases. In this study, we propose a multi-module deep learning framework named DLF-Sul for identification of S-sulfinylation sites in proteins. First, three types of features are extracted including binary encoding, BLOSUM62 and amino acid index. Then, sequential features are further extracted based on these three types of features using bidirectional long short-term memory network. Next, multi-head self-attention mechanism is utilized to filter the effective attribute information, and residual connection helps to reduce information loss. Furthermore, convolutional neural network is employed to extract local deep features information. Finally, fully connected layers acts as classifier that map samples to corresponding label. Performance metrics on independent test set, including sensitivity, specificity, accuracy, Matthews correlation coefficient and area under curve, reach 91.80%, 92.36%, 92.08%, 0.8416 and 96.40%, respectively. The results show that DLF-Sul is an effective tool for predicting S-sulfinylation sites. The source code is available on the website https://github.com/ningq669/DLF-Sul.
Collapse
Affiliation(s)
- Qiao Ning
- Information Science and Technology College, Dalian Maritime University, Dalian 116026, China
| | - Jinmou Li
- Information Science and Technology College, Dalian Maritime University, Dalian 116026, China
| |
Collapse
|
36
|
Al taweraqi N, King RD. Improved prediction of gene expression through integrating cell signalling models with machine learning. BMC Bioinformatics 2022; 23:323. [PMID: 35933367 PMCID: PMC9356471 DOI: 10.1186/s12859-022-04787-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 04/13/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND A key problem in bioinformatics is that of predicting gene expression levels. There are two broad approaches: use of mechanistic models that aim to directly simulate the underlying biology, and use of machine learning (ML) to empirically predict expression levels from descriptors of the experiments. There are advantages and disadvantages to both approaches: mechanistic models more directly reflect the underlying biological causation, but do not directly utilize the available empirical data; while ML methods do not fully utilize existing biological knowledge. RESULTS Here, we investigate overcoming these disadvantages by integrating mechanistic cell signalling models with ML. Our approach to integration is to augment ML with similarity features (attributes) computed from cell signalling models. Seven sets of different similarity feature were generated using graph theory. Each set of features was in turn used to learn multi-target regression models. All the features have significantly improved accuracy over the baseline model - without the similarity features. Finally, the seven multi-target regression models were stacked together to form an overall prediction model that was significantly better than the baseline on 95% of genes on an independent test set. The similarity features enable this stacking model to provide interpretable knowledge about cancer, e.g. the role of ERBB3 in the MCF7 breast cancer cell line. CONCLUSION Integrating mechanistic models as graphs helps to both improve the predictive results of machine learning models, and to provide biological knowledge about genes that can help in building state-of-the-art mechanistic models.
Collapse
Affiliation(s)
- Nada Al taweraqi
- Department of Computer Science, University of Manchester, Manchester, UK
- Department of Computer Science, Taif University, Taif, Saudi Arabia
| | - Ross D. King
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
- Alan Turing Institute, London, UK
| |
Collapse
|
37
|
Ali SD, Tayara H, Chong KT. Interpretable machine learning identification of arginine methylation sites. Comput Biol Med 2022; 147:105767. [DOI: 10.1016/j.compbiomed.2022.105767] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/12/2022] [Accepted: 06/18/2022] [Indexed: 11/25/2022]
|
38
|
Qiu XY, Wu H, Shao J. TALE-cmap: Protein function prediction based on a TALE-based architecture and the structure information from contact map. Comput Biol Med 2022; 149:105938. [DOI: 10.1016/j.compbiomed.2022.105938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 07/26/2022] [Accepted: 08/06/2022] [Indexed: 11/03/2022]
|
39
|
Wang J. Prediction of postoperative recovery in patients with acoustic neuroma using machine learning and SMOTE-ENN techniques. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2022; 19:10407-10423. [PMID: 36032000 DOI: 10.3934/mbe.2022487] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Acoustic neuroma is a common benign tumor that is frequently associated with postoperative complications such as facial nerve dysfunction, which greatly affects the physical and mental health of patients. In this paper, clinical data of patients with acoustic neuroma treated with microsurgery by the same operator at Xiangya Hospital of Central South University from June 2018 to March 2020 are used as the study object. Machine learning and SMOTE-ENN techniques are used to accurately predict postoperative facial nerve function recovery, thus filling a gap in auxiliary diagnosis within the field of facial nerve treatment in acoustic neuroma. First, raw clinical data are processed and dependent variables are identified based on clinical context and data characteristics. Secondly, data balancing is corrected using the SMOTE-ENN technique. Finally, XGBoost is selected to construct a prediction model for patients' postoperative recovery, and is also compared with a total of four machine learning models, LR, SVM, CART, and RF. We find that XGBoost can most accurately predict the postoperative facial nerve function recovery, with a prediction accuracy of 90.0% and an AUC value of 0.90. CART, RF, and XGBoost can further select the more important preoperative indicators and provide therapeutic assistance to physicians, thereby improving the patient's postoperative recovery. The results show that machine learning and SMOTE-ENN techniques can handle complex clinical data and achieve accurate predictions.
Collapse
Affiliation(s)
- Jianing Wang
- School of Mathematics and Statistics, Central South University, Changsha 410083, China
| |
Collapse
|
40
|
Fan Y, Peng B. StackEPI: identification of cell line-specific enhancer-promoter interactions based on stacking ensemble learning. BMC Bioinformatics 2022; 23:272. [PMID: 35820811 PMCID: PMC9277947 DOI: 10.1186/s12859-022-04821-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 07/01/2022] [Indexed: 11/10/2022] Open
Abstract
Background Understanding the regulatory role of enhancer–promoter interactions (EPIs) on specific gene expression in cells contributes to the understanding of gene regulation, cell differentiation, etc., and its identification has been a challenging task. On the one hand, using traditional wet experimental methods to identify EPIs often means a lot of human labor and time costs. On the other hand, although the currently proposed computational methods have good recognition effects, they generally require a long training time. Results In this study, we studied the EPIs of six human cell lines and designed a cell line-specific EPIs prediction method based on a stacking ensemble learning strategy, which has better prediction performance and faster training speed, called StackEPI. Specifically, by combining different encoding schemes and machine learning methods, our prediction method can extract the cell line-specific effective information of enhancer and promoter gene sequences comprehensively and in many directions, and make accurate recognition of cell line-specific EPIs. Ultimately, the source code to implement StackEPI and experimental data involved in the experiment are available at https://github.com/20032303092/StackEPI.git. Conclusions The comparison results show that our model can deliver better performance on the problem of identifying cell line-specific EPIs and outperform other state-of-the-art models. In addition, our model also has a more efficient computation speed. Supplementary Information The online version contains supplementary material available at 10.1186/s12859-022-04821-9.
Collapse
Affiliation(s)
- Yongxian Fan
- School of Computer Science and Information Security, Guilin University of Electronic Technology, Guilin, 541004, China.
| | - Binchao Peng
- School of Computer Science and Information Security, Guilin University of Electronic Technology, Guilin, 541004, China
| |
Collapse
|
41
|
Otović E, Njirjak M, Kalafatovic D, Mauša G. Sequential Properties Representation Scheme for Recurrent Neural Network-Based Prediction of Therapeutic Peptides. J Chem Inf Model 2022; 62:2961-2972. [PMID: 35704881 DOI: 10.1021/acs.jcim.2c00526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The discovery of therapeutic peptides is often accelerated by means of virtual screening supported by machine learning-based predictive models. The predictive performance of such models is sensitive to the choice of data and its representation scheme. While the peptide physicochemical and compositional representations fail to distinguish sequence permutations, the amino acid arrangement within the sequence lacks the important information contained in physicochemical, conformational, topological, and geometrical properties. In this paper, we propose a solution to the identified information gap by implementing a hybrid scheme that complements the best traits from both approaches with the aim of predicting antimicrobial and antiviral activities based on experimental data from DRAMP 2.0, AVPdb, and Uniprot data repositories. Using the Friedman test of statistical significance, we compared our hybrid, sequential properties approach to peptide properties, one-hot vector encoding, and word embedding schemes in the 10-fold cross-validation setting, with respect to the F1 score, Matthews correlation coefficient, geometric mean, recall, and precision evaluation metrics. Moreover, the sequence modeling neural network was employed to gain insight into the synergic effect of both properties- and amino acid order-based predictions. The results suggest that sequential properties significantly (P < 0.01) surpasses the aforementioned state-of-the-art representation schemes. This makes it a strong candidate for increasing the predictive power of screening methods based on machine learning, applicable to any category of peptides.
Collapse
Affiliation(s)
- Erik Otović
- University of Rijeka, Faculty of Engineering, 51000 Rijeka, Croatia
| | - Marko Njirjak
- University of Rijeka, Faculty of Engineering, 51000 Rijeka, Croatia
| | - Daniela Kalafatovic
- University of Rijeka, Department of Biotechnology, 51000 Rijeka, Croatia.,University of Rijeka, Center for Artificial Intelligence and Cybersecurity, 51000 Rijeka, Croatia
| | - Goran Mauša
- University of Rijeka, Faculty of Engineering, 51000 Rijeka, Croatia.,University of Rijeka, Center for Artificial Intelligence and Cybersecurity, 51000 Rijeka, Croatia
| |
Collapse
|
42
|
Jeon YJ, Hasan MM, Park HW, Lee KW, Manavalan B. TACOS: a novel approach for accurate prediction of cell-specific long noncoding RNAs subcellular localization. Brief Bioinform 2022; 23:6618237. [PMID: 35753698 PMCID: PMC9294414 DOI: 10.1093/bib/bbac243] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 11/14/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) are primarily regulated by their cellular localization, which is responsible for their molecular functions, including cell cycle regulation and genome rearrangements. Accurately identifying the subcellular location of lncRNAs from sequence information is crucial for a better understanding of their biological functions and mechanisms. In contrast to traditional experimental methods, bioinformatics or computational methods can be applied for the annotation of lncRNA subcellular locations in humans more effectively. In the past, several machine learning-based methods have been developed to identify lncRNA subcellular localization, but relevant work for identifying cell-specific localization of human lncRNA remains limited. In this study, we present the first application of the tree-based stacking approach, TACOS, which allows users to identify the subcellular localization of human lncRNA in 10 different cell types. Specifically, we conducted comprehensive evaluations of six tree-based classifiers with 10 different feature descriptors, using a newly constructed balanced training dataset for each cell type. Subsequently, the strengths of the AdaBoost baseline models were integrated via a stacking approach, with an appropriate tree-based classifier for the final prediction. TACOS displayed consistent performance in both the cross-validation and independent assessments compared with the other two approaches employed in this study. The user-friendly online TACOS web server can be accessed at https://balalab-skku.org/TACOS.
Collapse
Affiliation(s)
- Young-Jun Jeon
- Department of Integrative Biotechnology, College of Bioengineering and Biotechnology, Sungkyunkwan University, Suwon 16419, Korea
| | - Md Mehedi Hasan
- Tulane Center for Biomedical Informatics and Genomics, Division of Biomedical Informatics and Genomics, John W. Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Hyun Woo Park
- Department of Integrative Biotechnology, College of Bioengineering and Biotechnology, Sungkyunkwan University, Suwon 16419, Korea
| | - Ki Wook Lee
- Department of Integrative Biotechnology, College of Bioengineering and Biotechnology, Sungkyunkwan University, Suwon 16419, Korea
| | - Balachandran Manavalan
- Computational Biology and Bioinformatics laboratory, Department of Integrative Biotechnology, College of Bioengineering and Biotechnology, Sungkyunkwan University, Suwon 16419, Korea
| |
Collapse
|
43
|
Li Y, Li X, Liu Y, Yao Y, Huang G. MPMABP: A CNN and Bi-LSTM-Based Method for Predicting Multi-Activities of Bioactive Peptides. Pharmaceuticals (Basel) 2022; 15:707. [PMID: 35745625 PMCID: PMC9231127 DOI: 10.3390/ph15060707] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/23/2022] [Accepted: 05/30/2022] [Indexed: 12/30/2022] Open
Abstract
Bioactive peptides are typically small functional peptides with 2-20 amino acid residues and play versatile roles in metabolic and biological processes. Bioactive peptides are multi-functional, so it is vastly challenging to accurately detect all their functions simultaneously. We proposed a convolution neural network (CNN) and bi-directional long short-term memory (Bi-LSTM)-based deep learning method (called MPMABP) for recognizing multi-activities of bioactive peptides. The MPMABP stacked five CNNs at different scales, and used the residual network to preserve the information from loss. The empirical results showed that the MPMABP is superior to the state-of-the-art methods. Analysis on the distribution of amino acids indicated that the lysine preferred to appear in the anti-cancer peptide, the leucine in the anti-diabetic peptide, and the proline in the anti-hypertensive peptide. The method and analysis are beneficial to recognize multi-activities of bioactive peptides.
Collapse
Affiliation(s)
- You Li
- School of Electrical Engineering, Shaoyang University, Shaoyang 422000, China; (Y.L.); (X.L.)
| | - Xueyong Li
- School of Electrical Engineering, Shaoyang University, Shaoyang 422000, China; (Y.L.); (X.L.)
| | - Yuewu Liu
- College of Information and Intelligence, Hunan Agricultural University, Changsha 410128, China;
| | - Yuhua Yao
- School of Mathematics and Statistics, Hainan Normal University, Haikou 571158, China;
| | - Guohua Huang
- School of Electrical Engineering, Shaoyang University, Shaoyang 422000, China; (Y.L.); (X.L.)
| |
Collapse
|
44
|
Hosen MF, Mahmud SH, Ahmed K, Chen W, Moni MA, Deng HW, Shoombuatong W, Hasan MM. DeepDNAbP: A deep learning-based hybrid approach to improve the identification of deoxyribonucleic acid-binding proteins. Comput Biol Med 2022; 145:105433. [DOI: 10.1016/j.compbiomed.2022.105433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 03/11/2022] [Accepted: 03/20/2022] [Indexed: 11/03/2022]
|
45
|
Zou H. iAHTP-LH: Integrating Low-Order and High-Order Correlation Information for Identifying Antihypertensive Peptides. Int J Pept Res Ther 2022. [DOI: 10.1007/s10989-022-10414-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
46
|
Yan K, Lv H, Guo Y, Chen Y, Wu H, Liu B. TPpred-ATMV: therapeutic peptide prediction by adaptive multi-view tensor learning model. Bioinformatics 2022; 38:2712-2718. [PMID: 35561206 DOI: 10.1093/bioinformatics/btac200] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 03/17/2022] [Accepted: 04/06/2022] [Indexed: 11/12/2022] Open
Abstract
MOTIVATION Therapeutic peptide prediction is important for the discovery of efficient therapeutic peptides and drug development. Researchers have developed several computational methods to identify different therapeutic peptide types. However, these computational methods focus on identifying some specific types of therapeutic peptides, failing to predict the comprehensive types of therapeutic peptides. Moreover, it is still challenging to utilize different properties to predict the therapeutic peptides. RESULTS In this study, an adaptive multi-view based on the tensor learning framework TPpred-ATMV is proposed for predicting different types of therapeutic peptides. TPpred-ATMV constructs the class and probability information based on various sequence features. We constructed the latent subspace among the multi-view features and constructed an auto-weighted multi-view tensor learning model to utilize the high correlation based on the multi-view features. Experimental results showed that the TPpred-ATMV is better than or highly comparable with the other state-of-the-art methods for predicting eight types of therapeutic peptides. AVAILABILITY AND IMPLEMENTATION The code of TPpred-ATMV is accessed at: https://github.com/cokeyk/TPpred-ATMV. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Ke Yan
- School of Computer Science and Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Hongwu Lv
- School of Computer Science and Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Yichen Guo
- School of Computer Science and Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Yongyong Chen
- Bio-Computing Research Center, Harbin Institute of Technology, Shenzhen 518055, China
| | - Hao Wu
- School of Computer Science and Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Bin Liu
- School of Computer Science and Technology, Beijing Institute of Technology, Beijing 100081, China
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
47
|
Banna HU, Zanabli A, McMillan B, Lehmann M, Gupta S, Gerbo M, Palko J. Evaluation of machine learning algorithms for trabeculectomy outcome prediction in patients with glaucoma. Sci Rep 2022; 12:2473. [PMID: 35169235 PMCID: PMC8847459 DOI: 10.1038/s41598-022-06438-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 01/18/2022] [Indexed: 02/04/2023] Open
Abstract
The purpose of this study was to evaluate the performance of machine learning algorithms to predict trabeculectomy surgical outcomes. Preoperative systemic, demographic and ocular data from consecutive trabeculectomy surgeries from a single academic institution between January 2014 and December 2018 were incorporated into models using random forest, support vector machine, artificial neural networks and multivariable logistic regression. Mean area under the receiver operating characteristic curve (AUC) and accuracy were used to evaluate the discrimination of each model to predict complete success of trabeculectomy surgery at 1 year. The top performing model was optimized using recursive feature selection and hyperparameter tuning. Calibration and net benefit of the final models were assessed. Among the 230 trabeculectomy surgeries performed on 184 patients, 104 (45.2%) were classified as complete success. Random forest was found to be the top performing model with an accuracy of 0.68 and AUC of 0.74 using 5-fold cross-validation to evaluate the final optimized model. These results provide evidence that machine learning models offer value in predicting trabeculectomy outcomes in patients with refractory glaucoma.
Collapse
Affiliation(s)
- Hasan Ul Banna
- Department of Ophthalmology and Visual Sciences, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Ahmed Zanabli
- Department of Ophthalmology and Visual Sciences, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Brian McMillan
- Department of Ophthalmology and Visual Sciences, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Maria Lehmann
- Department of Ophthalmology and Visual Sciences, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Sumeet Gupta
- Department of Ophthalmology and Visual Sciences, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Michael Gerbo
- Department of Ophthalmology and Visual Sciences, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Joel Palko
- Department of Ophthalmology and Visual Sciences, West Virginia University School of Medicine, Morgantown, WV, 26506, USA.
| |
Collapse
|
48
|
Kabir M, Nantasenamat C, Kanthawong S, Charoenkwan P, Shoombuatong W. Large-scale comparative review and assessment of computational methods for phage virion proteins identification. EXCLI JOURNAL 2022; 21:11-29. [PMID: 35145365 PMCID: PMC8822302 DOI: 10.17179/excli2021-4411] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 11/29/2021] [Indexed: 12/11/2022]
Abstract
Phage virion proteins (PVPs) are effective at recognizing and binding to host cell receptors while having no deleterious effects on human or animal cells. Understanding their functional mechanisms is regarded as a critical goal that will aid in rational antibacterial drug discovery and development. Although high-throughput experimental methods for identifying PVPs are considered the gold standard for exploring crucial PVP features, these procedures are frequently time-consuming and labor-intensive. Thusfar, more than ten sequence-based predictors have been established for the in silico identification of PVPs in conjunction with traditional experimental approaches. As a result, a revised and more thorough assessment is extremely desirable. With this purpose in mind, we first conduct a thorough survey and evaluation of a vast array of 13 state-of-the-art PVP predictors. Among these PVP predictors, they can be classified into three groups according to the types of machine learning (ML) algorithms employed (i.e. traditional ML-based methods, ensemble-based methods and deep learning-based methods). Subsequently, we explored which factors are important for building more accurate and stable predictors and this included training/independent datasets, feature encoding algorithms, feature selection methods, core algorithms, performance evaluation metrics/strategies and web servers. Finally, we provide insights and future perspectives for the design and development of new and more effective computational approaches for the detection and characterization of PVPs.
Collapse
Affiliation(s)
- Muhammad Kabir
- School of Systems and Technology, Department of Computer Science, University of Management and Technology, Lahore, Pakistan, 54770
| | - Chanin Nantasenamat
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand, 10700
| | - Sakawrat Kanthawong
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand, 40002
| | - Phasit Charoenkwan
- Modern Management and Information Technology, College of Arts, Media and Technology, Chiang Mai University, Chiang Mai, Thailand, 50200
| | - Watshara Shoombuatong
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand, 10700
| |
Collapse
|
49
|
Han YM, Yang H, Huang QL, Sun ZJ, Li ML, Zhang JB, Deng KJ, Chen S, Lin H. Risk prediction of diabetes and pre-diabetes based on physical examination data. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2022; 19:3597-3608. [PMID: 35341266 DOI: 10.3934/mbe.2022166] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Diabetes is a metabolic disorder caused by insufficient insulin secretion and insulin secretion disorders. From health to diabetes, there are generally three stages: health, pre-diabetes and type 2 diabetes. Early diagnosis of diabetes is the most effective way to prevent and control diabetes and its complications. In this work, we collected the physical examination data from Beijing Physical Examination Center from January 2006 to December 2017, and divided the population into three groups according to the WHO (1999) Diabetes Diagnostic Standards: normal fasting plasma glucose (NFG) (FPG < 6.1 mmol/L), mildly impaired fasting plasma glucose (IFG) (6.1 mmol/L ≤ FPG < 7.0 mmol/L) and type 2 diabetes (T2DM) (FPG > 7.0 mmol/L). Finally, we obtained1,221,598 NFG samples, 285,965 IFG samples and 387,076 T2DM samples, with a total of 15 physical examination indexes. Furthermore, taking eXtreme Gradient Boosting (XGBoost), random forest (RF), Logistic Regression (LR), and Fully connected neural network (FCN) as classifiers, four models were constructed to distinguish NFG, IFG and T2DM. The comparison results show that XGBoost has the best performance, with AUC (macro) of 0.7874 and AUC (micro) of 0.8633. In addition, based on the XGBoost classifier, three binary classification models were also established to discriminate NFG from IFG, NFG from T2DM, IFG from T2DM. On the independent dataset, the AUCs were 0.7808, 0.8687, 0.7067, respectively. Finally, we analyzed the importance of the features and identified the risk factors associated with diabetes.
Collapse
Affiliation(s)
- Yu-Mei Han
- Beijing Physical Examination Center, Beijing, China
| | - Hui Yang
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Qin-Lai Huang
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Zi-Jie Sun
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | | | | | - Ke-Jun Deng
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Shuo Chen
- Beijing Physical Examination Center, Beijing, China
| | - Hao Lin
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China
| |
Collapse
|
50
|
Zulfiqar H, Huang QL, Lv H, Sun ZJ, Dao FY, Lin H. Deep-4mCGP: A Deep Learning Approach to Predict 4mC Sites in Geobacter pickeringii by Using Correlation-Based Feature Selection Technique. Int J Mol Sci 2022; 23:1251. [PMID: 35163174 PMCID: PMC8836036 DOI: 10.3390/ijms23031251] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 12/15/2022] Open
Abstract
4mC is a type of DNA alteration that has the ability to synchronize multiple biological movements, for example, DNA replication, gene expressions, and transcriptional regulations. Accurate prediction of 4mC sites can provide exact information to their hereditary functions. The purpose of this study was to establish a robust deep learning model to recognize 4mC sites in Geobacter pickeringii. In the anticipated model, two kinds of feature descriptors, namely, binary and k-mer composition were used to encode the DNA sequences of Geobacter pickeringii. The obtained features from their fusion were optimized by using correlation and gradient-boosting decision tree (GBDT)-based algorithm with incremental feature selection (IFS) method. Then, these optimized features were inserted into 1D convolutional neural network (CNN) to classify 4mC sites from non-4mC sites in Geobacter pickeringii. The performance of the anticipated model on independent data exhibited an accuracy of 0.868, which was 4.2% higher than the existing model.
Collapse
Affiliation(s)
| | | | | | | | | | - Hao Lin
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China; (H.Z.); (Q.-L.H.); (H.L.); (Z.-J.S.); (F.-Y.D.)
| |
Collapse
|