1
|
Menon AR, Prest RJ, Tobin DM, Champion PA. Mycobacterium marinum as a model for understanding principles of mycobacterial pathogenesis. J Bacteriol 2025:e0004725. [PMID: 40304497 DOI: 10.1128/jb.00047-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025] Open
Abstract
Mycobacterium marinum is a fish pathogen that has become a powerful and well-established model that has accelerated our understanding of the mechanisms of mycobacterial disease. M. marinum is a versatile surrogate for understanding the closely related human pathogen M. tuberculosis, which causes tuberculosis in humans. M. marinum has defined key mechanisms of pathogenesis, both shared with M. tuberculosis and unique to this species. In this review, we discuss the discovery of M. marinum as an occasional human pathogen, the shared aspects of pathogenesis with M. tuberculosis, and how M. marinum has been exploited as a model to define the molecular mechanisms of mycobacterial pathogenesis across several phases of infection.
Collapse
Affiliation(s)
- Aruna R Menon
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Rebecca J Prest
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| | - David M Tobin
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Patricia A Champion
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
- Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, USA
| |
Collapse
|
2
|
Spencer EK, Miller CR, Bull JJ. Standardized methods for rearing a moth larva, Manduca sexta, in a laboratory setting. PLoS One 2025; 20:e0316776. [PMID: 40299873 PMCID: PMC12040098 DOI: 10.1371/journal.pone.0316776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 03/14/2025] [Indexed: 05/01/2025] Open
Abstract
The larval tobacco hornworm, Manduca sexta, has been used in a laboratory setting for physiological studies and for pathogen virulence studies. This moth offers a much larger size than the commonly used wax moth (Galleria mellonella), and it can thus be used for a greater variety of assays, such as repeated sampling of the same individual, growth measurements, and tissue sampling. Yet their occasional use in research has led to a minimally documented set of rearing methods. To facilitate further adoption of this insect model, we expanded on previously reported protocols and developed our own rearing methods, which we report here. Our protocol requires little specialized equipment, with a cost less than $100/month for the feeding and maintenance of a laboratory colony of about five hundred larvae of differing instar phases. The low cost generalized equipment and supplies, and the simplification of the standardized protocols allows for an easy entry point for rearing tobacco hornworm populations. We also describe a few methods that are relevant to the uses of these organisms as infection models.
Collapse
Affiliation(s)
- Emma K. Spencer
- Department of Biological Sciences, University of Idaho, Moscow, Idaho, United States of America
| | - Craig R. Miller
- Department of Biological Sciences, University of Idaho, Moscow, Idaho, United States of America
- Institute for Modeling Collaboration and Innovation, University of Idaho, Moscow, Idaho, United States of America
| | - James J. Bull
- Department of Biological Sciences, University of Idaho, Moscow, Idaho, United States of America
- Institute for Modeling Collaboration and Innovation, University of Idaho, Moscow, Idaho, United States of America
| |
Collapse
|
3
|
Yeh JC, Yang CH, Hazam PK, Selvaraj SP, Lin YY, Hsieh CY, Liou JW, Chen JY. Combination of antimicrobial peptide and clinical antibiotic shows enhanced potency toward Acinetobacter baumannii infection. Eur J Pharmacol 2025; 999:177683. [PMID: 40288555 DOI: 10.1016/j.ejphar.2025.177683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 02/17/2025] [Accepted: 04/24/2025] [Indexed: 04/29/2025]
Abstract
The continued development of novel antimicrobial treatment strategies is crucial for maintaining an effective therapeutic arsenal, and antimicrobial peptides (AMPs) exhibit promising activities against a wide range of pathogens. In this study, we tested the combined effects of an AMP, TP4-3, and meropenem on Acinetobacter baumannii, which is responsible for many severe infections and is associated with high rates of overall mortality and morbidity. This study aimed to develop an effective combination therapy for microbial infections. TP4-3 and meropenem were found to act synergistically toward Acinetobacter baumannii and exhibit an activity profile better than those of the individual compounds. TP4-3 is an antimicrobial peptide with proven activity, low toxicity and extended stability. This AMP was combined with meropenem and tested for efficacy against A. baumannii using a panel of in vitro and in vivo tests. The combination of TP4-3 and meropenem exhibited robust activity against Acinetobacter baumannii pathogens. In particular, the combined treatment demonstrated significant antibiofilm properties and a lower degree of induced resistance than meropenem alone. Additionally, the combination showed an excellent activity profile in in vivo studies. Thus, the combination of TP4-3 and meropenem appears to be an effective strategy to mitigate the detrimental consequences of infections caused by this clinically relevant pathogen. Since the combination of TP4-3 and meropenem displayed better activity than the individual compounds, this strategy of combining AMPs with clinical antibiotics may be suitable for development of clinical treatments targeting various microbial infections.
Collapse
Affiliation(s)
- Jih-Chao Yeh
- Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, 23-10 Dawen Rd., Jiaushi, Ilan, 262, Taiwan
| | - Chin-Hao Yang
- Department of Biomedical Sciences and Engineering, College of Medicine, Tzu Chi University, Hualien, 970, Taiwan
| | - Prakash Kishore Hazam
- Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, 23-10 Dawen Rd., Jiaushi, Ilan, 262, Taiwan
| | - Sanjay Prasad Selvaraj
- Molecular and Biological Agricultural Science Program, Taiwan International Graduate Program, Academia Sinica, Taipei, 11529, Taiwan; Graduate Institute of Biotechnology, National Chung Hsing University, Taichung, 402, Taiwan
| | - You-Ying Lin
- Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, 23-10 Dawen Rd., Jiaushi, Ilan, 262, Taiwan
| | - Chu-Yi Hsieh
- Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, 23-10 Dawen Rd., Jiaushi, Ilan, 262, Taiwan
| | - Je-Wen Liou
- Department of Biomedical Sciences and Engineering, College of Medicine, Tzu Chi University, Hualien, 970, Taiwan
| | - Jyh-Yih Chen
- Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, 23-10 Dawen Rd., Jiaushi, Ilan, 262, Taiwan; The IEGG and Animal Biotechnology Center and the Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, 402, Taiwan.
| |
Collapse
|
4
|
Xiao L, Long Y, Gao B, Hu Y, Zhang T, Xiao Q, Qin X, Xia X, Wang X, Li Z, Dong Q. Evaluation of the virulence characteristics of ST11 Salmonella enterica from different sources using a 2D cell model. Int J Food Microbiol 2025; 434:111151. [PMID: 40073551 DOI: 10.1016/j.ijfoodmicro.2025.111151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025]
Abstract
This study aimed to evaluate the virulence characteristics of ST11 Salmonella enterica from various sources and explore its pathogenic mechanisms and the molecular basis of antimicrobial resistance. In total, 20 Salmonella isolates collected between 2017 and 2022 from environmental, animal, clinical, and food sources were analyzed. Comprehensive investigations were conducted using whole-genome sequencing, bioinformatic analysis, broth microdilution methods, a two-dimensional (2D) cell model (Caco-2 cells), and a Galleria mellonella infection model. All tested ST11 strains carried major pathogenicity islands (PAIs) SPI-1salmonella pathogenicity island-1 (SPI-1) to SPI-5, and 90 % of the isolates harbored three or more plasmids, facilitating the horizontal transfer of virulence genes. Expression levels of sopA, ssaV, sipA/sspA, and sipB/sspB virulence genes varied significantly among strains, with sipB/sspB playing a key role in the invasion of ST11 strains. The results of invasion assays using the 2D cell model were consistent with those from the Galleria mellonella infection model, validating the 2D model's effectiveness in evaluating Salmonella's virulence. The findings suggest that Salmonella's virulence is not directly associated with the source of the isolates, and plasmid diversity may impact adaptability and transmission patterns. This study provides new insights into the pathogenic mechanisms of ST11 Salmonella and lays the groundwork for developing a novel 3D cell model to assess bacterial virulence.
Collapse
Affiliation(s)
- Linlin Xiao
- Department of Laboratory Medicine, Affiliated Sixth People's Hospital South Campus, Shanghai University of Medicine and Health Sciences, Shanghai 201499, China; School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Yulin Long
- Department of Laboratory Medicine, Affiliated Sixth People's Hospital South Campus, Shanghai University of Medicine and Health Sciences, Shanghai 201499, China
| | - Binru Gao
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Ying Hu
- Anhui University of Science and Technology School of Medicine, Anhui 232001, China
| | - Tantao Zhang
- Anhui University of Science and Technology School of Medicine, Anhui 232001, China
| | - Qian Xiao
- Department of Laboratory Medicine, Affiliated Sixth People's Hospital South Campus, Shanghai University of Medicine and Health Sciences, Shanghai 201499, China
| | - Xiaojie Qin
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Xuejuan Xia
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Xiang Wang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Zhuosi Li
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Qingli Dong
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China.
| |
Collapse
|
5
|
Opris RV, Baciu AM, Filip GA, Florea A, Costache C. The use of Galleria mellonella in metal nanoparticle development: A systematic review. Chem Biol Interact 2025; 415:111511. [PMID: 40246051 DOI: 10.1016/j.cbi.2025.111511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/17/2025] [Accepted: 04/10/2025] [Indexed: 04/19/2025]
Abstract
Research on metal nanoparticles is crucial for their application in diverse fields, requiring detailed assessments of their effects and potential. Galleria mellonella larvae have emerged as a valuable model for studying the impacts of metal nanoparticles, offering ethical and logistical advantages over traditional models. This systematic review synthesizes evidence on the application of Galleria mellonella in evaluating the toxicity, distribution, and therapeutic potential of metal nanoparticles. Adhering to PRISMA guidelines, a comprehensive database search (MEDLINE, Embase, Cochrane, Scopus, Google Scholar, Science Citation Index Expanded) was conducted using keywords related to Galleria mellonella and metal nanoparticles. The SYRCLE's risk of bias tool (adapted for G. mellonella) was used for risk of bias assessment. Out of 1696 initially identified studies, 31 met the inclusion criteria, encompassing research from 2011 to 2024. The included studies effectively demonstrate G. mellonella's capacity to model the toxicity of metal nanoparticles, their therapeutic potential in treating infections, and the impact on the innate immune response, bridging the gap between simpler in vitro assays and more complex mammalian models. Galleria mellonella stands out as a critical model for the early-stage development and evaluation of metal nanoparticles, particularly in assessing toxicity, therapeutic efficacy in infection treatment, and interaction with immune systems. This review underscores the larvae's role in metal nanoparticle research, advocating for its broader use to streamline development processes while minimizing ethical concerns.
Collapse
Affiliation(s)
- Razvan Vlad Opris
- Department of Cell & Molecular Biology, "Iuliu Hatieganu" University of Medicine & Pharmacy, 6 Louis Pasteur Street, 400349, Cluj-Napoca, Romania; Department of Microbiology, "Iuliu Hatieganu" University of Medicine & Pharmacy, 6 Louis Pasteur Street, 400349, Cluj-Napoca, Romania.
| | - Alina Mihaela Baciu
- Department of Cell & Molecular Biology, "Iuliu Hatieganu" University of Medicine & Pharmacy, 6 Louis Pasteur Street, 400349, Cluj-Napoca, Romania; Department of Microbiology, "Iuliu Hatieganu" University of Medicine & Pharmacy, 6 Louis Pasteur Street, 400349, Cluj-Napoca, Romania.
| | - Gabriela Adriana Filip
- Department of Physiology, "Iuliu Hatieganu" University of Medicine & Pharmacy, 1-3 Clinicilor Street, 400006, Cluj-Napoca, Romania.
| | - Adrian Florea
- Department of Cell & Molecular Biology, "Iuliu Hatieganu" University of Medicine & Pharmacy, 6 Louis Pasteur Street, 400349, Cluj-Napoca, Romania.
| | - Carmen Costache
- Department of Microbiology, "Iuliu Hatieganu" University of Medicine & Pharmacy, 6 Louis Pasteur Street, 400349, Cluj-Napoca, Romania.
| |
Collapse
|
6
|
Prado A, de Souza Costa P, de Oliveira Brito R, Ferreira DG, Caris GAY, Negri M, Svidzinski TIE. Enhanced virulence of mixed-species Candida biofilms isolated from intragastric balloon patient: insights from larval model. Braz J Microbiol 2025:10.1007/s42770-025-01662-5. [PMID: 40210812 DOI: 10.1007/s42770-025-01662-5] [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: 01/10/2025] [Accepted: 03/18/2025] [Indexed: 04/12/2025] Open
Abstract
Candida species, particularly Candida albicans and C. tropicalis, are critical members of the human microbiota and are associated with systemic infections, including candidaemia. The pathogenicity is largely attributed to virulence factors such as biofilm formation, which enhances antifungal resistance and immune evasion. Despite extensive research on single-species biofilms, the dynamics of polymicrobial biofilms, especially those involving fungal‒fungal interactions, remain poorly understood. This study investigated the virulence of monomicrobial and polymicrobial biofilms of C. albicans and C. tropicalis formed in vitro, using Zophobas morio larvae as a model. Biofilms were formed from C. albicans and C. tropicalis obtained from the gastric mucosa of a patient with an intragastric balloon (IGB). The biomass and structure of the monomicrobial and mixed-species biofilms were characterized via crystal violet staining and fluorescence microscopy techniques. The virulence of suspended, adhered, and planktonic Z. morio larvae was evaluated via survival assays, monitored over 10 days. C. albicans single biofilms presented greater biomass and structural organization than C. tropicalis, while mixed-species biofilms produced the highest biomass and density. Fluorescence microscopy revealed enhanced interspecies interactions in mixed biofilms, suggesting synergistic effects. Yeasts from single biofilms impacted less on survival rates, particularly under suspended and adhered cell conditions. These findings suggest that mixed-species biofilms exhibit increased virulence due to synergistic interactions between both species. Moreover, they also suggest distinct functional roles within biofilms, where C. tropicalis contributes to cellular proliferation and C. albicans supports matrix production, collectively enhancing biofilm robustness and pathogenic potential. This study underscores the pathogenic significance of fungal‒fungal interactions in biofilms, particularly under mixed-species conditions. The enhanced virulence observed in mixed biofilms highlights the importance of targeting interspecies dynamics in antifungal strategies. The use of alternative models such as Z. morio larvae provides valuable insights into biofilm-mediated infections and potential therapeutic interventions.
Collapse
Affiliation(s)
- Andressa Prado
- Department of Clinical Analysis and Biomedicine, Laboratory of Medical Mycology, State University of Maringá, Avenida Colombo, 5790- Bloco T20, Sala 203, Maringá, Paraná, 87020-900, Brazil
| | - Polyana de Souza Costa
- Department of Clinical Analysis and Biomedicine, Laboratory of Medical Mycology, State University of Maringá, Avenida Colombo, 5790- Bloco T20, Sala 203, Maringá, Paraná, 87020-900, Brazil
| | - Rubens de Oliveira Brito
- Department of Diagnostic and Therapeutic Endoscopy, Mgastro Digestive Tract Medical Center, Maringá, Paraná, Brazil
| | - Deisiany Gomes Ferreira
- Department of Clinical Analysis and Biomedicine, Laboratory of Medical Mycology, State University of Maringá, Avenida Colombo, 5790- Bloco T20, Sala 203, Maringá, Paraná, 87020-900, Brazil
| | | | - Melyssa Negri
- Department of Clinical Analysis and Biomedicine, Laboratory of Medical Mycology, State University of Maringá, Avenida Colombo, 5790- Bloco T20, Sala 203, Maringá, Paraná, 87020-900, Brazil
| | - Terezinha Inez Estivalet Svidzinski
- Department of Clinical Analysis and Biomedicine, Laboratory of Medical Mycology, State University of Maringá, Avenida Colombo, 5790- Bloco T20, Sala 203, Maringá, Paraná, 87020-900, Brazil.
| |
Collapse
|
7
|
Quispe-Villegas G, Alcántara-Lozano GI, Cuicapuza D, Laureano R, Ayzanoa B, Tsukayama P, Tamariz J. In vivo evaluation of phage therapy against Klebsiella pneumoniae using the Galleria mellonella model and molecular characterization of a novel Drulisvirus phage species. Microbiol Spectr 2025:e0114524. [PMID: 40202337 DOI: 10.1128/spectrum.01145-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 03/07/2025] [Indexed: 04/10/2025] Open
Abstract
Multidrug-resistant (MDR) Klebsiella pneumoniae is challenging to treat with conventional antibiotic regimens, posing a threat to healthcare systems. Phage therapy presents a promising alternative treatment strategy; however, characterization of its efficacy and safety is required. Here, we describe the microbiological and molecular characterization of a novel bacteriophage with activity against MDR K. pneumoniae using a greater wax moth (Galleria mellonella) model system. A bacteriophage was isolated from hospital wastewater. Viral kinetics and phage stability were evaluated under varied pH and temperature conditions. The therapeutic efficacy of the phage was evaluated using MDR Klebsiella-infected G. mellonella larvae as an in vivo model. Phage titers and larva survival were compared in phage-treated and control groups. Genomic sequencing (Nanopore and Illumina) was used to classify the bacteriophage and identify any resistance genes or virulence factors present in its genome. Functional characterization demonstrated effective lytic activity, favorable burst size (161 PFU/cell), and an optimal MOI of 0.1. The phage demonstrated stability across a wide range of temperatures (8°C-40°C) and pH levels (4-8). Experiments using the G. mellonella model showed improved larval survival with phage treatment. The novel bacteriophage was identified as a new species within the genus Drulisvirus with no lysogeny-associated, antimicrobial resistance, or virulence genes detected. The new Drulisvirus phage identified is a promising candidate for treatment of infections caused by MDR K. pneumoniae.IMPORTANCEThe study describes a bacteriophage with potential for use in phage therapy against Klebsiella pneumoniae, one of the most clinically significant bacterial pathogens today. Microbiological and genomic characterization of the phage revealed advantageous properties for therapeutic applications, while also identifying a novel species within the Drulisvirus genus. These findings significantly contribute to our understanding of bacteriophage diversity and their utility in combating antibiotic-resistant infections. Moreover, the authors developed an in vivo preclinical model of MDR infection using Galleria mellonella larvae and successfully applied it to study the bacteriophage's therapeutic efficacy. This model offers a robust and efficient platform for preclinical testing.
Collapse
Affiliation(s)
- Gustavo Quispe-Villegas
- Laboratorio de Resistencia Antibiótica y Fagoterapia, Laboratorios de Investigación y Desarrollo, Facultad de Ciencias e Ingeniería, Universidad Peruana Cayetano Heredia, Lima, Peru
- Facultad de Medicina, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Gabriela I Alcántara-Lozano
- Laboratorio de Resistencia Antibiótica y Fagoterapia, Laboratorios de Investigación y Desarrollo, Facultad de Ciencias e Ingeniería, Universidad Peruana Cayetano Heredia, Lima, Peru
- Facultad de Medicina, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Diego Cuicapuza
- Laboratorio de Resistencia Antibiótica y Fagoterapia, Laboratorios de Investigación y Desarrollo, Facultad de Ciencias e Ingeniería, Universidad Peruana Cayetano Heredia, Lima, Peru
- Facultad de Medicina, Universidad Peruana Cayetano Heredia, Lima, Peru
- Laboratorio de Genómica Microbiana, Laboratorios de Investigación y Desarrollo, Facultad de Ciencias e Ingeniería, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Raúl Laureano
- Laboratorio de Resistencia Antibiótica y Fagoterapia, Laboratorios de Investigación y Desarrollo, Facultad de Ciencias e Ingeniería, Universidad Peruana Cayetano Heredia, Lima, Peru
- Laboratorio de Moléculas Individuales, Laboratorios de Investigación y Desarrollo, Facultad de Ciencias e Ingeniería, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Brenda Ayzanoa
- Laboratorio de Genómica Microbiana, Laboratorios de Investigación y Desarrollo, Facultad de Ciencias e Ingeniería, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Pablo Tsukayama
- Laboratorio de Genómica Microbiana, Laboratorios de Investigación y Desarrollo, Facultad de Ciencias e Ingeniería, Universidad Peruana Cayetano Heredia, Lima, Peru
- Instituto de Medicina Tropical Alexander von Humboldt, Universidad Peruana Cayetano Heredia, Lima, Peru
- Parasites and Microbes Programme, Wellcome Sanger Institute, Hinxton, United Kingdom
| | - Jesús Tamariz
- Laboratorio de Resistencia Antibiótica y Fagoterapia, Laboratorios de Investigación y Desarrollo, Facultad de Ciencias e Ingeniería, Universidad Peruana Cayetano Heredia, Lima, Peru
- Facultad de Medicina, Universidad Peruana Cayetano Heredia, Lima, Peru
| |
Collapse
|
8
|
Harshitha N, More SS, Mitra SD. Development of a lytic bacteriophage BPK01 impregnated biopolymer (chitosan) hydrogel for combating high-risk strains of carbapenem resistant Klebsiella pneumoniae (CRKP) pathogens- in vitro and in vivo evaluation. Int J Biol Macromol 2025; 304:140887. [PMID: 39947562 DOI: 10.1016/j.ijbiomac.2025.140887] [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: 09/15/2024] [Revised: 01/13/2025] [Accepted: 02/09/2025] [Indexed: 02/24/2025]
Abstract
Alternative strategies are urgently required to combat the rise of high-risk carbapenem-resistant Klebsiella pneumoniae (CRKP), including blaNDM-positive strains that produce carbapenemase enzymes, which deactivate beta-lactam antibiotics and result in poor treatment outcomes. In this study, we isolated a bacteriophage BPK01, targeting a high-risk strain of Klebsiella pneumoniae (carbapenem-resistant, blaNDM-positive, ST147, capsular type K64, biofilm former). BPK01 demonstrated strong lytic activity (84%) against a panel of genetically characterized CRKP strains (n = 59) from clinical specimens, including pus, urine, sputum, blood, and tracheal aspirates. BPK01 was classified as a Caudoviricetes phage, exhibiting a burst size of 220 virions and a short latent period of 10 min. It demonstrated stability across a range of conditions (temperature, pH, and organic solvents) and effectively disrupted biofilms on silicone catheters. In vivo, BPK01 improved survival rates in the Galleria mellonella infection model and reduced bacterial burden in a murine bacteremia model, underscoring its therapeutic potential. Subsequently, we developed a hydrogel by incorporating BPK01 into a chitosan biopolymer, which demonstrated efficient lytic activity (spot assay, scanning electron microscopy, time kill assay) against CRKP pathogens, stability of biological activity for 6 months of storage, and controlled release kinetics, with the mathematical model Korsmeyer - Peppas being the best fit (R2 = 0.9962). The hydrogel expedited the healing of CRKP-infected lesions in a murine model, suggesting its potential as an effective topical treatment. This study highlights BPK01 as a promising biotherapeutic candidate for treating CRKP infection, with the phage hydrogel offering an ecofriendly and sustainable solution for treating infected lesions. Further research could expand its use in phage cocktails and other formulations for broader CRKP infection management.
Collapse
Affiliation(s)
- N Harshitha
- Department of Biotechnology, School of Basic and Applied Sciences, Dayananda Sagar University, Bangalore 560068, India
| | - Sunil S More
- Department of Biotechnology, School of Basic and Applied Sciences, Dayananda Sagar University, Bangalore 560068, India
| | - Susweta Das Mitra
- Department of Biotechnology, School of Basic and Applied Sciences, Dayananda Sagar University, Bangalore 560068, India.
| |
Collapse
|
9
|
Anower MR, Dennis E, Chaturvedi S, Chaturvedi V. Candida auris isolates from New York outbreak are highly pathogenic with measurable experimental disease in Galleria mellonella. Microbiol Spectr 2025; 13:e0294223. [PMID: 39912700 DOI: 10.1128/spectrum.02942-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 10/14/2024] [Indexed: 02/07/2025] Open
Abstract
Candida auris causes prolonged colonization and bloodstream infections in hospitalized patients. Different C. auris clades vary in their geographic origin, disease spectrum, and antifungal resistance, but biological basis underlying such variations needs further examination. Therefore, we investigated susceptible and multidrug-resistant C. auris isolates, obtained early in the New York outbreak, to benchmark their pathogenic potential in caterpillar worms of greater wax moth Galleria mellonella. Healthy G. mellonella worms responded to C. auris in a strain-specific, dose-responsive pattern. Three drug-resistant, clade I C. auris 17-1, 18-1, and 18-2 isolates caused high mortality, while a control, drug-susceptible, clade II C. auris 16-1 caused lower mortality (P < 0.001) with graded inocula (1 × 104 to 1 × 107 CFU). Virulence correlated with fungal growth in hemolymph as C. auris 17-1, 18-1 and 18-2 reached high fungal cell load in the infected larvae, while C. auris 16-1 multiplied less proficiently. The degree of melanization was higher in worms infected with more pathogenic C. auris. Hemocoel histopathology showed more fungal elements and about 50 granulomas per profile for C. auris 17-1, 18-1 and 18-2 compared to 5 to 10 granulomas for C. auris 16-1. With more pathogenic C. auris isolates, the expression of gallerimycin, ceropin, and galiomycin increased significantly (P < 0.05). Thus, the induction of G. mellonella immune effector peptides, histopathological responses, and melanization are proportionate to C. auris pathogenic potential. However, G. mellonella from different vendors showed unpredictable quality upon delivery that impacted feasibility and reproducibility of planned studies. Our results indicate utility of G. mellonella experimental model for screening of C. auris pathogenesis depending upon quality of worms commercially available in the USA.IMPORTANCEThe New York metropolitan area continues to suffer from the largest, ongoing outbreak of drug-resistant Candida auris. It is necessary to gather more information to determine if C. auris isolates from this outbreak vary in their ability to cause more severe disease in affected patients. Therefore, we studied the experimental model of greater wax moth Galleria mellonella. We found that susceptible and drug-resistant NY C. auris isolates caused measurable disease in G. mellonella. There was a good correlation between pathogenic potential of C. auris and melanin in the infected worms, fungal elements in worm hemolymph, uptick in the genes involved in insect immunity, and histopathological changes. The worms sourced from various commercial vendors were of variable quality. We confirm that G. mellonella remains a facile model to study C. auris experimental disease.
Collapse
Affiliation(s)
- Md Rokebul Anower
- Mycology Laboratory, Wadsworth Center, New York State Department of Health, Albany, New York, USA
| | - Emily Dennis
- Mycology Laboratory, Wadsworth Center, New York State Department of Health, Albany, New York, USA
| | - Sudha Chaturvedi
- Mycology Laboratory, Wadsworth Center, New York State Department of Health, Albany, New York, USA
- Department of Biomedical Sciences, School of Public Health, University at Albany, Albany, New York, USA
| | - Vishnu Chaturvedi
- Mycology Laboratory, Wadsworth Center, New York State Department of Health, Albany, New York, USA
| |
Collapse
|
10
|
Oladipo EK, Adeyemo SF, Oluwasanya GJ, Adaramola EO, Akintola SB, Afolabi VO, Ajagbe JD, Ojo OH, Kolapo EP, Owoeye E, Jimah EM, Ayeleso AO, Onyeaka H. Novel antibacterial agents and emerging therapies in the fight against multidrug-resistant Acinetobacter baumannii. Microb Pathog 2025; 200:107361. [PMID: 39894233 DOI: 10.1016/j.micpath.2025.107361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/27/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
Acinetobacter baumannii, a multidrug-resistant pathogen, poses a critical challenge in healthcare settings due to its adaptability and limited treatment options. The global rise in antimicrobial resistance (AMR) has underscored the urgent need for novel therapeutic strategies to combat infections caused by extensively drug-resistant (EDR) and pan-drug-resistant (PDR) A. baumannii. Traditional antibiotic discovery methods, such as whole-cell screening, have fallen short, consistently identifying drugs prone to resistance. This review explores the discovery of new anti-bacterial agents targeting A. baumannii, focusing on emerging therapeutic approaches, including nanoparticle-based therapies, antimicrobial peptides, and antibiotic combination therapies. Nanoparticle-based approaches, leveraging enhanced penetration and multi-mechanistic action, show promise in overcoming resistance, though challenges such as toxicity and biocompatibility persist. Additionally, combination therapies, such as polymyxins with carbapenems, have demonstrated efficacy in clinical settings. This review also highlights the limitations of current therapies, the mechanisms of bacterial resistance, and the role of alternative strategies like bacteriophage therapy. Emphasis is placed on the need for further research into overcoming cross-resistance and enhancing therapeutic efficacy against A. baumannii. The review concludes by discussing the importance of advancing research into novel agents, optimizing dosage strategies, and addressing the challenges posed by toxicity to ensure the effective treatment of A. baumannii infections in both hospital and community settings.
Collapse
Affiliation(s)
- Elijah Kolawole Oladipo
- Division of Pharmacotherapies Design and Development, Helix Biogen Institute, Ogbomoso, Oyo State, Nigeria; Division of Vaccine Design and Development, Helix Biogen Institute, Ogbomoso, Oyo State, Nigeria; Department of Microbiology, Laboratory of Molecular Biology, Immunology and Bioinformatics, Adeleke University, Ede, Osun State, Nigeria; Department of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B12 2TT, UK.
| | - Stephen Feranmi Adeyemo
- Division of Vaccine Design and Development, Helix Biogen Institute, Ogbomoso, Oyo State, Nigeria.
| | - Glory Jesudara Oluwasanya
- Division of Pharmacotherapies Design and Development, Helix Biogen Institute, Ogbomoso, Oyo State, Nigeria
| | | | - Shalom Busayo Akintola
- Division of Vaccine Design and Development, Helix Biogen Institute, Ogbomoso, Oyo State, Nigeria; Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Victor Oluwatobi Afolabi
- Division of Vaccine Design and Development, Helix Biogen Institute, Ogbomoso, Oyo State, Nigeria; Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Joel Damilare Ajagbe
- Division of Vaccine Design and Development, Helix Biogen Institute, Ogbomoso, Oyo State, Nigeria; Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Oluwadara Hannah Ojo
- Division of Pharmacotherapies Design and Development, Helix Biogen Institute, Ogbomoso, Oyo State, Nigeria; Department of Pure and Applied Biology, Ladoke Akintola University of Technology, Ogbomoso, Oyo state, Nigeria
| | - Emmanuel Pelumi Kolapo
- Division of Vaccine Design and Development, Helix Biogen Institute, Ogbomoso, Oyo State, Nigeria
| | - Emmanuel Owoeye
- Division of Pharmacotherapies Design and Development, Helix Biogen Institute, Ogbomoso, Oyo State, Nigeria
| | - Esther Moradeyo Jimah
- Division of Pharmacotherapies Design and Development, Helix Biogen Institute, Ogbomoso, Oyo State, Nigeria
| | - Ademola O Ayeleso
- Biochemistry Programme, Bowen University, Iwo PMB 284, Osun State, Nigeria; Department of Life and Consumer Sciences, University of South Africa, Florida Park 1709, Roodepoort, South Africa
| | - Helen Onyeaka
- Department of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B12 2TT, UK
| |
Collapse
|
11
|
de Cena GL, Tada DB, Lucchi DB, Santos TA, Heras M, Juliano M, Torres Braconi C, Castanho MA, Lopes-Ferreira M, Conceição K. Design of Natterins-based peptides improves antimicrobial and antiviral activities. BIOTECHNOLOGY REPORTS (AMSTERDAM, NETHERLANDS) 2025; 45:e00867. [PMID: 39758971 PMCID: PMC11697409 DOI: 10.1016/j.btre.2024.e00867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/08/2024] [Accepted: 11/26/2024] [Indexed: 01/07/2025]
Abstract
The biochemical analysis of animal venoms has been intensifying over the years, enabling the prediction of new molecules derived from toxins, harnessing the therapeutic potential of these molecules. From the venom of the fish Thalassophryne nattereri, using in silico methods for predicting antimicrobial and cell-penetrating peptides, two peptides from Natterins with promising characteristics were synthesized and subjected to in vitro and in vivo analysis. The peptides were subjected to stability tests and antimicrobial assays, cytotoxicity in murine fibroblast cells, antiviral assays against the Chikungunya virus, and the toxicity on G. mellonella was also evaluated. The findings underscore the peptides' robust stability under varying temperatures and pH conditions and resistance to proteolytic degradation. The peptides demonstrated significant antimicrobial efficacy, minimal cytotoxicity, and low hemolytic activity. Although their antiviral efficacy was limited, they showed potential at specific stages of viral replication. The in vivo toxicity tests indicated a favorable safety profile. These findings suggest that this approach can aid in the development of antimicrobial agents, offering a faster and personalized method to combat microbial infections, and represent a promising discovery in venom biotechnology research.
Collapse
Affiliation(s)
- Gabrielle L. de Cena
- Laboratory of Peptide Biochemistry, Universidade Federal de São Paulo (UNIFESP), São José dos Campos, Brazil
| | - Dayane B. Tada
- Laboratory of Nanomaterials and Nanotoxicology, Universidade Federal de São Paulo (UNIFESP), São José dos Campos, Brazil
| | - Danilo B.M. Lucchi
- Department of Microbiology, Immunology and Parasitology, Escola Paulista de Medicina (UNIFESP), São Paulo, Brazil
| | - Tiago A.A. Santos
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Montserrat Heras
- Departament de Química, Universitat de Girona, Campus Montilivi, 17071 Girona, Spain
| | - Maria Juliano
- Department of Biophysics, Escola Paulista de Medicina (UNIFESP), São Paulo, Brazil
| | - Carla Torres Braconi
- Department of Microbiology, Immunology and Parasitology, Escola Paulista de Medicina (UNIFESP), São Paulo, Brazil
| | - Miguel A.R.B. Castanho
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Mônica Lopes-Ferreira
- Immunoregulation Unit, Laboratory of Applied Toxinology (CeTICs/FAPESP), Butantan Institute, São Paulo 05503900, Brazil
| | - Katia Conceição
- Laboratory of Peptide Biochemistry, Universidade Federal de São Paulo (UNIFESP), São José dos Campos, Brazil
| |
Collapse
|
12
|
Kaczmarek A, Boguś MI. The flavouring agent, 2-octenoic acid kills Galleria mellonella (Lepidoptera: Pyralidae) by affecting their immunocompetent cells and cuticular FFA profiles. JOURNAL OF INSECT PHYSIOLOGY 2025; 161:104779. [PMID: 39947514 DOI: 10.1016/j.jinsphys.2025.104779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/06/2025] [Accepted: 02/10/2025] [Indexed: 02/16/2025]
Abstract
This study investigates the effects of the naturally occurring flavouring agent, trans-2-octenoic acid, on the insect model Galleria mellonella by examining its impact on immunocompetent cells and free fatty acid (FFA) profiles in the cuticle. The value of LD50 for 2-octenoic acid has been calculated as 9.66 µg/mg of insect body mass, this value is outside the GHS scale, indicating that the compound is unlikely to cause acute toxicity after dermal application and is safe for humans and mammals. Thetreatment with 2-octenoic acid caused several changes in the insect defence mechanismes, viz. changes in cuticular FFA profiles and death of immunocompetent cells. In larvae, topical treatment of 2-octenoic acid increased the concentration of cuticular FFAs, particularly C6:0 (245 times higher), C15:0 (110 times higher), and C16:1 (1608 times higher), and 2-octenoic acid (C8:1) accumulated significantly on the surface of the cuticle. In adults, treatment resulted in lower cuticular C8:1concentrations than in larvae, which might indicate that 2-octenoic acid penetrates more effectively through the adult cuticle. The 2-octenoic acid application demonstrated considerable cytotoxicity against insect cell line Sf9 and G. mellonella hemocytes, with both in vivo and in vitro treatment. Our findings contribute to the broader understanding of how synthetic and naturally occurring chemicals may interact with the immune and physiological systems of insects, particularly focusing on G. mellonella as a model organism for toxicological studies. Given the increasing interest in the ecological and physiological impacts of food additives, our research provides novel insights into the biological interactions of 2-octenoic acid and its potential role as an insecticide.
Collapse
Affiliation(s)
- Agata Kaczmarek
- Museum and Institute of Zoology PAS, Twarda 51/55 00-818, Warsaw, Poland.
| | - Mieczysława Irena Boguś
- Museum and Institute of Zoology PAS, Twarda 51/55 00-818, Warsaw, Poland; Biomibo, Strzygłowska 15 04-872, Warsaw, Poland
| |
Collapse
|
13
|
Martinez MMB, Corleto M, Weschenfeller M, Urrea Montes S, Salomón CN, Gonzalez N, Garavaglia M, Faccone D, Maffía PC. In Vitro and In Vivo Evaluation of the De Novo Designed Antimicrobial Peptide P6.2 Against a KPC-Producing P. aeruginosa Clinical Isolate. Biomolecules 2025; 15:339. [PMID: 40149875 PMCID: PMC11940343 DOI: 10.3390/biom15030339] [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: 01/24/2025] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 03/29/2025] Open
Abstract
The antimicrobial peptide P6.2 was previously de novo designed as an alpha helix cationic amphipathic molecule. In previous work, we have shown that this peptide displayed significant antimicrobial activity against both Gram-positive (Staphylococcus aureus) and Gram-negative (Pseudomonas aeruginosa) bacteria. However, while P6.2 lacked biofilm-inhibiting properties against the P. aeruginosa strain PA01, it displayed anti-inflammatory effects in a murine acute lung infection model challenged with this pathogen. In this work, the peptide P6.2 antimicrobial activity and its possible synergy with meropenem were evaluated both in vitro and in vivo using a Galleria mellonella infection model against a carbapenem-resistant KPC-producing clinical isolate of P. aeruginosa. Firstly, the cytotoxic effect of the peptide on A549 and RAW264.7 cell lines was assayed, showing no cytotoxicity at 64 µg/mL and below. Then, the MIC (minimal inhibitory concentration) and bactericidal effect against the carbapenemase-producing strain P. aeruginosa M13513 strain were determined. P6.2 showed a MIC between 32 and 64 µg/mL, and a rapid bactericidal activity against this strain (less than 45 min). The peptide stability at different temperatures and in bovine serum at 37 °C was also analyzed, showing good stability and almost no degradation after 15 min of incubation at 100 °C or 24 h at 37 °C in serum, respectively. The antibiofilm activity was also evaluated, and although the peptide did not show biofilm inhibitory activity, it did demonstrate biofilm disruptive activity, together with bactericidal activity inside the pre-formed biofilm. The possible synergistic effect with the carbapenem meropenem was then analyzed in vitro by killing kinetics, revealing a synergistic interaction between P6.2 and the antibiotic against this strain. Finally, P6.2 was evaluated in vivo in the Galleria mellonella larvae infection model. Interestingly, in G. mellonella, P6.2 alone did not completely clear the infection caused by P. aeruginosa M13513. However, when combined with meropenem, P6.2 demonstrated a synergistic effect, leading to increased survival rates in infected larvae. The results presented here highlight the potential that this peptide displays when used in combination with carbapenems against a clinically relevant KPC-producing P. aeruginosa.
Collapse
Affiliation(s)
- Melina M. B. Martinez
- Laboratorio de Aplicaciones Biotecnológicas y Microbiología (LAByM), Secretaría de Investigación, Universidad Nacional de Hurlingham (UNAHUR), Hurlingham 1688, Buenos Aires, Argentina; (M.M.B.M.); (M.C.); (M.W.); (S.U.M.); (C.N.S.); (N.G.); (M.G.)
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires 1425, Argentina;
| | - Merlina Corleto
- Laboratorio de Aplicaciones Biotecnológicas y Microbiología (LAByM), Secretaría de Investigación, Universidad Nacional de Hurlingham (UNAHUR), Hurlingham 1688, Buenos Aires, Argentina; (M.M.B.M.); (M.C.); (M.W.); (S.U.M.); (C.N.S.); (N.G.); (M.G.)
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires 1425, Argentina;
| | - Melanie Weschenfeller
- Laboratorio de Aplicaciones Biotecnológicas y Microbiología (LAByM), Secretaría de Investigación, Universidad Nacional de Hurlingham (UNAHUR), Hurlingham 1688, Buenos Aires, Argentina; (M.M.B.M.); (M.C.); (M.W.); (S.U.M.); (C.N.S.); (N.G.); (M.G.)
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires 1425, Argentina;
| | - Santiago Urrea Montes
- Laboratorio de Aplicaciones Biotecnológicas y Microbiología (LAByM), Secretaría de Investigación, Universidad Nacional de Hurlingham (UNAHUR), Hurlingham 1688, Buenos Aires, Argentina; (M.M.B.M.); (M.C.); (M.W.); (S.U.M.); (C.N.S.); (N.G.); (M.G.)
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires 1425, Argentina;
| | - Camila N. Salomón
- Laboratorio de Aplicaciones Biotecnológicas y Microbiología (LAByM), Secretaría de Investigación, Universidad Nacional de Hurlingham (UNAHUR), Hurlingham 1688, Buenos Aires, Argentina; (M.M.B.M.); (M.C.); (M.W.); (S.U.M.); (C.N.S.); (N.G.); (M.G.)
- Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC), La Plata 1900, Buenos Aires, Argentina
| | - Natalia Gonzalez
- Laboratorio de Aplicaciones Biotecnológicas y Microbiología (LAByM), Secretaría de Investigación, Universidad Nacional de Hurlingham (UNAHUR), Hurlingham 1688, Buenos Aires, Argentina; (M.M.B.M.); (M.C.); (M.W.); (S.U.M.); (C.N.S.); (N.G.); (M.G.)
| | - Matías Garavaglia
- Laboratorio de Aplicaciones Biotecnológicas y Microbiología (LAByM), Secretaría de Investigación, Universidad Nacional de Hurlingham (UNAHUR), Hurlingham 1688, Buenos Aires, Argentina; (M.M.B.M.); (M.C.); (M.W.); (S.U.M.); (C.N.S.); (N.G.); (M.G.)
| | - Diego Faccone
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires 1425, Argentina;
- Servicio Antimicrobianos, National Reference Laboratory in Antimicrobial Resistance (NRLAR), National Institute of Infectious Diseases (INEI), ANLIS “Dr. Carlos G. Malbrán”, Ave. Velez Sarsfield, 563, Buenos Aires City 1281, Argentina
| | - Paulo C. Maffía
- Laboratorio de Aplicaciones Biotecnológicas y Microbiología (LAByM), Secretaría de Investigación, Universidad Nacional de Hurlingham (UNAHUR), Hurlingham 1688, Buenos Aires, Argentina; (M.M.B.M.); (M.C.); (M.W.); (S.U.M.); (C.N.S.); (N.G.); (M.G.)
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires 1425, Argentina;
| |
Collapse
|
14
|
Marena GD, Thomaz L, Nosanchuk JD, Taborda CP. Galleria mellonella as an Invertebrate Model for Studying Fungal Infections. J Fungi (Basel) 2025; 11:157. [PMID: 39997451 PMCID: PMC11856299 DOI: 10.3390/jof11020157] [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: 12/02/2024] [Revised: 01/24/2025] [Accepted: 02/15/2025] [Indexed: 02/26/2025] Open
Abstract
The incidence of fungal infections continues to increase and one of the factors responsible for these high rates is the emergence of multi-resistant species, hospitalizations, inappropriate or prolonged use of medications, and pandemics, such as the ongoing HIV/AIDS pandemic. The recent pandemic caused by the severe acute respiratory syndrome virus (SARS-CoV-2) has led to a significant increase in fungal infections, especially systemic mycoses caused by opportunistic fungi. There is a growing and urgent need to better understand how these microorganisms cause infection and develop resistance as well as to develop new therapeutic strategies to combat the diverse diseases caused by fungi. Non-mammalian hosts are increasingly used as alternative models to study microbial infections. Due to their low cost, simplicity of care, conserved innate immunity and reduced ethical issues, the greater wax moth Galleria mellonella is an excellent model host for studying fungal infections and it is currently widely used to study fungal pathogenesis and develop innovative strategies to mitigate the mycoses studied. G. mellonella can grow at 37 °C, which is similar to the mammalian temperature, and the anatomy of the larvae allows researchers to easily deliver pathogens, biological products, compounds and drugs. The aim of this review is to describe how G. mellonella is being used as a model system to study fungal infections as well as the importance of this model in evaluating the antifungal profile of potential drug candidates or new therapies against fungi.
Collapse
Affiliation(s)
- Gabriel Davi Marena
- Institute of Biomedical Science, Department of Microbiology, University of São Paulo (ICB II—USP), São Paulo 05508-900, Brazil;
| | - Luciana Thomaz
- Institute of Biomedical Science, Department of Microbiology, University of São Paulo (ICB II—USP), São Paulo 05508-900, Brazil;
| | - Joshua Daniel Nosanchuk
- Departments of Medicine and Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA;
| | - Carlos Pelleschi Taborda
- Institute of Biomedical Science, Department of Microbiology, University of São Paulo (ICB II—USP), São Paulo 05508-900, Brazil;
- Laboratory of Medical Mycology, Institute of Tropical Medicine of São Paulo/LIM53, School of Medicine, University of São Paulo, São Paulo 05403-000, Brazil
| |
Collapse
|
15
|
Muzahid NH, Ramesh A, Siew TH, Hasan MZ, Narayanan K, Rahman S. Comparison of the virulence of community- and hospital- isolated Acinetobacter baumannii in HeLa cell line and insect model, Galleria mellonella. Access Microbiol 2025; 7:000858.v3. [PMID: 39959467 PMCID: PMC11829074 DOI: 10.1099/acmi.0.000858.v3] [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: 06/03/2024] [Accepted: 01/21/2025] [Indexed: 02/18/2025] Open
Abstract
Acinetobacter baumannii is an important nosocomial pathogen causing high infections and morbidity among affected individuals, and most studies focus on nosocomial strains. However, A. baumannii can also be isolated from healthy community individuals. This study compared the pathogenicity of hospital and community A. baumannii isolates using Galleria mellonella and human cell cultures. The insect model, G. mellonella, and in vitro HeLa cell line were used with ten A. baumannii isolates (six community and four hospital isolates from Segamat, Malaysia). G. mellonella killing assays and HeLa cell adherence, invasion and cytotoxicity assays were performed to investigate the virulence and invasion potential of the isolates. Out of the ten isolates investigated, three community and two hospital isolates were found to be highly virulent in the G. mellonella infection model, killing 100% of larvae within 96 h. These strains were also found to be invasive and have significant cytotoxicity in HeLa cells. Our study revealed that community- and hospital-isolated A. baumannii could be equally virulent judged by both model systems. Undoubtedly, besides hospital settings, the presence of highly virulent A. baumannii in community reservoirs poses a significant public health risk and requires additional investigation.
Collapse
Affiliation(s)
- Nazmul Hasan Muzahid
- School of Science, Monash University Malaysia, 47500, Bandar Sunway, Selangor Darul Ehsan, Malaysia
| | - Aarthi Ramesh
- School of Science, Monash University Malaysia, 47500, Bandar Sunway, Selangor Darul Ehsan, Malaysia
| | - Tan Hock Siew
- School of Science, Monash University Malaysia, 47500, Bandar Sunway, Selangor Darul Ehsan, Malaysia
| | - Md Zobaer Hasan
- School of Science, Monash University Malaysia, 47500, Bandar Sunway, Selangor Darul Ehsan, Malaysia
| | - Kumaran Narayanan
- Jeffrey Cheah School of Medicine & Health Sciences, Monash University Malaysia, 47500, Bandar Sunway, Selangor Darul Ehsan, Malaysia
| | - Sadequr Rahman
- School of Science, Monash University Malaysia, 47500, Bandar Sunway, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
16
|
Andrade M, Neves J, Bento M, Marques J, Seabra SG, Silveira H, Rodrigues L, Armada A, Viveiros M, Couto I, Costa SS. Evaluation of Amlodipine and Imipramine Efficacy to Treat Galleria mellonella Infection by Biofilm-Producing and Antimicrobial-Resistant Staphylococcus aureus. Antibiotics (Basel) 2025; 14:183. [PMID: 40001426 PMCID: PMC11851413 DOI: 10.3390/antibiotics14020183] [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: 12/31/2024] [Revised: 01/30/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Antimicrobial-resistant Staphylococcus aureus is a growing threat to human health for which alternative therapeutic options are needed. In this study, we aimed to evaluate the efficacy of amlodipine (AML) and imipramine (IMI) to treat S. aureus infection in the Galleria mellonella larval model by targeting efflux and biofilms, which are relevant contributors to antimicrobial resistance and virulence in S. aureus. Methods: In-house reared G. mellonella were used in virulence assays to determine the infective dose of two S. aureus strains differing in the expression of norA (gene encoding the native NorA efflux pump). Toxicology assays were conducted to determine the drugs' LD50 for G. mellonella. Drug efficacy assays were performed to evaluate the potential of amlodipine, imipramine and the control drugs ciprofloxacin (CIP) and enalapril (ENA) to clear S. aureus infection in G. mellonella. Results: Survival analysis defined the infective dose as 1 × 107 CFU/larva for both strains. High LD50 values were determined (CIP: >1000 mg/kg; AML: >640 mg/kg; IMI: 1141 mg/kg; ENA: >1280 mg/kg), revealing a high tolerance of G. mellonella to these drugs. AML at 15 mg/kg and IMI at 100 mg/kg increased the larvae survival by 20% (p = 0.04) and 11% (p = 0.11), respectively, also positively affecting health score indexes. In agreement with the literature, ciprofloxacin at >100 mg/kg promoted larvae survival by >73%. Conclusions: Amlodipine and imipramine show mild potential as new therapeutic options for managing S. aureus infections but are promising as new lead molecules. This study also reinforces G. mellonella as a sustainable, reliable model for drug evaluation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Sofia Santos Costa
- Global Health and Tropical Medicine, GHTM, Associated Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, 1349-008 Lisbon, Portugal; (M.A.); (J.N.); (M.B.); (J.M.); (S.G.S.); (H.S.); (L.R.); (A.A.); (M.V.); (I.C.)
| |
Collapse
|
17
|
Axline CMR, Kochan TJ, Nozick S, Ward T, Afzal T, Niki I, Mitra SD, VanGosen E, Nelson J, Valdes A, Hynes D, Cheng W, Lee J, Prashanth P, Turner TL, Pincus NB, Scheetz MH, Bachta KER, Hauser AR. Refined methodology for quantifying Pseudomonas aeruginosa virulence using Galleria mellonella. Microbiol Spectr 2025; 13:e0166624. [PMID: 39665556 PMCID: PMC11792518 DOI: 10.1128/spectrum.01666-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 11/15/2024] [Indexed: 12/13/2024] Open
Abstract
Larvae of Galleria mellonella (the greater wax moth) are being increasingly used as a model to study microbial pathogenesis. In this model, bacterial virulence is typically measured by determining the 50% lethal dose (LD50) of a bacterial strain or mutant. The use of G. mellonella to study Pseudomonas aeruginosa pathogenesis, however, is challenging because of the extreme sensitivity of larvae to this bacterium. For some P. aeruginosa strains, as few as 1-5 colony-forming units are sufficient to kill G. mellonella, which poses challenges for determining LD50 values. For this reason, some groups have used time-to-death as a measure of P. aeruginosa virulence, but methodologies have not been standardized. We provide a detailed protocol for using the time at which 50% of larvae have died (LT50) at a particular inoculum as a measure of P. aeruginosa virulence. We also describe a quality control metric for enhancing the reproducibility of LT50 values. This approach provides an accurate and reproducible methodology for using G. mellonella larvae to measure and compare the virulence of P. aeruginosa strains.IMPORTANCEPseudomonas aeruginosa is a significant cause of morbidity and mortality. The invertebrate Galleria mellonella is used as a model to determine the virulence of P. aeruginosa strains. We provide a protocol and analytical approach for using a time-to-death metric to accurately quantify the virulence of P. aeruginosa strains in G. mellonella larvae. This methodology, which has several advantages over 50% lethal dose approaches, is a useful resource for the study of P. aeruginosa pathogenicity.
Collapse
Affiliation(s)
- Christopher M. R. Axline
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern University, Evanston, Illinois, USA
| | - Travis J. Kochan
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Sophie Nozick
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Timothy Ward
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Yale University, New Haven, Connecticut, USA
| | - Tania Afzal
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northeastern Illinois University, Chicago, Illinois, USA
| | - Issay Niki
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern University, Evanston, Illinois, USA
| | - Sumitra D. Mitra
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ethan VanGosen
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Tufts University, Medford, Massachusetts, USA
| | - Julia Nelson
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern University, Evanston, Illinois, USA
| | - Aliki Valdes
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - David Hynes
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Hamilton College, Clinton, New York, USA
| | - William Cheng
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Joanne Lee
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Prarthana Prashanth
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Illinois Mathematics and Science Academy, Aurora, Illinois, USA
| | - Timothy L. Turner
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Nathan B. Pincus
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Marc H. Scheetz
- Department of Pharmacy Practice, Midwestern University Colleges of Pharmacy and Pharmacology, Downers Grove, Illinois, USA
- Pharmacometrics Center of Excellence, Midwestern University, Downers Grove, Illinois, USA
| | - Kelly E. R. Bachta
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Division of Infectious Diseases, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Alan R. Hauser
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Division of Infectious Diseases, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
18
|
Jiang C, Jie J, Wang J, Deng X, Qiu J, Liu H. Sesamol hinders the proliferation of intracellular bacteria by promoting fatty acid metabolism and decreasing excessive inflammation. Int Immunopharmacol 2025; 146:113966. [PMID: 39733644 DOI: 10.1016/j.intimp.2024.113966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/15/2024] [Accepted: 12/26/2024] [Indexed: 12/31/2024]
Abstract
The extraintestinal pathogenic Escherichia coli (ExPEC) is a significant zoonotic bacterial pathogen that can cause severe infections and potentially cross-transmit between different hosts. The treatment of clinical bacterial infections is challenging because of the increasingly severe problem of drug resistance. The development of new strategies for managing bacterial infections is essential. Host-acting antibacterial compound (HAC)-based host-directed therapy (HDT) has emerged as a promising approach to combat bacterial infections by targeting host-pathogen interactions and bacterial intracellular survival strategies. In this study, we conducted a cell-based screening to identify compounds that can inhibit the survival and proliferation of ExPEC within host cells. Our screening revealed that sesamol effectively inhibited ExPEC proliferation but had no effect on the natural growth of bacteria. Analysis of the transcriptome data revealed that sesamol has the ability to increase the metabolism of host fatty acids while also suppressing excessive inflammation. Mechanistic studies have shown that sesamol-induced PPAR-β activation is crucial for increased fatty acid metabolism and clearance of intracellular bacteria. Furthermore, sesamol treatment demonstrated protective effects against ExPEC infection in both Galleria mellonella and mouse models, suggesting its potential use for treating diseases caused by intracellular bacterial pathogens and as a lead compound for further development of anti-infection drugs on the basis of the HDT strategy.
Collapse
Affiliation(s)
- Chenxiao Jiang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University; Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Jing Jie
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University; Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Jianfeng Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University; Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Xuming Deng
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University; Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Jiazhang Qiu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University; Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China.
| | - Hongtao Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University; Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
19
|
Alsharif A, Aldawsari L, Koukkidis G, Freestone P. The Impact of Compounds Released from Damaged Salad Leaves on the Growth and Virulence of Listeria monocytogenes. Microorganisms 2025; 13:252. [PMID: 40005619 PMCID: PMC11857465 DOI: 10.3390/microorganisms13020252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/09/2025] [Accepted: 01/18/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Fresh produce such as leafy green salads have recently become recognized as a potential source of food-borne infection by enteric pathogens This study investigated whether compounds released from damaged salad leaves were recognized by Listeria monocytogenes strain EGD and if they impacted its growth and virulence. METHODS The effects of extracts of salad leaves or salad bag fluids were tested on the growth, biofilm formation, and colonization of salad leaves and host cell virulence. RESULTS The presence of salad extract at a concentration of less than 0.5% v/v and salad bag fluids at a concentration of 10% v/v enhanced the growth in water and serum-based medium by more than 10,000 times over un-supplemented control cultures. Light and scanning electron microscopy, as well as eukaryotic Caco-2 and Galleria mellonella models of infection, showed that leafy green extracts from rocket, lettuce, spinach, and their salad bag fluids significantly increased the ability of Listeria to establish biofilms and infect host cells. CONCLUSIONS This investigation showed that salad leaf extracts can markedly enhance bacterial virulence, which has implications for bagged salad leaf consumer safety if the leaves become contaminated with pathogenic bacteria such as Listeria.
Collapse
Affiliation(s)
| | | | | | - Primrose Freestone
- Department of Respiratory Sciences, University of Leicester, University Road, Leicester LE1 7RH, UK; (A.A.); (L.A.); (G.K.)
| |
Collapse
|
20
|
Villani S, Calcagnile M, Demitri C, Alifano P. Galleria mellonella (Greater Wax Moth) as a Reliable Animal Model to Study the Efficacy of Nanomaterials in Fighting Pathogens. NANOMATERIALS (BASEL, SWITZERLAND) 2025; 15:67. [PMID: 39791825 PMCID: PMC11723170 DOI: 10.3390/nano15010067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 12/31/2024] [Accepted: 12/31/2024] [Indexed: 01/12/2025]
Abstract
The spread of multidrug-resistant microbes has made it necessary and urgent to develop new strategies to deal with the infections they cause. Some of these are based on nanotechnology, which has revolutionized many fields in medicine. Evaluating the safety and efficacy of these new antimicrobial strategies requires testing in animal models before being tested in clinical trials. In this context, Galleria mellonella could represent a valid alternative to traditional mammalian and non-mammalian animal models, due to its low cost, ease of handling, and valuable biological properties to investigate host-pathogen interactions. The purpose of this review is to provide an updated overview of the literature concerning the use of G. mellonella larvae as an animal model to evaluate safety and efficacy of nanoparticles and nanomaterials, particularly, of those that are used or are under investigation to combat microbial pathogens.
Collapse
Affiliation(s)
- Stefania Villani
- Department of Engineering for Innovation, University of Salento, Via Monteroni, 73100 Lecce, Italy;
| | - Matteo Calcagnile
- Department of Experimental Medicine, University of Salento, Via Monteroni, 73100 Lecce, Italy;
| | - Christian Demitri
- Department of Experimental Medicine, University of Salento, Via Monteroni, 73100 Lecce, Italy;
| | - Pietro Alifano
- Department of Experimental Medicine, University of Salento, Via Monteroni, 73100 Lecce, Italy;
| |
Collapse
|
21
|
Yan Y, Kong N, Niu Y, Sun K, Zhang W. Analysis of the virulence of a lethal, carbapenem-resistant hypervirulent KPC-33-producing Klebsiella pneumoniae: Emergence of ST11-KL64 hv-CRKP in ICU. Microb Pathog 2025; 198:107154. [PMID: 39586335 DOI: 10.1016/j.micpath.2024.107154] [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: 07/14/2024] [Revised: 11/17/2024] [Accepted: 11/22/2024] [Indexed: 11/27/2024]
Abstract
OBJECTIVE Hypervirulent and carbapenem-resistant Klebsiella pneumoniae (hv-CRKP) poses a serious threat to public health. Here, we analyse a case of systemic infection caused by a hv-CRKP, which ultimately led to the patient's death from sepsis. And a total of 30 CRKPs were analyzed to elucidate the molecular epidemiological features of CRKPs in the hospital, and to provide a basis for clinical anti-infective therapy. METHODS In this case, a total of 7 K. pneumoniae strains were isolated from the blood, sputum, urine, and feces of the patient. The Vitek-2 compact system was used to identify the strains and perform antimicrobial susceptibility testing. Biofilm formation, siderophore production assays and Galleria mellonella infection model were used to verify the virulence phenotypes of the strains in the case. Whole-genome sequencing was conducted on the four hv-CRKP isolated from different samples in the case and 26 other CRKP collected in our hospital from September to November in 2022, using the Illumina Hiseq 6000 high-throughput sequencing platform to analyse the resistance and virulence genes. RESULTS In the case, after 7 days of treatment with ceftazidime-avibactam (CZA), the resistance profile of the strains changed. The strain that was initially sensitive to CZA developed to resistant, resistant to imipenem (IPM) developed to sensitive, and resistant to meropenem (MEM) developed to intermediate. Whole-genome sequencing revealed that the four strains in the case were all ST11-KL64 K. pneumoniae, and the change in resistance phenotype was due to the mutation from blaKPC-2 to blaKPC-33. KPN7 had a total of six plasmids, with siderophore-related genes iucABCD and iutA, and mucoid phenotype-related gene rmpA2 located on plasmid p4-KPN7; resistance genes blaKPC-33, blaTEM-1B, and blaCTX-M-65 located on plasmid p5-KPN7; and virulence genes fim, irp, iutA, and ybt located on the chromosome. Biofilm formation and siderophore production assays confirmed that the seven K. pneumoniae strains isolated in this case had strong biofilm formation and siderophore production capabilities. Galleria mellonella Infection Model showed that KPN4 and KPN7 was phenotypically highly virulent and KPN7 performed lower virulence compared to KPN4. Apart from the 4 hv-CRKP strains, other 26 CRKP strains all carried blaKPC-2, and 69.2% (18/26) were ST-11 and 30.8%(8/26) were ST-15. And 83.3% (15/18) were ST11-KL64 strains, followed by ST11-KL25 strains 11.1%(2/18) and ST11-KL47 strain 5.6%(1/18). All the eight ST-15 strains were KL-19. CONCLUSION The ST11-KL64 hv-CRKP clone spread widely in ICU carried numerous resistance and virulence genes, and under antibiotic pressure, they easily underwent mutations resulting in changes in resistance phenotypes, especially in mutations of blaKPC-2 gene in acquiring resistance to CZA. Therefore, clinical attention should be paid to such strains, and the use of antibiotics should be adjusted promptly based on the susceptibility of the strains to antimicrobial agents.
Collapse
Affiliation(s)
- Yuzhong Yan
- Department of Scientific Research, Shanghai University of Medicine and Health Science Affiliated Zhoupu Hospital, Shanghai, China
| | - Nana Kong
- Department of Clinical Laboratory, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuxiao Niu
- Xinxiang Medical University, 601 Jinsui Road, Xinxiang, Henan, China
| | - Kangde Sun
- Department of Clinical Laboratory, Shanghai Ninth People's Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Wenxia Zhang
- Department of Clinical Laboratory, Shanghai University of Medicine and Health Science Affiliated Zhoupu Hospital, Shanghai, China.
| |
Collapse
|
22
|
Pilarczyk-Zurek M, Budziaszek J, Nandagopal K, Kurylek A, Kozinska A, Dmowski M, Sitkiewicz I, Kern-Zdanowicz I, Koziel J. Streptococcus anginosus orchestrates antibacterial potential of NETs facilitating survival of accompanying pathogens. Microbiol Res 2025; 290:127959. [PMID: 39489135 DOI: 10.1016/j.micres.2024.127959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/18/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024]
Abstract
Streptococcus anginosus is considered an emerging opportunistic pathogen causing life-threatening infections, including abscesses and empyema. Noticeably, clinical data revealed that S. anginosus also constitutes an important component of polymicrobial infections. Here, we showed for the first time that S. anginosus inactivates the antibacterial potential of neutrophil extracellular traps (NETs). The process is determined by a cell wall-anchored nuclease referred to as SanA, which high expression dominates in clinical strains isolated from severe infections. Nuclease activity protects S. anginosus against the antibacterial activity of NETs, supporting at the same time the survival of coexisting highly pathogenic species of Enterobacteriales. Obtained data suggest that SanA nuclease should be recognized as a critical S. anginosus virulence factor determining severe monospecies purulent infections but also shielding other pathogens promoting the development of polymicrobial infections.
Collapse
Affiliation(s)
- Magdalena Pilarczyk-Zurek
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology of Jagiellonian University, Krakow, Poland
| | - Joanna Budziaszek
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology of Jagiellonian University, Krakow, Poland
| | - Keerthanaa Nandagopal
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology of Jagiellonian University, Krakow, Poland
| | - Aleksandra Kurylek
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warszawa, Poland
| | - Aleksandra Kozinska
- Department of Drug Biotechnology and Bioinformatics, National Medicines Institute, Warszawa, Poland
| | - Michal Dmowski
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warszawa, Poland
| | - Izabela Sitkiewicz
- Institute of Biology, Warsaw University of Life Sciences-SGGW, Warszawa, Poland
| | | | - Joanna Koziel
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology of Jagiellonian University, Krakow, Poland.
| |
Collapse
|
23
|
De Lima Gualque MW, Vaso CO, dos Santos KS, Galeane MC, Gomes PC, Palma MS, Soares Mendes Giannini MJ, Moroz A, Fusco Almeida AM. Peptides from Galleria mellonella against Cryptococcus spp: toxicity in three-dimensional cell cultures and G. mellonella. Future Microbiol 2025; 20:11-21. [PMID: 39552598 PMCID: PMC11731228 DOI: 10.1080/17460913.2024.2421632] [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: 06/10/2024] [Accepted: 10/23/2024] [Indexed: 11/19/2024] Open
Abstract
Aim: This work aimed to test peptides against the planktonic and biofilm form of Cryptococcus spp. and in vitro toxicity using three-dimensional (3D) cells characterized and evaluate in vivo toxicity in Galleria mellonella.Materials & methods: Susceptibility tests were conducted on the planktonic form and biofilm formation. The toxicity of the peptides was evaluated in lung and brain cells in monolayer (2D) and 3D mono- and co-culture, in addition to in vivo analysis with G. mellonella.Results: Susceptibility values ranged from 31.25 to over 250 µg/ml with a fungicidal profile. Regarding toxicity, the PepM2 peptide was not toxic in 3D culture (500 µg/ml). G. mellonella, showed a survival rate of more than 85% In assays with brain and lung cell lines, concentrations ranged from 4 × 104 to 4 × 103 cells/well for brain cells and 1 × 103 cells/well for lung cells. Cocultures used 1 × 105 brain and 1 × 103 lung cells.Conclusion: This study shows that the peptides have great potential against cryptococcosis, and all spheroids were characterized as having a spheroidal and compact structure.
Collapse
Affiliation(s)
- Marcos William De Lima Gualque
- Laboratory of Mycology & Center of Proteomics, Department of Clinical Analysis, School of Pharmaceutical Sciences, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
| | - Carolina Orlando Vaso
- Laboratory of Mycology & Center of Proteomics, Department of Clinical Analysis, School of Pharmaceutical Sciences, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
| | - Kelvin Sousa dos Santos
- Laboratory for Monoclonal Antibodies, Department of Clinical Analysis, School of Pharmaceutical Sciences, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
| | - Mariana Cristina Galeane
- Laboratory of Mycology & Center of Proteomics, Department of Clinical Analysis, School of Pharmaceutical Sciences, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
| | - Paulo César Gomes
- Laboratory of Mycology & Center of Proteomics, Department of Clinical Analysis, School of Pharmaceutical Sciences, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
| | - Mario Sérgio Palma
- Department of Basic & Applied Biology/LSBZ, Institute of Biosciences, São Paulo State University-UNESP, Rio Claro, 13506-900, São Paulo, Brazil
| | - Maria José Soares Mendes Giannini
- Laboratory of Mycology & Center of Proteomics, Department of Clinical Analysis, School of Pharmaceutical Sciences, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
| | - Andrei Moroz
- Laboratory for Monoclonal Antibodies, Department of Clinical Analysis, School of Pharmaceutical Sciences, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
| | - Ana Marisa Fusco Almeida
- Laboratory of Mycology & Center of Proteomics, Department of Clinical Analysis, School of Pharmaceutical Sciences, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
| |
Collapse
|
24
|
Kaczmarek A, Wrońska AK, Sobich J, Boguś MI. The multifunctional role of IFN-γ in Galleria mellonella (Lepidoptera) immunocompetent cells. Cytokine 2025; 185:156804. [PMID: 39546818 DOI: 10.1016/j.cyto.2024.156804] [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: 09/13/2024] [Revised: 10/18/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024]
Abstract
Cytokines are highly conserved between mammals and insects. The present study examines the multiple effects of interferon-gamma (IFN-γ) application on the immunological defence mechanisms of Galleria mellonella larvae, invertebrates which are gaining popularity as a replacement for mammalian research models in immunological studies. G. mellonella hemolymph is known to contain an IFN-γ homolog that shares 33 % similarity with its mammalian analogue, and its level in insect hemocytes increases during exposition to entomopathogenic fungus Conidiobolus coronatus. The present research examines the impact of IFN-γ on larval development, the effectiveness of fungal infection, and the morphology and physiology of wax moth immunocompetent cells. Treatment with IFN-γ enhanced wound healing, chemotaxis activity and hemocyte impedance, while reducing hemocyte phagocytosis and oxidative stress in cultured immunocompetent cells; it also appears to increase the levels of Jak-2- and NF-κB-like molecules in hemocytes. Our findings suggest that IFN-γ demonstrated considerable similarity between mammals and humans, thus further demonstrating the evolutionary conservatism of cytokines.
Collapse
Affiliation(s)
- Agata Kaczmarek
- Museum and Institute of Zoology, Polish Academy of Sciences, Twarda 51/55, 00-818 Warszawa, Poland.
| | - Anna Katarzyna Wrońska
- Museum and Institute of Zoology, Polish Academy of Sciences, Twarda 51/55, 00-818 Warszawa, Poland
| | - Justyna Sobich
- Museum and Institute of Zoology, Polish Academy of Sciences, Twarda 51/55, 00-818 Warszawa, Poland
| | - Mieczysława Irena Boguś
- Museum and Institute of Zoology, Polish Academy of Sciences, Twarda 51/55, 00-818 Warszawa, Poland
| |
Collapse
|
25
|
Sun N, Deng X, Kong H, Zhi Z, Jiang G, Xiong J, Chen S, Li S, Yuan W, Wong WL. Magnolol as an Antibacterial Agent Against Drug-resistant Bacteria Targeting Filamentous Temperature-sensitive Mutant Z. Chem Biodivers 2024:e202402800. [PMID: 39714990 DOI: 10.1002/cbdv.202402800] [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: 10/29/2024] [Revised: 11/25/2024] [Accepted: 12/18/2024] [Indexed: 12/25/2024]
Abstract
The emergence of multiple drug-resistant bacteria poses critical health threats worldwide. It is urgently needed to develop potent and safe antibacterial agents with novel bactericidal mechanisms to treat these infections. In this study, magnolol was identified as a potential bacterial cell division inhibitor by a cell-based screening approach. This compound showed good antibacterial activity against a number of Gram-positive pathogens (minimum inhibitory concentration 8-16 µg/mL) including methicillin-resistant Staphylococcus aureus and vancomycin-resistant Enterococcus. Further results obtained from biochemical experiments demonstrated that magnolol could markedly disrupt GTPase activity and filamentous temperature-sensitive mutant Z (FtsZ) polymerization, consistent with the impediment to cell division in the bacteria tested. The in vivo antibacterial activity of magnolol was evaluated with a Galleria mellonella larvae model. The results showed that magnolol significantly increased the survival rate of larvae infected with methicillin-resistant S. aureus. The interaction pattern of magnolol with FtsZ was investigated through molecular docking. The finding may offer meaningful insights into the mechanism of action of the compound. The results point to magnolol as a promising antimicrobial compound that inhibits cell division by affecting FtsZ polymerization and has the potential to be developed into an effective antimicrobial drug by further structure modification.
Collapse
Affiliation(s)
- Ning Sun
- Guangzhou 11th People's Hospital, Guangzhou Cadre and Talent Health Management Center, Guangzhou, P. R. China
| | - Xin Deng
- Guangzhou 11th People's Hospital, Guangzhou Cadre and Talent Health Management Center, Guangzhou, P. R. China
| | - Hanqin Kong
- Guangzhou 11th People's Hospital, Guangzhou Cadre and Talent Health Management Center, Guangzhou, P. R. China
| | - Ziling Zhi
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, P. R. China
| | - Guli Jiang
- Guangzhou 11th People's Hospital, Guangzhou Cadre and Talent Health Management Center, Guangzhou, P. R. China
| | - Jing Xiong
- Guangzhou 11th People's Hospital, Guangzhou Cadre and Talent Health Management Center, Guangzhou, P. R. China
| | - Sisi Chen
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, P. R. China
| | - Song Li
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, P. R. China
| | - Wenchang Yuan
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, P. R. China
| | - Wing-Leung Wong
- State Key Laboratory of Chemical Biology and Drug Discovery, and Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, P. R. China
| |
Collapse
|
26
|
Spencer EK, Miller CR, Bull J. Standardized methods for rearing a moth larva, Manduca sexta, in a laboratory setting. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.18.629232. [PMID: 39763740 PMCID: PMC11702677 DOI: 10.1101/2024.12.18.629232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/14/2025]
Abstract
The larval tobacco hornworm, Manduca sexta, has been used in a laboratory setting for physiological studies and for pathogen virulence studies. This moth offers a much larger size than the commonly used wax moth (Galleria mellonella), and it can thus be used for a greater variety of assays, such as repeated sampling of the same individual, growth measurements, and tissue sampling. Yet their occasional use in research has led to a minimally documented set of rearing methods. To facilitate further adoption of this insect model, we expanded on previously reported protocols and developed our own rearing methods, which we report here. Our protocol requires little specialized equipment, with a cost less than $100/month for the feeding and maintenance of a laboratory colony of about five hundred larvae of differing instar phases. The low cost generalized equipment and supplies, and the simplification of the standardized protocols allows for an easy entry point for rearing tobacco hornworm populations. We also describe a few methods that are relevant to the uses of these organisms as infection models.
Collapse
Affiliation(s)
- Emma K. Spencer
- Department of Biological Sciences, University of Idaho, Moscow, Idaho, United States of America
| | - Craig R. Miller
- Department of Biological Sciences, University of Idaho, Moscow, Idaho, United States of America
- Institute for Modeling Collaboration and Innovation, University of Idaho, Moscow, Idaho, United States of America
| | - James Bull
- Department of Biological Sciences, University of Idaho, Moscow, Idaho, United States of America
- Institute for Modeling Collaboration and Innovation, University of Idaho, Moscow, Idaho, United States of America
| |
Collapse
|
27
|
Bah U, de Llanos Frutos R, Donnellan S, Smith A, Flockhart A, Singleton I, Wheelhouse N. The potential virulence of Listeria monocytogenes strains isolated from fresh produce processing facilities as determined by an invertebrate Galleria mellonella model. PLoS One 2024; 19:e0311839. [PMID: 39666623 PMCID: PMC11637379 DOI: 10.1371/journal.pone.0311839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 09/13/2024] [Indexed: 12/14/2024] Open
Abstract
Listeria monocytogenes, a bacterium responsible for listeriosis, is an environmental and food-borne pathogen that poses a particular risk to pregnant women and the elderly. While traditionally associated with animal products, ready-to-eat salads are increasingly recognised as a source of Listeria outbreaks. However, little is known about the potential virulence of Listeria isolates from the fresh produce environment. This study assessed the virulence potential of nine L. monocytogenes strains from the fresh produce chain using the Galleria mellonella invertebrate infection model. Larvae were infected with 106 CFU of each strain via their circulatory system and compared to a reference strain L. monocytogenes (EGD-e) and Listeria ivanovii. Virulence was evaluated by measuring mortality rates, health index score of larvae, viable bacterial counts in the larvae, and the larvae's immune. Significant differences in larval mortality were observed among strains. Strains NLmo4 and NLmo5 caused the highest mortality rates (98.8% and 96.7%, respectively at 7 days post-infection), while strain NLmo20 had a significantly lower mortality rate of 65% at the same time point (p<0.05). Six isolates that caused varied mortality rates were then selected and tested for their ability to replicate both in vitro and in vivo and their impact on larval haemocyte density. In vitro growth rates were not significantly different among L. monocytogenes strains or compared to Listeria ivanovii. However, L. monocytogenes strains persisted and replicated in larvae up to 7d days post-infection, whereas Listeria ivanovii was reduced by 5 logs CFU by day 7. The presence of these L. monocytogenes strains caused organ damage in larvae, indicated by increased melanisation and subsequent larval death. Haemocyte density showed insignificant fluctuations following infection. In conclusion, the results of this study suggest L. monocytogenes strains from fresh produce food chain have varying pathogenicity levels and can pose potential risk to human health.
Collapse
Affiliation(s)
- Umaru Bah
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, Scotland, United Kingdom
| | | | - Samantha Donnellan
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, Scotland, United Kingdom
| | - Alva Smith
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, Scotland, United Kingdom
| | - Allen Flockhart
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, Scotland, United Kingdom
| | - Ian Singleton
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, Scotland, United Kingdom
| | - Nick Wheelhouse
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, Scotland, United Kingdom
| |
Collapse
|
28
|
Vanhoffelen E, Vermoesen L, Michiels L, Lagrou K, Reséndiz-Sharpe A, Vande Velde G. Sensitive bioluminescence imaging of cryptococcosis in Galleria mellonella improves antifungal screening under in vivo conditions. Virulence 2024; 15:2327883. [PMID: 38465639 PMCID: PMC10939141 DOI: 10.1080/21505594.2024.2327883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 02/28/2024] [Indexed: 03/12/2024] Open
Abstract
Cryptococcus neoformans is an environmental yeast that primarily affects immunocompromised individuals, causing respiratory infections and life-threatening meningoencephalitis. Treatment is complicated by limited antifungal options, with concerns such as adverse effects, dose-limiting toxicity, blood-brain barrier permeability, and resistance development, emphasizing the critical need to optimize and expand current treatment options against invasive cryptococcosis. Galleria mellonella larvae have been introduced as an ethical intermediate for in vivo testing, bridging the gap between in vitro antifungal screening and mouse studies. However, current infection readouts in G. mellonella are indirect, insensitive, or invasive, which hampers the full potential of the model. To address the absence of a reliable non-invasive method for tracking infection, we longitudinally quantified the cryptococcal burden in G. mellonella using bioluminescence imaging (BLI). After infection with firefly luciferase-expressing C. neoformans, the resulting bioluminescence signal was quantitatively validated using colony-forming unit analysis. Longitudinal comparison of BLI to health and survival analysis revealed increased sensitivity of BLI in discriminating cryptococcal burden during early infection. Furthermore, BLI improved the detection of treatment efficacy using first-line antifungals, thereby benchmarking this model for antifungal testing. In conclusion, we introduced BLI as a real-time, quantitative readout of cryptococcal burden in G. mellonella over time, enabling more sensitive and reliable antifungal screening.
Collapse
Affiliation(s)
- Eliane Vanhoffelen
- Department of Imaging and Pathology, Biomedical MRI unit/MoSAIC, KULeuven, Leuven, Belgium
| | - Lori Vermoesen
- Department of Imaging and Pathology, Biomedical MRI unit/MoSAIC, KULeuven, Leuven, Belgium
| | - Lauren Michiels
- Department of Imaging and Pathology, Biomedical MRI unit/MoSAIC, KULeuven, Leuven, Belgium
| | - Katrien Lagrou
- Department of Microbiology, Immunology and Transplantation, Laboratory of Clinical Microbiology, KULeuven, Leuven, Belgium
- Department of Laboratory Medicine, National Reference Center for Mycosis, University Hospitals Leuven, Leuven, Belgium
| | | | - Greetje Vande Velde
- Department of Imaging and Pathology, Biomedical MRI unit/MoSAIC, KULeuven, Leuven, Belgium
| |
Collapse
|
29
|
Jiranantasak T, Bluhm AP, Chabot DJ, Friedlander A, Bowen R, McMillan IA, Hadfield TL, Hartwig A, Blackburn JK, Norris MH. Toxin and capsule production by Bacillus cereus biovar anthracis influence pathogenicity in macrophages and animal models. PLoS Negl Trop Dis 2024; 18:e0012779. [PMID: 39715264 PMCID: PMC11706511 DOI: 10.1371/journal.pntd.0012779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 01/07/2025] [Accepted: 12/11/2024] [Indexed: 12/25/2024] Open
Abstract
Bacillus cereus biovar anthracis (Bcbva) causes anthrax-like disease in animals, particularly in the non-human primates and great apes of West and Central Africa. Genomic analyses revealed Bcbva as a member of the B. cereus species that carries two plasmids, pBCXO1 and pBCXO2, which have high sequence homology to the B. anthracis toxin and polyglutamate capsule encoding plasmids pXO1 and pXO2, respectively. To date, only a few studies have investigated the effect of variations in Bcbva sporulation, toxin, and capsule synthesis on animal and macrophage pathogenicity compared to B. anthracis, therefore more research is needed to gain a better understanding of the pathogenesis of this emerging infection. Here, we report that Bcbva can multiply and vegetatively survive on nutrient-rich media for a minimum of six days while generating spores. Sporulation of Bcbva occurred faster and more extensively than B. anthracis Ames. Bcbva tended to secrete less protective antigen (PA) than B. anthracis Ames when cultured in growth medium. We found Bcbva produced a substantially higher amount of attached poly-ƴ-D-glutamic acid (PDGA) capsule than B. anthracis Ames when grown in medium supplemented with human serum and CO2. In a phagocytosis assay, Bcbva spores showed reduced internalization by mouse macrophages compared to B. anthracis Ames. Our research demonstrated that Bcbva is more virulent than B. anthracis Ames using two in vivo models, Galleria mellonella larvae and guinea pigs. Following that, the efficacy of the veterinary vaccine Sterne strain 34F2 against anthrax-like disease was assessed in guinea pigs. Sterne vaccinated guinea pigs had significantly increased anti-PA titers compared to the unvaccinated control group. Toxin neutralizing antibody titers in vaccinated guinea pigs correlated with anti-PA titers. This indicates the Sterne vaccine provides adequate protection against Bcbva infection in laboratory animals.
Collapse
Affiliation(s)
- Treenate Jiranantasak
- Spatial Epidemiology & Ecology Research Laboratory, Department of Geography, University of Florida, Gainesville, Florida, United States of America
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, United States of America
| | - Andrew P. Bluhm
- Spatial Epidemiology & Ecology Research Laboratory, Department of Geography, University of Florida, Gainesville, Florida, United States of America
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, United States of America
| | - Donald J. Chabot
- United States Army Medical Research Institute of Infectious Disease, Fort Detrick, Maryland, United States of America
| | - Arthur Friedlander
- United States Army Medical Research Institute of Infectious Disease, Fort Detrick, Maryland, United States of America
| | - Richard Bowen
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Ian A. McMillan
- Pathogen Analysis and Translational Health Group, School of Life Sciences, University of Hawaiʻi at Mānoa, Honolulu, Hawaiʻi, United States of America
| | - Ted L. Hadfield
- Spatial Epidemiology & Ecology Research Laboratory, Department of Geography, University of Florida, Gainesville, Florida, United States of America
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, United States of America
| | - Airn Hartwig
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Jason K. Blackburn
- Spatial Epidemiology & Ecology Research Laboratory, Department of Geography, University of Florida, Gainesville, Florida, United States of America
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, United States of America
| | - Michael H. Norris
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, United States of America
- Pathogen Analysis and Translational Health Group, School of Life Sciences, University of Hawaiʻi at Mānoa, Honolulu, Hawaiʻi, United States of America
| |
Collapse
|
30
|
Camilotti E, Furian TQ, Borges KA, Ortiz Granados OF, Zottis Chitolina G, de Brites Weber T, Tonini da Rocha D, Nascimento VPD, Souza Moraes HLD, Salle CTP. Galleria mellonella larvae as an alternative model to determine the pathogenicity of avian pathogenic Escherichia coli. Avian Pathol 2024; 53:507-519. [PMID: 38845537 DOI: 10.1080/03079457.2024.2365932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 06/27/2024]
Abstract
RESEARCH HIGHLIGHTS Galleria mellonella larvae are a viable model for determining APEC pathogenicity.Larval disease score is the main variable for determining APEC pathogenicity.Response variables should be evaluated up to 24 h post-inoculation.
Collapse
Affiliation(s)
- Elisar Camilotti
- Centro de Diagnóstico e Pesquisa em Patologia Aviária, Departamento de Medicina Animal, Faculdade de Medicina Veterinária, Universidade Federal do Rio Grande do Sul, Rio Grande do Sul, Brazil
| | - Thales Quedi Furian
- Centro de Diagnóstico e Pesquisa em Patologia Aviária, Departamento de Medicina Animal, Faculdade de Medicina Veterinária, Universidade Federal do Rio Grande do Sul, Rio Grande do Sul, Brazil
| | - Karen Apellanis Borges
- Centro de Diagnóstico e Pesquisa em Patologia Aviária, Departamento de Medicina Animal, Faculdade de Medicina Veterinária, Universidade Federal do Rio Grande do Sul, Rio Grande do Sul, Brazil
| | - Oscar Fernando Ortiz Granados
- Centro de Diagnóstico e Pesquisa em Patologia Aviária, Departamento de Medicina Animal, Faculdade de Medicina Veterinária, Universidade Federal do Rio Grande do Sul, Rio Grande do Sul, Brazil
| | - Gabriela Zottis Chitolina
- Centro de Diagnóstico e Pesquisa em Patologia Aviária, Departamento de Medicina Animal, Faculdade de Medicina Veterinária, Universidade Federal do Rio Grande do Sul, Rio Grande do Sul, Brazil
| | - Thaína de Brites Weber
- Centro de Diagnóstico e Pesquisa em Patologia Aviária, Departamento de Medicina Animal, Faculdade de Medicina Veterinária, Universidade Federal do Rio Grande do Sul, Rio Grande do Sul, Brazil
| | - Daniela Tonini da Rocha
- Centro de Diagnóstico e Pesquisa em Patologia Aviária, Departamento de Medicina Animal, Faculdade de Medicina Veterinária, Universidade Federal do Rio Grande do Sul, Rio Grande do Sul, Brazil
| | - Vladimir Pinheiro do Nascimento
- Centro de Diagnóstico e Pesquisa em Patologia Aviária, Departamento de Medicina Animal, Faculdade de Medicina Veterinária, Universidade Federal do Rio Grande do Sul, Rio Grande do Sul, Brazil
| | - Hamilton Luiz de Souza Moraes
- Centro de Diagnóstico e Pesquisa em Patologia Aviária, Departamento de Medicina Animal, Faculdade de Medicina Veterinária, Universidade Federal do Rio Grande do Sul, Rio Grande do Sul, Brazil
| | - Carlos Tadeu Pippi Salle
- Centro de Diagnóstico e Pesquisa em Patologia Aviária, Departamento de Medicina Animal, Faculdade de Medicina Veterinária, Universidade Federal do Rio Grande do Sul, Rio Grande do Sul, Brazil
| |
Collapse
|
31
|
Arévalo-Jaimes BV, Salinas-Pena M, Ponte I, Jordan A, Roque A, Torrents E. Antimicrobial and antibiofilm activity of human recombinant H1 histones against bacterial infections. mSystems 2024; 9:e0070424. [PMID: 39470247 PMCID: PMC11575268 DOI: 10.1128/msystems.00704-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 09/26/2024] [Indexed: 10/30/2024] Open
Abstract
Histones possess significant antimicrobial potential, yet their activity against biofilms remains underexplored. Moreover, concerns regarding adverse effects limit their clinical implementation. We investigated the antibacterial efficacy of human recombinant histone H1 subtypes against Pseudomonas aeruginosa PAO1, both planktonic and in biofilms. After the in vitro tests, toxicity and efficacy were assessed in a P. aeruginosa PAO1 infection model using Galleria mellonella larvae. Histones were also evaluated in combination with ciprofloxacin (Cpx) and gentamicin (Gm). Our results demonstrate antimicrobial activity of all three histones against P. aeruginosa PAO1, with H1.0 and H1.4 showing efficacy at lower concentrations. The bactericidal effect was associated with a mechanism of membrane disruption. In vitro studies using static and dynamic models showed that H1.4 had antibiofilm potential by reducing cell biomass. Neither H1.0 nor H1.4 showed toxicity in G. mellonella larvae, and both increased larvae survival when infected with P. aeruginosa PAO1. Although in vitro synergism was observed between ciprofloxacin and H1.0, no improvement over the antibiotic alone was noted in vivo. Differences in antibacterial and antibiofilm activity were attributed to sequence and structural variations among histone subtypes. Moreover, the efficacy of H1.0 and H1.4 was influenced by the presence and strength of the extracellular matrix. These findings suggest histones hold promise for combating acute and chronic infections caused by pathogens such as P. aeruginosa.IMPORTANCEThe constant increase of multidrug-resistant bacteria is a critical global concern. The inefficacy of current therapies to treat bacterial infections is attributed to multiple mechanisms of resistance, including the capacity to form biofilms. Therefore, the identification of novel and safe therapeutic strategies is imperative. This study confirms the antimicrobial potential of three histone H1 subtypes against both Gram-negative and Gram-positive bacteria. Furthermore, histones H1.0 and H1.4 demonstrated in vivo efficacy without associated toxicity in an acute infection model of Pseudomonas aeruginosa PAO1 in Galleria mellonella larvae. The bactericidal effect of these proteins also resulted in biomass reduction of P. aeruginosa PAO1 biofilms. Given the clinical significance of this opportunistic pathogen, our research provides a comprehensive initial evaluation of the efficacy, toxicity, and mechanism of action of a potential new therapeutic approach against acute and chronic bacterial infections.
Collapse
Affiliation(s)
- Betsy Verónica Arévalo-Jaimes
- Bacterial infections and antimicrobial therapies group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Microbiology Section, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | | | - Inmaculada Ponte
- Biochemistry and Molecular Biology Department, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Albert Jordan
- Molecular Biology Institute of Barcelona (IBMB-CSIC), Barcelona, Spain
| | - Alicia Roque
- Biochemistry and Molecular Biology Department, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Eduard Torrents
- Bacterial infections and antimicrobial therapies group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Microbiology Section, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| |
Collapse
|
32
|
Marinacci B, D'Agostino I, Angeli A, Carradori S, Melfi F, Grande R, Corsiani M, Ferraroni M, Agamennone M, Tondo AR, Zara S, Puca V, Pellegrini B, Vagaggini C, Dreassi E, Patrauchan MA, Capasso C, Nicolotti O, Carta F, Supuran CT. Inhibition of Pseudomonas aeruginosa Carbonic Anhydrases, Exploring Ciprofloxacin Functionalization Toward New Antibacterial Agents: An In-Depth Multidisciplinary Study. J Med Chem 2024; 67:19077-19102. [PMID: 39453626 DOI: 10.1021/acs.jmedchem.4c01555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Ciprofloxacin (CPX) is one of the most employed antibiotics in clinics to date. However, the rise of drug-resistant bacteria is dramatically impairing its efficacy, especially against life-threatening pathogens, such as Pseudomonas aeruginosa. This Gram-negative bacterium is an opportunistic pathogen, often infecting immuno-compromised patients with severe or fatal outcomes. The evidence of the possibility of exploiting Carbonic Anhydrase (CA, EC: 4.2.1.1) enzymes as pharmacological targets along with their role in P. aeruginosa virulence inspired the derivatization of CPX with peculiar CA-inhibiting chemotypes. Thus, a large library of CPX derivatives was synthesized and tested on a panel of bacterial CAs and human isoenzymes I and II. Selected derivatives were evaluated for antibacterial activity, revealing bactericidal and antibiofilm properties for some compounds. Importantly, promising preliminary absorption, distribution, metabolism, and excretion (ADME) properties in vitro were found and no cytotoxicity was detected for some representative compounds when tested in Galleria mellonella larvae.
Collapse
Affiliation(s)
- Beatrice Marinacci
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy
| | - Ilaria D'Agostino
- Department of Pharmacy, University of Pisa, via Bonanno 6, 56126 Pisa, Italy
| | - Andrea Angeli
- NEUROFARBA Department, University of Florence, 50019 Sesto Fiorentino, Florence, Italy
| | - Simone Carradori
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy
| | - Francesco Melfi
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy
| | - Rossella Grande
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy
| | - Micol Corsiani
- NEUROFARBA Department, University of Florence, 50019 Sesto Fiorentino, Florence, Italy
| | - Marta Ferraroni
- Department of Chemistry ″Ugo Schiff″, University of Florence, Via Della Lastruccia 3-13, 50019 Sesto Fiorentino, Italy
| | - Mariangela Agamennone
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy
| | - Anna Rita Tondo
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari Aldo Moro, 70121 Bari, Italy
| | - Susi Zara
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy
| | - Valentina Puca
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy
| | - Benedetta Pellegrini
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy
| | - Chiara Vagaggini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Elena Dreassi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Marianna A Patrauchan
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, Oklahoma 74078, United States
| | - Clemente Capasso
- Department of Biology, Agriculture and Food Sciences, CNR, Institute of Biosciences and Bioresources, 80131 Napoli, Italy
| | - Orazio Nicolotti
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari Aldo Moro, 70121 Bari, Italy
| | - Fabrizio Carta
- NEUROFARBA Department, University of Florence, 50019 Sesto Fiorentino, Florence, Italy
| | - Claudiu T Supuran
- NEUROFARBA Department, University of Florence, 50019 Sesto Fiorentino, Florence, Italy
| |
Collapse
|
33
|
Eiamthaworn K, Holthaus D, Suriyaprom S, Rickerts V, Tragoolpua Y. Immunomodulation and Protective Effects of Cordyceps militaris Extract Against Candida albicans Infection in Galleria mellonella Larvae. INSECTS 2024; 15:882. [PMID: 39590481 PMCID: PMC11595007 DOI: 10.3390/insects15110882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/03/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024]
Abstract
Cordyceps militaris-derived formulations are currently used for multiple purposes because of their medical properties, especially immune system modulation. This study analyzes the inhibitory effects of C. militaris aqueous extract on Candida albicans infections and the immune response in larvae of the greater wax moth Galleria mellonella (Lepidoptera: Pyralidae). Larvae exhibited melanization within 1 h of being infected with C. albicans inoculum at a concentration of 106 cells/larvae, and died within 24 h from a lethal dose. Aqueous extract of C. militaris proved to be nontoxic at concentrations of 0.25 and 0.125 mg/larvae, and had the greatest ability to prolong the survival of larvae infected with a sublethal dose of C. albicans at a concentration of 105 cells/larvae. Moreover, the number of hemocytes in the hemolymph of G. mellonella increased after infection with C. albicans and treatment with the aqueous extract of C. militaris at 1, 24, and 48 h by 1.21 × 107, 1.23 × 107, and 1.4 × 107 cells/100 µL, respectively. The highest number of hemocytes was recorded after treatment of infected G. mellonella with the extract for 48 h. Transcriptional upregulation of the immune system was observed in certain antimicrobial peptides (AMPs), showing that the relative expression of galiomicin, gallerimycin, and lysozyme genes were upregulated as early as 1 h after infection. Therefore, we conclude that C. militaris aqueous extract can modulate the immune system of G. mellonella and protect against infection from C. albicans.
Collapse
Affiliation(s)
- Kiratiya Eiamthaworn
- Department of Biology, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand; (K.E.); (S.S.)
| | - David Holthaus
- Department of Gynecology and Obstetrics, Universitätsklinikum Schleswig-Holstein, 24105 Kiel, Germany;
- Robert Koch Institute, 13353 Berlin, Germany;
| | - Sureeporn Suriyaprom
- Department of Biology, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand; (K.E.); (S.S.)
- Office of Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand
| | | | - Yingmanee Tragoolpua
- Department of Biology, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand; (K.E.); (S.S.)
- Natural Extracts and Innovative Products for Alternative Healthcare Research Group, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
34
|
Vidal M, Arch M, Fuentes E, Cardona PJ. Drosophila melanogaster experimental model to test new antimicrobials: a methodological approach. Front Microbiol 2024; 15:1478263. [PMID: 39568995 PMCID: PMC11576456 DOI: 10.3389/fmicb.2024.1478263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/24/2024] [Indexed: 11/22/2024] Open
Abstract
Given the increasing concern about antimicrobial resistance among the microorganisms that cause infections in our society, there is an urgent need for new drug discovery. Currently, this process involves testing many low-quality compounds, resulting from the in vivo testing, on mammal models, which not only wastes time, resources, and money, but also raises ethical questions. In this review, we have discussed the potential of D. melanogaster as an intermediary experimental model in this drug discovery timeline. We have tackled the topic from a methodological perspective, providing recommendations regarding the range of drug concentrations to test based on the mechanism of action of each compound; how to treat D. melanogaster, how to monitor that treatment, and what parameters we should consider when designing a drug screening protocol to maximize the study's benefits. We also discuss the necessary improvements needed to establish the D. melanogaster model of infection as a standard technique in the drug screening process. Overall, D. melanogaster has been demonstrated to be a manageable model for studying broad-spectrum infection treatment. It allows us to obtain valuable information in a cost-effective manner, which can improve the drug screening process and provide insights into our current major concern. This approach is also in line with the 3R policy in biomedical research, in particular on the replacement and reduce the use of vertebrates in preclinical development.
Collapse
Affiliation(s)
- Maria Vidal
- Microbiology Department, Laboratori Clínic Metropolitana Nord, Germans Trias i Pujol University Hospital (HUGTP), Badalona, Catalonia, Spain
- Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Bellaterra, Catalonia, Spain
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
| | - Marta Arch
- Microbiology Department, Laboratori Clínic Metropolitana Nord, Germans Trias i Pujol University Hospital (HUGTP), Badalona, Catalonia, Spain
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
| | - Esther Fuentes
- Microbiology Department, Laboratori Clínic Metropolitana Nord, Germans Trias i Pujol University Hospital (HUGTP), Badalona, Catalonia, Spain
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
- Comparative Medicine and Bioimage Centre of Catalonia (CMCiB), Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
| | - Pere-Joan Cardona
- Microbiology Department, Laboratori Clínic Metropolitana Nord, Germans Trias i Pujol University Hospital (HUGTP), Badalona, Catalonia, Spain
- Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Bellaterra, Catalonia, Spain
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
- Comparative Medicine and Bioimage Centre of Catalonia (CMCiB), Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
35
|
Lauman P, Dennis JJ. Prophylactic phage biocontrol prevents Burkholderia gladioli infection in a quantitative ex planta model of bacterial virulence. Appl Environ Microbiol 2024; 90:e0131724. [PMID: 39240081 PMCID: PMC11497830 DOI: 10.1128/aem.01317-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 08/09/2024] [Indexed: 09/07/2024] Open
Abstract
Agricultural crop yield losses and food destruction due to infections by phytopathogenic bacteria such as Burkholderia gladioli, which causes devastating diseases in onion, mushroom, corn, and rice crops, pose major threats to worldwide food security and cause enormous damage to the global economy. Biocontrol using bacteriophages has emerged as a promising strategy against a number of phytopathogenic species but has never been attempted against B. gladioli due to a lack of quantitative infection models and a scarcity of phages targeting this specific pathogen. In this study, we present a novel, procedurally straightforward, and highly generalizable fully quantitative ex planta maceration model and an accompanying quantitative metric, the ex planta maceration index (xPMI). In utilizing this model to test the ex planta virulence of a panel of 12 strains of B. gladioli in Allium cepa and Agaricus bisporus, we uncover substantial temperature-, host-, and strain-dependent diversity in the virulence of this fascinating pathogenic species. Crucially, we demonstrate that Burkholderia phages KS12 and AH2, respectively, prevent and reduce infection-associated onion tissue destruction, measured through significant (P < 0.0001) reductions in xPMI, by phytopathogenic strains of B. gladioli, thereby demonstrating the potential of agricultural phage biocontrol targeting this problematic microorganism.IMPORTANCEAgricultural crop destruction is increasing due to infections caused by bacteria such as Burkholderia gladioli, which causes plant tissue diseases in onion, mushroom, corn, and rice crops. These bacteria pose a major threat to worldwide food production, which, in turn, damages the global economy. One potential solution being investigated to prevent bacterial infections of plants is "biocontrol" using bacteriophages (or phages), which are bacterial viruses that readily infect and destroy bacterial cells. In this article, we demonstrate that Burkholderia phages KS12 and AH2 prevent or reduce infection-associated plant tissue destruction caused by strains of B. gladioli, thereby demonstrating the inherent potential of agricultural phage biocontrol.
Collapse
Affiliation(s)
- Philip Lauman
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Jonathan J. Dennis
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
36
|
Sousa M, Magalhães R, Ferreira V, Teixeira P. Current methodologies available to evaluate the virulence potential among Listeria monocytogenes clonal complexes. Front Microbiol 2024; 15:1425437. [PMID: 39493856 PMCID: PMC11528214 DOI: 10.3389/fmicb.2024.1425437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 09/26/2024] [Indexed: 11/05/2024] Open
Abstract
Listeria monocytogenes is a foodborne pathogen that causes listeriosis in humans, the severity of which depends on multiple factors, including intrinsic characteristics of the affected individuals and the pathogen itself. Additionally, emerging evidence suggests that epigenetic modifications may also modulate host susceptibility to infection. Therefore, different clinical outcomes can be expected, ranging from self-limiting gastroenteritis to severe central nervous system and maternal-neonatal infections, and bacteremia. Furthermore, L. monocytogenes is a genetically and phenotypically diverse species, resulting in a large variation in virulence potential between strains. Multilocus sequence typing (MLST) has been widely used to categorize the clonal structure of bacterial species and to define clonal complexes (CCs) of genetically related isolates. The combination of MLST and epidemiological data allows to distinguish hypervirulent CCs, which are notably more prevalent in clinical cases and typically associated with severe forms of the disease. Conversely, other CCs, termed hypovirulent, are predominantly isolated from food and food processing environments and are associated with the occurrence of listeriosis in immunosuppressed individuals. Reports of genetic traits associated with this diversity have been described. The Food and Agriculture Organization (FAO) is encouraging the search for virulence biomarkers to rapidly identify the main strains of concern to reduce food waste and economical losses. The aim of this review is to comprehensively collect, describe and discuss the methodologies used to discriminate the virulence potential of L. monocytogenes CCs. From the exploration of in vitro and in vivo models to the study of expression of virulence genes, each approach is critically explored to better understand its applicability and efficiency in distinguishing the virulence potential of the pathogen.
Collapse
Affiliation(s)
| | | | | | - Paula Teixeira
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina – Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, Porto, Portugal
| |
Collapse
|
37
|
Chu D, Lan J, Liang L, Xia K, Li L, Yang L, Liu H, Zhang T. The antibacterial activity of a novel highly thermostable endolysin, LysKP213, against Gram-negative pathogens is enhanced when combined with outer membrane permeabilizing agents. Front Microbiol 2024; 15:1454618. [PMID: 39439944 PMCID: PMC11493673 DOI: 10.3389/fmicb.2024.1454618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024] Open
Abstract
Phages and phage-encoded lytic enzymes are promising antimicrobial agents. In this study, we report the isolation and identification of bacteriophage KP2025 from Klebsiella pneumoniae. Bioinformatics analysis of KP2025 revealed a putative endolysin, LysKP213, containing a T4-like_lys domain. Purified LysKP213 was found to be highly thermostable, retaining approximately 44.4% of its lytic activity after 20 h of incubation at 95°C, and approximately 57.5% residual activity after 30 min at 121°C. Furthermore, when administered in combination with polymyxin B or fused at the N-terminus with the antimicrobial peptide cecropin A (CecA), LysKP213 exhibited increased antibacterial activity against Gram-negative pathogens, including K. pneumoniae, Pseudomonas aeruginosa, Acinetobacter baumannii, and Escherichia coli, both in vitro and in vivo. These results indicated that LysKP213 is a highly thermostable endolysin that, when combined with or fused with an outer membrane permeabilizer, has enhanced antibacterial activity and is a candidate agent for the control of infections by Gram-negative pathogens.
Collapse
Affiliation(s)
- Dingjian Chu
- Engineering Research Center of Health Medicine Biotechnology of Institution of Higher Education of Guizhou Province, School of Biology and Engineering (School of Modern Industry for Health and Medicine), Guizhou Medical University, Guiyang, China
| | - Jing Lan
- Engineering Research Center of Health Medicine Biotechnology of Institution of Higher Education of Guizhou Province, School of Biology and Engineering (School of Modern Industry for Health and Medicine), Guizhou Medical University, Guiyang, China
| | - Lu Liang
- Guiyang Maternal and Child Health Hospital, Guiyang, China
| | - Kaide Xia
- Guiyang Maternal and Child Health Hospital, Guiyang, China
| | - Linlin Li
- Shanghai Institute of Phage, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Lan Yang
- Shanghai Institute of Phage, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Hongmei Liu
- Engineering Research Center of Health Medicine Biotechnology of Institution of Higher Education of Guizhou Province, School of Biology and Engineering (School of Modern Industry for Health and Medicine), Guizhou Medical University, Guiyang, China
| | - Tingting Zhang
- Engineering Research Center of Health Medicine Biotechnology of Institution of Higher Education of Guizhou Province, School of Biology and Engineering (School of Modern Industry for Health and Medicine), Guizhou Medical University, Guiyang, China
| |
Collapse
|
38
|
Bosch C, Toplis B, Van Staden AD, Volschenk H, Smith C, Dicks L, Botha A. Exposure of Cryptococcus neoformans to low nitrogen levels enhances virulence. Int Microbiol 2024; 27:1587-1595. [PMID: 38483745 PMCID: PMC11452460 DOI: 10.1007/s10123-024-00504-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 02/18/2024] [Accepted: 03/07/2024] [Indexed: 10/05/2024]
Abstract
Previous studies have shown a correlation between nitrogen levels and Cryptococcus neoformans pathogenicity. Here we report on the in vivo effects of cryptococcal pre-exposure to ecologically relevant nitrogen levels. C. neoformans H99 was cultured in yeast carbon base (YCB) supplemented with 0.53 g/L NH4Cl and 0.21 g/L NH4Cl, respectively, and used to infect larvae of the Greater Wax moth, Galleria mellonella. Cells cultured in low nitrogen YCB (LN) were more virulent compared to cells cultured in high nitrogen YCB (HN). Microscopic examination of haemolymph collected from infected larvae revealed that cells cultured in LN were larger than cells cultured in HN, with the majority of LN cells exceeding 10 µm and possibly entering titanisation. Additionally, compared to HN-cultured cells, fewer LN-cultured cells were engulfed by macrophages. The enhanced virulence of LN-cultured cells was attributed to the increased cell size in vivo. In contrast, reduced macrophage uptake was attributed to increased capsule thickness of in vitro cells. Not only do these findings demonstrate the effects of culture conditions, specifically nitrogen levels, on C. neoformans virulence, but they also highlight the importance of isolate background in the cryptococcal-host interaction.
Collapse
Affiliation(s)
- Caylin Bosch
- Department of Microbiology, University of Stellenbosch, Private Bag X1, Matieland, Stellenbosch, 7602, South Africa
| | - Barbra Toplis
- Department of Microbiology, University of Stellenbosch, Private Bag X1, Matieland, Stellenbosch, 7602, South Africa
| | - Anton DuPreez Van Staden
- Department of Microbiology, University of Stellenbosch, Private Bag X1, Matieland, Stellenbosch, 7602, South Africa
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, University of Stellenbosch, Stellenbosch, South Africa
| | - Heinrich Volschenk
- Department of Microbiology, University of Stellenbosch, Private Bag X1, Matieland, Stellenbosch, 7602, South Africa
| | - Carine Smith
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, University of Stellenbosch, Stellenbosch, South Africa
| | - Leon Dicks
- Department of Microbiology, University of Stellenbosch, Private Bag X1, Matieland, Stellenbosch, 7602, South Africa
| | - Alfred Botha
- Department of Microbiology, University of Stellenbosch, Private Bag X1, Matieland, Stellenbosch, 7602, South Africa.
| |
Collapse
|
39
|
Gu Y, You Y, Yang Y, Liu X, Yang L, Li Y, Zhang C, Yang H, Sha Z, Ma Y, Pang Y, Liu Y. Multifunctional EGCG@ZIF-8 Nanoplatform with Photodynamic Therapy/Chemodynamic Therapy Antibacterial Properties Promotes Infected Wound Healing. ACS APPLIED MATERIALS & INTERFACES 2024; 16:50238-50250. [PMID: 39284745 DOI: 10.1021/acsami.4c08169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
Damaged skin is susceptible to invasion by harmful microorganisms, especially Staphylococcus aureus and Escherichia coli, which can delay healing. Epigallocatechin-3-gallate (EGCG) is a natural compound known for effectively promoting wound healing and its potent anti-inflammatory effects. However, its application is limited due to its susceptibility to oxidation and isomerization, which alter its structure. The use of zeolitic imidazolate framework-8 (ZIF-8) can effectively tackle these issues. This study introduces an oxygen (O2) and hydrogen peroxide (H2O2) self-supplying ZIF-8 nanoplatform designed to enhance the bioavailability of EGCG, combining photodynamic therapy (PDT) and chemodynamic therapy (CDT) to improve antibacterial properties and ultimately accelerate wound healing. For this purpose, EGCG and indocyanine green (ICG), a photosensitizer, were successively integrated into a ZIF-8, and coated with bovine serum albumin (BSA) to enhance biocompatibility. The outer layer of this construct was further modified with manganese dioxide (MnO2) to promote CDT and calcium peroxide (CaO2) to supply H2O2 and O2, resulting in the final nanoplatform EGCG-ICG@ZIF-8/BSA-MnO2/CaO2 (EIZBMC). In in vitro experiments under 808 nm laser, EIZBMC exhibited synergistic antibacterial effects through PDT and CDT. This combination effectively released reactive oxygen species (ROS), which mediated oxidative stress to inhibit the bacteria. Subsequently, in a murine model of wound infection, EIZBMC not only exerted antibacterial effects through PDT and CDT but also alleviated the inflammatory condition and promoted the regeneration of collagen fibers, which led to accelerated wound healing. Overall, this research presents a promising approach to enhancing the therapeutic efficacy of EGCG by leveraging the synergistic antibacterial effects of PDT and CDT. This multifunctional nanoplatform maximizes EGCG's anti-inflammatory properties, offering a potent solution for promoting infected wound healing.
Collapse
Affiliation(s)
- Yufan Gu
- Department of Biophysics, School of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Yuxin You
- Department of Biophysics, School of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Yijia Yang
- Department of Biophysics, School of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Xinyi Liu
- Department of Biophysics, School of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Luyuan Yang
- Department of Biophysics, School of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Yanzhu Li
- Department of Biophysics, School of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Chaoyi Zhang
- Department of Biophysics, School of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Huan Yang
- Department of Biophysics, School of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Ziqi Sha
- Department of Biophysics, School of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Youzhen Ma
- Department of Biophysics, School of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Yipeng Pang
- Department of Biophysics, School of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Yi Liu
- Department of Biophysics, School of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| |
Collapse
|
40
|
Jackson-Litteken CD, Di Venanzio G, Janet-Maitre M, Castro ÍA, Mackel JJ, Rosen DA, López CB, Feldman MF. A chronic murine model of pulmonary Acinetobacter baumannii infection enabling the investigation of late virulence factors, long-term antibiotic treatments, and polymicrobial infections. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.17.613469. [PMID: 39345519 PMCID: PMC11429896 DOI: 10.1101/2024.09.17.613469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Acinetobacter baumannii can cause prolonged infections that disproportionately affect immunocompromised populations. Our understanding of A. baumannii respiratory pathogenesis relies on an acute murine infection model with limited clinical relevance that employs an unnaturally high number of bacteria and requires the assessment of bacterial load at 24-36 hours post-infection. Here, we demonstrate that low intranasal inoculums in immunocompromised mice with a tlr4 mutation leads to reduced inflammation, allowing for persistent infections lasting at least 3 weeks. Using this "chronic infection model," we determined the adhesin InvL is an imperative virulence factor required during later stages of infection, despite being dispensable in the early phase. We also demonstrate that the chronic model enables the distinction between antibiotics that, although initially reduce bacterial burden, either lead to complete clearance or result in the formation of bacterial persisters. To illustrate how our model can be applied to study polymicrobial infections, we inoculated mice with an active A. baumannii infection with Staphylococcus aureus or Klebsiella pneumoniae. We found that S. aureus exacerbates the infection, while K. pneumoniae enhances A. baumannii clearance. In all, the chronic model overcomes some limitations of the acute pulmonary model, expanding our capabilities to study of A. baumannii pathogenesis and lays the groundwork for the development of similar models for other important opportunistic pathogens.
Collapse
Affiliation(s)
- Clay D Jackson-Litteken
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Gisela Di Venanzio
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Manon Janet-Maitre
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Ítalo A Castro
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Center for Women's Infectious Diseases Research, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Joseph J Mackel
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - David A Rosen
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Carolina B López
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Center for Women's Infectious Diseases Research, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Mario F Feldman
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| |
Collapse
|
41
|
Varney AM, Smitten KL, Southam HM, Fairbanks SD, Robertson CC, Thomas JA, McLean S. In Vitro and In Vivo Studies on a Mononuclear Ruthenium Complex Reveals It is a Highly Effective, Fast-Acting, Broad-Spectrum Antimicrobial in Physiologically Relevant Conditions. ACS Infect Dis 2024; 10:3346-3357. [PMID: 39106475 PMCID: PMC11406528 DOI: 10.1021/acsinfecdis.4c00447] [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] [Indexed: 08/09/2024]
Abstract
The crystal structure of a previously reported antimicrobial RuII complex that targets bacterial DNA is presented. Studies utilizing clinical isolates of Gram-negative bacteria that cause catheter-associated urinary tract infection, (CA)UTI, in media that model urine and plasma reveal that good antimicrobial activity is maintained in all conditions tested. Experiments with a series of Staphylococcus aureus clinical isolates show that, unlike the majority of previously reported RuII-based antimicrobial leads, the compound retains its potent activity even in MRSA strains. Furthermore, experiments using bacteria in early exponential growth and at different pHs reveal that the compound also retains its activity across a range of conditions that are relevant to those encountered in clinical settings. Combinatorial studies involving cotreatment with conventional antibiotics or a previously reported analogous dinuclear RuII complex showed no antagonistic effects. In fact, although all combinations show distinct additive antibacterial activity, in one case, this effect approaches synergy. It was found that the Galleria Mellonella model organism infected with a multidrug resistant strain of the ESKAPE pathogen Acinetobacter baumannii could be successfully treated and totally cleared within 48 h after a single dose of the lead complex with no detectable deleterious effect to the host.
Collapse
Affiliation(s)
- Adam M Varney
- School of Science and Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, U.K
- Medical Technologies Innovation Facility (MTIF), Clifton Lane, Nottingham NG11 8NS, U.K
| | - Kirsty L Smitten
- Department of Chemistry, University of Sheffield, Brook Hill, Sheffield S3 7HF, U.K
- School of Bioscience, The University of Sheffield, Western Bank, Sheffield S10 2TN, U.K
| | - Hannah M Southam
- School of Bioscience, The University of Sheffield, Western Bank, Sheffield S10 2TN, U.K
| | - Simon D Fairbanks
- Department of Chemistry, University of Sheffield, Brook Hill, Sheffield S3 7HF, U.K
| | - Craig C Robertson
- Department of Chemistry, University of Sheffield, Brook Hill, Sheffield S3 7HF, U.K
| | - Jim A Thomas
- Department of Chemistry, University of Sheffield, Brook Hill, Sheffield S3 7HF, U.K
| | - Samantha McLean
- School of Science and Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, U.K
| |
Collapse
|
42
|
Guarnieri A, Venditti N, Cutuli MA, Brancazio N, Salvatore G, Magnifico I, Pietrangelo L, Falcone M, Vergalito F, Nicolosi D, Scarsella F, Davinelli S, Scapagnini G, Petronio Petronio G, Di Marco R. Human breast milk isolated lactic acid bacteria: antimicrobial and immunomodulatory activity on the Galleria mellonella burn wound model. Front Cell Infect Microbiol 2024; 14:1428525. [PMID: 39310784 PMCID: PMC11412949 DOI: 10.3389/fcimb.2024.1428525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 08/16/2024] [Indexed: 09/25/2024] Open
Abstract
Introduction Managing burn injuries is a challenge in healthcare. Due to the alarming increase in antibiotic resistance, new prophylactic and therapeutic strategies are being sought. This study aimed to evaluate the potential of live Lactic Acid Bacteria for managing burn infections, using Galleria mellonella larvae as an alternative preclinical animal model and comparing the outcomes with a common antibiotic. Methods The antimicrobial activity of LAB isolated from human breast milk was assessed in vitro against Pseudomonas aeruginosa ATCC 27853. Additionally, the immunomodulatory effects of LAB were evaluated in vivo using the G. mellonella burn wound infection model. Results and discussion In vitro results demonstrated the antimicrobial activity of Lactic Acid Bacteria against P. aeruginosa. In vivo results show that their prophylactic treatment improves, statistically significant, larval survival and modulates the expression of immunity-related genes, Gallerimycin and Relish/NF-κB, strain-dependently. These findings lay the foundation and suggest a promising alternative for burn wound prevention and management, reducing the risk of antibiotic resistance, enhancing immune modulation, and validating the potential G. mellonella as a skin burn wound model.
Collapse
Affiliation(s)
- Antonio Guarnieri
- Università degli Studi del Molise Department of Medicina e Scienze della Salute “V. Tiberio”, Campobasso, Italy
| | - Noemi Venditti
- Università degli Studi del Molise Department of Medicina e Scienze della Salute “V. Tiberio”, Campobasso, Italy
- Unità Operativa (UO) Laboratorio Analisi, Responsible Research Hospital, Campobasso, Italy
| | - Marco Alfio Cutuli
- Università degli Studi del Molise Department of Medicina e Scienze della Salute “V. Tiberio”, Campobasso, Italy
| | - Natasha Brancazio
- Università degli Studi del Molise Department of Medicina e Scienze della Salute “V. Tiberio”, Campobasso, Italy
| | - Giovanna Salvatore
- Università degli Studi del Molise Department of Medicina e Scienze della Salute “V. Tiberio”, Campobasso, Italy
| | - Irene Magnifico
- Università degli Studi del Molise Department of Medicina e Scienze della Salute “V. Tiberio”, Campobasso, Italy
| | - Laura Pietrangelo
- Università degli Studi del Molise Department of Medicina e Scienze della Salute “V. Tiberio”, Campobasso, Italy
| | - Marilina Falcone
- Università degli Studi del Molise Department of Medicina e Scienze della Salute “V. Tiberio”, Campobasso, Italy
| | - Franca Vergalito
- Università degli Studi del Molise Department of Agricultural, Environmental and Food Sciences, Campobasso, Italy
| | - Daria Nicolosi
- Università degli Studi di Catania Department of Drug and Health Sciences, Catania, Italy
| | - Franco Scarsella
- Università degli Studi del Molise Department of Medicina e Scienze della Salute “V. Tiberio”, Campobasso, Italy
- ASReM-Azienda Sanitaria Regionale del Molise, Campobasso, Italy
| | - Sergio Davinelli
- Università degli Studi del Molise Department of Medicina e Scienze della Salute “V. Tiberio”, Campobasso, Italy
| | - Giovanni Scapagnini
- Università degli Studi del Molise Department of Medicina e Scienze della Salute “V. Tiberio”, Campobasso, Italy
| | - Giulio Petronio Petronio
- Università degli Studi del Molise Department of Medicina e Scienze della Salute “V. Tiberio”, Campobasso, Italy
| | - Roberto Di Marco
- Università degli Studi del Molise Department of Medicina e Scienze della Salute “V. Tiberio”, Campobasso, Italy
| |
Collapse
|
43
|
Bertorello S, Cei F, Fink D, Niccolai E, Amedei A. The Future Exploring of Gut Microbiome-Immunity Interactions: From In Vivo/Vitro Models to In Silico Innovations. Microorganisms 2024; 12:1828. [PMID: 39338502 PMCID: PMC11434319 DOI: 10.3390/microorganisms12091828] [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: 08/14/2024] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024] Open
Abstract
Investigating the complex interactions between microbiota and immunity is crucial for a fruitful understanding progress of human health and disease. This review assesses animal models, next-generation in vitro models, and in silico approaches that are used to decipher the microbiome-immunity axis, evaluating their strengths and limitations. While animal models provide a comprehensive biological context, they also raise ethical and practical concerns. Conversely, modern in vitro models reduce animal involvement but require specific costs and materials. When considering the environmental impact of these models, in silico approaches emerge as promising for resource reduction, but they require robust experimental validation and ongoing refinement. Their potential is significant, paving the way for a more sustainable and ethical future in microbiome-immunity research.
Collapse
Affiliation(s)
- Sara Bertorello
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (S.B.); (F.C.); (D.F.); (A.A.)
| | - Francesco Cei
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (S.B.); (F.C.); (D.F.); (A.A.)
| | - Dorian Fink
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (S.B.); (F.C.); (D.F.); (A.A.)
| | - Elena Niccolai
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (S.B.); (F.C.); (D.F.); (A.A.)
- Laboratorio Congiunto MIA-LAB (Microbiome-Immunity Axis Research for a Circular Health), University of Florence, 50134 Florence, Italy
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (S.B.); (F.C.); (D.F.); (A.A.)
- Laboratorio Congiunto MIA-LAB (Microbiome-Immunity Axis Research for a Circular Health), University of Florence, 50134 Florence, Italy
| |
Collapse
|
44
|
Barros MM, Castro J, Araújo D, Oliveira R, Campos AM, Silva S, Outor-Monteiro D, Almeida C. Application of DNA aptamers to block enterotoxigenic Escherichia coli toxicity in a Galleria mellonella larval model. Front Chem 2024; 12:1425903. [PMID: 39268007 PMCID: PMC11390681 DOI: 10.3389/fchem.2024.1425903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is the major bacterial cause of diarrheal diseases in pigs, particularly at young ages, resulting in significant costs to swine farming. The pathogenicity of ETEC is largely dependent on the presence of fimbriae and the ability to produce toxins. Fimbriae are responsible for their initial adhesion to the intestinal epithelial cells, leading to the onset of infection. In particular, the F4 type (K88) fimbriae are often attributed to neonatal infections and have also been associated with post-weaning diarrheal infections. This disease is traditionally prevented or treated with antibiotics, but their use is being severely restricted due to the emergence of resistant bacteria and their impact on human health. Emerging approaches such as aptamers that target the F4-type fimbriae and block the initial ETEC adhesion are a promising alternative. The aim of this study is to assess the effectiveness of two aptamers, Apt31 and Apt37, in controlling ETEC infection in the G. mellonella in vivo model. Initially, the dissociation constant (KD) of each aptamer against ETEC was established using real-time quantitative PCR methodology. Subsequently, different concentrations of the aptamers were injected into Galleria mellonella to study their toxicity. Afterwards, the anti-ETEC potential of Apt31 and Apt37 was assessed in the larvae model. The determined KD was 81.79 nM (95% CI: 31.21-199.4 nM) and 50.71 nM (95% CI: 26.52-96.15 nM) for the Apt31 and Apt37, respectively, showing no statistical difference. No toxicity was observed in G. mellonella following injection with both aptamers at any concentration. However, the administration of Apt31 together with ETEC-F4+ in G. mellonella resulted in a significant improvement of approximately 30% in both larvae survival and health index compared to ETEC-F4+ alone. These findings suggest that aptamers have promising inhibitory effect against ETEC infections and pave the way for additional in vivo studies.
Collapse
Affiliation(s)
- Maria Margarida Barros
- National Institute for Agrarian and Veterinariay Research (INIAV), Vairão, Portugal
- Veterinary and Animal Research Centre (CECAV), University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
| | - Joana Castro
- National Institute for Agrarian and Veterinariay Research (INIAV), Vairão, Portugal
- Centre of Biological Engineering, University of Minho, Braga, Portugal
| | - Daniela Araújo
- National Institute for Agrarian and Veterinariay Research (INIAV), Vairão, Portugal
- Centre of Biological Engineering, University of Minho, Braga, Portugal
- LABBELS-Associate Laboratory, Braga, Portugal
| | - Ricardo Oliveira
- National Institute for Agrarian and Veterinariay Research (INIAV), Vairão, Portugal
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Porto, Portugal
- ALiCE-Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Porto, Portugal
| | - Ana Maria Campos
- National Institute for Agrarian and Veterinariay Research (INIAV), Vairão, Portugal
| | - Sónia Silva
- National Institute for Agrarian and Veterinariay Research (INIAV), Vairão, Portugal
- Centre of Biological Engineering, University of Minho, Braga, Portugal
- LABBELS-Associate Laboratory, Braga, Portugal
| | - Divanildo Outor-Monteiro
- Veterinary and Animal Research Centre (CECAV), University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
| | - Carina Almeida
- National Institute for Agrarian and Veterinariay Research (INIAV), Vairão, Portugal
- Centre of Biological Engineering, University of Minho, Braga, Portugal
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Porto, Portugal
- ALiCE-Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Porto, Portugal
| |
Collapse
|
45
|
Costabile G, Baldassi D, Müller C, Groß B, Ungaro F, Schubert S, Firestine SM, Merkel OM. Antibiotic-loaded nanoparticles for the treatment of intracellular methicillin-resistant Staphylococcus Aureus infections: In vitro and in vivo efficacy of a novel antibiotic. J Control Release 2024; 374:454-465. [PMID: 39181163 DOI: 10.1016/j.jconrel.2024.08.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/29/2024] [Accepted: 08/19/2024] [Indexed: 08/27/2024]
Abstract
Antimicrobial resistance is considered one of the biggest threats to public health worldwide. Methicillin-resistant S. aureus is the causative agent of a number of infections and lung colonization in people suffering from cystic fibrosis. Moreover, a growing body of evidence links the microbiome to the development of cancer, as well as to the success of the treatment. In this view, the development of novel antibiotics is of critical importance, and SV7, a novel antibiotic active against MRSA at low concentrations, represents a promising candidate. However, the low aqueous solubility of SV7 hampers its therapeutic translation. In this study, SV7 was encapsulated in poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) to improve the solubility profile, to ensure sustained release and eventually support deposition in the airways. Furthermore, PLGA NPs were formulated as dry powder to extend their shelf-life and were shown to efficiently target intracellular infections. After identifying a formulation with suitable physico-chemical characteristics, SV7-loaded NPs were investigated in vitro in terms of inhibitory activity against MRSA, and their safety profile in lung epithelial cells. Subsequently, the activity against MRSA intracellular infections was investigated in a co-culture model of MRSA and macrophages. To test the translatability of our findings, SV7-loaded NPs were tested in vivo in a Galleria mellonella infection model. In conclusion, SV7-loaded NPs showed a safe profile and efficient inhibitory activity against MRSA at low concentrations. Furthermore, their activity against intracellular infections was confirmed, and was retained in vivo, rendering them a promising candidate for treatment of MRSA lung infections.
Collapse
Affiliation(s)
- Gabriella Costabile
- Department of Pharmacy, Pharmaceutical Technology & Biopharmaceutics, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377 Munich, DE, Germany; Department of Pharmacy, University of Napoli Federico II, Via Domenico Montesano 49, 80131 Napoli, IT, Italy
| | - Domizia Baldassi
- Department of Pharmacy, Pharmaceutical Technology & Biopharmaceutics, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377 Munich, DE, Germany
| | - Christoph Müller
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377 Munich, DE, Germany
| | - Birgit Groß
- Max von Pettenkofer-Institut Munich für Hygiene und Medizinische Mikrobiologie, Elisabeth-Winterhalter-Weg 6, 81377 Munich, DE, Germany
| | - Francesca Ungaro
- Department of Pharmacy, University of Napoli Federico II, Via Domenico Montesano 49, 80131 Napoli, IT, Italy
| | - Sören Schubert
- Max von Pettenkofer-Institut Munich für Hygiene und Medizinische Mikrobiologie, Elisabeth-Winterhalter-Weg 6, 81377 Munich, DE, Germany
| | - Steven M Firestine
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Eugene Applebaum College of Pharmacy and Health Sciences, 259 Mack Ave, Detroit, MI 48201, USA
| | - Olivia M Merkel
- Department of Pharmacy, Pharmaceutical Technology & Biopharmaceutics, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377 Munich, DE, Germany.
| |
Collapse
|
46
|
Berryhill BA, Gil-Gil T, Witzany C, Goldberg DA, Vega NM, Regoes RR, Levin BR. The dynamics of Staphylococcal infection and their treatment with antibiotics and bacteriophage in the Galleria mellonella model system. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.23.609294. [PMID: 39229007 PMCID: PMC11370618 DOI: 10.1101/2024.08.23.609294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Critical to our understanding of infections and their treatment is the role the innate immune system plays in controlling bacterial pathogens. Nevertheless, many in vivo systems are made or modified such that they do not have an innate immune response. Use of these systems denies the opportunity to examine the synergy between the immune system and antimicrobial agents. In this study we demonstrate that the larva of Galleria mellonella is an effective in vivo model for the study of the population and evolutionary biology of bacterial infections and their treatment. To do this we test three hypotheses concerning the role of the innate immune system during infection. We show: i) sufficiently high densities of bacteria are capable of saturating the innate immune system, ii) bacteriostatic drugs and bacteriophages are as effective as bactericidal antibiotics in preventing mortality and controlling bacterial densities, and iii) minority populations of bacteria resistant to a treating antibiotic will not ascend. Using a highly virulent strain of Staphylococcus aureus and a mathematical computer-simulation model, we further explore how the dynamics of the infection within the short term determine the ultimate infection outcome. We find that excess immune activation in response to high densities of bacteria leads to a strong but short-lived immune response which ultimately results in a high degree of mortality. Overall, our findings illustrate the utility of the G. mellonella model system in conjunction with established in vivo models in studying infectious disease progression and treatment.
Collapse
Affiliation(s)
- Brandon A. Berryhill
- Department of Biology, Emory University; Atlanta, Georgia, 30322, USA
- Program in Microbiology and Molecular Genetics, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University; Atlanta, GA, 30322, USA
| | - Teresa Gil-Gil
- Department of Biology, Emory University; Atlanta, Georgia, 30322, USA
| | | | - David A. Goldberg
- Department of Biology, Emory University; Atlanta, Georgia, 30322, USA
| | - Nic M. Vega
- Department of Biology, Emory University; Atlanta, Georgia, 30322, USA
| | - Roland R. Regoes
- Institute of Integrative Biology, ETH Zurich, Zurich, Switzerland
| | - Bruce R. Levin
- Department of Biology, Emory University; Atlanta, Georgia, 30322, USA
| |
Collapse
|
47
|
Banfi D, Bianchi T, Mastore M, Brivio MF. Optimization of Experimental Infection of the Animal Model Galleria mellonella Linnaeus 1758 (Lepidoptera: Pyralidae) with the Gram-Positive Bacterium Micrococcus luteus. INSECTS 2024; 15:618. [PMID: 39194822 DOI: 10.3390/insects15080618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024]
Abstract
The aim of this work was to develop an experimental protocol for the infection of Galleria mellonella with Gram-positive bacteria. Some physiological characteristics of these insects are comparable to those of vertebrates, therefore allowing the replacement of mammals in the preclinical phases of drug development. G. mellonella Linnaeus 1758 (Lepidoptera: Pyralidae) is accepted as an alternative model for the study of infectious diseases. Since data on infection procedures with different bacterial strains are scarce and sometimes conflicting, also due to different and non-uniform protocols, we developed an experimental protocol that would allow for controlled and repeatable infections, using the Gram-positive bacterium GRAS (Generally Regarded As Safe) Micrococcus luteus. After analyzing the morphology and defining the growth rate of M. luteus, doses of between 101 and 106 CFU/larvae were administered to late-stage larvae. The survival rate of the larvae was monitored up to 7 days and the LD50 determined. The bacterial clearance capacity of the larvae after injection with 103 and 105 CFU/larvae was assessed by hemolymph bacterial load analysis. The results made it possible to define the growth curve of M. luteus correlated with the CFU count; based on the LD50 (103.8 CFU/larvae) calculated on the survival of G. mellonella, infections were carried out to evaluate the immune efficiency of the larvae in bacterial clearance. This protocol, standardized on G. mellonella larvae, could provide a functional tool to study the course of bacterial infections.
Collapse
Affiliation(s)
- Davide Banfi
- Laboratory of Applied Entomology and Parasitology, Department of Theoretical and Applied Sciences (DiSTA), University of Insubria, 21100 Varese, Italy
| | - Tommaso Bianchi
- Laboratory of Applied Entomology and Parasitology, Department of Theoretical and Applied Sciences (DiSTA), University of Insubria, 21100 Varese, Italy
| | - Maristella Mastore
- Laboratory of Applied Entomology and Parasitology, Department of Theoretical and Applied Sciences (DiSTA), University of Insubria, 21100 Varese, Italy
| | - Maurizio Francesco Brivio
- Laboratory of Applied Entomology and Parasitology, Department of Theoretical and Applied Sciences (DiSTA), University of Insubria, 21100 Varese, Italy
| |
Collapse
|
48
|
Grünfeld M. Telling Ecopoetic Stories: Wax Worms, Care, and the Cultivation of Other Sensibilities. THE JOURNAL OF MEDICAL HUMANITIES 2024:10.1007/s10912-024-09878-6. [PMID: 39145849 DOI: 10.1007/s10912-024-09878-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/07/2024] [Indexed: 08/16/2024]
Abstract
Recently, a beekeeper discovered the metabolic wizardry of wax worms, their ability to decompose polyethylene. While this organism has usually been perceived as a model organism in science or a pest to beekeepers, it acquired a new mode of being as potentially probiotic, inviting us to dream of a future without plastic waste. In this paper, I explore how wax worms are entangled with material practices of care and narratives that give meaning to these practices. These stories, however, are marked by manipulation, exploitation, and extermination, and call for a questioning of our modes of caring. Consequently, I offer a counter-narrative that questions our anthropocentric practices of caring and the stories we attach to them. Borrowing Puig de la Bellacasa's notion of ecopoetics, I tell another story based on my participation in the making of an art installation hosting wax worms. The installation creates an opening of a world of curiosity and cultivates a sensibility for wax worms expanding their modes of being and our capabilities of appreciation. In the end, I argue that by mattering and storying differently, we have the opportunity to challenge anthropocentric interests and make a different world of caring and co-existence possible.
Collapse
Affiliation(s)
- Martin Grünfeld
- Department of Science Education, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
49
|
Schaefer S, Vij R, Sprague JL, Austermeier S, Dinh H, Judzewitsch PR, Müller-Loennies S, Lopes Silva T, Seemann E, Qualmann B, Hertweck C, Scherlach K, Gutsmann T, Cain AK, Corrigan N, Gresnigt MS, Boyer C, Lenardon MD, Brunke S. A synthetic peptide mimic kills Candida albicans and synergistically prevents infection. Nat Commun 2024; 15:6818. [PMID: 39122699 PMCID: PMC11315985 DOI: 10.1038/s41467-024-50491-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 07/11/2024] [Indexed: 08/12/2024] Open
Abstract
More than two million people worldwide are affected by life-threatening, invasive fungal infections annually. Candida species are the most common cause of nosocomial, invasive fungal infections and are associated with mortality rates above 40%. Despite the increasing incidence of drug-resistance, the development of novel antifungal formulations has been limited. Here we investigate the antifungal mode of action and therapeutic potential of positively charged, synthetic peptide mimics to combat Candida albicans infections. Our data indicates that these synthetic polymers cause endoplasmic reticulum stress and affect protein glycosylation, a mode of action distinct from currently approved antifungal drugs. The most promising polymer composition damaged the mannan layer of the cell wall, with additional membrane-disrupting activity. The synergistic combination of the polymer with caspofungin prevented infection of human epithelial cells in vitro, improved fungal clearance by human macrophages, and significantly increased host survival in a Galleria mellonella model of systemic candidiasis. Additionally, prolonged exposure of C. albicans to the synergistic combination of polymer and caspofungin did not lead to the evolution of tolerant strains in vitro. Together, this work highlights the enormous potential of these synthetic peptide mimics to be used as novel antifungal formulations as well as adjunctive antifungal therapy.
Collapse
Affiliation(s)
- Sebastian Schaefer
- School of Chemical Engineering, University of New South Wales (UNSW), Sydney, NSW, Australia
- Australian Centre for NanoMedicine, UNSW, Sydney, NSW, Australia
- School of Biotechnology and Biomolecular Sciences, UNSW, Sydney, NSW, Australia
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
| | - Raghav Vij
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
| | - Jakob L Sprague
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
| | - Sophie Austermeier
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
| | - Hue Dinh
- ARC Centre of Excellence in Synthetic Biology, School of Natural Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Peter R Judzewitsch
- School of Chemical Engineering, University of New South Wales (UNSW), Sydney, NSW, Australia
- Australian Centre for NanoMedicine, UNSW, Sydney, NSW, Australia
| | - Sven Müller-Loennies
- Division of Biophysics, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Taynara Lopes Silva
- Department of Biomolecular Chemistry, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
| | - Eric Seemann
- Institute of Biochemistry I, Jena University Hospital - Friedrich Schiller University Jena, Jena, Germany
| | - Britta Qualmann
- Institute of Biochemistry I, Jena University Hospital - Friedrich Schiller University Jena, Jena, Germany
| | - Christian Hertweck
- Department of Biomolecular Chemistry, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
- Faculty of Biological Sciences, Friedrich Schiller University Jena, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany
| | - Kirstin Scherlach
- Department of Biomolecular Chemistry, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
| | - Thomas Gutsmann
- Division of Biophysics, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
- Centre for Structural Systems Biology (CSSB), Hamburg, Germany
| | - Amy K Cain
- ARC Centre of Excellence in Synthetic Biology, School of Natural Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Nathaniel Corrigan
- School of Chemical Engineering, University of New South Wales (UNSW), Sydney, NSW, Australia
- Australian Centre for NanoMedicine, UNSW, Sydney, NSW, Australia
| | - Mark S Gresnigt
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
| | - Cyrille Boyer
- School of Chemical Engineering, University of New South Wales (UNSW), Sydney, NSW, Australia.
- Australian Centre for NanoMedicine, UNSW, Sydney, NSW, Australia.
| | - Megan D Lenardon
- School of Biotechnology and Biomolecular Sciences, UNSW, Sydney, NSW, Australia.
| | - Sascha Brunke
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany.
| |
Collapse
|
50
|
Zhou H, Du X, Wang Y, Kong J, Zhang X, Wang W, Sun Y, Zhou C, Zhou T, Ye J. Antimicrobial peptide A20L: in vitro and in vivo antibacterial and antibiofilm activity against carbapenem-resistant Klebsiella pneumoniae. Microbiol Spectr 2024; 12:e0397923. [PMID: 38980018 PMCID: PMC11302274 DOI: 10.1128/spectrum.03979-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 06/07/2024] [Indexed: 07/10/2024] Open
Abstract
Antimicrobial resistance has become a growing public health threat in recent years. Klebsiella pneumoniae is one of the priority pathogens listed by the World Health Organization. Antimicrobial peptides are considered promising alternatives to antibiotics due to their broad-spectrum antibacterial activity and low resistance. In this study, we investigated the antibacterial activity of antimicrobial peptide A20L against K. pneumoniae. In vitro antibacterial activity of A20L against K. pneumoniae was demonstrated by broth microdilution method. We confirmed the in vivo efficacy of A20L by Galleria mellonella infection model. In addition, we found that A20L also had certain antibiofilm activity by crystal violet staining. We also evaluated the safety and stability of A20L, and the results revealed that at a concentration of ≤128 µg/mL, A20L exhibited negligible toxicity to RAW264.7 cells and no substantial toxicity to G. mellonella. A20L was stable at different temperatures and with low concentration of serum [5% fetal bovine serum (FBS)]; however, Ca2+, Mg2+, and high serum concentrations reduced the antibacterial activity of A20L. Scanning electron microscope (SEM) and membrane permeability tests revealed that A20L may exhibit antibacterial action by damaging bacterial cell membranes and increasing the permeability of outer membrane. Taken together, our results suggest that A20L has significant development potential as a therapeutic antibiotic alternative, which provides ideas for the treatment of K. pneumoniae infection. IMPORTANCE A20L showed antibacterial and anti-infective efficacy in vitro and in vivo against Klebsiella pneumoniae. It can have an antibacterial effect by disrupting the integrity of cell membranes. A20L displayed anti-biofilm and anti-inflammatory activity against carbapenem-resistant K. pneumoniae and certain application potential in vivo, which provides a new idea for the clinical treatment of biofilm-associated infections.
Collapse
Affiliation(s)
- Huijing Zhou
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University; Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Xin Du
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University; Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Yue Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University; Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Jingchun Kong
- Department of Medical Lab Science, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaodong Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University; Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Weixiang Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University; Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Yao Sun
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University; Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Cui Zhou
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University; Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Tieli Zhou
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University; Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Jianzhong Ye
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University; Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China
| |
Collapse
|