1
|
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
|
2
|
Cremelie E, Vázquez R, Briers Y. A comparative guide to expression systems for phage lysin production. Essays Biochem 2024; 68:645-659. [PMID: 39290148 DOI: 10.1042/ebc20240019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 09/19/2024]
Abstract
Phage lysins, bacteriophage-encoded enzymes tasked with degrading their host's cell wall, are increasingly investigated and engineered as novel antibacterials across diverse applications. Their rapid action, tuneable specificity, and low likelihood of resistance development make them particularly interesting. Despite numerous application-focused lysin studies, the art of their recombinant production remains relatively undiscussed. Here, we provide an overview of the available expression systems for phage lysin production and discuss key considerations guiding the choice of a suitable recombinant host. We systematically surveyed recent literature to evaluate the hosts used in the lysin field and cover various recombinant systems, including the well-known bacterial host Escherichia coli or yeast Saccharomyces cerevisiae, as well as plant, mammalian, and cell-free systems. Careful analysis of the limited studies expressing lysins in various hosts suggests a host-dependent effect on activity. Nonetheless, the multitude of available expression systems should be further leveraged to accommodate the growing interest in phage lysins and their expanding range of applications.
Collapse
Affiliation(s)
- Emma Cremelie
- Laboratory of Applied Biotechnology, Department of Biotechnology, Ghent University, Ghent, Belgium
| | - Roberto Vázquez
- Laboratory of Applied Biotechnology, Department of Biotechnology, Ghent University, Ghent, Belgium
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Yves Briers
- Laboratory of Applied Biotechnology, Department of Biotechnology, Ghent University, Ghent, Belgium
| |
Collapse
|
3
|
Eghbalpoor F, Gorji M, Alavigeh MZ, Moghadam MT. Genetically engineered phages and engineered phage-derived enzymes to destroy biofilms of antibiotics resistance bacteria. Heliyon 2024; 10:e35666. [PMID: 39170521 PMCID: PMC11336853 DOI: 10.1016/j.heliyon.2024.e35666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/27/2024] [Accepted: 08/01/2024] [Indexed: 08/23/2024] Open
Abstract
"An impregnable stronghold where one or more warrior clans can evade enemy attacks" may serve as a description of bacterial biofilm on a smaller level than human conflicts. Consider this hypothetical conflict: who would emerge victorious? The occupants of secure trenches or those carrying out relentless assault? Either faction has the potential for triumph; the defenders will prevail if they can fortify the trench with unwavering resolve, while the assailants will succeed if they can devise innovative means to breach the trench. Hence, bacterial biofilms pose a significant challenge and are formidable adversaries for medical professionals, often leading to the failure of antibiotic treatments in numerous hospital infections. Phage engineering has become the foundation for the targeted enhancement of various phage properties, facilitating the eradication of biofilms. Researchers across the globe have studied the impact of engineered phages and phage-derived enzymes on biofilms formed by difficult-to-treat bacteria. These novel biological agents have shown promising results in addressing biofilm-related challenges. The compilation of research findings highlights the impressive capabilities of engineered phages in combating antibiotic-resistant bacteria, superbugs, and challenging infections. Specifically, these engineered phages exhibit enhanced biofilm destruction, penetration, and prevention capabilities compared to their natural counterparts. Additionally, the engineered enzymes derived from phages demonstrate improved effectiveness in addressing bacterial biofilms. As a result, these novel solutions, which demonstrate high penetration, destruction, and inhibition of biofilms, can be regarded as a viable option for addressing infectious biofilms in the near future.
Collapse
Affiliation(s)
- Fatemeh Eghbalpoor
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran
| | - Mahdieh Gorji
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Maryam Zamani Alavigeh
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
4
|
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
|
5
|
Cho Y, Park K, Park J, An J, Myung H, Yoon H. Exploring the therapeutic potential of endolysin CD27L_EAD against Clostridioides difficile infection. Int J Antimicrob Agents 2024; 64:107222. [PMID: 38810936 DOI: 10.1016/j.ijantimicag.2024.107222] [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: 09/26/2023] [Revised: 04/06/2024] [Accepted: 05/20/2024] [Indexed: 05/31/2024]
Abstract
OBJECTIVES Clostridioides difficile has emerged as a major cause of life-threatening diarrheal disease. Conventional antibiotics used in current standards of care exacerbate the emergence of antibiotic-resistant strains and pose a risk of recurrent C. difficile infection (CDI). Thus, there is an urgent need for alternative therapeutics that selectively eliminate C. difficile without disturbing the commensal microbiota. This study aimed to explore the potential of endolysins as an alternative therapeutic agent to antibiotics. Endolysin is a bacteriophage-derived peptidoglycan hydrolase that aids in the release of phage progeny during the final stage of infection. METHODS In order to exploit endolysin as a therapeutic agent against CDI, the bactericidal activity of 23 putative endolysins was compared and ΦCD27 endolysin CD27L was selected and modified to CD27L_EAD by cleaving the cell-wall binding domain of CD27L. RESULTS CD27L_EAD exhibited greater bacteriolytic activity than CD27L and its activity was stable over a wide range of salt concentrations and pH conditions. CD27L_EAD was added to a co-culture of human gut microbiota with C. difficile and the bacterial community structure was analyzed. CD27L_EAD did not impair the richness and diversity of the bacterial population but remarkably attenuated the abundance of C. difficile. Furthermore, the co-administration of vancomycin exerted synergistic bactericidal activity against C. difficile. β-diversity analysis revealed that CD27L_EAD did not significantly disturb the composition of the microbial community, whereas the abundance of some species belonging to the family Lachnospiraceae decreased after CD27L_EAD treatment. CONCLUSIONS This study provides insights into endolysin as a prospective therapeutic agent for the treatment of CDI without damaging the normal gut microbiota.
Collapse
Affiliation(s)
- Youngjin Cho
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Kyungah Park
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Jeongseok Park
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Jieun An
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Heejoon Myung
- LyseNTech Co., Ltd., Seongnam, South Korea; Department of Bioscience and Biotechnology, Hankuk University of Foreign Studies, Yongin, South Korea
| | - Hyunjin Yoon
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea; Department of Applied Chemistry and Biological Engineering, Ajou University, Suwon, South Korea.
| |
Collapse
|
6
|
Hassannia M, Naderifar M, Salamy S, Akbarizadeh MR, Mohebi S, Moghadam MT. Engineered phage enzymes against drug-resistant pathogens: a review on advances and applications. Bioprocess Biosyst Eng 2024; 47:301-312. [PMID: 37962644 DOI: 10.1007/s00449-023-02938-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 10/16/2023] [Indexed: 11/15/2023]
Abstract
In recent decades, the expansion of multi and extensively drug-resistant (MDR and XDR) bacteria has reached an alarming rate, causing serious health concerns. Infections caused by drug-resistant bacteria have been associated with morbidity and mortality, making tackling bacterial resistance an urgent and unmet challenge that needs to be addressed properly. Endolysins are phage-encoded enzymes that can specifically degrade the bacterial cell wall and lead to bacterial death. There is remarkable evidence that corroborates the unique ability of endolysins to rapidly digest the peptidoglycan particular bonds externally without the assistance of phage. Thus, their modulation in therapeutic approaches has opened new options for therapeutic applications in the fight against bacterial infections in the human and veterinary sectors, as well as within the agricultural and biotechnology areas. The use of genetically engineered phage enzymes (EPE) promises to generate endolysin variants with unique properties for prophylactic and therapeutic applications. These approaches have gained momentum to accelerate basic as well as translational phage research and the potential development of therapeutics in the near future. This review will focus on the novel knowledge into EPE and demonstrate that EPE has far better performance than natural endolysins and phages in dealing with antibiotic-resistant infections. Therefore, it provides essential information for clinical trials involving EPE.
Collapse
Affiliation(s)
- Mohadeseh Hassannia
- Department of Genetic, Faculty of Science, Islamic Azad University, Tehran, Iran
| | - Mahin Naderifar
- School of Medicine, Zabol University of Medical Sciences, Zabol, Iran
| | - Shakiba Salamy
- Department of Microbiology, Faculty of Pharmacy, Islamic Azad University, Tehran, Iran
| | | | - Samane Mohebi
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | |
Collapse
|
7
|
Sánchez C, Garde S, Landete JM, Calzada J, Baker DJ, Evans R, Narbad A, Mayer MJ, Ávila M. Identification, activity and delivery of new LysFA67 endolysin to target cheese spoilage Clostridium tyrobutyricum. Food Microbiol 2024; 117:104401. [PMID: 37919009 DOI: 10.1016/j.fm.2023.104401] [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: 06/20/2023] [Revised: 10/03/2023] [Accepted: 10/07/2023] [Indexed: 11/04/2023]
Abstract
Bacteriophages and their endolysins are potential biocontrol agents for the anaerobic spoilage organism Clostridium tyrobutyricum, which causes cheese late blowing defect. This study sequenced and compared the genomes of eight bacteriophages from Spanish dairy farms that were active against C. tyrobutyricum, to identify novel species and phage proteins. Phages vB_CtyS-FA67 and vB_CtyS-FA70 shared >94% intergenomic similarity to each other but neither phage had significant similarity to ΦCTP1, the unique C. tyrobutyricum phage sequenced to date. Taxonomic analysis indicated that both phages belong to the class Caudoviricetes and are related to dsDNA viruses with long non-contractile tails. vB_CtyS-FA67 had no other close relatives and encoded a novel endolysin, LysFA67, predicted to belong to the glycoside hydrolase GH24 family. LysFA67 lysed 93% of C. tyrobutyricum cells after 4 min in turbidity reduction assays, retaining lytic activity at pHs 4.2-8.1 and at 30-45 °C. The endolysin remained stable after 30 d storage at 4, 12 and 25 °C, while its activity decreased at -20 °C. LysFA67 lysed several clostridia species, while common dairy bacteria were not affected. Lactococcus lactis INIA 437, used as a cheese starter, was engineered to deliver LysFA67 and red fluorescent LysFA67-mCherry to dairy products. We demonstrated that these engineered strains were able to maintain lytic activity and fluorescence without affecting their technological properties in milk.
Collapse
Affiliation(s)
- Carmen Sánchez
- Departamento de Tecnología de Alimentos, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Carretera de La Coruña km 7, 28040, Madrid, Spain
| | - Sonia Garde
- Departamento de Tecnología de Alimentos, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Carretera de La Coruña km 7, 28040, Madrid, Spain.
| | - José María Landete
- Departamento de Tecnología de Alimentos, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Carretera de La Coruña km 7, 28040, Madrid, Spain
| | - Javier Calzada
- Departamento de Tecnología de Alimentos, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Carretera de La Coruña km 7, 28040, Madrid, Spain
| | - Dave J Baker
- Science Operations, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
| | - Rhiannon Evans
- Science Operations, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
| | - Arjan Narbad
- Food, Microbiome and Health Institute Strategic Programme, Quadram Institute Bioscience, Colney, Norwich, NR4 7UA, UK
| | - Melinda J Mayer
- Food, Microbiome and Health Institute Strategic Programme, Quadram Institute Bioscience, Colney, Norwich, NR4 7UA, UK.
| | - Marta Ávila
- Departamento de Tecnología de Alimentos, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Carretera de La Coruña km 7, 28040, Madrid, Spain.
| |
Collapse
|
8
|
Pottie I, Vázquez Fernández R, Van de Wiele T, Briers Y. Phage lysins for intestinal microbiome modulation: current challenges and enabling techniques. Gut Microbes 2024; 16:2387144. [PMID: 39106212 PMCID: PMC11305034 DOI: 10.1080/19490976.2024.2387144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/05/2024] [Accepted: 07/26/2024] [Indexed: 08/09/2024] Open
Abstract
The importance of the microbiota in the intestinal tract for human health has been increasingly recognized. In this perspective, microbiome modulation, a targeted alteration of the microbial composition, has gained interest. Phage lysins, peptidoglycan-degrading enzymes encoded by bacteriophages, are a promising new class of antibiotics currently under clinical development for treating bacterial infections. Due to their high specificity, lysins are considered microbiome-friendly. This review explores the opportunities and challenges of using lysins as microbiome modulators. First, the high specificity of endolysins, which can be further modulated using protein engineering or targeted delivery methods, is discussed. Next, obstacles and possible solutions to assess the microbiome-friendliness of lysins are considered. Finally, lysin delivery to the intestinal tract is discussed, including possible delivery methods such as particle-based and probiotic vehicles. Mapping the hurdles to developing lysins as microbiome modulators and identifying possible ways to overcome these hurdles can help in their development. In this way, the application of these innovative antimicrobial agents can be expanded, thereby taking full advantage of their characteristics.
Collapse
Affiliation(s)
- Iris Pottie
- Laboratory of Applied Biotechnology, Department of Biotechnology, Ghent University, Gent, Belgium
- Center for Microbial Ecology and Technology (CMET), Faculty of Bioscience Engineering, Ghent University, Gent, Belgium
| | - Roberto Vázquez Fernández
- Laboratory of Applied Biotechnology, Department of Biotechnology, Ghent University, Gent, Belgium
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Tom Van de Wiele
- Center for Microbial Ecology and Technology (CMET), Faculty of Bioscience Engineering, Ghent University, Gent, Belgium
| | - Yves Briers
- Laboratory of Applied Biotechnology, Department of Biotechnology, Ghent University, Gent, Belgium
| |
Collapse
|
9
|
Zhao X, Li L, Zhang Q, Li M, Hu M, Luo Y, Xu X, Chen Y, Liu Y. Characterization of the Clostridium perfringens phage endolysin cpp-lys and its application on lettuce. Int J Food Microbiol 2023; 405:110343. [PMID: 37523902 DOI: 10.1016/j.ijfoodmicro.2023.110343] [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: 05/04/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/02/2023]
Abstract
Clostridium perfringens is an important foodborne pathogen that can have severe consequences, including mortality and economic losses. In this study, the gene encoding cpp-lys, an endolysin from the C. perfringens phage cpp has been cloned and overexpressed. The encoded protein was characterized, and then its efficacy in controlling C. perfringens on lettuce was evaluated. The endolysin cpp-lys presented lytic activity against seven strains of C. perfringens that produce different types of toxins. It maintained stability across a wide range of temperatures (4 °C - 50 °C), and demonstrated tolerance to varying pH levels (4-9). Storage of endolysin cpp-lys under room-temperature conditions (16 °C-25 °C) and cold-temperature conditions (4 °C, -20 °C, and -80 °C) for 30 days did not affect its lytic activity. However, the lytic activity of cpp-lys decreased by 40 % and 18 % after storage for 30 d at 42 °C and 37 °C, respectively. The endolysin cpp-lys did not display cytotoxic activity against normal eukaryotic cells. The bacterial viability on lettuce was significantly lower in the group treated with endolysin cpp-lys than in the PBS group, and >4-log of C. perfringens J1 were removed within 15 min. Cpp-lys plus Zn2+ inhibited the activity of cpp-lys. The EDTA-treated cpp-lys significantly reduced the number of bacteria by up to 0.6-log CFU compared with the endolysin cpp-lys group. The findings of this study demonstrated that endolysin cpp-lys has potential applications in controlling C. perfringens in the food industry.
Collapse
Affiliation(s)
- Xiaonan Zhao
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, PR China; Key Laboratory of Livestock and Poultry Multi-omics of MARA, PR China
| | - Lulu Li
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, PR China; Key Laboratory of Livestock and Poultry Multi-omics of MARA, PR China
| | - Qing Zhang
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, PR China; Key Laboratory of Livestock and Poultry Multi-omics of MARA, PR China
| | - Mengxuan Li
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, PR China; Key Laboratory of Livestock and Poultry Multi-omics of MARA, PR China
| | - Ming Hu
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, PR China; Key Laboratory of Livestock and Poultry Multi-omics of MARA, PR China
| | - Yanbo Luo
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, PR China; Key Laboratory of Livestock and Poultry Multi-omics of MARA, PR China
| | - Xiaohui Xu
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, PR China; Key Laboratory of Livestock and Poultry Multi-omics of MARA, PR China
| | - Yibao Chen
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, PR China; Key Laboratory of Livestock and Poultry Multi-omics of MARA, PR China.
| | - Yuqing Liu
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, PR China; Key Laboratory of Livestock and Poultry Multi-omics of MARA, PR China.
| |
Collapse
|
10
|
Takala TM, Mokhtari S, Ahonen SL, Wan X, Saris PEJ. Wild-type Lactococcus lactis producing bacteriocin-like prophage lysins. Front Microbiol 2023; 14:1219723. [PMID: 37520360 PMCID: PMC10377672 DOI: 10.3389/fmicb.2023.1219723] [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: 05/09/2023] [Accepted: 06/26/2023] [Indexed: 08/01/2023] Open
Abstract
Introduction Lactococcus is a genus of lactic acid bacteria used in the dairy industry as a starter. Lactococci have been found to produce altogether more than 40 different bacteriocins, ribosomally synthesized antimicrobial proteins. All known Lactococcus spp. bacteriocins belong to classes I and II, which are mainly heat-resistant peptides. No class III bacteriocins, bigger heat-sensitive proteins, including phage tail-like bacteriocins, have been found from the Lactococcus spp. Unlike phage tail-like bacteriocins, prophage lysins have not been regarded as bacteriocins, possibly because phage lysins contribute to autolysis, degrading the host's own cell wall. Methods Wild-type Lactococcus lactis strain LAC460, isolated from spontaneously fermented idli batter, was examined for its antimicrobial activity. We sequenced the genome, searched phage lysins from the culture supernatant, and created knock-out mutants to find out the source of the antimicrobial activity. Results and discussion The strain LAC460 was shown to kill other Lactococcus strains with protease- and heat-sensitive lytic activity. Three phage lysins were identified in the culture supernatant. The genes encoding the three lysins were localized in different prophage regions in the chromosome. By knock-out mutants, two of the lysins, namely LysL and LysP, were demonstrated to be responsible for the antimicrobial activity. The strain LAC460 was found to be resistant to the lytic action of its own culture supernatant, and as a consequence, the phage lysins could behave like bacteriocins targeting and killing other closely related bacteria. Hence, similar to phage tail-like bacteriocins, phage lysin-like bacteriocins could be regarded as a novel type of class III bacteriocins.
Collapse
Affiliation(s)
- Timo M. Takala
- Department of Microbiology, Faculty of Agriculture and Forestry, University of Helsinki, Helsinki, Finland
| | - Samira Mokhtari
- Department of Microbiology, Faculty of Agriculture and Forestry, University of Helsinki, Helsinki, Finland
| | - Susanna L. Ahonen
- Department of Microbiology, Faculty of Agriculture and Forestry, University of Helsinki, Helsinki, Finland
- Expert Microbiology Unit, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Xing Wan
- Department of Microbiology, Faculty of Agriculture and Forestry, University of Helsinki, Helsinki, Finland
| | - Per E. J. Saris
- Department of Microbiology, Faculty of Agriculture and Forestry, University of Helsinki, Helsinki, Finland
| |
Collapse
|
11
|
Bacteriophages as Biocontrol Agents in Livestock Food Production. Microorganisms 2022; 10:microorganisms10112126. [PMID: 36363718 PMCID: PMC9692620 DOI: 10.3390/microorganisms10112126] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/19/2022] [Accepted: 10/25/2022] [Indexed: 12/03/2022] Open
Abstract
Bacteriophages have been regarded as biocontrol agents that can be used in the food industry. They can be used in various applications, such as pathogen detection and bio-preservation. Their potential to improve the quality of food and prevent foodborne illness is widespread. These bacterial viruses can also be utilized in the preservation of various other food products. The specificity and high sensitivity of bacteriophages when they lyse bacterial targets have been regarded as important factors that contribute to their great potential utility in the food industry. This review will provide an overview of their current and potential applications.
Collapse
|
12
|
Lee W, Matthews A, Moore D. Safety Evaluation of a Novel Algal Feed Additive for Poultry Production. Avian Dis 2022; 66:1-11. [PMID: 36214407 DOI: 10.1637/aviandiseases-d-22-00043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/11/2022] [Indexed: 12/14/2022]
Abstract
Feed additives are critical components for poultry health and the economic viability of antibiotic-free poultry production. The aim of the present study is to evaluate the safety of a novel algal-derived feed additive, a dried biomass powder produced from Chlamydomonas reinhardtii strain crAL082, modified to express an N-acetylmuramoyl-L-alanine amidase (EC 3.5.1.28) and a lysozyme-type enzyme (EC 3.2.1.17). A 42-day oral toxicity study showed that the crAL082 dried biomass powder was fully tolerated by broiler chicken based on the lack of detrimental effects found in performance, mortality, hematology, blood clinical chemistry, and histopathologic results compared with those of a nontreated control group, resulting in a "No Observed Adverse Effect Level" of 5000 ppm, the highest dose tested. The study demonstrates the first-ever safety result of a C. reinhardtii microalgae dried biomass powder used as a feed additive in broiler chickens. Furthermore, safety is shown for the two additional enzymes expressed within the C. reinhardtii crAL082 strain and ingested by the birds.
Collapse
Affiliation(s)
- Weiluo Lee
- Axitan Ltd., Ground Floor Offices, Whittle Way, SG1 2FS, Stevenage, United Kingdom,
| | | | - Daniel Moore
- Colorado Quality Research, Inc., Wellington, CO 80549
| |
Collapse
|
13
|
Venhorst J, van der Vossen JMBM, Agamennone V. Battling Enteropathogenic Clostridia: Phage Therapy for Clostridioides difficile and Clostridium perfringens. Front Microbiol 2022; 13:891790. [PMID: 35770172 PMCID: PMC9234517 DOI: 10.3389/fmicb.2022.891790] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 04/19/2022] [Indexed: 12/17/2022] Open
Abstract
The pathogenic Clostridioides difficile and Clostridium perfringens are responsible for many health care-associated infections as well as systemic and enteric diseases. Therefore, they represent a major health threat to both humans and animals. Concerns regarding increasing antibiotic resistance (related to C. difficile and C. perfringens) have caused a surge in the pursual of novel strategies that effectively combat pathogenic infections, including those caused by both pathogenic species. The ban on antibiotic growth promoters in the poultry industry has added to the urgency of finding novel antimicrobial therapeutics for C. perfringens. These efforts have resulted in various therapeutics, of which bacteriophages (in short, phages) show much promise, as evidenced by the Eliava Phage Therapy Center in Tbilisi, Georgia (https://eptc.ge/). Bacteriophages are a type of virus that infect bacteria. In this review, the (clinical) impact of clostridium infections in intestinal diseases is recapitulated, followed by an analysis of the current knowledge and applicability of bacteriophages and phage-derived endolysins in this disease indication. Limitations of phage and phage endolysin therapy were identified and require considerations. These include phage stability in the gastrointestinal tract, influence on gut microbiota structure/function, phage resistance development, limited host range for specific pathogenic strains, phage involvement in horizontal gene transfer, and-for phage endolysins-endolysin resistance, -safety, and -immunogenicity. Methods to optimize features of these therapeutic modalities, such as mutagenesis and fusion proteins, are also addressed. The future success of phage and endolysin therapies require reliable clinical trial data for phage(-derived) products. Meanwhile, additional research efforts are essential to expand the potential of exploiting phages and their endolysins for mitigating the severe diseases caused by C. difficile and C. perfringens.
Collapse
Affiliation(s)
- Jennifer Venhorst
- Biomedical Health, Netherlands Organisation for Applied Scientific Research (TNO), Utrecht, Netherlands
| | - Jos M. B. M. van der Vossen
- Microbiology and Systems Biology, Netherlands Organisation for Applied Scientific Research (TNO), Zeist, Netherlands
| | - Valeria Agamennone
- Microbiology and Systems Biology, Netherlands Organisation for Applied Scientific Research (TNO), Zeist, Netherlands
| |
Collapse
|
14
|
Cruz KCP, Enekegho LO, Stuart DT. Bioengineered Probiotics: Synthetic Biology Can Provide Live Cell Therapeutics for the Treatment of Foodborne Diseases. Front Bioeng Biotechnol 2022; 10:890479. [PMID: 35656199 PMCID: PMC9152101 DOI: 10.3389/fbioe.2022.890479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 04/29/2022] [Indexed: 11/15/2022] Open
Abstract
The rising prevalence of antibiotic resistant microbial pathogens presents an ominous health and economic challenge to modern society. The discovery and large-scale development of antibiotic drugs in previous decades was transformational, providing cheap, effective treatment for what would previously have been a lethal infection. As microbial strains resistant to many or even all antibiotic drug treatments have evolved, there is an urgent need for new drugs or antimicrobial treatments to control these pathogens. The ability to sequence and mine the genomes of an increasing number of microbial strains from previously unexplored environments has the potential to identify new natural product antibiotic biosynthesis pathways. This coupled with the power of synthetic biology to generate new production chassis, biosensors and “weaponized” live cell therapeutics may provide new means to combat the rapidly evolving threat of drug resistant microbial pathogens. This review focuses on the application of synthetic biology to construct probiotic strains that have been endowed with functionalities allowing them to identify, compete with and in some cases kill microbial pathogens as well as stimulate host immunity. Weaponized probiotics may have the greatest potential for use against pathogens that infect the gastrointestinal tract: Vibrio cholerae, Staphylococcus aureus, Clostridium perfringens and Clostridioides difficile. The potential benefits of engineered probiotics are highlighted along with the challenges that must still be met before these intriguing and exciting new therapeutic tools can be widely deployed.
Collapse
|
15
|
Lee C, Kim H, Ryu S. Bacteriophage and endolysin engineering for biocontrol of food pathogens/pathogens in the food: recent advances and future trends. Crit Rev Food Sci Nutr 2022; 63:8919-8938. [PMID: 35400249 DOI: 10.1080/10408398.2022.2059442] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Despite advances in modern technologies, various foodborne outbreaks have continuously threatened the food safety. The overuse of and abuse/misuse of antibiotics have escalated this threat due to the prevalence of multidrug-resistant (MDR) pathogens. Therefore, the development of new methodologies for controlling microbial contamination is extremely important to ensure the food safety. As an alternative to antibiotics, bacteriophages(phages) and derived endolysins have been proposed as novel, effective, and safe antimicrobial agents and applied for the prevention and/or eradication of bacterial contaminants even in foods and food processing facilities. In this review, we describe recent genetic and protein engineering tools for phages and endolysins. The major aim of engineering is to overcome limitations such as a narrow host range, low antimicrobial activity, and low stability of phages and endolysins. Phage engineering also aims to deter the emergence of phage resistance. In the case of endolysin engineering, enhanced antibacterial ability against Gram-negative and Gram-positive bacteria is another important goal. Here, we summarize the successful studies of phages and endolysins treatment in different types of food. Moreover, this review highlights the recent advances in engineering techniques for phages and endolysins, discusses existing challenges, and suggests technical opportunities for further development, especially in terms of antimicrobial agents in the food industry.
Collapse
Affiliation(s)
- Chanyoung Lee
- Department of Food and Animal Biotechnology, College of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
- Center for Food and Bioconvergence, Seoul National University, Seoul, Republic of Korea
| | - Hyeongsoon Kim
- Department of Food and Animal Biotechnology, College of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Sangryeol Ryu
- Department of Food and Animal Biotechnology, College of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
- Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
- Center for Food and Bioconvergence, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
16
|
Biofunctionalization of Endolysins with Oligosacharides: Formulation of Therapeutic Agents to Combat Multi-Resistant Bacteria and Potential Strategies for Their Application. POLYSACCHARIDES 2022. [DOI: 10.3390/polysaccharides3020018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
In the aquaculture sector, the biofunctionalization of biomaterials is discussed using materials from algae and analyzed as a possible potential strategy to overcome the challenges that hinder the future development of the application of endolysins in this field. Derived from years of analysis, endolysins have recently been considered as potential alternative therapeutic antibacterial agents, due to their attributes and ability to combat multi-resistant bacterial cells when applied externally. On the other hand, although the aquaculture sector has been characterized by its high production rates, serious infectious diseases have led to significant economic losses that persist to this day. Although there are currently interesting data from studies under in vitro conditions on the application of endolysins in this sector, there is little or no information on in vivo studies. This lack of analysis can be attributed to the relatively low stability of endolysins in marine conditions and to the complex gastrointestinal conditions of the organisms. This review provides updated information regarding the application of endolysins against multi-resistant bacteria of clinical and nutritional interest, previously addressing their important characteristics (structure, properties and stability). In addition, regarding the aquaculture sector, the biofunctionalization of biomaterials is discussed using materials from algae and analyzed as a possible potential strategy to overcome the challenges that hinder the future development of the application of endolysins in this field.
Collapse
|
17
|
In Vivo Recovery of Bacteriophages and Their Effects on Clostridium perfringens-Infected Broiler Chickens. Vet Sci 2022; 9:vetsci9030119. [PMID: 35324847 PMCID: PMC8953289 DOI: 10.3390/vetsci9030119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 03/03/2022] [Accepted: 03/04/2022] [Indexed: 11/20/2022] Open
Abstract
The objectives of this study were to recover bacteriophages (BPs) from the intestinal digesta of BP-fed broilers and to evaluate the antibacterial effects of encapsulated or powdered BPs in broiler chickens challenged with Clostridium perfringens. Day-old broiler chicks (n = 320/experiment) were randomly assigned to 32 pens (n = 10 broilers/pen) and allocated to one of four dietary groups: (1) unchallenged group (NEG); (2) C. perfringens-challenged group (POS); (3) POS group fed a diet supplemented with powdered BPs; and (4) POS group fed a diet supplemented with encapsulated BPs. On days 21, 22, and 23 post-hatch, all chickens except NEG were orally inoculated twice a day with 2 mL C. perfringens (1.0 × 108 cfu/mL). Varying BP levels were detected in gut digesta at all ages and were numerically or significantly higher in the encapsulated BP group than in the powdered BP group. Dietary powder or encapsulated BPs reversed the C. perfringens-mediated increase in crypt depth. In addition, villus height to crypt depth ratio was elevated in the NEG and BP-treated/challenged groups compared with that in the POS group. C. perfringens counts in the cecum were significantly lower in the BP-fed chickens than in the POS group. The encapsulated BP-supplemented diet-fed chickens had the highest serum IgA levels. Collectively, our results suggest that dietary BP remains viable in intestinal digesta upon ingestion and can inhibit cecal C. perfringens counts.
Collapse
|
18
|
Vasina DV, Antonova NP, Vorobev AM, Laishevtsev AI, Kapustin AV, Zulkarneev ER, Bochkareva SS, Kiseleva IA, Anurova MN, Aleshkin AV, Tkachuk AP, Gushchin VA. Efficacy of the Endolysin-Based Antibacterial Gel for Treatment of Anaerobic Infection Caused by Fusobacterium necrophorum. Antibiotics (Basel) 2021; 10:1260. [PMID: 34680839 PMCID: PMC8532708 DOI: 10.3390/antibiotics10101260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/30/2021] [Accepted: 10/14/2021] [Indexed: 11/25/2022] Open
Abstract
Abscess formation is a common complication of severe life-threatening infections caused by obligate anaerobes. Fusobacterium necrophorum is among the frequently detected anaerobic pathogens from clinical specimens associated with liver abscesses, skin and soft tissue infections, or oral abscesses. The antimicrobial therapy for this kind of infection needs to be optimized. Here, we examined the possibility of treating F. necrophorum-induced abscess wound infections with candidate therapeutics based on three endolysins with activity against a broad spectrum of aerobe Gram-negative pathogens. Antibacterial gel containing three Gram-negative bacteria-targeting endolysins, LysAm24, LysAp22, and LysECD7, was formulated for topical use. Abscess formation was induced in rabbits with F. necrophorum and caused systemic infection. The survival and lifespan of the animals, general parameters, and biochemical and hematological blood tests were analyzed to assess the effectiveness of the gel treatment for the wound infection. The administration of the investigated gel twice per day for 5 days resulted in less acute inflammation, with decreased leukocytes and segmented neutrophils in the blood, retardation of infection progression, and an almost two-fold increase in the lifespan of the animals compared to the placebo group. The results indicate that endolysin-based therapy is an effective approach to treat anaerobic bacterial infections. The use of endolysins as independent pharmaceuticals, or their combination with antibiotics, could significantly reduce the development of complications in infectious diseases caused by sensitive bacterial species.
Collapse
Affiliation(s)
- Daria V. Vasina
- N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (N.P.A.); (A.P.T.)
| | - Nataliia P. Antonova
- N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (N.P.A.); (A.P.T.)
| | - Aleksei M. Vorobev
- G.N. Gabrichevsky Moscow Research Institute for Epidemiology and Microbiology, 125212 Moscow, Russia; (A.M.V.); (E.R.Z.); (S.S.B.); (I.A.K.); (A.V.A.)
| | - Aleksei I. Laishevtsev
- Federal State Budget Scientific Institution “Federal Scientific Centre VIEV” (FSC VIEV), 117218 Moscow, Russia; (A.I.L.); (A.V.K.)
| | - Andrei V. Kapustin
- Federal State Budget Scientific Institution “Federal Scientific Centre VIEV” (FSC VIEV), 117218 Moscow, Russia; (A.I.L.); (A.V.K.)
| | - Eldar R. Zulkarneev
- G.N. Gabrichevsky Moscow Research Institute for Epidemiology and Microbiology, 125212 Moscow, Russia; (A.M.V.); (E.R.Z.); (S.S.B.); (I.A.K.); (A.V.A.)
| | - Svetlana S. Bochkareva
- G.N. Gabrichevsky Moscow Research Institute for Epidemiology and Microbiology, 125212 Moscow, Russia; (A.M.V.); (E.R.Z.); (S.S.B.); (I.A.K.); (A.V.A.)
| | - Irina A. Kiseleva
- G.N. Gabrichevsky Moscow Research Institute for Epidemiology and Microbiology, 125212 Moscow, Russia; (A.M.V.); (E.R.Z.); (S.S.B.); (I.A.K.); (A.V.A.)
| | - Mariia N. Anurova
- Department of Pharmaceutical Technology, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia;
| | - Andrei V. Aleshkin
- G.N. Gabrichevsky Moscow Research Institute for Epidemiology and Microbiology, 125212 Moscow, Russia; (A.M.V.); (E.R.Z.); (S.S.B.); (I.A.K.); (A.V.A.)
| | - Artem P. Tkachuk
- N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (N.P.A.); (A.P.T.)
| | - Vladimir A. Gushchin
- N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (N.P.A.); (A.P.T.)
- Department of Virology, Biological Faculty, Lomonosov Moscow State University, 119991 Moscow, Russia
| |
Collapse
|
19
|
Fermentative production of alternative antimicrobial peptides and enzymes. BIOCATALYSIS AND AGRICULTURAL BIOTECHNOLOGY 2021. [DOI: 10.1016/j.bcab.2021.102189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
20
|
Morzywolek A, Plotka M, Kaczorowska AK, Szadkowska M, Kozlowski LP, Wyrzykowski D, Makowska J, Waters JJ, Swift SM, Donovan DM, Kaczorowski T. Novel Lytic Enzyme of Prophage Origin from Clostridium botulinum E3 Strain Alaska E43 with Bactericidal Activity against Clostridial Cells. Int J Mol Sci 2021; 22:ijms22179536. [PMID: 34502443 PMCID: PMC8430805 DOI: 10.3390/ijms22179536] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 01/13/2023] Open
Abstract
Clostridium botulinum is a Gram-positive, anaerobic, spore-forming bacterium capable of producing botulinum toxin and responsible for botulism of humans and animals. Phage-encoded enzymes called endolysins, which can lyse bacteria when exposed externally, have potential as agents to combat bacteria of the genus Clostridium. Bioinformatics analysis revealed in the genomes of several Clostridium species genes encoding putative N-acetylmuramoyl-l-alanine amidases with anti-clostridial potential. One such enzyme, designated as LysB (224-aa), from the prophage of C. botulinum E3 strain Alaska E43 was chosen for further analysis. The recombinant 27,726 Da protein was expressed and purified from E. coli Tuner(DE3) with a yield of 37.5 mg per 1 L of cell culture. Size-exclusion chromatography and analytical ultracentrifugation experiments showed that the protein is dimeric in solution. Bioinformatics analysis and results of site-directed mutagenesis studies imply that five residues, namely H25, Y54, H126, S132, and C134, form the catalytic center of the enzyme. Twelve other residues, namely M13, H43, N47, G48, W49, A50, L73, A75, H76, Q78, N81, and Y182, were predicted to be involved in anchoring the protein to the lipoteichoic acid, a significant component of the Gram-positive bacterial cell wall. The LysB enzyme demonstrated lytic activity against bacteria belonging to the genera Clostridium, Bacillus, Staphylococcus, and Deinococcus, but did not lyse Gram-negative bacteria. Optimal lytic activity of LysB occurred between pH 4.0 and 7.5 in the absence of NaCl. This work presents the first characterization of an endolysin derived from a C. botulinum Group II prophage, which can potentially be used to control this important pathogen.
Collapse
Affiliation(s)
- Agnieszka Morzywolek
- Laboratory of Extremophiles Biology, Department of Microbiology, Faculty of Biology, University of Gdansk, 80-822 Gdansk, Poland; (A.M.); (M.S.)
| | - Magdalena Plotka
- Laboratory of Extremophiles Biology, Department of Microbiology, Faculty of Biology, University of Gdansk, 80-822 Gdansk, Poland; (A.M.); (M.S.)
- Correspondence: (M.P.); (T.K.)
| | - Anna-Karina Kaczorowska
- Collection of Plasmids and Microorganisms, Faculty of Biology, University of Gdansk, 80-308 Gdansk, Poland;
| | - Monika Szadkowska
- Laboratory of Extremophiles Biology, Department of Microbiology, Faculty of Biology, University of Gdansk, 80-822 Gdansk, Poland; (A.M.); (M.S.)
| | - Lukasz P. Kozlowski
- Institute of Informatics, Faculty of Mathematics, Informatics and Mechanics, University of Warsaw, 02-097 Warsaw, Poland;
| | - Dariusz Wyrzykowski
- Department of General and Inorganic Chemistry, Faculty of Chemistry, University of Gdansk, 80-308 Gdansk, Poland; (D.W.); (J.M.)
| | - Joanna Makowska
- Department of General and Inorganic Chemistry, Faculty of Chemistry, University of Gdansk, 80-308 Gdansk, Poland; (D.W.); (J.M.)
| | - Jerel J. Waters
- Animal Biosciences and Biotechnology Laboratory, ARS, NEA, USDA, Beltsville, MD 20705-2350, USA; (J.J.W.); (S.M.S.); (D.M.D.)
| | - Steven M. Swift
- Animal Biosciences and Biotechnology Laboratory, ARS, NEA, USDA, Beltsville, MD 20705-2350, USA; (J.J.W.); (S.M.S.); (D.M.D.)
| | - David M. Donovan
- Animal Biosciences and Biotechnology Laboratory, ARS, NEA, USDA, Beltsville, MD 20705-2350, USA; (J.J.W.); (S.M.S.); (D.M.D.)
| | - Tadeusz Kaczorowski
- Laboratory of Extremophiles Biology, Department of Microbiology, Faculty of Biology, University of Gdansk, 80-822 Gdansk, Poland; (A.M.); (M.S.)
- Correspondence: (M.P.); (T.K.)
| |
Collapse
|
21
|
Xu Y. Phage and phage lysins: New era of bio-preservatives and food safety agents. J Food Sci 2021; 86:3349-3373. [PMID: 34302296 DOI: 10.1111/1750-3841.15843] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 06/09/2021] [Accepted: 06/14/2021] [Indexed: 01/21/2023]
Abstract
There has been an increase in the search and application of new antimicrobial agents as alternatives to use of chemical preservatives and antibiotic-like compounds by the food industry. The massive use of antibiotic has created a reservoir of antibiotic-resistant bacteria that find their way from farm to humans. Thus, there exists an imperative need to explore new antibacterial options and bacteriophages perfectly fit into the class of safe and potent antimicrobials. Phage bio-control has come a long way owing to advances with use of phage cocktails, recombinant phages, and phage lysins; however, there still exists unmet challenges that restrict the number of phage-based products reaching the market. Hence, further studies are required to explore for more efficient phage-based bio-control strategies that can become an integral part of food safety protocols. This review thus aims to highlight the recent developments made in the application of phages and phage enzymes covering pre-harvest as well as post-harvest usage. It further focuses on the major issues in both phage and phage lysin research hindering their optimum use while detailing out the advances made by researchers lately in this direction for full exploitation of phages and phage lysins in the food sector.
Collapse
Affiliation(s)
- Yingmin Xu
- Food Technology College Jiangsu Vocational College of Agriculture and Forestry, China
| |
Collapse
|
22
|
Jiang Y, Xu D, Wang L, Qu M, Li F, Tan Z, Yao L. Characterization of a broad-spectrum endolysin LysSP1 encoded by a Salmonella bacteriophage. Appl Microbiol Biotechnol 2021; 105:5461-5470. [PMID: 34241646 DOI: 10.1007/s00253-021-11366-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 05/16/2021] [Accepted: 05/25/2021] [Indexed: 12/17/2022]
Abstract
Foodborne pathogens have caused many public health incidents and heavy economic burden. Endolysins have been proven to have efficient bactericidal activity against pathogens with low incidence of resistance. In this study, the recombinant endolysin LysSP1 encoded by Salmonella Typhimurium lytic bacteriophage SLMP1 was obtained by prokaryotic expression, and its characteristics were analyzed. Ethylenediaminetetraacetic acid (EDTA) can be used as the outer membrane permeabilizer to increase the bactericidal activity of LysSP1. Under the synergism of 5 mmol/L EDTA, LysSP1 exhibited a strong bactericidal activity against Salmonella Typhimurium ATCC14028. LysSP1 was stable at 4°C for 7 days and at -20°C for 180 days. LysSP1 remained the optimal activity at 40°C and was efficiently active at alkaline condition (pH 8.0-10.0). Divalent metal ions could not enhance the bactericidal activity of LysSP1 and even caused the significant reduction of bactericidal activity. LysSP1 not only could lyse Salmonella, but also could lyse other Gram-negative strains and Gram-positive strains. These results indicated that LysSP1 is a broad-spectrum endolysin and has potential as an antimicrobial agent against Salmonella and other foodborne pathogens. KEY POINTS: • Recombinant endolysin LysSP1 can be prepared by prokaryotic expression. • LysSP1 has stable nature and strong bactericidal activity on Salmonella Typhimurium with EDTA. • LysSP1 has a broad range of hosts including Gram-negative bacteria and Gram-positive bacteria.
Collapse
Affiliation(s)
- Yanhua Jiang
- Key Laboratory of Testing and Evaluation for Aquatic Product Safety and Quality, Ministry of Agriculture and Rural Affairs, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, No. 106, Nanjing Road, Qingdao, 266071, People's Republic of China
| | - Dongqin Xu
- Key Laboratory of Testing and Evaluation for Aquatic Product Safety and Quality, Ministry of Agriculture and Rural Affairs, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, No. 106, Nanjing Road, Qingdao, 266071, People's Republic of China
- College of Food Sciences and Technology, Shanghai Ocean University, Shanghai, 201306, People's Republic of China
| | - Lianzhu Wang
- Key Laboratory of Testing and Evaluation for Aquatic Product Safety and Quality, Ministry of Agriculture and Rural Affairs, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, No. 106, Nanjing Road, Qingdao, 266071, People's Republic of China
| | - Meng Qu
- Key Laboratory of Testing and Evaluation for Aquatic Product Safety and Quality, Ministry of Agriculture and Rural Affairs, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, No. 106, Nanjing Road, Qingdao, 266071, People's Republic of China
| | - Fengling Li
- Key Laboratory of Testing and Evaluation for Aquatic Product Safety and Quality, Ministry of Agriculture and Rural Affairs, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, No. 106, Nanjing Road, Qingdao, 266071, People's Republic of China
| | - Zhijun Tan
- Key Laboratory of Testing and Evaluation for Aquatic Product Safety and Quality, Ministry of Agriculture and Rural Affairs, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, No. 106, Nanjing Road, Qingdao, 266071, People's Republic of China
- Pilot National Laboratory for Marine Science and Technology, Qingdao, 266033, People's Republic of China
| | - Lin Yao
- Key Laboratory of Testing and Evaluation for Aquatic Product Safety and Quality, Ministry of Agriculture and Rural Affairs, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, No. 106, Nanjing Road, Qingdao, 266071, People's Republic of China.
| |
Collapse
|
23
|
Benameur Q, Gervasi T, Pellizzeri V, Pľuchtová M, Gruľová D, Cicero N, Meriem-Hind B. Comparison of sensitivity to a commercial Origanum vulgare essential oil between extended-spectrum β-lactamases (ESBL-) and non-ESBL-producing Enterobacteriaceae isolates. Nat Prod Res 2021; 36:2830-2835. [PMID: 34121535 DOI: 10.1080/14786419.2021.1933969] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
This study aimed to evaluate the antibacterial effect of Origanum vulgare essential oil (OVEO) against ESBL- and non-ESBL-producing multidrug-resistant (MDR) Enterobacteriaceae isolates. OVEO composition was determined using Gas chromatograph-mass spectrometer (GC/MS). In the current study, the effect of OVEO was studied on seven MDR Enterobacteriaceae isolates. Antibacterial activity of OVEO was investigated by the disc diffusion assay and twofold serial dilution method. GC/MS analysis identified thymol (78.21%) as the single major component present in the OVEO. This EO showed an unexpectedly high antibacterial activity against all the studied MDR Enterobacteriaceae isolates, with inhibition zone diameters and minimum inhibitory concentration (MIC) values ranging from 28 ± 0.6 to 35 ± 0.6 mm and 0.31 ± 0.0 to 5 ± 0.0 μL/mL, respectively. However, ESBL-producing isolates were more susceptible to OVEO than non-ESBL producing isolates. This study compared, for the first time, the sensitivity to OVEO between ESBL and non-ESBL-producing Enterobacteriaceae isolates.
Collapse
Affiliation(s)
- Qada Benameur
- Nursing Department, Faculty of Nature and Life Sciences, University of Abdelhamid Ibn Badis of Mostaganem, Mostaganem, Algeria.,Research Laboratory 'Health and Animal Productions', Higher National Veterinary School, Algiers, Algeria
| | - Teresa Gervasi
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Italy
| | - Vito Pellizzeri
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Italy
| | - Mária Pľuchtová
- Department of Ecology, Faculty of Humanities and Natural Sciences, University of Prešov, Slovakia
| | - Daniela Gruľová
- Department of Ecology, Faculty of Humanities and Natural Sciences, University of Prešov, Slovakia
| | - Nicola Cicero
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Italy
| | - Benmahdi Meriem-Hind
- Research Laboratory 'Health and Animal Productions', Higher National Veterinary School, Algiers, Algeria
| |
Collapse
|
24
|
Stable Recombinant-Gene Expression from a Ligilactobacillus Live Bacterial Vector via Chromosomal Integration. Appl Environ Microbiol 2021; 87:AEM.00392-21. [PMID: 33741626 DOI: 10.1128/aem.00392-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 03/08/2021] [Indexed: 01/08/2023] Open
Abstract
Disease control in animal production systems requires constant vigilance. Historically, the application of in-feed antibiotics to control bacteria and improve performance has been a much-used approach to maintain animal health and welfare. However, the widespread use of in-feed antibiotics is thought to increase the risk of antibiotic resistance developing. Alternative methods to control disease and maintain productivity need to be developed. Live vaccination is useful in preventing colonization of mucosa-dwelling pathogens by inducing a mucosal immune response. Native poultry isolate Ligilactobacillus agilis La3 (previously Lactobacillus agilis) has been identified as a candidate for use as a live vector to deliver therapeutic proteins such as bacteriocins, phage endolysins, or vaccine antigens to the gastrointestinal tract of chickens. In this study, the complete genome sequence of L. agilis La3 was determined and transcriptome analysis was undertaken to identify highly expressed genes. Predicted promoter regions and ribosomal binding sites from constitutively expressed genes were used to construct recombinant protein expression cassettes. A series of double-crossover shuttle plasmids were constructed to facilitate rapid selectable integration of expression cassettes into the L agilis La3 chromosome via homologous recombination. Inserts showed 100% stable integration over 100 generations without selection. A positive relationship was found between protein expression levels and the predicted strength of the promoters. Using this system, stable chromosomal expression of a Clostridium perfringens antigen, rNetB, was demonstrated without selection. Finally, two recombinant strains, L agilis La3::P eft -rnetB and L agilis La3::P cwah -rnetB, were constructed and characterized, and they showed potential for future application as live vaccines in chickens.IMPORTANCE Therapeutic proteins such as antigens can be used to prevent infectious diseases in poultry. However, traditional vaccine delivery by intramuscular or subcutaneous injection generally has not proven effective for mucosa-dwelling microorganisms that live within the gastrointestinal tract. Utilizing live bacteria to deliver vaccine antigens directly to the gut immune system can overcome some of the limitations of conventional vaccination. In this work, Ligilactobacillus agilis La3, an especially effective gut colonizer, has been analyzed and engineered with modular and stable expression systems to produce recombinant proteins. To demonstrate the effectiveness of the system, expression of a vaccine antigen from poultry pathogen Clostridium perfringens was monitored over 100 generations without selection and found to be completely stable. This study demonstrates the development of genetic tools and novel constitutive expression systems and further development of L. agilis La3 as a live delivery vehicle for recombinant proteins.
Collapse
|
25
|
Zhang Z, Lahti M, Douillard FP, Korkeala H, Lindström M. Phage lysin that specifically eliminates Clostridium botulinum Group I cells. Sci Rep 2020; 10:21571. [PMID: 33299101 PMCID: PMC7725837 DOI: 10.1038/s41598-020-78622-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 11/23/2020] [Indexed: 01/21/2023] Open
Abstract
Clostridium botulinum poses a serious threat to food safety and public health by producing potent neurotoxin during its vegetative growth and causing life-threatening neuroparalysis, botulism. While high temperature can be utilized to eliminate C. botulinum spores and the neurotoxin, non-thermal elimination of newly germinated C. botulinum cells before onset of toxin production could provide an alternative or additional factor controlling the risk of botulism in some applications. Here we introduce a putative phage lysin that specifically lyses vegetative C. botulinum Group I cells. This lysin, called CBO1751, efficiently kills cells of C. botulinum Group I strains at the concentration of 5 µM, but shows little or no lytic activity against C. botulinum Group II or III or other Firmicutes strains. CBO1751 is active at pH from 6.5 to 10.5. The lytic activity of CBO1751 is tolerant to NaCl (200 mM), but highly susceptible to divalent cations Ca2+ and Mg2+ (50 mM). CBO1751 readily and effectively eliminates C. botulinum during spore germination, an early stage preceding vegetative growth and neurotoxin production. This is the first report of an antimicrobial lysin against C. botulinum, presenting high potential for developing a novel antibotulinal agent for non-thermal applications in food and agricultural industries.
Collapse
Affiliation(s)
- Zhen Zhang
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, P. O. Box 66, 00014, Helsinki, Finland
| | - Meeri Lahti
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, P. O. Box 66, 00014, Helsinki, Finland
| | - François P Douillard
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, P. O. Box 66, 00014, Helsinki, Finland
| | - Hannu Korkeala
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, P. O. Box 66, 00014, Helsinki, Finland
| | - Miia Lindström
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, P. O. Box 66, 00014, Helsinki, Finland.
| |
Collapse
|
26
|
Mondal SI, Draper LA, Ross RP, Hill C. Bacteriophage endolysins as a potential weapon to combat Clostridioides difficile infection. Gut Microbes 2020; 12:1813533. [PMID: 32985336 PMCID: PMC7524323 DOI: 10.1080/19490976.2020.1813533] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Clostridioides difficile is the leading cause of health-care-associated infection throughout the developed world and contributes significantly to patient morbidity and mortality. Typically, antibiotics are used for the primary treatment of C. difficile infections (CDIs), but they are not universally effective for all ribotypes and can result in antibiotic resistance and recurrent infection, while also disrupting the microbiota. Novel targeted therapeutics are urgently needed to combat CDI. Bacteriophage-derived endolysins are required to disrupt the bacterial cell wall of their target bacteria and are possible alternatives to antibiotics. These lytic proteins could potentially replace or augment antibiotics in CDI treatment. We discuss candidate therapeutic lysins derived from phages/prophages of C. difficile and their potential as antimicrobials against CDI. Additionally, we review the antibacterial potential of some recently identified homologues of C. difficile endolysins. Finally, the challenges of endolysins are considered with respect to the development of novel lysin-based therapies.
Collapse
Affiliation(s)
- Shakhinur Islam Mondal
- APC Microbiome Ireland, University College Cork, Cork, Ireland,Genetic Engineering and Biotechnology Department, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Lorraine A. Draper
- APC Microbiome Ireland, University College Cork, Cork, Ireland,School of Microbiology, University College Cork, Cork, Ireland
| | - R Paul Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland,School of Microbiology, University College Cork, Cork, Ireland,Teagasc Food Research Centre, Moorepark, Cork, Ireland
| | - Colin Hill
- APC Microbiome Ireland, University College Cork, Cork, Ireland,School of Microbiology, University College Cork, Cork, Ireland,CONTACT Colin Hill APC Microbiome Ireland & School of Microbiology, University College Cork, Cork, Ireland
| |
Collapse
|
27
|
Cho JH, Kwon JG, O'Sullivan DJ, Ryu S, Lee JH. Development of an endolysin enzyme and its cell wall-binding domain protein and their applications for biocontrol and rapid detection of Clostridium perfringens in food. Food Chem 2020; 345:128562. [PMID: 33189482 DOI: 10.1016/j.foodchem.2020.128562] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 10/28/2020] [Accepted: 11/01/2020] [Indexed: 01/10/2023]
Abstract
Clostridium perfringens is a well-known pathogen that causes food-borne illnesses. Although bacteriophages can be effective natural food preservatives, phage endolysin and cell wall-binding domain (CBD) provide useful materials for lysis of C. perfringens and rapid detection. The genome of phage CPAS-15 consists of 51.8-kb double-stranded circular DNA with 78 open reading frames, including an endolysin gene. The apparent absence of a virulence factor or toxin gene suggests its safety in food applications. C. perfringens endolysin (LysCPAS15) inhibits host cells by up to a 3-log reduction in 2 h, and enhanced green fluorescent protein (EGFP)-fused CBD protein (EGFP-LysCPAS15_CBD1) detects C. perfringens within 5 min. Both exhibit broader host range spectra and higher stabilities than a bacteriophage. Tests in milk show the same host lysis and specific detection activities, with no hindrance effect from food matrices, indicating that endolysin and its CBD can provide food extended protection from C. perfringens contamination.
Collapse
Affiliation(s)
- Jae-Hyun Cho
- Department of Food Science and Biotechnology, Graduate School of Biotechnology, Kyung Hee University, Yongin, South Korea
| | - Joon-Gi Kwon
- Department of Food and Animal Biotechnology, Department of Agricultural Biotechnology, Center for Food and Bioconvergence, Seoul National University, Seoul, South Korea
| | - Daniel J O'Sullivan
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN 55108, USA
| | - Sangryeol Ryu
- Department of Food and Animal Biotechnology, Department of Agricultural Biotechnology, Center for Food and Bioconvergence, Seoul National University, Seoul, South Korea
| | - Ju-Hoon Lee
- Department of Food and Animal Biotechnology, Department of Agricultural Biotechnology, Center for Food and Bioconvergence, Seoul National University, Seoul, South Korea.
| |
Collapse
|
28
|
De Maesschalck V, Gutiérrez D, Paeshuyse J, Lavigne R, Briers Y. Advanced engineering of third-generation lysins and formulation strategies for clinical applications. Crit Rev Microbiol 2020; 46:548-564. [PMID: 32886565 DOI: 10.1080/1040841x.2020.1809346] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
One of the possible solutions for the current antibiotic resistance crisis may be found in (often bacteriophage-derived) peptidoglycan hydrolases. The first clinical trials of these natural enzymes, coined here as first-generation lysins, are currently ongoing. Moving beyond natural endolysins with protein engineering established the second generation of lysins. In second-generation lysins, the focus lies on improving antibacterial and biochemical properties such as antimicrobial activity and stability, as well as expanding their activities towards Gram-negative pathogens. However, solutions to particular key challenges regarding clinical applications are only beginning to emerge in the third generation of lysins, in which protein and biochemical engineering efforts focus on improving properties relevant under clinical conditions. In addition, increasingly advanced formulation strategies are developed to increase the bioavailability, antibacterial activity, and half-life, and to reduce pro-inflammatory responses. This review focuses on third-generation and advanced formulation strategies that are developed to treat infections, ranging from topical to systemic applications. Together, these efforts may fully unlock the potential of lysin therapy and will propel it as a true antibiotic alternative or supplement.
Collapse
Affiliation(s)
- Vincent De Maesschalck
- Department of Biosystems, KU Leuven, Leuven, Belgium.,Department of Biotechnology, Ghent University, Gent, Belgium
| | - Diana Gutiérrez
- Department of Biotechnology, Ghent University, Gent, Belgium
| | - Jan Paeshuyse
- Department of Biosystems, KU Leuven, Leuven, Belgium
| | - Rob Lavigne
- Department of Biosystems, KU Leuven, Leuven, Belgium
| | - Yves Briers
- Department of Biotechnology, Ghent University, Gent, Belgium
| |
Collapse
|
29
|
Effect of Lactococcus lactis expressing phage endolysin on the late blowing defect of cheese caused by Clostridium tyrobutyricum. Int J Food Microbiol 2020; 329:108686. [DOI: 10.1016/j.ijfoodmicro.2020.108686] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 05/13/2020] [Accepted: 05/22/2020] [Indexed: 01/28/2023]
|
30
|
Żbikowska K, Michalczuk M, Dolka B. The Use of Bacteriophages in the Poultry Industry. Animals (Basel) 2020; 10:E872. [PMID: 32443410 PMCID: PMC7278383 DOI: 10.3390/ani10050872] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 05/15/2020] [Accepted: 05/16/2020] [Indexed: 12/13/2022] Open
Abstract
The emergence of multidrug-resistant infections and antibiotic failures have raised concerns over human and veterinary medicine worldwide. Poultry production has had to confront the problems of an alarming increase in bacterial resistance, including zoonotic pathogens. According to the European Food Safety Authority (EFSA), campylobacteriosis and salmonellosis have been the most frequently reported human foodborne diseases linked to poultry. This situation has strongly stimulated a renewal of scientists' interest in bacteriophages (phages) since the beginning of the 21st century. Bacteriophages are the viruses of bacteria. They are abundant in nature, and accompany bacteria in each environment they colonize, including human microbiota. In this review, we focused on the use of bacteriophages as therapeutic agents to treat infections and reduce counts of pathogenic bacteria in poultry, as biocontrol agents to eliminate foodborne pathogens on/in food, and also as disinfectants to reduce contamination on food-contact surfaces or poultry carcasses in industrial conditions. Most of the phage-based products are targeted against the main foodborne pathogens, such as Campylobacter jejuni, Salmonella spp., Escherichia coli, Listeria monocytogenes, Staphylococcus aureus, and Clostridium perfringens. Phages are currently addressed at all stages of the poultry production "from farm to fork", however, their implementation into live birds and food products still provokes discussions especially in the context of the current legal framework, limitations, as well as public health and safety.
Collapse
Affiliation(s)
- Katarzyna Żbikowska
- Department of Animal Breeding, Institute of Animal Sciences, Warsaw University of Life Sciences—SGGW, Ciszewskiego 8 St., 02-786 Warsaw, Poland; (K.Ż.); (M.M.)
| | - Monika Michalczuk
- Department of Animal Breeding, Institute of Animal Sciences, Warsaw University of Life Sciences—SGGW, Ciszewskiego 8 St., 02-786 Warsaw, Poland; (K.Ż.); (M.M.)
| | - Beata Dolka
- Department of Pathology and Veterinary Diagnostics, Institute of Veterinary Medicine, Warsaw University of Life Sciences—SGGW, Nowoursynowska 159c St., 02-776 Warsaw, Poland
| |
Collapse
|
31
|
Hammond RW, Swift SM, Foster-Frey JA, Kovalskaya NY, Donovan DM. Optimized production of a biologically active Clostridium perfringens glycosyl hydrolase phage endolysin PlyCP41 in plants using virus-based systemic expression. BMC Biotechnol 2019; 19:101. [PMID: 31864319 PMCID: PMC6925876 DOI: 10.1186/s12896-019-0594-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 12/10/2019] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Clostridium perfringens, a gram-positive, anaerobic, rod-shaped bacterium, is the third leading cause of human foodborne bacterial disease and a cause of necrotic enteritis in poultry. It is controlled using antibiotics, widespread use of which may lead to development of drug-resistant bacteria. Bacteriophage-encoded endolysins that degrade peptidoglycans in the bacterial cell wall are potential replacements for antibiotics. Phage endolysins have been identified that exhibit antibacterial activities against several Clostridium strains. RESULTS An Escherichia coli codon-optimized gene encoding the glycosyl hydrolase endolysin (PlyCP41) containing a polyhistidine tag was expressed in E. coli. In addition, The E. coli optimized endolysin gene was engineered for expression in plants (PlyCP41p) and a plant codon-optimized gene (PlyCP41pc), both containing a polyhistidine tag, were expressed in Nicotiana benthamiana plants using a potato virus X (PVX)-based transient expression vector. PlyCP41p accumulated to ~ 1% total soluble protein (100μg/gm f. wt. leaf tissue) without any obvious toxic effects on plant cells, and both the purified protein and plant sap containing the protein lysed C. perfringens strain Cp39 in a plate lysis assay. Optimal systemic expression of PlyCP41p was achieved at 2 weeks-post-infection. PlyCP41pc did not accumulate to higher levels than PlyCP41p in infected tissue. CONCLUSION We demonstrated that functionally active bacteriophage PlyCP41 endolysin can be produced in systemically infected plant tissue with potential for use of crude plant sap as an effective antimicrobial agent against C. perfringens.
Collapse
Affiliation(s)
- Rosemarie W Hammond
- USDA ARS NEA BARC Molecular Plant Pathology Laboratory, Beltsville, MD, 20705, USA.
| | - Steven M Swift
- USDA ARS NEA BARC Animal Biosciences and Biotechnology Laboratory, Beltsville, MD, 20705, USA
| | - Juli A Foster-Frey
- USDA ARS NEA BARC Animal Biosciences and Biotechnology Laboratory, Beltsville, MD, 20705, USA
| | - Natalia Y Kovalskaya
- USDA ARS NEA BARC Molecular Plant Pathology Laboratory, Beltsville, MD, 20705, USA
- Oak Ridge Institute for Science and Education, ORISE, Beltsville, MD, 20705, USA
| | - David M Donovan
- USDA ARS NEA BARC Animal Biosciences and Biotechnology Laboratory, Beltsville, MD, 20705, USA
| |
Collapse
|
32
|
Gigante A, Atterbury RJ. Veterinary use of bacteriophage therapy in intensively-reared livestock. Virol J 2019; 16:155. [PMID: 31831017 PMCID: PMC6909661 DOI: 10.1186/s12985-019-1260-3] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 11/22/2019] [Indexed: 02/06/2023] Open
Abstract
Zoonoses are infectious diseases transmitted directly or indirectly between animals and humans. Several important zoonotic pathogens colonize farm animals asymptomatically, which may lead to contamination of the food chain and public health hazards. Moreover, routine sampling of carcasses at retail by government authorities over the past 20 years suggests the prevalence of antibiotic resistance in foodborne pathogens has increased. If this continues, antibiotics may be ineffective against such pathogens in the future and alternative approaches, such as phage therapy, may be necessary. Intensive livestock farming is the only realistic way of meeting the demand for meat from an increasing global population and growth in middle class consumers in developing countries, particularly in Asia. This review elaborates on the use of phages to control zoonotic pathogens in intensively-reared livestock (poultry and pigs).
Collapse
Affiliation(s)
- Adriano Gigante
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, Leicestershire, LE12 5RD UK
| | - Robert J Atterbury
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, Leicestershire, LE12 5RD UK
| |
Collapse
|
33
|
Swift SM, Waters JJ, Rowley DT, Oakley BB, Donovan DM. Characterization of two glycosyl hydrolases, putative prophage endolysins, that target Clostridium perfringens. FEMS Microbiol Lett 2019; 365:5053808. [PMID: 30010898 DOI: 10.1093/femsle/fny179] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 07/11/2018] [Indexed: 12/31/2022] Open
Abstract
Clostridium perfringens, a spore-forming anaerobic bacterium, causes food poisoning and gas gangrene in humans and is an agent of necrotizing enteritis in poultry, swine and cattle. Endolysins are peptidoglycan hydrolases from bacteriophage that degrade the bacterial host cell wall causing lysis and thus harbor antimicrobial therapy potential. The genes for the PlyCP10 and PlyCP41 endolysins were found in prophage regions of the genomes from C. perfringens strains Cp10 and Cp41, respectively. The gene for PlyCP10 encodes a protein of 351 amino acids, while the gene for PlyCP41 encodes a protein of 335 amino acids. Both proteins harbor predicted glycosyl hydrolase domains. Recombinant PlyCP10 and PlyCP41 were expressed in E. coli with C-terminal His-tags, purified by nickel chromatography and characterized in vitro. PlyCP10 activity was greatest at pH 6.0, and between 50 and 100 mM NaCl. PlyCP41 activity was greatest between pH 6.5 and 7.0, and at 50 mM NaCl, with retention of activity as high as 600 mM NaCl. PlyCP10 lost most of its activity above 42°C, whereas PlyCP41 survived at 50°C for 30 min and still retained >60% activity. Both enzymes had lytic activity against 75 C. perfringens strains (isolates from poultry, swine and cattle) suggesting therapeutic potential.
Collapse
Affiliation(s)
- Steven M Swift
- Animal Biosciences and Biotechnology Laboratory, Agricultural Research Service, BARC, USDA, 10300 Baltimore Ave., Beltsville, MD, USA
| | - Jerel J Waters
- Animal Biosciences and Biotechnology Laboratory, Agricultural Research Service, BARC, USDA, 10300 Baltimore Ave., Beltsville, MD, USA
| | - D Treva Rowley
- Animal Biosciences and Biotechnology Laboratory, Agricultural Research Service, BARC, USDA, 10300 Baltimore Ave., Beltsville, MD, USA
| | - Brian B Oakley
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA, USA
| | - David M Donovan
- Animal Biosciences and Biotechnology Laboratory, Agricultural Research Service, BARC, USDA, 10300 Baltimore Ave., Beltsville, MD, USA
| |
Collapse
|
34
|
Jończyk-Matysiak E, Łodej N, Kula D, Owczarek B, Orwat F, Międzybrodzki R, Neuberg J, Bagińska N, Weber-Dąbrowska B, Górski A. Factors determining phage stability/activity: challenges in practical phage application. Expert Rev Anti Infect Ther 2019; 17:583-606. [PMID: 31322022 DOI: 10.1080/14787210.2019.1646126] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Introduction: Phages consist of nucleic acids and proteins that may lose their activity under different physico-chemical conditions. The production process of phage formulations may decrease phage infectivity. Ingredients present in the preparation may influence phage particles, although preparation and storage conditions may also cause variations in phage titer. Significant factors are the manner of phage application, the patient's immune system status, the type of medication being taken, and diet. Areas covered: We discuss factors determining phage activity and stability, which is relevant for the preparation and application of phage formulations with the highest therapeutic efficacy. Our article should be helpful for more insightful implementation of clinical trials, which could pave the way for successful phage therapy. Expert opinion: The number of naturally occurring phages is practically unlimited and phages vary in their susceptibility to external factors. Modern methods offer engineering techniques which should lead to enhanced precision in phage delivery and anti-bacterial activity. Recent data suggesting that phages may also be used in treating nonbacterial infections as well as anti-inflammatory and immunomodulatory agents add further weight to such studies. It may be anticipated that different phage activities could have varying susceptibility to factors determining their actions.
Collapse
Affiliation(s)
- Ewa Jończyk-Matysiak
- a Bacteriophage Laboratory, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences , Wroclaw , Poland
| | - Norbert Łodej
- a Bacteriophage Laboratory, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences , Wroclaw , Poland
| | - Dominika Kula
- a Bacteriophage Laboratory, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences , Wroclaw , Poland
| | - Barbara Owczarek
- a Bacteriophage Laboratory, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences , Wroclaw , Poland
| | - Filip Orwat
- a Bacteriophage Laboratory, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences , Wroclaw , Poland
| | - Ryszard Międzybrodzki
- a Bacteriophage Laboratory, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences , Wroclaw , Poland.,b Department of Clinical Immunology, Transplantation Institute, Medical University of Warsaw , Warsaw , Poland.,c Phage Therapy Unit, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences , Wroclaw , Poland
| | - Joanna Neuberg
- a Bacteriophage Laboratory, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences , Wroclaw , Poland
| | - Natalia Bagińska
- a Bacteriophage Laboratory, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences , Wroclaw , Poland
| | - Beata Weber-Dąbrowska
- a Bacteriophage Laboratory, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences , Wroclaw , Poland.,c Phage Therapy Unit, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences , Wroclaw , Poland
| | - Andrzej Górski
- a Bacteriophage Laboratory, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences , Wroclaw , Poland.,b Department of Clinical Immunology, Transplantation Institute, Medical University of Warsaw , Warsaw , Poland.,c Phage Therapy Unit, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences , Wroclaw , Poland
| |
Collapse
|
35
|
Aghraz A, Albergamo A, Benameur Q, Salvo A, Larhsini M, Markouk M, Gervasi T, Cicero N. Polyphenols contents, heavy metals analysis and in vitro antibacterial activity of extracts from Cladanthus arabicus and Bubonium imbricatum of Moroccan Origin. Nat Prod Res 2019; 34:63-70. [DOI: 10.1080/14786419.2019.1573424] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Abdellah Aghraz
- Laboratory of Biotechnology, Protection and Valorisation of Plant Ressources (URAC35 Association Unit), Cadi Ayyad University, Marrakesh, Morocco
| | - Ambrogina Albergamo
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Qada Benameur
- Nursing Department Faculty of Natural Sciences and Life, Abdelhamid Ibn Badis University, Mostaganem, Algeria
- Ecole Nationale Supérieure Vétérinaire d’El-Harrach, Laboratoire de Recherche «Santé et Production Animale», Algiers, Algeria
| | - Andrea Salvo
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Mustapha Larhsini
- Laboratory of Biotechnology, Protection and Valorisation of Plant Ressources (URAC35 Association Unit), Cadi Ayyad University, Marrakesh, Morocco
| | - Mohamed Markouk
- Laboratory of Biotechnology, Protection and Valorisation of Plant Ressources (URAC35 Association Unit), Cadi Ayyad University, Marrakesh, Morocco
| | - Teresa Gervasi
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Nicola Cicero
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| |
Collapse
|
36
|
Kazanavičiūtė V, Misiūnas A, Gleba Y, Giritch A, Ražanskienė A. Plant-expressed bacteriophage lysins control pathogenic strains of Clostridium perfringens. Sci Rep 2018; 8:10589. [PMID: 30002425 PMCID: PMC6043497 DOI: 10.1038/s41598-018-28838-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 07/02/2018] [Indexed: 01/10/2023] Open
Abstract
The anaerobic spore-forming bacterium Clostridium perfringens is a source of one of the most common food-borne illnesses in the United States and Europe. The costs associated with disease management are high and interventions are limited; therefore, effective and safe antimicrobials are needed to control food contamination by C. perfringens. A viable solution to this problem could be bacteriophage lysins used as food additives or food processing aids. Such antimicrobials could be produced cost-effectively and in ample supply in green plants. By using edible plant species as production hosts the need for expensive product purification can be reduced or obviated. We describe the first successful expression in plants of C. perfringens-specific bacteriophage lysins. We demonstrate that six lysins belonging to two different families (N-acetylmuramoyl-L-alanine amidase and glycosyl hydrolase 25) are active against a panel of enteropathogenic C. perfringens strains under salinity and acidity conditions relevant to food preparation environments. We also demonstrate that plant-expressed lysins prevent multiplication of C. perfringens on cooked meat matrices far better than nisin, the only currently approved bacteriocin food preservative to control this pathogen.
Collapse
Affiliation(s)
| | | | - Yuri Gleba
- Nomad Bioscience GmbH, Biozentrum Halle, Weinbergweg 22, D-06120, Halle (Saale), Germany
| | - Anatoli Giritch
- Nomad Bioscience GmbH, Biozentrum Halle, Weinbergweg 22, D-06120, Halle (Saale), Germany
| | | |
Collapse
|
37
|
Structure of an Acinetobacter Broad-Range Prophage Endolysin Reveals a C-Terminal α-Helix with the Proposed Role in Activity against Live Bacterial Cells. Viruses 2018; 10:v10060309. [PMID: 29882827 PMCID: PMC6024848 DOI: 10.3390/v10060309] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 05/28/2018] [Accepted: 05/31/2018] [Indexed: 01/07/2023] Open
Abstract
Proteins that include enzymatic domain degrading the bacterial cell wall and a domain providing transport through the bacterial outer membrane are considered as prospective compounds to combat pathogenic Gram-negative bacteria. This paper presents an isolation and study of an enzyme of this class naturally encoded in the prophage region of Acinetobacter baumannii AB 5075 genome. Recombinant protein expressed in E. coli exhibits an antimicrobial activity with respect to live cultures of Gram-negative bacteria reducing the population of viable bacteria by 1.5⁻2 log colony forming units (CFU)/mL. However the protein becomes rapidly inactivated and enables the bacteria to restore the population. AcLys structure determined by X-ray crystallography reveals a predominantly α—helical fold similar to bacteriophage P22 lysozyme. The С-terminal part of AcLys polypeptide chains forms an α—helix enriched by Lys and Arg residues exposed outside of the protein globule. Presumably this type of structure of the C-terminal α—helix has evolved evolutionally enabling the endolysin to pass the inner membrane during the host lysis or, potentially, to penetrate the outer membrane of the Gram-negative bacteria.
Collapse
|
38
|
Aghraz A, Benameur Q, Gervasi T, Ait Dra L, Ben-Mahdi MH, Larhsini M, Markouk M, Cicero N. Antibacterial activity of Cladanthus arabicus and Bubonium imbricatum essential oils alone and in combination with conventional antibiotics against Enterobacteriaceae isolates. Lett Appl Microbiol 2018; 67:175-182. [PMID: 29763981 DOI: 10.1111/lam.13007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 05/01/2018] [Accepted: 05/02/2018] [Indexed: 11/30/2022]
Abstract
Multidrug-resistant bacteria have become common all over the world, necessitating the development of new therapeutic strategies. Synergistic interactions between conventional antibiotics and natural bioactive may have therapeutic benefits in a clinical setting. There are plenty of medicinal plants that have proven efficacy against broad spectrum of micro-organisms. The aim of the work was to assess the antibacterial activity of Cladanthus arabicus, a Moroccan medicinal plant, and Bubonium imbricatum, a Moroccan endemic plant. The evaluation of the synergistic effect of extracted essential oils (EOs) together with some conventional antibiotics was also investigated. Checkerboard test was used to evaluate the interaction of EOs in combination with amoxicillin and neomycin. The results showed that EOs contain a potent activity against the tested Enterobacteriaceae isolates, with inhibition zones values in the range of 8·05 ± 0·1 and 13·1 ± 0·11 mm and MIC values between 200 μg ml-1 to 800 μg ml-1 for C. arabicus and from 400 μg ml-1 to 1600 μg ml-1 for B. imbricatum, respectively. Moreover, the current study allowed concluding that both EOs showed not only satisfactory antibacterial properties but also active effects combined with conventional antibiotics demonstrated by the Fractional Inhibitory Concentration Index (FICI). These findings are very interesting since there are no previous studies on synergistic interactions of these two plants with antibiotics. SIGNIFICANCE AND IMPACT OF THE STUDY The development of antibiotic resistance is multifactorial, including the specific nature of the relationship of bacteria to antibiotics. This situation has forced scientists to search for new antimicrobial substances from various sources as novel antimicrobial chemotherapeutic agents. Recently, medicinal plants and their derivatives (essential oils, extracts) have become very important in therapeutics because they encounter minimal challenges of the emergence of resistance. In this direction, the antimicrobial activity of the endemic Bubonium imbricatum plant and medicinal Cladanthus arabicus plant essential oils against multidrug-resistant Enterobacteriaceae strains was demonstrated.
Collapse
Affiliation(s)
- A Aghraz
- Laboratory of Biotechnology, Protection and Valorisation of Plant Resources (URAC35 Association Unit), Department of Biology, Faculty of Sciences, Cadi Ayyad University, Marrakesh, Morocco
| | - Q Benameur
- Research Laboratory "Animal Health and Production", Higher National Veterinary School, Algiers, Algeria.,Nursing Department, Faculty of Natural Sciences and Life, Abdelhamid Ibn Badis University, Mostaganem, Algeria
| | - T Gervasi
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - L Ait Dra
- Laboratory of Biotechnology, Protection and Valorisation of Plant Resources (URAC35 Association Unit), Department of Biology, Faculty of Sciences, Cadi Ayyad University, Marrakesh, Morocco
| | - M H Ben-Mahdi
- Research Laboratory "Animal Health and Production", Higher National Veterinary School, Algiers, Algeria
| | - M Larhsini
- Laboratory of Biotechnology, Protection and Valorisation of Plant Resources (URAC35 Association Unit), Department of Biology, Faculty of Sciences, Cadi Ayyad University, Marrakesh, Morocco
| | - M Markouk
- Laboratory of Biotechnology, Protection and Valorisation of Plant Resources (URAC35 Association Unit), Department of Biology, Faculty of Sciences, Cadi Ayyad University, Marrakesh, Morocco
| | - N Cicero
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy.,Science4Life, Spin Off Company, University of Messina, Messina, Italy
| |
Collapse
|
39
|
Ha E, Son B, Ryu S. Clostridium perfringens Virulent Bacteriophage CPS2 and Its Thermostable Endolysin LysCPS2. Viruses 2018; 10:v10050251. [PMID: 29751651 PMCID: PMC5977244 DOI: 10.3390/v10050251] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 05/10/2018] [Accepted: 05/11/2018] [Indexed: 11/30/2022] Open
Abstract
Clostridium perfringens is one of the most common causes of food-borne illness. The increasing prevalence of multidrug-resistant bacteria requires the development of alternatives to typical antimicrobial treatments. Here, we isolated and characterized a C. perfringens-specific virulent bacteriophage CPS2 from chicken feces. The CPS2 phage contains a 17,961 bp double-stranded DNA genome with 25 putative ORFs, and belongs to the Picovirinae, subfamily of Podoviridae. Bioinformatic analysis of the CPS2 genome revealed a putative endolysin, LysCPS2, which is homologous to the endolysin of Clostridium phage phiZP2 and phiCP7R. The enzyme showed strong lytic activity against C. perfringens with optimum conditions at pH 7.5–10, 25–65 °C, and over a broad range of NaCl concentrations. Interestingly, LysCPS2 was found to be highly thermostable, with up to 30% of its lytic activity remaining after 10 min of incubation at 95 °C. The cell wall binding domain in the C-terminal region of LysCPS2 showed a binding spectrum specific to C. perfringens strains. This is the first report to characterize highly thermostable endolysin isolated from virulent C. perfringens bacteriophage. The enzyme can be used as an alternative biocontrol and detection agent against C. perfringens.
Collapse
Affiliation(s)
- Eunsu Ha
- Department of Food and Animal Biotechnology, Seoul National University, Seoul 08826, Korea.
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea.
| | - Bokyung Son
- Department of Food and Animal Biotechnology, Seoul National University, Seoul 08826, Korea.
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea.
| | - Sangryeol Ryu
- Department of Food and Animal Biotechnology, Seoul National University, Seoul 08826, Korea.
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea.
| |
Collapse
|
40
|
Gerstmans H, Criel B, Briers Y. Synthetic biology of modular endolysins. Biotechnol Adv 2018; 36:624-640. [DOI: 10.1016/j.biotechadv.2017.12.009] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 12/08/2017] [Accepted: 12/13/2017] [Indexed: 01/15/2023]
|
41
|
Won G, Kim B, Lee JH. A novel method to generate Salmonella Typhi Ty21a ghosts exploiting the λ phage holin-endolysin system. Oncotarget 2018. [PMID: 28637001 PMCID: PMC5564637 DOI: 10.18632/oncotarget.18383] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Human typhoid fever caused by Salmonella Typhi still poses a severe global disease burden in developing countries despite the availability of commercial vaccines. In this study, we constructed a non-living S. Typhi Ty21a vaccine candidate by employing a lambda (λ) phage-derived holin-endolysin system to efficiently construct bacterial ghosts. The lysis plasmid pJHL464 harbors an R lysis cassette that is stringently regulated by dual promoters containing cI857/λPR and ParaBAD/araC components. The plasmid was introduced into an asd gene-deleted S. Typhi Ty21a strain designated JOL1675. The in vitro expression of endolysin (~17.76 kDa) in the subsequent JOL1675 vaccine construct when grown under lysis inducible conditions was validated by immunoblotting. In scanning electron microscopy analysis, surface transmembrane tunnels and a collapsed body were visualized in the ghosts. Following 48 h of lysis, no viable JOL1675 cells remained, indicating that lysis of all cells was achieved. Subcutaneous immunizations of mice with the JOL1675 ghosts produced significantly increasing titers of serum IgG and vaginal wash secretory IgA antibodies against JOL1675 outer membrane proteins during the observational period. Further, serum collected at 6 weeks post-immunization of rabbits exhibited effective bactericidal activity against wild type S. Typhi in the presence of complement. These data showed that JOL1675 ghosts are highly immunogenic and elicit humoral and mucosal responses expected to correlate with protective immunity against S. typhi. Collectively, our findings support the conclusion that incorporating a λ phage holin-endolysin-mediated lysis construct into S. Typhi is an efficient strategy for developing a novel and safe non-living typhoid vaccine candidate.
Collapse
Affiliation(s)
- Gayeon Won
- College of Veterinary Medicine, Chonbuk National University, Iksan Campus, Iksan, Republic of Korea
| | - Boram Kim
- College of Veterinary Medicine, Chonbuk National University, Iksan Campus, Iksan, Republic of Korea
| | - John Hwa Lee
- College of Veterinary Medicine, Chonbuk National University, Iksan Campus, Iksan, Republic of Korea
| |
Collapse
|
42
|
Maciejewska B, Olszak T, Drulis-Kawa Z. Applications of bacteriophages versus phage enzymes to combat and cure bacterial infections: an ambitious and also a realistic application? Appl Microbiol Biotechnol 2018; 102:2563-2581. [PMID: 29442169 PMCID: PMC5847195 DOI: 10.1007/s00253-018-8811-1] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 01/22/2018] [Accepted: 01/23/2018] [Indexed: 01/21/2023]
Abstract
Bacteriophages (phages) are viruses that infect bacteria. The "predator-prey" interactions are recognized as a potentially effective way to treat infections. Phages, as well as phage-derived proteins, especially enzymes, are intensively studied to become future alternative or supportive antibacterials used alone or in combination with standard antibiotic regimens treatment. There are many publications presenting phage therapy aspects, and some papers focused separately on the application of phage-derived enzymes. In this review, we discuss advantages and limitations of both agents concerning their specificity, mode of action, structural issues, resistance development, pharmacokinetics, product preparation, and interactions with the immune system. Finally, we describe the current regulations for phage-based product application.
Collapse
Affiliation(s)
- Barbara Maciejewska
- Department of Pathogen Biology and Immunology, Institute of Genetics and Microbiology, University of Wroclaw, S. Przybyszewskiego 63/77, 51-148, Wroclaw, Poland
| | - Tomasz Olszak
- Department of Pathogen Biology and Immunology, Institute of Genetics and Microbiology, University of Wroclaw, S. Przybyszewskiego 63/77, 51-148, Wroclaw, Poland
| | - Zuzanna Drulis-Kawa
- Department of Pathogen Biology and Immunology, Institute of Genetics and Microbiology, University of Wroclaw, S. Przybyszewskiego 63/77, 51-148, Wroclaw, Poland.
| |
Collapse
|
43
|
Love MJ, Bhandari D, Dobson RCJ, Billington C. Potential for Bacteriophage Endolysins to Supplement or Replace Antibiotics in Food Production and Clinical Care. Antibiotics (Basel) 2018; 7:E17. [PMID: 29495476 PMCID: PMC5872128 DOI: 10.3390/antibiotics7010017] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 02/06/2018] [Accepted: 02/23/2018] [Indexed: 01/21/2023] Open
Abstract
There is growing concern about the emergence of bacterial strains showing resistance to all classes of antibiotics commonly used in human medicine. Despite the broad range of available antibiotics, bacterial resistance has been identified for every antimicrobial drug developed to date. Alarmingly, there is also an increasing prevalence of multidrug-resistant bacterial strains, rendering some patients effectively untreatable. Therefore, there is an urgent need to develop alternatives to conventional antibiotics for use in the treatment of both humans and food-producing animals. Bacteriophage-encoded lytic enzymes (endolysins), which degrade the cell wall of the bacterial host to release progeny virions, are potential alternatives to antibiotics. Preliminary studies show that endolysins can disrupt the cell wall when applied exogenously, though this has so far proven more effective in Gram-positive bacteria compared with Gram-negative bacteria. Their potential for development is furthered by the prospect of bioengineering, and aided by the modular domain structure of many endolysins, which separates the binding and catalytic activities into distinct subunits. These subunits can be rearranged to create novel, chimeric enzymes with optimized functionality. Furthermore, there is evidence that the development of resistance to these enzymes may be more difficult compared with conventional antibiotics due to their targeting of highly conserved bonds.
Collapse
Affiliation(s)
- Michael J Love
- Biomolecular Interaction Centre and School of Biological Sciences, University of Canterbury, Christchurch 8041, New Zealand.
| | - Dinesh Bhandari
- Biomolecular Interaction Centre and School of Biological Sciences, University of Canterbury, Christchurch 8041, New Zealand.
- Institute of Environmental Science and Research, Christchurch 8041, New Zealand.
| | - Renwick C J Dobson
- Biomolecular Interaction Centre and School of Biological Sciences, University of Canterbury, Christchurch 8041, New Zealand.
- Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne 3052, Australia.
| | - Craig Billington
- Biomolecular Interaction Centre and School of Biological Sciences, University of Canterbury, Christchurch 8041, New Zealand.
- Institute of Environmental Science and Research, Christchurch 8041, New Zealand.
| |
Collapse
|
44
|
Gervasi T, Pellizzeri V, Benameur Q, Gervasi C, Santini A, Cicero N, Dugo G. Valorization of raw materials from agricultural industry for astaxanthin and β-carotene production by Xanthophyllomyces dendrorhous. Nat Prod Res 2017; 32:1554-1561. [DOI: 10.1080/14786419.2017.1385024] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Teresa Gervasi
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Polo Universitario Annunziata, Messina, Italy
| | - Vito Pellizzeri
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Polo Universitario Annunziata, Messina, Italy
| | - Qada Benameur
- Faculty of Natural Sciences and Life, Nursing Department, University of Mostaganem, Mostaganem, Algeria
| | - Claudio Gervasi
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Polo Universitario Annunziata, Messina, Italy
| | - Antonello Santini
- Dipartimento di Farmacia, Università di Napoli Federico II, Napoli, Italy
| | - Nicola Cicero
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Polo Universitario Annunziata, Messina, Italy
- Science4Life, Spin Off Company, University of Messina, Messina, Italy
| | - Giacomo Dugo
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Polo Universitario Annunziata, Messina, Italy
- Science4Life, Spin Off Company, University of Messina, Messina, Italy
| |
Collapse
|
45
|
Recent advances in therapeutic delivery systems of bacteriophage and bacteriophage-encoded endolysins. Ther Deliv 2017. [DOI: 10.4155/tde-2017-0040] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Antibiotics have been the cornerstone of clinical management of bacterial infection since their discovery in the early 20th century. However, their widespread and often indiscriminate use has now led to reports of multidrug resistance becoming globally commonplace. Bacteriophage therapy has undergone a recent revival in battle against pathogenic bacteria, as the self-replicating and co-evolutionary features of these predatory virions offer several advantages over conventional therapeutic agents. In particular, the use of targeted bacteriophage therapy from specialized delivery platforms has shown particular promise owing to the control of delivery location, administration conditions and dosage of the therapeutic cargo. This review presents an overview of the recent formulations and applications of such delivery vehicles as an innovative and elegant tool for bacterial control.
Collapse
|
46
|
Gervasi T, Pellizzeri V, Calabrese G, Di Bella G, Cicero N, Dugo G. Production of single cell protein (SCP) from food and agricultural waste by using Saccharomyces cerevisiae. Nat Prod Res 2017; 32:648-653. [DOI: 10.1080/14786419.2017.1332617] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Teresa Gervasi
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Vito Pellizzeri
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Giorgio Calabrese
- Department of Pharmacy, Università degli Studi di Napoli Federico II, Napoli, Italy
| | - Giuseppa Di Bella
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Nicola Cicero
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
- Department of Pharmacy, Università degli Studi di Napoli Federico II, Napoli, Italy
| | - Giacomo Dugo
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
- Science4Life, Spin Off Company, University of Messina, Messina, Italy
| |
Collapse
|
47
|
Multivalent Chromosomal Expression of the Clostridium botulinum Serotype A Neurotoxin Heavy-Chain Antigen and the Bacillus anthracis Protective Antigen in Lactobacillus acidophilus. Appl Environ Microbiol 2016; 82:6091-6101. [PMID: 27496774 PMCID: PMC5068166 DOI: 10.1128/aem.01533-16] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 07/28/2016] [Indexed: 02/02/2023] Open
Abstract
Clostridium botulinum and Bacillus anthracis produce potent toxins that cause severe disease in humans. New and improved vaccines are needed for both of these pathogens. For mucosal vaccine delivery using lactic acid bacteria, chromosomal expression of antigens is preferred over plasmid-based expression systems, as chromosomal expression circumvents plasmid instability and the need for antibiotic pressure. In this study, we constructed three strains of Lactobacillus acidophilus NCFM expressing from the chromosome (i) the nontoxic host receptor-binding domain of the heavy chain of Clostridium botulinum serotype A neurotoxin (BoNT/A-Hc), (ii) the anthrax protective antigen (PA), and (iii) both the BoNT/A-Hc and the PA. The BoNT/A-Hc vaccine cassette was engineered to contain the signal peptide from the S-layer protein A from L. acidophilus and a dendritic-cell-targeting peptide. A chromosomal region downstream of lba0889 carrying a highly expressed enolase gene was selected for insertion of the vaccine cassettes. Western blot analysis confirmed the heterologous expression of the two antigens from plasmid and chromosome locations. Stability assays demonstrated loss of the vaccine cassettes from expression plasmids without antibiotic maintenance. RNA sequencing showed high expression of each antigen and that insertion of the vaccine cassettes had little to no effect on the transcription of other genes in the chromosome. This study demonstrated that chromosomal integrative recombinant strains are promising vaccine delivery vehicles when targeted into high-expression chromosomal regions. Levels of expression match high-copy-number plasmids and eliminate the requirement for antibiotic selective maintenance of recombinant plasmids. IMPORTANCEClostridium botulinum and Bacillus anthracis produce potent neurotoxins that pose a biochemical warfare concern; therefore, effective vaccines against these bacteria are required. Chromosomal expression of antigens is preferred over plasmid-based expression systems since expressing antigens from a chromosomal location confers an advantage to the vaccine strains by eliminating the antibiotic maintenance required for plasmids and negates issues with plasmid instability that would result in loss of the antigen. Lactic acid bacteria, including Lactobacillus acidophilus, have shown potential for mucosal vaccine delivery, as L. acidophilus is bile and acid tolerant, allowing transit through the gastrointestinal tract where cells interact with host epithelial and immune cells, including dendritic cells. In this study, we successfully expressed C. botulinum and B. anthracis antigens in the probiotic L. acidophilus strain NCFM. Both antigens were highly expressed individually or in tandem from the chromosome of L. acidophilus.
Collapse
|
48
|
Davison M, Treangen TJ, Koren S, Pop M, Bhaya D. Diversity in a Polymicrobial Community Revealed by Analysis of Viromes, Endolysins and CRISPR Spacers. PLoS One 2016; 11:e0160574. [PMID: 27611571 PMCID: PMC5017753 DOI: 10.1371/journal.pone.0160574] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 07/21/2016] [Indexed: 12/13/2022] Open
Abstract
The polymicrobial biofilm communities in Mushroom and Octopus Spring in Yellowstone National Park (YNP) are well characterized, yet little is known about the phage populations. Dominant species, Synechococcus sp. JA-2-3B'a(2–13), Synechococcus sp. JA-3-3Ab, Chloroflexus sp. Y-400-fl, and Roseiflexus sp. RS-1, contain multiple CRISPR-Cas arrays, suggesting complex interactions with phage predators. To analyze phage populations from Octopus Spring biofilms, we sequenced a viral enriched fraction. To assemble and analyze phage metagenomic data, we developed a custom module, VIRITAS, implemented within the MetAMOS framework. This module bins contigs into groups based on tetranucleotide frequencies and CRISPR spacer-protospacer matching and ORF calling. Using this pipeline we were able to assemble phage sequences into contigs and bin them into three clusters that corroborated with their potential host range. The virome contained 52,348 predicted ORFs; some were clearly phage-like; 9319 ORFs had a recognizable Pfam domain while the rest were hypothetical. Of the recognized domains with CRISPR spacer matches, was the phage endolysin used by lytic phage to disrupt cells. Analysis of the endolysins present in the thermophilic cyanophage contigs revealed a subset of characterized endolysins as well as a Glyco_hydro_108 (PF05838) domain not previously associated with sequenced cyanophages. A search for CRISPR spacer matches to all identified phage endolysins demonstrated that a majority of endolysin domains were targets. This strategy provides a general way to link host and phage as endolysins are known to be widely distributed in bacteriophage. Endolysins can also provide information about host cell wall composition and have the additional potential to be used as targets for novel therapeutics.
Collapse
Affiliation(s)
- Michelle Davison
- Carnegie Institution for Science, Department of Plant Biology, Stanford, CA, 94305, United States of America
- Stanford University, Department of Biology, Stanford, CA, 94305, United States of America
- * E-mail: (MD); (DB)
| | - Todd J. Treangen
- Center for Bioinformatics and Computational Biology, Biomolecular Sciences Building, College Park, MD, 20742, United States of America
| | - Sergey Koren
- Center for Bioinformatics and Computational Biology, Biomolecular Sciences Building, College Park, MD, 20742, United States of America
| | - Mihai Pop
- Center for Bioinformatics and Computational Biology, Biomolecular Sciences Building, College Park, MD, 20742, United States of America
- Department of Computer Science, University of Maryland, College Park, MD, 20742, United States of America
| | - Devaki Bhaya
- Carnegie Institution for Science, Department of Plant Biology, Stanford, CA, 94305, United States of America
- Stanford University, Department of Biology, Stanford, CA, 94305, United States of America
- * E-mail: (MD); (DB)
| |
Collapse
|
49
|
Ajuebor J, McAuliffe O, O'Mahony J, Ross RP, Hill C, Coffey A. Bacteriophage endolysins and their applications. Sci Prog 2016; 99:183-199. [PMID: 28742472 PMCID: PMC10365499 DOI: 10.3184/003685016x14627913637705] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Endolysins (lysins) are bacteriophage-encoded enzymes that have evolved to degrade specific bonds within the bacterial cell wall. These enzymes represent a novel class of antibacterial agents against infectious pathogens, especially in light of multidrug-resistant bacteria, which have made antibiotic therapy increasingly redundant. Lysins have been used successfully to eliminate/control bacterial pathogens in various anatomical locations in mouse and other animal models. Engineering tactics have also been successfully applied to improve lysin function. This review discusses the structure and function of lysins. It highlights protein-engineering tactics utilised to improve lysin activity. It also reviews the applications of lysins towards food biopreservation, therapeutics, biofilm elimination and diagnostics.
Collapse
Affiliation(s)
| | | | - Jim O'Mahony
- Cork Institute of Technology (CIT) at the Department of Biological Sciences
| | - R. Paul Ross
- Dean of the College of Science Engineering and Food Science at University College Cork
| | - Colin Hill
- University College Cork and a Principal Investigator in the Alimentary Pharmabiotic Centre
| | - Aidan Coffey
- Cork Institute of Technology at the Department of Biological Sciences and Head of the BioExplore Research Centre
| |
Collapse
|
50
|
Dunne M, Leicht S, Krichel B, Mertens HDT, Thompson A, Krijgsveld J, Svergun DI, Gómez-Torres N, Garde S, Uetrecht C, Narbad A, Mayer MJ, Meijers R. Crystal Structure of the CTP1L Endolysin Reveals How Its Activity Is Regulated by a Secondary Translation Product. J Biol Chem 2016; 291:4882-93. [PMID: 26683375 PMCID: PMC4777826 DOI: 10.1074/jbc.m115.671172] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 12/16/2015] [Indexed: 11/18/2022] Open
Abstract
Bacteriophages produce endolysins, which lyse the bacterial host cell to release newly produced virions. The timing of lysis is regulated and is thought to involve the activation of a molecular switch. We present a crystal structure of the activated endolysin CTP1L that targets Clostridium tyrobutyricum, consisting of a complex between the full-length protein and an N-terminally truncated C-terminal cell wall binding domain (CBD). The truncated CBD is produced through an internal translation start site within the endolysin gene. Mutants affecting the internal translation site change the oligomeric state of the endolysin and reduce lytic activity. The activity can be modulated by reconstitution of the full-length endolysin-CBD complex with free CBD. The same oligomerization mechanism applies to the CD27L endolysin that targets Clostridium difficile and the CS74L endolysin that targets Clostridium sporogenes. When the CTP1L endolysin gene is introduced into the commensal bacterium Lactococcus lactis, the truncated CBD is also produced, showing that the alternative start codon can be used in other bacterial species. The identification of a translational switch affecting oligomerization presented here has implications for the design of effective endolysins for the treatment of bacterial infections.
Collapse
Affiliation(s)
- Matthew Dunne
- From the European Molecular Biology Laboratory, Notkestrasse 85, 22607 Hamburg, Germany
| | - Stefan Leicht
- the European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Boris Krichel
- the Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Martinistrasse 52, 20251 Hamburg, Germany
| | - Haydyn D T Mertens
- From the European Molecular Biology Laboratory, Notkestrasse 85, 22607 Hamburg, Germany
| | - Andrew Thompson
- the Synchrotron Soleil, L'Orme des Merisiers, BP 48, Saint Aubin, 91192 Gif sur Yvette, France
| | - Jeroen Krijgsveld
- the European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Dmitri I Svergun
- From the European Molecular Biology Laboratory, Notkestrasse 85, 22607 Hamburg, Germany
| | - Natalia Gómez-Torres
- the Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Departamento de Tecnología de Alimentos, Carretera de La Coruña km 7, 28040 Madrid, Spain
| | - Sonia Garde
- the Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Departamento de Tecnología de Alimentos, Carretera de La Coruña km 7, 28040 Madrid, Spain
| | - Charlotte Uetrecht
- the Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Martinistrasse 52, 20251 Hamburg, Germany, the European XFEL GmbH, Notkestrasse 85, 22607 Hamburg, Germany, and
| | - Arjan Narbad
- the Institute of Food Research, Colney, Norwich NR4 7UA, United Kingdom
| | - Melinda J Mayer
- the Institute of Food Research, Colney, Norwich NR4 7UA, United Kingdom
| | - Rob Meijers
- From the European Molecular Biology Laboratory, Notkestrasse 85, 22607 Hamburg, Germany,
| |
Collapse
|