51
|
Sengupta B, Alrubayan M, Wang Y, Mallet E, Torres A, Solis R, Wang H, Pradhan P. An AI-directed analytical study on the optical transmission microscopic images of Pseudomonas aeruginosa in planktonic and biofilm states. ARXIV 2024:arXiv:2412.18205v1. [PMID: 39764404 PMCID: PMC11703328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2025]
Abstract
Biofilms are resistant microbial cell aggregates that pose risks to health and food industries and produce environmental contamination. Accurate and efficient detection and prevention of biofilms are challenging and demand interdisciplinary approaches. This multidisciplinary research reports the application of a deep learning-based artificial intelligence (AI) model for detecting biofilms produced by Pseudomonas aeruginosa with high accuracy. Aptamer DNA templated silver nanocluster (Ag-NC) was used to prevent biofilm formation, which produced images of the planktonic states of the bacteria. Large-volume bright field images of bacterial biofilms were used to design the AI model. In particular, we used U-Net with ResNet encoder enhancement to segment biofilm images for AI analysis. Different degrees of biofilm structures can be efficiently detected using ResNet18 and ResNet34 backbones. The potential applications of this technique are also discussed.
Collapse
Affiliation(s)
- Bidisha Sengupta
- Department of Chemistry & Biochemistry, Stephen F. Austin State University, Nacogdoches, TX, 75962
- These authors have equal contributions
| | - Mousa Alrubayan
- Department of Physics and Astronomy, Mississippi State University, Mississippi State, MS 39762
- These authors have equal contributions
| | - Yibin Wang
- Department of Industrial Engineering, Mississippi State University, Mississippi State, MS 39762
- These authors have equal contributions
| | - Esther Mallet
- Department of Chemistry & Biochemistry, Stephen F. Austin State University, Nacogdoches, TX, 75962
| | - Angel Torres
- Department of Chemistry & Biochemistry, Stephen F. Austin State University, Nacogdoches, TX, 75962
| | - Ravyn Solis
- Department of Chemistry & Biochemistry, Stephen F. Austin State University, Nacogdoches, TX, 75962
| | - Haifeng Wang
- Department of Industrial Engineering, Mississippi State University, Mississippi State, MS 39762
| | - Prabhakar Pradhan
- Department of Physics and Astronomy, Mississippi State University, Mississippi State, MS 39762
| |
Collapse
|
52
|
Tang D, Liu Y, Yao H, Lin Y, Xi Y, Li M, Mao A. Transcriptome Analysis Reveals the Mechanism of Y0-C10-HSL on Biofilm Formation and Motility of Pseudomonas aeruginosa. Pharmaceuticals (Basel) 2024; 17:1719. [PMID: 39770562 PMCID: PMC11678461 DOI: 10.3390/ph17121719] [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: 08/26/2024] [Revised: 10/12/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Background:Pseudomonas aeruginosa (P. aeruginosa) is a type of pathogen that takes advantage of opportunities to infect and form biofilm during infection. Inhibiting biofilm formation is a promising approach for the treatment of biofilm-related infections. Methods: Here, Y0-C10-HSL (N-cyclopentyl-n-decanamide) was designed, synthesized, and tested for its effect on biofilm formation, motility, and the Caenorhabditis elegans (C. elegans) survival assay. In addition, the molecular mechanism of Y0-C10-HSL on P. aeruginosa biofilm formation was explored using transcriptome analysis. Results: At a concentration of 200 μmol/L Y0-C10-HSL, biofilm and exopolysaccharides were decreased by 38.5% and 29.3%, respectively; Y0-C10-HSL effectively dispersed the pre-formed biofilm and inhibited the motility ability of P. aeruginosa; and the C. elegans survival assay showed that Y0-C10-HSL was safe and provided protection to C. elegans against P. aeruginosa infection (the survival rates of C. elegans were higher than 74% and increased by 39%, 35.1%, and 47.5%, respectively, when treated with 200 μmol/L Y0-C10-HSL at 24, 48, and 80 h). Transcriptome analysis showed that 585 differentially expressed genes (DEGs) were found after treatment with 200 μmol/L Y0-C10-HSL, including 254 up-regulated DEGs and 331 down-regulated DEGs. The genes involved in the quorum sensing system and biofilm formation were down-regulated. Conclusions: Y0-C10-HSL inhibited the biofilm formation and dispersed the pre-formed biofilm of P. aeruginosa through down-regulated genes related to quorum sensing pathways and biofilm formation. These findings provide a theoretical foundation for the treatment and prevention of antibiotic resistance in clinical and environmental microorganisms such as P. aeruginosa.
Collapse
Affiliation(s)
- Deping Tang
- School of Biological & Pharmaceutical Engineering, Lanzhou Jiaotong University, Lanzhou 730070, China; (D.T.)
| | - Yali Liu
- School of Biological & Pharmaceutical Engineering, Lanzhou Jiaotong University, Lanzhou 730070, China; (D.T.)
| | - Huihui Yao
- School of Biological & Pharmaceutical Engineering, Lanzhou Jiaotong University, Lanzhou 730070, China; (D.T.)
| | - Yanyan Lin
- School of Biological & Pharmaceutical Engineering, Lanzhou Jiaotong University, Lanzhou 730070, China; (D.T.)
| | - Yanpeng Xi
- School of Biological & Pharmaceutical Engineering, Lanzhou Jiaotong University, Lanzhou 730070, China; (D.T.)
| | - Mengjiao Li
- School of Biological & Pharmaceutical Engineering, Lanzhou Jiaotong University, Lanzhou 730070, China; (D.T.)
| | - Aihong Mao
- School of Biological & Pharmaceutical Engineering, Lanzhou Jiaotong University, Lanzhou 730070, China; (D.T.)
- Gansu Provincial Academic Institute for Medical Research, Lanzhou 730050, China
| |
Collapse
|
53
|
Teixeira FL, Pauer H, Valente GLC, de Paula GR. Comparative analysis of virulence factors produced by Pseudomonas aeruginosa strains isolated from chronic wounds or bloodstream infections. MICROPUBLICATION BIOLOGY 2024; 2024:10.17912/micropub.biology.001413. [PMID: 39748833 PMCID: PMC11688615 DOI: 10.17912/micropub.biology.001413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/12/2024] [Accepted: 12/17/2024] [Indexed: 01/04/2025]
Abstract
Pseudomonas aeruginosa is an important pathogen associated with both chronic wounds and bloodstream infections. Virulence factors required for the establishment of acute and chronic infections differ substantially. Since bacteremia can be a severe outcome of wound colonization, we performed a comparative analysis of virulence between P. aeruginosa strains isolated from the bloodstream and chronic wounds. Our results show that, in general, P. aeruginosa strains isolated from bloodstream infections are more virulent than strains that colonize chronic wounds.
Collapse
Affiliation(s)
- Felipe L. Teixeira
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, United States
- Departamento de Tecnologia Farmacêutica, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Heidi Pauer
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, United States
- Instituto Nacional de Ciência e Tecnologia de Inovação em Doenças de Populações Negligenciadas, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gabriel Luis C. Valente
- Departamento de Tecnologia Farmacêutica, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Geraldo Renato de Paula
- Departamento de Tecnologia Farmacêutica, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| |
Collapse
|
54
|
Wang M, Wang Y, Chen G, Gao H, Peng Q. Chitosan-Based Multifunctional Biomaterials as Active Agents or Delivery Systems for Antibacterial Therapy. Bioengineering (Basel) 2024; 11:1278. [PMID: 39768096 PMCID: PMC11673874 DOI: 10.3390/bioengineering11121278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/07/2024] [Accepted: 12/12/2024] [Indexed: 01/04/2025] Open
Abstract
Antibiotic therapy has been a common method for treating bacterial infections over the past century, but with the rise in bacterial resistance caused by antibiotic abuse, better control and more rational use of antibiotics have been increasingly demanded. At the same time, a journey to explore alternatives to antibiotic therapies has also been undertaken. Chitosan and its derivatives, materials with good biocompatibility, biodegradability, and excellent antibacterial properties, have garnered significant attention, and more and more studies on chitosan and its derivatives have been conducted in recent years. In this work, we aim to elucidate the biological properties of chitosan and its derivatives and to track their clinical applications, as well as to propose issues that need to be addressed and possible solutions to further their future development and application.
Collapse
Affiliation(s)
| | | | | | | | - Qiang Peng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
55
|
Xue F, Ragno M, Blackburn SA, Fasseas M, Maitra S, Liang M, Rai S, Mastroianni G, Tholozan F, Thompson R, Sellars L, Hall R, Saunter C, Weinkove D, Ezcurra M. New tools to monitor Pseudomonas aeruginosa infection and biofilms in vivo in C. elegans. Front Cell Infect Microbiol 2024; 14:1478881. [PMID: 39737329 PMCID: PMC11683784 DOI: 10.3389/fcimb.2024.1478881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 11/08/2024] [Indexed: 01/01/2025] Open
Abstract
Introduction Antimicrobial resistance is a growing health problem. Pseudomonas aeruginosa is a pathogen of major concern because of its multidrug resistance and global threat, especially in health-care settings. The pathogenesis and drug resistance of P. aeruginosa depends on its ability to form biofilms, making infections chronic and untreatable as the biofilm protects against antibiotics and host immunity. A major barrier to developing new antimicrobials is the lack of in vivo biofilm models. Standard microbiological testing is usually performed in vitro using planktonic bacteria, without representation of biofilms, reducing translatability. Here we develop tools to study both infection and biofilm formation by P. aeruginosa in vivo to accelerate development of strategies targeting infection and pathogenic biofilms. Methods Biofilms were quantified in vitro using Crystal Violet staining and fluorescence biofilm assays. For in vivo assays, C. elegans were infected with P. aeruginosa strains. Pathogenicity was quantified by measuring healthspan, survival and GFP fluorescence. Healthspan assays were performed using the WormGazerTM automated imaging technology. Results Using the nematode Caenorhabditis elegans and P. aeruginosa reporters combined with in vivo imaging we show that fluorescent P. aeruginosa reporters that form biofilms in vitro can be used to visualize tissue infection. Using automated tracking of C. elegans movement, we find that that the timing of this infection corresponds with a decline in health endpoints. In a mutant strain of P. aeruginosa lacking RhlR, a transcription factor that controls quorum sensing and biofilm formation, we find reduced capacity of P. aeruginosa to form biofilms, invade host tissues and negatively impact healthspan and survival. Discussion Our findings suggest that RhlR could be a new antimicrobial target to reduce P. aeruginosa biofilms and virulence in vivo and C. elegans could be used to more effectively screen for new drugs to combat antimicrobial resistance.
Collapse
Affiliation(s)
- Feng Xue
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| | - Martina Ragno
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| | | | - Michael Fasseas
- Magnitude Biosciences Limited, NETPark Plexus, Sedgefield, United Kingdom
| | - Sushmita Maitra
- Magnitude Biosciences Limited, NETPark Plexus, Sedgefield, United Kingdom
| | - Mingzhi Liang
- School of Biosciences, University of Kent, Canterbury, United Kingdom
- Quadram Institute Bioscience, Norwich Research Park, Norwich, United Kingdom
| | - Subash Rai
- The NanoVision Centre, Queen Mary University of London, London, United Kingdom
| | - Giulia Mastroianni
- The NanoVision Centre, Queen Mary University of London, London, United Kingdom
| | | | - Rachel Thompson
- Perfectus Biomed Group, Sci-Tech Daresbury, Chesire, United Kingdom
| | - Laura Sellars
- Perfectus Biomed Group, Sci-Tech Daresbury, Chesire, United Kingdom
| | - Rebecca Hall
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| | - Chris Saunter
- Magnitude Biosciences Limited, NETPark Plexus, Sedgefield, United Kingdom
| | - David Weinkove
- Magnitude Biosciences Limited, NETPark Plexus, Sedgefield, United Kingdom
- Department of Biosciences, Durham University, Durham, United Kingdom
| | - Marina Ezcurra
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| |
Collapse
|
56
|
Wale YM, Roberts JA, Sime FB. Dynamic In Vitro PK/PD Infection Models for the Development and Optimisation of Antimicrobial Regimens: A Narrative Review. Antibiotics (Basel) 2024; 13:1201. [PMID: 39766591 PMCID: PMC11672834 DOI: 10.3390/antibiotics13121201] [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: 11/07/2024] [Revised: 12/05/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
The antimicrobial concentration-time profile in humans affects antimicrobial activity, and as such, it is critical for preclinical infection models to simulate human-like dynamic concentration-time profiles for maximal translatability. This review discusses the setup, principle, and application of various dynamic in vitro PK/PD infection models commonly used in the development and optimisation of antimicrobial treatment regimens. It covers the commonly used dynamic in vitro infection models, including the one-compartment model, hollow fibre infection model, biofilm model, bladder infection model, and aspergillus infection model. It summarises the mathematical methods for the simulation of the pharmacokinetic profile of single or multiple antimicrobials when using the serial or parallel configurations of in vitro systems. Dynamic in vitro models offer reliable pharmacokinetic/pharmacodynamic data to help define the initial dosing regimens of new antimicrobials that can be developed further in clinical trials. They can also help in the optimisation of dosing regimens for existing antimicrobials, especially in the presence of emerging antimicrobial resistance. In conclusion, dynamic in vitro infection models replicate the interactions that occur between microorganisms and dynamic antimicrobial exposures in the human body to generate data highly predictive of the clinical efficacy. They are particularly useful for the development new treatment strategies against antimicrobial-resistant pathogens.
Collapse
Affiliation(s)
- Yalew M. Wale
- Centre for Clinical Research (UQCCR), Faculty of Medicine, The University of Queensland, Brisbane, QLD 4029, Australia
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos P.O. Box 269, Ethiopia
| | - Jason A. Roberts
- Centre for Clinical Research (UQCCR), Faculty of Medicine, The University of Queensland, Brisbane, QLD 4029, Australia
- Departments of Pharmacy and Intensive Care Medicine, Royal Brisbane and Women’s Hospital, Brisbane, QLD 4006, Australia
- Division of Anesthesia Critical Care and Emergency and Pain Medicine, Nimes University Hospital, University of Montpellier, UR UM 103, 34090 Nimes, France
- Herston Infectious Diseases Institute (HeIDI), Metro North Health, Brisbane, QLD 4006, Australia
| | - Fekade B. Sime
- Centre for Clinical Research (UQCCR), Faculty of Medicine, The University of Queensland, Brisbane, QLD 4029, Australia
| |
Collapse
|
57
|
Ma Z, Zeng W, Liu H, Chen H, Ye L, Liu S, Qian C, Zhou T, Cao J. Characterization of novel sequence type 12531 and O8:H7 serotype carbapenem-resistant Escherichia coli with strong swimming and intestinal epithelial cell barrier migration abilities. Antimicrob Agents Chemother 2024; 68:e0080524. [PMID: 39440955 PMCID: PMC11619422 DOI: 10.1128/aac.00805-24] [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: 06/02/2024] [Accepted: 09/19/2024] [Indexed: 10/25/2024] Open
Abstract
Carbapenem-resistant Enterobacteriaceae have become widely prevalent globally because of antibiotic misuse and the spread of drug-resistant plasmids, where carbapenem-resistant Escherichia coli (CREC) is one of the most common and prevalent pathogens. Furthermore, E. coli has been identified as a member of normal gut flora and does not cause disease under normal circumstances. However, certain strains of E. coli, due to the expression of virulence genes, can cause severe intestinal and extra-intestinal infections. Therefore, clinically, drug resistance and pathogenic E. coli strains are significantly challenging to treat. In this study, a novel CREC strain DC8855 was isolated from the ascites of a patient with intestinal perforation, identified as a novel sequence type 12531 (ST12531) and an unreported serotype O8:H7. It was revealed that the resistance of ST12531 CREC was predominantly conferred by an IncFII(K) plasmid carrying blaNDM-4. Furthermore, phylogenetic analysis indicated that this is the first discovery of such plasmids in China and the first identification in E. coli. Moreover, regarding virulence, the swimming assays, qRT-PCR, and in vitro intestinal barrier model indicated that DC8855 had significantly higher motility, flagella gene expression, and intestinal epithelial cell barrier migration ability than the other sequence types CREC strains (ST167 and ST410). In conclusion, this study identified novel CREC which was multidrug resistant as well as enteropathogenic and therefore requires continuous monitoring.
Collapse
Affiliation(s)
- Zhexiao Ma
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- 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 Province, China
| | - Weiliang Zeng
- 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 Province, China
| | - Haifeng Liu
- 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 Province, China
| | - Huanchang Chen
- 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 Province, China
| | - Lulu 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 Province, China
| | - Sichen Liu
- 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 Province, China
| | - Changrui Qian
- 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 Province, 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 Province, China
| | - Jianming Cao
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| |
Collapse
|
58
|
Thakur B, Kaur S, Tripathi M, Upadhyay SK. Exploring the potential of lactic acid bacteria and its molecular mechanism of action in the development of biosurfactants: Current finding and future outlook. Biotechnol Genet Eng Rev 2024; 40:4737-4768. [PMID: 37226486 DOI: 10.1080/02648725.2023.2216421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/15/2023] [Indexed: 05/26/2023]
Abstract
Biosurfactants generated from lactic acid bacteria (LAB) offer an advantage over standard microbial surfactants due to their antifungal, antibacterial and antiviral capabilities. Many LAB strains have been related to the manufacture of biosurfactant, an essential chemical with uses in the treatment of a number of illnesses. Furthermore, their effectiveness as anti-adhesive agents against a diverse variety of pathogens proves their utility as anti-adhesive coating agents for medical insertional materials, reducing hospital infections without the need of synthetic drugs and chemicals. LAB produces both low and high molecular weight biosurfactants. Biosurfactants from L. pentosus, L. gasseri and L. jensenii have been reported to produce glycolipopeptides that comprise carbohydrates, proteins and lipids in the ratio of 1:3:6 with palmitic, stearic acid, and linoelaidic acid as the major fatty acid component, whereas L. plantarum has been reported to make surlactin due to the presence of non-ribosomal peptide synthetase genes (NRPS) genes. Antimicrobial activity of sophorolipids and rhamnolipids generated from LAB against B. subtilis, P. aeruginosa, S. epidermidis, Propionibacterium acnes and E. coli has been demonstrated. The safety of biosurfactants is being evaluated in compliance with a number of regulatory standards that emphasize the importance of safety in the pharmaceutical industry. This review attempts, for the first time, to provide a comprehensive evaluation of several approaches for the synthesis of biosurfactant-mediated molecular modulation in terms of their biological value. Future biosurfactant directions, as well as regulatory considerations that are crucial for the synthesis of biosurfactants from novel LAB, have also been explored.
Collapse
Affiliation(s)
- Babita Thakur
- Department of Biotechnology, Chandigarh University, Mohali, India
| | | | - Manikant Tripathi
- Biotechnology Program, Dr. Rammanohar Lohia Avadh University, Ayodhya, India
| | - Sudhir K Upadhyay
- Department of Environmental Science, Veer Bahadur Singh Purvanchal University, Jaunpur, India
| |
Collapse
|
59
|
Ferreira Gomes LH, Corrêa PR, Schwarz MGA, Mendonça-Lima L. Functional impact of a deletion in Mycobacterium bovis BCG Moreau celA1 gene. Tuberculosis (Edinb) 2024; 149:102576. [PMID: 39546869 DOI: 10.1016/j.tube.2024.102576] [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: 09/17/2024] [Revised: 11/05/2024] [Accepted: 11/10/2024] [Indexed: 11/17/2024]
Abstract
Several mycobacterial species are known to cause human diseases, such as tuberculosis and leprosy. In addition to these pathogenic species, there are also saprophytic representatives, which occasionally cause opportunistic infections. It is well established that numerous mycobacteria produce biofilms containing cellulose, and their genomes frequently harbor genes involved in cellulose degradation, such as celA1. Notably, the BCG Moreau vaccine strain carries a specific deletion of two-base pairs, resulting in a predicted protein with fewer than 100 amino acids in the catalytic portion at the C-terminal end. We investigated the functional consequences of this polymorphism and observed that recombinant enzyme from the Moreau strain lack catalytic activity. Furthermore, compared to the Pasteur strain, Moreau is unable to utilize carboxymethylcellulose (CMC) as the sole carbon source. These findings suggest an absence of cellulolytic activity in this strain, which may influence the bacterium virulence.
Collapse
Affiliation(s)
| | - Paloma Rezende Corrêa
- Laboratório de Genômica Funcional e Bioinformática, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil.
| | | | - Leila Mendonça-Lima
- Laboratório de Genômica Funcional e Bioinformática, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil.
| |
Collapse
|
60
|
Miller E, Jamal H, Patel P. Recurrent Bacteremia in the Setting of Pseudomonas Endocarditis of the Tricuspid Valve and Indwelling Medical Devices. Cureus 2024; 16:e76368. [PMID: 39867067 PMCID: PMC11759660 DOI: 10.7759/cureus.76368] [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] [Accepted: 12/24/2024] [Indexed: 01/28/2025] Open
Abstract
This case report presents a complex and challenging scenario of recurrent Pseudomonas aeruginosa (P. aeruginosa) bacteremia and tricuspid valve endocarditis in a 77-year-old male patient with multiple comorbidities and indwelling medical devices. The patient's medical history was significant for T4 paraplegia, neurogenic bladder requiring a chronic indwelling suprapubic catheter, heart block status post-permanent pacemaker placement, type 2 diabetes mellitus, chronic kidney disease, and chronic sacral wounds. The case highlights the difficulties in managing antibiotic-resistant P. aeruginosa infections, particularly in patients with implantable devices and chronic wounds. The patient's clinical course was marked by multiple hospital admissions, each time presenting with fever, confusion, and positive blood cultures for P. aeruginosa. Despite aggressive antibiotic treatment and interventions such as pacemaker replacement and tricuspid valve debulking, the patient's condition continued to deteriorate. The recurring nature of the infection, despite therapeutic interventions, underscores the risk of bacterial seeding of indwelling medical devices and the challenges posed by antibiotic resistance. This case also draws attention to the significance of P. aeruginosa as a causative agent of severe nosocomial infections, particularly in immunocompromised individuals, and its growing resistance to antibiotics through mechanisms such as biofilm formation. Ultimately, the patient developed septic shock and transitioned to comfort care due to treatment failure, highlighting the difficult clinical decisions required in the face of chronic infections refractory to repeated interventions. This case serves as a reminder of the need for continued vigilance and innovative strategies, such as multifunctional antibacterial-coated devices, in preventing and managing device-associated infections. This is particularly important in the context of increasing antibiotic resistance and the complications associated with biofilm formation.
Collapse
Affiliation(s)
- Ethan Miller
- Internal Medicine, Cooper Medical School of Rowan University, Camden, USA
| | - Hera Jamal
- Internal Medicine, Cooper University Hospital, Camden, USA
| | - Parita Patel
- Internal Medicine, Cooper University Hospital, Camden, USA
| |
Collapse
|
61
|
Schlichter Kadosh Y, Muthuraman S, Nisaa K, Ben-Zvi A, Karsagi Byron DL, Shagan M, Brandis A, Mehlman T, Gopas J, Saravana Kumar R, Kushmaro A. Pseudomonas aeruginosa quorum sensing and biofilm attenuation by a di-hydroxy derivative of piperlongumine (PL-18). Biofilm 2024; 8:100215. [PMID: 39148892 PMCID: PMC11326495 DOI: 10.1016/j.bioflm.2024.100215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 07/01/2024] [Accepted: 07/13/2024] [Indexed: 08/17/2024] Open
Abstract
Bacterial communication, Quorum Sensing (QS), is a target against virulence and prevention of antibiotic-resistant infections. 16 derivatives of Piperlongumine (PL), an amide alkaloid from Piper longum L., were screened for QS inhibition. PL-18 had the best QSI activity. PL-18 inhibited the lasR-lasI, rhlR-rhlI, and pqs QS systems of Pseudomonas aeruginosa. PL-18 inhibited pyocyanin and rhamnolipids that are QS-controlled virulence elements. Iron is an essential element for pathogenicity, biofilm formation and resilience in harsh environments, its uptake was inhibited by PL-18. Pl-18 significantly reduced the biofilm biovolume including in established biofilms. PL-18-coated silicon tubes significantly inhibited biofilm formation. The transcriptome study of treated P. aeruginosa showed that PL-18 indeed reduced the expression of QS and iron homeostasis related genes, and up regulated sulfur metabolism related genes. Altogether, PL-18 inhibits QS, virulence, iron uptake, and biofilm formation. Thus, PL-18 should be further developed against bacterial infection, antibiotic resistance, and biofilm formation.
Collapse
Affiliation(s)
- Yael Schlichter Kadosh
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben Gurion University of the Negev, Beer Sheva, Israel
| | | | - Khairun Nisaa
- Department of Life Science, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Anat Ben-Zvi
- Department of Life Science, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Danit Lisa Karsagi Byron
- Department of Civil and Environmental Engineering, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Marilou Shagan
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Alexander Brandis
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Tevie Mehlman
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Jacob Gopas
- Department of Microbiology, Immunology and Genetics Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel
| | | | - Ariel Kushmaro
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben Gurion University of the Negev, Beer Sheva, Israel
- The Ilse Katz Center for Nanoscale Science and Technology, Ben Gurion University of the Negev, Beer Sheva, Israel
- School of Sustainability and Climate Change, Ben Gurion University of the Negev, Beer Sheva, Israel
| |
Collapse
|
62
|
Sathishkumar P, Khan F. Leveraging bacteria-inspired nanomaterials for targeted controlling biofilm and virulence properties of Pseudomonas aeruginosa. Microb Pathog 2024; 197:107103. [PMID: 39505089 DOI: 10.1016/j.micpath.2024.107103] [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: 09/12/2024] [Revised: 10/24/2024] [Accepted: 11/03/2024] [Indexed: 11/08/2024]
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen designated as a high-priority pathogen because of its role in major healthcare-associated and nosocomial infections. Biofilm production by these bacteria is one of the adaptive resistance mechanisms to traditional antibiotics, making treatment challenging, especially for immunocompromised patients. P. aeruginosa also produces a variety of virulence factors, which aid in invasion, adhesion, persistence, and immune system protection. Recent advances in nanotechnology-based therapy, notably the application of bioinspired metal and metal-oxide nanomaterials, have been seen as a viable way to control P. aeruginosa biofilm and virulence. Because of its ease of growth and culture, synthesizing metal and metal-oxide nanomaterials using bacterial species has become one of the most environmentally benign green synthesis options. The application of bacterial-inspired nanomaterials is particularly successful for targeted control of P. aeruginosa infection due to interactions with cell membrane components and transport systems. This paper delves into and provides a complete overview of the application of bacterial-inspired metal and metal-oxide nanomaterials to treat P. aeruginosa infection by targeting biofilm and virulence characteristics. The review focused on synthesizing and applying gold, silver, copper, iron, magnetite, and zinc oxide nanomaterials to mitigate P. aeruginosa biofilm and virulence. The underlying mechanism of these metal and metal-oxide nanoparticles in relation to biofilm and virulence features has also been thoroughly discussed. The current review introduces novel approaches to treating and controlling drug-resistant P. aeruginosa using bacterial-inspired nanomaterials as a targeted therapeutic strategy.
Collapse
Affiliation(s)
- Palanivel Sathishkumar
- Green Lab, Department of Prosthodontics, Saveetha Dental College and Hospitals, SIMATS, Saveetha University, Chennai, 600077, Tamil Nadu, India.
| | - Fazlurrahman Khan
- Ocean and Fisheries Development International Cooperation Institute, Pukyong National University. Busan 48513, Republic of Korea; International Graduate Program of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea; Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea.
| |
Collapse
|
63
|
Li R, Zhu X, Zhang P, Wu X, Jin Q, Pan J. Ser/Thr protein kinase Stk1 phosphorylates the key transcriptional regulator AlgR to modulate virulence and resistance in Pseudomonas aeruginosa. Virulence 2024; 15:2367649. [PMID: 38898809 PMCID: PMC11197903 DOI: 10.1080/21505594.2024.2367649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 06/09/2024] [Indexed: 06/21/2024] Open
Abstract
Pseudomonas aeruginosa is one of the leading causes of nosocomial infections worldwide and has emerged as a serious public health threat, due in large part to its multiple virulence factors and remarkable resistance capabilities. Stk1, a eukaryotic-type Ser/Thr protein kinase, has been shown in our previous work to be involved in the regulation of several signalling pathways and biological processes. Here, we demonstrate that deletion of stk1 leads to alterations in several virulence- and resistance-related physiological functions, including reduced pyocyanin and pyoverdine production, attenuated twitching motility, and enhanced biofilm production, extracellular polysaccharide secretion, and antibiotic resistance. Moreover, we identified AlgR, an important transcriptional regulator, as a substrate for Stk1, with its phosphorylation at the Ser143 site catalysed by Stk1. Intriguingly, both the deletion of stk1 and the mutation of Ser143 of AlgR to Ala result in similar changes in the above-mentioned physiological functions. Furthermore, assays of algR expression in these strains suggest that changes in the phosphorylation state of AlgR, rather than its expression level, underlie changes in these physiological functions. These findings uncover Stk1-mediated phosphorylation of AlgR as an important mechanism for regulating virulence and resistance in P. aeruginosa.
Collapse
Affiliation(s)
- Rui Li
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xuan Zhu
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Pengfei Zhang
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xuan Wu
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Qian Jin
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Jianyi Pan
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| |
Collapse
|
64
|
Gartika M, Tumilaar SG, Dharsono HDA, Nurdin D, Kurnia D. Exploring the Inhibitory Potential of M. pendans Compounds Against N-Acetylglucosamine (Mur) Receptor: In Silico Insights Into Antibacterial Activity and Drug-Likeness. ScientificWorldJournal 2024; 2024:3569811. [PMID: 39654692 PMCID: PMC11628175 DOI: 10.1155/tswj/3569811] [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: 03/23/2024] [Revised: 09/27/2024] [Accepted: 10/25/2024] [Indexed: 12/12/2024] Open
Abstract
Oral diseases are often caused by bacterial infections, making the inhibition of receptors like N-acetylglucosamine critical in preventing bacterial formation. The plant Myrmecodia pendans (M. pendans) is known for its diverse bioactivities and may serve as a promising source for developing new antibacterial agents. This study employs in silico methods to predict the inhibitory mechanisms, pharmacokinetics, and drug-likeness of compounds isolated from M. pendans. Three compounds were evaluated for their inhibitory effects on the MurA and MurB receptors using the AutoDock4 molecular docking software, with visualizations performed using the BIOVIA Discovery Studio Visualizer. The binding affinities obtained for compounds 1, 2, and 3 to the MurA receptor were -9.42, -9.57, and -6.84 kcal/mol, respectively, while their binding affinities to the MurB receptor were -11.25, -10.55, and -8.69 kcal/mol. These affinities were found to be stronger than those of fosfomycin (benchmark compound) but weaker than the native ligands of the respective receptors. Key amino acid residues involved in the binding to MurA were identified as Cys115 and Asp305, while Ser82 and Asn83 were noted for MurB. In the ADMET prediction and drug-likeness analysis, some compounds met the necessary criteria, whereas others did not. Although all the three compounds demonstrated strong predicted inhibitory activity against MurA and MurB receptors, the analysis suggests that Compound 2 may hold the most promise as a potential antibacterial agent, warranting further investigation.
Collapse
Affiliation(s)
- Meirina Gartika
- Department of Pediatric Dentistry, Faculty of Dentistry, Universitas Padjadjaran, Bandung, Indonesia
| | - Sefren Geiner Tumilaar
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Bandung, Indonesia
| | | | - Denny Nurdin
- Department of Conservative Dentistry, Faculty of Dentistry, Universitas Padjadjaran, Bandung, Indonesia
| | - Dikdik Kurnia
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Bandung, Indonesia
| |
Collapse
|
65
|
Parra B, Sandoval M, Arriagada V, Amsteins L, Aguayo C, Opazo-Capurro A, Dechesne A, González-Rocha G. Isolation and Characterization of Lytic Bacteriophages Capable of Infecting Diverse Multidrug-Resistant Strains of Pseudomonas aeruginosa: PaCCP1 and PaCCP2. Pharmaceuticals (Basel) 2024; 17:1616. [PMID: 39770458 PMCID: PMC11728774 DOI: 10.3390/ph17121616] [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: 10/20/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND/OBJECTIVES Antimicrobial resistance (AMR) is a major public health threat, which is exacerbated by the lack of new antibiotics and the emergence of multidrug-resistant (MDR) superbugs. Comprehensive efforts and alternative strategies to combat AMR are urgently needed to prevent social, medical, and economic consequences. Pseudomonas aeruginosa is a pathogen responsible for a wide range of infections, from soft tissue infections to life-threatening conditions such as bacteremia and pneumonia. Bacteriophages have been considered as a potential therapeutic option to treat bacterial infections. Our aim was to isolate phages able to infect MDR P. aeruginosa strains. METHODS We isolated two lytic phages, using the conventional double layer agar technique (DLA), from samples obtained from the influent of a wastewater treatment plant in Concepción, Chile. The phages, designated as PaCCP1 and PaCCP2, were observed by electron microscopy and their host range was determined against multiple P. aeruginosa strains using DLA. Moreover, their genomes were sequenced and analyzed. RESULTS Phage PaCCP1 is a member of the Septimatrevirus genus and phage PaCCP2 is a member of the Pbunavirus genus. Both phages are tailed and contain dsDNA. The genome of PaCCP1 is 43,176 bp in length with a GC content of 54.4%, encoding 59 ORFs, one of them being a tRNA gene. The genome of PaCCP2 is 66,333 bp in length with a GC content of 55.6%, encoding 102 non-tRNA ORFs. PaCCP1 is capable of infecting five strains of P. aeruginosa, whereas phage PaCCP2 is capable of infecting three strains of P. aeruginosa. Both phages do not contain bacterial virulence or AMR genes and contain three and six putative Anti-CRISPR proteins. CONCLUSIONS Phages PaCCP1 and PaCCP2 show promise as effective treatments for MDR P. aeruginosa strains, offering a potential strategy for controlling this clinically important pathogen through phage therapy.
Collapse
Affiliation(s)
- Boris Parra
- Laboratorio de Investigación en Agentes Antibacterianos (LIAA), Departamento de Microbiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción 4070409, Chile
- Grupo de Estudio en Resistencia Antimicrobiana (GRAM), Universidad de Concepción, Concepción 4070409, Chile
- Facultad de Medicina Veterinaria y Agronomía, Instituto de Ciencias Naturales, Universidad de las Américas, Av. Jorge Alessandri 1160, Campus El Boldal, Concepción 4070409, Chile
| | - Maximiliano Sandoval
- Laboratorio de Investigación en Agentes Antibacterianos (LIAA), Departamento de Microbiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción 4070409, Chile
- Grupo de Estudio en Resistencia Antimicrobiana (GRAM), Universidad de Concepción, Concepción 4070409, Chile
| | - Vicente Arriagada
- Laboratorio de Investigación en Agentes Antibacterianos (LIAA), Departamento de Microbiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción 4070409, Chile
- Grupo de Estudio en Resistencia Antimicrobiana (GRAM), Universidad de Concepción, Concepción 4070409, Chile
| | - Luis Amsteins
- Laboratorio de Investigación en Agentes Antibacterianos (LIAA), Departamento de Microbiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción 4070409, Chile
- Grupo de Estudio en Resistencia Antimicrobiana (GRAM), Universidad de Concepción, Concepción 4070409, Chile
| | - Cristobal Aguayo
- Laboratorio de Investigación en Agentes Antibacterianos (LIAA), Departamento de Microbiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción 4070409, Chile
- Grupo de Estudio en Resistencia Antimicrobiana (GRAM), Universidad de Concepción, Concepción 4070409, Chile
| | - Andrés Opazo-Capurro
- Laboratorio de Investigación en Agentes Antibacterianos (LIAA), Departamento de Microbiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción 4070409, Chile
- Grupo de Estudio en Resistencia Antimicrobiana (GRAM), Universidad de Concepción, Concepción 4070409, Chile
| | - Arnaud Dechesne
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofs Plads, Building 221, 2800 Kongens Lyngby, Denmark
| | - Gerardo González-Rocha
- Laboratorio de Investigación en Agentes Antibacterianos (LIAA), Departamento de Microbiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción 4070409, Chile
- Grupo de Estudio en Resistencia Antimicrobiana (GRAM), Universidad de Concepción, Concepción 4070409, Chile
| |
Collapse
|
66
|
Ciemniecki JA, Ho CL, Horak RD, Okamoto A, Newman DK. Mechanistic study of a low-power bacterial maintenance state using high-throughput electrochemistry. Cell 2024; 187:6882-6895.e8. [PMID: 39447571 PMCID: PMC11606744 DOI: 10.1016/j.cell.2024.09.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 07/03/2024] [Accepted: 09/26/2024] [Indexed: 10/26/2024]
Abstract
Mechanistic studies of life's lower metabolic limits have been limited due to a paucity of tractable experimental systems. Here, we show that redox-cycling of phenazine-1-carboxamide (PCN) by Pseudomonas aeruginosa supports cellular maintenance in the absence of growth with a low mass-specific metabolic rate of 8.7 × 10-4 W (g C)-1 at 25°C. Leveraging a high-throughput electrochemical culturing device, we find that non-growing cells cycling PCN tolerate conventional antibiotics but are susceptible to those that target membrane components. Under these conditions, cells conserve energy via a noncanonical, facilitated fermentation that is dependent on acetate kinase and NADH dehydrogenases. Across PCN concentrations that limit cell survival, the cell-specific metabolic rate is constant, indicating the cells are operating near their bioenergetic limit. This quantitative platform opens the door to further mechanistic investigations of maintenance, a physiological state that underpins microbial survival in nature and disease.
Collapse
Affiliation(s)
- John A Ciemniecki
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Chia-Lun Ho
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba 305-0044, Ibaraki, Japan; School of Chemical Sciences and Engineering, Hokkaido University, 13 Kita, 8 Nishi, Kita-ku, Sapporo 060-8628, Hokkaido, Japan
| | - Richard D Horak
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Akihiro Okamoto
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba 305-0044, Ibaraki, Japan; Faculty of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8577, Ibaraki, Japan; School of Chemical Sciences and Engineering, Hokkaido University, 13 Kita, 8 Nishi, Kita-ku, Sapporo 060-8628, Hokkaido, Japan; Living Systems Materialogy (LiSM) Research Group, International Research Frontiers Initiative (IRFI), Tokyo Institute of Technology, Yokohama 226-8501, Kanagawa, Japan.
| | - Dianne K Newman
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA; Division of Geological & Planetary Sciences, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
67
|
Al-Momani H, Albalawi H, Al Balawi D, Khleifat KM, Aolymat I, Hamed S, Albiss BA, Khasawneh AI, Ebbeni O, Alsheikh A, Zueter AM, Pearson JP, Ward C. Enhanced Efficacy of Some Antibiotics in the Presence of Silver Nanoparticles Against Clinical Isolate of Pseudomonas aeruginosa Recovered from Cystic Fibrosis Patients. Int J Nanomedicine 2024; 19:12461-12481. [PMID: 39611007 PMCID: PMC11602434 DOI: 10.2147/ijn.s479937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 11/01/2024] [Indexed: 11/30/2024] Open
Abstract
Introduction Given the increasing frequency of drug-resistant bacteria and the limited progress in developing new antibiotics, it is necessary to explore new methods of combating microbial infections. Nanoparticles, particularly silver nanoparticles (Ag-NPs), have shown exceptional antibacterial characteristics; however, elevated concentrations of Ag-NPs can produce noticeable levels of toxicity in mammalian cells. Aim This study examined the potential synergistic effect of combining a low dosage of Ag-NPs and anti-pseudomonas drugs against Pseudomonas aeruginosa (ATCC strain) and eleven clinical isolates from cystic fibrosis patients. Methods The Ag-NPs were chemically produced by utilizing a seed extract from Peganum Harmala and characterized via ultraviolet-visible spectroscopy and scanning electron microscopy. The broth microdilution technique was utilized to investigate the minimum inhibitory concentration (MIC) of Ag-NPs and eight antibiotics (Piperacillin, Ciprofloxacin, Levofloxacin, Meropenem, Amikacin, Ceftazidime, Gentamicin, Aztreonam). The fractional inhibitory concentration index (FICI) was determined via the checkerboard method to evaluate the synergistic effects of Ag-NPs and various antibiotics. Results The biosynthesized Ag-NPs were uniformly spherical and measured around 15 nm in size. When combined with antibiotics, Ag-NP produced statistically significant reductions in the amount of antibiotics required to completely prevent P. aeruginosa growth for all strains. The findings revealed that the MIC of Ag-NPs was 15 ug/mL for all strains which decreased substantially when administered with antibiotics at a dose of 1.875-7.5 ug/mL. The majority of Ag-NP and antibiotic combinations exhibited a synergistic or partially synergistic impact. This was particularly noticeable in combinations containing Meropenem, Ciprofloxacin, and Aztreonam (in which the FIC index was less than or equal to 0.5). Conclusion The findings revealed that combining Ag-NPs with antibiotics was more effective than using Ag-NPs or antibiotics in isolation and that combinations of Ag-NPs and antimicrobial agents displayed synergistic activity against the majority of strains assessed.
Collapse
Affiliation(s)
- Hafez Al-Momani
- Department of Microbiology, Pathology and Forensic Medicine, Faculty of Medicine, The Hashemite University, Zarqa, 13133, Jordan
| | - Hadeel Albalawi
- Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, 13133, Jordan
| | - Dua’a Al Balawi
- Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, 13133, Jordan
| | - Khaled M Khleifat
- Biology Department, College of Science, Mutah University, Mutah, Karak, 61710, Jordan
| | - Iman Aolymat
- Department of Anatomy, Physiology and Biochemistry, Faculty of Medicine, The Hashemite University, Zarqa, 13133, Jordan
| | - Saja Hamed
- Department of Pharmaceutics & Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa, 13133, Jordan
| | - Borhan Aldeen Albiss
- Nanotechnology Institute, Jordan University of Science & Technology, Irbid, 22110, Jordan
| | - Ashraf I Khasawneh
- Department of Microbiology, Pathology and Forensic Medicine, Faculty of Medicine, The Hashemite University, Zarqa, 13133, Jordan
| | - Ola Ebbeni
- Department of Pharmacology and Public Health, Faculty of Medicine, The Hashemite University, Zarqa, 13133, Jordan
| | - Ayman Alsheikh
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Zarqa University, Zarqa, 13110, Jordan
| | - AbdelRahman M Zueter
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, 13133, Jordan
| | | | - Christopher Ward
- Translational and Clinical Research Institute, Newcastle University Medical School, Newcastle Upon Tyne, NE2 4HHUK
| |
Collapse
|
68
|
Oh JW, Shin MK, Park HR, Kim S, Lee B, Yoo JS, Chi WJ, Sung JS. PA-Win2: In Silico-Based Discovery of a Novel Peptide with Dual Antibacterial and Anti-Biofilm Activity. Antibiotics (Basel) 2024; 13:1113. [PMID: 39766503 PMCID: PMC11672609 DOI: 10.3390/antibiotics13121113] [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: 10/15/2024] [Revised: 11/15/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025] Open
Abstract
Background: The emergence and prevalence of antibiotic-resistant bacteria (ARBs) have become a serious global threat, as the morbidity and mortality associated with ARB infections are continuously rising. The activation of quorum sensing (QS) genes can promote biofilm formation, which contributes to the acquisition of drug resistance and increases virulence. Therefore, there is an urgent need to develop new antimicrobial agents to control ARB and prevent further development. Antimicrobial peptides (AMPs) are naturally occurring defense molecules in organisms known to suppress pathogens through a broad range of antimicrobial mechanisms. Methods: In this study, we utilized a previously developed deep-learning model to identify AMP candidates from the venom gland transcriptome of the spider Pardosa astrigera, followed by experimental validation. Results: PA-Win2 was among the top-scoring predicted peptides and was selected based on physiochemical features. Subsequent experimental validation demonstrated that PA-Win2 inhibits the growth of Bacillus subtilis, Escherichia coli, Staphylococcus aureus, Staphylococcus epidermidis, Pseudomonas aeruginosa, and multidrug-resistant P. aeruginosa (MRPA) strain CCARM 2095. The peptide exhibited strong bactericidal activity against P. aeruginosa, and MRPA CCARM 2095 through the depolarization of bacterial cytoplasmic membranes and alteration of gene expression associated with bacterial survival. In addition, PA-Win2 effectively inhibited biofilm formation and degraded pre-formed biofilms of P. aeruginosa. The gene expression study showed that the peptide treatment led to the downregulation of QS genes in the Las, Pqs, and Rhl systems. Conclusions: These findings suggest PA-Win2 as a promising drug candidate against ARB and demonstrate the potential of in silico methods in discovering functional peptides from biological data.
Collapse
Affiliation(s)
- Jin Wook Oh
- Department of Life Science, Dongguk University-Seoul, Goyang 10326, Republic of Korea; (J.W.O.); (M.K.S.); (H.-R.P.); (S.K.)
| | - Min Kyoung Shin
- Department of Life Science, Dongguk University-Seoul, Goyang 10326, Republic of Korea; (J.W.O.); (M.K.S.); (H.-R.P.); (S.K.)
| | - Hye-Ran Park
- Department of Life Science, Dongguk University-Seoul, Goyang 10326, Republic of Korea; (J.W.O.); (M.K.S.); (H.-R.P.); (S.K.)
| | - Sejun Kim
- Department of Life Science, Dongguk University-Seoul, Goyang 10326, Republic of Korea; (J.W.O.); (M.K.S.); (H.-R.P.); (S.K.)
| | - Byungjo Lee
- Research Institute, National Cancer Center, Goyang 10408, Republic of Korea;
| | - Jung Sun Yoo
- Wildlife Quarantine Center, National Institute of Wildlife Disease Control and Prevention, Incheon 22382, Republic of Korea;
| | - Won-Jae Chi
- Species Diversity Research Division, National Institute of Biological Resources, Incheon 22689, Republic of Korea;
| | - Jung-Suk Sung
- Department of Life Science, Dongguk University-Seoul, Goyang 10326, Republic of Korea; (J.W.O.); (M.K.S.); (H.-R.P.); (S.K.)
| |
Collapse
|
69
|
Kuhn EMA, Sominsky LA, Chittò M, Schwarz EM, Moriarty TF. Antibacterial Mechanisms and Clinical Impact of Sitafloxacin. Pharmaceuticals (Basel) 2024; 17:1537. [PMID: 39598446 PMCID: PMC11597390 DOI: 10.3390/ph17111537] [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: 10/14/2024] [Revised: 11/09/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
Sitafloxacin is a 4th generation fluoroquinolone antibiotic with broad activity against a wide range of Gram-negative and Gram-positive bacteria. It is approved in Japan and used to treat pneumonia and urinary tract infections (UTIs) as well as other upper and lower respiratory infections, genitourinary infections, oral infections and otitis media. Compared to other fluoroquinolones, sitafloxacin displays a low minimal inhibitory concentration (MIC) for many bacterial species but also activity against anaerobes, intracellular bacteria, and persisters. Furthermore, it has also shown strong activity against biofilms of P. aeruginosa and S. aureus in vitro, which was recently validated in vivo with murine models of S. aureus implant-associated bone infection. Although limited in scale at present, the published literature supports the further evaluation of sitafloxacin in implant-related infections and other biofilm-related infections. The aim of this review is to summarize the chemical-positioning-based mechanisms, activity, resistance profile, and future clinical potential of sitafloxacin.
Collapse
Affiliation(s)
- Elian M. A. Kuhn
- AO Research Institute Davos, 7270 Davos, Switzerland; (E.M.A.K.); (M.C.)
- Infection Biology, Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Levy A. Sominsky
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA (E.M.S.)
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
- Medical Scientist Training Program, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Marco Chittò
- AO Research Institute Davos, 7270 Davos, Switzerland; (E.M.A.K.); (M.C.)
| | - Edward M. Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA (E.M.S.)
| | - T. Fintan Moriarty
- AO Research Institute Davos, 7270 Davos, Switzerland; (E.M.A.K.); (M.C.)
| |
Collapse
|
70
|
Huang Y, Chen P, Cao H, Zhou Z, Xu T. Characterization of Pseudomonas aeruginosa Isolated from Bovine Mastitis in Northern Jiangsu Province and Correlation to Drug Resistance and Biofilm Formability. Animals (Basel) 2024; 14:3290. [PMID: 39595342 PMCID: PMC11590879 DOI: 10.3390/ani14223290] [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: 08/31/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
This study aimed to provide experimental support for the prevention and treatment of Pseudomonas aeruginosa infections and to elucidate the epidemiological distribution of resistance and virulence genes of Pseudomonas aeruginosa from mastitis in dairy cows in the northern part of Jiangsu Province and their relationship with the biofilm-forming ability of the strains. Mastitis presents a significant challenge within dairy farming, adversely impacting the health of dairy cows and precipitating substantial economic losses in milk production. In this study, Pseudomonas aeruginosa (PA) was isolated and identified from mastitis milk samples in Jiangsu Province, China. In order to characterize the isolates, multilocus sequence typing (MLST), drug resistance phenotypes, virulence genes, and biofilm formations were detected. The isolation and identification of pathogenic bacteria from 168 clinical mastitis milk samples using 16S rRNA and PCR revealed 63 strains of Pseudomonas aeruginosa, which were determined to be highly homologous according to phylogenetic tree analysis. In addition, the MLST indicated five major ST types, namely ST277, ST450, ST571, ST641, and ST463. The susceptibility to 10 antimicrobials was determined, and it was found that 63 strains of Pseudomonas aeruginosa did not have a strong resistance to the antimicrobials in general. However, there were differences in the phenotypes' resistance to antimicrobials among the different ST types. It was also found that the more resistant the strains were to antimicrobials, the lower the carriage of virulence genes detected. The biofilm content was measured using the semi-quantitative crystal violet method. It was found that there were a few strains with medium or strong biofilm-forming abilities. However, the number of virulence genes carried by the 63 strains of Pseudomonas aeruginosa was inversely proportional to the biofilm-forming ability. It was also found that there were significantly more Pseudomonas aeruginosa in the biofilm state than in the planktonic state and that strains with strong biofilm-forming abilities were more resistant to antimicrobials.
Collapse
Affiliation(s)
- Yicai Huang
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Pengqiang Chen
- Fujian Nanxing Animal Health Products Co., Ltd., Nanping 353000, China
| | - Hainan Cao
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Zheng Zhou
- Department of Animal Science, Michigan State University, East Lansing, MI 48824, USA
| | - Tianle Xu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- International Joint Research Laboratory in Universities of Jiangsu Province of China for Domestic Animal Germplasm Resources and Genetic Improvement, Yangzhou 225009, China
| |
Collapse
|
71
|
Rodriguez Gonzalez C, Basílio-Queirós D, Neehus AL, Merkert S, Tschritter D, Ünal S, Hegermann J, Mörgelin M, Bustamante J, Nietert MM, Martin U, Tümmler B, Munder A, Lachmann N. Human CFTR deficient iPSC-macrophages reveal impaired functional and transcriptomic response upon Pseudomonas aeruginosa infection. Front Immunol 2024; 15:1397886. [PMID: 39606224 PMCID: PMC11601075 DOI: 10.3389/fimmu.2024.1397886] [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: 03/08/2024] [Accepted: 08/27/2024] [Indexed: 11/29/2024] Open
Abstract
Introduction Cystic fibrosis (CF) is a hereditary autosomal recessive disease driven by deleterious variants of the CFTR gene, leading, among other symptoms, to increased lung infection susceptibility. Mucus accumulation in the CF lung is, as of yet, considered as one important factor contributing to its colonization by opportunistic pathogens such as Pseudomonas aeruginosa. However, in recent years evidence was provided that alveolar macrophages, which form the first line of defense against airborne pathogens, seem to be intrinsically defective with regard to bactericidal functionality in the CF lung. To assess the impact of CFTR deficiency in human macrophages only insufficient systems are available. Methods To address this problem and to evaluate the role of CFTR in human macrophages, we successfully differentiated human induced pluripotent stem cells (iPSC) from a CF p.Phe508del homozygous individual and a healthy donor into primitive macrophages (iMacΔF508 and iMacWT), respectively, and compared the bactericidal functionality in the relevant cell type. Results iMacΔF508 showed impaired P. aeruginosa clearance and intracellular killing capacity in comparison to iMacWT. Furthermore, iMacΔF508 exhibited a less acidic lysosomal pH, and upon P. aeruginosa infection, there were signs of mitochondrial fragmentation and autophagosome formation together with a hyperinflammatory phenotype and deficient type I interferon response. Conclusion In summary, we present a defective phenotype in iMacΔF508 upon P. aeruginosa infection, which will constitute an ideal platform to further study the role of macrophages in the context of CF.
Collapse
Affiliation(s)
- Claudio Rodriguez Gonzalez
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Débora Basílio-Queirós
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Anna-Lena Neehus
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
- Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Sylvia Merkert
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
- REBIRTH, Research Center for Translational and Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - David Tschritter
- Department of Medical Bioinformatics, University Medical Center Göttingen, Göttingen, Germany
| | - Sinem Ünal
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Jan Hegermann
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
- Research Core Unit Electron Microscopy, Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
| | | | - Jacinta Bustamante
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, Assistance Publique-Hôpitaux de Paris AP-HP, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, United States
| | - Manuel Manfred Nietert
- Department of Medical Bioinformatics, University Medical Center Göttingen, Göttingen, Germany
| | - Ulrich Martin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
- REBIRTH, Research Center for Translational and Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Burkhard Tümmler
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Antje Munder
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Nico Lachmann
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
- REBIRTH, Research Center for Translational and Regenerative Medicine, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| |
Collapse
|
72
|
Foksiński P, Blank A, Kaczorek-Łukowska E, Małaczewska J, Wróbel M, Wójcik EA, Sowińska P, Pietrzyk N, Matusiak R, Wójcik R. Does Every Strain of Pseudomonas aeruginosa Attack the Same? Results of a Study of the Prevalence of Virulence Factors of Strains Obtained from Different Animal Species in Northeastern Poland. Pathogens 2024; 13:979. [PMID: 39599532 PMCID: PMC11597259 DOI: 10.3390/pathogens13110979] [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: 09/17/2024] [Revised: 10/29/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Pseudomonas aeruginosa is a pathogen that causes infections in animals and humans, with veterinary implications including ear infections in dogs, respiratory diseases in cats, and mastitis in ruminants. In humans, it causes severe hospital-acquired infections, particularly in immunosuppressed patients. This study aimed to identify and assess the prevalence of specific virulence factors in Pseudomonas aeruginosa isolates. METHODS We analyzed 98 Pseudomonas aeruginosa isolates from various animal samples (dogs, cats, ruminants, fowl) from northeastern Poland in 2019-2022 for virulence-related genes (toxA, exoU, exoT, exoS, lasB, plcN, plcH, pldA, aprA, gacA, algD, pelA, endA, and oprF) by PCR and assessed biofilm formation at 48 and 72 h. Genomic diversity was assessed by ERIC-PCR. RESULTS The obtained results showed that all strains harbored the pelA gene (100%), while the lowest prevalence was found for pldA (24%) and exoU (36%). Regardless of the animal species, strong biofilm forming ability was prevalent among the strains after both 48 h (75%) and 72 h (74%). We obtained as many as 87 different genotyping profiles, where the dominant one was profile ERIC-48, observed in four strains. CONCLUSIONS No correlation was found between presence or absence of determined genes and the nature of infection. Similarly, no correlation was found between biofilm-forming genes and biofilm strength. The high genetic diversity indicates challenges for effective prevention, emphasizing the need for ongoing monitoring and research.
Collapse
Affiliation(s)
- Paweł Foksiński
- Department of Microbiology and Clinical Immunology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego 13, 10-719 Olsztyn, Poland; (A.B.); (E.K.-Ł.); (J.M.); (M.W.); (R.W.)
| | - Alicja Blank
- Department of Microbiology and Clinical Immunology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego 13, 10-719 Olsztyn, Poland; (A.B.); (E.K.-Ł.); (J.M.); (M.W.); (R.W.)
| | - Edyta Kaczorek-Łukowska
- Department of Microbiology and Clinical Immunology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego 13, 10-719 Olsztyn, Poland; (A.B.); (E.K.-Ł.); (J.M.); (M.W.); (R.W.)
| | - Joanna Małaczewska
- Department of Microbiology and Clinical Immunology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego 13, 10-719 Olsztyn, Poland; (A.B.); (E.K.-Ł.); (J.M.); (M.W.); (R.W.)
| | - Małgorzata Wróbel
- Department of Microbiology and Clinical Immunology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego 13, 10-719 Olsztyn, Poland; (A.B.); (E.K.-Ł.); (J.M.); (M.W.); (R.W.)
| | - Ewelina A. Wójcik
- Proteon Pharmaceuticals, Tylna 3a, 90-364 Łódź, Poland; (E.A.W.); (P.S.); (N.P.); (R.M.)
| | - Patrycja Sowińska
- Proteon Pharmaceuticals, Tylna 3a, 90-364 Łódź, Poland; (E.A.W.); (P.S.); (N.P.); (R.M.)
| | - Nina Pietrzyk
- Proteon Pharmaceuticals, Tylna 3a, 90-364 Łódź, Poland; (E.A.W.); (P.S.); (N.P.); (R.M.)
| | - Rafał Matusiak
- Proteon Pharmaceuticals, Tylna 3a, 90-364 Łódź, Poland; (E.A.W.); (P.S.); (N.P.); (R.M.)
| | - Roman Wójcik
- Department of Microbiology and Clinical Immunology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego 13, 10-719 Olsztyn, Poland; (A.B.); (E.K.-Ł.); (J.M.); (M.W.); (R.W.)
| |
Collapse
|
73
|
Chen J, Su Z, Li F, Cao F, Xiong F, Jiang B, Xing Y, Wen D. The variation of resistome, mobilome and pathogen in domestic and industrial wastewater treatment systems. ENVIRONMENT INTERNATIONAL 2024; 193:109051. [PMID: 39418785 DOI: 10.1016/j.envint.2024.109051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/18/2024] [Accepted: 10/03/2024] [Indexed: 10/19/2024]
Abstract
Wastewater treatment plants (WWTPs), including both domestic and industrial facilities, are key contributors to antibiotic resistance genes (ARGs) and human pathogens in the environment. However, the characteristics and dissemination mechanisms of ARGs in domestic (SD) and industrial (SI) wastewater treatment systems remain unclear, leading to uncertainties in risk assessment. Based on metagenomic analysis, we observed significant differences in the compositions of resistome (ARGs and metal resistance genes, MRGs), mobilome (mobile genetic elements, MGEs), and bacterial community between SD and SI. SI exhibited lower diversity of ARGs but higher abundance of MRGs compared to SD. The removal efficiency of resistome was lower in the SI than that in the SD. MGEs emerged as the primary driver of ARG dissemination in the WWTPs, followed by the bacterial community. Environmental conditions (physicochemical parameters, heavy metals, and antibiotics) indirectly influenced the variation of resistome. Significantly, environmental conditions and MGEs highly influenced the composition of resistome in the SI, while bacterial community more associated with resistome in the SD. Additionally, we identified 36 human bacterial pathogens as potential hosts of ARGs, MRGs, and MGEs in wastewater samples. This study provides new insights on the dissemination mechanisms and risk assessment of antimicrobial resistance in the different types of WWTPs.
Collapse
Affiliation(s)
- Jiayu Chen
- School of Energy and Environmental Engineering, University of Science and Technology, Beijing 100083, China
| | - Zhiguo Su
- School of Environment, Tsinghua University, Beijing 100084, China.
| | - Feifei Li
- School of Environment, Tsinghua University, Beijing 100084, China
| | - Feng Cao
- College of Environmental Sciences and Engineering, Peking University, Beijing 100871, China
| | - Fuzhong Xiong
- College of Environmental Sciences and Engineering, Peking University, Beijing 100871, China
| | - Bo Jiang
- School of Energy and Environmental Engineering, University of Science and Technology, Beijing 100083, China.
| | - Yi Xing
- School of Energy and Environmental Engineering, University of Science and Technology, Beijing 100083, China
| | - Donghui Wen
- College of Environmental Sciences and Engineering, Peking University, Beijing 100871, China
| |
Collapse
|
74
|
Li Y, Farhan MHR, Yang X, Guo Y, Sui Y, Chu J, Huang L, Cheng G. A review on the development of bacterial multi-epitope recombinant protein vaccines via reverse vaccinology. Int J Biol Macromol 2024; 282:136827. [PMID: 39476887 DOI: 10.1016/j.ijbiomac.2024.136827] [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: 07/05/2024] [Revised: 10/04/2024] [Accepted: 10/21/2024] [Indexed: 11/10/2024]
Abstract
Bacterial vaccines play a crucial role in combating bacterial infectious diseases. Apart from the prevention of disease, bacterial vaccines also help to reduce the mortality rates in infected populations. Advancements in vaccine development technologies have addressed the constraints of traditional vaccine design, providing novel approaches for the development of next-generation vaccines. Advancements in reverse vaccinology, bioinformatics, and comparative proteomics have opened horizons in vaccine development. Specifically, the use of protein structural data in crafting multi-epitope vaccines (MEVs) to target pathogens has become an important research focus in vaccinology. In this review, we focused on describing the methodologies and tools for epitope vaccine development, along with recent progress in this field. Moreover, this article also discusses the challenges in epitope vaccine development, providing insights for the future development of bacterial multi-epitope genetically engineered vaccines.
Collapse
Affiliation(s)
- Yuxin Li
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Muhammad Haris Raza Farhan
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Xiaohan Yang
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Ying Guo
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Yuxin Sui
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Jinhua Chu
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Lingli Huang
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Huazhong Agricultural University, Wuhan, Hubei 430070, PR China; MOA Laboratory of Risk Assessment for Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Guyue Cheng
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Huazhong Agricultural University, Wuhan, Hubei 430070, PR China; MOA Laboratory of Risk Assessment for Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, PR China.
| |
Collapse
|
75
|
Tian M, Yan B, Jiang R, Liu C, Li Y, Xu B, Guo S, Li X. Activity of polymyxin B combined with cefepime-avibactam against the biofilms of polymyxin B-resistant Pseudomonas aeruginosa and Klebsiella pneumoniae in in vitro and in vivo models. BMC Microbiol 2024; 24:409. [PMID: 39407114 PMCID: PMC11481319 DOI: 10.1186/s12866-024-03571-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
Bacterial biofilms, often forming on medical devices, can lead to treatment failure due to their increased antimicrobial resistance. Cefepime-avibactam (CFP-AVI) exhibits potent activities against Pseudomonas aeruginosa (P. aeruginosa) and Klebsiella pneumoniae (K. pneumoniae) when used with polymyxin B (PMB). However, its efficacy in biofilm-related infections is unknown. The present study aimed to evaluate the activity of PMB combined with CFP-AVI against the biofilms of PMB-resistant Gram-negative bacteria. Five K. pneumoniae strains and three P. aeruginosa strains known to be PMB-resistant and prone to biofilm formation were selected and evaluated. Antimicrobial susceptibility assays demonstrated that the minimal biofilm inhibitory and eradication concentrations of PMB and CFP-AVI for biofilms formed by the eight strains were significantly higher than the minimal inhibitory concentrations of the antibiotics for planktonic cells. The biofilm formation inhibition and eradication assays showed that PMB combined with CFP-AVI cannot only suppress the formation of biofilm but also effectively eradicate the preformed mature biofilms. In a modified in vitro pharmacokinetic/pharmacodynamic biofilm model, CFP-AVI monotherapy exhibited a bacteriostatic or effective activity against the biofilms of seven strains, whereas PMB monotherapy did not have any activity at 72 h. However, PMB combined with CFP-AVI demonstrated bactericidal activity against the biofilms of all strains at 72 h. In an in vivo Galleria mellonella infection model, the 7-day survival rates of larvae infected with biofilm implants of K. pneumoniae or P. aeruginosa were 0-6.7%, 40.0-63.3%, and 46.7-90.0%, respectively, for PMB alone, CFP-AVI alone, and PMB combined with CFP-AVI; the combination therapy increased the rate by 6.7-33.3% (P < 0.05, n = 6), compared to CFP-AVI monotherapy. It is concluded that PMB combined with CFP-AVI exhibits effective anti-biofilm activities against PMB-resistant K. pneumoniae and P. aeruginosa both in vitro and in vivo, and thus may be a promising therapeutic strategy to treat biofilm-related infections.
Collapse
Affiliation(s)
- Miaomei Tian
- Department of Pharmacy, The Third Hospital of Changsha, 176 Western Laodong Road, Tianxin District, Changsha, Hunan Province, 410015, People's Republic of China
- Hunan Provincial Key Laboratory of Anti-Resistance Microbial Drugs, Changsha, Hunan Province, People's Republic of China
| | - Bingqian Yan
- Department of Pharmacy, The Third Hospital of Changsha, 176 Western Laodong Road, Tianxin District, Changsha, Hunan Province, 410015, People's Republic of China
- Hunan Provincial Key Laboratory of Anti-Resistance Microbial Drugs, Changsha, Hunan Province, People's Republic of China
| | - Rong Jiang
- Department of Pharmacy, The Third Hospital of Changsha, 176 Western Laodong Road, Tianxin District, Changsha, Hunan Province, 410015, People's Republic of China
- Institute of Clinical Application of Antibiotics, Changsha, Hunan Province, People's Republic of China
- Hunan Provincial Key Laboratory of Anti-Resistance Microbial Drugs, Changsha, Hunan Province, People's Republic of China
| | - Candi Liu
- Hunan Drug Inspection Center, Changsha, Hunan Province, People's Republic of China
| | - You Li
- Department of Pharmacy, The Third Hospital of Changsha, 176 Western Laodong Road, Tianxin District, Changsha, Hunan Province, 410015, People's Republic of China
| | - Bing Xu
- Department of Pharmacy, The Third Hospital of Changsha, 176 Western Laodong Road, Tianxin District, Changsha, Hunan Province, 410015, People's Republic of China
- Institute of Clinical Application of Antibiotics, Changsha, Hunan Province, People's Republic of China
- Hunan Provincial Key Laboratory of Anti-Resistance Microbial Drugs, Changsha, Hunan Province, People's Republic of China
| | - Siwei Guo
- Department of Pharmacy, The Third Hospital of Changsha, 176 Western Laodong Road, Tianxin District, Changsha, Hunan Province, 410015, People's Republic of China.
- Institute of Clinical Application of Antibiotics, Changsha, Hunan Province, People's Republic of China.
- Hunan Provincial Key Laboratory of Anti-Resistance Microbial Drugs, Changsha, Hunan Province, People's Republic of China.
| | - Xin Li
- Department of Pharmacy, The Third Hospital of Changsha, 176 Western Laodong Road, Tianxin District, Changsha, Hunan Province, 410015, People's Republic of China.
- Institute of Clinical Application of Antibiotics, Changsha, Hunan Province, People's Republic of China.
- Hunan Provincial Key Laboratory of Anti-Resistance Microbial Drugs, Changsha, Hunan Province, People's Republic of China.
| |
Collapse
|
76
|
Theodorakis N, Feretzakis G, Hitas C, Kreouzi M, Kalantzi S, Spyridaki A, Kollia Z, Verykios VS, Nikolaou M. Immunosenescence: How Aging Increases Susceptibility to Bacterial Infections and Virulence Factors. Microorganisms 2024; 12:2052. [PMID: 39458361 PMCID: PMC11510421 DOI: 10.3390/microorganisms12102052] [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: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
The process of aging leads to a progressive decline in the immune system function, known as immunosenescence, which compromises both innate and adaptive responses. This includes impairments in phagocytosis and decreased production, activation, and function of T- and B-lymphocytes, among other effects. Bacteria exploit immunosenescence by using various virulence factors to evade the host's defenses, leading to severe and often life-threatening infections. This manuscript explores the complex relationship between immunosenescence and bacterial virulence, focusing on the underlying mechanisms that increase vulnerability to bacterial infections in the elderly. Additionally, it discusses how machine learning methods can provide accurate modeling of interactions between the weakened immune system and bacterial virulence mechanisms, guiding the development of personalized interventions. The development of vaccines, novel antibiotics, and antivirulence therapies for multidrug-resistant bacteria, as well as the investigation of potential immune-boosting therapies, are promising strategies in this field. Future research should focus on how machine learning approaches can be integrated with immunological, microbiological, and clinical data to develop personalized interventions that improve outcomes for bacterial infections in the growing elderly population.
Collapse
Affiliation(s)
- Nikolaos Theodorakis
- Department of Cardiology, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (N.T.); (C.H.); (M.N.)
- 65+ Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (M.K.); (S.K.); (A.S.); (Z.K.)
- School of Medicine, National and Kapodistrian University of Athens, 75 Mikras Asias, 11527 Athens, Greece
| | - Georgios Feretzakis
- School of Science and Technology, Hellenic Open University, 18 Aristotelous Str., 26335 Patras, Greece;
| | - Christos Hitas
- Department of Cardiology, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (N.T.); (C.H.); (M.N.)
- 65+ Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (M.K.); (S.K.); (A.S.); (Z.K.)
| | - Magdalini Kreouzi
- 65+ Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (M.K.); (S.K.); (A.S.); (Z.K.)
- Department of Internal Medicine, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece
| | - Sofia Kalantzi
- 65+ Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (M.K.); (S.K.); (A.S.); (Z.K.)
- Department of Internal Medicine, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece
| | - Aikaterini Spyridaki
- 65+ Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (M.K.); (S.K.); (A.S.); (Z.K.)
- Department of Internal Medicine, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece
| | - Zoi Kollia
- 65+ Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (M.K.); (S.K.); (A.S.); (Z.K.)
| | - Vassilios S. Verykios
- School of Science and Technology, Hellenic Open University, 18 Aristotelous Str., 26335 Patras, Greece;
| | - Maria Nikolaou
- Department of Cardiology, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (N.T.); (C.H.); (M.N.)
- 65+ Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (M.K.); (S.K.); (A.S.); (Z.K.)
| |
Collapse
|
77
|
Pérez LM, Havryliuk O, Infante N, Muniesa M, Morató J, Mariychuk R, Tzanov T. Biofilm Prevention and Removal in Non-Target Pseudomonas Strain by Siphovirus-like Coliphage. Biomedicines 2024; 12:2291. [PMID: 39457603 PMCID: PMC11504082 DOI: 10.3390/biomedicines12102291] [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: 09/24/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives. Bacteriophages have gained significant interest as a potential solution to combat harmful bacteria, especially in the fight against antimicrobial resistance. With the rise in drug-resistant microorganisms, the medical community is increasingly exploring new alternatives to traditional antibiotics, and bacteriophages offer several advantages in this regard. However, phage applications still face some challenges, such as host specificity. Methods. In this study, a somatic Siphovirus-like coliphage (SOM7) was tested for inhibiting the biofilm-forming capacity of the non-target strain Pseudomonas aeruginosa (ATTC 10145). The phage-sensitive strain E. coli WG5 was used as a control. The selected microorganisms were first tested for growth in the presence of SOM7 at three different concentrations (105, 107, and 109 PFU/mL). Results. As expected, the phage-sensitive E. coli WG5 was fully inhibited by the coliphage, and no phage-related affection on the growth rate was observed for the SOM7-resistant P. aeruginosa. More notably, increasing concentrations of SOM7 significantly reduced both the biofilm-forming capacity and the amount of pre-established bacterial biofilm of the phage-insensitive P. aeruginosa (24.9% and 38.8% reduction in the biofilm-forming ability, and 18.8% and 28.0% biofilm degradation for 107 PFU/mL and 109 PFU/mL SOM7, respectively; p < 0.05). These results were supported by transmission electron microscopy (TEM) imaging, providing unprecedent evidence for the interaction of the somatic coliphage with the non-host strain. Conclusions. Although more studies in other biofilm models are necessary, our results show for the very first time that bacteriophages could potentially be used as an alternative to achieve desired anti-biofilm and biofilm-degrading activity in non-host bacterial strains.
Collapse
Affiliation(s)
- Leonardo Martín Pérez
- Laboratory of Sanitary and Environmental Microbiology (MSMLab)-UNESCO Chair on Sustainability, Universitat Politècnica de Catalunya-BarcelonaTech, R/Sant Nebridi, 22, GAIA Building (TR14), 08222 Terrassa, Spain; (L.M.P.); (O.H.); (N.I.); (J.M.)
- Grup de Biotecnologia Molecular i Industrial, Departament d’Enginyeria Química, Universitat Politècnica de Catalunya (UPC-BarcelonaTech), Rambla de Sant Nebridi 22, 08222 Terrassa, Spain;
| | - Olesia Havryliuk
- Laboratory of Sanitary and Environmental Microbiology (MSMLab)-UNESCO Chair on Sustainability, Universitat Politècnica de Catalunya-BarcelonaTech, R/Sant Nebridi, 22, GAIA Building (TR14), 08222 Terrassa, Spain; (L.M.P.); (O.H.); (N.I.); (J.M.)
- Department of Extremophilic Microorganisms Biology, Zabolotny Institute of Microbiology and Virology, National Academy of Sciences of Ukraine, 03143 Kyiv, Ukraine
| | - Nury Infante
- Laboratory of Sanitary and Environmental Microbiology (MSMLab)-UNESCO Chair on Sustainability, Universitat Politècnica de Catalunya-BarcelonaTech, R/Sant Nebridi, 22, GAIA Building (TR14), 08222 Terrassa, Spain; (L.M.P.); (O.H.); (N.I.); (J.M.)
| | - Maite Muniesa
- Departament de Genètica, Microbiologia i Estadística, Universitat de Barcelona, Diagonal 643 (Annex. Floor 0), 08028 Barcelona, Spain;
| | - Jordi Morató
- Laboratory of Sanitary and Environmental Microbiology (MSMLab)-UNESCO Chair on Sustainability, Universitat Politècnica de Catalunya-BarcelonaTech, R/Sant Nebridi, 22, GAIA Building (TR14), 08222 Terrassa, Spain; (L.M.P.); (O.H.); (N.I.); (J.M.)
| | - Ruslan Mariychuk
- Department of Ecology, Faculty of Humanities and Natural Sciences, University of Presov, 08001 Presov, Slovakia
| | - Tzanko Tzanov
- Grup de Biotecnologia Molecular i Industrial, Departament d’Enginyeria Química, Universitat Politècnica de Catalunya (UPC-BarcelonaTech), Rambla de Sant Nebridi 22, 08222 Terrassa, Spain;
| |
Collapse
|
78
|
Zhang L, Xu Q, Tan FC, Deng Y, Hakki M, Shelburne SA, Kirienko NV. Role of R5 Pyocin in the Predominance of High-Risk Pseudomonas aeruginosa Isolates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.07.616987. [PMID: 39416193 PMCID: PMC11483031 DOI: 10.1101/2024.10.07.616987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Infections with antimicrobial resistant pathogens, such as Pseudomonas aeruginosa, are a frequent occurrence in healthcare settings. Human P. aeruginosa infections are predominantly caused by a small number of sequence types (ST), such as ST235, ST111, and ST175. Although ST111 is recognized as one of the most prevalent high-risk P. aeruginosa clones worldwide and frequently exhibits multidrug-resistant or extensively drug-resistant phenotypes, the basis for this dominance remains unclear. In this study, we used a genome-wide transposon insertion library screen to discover that the competitive advantage of ST111 strains over certain non-ST111 strains is through production of R pyocins. We confirmed this finding by showing that competitive dominance was lost by ST111 mutants with R pyocin gene deletions. Further investigation showed that sensitivity to ST111 R pyocin (specifically R5 pyocin) is caused by deficiency in the O-antigen ligase waaL, which leaves lipopolysaccharide (LPS) bereft of O antigen, enabling pyocins to bind the LPS core. In contrast, sensitivity of waaL mutants to R1 or R2 pyocins depended on additional genomic changes. In addition, we found the PA14 mutants in lipopolysaccharide biosynthesis (waaL, wbpL, wbpM) that cause high susceptibility to R pyocins also exhibit poor swimming motility. Analysis of 5,135 typed P. aeruginosa strains revealed that several international, high-risk sequence types (including ST235, ST111, and ST175) are enriched for R5 pyocin production, indicating a correlation between these phenotypes and suggesting a novel approach for evaluating risk from emerging prevalent P. aeruginosa strains. Overall, our study sheds light on the mechanisms underlying the dominance of ST111 strains and highlighting the role of waaL in extending spectrum of R pyocin susceptibility.
Collapse
Affiliation(s)
- Liyang Zhang
- Department of BioSciences, Rice University, Houston, TX, United States
| | - Qi Xu
- Department of BioSciences, Rice University, Houston, TX, United States
- Department of Bioengineering, Rice University, Houston, TX, United States
| | - Filemon C Tan
- Department of BioSciences, Rice University, Houston, TX, United States
| | - Yanhan Deng
- Department of BioSciences, Rice University, Houston, TX, United States
| | - Morgan Hakki
- Division of Infectious Diseases, Department of Medicine, Oregon Health and Science University, Portland, OR, United States
| | - Samuel A. Shelburne
- Departments of Infectious Diseases and Genomic Medicine, MD Anderson Cancer Center, Houston TX
| | | |
Collapse
|
79
|
Cabal A, Hörtenhuber A, Salaheddin Y, Stöger A, Springer B, Bletz S, Mellmann A, Hyden P, Hartl R, Weinberger J, Conzemius R, Hell M, Daza-Prieto B, Lippert K, Steindl G, Köberl-Jelovcan S, Ruppitsch W. Three prolonged outbreaks of metallo-β-lactamase-producing Pseudomonas aeruginosa in an Upper Austrian hospital, 2017-2023. Microbiol Spectr 2024; 12:e0074024. [PMID: 39162508 PMCID: PMC11448029 DOI: 10.1128/spectrum.00740-24] [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: 03/22/2024] [Accepted: 07/01/2024] [Indexed: 08/21/2024] Open
Abstract
In spring 2022, an increase in metallo-β-lactamase-producing Pseudomonas aeruginosa (MBL-Pa) infections was detected in a hospital in Upper Austria. To identify the source of infection and to stop further transmissions, an epidemiological outbreak investigation including whole-genome sequencing (WGS)-based typing was conducted. The final case definition included cases admitted to the hospital between 2020 and 2023 with an MBL-Pa in one of the three genomic clusters identified. In addition, the investigation was extended to include historical cases from 2017. Core genome multilocus sequence typing was performed to assess the genetic relatedness between the isolates. Fifty-four clinical P. aeruginosa isolates and eight P. aeruginosa isolates from the hospital environment were obtained. All but nine isolates grouped into one of three genomic clusters (ST235/blaVIM-1, ST111/blaVIM-2, or ST621/blaIMP-13), which were considered to be distinct, prolonged outbreaks involving 47 out of 52 cases. The most likely source of infection for cluster 1 (ST111/blaVIM-2) and cluster 2 (ST235/blaVIM-1) was sinks in the intensive care unit (ICU) washroom. Cluster 3 clone (ST621/blaIMP-13) could have originated in the urology ward in 2020 and then spread to the ICU years later. However, the nosocomial origin of this clone could not be proven. In March 2023, following the implementation of control measures (gowning, patient isolation, screening, and daily disinfection), no further MLB-Pa was detected, and the outbreaks were considered to be over. As ICUs play an important role in the transmission of P. aeruginosa, emphasis should be placed on genomic surveillance, infection prevention, and control in such wards. IMPORTANCE The significance of our work lies in the successful resolution of three prolonged outbreaks of MBL-Pa infections in a hospital in Upper Austria. Through a comprehensive epidemiological investigation coupled with WGS-based typing of P. aeruginosa isolates, the study identified three distinct genomic clusters responsible for prolonged outbreaks involving 47 cases. The investigation pinpointed sinks in the ICU washroom as the likely source of infection for two of the clusters. The study demonstrates the effectiveness of control measures such as hand hygiene, gowning, patient isolation, screening, and disinfection in stopping further transmission and bringing the outbreaks to a close. This underscores the critical role of genomic surveillance and control measures, particularly in high-risk settings like ICUs, in reducing nosocomial transmission of MBL-Pa infections.
Collapse
Affiliation(s)
- Adriana Cabal
- Division for Public Health, Institute for Medical Microbiology and Hygiene, Austrian Agency for Health and Food Safety, Vienna, Austria
| | - Anna Hörtenhuber
- Institute of Pathology, Upper Austrian Health Holding GmbH, Pyhrn-Eisenwurzen Clinical Centre Kirchdorf Steyr, Steyr, Austria
| | - Yarub Salaheddin
- Institute of Pathology, Upper Austrian Health Holding GmbH, Pyhrn-Eisenwurzen Clinical Centre Kirchdorf Steyr, Steyr, Austria
| | - Anna Stöger
- Division for Public Health, Institute for Medical Microbiology and Hygiene, Austrian Agency for Health and Food Safety, Vienna, Austria
| | - Burkhard Springer
- Division for Public Health, Institute for Medical Microbiology and Hygiene, Austrian Agency for Health and Food Safety, Graz, Austria
| | - Stefan Bletz
- Institute of Hygiene, University Hospital Muenster and University of Muenster, Muenster, Germany
| | - Alexander Mellmann
- Institute of Hygiene, University Hospital Muenster and University of Muenster, Muenster, Germany
| | - Patrick Hyden
- Department of Statistics and Analytical Epidemiology, Austrian Agency for Health and Food Safety, Vienna, Austria
| | - Rainer Hartl
- National Reference Center for Antimicrobial Resistance, Institute for Hygiene, Microbiology and Tropical Medicine, Ordensklinikum Linz Elisabethinen, Linz, Austria
- Johannes Kepler University Linz, Medical Faculty, Linz, Austria
| | | | | | - Markus Hell
- MEDILAB, Teaching Laboratory of the Paracelsus Medical University, Salzburg, Austria
| | - Beatriz Daza-Prieto
- Division for Public Health, Institute for Medical Microbiology and Hygiene, Austrian Agency for Health and Food Safety, Vienna, Austria
| | - Kathrin Lippert
- Division for Public Health, Institute for Medical Microbiology and Hygiene, Austrian Agency for Health and Food Safety, Vienna, Austria
| | - Georg Steindl
- Institute for Hospital Hygiene and Microbiology (IKM), Graz, Austria
| | - Sandra Köberl-Jelovcan
- Division for Public Health, Institute for Medical Microbiology and Hygiene, Austrian Agency for Health and Food Safety, Graz, Austria
| | - Werner Ruppitsch
- Division for Public Health, Institute for Medical Microbiology and Hygiene, Austrian Agency for Health and Food Safety, Vienna, Austria
| |
Collapse
|
80
|
Chen KZM, Vu LM, Vollmer AC. Cultivation in long-term simulated microgravity is detrimental to pyocyanin production and subsequent biofilm formation ability of Pseudomonas aeruginosa. Microbiol Spectr 2024; 12:e0021124. [PMID: 39162544 PMCID: PMC11448113 DOI: 10.1128/spectrum.00211-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 06/27/2024] [Indexed: 08/21/2024] Open
Abstract
Pseudomonas aeruginosa forms aggregates known as biofilms. Previous studies have shown that when P. aeruginosa is cultivated in space, thicker and structurally different biofilms are formed than from those grown on Earth. We investigated how microgravity, simulated in a laboratory setting, influenced the growth, colonization, and virulence potentials of a P. aeruginosa PA14 wild-type strain, as well as two surface attachment-defective (sad) mutants altered at crucial biofilm-forming steps: flgK and pelA. Using high-aspect ratio rotating-wall vessel (HARV) bioreactors, P. aeruginosa bacteria were grown to stationary phase under prolonged (6 days) exposure to simulated microgravity or normal gravity conditions. After the exposure, the capacity of the culture to form biofilms was measured. Additionally, pigment (pyocyanin) formed by each culture during the incubation was extracted and quantified. We demonstrate that the first prolonged exposure to low-shear modeled microgravity (LSMMG) and without nutrient replenishment significantly diminishes wild-type P. aeruginosa PA14 biofilm formation abilities after exposure and pyocyanin production during exposure, while the mutant strains exhibit differing outcomes for both properties. IMPORTANCE Given plans for humans to engage in prolonged space travel, we investigated biofilm and pigment/virulence factor formation in Pseudomonas aeruginosa when cultivated in microgravity. These bacteria are opportunistic pathogens in immunocompromised individuals. Previous studies of space travelers have shown some immune system diminutions. Hence, our studies shed some light on how prolonged cultivation of bacteria in simulated microgravity conditions affect their growth characteristics.
Collapse
Affiliation(s)
| | - Linda My Vu
- Department of Biology, Swarthmore College, Swarthmore, Pennsylvania, USA
- Department of Microbial Pathogenesis, University of Maryland—Baltimore, Baltimore, Maryland, USA
| | - Amy Cheng Vollmer
- Department of Biology, Swarthmore College, Swarthmore, Pennsylvania, USA
| |
Collapse
|
81
|
da Cruz Nizer WS, Allison KN, Adams ME, Vargas MA, Ahmed D, Beaulieu C, Raju D, Cassol E, Howell PL, Overhage J. The role of exopolysaccharides Psl and Pel in resistance of Pseudomonas aeruginosa to the oxidative stressors sodium hypochlorite and hydrogen peroxide. Microbiol Spectr 2024; 12:e0092224. [PMID: 39194290 PMCID: PMC11448232 DOI: 10.1128/spectrum.00922-24] [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: 04/11/2024] [Accepted: 07/01/2024] [Indexed: 08/29/2024] Open
Abstract
Pseudomonas aeruginosa is well-known for its antimicrobial resistance and the ability to survive in harsh environmental conditions due to an abundance of resistance mechanisms, including the formation of biofilms and the production of exopolysaccharides. Exopolysaccharides are among the major components of the extracellular matrix in biofilms and aggregates of P. aeruginosa. Although their contribution to antibiotic resistance has been previously shown, their roles in resistance to oxidative stressors remain largely elusive. Here, we studied the function of the exopolysaccharides Psl and Pel in the resistance of P. aeruginosa to the commonly used disinfectants and strong oxidizing agents NaOCl and H2O2. We observed that the simultaneous inactivation of Psl and Pel in P. aeruginosa PAO1 mutant strain ∆pslA pelF resulted in a significant increase in susceptibility to both NaOCl and H2O2. Further analyses revealed that Pel is more important for oxidative stress resistance in P. aeruginosa and that the form of Pel (i.e., cell-associated or cell-free) did not affect NaOCl susceptibility. Additionally, we show that Psl/Pel-negative strains are protected against oxidative stress in co-culture biofilms with P. aeruginosa PAO1 WT. Taken together, our results demonstrate that the EPS matrix and, more specifically, Pel exhibit protective functions against oxidative stressors such as NaOCl and H2O2 in P. aeruginosa. IMPORTANCE Biofilms are microbial communities of cells embedded in a self-produced polymeric matrix composed of polysaccharides, proteins, lipids, and extracellular DNA. Biofilm bacteria have been shown to possess unique characteristics, including increased stress resistance and higher antimicrobial tolerance, leading to failures in bacterial eradication during chronic infections or in technical settings, including drinking and wastewater industries. Previous studies have shown that in addition to conferring structure and stability to biofilms, the polysaccharides Psl and Pel are also involved in antibiotic resistance. This work provides evidence that these biofilm matrix components also contribute to the resistance of Pseudomonas aeruginosa to oxidative stressors including the widely used disinfectant NaOCl. Understanding the mechanisms by which bacteria escape antimicrobial agents, including strong oxidants, is urgently needed in the fight against antimicrobial resistance and will help in developing new strategies to eliminate resistant strains in any environmental, industrial, and clinical setting.
Collapse
Affiliation(s)
| | - Kira N. Allison
- Department of Health Sciences, Carleton University, Ottawa, Ontario, Canada
| | - Madison E. Adams
- Department of Health Sciences, Carleton University, Ottawa, Ontario, Canada
| | - Mario A. Vargas
- Program in Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Duale Ahmed
- Department of Health Sciences, Carleton University, Ottawa, Ontario, Canada
| | - Carole Beaulieu
- Department of Health Sciences, Carleton University, Ottawa, Ontario, Canada
| | - Deepa Raju
- Program in Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Edana Cassol
- Department of Health Sciences, Carleton University, Ottawa, Ontario, Canada
| | - P. Lynne Howell
- Program in Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Joerg Overhage
- Department of Health Sciences, Carleton University, Ottawa, Ontario, Canada
| |
Collapse
|
82
|
Kunisch F, Campobasso C, Wagemans J, Yildirim S, Chan BK, Schaudinn C, Lavigne R, Turner PE, Raschke MJ, Trampuz A, Gonzalez Moreno M. Targeting Pseudomonas aeruginosa biofilm with an evolutionary trained bacteriophage cocktail exploiting phage resistance trade-offs. Nat Commun 2024; 15:8572. [PMID: 39362854 PMCID: PMC11450229 DOI: 10.1038/s41467-024-52595-w] [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: 05/29/2023] [Accepted: 09/12/2024] [Indexed: 10/05/2024] Open
Abstract
Spread of multidrug-resistant Pseudomonas aeruginosa strains threatens to render currently available antibiotics obsolete, with limited prospects for the development of new antibiotics. Lytic bacteriophages, the viruses of bacteria, represent a path to combat this threat. In vitro-directed evolution is traditionally applied to expand the bacteriophage host range or increase bacterial suppression in planktonic cultures. However, while up to 80% of human microbial infections are biofilm-associated, research towards targeted improvement of bacteriophages' ability to combat biofilms remains scarce. This study aims at an in vitro biofilm evolution assay to improve multiple bacteriophage parameters in parallel and the optimisation of bacteriophage cocktail design by exploiting a bacterial bacteriophage resistance trade-off. The evolved bacteriophages show an expanded host spectrum, improved antimicrobial efficacy and enhanced antibiofilm performance, as assessed by isothermal microcalorimetry and quantitative polymerase chain reaction, respectively. Our two-phage cocktail reveals further improved antimicrobial efficacy without incurring dual-bacteriophage-resistance in treated bacteria. We anticipate this assay will allow a better understanding of phenotypic-genomic relationships in bacteriophages and enable the training of bacteriophages against other desired pathogens. This, in turn, will strengthen bacteriophage therapy as a treatment adjunct to improve clinical outcomes of multidrug-resistant bacterial infections.
Collapse
Affiliation(s)
- Fabian Kunisch
- Faculty of Medicine, Universität Münster, Münster, Germany
- Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, USA
- Center for Phage Biology and Therapy, Yale University, New Haven, CT, USA
| | - Claudia Campobasso
- Department of Biosystems, KU Leuven, Leuven, Belgium
- Department of Biology, Università di Pisa, Pisa, Italy
| | | | - Selma Yildirim
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Benjamin K Chan
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, USA
- Center for Phage Biology and Therapy, Yale University, New Haven, CT, USA
| | - Christoph Schaudinn
- Advanced Light and Electron Microscopy (Zentrum für Biologische Gefahren und Spezielle Pathogene 4), Robert Koch Institute, Berlin, Germany
| | - Rob Lavigne
- Department of Biosystems, KU Leuven, Leuven, Belgium
| | - Paul E Turner
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, USA
- Center for Phage Biology and Therapy, Yale University, New Haven, CT, USA
- Program in Microbiology, Yale School of Medicine, New Haven, CT, USA
| | - Michael J Raschke
- Faculty of Medicine, Universität Münster, Münster, Germany
- Department of Trauma, Hand and Reconstructive Surgery, Universitätsklinikum Münster, Münster, Germany
| | - Andrej Trampuz
- Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany.
| | - Mercedes Gonzalez Moreno
- Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| |
Collapse
|
83
|
Ren Y, You X, Zhu R, Li D, Wang C, He Z, Hu Y, Li Y, Liu X, Li Y. Mutation of Pseudomonas aeruginosa lasI/rhlI diminishes its cytotoxicity, oxidative stress, inflammation, and apoptosis on THP-1 macrophages. Microbiol Spectr 2024; 12:e0414623. [PMID: 39162513 PMCID: PMC11448257 DOI: 10.1128/spectrum.04146-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 06/27/2024] [Indexed: 08/21/2024] Open
Abstract
The management of Pseudomonas aeruginosa (P. aeruginosa) infections presents a substantial challenge to clinics and public health, emphasizing the urgent need for innovative strategies to address this issue. Quorum sensing (QS) is an intercellular communication mechanism that coordinates bacterial activities involved in various virulence mechanisms, such as acquiring host nutrients, facilitating biofilm formation, enhancing motility, secreting virulence factors, and evading host immune responses, all of which play a crucial role in the colonization and infection of P. aeruginosa. The LasI/R and RhlI/R sub-systems dominate in the QS system of P. aeruginosa. Macrophages play a pivotal role in the host's innate immune response to P. aeruginosa invasion, particularly through phagocytosis as the initial host defense mechanism. This study investigated the effects of P. aeruginosa's QS system on THP-1 macrophages. Mutants of PAO1 with lasI/rhlI deletion, as well as their corresponding complemented strains, were obtained, and significant downregulation of QS-related genes was observed in the mutants. Furthermore, the ΔlasI and ΔlasIΔrhlI mutants exhibited significantly attenuated virulence in terms of biofilm formation, extracellular polymeric substances synthesis, bacterial adhesion, motility, and virulence factors production. When infected with ΔlasI and ΔlasIΔrhlI mutants, THP-1 macrophages exhibited enhanced scavenging ability against the mutants and demonstrated resistance to cytotoxicity, oxidative stress, inflammatory response, and apoptosis induced by the culture supernatants of these mutant strains. These findings offer novel insights into the mechanisms underlying how the lasI/rhlI mutation attenuates cytotoxicity, oxidative stress, inflammation, and apoptosis in macrophages induced by P. aeruginosa.IMPORTANCEP. aeruginosa is classified as one of the ESKAPE pathogens and poses a global public health concern. The QS system of this versatile pathogen contributes to a broad spectrum of virulence, thereby constraining therapeutic options for serious infections. This study illustrated that the lasI/rhlI mutation of the QS system plays a prominent role in attenuating the virulence of P. aeruginosa by affecting bacterial adhesion, biofilm formation, extracellular polymeric substances synthesis, bacterial motility, and virulence factors' production. Notably, THP-1 macrophages infected with mutant strains exhibited increased phagocytic activity in eliminating intracellular bacteria and enhanced resistance to cytotoxicity, oxidative stress, inflammation, and apoptosis. These findings suggest that targeted intervention toward the QS system is anticipated to diminish the pathogenicity of P. aeruginosa to THP-1 macrophages.
Collapse
Affiliation(s)
- Yanying Ren
- Dazhou integrated Traditional Chinese Medicine & Western Medicine Hospital, Dazhou Second People's Hospital, Dazhou, China
- Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiaojuan You
- Henan University of Chinese Medicine, Zhengzhou, China
- Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhenghzhou, China
| | - Rui Zhu
- Henan University of Chinese Medicine, Zhengzhou, China
- Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhenghzhou, China
| | - Dengzhou Li
- Henan University of Chinese Medicine, Zhengzhou, China
- Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhenghzhou, China
| | - Chunxia Wang
- Henan University of Chinese Medicine, Zhengzhou, China
- Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhenghzhou, China
| | - Zhiqiang He
- Henan University of Chinese Medicine, Zhengzhou, China
- Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhenghzhou, China
| | - Yue Hu
- Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhenghzhou, China
| | - Yifan Li
- Henan University of Chinese Medicine, Zhengzhou, China
| | - Xinwei Liu
- Henan University of Chinese Medicine, Zhengzhou, China
- Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhenghzhou, China
| | - Yongwei Li
- Henan University of Chinese Medicine, Zhengzhou, China
- Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhenghzhou, China
- The Key Laboratory of Pathogenic Microbes &Antimicrobial Resistance Surveillance of Zhengzhou, Zhengzhou, China
- Henan Engineering Research Center for Identification of Pathogenic Microbes, Zhengzhou, China
- Henan Provincial Key Laboratory of Antibiotics-Resistant Bacterial Infection Prevention & Therapy with Traditional Chinese Medicine, Zhengzhou, China
| |
Collapse
|
84
|
Kungwani NA, Panda J, Mishra AK, Chavda N, Shukla S, Vikhe K, Sharma G, Mohanta YK, Sharifi-Rad M. Combating bacterial biofilms and related drug resistance: Role of phyto-derived adjuvant and nanomaterials. Microb Pathog 2024; 195:106874. [PMID: 39181190 DOI: 10.1016/j.micpath.2024.106874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 08/27/2024]
Abstract
The emergence of antimicrobial resistance (AMR) in clinical microbes has led to a search for novel antibiotics for combating bacterial infections. The treatment of bacterial infections becomes more challenging with the onset of biofilm formation. AMR is further accelerated by biofilm physiology and differential gene expression in bacteria with an inherent resistance to conventional antibiotics. In the search for innovative strategies to control the spread of AMR in clinical isolates, plant-derived therapeutic metabolites can be repurposed to control biofilm-associated drug resistance. Unlike antibiotics, designed to act on a single cellular process, phytochemicals can simultaneously target multiple cellular components. Furthermore, they can disrupt biofilm formation and inhibit quorum sensing, offering a comprehensive approach to combat bacterial infections. In bacterial biofilms, the first line of AMR is due to biofilms associated with the extracellular matrix, diffusion barriers, quorum sensing, and persister cells. These extracellular barriers can be overcome using phytochemical-based antibiotic adjuvants to increase the efficacy of antibiotic treatment and restrict the spread of AMR. Furthermore, phytochemicals can be used to target bacterial intracellular machinery such as DNA replication, protein synthesis, efflux pumps, and degrading enzymes. In parallel with pristine phytochemicals, phyto-derived nanomaterials have emerged as an effective means of fighting bacterial biofilms. These nanomaterials can be formulated to cross the biofilm barriers and function on cellular targets. This review focuses on the synergistic effects of phytochemicals and phyto-derived nanomaterials in controlling the progression of biofilm-related AMR. IT provides comprehensive insights into recent advancements and the underlying mechanisms of the use of phyto-derived adjuvants and nanomaterials.
Collapse
Affiliation(s)
- Neelam Amit Kungwani
- Department of Environmental Biotechnology, Gujarat Biotechnology University, Gandhinagar, Gujarat, 382355, India.
| | - Jibanjyoti Panda
- Nano-biotechnology and Translational Knowledge Laboratory, Department of Applied Biology, School of Biological Sciences, University of Science and Technology Meghalaya, Techno City, 9th Mile, Baridua, Ri-Bhoi, 793101, Meghalaya, India
| | - Awdhesh Kumar Mishra
- Department of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk, 38541, Republic of Korea
| | | | - Sudhir Shukla
- Homi Bhabha National Institute, Biofouling and Biofilm Processes Section, WSCD, Chemistry Group, Bhabha Atomic Research Centre, Kalpakkam, Tamilnadu, 603102, India
| | - Kalyani Vikhe
- Department of Environmental Biotechnology, Gujarat Biotechnology University, Gandhinagar, Gujarat, 382355, India
| | - Gunjan Sharma
- Department of Plant Biotechnology, Gujarat Biotechnology University, Gandhinagar, Gujarat, 382355, India
| | - Yugal Kishore Mohanta
- Nano-biotechnology and Translational Knowledge Laboratory, Department of Applied Biology, School of Biological Sciences, University of Science and Technology Meghalaya, Techno City, 9th Mile, Baridua, Ri-Bhoi, 793101, Meghalaya, India; Centre for Herbal Pharmacology and Environmental Sustainability, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, 603103, Tamil Nadu, India.
| | - Majid Sharifi-Rad
- Department of Range and Watershed Management, Faculty of Water and Soil, University of Zabol, Zabol, 98613-35856, Iran.
| |
Collapse
|
85
|
Mazloumi Jourkouyeh E, Taslimi Eshkalak M, Faezi Ghasemi M, Zahmatkesh H, Rasti B, Zamani H. Diclofenac Sodium and Gentamicin Co-Encapsulated PLGA Nanoparticles: Targeting Extracellular Matrix Components to Combat Biofilm Formation in Pseudomonas aeruginosa PAO1. J CLUST SCI 2024; 35:2475-2488. [DOI: 10.1007/s10876-024-02675-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 07/23/2024] [Indexed: 01/05/2025]
|
86
|
Quang MT, Vo DHT, Nguyen MT. Establishment of a Pseudomonas aeruginosa Biofilm Model Using a Drip Flow Reactor: Evaluation of Antibiotic Efficacy against Biofilm Formation and Destruction. BIOMEDICAL AND BIOTECHNOLOGY RESEARCH JOURNAL 2024; 8:434-439. [DOI: 10.4103/bbrj.bbrj_281_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/19/2024] [Indexed: 01/04/2025]
Abstract
Abstract
Background:
Pseudomonas aeruginosa (PA) is a significant cause of nosocomial infections, particularly because its ability to form biofilms on medical devices complicates treatment. This study developed a biofilm model using a drip flow reactor (DFR) system and apply it to investigate the effects of antibiotics on biofilm formation.
Methods:
The biofilm-forming capacity of PA strains ATCC 9027, ATCC 27853, and PA 01 (a PA isolate from wastewater samples) was evaluated using a DFR system. This model was then used to assess biofilm resistance to antibiotics through flow cytometry analysis.
Results:
A PA biofilm model was successfully established in the DFR system, as evidenced by images of the carrier and flow cytometry analysis results. Significant differences in biofilm formation were observed among the tested strains (one-way ANOVA, P < 0.01). The timing of antibiotic administration was found to influence biofilm formation. The combination of colistin and rifampicin exerted a synergistic effect, significantly enhancing both the inhibition of biofilm formation and the destruction of existing biofilms compared with individual antibiotic treatments (one-way ANOVA, P < 0.01).
Conclusion:
This study successfully developed a PA biofilm model using the DFR system and applied it to evaluate the efficacy of colistin and rifampicin, both individually and in combination, against PA biofilms. The DFR model, coupled with flow cytometry analysis, is a valuable tool for investigating biofilm formation kinetics and assessing the impact of antibiotics on biofilm development and eradication.
Collapse
Affiliation(s)
- Minh Trong Quang
- Department of Microbiology and Parasitology, Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Duong Hai Thi Vo
- Department of Microbiology and Parasitology, Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Minh Thai Nguyen
- Department of Microbiology and Parasitology, Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| |
Collapse
|
87
|
Mohan A, Rajan PP, Kumar P, Jayakumar D, Mini M, Asha S, Vaikkathillam P. Theophylline as a quorum sensing and biofilm inhibitor in Pseudomonas aeruginosa and Chromobacterium violaceum. Int Microbiol 2024; 27:1457-1471. [PMID: 38342794 DOI: 10.1007/s10123-024-00487-w] [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: 10/14/2023] [Revised: 01/15/2024] [Accepted: 01/19/2024] [Indexed: 02/13/2024]
Abstract
Quorum sensing (QS) is pivotal in coordinating virulence factors and biofilm formation in various pathogenic bacteria, making it a prime target for disrupting bacterial communication. Pseudomonas aeruginosa is a member of the "ESKAPE" group of bacterial pathogens known for their association with antimicrobial resistance and biofilm formation. The current antibiotic arsenal falls short of addressing biofilm-related infections effectively, highlighting the urgent need for novel therapeutic agents. In this study, we explored the anti-QS and anti-biofilm properties of theophylline against two significant pathogens, Chromobacterium violaceum and P. aeruginosa. The production of violacein, pyocyanin, rhamnolipid, and protease was carried out, along with the evaluation of biofilm formation through methods including crystal violet staining, triphenyl tetrazolium chloride assay, and fluorescence microscopy. Furthermore, computational analyses were conducted to predict the targets of theophylline in the QS pathways of P. aeruginosa and C. violaceum. Our study demonstrated that theophylline effectively inhibits QS activity and biofilm formation in C. violaceum and P. aeruginosa. In P. aeruginosa, theophylline inhibited the production of key virulence factors, including pyocyanin, rhamnolipid, protease, and biofilm formation. The computational analyses suggest that theophylline exhibits robust binding affinity to CviR in C. violaceum and RhlR in P. aeruginosa, key participants in the QS-mediated biofilm pathways. Furthermore, theophylline also displays promising interactions with LasR and QscR in P. aeruginosa. Our study highlights theophylline as a versatile anti-QS agent and offers a promising avenue for future research to develop novel therapeutic strategies against biofilm-associated infections.
Collapse
Affiliation(s)
- Aparna Mohan
- Department of Zoology, Government College for Women, Thiruvananthapuram, -695014, Kerala, India
| | - Pooja P Rajan
- Department of Zoology, Government College for Women, Thiruvananthapuram, -695014, Kerala, India
| | - Praveen Kumar
- Department of Zoology, Government College for Women, Thiruvananthapuram, -695014, Kerala, India.
| | - Devi Jayakumar
- Department of Zoology, Government College for Women, Thiruvananthapuram, -695014, Kerala, India
| | - Minsa Mini
- Department of Zoology, Government College for Women, Thiruvananthapuram, -695014, Kerala, India
| | - Sneha Asha
- Department of Zoology, Government College for Women, Thiruvananthapuram, -695014, Kerala, India
| | - Parvathi Vaikkathillam
- Department of Zoology, Government College for Women, Thiruvananthapuram, -695014, Kerala, India
| |
Collapse
|
88
|
Theodorakis N, Feretzakis G, Hitas C, Kreouzi M, Kalantzi S, Spyridaki A, Boufeas IZ, Sakagianni A, Paxinou E, Verykios VS, Nikolaou M. Antibiotic Resistance in the Elderly: Mechanisms, Risk Factors, and Solutions. Microorganisms 2024; 12:1978. [PMID: 39458286 PMCID: PMC11509523 DOI: 10.3390/microorganisms12101978] [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: 09/22/2024] [Revised: 09/27/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
Antibiotic resistance presents a critical challenge in healthcare, particularly among the elderly, where multidrug-resistant organisms (MDROs) contribute to increased morbidity, mortality, and healthcare costs. This review focuses on the mechanisms underlying resistance in key bacterial pathogens and highlights how aging-related factors like immunosenescence, frailty, and multimorbidity increase the burden of infections from MDROs in this population. Novel strategies to mitigate resistance include the development of next-generation antibiotics like teixobactin and cefiderocol, innovative therapies such as bacteriophage therapy and antivirulence treatments, and the implementation of antimicrobial stewardship programs to optimize antibiotic use. Furthermore, advanced molecular diagnostic techniques, including nucleic acid amplification tests and next-generation sequencing, allow for faster and more precise identification of resistant pathogens. Vaccine development, particularly through innovative approaches like multi-epitope vaccines and nanoparticle-based platforms, holds promise in preventing MDRO infections among the elderly. The role of machine learning (ML) in predicting resistance patterns and aiding in vaccine and antibiotic development is also explored, offering promising solutions for personalized treatment and prevention strategies in the elderly. By integrating cutting-edge diagnostics, therapeutic innovations, and ML-based approaches, this review underscores the importance of multidisciplinary efforts to address the global challenge of antibiotic resistance in aging populations.
Collapse
Affiliation(s)
- Nikolaos Theodorakis
- Department of Cardiology, 65+ Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (N.T.); (C.H.); (M.N.)
- School of Medicine, National and Kapodistrian University of Athens, 75 Mikras Asias, 11527 Athens, Greece
| | - Georgios Feretzakis
- School of Science and Technology, Hellenic Open University, 18 Aristotelous Str., 26335 Patras, Greece; (G.F.); (E.P.)
| | - Christos Hitas
- Department of Cardiology, 65+ Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (N.T.); (C.H.); (M.N.)
| | - Magdalini Kreouzi
- Department of Internal Medicine, 65+ Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (M.K.); (S.K.); (A.S.)
| | - Sofia Kalantzi
- Department of Internal Medicine, 65+ Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (M.K.); (S.K.); (A.S.)
| | - Aikaterini Spyridaki
- Department of Internal Medicine, 65+ Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (M.K.); (S.K.); (A.S.)
| | - Iris Zoe Boufeas
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London, 64 Turner Street, London E1 2AD, UK;
| | - Aikaterini Sakagianni
- Intensive Care Unit, Sismanogelio General Hospital, 37 Sismanogleiou Str., 15126 Marousi, Greece;
| | - Evgenia Paxinou
- School of Science and Technology, Hellenic Open University, 18 Aristotelous Str., 26335 Patras, Greece; (G.F.); (E.P.)
| | - Vassilios S. Verykios
- School of Science and Technology, Hellenic Open University, 18 Aristotelous Str., 26335 Patras, Greece; (G.F.); (E.P.)
| | - Maria Nikolaou
- Department of Cardiology, 65+ Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (N.T.); (C.H.); (M.N.)
| |
Collapse
|
89
|
Kim C, Oh KK, Jothi R, Park DS. An innovative approach to decoding genetic variability in Pseudomonas aeruginosa via amino acid repeats and gene structure profiles. Sci Rep 2024; 14:22610. [PMID: 39349595 PMCID: PMC11443150 DOI: 10.1038/s41598-024-73031-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 09/12/2024] [Indexed: 10/04/2024] Open
Abstract
Pseudomonas aeruginosa, a common pathogen in nosocomial infections, presents significant global health challenges due to its high prevalence and mortality rates. However, the origins and distribution of this bacterium remain unclear, partly due to the lack of effective gene typing methods. This situation necessitates the establishment of trustworthy and high-resolution protocol for differentiating closely related P. aeruginosa strains. In this context, the present study attempted to undertake a comparative genomic analysis of multiple P. aeruginosa strains available in the public database NCBI, with the goal of identifying potential genetic markers for measuring the genetic diversity. The preliminary comparative analysis of 816 P. aeruginosa strains revealed notable variations in two genes-specifically, the CDF family iron/cobalt efflux transporter AitP and the protease modulator HflC-across 44 strains. These variations were associated with single amino acid repeats (SHRs) that responsible for encoding histidine residue. Additionally, comparative gene map analysis revealed differential clustering patterns in the Rsx and TAXI genes among 16 strains. Interestingly, the gene structure pattern observed in TAXI groups displayed a strong correlation with the SHRs pattern in the CDF and HflC groups. In addition, the SHRs pattern of CDF and HflC were strongly correlated with MLST sequence type number. Overall, the study present a novel genetic markers based on SHRs and gene cluster patterns, offering a reliable method for genotyping of P. aeruginosa.
Collapse
Affiliation(s)
- Chaerin Kim
- Microbial Safety Division, Rural Development Administration, National Institute of Agricultural Sciences, Wanju, 55365, Republic of Korea
| | - Kwang-Kyo Oh
- Microbial Safety Division, Rural Development Administration, National Institute of Agricultural Sciences, Wanju, 55365, Republic of Korea
| | - Ravi Jothi
- Microbial Safety Division, Rural Development Administration, National Institute of Agricultural Sciences, Wanju, 55365, Republic of Korea
| | - Dong Suk Park
- Microbial Safety Division, Rural Development Administration, National Institute of Agricultural Sciences, Wanju, 55365, Republic of Korea.
| |
Collapse
|
90
|
Otava UE, Tervo L, Havela R, Vuotari L, Ylänne M, Asplund A, Patpatia S, Kiljunen S. Phage-Antibiotic Combination Therapy against Recurrent Pseudomonas Septicaemia in a Patient with an Arterial Stent. Antibiotics (Basel) 2024; 13:916. [PMID: 39452183 PMCID: PMC11504013 DOI: 10.3390/antibiotics13100916] [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: 08/27/2024] [Revised: 09/16/2024] [Accepted: 09/20/2024] [Indexed: 10/26/2024] Open
Abstract
Background: Intravascular stent infections are often associated with high risks of morbidity and mortality. We report here a case of a patient with an arterial stent and recurrent Pseudomonas septicaemias successfully treated with phage-meropenem combination therapy. Methods: A 75-year-old female with arteriosclerosis and comorbidities went through a femoropopliteal bypass with prosthesis in the right inguinal area. After the bypass, she developed a recurring Pseudomonas aeruginosa infection and also neutropenia during different antibiotics. A rapidly growing pseudoaneurysm in the right inguinal area led to an emergency intra-arterial stent placement during blood stream infection, later suspected to host a P. aeruginosa biofilm. Removing the stent was deemed precarious, and phage therapy was considered as a compassionate treatment option. A three-phage cocktail infecting the P. aeruginosa strain was prepared and administered intravenously together with meropenem for two weeks, after which, a ten-month follow-up was carried out. Results: No adverse reactions occurred during the phage therapy treatment, while infection markers were normalized. In addition, recovery was seen in a PET-CT scan. During the 10-month follow-up, no further P. aeruginosa septicaemias occurred. Conclusions: Phage-meropenem combination therapy was thus found safe and effective in the treatment of recurrent Pseudomonas septicaemia in a patient with an arterial stent.
Collapse
Affiliation(s)
- Ulla Elina Otava
- Department of Internal Medicine, Tampere University Hospital, 33520 Tampere, Finland (L.T.); (R.H.)
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland;
| | - Laura Tervo
- Department of Internal Medicine, Tampere University Hospital, 33520 Tampere, Finland (L.T.); (R.H.)
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland;
| | - Riikka Havela
- Department of Internal Medicine, Tampere University Hospital, 33520 Tampere, Finland (L.T.); (R.H.)
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland;
| | - Liisa Vuotari
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland;
- Department of Clinical Physiology and Nuclear Medicine, Tampere University Hospital, 33520 Tampere, Finland
| | - Matti Ylänne
- Human Microbiome Research Program, Research Programs Unit and Medicum, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland (S.P.)
| | - Annette Asplund
- Human Microbiome Research Program, Research Programs Unit and Medicum, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland (S.P.)
| | - Sheetal Patpatia
- Human Microbiome Research Program, Research Programs Unit and Medicum, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland (S.P.)
| | - Saija Kiljunen
- Human Microbiome Research Program, Research Programs Unit and Medicum, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland (S.P.)
- PrecisionPhage Ltd., 40500 Jyväskylä, Finland
| |
Collapse
|
91
|
Hourihane E, Hixon KR. Nanoparticles as Drug Delivery Vehicles for People with Cystic Fibrosis. Biomimetics (Basel) 2024; 9:574. [PMID: 39329596 PMCID: PMC11430251 DOI: 10.3390/biomimetics9090574] [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: 07/29/2024] [Revised: 08/29/2024] [Accepted: 09/11/2024] [Indexed: 09/28/2024] Open
Abstract
Cystic Fibrosis (CF) is a life-shortening, genetic disease that affects approximately 145,000 people worldwide. CF causes a dehydrated mucus layer in the lungs, leading to damaging infection and inflammation that eventually result in death. Nanoparticles (NPs), drug delivery vehicles intended for inhalation, have become a recent source of interest for treating CF and CF-related conditions, and many formulations have been created thus far. This paper is intended to provide an overview of CF and the effect it has on the lungs, the barriers in using NP drug delivery vehicles for treatment, and three common material class choices for these NP formulations: metals, polymers, and lipids. The materials to be discussed include gold, silver, and iron oxide metallic NPs; polyethylene glycol, chitosan, poly lactic-co-glycolic acid, and alginate polymeric NPs; and lipid-based NPs. The novelty of this review comes from a less specific focus on nanoparticle examples, with the focus instead being on the general theory behind material function, why or how a material might be used, and how it may be preferable to other materials used in treating CF. Finally, this paper ends with a short discussion of the two FDA-approved NPs for treatment of CF-related conditions and a recommendation for the future usage of NPs in people with Cystic Fibrosis (pwCF).
Collapse
Affiliation(s)
- Eoin Hourihane
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA;
| | - Katherine R. Hixon
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA;
- Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA
| |
Collapse
|
92
|
Gómez SG, Ginebra MP, Gil FJ, Barraquer RI, Manero JM. Antibacterial and cytocompatible silver coating for titanium Boston Keratoprosthesis. Front Bioeng Biotechnol 2024; 12:1421706. [PMID: 39364264 PMCID: PMC11446748 DOI: 10.3389/fbioe.2024.1421706] [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: 04/22/2024] [Accepted: 09/02/2024] [Indexed: 10/05/2024] Open
Abstract
The Boston Keratoprosthesis (BKPro) serves as a medical solution for restoring vision in complex cases of corneal blindness. Comprising a front plate made of polymethylmethacrylate (PMMA) and a back plate of titanium (Ti), this device utilizes the beneficial biomaterial properties of Ti. While BKPro demonstrates promising retention rates, infection emerges as a significant concern that impacts its long-term efficacy. However, limited research exists on enhancement of BKPros through intrinsic infection-preventing mechanisms. In this regard, metal ions, especially the well-known Ag+ ions, are a promising alternative to obtain implants with innate antibacterial properties. However, little information is available about the effects of Ag in corneal tissue, especially within human corneal keratocytes (HCKs). In this work, an electrodeposition treatment using a constant pulse is proposed to attach Ag complexes onto rough Ti surfaces, thus providing antibacterial properties without inducing cytotoxicity. Complete physicochemical characterization and ion release studies were carried out with both control and Ag-treated samples. The possible cytotoxic effects in the short and long term were evaluated in vitro with HCKs. Moreover, the antibacterial properties of the silver-treated surfaces were tested against the gram-negative bacterial strain Pseudomonas aeruginosa and the gram-positive strain Staphylococcus epidermidis, that are common contributors to infections in BKPros. Physicochemical characterization confirmed the presence of silver, predominantly in oxide form, with low release of Ag+ ions. Ag-treated surfaces demonstrated no cytotoxicity and promoted long-term proliferation of HCKs. Furthermore, the silver-treated surfaces exhibited a potent antibacterial effect, causing a reduction in bacterial adhesion and evident damage to the bacterial cell walls of P. aeruginosa and S. epidermidis. The low release of Ag+ ions suggested reactive oxygen species (ROS)-mediated oxidative stress imbalance as the bactericidal mechanism of the silver deposits. In conclusion, the proposed electrodeposition technique confers antibacterial protection to the Ti backplate of BKPro, mitigating implant-threatening infections while ensuring non-cytotoxicity within the corneal tissue.
Collapse
Affiliation(s)
- Silvia González Gómez
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya. Barcelona Tech (UPC), Barcelona East School of Engineering (EEBE), Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, UPC, EEBE, Barcelona, Spain
| | - Maria-Pau Ginebra
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya. Barcelona Tech (UPC), Barcelona East School of Engineering (EEBE), Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, UPC, EEBE, Barcelona, Spain
- Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, Spain
| | - Francisco Javier Gil
- Bioengineering Institute of Technology. Universitat Internacional de Catalunya. Barcelona, Barcelona, Spain
| | - Rafael I Barraquer
- Centro de Oftalmología Barraquer, Barcelona, Spain
- Institut Universitari Barraquer, Universitat Autònoma de Barcelona, Barcelona, Spain
- Universitat Internacional de Catalunya (UIC), Barcelona, Spain
| | - José María Manero
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya. Barcelona Tech (UPC), Barcelona East School of Engineering (EEBE), Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, UPC, EEBE, Barcelona, Spain
| |
Collapse
|
93
|
Arefian Jazi M, Hajikhani B, Goudarzi M, Ebrahimipour G. Exploiting immunopotential PAPI-1 encoded type IVb major pilin targeting Pseudomonas aeruginosa. Heliyon 2024; 10:e36859. [PMID: 39281519 PMCID: PMC11401190 DOI: 10.1016/j.heliyon.2024.e36859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 09/18/2024] Open
Abstract
Pseudomonas aeruginosa (P. aeruginosa) significantly contributes to nosocomial infections and necessitates research into novel treatment methods. For the first time, this research evaluated the immunoprotective potential of recombinant PAPI-1 encoded type IV pili targeting P. aeruginosa in BALB/C mice. The target sequence was identified, and a PilS2-encoding vector was constructed. The vector was then expressed and purified in E. coli BL21 (DE3). The PilS2 protein was inoculated into BALB/C mice in four groups, with or without alum, to measure total IgG, its subclasses, and cytokines. MTT and opsonophagocytosis tests were used to examine the immunological response. PilS2, especially when paired with alum, boosts the humoral immune response by enhancing IgG and IL-4 levels. However, PilS2 did not affect IL-17 or IFN-γ and only increased lymphocyte proliferation. Antibodies targeting PilS2 increased phagocytic cell death of P. aeruginosa by over 95 %, indicating possible therapies for P. aeruginosa infections. Our study on the immunopotentiation of P. aeruginosa PilS2 paves the way for pilin-based vaccines and immunotherapy targeting this pathogen.
Collapse
Affiliation(s)
- Mojgan Arefian Jazi
- Department of Microbiology and Microbial Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
- Department of Microbiology, School of Medicine Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bahareh Hajikhani
- Department of Microbiology, School of Medicine Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Goudarzi
- Department of Microbiology, School of Medicine Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Gholamhossein Ebrahimipour
- Department of Microbiology and Microbial Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| |
Collapse
|
94
|
Thakur B, Kaur S, Dwibedi V, Albadrani GM, Al-Ghadi MQ, Abdel-Daim MM. Unveiling the antimicrobial and antibiofilm potential of biosurfactant produced by newly isolated Lactiplantibacillus plantarum strain 1625. Front Microbiol 2024; 15:1459388. [PMID: 39318434 PMCID: PMC11420119 DOI: 10.3389/fmicb.2024.1459388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 08/09/2024] [Indexed: 09/26/2024] Open
Abstract
The present study aimed to characterize the biosurfactants synthesized by lactic acid bacteria (LAB) obtained from fermented foods, optimize the conditions for increasing the yield of biosurfactants and explore their antimicrobial and antibiofilm potential. Out of the 26 LAB isolates, isolate BS2 showed the highest biosurfactant production as indicated in the oil displacement test, drop collapse and emulsification activity. BS2 was identified as Lactiplantibacillus plantarum 1625 using 16S-rRNA gene sequencing and phylogenetic analysis. The biosurfactant produced by BS2 was identified as an anionic glycol-lipo-proteins by employing Fourier Transform Infrared Spectroscopy (FTIR) and Gas Chromatography-Mass Spectrometry (GC-MS) analysis. The biosurfactants produced by L. plantarum 1625 demonstrated strong antibacterial and antibiofilm characteristics against pathogenic strains such as Staphylococcus aureus MTCC 1049, Escherichia coli MTCC 1587, and Pseudomonas putida MTCC 1655. The minimal inhibition concentration value of antibacterial activity was found to be 0.1 mg/mL with the inhibition percentage ranging from 90 to 95%. Further, the effect of temperature, pH, and substrate composition on biosurfactant production was also studied to enhance it production using the Box-Behnken Design approach of Response surface methodology (RSM). Application of biosurfactant led to a considerable decrease in biofilm-forming harmful bacteria, as proven by scanning electron microscopy analysis. The results highlight the potential uses of biosurfactants in distinct industries, and biotechnological contexts, especially in the creation of new antimicrobial and antibiofilm agents.
Collapse
Affiliation(s)
- Babita Thakur
- Department of Biotechnology, University Institute of Biotechnology, Chandigarh University, Mohali, Punjab, India
| | - Sukhminderjit Kaur
- Department of Biotechnology, University Institute of Biotechnology, Chandigarh University, Mohali, Punjab, India
| | - Vagish Dwibedi
- Department of Biotechnology, University Institute of Biotechnology, Chandigarh University, Mohali, Punjab, India
| | - Ghadeer M. Albadrani
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Muath Q. Al-Ghadi
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mohamed M. Abdel-Daim
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, Jeddah, Saudi Arabia
- Department of Pharmacology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
95
|
Liu YS, Zhang C, Khoo BL, Hao P, Chua SL. Dual-species proteomics and targeted intervention of animal-pathogen interactions. J Adv Res 2024:S2090-1232(24)00383-7. [PMID: 39233003 DOI: 10.1016/j.jare.2024.08.038] [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: 06/15/2024] [Revised: 08/30/2024] [Accepted: 08/31/2024] [Indexed: 09/06/2024] Open
Abstract
INTRODUCTION Host-microbe interactions are important to human health and ecosystems globally, so elucidating the complex host-microbe interactions and associated protein expressions drives the need to develop sensitive and accurate biochemical techniques. Current proteomics techniques reveal information from the point of view of either the host or microbe, but do not provide data on the corresponding partner. Moreover, it remains challenging to simultaneously study host-microbe proteomes that reflect the direct competition between host and microbe. This raises the need to develop a dual-species proteomics method for host-microbe interactions. OBJECTIVES We aim to establish a forward + reverse Stable Isotope Labeling with Amino acids in Cell culture (SILAC) proteomics approach to simultaneously label and quantify newly-expressed proteins of host and microbe without physical isolation, for investigating mechanisms in direct host-microbe interactions. METHODS Using Caenorhabditis elegans-Pseudomonas aeruginosa infection model as proof-of-concept, we employed SILAC proteomics and molecular pathway analysis to characterize the differentially-expressed microbial and host proteins. We then used molecular docking and chemical characterization to identify chemical inhibitors that intercept host-microbe interactions and eliminate microbial infection. RESULTS Based on our proteomics results, we studied the iron competition between pathogen iron scavenger and host iron uptake protein, where P. aeruginosa upregulated pyoverdine synthesis protein (PvdA) (fold-change of 5.2313) and secreted pyoverdine, and C. elegans expressed ferritin (FTN-2) (fold-change of 3.4057). Targeted intervention of iron competition was achieved using Galangin, a ginger-derived phytochemical that inhibited pyoverdine production and biofilm formation in P. aeruginosa. The Galangin-ciprofloxacin combinatorial therapy could eliminate P. aeruginosa biofilms in a fish wound infection model, and enabled animal survival. CONCLUSION Our work provides a novel SILAC-based proteomics method that can simultaneously evaluate host and microbe proteomes, with future applications in higher host organisms and other microbial species. It also provides insights into the mechanisms dictating host-microbe interactions, offering novel strategies for anti-infective therapy.
Collapse
Affiliation(s)
- Yang Sylvia Liu
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong Special Administrative Region
| | - Chengqian Zhang
- School of Life Science and Technology, ShanghaiTech University, China
| | - Bee Luan Khoo
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong Special Administrative Region; Hong Kong Center for Cerebro-Cardiovascular Health Engineering (COCHE), Hong Kong Special Administrative Region; City University of Hong Kong-Shenzhen Futian Research Institute, Shenzhen, China
| | - Piliang Hao
- School of Life Science and Technology, ShanghaiTech University, China.
| | - Song Lin Chua
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong Special Administrative Region; State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Kowloon, Hong Kong Special Administrative Region; Research Centre for Deep Space Explorations (RCDSE), The Hong Kong Polytechnic University, Kowloon, Hong Kong Special Administrative Region.
| |
Collapse
|
96
|
Tuytschaever T, Faille C, Raes K, Sampers I. Influence of slope, material, and temperature on Listeria monocytogenes and Pseudomonas aeruginosa mono- and dual-species biofilms. BIOFOULING 2024; 40:467-482. [PMID: 39054784 DOI: 10.1080/08927014.2024.2380410] [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: 03/19/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/27/2024]
Abstract
Understanding factors influencing Listeria monocytogenes biofilms aid in developing more effective elimination/prevention strategies. This study examined the effect of temperature (4 °C, 21 °C, 30 °C), materials (stainless steel 316 L with 2B and 2 R finishes, glass, and polypropylene), and slope (0°/horizontal or 90°/vertical) on mono- and dual-species biofilms using two L. monocytogenes strains and one Pseudomonas aeruginosa strain. All biofilms were grown in 10% TSB for 24 h and analyzed using culture-based methods. Additionally, the architecture of monospecies biofilms was studied using fluorescence microscopy. Overall, P. aeruginosa showed higher biofilm formation potential (6.2 log CFU/cm2) than L. monocytogenes (4.0 log CFU/cm2). Temperature greatly influenced P. aeruginosa and varied for L. monocytogenes. The slope predominantly influenced L. monocytogenes monospecies biofilms, with cell counts increasing by up to 2 log CFU/cm2. Surface material had little impact on biofilm formation. The study highlights the varying effects of different parameters on multispecies biofilms and the importance of surface geometry.
Collapse
Affiliation(s)
- Tessa Tuytschaever
- Research Unit VEG-i-TEC, Department of Food Technology, Safety, and Health, Faculty of Bioscience Engineering, Ghent University, Campus Kortrijk, Kortrijk, Belgium
| | - Christine Faille
- Univ. Lille, CNRS, INRAE, Centrale Lille, UMR 8207 - UMET - Unité Matériaux et Transformations, Lille, France
| | - Katleen Raes
- Research Unit VEG-i-TEC, Department of Food Technology, Safety, and Health, Faculty of Bioscience Engineering, Ghent University, Campus Kortrijk, Kortrijk, Belgium
| | - Imca Sampers
- Research Unit VEG-i-TEC, Department of Food Technology, Safety, and Health, Faculty of Bioscience Engineering, Ghent University, Campus Kortrijk, Kortrijk, Belgium
| |
Collapse
|
97
|
Pintea-Simon IA, Bancu L, Mare AD, Ciurea CN, Toma F, Man A. Rapid Molecular Diagnostics of Pneumonia Caused by Gram-Negative Bacteria: A Clinician's Review. Antibiotics (Basel) 2024; 13:805. [PMID: 39334980 PMCID: PMC11429159 DOI: 10.3390/antibiotics13090805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 08/21/2024] [Accepted: 08/24/2024] [Indexed: 09/30/2024] Open
Abstract
With approximately half a billion events per year, lower respiratory tract infections (LRTIs) represent a major challenge for the global public health. Among LRTI cases, those caused by Gram-negative bacteria (GNB) are associated with a poorer prognostic. Standard-of-care etiologic diagnostics is lengthy and difficult to establish, with more than half of cases remaining microbiologically undocumented. Recently, syndromic molecular diagnostic panels became available, enabling simultaneous detection of tens of pathogen-related and antimicrobial-resistance genetic markers within a few hours. In this narrative review, we summarize the available data on the performance of molecular diagnostics in GNB pneumonia, highlighting the main strengths and limitations of these assays, as well as the main factors influencing their clinical utility. We searched MEDLINE and Web of Science databases for relevant English-language articles. Molecular assays have higher analytical sensitivity than cultural methods, and show good agreement with standard-of-care diagnostics regarding detection of respiratory pathogens, including GNB, and identification of frequent patterns of resistance to antibiotics. Clinical trials reported encouraging results on the usefulness of molecular assays in antibiotic stewardship. By providing early information on the presence of pathogens and their probable resistance phenotypes, these assays assist in the choice of targeted therapy, in shortening the time from sample collection to appropriate antimicrobial treatment, and in reducing unnecessary antibiotic use.
Collapse
Affiliation(s)
- Ionela-Anca Pintea-Simon
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mures, 540142 Târgu Mures, Romania
- Department of Internal Medicine M3, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mures, 540142 Târgu Mures, Romania
| | - Ligia Bancu
- Department of Internal Medicine M3, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mures, 540142 Târgu Mures, Romania
| | - Anca Delia Mare
- Department of Microbiology, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540142 Târgu Mures, Romania
| | - Cristina Nicoleta Ciurea
- Department of Microbiology, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540142 Târgu Mures, Romania
| | - Felicia Toma
- Department of Microbiology, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540142 Târgu Mures, Romania
| | - Adrian Man
- Department of Microbiology, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540142 Târgu Mures, Romania
| |
Collapse
|
98
|
Li S, Ren A, Li M, Li G, Yang L, Jia T. Extraction of Bacterial Membrane Vesicle and Phage Complex by Density Gradient Ultracentrifugation. Bio Protoc 2024; 14:e5050. [PMID: 39210957 PMCID: PMC11349496 DOI: 10.21769/bioprotoc.5050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 09/04/2024] Open
Abstract
The bacterial membrane vesicles (MVs) are non-replicative, nanoscale structures that carry specific cargos and play multiple roles in microbe-host interactions. An appropriate MV isolation method that mimics complex pathogen infections in vivo is needed. After bacterial MVs extraction, flagella or pili can be frequently observed along with MVs by transmission electron microscope (TEM). Recently, MVs from Pseudomonas aeruginosa were found to coexist with Pf4 phages, and this MV-phages complex exhibited a different impact on host cell innate immunity compared with MVs or phages solely. The presence of this MVs-phages complex simulates the real condition of complex pathogen infections within the host. This protocol outlines the extraction of the MVs and Pf4 phages complex of P. aeruginosa PAO1, including the respective isolation and qualification approaches. Our step-by-step bacterial MVs-phages complex extraction protocol provides valuable insights for further studying microbe-host cell interactions and the development of novel phage therapies. Key features • Detailed density gradient extraction procedures of MVs-phages complex • TEM, plaque assay, and PCR to verify the coexistence of MVs and phages • The obtained MVs-phages complex can be used for exploring phage-microbe-host cell interactions Graphical overview.
Collapse
Affiliation(s)
- Shangru Li
- Shenzhen National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Anmin Ren
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Menglu Li
- Shenzhen National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Guobao Li
- Shenzhen National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Liang Yang
- Shenzhen National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Tianyuan Jia
- Shenzhen National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
99
|
Jantaruk P, Teerapo K, Charoenwutthikun S, Roytrakul S, Kunthalert D. Anti-Biofilm and Anti-Inflammatory Properties of the Truncated Analogs of the Scorpion Venom-Derived Peptide IsCT against Pseudomonas aeruginosa. Antibiotics (Basel) 2024; 13:775. [PMID: 39200075 PMCID: PMC11352108 DOI: 10.3390/antibiotics13080775] [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: 07/16/2024] [Revised: 08/05/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen in humans and a frequent cause of severe nosocomial infections and fatal infections in immunocompromised individuals. Its ability to form biofilms has been the main driving force behind its resistance to almost all conventional antibiotics, thereby limiting treatment efficacy. In an effort to discover novel therapeutic agents to fight P. aeruginosa-associated biofilm infections, the truncated analogs of scorpion venom-derived peptide IsCT were synthesized and their anti-biofilm properties were examined. Among the investigated peptides, the IsCT-Δ6-8 peptide evidently showed the most potential anti-P. aeruginosa biofilm activity and the effect was not due to bacterial growth inhibition. The IsCT-Δ6-8 peptide also exhibited inhibitory activity against the production of pyocyanin, an important virulence factor of P. aeruginosa. Furthermore, the IsCT-Δ6-8 peptide significantly suppressed the production of inflammatory mediators nitric oxide and interleukin-6 in P. aeruginosa LPS-induced macrophages. Due to its low cytotoxicity to mammalian cells, the IsCT-Δ6-8 peptide emerges as a promising candidate with significant anti-biofilm and anti-inflammatory properties. These findings highlight its potential application in treating P. aeruginosa-related biofilm infections.
Collapse
Affiliation(s)
- Pornpimon Jantaruk
- Department of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand; (P.J.); (K.T.); (S.C.)
| | - Kittitat Teerapo
- Department of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand; (P.J.); (K.T.); (S.C.)
| | - Supattra Charoenwutthikun
- Department of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand; (P.J.); (K.T.); (S.C.)
| | - Sittiruk Roytrakul
- Functional Proteomics Technology Laboratory, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Thailand Science Park, Pathumthani 12120, Thailand;
| | - Duangkamol Kunthalert
- Department of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand; (P.J.); (K.T.); (S.C.)
- Centre of Excellence in Medical Biotechnology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
| |
Collapse
|
100
|
George M, Narayanan S, Tejada-Arranz A, Plack A, Basler M. Initiation of H1-T6SS dueling between Pseudomonas aeruginosa. mBio 2024; 15:e0035524. [PMID: 38990002 PMCID: PMC11323562 DOI: 10.1128/mbio.00355-24] [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: 02/03/2024] [Accepted: 06/10/2024] [Indexed: 07/12/2024] Open
Abstract
The Type VI secretion system (T6SS) is a multicomponent apparatus, present in many Gram-negative bacteria, which can inhibit bacterial prey in various ecological niches. Pseudomonas aeruginosa assembles one of its three T6SS (H1-T6SS) to respond to attacks from adjacent competing bacteria. Surprisingly, repeated assemblies of the H1-T6SS, termed dueling, were described in a monoculture in the absence of an attacker strain; however, the underlying mechanism was unknown. Here, we explored the role of H2-T6SS of P. aeruginosa in triggering H1-T6SS assembly. We show that H2-T6SS inactivation in P. aeruginosa causes a significant reduction in H1-T6SS dueling and that H2-T6SS activity directly triggers retaliation by the H1-T6SS. Intraspecific competition experiments revealed that elimination of H2-T6SS in non-immune prey cells conferred protection from H1-T6SS. Moreover, we show that the H1-T6SS response is triggered independently of the characterized lipase effectors of the H2-T6SS, as well as those of Acinetobacter baylyi and Vibrio cholerae. Our results suggest that H1-T6SS response to H2-T6SS in P. aeruginosa can impact intraspecific competition, particularly when the H1-T6SS effector-immunity pairs differ between strains, and could determine the outcome of multistrain colonization.IMPORTANCEThe opportunistic pathogen Pseudomonas aeruginosa harbors three different Type VI secretion systems (H1, H2, and H3-T6SS), which can translocate toxins that can inhibit bacterial competitors or inflict damage to eukaryotic host cells. Unlike the unregulated T6SS assembly in other Gram-negative bacteria, the H1-T6SS in P. aeruginosa is precisely assembled as a response to various cell damaging attacks from neighboring bacterial cells. Surprisingly, it was observed that neighboring P. aeruginosa cells repeatedly assemble their H1-T6SS toward each other. Mechanisms triggering this "dueling" behavior between sister cells were unknown. In this report, we used a combination of microscopy, genetic and intraspecific competition experiments to show that H2-T6SS initiates H1-T6SS dueling. Our study highlights the interplay between different T6SS clusters in P. aeruginosa, which may influence the outcomes of multistrain competition in various ecological settings such as biofilm formation and colonization of cystic fibrosis lungs.
Collapse
Affiliation(s)
- M. George
- Biozentrum, University of Basel, Basel, Switzerland
| | - S. Narayanan
- Biozentrum, University of Basel, Basel, Switzerland
| | | | - A. Plack
- Biozentrum, University of Basel, Basel, Switzerland
| | - M. Basler
- Biozentrum, University of Basel, Basel, Switzerland
| |
Collapse
|