1
|
Li M, Zhao P, Wang J, Zhang X, Li J. Functional antimicrobial peptide-loaded 3D scaffolds for infected bone defect treatment with AI and multidimensional printing. MATERIALS HORIZONS 2025; 12:20-36. [PMID: 39484845 DOI: 10.1039/d4mh01124d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Infection is the most prevalent complication of fractures, particularly in open fractures, and often leads to severe consequences. The emergence of bacterial resistance has significantly exacerbated the burden of infection in clinical practice, making infection control a significant treatment challenge for infectious bone defects. The implantation of a structural stent is necessary to treat large bone defects despite the increased risk of infection. Therefore, there is a need for the development of novel antibacterial therapies. The advancement in antibacterial biomaterials and new antimicrobial drugs offers fresh perspectives on antibacterial treatment. Although antimicrobial 3D scaffolds are currently under intense research focus, relying solely on material properties or antibiotic action remains insufficient. Antimicrobial peptides (AMPs) are one of the most promising new antibacterial therapy approaches. This review discusses the underlying mechanisms behind infectious bone defects and presents research findings on antimicrobial peptides, specifically emphasizing their mechanisms and optimization strategies. We also explore the potential prospects of utilizing antimicrobial peptides in treating infectious bone defects. Furthermore, we propose that artificial intelligence (AI) algorithms can be utilized for predicting the pharmacokinetic properties of AMPs, including absorption, distribution, metabolism, and excretion, and by combining information from genomics, proteomics, metabolomics, and clinical studies with computational models driven by machine learning algorithms, scientists can gain a comprehensive understanding of AMPs' mechanisms of action, therapeutic potential, and optimizing treatment strategies tailored to individual patients, and through interdisciplinary collaborations between computer scientists, biologists, and clinicians, the full potential of AI in accelerating the discovery and development of novel AMPs will be realized. Besides, with the continuous advancements in 3D/4D/5D/6D technology and its integration into bone scaffold materials, we anticipate remarkable progress in the field of regenerative medicine. This review summarizes relevant research on the optimal future for the treatment of infectious bone defects, provides guidance for future novel treatment strategies combining multi-dimensional printing with new antimicrobial agents, and provides a novel and effective solution to the current challenges in the field of bone regeneration.
Collapse
Affiliation(s)
- Mengmeng Li
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
- Trauma Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Peizhang Zhao
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
- Trauma Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Jingwen Wang
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
- Trauma Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xincai Zhang
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA.
| | - Jun Li
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
- Trauma Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| |
Collapse
|
2
|
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 DOI: 10.1111/1751-7915.70072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [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)
- Carlos A Brizuela
- Department of Computer Science, CICESE Research Center, Ensenada, Mexico
| | - Gary Liu
- Department of Biochemistry and Biomedical Sciences, Michael G. DeGroote Institute for Infectious Disease Research, David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, Ontario, Canada
| | - Jonathan M Stokes
- Department of Biochemistry and Biomedical Sciences, Michael G. DeGroote Institute for Infectious Disease Research, David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, Ontario, Canada
| | - 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 Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department 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
|
3
|
Chen S, Qi H, Zhu X, Liu T, Fan Y, Su Q, Gong Q, Jia C, Liu T. Screening and identification of antimicrobial peptides from the gut microbiome of cockroach Blattella germanica. MICROBIOME 2024; 12:272. [PMID: 39709489 DOI: 10.1186/s40168-024-01985-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 11/21/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND The overuse of antibiotics has led to lethal multi-antibiotic-resistant microorganisms around the globe, with restricted availability of novel antibiotics. Compared to conventional antibiotics, evolutionarily originated antimicrobial peptides (AMPs) are promising alternatives to address these issues. The gut microbiome of Blattella germanica represents a previously untapped resource of naturally evolving AMPs for developing antimicrobial agents. RESULTS Using the in-house designed tool "AMPidentifier," AMP candidates were mined from the gut microbiome of B. germanica, and their activities were validated both in vitro and in vivo. Among filtered candidates, AMP1, derived from the symbiotic microorganism Blattabacterium cuenoti, demonstrated broad-spectrum antibacterial activity, low cytotoxicity towards mammalian cells, and a lack of hemolytic effects. Mechanistic studies revealed that AMP1 rapidly permeates the bacterial cell and accumulates intracellularly, resulting in a gradual and mild depolarization of the cell membrane during the initial incubation period, suggesting minimal direct impact on membrane integrity. Furthermore, observations from fluorescence microscopy and scanning electron microscopy indicated abnormalities in bacterial binary fission and compromised cell structure. These findings led to the hypothesis that AMP1 may inhibit bacterial cell wall synthesis. Furthermore, AMP1 showed potent antibacterial and wound healing effects in mice, with comparable performances of vancomycin. CONCLUSIONS This study exemplifies an interdisciplinary approach to screening safe and effective AMPs from natural biological tissues, and our identified AMP 1 holds promising potential for clinical application.
Collapse
Affiliation(s)
- Sizhe Chen
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, 116024, China
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- The Department of Medicine & Therapeutics, The Chinese University of Hong Kong, ShatinHong Kong SAR, NT, China
| | - Huitang Qi
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, 116024, China
| | - Xingzhuo Zhu
- Department of Thoracic Surgery, The First Affiliated Hospital of Xiaan Jiaotong University, Xian, 710061, China
| | - Tianxiang Liu
- School of Science, Dalian Maritime University, Dalian, 116026, China
| | - Yuting Fan
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, 116024, China
| | - Qi Su
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- The Department of Medicine & Therapeutics, The Chinese University of Hong Kong, ShatinHong Kong SAR, NT, China
| | - Qiuyu Gong
- Department of Thoracic Surgery, The First Affiliated Hospital of Xiaan Jiaotong University, Xian, 710061, China.
| | - Cangzhi Jia
- School of Science, Dalian Maritime University, Dalian, 116026, China.
| | - Tian Liu
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, 116024, China.
| |
Collapse
|
4
|
Al-Omari AM, Akkam YH, Zyout A, Younis S, Tawalbeh SM, Al-Sawalmeh K, Al Fahoum A, Arnold J. Accelerating antimicrobial peptide design: Leveraging deep learning for rapid discovery. PLoS One 2024; 19:e0315477. [PMID: 39705302 DOI: 10.1371/journal.pone.0315477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 11/26/2024] [Indexed: 12/22/2024] Open
Abstract
Antimicrobial peptides (AMPs) are excellent at fighting many different infections. This demonstrates how important it is to make new AMPs that are even better at eliminating infections. The fundamental transformation in a variety of scientific disciplines, which led to the emergence of machine learning techniques, has presented significant opportunities for the development of antimicrobial peptides. Machine learning and deep learning are used to predict antimicrobial peptide efficacy in the study. The main purpose is to overcome traditional experimental method constraints. Gram-negative bacterium Escherichia coli is the model organism in this study. The investigation assesses 1,360 peptide sequences that exhibit anti- E. coli activity. These peptides' minimal inhibitory concentrations have been observed to be correlated with a set of 34 physicochemical characteristics. Two distinct methodologies are implemented. The initial method involves utilizing the pre-computed physicochemical attributes of peptides as the fundamental input data for a machine-learning classification approach. In the second method, these fundamental peptide features are converted into signal images, which are then transmitted to a deep learning neural network. The first and second methods have accuracy of 74% and 92.9%, respectively. The proposed methods were developed to target a single microorganism (gram negative E.coli), however, they offered a framework that could potentially be adapted for other types of antimicrobial, antiviral, and anticancer peptides with further validation. Furthermore, they have the potential to result in significant time and cost reductions, as well as the development of innovative AMP-based treatments. This research contributes to the advancement of deep learning-based AMP drug discovery methodologies by generating potent peptides for drug development and application. This discovery has significant implications for the processing of biological data and the computation of pharmacology.
Collapse
Affiliation(s)
- Ahmad M Al-Omari
- Biomedical Systems and Informatics Engineering Department, College of Engineering, Yarmouk University, Irbid, Jordan
| | - Yazan H Akkam
- Medicinal Chemistry and Pharmacognosy Department, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan
| | - Ala'a Zyout
- Biomedical Systems and Informatics Engineering Department, College of Engineering, Yarmouk University, Irbid, Jordan
| | - Shayma'a Younis
- Biomedical Systems and Informatics Engineering Department, College of Engineering, Yarmouk University, Irbid, Jordan
| | - Shefa M Tawalbeh
- Biomedical Systems and Informatics Engineering Department, College of Engineering, Yarmouk University, Irbid, Jordan
| | - Khaled Al-Sawalmeh
- Department of Basic Pathological Sciences, College of Medicine, Yarmouk University, Irbid, Jordan
| | - Amjed Al Fahoum
- Biomedical Systems and Informatics Engineering Department, College of Engineering, Yarmouk University, Irbid, Jordan
| | - Jonathan Arnold
- Genetics Department, University of Georgia, Athens, GA, United States of America
| |
Collapse
|
5
|
Gao Q, Ge L, Wang Y, Zhu Y, Liu Y, Zhang H, Huang J, Qin Z. An explainable few-shot learning model for the directed evolution of antimicrobial peptides. Int J Biol Macromol 2024; 285:138272. [PMID: 39631577 DOI: 10.1016/j.ijbiomac.2024.138272] [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: 10/04/2024] [Revised: 11/20/2024] [Accepted: 11/30/2024] [Indexed: 12/07/2024]
Abstract
Due to the persistent threat of antibiotic resistance posed by Gram-negative pathogens, the discovery of new antimicrobial agents is of critical importance. In this study, we employed deep learning-guided directed evolution to explore the chemical space of antimicrobial peptides (AMPs), which present promising alternatives to traditional small-molecule antibiotics. Utilizing a fine-tuned protein language model tailored for small dataset learning, we achieved structural modifications of the lipopolysaccharide-binding domain (LBD) derived from Marsupenaeus japonicus, a prawn species of considerable value in aquaculture and commercial fisheries. The engineered LBDs demonstrated exceptional activity against a range of Gram-negative pathogens. Drawing inspiration from evolutionary principles, we elucidated the bactericidal mechanism through molecular dynamics simulations and mapped the directed evolution pathways using a ladderpath framework. This work highlights the efficacy of explainable few-shot learning in the rational design of AMPs through directed evolution.
Collapse
Affiliation(s)
- Qiandi Gao
- Center for Biological Science and Technology, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, Guangdong 519087, China
| | - Liangjun Ge
- Center for Biological Science and Technology, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, Guangdong 519087, China
| | - Yihan Wang
- Center for Biological Science and Technology, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, Guangdong 519087, China
| | - Yanran Zhu
- Center for Biological Science and Technology, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, Guangdong 519087, China
| | - Yu Liu
- International Academic Center of Complex Systems, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, Guangdong 519087, China
| | - Heqian Zhang
- Center for Biological Science and Technology, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, Guangdong 519087, China.
| | - Jiaquan Huang
- Center for Biological Science and Technology, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, Guangdong 519087, China.
| | - Zhiwei Qin
- Center for Biological Science and Technology, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, Guangdong 519087, China.
| |
Collapse
|
6
|
Bello-Madruga R, Torrent Burgas M. The limits of prediction: Why intrinsically disordered regions challenge our understanding of antimicrobial peptides. Comput Struct Biotechnol J 2024; 23:972-981. [PMID: 38404711 PMCID: PMC10884422 DOI: 10.1016/j.csbj.2024.02.008] [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: 11/28/2023] [Revised: 02/10/2024] [Accepted: 02/10/2024] [Indexed: 02/27/2024] Open
Abstract
Antimicrobial peptides (AMPs) are molecules found in most organisms, playing a vital role in innate immune defense against pathogens. Their mechanism of action involves the disruption of bacterial cell membranes, causing leakage of cellular contents and ultimately leading to cell death. While AMPs typically lack a defined structure in solution, they often assume a defined conformation when interacting with bacterial membranes. Given this structural flexibility, we investigated whether intrinsically disordered regions (IDRs) with AMP-like properties could exhibit antimicrobial activity. We tested 14 peptides from different IDRs predicted to have antimicrobial activity and found that nearly all of them did not display the anticipated effects. These peptides failed to adopt a defined secondary structure and had compromised membrane interactions, resulting in a lack of antimicrobial activity. We hypothesize that evolutionary constraints may prevent IDRs from folding, even in membrane-like environments, limiting their antimicrobial potential. Moreover, our research reveals that current antimicrobial predictors fail to accurately capture the structural features of peptides when dealing with intrinsically unstructured sequences. Hence, the results presented here may have far-reaching implications for designing and improving antimicrobial strategies and therapies against infectious diseases.
Collapse
Affiliation(s)
- Roberto Bello-Madruga
- The Systems Biology of Infection Lab, Department of Biochemistry and Molecular Biology, Biosciences Faculty, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
| | - Marc Torrent Burgas
- The Systems Biology of Infection Lab, Department of Biochemistry and Molecular Biology, Biosciences Faculty, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
| |
Collapse
|
7
|
Wang Y, Song M, Chang W. Antimicrobial peptides and proteins against drug-resistant pathogens. Cell Surf 2024; 12:100135. [PMID: 39687062 PMCID: PMC11646788 DOI: 10.1016/j.tcsw.2024.100135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/24/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
The rise of drug-resistant pathogens, driven by the misuse and overuse of antibiotics, has created a formidable challenge for global public health. Antimicrobial peptides and proteins have garnered considerable attention as promising candidates for novel antimicrobial agents. These bioactive molecules, whether derived from natural sources, designed synthetically, or predicted using artificial intelligence, can induce lethal effects on pathogens by targeting key microbial structures or functional components, such as cell membranes, cell walls, biofilms, and intracellular components. Additionally, they may enhance overall immune defenses by modulating innate or adaptive immune responses in the host. Of course, development of antimicrobial peptides and proteins also face some limitations, including high toxicity, lack of selectivity, insufficient stability, and potential immunogenicity. Despite these challenges, they remain a valuable resource in the fight against drug-resistant pathogens. Future research should focus on overcoming these limitations to fully realize the therapeutic potential of antimicrobial peptides in the infection control.
Collapse
Affiliation(s)
- Yeji Wang
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Minghui Song
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wenqiang Chang
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
8
|
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
|
9
|
Sun S. Progress in the Identification and Design of Novel Antimicrobial Peptides Against Pathogenic Microorganisms. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10402-4. [PMID: 39557756 DOI: 10.1007/s12602-024-10402-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [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
|
10
|
Li S, Peng L, Chen L, Que L, Kang W, Hu X, Ma J, Di Z, Liu Y. Discovery of Highly Bioactive Peptides through Hierarchical Structural Information and Molecular Dynamics Simulations. J Chem Inf Model 2024; 64:8164-8175. [PMID: 39466714 DOI: 10.1021/acs.jcim.4c01006] [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: 10/30/2024]
Abstract
Peptide drugs play an essential role in modern therapeutics, but the computational design of these molecules is hindered by several challenges. Traditional methods like molecular docking and molecular dynamics (MD) simulation, as well as recent deep learning approaches, often face limitations related to computational resource demands, complex binding affinity assessments, extensive data requirements, and poor model interpretability. Here, we introduce PepHiRe, an innovative methodology that utilizes the hierarchical structural information in peptide sequences and employs a novel strategy called Ladderpath, rooted in algorithmic information theory, to rapidly generate and enhance the efficiency and clarity of novel peptide design. We applied PepHiRe to develop BH3-like peptide inhibitors targeting myeloid cell leukemia-1, a protein associated with various cancers. By analyzing just eight known bioactive BH3 peptide sequences, PepHiRe effectively derived a hierarchy of subsequences used to create new BH3-like peptides. These peptides underwent screening through MD simulations, leading to the selection of five candidates for synthesis and subsequent in vitro testing. Experimental results demonstrated that these five peptides possess high inhibitory activity, with IC50 values ranging from 28.13 ± 7.93 to 167.42 ± 22.15 nM. Our study explores a white-box model driven technique and a structured screening pipeline for identifying and generating novel peptides with potential bioactivity.
Collapse
Affiliation(s)
- Shu Li
- Centre of Artificial Intelligence Driven Drug Discovery, Faculty of Applied Science, Macao Polytechnic University, Macao SAR 999078, China
| | - Lu Peng
- Department of Systems Science, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai 519087, China
- International Academic Center of Complex Systems, Beijing Normal University, Zhuhai 519087, China
- School of Systems Science, Beijing Normal University, Beijing 100875, China
| | - Liuqing Chen
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Linjie Que
- Department of Systems Science, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai 519087, China
- International Academic Center of Complex Systems, Beijing Normal University, Zhuhai 519087, China
| | - Wenqingqing Kang
- Centre of Artificial Intelligence Driven Drug Discovery, Faculty of Applied Science, Macao Polytechnic University, Macao SAR 999078, China
| | - Xiaojun Hu
- Department of Systems Science, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai 519087, China
- International Academic Center of Complex Systems, Beijing Normal University, Zhuhai 519087, China
- School of Systems Science, Beijing Normal University, Beijing 100875, China
| | - Jun Ma
- Department of Systems Science, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai 519087, China
- International Academic Center of Complex Systems, Beijing Normal University, Zhuhai 519087, China
- School of Systems Science, Beijing Normal University, Beijing 100875, China
| | - Zengru Di
- Department of Systems Science, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai 519087, China
- International Academic Center of Complex Systems, Beijing Normal University, Zhuhai 519087, China
| | - Yu Liu
- Department of Systems Science, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai 519087, China
- International Academic Center of Complex Systems, Beijing Normal University, Zhuhai 519087, China
| |
Collapse
|
11
|
Fang Y, Ma Y, Yu K, Dong J, Zeng W. Integrated computational approaches for advancing antimicrobial peptide development. Trends Pharmacol Sci 2024; 45:1046-1060. [PMID: 39490363 DOI: 10.1016/j.tips.2024.09.011] [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: 08/23/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 11/05/2024]
Abstract
The increasing prevalence of antimicrobial resistance has intensified the need for novel antimicrobial drugs. Antimicrobial peptides (AMPs) are promising alternative antibiotics due to their broad-spectrum activity and slower resistance development. However, the time-consuming, costly development and challenge of systematic optimization limit their translation into the clinic. Recently, integrating computational methods have led to breakthroughs in the precise design and optimization of AMPs, reduced resource consumption, and accelerated AMP development process. We highlight the application of these integrated approaches in AMP molecule discovery, optimization, and delivery and demonstrate the synergy of these strategies to fuel AMP development.
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
| | - Yeshuo Ma
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410083, PR China; The Third Xiangya Hospital, Central South University, Changsha 410083, PR China
| | - Kunqian Yu
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, 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
|
12
|
Wang XF, Tang JY, Sun J, Dorje S, Sun TQ, Peng B, Ji XW, Li Z, Zhang XE, Wang DB. ProT-Diff: A Modularized and Efficient Strategy for De Novo Generation of Antimicrobial Peptide Sequences by Integrating Protein Language and Diffusion Models. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406305. [PMID: 39319609 DOI: 10.1002/advs.202406305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 09/08/2024] [Indexed: 09/26/2024]
Abstract
Antimicrobial peptides (AMPs) are a promising solution for treating antibiotic-resistant pathogens. However, efficient generation of diverse AMPs without prior knowledge of peptide structures or sequence alignments remains a challenge. Here, ProT-Diff is introduced, a modularized deep generative approach that combines a pretrained protein language model with a diffusion model for the de novo generation of AMPs sequences. ProT-Diff generates thousands of AMPs with diverse lengths and structures within a few hours. After silico physicochemical screening, 45 peptides are selected for experimental validation. Forty-four peptides showed antimicrobial activity against both gram-positive or gram-negative bacteria. Among broad-spectrum peptides, AMP_2 exhibited potent antimicrobial activity, low hemolysis, and minimal cytotoxicity. An in vivo assessment demonstrated its effectiveness against a drug-resistant E. coli strain in acute peritonitis. This study not only introduces a viable and user-friendly strategy for de novo generation of antimicrobial peptides, but also provides potential antimicrobial drug candidates with excellent activity. It is believed that this study will facilitate the development of other peptide-based drug candidates in the future, as well as proteins with tailored characteristics.
Collapse
Affiliation(s)
- Xue-Fei Wang
- Precision Scientific (Beijing) Co. Ltd., Beijing, 100085, China
| | - Jing-Ya Tang
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Science, Beijing, 100049, China
| | - Jing Sun
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- Department of Biotechnology, School of Life Sciences, Shandong Normal University, Jinan, 250014, China
| | - Sonam Dorje
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Science, Beijing, 100049, China
| | - Tian-Qi Sun
- Precision Scientific (Beijing) Co. Ltd., Beijing, 100085, China
| | - Bo Peng
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Science, Beijing, 100049, China
| | - Xu-Wo Ji
- Precision Scientific (Beijing) Co. Ltd., Beijing, 100085, China
| | - Zhe Li
- Precision Scientific (Beijing) Co. Ltd., Beijing, 100085, China
| | - Xian-En Zhang
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- Faculty of Synthetic Biology, Shenzhen Institute of Advances Technology, Shenzhen, 518055, China
| | - Dian-Bing Wang
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
13
|
Sun X, Li Y, Wang M, Amakye WK, Ren J, Matsui T, Wang W, Tsopmo A, Udenigwe CC, Giblin L, Du M, Mine Y, De Mejia E, Aluko RE, Wu J. Research Progress on Food-Derived Bioactive Peptides: An Overview of the 3rd International Symposium on Bioactive Peptides. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:23709-23715. [PMID: 39405493 DOI: 10.1021/acs.jafc.4c02854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/01/2024]
Abstract
Interest in food-derived bioactive peptides is on the rise. In 2023, the 3rd International Symposium on Bioactive Peptides (ISBP) was held in Niagara Falls, Canada, to provide a platform for knowledge exchange, networking, and collaboration among researchers in this field. This article aims to provide a high-level overview of the key progress and emerging trends in bioactive peptides based on the 3rd ISBP. This review highlights the production of bioactive peptides from sustainable sources through the integration of artificial intelligence and wet-lab research, the emerging roles of bioactive peptides in cognitive function, and the ability of peptides to act as taste modifiers. The emerging research trend in bioactive peptides focuses on utilizing novel processing technologies, understanding peptide-receptor interactions, applying omics in mechanistic studies, conducting clinical trials, and facilitating product development and commercialization.
Collapse
Affiliation(s)
- Xiaohong Sun
- Department of Plant, Food and Environmental Sciences, Faculty of Agriculture, Dalhousie University, Truro, Nova Scotia B2N 5E3, Canada
| | - Yonghui Li
- Department of Grain Science and Technology, Kansas State University, Manhattan, Kansas 66506, United States
| | - Min Wang
- School of Food Sciences and Engineering, South China University of Technology, Guangzhou 510641, China
| | - William Kwame Amakye
- School of Food Sciences and Engineering, South China University of Technology, Guangzhou 510641, China
| | - Jiaoyan Ren
- School of Food Sciences and Engineering, South China University of Technology, Guangzhou 510641, China
| | - Toshiro Matsui
- Faculty of Agriculture, Kyushu University, 744 Mototoka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Wenli Wang
- Department of Food Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Apollinaire Tsopmo
- Department of Chemistry, Carleton University, Ottawa, ON K1S 5B6, Canada
| | - Chibuike C Udenigwe
- School of Nutrition Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Linda Giblin
- Teagasc Food Research Centre, Moorepark, Fermoy, County Cork P61 C996, Ireland
| | - Ming Du
- School of Food Science and Technology, Collaborative Innovation Center of Seafood Deep Processing, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, China
| | - Yoshinori Mine
- Department of Food Science, University of Guelph, Guelph, ON N1G2W1, Canada
| | - Elvira De Mejia
- Department of Food Science & Human Nutrition, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Rotimi E Aluko
- Department of Food & Human Nutritional Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Jianping Wu
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| |
Collapse
|
14
|
Vladkova TG, Smani Y, Martinov BL, Gospodinova DN. Recent Progress in Terrestrial Biota Derived Antibacterial Agents for Medical Applications. Molecules 2024; 29:4889. [PMID: 39459256 PMCID: PMC11510244 DOI: 10.3390/molecules29204889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/07/2024] [Accepted: 10/13/2024] [Indexed: 10/28/2024] Open
Abstract
Conventional antibiotic and multidrug treatments are becoming less and less effective and the discovery of new effective and safe antibacterial agents is becoming a global priority. Returning to a natural antibacterial product is a relatively new current trend. Terrestrial biota is a rich source of biologically active substances whose antibacterial potential has not been fully utilized. The aim of this review is to present the current state-of-the-art terrestrial biota-derived antibacterial agents inspired by natural treatments. It summarizes the most important sources and newly identified or modified antibacterial agents and treatments from the last five years. It focuses on the significance of plant- animal- and bacteria-derived biologically active agents as powerful alternatives to antibiotics, as well as the advantages of utilizing natural antibacterial molecules alone or in combination with antibiotics. The main conclusion is that terrestrial biota-derived antibacterial products and substances open a variety of new ways for modern improved therapeutic strategies. New terrestrial sources of known antibacterial agents and new antibacterial agents from terrestrial biota were discovered during the last 5 years, which are under investigation together with some long-ago known but now experiencing their renaissance for the development of new medical treatments. The use of natural antibacterial peptides as well as combinational therapy by commercial antibiotics and natural products is outlined as the most promising method for treating bacterial infections. In vivo testing and clinical trials are necessary to reach clinical application.
Collapse
Affiliation(s)
- Todorka G. Vladkova
- Department of Polymer Engineering, University of Chemical Technology and Metallurgy, 8 “Kl. Ohridski” Blvd, 1756 Sofia, Bulgaria
| | - Younes Smani
- Andalusian Center of Developmental Biology, CSIC, Junta de Andalusia, University of Pablo de Olavide, 41013 Seville, Spain;
- Department of Molecular Biology and Biochemical Engineering, Andalusian Center of Developmental Biology, CSIC, Junta de Andalusia, University of Pablo de Olavide, 41013 Seville, Spain
| | - Boris L. Martinov
- Department of Biotechnology, University of Chemical Technology and Metallurgy, 8 “Kl. Ohridski” Blvd, 1756 Sofia, Bulgaria;
| | - Dilyana N. Gospodinova
- Faculty of Electrical Engineering, Technical University of Sofia, 8 “Kl. Ohridski” Blvd, 1756 Sofia, Bulgaria;
| |
Collapse
|
15
|
Yoon J, Jo Y, Shin S. Understanding Antimicrobial Peptide Synergy: Differential Binding Interactions and Their Impact on Membrane Integrity. J Phys Chem B 2024; 128:9756-9771. [PMID: 39347577 DOI: 10.1021/acs.jpcb.4c03766] [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: 10/01/2024]
Abstract
Research on antimicrobial peptides (AMPs) has been conducted as a solution to overcome antibiotic resistance. In particular, the synergistic effect that appears when two or more AMPs are used in combination has been observed. To find an effective synergistic combination, it is necessary to understand the underlying mechanism. However, a consistent explanation for this phenomenon has not yet been provided due to limitations in experimentally determining or predicting the structure of the heteroaggregates formed by the interactions between different AMPs and the interaction of the aggregate surface with the lipid membrane surface. In this study, we conducted molecular dynamics simulations for two heterogeneous aggregates of melittin-indolicidin and pexiganan-indolicidin to observe their structures in the solution phase and their interactions with the lipid membrane. We aimed to determine how the surfaces of these aggregates interact with the lipid membrane. Due to the different amino acid residue sequence characteristics of melittin and pexiganan, we found that when the two AMPs bind to indolicidin, they form aggregates with completely different structural characteristics. Accordingly, the sequence characteristics of pexiganan, which exhibits a relatively unstable structure compared to melittin in aqueous solution or on lipid membranes, allow for a more stable interaction with the lipid membrane when forming aggregates with indolicidin, effectively inhibiting the integrity of the lipid membranes. We also found that the amino acid residues forming the surface of the AMP aggregate show differential binding strengths to different lipid species forming the lipid membrane, thereby disrupting the membrane in a way that weakens its integrity. Through this, we provided insight into the basic principle of how the synergistic effect of AMPs occurs.
Collapse
Affiliation(s)
- Jeseong Yoon
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Youngbeom Jo
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Seokmin Shin
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
16
|
Yu W, Guo X, Li X, Wei Y, Lyu Y, Zhang L, Wang J, Shan A. Novel multidomain peptide self-assembly biomaterials based on bola structure and terminal anchoring: Nanotechnology meets antimicrobial therapy. Mater Today Bio 2024; 28:101183. [PMID: 39221200 PMCID: PMC11363844 DOI: 10.1016/j.mtbio.2024.101183] [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: 03/26/2024] [Revised: 06/28/2024] [Accepted: 08/03/2024] [Indexed: 09/04/2024] Open
Abstract
To ameliorate the diminished antimicrobial efficiency and physiological stability associated with monomeric antimicrobial peptides (AMPs) molecules, future research will focus on the artificial design of self-assembling peptides to replace monomeric entities, aiming to combat the antibiotic resistance crisis caused by microbial infections. In this study, the "bola" structure was used as the foundational architecture driving molecular self-assembly, with hydrophobic amino acids at the termini to anchor and finely adjust the sequence, thereby organizing a range of novel multidomain peptides (MDPs) templates into an ABA block motif. The results indicate that FW2 (GMSI = 53.94) exhibits the highest selectivity index among all MDPs and can form spherical micelles in an aqueous medium without the addition of any exogenous additives. FW2 exhibited high stability in vitro in the presence of physiological salt ions, serum, and various pH conditions. It exhibited excellent biocompatibility and efficacy both in vivo and in vitro. Furthermore, FW2 strongly interacts with the lipid membrane and employs various synergistic mechanisms, such as reactive oxygen species (ROS) accumulation, collectively driving cellular apoptosis. This study demonstrates a straightforward strategy for designing self-assembling peptides and promotes the advancement of peptide-based biomaterials integration progress with nanotechnology.
Collapse
Affiliation(s)
- Weikang Yu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Xu Guo
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Xuefeng Li
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Yingxin Wei
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Yinfeng Lyu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Licong Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Jiajun Wang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Anshan Shan
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| |
Collapse
|
17
|
Chen J, Jia Y, Sun Y, Liu K, Zhou C, Liu C, Li D, Liu G, Zhang C, Yang T, Huang L, Zhuang Y, Wang D, Xu D, Zhong Q, Guo Y, Li A, Seim I, Jiang L, Wang L, Lee SMY, Liu Y, Wang D, Zhang G, Liu S, Wei X, Yue Z, Zheng S, Shen X, Wang S, Qi C, Chen J, Ye C, Zhao F, Wang J, Fan J, Li B, Sun J, Jia X, Xia Z, Zhang H, Liu J, Zheng Y, Liu X, Wang J, Yang H, Kristiansen K, Xu X, Mock T, Li S, Zhang W, Fan G. Global marine microbial diversity and its potential in bioprospecting. Nature 2024; 633:371-379. [PMID: 39232160 PMCID: PMC11390488 DOI: 10.1038/s41586-024-07891-2] [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: 09/09/2023] [Accepted: 07/31/2024] [Indexed: 09/06/2024]
Abstract
The past two decades has witnessed a remarkable increase in the number of microbial genomes retrieved from marine systems1,2. However, it has remained challenging to translate this marine genomic diversity into biotechnological and biomedical applications3,4. Here we recovered 43,191 bacterial and archaeal genomes from publicly available marine metagenomes, encompassing a wide range of diversity with 138 distinct phyla, redefining the upper limit of marine bacterial genome size and revealing complex trade-offs between the occurrence of CRISPR-Cas systems and antibiotic resistance genes. In silico bioprospecting of these marine genomes led to the discovery of a novel CRISPR-Cas9 system, ten antimicrobial peptides, and three enzymes that degrade polyethylene terephthalate. In vitro experiments confirmed their effectiveness and efficacy. This work provides evidence that global-scale sequencing initiatives advance our understanding of how microbial diversity has evolved in the oceans and is maintained, and demonstrates how such initiatives can be sustainably exploited to advance biotechnology and biomedicine.
Collapse
Affiliation(s)
- Jianwei Chen
- BGI Research, Qingdao, China
- BGI Research, Shenzhen, China
- Qingdao Key Laboratory of Marine Genomics and Qingdao-Europe Advanced Institute for Life Sciences, BGI Research, Qingdao, China
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | | | - Ying Sun
- BGI Research, Qingdao, China.
- Qingdao Key Laboratory of Marine Genomics and Qingdao-Europe Advanced Institute for Life Sciences, BGI Research, Qingdao, China.
| | - Kun Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | | | - Chuan Liu
- BGI Research, Shenzhen, China
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Chengsong Zhang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Tao Yang
- China National GeneBank, BGI Research, Shenzhen, China
- Guangdong Genomics Data Center, BGI Research, Shenzhen, China
| | | | - Yunyun Zhuang
- Key Laboratory of Environment and Ecology, Ministry of Education, Ocean University of China, Qingdao, China
| | - Dazhi Wang
- State Key Laboratory of Marine Environmental Science, College of the Environment and Ecology, Xiamen University, Xiamen, China
| | | | | | - Yang Guo
- BGI Research, Qingdao, China
- Center of Deep-Sea Research, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
| | | | - Inge Seim
- Marine Mammal and Marine Bioacoustics Laboratory, Institute of Deep-Sea Science and Engineering, Chinese Academy of Sciences, Sanya, China
| | - Ling Jiang
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing, China
| | - Lushan Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Simon Ming Yuen Lee
- Department of Food Science and Nutrition, and PolyU-BGI Joint Research Centre for Genomics and Synthetic Biology in Global Deep Ocean Resource, The Hong Kong Polytechnic University, Hong Kong, China
| | - Yujing Liu
- BGI Research, Qingdao, China
- Qingdao Key Laboratory of Marine Genomics and Qingdao-Europe Advanced Institute for Life Sciences, BGI Research, Qingdao, China
| | | | - Guoqiang Zhang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | | | - Xiaofeng Wei
- China National GeneBank, BGI Research, Shenzhen, China
- Guangdong Genomics Data Center, BGI Research, Shenzhen, China
| | | | - Shanmin Zheng
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | | | - Sen Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Chen Qi
- BGI Research, Shenzhen, China
| | - Jing Chen
- Guangdong Genomics Data Center, BGI Research, Shenzhen, China
| | - Chen Ye
- BGI Research, Shenzhen, China
| | | | | | - Jie Fan
- BGI Research, Qingdao, China
- Qingdao Key Laboratory of Marine Genomics and Qingdao-Europe Advanced Institute for Life Sciences, BGI Research, Qingdao, China
| | | | | | - Xiaodong Jia
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, China
| | - Zhangyong Xia
- Department of Neurology, The Second People's Hospital of Liaocheng, Liaocheng, China
| | - He Zhang
- BGI Research, Qingdao, China
- BGI Research, Shenzhen, China
| | | | | | - Xin Liu
- BGI Research, Qingdao, China
- BGI Research, Shenzhen, China
| | | | | | - Karsten Kristiansen
- BGI Research, Shenzhen, China
- Qingdao Key Laboratory of Marine Genomics and Qingdao-Europe Advanced Institute for Life Sciences, BGI Research, Qingdao, China
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Xun Xu
- BGI Research, Qingdao, China
- BGI Research, Shenzhen, China
- Qingdao Key Laboratory of Marine Genomics and Qingdao-Europe Advanced Institute for Life Sciences, BGI Research, Qingdao, China
- State Key Laboratory of Agricultural Genomics, BGI Research, Shenzhen, China
| | - Thomas Mock
- School of Environmental Sciences, University of East Anglia, Norwich Research Park, Norwich, UK.
| | - Shengying Li
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China.
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China.
| | - Wenwei Zhang
- BGI Research, Shenzhen, China.
- State Key Laboratory of Agricultural Genomics, BGI Research, Shenzhen, China.
| | - Guangyi Fan
- BGI Research, Qingdao, China.
- BGI Research, Shenzhen, China.
- Qingdao Key Laboratory of Marine Genomics and Qingdao-Europe Advanced Institute for Life Sciences, BGI Research, Qingdao, China.
- Department of Food Science and Nutrition, and PolyU-BGI Joint Research Centre for Genomics and Synthetic Biology in Global Deep Ocean Resource, The Hong Kong Polytechnic University, Hong Kong, China.
- State Key Laboratory of Agricultural Genomics, BGI Research, Shenzhen, China.
| |
Collapse
|
18
|
Li T, Ren X, Luo X, Wang Z, Li Z, Luo X, Shen J, Li Y, Yuan D, Nussinov R, Zeng X, Shi J, Cheng F. A Foundation Model Identifies Broad-Spectrum Antimicrobial Peptides against Drug-Resistant Bacterial Infection. Nat Commun 2024; 15:7538. [PMID: 39214978 PMCID: PMC11364768 DOI: 10.1038/s41467-024-51933-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
Development of potent and broad-spectrum antimicrobial peptides (AMPs) could help overcome the antimicrobial resistance crisis. We develop a peptide language-based deep generative framework (deepAMP) for identifying potent, broad-spectrum AMPs. Using deepAMP to reduce antimicrobial resistance and enhance the membrane-disrupting abilities of AMPs, we identify, synthesize, and experimentally test 18 T1-AMP (Tier 1) and 11 T2-AMP (Tier 2) candidates in a two-round design and by employing cross-optimization-validation. More than 90% of the designed AMPs show a better inhibition than penetratin in both Gram-positive (i.e., S. aureus) and Gram-negative bacteria (i.e., K. pneumoniae and P. aeruginosa). T2-9 shows the strongest antibacterial activity, comparable to FDA-approved antibiotics. We show that three AMPs (T1-2, T1-5 and T2-10) significantly reduce resistance to S. aureus compared to ciprofloxacin and are effective against skin wound infection in a female wound mouse model infected with P. aeruginosa. In summary, deepAMP expedites discovery of effective, broad-spectrum AMPs against drug-resistant bacteria.
Collapse
Affiliation(s)
- Tingting Li
- Affiliated Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, China
- Greater Bay Area Institute for Innovation, Hunan University, Guangzhou, 511300, Guangdong Province, China
| | - Xuanbai Ren
- College of Information Science and Engineering, Hunan University, Changsha, China
| | - Xiaoli Luo
- College of Information Science and Engineering, Hunan University, Changsha, China
| | - Zhuole Wang
- Affiliated Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, China
- Greater Bay Area Institute for Innovation, Hunan University, Guangzhou, 511300, Guangdong Province, China
| | - Zhenlu Li
- School of Life Science, Tianjin University, Tianjin, 300072, China
| | - Xiaoyan Luo
- College of Information Science and Engineering, Hunan University, Changsha, China
| | - Jun Shen
- Affiliated Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, China
- Greater Bay Area Institute for Innovation, Hunan University, Guangzhou, 511300, Guangdong Province, China
| | - Yun Li
- Department of Ophthalmology, The 2nd Xiangya Hospital of Central South University, Changsha, China
| | - Dan Yuan
- Affiliated Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, China
- Greater Bay Area Institute for Innovation, Hunan University, Guangzhou, 511300, Guangdong Province, China
| | - Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Laboratory of Cancer Immunometabolism, National Cancer Institute, Frederick, MD, 21702, USA
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Xiangxiang Zeng
- College of Information Science and Engineering, Hunan University, Changsha, China.
| | - Junfeng Shi
- Affiliated Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, China.
- Greater Bay Area Institute for Innovation, Hunan University, Guangzhou, 511300, Guangdong Province, China.
| | - Feixiong Cheng
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
- Genome Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA.
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
19
|
Hao Y, Liu X, Fu H, Shao X, Cai W. PGAT-ABPp: harnessing protein language models and graph attention networks for antibacterial peptide identification with remarkable accuracy. Bioinformatics 2024; 40:btae497. [PMID: 39120878 PMCID: PMC11338452 DOI: 10.1093/bioinformatics/btae497] [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: 03/29/2024] [Revised: 07/24/2024] [Accepted: 08/08/2024] [Indexed: 08/10/2024] Open
Abstract
MOTIVATION The emergence of drug-resistant pathogens represents a formidable challenge to global health. Using computational methods to identify the antibacterial peptides (ABPs), an alternative antimicrobial agent, has demonstrated advantages in further drug design studies. Most of the current approaches, however, rely on handcrafted features and underutilize structural information, which may affect prediction performance. RESULTS To present an ultra-accurate model for ABP identification, we propose a novel deep learning approach, PGAT-ABPp. PGAT-ABPp leverages structures predicted by AlphaFold2 and a pretrained protein language model, ProtT5-XL-U50 (ProtT5), to construct graphs. Then the graph attention network (GAT) is adopted to learn global discriminative features from the graphs. PGAT-ABPp outperforms the other fourteen state-of-the-art models in terms of accuracy, F1-score and Matthews Correlation Coefficient on the independent test dataset. The results show that ProtT5 has significant advantages in the identification of ABPs and the introduction of spatial information further improves the prediction performance of the model. The interpretability analysis of key residues in known active ABPs further underscores the superiority of PGAT-ABPp. AVAILABILITY AND IMPLEMENTATION The datasets and source codes for the PGAT-ABPp model are available at https://github.com/moonseter/PGAT-ABPp/.
Collapse
Affiliation(s)
- Yuelei Hao
- Research Center for Analytical Sciences, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
| | - Xuyang Liu
- Research Center for Analytical Sciences, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
| | - Haohao Fu
- Research Center for Analytical Sciences, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
| | - Xueguang Shao
- Research Center for Analytical Sciences, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
| | - Wensheng Cai
- Research Center for Analytical Sciences, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
| |
Collapse
|
20
|
Deka H, Pawar A, Battula M, Ghfar AA, Assal ME, Chikhale RV. Identification and Design of Novel Potential Antimicrobial Peptides Targeting Mycobacterial Protein Kinase PknB. Protein J 2024; 43:858-868. [PMID: 39014259 PMCID: PMC11345320 DOI: 10.1007/s10930-024-10218-9] [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] [Accepted: 06/21/2024] [Indexed: 07/18/2024]
Abstract
Antimicrobial peptides have gradually gained advantages over small molecule inhibitors for their multifunctional effects, synthesising accessibility and target specificity. The current study aims to determine an antimicrobial peptide to inhibit PknB, a serine/threonine protein kinase (STPK), by binding efficiently at the helically oriented hinge region. A library of 5626 antimicrobial peptides from publicly available repositories has been prepared and categorised based on the length. Molecular docking using ADCP helped to find the multiple conformations of the subjected peptides. For each peptide served as input the tool outputs 100 poses of the subjected peptide. To maintain an efficient binding for relatively a longer duration, only those peptides were chosen which were seen to bind constantly to the active site of the receptor protein over all the poses observed. Each peptide had different number of constituent amino acid residues; the peptides were classified based on the length into five groups. In each group the peptide length incremented upto four residues from the initial length form. Five peptides were selected for Molecular Dynamic simulation in Gromacs based on higher binding affinity. Post-dynamic analysis and the frame comparison inferred that neither the shorter nor the longer peptide but an intermediate length of 15 mer peptide bound well to the receptor. Residual substitution to the selected peptides was performed to enhance the targeted interaction. The new complexes considered were further analysed using the Elastic Network Model (ENM) for the functional site's intrinsic dynamic movement to estimate the new peptide's role. The study sheds light on prospects that besides the length of peptides, the combination of constituent residues equally plays a pivotal role in peptide-based inhibitor generation. The study envisages the challenges of fine-tuned peptide recovery and the scope of Machine Learning (ML) and Deep Learning (DL) algorithm development. As the study was primarily meant for generation of therapeutics for Tuberculosis (TB), the peptide proposed by this study demands meticulous invitro analysis prior to clinical applications.
Collapse
Affiliation(s)
- Hemchandra Deka
- SilicoScientia Private Limited, Nagananda Commercial Complex, No. 07/3, 15/1, 18th Main Road, Jayanagar 9th Block, Bengaluru, 5600413, India
| | - Atul Pawar
- SilicoScientia Private Limited, Nagananda Commercial Complex, No. 07/3, 15/1, 18th Main Road, Jayanagar 9th Block, Bengaluru, 5600413, India
| | - Monishka Battula
- Department of Bioinformatics, Rajiv Gandhi Institute of IT and Biotechnology, Bharati Vidyapeeth Deemed to be University, Pune-Satara Road, Pune, India
| | - Ayman A Ghfar
- Chemistry Department, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Mohamed E Assal
- Chemistry Department, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Rupesh V Chikhale
- Department of Pharmaceutical and Biological Chemistry, School of Pharmacy, University College London, Brunswick Square, London, UK.
| |
Collapse
|
21
|
Luo X, Hu C, Yin Q, Zhang X, Liu Z, Zhou C, Zhang J, Chen W, Yang Y. Dual-Mechanism Peptide SR25 has Broad Antimicrobial Activity and Potential Application for Healing Bacteria-infected Diabetic Wounds. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401793. [PMID: 38874469 PMCID: PMC11321617 DOI: 10.1002/advs.202401793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/12/2024] [Indexed: 06/15/2024]
Abstract
The rise of antibiotic resistance poses a significant public health crisis, particularly due to limited antimicrobial options for the treatment of infections with Gram-negative pathogens. Here, an antimicrobial peptide (AMP) SR25 is characterized, which effectively kills both Gram-negative and Gram-positive bacteria through a unique dual-targeting mechanism without detectable resistance. Meanwhile, an SR25-functionalized hydrogel is developed for the efficient treatment of infected diabetic wounds. SR25 is obtained through genome mining from an uncultured bovine enteric actinomycete named Nonomuraea Jilinensis sp. nov. Investigations reveal that SR25 has two independent cellular targets, disrupting bacterial membrane integrity and restraining the activity of succinate:quinone oxidoreductase (SQR). In a diabetic mice wound infection model, the SR25-incorporated hydrogel exhibits high efficacy against mixed infections of Escherichia coli (E. coli) and methicillin-resistant Staphylococcus aureus (MRSA), accelerating wound healing. Overall, these findings demonstrate the therapeutic potential of SR25 and highlight the value of mining drugs with multiple mechanisms from uncultured animal commensals for combating challenging bacterial pathogens.
Collapse
Affiliation(s)
- Xue‐Yue Luo
- Department of Preventive Veterinary MedicineCollege of Veterinary MedicineJilin UniversityChangchunJilin130062P. R. China
| | - Chun‐Mei Hu
- Department of Preventive Veterinary MedicineCollege of Veterinary MedicineJilin UniversityChangchunJilin130062P. R. China
| | - Qi Yin
- Department of Preventive Veterinary MedicineCollege of Veterinary MedicineJilin UniversityChangchunJilin130062P. R. China
| | - Xiao‐Mei Zhang
- Department of Preventive Veterinary MedicineCollege of Veterinary MedicineJilin UniversityChangchunJilin130062P. R. China
| | - Zhen‐Zhen Liu
- Department of Preventive Veterinary MedicineCollege of Veterinary MedicineJilin UniversityChangchunJilin130062P. R. China
| | - Cheng‐Kai Zhou
- Department of Preventive Veterinary MedicineCollege of Veterinary MedicineJilin UniversityChangchunJilin130062P. R. China
| | - Jian‐Gang Zhang
- Department of Preventive Veterinary MedicineCollege of Veterinary MedicineJilin UniversityChangchunJilin130062P. R. China
| | - Wei Chen
- Department of Preventive Veterinary MedicineCollege of Veterinary MedicineJilin UniversityChangchunJilin130062P. R. China
| | - Yong‐Jun Yang
- Department of Preventive Veterinary MedicineCollege of Veterinary MedicineJilin UniversityChangchunJilin130062P. R. China
| |
Collapse
|
22
|
Liang Q, Liu Z, Liang Z, Zhu C, Li D, Kong Q, Mou H. Development strategies and application of antimicrobial peptides as future alternatives to in-feed antibiotics. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 927:172150. [PMID: 38580107 DOI: 10.1016/j.scitotenv.2024.172150] [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: 01/12/2024] [Revised: 03/14/2024] [Accepted: 03/30/2024] [Indexed: 04/07/2024]
Abstract
The use of in-feed antibiotics has been widely restricted due to the significant environmental pollution and food safety concerns they have caused. Antimicrobial peptides (AMPs) have attracted widespread attention as potential future alternatives to in-feed antibiotics owing to their demonstrated antimicrobial activity and environment friendly characteristics. However, the challenges of weak bioactivity, immature stability, and low production yields of natural AMPs impede practical application in the feed industry. To address these problems, efforts have been made to develop strategies for approaching the AMPs with enhanced properties. Herein, we summarize approaches to improving the properties of AMPs as potential alternatives to in-feed antibiotics, mainly including optimization of structural parameters, sequence modification, selection of microbial hosts, fusion expression, and industrially fermentation control. Additionally, the potential for application of AMPs in animal husbandry is discussed. This comprehensive review lays a strong theoretical foundation for the development of in-feed AMPs to achieve the public health globally.
Collapse
Affiliation(s)
- Qingping Liang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266404, China
| | - Zhemin Liu
- Fundamental Science R&D Center of Vazyme Biotech Co. Ltd., Nanjing 210000, China
| | - Ziyu Liang
- Section of Neurobiology, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Changliang Zhu
- College of Food Science and Engineering, Ocean University of China, Qingdao 266404, China
| | - Dongyu Li
- College of Food Science and Engineering, Ocean University of China, Qingdao 266404, China
| | - Qing Kong
- College of Food Science and Engineering, Ocean University of China, Qingdao 266404, China
| | - Haijin Mou
- College of Food Science and Engineering, Ocean University of China, Qingdao 266404, China.
| |
Collapse
|
23
|
Zhang J, Sun X, Zhao H, Zhou X, Zhang Y, Xie F, Li B, Guo G. In Silico Design and Synthesis of Antifungal Peptides Guided by Quantitative Antifungal Activity. J Chem Inf Model 2024; 64:4277-4285. [PMID: 38743449 DOI: 10.1021/acs.jcim.4c00142] [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: 05/16/2024]
Abstract
Antifungal peptides (AFPs) are emerging as promising candidates for advanced antifungal therapies because of their broad-spectrum efficacy and reduced resistance development. In silico design of AFPs, however, remains challenging, due to the lack of an efficient and well-validated quantitative assessment of antifungal activity. This study introduced an AFP design approach that leverages an innovative quantitative metric, named the antifungal index (AFI), through a three-step process, i.e., segmentation, single-point mutation, and global multipoint optimization. An exhaustive search of 100 putative AFP sequences indicated that random modifications without guidance only have a 5.97-20.24% chance of enhancing antifungal activity. Analysis of the search results revealed that (1) N-terminus truncation is more effective in enhancing antifungal activity than the modifications at the C-terminus or both ends, (2) introducing the amino acids within the 10-60% sequence region that enhance aromaticity and hydrophobicity are more effective in increasing antifungal efficacy, and (3) incorporating alanine, cysteine, and phenylalanine during multiple point mutations has a synergistic effect on enhancing antifungal activity. Subsequently, 28 designed peptides were synthesized and tested against four typical fungal strains. The success rate for developing promising AFPs, with a minimal inhibitory concentration of ≤5.00 μM, was an impressive 82.14%. The predictive and design tool is accessible at https://antifungipept.chemoinfolab.com.
Collapse
Affiliation(s)
- Jin Zhang
- School of Public Health/Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang 561113, China
| | - Xinhao Sun
- School of Public Health/Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang 561113, China
| | - Hongwei Zhao
- School of Public Health/Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang 561113, China
| | - Xu Zhou
- School of Public Health/Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang 561113, China
| | - Yiling Zhang
- School of Public Health/Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang 561113, China
| | - Feng Xie
- Moutai Institute, Renhuai 564507, China
| | - Boyan Li
- School of Public Health/Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang 561113, China
| | - Guo Guo
- The Key and Characteristic Laboratory of Modern Pathogen Biology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang 561113, China
| |
Collapse
|
24
|
Goles M, Daza A, Cabas-Mora G, Sarmiento-Varón L, Sepúlveda-Yañez J, Anvari-Kazemabad H, Davari MD, Uribe-Paredes R, Olivera-Nappa Á, Navarrete MA, Medina-Ortiz D. Peptide-based drug discovery through artificial intelligence: towards an autonomous design of therapeutic peptides. Brief Bioinform 2024; 25:bbae275. [PMID: 38856172 PMCID: PMC11163380 DOI: 10.1093/bib/bbae275] [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: 02/08/2024] [Revised: 04/23/2024] [Accepted: 06/04/2024] [Indexed: 06/11/2024] Open
Abstract
With their diverse biological activities, peptides are promising candidates for therapeutic applications, showing antimicrobial, antitumour and hormonal signalling capabilities. Despite their advantages, therapeutic peptides face challenges such as short half-life, limited oral bioavailability and susceptibility to plasma degradation. The rise of computational tools and artificial intelligence (AI) in peptide research has spurred the development of advanced methodologies and databases that are pivotal in the exploration of these complex macromolecules. This perspective delves into integrating AI in peptide development, encompassing classifier methods, predictive systems and the avant-garde design facilitated by deep-generative models like generative adversarial networks and variational autoencoders. There are still challenges, such as the need for processing optimization and careful validation of predictive models. This work outlines traditional strategies for machine learning model construction and training techniques and proposes a comprehensive AI-assisted peptide design and validation pipeline. The evolving landscape of peptide design using AI is emphasized, showcasing the practicality of these methods in expediting the development and discovery of novel peptides within the context of peptide-based drug discovery.
Collapse
Affiliation(s)
- Montserrat Goles
- Departamento de Ingeniería en Computación, Universidad de Magallanes, Av. Pdte. Manuel Bulnes 01855, 6210427, Punta Arenas, Chile
- Departamento de Ingeniería Química, Biotecnología y Materiales, Universidad de Chile, Beauchef 851, 8370456, Santiago, Chile
| | - Anamaría Daza
- Centre for Biotechnology and Bioengineering, CeBiB, Universidad de Chile, Beauchef 851, 8370456, Santiago, Chile
| | - Gabriel Cabas-Mora
- Departamento de Ingeniería en Computación, Universidad de Magallanes, Av. Pdte. Manuel Bulnes 01855, 6210427, Punta Arenas, Chile
| | - Lindybeth Sarmiento-Varón
- Centro Asistencial de Docencia e Investigación, CADI, Universidad de Magallanes, Av. Los Flamencos 01364, 6210005, Punta Arenas, Chile
| | - Julieta Sepúlveda-Yañez
- Facultad de Ciencias de la Salud, Universidad de Magallanes, Av. Pdte. Manuel Bulnes 01855, 6210427, Punta Arenas, Chile
| | - Hoda Anvari-Kazemabad
- Departamento de Ingeniería en Computación, Universidad de Magallanes, Av. Pdte. Manuel Bulnes 01855, 6210427, Punta Arenas, Chile
| | - Mehdi D Davari
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, 06120, Halle, Germany
| | - Roberto Uribe-Paredes
- Departamento de Ingeniería en Computación, Universidad de Magallanes, Av. Pdte. Manuel Bulnes 01855, 6210427, Punta Arenas, Chile
| | - Álvaro Olivera-Nappa
- Centre for Biotechnology and Bioengineering, CeBiB, Universidad de Chile, Beauchef 851, 8370456, Santiago, Chile
| | - Marcelo A Navarrete
- Centro Asistencial de Docencia e Investigación, CADI, Universidad de Magallanes, Av. Los Flamencos 01364, 6210005, Punta Arenas, Chile
- Escuela de Medicina, Universidad de Magallanes, Av. Pdte. Manuel Bulnes 01855, 6210427, Punta Arenas, Chile
| | - David Medina-Ortiz
- Departamento de Ingeniería en Computación, Universidad de Magallanes, Av. Pdte. Manuel Bulnes 01855, 6210427, Punta Arenas, Chile
- Centre for Biotechnology and Bioengineering, CeBiB, Universidad de Chile, Beauchef 851, 8370456, Santiago, Chile
| |
Collapse
|
25
|
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
|
26
|
Zervou MA, Doutsi E, Pantazis Y, Tsakalides P. De Novo Antimicrobial Peptide Design with Feedback Generative Adversarial Networks. Int J Mol Sci 2024; 25:5506. [PMID: 38791544 PMCID: PMC11122239 DOI: 10.3390/ijms25105506] [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: 04/15/2024] [Revised: 05/10/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Antimicrobial peptides (AMPs) are promising candidates for new antibiotics due to their broad-spectrum activity against pathogens and reduced susceptibility to resistance development. Deep-learning techniques, such as deep generative models, offer a promising avenue to expedite the discovery and optimization of AMPs. A remarkable example is the Feedback Generative Adversarial Network (FBGAN), a deep generative model that incorporates a classifier during its training phase. Our study aims to explore the impact of enhanced classifiers on the generative capabilities of FBGAN. To this end, we introduce two alternative classifiers for the FBGAN framework, both surpassing the accuracy of the original classifier. The first classifier utilizes the k-mers technique, while the second applies transfer learning from the large protein language model Evolutionary Scale Modeling 2 (ESM2). Integrating these classifiers into FBGAN not only yields notable performance enhancements compared to the original FBGAN but also enables the proposed generative models to achieve comparable or even superior performance to established methods such as AMPGAN and HydrAMP. This achievement underscores the effectiveness of leveraging advanced classifiers within the FBGAN framework, enhancing its computational robustness for AMP de novo design and making it comparable to existing literature.
Collapse
Affiliation(s)
- Michaela Areti Zervou
- Department of Computer Science, University of Crete, 700 13 Heraklion, Greece
- Institute of Computer Science, Foundation for Research and Technology-Hellas, 700 13 Heraklion, Greece;
| | - Effrosyni Doutsi
- Institute of Computer Science, Foundation for Research and Technology-Hellas, 700 13 Heraklion, Greece;
| | - Yannis Pantazis
- Institute of Applied and Computational Mathematics, Foundation for Research and Technology-Hellas, 700 13 Heraklion, Greece;
| | - Panagiotis Tsakalides
- Department of Computer Science, University of Crete, 700 13 Heraklion, Greece
- Institute of Computer Science, Foundation for Research and Technology-Hellas, 700 13 Heraklion, Greece;
| |
Collapse
|
27
|
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
|
28
|
Zhang H, Wang Y, Zhu Y, Huang P, Gao Q, Li X, Chen Z, Liu Y, Jiang J, Gao Y, Huang J, Qin Z. Machine learning and genetic algorithm-guided directed evolution for the development of antimicrobial peptides. J Adv Res 2024:S2090-1232(24)00078-X. [PMID: 38431124 DOI: 10.1016/j.jare.2024.02.016] [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: 10/26/2023] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/05/2024] Open
Abstract
INTRODUCTION Antimicrobial peptides (AMPs) are valuable alternatives to traditional antibiotics, possess a variety of potent biological activities and exhibit immunomodulatory effects that alleviate difficult-to-treat infections. Clarifying the structure-activity relationships of AMPs can direct the synthesis of desirable peptide therapeutics. OBJECTIVES In this study, the lipopolysaccharide-binding domain (LBD) was identified through machine learning-guided directed evolution, which acts as a functional domain of the anti-lipopolysaccharide factor family of AMPs identified from Marsupenaeus japonicus. METHODS LBDA-D was identified as an output of this algorithm, in which the original LBDMj sequence was the input, and the three-dimensional solution structure of LBDB was determined using nuclear magnetic resonance. Furthermore, our study involved a comprehensive series of experiments, including morphological studies and in vitro and in vivo antibacterial tests. RESULTS The NMR solution structure showed that LBDB possesses a circular extended structure with a disulfide crosslink at the terminus and two 310-helices and exhibits a broad antimicrobial spectrum. In addition, scanning electron microscopy (SEM) and transmission electron microscopy (TEM) showed that LBDB induced the formation of a cluster of bacteria wrapped in a flexible coating that ruptured and consequently killed the bacteria. Finally, coinjection of LBDB, Vibrio alginolyticus and Staphylococcus aureus in vivo improved the survival of M. japonicus, demonstrating the promising therapeutic role of LBDB for treating infectious disease. CONCLUSIONS The findings of this study pave the way for the rational drug design of activity-enhanced peptide antibiotics.
Collapse
Affiliation(s)
- Heqian Zhang
- Center for Biological Science and Technology, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, Guangdong 519087, China
| | - Yihan Wang
- Center for Biological Science and Technology, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, Guangdong 519087, China
| | - Yanran Zhu
- Center for Biological Science and Technology, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, Guangdong 519087, China
| | - Pengtao Huang
- Center for Biological Science and Technology, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, Guangdong 519087, China
| | - Qiandi Gao
- Center for Biological Science and Technology, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, Guangdong 519087, China
| | - Xiaojie Li
- Center for Biological Science and Technology, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, Guangdong 519087, China
| | - Zhaoying Chen
- Center for Biological Science and Technology, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, Guangdong 519087, China
| | - Yu Liu
- International Academic Center of Complex Systems, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, Guangdong 519087, China
| | - Jiakun Jiang
- Center for Statistics and Data Science, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, Guangdong 519087, China
| | - Yuan Gao
- Instrumentation and Service Center for Science and Technology, Beijing Normal University, Zhuhai, Guangdong 519087, China
| | - Jiaquan Huang
- Center for Biological Science and Technology, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, Guangdong 519087, China.
| | - Zhiwei Qin
- Center for Biological Science and Technology, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, Guangdong 519087, China.
| |
Collapse
|
29
|
de la Fuente-Nunez C. AI in infectious diseases: The role of datasets. Drug Resist Updat 2024; 73:101067. [PMID: 38387282 PMCID: PMC11537278 DOI: 10.1016/j.drup.2024.101067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/07/2024] [Accepted: 02/08/2024] [Indexed: 02/24/2024]
Affiliation(s)
- 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, United States of America; Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States of America; Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, United States of America; Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA, United States of America.
| |
Collapse
|
30
|
Liu GY, Yu D, Fan MM, Zhang X, Jin ZY, Tang C, Liu XF. Antimicrobial resistance crisis: could artificial intelligence be the solution? Mil Med Res 2024; 11:7. [PMID: 38254241 PMCID: PMC10804841 DOI: 10.1186/s40779-024-00510-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Antimicrobial resistance is a global public health threat, and the World Health Organization (WHO) has announced a priority list of the most threatening pathogens against which novel antibiotics need to be developed. The discovery and introduction of novel antibiotics are time-consuming and expensive. According to WHO's report of antibacterial agents in clinical development, only 18 novel antibiotics have been approved since 2014. Therefore, novel antibiotics are critically needed. Artificial intelligence (AI) has been rapidly applied to drug development since its recent technical breakthrough and has dramatically improved the efficiency of the discovery of novel antibiotics. Here, we first summarized recently marketed novel antibiotics, and antibiotic candidates in clinical development. In addition, we systematically reviewed the involvement of AI in antibacterial drug development and utilization, including small molecules, antimicrobial peptides, phage therapy, essential oils, as well as resistance mechanism prediction, and antibiotic stewardship.
Collapse
Affiliation(s)
- Guang-Yu Liu
- Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Hangzhou Normal University, Key Laboratory of Aging and Cancer Biology of Zhejiang Province, Key Laboratory of Inflammation and Immunoregulation of Hangzhou, Hangzhou Normal University, Hangzhou, 311121, China
| | - Dan Yu
- National Key Discipline of Pediatrics Key Laboratory of Major Diseases in Children Ministry of Education, Laboratory of Dermatology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Mei-Mei Fan
- Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Hangzhou Normal University, Key Laboratory of Aging and Cancer Biology of Zhejiang Province, Key Laboratory of Inflammation and Immunoregulation of Hangzhou, Hangzhou Normal University, Hangzhou, 311121, China
| | - Xu Zhang
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Ze-Yu Jin
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Christoph Tang
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK.
| | - Xiao-Fen Liu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission of the People's Republic of China, National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
31
|
Purohit K, Reddy N, Sunna A. Exploring the Potential of Bioactive Peptides: From Natural Sources to Therapeutics. Int J Mol Sci 2024; 25:1391. [PMID: 38338676 PMCID: PMC10855437 DOI: 10.3390/ijms25031391] [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: 12/01/2023] [Revised: 01/18/2024] [Accepted: 01/21/2024] [Indexed: 02/12/2024] Open
Abstract
Bioactive peptides, specific protein fragments with positive health effects, are gaining traction in drug development for advantages like enhanced penetration, low toxicity, and rapid clearance. This comprehensive review navigates the intricate landscape of peptide science, covering discovery to functional characterization. Beginning with a peptidomic exploration of natural sources, the review emphasizes the search for novel peptides. Extraction approaches, including enzymatic hydrolysis, microbial fermentation, and specialized methods for disulfide-linked peptides, are extensively covered. Mass spectrometric analysis techniques for data acquisition and identification, such as liquid chromatography, capillary electrophoresis, untargeted peptide analysis, and bioinformatics, are thoroughly outlined. The exploration of peptide bioactivity incorporates various methodologies, from in vitro assays to in silico techniques, including advanced approaches like phage display and cell-based assays. The review also discusses the structure-activity relationship in the context of antimicrobial peptides (AMPs), ACE-inhibitory peptides (ACEs), and antioxidative peptides (AOPs). Concluding with key findings and future research directions, this interdisciplinary review serves as a comprehensive reference, offering a holistic understanding of peptides and their potential therapeutic applications.
Collapse
Affiliation(s)
- Kruttika Purohit
- School of Natural Sciences, Macquarie University, Sydney, NSW 2109, Australia;
- Australian Research Council Industrial Transformation Training Centre for Facilitated Advancement of Australia’s Bioactives (FAAB), Sydney, NSW 2109, Australia;
| | - Narsimha Reddy
- Australian Research Council Industrial Transformation Training Centre for Facilitated Advancement of Australia’s Bioactives (FAAB), Sydney, NSW 2109, Australia;
- School of Science, Parramatta Campus, Western Sydney University, Penrith, NSW 2751, Australia
| | - Anwar Sunna
- School of Natural Sciences, Macquarie University, Sydney, NSW 2109, Australia;
- Australian Research Council Industrial Transformation Training Centre for Facilitated Advancement of Australia’s Bioactives (FAAB), Sydney, NSW 2109, Australia;
- Biomolecular Discovery Research Centre, Macquarie University, Sydney, NSW 2109, Australia
| |
Collapse
|
32
|
Wang R, Wang T, Zhuo L, Wei J, Fu X, Zou Q, Yao X. Diff-AMP: tailored designed antimicrobial peptide framework with all-in-one generation, identification, prediction and optimization. Brief Bioinform 2024; 25:bbae078. [PMID: 38446739 PMCID: PMC10939340 DOI: 10.1093/bib/bbae078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/25/2024] [Accepted: 02/08/2024] [Indexed: 03/08/2024] Open
Abstract
Antimicrobial peptides (AMPs), short peptides with diverse functions, effectively target and combat various organisms. The widespread misuse of chemical antibiotics has led to increasing microbial resistance. Due to their low drug resistance and toxicity, AMPs are considered promising substitutes for traditional antibiotics. While existing deep learning technology enhances AMP generation, it also presents certain challenges. Firstly, AMP generation overlooks the complex interdependencies among amino acids. Secondly, current models fail to integrate crucial tasks like screening, attribute prediction and iterative optimization. Consequently, we develop a integrated deep learning framework, Diff-AMP, that automates AMP generation, identification, attribute prediction and iterative optimization. We innovatively integrate kinetic diffusion and attention mechanisms into the reinforcement learning framework for efficient AMP generation. Additionally, our prediction module incorporates pre-training and transfer learning strategies for precise AMP identification and screening. We employ a convolutional neural network for multi-attribute prediction and a reinforcement learning-based iterative optimization strategy to produce diverse AMPs. This framework automates molecule generation, screening, attribute prediction and optimization, thereby advancing AMP research. We have also deployed Diff-AMP on a web server, with code, data and server details available in the Data Availability section.
Collapse
Affiliation(s)
- Rui Wang
- School of Data Science and Artificial Intelligence, Wenzhou University of Technology, 325000 Wenzhou, China
| | - Tao Wang
- School of Data Science and Artificial Intelligence, Wenzhou University of Technology, 325000 Wenzhou, China
| | - Linlin Zhuo
- School of Data Science and Artificial Intelligence, Wenzhou University of Technology, 325000 Wenzhou, China
| | - Jinhang Wei
- School of Data Science and Artificial Intelligence, Wenzhou University of Technology, 325000 Wenzhou, China
| | - Xiangzheng Fu
- College of Computer Science and Electronic Engineering, Hunan University, 410012 Changsha, China
| | - Quan Zou
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, 611730 Chengdu, China
| | - Xiaojun Yao
- Faculty of Applied Sciences, Macao Polytechnic University, 999078 Macao, China
| |
Collapse
|
33
|
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: 0] [Impact Index Per Article: 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
|
34
|
Szymczak P, Szczurek E. Artificial intelligence-driven antimicrobial peptide discovery. Curr Opin Struct Biol 2023; 83:102733. [PMID: 37992451 DOI: 10.1016/j.sbi.2023.102733] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/06/2023] [Accepted: 10/30/2023] [Indexed: 11/24/2023]
Abstract
Antimicrobial peptides (AMPs) emerge as promising agents against antimicrobial resistance, providing an alternative to conventional antibiotics. Artificial intelligence (AI) revolutionized AMP discovery through both discrimination and generation approaches. The discriminators aid in the identification of promising candidates by predicting key peptide properties such as activity and toxicity, while the generators learn the distribution of peptides and enable sampling novel AMP candidates, either de novo or as analogs of a prototype peptide. Moreover, the controlled generation of AMPs with desired properties is achieved by discriminator-guided filtering, positive-only learning, latent space sampling, as well as conditional and optimized generation. Here we review recent achievements in AI-driven AMP discovery, highlighting the most exciting directions.
Collapse
Affiliation(s)
- Paulina Szymczak
- Faculty of Mathematics, Informatics and Mechanics, University of Warsaw, Banacha 2, 02-097, Warsaw, Poland.
| | - Ewa Szczurek
- Faculty of Mathematics, Informatics and Mechanics, University of Warsaw, Banacha 2, 02-097, Warsaw, Poland.
| |
Collapse
|
35
|
Gallardo-Becerra L, Cervantes-Echeverría M, Cornejo-Granados F, Vazquez-Morado LE, Ochoa-Leyva A. Perspectives in Searching Antimicrobial Peptides (AMPs) Produced by the Microbiota. MICROBIAL ECOLOGY 2023; 87:8. [PMID: 38036921 PMCID: PMC10689560 DOI: 10.1007/s00248-023-02313-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 10/24/2023] [Indexed: 12/02/2023]
Abstract
Changes in the structure and function of the microbiota are associated with various human diseases. These microbial changes can be mediated by antimicrobial peptides (AMPs), small peptides produced by the host and their microbiota, which play a crucial role in host-bacteria co-evolution. Thus, by studying AMPs produced by the microbiota (microbial AMPs), we can better understand the interactions between host and bacteria in microbiome homeostasis. Additionally, microbial AMPs are a new source of compounds against pathogenic and multi-resistant bacteria. Further, the growing accessibility to metagenomic and metatranscriptomic datasets presents an opportunity to discover new microbial AMPs. This review examines the structural properties of microbiota-derived AMPs, their molecular action mechanisms, genomic organization, and strategies for their identification in any microbiome data as well as experimental testing. Overall, we provided a comprehensive overview of this important topic from the microbial perspective.
Collapse
Affiliation(s)
- Luigui Gallardo-Becerra
- Departamento de Microbiologia Molecular, Instituto de Biotecnologia, Universidad Nacional Autonoma de Mexico (UNAM), Avenida Universidad 2001, C.P. 62210, Cuernavaca, Morelos, Mexico
| | - Melany Cervantes-Echeverría
- Departamento de Microbiologia Molecular, Instituto de Biotecnologia, Universidad Nacional Autonoma de Mexico (UNAM), Avenida Universidad 2001, C.P. 62210, Cuernavaca, Morelos, Mexico
| | - Fernanda Cornejo-Granados
- Departamento de Microbiologia Molecular, Instituto de Biotecnologia, Universidad Nacional Autonoma de Mexico (UNAM), Avenida Universidad 2001, C.P. 62210, Cuernavaca, Morelos, Mexico
| | - Luis E Vazquez-Morado
- Departamento de Microbiologia Molecular, Instituto de Biotecnologia, Universidad Nacional Autonoma de Mexico (UNAM), Avenida Universidad 2001, C.P. 62210, Cuernavaca, Morelos, Mexico
| | - Adrian Ochoa-Leyva
- Departamento de Microbiologia Molecular, Instituto de Biotecnologia, Universidad Nacional Autonoma de Mexico (UNAM), Avenida Universidad 2001, C.P. 62210, Cuernavaca, Morelos, Mexico.
| |
Collapse
|
36
|
Pandi A, Adam D, Zare A, Trinh VT, Schaefer SL, Burt M, Klabunde B, Bobkova E, Kushwaha M, Foroughijabbari Y, Braun P, Spahn C, Preußer C, Pogge von Strandmann E, Bode HB, von Buttlar H, Bertrams W, Jung AL, Abendroth F, Schmeck B, Hummer G, Vázquez O, Erb TJ. Cell-free biosynthesis combined with deep learning accelerates de novo-development of antimicrobial peptides. Nat Commun 2023; 14:7197. [PMID: 37938588 PMCID: PMC10632401 DOI: 10.1038/s41467-023-42434-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 10/10/2023] [Indexed: 11/09/2023] Open
Abstract
Bioactive peptides are key molecules in health and medicine. Deep learning holds a big promise for the discovery and design of bioactive peptides. Yet, suitable experimental approaches are required to validate candidates in high throughput and at low cost. Here, we established a cell-free protein synthesis (CFPS) pipeline for the rapid and inexpensive production of antimicrobial peptides (AMPs) directly from DNA templates. To validate our platform, we used deep learning to design thousands of AMPs de novo. Using computational methods, we prioritized 500 candidates that we produced and screened with our CFPS pipeline. We identified 30 functional AMPs, which we characterized further through molecular dynamics simulations, antimicrobial activity and toxicity. Notably, six de novo-AMPs feature broad-spectrum activity against multidrug-resistant pathogens and do not develop bacterial resistance. Our work demonstrates the potential of CFPS for high throughput and low-cost production and testing of bioactive peptides within less than 24 h.
Collapse
Affiliation(s)
- Amir Pandi
- Department of Biochemistry and Synthetic Metabolism, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany.
| | - David Adam
- Department of Biochemistry and Synthetic Metabolism, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
- Bundeswehr Institute of Microbiology, Munich, Germany
| | - Amir Zare
- Department of Biochemistry and Synthetic Metabolism, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Van Tuan Trinh
- Department of Chemistry, Philipps-University Marburg, Marburg, Germany
| | - Stefan L Schaefer
- Department of Theoretical Biophysics, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Marie Burt
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, German Center for Lung Research (DZL), Marburg, Germany
| | - Björn Klabunde
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, German Center for Lung Research (DZL), Marburg, Germany
| | - Elizaveta Bobkova
- Department of Biochemistry and Synthetic Metabolism, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Manish Kushwaha
- Université Paris-Saclay, INRAe, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Yeganeh Foroughijabbari
- Department of Biochemistry and Synthetic Metabolism, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Peter Braun
- Bundeswehr Institute of Microbiology, Munich, Germany
- German Center for Infection Research (DZIF), Munich, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Immunology, Infection and Pandemic Research, Munich, Germany
| | - Christoph Spahn
- Department of Natural Products in Organismic Interactions, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Christian Preußer
- Institute for Tumor Immunology, Center for Tumor Biology and Immunology, Philipps-University Marburg, Marburg, Germany
- Core Facility Extracellular Vesicles, Center for Tumor Biology and Immunology, Philipps-University of Marburg, Marburg, Germany
| | - Elke Pogge von Strandmann
- Institute for Tumor Immunology, Center for Tumor Biology and Immunology, Philipps-University Marburg, Marburg, Germany
- Core Facility Extracellular Vesicles, Center for Tumor Biology and Immunology, Philipps-University of Marburg, Marburg, Germany
| | - Helge B Bode
- Department of Natural Products in Organismic Interactions, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
- Molecular Biotechnology, Department of Biosciences, Goethe University Frankfurt, Frankfurt am Main, Germany
- Department of Chemistry, Chemical Biology, Philipps-University Marburg, Marburg, Germany
- Senckenberg Gesellschaft für Naturforschung, Frankfurt, Germany
- SYNMIKRO Center of Synthetic Microbiology, Marburg, Germany
| | - Heiner von Buttlar
- Bundeswehr Institute of Microbiology, Munich, Germany
- German Center for Infection Research (DZIF), Munich, Germany
| | - Wilhelm Bertrams
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, German Center for Lung Research (DZL), Marburg, Germany
| | - Anna Lena Jung
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, German Center for Lung Research (DZL), Marburg, Germany
- Core Facility Flow Cytometry - Bacterial Vesicles, Philipps-University Marburg, Marburg, Germany
| | - Frank Abendroth
- Department of Chemistry, Philipps-University Marburg, Marburg, Germany
| | - Bernd Schmeck
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, German Center for Lung Research (DZL), Marburg, Germany
- SYNMIKRO Center of Synthetic Microbiology, Marburg, Germany
- Core Facility Flow Cytometry - Bacterial Vesicles, Philipps-University Marburg, Marburg, Germany
- Department of Medicine, Pulmonary and Critical Care Medicine, University Medical Center Marburg, Philipps-University Marburg, Marburg, Germany
- Institute for Lung Health (ILH), Giessen, Germany
- Member of the German Center for Infectious Disease Research (DZIF), Marburg, Germany
| | - Gerhard Hummer
- Department of Theoretical Biophysics, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
- Institute for Biophysics, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Olalla Vázquez
- Department of Chemistry, Philipps-University Marburg, Marburg, Germany
- SYNMIKRO Center of Synthetic Microbiology, Marburg, Germany
| | - Tobias J Erb
- Department of Biochemistry and Synthetic Metabolism, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany.
- SYNMIKRO Center of Synthetic Microbiology, Marburg, Germany.
| |
Collapse
|
37
|
Guo L, Tang M, Luo S, Zhou X. Screening and Functional Analyses of Novel Cecropins from Insect Transcriptome. INSECTS 2023; 14:794. [PMID: 37887806 PMCID: PMC10607850 DOI: 10.3390/insects14100794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/27/2023] [Accepted: 09/27/2023] [Indexed: 10/28/2023]
Abstract
Antibiotic resistance is a significant and growing threat to global public health. However, antimicrobial peptides (AMPs) have shown promise as they exhibit a broad spectrum of antibacterial activities with low potential for resistance development. Insects, which inhabit a wide range of environments and are incredibly diverse, remain largely unexplored as a source of novel AMPs. To address this, we conducted a screening of the representative transcriptomes from the 1000 Insect Transcriptome Evolution (1KITE) dataset, focusing on the homologous reference genes of Cecropins, the first identified AMPs in insects known for its high efficiency. Our analysis identified 108 Cecropin genes from 105 insect transcriptomes, covering all major hexapod lineages. We validated the gene sequences and synthesized mature peptides for three identified Cecropin genes. Through minimal inhibition concentration and agar diffusion assays, we confirmed that these peptides exhibited antimicrobial activities against Gram-negative bacteria. Similar to the known Cecropin, the three Cecropins adopted an alpha-helical conformation in membrane-like environments, efficiently disrupting bacterial membranes through permeabilization. Importantly, none of the three Cecropins demonstrated cytotoxicity in erythrocyte hemolysis tests, suggesting their safety in real-world applications. Overall, this newly developed methodology provides a high-throughput bioinformatic pipeline for the discovery of AMP, taking advantage of the expanding genomic resources available for diverse organisms.
Collapse
Affiliation(s)
- Lizhen Guo
- Department of Entomology, College of Plant Protection, China Agricultural University, Beijing 100193, China; (L.G.); (M.T.)
- Sanya Institute of China Agricultural University, Sanya 572000, China
| | - Min Tang
- Department of Entomology, College of Plant Protection, China Agricultural University, Beijing 100193, China; (L.G.); (M.T.)
- Department of Biological Sciences, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China
| | - Shiqi Luo
- Department of Entomology, College of Plant Protection, China Agricultural University, Beijing 100193, China; (L.G.); (M.T.)
| | - Xin Zhou
- Department of Entomology, College of Plant Protection, China Agricultural University, Beijing 100193, China; (L.G.); (M.T.)
- Sanya Institute of China Agricultural University, Sanya 572000, China
| |
Collapse
|
38
|
Szot-Karpińska K, Kudła P, Orzeł U, Narajczyk M, Jönsson-Niedziółka M, Pałys B, Filipek S, Ebner A, Niedziółka-Jönsson J. Investigation of Peptides for Molecular Recognition of C-Reactive Protein-Theoretical and Experimental Studies. Anal Chem 2023; 95:14475-14483. [PMID: 37695838 PMCID: PMC10535004 DOI: 10.1021/acs.analchem.3c03127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 08/29/2023] [Indexed: 09/13/2023]
Abstract
We investigate the interactions between C-reactive protein (CRP) and new CRP-binding peptide materials using experimental (biological and physicochemical) methods with the support of theoretical simulations (computational modeling analysis). Three specific CRP-binding peptides (P2, P3, and P9) derived from an M13 bacteriophage have been identified using phage-display technology. The binding efficiency of the peptides exposed on phages toward the CRP protein was demonstrated via biological methods. Fibers of the selected phages/peptides interact differently due to different compositions of amino acid sequences on the exposed peptides, which was confirmed by transmission electron microscopy. Numerical and experimental studies consistently showed that the P3 peptide is the best CRP binder. A combination of theoretical and experimental methods demonstrates that identifying the best binder can be performed simply, cheaply, and fast. Such an approach has not been reported previously for peptide screening and demonstrates a new trend in science where calculations can replace or support laborious experimental techniques. Finally, the best CRP binder─the P3 peptide─was used for CRP recognition on silicate-modified indium tin oxide-coated glass electrodes. The obtained electrodes exhibit a wide range of operation (1.0-100 μg mL-1) with a detection limit (LOD = 3σ/S) of 0.34 μg mL-1. Moreover, the dissociation constant Kd of 4.2 ± 0.144 μg mL-1 (35 ± 1.2 nM) was evaluated from the change in the current. The selectivity of the obtained electrode was demonstrated in the presence of three interfering proteins. These results prove that the presented P3 peptide is a potential candidate as a receptor for CRP, which can replace specific antibodies.
Collapse
Affiliation(s)
- Katarzyna Szot-Karpińska
- Institute
of Physical Chemistry, Polish Academy of
Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland
| | - Patryk Kudła
- Institute
of Physical Chemistry, Polish Academy of
Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland
| | - Urszula Orzeł
- Biological
and Chemical Research Centre, University
of Warsaw, Zwirki i Wigury 101, 02-089 Warsaw, Poland
- Faculty
of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland
| | - Magdalena Narajczyk
- Department
of Electron Microscopy, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| | | | - Barbara Pałys
- Faculty
of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland
| | - Sławomir Filipek
- Biological
and Chemical Research Centre, University
of Warsaw, Zwirki i Wigury 101, 02-089 Warsaw, Poland
- Faculty
of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland
| | - Andreas Ebner
- Institute
of Biophysics, Johannes Kepler University, Gruberstrasse 40, 4020 Linz, Austria
| | | |
Collapse
|
39
|
Cesaro A, Bagheri M, Torres MDT, Wan F, de la Fuente-Nunez C. Deep learning tools to accelerate antibiotic discovery. Expert Opin Drug Discov 2023; 18:1245-1257. [PMID: 37794737 PMCID: PMC10790350 DOI: 10.1080/17460441.2023.2250721] [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: 05/19/2023] [Accepted: 08/18/2023] [Indexed: 10/06/2023]
Abstract
INTRODUCTION As machine learning (ML) and artificial intelligence (AI) expand to many segments of our society, they are increasingly being used for drug discovery. Recent deep learning models offer an efficient way to explore high-dimensional data and design compounds with desired properties, including those with antibacterial activity. AREAS COVERED This review covers key frameworks in antibiotic discovery, highlighting physicochemical features and addressing dataset limitations. The deep learning approaches here described include discriminative models such as convolutional neural networks, recurrent neural networks, graph neural networks, and generative models like neural language models, variational autoencoders, generative adversarial networks, normalizing flow, and diffusion models. As the integration of these approaches in drug discovery continues to evolve, this review aims to provide insights into promising prospects and challenges that lie ahead in harnessing such technologies for the development of antibiotics. EXPERT OPINION Accurate antimicrobial prediction using deep learning faces challenges such as imbalanced data, limited datasets, experimental validation, target strains, and structure. The integration of deep generative models with bioinformatics, molecular dynamics, and data augmentation holds the potential to overcome these challenges, enhance model performance, and utlimately accelerate antimicrobial discovery.
Collapse
Affiliation(s)
- Angela Cesaro
- 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
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Mojtaba Bagheri
- 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
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - 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
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - 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
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - 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
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|