1
|
Fang Y, Fan D, Feng B, Zhu Y, Xie R, Tan X, Liu Q, Dong J, Zeng W. Harnessing advanced computational approaches to design novel antimicrobial peptides against intracellular bacterial infections. Bioact Mater 2025; 50:510-524. [PMID: 40342489 PMCID: PMC12059401 DOI: 10.1016/j.bioactmat.2025.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 04/06/2025] [Accepted: 04/15/2025] [Indexed: 05/11/2025] Open
Abstract
Intracellular bacterial infections pose a significant challenge to current therapeutic strategies due to the limited penetration of antibiotics through host cell membranes. This study presents a novel computational framework for efficiently screening candidate peptides against these infections. The proposed strategy comprehensively evaluates the essential properties for the clinical application of candidate peptides, including antimicrobial activity, permeation efficiency, and biocompatibility, while also taking into account the speed and reliability of the screening process. A combination of multiple AI-based activity prediction models allows for a thorough assessment of sequences in the cell-penetrating peptides (CPPs) database and quickly identifies candidate peptides with target properties. On this basis, the CPP microscopic dynamics research system was constructed. Exploration of the mechanism of action at the atomic level provides strong support for the discovery of promising candidate peptides. Promising candidates are subsequently validated through in vitro and in vivo experiments. Finally, Crot-1 was rapidly identified from the CPPsite 2.0 database. Crot-1 effectively eradicated intracellular MRSA, demonstrating significantly greater efficacy than vancomycin. Moreover, it exhibited no apparent cytotoxicity to host cells, highlighting its potential for clinical application. This work offers a promising new avenue for developing novel antimicrobial materials to combat intracellular bacterial infections.
Collapse
Affiliation(s)
- Yanpeng Fang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410083, PR China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha 410078, PR China
| | - Duoyang Fan
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410083, PR China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha 410078, PR China
| | - Bin Feng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410083, PR China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha 410078, PR China
| | - Yingli Zhu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410083, PR China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha 410078, PR China
| | - Ruyan Xie
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410083, PR China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha 410078, PR China
| | - Xiaorong Tan
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410083, PR China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha 410078, PR China
| | - Qianhui Liu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410083, PR China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha 410078, PR China
| | - Jie Dong
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410083, PR China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha 410078, PR China
| | - Wenbin Zeng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410083, PR China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha 410078, PR China
| |
Collapse
|
2
|
Zhang L, Xiong S, Xu L, Liang J, Zhao X, Zhang H, Tan X. Leveraging protein language models for robust antimicrobial peptide detection. Methods 2025; 238:19-26. [PMID: 40049432 DOI: 10.1016/j.ymeth.2025.03.002] [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: 01/15/2025] [Revised: 02/09/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025] Open
Abstract
Antimicrobial peptides (AMPs) are promising candidates for addressing the global challenge of antibiotic resistance due to their broad-spectrum antimicrobial properties. Traditional AMP identification methods, while effective, are labor-intensive and time-consuming. Recent advancements in deep learning and large language models (LLMs), especially protein language models (PLMs) present a transformative approach for AMP prediction. In this study, we propose PLAPD, a novel framework leveraging a pre-trained ESM2 protein language model for AMP classification. Besides, PLAPD combines local feature extraction via convolutional layers and global feature extraction with a residual Transformer module. We benchmarked PLAPD against state-of-the-art AMP prediction models using a dataset comprising 8,268 peptide sequences, achieving superior performance in Accuracy (0.87), Precision (0.9359), Specificity (0.9456), MCC (0.7486), and AUC (0.9225). The results highlight the potential of PLAPD as a high-throughput and accurate tool for AMP discovery.
Collapse
Affiliation(s)
- Lichao Zhang
- School of Intelligent Manufacturing and Equipment, Shenzhen Institute of Information Technology, Shenzhen 518172, China.
| | - Shuwen Xiong
- Faculty of Applied Sciences, Macao Polytechnic University, R. de Luís Gonzaga Gomes, Macao 999078, China
| | - Lei Xu
- School of Electronic and Communication Engineering, Shenzhen Polytechnic University, Shenzhen 518055, China
| | - Junwei Liang
- School of Computer and Software, Shenzhen Institute of Information Technology, Shenzhen 518172, China
| | - Xuehua Zhao
- School of Digital Media, Shenzhen Institute of Information Technology, Shenzhen 518172, China
| | - Honglai Zhang
- Thyroid Surgery Department, The Affiliated Hospital of Qingdao University, Qingdao 266035, China
| | - Xu Tan
- School of Artificial Intelligence, Shenzhen Institute of Information Technology, Shenzhen 518172, China.
| |
Collapse
|
3
|
Olayo-Alarcon R, Amstalden MK, Zannoni A, Bajramovic M, Sharma CM, Brochado AR, Rezaei M, Müller CL. Pre-trained molecular representations enable antimicrobial discovery. Nat Commun 2025; 16:3420. [PMID: 40210659 PMCID: PMC11986102 DOI: 10.1038/s41467-025-58804-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 04/02/2025] [Indexed: 04/12/2025] Open
Abstract
The rise in antimicrobial resistance poses a worldwide threat, reducing the efficacy of common antibiotics. Determining the antimicrobial activity of new chemical compounds through experimental methods remains time-consuming and costly. While compound-centric deep learning models promise to accelerate this search and prioritization process, current strategies require large amounts of custom training data. Here, we introduce a lightweight computational strategy for antimicrobial discovery that builds on MolE (Molecular representation through redundancy reduced Embedding), a self-supervised deep learning framework that leverages unlabeled chemical structures to learn task-independent molecular representations. By combining MolE representation learning with available, experimentally validated compound-bacteria activity data, we design a general predictive model that enables assessing compounds with respect to their antimicrobial potential. Our model correctly identifies recent growth-inhibitory compounds that are structurally distinct from current antibiotics. Using this approach, we discover de novo, and experimentally confirm, three human-targeted drugs as growth inhibitors of Staphylococcus aureus. This framework offers a viable, cost-effective strategy to accelerate antibiotic discovery.
Collapse
Affiliation(s)
- Roberto Olayo-Alarcon
- Department of Statistics, Ludwig-Maximilians-Universität München, Munich, Germany.
- Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany.
| | - Martin K Amstalden
- Department of Microbiology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Annamaria Zannoni
- Department of Molecular Infection Biology II, Institute of Molecular Infection Biology (IMIB), Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Medina Bajramovic
- Department of Statistics, Ludwig-Maximilians-Universität München, Munich, Germany
- Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
| | - Cynthia M Sharma
- Department of Molecular Infection Biology II, Institute of Molecular Infection Biology (IMIB), Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Ana Rita Brochado
- Department of Microbiology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), University of Tübingen, Tübingen, Germany
- Cluster of Excellence 'Controlling Microbes to Fight Infections' (CMFI), University of Tübingen, Tübingen, Germany
| | - Mina Rezaei
- Department of Statistics, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Christian L Müller
- Department of Statistics, Ludwig-Maximilians-Universität München, Munich, Germany.
- Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany.
- Center for Computational Mathematics, Flatiron Institute, New York, USA.
| |
Collapse
|
4
|
Khalaf WS, Morgan RN, Elkhatib WF. Clinical microbiology and artificial intelligence: Different applications, challenges, and future prospects. J Microbiol Methods 2025; 232-234:107125. [PMID: 40188989 DOI: 10.1016/j.mimet.2025.107125] [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: 11/07/2024] [Revised: 03/10/2025] [Accepted: 04/03/2025] [Indexed: 04/10/2025]
Abstract
Conventional clinical microbiological techniques are enhanced by the introduction of artificial intelligence (AI). Comprehensive data processing and analysis enabled the development of curated datasets that has been effectively used in training different AI algorithms. Recently, a number of machine learning (ML) and deep learning (DL) algorithms are developed and evaluated using diverse microbiological datasets. These datasets included spectral analysis (Raman and MALDI-TOF spectroscopy), microscopic images (Gram and acid fast stains), and genomic and protein sequences (whole genome sequencing (WGS) and protein data banks (PDBs)). The primary objective of these algorithms is to minimize the time, effort, and expenses linked to conventional analytical methods. Furthermore, AI algorithms are incorporated with quantitative structure-activity relationship (QSAR) models to predict novel antimicrobial agents that address the continuing surge of antimicrobial resistance. During the COVID-19 pandemic, AI algorithms played a crucial role in vaccine developments and the discovery of new antiviral agents, and introduced potential drug candidates via drug repurposing. However, despite their significant benefits, the implementation of AI encounters various challenges, including ethical considerations, the potential for bias, and errors related to data training. This review seeks to provide an overview of the most recent applications of artificial intelligence in clinical microbiology, with the intention of educating a wider audience of clinical practitioners regarding the current uses of machine learning algorithms and encouraging their implementation. Furthermore, it will discuss the challenges related to the incorporation of AI into clinical microbiology laboratories and examine future opportunities for AI within the realm of infectious disease epidemiology.
Collapse
Affiliation(s)
- Wafaa S Khalaf
- Department of Microbiology and Immunology, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr city, Cairo 11751, Egypt.
| | - Radwa N Morgan
- National Centre for Radiation Research and Technology (NCRRT), Drug Radiation Research Department, Egyptian Atomic Energy Authority (EAEA), Cairo 11787, Egypt.
| | - Walid F Elkhatib
- Department of Microbiology & Immunology, Faculty of Pharmacy, Galala University, New Galala City, Suez, Egypt; Microbiology and Immunology Department, Faculty of Pharmacy, Ain Shams University, African Union Organization St., Abbassia, Cairo 11566, Egypt.
| |
Collapse
|
5
|
Miao H, Wang L, Wu Q, Huang Z. Antimicrobial Peptides: Mechanism, Expressions, and Optimization Strategies. Probiotics Antimicrob Proteins 2025; 17:857-872. [PMID: 39528853 DOI: 10.1007/s12602-024-10391-4] [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] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
Antimicrobial peptides (AMPs) are favoured because of their broad-spectrum antimicrobial properties and because they do not easily develop microbial resistance. However, the low yield and difficult extraction processes of AMPs have become bottlenecks in large-scale industrial applications and scientific research. Microbial recombinant production may be the most economical and effective method of obtaining AMPs in large quantities. In this paper, we review the mechanism, summarize the current status of microbial recombinant production, and focus on strategies to improve the yield and activity of AMPs, in order to provide a reference for their large-scale production.
Collapse
Affiliation(s)
- Huabiao Miao
- School of Life Science, Yunnan Normal University, Kunming, 650500, China
- Engineering Research Center for Efficient Utilization of Characteristic Biological Resources in Yunnan, Ministry of Education, Kunming, 650500, China
- Key Laboratory of Yunnan for Biomass Energy and Biotechnology of Environment, Kunming, 650500, China
| | - Lu Wang
- School of Life Science, Yunnan Normal University, Kunming, 650500, China
| | - Qian Wu
- School of Life Science, Yunnan Normal University, Kunming, 650500, China
- Engineering Research Center for Efficient Utilization of Characteristic Biological Resources in Yunnan, Ministry of Education, Kunming, 650500, China
- Key Laboratory of Yunnan for Biomass Energy and Biotechnology of Environment, Kunming, 650500, China
| | - Zunxi Huang
- School of Life Science, Yunnan Normal University, Kunming, 650500, China.
- Engineering Research Center for Efficient Utilization of Characteristic Biological Resources in Yunnan, Ministry of Education, Kunming, 650500, China.
- Key Laboratory of Yunnan for Biomass Energy and Biotechnology of Environment, Kunming, 650500, China.
| |
Collapse
|
6
|
Sun S. Progress in the Identification and Design of Novel Antimicrobial Peptides Against Pathogenic Microorganisms. Probiotics Antimicrob Proteins 2025; 17:918-936. [PMID: 39557756 PMCID: PMC11925980 DOI: 10.1007/s12602-024-10402-4] [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] [Accepted: 11/11/2024] [Indexed: 11/20/2024]
Abstract
The occurrence and spread of antimicrobial resistance (AMR) pose a looming threat to human health around the world. Novel antibiotics are urgently needed to address the AMR crisis. In recent years, antimicrobial peptides (AMPs) have gained increasing attention as potential alternatives to conventional antibiotics due to their abundant sources, structural diversity, broad-spectrum antimicrobial activity, and ease of production. Given its significance, there has been a tremendous advancement in the research and development of AMPs. Numerous AMPs have been identified from various natural sources (e.g., plant, animal, human, microorganism) based on either well-established isolation or bioinformatic pipelines. Moreover, computer-assisted strategies (e.g., machine learning (ML) and deep learning (DL)) have emerged as a powerful and promising technology for the accurate prediction and design of new AMPs. It may overcome some of the shortcomings of traditional antibiotic discovery and contribute to the rapid development and translation of AMPs. In these cases, this review aims to appraise the latest advances in identifying and designing AMPs and their significant antimicrobial activities against a wide range of bacterial pathogens. The review also highlights the critical challenges in discovering and applying AMPs.
Collapse
Affiliation(s)
- Shengwei Sun
- School of Engineering Sciences in Chemistry, Biotechnology and Health, Department of Fibre and Polymer Technology, KTH Royal Institute of Technology, 100 44, Stockholm, Sweden.
- School of Engineering Sciences in Chemistry, Biotechnology and Health, Science for Life Laboratory, Tomtebodavägen 23, 171 65, Solna, Sweden.
| |
Collapse
|
7
|
Li C, Sutherland D, Salehi A, Richter A, Lin D, Aninta SI, Ebrahimikondori H, Yanai A, Coombe L, Warren RL, Kotkoff M, Hoang LMN, Helbing CC, Birol I. Mining the UniProtKB/Swiss-Prot database for antimicrobial peptides. Protein Sci 2025; 34:e70083. [PMID: 40100125 PMCID: PMC11917140 DOI: 10.1002/pro.70083] [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: 10/11/2024] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 03/20/2025]
Abstract
The ever-growing global health threat of antibiotic resistance is compelling researchers to explore alternatives to conventional antibiotics. Antimicrobial peptides (AMPs) are emerging as a promising solution to fill this need. Naturally occurring AMPs are produced by all forms of life as part of the innate immune system. High-throughput bioinformatics tools have enabled fast and large-scale discovery of AMPs from genomic, transcriptomic, and proteomic resources of selected organisms. Public protein sequence databases, comprising over 200 million records and growing, serve as comprehensive compendia of sequences from a broad range of source organisms. Yet, large-scale in silico probing of those databases for novel AMP discovery using modern deep learning techniques has rarely been reported. In the present study, we propose an AMP mining workflow to predict novel AMPs from the UniProtKB/Swiss-Prot database using the AMP prediction tool, AMPlify, as its discovery engine. Using this workflow, we identified 8008 novel putative AMPs from all eukaryotic sequences in the database. Focusing on the practical use of AMPs as suitable antimicrobial agents with applications in the poultry industry, we prioritized 40 of those AMPs based on their similarities to known chicken AMPs in predicted structures. In our tests, 13 out of the 38 successfully synthesized peptides showed antimicrobial activity against Escherichia coli and/or Staphylococcus aureus. AMPlify and the companion scripts supporting the AMP mining workflow presented herein are publicly available at https://github.com/bcgsc/AMPlify.
Collapse
Affiliation(s)
- Chenkai Li
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
- Bioinformatics Graduate ProgramUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Darcy Sutherland
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
- Public Health LaboratoryBritish Columbia Centre for Disease ControlVancouverBritish ColumbiaCanada
- Department of Pathology and Laboratory MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Ali Salehi
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
- Public Health LaboratoryBritish Columbia Centre for Disease ControlVancouverBritish ColumbiaCanada
| | - Amelia Richter
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
- Public Health LaboratoryBritish Columbia Centre for Disease ControlVancouverBritish ColumbiaCanada
| | - Diana Lin
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
| | - Sambina Islam Aninta
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
| | - Hossein Ebrahimikondori
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
- Bioinformatics Graduate ProgramUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Anat Yanai
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
- Public Health LaboratoryBritish Columbia Centre for Disease ControlVancouverBritish ColumbiaCanada
| | - Lauren Coombe
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
| | - René L. Warren
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
| | - Monica Kotkoff
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
| | - Linda M. N. Hoang
- Public Health LaboratoryBritish Columbia Centre for Disease ControlVancouverBritish ColumbiaCanada
- Department of Pathology and Laboratory MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Caren C. Helbing
- Department of Biochemistry and MicrobiologyUniversity of VictoriaVictoriaBritish ColumbiaCanada
| | - Inanc Birol
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
- Public Health LaboratoryBritish Columbia Centre for Disease ControlVancouverBritish ColumbiaCanada
- Department of Pathology and Laboratory MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
- Department of Medical GeneticsUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| |
Collapse
|
8
|
Zhenghui L, Wenxing H, Yan W, Jihong Z, Xiaojun X, Lixin G, Mengshan L. Ensemble learning based on bi-directional gated recurrent unit and convolutional neural network with word embedding module for bioactive peptide prediction. Food Chem 2025; 468:142464. [PMID: 39675273 DOI: 10.1016/j.foodchem.2024.142464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 11/12/2024] [Accepted: 12/11/2024] [Indexed: 12/17/2024]
Abstract
Bioactive peptides, as small protein fragments, are essential mediators of diverse physiological activities, such as antimicrobial, anti-inflammatory, anticancer, antioxidant, and immunomodulatory functions. Despite their substantial potential in pharmaceuticals and the food industry, conventional methods for peptide classification and activity prediction are limited by high costs, time-intensive procedures, and extensive data processing requirements. Here, we present BioPepPred-DLEmb, a novel computational model integrating Convolutional Neural Networks (CNNs) and Bidirectional Gated Recurrent Units (BiGRUs), augmented with natural language processing to encode amino acids into information-dense vectors. Evaluated across nine bioactive peptide datasets, BioPepPred-DLEmb demonstrates superior predictive accuracy (0.909) and sensitivity (0.911) compared to traditional methods. Through UMAP visualization and Kplogo analysis, the model effectively differentiates peptide activity states and identifies key biomarkers. The predicted antimicrobial peptides (Pred-AMPs) exhibit potent efficacy in vitro, achieving low micromolar inhibitory concentrations (2-16 μmol/L) against pathogens such as Escherichia coli and Acinetobacter baumannii. These findings establish a robust foundation for bioactive peptide development, with implications for advancements in precision medicine, personalized therapies, and functional food innovations.
Collapse
Affiliation(s)
- Lai Zhenghui
- College of Physics and Electronic Information, Gannan Normal University, Ganzhou 341000, Jiangxi, China
| | - Hu Wenxing
- College of Physics and Electronic Information, Gannan Normal University, Ganzhou 341000, Jiangxi, China
| | - Wu Yan
- College of Physics and Electronic Information, Gannan Normal University, Ganzhou 341000, Jiangxi, China
| | - Zhu Jihong
- College of Physics and Electronic Information, Gannan Normal University, Ganzhou 341000, Jiangxi, China
| | - Xie Xiaojun
- College of Physics and Electronic Information, Gannan Normal University, Ganzhou 341000, Jiangxi, China
| | - Guan Lixin
- College of Physics and Electronic Information, Gannan Normal University, Ganzhou 341000, Jiangxi, China
| | - Li Mengshan
- College of Physics and Electronic Information, Gannan Normal University, Ganzhou 341000, Jiangxi, China.
| |
Collapse
|
9
|
Dong R, Liu R, Liu Z, Liu Y, Zhao G, Li H, Hou S, Ma X, Kang H, Liu J, Guo F, Zhao P, Wang J, Wang C, Wu X, Ye S, Zhu C. Exploring the repository of de novo-designed bifunctional antimicrobial peptides through deep learning. eLife 2025; 13:RP97330. [PMID: 40079572 PMCID: PMC11906162 DOI: 10.7554/elife.97330] [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] [Indexed: 03/15/2025] Open
Abstract
Antimicrobial peptides (AMPs) are attractive candidates to combat antibiotic resistance for their capability to target biomembranes and restrict a wide range of pathogens. It is a daunting challenge to discover novel AMPs due to their sparse distributions in a vast peptide universe, especially for peptides that demonstrate potencies for both bacterial membranes and viral envelopes. Here, we establish a de novo AMP design framework by bridging a deep generative module and a graph-encoding activity regressor. The generative module learns hidden 'grammars' of AMP features and produces candidates sequentially pass antimicrobial predictor and antiviral classifiers. We discovered 16 bifunctional AMPs and experimentally validated their abilities to inhibit a spectrum of pathogens in vitro and in animal models. Notably, P076 is a highly potent bactericide with the minimal inhibitory concentration of 0.21 μM against multidrug-resistant Acinetobacter baumannii, while P002 broadly inhibits five enveloped viruses. Our study provides feasible means to uncover the sequences that simultaneously encode antimicrobial and antiviral activities, thus bolstering the function spectra of AMPs to combat a wide range of drug-resistant infections.
Collapse
Affiliation(s)
- Ruihan Dong
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life Sciences, Faculty of Medicine, Tianjin UniversityTianjinChina
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking UniversityBeijingChina
| | - Rongrong Liu
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical UniversityShaanxiChina
| | - Ziyu Liu
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical UniversityShaanxiChina
| | - Yangang Liu
- Department of Microbiology, Second Military Medical UniversityShanghaiChina
| | - Gaomei Zhao
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Third Military Medical University (Army Medical University)ChongqingChina
| | - Honglei Li
- Tianjin Cancer Hospital Airport HospitalTianjinChina
| | - Shiyuan Hou
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical UniversityShaanxiChina
| | - Xiaohan Ma
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical UniversityShaanxiChina
| | - Huarui Kang
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical UniversityShaanxiChina
| | - Jing Liu
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical UniversityShaanxiChina
| | - Fei Guo
- School of Computer Science and Engineering, Central South UniversityChangshaChina
| | - Ping Zhao
- Department of Microbiology, Second Military Medical UniversityShanghaiChina
| | - Junping Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Third Military Medical University (Army Medical University)ChongqingChina
| | - Cheng Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Third Military Medical University (Army Medical University)ChongqingChina
| | - Xingan Wu
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical UniversityShaanxiChina
| | - Sheng Ye
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life Sciences, Faculty of Medicine, Tianjin UniversityTianjinChina
| | - Cheng Zhu
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life Sciences, Faculty of Medicine, Tianjin UniversityTianjinChina
| |
Collapse
|
10
|
Wang J, Feng J, Kang Y, Pan P, Ge J, Wang Y, Wang M, Wu Z, Zhang X, Yu J, Zhang X, Wang T, Wen L, Yan G, Deng Y, Shi H, Hsieh CY, Jiang Z, Hou T. Discovery of antimicrobial peptides with notable antibacterial potency by an LLM-based foundation model. SCIENCE ADVANCES 2025; 11:eads8932. [PMID: 40043127 DOI: 10.1126/sciadv.ads8932] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 01/29/2025] [Indexed: 05/13/2025]
Abstract
Large language models (LLMs) have shown remarkable advancements in chemistry and biomedical research, acting as versatile foundation models for various tasks. We introduce AMP-Designer, an LLM-based approach, for swiftly designing antimicrobial peptides (AMPs) with desired properties. Within 11 days, AMP-Designer achieved the de novo design of 18 AMPs with broad-spectrum activity against Gram-negative bacteria. In vitro validation revealed a 94.4% success rate, with two candidates demonstrating exceptional antibacterial efficacy, minimal hemotoxicity, stability in human plasma, and low potential to induce resistance, as evidenced by significant bacterial load reduction in murine lung infection experiments. The entire process, from design to validation, concluded in 48 days. AMP-Designer excels in creating AMPs targeting specific strains despite limited data availability, with a top candidate displaying a minimum inhibitory concentration of 2.0 micrograms per milliliter against Propionibacterium acnes. Integrating advanced machine learning techniques, AMP-Designer demonstrates remarkable efficiency, paving the way for innovative solutions to antibiotic resistance.
Collapse
Affiliation(s)
- Jike Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
- CarbonSilicon AI Technology Co. Ltd., Hangzhou 310018, Zhejiang, China
| | - Jianwen Feng
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yu Kang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Peichen Pan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Jingxuan Ge
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Yan Wang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Mingyang Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Zhenxing Wu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Xingcai Zhang
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
- World Tea Organization, Cambridge, MA 02139, USA
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA
| | - Jiameng Yu
- Graduate School, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong, China
| | - Xujun Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Tianyue Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Lirong Wen
- School of Pharmaceutical Sciences, Dali University, Dali 671003, Yunan, China
| | - Guangning Yan
- Department of Pathology, General Hospital of Southern Theatre Command, Guangzhou 510010, China
| | - Yafeng Deng
- CarbonSilicon AI Technology Co. Ltd., Hangzhou 310018, Zhejiang, China
| | - Hui Shi
- CarbonSilicon AI Technology Co. Ltd., Hangzhou 310018, Zhejiang, China
| | - Chang-Yu Hsieh
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Zhihui Jiang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
- Graduate School, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong, China
- Department of Pharmacy, General Hospital of Southern Theatre Command, Guangzhou 510010, Guangdong, China
| | - Tingjun Hou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| |
Collapse
|
11
|
Cao J, Zhou W, Yu Q, Ji J, Zhang J, He S, Zhu Z. MDTL-ACP: Anticancer Peptides Prediction Based on Multi-Domain Transfer Learning. IEEE J Biomed Health Inform 2025; 29:1714-1725. [PMID: 38147420 DOI: 10.1109/jbhi.2023.3347138] [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: 12/28/2023]
Abstract
Anticancer peptides (ACPs) have emerged as one of the most promising therapeutic agents for cancer treatment. They are bioactive peptides featuring broad-spectrum activity and low drug-resistance. The discovery of ACPs via traditional biochemical methods is laborious and costly. Accordingly, various computational methods have been developed to facilitate the discovery of ACPs. However, the data resources and knowledge of ACPs are still very scarce, and only a few of them are clinically verified, which limits the competence of computational methods. To address this issue, in this article, we propose an ACP prediction model based on multi-domain transfer learning, namely MDTL-ACP, to discriminate novel ACPs from plentiful inactive peptides. In particular, we collect abundant antimicrobial peptides (AMPs) from four well-studied peptide domains and extract their inherent features as the input of MDTL-ACP. The features learned from multiple source domains of AMPs are then transferred into the target prediction task of ACPs via artificial neural network-based shared-extractor and task-specific classifiers in MDTL-ACP. The knowledge captured in the transferred features enhances the prediction of ACPs in the target domain. Experimental results demonstrate that MDTL-ACP can outperform the traditional and state-of-the-art ACP prediction methods.
Collapse
|
12
|
Osiro KO, Gil-Ley A, Fernandes FC, de Oliveira KBS, de la Fuente-Nunez C, Franco OL. Paving the way for new antimicrobial peptides through molecular de-extinction. MICROBIAL CELL (GRAZ, AUSTRIA) 2025; 12:1-8. [PMID: 40012704 PMCID: PMC11853161 DOI: 10.15698/mic2025.02.841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 01/02/2025] [Accepted: 01/07/2025] [Indexed: 02/28/2025]
Abstract
Molecular de-extinction has emerged as a novel strategy for studying biological molecules throughout evolutionary history. Among the myriad possibilities offered by ancient genomes and proteomes, antimicrobial peptides (AMPs) stand out as particularly promising alternatives to traditional antibiotics. Various strategies, including software tools and advanced deep learning models, have been used to mine these host defense peptides. For example, computational analysis of disulfide bond patterns has led to the identification of six previously uncharacterized β-defensins in extinct and critically endangered species. Additionally, artificial intelligence and machine learning have been utilized to uncover ancient antibiotics, revealing numerous candidates, including mammuthusin, and elephasin, which display inhibitory effects toward pathogens in vitro and in vivo. These innovations promise to discover novel antibiotics and deepen our insight into evolutionary processes.
Collapse
Affiliation(s)
- Karen O Osiro
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília70790-160Brazil
| | - Abel Gil-Ley
- S-Inova Biotech, Pós-graduação em Biotecnologia, Universidade Católica Dom BoscoCampo Grande, Mato Grosso do SulBrazil
| | - Fabiano C Fernandes
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília70790-160Brazil
- Departamento de Ciência da Computação, Instituto Federal de Brasília, Campus Taguatinga, Brasília, Brazil
| | - Kamila B S de Oliveira
- S-Inova Biotech, Pós-graduação em Biotecnologia, Universidade Católica Dom BoscoCampo Grande, Mato Grosso do SulBrazil
| | - Cesar de la Fuente-Nunez
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of PennsylvaniaPhiladelphia, PennsylvaniaUnited States of America
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of PennsylvaniaPhiladelphia, PennsylvaniaUnited States of America
- Department of Chemistry, School of Arts and Sciences, University of PennsylvaniaPhiladelphia, PennsylvaniaUnited States of America
- Penn Institute for Computational Science, University of PennsylvaniaPhiladelphia, PennsylvaniaUnited States of America
| | - Octavio L Franco
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília70790-160Brazil
- S-Inova Biotech, Pós-graduação em Biotecnologia, Universidade Católica Dom BoscoCampo Grande, Mato Grosso do SulBrazil
| |
Collapse
|
13
|
Wang B, Lin P, Zhong Y, Tan X, Shen Y, Huang Y, Jin K, Zhang Y, Zhan Y, Shen D, Wang M, Yu Z, Wu Y. Explainable deep learning and virtual evolution identifies antimicrobial peptides with activity against multidrug-resistant human pathogens. Nat Microbiol 2025; 10:332-347. [PMID: 39825096 DOI: 10.1038/s41564-024-01907-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 12/04/2024] [Indexed: 01/20/2025]
Abstract
Artificial intelligence (AI) is a promising approach to identify new antimicrobial compounds in diverse microbial species. Here we developed an AI-based, explainable deep learning model, EvoGradient, that predicts the potency of antimicrobial peptides (AMPs) and virtually modifies peptide sequences to produce more potent AMPs, akin to in silico directed evolution. We applied this model to peptides encoded in low-abundance human oral bacteria, resulting in the virtual evolution of 32 peptides into potent AMPs. Of these, the 6 most effective were synthesized and tested against multidrug-resistant pathogens and demonstrated activity against carbapenem-resistant species Escherichia coli, Klebsiella pneumoniae and Acinetobacter baumannii, and vancomycin-resistant Enterococcus faecium. The most potent AMP, pep-19-mod, was validated in vivo, achieving over 95% reduction in bacterial loads in mouse models of thigh infection through both systemic and local administration. Our approach advances the automatic identification and optimization of AMPs.
Collapse
Affiliation(s)
- Beilun Wang
- School of Computer Science and Engineering, Southeast University, Nanjing, China.
| | - Peijun Lin
- School of Computer Science and Engineering, Southeast University, Nanjing, China
| | - Yuwei Zhong
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, China
| | - Xiao Tan
- School of Computer Science and Engineering, Southeast University, Nanjing, China
- Department of Data Science and AI, Monash University, Melbourne, Victoria, Australia
| | - Yangyang Shen
- School of Computer Science and Engineering, Southeast University, Nanjing, China
| | - Yi Huang
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, China
| | - Kai Jin
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, China
| | - Yan Zhang
- School of Computer Science and Engineering, Southeast University, Nanjing, China
| | - Ying Zhan
- School of Computer Science and Engineering, Southeast University, Nanjing, China
| | - Dian Shen
- School of Computer Science and Engineering, Southeast University, Nanjing, China
| | - Meng Wang
- XAI Lab, College of Design and Innovation, Tongji University, Shanghai, China
| | - Zhou Yu
- Computer Science Department, Columbia University, New York, NY, USA.
| | - Yihan Wu
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, China.
| |
Collapse
|
14
|
Torres MDT, Chen T, Wan F, Chatterjee P, de la Fuente-Nunez C. Generative latent diffusion language modeling yields anti-infective synthetic peptides. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.31.636003. [PMID: 39975107 PMCID: PMC11838489 DOI: 10.1101/2025.01.31.636003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Generative artificial intelligence (AI) offers a powerful avenue for peptide design, yet this process remains challenging due to the vast sequence space, complex structure-activity relationships, and the need to balance antimicrobial potency with low toxicity. Traditional approaches often rely on trial-and-error screening and fail to efficiently navigate the immense diversity of potential sequences. Here, we introduce AMP-Diffusion, a novel latent diffusion model fine-tuned on antimicrobial peptide (AMP) sequences using embeddings from protein language models. By systematically exploring sequence space, AMP-Diffusion enables the rapid discovery of promising antibiotic candidates. We generated 50,000 candidate sequences, which were subsequently filtered and ranked using our APEX predictor model. From these, 46 top candidates were synthesized and experimentally validated. The resulting AMP-Diffusion peptides demonstrated broad-spectrum antibacterial activity, targeting clinically relevant pathogens-including multidrug-resistant strains-while exhibiting low cytotoxicity in human cell assays. Mechanistic studies revealed bacterial killing via membrane permeabilization and depolarization, and the peptides showed favorable physicochemical profiles. In preclinical mouse models of infection, lead peptides effectively reduced bacterial burdens, displaying efficacy comparable to polymyxin B and levofloxacin, with no detectable adverse effects. This study highlights the potential of AMP-Diffusion as a robust generative platform for designing novel antibiotics and bioactive peptides, offering a promising strategy to address the escalating challenge of antimicrobial resistance.
Collapse
Affiliation(s)
- Marcelo D. T. Torres
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Tianlai Chen
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Fangping Wan
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Pranam Chatterjee
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Department of Computer Science, Duke University, Durham, NC, USA
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - Cesar de la Fuente-Nunez
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
15
|
Alexander J, Liu G, Stokes JM. Explainable artificial intelligence evolves antimicrobial peptides. Nat Microbiol 2025; 10:267-269. [PMID: 39856390 DOI: 10.1038/s41564-024-01919-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2025]
Affiliation(s)
- Jeremie Alexander
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, Ontario, Canada
| | - Gary Liu
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, Ontario, Canada
| | - Jonathan M Stokes
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada.
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada.
- David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
16
|
Barroso RA, Agüero-Chapin G, Sousa R, Marrero-Ponce Y, Antunes A. Unlocking Antimicrobial Peptides: In Silico Proteolysis and Artificial Intelligence-Driven Discovery from Cnidarian Omics. Molecules 2025; 30:550. [PMID: 39942653 PMCID: PMC11820242 DOI: 10.3390/molecules30030550] [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: 12/07/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Overcoming the growing challenge of antimicrobial resistance (AMR), which affects millions of people worldwide, has driven attention for the exploration of marine-derived antimicrobial peptides (AMPs) for innovative solutions. Cnidarians, such as corals, sea anemones, and jellyfish, are a promising valuable resource of these bioactive peptides due to their robust innate immune systems yet are still poorly explored. Hence, we employed an in silico proteolysis strategy to search for novel AMPs from omics data of 111 Cnidaria species. Millions of peptides were retrieved and screened using shallow- and deep-learning models, prioritizing AMPs with a reduced toxicity and with a structural distinctiveness from characterized AMPs. After complex network analysis, a final dataset of 3130 Cnidaria singular non-haemolytic and non-toxic AMPs were identified. Such unique AMPs were mined for their putative antibacterial activity, revealing 20 favourable candidates for in vitro testing against important ESKAPEE pathogens, offering potential new avenues for antibiotic development.
Collapse
Affiliation(s)
- Ricardo Alexandre Barroso
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, 4450-208 Porto, Portugal; (R.A.B.); (G.A.-C.); (R.S.)
- Department of Biology, Faculty of Sciences of University of Porto (FCUP), Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Guillermin Agüero-Chapin
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, 4450-208 Porto, Portugal; (R.A.B.); (G.A.-C.); (R.S.)
- Department of Biology, Faculty of Sciences of University of Porto (FCUP), Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Rita Sousa
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, 4450-208 Porto, Portugal; (R.A.B.); (G.A.-C.); (R.S.)
- Department of Biology, Faculty of Sciences of University of Porto (FCUP), Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Yovani Marrero-Ponce
- Facultad de Ingeniería, Universidad Panamericana, Augusto Rodin No. 498, Insurgentes Mixcoac, Benito Juárez, Ciudad de Mexico 03920, Mexico;
- Grupo de Medicina Molecular y Traslacional (MeM&T), Colegio de Ciencias de la Salud (COCSA), Escuela de Medicina, Edificio de Especialidades Médicas, Instituto de Simulación Computacional (ISC-USFQ), Universidad San Francisco de Quito (USFQ), Diego de Robles y vía Interoceánica, Quito 170157, Ecuador
| | - Agostinho Antunes
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, 4450-208 Porto, Portugal; (R.A.B.); (G.A.-C.); (R.S.)
- Department of Biology, Faculty of Sciences of University of Porto (FCUP), Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| |
Collapse
|
17
|
Chen Z, Ji C, Xu W, Gao J, Huang J, Xu H, Qian G, Huang J. UniAMP: enhancing AMP prediction using deep neural networks with inferred information of peptides. BMC Bioinformatics 2025; 26:10. [PMID: 39799358 PMCID: PMC11725221 DOI: 10.1186/s12859-025-06033-3] [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: 08/21/2024] [Accepted: 01/02/2025] [Indexed: 01/15/2025] Open
Abstract
Antimicrobial peptides (AMPs) have been widely recognized as a promising solution to combat antimicrobial resistance of microorganisms due to the increasing abuse of antibiotics in medicine and agriculture around the globe. In this study, we propose UniAMP, a systematic prediction framework for discovering AMPs. We observe that feature vectors used in various existing studies constructed from peptide information, such as sequence, composition, and structure, can be augmented and even replaced by information inferred by deep learning models. Specifically, we use a feature vector with 2924 values inferred by two deep learning models, UniRep and ProtT5, to demonstrate that such inferred information of peptides suffice for the task, with the help of our proposed deep neural network model composed of fully connected layers and transformer encoders for predicting the antibacterial activity of peptides. Evaluation results demonstrate superior performance of our proposed model on both balanced benchmark datasets and imbalanced test datasets compared with existing studies. Subsequently, we analyze the relations among peptide sequences, manually extracted features, and automatically inferred information by deep learning models, leading to observations that the inferred information is more comprehensive and non-redundant for the task of predicting AMPs. Moreover, this approach alleviates the impact of the scarcity of positive data and demonstrates great potential in future research and applications.
Collapse
Affiliation(s)
- Zixin Chen
- College of Artificial Intelligence, Nanjing Agricultural University, Weigang No.1, Nanjing, 210095, Jiangsu, China
| | - Chengming Ji
- College of Artificial Intelligence, Nanjing Agricultural University, Weigang No.1, Nanjing, 210095, Jiangsu, China
| | - Wenwen Xu
- College of Artificial Intelligence, Nanjing Agricultural University, Weigang No.1, Nanjing, 210095, Jiangsu, China
| | - Jianfeng Gao
- StarHelix Inc, Jiangmiao Road, Nanjing, 210000, Jiangsu, China
| | - Ji Huang
- College of Agriculture, Nanjing Agricultural University, Weigang No.1, Nanjing, 210095, Jiangsu, China
| | - Huanliang Xu
- College of Artificial Intelligence, Nanjing Agricultural University, Weigang No.1, Nanjing, 210095, Jiangsu, China
| | - Guoliang Qian
- College of Plant Protection, Nanjing Agricultural University, Weigang No.1, Nanjing, 210095, Jiangsu, China.
| | - Junxian Huang
- College of Artificial Intelligence, Nanjing Agricultural University, Weigang No.1, Nanjing, 210095, Jiangsu, China.
| |
Collapse
|
18
|
Brizuela CA, Liu G, Stokes JM, de la Fuente‐Nunez C. AI Methods for Antimicrobial Peptides: Progress and Challenges. Microb Biotechnol 2025; 18:e70072. [PMID: 39754551 PMCID: PMC11702388 DOI: 10.1111/1751-7915.70072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 11/18/2024] [Accepted: 12/16/2024] [Indexed: 01/06/2025] Open
Abstract
Antimicrobial peptides (AMPs) are promising candidates to combat multidrug-resistant pathogens. However, the high cost of extensive wet-lab screening has made AI methods for identifying and designing AMPs increasingly important, with machine learning (ML) techniques playing a crucial role. AI approaches have recently revolutionised this field by accelerating the discovery of new peptides with anti-infective activity, particularly in preclinical mouse models. Initially, classical ML approaches dominated the field, but recently there has been a shift towards deep learning (DL) models. Despite significant contributions, existing reviews have not thoroughly explored the potential of large language models (LLMs), graph neural networks (GNNs) and structure-guided AMP discovery and design. This review aims to fill that gap by providing a comprehensive overview of the latest advancements, challenges and opportunities in using AI methods, with a particular emphasis on LLMs, GNNs and structure-guided design. We discuss the limitations of current approaches and highlight the most relevant topics to address in the coming years for AMP discovery and design.
Collapse
Affiliation(s)
| | - Gary Liu
- Department of Biochemistry and Biomedical Sciences, Michael G. DeGroote Institute for Infectious Disease Research, David Braley Centre for Antibiotic DiscoveryMcMaster UniversityHamiltonOntarioCanada
| | - Jonathan M. Stokes
- Department of Biochemistry and Biomedical Sciences, Michael G. DeGroote Institute for Infectious Disease Research, David Braley Centre for Antibiotic DiscoveryMcMaster UniversityHamiltonOntarioCanada
| | - Cesar de la Fuente‐Nunez
- Machine Biology Group, Department of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied ScienceUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of Chemistry, School of Arts and SciencesUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Penn Institute for Computational ScienceUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
19
|
Rios TB, Rezende SB, Maximiano MR, Cardoso MH, Malmsten M, de la Fuente-Nunez C, Franco OL. Computational Approaches for Antimicrobial Peptide Delivery. Bioconjug Chem 2024; 35:1873-1882. [PMID: 39541149 DOI: 10.1021/acs.bioconjchem.4c00406] [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: 11/16/2024]
Abstract
Peptides constitute alternative molecules for the treatment of infections caused by bacteria, viruses, fungi, and protozoa. However, their therapeutic effectiveness is often limited by enzymatic degradation, chemical and physical instability, and toxicity toward healthy human cells. To improve their pharmacokinetic (PK) and pharmacodynamic (PD) profiles, novel routes of administration are being explored. Among these, nanoparticles have shown promise as potential carriers for peptides, although the design of delivery vehicles remains a slow and painstaking process, heavily reliant on trial and error. Recently, computational approaches have been introduced to accelerate the development of effective drug delivery systems for peptides. Here we present an overview of some of these computational strategies and discuss their potential to optimize drug development and delivery.
Collapse
Affiliation(s)
- Thuanny Borba Rios
- S-Inova Biotech, Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul 70990-160, Brazil
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Distrito Federal 71966-700, Brazil
| | - Samilla Beatriz Rezende
- S-Inova Biotech, Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul 70990-160, Brazil
| | - Mariana Rocha Maximiano
- S-Inova Biotech, Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul 70990-160, Brazil
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Distrito Federal 71966-700, Brazil
| | - Marlon Henrique Cardoso
- S-Inova Biotech, Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul 70990-160, Brazil
| | - Martin Malmsten
- Department of Pharmacy, University of Copenhagen, DK-2100 Copenhagen, Denmark
- Physical Chemistry 1, University of Lund, S-221 00 Lund, Sweden
| | - Cesar de la Fuente-Nunez
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Octávio Luiz Franco
- S-Inova Biotech, Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul 70990-160, Brazil
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Distrito Federal 71966-700, Brazil
| |
Collapse
|
20
|
Fan S, Qin P, Lu J, Wang S, Zhang J, Wang Y, Cheng A, Cao Y, Ding W, Zhang W. Bioprospecting of culturable marine biofilm bacteria for novel antimicrobial peptides. IMETA 2024; 3:e244. [PMID: 39742298 PMCID: PMC11683478 DOI: 10.1002/imt2.244] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 01/03/2025]
Abstract
Antimicrobial peptides (AMPs) have become a viable source of novel antibiotics that are effective against human pathogenic bacteria. In this study, we construct a bank of culturable marine biofilm bacteria constituting 713 strains and their nearly complete genomes and predict AMPs using ribosome profiling and deep learning. Compared with previous approaches, ribosome profiling has improved the identification and validation of small open reading frames (sORFs) for AMP prediction. Among the 80,430 expressed sORFs, 341 are identified as candidate AMPs with high probability. Most potential AMPs have less than 40% similarity in their amino acid sequence compared to those listed in public databases. Furthermore, these AMPs are associated with bacterial groups that are not previously known to produce AMPs. Therefore, our deep learning model has acquired characteristics of unfamiliar AMPs. Chemical synthesis of 60 potential AMP sequences yields 54 compounds with antimicrobial activity, including potent inhibitory effects on various drug-resistant human pathogens. This study extends the range of AMP compounds by investigating marine biofilm microbiomes using a novel approach, accelerating AMP discovery.
Collapse
Affiliation(s)
- Shen Fan
- MOE Key Laboratory of Evolution & Marine Biodiversity and Institute of Evolution & Marine BiodiversityOcean University of ChinaQingdaoChina
| | - Peng Qin
- MOE Key Laboratory of Evolution & Marine Biodiversity and Institute of Evolution & Marine BiodiversityOcean University of ChinaQingdaoChina
| | - Jie Lu
- MOE Key Laboratory of Evolution & Marine Biodiversity and Institute of Evolution & Marine BiodiversityOcean University of ChinaQingdaoChina
| | - Shuaitao Wang
- MOE Key Laboratory of Evolution & Marine Biodiversity and Institute of Evolution & Marine BiodiversityOcean University of ChinaQingdaoChina
| | - Jie Zhang
- MOE Key Laboratory of Evolution & Marine Biodiversity and Institute of Evolution & Marine BiodiversityOcean University of ChinaQingdaoChina
| | - Yan Wang
- MOE Key Laboratory of Evolution & Marine Biodiversity and Institute of Evolution & Marine BiodiversityOcean University of ChinaQingdaoChina
| | - Aifang Cheng
- Department of Biomedical Sciences, Faculty of Health SciencesUniversity of MacauTaipaMacao SARChina
| | - Yan Cao
- College of Pulmonary & Critical Care MedicineChinese PLA General HospitalBeijingChina
| | - Wei Ding
- MOE Key Laboratory of Marine Genetics & Breeding and College of Marine Life SciencesOcean University of ChinaQingdaoChina
| | - Weipeng Zhang
- MOE Key Laboratory of Evolution & Marine Biodiversity and Institute of Evolution & Marine BiodiversityOcean University of ChinaQingdaoChina
| |
Collapse
|
21
|
Varela-Rodríguez H, Guzman-Pando A, Camarillo-Cisneros J. Screening and computational characterization of novel antimicrobial cathelicidins from amphibian transcriptomic data. Comput Biol Chem 2024; 113:108276. [PMID: 39546857 DOI: 10.1016/j.compbiolchem.2024.108276] [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: 09/05/2024] [Revised: 10/15/2024] [Accepted: 11/03/2024] [Indexed: 11/17/2024]
Abstract
As cold-blooded organisms living in damp and dark environments, amphibians have evolved robust defense mechanisms to protect themselves from predators and infections. Among the wide repertoire of bioactive compounds they produce are antimicrobial peptides (AMPs), which are required as part of innate immunity. One important class of AMPs is cathelicidins, known for their broad-spectrum activity against pathogens and their immunoregulatory roles. However, despite their promising biomedical potential and the increasing availability of omics data, few cathelicidins have been studied in amphibians, mostly through conventional experimental techniques. Here, we present 210 novel cathelicidin sequences from amphibian transcriptomes, identified through a comprehensive computational pipeline, which employed HMMER and BLAST tools to screen cathelicidin domains. These sequences reveal a typical tripartite domain architecture that was confirmed by SignalP and InterProScan analysis. Phylogenetic inference with IQ-TREE classified the sequences into six categories based on evolutionary relationships. Compared to cathelicidins from other vertebrates, amphibian mature peptides exhibit longer average lengths (around 50 amino acids), fewer aromatic and hydrophobic residues, and reduced thermal stability. Furthermore, these amphibian cathelicidins were characterized for their physicochemical and biological properties, revealing significant antimicrobial potential with lower hemolytic capability, especially in anurans, which suggests a balance between their antimicrobial and hemolytic activities predicted through AMPlify, ampir, AmpGram, and HemoPI. Secondary structure estimations, including three-dimensional modeling using AlphaFold2, indicate that amphibian cathelicidins predominantly feature α-helices and coils. Some representative models also display a high α-helix composition with amphipathic topology, facilitating interactions with simulated bacterial membranes as assessed by the PPM approach. Thus, these findings highlight the functional role of cathelicidins in amphibian immunity and their promising biomedical applicability, emphasizing the importance of applying computational methods to expand the scope and reveal the diverse landscape of cathelicidins across vertebrates.
Collapse
Affiliation(s)
- H Varela-Rodríguez
- Computational Chemistry Physics Laboratory, Facultad de Medicina y Ciencias Biomédicas, Universidad Autónoma de Chihuahua (UACH), Chihuahua 31125, Chihuahua, Mexico
| | - A Guzman-Pando
- Computational Chemistry Physics Laboratory, Facultad de Medicina y Ciencias Biomédicas, Universidad Autónoma de Chihuahua (UACH), Chihuahua 31125, Chihuahua, Mexico
| | - J Camarillo-Cisneros
- Computational Chemistry Physics Laboratory, Facultad de Medicina y Ciencias Biomédicas, Universidad Autónoma de Chihuahua (UACH), Chihuahua 31125, Chihuahua, Mexico.
| |
Collapse
|
22
|
Zhong G, Liu H, Deng L. Ensemble Machine Learning and Predicted Properties Promote Antimicrobial Peptide Identification. Interdiscip Sci 2024; 16:951-965. [PMID: 38972032 DOI: 10.1007/s12539-024-00640-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: 01/22/2024] [Revised: 06/04/2024] [Accepted: 06/07/2024] [Indexed: 07/08/2024]
Abstract
The emergence of antibiotic-resistant microbes raises a pressing demand for novel alternative treatments. One promising alternative is the antimicrobial peptides (AMPs), a class of innate immunity mediators within the therapeutic peptide realm. AMPs offer salient advantages such as high specificity, cost-effective synthesis, and reduced toxicity. Although some computational methodologies have been proposed to identify potential AMPs with the rapid development of artificial intelligence techniques, there is still ample room to improve their performance. This study proposes a predictive framework which ensembles deep learning and statistical learning methods to screen peptides with antimicrobial activity. We integrate multiple LightGBM classifiers and convolution neural networks which leverages various predicted sequential, structural and physicochemical properties from their residue sequences extracted by diverse machine learning paradigms. Comparative experiments exhibit that our method outperforms other state-of-the-art approaches on an independent test dataset, in terms of representative capability measures. Besides, we analyse the discrimination quality under different varieties of attribute information and it reveals that combination of multiple features could improve prediction. In addition, a case study is carried out to illustrate the exemplary favorable identification effect. We establish a web application at http://amp.denglab.org to provide convenient usage of our proposal and make the predictive framework, source code, and datasets publicly accessible at https://github.com/researchprotein/amp .
Collapse
Affiliation(s)
- Guolun Zhong
- School of Computer Science and Engineering, Central South University, Changsha, 410083, China
| | - Hui Liu
- College of Computer and Information Engineering, Nanjing Tech University, Nanjing, 211816, China.
| | - Lei Deng
- School of Computer Science and Engineering, Central South University, Changsha, 410083, China.
| |
Collapse
|
23
|
Hashemi S, Vosough P, Taghizadeh S, Savardashtaki A. Therapeutic peptide development revolutionized: Harnessing the power of artificial intelligence for drug discovery. Heliyon 2024; 10:e40265. [PMID: 39605829 PMCID: PMC11600032 DOI: 10.1016/j.heliyon.2024.e40265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 10/07/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Abstract
Due to the spread of antibiotic resistance, global attention is focused on its inhibition and the expansion of effective medicinal compounds. The novel functional properties of peptides have opened up new horizons in personalized medicine. With artificial intelligence methods combined with therapeutic peptide products, pharmaceuticals and biotechnology advance drug development rapidly and reduce costs. Short-chain peptides inhibit a wide range of pathogens and have great potential for targeting diseases. To address the challenges of synthesis and sustainability, artificial intelligence methods, namely machine learning, must be integrated into their production. Learning methods can use complicated computations to select the active and toxic compounds of the drug and its metabolic activity. Through this comprehensive review, we investigated the artificial intelligence method as a potential tool for finding peptide-based drugs and providing a more accurate analysis of peptides through the introduction of predictable databases for effective selection and development.
Collapse
Affiliation(s)
- Samaneh Hashemi
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Parisa Vosough
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeed Taghizadeh
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Science Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
24
|
Cao J, Zhang J, Yu Q, Ji J, Li J, He S, Zhu Z. TG-CDDPM: text-guided antimicrobial peptides generation based on conditional denoising diffusion probabilistic model. Brief Bioinform 2024; 26:bbae644. [PMID: 39668337 PMCID: PMC11637771 DOI: 10.1093/bib/bbae644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 11/13/2024] [Accepted: 11/27/2024] [Indexed: 12/14/2024] Open
Abstract
Antimicrobial peptides (AMPs) have emerged as a promising substitution to antibiotics thanks to their boarder range of activities, less likelihood of drug resistance, and low toxicity. Traditional biochemical methods for AMP discovery are costly and inefficient. Deep generative models, including the long-short term memory model, variational autoencoder model, and generative adversarial model, have been widely introduced to expedite AMP discovery. However, these models tend to suffer from the lack of diversity in generating AMPs. The denoising diffusion probabilistic model serves as a good candidate for solving this issue. We proposed a three-stage Text-Guided Conditional Denoising Diffusion Probabilistic Model (TG-CDDPM) to generate novel and homologous AMPs. In the first two stages, contrastive learning and inferring models are crafted to create better conditions for guiding AMP generation, respectively. In the last stage, a pre-trained conditional denoising diffusion probabilistic model is leveraged to enrich the peptide knowledge and fine-tuned to learn feature representation in downstream. TG-CDDPM was compared to the state-of-the-art generative models for AMP generation, and it demonstrated competitive or better performance with the assistance of text description as supervised information. The membrane penetration capabilities of the identified candidate AMPs by TG-CDDPM were also validated through molecular weight dynamics experiments.
Collapse
Affiliation(s)
- Junhang Cao
- College of Computer Science and Software Engineering, Shenzhen University, Shenzhen 518060, China
| | - Jun Zhang
- National Engineering Laboratory for Big Data System Computing Technology, Shenzhen University, Shenzhen 518060, China
| | - Qiyuan Yu
- College of Computer Science and Software Engineering, Shenzhen University, Shenzhen 518060, China
| | - Junkai Ji
- National Engineering Laboratory for Big Data System Computing Technology, Shenzhen University, Shenzhen 518060, China
| | - Jianqiang Li
- National Engineering Laboratory for Big Data System Computing Technology, Shenzhen University, Shenzhen 518060, China
| | - Shan He
- School of Computer Science, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Zexuan Zhu
- National Engineering Laboratory for Big Data System Computing Technology, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
25
|
Liu X, Luo J, Wang X, Zhang Y, Chen J. Directed evolution of antimicrobial peptides using multi-objective zeroth-order optimization. Brief Bioinform 2024; 26:bbae715. [PMID: 39800873 PMCID: PMC11725395 DOI: 10.1093/bib/bbae715] [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: 08/01/2024] [Revised: 12/08/2024] [Accepted: 12/27/2024] [Indexed: 01/16/2025] Open
Abstract
Antimicrobial peptides (AMPs) emerge as a type of promising therapeutic compounds that exhibit broad spectrum antimicrobial activity with high specificity and good tolerability. Natural AMPs usually need further rational design for improving antimicrobial activity and decreasing toxicity to human cells. Although several algorithms have been developed to optimize AMPs with desired properties, they explored the variations of AMPs in a discrete amino acid sequence space, usually suffering from low efficiency, lack diversity, and local optimum. In this work, we propose a novel directed evolution method, named PepZOO, for optimizing multi-properties of AMPs in a continuous representation space guided by multi-objective zeroth-order optimization. PepZOO projects AMPs from a discrete amino acid sequence space into continuous latent representation space by a variational autoencoder. Subsequently, the latent embeddings of prototype AMPs are taken as start points and iteratively updated according to the guidance of multi-objective zeroth-order optimization. Experimental results demonstrate PepZOO outperforms state-of-the-art methods on improving the multi-properties in terms of antimicrobial function, activity, toxicity, and binding affinity to the targets. Molecular docking and molecular dynamics simulations are further employed to validate the effectiveness of our method. Moreover, PepZOO can reveal important motifs which are required to maintain a particular property during the evolution by aligning the evolutionary sequences. PepZOO provides a novel research paradigm that optimizes AMPs by exploring property change instead of exploring sequence mutations, accelerating the discovery of potential therapeutic peptides.
Collapse
Affiliation(s)
- Xianliang Liu
- School of Computer Science and Technology, Harbin Institute of Technology, HIT Campus, Shenzhen University Town, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Jiawei Luo
- School of Computer Science and Technology, Harbin Institute of Technology, HIT Campus, Shenzhen University Town, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Xinyan Wang
- Core Research Facility, Southern University of Science and Technology, No. 1088 Xueyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Yang Zhang
- School of Science, Harbin Institute of Technology, HIT Campus, Shenzhen University Town, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Junjie Chen
- School of Computer Science and Technology, Harbin Institute of Technology, HIT Campus, Shenzhen University Town, Nanshan District, Shenzhen 518055, Guangdong, China
| |
Collapse
|
26
|
Meng Q, Chen G, Lin B, Zheng S, Lin Y, Tang J, Guo F. DMAMP: A Deep-Learning Model for Detecting Antimicrobial Peptides and Their Multi-Activities. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2024; 21:2025-2034. [PMID: 39106141 DOI: 10.1109/tcbb.2024.3439541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/09/2024]
Abstract
Due to the broad-spectrum and high-efficiency antibacterial activity, antimicrobial peptides (AMPs) and their functions have been studied in the field of drug discovery. Using biological experiments to detect the AMPs and corresponding activities require a high cost, whereas computational technologies do so for much less. Currently, most computational methods solve the identification of AMPs and their activities as two independent tasks, which ignore the relationship between them. Therefore, the combination and sharing of patterns for two tasks is a crucial problem that needs to be addressed. In this study, we propose a deep learning model, called DMAMP, for detecting AMPs and activities simultaneously, which is benefited from multi-task learning. The first stage is to utilize convolutional neural network models and residual blocks to extract the sharing hidden features from two related tasks. The next stage is to use two fully connected layers to learn the distinct information of two tasks. Meanwhile, the original evolutionary features from the peptide sequence are also fed to the predictor of the second task to complement the forgotten information. The experiments on the independent test dataset demonstrate that our method performs better than the single-task model with 4.28% of Matthews Correlation Coefficient (MCC) on the first task, and achieves 0.2627 of an average MCC which is higher than the single-task model and two existing methods for five activities on the second task. To understand whether features derived from the convolutional layers of models capture the differences between target classes, we visualize these high-dimensional features by projecting into 3D space. In addition, we show that our predictor has the ability to identify peptides that achieve activity against Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2). We hope that our proposed method can give new insights into the discovery of novel antiviral peptide drugs.
Collapse
|
27
|
Singh A, Tanwar M, Singh TP, Sharma S, Sharma P. An escape from ESKAPE pathogens: A comprehensive review on current and emerging therapeutics against antibiotic resistance. Int J Biol Macromol 2024; 279:135253. [PMID: 39244118 DOI: 10.1016/j.ijbiomac.2024.135253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/09/2024]
Abstract
The rise of antimicrobial resistance has positioned ESKAPE pathogens as a serious global health threat, primarily due to the limitations and frequent failures of current treatment options. This growing risk has spurred the scientific community to seek innovative antibiotic therapies and improved oversight strategies. This review aims to provide a comprehensive overview of the origins and resistance mechanisms of ESKAPE pathogens, while also exploring next-generation treatment strategies for these infections. In addition, it will address both traditional and novel approaches to combating antibiotic resistance, offering insights into potential new therapeutic avenues. Emerging research underscores the urgency of developing new antimicrobial agents and strategies to overcome resistance, highlighting the need for novel drug classes and combination therapies. Advances in genomic technologies and a deeper understanding of microbial pathogenesis are crucial in identifying effective treatments. Integrating precision medicine and personalized approaches could enhance therapeutic efficacy. The review also emphasizes the importance of global collaboration in surveillance and stewardship, as well as policy reforms, enhanced diagnostic tools, and public awareness initiatives, to address resistance on a worldwide scale.
Collapse
Affiliation(s)
- Anamika Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Mansi Tanwar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - T P Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Sujata Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India.
| | - Pradeep Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India.
| |
Collapse
|
28
|
Anwer F, Navid A, Faiz F, Haider U, Nasir S, Farooq M, Zahra M, Bano A, Bashir HH, Ahmad M, Abbas SA, Room SE, Saeed MT, Ali A. AbAMPdb: a database of Acinetobacter baumannii specific antimicrobial peptides. Database (Oxford) 2024; 2024:baae096. [PMID: 39395188 PMCID: PMC11470754 DOI: 10.1093/database/baae096] [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: 01/08/2024] [Revised: 07/26/2024] [Accepted: 10/07/2024] [Indexed: 10/14/2024]
Abstract
Acinetobacter baumannii has emerged as a prominent nosocomial pathogen, exhibiting a progressive rise in resistance to therapeutic interventions. This rise in resistance calls for alternative strategies. Here, we propose an alternative yet specialized resource on antimicrobial peptides (AMPs) against A. baumannii. Database 'AbAMPdb' is the manually curated collection of 300 entries containing the 250 experimental AMP sequences and 50 corresponding synthetic or mutated AMP sequences. The mutated sequences were modified with reported amino acid substitutions intended for decreasing the toxicity and increasing the antimicrobial potency. AbAMPdb also provides 3D models of all 300 AMPs, comprising 250 natural and 50 synthetic or mutated AMPs. Moreover, the database offers docked complexes comprising 5000 AMPs and their corresponding A. baumannii target proteins. These complexes, accessible in Protein Data Bank format, enable the 2D visualization of the interacting amino acid residues. We are confident that this comprehensive resource furnishes vital information concerning AMPs, encompassing their docking interactions with virulence factors and antibiotic resistance proteins of A. baumannii. To enhance clinical relevance, the characterized AMPs could undergo further investigation both in vitro and in vivo. Database URL: https://abampdb.mgbio.tech/.
Collapse
Affiliation(s)
- Farha Anwer
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Sector H-12, Islamabad 44000, Pakistan
| | - Ahmad Navid
- School of Interdisciplinary Engineering & Sciences (SINES), National University of Sciences and Technology (NUST), Sector H-12, Islamabad 44000, Pakistan
| | - Fiza Faiz
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Sector H-12, Islamabad 44000, Pakistan
| | - Uzair Haider
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Sector H-12, Islamabad 44000, Pakistan
| | - Samavi Nasir
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Sector H-12, Islamabad 44000, Pakistan
| | - Muhammad Farooq
- Department of Medical Lab Technology, BIC, University of Harīpur, Haripur, Khyber Pakhtunkhwa 22620, Pakistan
| | - Maryam Zahra
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Sector H-12, Islamabad 44000, Pakistan
| | - Anosh Bano
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Sector H-12, Islamabad 44000, Pakistan
| | - Hafiza Hira Bashir
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Sector H-12, Islamabad 44000, Pakistan
| | - Madiha Ahmad
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Sector H-12, Islamabad 44000, Pakistan
| | - Syeda Aleena Abbas
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Sector H-12, Islamabad 44000, Pakistan
| | - Shah E Room
- Xylexa Inc, National University of Sciences and Technology (NUST), Sector H-12, Islamabad 44000, Pakistan
| | - Muhammad Tariq Saeed
- School of Interdisciplinary Engineering & Sciences (SINES), National University of Sciences and Technology (NUST), Sector H-12, Islamabad 44000, Pakistan
| | - Amjad Ali
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Sector H-12, Islamabad 44000, Pakistan
- MGBIO (SMC-PRIVATE) Limited, C4 H Building 1, National Science and Technology Park, NUST, H-12, Islamabad 44000, Pakistan
| |
Collapse
|
29
|
Han J, Kong T, Liu J. PepNet: an interpretable neural network for anti-inflammatory and antimicrobial peptides prediction using a pre-trained protein language model. Commun Biol 2024; 7:1198. [PMID: 39341947 PMCID: PMC11438969 DOI: 10.1038/s42003-024-06911-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
Identifying anti-inflammatory peptides (AIPs) and antimicrobial peptides (AMPs) is crucial for the discovery of innovative and effective peptide-based therapies targeting inflammation and microbial infections. However, accurate identification of AIPs and AMPs remains a computational challenge mainly due to limited utilization of peptide sequence information. Here, we propose PepNet, an interpretable neural network for predicting both AIPs and AMPs by applying a pre-trained protein language model to fully utilize the peptide sequence information. It first captures the information of residue arrangements and physicochemical properties using a residual dilated convolution block, and then seizes the function-related diverse information by introducing a residual Transformer block to characterize the residue representations generated by a pre-trained protein language model. After training and testing, PepNet demonstrates great superiority over other leading AIP and AMP predictors and shows strong interpretability of its learned peptide representations. A user-friendly web server for PepNet is freely available at http://liulab.top/PepNet/server .
Collapse
Affiliation(s)
- Jiyun Han
- School of Mathematics and Statistics, Shandong University, 264209, Weihai, China
| | - Tongxin Kong
- School of Mathematics and Statistics, Shandong University, 264209, Weihai, China
| | - Juntao Liu
- School of Mathematics and Statistics, Shandong University, 264209, Weihai, China.
| |
Collapse
|
30
|
Shariati A, Kashi M, Chegini Z, Hosseini SM. Antibiotics-free compounds for managing carbapenem-resistant bacteria; a narrative review. Front Pharmacol 2024; 15:1467086. [PMID: 39355778 PMCID: PMC11442292 DOI: 10.3389/fphar.2024.1467086] [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: 07/19/2024] [Accepted: 09/04/2024] [Indexed: 10/03/2024] Open
Abstract
Carbapenem-resistant (CR) Gram-negative bacteria have become a significant public health problem in the last decade. In recent years, the prevalence of CR bacteria has increased. The resistance to carbapenems could result from different mechanisms such as loss of porin, penicillin-binding protein alteration, carbapenemase, efflux pump, and biofilm community. Additionally, genetic variations like insertion, deletion, mutation, and post-transcriptional modification of corresponding coding genes could decrease the susceptibility of bacteria to carbapenems. In this regard, scientists are looking for new approaches to inhibit CR bacteria. Using bacteriophages, natural products, nanoparticles, disulfiram, N-acetylcysteine, and antimicrobial peptides showed promising inhibitory effects against CR bacteria. Additionally, the mentioned compounds could destroy the biofilm community of CR bacteria. Using them in combination with conventional antibiotics increases the efficacy of antibiotics, decreases their dosage and toxicity, and resensitizes CR bacteria to antibiotics. Therefore, in the present review article, we have discussed different aspects of non-antibiotic approaches for managing and inhibiting the CR bacteria and various methods and procedures used as an alternative for carbapenems against these bacteria.
Collapse
Affiliation(s)
- Aref Shariati
- Infectious Diseases Research Center (IDRC), Arak University of Medical Sciences, Arak, Iran
| | - Milad Kashi
- Student research committee, Arak University of Medical Sciences, Arak, Iran
| | - Zahra Chegini
- Infectious Disease Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Seyed Mostafa Hosseini
- Infectious Disease Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
31
|
de la Lastra JMP, Wardell SJT, Pal T, de la Fuente-Nunez C, Pletzer D. From Data to Decisions: Leveraging Artificial Intelligence and Machine Learning in Combating Antimicrobial Resistance - a Comprehensive Review. J Med Syst 2024; 48:71. [PMID: 39088151 PMCID: PMC11294375 DOI: 10.1007/s10916-024-02089-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 07/12/2024] [Indexed: 08/02/2024]
Abstract
The emergence of drug-resistant bacteria poses a significant challenge to modern medicine. In response, Artificial Intelligence (AI) and Machine Learning (ML) algorithms have emerged as powerful tools for combating antimicrobial resistance (AMR). This review aims to explore the role of AI/ML in AMR management, with a focus on identifying pathogens, understanding resistance patterns, predicting treatment outcomes, and discovering new antibiotic agents. Recent advancements in AI/ML have enabled the efficient analysis of large datasets, facilitating the reliable prediction of AMR trends and treatment responses with minimal human intervention. ML algorithms can analyze genomic data to identify genetic markers associated with antibiotic resistance, enabling the development of targeted treatment strategies. Additionally, AI/ML techniques show promise in optimizing drug administration and developing alternatives to traditional antibiotics. By analyzing patient data and clinical outcomes, these technologies can assist healthcare providers in diagnosing infections, evaluating their severity, and selecting appropriate antimicrobial therapies. While integration of AI/ML in clinical settings is still in its infancy, advancements in data quality and algorithm development suggest that widespread clinical adoption is forthcoming. In conclusion, AI/ML holds significant promise for improving AMR management and treatment outcome.
Collapse
Affiliation(s)
- José M Pérez de la Lastra
- Biotechnology of Macromolecules, Instituto de Productos Naturales y Agrobiología, IPNA (CSIC), Avda. Astrofísico Francisco Sánchez, 3, 38206, San Cristóbal de la Laguna, (Santa Cruz de Tenerife), Spain.
| | - Samuel J T Wardell
- Department of Microbiology and Immunology, School of Biomedical Sciences, University of Otago, 9054, Dunedin, New Zealand
| | - Tarun Pal
- School of Bioengineering and Food Technology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, 173229, Himachal Pradesh, India
| | - Cesar de la Fuente-Nunez
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel Pletzer
- Department of Microbiology and Immunology, School of Biomedical Sciences, University of Otago, 9054, Dunedin, New Zealand.
| |
Collapse
|
32
|
Li C, Sutherland D, Richter A, Coombe L, Yanai A, Warren RL, Kotkoff M, Hof F, Hoang LMN, Helbing CC, Birol I. De novo synthetic antimicrobial peptide design with a recurrent neural network. Protein Sci 2024; 33:e5088. [PMID: 38988311 PMCID: PMC11237553 DOI: 10.1002/pro.5088] [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: 12/18/2023] [Revised: 05/16/2024] [Accepted: 06/10/2024] [Indexed: 07/12/2024]
Abstract
Antibiotic resistance is recognized as an imminent and growing global health threat. New antimicrobial drugs are urgently needed due to the decreasing effectiveness of conventional small-molecule antibiotics. Antimicrobial peptides (AMPs), a class of host defense peptides, are emerging as promising candidates to address this need. The potential sequence space of amino acids is combinatorially vast, making it possible to extend the current arsenal of antimicrobial agents with a practically infinite number of new peptide-based candidates. However, mining naturally occurring AMPs, whether directly by wet lab screening methods or aided by bioinformatics prediction tools, has its theoretical limit regarding the number of samples or genomic/transcriptomic resources researchers have access to. Further, manually designing novel synthetic AMPs requires prior field knowledge, restricting its throughput. In silico sequence generation methods are gaining interest as a high-throughput solution to the problem. Here, we introduce AMPd-Up, a recurrent neural network based tool for de novo AMP design, and demonstrate its utility over existing methods. Validation of candidates designed by AMPd-Up through antimicrobial susceptibility testing revealed that 40 of the 58 generated sequences possessed antimicrobial activity against Escherichia coli and/or Staphylococcus aureus. These results illustrate that AMPd-Up can be used to design novel synthetic AMPs with potent activities.
Collapse
Affiliation(s)
- Chenkai Li
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
- Bioinformatics Graduate ProgramUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Darcy Sutherland
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
- Public Health LaboratoryBritish Columbia Centre for Disease ControlVancouverBritish ColumbiaCanada
- Department of Pathology and Laboratory MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Amelia Richter
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
- Public Health LaboratoryBritish Columbia Centre for Disease ControlVancouverBritish ColumbiaCanada
| | - Lauren Coombe
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
| | - Anat Yanai
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
- Public Health LaboratoryBritish Columbia Centre for Disease ControlVancouverBritish ColumbiaCanada
| | - René L. Warren
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
| | - Monica Kotkoff
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
| | - Fraser Hof
- Department of Chemistry and the Centre for Advanced Materials and Related TechnologyUniversity of VictoriaVictoriaBritish ColumbiaCanada
| | - Linda M. N. Hoang
- Public Health LaboratoryBritish Columbia Centre for Disease ControlVancouverBritish ColumbiaCanada
- Department of Pathology and Laboratory MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Caren C. Helbing
- Department of Biochemistry and MicrobiologyUniversity of VictoriaVictoriaBritish ColumbiaCanada
| | - Inanc Birol
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
- Public Health LaboratoryBritish Columbia Centre for Disease ControlVancouverBritish ColumbiaCanada
- Department of Pathology and Laboratory MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
- Department of Medical GeneticsUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| |
Collapse
|
33
|
Ramos-Llorens M, Bello-Madruga R, Valle J, Andreu D, Torrent M. PyAMPA: a high-throughput prediction and optimization tool for antimicrobial peptides. mSystems 2024; 9:e0135823. [PMID: 38934543 PMCID: PMC11264690 DOI: 10.1128/msystems.01358-23] [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: 12/23/2023] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
The alarming rise of antibiotic-resistant bacterial infections is driving efforts to develop alternatives to conventional antibiotics. In this context, antimicrobial peptides (AMPs) have emerged as promising candidates for their ability to target a broad range of microorganisms. However, the development of AMPs with optimal potency, selectivity, and/or stability profiles remains a challenge. To address it, computational tools for predicting AMP properties and designing novel peptides have gained increasing attention. PyAMPA is a novel platform for AMP discovery. It consists of five modules, namely AMPScreen, AMPValidate, AMPSolve, AMPMutate, and AMPOptimize, that allow high-throughput proteome inspection, candidate screening, and optimization through point-mutation and genetic algorithms. The platform also offers additional tools for predicting and evaluating AMP properties, including antimicrobial and cytotoxic activity, and peptide half-life. By providing innovative and accessible inroads into AMP motifs in proteomes, PyAMPA will enable advances in AMP development and potential translation into clinically useful molecules. PyAMPA is available at: https://github.com/SysBioUAB/PyAMPA. IMPORTANCE This paper introduces PyAMPA, a new bioinformatics platform designed for the discovery and optimization of antimicrobial peptides (AMPs). It addresses the urgent need for new antimicrobials due to the rise of antibiotic-resistant infections. PyAMPA, with its five predictive modules -AMPScreen, AMPValidate, AMPSolve, AMPMutate and AMPOptimize, enables high-throughput screening of proteomes to identify potential AMP motifs and optimize them for clinical use. Its unique approach, combining prediction, design, and optimization tools, makes PyAMPA a robust solution for developing new AMP-based therapies, offering a significant advance in combatting antibiotic resistance.
Collapse
Affiliation(s)
- Marc Ramos-Llorens
- Systems Biology of Infection Lab, Department of Biochemistry and Molecular Biology, Biosciences Faculty, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Roberto Bello-Madruga
- Systems Biology of Infection Lab, Department of Biochemistry and Molecular Biology, Biosciences Faculty, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Javier Valle
- Barcelona Biomedical Research Park, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - David Andreu
- Barcelona Biomedical Research Park, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Marc Torrent
- Systems Biology of Infection Lab, Department of Biochemistry and Molecular Biology, Biosciences Faculty, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
34
|
Zhao F, Qiu J, Xiang D, Jiao P, Cao Y, Xu Q, Qiao D, Xu H, Cao Y. deepAMPNet: a novel antimicrobial peptide predictor employing AlphaFold2 predicted structures and a bi-directional long short-term memory protein language model. PeerJ 2024; 12:e17729. [PMID: 39040937 PMCID: PMC11262304 DOI: 10.7717/peerj.17729] [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: 02/28/2024] [Accepted: 06/20/2024] [Indexed: 07/24/2024] Open
Abstract
Background Global public health is seriously threatened by the escalating issue of antimicrobial resistance (AMR). Antimicrobial peptides (AMPs), pivotal components of the innate immune system, have emerged as a potent solution to AMR due to their therapeutic potential. Employing computational methodologies for the prompt recognition of these antimicrobial peptides indeed unlocks fresh perspectives, thereby potentially revolutionizing antimicrobial drug development. Methods In this study, we have developed a model named as deepAMPNet. This model, which leverages graph neural networks, excels at the swift identification of AMPs. It employs structures of antimicrobial peptides predicted by AlphaFold2, encodes residue-level features through a bi-directional long short-term memory (Bi-LSTM) protein language model, and constructs adjacency matrices anchored on amino acids' contact maps. Results In a comparative study with other state-of-the-art AMP predictors on two external independent test datasets, deepAMPNet outperformed in accuracy. Furthermore, in terms of commonly accepted evaluation matrices such as AUC, Mcc, sensitivity, and specificity, deepAMPNet achieved the highest or highly comparable performances against other predictors. Conclusion deepAMPNet interweaves both structural and sequence information of AMPs, stands as a high-performance identification model that propels the evolution and design in antimicrobial peptide pharmaceuticals. The data and code utilized in this study can be accessed at https://github.com/Iseeu233/deepAMPNet.
Collapse
Affiliation(s)
- Fei Zhao
- Microbiology and Metabolic Engineering Laboratory of Sichuan Province, College of Life Science, Sichuan University, Chengdu, Sichuan, China
| | - Junhui Qiu
- Microbiology and Metabolic Engineering Laboratory of Sichuan Province, College of Life Science, Sichuan University, Chengdu, Sichuan, China
| | - Dongyou Xiang
- Microbiology and Metabolic Engineering Laboratory of Sichuan Province, College of Life Science, Sichuan University, Chengdu, Sichuan, China
| | - Pengrui Jiao
- Microbiology and Metabolic Engineering Laboratory of Sichuan Province, College of Life Science, Sichuan University, Chengdu, Sichuan, China
| | - Yu Cao
- Microbiology and Metabolic Engineering Laboratory of Sichuan Province, College of Life Science, Sichuan University, Chengdu, Sichuan, China
| | - Qingrui Xu
- Microbiology and Metabolic Engineering Laboratory of Sichuan Province, College of Life Science, Sichuan University, Chengdu, Sichuan, China
| | - Dairong Qiao
- Microbiology and Metabolic Engineering Laboratory of Sichuan Province, College of Life Science, Sichuan University, Chengdu, Sichuan, China
| | - Hui Xu
- Microbiology and Metabolic Engineering Laboratory of Sichuan Province, College of Life Science, Sichuan University, Chengdu, Sichuan, China
| | - Yi Cao
- Microbiology and Metabolic Engineering Laboratory of Sichuan Province, College of Life Science, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
35
|
Santos-Júnior CD, Torres MDT, Duan Y, Rodríguez Del Río Á, Schmidt TSB, Chong H, Fullam A, Kuhn M, Zhu C, Houseman A, Somborski J, Vines A, Zhao XM, Bork P, Huerta-Cepas J, de la Fuente-Nunez C, Coelho LP. Discovery of antimicrobial peptides in the global microbiome with machine learning. Cell 2024; 187:3761-3778.e16. [PMID: 38843834 PMCID: PMC11666328 DOI: 10.1016/j.cell.2024.05.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 04/11/2024] [Accepted: 05/06/2024] [Indexed: 06/25/2024]
Abstract
Novel antibiotics are urgently needed to combat the antibiotic-resistance crisis. We present a machine-learning-based approach to predict antimicrobial peptides (AMPs) within the global microbiome and leverage a vast dataset of 63,410 metagenomes and 87,920 prokaryotic genomes from environmental and host-associated habitats to create the AMPSphere, a comprehensive catalog comprising 863,498 non-redundant peptides, few of which match existing databases. AMPSphere provides insights into the evolutionary origins of peptides, including by duplication or gene truncation of longer sequences, and we observed that AMP production varies by habitat. To validate our predictions, we synthesized and tested 100 AMPs against clinically relevant drug-resistant pathogens and human gut commensals both in vitro and in vivo. A total of 79 peptides were active, with 63 targeting pathogens. These active AMPs exhibited antibacterial activity by disrupting bacterial membranes. In conclusion, our approach identified nearly one million prokaryotic AMP sequences, an open-access resource for antibiotic discovery.
Collapse
Affiliation(s)
- Célio Dias Santos-Júnior
- Institute of Science and Technology for Brain-Inspired Intelligence - ISTBI, Fudan University, Shanghai 200433, China; Laboratory of Microbial Processes & Biodiversity - LMPB, Department of Hydrobiology, Universidade Federal de São Carlos - UFSCar, São Carlos, São Paulo 13565-905, Brazil
| | - Marcelo D T Torres
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA; Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA; Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Yiqian Duan
- Institute of Science and Technology for Brain-Inspired Intelligence - ISTBI, Fudan University, Shanghai 200433, China
| | - Álvaro Rodríguez Del Río
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid (UPM) - Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Campus de Montegancedo-UPM, Pozuelo de Alarcón, 28223 Madrid, Spain
| | - Thomas S B Schmidt
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany; APC Microbiome & School of Medicine, University College Cork, Cork, Ireland
| | - Hui Chong
- Institute of Science and Technology for Brain-Inspired Intelligence - ISTBI, Fudan University, Shanghai 200433, China
| | - Anthony Fullam
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Michael Kuhn
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Chengkai Zhu
- Institute of Science and Technology for Brain-Inspired Intelligence - ISTBI, Fudan University, Shanghai 200433, China
| | - Amy Houseman
- Institute of Science and Technology for Brain-Inspired Intelligence - ISTBI, Fudan University, Shanghai 200433, China
| | - Jelena Somborski
- Institute of Science and Technology for Brain-Inspired Intelligence - ISTBI, Fudan University, Shanghai 200433, China
| | - Anna Vines
- Institute of Science and Technology for Brain-Inspired Intelligence - ISTBI, Fudan University, Shanghai 200433, China
| | - Xing-Ming Zhao
- Institute of Science and Technology for Brain-Inspired Intelligence - ISTBI, Fudan University, Shanghai 200433, China; Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China; State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai, China; MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Peer Bork
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany; Max Delbrück Centre for Molecular Medicine, Berlin, Germany; Department of Bioinformatics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Jaime Huerta-Cepas
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid (UPM) - Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Campus de Montegancedo-UPM, Pozuelo de Alarcón, 28223 Madrid, Spain
| | - Cesar de la Fuente-Nunez
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA; Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA; Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA, USA.
| | - Luis Pedro Coelho
- Institute of Science and Technology for Brain-Inspired Intelligence - ISTBI, Fudan University, Shanghai 200433, China; Centre for Microbiome Research, School of Biomedical Sciences, Queensland University of Technology, Translational Research Institute, Woolloongabba, QLD, Australia.
| |
Collapse
|
36
|
Kim N, Ma J, Kim W, Kim J, Belenky P, Lee I. Genome-resolved metagenomics: a game changer for microbiome medicine. Exp Mol Med 2024; 56:1501-1512. [PMID: 38945961 PMCID: PMC11297344 DOI: 10.1038/s12276-024-01262-7] [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: 12/13/2023] [Revised: 03/06/2024] [Accepted: 03/25/2024] [Indexed: 07/02/2024] Open
Abstract
Recent substantial evidence implicating commensal bacteria in human diseases has given rise to a new domain in biomedical research: microbiome medicine. This emerging field aims to understand and leverage the human microbiota and derivative molecules for disease prevention and treatment. Despite the complex and hierarchical organization of this ecosystem, most research over the years has relied on 16S amplicon sequencing, a legacy of bacterial phylogeny and taxonomy. Although advanced sequencing technologies have enabled cost-effective analysis of entire microbiota, translating the relatively short nucleotide information into the functional and taxonomic organization of the microbiome has posed challenges until recently. In the last decade, genome-resolved metagenomics, which aims to reconstruct microbial genomes directly from whole-metagenome sequencing data, has made significant strides and continues to unveil the mysteries of various human-associated microbial communities. There has been a rapid increase in the volume of whole metagenome sequencing data and in the compilation of novel metagenome-assembled genomes and protein sequences in public depositories. This review provides an overview of the capabilities and methods of genome-resolved metagenomics for studying the human microbiome, with a focus on investigating the prokaryotic microbiota of the human gut. Just as decoding the human genome and its variations marked the beginning of the genomic medicine era, unraveling the genomes of commensal microbes and their sequence variations is ushering us into the era of microbiome medicine. Genome-resolved metagenomics stands as a pivotal tool in this transition and can accelerate our journey toward achieving these scientific and medical milestones.
Collapse
Affiliation(s)
- Nayeon Kim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Junyeong Ma
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Wonjong Kim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jungyeon Kim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Peter Belenky
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912, USA.
| | - Insuk Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea.
- POSTECH Biotech Center, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea.
| |
Collapse
|
37
|
Ebrahimikondori H, Sutherland D, Yanai A, Richter A, Salehi A, Li C, Coombe L, Kotkoff M, Warren RL, Birol I. Structure-aware deep learning model for peptide toxicity prediction. Protein Sci 2024; 33:e5076. [PMID: 39196703 PMCID: PMC11193153 DOI: 10.1002/pro.5076] [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: 01/09/2024] [Revised: 04/26/2024] [Accepted: 05/28/2024] [Indexed: 08/30/2024]
Abstract
Antimicrobial resistance is a critical public health concern, necessitating the exploration of alternative treatments. While antimicrobial peptides (AMPs) show promise, assessing their toxicity using traditional wet lab methods is both time-consuming and costly. We introduce tAMPer, a novel multi-modal deep learning model designed to predict peptide toxicity by integrating the underlying amino acid sequence composition and the three-dimensional structure of peptides. tAMPer adopts a graph-based representation for peptides, encoding ColabFold-predicted structures, where nodes represent amino acids and edges represent spatial interactions. Structural features are extracted using graph neural networks, and recurrent neural networks capture sequential dependencies. tAMPer's performance was assessed on a publicly available protein toxicity benchmark and an AMP hemolysis data we generated. On the latter, tAMPer achieves an F1-score of 68.7%, outperforming the second-best method by 23.4%. On the protein benchmark, tAMPer exhibited an improvement of over 3.0% in the F1-score compared to current state-of-the-art methods. We anticipate tAMPer to accelerate AMP discovery and development by reducing the reliance on laborious toxicity screening experiments.
Collapse
Affiliation(s)
- Hossein Ebrahimikondori
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
- Bioinformatics Graduate ProgramUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Darcy Sutherland
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
- Public Health LaboratoryBritish Columbia Centre for Disease ControlVancouverBritish ColumbiaCanada
- Department of Pathology and Laboratory MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Anat Yanai
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
- Public Health LaboratoryBritish Columbia Centre for Disease ControlVancouverBritish ColumbiaCanada
| | - Amelia Richter
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
- Public Health LaboratoryBritish Columbia Centre for Disease ControlVancouverBritish ColumbiaCanada
| | - Ali Salehi
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
- Public Health LaboratoryBritish Columbia Centre for Disease ControlVancouverBritish ColumbiaCanada
| | - Chenkai Li
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
- Bioinformatics Graduate ProgramUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Lauren Coombe
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
| | - Monica Kotkoff
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
| | - René L. Warren
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
| | - Inanc Birol
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
- Public Health LaboratoryBritish Columbia Centre for Disease ControlVancouverBritish ColumbiaCanada
- Department of Pathology and Laboratory MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
- Department of Medical GeneticsUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| |
Collapse
|
38
|
Nguyen QH, Nguyen-Vo TH, Do TTT, Nguyen BP. An efficient hybrid deep learning architecture for predicting short antimicrobial peptides. Proteomics 2024; 24:e2300382. [PMID: 38837544 DOI: 10.1002/pmic.202300382] [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: 10/02/2023] [Revised: 05/02/2024] [Accepted: 05/07/2024] [Indexed: 06/07/2024]
Abstract
Short-length antimicrobial peptides (AMPs) have been demonstrated to have intensified antimicrobial activities against a wide spectrum of microbes. Therefore, exploration of novel and promising short AMPs is highly essential in developing various types of antimicrobial drugs or treatments. In addition to experimental approaches, computational methods have been developed to improve screening efficiency. Although existing computational methods have achieved satisfactory performance, there is still much room for model improvement. In this study, we proposed iAMP-DL, an efficient hybrid deep learning architecture, for predicting short AMPs. The model was constructed using two well-known deep learning architectures: the long short-term memory architecture and convolutional neural networks. To fairly assess the performance of the model, we compared our model with existing state-of-the-art methods using the same independent test set. Our comparative analysis shows that iAMP-DL outperformed other methods. Furthermore, to assess the robustness and stability of our model, the experiments were repeated 10 times to observe the variation in prediction efficiency. The results demonstrate that iAMP-DL is an effective, robust, and stable framework for detecting promising short AMPs. Another comparative study of different negative data sampling methods also confirms the effectiveness of our method and demonstrates that it can also be used to develop a robust model for predicting AMPs in general. The proposed framework was also deployed as an online web server with a user-friendly interface to support the research community in identifying short AMPs.
Collapse
Affiliation(s)
- Quang H Nguyen
- School of Information and Communication Technology, Hanoi University of Science and Technology, Hanoi, Vietnam
| | - Thanh-Hoang Nguyen-Vo
- School of Mathematics and Statistics, Victoria University of Wellington, Wellington, New Zealand
- School of Innovation, Design and Technology, Wellington Institute of Technology, Lower Hutt, New Zealand
| | - Trang T T Do
- Faculty of Information Technology, Ho Chi Minh City Open University, Ho Chi Minh City, Vietnam
| | - Binh P Nguyen
- School of Mathematics and Statistics, Victoria University of Wellington, Wellington, New Zealand
- Faculty of Information Technology, Ho Chi Minh City Open University, Ho Chi Minh City, Vietnam
| |
Collapse
|
39
|
Groover KE, Randall JR, Davies BW. Development of a Selective and Stable Antimicrobial Peptide. ACS Infect Dis 2024; 10:2151-2160. [PMID: 38712889 PMCID: PMC11185160 DOI: 10.1021/acsinfecdis.4c00142] [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: 02/23/2024] [Revised: 04/17/2024] [Accepted: 04/26/2024] [Indexed: 05/08/2024]
Abstract
Antimicrobial peptides (AMPs) are presented as potential scaffolds for antibiotic development due to their desirable qualities including broad-spectrum activity, rapid action, and general lack of susceptibility to current resistance mechanisms. However, they often lose antibacterial activity under physiological conditions and/or display mammalian cell toxicity, which limits their potential use. Identification of AMPs that overcome these barriers will help develop rules for how this antibacterial class can be developed to treat infection. Here we describe the development of our novel synthetic AMP, from discovery through in vivo application. Our evolved AMP, DTr18-dab, has broad-spectrum antibacterial activity and is nonhemolytic. It is active against planktonic bacteria and biofilm, is unaffected by colistin resistance, and importantly is active in both human serum and a Galleria mellonella infection model. Several modifications, including the incorporation of noncanonical amino acids, were used to arrive at this robust sequence. We observed that the impact on antibacterial activity with noncanonical amino acids was dependent on assay conditions and therefore not entirely predictable. Overall, our results demonstrate how a relatively weak lead can be developed into a robust AMP with qualities important for potential therapeutic translation.
Collapse
Affiliation(s)
- Kyra E. Groover
- Department
of Molecular Biosciences, The University
of Texas at Austin, Austin, Texas 78712, United States
| | - Justin R. Randall
- Department
of Molecular Biosciences, The University
of Texas at Austin, Austin, Texas 78712, United States
| | - Bryan W. Davies
- Department
of Molecular Biosciences, The University
of Texas at Austin, Austin, Texas 78712, United States
- John
Ring LaMontagne Center for Infectious Diseases, The University of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|
40
|
Shaon MSH, Karim T, Sultan MF, Ali MM, Ahmed K, Hasan MZ, Moustafa A, Bui FM, Al-Zahrani FA. AMP-RNNpro: a two-stage approach for identification of antimicrobials using probabilistic features. Sci Rep 2024; 14:12892. [PMID: 38839785 PMCID: PMC11153637 DOI: 10.1038/s41598-024-63461-6] [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: 02/16/2024] [Accepted: 05/29/2024] [Indexed: 06/07/2024] Open
Abstract
Antimicrobials are molecules that prevent the formation of microorganisms such as bacteria, viruses, fungi, and parasites. The necessity to detect antimicrobial peptides (AMPs) using machine learning and deep learning arises from the need for efficiency to accelerate the discovery of AMPs, and contribute to developing effective antimicrobial therapies, especially in the face of increasing antibiotic resistance. This study introduced AMP-RNNpro based on Recurrent Neural Network (RNN), an innovative model for detecting AMPs, which was designed with eight feature encoding methods that are selected according to four criteria: amino acid compositional, grouped amino acid compositional, autocorrelation, and pseudo-amino acid compositional to represent the protein sequences for efficient identification of AMPs. In our framework, two-stage predictions have been conducted. Initially, this study analyzed 33 models on these feature extractions. Then, we selected the best six models from these models using rigorous performance metrics. In the second stage, probabilistic features have been generated from the selected six models in each feature encoding and they are aggregated to be fed into our final meta-model called AMP-RNNpro. This study also introduced 20 features with SHAP, which are crucial in the drug development fields, where we discover AAC, ASDC, and CKSAAGP features are highly impactful for detection and drug discovery. Our proposed framework, AMP-RNNpro excels in the identification of novel Amps with 97.15% accuracy, 96.48% sensitivity, and 97.87% specificity. We built a user-friendly website for demonstrating the accurate prediction of AMPs based on the proposed approach which can be accessed at http://13.126.159.30/ .
Collapse
Affiliation(s)
- Md Shazzad Hossain Shaon
- Department of Computer Science and Engineering, Daffodil International University, Daffodil Smart City, Birulia, Dhaka, 1216, Bangladesh
| | - Tasmin Karim
- Department of Computer Science and Engineering, Daffodil International University, Daffodil Smart City, Birulia, Dhaka, 1216, Bangladesh
| | - Md Fahim Sultan
- Department of Computer Science and Engineering, Daffodil International University, Daffodil Smart City, Birulia, Dhaka, 1216, Bangladesh
| | - Md Mamun Ali
- Health Informatics Research Lab, Department of Computer Science and Engineering, Daffodil International University, Daffodil Smart City, Birulia, Dhaka, 1216, Bangladesh
- Division of Biomedical Engineering, University of Saskatchewan, 57 Campus Drive, Saskatoon, SK, S7N 5A9, Canada
- Department of Software Engineering, Daffodil International University, Daffodil Smart City (DSC), Birulia, Savar, Dhaka, 1216, Bangladesh
| | - Kawsar Ahmed
- Health Informatics Research Lab, Department of Computer Science and Engineering, Daffodil International University, Daffodil Smart City, Birulia, Dhaka, 1216, Bangladesh.
- Department of Electrical and Computer Engineering, University of Saskatchewan, 57 Campus Drive, Saskatoon, SK, S7N 5A9, Canada.
- Group of Bio-photomatiχ, Information and Communication Technology, Mawlana Bhashani Science and Technology University, Santosh, Tangail, 1902, Bangladesh.
| | - Md Zahid Hasan
- Department of Computer Science and Engineering, Daffodil International University, Daffodil Smart City, Birulia, Dhaka, 1216, Bangladesh
- Health Informatics Research Lab, Department of Computer Science and Engineering, Daffodil International University, Daffodil Smart City, Birulia, Dhaka, 1216, Bangladesh
| | - Ahmed Moustafa
- Department of Human Anatomy and Physiology, The Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
- School of Psychology, Centre for Data Analytics, Bond University, Gold Coast, QLD, Australia
| | - Francis M Bui
- Department of Electrical and Computer Engineering, University of Saskatchewan, 57 Campus Drive, Saskatoon, SK, S7N 5A9, Canada
| | | |
Collapse
|
41
|
Yin K, Xu W, Ren S, Xu Q, Zhang S, Zhang R, Jiang M, Zhang Y, Xu D, Li R. Machine Learning Accelerates De Novo Design of Antimicrobial Peptides. Interdiscip Sci 2024; 16:392-403. [PMID: 38416364 DOI: 10.1007/s12539-024-00612-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/17/2024] [Accepted: 01/23/2024] [Indexed: 02/29/2024]
Abstract
Efficient and precise design of antimicrobial peptides (AMPs) is of great importance in the field of AMP development. Computing provides opportunities for peptide de novo design. In the present investigation, a new machine learning-based AMP prediction model, AP_Sin, was trained using 1160 AMP sequences and 1160 non-AMP sequences. The results showed that AP_Sin correctly classified 94.61% of AMPs on a comprehensive dataset, outperforming the mainstream and open-source models (Antimicrobial Peptide Scanner vr.2, iAMPpred and AMPlify) and being effective in identifying AMPs. In addition, a peptide sequence generator, AP_Gen, was devised based on the concept of recombining dominant amino acids and dipeptide compositions. After inputting the parameters of the 71 tridecapeptides from antimicrobial peptides database (APD3) into AP_Gen, a tridecapeptide bank consisting of de novo designed 17,496 tridecapeptide sequences were randomly generated, from which 2675 candidate AMP sequences were identified by AP_Sin. Chemical synthesis was performed on 180 randomly selected candidate AMP sequences, of which 18 showed high antimicrobial activities against a wide range of the tested pathogenic microorganisms, and 16 of which had a minimal inhibitory concentration of less than 10 μg/mL against at least one of the tested pathogenic microorganisms. The method established in this research accelerates the discovery of valuable candidate AMPs and provides a novel approach for de novo design of antimicrobial peptides.
Collapse
Affiliation(s)
- Kedong Yin
- Key Laboratory of Functional Molecules for Biomedical Research, Henan University of Technology, 100 Lianhua Street, Zhengzhou, 450001, Henan, People's Republic of China
- College of Information Science and Engineering, Henan University of Technology, 100 Lianhua Street, Zhengzhou, 450001, Henan, People's Republic of China
| | - Wen Xu
- Key Laboratory of Functional Molecules for Biomedical Research, Henan University of Technology, 100 Lianhua Street, Zhengzhou, 450001, Henan, People's Republic of China.
- Law College, Henan University of Technology, Zhengzhou, 450001, Henan, People's Republic of China.
| | - Shiming Ren
- Key Laboratory of Functional Molecules for Biomedical Research, Henan University of Technology, 100 Lianhua Street, Zhengzhou, 450001, Henan, People's Republic of China
- College of Biological Engineering, Henan University of Technology, 100 Lianhua Street, Zhengzhou, 450001, Henan, People's Republic of China
| | - Qingpeng Xu
- Key Laboratory of Functional Molecules for Biomedical Research, Henan University of Technology, 100 Lianhua Street, Zhengzhou, 450001, Henan, People's Republic of China
- School of Artificial Intelligence and Big Data, Henan University of Technology, Zhengzhou, 450001, Henan, People's Republic of China
| | - Shaojie Zhang
- Key Laboratory of Functional Molecules for Biomedical Research, Henan University of Technology, 100 Lianhua Street, Zhengzhou, 450001, Henan, People's Republic of China
- College of Biological Engineering, Henan University of Technology, 100 Lianhua Street, Zhengzhou, 450001, Henan, People's Republic of China
| | - Ruiling Zhang
- Key Laboratory of Functional Molecules for Biomedical Research, Henan University of Technology, 100 Lianhua Street, Zhengzhou, 450001, Henan, People's Republic of China
- School of Economics and Trade, Henan University of Technology, Zhengzhou, 450001, Henan, People's Republic of China
| | - Mengwan Jiang
- School of Artificial Intelligence and Big Data, Henan University of Technology, Zhengzhou, 450001, Henan, People's Republic of China
| | - Yuhong Zhang
- School of Artificial Intelligence and Big Data, Henan University of Technology, Zhengzhou, 450001, Henan, People's Republic of China
| | - Degang Xu
- College of Information Science and Engineering, Henan University of Technology, 100 Lianhua Street, Zhengzhou, 450001, Henan, People's Republic of China.
| | - Ruifang Li
- Key Laboratory of Functional Molecules for Biomedical Research, Henan University of Technology, 100 Lianhua Street, Zhengzhou, 450001, Henan, People's Republic of China.
- College of Biological Engineering, Henan University of Technology, 100 Lianhua Street, Zhengzhou, 450001, Henan, People's Republic of China.
| |
Collapse
|
42
|
Coelho LP, Santos-Júnior CD, de la Fuente-Nunez C. Challenges in computational discovery of bioactive peptides in 'omics data. Proteomics 2024; 24:e2300105. [PMID: 38458994 PMCID: PMC11537280 DOI: 10.1002/pmic.202300105] [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: 10/13/2023] [Revised: 02/06/2024] [Accepted: 02/06/2024] [Indexed: 03/10/2024]
Abstract
Peptides have a plethora of activities in biological systems that can potentially be exploited biotechnologically. Several peptides are used clinically, as well as in industry and agriculture. The increase in available 'omics data has recently provided a large opportunity for mining novel enzymes, biosynthetic gene clusters, and molecules. While these data primarily consist of DNA sequences, other types of data provide important complementary information. Due to their size, the approaches proven successful at discovering novel proteins of canonical size cannot be naïvely applied to the discovery of peptides. Peptides can be encoded directly in the genome as short open reading frames (smORFs), or they can be derived from larger proteins by proteolysis. Both of these peptide classes pose challenges as simple methods for their prediction result in large numbers of false positives. Similarly, functional annotation of larger proteins, traditionally based on sequence similarity to infer orthology and then transferring functions between characterized proteins and uncharacterized ones, cannot be applied for short sequences. The use of these techniques is much more limited and alternative approaches based on machine learning are used instead. Here, we review the limitations of traditional methods as well as the alternative methods that have recently been developed for discovering novel bioactive peptides with a focus on prokaryotic genomes and metagenomes.
Collapse
Affiliation(s)
- Luis Pedro Coelho
- Centre for Microbiome Research, School of Biomedical Sciences, Queensland University of Technology, Woolloongabba, Queensland, Australia
- Institute of Science and Technology for Brain-Inspired Intelligence – ISTBI, Fudan University, Shanghai, China
| | - Célio Dias Santos-Júnior
- Institute of Science and Technology for Brain-Inspired Intelligence – ISTBI, Fudan University, Shanghai, China
- Laboratory of Microbial Processes & Biodiversity – LMPB, Hydrobiology Department, Federal University of São Carlos – UFSCar, São Paulo, Brazil
| | - Cesar de la Fuente-Nunez
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
43
|
Corrie LM, Kuecks-Winger H, Ebrahimikondori H, Birol I, Helbing CC. Transcriptomic profiling of Rana [Lithobates] catesbeiana back skin during natural and thyroid hormone-induced metamorphosis under different temperature regimes with particular emphasis on innate immune system components. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2024; 50:101238. [PMID: 38714098 DOI: 10.1016/j.cbd.2024.101238] [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: 01/26/2024] [Revised: 04/28/2024] [Accepted: 04/30/2024] [Indexed: 05/09/2024]
Abstract
As amphibians undergo thyroid hormone (TH)-dependent metamorphosis from an aquatic tadpole to the terrestrial frog, their innate immune system must adapt to the new environment. Skin is a primary line of defense, yet this organ undergoes extensive remodelling during metamorphosis and how it responds to TH is poorly understood. Temperature modulation, which regulates metamorphic timing, is a unique way to uncover early TH-induced transcriptomic events. Metamorphosis of premetamorphic tadpoles is induced by exogenous TH administration at 24 °C but is paused at 5 °C. However, at 5 °C a "molecular memory" of TH exposure is retained that results in an accelerated metamorphosis upon shifting to 24 °C. We used RNA-sequencing to identify changes in Rana (Lithobates) catesbeiana back skin gene expression during natural and TH-induced metamorphosis. During natural metamorphosis, significant differential expression (DE) was observed in >6500 transcripts including classic TH-responsive transcripts (thrb and thibz), heat shock proteins, and innate immune system components: keratins, mucins, and antimicrobial peptides (AMPs). Premetamorphic tadpoles maintained at 5 °C showed 83 DE transcripts within 48 h after TH administration, including thibz which has previously been identified as a molecular memory component in other tissues. Over 3600 DE transcripts were detected in TH-treated tadpoles at 24 °C or when tadpoles held at 5 °C were shifted to 24 °C. Gene ontology (GO) terms related to transcription, RNA metabolic processes, and translation were enriched in both datasets and immune related GO terms were observed in the temperature-modulated experiment. Our findings have implications on survival as climate change affects amphibia worldwide.
Collapse
Affiliation(s)
- Lorissa M Corrie
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Haley Kuecks-Winger
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Hossein Ebrahimikondori
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC V5Z 4S6, Canada
| | - Inanc Birol
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC V5Z 4S6, Canada
| | - Caren C Helbing
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC V8P 5C2, Canada.
| |
Collapse
|
44
|
Chen N, Yu J, Zhe L, Wang F, Li X, Wong KC. TP-LMMSG: a peptide prediction graph neural network incorporating flexible amino acid property representation. Brief Bioinform 2024; 25:bbae308. [PMID: 38920345 PMCID: PMC11200197 DOI: 10.1093/bib/bbae308] [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: 04/08/2024] [Revised: 05/28/2024] [Accepted: 06/10/2024] [Indexed: 06/27/2024] Open
Abstract
Bioactive peptide therapeutics has been a long-standing research topic. Notably, the antimicrobial peptides (AMPs) have been extensively studied for its therapeutic potential. Meanwhile, the demand for annotating other therapeutic peptides, such as antiviral peptides (AVPs) and anticancer peptides (ACPs), also witnessed an increase in recent years. However, we conceive that the structure of peptide chains and the intrinsic information between the amino acids is not fully investigated among the existing protocols. Therefore, we develop a new graph deep learning model, namely TP-LMMSG, which offers lightweight and easy-to-deploy advantages while improving the annotation performance in a generalizable manner. The results indicate that our model can accurately predict the properties of different peptides. The model surpasses the other state-of-the-art models on AMP, AVP and ACP prediction across multiple experimental validated datasets. Moreover, TP-LMMSG also addresses the challenges of time-consuming pre-processing in graph neural network frameworks. With its flexibility in integrating heterogeneous peptide features, our model can provide substantial impacts on the screening and discovery of therapeutic peptides. The source code is available at https://github.com/NanjunChen37/TP_LMMSG.
Collapse
Affiliation(s)
- Nanjun Chen
- Department of Computer Science, City University of Hong Kong, 83 Tat Chee Ave, Kowloon Tong, Kowloon, Hong Kong SAR
| | - Jixiang Yu
- Department of Computer Science, City University of Hong Kong, 83 Tat Chee Ave, Kowloon Tong, Kowloon, Hong Kong SAR
| | - Liu Zhe
- Department of Computer Science, City University of Hong Kong, 83 Tat Chee Ave, Kowloon Tong, Kowloon, Hong Kong SAR
| | - Fuzhou Wang
- Department of Computer Science, City University of Hong Kong, 83 Tat Chee Ave, Kowloon Tong, Kowloon, Hong Kong SAR
| | - Xiangtao Li
- School of Artificial Intelligence, Jilin University, Chang Chun, Ji Lin, China
| | - Ka-Chun Wong
- Department of Computer Science, City University of Hong Kong, 83 Tat Chee Ave, Kowloon Tong, Kowloon, Hong Kong SAR
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, Guang Dong, China
| |
Collapse
|
45
|
Rusic D, Kumric M, Seselja Perisin A, Leskur D, Bukic J, Modun D, Vilovic M, Vrdoljak J, Martinovic D, Grahovac M, Bozic J. Tackling the Antimicrobial Resistance "Pandemic" with Machine Learning Tools: A Summary of Available Evidence. Microorganisms 2024; 12:842. [PMID: 38792673 PMCID: PMC11123121 DOI: 10.3390/microorganisms12050842] [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: 03/16/2024] [Revised: 04/16/2024] [Accepted: 04/19/2024] [Indexed: 05/26/2024] Open
Abstract
Antimicrobial resistance is recognised as one of the top threats healthcare is bound to face in the future. There have been various attempts to preserve the efficacy of existing antimicrobials, develop new and efficient antimicrobials, manage infections with multi-drug resistant strains, and improve patient outcomes, resulting in a growing mass of routinely available data, including electronic health records and microbiological information that can be employed to develop individualised antimicrobial stewardship. Machine learning methods have been developed to predict antimicrobial resistance from whole-genome sequencing data, forecast medication susceptibility, recognise epidemic patterns for surveillance purposes, or propose new antibacterial treatments and accelerate scientific discovery. Unfortunately, there is an evident gap between the number of machine learning applications in science and the effective implementation of these systems. This narrative review highlights some of the outstanding opportunities that machine learning offers when applied in research related to antimicrobial resistance. In the future, machine learning tools may prove to be superbugs' kryptonite. This review aims to provide an overview of available publications to aid researchers that are looking to expand their work with new approaches and to acquaint them with the current application of machine learning techniques in this field.
Collapse
Affiliation(s)
- Doris Rusic
- Department of Pharmacy, University of Split School of Medicine, Soltanska 2A, 21000 Split, Croatia; (D.R.); (A.S.P.); (D.L.); (J.B.); (D.M.)
| | - Marko Kumric
- Department of Pathophysiology, University of Split School of Medicine, Soltanska 2A, 21000 Split, Croatia; (M.K.); (M.V.); (J.V.); (D.M.)
- Laboratory for Cardiometabolic Research, University of Split School of Medicine, Soltanska 2A, 21000 Split, Croatia
| | - Ana Seselja Perisin
- Department of Pharmacy, University of Split School of Medicine, Soltanska 2A, 21000 Split, Croatia; (D.R.); (A.S.P.); (D.L.); (J.B.); (D.M.)
| | - Dario Leskur
- Department of Pharmacy, University of Split School of Medicine, Soltanska 2A, 21000 Split, Croatia; (D.R.); (A.S.P.); (D.L.); (J.B.); (D.M.)
| | - Josipa Bukic
- Department of Pharmacy, University of Split School of Medicine, Soltanska 2A, 21000 Split, Croatia; (D.R.); (A.S.P.); (D.L.); (J.B.); (D.M.)
| | - Darko Modun
- Department of Pharmacy, University of Split School of Medicine, Soltanska 2A, 21000 Split, Croatia; (D.R.); (A.S.P.); (D.L.); (J.B.); (D.M.)
| | - Marino Vilovic
- Department of Pathophysiology, University of Split School of Medicine, Soltanska 2A, 21000 Split, Croatia; (M.K.); (M.V.); (J.V.); (D.M.)
- Laboratory for Cardiometabolic Research, University of Split School of Medicine, Soltanska 2A, 21000 Split, Croatia
| | - Josip Vrdoljak
- Department of Pathophysiology, University of Split School of Medicine, Soltanska 2A, 21000 Split, Croatia; (M.K.); (M.V.); (J.V.); (D.M.)
- Laboratory for Cardiometabolic Research, University of Split School of Medicine, Soltanska 2A, 21000 Split, Croatia
| | - Dinko Martinovic
- Department of Pathophysiology, University of Split School of Medicine, Soltanska 2A, 21000 Split, Croatia; (M.K.); (M.V.); (J.V.); (D.M.)
- Department of Maxillofacial Surgery, University Hospital of Split, Spinciceva 1, 21000 Split, Croatia
| | - Marko Grahovac
- Department of Pharmacology, University of Split School of Medicine, Soltanska 2A, 21000 Split, Croatia;
| | - Josko Bozic
- Department of Pathophysiology, University of Split School of Medicine, Soltanska 2A, 21000 Split, Croatia; (M.K.); (M.V.); (J.V.); (D.M.)
- Laboratory for Cardiometabolic Research, University of Split School of Medicine, Soltanska 2A, 21000 Split, Croatia
| |
Collapse
|
46
|
La Corte C, Catania V, Dara M, Parrinello D, Staropoli M, Trapani MR, Cammarata M, Parisi MG. Equinins as Novel Broad-Spectrum Antimicrobial Peptides Isolated from the Cnidarian Actinia equina (Linnaeus, 1758). Mar Drugs 2024; 22:172. [PMID: 38667789 PMCID: PMC11051070 DOI: 10.3390/md22040172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/05/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Sea anemones are valuable for therapeutic research as a diversified source of bioactive molecules, due to their diverse bioactive molecules linked to predation and defence mechanisms involving toxins and antimicrobial peptides. Acid extracts from Actinia equina tentacles and body were examined for antibacterial activity against Gram-positive, Gram-negative bacteria, and fungi. The peptide fractions showed interesting minimum inhibitory concentration (MIC) values (up to 0.125 µg/mL) against the tested pathogens. Further investigation and characterization of tentacle acid extracts with significant antimicrobial activity led to the purification of peptides through reverse phase chromatography on solid phase and HPLC. Broad-spectrum antimicrobial peptide activity was found in 40% acetonitrile fractions. The resulting peptides had a molecular mass of 2612.91 and 3934.827 Da and MIC ranging from 0.06 to 0.20 mg/mL. Sequencing revealed similarities to AMPs found in amphibians, fish, and Cnidaria, with anti-Gram+, Gram-, antifungal, candidacidal, anti-methicillin-resistant Staphylococcus aureus, carbapenemase-producing, vancomycin-resistant bacteria, and multi-drug resistant activity. Peptides 6.2 and 7.3, named Equinin A and B, respectively, were synthesized and evaluated in vitro towards the above-mentioned bacterial pathogens. Equinin B exerted interesting antibacterial activity (MIC and bactericidal concentrations of 1 mg/mL and 0.25 mg/mL, respectively) and gene organization supporting its potential in applied research.
Collapse
Affiliation(s)
- Claudia La Corte
- Marine Immunobiology Laboratory, Department of Earth and Marine Sciences (DiSTeM), University of Palermo, Viale delle Scienze, Ed. 16, 90128 Palermo, Italy; (C.L.C.); (M.D.); (D.P.); (M.S.); (M.R.T.); (M.G.P.)
- NBFC—National Biodiversity Future Center, Piazza Marina 61, 90133 Palermo, Italy;
| | - Valentina Catania
- NBFC—National Biodiversity Future Center, Piazza Marina 61, 90133 Palermo, Italy;
- Department of Earth and Marine Sciences (DiSTeM), University of Palermo, Viale delle Scienze, Ed. 16, 90128 Palermo, Italy
| | - Mariano Dara
- Marine Immunobiology Laboratory, Department of Earth and Marine Sciences (DiSTeM), University of Palermo, Viale delle Scienze, Ed. 16, 90128 Palermo, Italy; (C.L.C.); (M.D.); (D.P.); (M.S.); (M.R.T.); (M.G.P.)
- NBFC—National Biodiversity Future Center, Piazza Marina 61, 90133 Palermo, Italy;
| | - Daniela Parrinello
- Marine Immunobiology Laboratory, Department of Earth and Marine Sciences (DiSTeM), University of Palermo, Viale delle Scienze, Ed. 16, 90128 Palermo, Italy; (C.L.C.); (M.D.); (D.P.); (M.S.); (M.R.T.); (M.G.P.)
- NBFC—National Biodiversity Future Center, Piazza Marina 61, 90133 Palermo, Italy;
| | - Mariele Staropoli
- Marine Immunobiology Laboratory, Department of Earth and Marine Sciences (DiSTeM), University of Palermo, Viale delle Scienze, Ed. 16, 90128 Palermo, Italy; (C.L.C.); (M.D.); (D.P.); (M.S.); (M.R.T.); (M.G.P.)
- NBFC—National Biodiversity Future Center, Piazza Marina 61, 90133 Palermo, Italy;
| | - Maria Rosa Trapani
- Marine Immunobiology Laboratory, Department of Earth and Marine Sciences (DiSTeM), University of Palermo, Viale delle Scienze, Ed. 16, 90128 Palermo, Italy; (C.L.C.); (M.D.); (D.P.); (M.S.); (M.R.T.); (M.G.P.)
| | - Matteo Cammarata
- Marine Immunobiology Laboratory, Department of Earth and Marine Sciences (DiSTeM), University of Palermo, Viale delle Scienze, Ed. 16, 90128 Palermo, Italy; (C.L.C.); (M.D.); (D.P.); (M.S.); (M.R.T.); (M.G.P.)
- NBFC—National Biodiversity Future Center, Piazza Marina 61, 90133 Palermo, Italy;
| | - Maria Giovanna Parisi
- Marine Immunobiology Laboratory, Department of Earth and Marine Sciences (DiSTeM), University of Palermo, Viale delle Scienze, Ed. 16, 90128 Palermo, Italy; (C.L.C.); (M.D.); (D.P.); (M.S.); (M.R.T.); (M.G.P.)
- NBFC—National Biodiversity Future Center, Piazza Marina 61, 90133 Palermo, Italy;
| |
Collapse
|
47
|
Li C, Zou Q, Jia C, Zheng J. AMPpred-MFA: An Interpretable Antimicrobial Peptide Predictor with a Stacking Architecture, Multiple Features, and Multihead Attention. J Chem Inf Model 2024; 64:2393-2404. [PMID: 37799091 DOI: 10.1021/acs.jcim.3c01017] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/07/2023]
Abstract
Antimicrobial peptides (AMPs) are small molecular polypeptides that can be widely used in the prevention and treatment of microbial infections. Although many computational models have been proposed to help identify AMPs, a high-performance and interpretable model is still lacking. In this study, new benchmark data sets are collected and processed, and a stacking deep architecture named AMPpred-MFA is carefully designed to discover and identify AMPs. Multiple features and a multihead attention mechanism are utilized on the basis of a bidirectional long short-term memory (LSTM) network and a convolutional neural network (CNN). The effectiveness of AMPpred-MFA is verified through five independent tests conducted in batches. Experimental results show that AMPpred-MFA achieves a state-of-the-art performance. The visualization interpretability analyses and ablation experiments offer a further understanding of the model behavior and performance, validating the importance of our feature representation and stacking architecture, especially the multihead attention mechanism. Therefore, AMPpred-MFA can be considered a reliable and efficient approach to understanding and predicting AMPs.
Collapse
Affiliation(s)
- Changjiang Li
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Quan Zou
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Cangzhi Jia
- School of Science, Dalian Maritime University, Dalian 116026, China
| | - Jia Zheng
- School of Science, Dalian Maritime University, Dalian 116026, China
| |
Collapse
|
48
|
Salvati B, Flórez-Castillo JM, Santagapita PR, Barja BC, Perullini M. One-pot synthesis of alginate-antimicrobial peptide nanogel. Photochem Photobiol Sci 2024; 23:665-679. [PMID: 38443738 DOI: 10.1007/s43630-024-00542-5] [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: 10/09/2023] [Accepted: 01/23/2024] [Indexed: 03/07/2024]
Abstract
Nanosized alginate-based particles (NAPs) were obtained in a one-pot solvent-free synthesis procedure, achieving the design of a biocompatible nanocarrier for the encapsulation of IbM6 antimicrobial peptide (IbM6). IbM6 is integrated in the nascent nanosized hydrogel self-assembly guided by electrostatic interactions and by weak interactions, typical of soft matter. The formation of the nanogel is a dynamic and complex process, which presents an interesting temporal evolution. In this work, we optimized the synthesis conditions of IbM6-NAPs based on small-angle X-ray scattering (SAXS) measurements and evaluated its time evolution over several weeks by sensing the IbM6 environment in IbM6-NAPs from photochemical experiments. Fluorescence deactivation experiments revealed that the accessibility of different quenchers to the IbM6 peptide embedded in NAPs is dependent on the aging time of the alginate network. Lifetimes measurements indicate that the deactivation paths of the excited state of the IbM6 in the nanoaggregates are reduced when compared with those exhibited by the peptide in aqueous solution, and are also dependent on the aging time of the nanosized alginate network. Finally, the entrapment of IbM6 in NAPs hinders the degradation of the peptide by trypsin, increasing its antimicrobial activity against Escherichia coli K-12 in simulated operation conditions.
Collapse
Affiliation(s)
- Brianne Salvati
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Inorgánica, Analítica y Química Física (DQIAQF), Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Química de Materiales medio Ambiente y Energía (INQUIMAE), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Johanna Marcela Flórez-Castillo
- Universidad de Magdalena, Santa Marta, Colombia
- Universidad de Santander UDES, Grupo de Investigación en Ciencias Básicas y Aplicadas para la Sostenibilidad-CIBAS, Santander, Colombia
| | - Patricio Román Santagapita
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Orgánica, Universidad de Buenos Aires, Buenos Aires, Argentina
- Centro de Investigaciones en Hidratos de Carbono (CIHIDECAR), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Beatriz C Barja
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Inorgánica, Analítica y Química Física (DQIAQF), Universidad de Buenos Aires, Buenos Aires, Argentina.
- Instituto de Química de Materiales medio Ambiente y Energía (INQUIMAE), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina.
| | - Mercedes Perullini
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Inorgánica, Analítica y Química Física (DQIAQF), Universidad de Buenos Aires, Buenos Aires, Argentina.
- Instituto de Química de Materiales medio Ambiente y Energía (INQUIMAE), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
49
|
Tsai CT, Lin CW, Ye GL, Wu SC, Yao P, Lin CT, Wan L, Tsai HHG. Accelerating Antimicrobial Peptide Discovery for WHO Priority Pathogens through Predictive and Interpretable Machine Learning Models. ACS OMEGA 2024; 9:9357-9374. [PMID: 38434814 PMCID: PMC10905719 DOI: 10.1021/acsomega.3c08676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/19/2023] [Accepted: 01/19/2024] [Indexed: 03/05/2024]
Abstract
The escalating menace of multidrug-resistant (MDR) pathogens necessitates a paradigm shift from conventional antibiotics to innovative alternatives. Antimicrobial peptides (AMPs) emerge as a compelling contender in this arena. Employing in silico methodologies, we can usher in a new era of AMP discovery, streamlining the identification process from vast candidate sequences, thereby optimizing laboratory screening expenditures. Here, we unveil cutting-edge machine learning (ML) models that are both predictive and interpretable, tailored for the identification of potent AMPs targeting World Health Organization's (WHO) high-priority pathogens. Furthermore, we have developed ML models that consider the hemolysis of human erythrocytes, emphasizing their therapeutic potential. Anchored in the nuanced physical-chemical attributes gleaned from the three-dimensional (3D) helical conformations of AMPs, our optimized models have demonstrated commendable performance-boasting an accuracy exceeding 75% when evaluated against both low-sequence-identified peptides and recently unveiled AMPs. As a testament to their efficacy, we deployed these models to prioritize peptide sequences stemming from PEM-2 and subsequently probed the bioactivity of our algorithm-predicted peptides vis-à-vis WHO's priority pathogens. Intriguingly, several of these new AMPs outperformed the native PEM-2 in their antimicrobial prowess, thereby underscoring the robustness of our modeling approach. To elucidate ML model outcomes, we probe via Shapley Additive exPlanations (SHAP) values, uncovering intricate mechanisms guiding diverse actions against bacteria. Our state-of-the-art predictive models expedite the design of new AMPs, offering a robust countermeasure to antibiotic resistance. Our prediction tool is available to the public at https://ai-meta.chem.ncu.edu.tw/amp-meta.
Collapse
Affiliation(s)
- Cheng-Ting Tsai
- Department
of Chemistry, National Central University, No. 300, Zhongda Road, Zhongli District, Taoyuan 32001, Taiwan
| | - Chia-Wei Lin
- Department
of Chemistry, National Central University, No. 300, Zhongda Road, Zhongli District, Taoyuan 32001, Taiwan
| | - Gen-Lin Ye
- Department
of Chemistry, National Central University, No. 300, Zhongda Road, Zhongli District, Taoyuan 32001, Taiwan
| | - Shao-Chi Wu
- Department
of Chemistry, National Central University, No. 300, Zhongda Road, Zhongli District, Taoyuan 32001, Taiwan
| | - Philip Yao
- Aurora
High School, 109 W Pioneer Trail, Aurora, Ohio 44202, United States
| | - Ching-Ting Lin
- School
of Chinese Medicine, China Medical University, No. 91 Hsueh-Shih Road, Taichung 40402, Taiwan
| | - Lei Wan
- School
of Chinese Medicine, China Medical University, No. 91 Hsueh-Shih Road, Taichung 40402, Taiwan
| | - Hui-Hsu Gavin Tsai
- Department
of Chemistry, National Central University, No. 300, Zhongda Road, Zhongli District, Taoyuan 32001, Taiwan
- Research
Center of New Generation Light Driven Photovoltaic Modules, National Central University, Taoyuan 32001, Taiwan
| |
Collapse
|
50
|
Aguilera-Puga MDC, Cancelarich NL, Marani MM, de la Fuente-Nunez C, Plisson F. Accelerating the Discovery and Design of Antimicrobial Peptides with Artificial Intelligence. Methods Mol Biol 2024; 2714:329-352. [PMID: 37676607 DOI: 10.1007/978-1-0716-3441-7_18] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
Peptides modulate many processes of human physiology targeting ion channels, protein receptors, or enzymes. They represent valuable starting points for the development of new biologics against communicable and non-communicable disorders. However, turning native peptide ligands into druggable materials requires high selectivity and efficacy, predictable metabolism, and good safety profiles. Machine learning models have gradually emerged as cost-effective and time-saving solutions to predict and generate new proteins with optimal properties. In this chapter, we will discuss the evolution and applications of predictive modeling and generative modeling to discover and design safe and effective antimicrobial peptides. We will also present their current limitations and suggest future research directions, applicable to peptide drug design campaigns.
Collapse
Affiliation(s)
- Mariana D C Aguilera-Puga
- Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), Unidad de Genómica Avanzada, Laboratorio Nacional de Genómica para la Biodiversidad (Langebio), Irapuato, Guanajuato, Mexico
- CINVESTAV-IPN, Unidad Irapuato, Departamento de Biotecnología y Bioquímica, Irapuato, Guanajuato, Mexico
| | - Natalia L Cancelarich
- Instituto Patagónico para el Estudio de los Ecosistemas Continentales (IPEEC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Puerto Madryn, Argentina
| | - Mariela M Marani
- Instituto Patagónico para el Estudio de los Ecosistemas Continentales (IPEEC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Puerto Madryn, Argentina
| | - Cesar de la Fuente-Nunez
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA.
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA, USA.
| | - Fabien Plisson
- Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), Unidad de Genómica Avanzada, Laboratorio Nacional de Genómica para la Biodiversidad (Langebio), Irapuato, Guanajuato, Mexico.
- CINVESTAV-IPN, Unidad Irapuato, Departamento de Biotecnología y Bioquímica, Irapuato, Guanajuato, Mexico.
| |
Collapse
|