1
|
Kim J, Liao X, Zhang S, Ding T, Ahn J. Application of phage-derived enzymes for enhancing food safety. Food Res Int 2025; 209:116318. [PMID: 40253159 DOI: 10.1016/j.foodres.2025.116318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 03/13/2025] [Accepted: 03/17/2025] [Indexed: 04/21/2025]
Abstract
Foodborne pathogens such as Salmonella, Escherichia coli, Listeria monocytogenes, and Staphylococcus aureus present significant public health threats, causing widespread illness and economic loss. Contaminated food is responsible for an estimated 600 million illnesses and 420,000 deaths annually, with low- and middle-income countries facing losses of approximately $110 billion each year. Traditional methods to ensure food safety, including antimicrobials and preservatives, can contribute to the development of antimicrobial-resistant bacteria, highlighting the need for alternative strategies. Bacteriophages are gaining renewed attention as promising alternatives to conventional antibiotics due to their specifically target bacteria and their lower potential for causing adverse effects. However, their practical application is limited by challenges such as narrow host ranges, the emergence of phage-resistant bacteria, and stability issues. Recent research has shifted focus towards phage-derived enzymes, including endolysins, depolymerases, holins, and spanins, which are involved in the phage lytic cycle. These enzymes, as potential approaches to food safety, have demonstrated significant efficacy in targeting and lysing bacterial pathogens, making them suitable for controlling foodborne pathogens and preventing foodborne illnesses. Phage-derived enzymes also show promise in controlling biofilms and enhancing antimicrobial activity when combined with other antimicrobials. Therefore, this review emphasizes recent advancements in the use of the phage-derived enzymes for food safety, addresses their limitations, and suggests strategies to enhance their effectiveness in food processing and storage environments.
Collapse
Affiliation(s)
- Junhwan Kim
- Department of Biomedical Science, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Xinyu Liao
- Future Food Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, Zhejiang 314100, China
| | - Song Zhang
- Department of Biomedical Science, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Tian Ding
- Future Food Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, Zhejiang 314100, China; College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| | - Juhee Ahn
- Department of Biomedical Science, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea; Future Food Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, Zhejiang 314100, China.
| |
Collapse
|
2
|
Abbas S, Kanwar R, Ullah K, Kanwal R, Tajamal M, Aslam MA, Ahmad A, Qadeer A, Huang HY, Chen CC. Bacteriophage therapy: a possible alternative therapy against antibiotic-resistant strains of Klebsiella pneumoniae. Front Microbiol 2025; 16:1443430. [PMID: 40231234 PMCID: PMC11994585 DOI: 10.3389/fmicb.2025.1443430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 03/07/2025] [Indexed: 04/16/2025] Open
Abstract
Klebsiella pneumoniae is a notorious, Gram-negative pathogen and is a leading cause of healthcare settings and community-acquired infections. This is the commensal of human microbiota and can invade and cause infections in different body parts. The global emergence of antibiotic resistance in K. pneumoniae has become a major challenge in the whole medical community. Alternative paths to treat the infections caused by these MDR pathogens are needed as these bacteria become resistant to last-resort antibiotics like colistin. The lytic bacteriophages (phages) are the bacteria's natural predators and can rapidly eliminate the bacterial cells. Phages are abundant in nature and have recently been found to be effective tools in modern biotechnology. They can be used to control the bacterial infectious diseases. They can be manipulated easily and potentially used in therapeutics, biotechnology, and research. Several studies, both in vitro and in vivo, have demonstrated the possible applications of the lytic phages in treating K. pneumoniae superbug strains. Phage endolysins have drawn the scientific world's attention because of their involvement in phage adsorption and bacterial capsules digestion. These phage-encoded enzymes digest the polysaccharide components of bacterial cell walls by recognizing and binding them. Phage lysins, being strong biological agents, are capable of effectively and swiftly eliminating bacteria. This review summarizes the information on phages of K. pneumoniae and phage-based therapies to target their bacterial hosts.
Collapse
Affiliation(s)
- Sadia Abbas
- Institute of Microbiology, University of Agriculture Faisalabad, Faisalabad, Pakistan
| | - Rabia Kanwar
- Institute of Microbiology, University of Agriculture Faisalabad, Faisalabad, Pakistan
| | - Kaleem Ullah
- Directorate General (Research) Livestock & Dairy Development Department Peshawar, Peshawar, Khyber Pakhtunkhwa, Pakistan
| | - Rimsha Kanwal
- Institute of Microbiology, University of Agriculture Faisalabad, Faisalabad, Pakistan
| | - Mamoon Tajamal
- Institute of Microbiology, University of Agriculture Faisalabad, Faisalabad, Pakistan
| | - Muhammad Aamir Aslam
- Institute of Microbiology, University of Agriculture Faisalabad, Faisalabad, Pakistan
| | - Abid Ahmad
- Department of Animal Nutrition, The University of Agriculture Peshawar, Peshawar, Khyber Pakhtunkhwa, Pakistan
| | - Abdul Qadeer
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Hsun-Yu Huang
- Division of Endodontics, Department of Stomatology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, Taiwan
| | - Chien-Chin Chen
- Department of Pathology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, Taiwan
- Department of Cosmetic Science, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
- Doctoral Program in Translational Medicine, Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
- Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
3
|
Choi D, Ryu S, Kong M. Phage-derived proteins: Advancing food safety through biocontrol and detection of foodborne pathogens. Compr Rev Food Sci Food Saf 2025; 24:e70124. [PMID: 39898971 DOI: 10.1111/1541-4337.70124] [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: 10/03/2024] [Revised: 01/01/2025] [Accepted: 01/10/2025] [Indexed: 02/04/2025]
Abstract
The emergence of antimicrobial-resistant foodborne pathogens poses a continuous health risk and economic burden as they can easily spread through contaminated food. Therefore, the demand for new antimicrobial agents to address this problem is steadily increasing. Similarly, the development of rapid, sensitive, and accurate pathogen detection tools is a prerequisite for ensuring food safety. Phage-derived proteins have become innovative tools for combating these pathogens because of their potent antimicrobial activity and host specificity. Phage proteins are relatively free from regulation compared to phages per se, and there are no concerns about the transduction of harmful genes. With recent progress in next-generation sequencing technology, the analysis of phage genomes has become more accessible, and numerous phage proteins with potential for biocontrol and detection have been identified. This review provides a comprehensive overview of phage protein research on food safety from 2006 to the present, a pivotal period marked by the certification of phages as Generally Recognized As Safe (GRAS). Emphasizing recent advancements, we investigated the diverse applications of various phage proteins for biocontrol and detection purposes. While highlighting the successful implementation of these proteins, we also address the current bottlenecks and propose strategies to overcome these challenges. By summarizing the current state of research on phage-derived proteins, this review contributes to a deeper understanding of their potential as effective antimicrobial agents and tools for detecting foodborne pathogens.
Collapse
Affiliation(s)
- Dahee Choi
- Department of Food Science and Biotechnology, Institute of Food and Biotechnology, Seoul National University of Science and Technology, Seoul, South Korea
| | - Sangryeol Ryu
- Department of Food and Animal Biotechnology, Department of Agricultural Biotechnology, Seoul National University, Seoul, South Korea
| | - Minsuk Kong
- Department of Food Science and Biotechnology, Institute of Food and Biotechnology, Seoul National University of Science and Technology, Seoul, South Korea
| |
Collapse
|
4
|
Rajab AAH, Khafagy ES, Lila ASA, Yousef N, Askoura M. Combating enteropathogenic and multidrug resistant Escherichia coli using the lytic bacteriophage vB_EcoM_ECO78, which disrupts bacterial biofilm formation and exhibits a remarkable environmental stability. J Appl Microbiol 2025; 136:lxaf028. [PMID: 39919762 DOI: 10.1093/jambio/lxaf028] [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: 11/05/2024] [Revised: 01/31/2025] [Accepted: 02/06/2025] [Indexed: 02/09/2025]
Abstract
AIM The current study aimed to establish a phenotypic and genotypic characterization record of a novel lytic bacteriophage (phage) against multidrug-resistant (MDR) Escherichia coli (E. coli) infections. METHODS AND RESULTS Phenotypic characterization of the isolated phage included the assessment of phage morphology, host range, stability, and antibiofilm activity. The isolated phage vB_EcoM_ECO78 demonstrated a high lytic activity against MDR E. coli and E. coli serotypes O78: K80: H12 and O26: H11. Additionally, it showed a marked antibiofilm activity and high physical stability at a wide range of temperatures and pH. Genotypic investigations identified a double-stranded DNA genome of 165 912 base pairs (bp) spanning 258 open reading frames (ORFs), out of which 149 ORFs were identified and annotated. In vivo analysis further confirmed the therapeutic potential of vB_EcoM_ECO78 which effectively increased the survival of mice infected with MDR E. coli. CONCLUSION The isolated phage vB_EcoM_ECO78 exhibits considerable stability and antibiofilm activity against MDR E. coli isolates, supported by notable environmental fitness and in vivo antibacterial capability.
Collapse
Affiliation(s)
- Azza A H Rajab
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - El-Sayed Khafagy
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Amr S Abu Lila
- Department of Pharmaceutics, College of Pharmacy, University of Hail, Hail 81442, Saudi Arabia
| | - Nehal Yousef
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Momen Askoura
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
5
|
Kogawa M, Yoda T, Matsuhashi A, Matsushita A, Otsuka Y, Shibagaki S, Hosokawa M, Tsuda S. Development of Chimera AMP-Endolysin with Wider Spectra Against Gram-Negative Bacteria Using High-Throughput Assay. Viruses 2025; 17:200. [PMID: 40006955 PMCID: PMC11860666 DOI: 10.3390/v17020200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/25/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
Bacteriophage-derived endolysins are being developed as an alternative to antimicrobials. The development of endolysins against Gram-negative bacteria requires the discovery of effective endolysins against the target species and the capability to penetrate the outer membrane of bacteria by endolysin. Here, we propose an efficient endolysin development approach that combines a data-driven endolysin search utilizing bacterial genomes with high-throughput laboratory assays. As a proof of concept, we analyzed endolysin genes detected in 273 bacterial genomes of Acinetobacter, Pseudomonas, and Escherichia. Firstly, we conducted assays of 192 recombinants of endolysin genes obtained through in silico search from bacterial genomes and identified natural endolysins degrading peptidoglycan of Acinetobacter baumannii. Then, we performed high-throughput screening against Gram-negative bacteria for hundreds of chimera AMP-endolysins, natural endolysin conjugated with antimicrobial peptide. As a result, we obtained four chimera AMP-endolysins against A. baumannii, which demonstrated the minimum inhibitory concentration ranging from 4 to 8 μg/mL. Moreover, we assessed the antimicrobial spectra of these chimera AMP-endolysins, validating that two endolysins exhibited antimicrobial efficacy against Pseudomonas aeruginosa and Escherichia coli with <32 μg/mL of concentration. This endolysin development approach can be applied to other Gram-negative bacterial targets and is expected to facilitate the acquisition of effective novel endolysins.
Collapse
Affiliation(s)
- Masato Kogawa
- bitBiome, Inc., 513 Wasedatsurumaki-Cho, Shinjuku-Ku 162-0041, Tokyo, Japan
| | - Takuya Yoda
- bitBiome, Inc., 513 Wasedatsurumaki-Cho, Shinjuku-Ku 162-0041, Tokyo, Japan
| | - Ayumi Matsuhashi
- bitBiome, Inc., 513 Wasedatsurumaki-Cho, Shinjuku-Ku 162-0041, Tokyo, Japan
| | - Ai Matsushita
- bitBiome, Inc., 513 Wasedatsurumaki-Cho, Shinjuku-Ku 162-0041, Tokyo, Japan
| | - Yoshiki Otsuka
- bitBiome, Inc., 513 Wasedatsurumaki-Cho, Shinjuku-Ku 162-0041, Tokyo, Japan
| | - Shohei Shibagaki
- bitBiome, Inc., 513 Wasedatsurumaki-Cho, Shinjuku-Ku 162-0041, Tokyo, Japan
| | - Masahito Hosokawa
- bitBiome, Inc., 513 Wasedatsurumaki-Cho, Shinjuku-Ku 162-0041, Tokyo, Japan
- Graduate School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsu-Cho, Shinjuku-Ku 162-8480, Tokyo, Japan
- Computational Bio Big-Data Open Innovation Laboratory, National Institute of Advanced Industrial Science and Technology, 3-4-1 Okubo, Shinjuku-Ku 169-8555, Tokyo, Japan
- Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, 3-4-1 Okubo, Shinjuku-Ku 169-8555, Tokyo, Japan
- Research Organization for Nano and Life Innovation, Waseda University, 513 Wasedatsurumaki-Cho, Shinjuku-Ku 162-0041, Tokyo, Japan
| | - Soichiro Tsuda
- bitBiome, Inc., 513 Wasedatsurumaki-Cho, Shinjuku-Ku 162-0041, Tokyo, Japan
| |
Collapse
|
6
|
Sabur A, Khan A, Borphukan B, Razzak A, Salimullah M, Khatun M. The Unique Capability of Endolysin to Tackle Antibiotic Resistance: Cracking the Barrier. J Xenobiot 2025; 15:19. [PMID: 39997362 PMCID: PMC11856723 DOI: 10.3390/jox15010019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 01/17/2025] [Accepted: 01/18/2025] [Indexed: 02/26/2025] Open
Abstract
The lack of new antibacterial medicines and the rapid rise in bacterial resistance to antibiotics pose a major threat to individuals and healthcare systems. Despite the availability of various antibiotics, bacterial resistance has emerged for almost every antibiotic discovered to date. The increasing prevalence of multidrug-resistant bacterial strains has rendered some infections nearly untreatable, posing severe challenges to health care. Thus, the development of alternatives to conventional antibiotics is critical for the treatment of both humans and food-producing animals. Endolysins, which are peptidoglycan hydrolases encoded by bacteriophages, represent a promising new class of antimicrobials. Preliminary research suggests that endolysins are more effective against Gram-positive bacteria than Gram-negative bacteria when administered exogenously, although they can still damage the cell wall of Gram-negative bacteria. Numerous endolysins have a modular domain structure that divides their binding and catalytic activity into distinct subunits, which helps maximize their bioengineering and potential drug development. Endolysins and endolysin-derived antimicrobials offer several advantages as antibiotic substitutes. They have a unique mechanism of action and efficacy against bacterial persisters (without requiring an active host metabolism); subsequently, they target both Gram-positive and Gram-negative bacteria (including antibiotic-resistant strains), and mycobacteria. Furthermore, there has been limited evidence of endolysin being resistant. Because these enzymes target highly conserved links, resistance may develop more slowly compared to traditional antibiotics. This review provides an overview and insight of the potential applications of endolysins as novel antimicrobials.
Collapse
Affiliation(s)
- Abdus Sabur
- Animal Biotechnology Division, National Institute of Biotechnology, Savar, Dhaka 1349, Bangladesh;
| | - Angkan Khan
- Infectious Diseases Division, International Centre for Diarrheal Disease Research, Bangladesh, Mohakhali, Dhaka 1212, Bangladesh;
| | - B. Borphukan
- Department of Crop and Soil Sciences, Washington State University, Pullman, WA 99163, USA;
| | - Abdur Razzak
- Bioassay Department, Eurofins Biopharma, Columbia, MO 65201, USA;
| | - M. Salimullah
- Molecular Biotechnology Division, National Institute of Biotechnology, Savar, Dhaka 1349, Bangladesh;
| | - Muslima Khatun
- Molecular Biotechnology Division, National Institute of Biotechnology, Savar, Dhaka 1349, Bangladesh;
| |
Collapse
|
7
|
Seo S, Son B, Kong M. Characterization of Clostridium perfringens Phage Endolysin PlyDolk21. Antibiotics (Basel) 2025; 14:81. [PMID: 39858367 PMCID: PMC11762992 DOI: 10.3390/antibiotics14010081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/31/2024] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Background:Clostridium perfringens is a significant cause of food poisoning. Broad-spectrum antibiotics, commonly used to control C. perfringens, are becoming less effective due to the rise of antibiotic-resistant strains, necessitating alternative control strategies. Methods: A C. perfringens-infecting bacteriophage, Dolk21, and its endolysin, PlyDolk21, were isolated and characterized. The lytic activity of PlyDolk21 was assessed in comparison to its catalytic domain alone. Both PlyDolk21 and its cell wall binding domain (CBD) were evaluated in beef and milk for their antimicrobial activity and cell wall binding activity, respectively. Results: While phage Dolk21 was specific to certain C. perfringens strains, PlyDolk21 exhibited lytic activity against all C. perfringens strains tested. The full-length PlyDolk21 showed stronger lytic activity compared to its catalytic domain alone. PlyDolk21_CBD successfully bound to C. perfringens in vitro and in foods. Additionally, PlyDolk21 effectively reduced the viable cell counts of C. perfringens by 3-log in beef soup and milk samples. Conclusions: This study demonstrates that PlyDolk21 and its CBD hold potential as a biocontrol and detection agent targeting C. perfringens in various food matrices.
Collapse
Affiliation(s)
- Suyoung Seo
- Department of Food Science and Biotechnology, Research Institute of Food and Biotechnology, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea;
| | - Bokyung Son
- Department of Food Biotechnology, Dong-A University, Busan 49315, Republic of Korea;
| | - Minsuk Kong
- Department of Food Science and Biotechnology, Research Institute of Food and Biotechnology, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea;
| |
Collapse
|
8
|
Golban M, Charostad J, Kazemian H, Heidari H. Phage-Derived Endolysins Against Resistant Staphylococcus spp.: A Review of Features, Antibacterial Activities, and Recent Applications. Infect Dis Ther 2025; 14:13-57. [PMID: 39549153 PMCID: PMC11782739 DOI: 10.1007/s40121-024-01069-z] [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: 08/19/2024] [Accepted: 10/22/2024] [Indexed: 11/18/2024] Open
Abstract
Antimicrobial resistance is a significant global public health issue, and the dissemination of antibiotic resistance in Gram-positive bacterial pathogens has significantly increased morbidity, mortality rates, and healthcare costs. Among them, Staphylococcus, especially methicillin-resistant Staphylococcus aureus (MRSA), causes a wide range of diseases due to its diverse pathogenic factors and infection strategies. These bacteria also present significant issues in veterinary medicine and food safety. Effectively managing staphylococci-related problems necessitates a concerted effort to implement preventive measures, rapidly detect the pathogen, and develop new and safe antimicrobial therapies. In recent years, there has been growing interest in using endolysins to combat bacterial infections. These enzymes, which are also referred to as lysins, are a unique class of hydrolytic enzymes synthesized by double-stranded DNA bacteriophages. They possess glycosidase, lytic transglycosylase, amidase, and endopeptidase activities, effectively destroying the peptidoglycan layer and resulting in bacterial lysis. This unique property makes endolysins powerful antimicrobial agents, particularly against Gram-positive organisms with more accessible peptidoglycan layers. Therefore, considering the potential benefits of endolysins compared to conventional antibiotics, we have endeavored to gather and review the characteristics and uses of endolysins derived from staphylococcal bacteriophages, as well as their antibacterial effectiveness against Staphylococcus spp. based on conducted experiments and trials.
Collapse
Affiliation(s)
- Mina Golban
- Department of Microbiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Javad Charostad
- Department of Microbiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hossein Kazemian
- Clinical Microbiology Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Hamid Heidari
- Department of Microbiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
9
|
Wang Q, Chen R, Liu H, Liu Y, Li J, Wang Y, Jin Y, Bai Y, Song Z, Lu X, Wang C, Hao Y. Isolation and characterization of lytic bacteriophage vB_KpnP_23: A promising antimicrobial candidate against carbapenem-resistant Klebsiella pneumoniae. Virus Res 2024; 350:199473. [PMID: 39332682 PMCID: PMC11474366 DOI: 10.1016/j.virusres.2024.199473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/08/2024] [Accepted: 09/21/2024] [Indexed: 09/29/2024]
Abstract
The global health threat posed by carbapenem-resistant Klebsiella pneumoniae (CRKP) is exacerbated by the limited availability of effective treatments. Bacteriophages are promising alternatives to conventional antimicrobial agents. However, current phage databases are limited. Thus, identifying and characterizing new phages could provide biological options for the treatment of multi-drug resistant bacterial infections. Here, we report the characterization of a novel lytic phage, vB_KpnP_23, isolated from hospital sewage. This phage exhibited potent activity against carbapenemase-producing CRKP strains and was characterised by an icosahedral head, a retractable tail, and a genome comprising 40,987 base pairs, with a G + C content of 51 %. Capable of targeting and lysing nine different capsule types (K-types) of CRKP, including the clinically relevant ST11-K64, it demonstrated both high bacteriolytic efficiency and stability in various environmental contexts. Crucially, vB_KpnP_23 lacks virulence factors, antimicrobial resistance genes, or tRNA, aligning with the key criteria for therapeutic application. In vitro evaluation of phage-antibiotic combinations revealed a significant synergistic effect between vB_KpnP_23 and meropenem, levofloxacin, or amikacin. This synergy could lead to an 8-fold reduction in the minimum inhibitory concentration (MIC), suggesting that integrated treatments combining this phage with the aforementioned antibiotics may substantially enhance drug effectiveness. This approach not only extends the clinical utility of these antibiotics but also presents a strategic advance in combating antibiotic resistance. Specifically, it underscores the potential of phage-antibiotic combinations as a powerful tool in the treatment of infections caused by CRKP, offering a promising avenue to mitigate the public health challenges of antibiotic-resistant pathogens.
Collapse
Affiliation(s)
- Qian Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ran Chen
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Hui Liu
- Department of Clinical Laboratory, Maternal and Child Health Care Hospital of Zaozhuang, Zaozhuang, Shandong, 277100, China
| | - Yue Liu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jinmei Li
- Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo college of Medicine, Shandong University, Jinan, Shandong, China; Department of Clinical Laboratory, Jinan Seventh People's Hospital, Jinan, Shandong, 250021, China
| | - Yueling Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yan Jin
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yuanyuan Bai
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhen Song
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xinglun Lu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Changyin Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
| | - Yingying Hao
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China; Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo college of Medicine, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
10
|
Wójcicki M, Shymialevich D, Średnicka P, Emanowicz P, Ostrowska A, Cieślak H, Sokołowska B. Phenotypic Characterization and Genome Analysis of New Broad-Spectrum Virulent Salmophage, Salmonella Phage KKP_3822, for Biocontrol of Multidrug-Resistant Salmonella enterica Strains. Int J Mol Sci 2024; 25:12930. [PMID: 39684641 DOI: 10.3390/ijms252312930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/30/2024] [Accepted: 11/30/2024] [Indexed: 12/18/2024] Open
Abstract
Salmonella is one of the main foodborne pathogens. Irrational antibiotic management has led to an increase in the incidence of multidrug-resistant strains. Bacteriophages may be an alternative method of food biopreservation and contribute to reducing the number of food poisonings requiring pharmacotherapy. This study aimed to isolate a bacteriophage (phage) targeting indigenous multidrug-resistant (MDR) Salmonella strains, followed by their biological, morphological, and genomic characterization. In this study we isolated Salmonella phage KKP_3822, targeting MDR Salmonella Manchester strain KKP 1213. Salmonella phage KKP_3822 retained high activity in the temperature range from -20 °C to 40 °C and active acidity from pH 3 to 11. Temperatures of 70 °C and 80 °C and extreme pH values (2 and 12) significantly reduced the phage titer. Its activity decreased proportionally to the time of UV exposure. Genome analysis (linear dsDNA with a length of 114,843 bp) revealed the presence of 27 tRNA genes. Proteins encoded by the vB_Sen-IAFB3822 phage were divided into functional modules related to (i) phage structure/assembly, (ii) DNA replication/modification/regulation, (iii) phage lysis, and (iv) DNA packaging into the capsid. No genes associated with antibiotic resistance or integration into the host genome, markers of temperate bacteriophages, were annotated in the Salmonella phage KKP_3822 genome. Based on morphological features and whole-genome sequence analysis, the newly isolated Salmonella phage KKP_3822 shows the greatest similarity to representatives of tailed phages from the Caudoviricetes class, Demerecviridae family, and Epseptimavirus genus. Genome analysis confirmed the virulent nature of the Salmonella phage KKP_3822, making it a potential candidate for food biocontrol.
Collapse
Affiliation(s)
- Michał Wójcicki
- Department of Microbiology, Prof. Wacław Dąbrowski Institute of Agricultural and Food Biotechnology-State Research Institute, Rakowiecka 36 Str., 02-532 Warsaw, Poland
| | - Dziyana Shymialevich
- Department of Microbiology, Prof. Wacław Dąbrowski Institute of Agricultural and Food Biotechnology-State Research Institute, Rakowiecka 36 Str., 02-532 Warsaw, Poland
| | - Paulina Średnicka
- Department of Microbiology, Prof. Wacław Dąbrowski Institute of Agricultural and Food Biotechnology-State Research Institute, Rakowiecka 36 Str., 02-532 Warsaw, Poland
| | - Paulina Emanowicz
- Department of Microbiology, Prof. Wacław Dąbrowski Institute of Agricultural and Food Biotechnology-State Research Institute, Rakowiecka 36 Str., 02-532 Warsaw, Poland
| | - Agnieszka Ostrowska
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences (WULS-SGGW), Ciszewskiego 8 Str., 02-786 Warsaw, Poland
| | - Hanna Cieślak
- Department of Microbiology, Prof. Wacław Dąbrowski Institute of Agricultural and Food Biotechnology-State Research Institute, Rakowiecka 36 Str., 02-532 Warsaw, Poland
| | - Barbara Sokołowska
- Department of Microbiology, Prof. Wacław Dąbrowski Institute of Agricultural and Food Biotechnology-State Research Institute, Rakowiecka 36 Str., 02-532 Warsaw, Poland
| |
Collapse
|
11
|
Nakonieczna A, Topolska-Woś A, Łobocka M. New bacteriophage-derived lysins, LysJ and LysF, with the potential to control Bacillus anthracis. Appl Microbiol Biotechnol 2024; 108:76. [PMID: 38194144 PMCID: PMC10776502 DOI: 10.1007/s00253-023-12839-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 11/13/2023] [Accepted: 11/21/2023] [Indexed: 01/10/2024]
Abstract
Bacillus anthracis is an etiological agent of anthrax, a severe zoonotic disease that can be transmitted to people and cause high mortalities. Bacteriophages and their lytic enzymes, endolysins, have potential therapeutic value in treating infections caused by this bacterium as alternatives or complements to antibiotic therapy. They can also be used to identify and detect B. anthracis. Endolysins of two B. anthracis Wbetavirus phages, J5a and F16Ba which were described by us recently, differ significantly from the best-known B. anthracis phage endolysin PlyG from Wbetavirus genus bacteriophage Gamma and a few other Wbetavirus genus phages. They are larger than PlyG (351 vs. 233 amino acid residues), contain a signal peptide at their N-termini, and, by prediction, have a different fold of cell binding domain suggesting different structural basis of cell epitope recognition. We purified in a soluble form the modified versions of these endolysins, designated by us LysJ and LysF, respectively, and depleted of signal peptides. Both modified endolysins could lyse the B. anthracis cell wall in zymogram assays. Their activity against the living cells of B. anthracis and other species of Bacillus genus was tested by spotting on the layers of bacteria in soft agar and by assessing the reduction of optical density of bacterial suspensions. Both methods proved the effectiveness of LysJ and LysF in killing the anthrax bacilli, although the results obtained by each method differed. Additionally, the lytic efficiency of both proteins was different, which apparently correlates with differences in their amino acid sequence. KEY POINTS: • LysJ and LysF are B. anthracis-targeting lysins differing from lysins studied so far • LysJ and LysF could be overproduced in E. coli in soluble and active forms • LysJ and LysF are active in killing cells of B. anthracis virulent strains.
Collapse
Affiliation(s)
- Aleksandra Nakonieczna
- Military Institute of Hygiene and Epidemiology, Biological Threats Identification and Countermeasure Center, 24-100, Puławy, Poland.
| | | | - Małgorzata Łobocka
- Institute of Biochemistry and Biophysics of the Polish Academy of Sciences, 02-106, Warsaw, Poland
| |
Collapse
|
12
|
Palma M, Qi B. Advancing Phage Therapy: A Comprehensive Review of the Safety, Efficacy, and Future Prospects for the Targeted Treatment of Bacterial Infections. Infect Dis Rep 2024; 16:1127-1181. [PMID: 39728014 DOI: 10.3390/idr16060092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/13/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Phage therapy, a treatment utilizing bacteriophages to combat bacterial infections, is gaining attention as a promising alternative to antibiotics, particularly for managing antibiotic-resistant bacteria. This study aims to provide a comprehensive review of phage therapy by examining its safety, efficacy, influencing factors, future prospects, and regulatory considerations. The study also seeks to identify strategies for optimizing its application and to propose a systematic framework for its clinical implementation. METHODS A comprehensive analysis of preclinical studies, clinical trials, and regulatory frameworks was undertaken to evaluate the therapeutic potential of phage therapy. This included an in-depth assessment of key factors influencing clinical outcomes, such as infection site, phage-host specificity, bacterial burden, and immune response. Additionally, innovative strategies-such as combination therapies, bioengineered phages, and phage cocktails-were explored to enhance efficacy. Critical considerations related to dosing, including inoculum size, multiplicity of infection, therapeutic windows, and personalized medicine approaches, were also examined to optimize treatment outcomes. RESULTS Phage therapy has demonstrated a favorable safety profile in both preclinical and clinical settings, with minimal adverse effects. Its ability to specifically target harmful bacteria while preserving beneficial microbiota underpins its efficacy in treating a range of infections. However, variable outcomes in some studies highlight the importance of addressing critical factors that influence therapeutic success. Innovative approaches, including combination therapies, bioengineered phages, expanded access to diverse phage banks, phage cocktails, and personalized medicine, hold significant promise for improving efficacy. Optimizing dosing strategies remains a key area for enhancement, with critical considerations including inoculum size, multiplicity of infection, phage kinetics, resistance potential, therapeutic windows, dosing frequency, and patient-specific factors. To support the clinical application of phage therapy, a streamlined four-step guideline has been developed, providing a systematic framework for effective treatment planning and implementation. CONCLUSION Phage therapy offers a highly adaptable, targeted, and cost-effective approach to addressing antibiotic-resistant infections. While several critical factors must be thoroughly evaluated to optimize treatment efficacy, there remains significant potential for improvement through innovative strategies and refined methodologies. Although phage therapy has yet to achieve widespread approval in the U.S. and Europe, its accessibility through Expanded Access programs and FDA authorizations for food pathogen control underscores its promise. Established practices in countries such as Poland and Georgia further demonstrate its clinical feasibility. To enable broader adoption, regulatory harmonization and advancements in production, delivery, and quality control will be essential. Notably, the affordability and scalability of phage therapy position it as an especially valuable solution for developing regions grappling with escalating rates of antibiotic resistance.
Collapse
Affiliation(s)
- Marco Palma
- Institute for Globally Distributed Open Research and Education (IGDORE), 03181 Torrevieja, Spain
- R&D Drug Discovery, Protheragen Inc., Holbrook, NY 11741, USA
| | - Bowen Qi
- Drug Discovery and Development, Creative Biolabs Inc., Shirley, NY 11967, USA
| |
Collapse
|
13
|
Arakelian AG, Chuev GN, Mamedov TV. Molecular Docking of Endolysins for Studying Peptidoglycan Binding Mechanism. Molecules 2024; 29:5386. [PMID: 39598776 PMCID: PMC11597070 DOI: 10.3390/molecules29225386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 11/09/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024] Open
Abstract
Endolysins of bacteriophages, which degrade the bacterial cell wall peptidoglycan, are applicable in many industries to deal with biofilms and bacterial infections. While multi-domain endolysins have both enzymatically active and cell wall-binding domains, single-domain endolysins consist only of an enzymatically active domain, and their mechanism of peptidoglycan binding remains unexplored, for this is a challenging task experimentally. This research aimed to explore the binding mechanism of endolysins using computational approaches, namely molecular docking and bioinformatical tools, and analyze the performance of these approaches. The docking engine Autodock Vina 1.1.2 and the 3D-RISM module of AmberTools 24 were studied in the current work and used for receptor-ligand affinity and binding energy calculations, respectively. Two possible mechanisms of single-domain endolysin-ligand binding were predicted by Autodock Vina and verified by the 3D-RISM. As a result, the previously obtained experimental results on peptidoglycan binding of the isolated gamma phage endolysin PlyG enzymatically active domain were supported by molecular docking. Both methods predicted that single-domain endolysins are able to bind peptidoglycan, with Autodock Vina being able to give accurate numerical estimates of protein-ligand affinities and 3D-RISM providing comparative values.
Collapse
Affiliation(s)
- Arina G. Arakelian
- Institute of Theoretical and Experimental Biophysics, RAS, Institutskaya ul., 3, 142290 Pushchino, Moscow Oblast, Russia; (G.N.C.)
| | | | | |
Collapse
|
14
|
Ajose DJ, Adekanmbi AO, Kamaruzzaman NF, Ateba CN, Saeed SI. Combating antibiotic resistance in a one health context: a plethora of frontiers. ONE HEALTH OUTLOOK 2024; 6:19. [PMID: 39487542 PMCID: PMC11531134 DOI: 10.1186/s42522-024-00115-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 08/20/2024] [Indexed: 11/04/2024]
Abstract
One of the most significant medical advancements of the 20th century was the discovery of antibiotics, which continue to play a vital tool in the treatment and prevention of diseases in humans and animals. However, the imprudent use of antibiotics in all fields of One-Health and concerns about antibiotic resistance among bacterial pathogens have raised interest in antibiotic use restrictions on a global scale. Despite the failure of conventional antimicrobial agents, only about 15 new antibiotics have been introduced clinically since year 2000 to date. Moreover, there has been reports of resistance to some of these new antibiotics. This has necessitated a need to search for alternative strategies to combat antimicrobial resistant pathogens. Thus, this review compiles and evaluates the approaches-natural compounds, phage treatment, and nanomaterials-that are being used and/or suggested as the potential substitutes for conventional antibiotics.
Collapse
Affiliation(s)
- Daniel Jesuwenu Ajose
- Antimicrobial Resistance and Phage Biocontrol Research Group (AREPHABREG), Department of Microbiology, School of Biological Sciences, Faculty of Natural and Agricultural Sciences, North-West University, Private Mail Bag X2046, Mmabatho, 2735, South Africa.
- Food Security and Safety Focus Area, Faculty of Natural and Agricultural Sciences, North-West University, Private Bag X2046, Mmabatho, 2735, South Africa.
| | - Abimbola Olumide Adekanmbi
- Environmental Microbiology and Biotechnology Laboratory, Department of Microbiology, University of Ibadan, Ibadan, Nigeria
| | - Nor Fadhilah Kamaruzzaman
- Nanotechnology Research Group Faculty of Veterinary Medicine, Universiti Malaysia Kelantan, Kota Bharu, Kelantan, 16100, Malaysia
| | - Collins Njie Ateba
- Antimicrobial Resistance and Phage Biocontrol Research Group (AREPHABREG), Department of Microbiology, School of Biological Sciences, Faculty of Natural and Agricultural Sciences, North-West University, Private Mail Bag X2046, Mmabatho, 2735, South Africa
- Food Security and Safety Focus Area, Faculty of Natural and Agricultural Sciences, North-West University, Private Bag X2046, Mmabatho, 2735, South Africa
| | - Shamsaldeen Ibrahim Saeed
- College of Veterinary Medicine, University of Juba, P.O. Box 82, Juba, Central Equatoria, South Sudan.
- Department of microbiology, Faculty of Veterinary Science, University of Nyala, P.O. Box 155, Nyala, Sudan.
- Nanotechnology Research Group Faculty of Veterinary Medicine, Universiti Malaysia Kelantan, Kota Bharu, Kelantan, 16100, Malaysia.
| |
Collapse
|
15
|
Antonova NP, Vasina DV, Grigoriev IV, Laishevtsev AI, Kapustin AV, Savinov VA, Vorobev AM, Aleshkin AV, Zackharova AA, Remizov TA, Makarov VV, Yudin SM, Gushchin VA. Pharmacokinetic and preclinical safety studies of endolysin-based therapeutic for intravenous administration. Int J Antimicrob Agents 2024; 64:107328. [PMID: 39244166 DOI: 10.1016/j.ijantimicag.2024.107328] [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: 04/03/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/09/2024]
Abstract
Pharmacokinetics and safety studies of innovative drugs is an essential part of drug development process. Previously we have developed a novel drug for intravenous administration (lyophilizate) containing modified endolysin LysECD7-SMAP that showed notable antibacterial effect in different animal models of systemic infections. Here we present data on pharmacokinetics of endolysin in mice after single and multiple injections. Time-concentration curves were obtained, and pharmacokinetic parameters for preparation (C0, kel t1/2, AUC0-∞, MRT, ClT, Vss) were calculated. It was shown that although endolysin is rather short-lived in blood serum (t1/2 = 12.5 min), the therapeutic concentrations of LysECD7-SMAP (in degraded and non-degraded form) were detected for 60 minutes after injection that is sufficient for antibacterial effect. Based on the obtained data, it was proposed that endolysin distributes presumably in murine blood, degrades in blood and liver, and is eliminated via glomerular filtration. Safety profile of the preparation relating to general toxicity, immunotoxicity and allergenicity was assessed in rodents. It was demonstrated that LysECD7-SMAP in potential therapeutic (12.5 mg/kg), 10-fold (125 mg/kg) and 40-fold (500 mg/kg) doses showed no signs of intoxication and significant abnormalities after single and repeated i.v. administrations, preparation was non-immunogenic and induced minor and reversible allergic reaction in animals.
Collapse
Affiliation(s)
- Nataliia P Antonova
- Laboratory of Pathogen Population Variability Mechanisms, N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Daria V Vasina
- Laboratory of Pathogen Population Variability Mechanisms, N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Igor V Grigoriev
- Translational Biomedicine Laboratory, N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Aleksei I Laishevtsev
- Federal State Budget Scientific Institution "Federal Scientific Centre VIEV" (FSC VIEV), Moscow, Russia
| | - Andrey V Kapustin
- Federal State Budget Scientific Institution "Federal Scientific Centre VIEV" (FSC VIEV), Moscow, Russia
| | - Vasiliy A Savinov
- Federal State Budget Scientific Institution "Federal Scientific Centre VIEV" (FSC VIEV), Moscow, Russia
| | - Aleksei M Vorobev
- Laboratory of Clinical Microbiology and Biotechnology of Bacteriophages, G.N. Gabrichevsky Moscow Research Institute for Epidemiology and Microbiology, Moscow, Russia
| | - Andrei V Aleshkin
- Laboratory of Clinical Microbiology and Biotechnology of Bacteriophages, G.N. Gabrichevsky Moscow Research Institute for Epidemiology and Microbiology, Moscow, Russia
| | - Anastasia A Zackharova
- Translational Biomedicine Laboratory, N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Timofey A Remizov
- Translational Biomedicine Laboratory, N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Valentine V Makarov
- Centre for Strategic Planning and Management of Biomedical Health Risks of the Federal Medical Biological Agency, Moscow, Russia
| | - Sergey M Yudin
- Centre for Strategic Planning and Management of Biomedical Health Risks of the Federal Medical Biological Agency, Moscow, Russia
| | - Vladimir A Gushchin
- Laboratory of Pathogen Population Variability Mechanisms, N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia.
| |
Collapse
|
16
|
Köhne M, Hüsch R, Tönissen A, Schmidt M, Müsken M, Böttcher D, Hirnet J, Plötz M, Kittler S, Sieme H. Isolation and characterization of bacteriophages specific to Streptococcus equi subspecies zooepidemicus and evaluation of efficacy ex vivo. Front Microbiol 2024; 15:1448958. [PMID: 39529671 PMCID: PMC11550937 DOI: 10.3389/fmicb.2024.1448958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024] Open
Abstract
Streptococcus (S.) equi subspecies (subsp.) zooepidemicus is an important facultative pathogen in horses and can cause severe infections in other species including humans. Facing the post-antibiotic era, novel antimicrobials are needed for fighting bacterial infections. Bacteriophages (phages) are the natural predators of bacteria and discussed as a promising antimicrobial treatment option. The objective of this study was to isolate and characterize S. equi subsp. zooepidemicus-specific phages for the first time and to evaluate their efficacy in vitro and ex vivo. In total, 13 phages with lytic activity were isolated and host ranges were determined. Two phages with broad host ranges and high efficiency of plating (vB_SeqZP_LmqsRe26-2 (lytic activity: 30/37 bacterial isolates) and vB_SeqZP_LmqsRe26-3 (lytic activity: 29/37 bacterial isolates)) and one phage with relatively low efficiency of plating (vB_SeqZP_LmqsRe26-1) were selected for further characterization, including electron microscopy and whole genome sequencing. In in vitro planktonic killing assays at two tested multiplicities of infection (MOI 1 and MOI 10), significant bacterial growth reduction was observed when the phages vB_SeqZP_LmqsRe26-2 and vB_SeqZP_LmqsRe26-3 were added. These phages were subsequently co-incubated with clinical S. equi subsp. zooepidemicus isolates in an equine endometrial explant model but did not achieve bacterial growth reduction at MOI 1 and MOI 10. However, helium ion microscopy revealed presence of particles adherent to the bacteria on the explant after incubation (25 h), suggesting possible phage-bacteria interactions. In conclusion, phages against S. equi subsp. zooepidemicus were successfully isolated and characterized. Promising results were observed in in vitro but no significant reduction was detected in ex vivo experiments, requiring additional investigations. However, after further adaptations (e.g., optimization of MOIs and phage administration or use of phage-antibiotic combination), phages could be a potential antimicrobial tool for future therapeutic use in S. equi subsp. zooepidemicus infections, although the available results do not currently support the therapeutic usage.
Collapse
Affiliation(s)
- Martin Köhne
- Unit for Reproductive Medicine – Clinic for Horses, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Ronja Hüsch
- Unit for Reproductive Medicine – Clinic for Horses, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Anna Tönissen
- Unit for Reproductive Medicine – Clinic for Horses, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Matthias Schmidt
- Department of Technical Biogeochemistry, Helmholtz Centre for Environmental Research –UFZ, Leipzig, Germany
| | - Mathias Müsken
- Central Facility for Microscopy, Helmholtz Centre for Infection Research – HZI, Braunschweig, Germany
| | - Denny Böttcher
- Institute for Veterinary Pathology, Faculty of Veterinary Medicine, Leipzig University, Leipzig, Germany
| | - Juliane Hirnet
- Institute of Food Quality and Food Safety, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Madeleine Plötz
- Institute of Food Quality and Food Safety, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Sophie Kittler
- Institute of Food Quality and Food Safety, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Harald Sieme
- Unit for Reproductive Medicine – Clinic for Horses, University of Veterinary Medicine, Foundation, Hannover, Germany
| |
Collapse
|
17
|
Rastegar S, Skurnik M, Tadjrobehkar O, Samareh A, Samare-Najaf M, Lotfian Z, Khajedadian M, Hosseini-Nave H, Sabouri S. Synergistic effects of bacteriophage cocktail and antibiotics combinations against extensively drug-resistant Acinetobacter baumannii. BMC Infect Dis 2024; 24:1208. [PMID: 39455951 PMCID: PMC11515142 DOI: 10.1186/s12879-024-10081-0] [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: 07/14/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND The extensively drug-resistant (XDR) strains of Acinetobacter baumannii have become a major cause of nosocomial infections, increasing morbidity and mortality worldwide. Many different treatments, including phage therapy, are attractive ways to overcome the challenges of antibiotic resistance. METHODS This study investigates the biofilm formation ability of 30 XDR A. baumannii isolates and the efficacy of a cocktail of four tempetate bacteriophages (SA1, Eve, Ftm, and Gln) and different antibiotics (ampicillin/sulbactam, meropenem, and colistin) in inhibiting and degrading the biofilms of these strains. RESULTS The majority (83.3%) of the strains exhibited strong biofilm formation. The bacteriophage cocktail showed varying degrees of effectiveness against A. baumannii biofilms, with higher concentrations generally leading to more significant inhibition and degradation rates. The antibiotics-bacteriophage cocktail combinations also enhanced the inhibition and degradation of biofilms. CONCLUSION The findings suggested that the bacteriophage cocktail is an effective tool in combating A. baumannii biofilms, with its efficacy depending on the concentration. Combining antibiotics with the bacteriophage cocktail improved the inhibition and removal of biofilms, indicating a promising strategy for managing A. baumannii infections. These results contribute to our understanding of biofilm dynamics and the potential of bacteriophage cocktails as a novel therapeutic approach to combat antibiotic-resistant bacteria.
Collapse
Affiliation(s)
- Sanaz Rastegar
- Student Research Committee, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Medical Mycology and Bacteriology Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Mikael Skurnik
- Department of Bacteriology and Immunology, Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Omid Tadjrobehkar
- Medical Mycology and Bacteriology Research Center, Kerman University of Medical Sciences, Kerman, Iran
- Department of Medical Microbiology(Bacteriology and Virology), Afzalipour School of Medicine, Kerman, Iran
| | - Ali Samareh
- Department of Biochemistry, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Samare-Najaf
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Zahra Lotfian
- Medical Mycology and Bacteriology Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Maryam Khajedadian
- Medical Mycology and Bacteriology Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Hossein Hosseini-Nave
- Medical Mycology and Bacteriology Research Center, Kerman University of Medical Sciences, Kerman, Iran.
- Department of Medical Microbiology(Bacteriology and Virology), Afzalipour School of Medicine, Kerman, Iran.
- Department of Microbiology and Virology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| | - Salehe Sabouri
- Extremophile and Productive Microorganisms Research Center, Kerman University of Medical Sciences, Kerman, Iran.
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
18
|
Gangakhedkar R, Jain V. Elucidating the molecular properties and anti-mycobacterial activity of cysteine peptidase domain of D29 mycobacteriophage endolysin. J Virol 2024; 98:e0132824. [PMID: 39287392 PMCID: PMC11494882 DOI: 10.1128/jvi.01328-24] [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: 07/30/2024] [Accepted: 08/20/2024] [Indexed: 09/19/2024] Open
Abstract
Emergence of antibiotic resistance in pathogenic Mycobacterium tuberculosis (Mtb) has elevated tuberculosis to a serious global threat, necessitating alternate solutions for its eradication. D29 mycobacteriophage can infect and kill several mycobacterial species including Mtb. It encodes an endolysin LysA to hydrolyze host bacteria peptidoglycan for progeny release. We previously showed that out of the two catalytically active domains of LysA [N-terminal domain (NTD) and lysozyme-like domain], NTD, when ectopically expressed in Mycobacterium smegmatis (Msm), is able to kill the bacterium nearly as efficiently as full-length LysA. Here, we dissected the functioning of NTD to develop it as a phage-derived small molecule anti-mycobacterial therapeutic. We performed a large-scale site-directed mutagenesis of the conserved residues in NTD and examined its structure, stability, and function using molecular dynamic simulations coupled with biophysical and biochemical experiments. Our data show that NTD functions as a putative cysteine peptidase with a catalytic triad composed of Cys41, His112, and Glu137, acting as nucleophile, base, and acid, respectively, and showing characteristics similar to the NlpC/P60 family of cysteine peptidases. Additionally, our peptidoglycan hydrolysis assays suggested that NTD hydrolyzes only mycobacterial peptidoglycan and does not act on Gram-positive and Gram-negative bacterial peptidoglycans. More importantly, the combined activity of exogenously added NTD and sub-lethal doses of anti-mycobacterial drugs kills Msm in vitro and exhibits disruption of pre-formed mycobacterial biofilm. We additionally show that NTD treatment increases the permeability of antibiotics in Msm, which reduces the minimum inhibitory concentration of the antibiotics. Collectively, we present NTD as a promising phage-derived therapeutic against mycobacteria.IMPORTANCEMycobacteriophages are the viruses that use mycobacteria as host for their progeny production and, in the process, kill them. Mycobacteriophages are, therefore, considered as promising alternatives to antibiotics for killing pathogenic Mycobacterium tuberculosis. The endolysin LysA produced by mycobacteriophage D29 plays an important role in host cell lysis and virion release. Our work presented here highlights the functioning of LysA's N-terminal catalytic domain (NTD) in order to develop it as phage-derived small molecule therapeutics. We show that combined treatment of exogenously added NTD and sub-lethal doses of anti-mycobacterial drugs kills M. smegmatis, shows synergism by reducing the minimum inhibitory concentration of these antibiotics, and exhibits disruption of pre-formed mature biofilm. These outcomes and our detailed biochemical and biophysical dissection of the protein further pave the way toward engineering and development of NTD as a promising therapeutic against mycobacterial infections such as tuberculosis.
Collapse
Affiliation(s)
- Rutuja Gangakhedkar
- Microbiology and Molecular Biology Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, India
| | - Vikas Jain
- Microbiology and Molecular Biology Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, India
| |
Collapse
|
19
|
Chu D, Lan J, Liang L, Xia K, Li L, Yang L, Liu H, Zhang T. The antibacterial activity of a novel highly thermostable endolysin, LysKP213, against Gram-negative pathogens is enhanced when combined with outer membrane permeabilizing agents. Front Microbiol 2024; 15:1454618. [PMID: 39439944 PMCID: PMC11493673 DOI: 10.3389/fmicb.2024.1454618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024] Open
Abstract
Phages and phage-encoded lytic enzymes are promising antimicrobial agents. In this study, we report the isolation and identification of bacteriophage KP2025 from Klebsiella pneumoniae. Bioinformatics analysis of KP2025 revealed a putative endolysin, LysKP213, containing a T4-like_lys domain. Purified LysKP213 was found to be highly thermostable, retaining approximately 44.4% of its lytic activity after 20 h of incubation at 95°C, and approximately 57.5% residual activity after 30 min at 121°C. Furthermore, when administered in combination with polymyxin B or fused at the N-terminus with the antimicrobial peptide cecropin A (CecA), LysKP213 exhibited increased antibacterial activity against Gram-negative pathogens, including K. pneumoniae, Pseudomonas aeruginosa, Acinetobacter baumannii, and Escherichia coli, both in vitro and in vivo. These results indicated that LysKP213 is a highly thermostable endolysin that, when combined with or fused with an outer membrane permeabilizer, has enhanced antibacterial activity and is a candidate agent for the control of infections by Gram-negative pathogens.
Collapse
Affiliation(s)
- Dingjian Chu
- Engineering Research Center of Health Medicine Biotechnology of Institution of Higher Education of Guizhou Province, School of Biology and Engineering (School of Modern Industry for Health and Medicine), Guizhou Medical University, Guiyang, China
| | - Jing Lan
- Engineering Research Center of Health Medicine Biotechnology of Institution of Higher Education of Guizhou Province, School of Biology and Engineering (School of Modern Industry for Health and Medicine), Guizhou Medical University, Guiyang, China
| | - Lu Liang
- Guiyang Maternal and Child Health Hospital, Guiyang, China
| | - Kaide Xia
- Guiyang Maternal and Child Health Hospital, Guiyang, China
| | - Linlin Li
- Shanghai Institute of Phage, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Lan Yang
- Shanghai Institute of Phage, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Hongmei Liu
- Engineering Research Center of Health Medicine Biotechnology of Institution of Higher Education of Guizhou Province, School of Biology and Engineering (School of Modern Industry for Health and Medicine), Guizhou Medical University, Guiyang, China
| | - Tingting Zhang
- Engineering Research Center of Health Medicine Biotechnology of Institution of Higher Education of Guizhou Province, School of Biology and Engineering (School of Modern Industry for Health and Medicine), Guizhou Medical University, Guiyang, China
| |
Collapse
|
20
|
Mayorga-Ramos A, Carrera-Pacheco SE, Barba-Ostria C, Guamán LP. Bacteriophage-mediated approaches for biofilm control. Front Cell Infect Microbiol 2024; 14:1428637. [PMID: 39435185 PMCID: PMC11491440 DOI: 10.3389/fcimb.2024.1428637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 07/29/2024] [Indexed: 10/23/2024] Open
Abstract
Biofilms are complex microbial communities in which planktonic and dormant bacteria are enveloped in extracellular polymeric substances (EPS) such as exopolysaccharides, proteins, lipids, and DNA. These multicellular structures present resistance to conventional antimicrobial treatments, including antibiotics. The formation of biofilms raises considerable concern in healthcare settings, biofilms can exacerbate infections in patients and compromise the integrity of medical devices employed during treatment. Similarly, certain bacterial species contribute to bulking, foaming, and biofilm development in water environments such as wastewater treatment plants, water reservoirs, and aquaculture facilities. Additionally, food production facilities provide ideal conditions for establishing bacterial biofilms, which can serve as reservoirs for foodborne pathogens. Efforts to combat antibiotic resistance involve exploring various strategies, including bacteriophage therapy. Research has been conducted on the effects of phages and their individual proteins to assess their potential for biofilm removal. However, challenges persist, prompting the examination of refined approaches such as drug-phage combination therapies, phage cocktails, and genetically modified phages for clinical applications. This review aims to highlight the progress regarding bacteriophage-based approaches for biofilm eradication in different settings.
Collapse
Affiliation(s)
- Arianna Mayorga-Ramos
- Universidad UTE, Centro de Investigación Biomédica (CENBIO), Facultad de Ciencias de la Salud Eugenio Espejo, Quito, Ecuador
| | - Saskya E. Carrera-Pacheco
- Universidad UTE, Centro de Investigación Biomédica (CENBIO), Facultad de Ciencias de la Salud Eugenio Espejo, Quito, Ecuador
| | - Carlos Barba-Ostria
- Escuela de Medicina, Colegio de Ciencias de la Salud Quito, Universidad San Francisco de Quito USFQ, Quito, Ecuador
- Instituto de Microbiología, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| | - Linda P. Guamán
- Universidad UTE, Centro de Investigación Biomédica (CENBIO), Facultad de Ciencias de la Salud Eugenio Espejo, Quito, Ecuador
| |
Collapse
|
21
|
Ghate MM, Gulati K, Poluri KM. Alginate binding enhances the structural stability and potentiates the lytic activity of bacteriophage endolysin's partially folded conformation. Arch Biochem Biophys 2024; 760:110129. [PMID: 39159898 DOI: 10.1016/j.abb.2024.110129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 08/21/2024]
Abstract
Polysaccharide polymers are increasingly being used as chaperon-like macromolecules in assisting protein folding of unfolded protein molecules. They interact with unfolded or partially folded proteins in a charge and conformation specific manner that results in the formation of stable protein-polysaccharide complexes. In most of the cases, the complex formation of protein-polysaccharide is driven via non-covalent interactions that have found to endorse the activity of proteins. T4L (18.7 kDa) and T7L (17 kDa) endolysins belong to the hydrolase and amidase class of peptidoglycan degrading enzymes. Both T4L and T7L exist in partially folded forms and are devoid of lytic activity at low pH conditions. In the current study, we assessed the binding of alginate with T4L and T7L at pH 7 and 3 using variety of biophysical and biochemical techniques. Spectroscopic studies revealed differential structural modulations of partially folded T4L and T7L upon their interaction with alginate. Further, the complex formation of alginate with partially folded T4L/T7L was confirmed by ITC and STEM. Additionally, the formed complexes of alginate with both T4L/T7L PF endolysins were found to be chemically and enzymatically stable. Moreover, such complexes were also marked with differential enhancement in their lytic activities at acidic pH conditions. This implied the potency of alginate as an excellent choice of matrix to preserve the structural and functional integrity of partially folded forms of T4L and T7L at highly acidic conditions.
Collapse
Affiliation(s)
- Mayur Mohan Ghate
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee-247667, Uttarakhand, India
| | - Khushboo Gulati
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee-247667, Uttarakhand, India
| | - Krishna Mohan Poluri
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee-247667, Uttarakhand, India; Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee-247667, Uttarakhand, India.
| |
Collapse
|
22
|
Pattnaik A, Pati S, Samal SK. Bacteriophage as a potential biotherapeutics to combat present-day crisis of multi-drug resistant pathogens. Heliyon 2024; 10:e37489. [PMID: 39309956 PMCID: PMC11416503 DOI: 10.1016/j.heliyon.2024.e37489] [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: 05/13/2024] [Revised: 08/15/2024] [Accepted: 09/04/2024] [Indexed: 09/25/2024] Open
Abstract
The rise of Multi-Drug Resistant (MDR) bacterial pathogens to most, if not all, currently available antibacterial agents has become a global threat. As a consequence of the antibiotic resistance epidemic, phage therapy has emerged as a potential alternative to conventional antibiotics. Despite the high therapeutic advantages of phage therapy, they have not yet been successfully used in the clinic due to various limitations of narrow host specificity compared to antibiotics, poor adhesion on biofilm surface, and susceptibility to both human and bacterial defences. This review focuses on the antibacterial effect of bacteriophage and their recent clinical trials with a special emphasis on the underlying mechanism of lytic phage action with the help of endolysin and holin. Furthermore, recent clinical trials of natural and modified endolysins and some marketed products have also been emphasized with future prospective.
Collapse
Affiliation(s)
- Ananya Pattnaik
- ICMR-Regional Medical Research Center, Bhubaneswar, Odisha, India
- KSBT, Kalinga Institute of Industrial Technology, Bhubaneswar, Odisha, India
| | - Sanghamitra Pati
- ICMR-Regional Medical Research Center, Bhubaneswar, Odisha, India
| | | |
Collapse
|
23
|
Zhang M, Xu X, Lv L, Luo J, Ahmed T, Alsakkaf WAA, Ali HM, Bi J, Yan C, Gu C, Shou L, Li B. Genomic Characterization of Phage ZP3 and Its Endolysin LysZP with Antimicrobial Potential against Xanthomonas oryzae pv. oryzae. Viruses 2024; 16:1450. [PMID: 39339926 PMCID: PMC11437452 DOI: 10.3390/v16091450] [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: 07/09/2024] [Revised: 09/08/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Xanthomonas oryzae pv. oryzae (Xoo) is a significant bacterial pathogen responsible for outbreaks of bacterial leaf blight in rice, posing a major threat to rice cultivation worldwide. Effective management of this pathogen is crucial for ensuring rice yield and food security. In this study, we identified and characterized a novel Xoo phage, ZP3, isolated from diseased rice leaves in Zhejiang, China, which may offer new insights into biocontrol strategies against Xoo and contribute to the development of innovative approaches to combat bacterial leaf blight. Transmission electron microscopy indicated that ZP3 had a short, non-contractile tail. Genome sequencing and bioinformatic analysis showed that ZP3 had a double-stranded DNA genome with a length of 44,713 bp, a G + C content of 52.2%, and 59 predicted genes, which was similar to other OP1-type Xoo phages belonging to the genus Xipdecavirus. ZP3's endolysin LysZP was further studied for its bacteriolytic action, and the N-terminal transmembrane domain of LysZP is suggested to be a signal-arrest-release sequence that mediates the translocation of LysZP to the periplasm. Our study contributes to the understanding of phage-Xoo interactions and suggests that phage ZP3 and its endolysin LysZP could be developed into biocontrol agents against this phytopathogen.
Collapse
Affiliation(s)
- Muchen Zhang
- State Key Laboratory of Rice Biology and Breeding, Ministry of Agriculture Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Zhejiang Key Laboratory of Biology and Ecological Regulation of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou 310058, China; (M.Z.); (X.X.); (L.L.); (T.A.)
- Food Quality Supervision, Inspection and Testing Center of the Ministry of Agriculture and Rural Affairs (Shanghai), Shanghai Center of Agricultural Products Quality Safety, Shanghai 201708, China
| | - Xinyan Xu
- State Key Laboratory of Rice Biology and Breeding, Ministry of Agriculture Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Zhejiang Key Laboratory of Biology and Ecological Regulation of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou 310058, China; (M.Z.); (X.X.); (L.L.); (T.A.)
| | - Luqiong Lv
- State Key Laboratory of Rice Biology and Breeding, Ministry of Agriculture Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Zhejiang Key Laboratory of Biology and Ecological Regulation of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou 310058, China; (M.Z.); (X.X.); (L.L.); (T.A.)
| | - Jinyan Luo
- Department of Plant Quarantine, Shanghai Extension and Service Center of Agriculture Technology, Shanghai 201103, China;
| | - Temoor Ahmed
- State Key Laboratory of Rice Biology and Breeding, Ministry of Agriculture Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Zhejiang Key Laboratory of Biology and Ecological Regulation of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou 310058, China; (M.Z.); (X.X.); (L.L.); (T.A.)
- Department of Life Sciences, Western Caspian University, Baku AZ1001, Azerbaijan
- MEU Research Unit, Middle East University, Amman 11192, Jordan
| | - Waleed A. A. Alsakkaf
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (W.A.A.A.); (H.M.A.)
| | - Hayssam M. Ali
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (W.A.A.A.); (H.M.A.)
| | - Ji’an Bi
- Crop Institute, Ningbo Academy of Agricultural Sciences, Ningbo 315040, China; (J.B.); (C.Y.)
| | - Chengqi Yan
- Crop Institute, Ningbo Academy of Agricultural Sciences, Ningbo 315040, China; (J.B.); (C.Y.)
| | - Chunyan Gu
- Anhui Province Key Laboratory of Pesticide Resistance Management on Grain and Vegetable Pests, Institute of Plant Protection and Agro-Products Safety, Anhui Academy of Agricultural Sciences, Hefei 230031, China
| | - Linfei Shou
- Station for the Plant Protection & Quarantine and Control of Agrochemicals Zhejiang Province, Hangzhou 310004, China
| | - Bin Li
- State Key Laboratory of Rice Biology and Breeding, Ministry of Agriculture Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Zhejiang Key Laboratory of Biology and Ecological Regulation of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou 310058, China; (M.Z.); (X.X.); (L.L.); (T.A.)
| |
Collapse
|
24
|
Choi YJ, Kim S, Dahal RH, Kim J. A Novel Truncated CHAP Modular Endolysin, CHAP SAP26-161, That Lyses Staphylococcus aureus, Acinetobacter baumannii, and Clostridioides difficile, and Exhibits Therapeutic Effects in a Mouse Model of A. baumannii Infection. J Microbiol Biotechnol 2024; 34:1718-1726. [PMID: 39081246 PMCID: PMC11380504 DOI: 10.4014/jmb.2402.02042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/27/2024] [Accepted: 05/29/2024] [Indexed: 08/29/2024]
Abstract
Development of novel antibacterial agents is imperative due to the increasing threat of antibiotic-resistant pathogens. This study aimed to develop the enhanced antibacterial activity and in-vivo efficacy of a novel truncated endolysin, CHAPSAP26-161, derived from the endolysin LysSAP26, against multidrug-resistant bacteria. CHAPSAP26-161 exhibited higher protein purification efficiency in E. coli and antibacterial activity than LysSAP26. Moreover, CHAPSAP26-161 showed the higher lytic activity against A. baumannii with minimal bactericidal concentrations (MBCs) of 5-10 μg/ml, followed by Staphylococcus aureus with MBCs of 10-25 μg/ml. Interestingly, CHAPSAP26-161 could lyse anaerobic bacteria, such as Clostridioides difficile, with MBCs of 25-50 μg/ml. At pH 4-8 and temperatures of 4°C-45°C, CHAPSAP26-161 maintained antibacterial activity without remarkable difference. The lytic activity of CHAPSAP26-161 was increased with Zn2+. In vivo tests demonstrated the therapeutic effects of CHAPSAP26-161 in murine systemic A. baumannii infection model. In conclusion, CHAPSAP26-161, a truncated endolysin that retains only the CHAP domain from LysSAP26, demonstrated enhanced protein purification efficiency and antibacterial activity compared to LysSAP26. It further displayed broad-spectrum antibacterial effects against S. aureus, A. baumannii, and C. difficile. Our in vitro and in-vivo results of CHAPSAP26-161 highlights its promise as an innovative therapeutic option against those bacteria with multiple antibiotic resistance.
Collapse
Affiliation(s)
- Yoon-Jung Choi
- Department of Microbiology, School of Medicine, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Shukho Kim
- Department of Microbiology, School of Medicine, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Ram Hari Dahal
- Department of Microbiology, School of Medicine, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Jungmin Kim
- Department of Microbiology, School of Medicine, Kyungpook National University, Daegu 41566, Republic of Korea
| |
Collapse
|
25
|
Awad MM, Suraweera CD, Vidor CJ, Ye-Lin AY, Williams GC, Mileto SJ, Barlow CK, McGowan S, Lyras D. A Clostridioides difficile endolysin modulates toxin secretion without cell lysis. Commun Biol 2024; 7:1044. [PMID: 39179651 PMCID: PMC11344133 DOI: 10.1038/s42003-024-06730-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 08/13/2024] [Indexed: 08/26/2024] Open
Abstract
The Clostridia produce and secrete Large Clostridial Glucosylating Toxins (LCGTs) responsible for disease symptoms, but the secretion mechanism is largely unknown. Recently, a holin-like protein was shown to be essential for toxin secretion. Holins, typically bacteriophage-specific proteins, are part of the holin-endo(lysin) system that releases phage progeny. To determine if the clostridia also use a lysin, we investigated two conserved putative lysins, M7404_01910 and M7404_02200, in the release of the LCGTs TcdA and TcdB from a Clostridioides difficile ribotype 027 strain, M7404. Sequence analysis and structural modelling indicates that both proteins are related to N-acetylmuramoyl-l-alanine amidases, similar to CD27L, a lysin from the C. difficile phage ΦCD27. Disruption of these genes reveal that only M7404_02200 contributes to toxin secretion and does so in a non-lytic fashion. Peptidoglycan hydrolysis assays show that recombinant M7404_02200 is an active peptidoglycan amidase, confirming its role in TcdA and TcdB secretion in C. difficile M7404.
Collapse
Affiliation(s)
- Milena M Awad
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, 3800, Australia
| | - Chathura D Suraweera
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, 3800, Australia
| | - Callum J Vidor
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, 3800, Australia
| | - Auberon Y Ye-Lin
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, 3800, Australia
| | - Galain C Williams
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, 3800, Australia
| | - Steven J Mileto
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, 3800, Australia
| | - Christopher K Barlow
- Department of Biochemistry, Biomedicine Discovery Institute, Monash University, Clayton, 3800, Australia
- Monash Proteomics & Metabolomics Platform, Monash University, Clayton, 3800, Australia
| | - Sheena McGowan
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, 3800, Australia
| | - Dena Lyras
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, 3800, Australia.
| |
Collapse
|
26
|
Yoda T, Matsuhashi A, Matsushita A, Shibagaki S, Sasakura Y, Aoki K, Hosokawa M, Tsuda S. Uncovering Endolysins against Methicillin-Resistant Staphylococcus aureus Using a Microbial Single-Cell Genome Database. ACS Infect Dis 2024; 10:2679-2689. [PMID: 38906534 PMCID: PMC11320564 DOI: 10.1021/acsinfecdis.4c00039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 04/18/2024] [Accepted: 05/22/2024] [Indexed: 06/23/2024]
Abstract
Endolysins, peptidoglycan hydrolases derived from bacteriophages (phages), are being developed as a promising alternative to conventional antibiotics. To obtain highly active endolysins, a diverse library of these endolysins is vital. We propose here microbial single-cell genome sequencing as an efficient tool to discover dozens of previously unknown endolysins, owing to its culture-independent sequencing method. As a proof of concept, we analyzed and recovered endolysin genes within prophage regions of Staphylococcus single-amplified genomes in human skin microbiome samples. We constructed a library of chimeric endolysins by shuffling domains of the natural endolysins and performed high-throughput screening against Staphylococcus aureus. One of the lead endolysins, bbst1027, exhibited desirable antimicrobial properties, such as rapid bactericidal activity, no detectable resistance development, and in vivo efficacy. We foresee that this endolysin discovery pipeline is in principle applicable to any bacterial target and boost the development of novel antimicrobial agents.
Collapse
Affiliation(s)
- Takuya Yoda
- bitBiome,
Inc., 513 Wasedatsurumaki-cho, Shinjuku-ku, Tokyo 162-0041, Japan
| | - Ayumi Matsuhashi
- bitBiome,
Inc., 513 Wasedatsurumaki-cho, Shinjuku-ku, Tokyo 162-0041, Japan
| | - Ai Matsushita
- bitBiome,
Inc., 513 Wasedatsurumaki-cho, Shinjuku-ku, Tokyo 162-0041, Japan
| | - Shohei Shibagaki
- bitBiome,
Inc., 513 Wasedatsurumaki-cho, Shinjuku-ku, Tokyo 162-0041, Japan
| | - Yukie Sasakura
- bitBiome,
Inc., 513 Wasedatsurumaki-cho, Shinjuku-ku, Tokyo 162-0041, Japan
| | - Kazuteru Aoki
- bitBiome,
Inc., 513 Wasedatsurumaki-cho, Shinjuku-ku, Tokyo 162-0041, Japan
| | - Masahito Hosokawa
- bitBiome,
Inc., 513 Wasedatsurumaki-cho, Shinjuku-ku, Tokyo 162-0041, Japan
- Department
of Life Science and Medical Bioscience, Waseda University, 2-2
Wakamatsu-cho, Shinjuku-ku, Tokyo 162-8480, Japan
- Research
Organization for Nano and Life Innovation, Waseda University, 513
Wasedatsurumaki-cho, Shinjuku-ku, Tokyo 162-0041, Japan
- Institute
for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan
| | - Soichiro Tsuda
- bitBiome,
Inc., 513 Wasedatsurumaki-cho, Shinjuku-ku, Tokyo 162-0041, Japan
| |
Collapse
|
27
|
Arora R, Nadar K, Bajpai U. Discovery and characterization of a novel LysinB from F2 sub-cluster mycobacteriophage RitSun. Sci Rep 2024; 14:18073. [PMID: 39103410 PMCID: PMC11300654 DOI: 10.1038/s41598-024-68636-9] [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] [Received: 03/11/2024] [Accepted: 07/25/2024] [Indexed: 08/07/2024] Open
Abstract
The escalating antibiotic resistance in mycobacterial species poses a significant threat globally, necessitating an urgent need to find alternative solutions. Bacteriophage-derived endolysins, which facilitate phage progeny release by attacking bacterial cell walls, present promising antibacterial candidates due to their rapid lytic action, high specificity and low risk of resistance development. In mycobacteria, owing to the complex, hydrophobic cell wall, mycobacteriophages usually synthesize two endolysins: LysinA, which hydrolyzes peptidoglycan; LysinB, which delinks mycolic acid-containing outer membrane and arabinogalactan, releasing free mycolic acid. In this study, we conducted domain analysis and functional characterization of a novel LysinB from RitSun, an F2 sub-cluster mycobacteriophage from our phage collection. Several key properties of RitSun LysinB make it an important antimycobacterial agent: its ability to lyse Mycobacterium smegmatis from without, a higher than previously reported specific activity of 1.36 U/mg and its inhibitory effect on biofilm formation. Given the impermeable nature of the mycobacterial cell envelope, dissecting RitSun LysinB at the molecular level to identify its cell wall-destabilizing sequence could be utilized to engineer other native lysins as fusion proteins, broadening their activity spectrum.
Collapse
Affiliation(s)
- Ritu Arora
- Department of Biomedical Science, Acharya Narendra Dev College, University of Delhi, Govindpuri, Kalkaji, New Delhi, 110019, India
| | - Kanika Nadar
- Department of Biomedical Science, Acharya Narendra Dev College, University of Delhi, Govindpuri, Kalkaji, New Delhi, 110019, India
| | - Urmi Bajpai
- Department of Biomedical Science, Acharya Narendra Dev College, University of Delhi, Govindpuri, Kalkaji, New Delhi, 110019, India.
| |
Collapse
|
28
|
Hasan M, Kim J, Liao X, Ding T, Ahn J. Antibacterial activity of bacteriophage-encoded endolysins against planktonic and biofilm cells of pathogenic Escherichia coli. Microb Pathog 2024; 193:106780. [PMID: 38969189 DOI: 10.1016/j.micpath.2024.106780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 06/25/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
This study was designed to assess the possibility of using bacteriophage-encoded endolysins for controlling planktonic and biofilm cells. The endolysins, LysEP114 and LysEP135, were obtained from plasmid vectors containing the endolysin genes derived from Escherichia coli phages. The high identity (>96 %) was observed between LysEP114 and LysEP135. LysEP114 and LysEP135 were characterized by pH, thermal, and lactic acid stability, lytic spectrum, antibacterial activity, and biofilm eradication. The molecular masses of LysEP114 and LysEP135 were 18.2 kDa, identified as muramidases. LysEP114 and LysEP135 showed high lytic activity against the outer membrane-permeabilized E. coli KCCM 40405 at below 37 °C, between pH 5 to 11, and below 70 mM of lactic acid. LysEP114 and LysEP135 showed the broad rang of lytic activity against E. coli KACC 10115, S. Typhimurium KCCM 40253, S. Typhimurium CCARM 8009, tetracycline-resistant S. Typhimurium, polymyxin B-resistant S. Typhimurium, chloramphenicol-resistant S. Typhimurium, K. pneumoniae ATCC 23357, K. pneumoniae CCARM 10237, and Shigella boydii KACC 10792. LysEP114 and LysEP135 effectively reduced the numbers of planktonic E. coli KCCM by 1.7 and 2.1 log, respectively, when treated with 50 mM lactic acid. The numbers of biofilm cells were reduced from 7.3 to 4.1 log CFU/ml and 2.2 log CFU/ml, respectively, when treated with LysEP114- and LysEP135 in the presence of 50 mM lactic acid. The results suggest that the endolysins in combination with lactic acid could be potential alternative therapeutic agents for controlling planktonic and biofilm cells.
Collapse
Affiliation(s)
- Mahadi Hasan
- Department of Biomedical Science, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Junhwan Kim
- Department of Biomedical Science, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Xinyu Liao
- Future Food Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, Zhejiang, 314100, China
| | - Tian Ding
- Future Food Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, Zhejiang, 314100, China; Department of Food Science and Nutrition, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Juhee Ahn
- Department of Biomedical Science, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea; Future Food Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, Zhejiang, 314100, China.
| |
Collapse
|
29
|
Bałdysz S, Nawrot R, Barylski J. "Tear down that wall"-a critical evaluation of bioinformatic resources available for lysin researchers. Appl Environ Microbiol 2024; 90:e0236123. [PMID: 38842338 PMCID: PMC11267937 DOI: 10.1128/aem.02361-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2024] Open
Abstract
Lytic enzymes, or lysins for short, break down peptidoglycan and interrupt the continuity of the cell wall, which, in turn, causes osmotic lysis of the bacterium. Their ability to destroy bacteria from within makes them promising antimicrobial agents that can be used as alternatives or supplements to antibiotics. In this paper, we briefly summarize basic terms and concepts used to describe lysin sequences and delineate major lysin groups. More importantly, we describe the domain repertoire found in lysins and critically review bioinformatic tools or databases which are used in studies of these enzymes (with particular emphasis on the repositories of Hidden Markov models). Finally, we present a novel comprehensive, meticulously curated set of lysin-related family and domain models, sort them into clusters that reflect major families, and demonstrate that the selected models can be used to efficiently search for new lysins.
Collapse
Affiliation(s)
- Sophia Bałdysz
- Department of Molecular Virology, Institute of Experimental Biology, Adam Mickiewicz University, Poznań, Poland
| | - Robert Nawrot
- Department of Molecular Virology, Institute of Experimental Biology, Adam Mickiewicz University, Poznań, Poland
| | - Jakub Barylski
- Department of Molecular Virology, Institute of Experimental Biology, Adam Mickiewicz University, Poznań, Poland
| |
Collapse
|
30
|
Xu L, Li X, Yang X, Zhao Y, Niu J, Jiang S, Ma J, Zhang X. Identification and Characterization of a Novel Prophage Lysin against Streptococcus dysgalactiae. Molecules 2024; 29:3411. [PMID: 39064988 PMCID: PMC11279900 DOI: 10.3390/molecules29143411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/13/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Streptococcus dysgalactiae infection can cause bovine mastitis and lead to huge economic losses for the dairy industry. The abuse of antibiotics has resulted in growing drug resistance of S. dysgalactiae, which causes hard-to-treat infections. Bacteriophage lysin, as a novel antibacterial agent, has great potential for application against drug-resistant gram-positive bacteria. However, few studies have been conducted on the prophage lysin of S. dysgalactiae. In this study, we mined a novel prophage lysin, named Lys1644, from a clinical S. dysgalactiae isolate by genome sequencing and bioinformatic analysis. Lys1644 was expressed and purified, and the lytic activity, antibacterial spectrum, optimal pH and temperature, lytic activity in milk in vitro, and synergistic bacteriostasis with antibiotics were assessed. The Lys1644 prophage lysin showed high bacteriolysis activity specifically on S. dysgalactiae, which resulted in CFU 100-fold reduction in milk. Moreover, Lys1644 maintained high activity over a wide pH range (pH 5-10) and a wide temperature range (4-42 °C). Synergistic bacteriostatic experiments showed that the combination of low-dose Lys1644 (50 μg/mL) with a subinhibitory concentration of aminoglycoside antibiotics (kanamycin or spectinomycin) can completely inhibit bacterial growth, suggesting that the combination of Lys1644 and antibiotics could be an effective therapeutic strategy against S. dysgalactiae infection.
Collapse
Affiliation(s)
- Linan Xu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai’an 271018, China; (L.X.)
- College of Agriculture and Forestry, Linyi University, Linyi 276005, China
| | - Xingshuai Li
- College of Agriculture and Forestry, Linyi University, Linyi 276005, China
| | - Xiangpeng Yang
- College of Agriculture and Forestry, Linyi University, Linyi 276005, China
| | - Yuzhong Zhao
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai’an 271018, China; (L.X.)
- College of Agriculture and Forestry, Linyi University, Linyi 276005, China
| | - Jianrui Niu
- College of Agriculture and Forestry, Linyi University, Linyi 276005, China
| | - Shijin Jiang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai’an 271018, China; (L.X.)
| | - Junfei Ma
- College of Agriculture and Forestry, Linyi University, Linyi 276005, China
| | - Xinglin Zhang
- College of Agriculture and Forestry, Linyi University, Linyi 276005, China
| |
Collapse
|
31
|
Mokhtari S, Li Y, Saris PEJ, Takala TM. Analysis of the cell wall binding domain in bacteriocin-like lysin LysL from Lactococcus lactis LAC460. Arch Microbiol 2024; 206:336. [PMID: 38954047 PMCID: PMC11219366 DOI: 10.1007/s00203-024-04066-5] [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: 03/27/2024] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 07/04/2024]
Abstract
Wild-type Lactococcus lactis strain LAC460 secretes prophage-encoded bacteriocin-like lysin LysL, which kills some Lactococcus strains, but has no lytic effect on the producer. LysL carries two N-terminal enzymatic active domains (EAD), and an unknown C-terminus without homology to known domains. This study aimed to determine whether the C-terminus of LysL carries a cell wall binding domain (CBD) for target specificity of LysL. The C-terminal putative CBD region of LysL was fused with His-tagged green fluorescent protein (HGFPuv). The HGFPuv_CBDlysL gene fusion was ligated into the pASG-IBA4 vector, and introduced into Escherichia coli. The fusion protein was produced and purified with affinity chromatography. To analyse the binding of HGFPuv_CBDLysL to Lactococcus cells, the protein was mixed with LysL-sensitive and LysL-resistant strains, including the LysL-producer LAC460, and the fluorescence of the cells was analysed. As seen in fluorescence microscope, HGFPuv_CBDLysL decorated the cell surface of LysL-sensitive L. cremoris MG1614 with green fluorescence, whereas the resistant L. lactis strains LM0230 and LAC460 remained unfluorescent. The fluorescence plate reader confirmed the microscopy results detecting fluorescence only from four tested LysL-sensitive strains but not from 11 tested LysL-resistant strains. Specific binding of HGFPuv_CBDLysL onto the LysL-sensitive cells but not onto the LysL-resistant strains indicates that the C-terminus of LysL contains specific CBD. In conclusion, this report presents experimental evidence of the presence of a CBD in a lactococcal phage lysin. Moreover, the inability of HGFPuv_CBDLysL to bind to the LysL producer LAC460 may partly explain the host's resistance to its own prophage lysin.
Collapse
Affiliation(s)
- Samira Mokhtari
- Department of Microbiology, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland.
| | - Yanru Li
- Department of Microbiology, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| | - Per E J Saris
- Department of Microbiology, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| | - Timo M Takala
- Department of Microbiology, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| |
Collapse
|
32
|
Raman SK, Siva Reddy DV, Jain V, Bajpai U, Misra A, Singh AK. Mycobacteriophages: therapeutic approach for mycobacterial infections. Drug Discov Today 2024; 29:104049. [PMID: 38830505 DOI: 10.1016/j.drudis.2024.104049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 05/07/2024] [Accepted: 05/29/2024] [Indexed: 06/05/2024]
Abstract
Tuberculosis (TB) is a significant global health threat, and cases of infection with non-tuberculous mycobacteria (NTM) causing lung disease (NTM-LD) are rising. Bacteriophages and their gene products have garnered interest as potential therapeutic options for bacterial infections. Here, we have compiled information on bacteriophages and their products that can kill Mycobacterium tuberculosis or NTM. We summarize the mechanisms whereby viable phages can access macrophage-resident bacteria and not elicit immune responses, review methodologies of pharmaceutical product development containing mycobacteriophages and their gene products, mainly lysins, in the context of drug regulatory requirements and we discuss industrially relevant methods for producing pharmaceutical products comprising mycobacteriophages, emphasizing delivery of mycobacteriophages to the lungs. We conclude with an outline of some recent case studies on mycobacteriophage therapy.
Collapse
Affiliation(s)
- Sunil Kumar Raman
- Pharmaceutics and Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - D V Siva Reddy
- Pharmaceutics and Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Vikas Jain
- Microbiology and Molecular Biology Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal 462066, India
| | - Urmi Bajpai
- Department of Biomedical Science, Acharya Narendra Dev College, University of Delhi, Govindpuri, Kalkaji , New Delhi 110019, India
| | - Amit Misra
- Pharmaceutics and Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Amit Kumar Singh
- Experimental Animal Facility, ICMR-National JALMA Institute for Leprosy & Other Mycobacterial Diseases, M. Miyazaki Marg, Tajganj, Agra 282004, Uttar Pradesh, India.
| |
Collapse
|
33
|
Choi YJ, Kim S, Shin M, Kim J. Isolation and Characterization of Novel Bacteriophages to Target Carbapenem-Resistant Acinetobacter baumannii. Antibiotics (Basel) 2024; 13:610. [PMID: 39061292 PMCID: PMC11273472 DOI: 10.3390/antibiotics13070610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
The spread of multidrug-resistant Acinetobacter baumannii in hospitals and nursing homes poses serious healthcare challenges. Therefore, we aimed to isolate and characterize lytic bacteriophages targeting carbapenem-resistant Acinetobacter baumannii (CRAB). Of the 21 isolated A. baumannii phages, 11 exhibited potent lytic activities against clinical isolates of CRAB. Based on host spectrum and RAPD-PCR results, 11 phages were categorized into four groups. Three phages (vB_AbaP_W8, vB_AbaSi_W9, and vB_AbaSt_W16) were further characterized owing to their antibacterial efficacy, morphology, and whole-genome sequence and were found to lyse 37.93%, 89.66%, and 37.93%, respectively, of the 29 tested CRAB isolates. The lytic spectrum of phages varied depending on the multilocus sequence type (MLST) of the CRAB isolates. The three phages contained linear double-stranded DNA genomes, with sizes of 41,326-166,741 bp and GC contents of 34.4-35.6%. Genome-wide phylogenetic analysis and single gene-based tree construction revealed no correlation among the three phages. Moreover, no genes were associated with lysogeny, antibiotic resistance, or bacterial toxins. Therefore, the three novel phages represent potential candidates for phage therapy against CRAB infections.
Collapse
Affiliation(s)
| | | | | | - Jungmin Kim
- Department of Microbiology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; (Y.-J.C.); (S.K.); (M.S.)
| |
Collapse
|
34
|
Kairamkonda M, Saxena H, Gulati K, Poluri KM. Analyzing the impact of T7L variants overexpression on the metabolic profile of Escherichia coli. Metabolomics 2024; 20:68. [PMID: 38941046 DOI: 10.1007/s11306-024-02133-y] [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: 03/08/2024] [Accepted: 05/16/2024] [Indexed: 06/29/2024]
Abstract
INTRODUCTION Exploring metabolic changes within host E. coli through an untargeted metabolomic study of T7L variants overexpression to optimize engineered endolysins for clinical/therapeutic use. AIM AND OBJECTIVE This study aims to assess the impact of overexpressing T7L variants on the metabolic profiles of E. coli. The two variants considered include T7L-H37A, which has enhanced lytic activity compared to its wild-type protein, and T7L-H48K, a dead mutant with no significant activity. METHODS 1H NMR-based metabolomics was employed to compare the metabolic profiles of E. coli cells overexpressing T7L wild-type protein and its variants. RESULTS Overexpression of the T7L wild-type (T7L-WT) protein and its variants (T7L-H48K and T7L-H37A) was compared to RNAP overexpression in E. coli cells using 1H NMR-based metabolomics, analyzing a total of 75 annotated metabolites, including organic acids, amino acids, sugars, and nucleic acids. The results showed distinct clustering patterns for the two T7L variant groups compared with the WT, in which the dead mutant (H48K) group showed clustering close to that of RNAP. Pathway impact analysis revealed different effects of T7L variants on E. coli metabolic profiles, with T7L-H48K showing minimal alterations in energy and amino acid pathways linked to osmotic stress compared to noticeable alterations in these pathways for both T7L-H37A and T7L-WT. CONCLUSIONS This study uncovered distinct metabolic fingerprints when comparing the overexpression of active and inactive mutants of T7L lytic enzymes in E. coli cells. These findings could contribute to the optimization and enhancement of suitable endolysins as potential alternatives to antibiotics.
Collapse
Affiliation(s)
- Manikyaprabhu Kairamkonda
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India
| | - Harshi Saxena
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India
| | - Khushboo Gulati
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India
| | - Krishna Mohan Poluri
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India.
- Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India.
| |
Collapse
|
35
|
Nakonieczna A, Abramowicz K, Kwiatek M, Kowalczyk E. Lysins as a powerful alternative to combat Bacillus anthracis. Appl Microbiol Biotechnol 2024; 108:366. [PMID: 38850320 PMCID: PMC11162388 DOI: 10.1007/s00253-024-13194-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 05/14/2024] [Accepted: 05/21/2024] [Indexed: 06/10/2024]
Abstract
This review gathers all, to the best of our current knowledge, known lysins, mainly bacteriophage-derived, that have demonstrated activity against Bacillus anthracis strains. B. anthracis is a spore-forming, toxin-producing bacteria, naturally dwelling in soil. It is best known as a potential biowarfare threat, an etiological agent of anthrax, and a severe zoonotic disease. Anthrax can be treated with antibiotics (ciprofloxacin, penicillin, doxycycline); however, their administration may take up even to 60 days, and different factors can compromise their effectiveness. Bacterial viruses, bacteriophages (phages), are natural enemies of bacteria and use their lytic enzymes, endolysins (lysins), to specifically kill bacterial cells. Harnessing the potential of lysins to combat bacterial infections holds promise for diminishing antibiotic usage and, consequently, addressing the escalating antibiotic resistance in bacteria. In this context, we list the lysins with the activity against B. anthracis, providing a summary of their lytic properties in vitro and the outcomes observed in animal models. Bacillus cereus strain ATCC 4342/RSVF1, a surrogate for B. anthracis, was also included as a target bacteria. KEY POINTS: • More than a dozen different B. anthracis lysins have been identified and studied. • They fall into three blocks regarding their amino acid sequence similarity and most of them are amidases. • Lysins could be used in treating B. anthracis infections.
Collapse
Affiliation(s)
- Aleksandra Nakonieczna
- Military Institute of Hygiene and Epidemiology, Biological Threats Identification and Countermeasure Center, Puławy, 24-100, Poland.
| | - Karolina Abramowicz
- Military Institute of Hygiene and Epidemiology, Biological Threats Identification and Countermeasure Center, Puławy, 24-100, Poland
| | - Magdalena Kwiatek
- Military Institute of Hygiene and Epidemiology, Biological Threats Identification and Countermeasure Center, Puławy, 24-100, Poland
| | | |
Collapse
|
36
|
Tyagi JL, Gupta P, Ghate MM, Kumar D, Poluri KM. Assessing the synergistic potential of bacteriophage endolysins and antimicrobial peptides for eradicating bacterial biofilms. Arch Microbiol 2024; 206:272. [PMID: 38772980 DOI: 10.1007/s00203-024-04003-6] [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: 03/13/2024] [Accepted: 05/14/2024] [Indexed: 05/23/2024]
Abstract
Phage-encoded endolysins have emerged as a potential substitute to conventional antibiotics due to their exceptional benefits including host specificity, rapid host killing, least risk of resistance. In addition to their antibacterial potency and biofilm eradication properties, endolysins are reported to exhibit synergism with other antimicrobial agents. In this study, the synergistic potency of endolysins was dissected with antimicrobial peptides to enhance their therapeutic effectiveness. Recombinantly expressed and purified bacteriophage endolysin [T7 endolysin (T7L); and T4 endolysin (T4L)] proteins have been used to evaluate the broad-spectrum antibacterial efficacy using different bacterial strains. Antibacterial/biofilm eradication studies were performed in combination with different antimicrobial peptides (AMPs) such as colistin, nisin, and polymyxin B (PMB) to assess the endolysin's antimicrobial efficacy and their synergy with AMPs. In combination with T7L, polymyxin B and colistin effectively eradicated the biofilm of Pseudomonas aeruginosa and exhibited a synergistic effect. Further, a combination of T4L and nisin displayed a synergistic effect against Staphylococcus aureus biofilms. In summary, the obtained results endorse the theme of combinational therapy consisting of endolysins and AMPs as an effective remedy against the drug-resistant bacterial biofilms that are a serious concern in healthcare settings.
Collapse
Affiliation(s)
- Jaya Lakshmi Tyagi
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India
| | - Payal Gupta
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India
- Department of Biotechnology, Graphic Era Deemed to be University, Dehradun, Uttarakhand, 248001, India
| | - Mayur Mohan Ghate
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India
| | - Dinesh Kumar
- Centre of Bio-Medical Research, SGPGIMS, Lucknow, Uttar Pradesh, 226014, India
| | - Krishna Mohan Poluri
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India.
- Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India.
| |
Collapse
|
37
|
Khan FM, Rasheed F, Yang Y, Liu B, Zhang R. Endolysins: a new antimicrobial agent against antimicrobial resistance. Strategies and opportunities in overcoming the challenges of endolysins against Gram-negative bacteria. Front Pharmacol 2024; 15:1385261. [PMID: 38831886 PMCID: PMC11144922 DOI: 10.3389/fphar.2024.1385261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 04/16/2024] [Indexed: 06/05/2024] Open
Abstract
Antibiotic-resistant bacteria are rapidly emerging, and the increasing prevalence of multidrug-resistant (MDR) Acinetobacter baumannii poses a severe threat to humans and healthcare organizations, due to the lack of innovative antibacterial drugs. Endolysins, which are peptidoglycan hydrolases encoded by a bacteriophage, are a promising new family of antimicrobials. Endolysins have been demonstrated as an effective therapeutic agent against bacterial infections of A. baumannii and many other Gram-positive and Gram-negative bacteria. Endolysin research has progressed from basic in vitro characterization to sophisticated protein engineering methodologies, including advanced preclinical and clinical testing. Endolysin are therapeutic agent that shows antimicrobial properties against bacterial infections caused by drug-resistant Gram-negative bacteria, there are still barriers to their implementation in clinical settings, such as safety concerns with outer membrane permeabilizers (OMP) use, low efficiency against stationary phase bacteria, and stability issues. The application of protein engineering and formulation techniques to improve enzyme stability, as well as combination therapy with other types of antibacterial drugs to optimize their medicinal value, have been reviewed as well. In this review, we summarize the clinical development of endolysin and its challenges and approaches for bringing endolysin therapies to the clinic. This review also discusses the different applications of endolysins.
Collapse
Affiliation(s)
- Fazal Mehmood Khan
- College of Civil and Transportation Engineering, Shenzhen University, Shenzhen, China
- Institute for Advanced Study, Shenzhen University, Shenzhen, China
| | - Fazal Rasheed
- Institute of Microscale Optoelectronics, Shenzhen University, Shenzhen, China
| | - Yunlan Yang
- Institute for Advanced Study, Shenzhen University, Shenzhen, China
| | - Bin Liu
- Institute for Advanced Study, Shenzhen University, Shenzhen, China
| | - Rui Zhang
- Institute for Advanced Study, Shenzhen University, Shenzhen, China
- Southern Marine Science and Engineering Guangdong Laboratory, Zhuhai, China
| |
Collapse
|
38
|
Chen W, Han LM, Chen XZ, Yi PC, Li H, Ren YY, Gao JH, Zhang CY, Huang J, Wang WX, Hu ZL, Hu CM. Engineered endolysin of Klebsiella pneumoniae phage is a potent and broad-spectrum bactericidal agent against "ESKAPEE" pathogens. Front Microbiol 2024; 15:1397830. [PMID: 38784808 PMCID: PMC11112412 DOI: 10.3389/fmicb.2024.1397830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 04/22/2024] [Indexed: 05/25/2024] Open
Abstract
The rise of antimicrobial resistance in ESKAPEE pathogens poses significant clinical challenges, especially in polymicrobial infections. Bacteriophage-derived endolysins offer promise in combating this crisis, but face practical hurdles. Our study focuses on engineering endolysins from a Klebsiella pneumoniae phage, fusing them with ApoE23 and COG133 peptides. We assessed the resulting chimeric proteins' bactericidal activity against ESKAPEE pathogens in vitro. ApoE23-Kp84B (CHU-1) reduced over 3 log units of CFU for A. baumannii, E. faecalis, K. pneumoniae within 1 h, while COG133-Kp84B (CHU-2) showed significant efficacy against S. aureus. COG133-L1-Kp84B, with a GS linker insertion in CHU-2, exhibited outstanding bactericidal activity against E. cloacae and P. aeruginosa. Scanning electron microscopy revealed alterations in bacterial morphology after treatment with engineered endolysins. Notably, CHU-1 demonstrated promising anti-biofilm and anti-persister cell activity against A. baumannii and E. faecalis but had limited efficacy in a bacteremia mouse model of their coinfection. Our findings advance the field of endolysin engineering, facilitating the customization of these proteins to target specific bacterial pathogens. This approach holds promise for the development of personalized therapies tailored to combat ESKAPEE infections effectively.
Collapse
Affiliation(s)
- Wei Chen
- Department of Tuberculosis, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Li-Mei Han
- Department of Tuberculosis, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
- Clinical Research Center, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiu-Zhen Chen
- Department of Infectious Diseases, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Peng-Cheng Yi
- Department of Tuberculosis, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Hui Li
- Department of Laboratory Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yun-Yao Ren
- Department of Tuberculosis, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Jing-Han Gao
- Clinical Research Center, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Cai-Yun Zhang
- Clinical Research Center, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Jing Huang
- Department of Clinical Laboratory, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Wei-Xiao Wang
- Department of Infectious Diseases, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhi-Liang Hu
- Department of Infectious Diseases, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Chun-Mei Hu
- Department of Tuberculosis, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
- Innovation Center for Infectious Diseases of Jiangsu Province, Nanjing, China
| |
Collapse
|
39
|
Szymczak M, Pankowski JA, Kwiatek A, Grygorcewicz B, Karczewska-Golec J, Sadowska K, Golec P. An effective antibiofilm strategy based on bacteriophages armed with silver nanoparticles. Sci Rep 2024; 14:9088. [PMID: 38643290 PMCID: PMC11032367 DOI: 10.1038/s41598-024-59866-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 04/15/2024] [Indexed: 04/22/2024] Open
Abstract
The emerging antibiotic resistance in pathogenic bacteria is a key problem in modern medicine that has led to a search for novel therapeutic strategies. A potential approach for managing such bacteria involves the use of their natural killers, namely lytic bacteriophages. Another effective method involves the use of metal nanoparticles with antimicrobial properties. However, the use of lytic phages armed with nanoparticles as an effective antimicrobial strategy, particularly with respect to biofilms, remains unexplored. Here, we show that T7 phages armed with silver nanoparticles exhibit greater efficacy in terms of controlling bacterial biofilm, compared with phages or nanoparticles alone. We initially identified a novel silver nanoparticle-binding peptide, then constructed T7 phages that successfully displayed the peptide on the outer surface of the viral head. These recombinant, AgNP-binding phages could effectively eradicate bacterial biofilm, even when used at low concentrations. Additionally, when used at concentrations that could eradicate bacterial biofilm, T7 phages armed with silver nanoparticles were not toxic to eukaryotic cells. Our results show that the novel combination of lytic phages with phage-bound silver nanoparticles is an effective, synergistic and safe strategy for the treatment of bacterial biofilms.
Collapse
Affiliation(s)
- Mateusz Szymczak
- Department of Molecular Virology, Faculty of Biology, Institute of Microbiology, University of Warsaw, Miecznikowa 1, 02-096, Warsaw, Poland
| | - Jarosław A Pankowski
- Department of Molecular Virology, Faculty of Biology, Institute of Microbiology, University of Warsaw, Miecznikowa 1, 02-096, Warsaw, Poland
- Dioscuri Centre for Physics and Chemistry of Bacteria, Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224, Warsaw, Poland
| | - Agnieszka Kwiatek
- Department of Molecular Virology, Faculty of Biology, Institute of Microbiology, University of Warsaw, Miecznikowa 1, 02-096, Warsaw, Poland
| | - Bartłomiej Grygorcewicz
- Department of Molecular Virology, Faculty of Biology, Institute of Microbiology, University of Warsaw, Miecznikowa 1, 02-096, Warsaw, Poland
| | - Joanna Karczewska-Golec
- Department of Molecular Virology, Faculty of Biology, Institute of Microbiology, University of Warsaw, Miecznikowa 1, 02-096, Warsaw, Poland
| | - Kamila Sadowska
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Ks. Trojdena 4, 02-109, Warsaw, Poland
| | - Piotr Golec
- Department of Molecular Virology, Faculty of Biology, Institute of Microbiology, University of Warsaw, Miecznikowa 1, 02-096, Warsaw, Poland.
| |
Collapse
|
40
|
Zheng T, Zhang C. Engineering strategies and challenges of endolysin as an antibacterial agent against Gram-negative bacteria. Microb Biotechnol 2024; 17:e14465. [PMID: 38593316 PMCID: PMC11003714 DOI: 10.1111/1751-7915.14465] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 03/09/2024] [Accepted: 03/21/2024] [Indexed: 04/11/2024] Open
Abstract
Bacteriophage endolysin is a novel antibacterial agent that has attracted much attention in the prevention and control of drug-resistant bacteria due to its unique mechanism of hydrolysing peptidoglycans. Although endolysin exhibits excellent bactericidal effects on Gram-positive bacteria, the presence of the outer membrane of Gram-negative bacteria makes it difficult to lyse them extracellularly, thus limiting their application field. To enhance the extracellular activity of endolysin and facilitate its crossing through the outer membrane of Gram-negative bacteria, researchers have adopted physical, chemical, and molecular methods. This review summarizes the characterization of endolysin targeting Gram-negative bacteria, strategies for endolysin modification, and the challenges and future of engineering endolysin against Gram-negative bacteria in clinical applications, to promote the application of endolysin in the prevention and control of Gram-negative bacteria.
Collapse
Affiliation(s)
- Tianyu Zheng
- Bathurst Future Agri‐Tech InstituteQingdao Agricultural UniversityQingdaoChina
| | - Can Zhang
- College of Veterinary MedicineQingdao Agricultural UniversityQingdaoChina
| |
Collapse
|
41
|
Yang D, Xiang Y, Song F, Li H, Ji X. Phage therapy: A renewed approach against oral diseases caused by Enterococcus faecalis infections. Microb Pathog 2024; 189:106574. [PMID: 38354990 DOI: 10.1016/j.micpath.2024.106574] [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: 09/27/2023] [Revised: 02/12/2024] [Accepted: 02/12/2024] [Indexed: 02/16/2024]
Abstract
Antibiotics play an important role in the treatment of infectious diseases. Long-term overuse or misuse of antibiotics, however, has triggered the global crisis of antibiotic resistance, bringing challenges to treating clinical infection. Bacteriophages (phages) are the viruses infecting bacterial cells. Due to high host specificity, high bactericidal activity, and good biosafety, phages have been used as natural alternative antibacterial agents to fight against multiple drug-resistant bacteria. Enterococcus faecalis is the main species detected in secondary persistent infection caused by failure of root canal therapy. Due to strong tolerance and the formation of biofilm, E. faecalis can survive the changes in pH, temperature, and osmotic pressure in the mouth and thus is one of the main causes of periapical lesions. This paper summarizes the advantages of phage therapy, its applications in treating oral diseases caused by E. faecalis infections, and the challenges it faces. It offers a new perspective on phage therapy in oral diseases.
Collapse
Affiliation(s)
- Dan Yang
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
| | - Yingying Xiang
- Department of Stomatology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, 650031, China
| | - Fei Song
- Department of Minimally Invasive Intervention, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, China
| | - Haiyan Li
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
| | - Xiuling Ji
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China.
| |
Collapse
|
42
|
Wang J, Zhang M, Pei J, Yi W, Fan L, Wang C, Xiao X. Isolation and identification of a novel phage targeting clinical multidrug-resistant Corynebacterium striatum isolates. Front Cell Infect Microbiol 2024; 14:1361045. [PMID: 38572320 PMCID: PMC10987712 DOI: 10.3389/fcimb.2024.1361045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 02/26/2024] [Indexed: 04/05/2024] Open
Abstract
Introduction Over the past decade, Corynebacterium striatum (C. striatum), an emerging multidrug-resistant (MDR) pathogen, has significantly challenged healthcare settings, especially those involving individuals with weakened immune systems. The rise of these superbugs necessitates innovative solutions. Methods This study aimed to isolate and characterize bacteriophages targeting MDR-C. striatum. Utilizing 54 MDR-C. striatum isolates from a local hospital as target strains, samples were collected from restroom puddles for phage screening. Dot Plaque and Double-layer plate Assays were employed for screening. Results A novel temperate bacteriophage, named CSP1, was identified through a series of procedures, including purification, genome extraction, sequencing, and one-step growth curves. CSP1 possesses a 39,752 base pair circular double-stranded DNA genome with HK97-like structural proteins and potential for site-specific recombination. It represents a new species within the unclassified Caudoviricetes class, as supported by transmission electron microscopy, genomic evolutionary analysis, and collinearity studies. Notably, CSP1 infected and lysed 21 clinical MDR-C. striatum isolates, demonstrating a wide host range. The phage remained stable in conditions ranging from -40 to 55°C, pH 4 to 12, and in 0.9% NaCl buffer, showing no cytotoxicity. Discussion The identification of CSP1 as the first phage targeting clinical C. striatum strains opens new possibilities in bacteriophage therapy research, and the development of diagnostic and therapeutic tools against pathogenic bacteria.
Collapse
Affiliation(s)
- Jiao Wang
- Department of Pathogen Biology, School of Basic Medicine, Hubei University of Arts and Science, Xiangyang, China
| | - Meng Zhang
- Department of Pathogen Biology, School of Basic Medicine, Hubei University of Arts and Science, Xiangyang, China
| | - Jiao Pei
- Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Wei Yi
- Department of Pathogen Biology, School of Basic Medicine, Hubei University of Arts and Science, Xiangyang, China
| | - Li Fan
- Department of Pathogen Biology, School of Basic Medicine, Hubei University of Arts and Science, Xiangyang, China
| | - Chunhua Wang
- Department of Clinical Laboratory, Xiangyang No.1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
| | - Xiao Xiao
- Department of Pathogen Biology, School of Basic Medicine, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
43
|
Wang M, Zhang J, Wei J, Jiang L, Jiang L, Sun Y, Zeng Z, Wang Z. Phage-inspired strategies to combat antibacterial resistance. Crit Rev Microbiol 2024; 50:196-211. [PMID: 38400715 DOI: 10.1080/1040841x.2023.2181056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 02/07/2023] [Indexed: 02/25/2023]
Abstract
Antimicrobial resistance (AMR) in clinically priority pathogensis now a major threat to public health worldwide. Phages are bacterial parasites that efficiently infect or kill specific strains and represent the most abundant biological entities on earth, showing great attraction as potential antibacterial therapeutics in combating AMR. This review provides a summary of phage-inspired strategies to combat AMR. We firstly cover the phage diversity, and then explain the biological principles of phage therapy that support the use of phages in the post-antimicrobial era. Furthermore, we state the versatility methods of phage therapy both from direct access as well as collateral access. Among the direct access approaches, we discuss the use of phage cocktail therapy, phage-encoded endolysins and the bioengineering for function improvement of used phages or endolysins. On the other hand, we introduce the collateral access, including the phages antimicrobial immunity combined therapy and phage-based novel antibacterial mimic molecules. Nowadays, more and more talented and enthusiastic scientist, doctors, pharmacists, media, authorities, and industry are promoting the progress of phage therapy, and proposed more phages-inspired strategy to make them more tractable to combat AMR and benefit more people, more animal and diverse environment in "one health" framework.
Collapse
Affiliation(s)
- Mianzhi Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Junxuan Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Jingyi Wei
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Lei Jiang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Li Jiang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Yongxue Sun
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Zhenling Zeng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Zhiqiang Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou, China
| |
Collapse
|
44
|
Erol HB, Kaskatepe B, Yildiz S, Altanlar N, Bayrakdar F. Characterization of two bacteriophages specific to Acinetobacter baumannii and their effects on catheters biofilm. Cell Biochem Funct 2024; 42:e3966. [PMID: 38444208 DOI: 10.1002/cbf.3966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/07/2024]
Abstract
Multidrug-resistant strains of Acinetobacter baumannii cause major nosocomial infections. Bacteriophages that are specific to the bacterial species and destroy bacteria can be effectively used for treatment. In this study, we characterized lytic bacteriophages specific to A. baumannii strains. We isolated lytic bacteriophages from environmental water samples and then investigated their morphology, host range, growth characteristics, stability, genome analysis, and biofilm destruction on the catheter surface. Our results showed that the efficacy of the phages varied between 32% and 78%, tested on 78 isolates of A. baumannii; 80 phages were isolated, and two lytic bacteriophages, vB_AbaP_HB01 (henceforth called C2 phage) and vB_AbaM_HB02 (henceforth called K3 phage), were selected for characterization. Electron microscopy scans revealed that the C2 and K3 phages were members of the Podoviridae and Myoviridae families, respectively. Whole-genome sequencing revealed that the sequence of the C2 phage is available in the NCBI database (accession number: OP917929.1), and it was found sequence identity with Acinetobacter phage AB1 18%, the K3 phage DNA sequence is closely related to Acinetobacter phage vB_AbaM_phiAbaA1 (94% similarity). The cocktail of C2 and K3 phages demonstrated a promising decrease in the bacterial cell counts of the biofilm after 4 h. Under a scanning electron microscope, the cocktail treatment destructed the biofilm on the catheter. We propose that the phage cocktail could be a strong alternative to antibiotics to control the A. baumannii biofilm in catheter infections.
Collapse
Affiliation(s)
- Hilal Basak Erol
- Department of Pharmaceutical Microbiology, Ankara University Faculty of Pharmacy, Ankara, Turkey
- Ankara University Graduate School of Health Science, Ankara, Turkey
| | - Banu Kaskatepe
- Department of Pharmaceutical Microbiology, Ankara University Faculty of Pharmacy, Ankara, Turkey
| | - Sulhiye Yildiz
- Department of Pharmaceutical Microbiology, Lokman Hekim University Faculty of Pharmacy, Ankara, Turkey
| | - Nurten Altanlar
- Department of Pharmaceutical Microbiology, Ankara University Faculty of Pharmacy, Ankara, Turkey
| | - Fatma Bayrakdar
- Ministry of Health, General Directorate of Public Health, Microbiology References Laboratory, Ankara, Turkey
| |
Collapse
|
45
|
Golosova NN, Matveev AL, Tikunova NV, Khlusevich YA, Kozlova YN, Morozova VV, Babkin IV, Ushakova TA, Zhirakovskaya EV, Panina EA, Ryabchikova EI, Tikunov AY. Bacteriophage vB_SepP_134 and Endolysin LysSte_134_1 as Potential Staphylococcus-Biofilm-Removing Biological Agents. Viruses 2024; 16:385. [PMID: 38543751 PMCID: PMC10975630 DOI: 10.3390/v16030385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/19/2024] [Accepted: 02/28/2024] [Indexed: 05/23/2024] Open
Abstract
Bacteria of the genus Staphylococcus are significant challenge for medicine, as many species are resistant to multiple antibiotics and some are even to all of the antibiotics we use. One of the approaches to developing new therapeutics to treat staphylococcal infections is the use of bacteriophages specific to these bacteria or the lytic enzymes of such bacteriophages, which are capable of hydrolyzing the cell walls of these bacteria. In this study, a new bacteriophage vB_SepP_134 (St 134) specific to Staphylococcus epidermidis was described. This podophage, with a genome of 18,275 bp, belongs to the Andhravirus genus. St 134 was able to infect various strains of 12 of the 21 tested coagulase-negative Staphylococcus species and one clinical strain from the Staphylococcus aureus complex. The genes encoding endolysin (LysSte134_1) and tail tip lysin (LysSte134_2) were identified in the St 134 genome. Both enzymes were cloned and produced in Escherichia coli cells. The endolysin LysSte134_1 demonstrated catalytic activity against peptidoglycans isolated from S. aureus, S. epidermidis, Staphylococcus haemolyticus, and Staphylococcus warneri. LysSte134_1 was active against S. aureus and S. epidermidis planktonic cells and destroyed the biofilms formed by clinical strains of S. aureus and S. epidermidis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Artem Y. Tikunov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (N.N.G.); (N.V.T.); (Y.A.K.); (Y.N.K.); (V.V.M.); (I.V.B.); (T.A.U.); (E.A.P.); (E.I.R.)
| |
Collapse
|
46
|
Grigore-Gurgu L, Bucur FI, Mihalache OA, Nicolau AI. Comprehensive Review on the Biocontrol of Listeria monocytogenes in Food Products. Foods 2024; 13:734. [PMID: 38472848 DOI: 10.3390/foods13050734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/21/2024] [Accepted: 02/25/2024] [Indexed: 03/14/2024] Open
Abstract
Listeria monocytogenes is a foodborne pathogen that causes listeriosis, a group of human illnesses that appear more frequently in countries with better-developed food supply systems. This review discusses the efficacy of actual biocontrol methods combined with the main types of food involved in illnesses. Comments on bacteriophages, lactic acid bacteria, bacteriocins, essential oils, and endolysins and derivatives, as main biological antilisterial agents, are made bearing in mind that, using them, food processors can intervene to protect consumers. Both commercially available antilisterial products and solutions presented in scientific papers for mitigating the risk of contamination are emphasized. Potential combinations between different types of antilisterial agents are highlighted for their synergic effects (bacteriocins and essential oils, phages and bacteriocins, lactic acid bacteria with natural or synthetic preservatives, etc.). The possibility to use various antilisterial biological agents in active packaging is also presented to reveal the diversity of means that food processors may adopt to assure the safety of their products. Integrating biocontrol solutions into food processing practices can proactively prevent outbreaks and reduce the occurrences of L. monocytogenes-related illnesses.
Collapse
Affiliation(s)
- Leontina Grigore-Gurgu
- Faculty of Food Science and Engineering, Dunarea de Jos University of Galati, 47 Domneasca Street, 800008 Galati, Romania
| | - Florentina Ionela Bucur
- Faculty of Food Science and Engineering, Dunarea de Jos University of Galati, 47 Domneasca Street, 800008 Galati, Romania
| | - Octavian Augustin Mihalache
- Faculty of Food Science and Engineering, Dunarea de Jos University of Galati, 47 Domneasca Street, 800008 Galati, Romania
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 17/A, 43124 Parma, Italy
| | - Anca Ioana Nicolau
- Faculty of Food Science and Engineering, Dunarea de Jos University of Galati, 47 Domneasca Street, 800008 Galati, Romania
| |
Collapse
|
47
|
Eiselt VA, Bereswill S, Heimesaat MM. Phage therapy in lung infections caused by multidrug-resistant Pseudomonas aeruginosa - A literature review. Eur J Microbiol Immunol (Bp) 2024; 14:1-12. [PMID: 38261031 PMCID: PMC10895363 DOI: 10.1556/1886.2023.00060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
Pulmonary infections of patients with cystic fibrosis (CF) or in intensive care units are frequently caused by the Gram-negative opportunistic pathogen Pseudomonas aeruginosa. Since these bacteria are commonly inherently multidrug-resistant (MDR) and hence, antibiotic treatment options are limited, bacteriophages may provide alternative therapeutic and prophylactic measures in the combat of pneumonia caused by P. aeruginosa. This prompted us to perform a comprehensive literature survey of current knowledge regarding effects of phages applied against pulmonary P. aeruginosa infections. The included 23 studies revealed that P. aeruginosa specific phages lyse and eliminate the bacteria even in case of biofilm production in vitro, whereas application to mice and men resulted in mitigated P. aeruginosa induced clinical signs and enhanced survival. Besides distinct host immune responses, no major adverse effects limiting therapeutic and/or prophylactic phage application were noted. However, the immune system and antibiotics generate synergies with phages due to the mutable sensitivity of P. aeruginosa. In conclusion, results summarized in this review provide evidence that phages constitute promising alternative treatment options for lung infections caused by MDR P. aeruginosa. Further studies are needed, however, to underscore the efficacy and safety aspects of phages application to infected patients including immune-compromised individuals.
Collapse
Affiliation(s)
- Vincent A Eiselt
- Gastrointestinal Microbiology Research Group, Institute of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Stefan Bereswill
- Gastrointestinal Microbiology Research Group, Institute of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Markus M Heimesaat
- Gastrointestinal Microbiology Research Group, Institute of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
48
|
Nazir A, Li L, Li F, Tong Y, Liu Y, Chen Y. Characterization, taxonomic classification, and genomic analysis of two newly isolated bacteriophages with potential to infect Escherichia coli. Microbiol Spectr 2024:e0223023. [PMID: 38376266 DOI: 10.1128/spectrum.02230-23] [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: 05/26/2023] [Accepted: 01/29/2024] [Indexed: 02/21/2024] Open
Abstract
Escherichia coli is a pathogenic bacterium that is widely distributed and can lead to serious illnesses in both humans and animals. As there is rising incidence of multidrug resistance among these bacteria, it has become imperative to discover alternative therapies beyond antibiotics to effectively treat such infections. Bacteriophage (phage) therapy has the potential to treat infections caused by E. coli, as phages contain enzymes that can cause lysis or destruction of bacterial cells. Simultaneously, the easy accessibility and cost-effectiveness of next-generation sequencing technologies have led to the accumulation of a vast amount of phage sequence data. Here, phages IME177 and IME267 were isolated from sewage water of a hospital in China. Modern phylogenetic approaches and key findings from the genomic analysis revealed that phages IME177 and IME267 are classified as members of the Kayfunavirus genus, Autographiviridae family, and a newly proposed Suseptimavirus genus under subfamily Gordonclarkvirinae, respectively. Further, the Kuravirus genus reshaped into three different genera: Kuravirus, Nieuwekanaalvirus, and Suspeptimavirus, which are classified together under a higher taxonomic rank (subfamily) named Gordonclarkvirinae. No genes related to virulence were detected in the genomes of the phages IME177 and IME267. Both phages exhibited a high degree of resilience to a wide range of conditions, including pH, temperature, exposure to chloroform, and UV radiation. Phages IME177 and IME267 are promising biological agents that can infect E. coli, making them suitable candidates for use in phage therapies.IMPORTANCEBiological and taxonomic characterization of phages is essential for facilitating the development of effective strategies for phage therapy and disease control. Escherichia coli phages are incredibly diverse, and their isolation and classification help us understand the scope and nature of this diversity. By identifying new phages and grouping them into families, we can better understand the genetic and structural variations between phages and how they affect their infectivity and interactions with bacteria. Overall, the isolation and classification of E. coli phages have broad implications for both basic and applied research, clinical practice, and public health.
Collapse
Affiliation(s)
- Amina Nazir
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, Shandong, China
- Key Laboratory of Livestock and Poultry Multi-omics of MARA, China-UK Joint Laboratory of Bacteriophage Engineering, Jinan, China
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Lulu Li
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, Shandong, China
- Key Laboratory of Livestock and Poultry Multi-omics of MARA, China-UK Joint Laboratory of Bacteriophage Engineering, Jinan, China
| | - Fei Li
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Yigang Tong
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Yuqing Liu
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, Shandong, China
- Key Laboratory of Livestock and Poultry Multi-omics of MARA, China-UK Joint Laboratory of Bacteriophage Engineering, Jinan, China
| | - Yibao Chen
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, Shandong, China
- Key Laboratory of Livestock and Poultry Multi-omics of MARA, China-UK Joint Laboratory of Bacteriophage Engineering, Jinan, China
| |
Collapse
|
49
|
Blanch-Asensio M, Tadimarri VS, Wilk A, Sankaran S. Discovery of a high-performance phage-derived promoter/repressor system for probiotic lactobacillus engineering. Microb Cell Fact 2024; 23:42. [PMID: 38326819 PMCID: PMC10848424 DOI: 10.1186/s12934-024-02302-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 01/10/2024] [Indexed: 02/09/2024] Open
Abstract
BACKGROUND The Lactobacillaceae family comprises many species of great importance for the food and healthcare industries, with numerous strains identified as beneficial for humans and used as probiotics. Hence, there is a growing interest in engineering these probiotic bacteria as live biotherapeutics for animals and humans. However, the genetic parts needed to regulate gene expression in these bacteria remain limited compared to model bacteria like E. coli or B. subtilis. To address this deficit, in this study, we selected and tested several bacteriophage-derived genetic parts with the potential to regulate transcription in lactobacilli. RESULTS We screened genetic parts from 6 different lactobacilli-infecting phages and identified one promoter/repressor system with unprecedented functionality in Lactiplantibacillus plantarum WCFS1. The phage-derived promoter was found to achieve expression levels nearly 9-fold higher than the previously reported strongest promoter in this strain and the repressor was able to almost completely repress this expression by reducing it nearly 500-fold. CONCLUSIONS The new parts and insights gained from their engineering will enhance the genetic programmability of lactobacilli for healthcare and industrial applications.
Collapse
Affiliation(s)
- Marc Blanch-Asensio
- Bioprogrammable Materials, INM - Leibniz Institute for New Materials, Campus D2 2, 66123, Saarbrücken, Germany
- Saarland University, 66123, Saarbrücken, Germany
| | - Varun Sai Tadimarri
- Bioprogrammable Materials, INM - Leibniz Institute for New Materials, Campus D2 2, 66123, Saarbrücken, Germany
- Saarland University, 66123, Saarbrücken, Germany
| | - Alina Wilk
- Bioprogrammable Materials, INM - Leibniz Institute for New Materials, Campus D2 2, 66123, Saarbrücken, Germany
- Saarland University, 66123, Saarbrücken, Germany
| | - Shrikrishnan Sankaran
- Bioprogrammable Materials, INM - Leibniz Institute for New Materials, Campus D2 2, 66123, Saarbrücken, Germany.
| |
Collapse
|
50
|
Szymanski CM. Bacteriophages and their unique components provide limitless resources for exploitation. Front Microbiol 2024; 15:1342544. [PMID: 38380101 PMCID: PMC10877033 DOI: 10.3389/fmicb.2024.1342544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 01/22/2024] [Indexed: 02/22/2024] Open
Affiliation(s)
- Christine M. Szymanski
- Department of Microbiology, Complex Carbohydrate Research Center, University of Georgia, Athens, GA, United States
| |
Collapse
|