1
|
Monteiro R, Sousa AM, Pereira MO. Aspartic acid unveils as antibiofilm agent and tobramycin adjuvant against mucoid and small colony variants of Pseudomonas aeruginosa isolates in vitro within cystic fibrosis airway mucus. Biofilm 2025; 9:100252. [PMID: 39866543 PMCID: PMC11759549 DOI: 10.1016/j.bioflm.2024.100252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 12/07/2024] [Accepted: 12/29/2024] [Indexed: 01/28/2025] Open
Abstract
Antibiotics are central to managing airway infections in cystic fibrosis (CF), yet current treatments often fail due to the presence of Pseudomonas aeruginosa biofilms, settling down the need for seeking therapies targeting biofilms. This study aimed to investigate the antibiofilm activity of aspartic acid and its potential as an adjuvant to tobramycin against P. aeruginosa biofilms formed by mucoid and small colony variant (SCV) tobramycin tolerant strain. We assessed the effect of aspartic acid on both surface-attached and suspended P. aeruginosa biofilms within CF artificial mucus and investigated the synergistic impact of combining it with non-lethal tobramycin concentrations. Our findings showed that aspartic acid inhibited planktonic P. aeruginosa without affecting its viability and prevented biofilm formation by hindering bacterial adhesion or interfering with EPS production, depending on the experimental conditions. In CF mucus, aspartic acid significantly reduced bacterial growth, with the highest inhibition observed when combined with tobramycin, showing notable effects against the mucoid and tolerant SCV strain. Despite these reductions, P. aeruginosa repopulated the mucus within 24 h of stress withdrawal. Additional strategies, including delayed tobramycin application and a second dose of co-application of aspartic acid and tobramycin were explored to address bacterial survival and recovery. Although none of the strategies eradicated P. aeruginosa, the second co-application resulted in slower bacterial recovery rates. In conclusion, this study highlighted aspartic acid as an effective antibiofilm agent and demonstrated for the first time its potential as an adjuvant to tobramycin. The combined use of aspartic acid and tobramycin offers a promising advancement in CF therapeutics, particularly against P. aeruginosa biofilms formed by mucoid and SCV strains, mitigating their antibiotic resistance.
Collapse
Affiliation(s)
- Rosana Monteiro
- CEB - Centre of Biological Engineering, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal
| | - Ana Margarida Sousa
- CEB - Centre of Biological Engineering, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal
- LABBELS – Associate Laboratory, Braga/Guimarães, Portugal
| | - Maria Olívia Pereira
- CEB - Centre of Biological Engineering, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal
- LABBELS – Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
2
|
Wang Z, Giedraitis E, Knoop C, Breiner DJ, Phelan VV, Van Bambeke F. Modeling reciprocal adaptation of Staphylococcus aureus and Pseudomonas aeruginosa co-isolates in artificial sputum medium. Biofilm 2025; 9:100279. [PMID: 40290724 PMCID: PMC12033965 DOI: 10.1016/j.bioflm.2025.100279] [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: 01/19/2025] [Revised: 04/01/2025] [Accepted: 04/07/2025] [Indexed: 04/30/2025] Open
Abstract
Co-infections by Staphylococcus aureus and Pseudomonas aeruginosa are frequent in the airways of patients with cystic fibrosis. These co-infections show higher antibiotic tolerance in vitro compared to mono-infections. In vitro models have been developed to study the interspecies interactions between P. aeruginosa and S. aureus. However, these model systems fail to incorporate clinical isolates with diverse phenotypes, do not reflect the nutritional environment of the CF airway mucus, and/or do not model the biofilm mode of growth observed in the CF airways. Here, we established a dual-species biofilm model grown in artificial sputum medium, where S. aureus was inoculated before P. aeruginosa to facilitate the maintenance of both species over time. It was successfully applied to ten pairs of clinical isolates exhibiting different phenotypes. Co-isolates from individual patients led to robust, stable co-cultures, supporting the theory of cross-adaptation in vivo. Investigation into the cross-adaptation of the VBB496 co-isolate pair revealed that both the P. aeruginosa and S. aureus isolates had reduced antagonism, in part due to reduced production of P. aeruginosa secondary metabolites as well as higher tolerance to those metabolites by S. aureus. Together, these results indicate that the two-species biofilm model system provides a useful tool for exploring interspecies interactions of P. aeruginosa and S. aureus in the context of CF airway infections.
Collapse
Affiliation(s)
- Zhifen Wang
- Pharmacologie Cellulaire et moléculaire, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Emily Giedraitis
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Christiane Knoop
- Erasme Hospital, Université libre de Bruxelles, Brussels, Belgium
| | - Daniel J. Breiner
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Vanessa V. Phelan
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Françoise Van Bambeke
- Pharmacologie Cellulaire et moléculaire, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
3
|
Garcia-Maset R, Chu V, Yuen N, Blumgart D, Yoon J, Murray BO, Joseph AA, Rohn JL. Effect of host microenvironment and bacterial lifestyles on antimicrobial sensitivity and implications for susceptibility testing. NPJ ANTIMICROBIALS AND RESISTANCE 2025; 3:42. [PMID: 40399473 PMCID: PMC12095824 DOI: 10.1038/s44259-025-00113-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 05/01/2025] [Indexed: 05/23/2025]
Abstract
Bacterial infections remain a major global health issue, with antimicrobial resistance (AMR) worsening the crisis. However, treatment failure can occur even when bacteria show antibiotic susceptibility in diagnostic tests. We explore factors such as phenotypic resilience, bacterial lifestyles such as biofilms, and differences between laboratory tests and real infection sites, highlighting the need for improved platforms to better predict treatment outcomes, and reviewing emerging technologies aimed at improving susceptibility testing.
Collapse
Affiliation(s)
- Ramon Garcia-Maset
- Centre for Urological Biology, Department of Renal Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK.
| | - Victoria Chu
- Centre for Urological Biology, Department of Renal Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK
| | - Nicholas Yuen
- Centre for Urological Biology, Department of Renal Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK
| | - Dalia Blumgart
- Centre for Urological Biology, Department of Renal Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK
| | - Jenny Yoon
- Centre for Urological Biology, Department of Renal Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK
| | - Benjamin O Murray
- Centre for Urological Biology, Department of Renal Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK
| | - Amelia A Joseph
- Nottingham University Hospitals NHS Trust, Nottingham, NG5 1PB, UK
| | - Jennifer L Rohn
- Centre for Urological Biology, Department of Renal Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK.
| |
Collapse
|
4
|
Saleh NM, Sudan ESF, Mohamed SH, El-Shahed MM, Hamouda RE, El-Gendy AO, Farag AA. Pathogen Partnerships or Power Struggles? Pseudomonas aeruginosa, and Staphylococcus aureus Dynamics in Cystic Fibrosis. Curr Microbiol 2025; 82:236. [PMID: 40198369 DOI: 10.1007/s00284-025-04167-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 02/26/2025] [Indexed: 04/10/2025]
Abstract
Cystic fibrosis (CF) is a polymicrobial infection characterized by interactions among various bacterial species that affect one another's cohabitation. The investigation of interspecies interactions in dual infections is essential to understand their reaction in the environment better and assist in the development of treatment regimens and innovative disease control approaches. Our hypothesis posits that co-infection interactions promote the adaptation of Staphylococcus aureus and Pseudomonas aeruginosa, potentially leading to synergistic action. To explore this, we examined dual-species interactions in co-isolated pairs of these organisms from Egyptian CF patients using laboratory media and artificial sputum media (ASM). Based on demographic data, 82 collected bacterial isolates from single, dual, and triple cultures were identified from 50 enrolled patients. In the interaction of the pairs in mimic media, P. aeruginosa exo-products significantly enhanced the biofilm formation and growth of S. aureus. Conversely, S. aureus did not inhibit P. aeruginosa biofilm formation. Furthermore, the biofilm mode of dual-organism growth provides protection in the CF context, as bacterial biofilms can withstand much higher antimicrobial levels compared to planktonically grown bacteria. Additionally, key biofilm genes regulated by quorum sensing were differentially expressed in both species in an isolate-dependent manner, highlighting their significant role in coexistence dual-species biofilm coexistence. In conclusion, our study illuminates the competitive and cooperative interactions between these two pathogens, which impact their coexistence and encourage biofilm production. This, in turn, accelerates disease progression and compromises patient health.
Collapse
Affiliation(s)
- Neveen M Saleh
- Department of Microbiology, Egyptian Drug Authority (Former National Organization for Drug Control and Research (NODCAR)), Giza, Egypt.
- College of Public Health, University of Nebraska Medical Center (UNMC), Omaha, United States.
| | - Esraa S F Sudan
- Department of Microbiology, Egyptian Drug Authority (Former National Organization for Drug Control and Research (NODCAR)), Giza, Egypt
| | - Sara H Mohamed
- Department of Microbiology, Egyptian Drug Authority (Former National Organization for Drug Control and Research (NODCAR)), Giza, Egypt
| | - Maram M El-Shahed
- Department of Microbiology, Egyptian Drug Authority (Former National Organization for Drug Control and Research (NODCAR)), Giza, Egypt
| | - Reda E Hamouda
- Department of Animal Production Systems Research, Animal Production Research Institute, Agricultural Research Center, Ministry of Agriculture and Land Reclamation, Giza, Egypt
| | - Ahmed Osama El-Gendy
- Microbiology and Immunology Department, Faculty of Pharmacy, Beni-Suef University, Beni Suef, 62514, Egypt
| | - Ahmed A Farag
- Microbiology and Immunology Department, Faculty of Pharmacy, Beni-Suef University, Beni Suef, 62514, Egypt
| |
Collapse
|
5
|
Fang P, Konyali D, Fischer E, Mayer RP, Huang J, Elena AX, Orzechowski GH, Tony-Odigie A, Kneis D, Dalpke A, Krebs P, Li B, Berendonk TU, Klümper U. Effects of Cigarette-Derived Compounds on the Spread of Antimicrobial Resistance in Artificial Human Lung Sputum Medium, Simulated Environmental Media, and Wastewater. ENVIRONMENTAL HEALTH PERSPECTIVES 2025; 133:47003. [PMID: 40032488 PMCID: PMC11980918 DOI: 10.1289/ehp14704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 11/20/2024] [Accepted: 01/21/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND Antimicrobial resistance (AMR) and smoking of tobacco products are two of the most important threats to global human health. Both are associated with millions of deaths every year. Surprisingly, the immediate interactions between these two threats remain poorly understood. OBJECTIVES We aimed to elucidate the effect of toxic compounds from cigarette smoke, ashes, and filters on the spread of antibiotic resistance genes in human lung and environmental microbiomes. METHODS Conjugation experiments using donor and recipient strain pairs of either Pseudomonas putida or Escherichia coli and AMR-encoding plasmids were conducted under exposure to different concentrations of cigarette smoke condensate in lung sputum medium, as well as cigarette ash and filter leachate in environmental media. We further measured reactive oxygen species (ROS) production of the donor strain under exposure to the cigarette-derived compounds to explore whether stress experienced by the bacteria could be one of the underlying mechanisms of change in plasmid transfer frequencies. Furthermore, used cigarette filters were submerged in a wastewater stream for several weeks, and the colonizing communities were analyzed using high-throughput sequencing and high-throughput quantitative polymerase chain reaction and compared with communities colonizing unused control filters. RESULTS Exposure to cigarette smoke condensate at relevant concentrations resulted in > 2 -fold higher transfer rates of a multidrug-resistance-encoding plasmid in artificial lung sputum medium. This was associated with higher ROS production as part of the bacterial stress response when exposed to cigarette-derived toxicants. Similar results were obtained for cigarette ash leachate in an environmental medium. Further, used cigarette filters were colonized by different microbial communities compared with unused filters. Those communities were significantly enriched with potential human pathogens and AMR. DISCUSSION The results of this study suggest that cigarette-derived compounds can indeed promote the spread of AMR within simulated human lung and environmental conditions. This study highlights that the consumption of cigarettes has not only direct but may also have indirect adverse effects on human health by promoting AMR. https://doi.org/10.1289/EHP14704.
Collapse
Affiliation(s)
- Peiju Fang
- Institute of Hydrobiology, Technische Universität (TU) Dresden, Dresden, Germany
- Institute of Environment and Ecology, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Diala Konyali
- Institute of Hydrobiology, Technische Universität (TU) Dresden, Dresden, Germany
| | - Emily Fischer
- Institute of Hydrobiology, Technische Universität (TU) Dresden, Dresden, Germany
| | - Robin Pascal Mayer
- Institute of Urban and Industrial Water Management, TU Dresden, Dresden, Germany
| | - Jin Huang
- Institute of Environment and Ecology, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Alan Xavier Elena
- Institute of Hydrobiology, Technische Universität (TU) Dresden, Dresden, Germany
| | | | - Andrew Tony-Odigie
- Institute of Medical Microbiology and Virology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
- Department of Infectious Diseases, Medical Microbiology and Hygiene, University Hospital Heidelberg, Heidelberg, Germany
| | - David Kneis
- Institute of Hydrobiology, Technische Universität (TU) Dresden, Dresden, Germany
| | - Alexander Dalpke
- Institute of Medical Microbiology and Virology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
- Department of Infectious Diseases, Medical Microbiology and Hygiene, University Hospital Heidelberg, Heidelberg, Germany
| | - Peter Krebs
- Institute of Urban and Industrial Water Management, TU Dresden, Dresden, Germany
| | - Bing Li
- Institute of Environment and Ecology, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Thomas U. Berendonk
- Institute of Hydrobiology, Technische Universität (TU) Dresden, Dresden, Germany
| | - Uli Klümper
- Institute of Hydrobiology, Technische Universität (TU) Dresden, Dresden, Germany
| |
Collapse
|
6
|
Cai YM, Hong F, De Craemer A, Malone JG, Crabbé A, Coenye T. Echinacoside reduces intracellular c-di-GMP levels and potentiates tobramycin activity against Pseudomonas aeruginosa biofilm aggregates. NPJ Biofilms Microbiomes 2025; 11:40. [PMID: 40055321 PMCID: PMC11889090 DOI: 10.1038/s41522-025-00673-2] [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: 09/16/2024] [Accepted: 02/25/2025] [Indexed: 05/13/2025] Open
Abstract
Cyclic diguanylate (c-di-GMP) is a central biofilm regulator in Pseudomonas aeruginosa, where increased intracellular levels promote biofilm formation and antibiotic tolerance. Targeting the c-di-GMP network may be a promising anti-biofilm approach, but most strategies studied so far aimed at eliminating surface-attached biofilms, while in vivo P. aeruginosa biofilms often occur as suspended aggregates. Here, the expression profile of c-di-GMP metabolism-related genes was analysed among 32 P. aeruginosa strains grown as aggregates in synthetic cystic fibrosis sputum. The diguanylate cyclase SiaD proved essential for auto-aggregation under in vivo-like conditions. Virtual screening predicted a high binding affinity of echinacoside towards the active site of SiaD. Echinacoside reduced c-di-GMP levels and aggregate sizes and potentiated tobramycin activity against aggregates in >80% of strains tested. This synergism was also observed in P. aeruginosa-infected 3-D alveolar epithelial cells and murine lungs, demonstrating echinacoside's potential as an adjunctive therapy for recalcitrant P. aeruginosa infections.
Collapse
Affiliation(s)
- Yu-Ming Cai
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium.
- John Innes Centre, Norwich Research Park, Colney Lane, Norwich, UK.
| | - Feng Hong
- Group of Microbiological Engineering and Biomedical Materials, College of Biological Science and Medical Engineering, Donghua University, North Ren Min Road 2999, 201620, Shanghai, China
- National Advanced Functional Fiber Innovation Centre, Wu Jiang, Su Zhou, China
| | - Amber De Craemer
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | - Jacob George Malone
- John Innes Centre, Norwich Research Park, Colney Lane, Norwich, UK
- School of Biological Sciences, University of East Anglia, Norwich, UK
| | - Aurélie Crabbé
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | - Tom Coenye
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium.
| |
Collapse
|
7
|
Jouault A, Jeguirim I, Kaddour IBH, Touqui L. Assessment of the efficacy of an antimicrobial peptide in the context of cystic fibrosis airways. CURRENT RESEARCH IN MICROBIAL SCIENCES 2025; 8:100367. [PMID: 40129463 PMCID: PMC11931299 DOI: 10.1016/j.crmicr.2025.100367] [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] [Indexed: 03/26/2025] Open
Abstract
Antimicrobial peptides (AMPs) offer a promising alternative to control airway infections with multi-resistant bacteria, such as methicillin-resistant Staphylococcus aureus (MRSA), which commonly infects patients with cystic fibrosis (CF). However, the behavior of AMPs in the CF context has yet to be fully elucidated. CF airways produce large amounts of proteases and viscous mucus (sputum), which may affect the efficacy of AMPs. The present work aimed to determine whether CF conditions affect the bactericidal efficacy of CAMA, a promising AMP known to kill clinical MRSA strains efficiently. Using a killing assay, we quantified CAMA bactericidal activity on a CF clinical MRSA strain in the presence of several compounds of CF airways, including sputum and bronchial epithelial cells (BECs). Our results indicate that CF sputum impairs the bactericidal efficacy of CAMA. Similar results were observed when CAMA was incubated with an artificial sputum medium (ASM). When used separately, sputum components (DNA, lipids, and mucins) reproduced the inhibitory effects of ASM. Additionally, the bactericidal efficacy of CAMA was also slightly altered when planktonic S. aureus strains were co-cultured with CF BECs. However, CAMA was not active against S. aureus cultured on BEC in biofilm mode, characteristic of chronic infections in CF patients. These findings suggest that although CAMA represents a promising tool to treat MRSA strains, the CF environment may impair the efficacy of this AMP. Identifying strategies to protect AMPs from the deleterious effects of CF sputum is a key priority.
Collapse
Affiliation(s)
- Albane Jouault
- Institut Pasteur, Université de Paris Cité, Mucoviscidose et Bronchopathies Chroniques, Paris, France
- Centre de Recherche Saint-Antoine, Sorbonne Université, Inserm, Paris, France
| | - Inès Jeguirim
- Institut Pasteur, Université de Paris Cité, Mucoviscidose et Bronchopathies Chroniques, Paris, France
- Centre de Recherche Saint-Antoine, Sorbonne Université, Inserm, Paris, France
| | - Inès Ben Hadj Kaddour
- Institut Pasteur, Université de Paris Cité, Mucoviscidose et Bronchopathies Chroniques, Paris, France
- Centre de Recherche Saint-Antoine, Sorbonne Université, Inserm, Paris, France
| | - Lhousseine Touqui
- Institut Pasteur, Université de Paris Cité, Mucoviscidose et Bronchopathies Chroniques, Paris, France
- Centre de Recherche Saint-Antoine, Sorbonne Université, Inserm, Paris, France
| |
Collapse
|
8
|
Bellavita R, Casciaro B, Nocerino V, Palladino S, Loffredo MR, Dardano P, De Stefano L, Falcigno L, D'Auria G, Galdiero S, Falanga A. Myxinidin-analogs able to sequester Fe(III): Metal-based gun to combat Pseudomonas aeruginosa biofilm. J Inorg Biochem 2025; 263:112774. [PMID: 39561505 DOI: 10.1016/j.jinorgbio.2024.112774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/04/2024] [Accepted: 11/11/2024] [Indexed: 11/21/2024]
Abstract
Bacteria have developed a tendency to form biofilms, where bacteria live in organized structures embedded in a self-produced matrix of DNA, proteins, and polysaccharides. Additionally, bacteria need iron(III) as an essential nutrient for bacterial growth and secrete siderophore groups that sequester it from the environment. To design a molecule able both to inhibit the bacteria and to sequester iron, we developed two hydroxamate-based peptides derived from an analog (WMR-4), previously developed in our lab, of the antimicrobial peptide myxinidin. In detail, we proposed a combination of WMR-4 with the hydroxamic acid resulting in the peptides WMR-7 and WMR-16 which differ for the length of the linker between the antimicrobial moiety and the siderophore. Both peptides were characterized through a set of different biophysical experiments to investigate their ability to sequester Fe3+. The peptide‑iron(III) complexes were studied through the UV-visible spectroscopy in organic solvent to eliminate water competition, and in acidic water to avoid iron precipitation. The complexes were also characterized by performing electrochemistry, circular dichroism and NMR spectroscopy experiments. In addition, we demonstrated the ability of peptide‑iron(III) complexes to inhibit the biofilm of Pseudomonas aeruginosa and to have an impact on the cell motility. This metal-based approach consisting in a hydroxamic acid conjugation represents a promising strategy to enhance the antibiofilm activity of antimicrobial peptides against one of most dangerous bacteria such as Pseudomonas aeruginosa.
Collapse
Affiliation(s)
- Rosa Bellavita
- Department of Pharmacy, School of Medicine, University of Naples 'Federico II', Via Domenico Montesano 49, 80131 Naples, Italy
| | - Bruno Casciaro
- Laboratory Affiliated to Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences, Sapienza University of Rome, Rome, Italy
| | - Valeria Nocerino
- National Research Council (ISASI-CNR), Institute of Applied Sciences and Intelligent Systems, Napoli, Italy
| | - Sara Palladino
- Department of Pharmacy, School of Medicine, University of Naples 'Federico II', Via Domenico Montesano 49, 80131 Naples, Italy
| | - Maria Rosa Loffredo
- Laboratory Affiliated to Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences, Sapienza University of Rome, Rome, Italy
| | - Principia Dardano
- National Research Council (ISASI-CNR), Institute of Applied Sciences and Intelligent Systems, Napoli, Italy
| | - Luca De Stefano
- National Research Council (ISASI-CNR), Institute of Applied Sciences and Intelligent Systems, Napoli, Italy
| | - Lucia Falcigno
- Department of Pharmacy, School of Medicine, University of Naples 'Federico II', Via Domenico Montesano 49, 80131 Naples, Italy
| | - Gabriella D'Auria
- Department of Pharmacy, School of Medicine, University of Naples 'Federico II', Via Domenico Montesano 49, 80131 Naples, Italy
| | - Stefania Galdiero
- Department of Pharmacy, School of Medicine, University of Naples 'Federico II', Via Domenico Montesano 49, 80131 Naples, Italy
| | - Annarita Falanga
- Department of Agricultural Science, University of Naples 'Federico II', Via Università 100, Portici, 80055 Portici, Italy.
| |
Collapse
|
9
|
Farha MA, Tu MM, Brown ED. Important challenges to finding new leads for new antibiotics. Curr Opin Microbiol 2025; 83:102562. [PMID: 39603107 DOI: 10.1016/j.mib.2024.102562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/15/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024]
Abstract
Identification of new antibiotics remains a huge challenge. The last antibiotic of new chemical class and mechanism was discovered more than 30 years ago. Advances since have been largely incremental modifications to a limited number of chemical scaffolds. Discovering and developing truly new antibiotics is challenging: the science is complex, and the development process is time consuming and expensive. Herein, we focus on the discovery phase of modern antibacterial research and development. We argue that antibacterial discovery has been challenged by a poor understanding of bacterial permeability, by generic in vitro conventions that ignore the host, and by the inherent complexity of bacterial systems. Together, these factors have colluded to challenge modern, industrial, and reductionist approaches to antibiotic discovery. Nevertheless, advances in our understanding of many of these obstacles, including a new appreciation for the complexity of both host and pathogen biology, bode well for future efforts.
Collapse
Affiliation(s)
- Maya A Farha
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada; Michael G. DeGroote Institute of Infectious Disease Research, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Megan M Tu
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada; Michael G. DeGroote Institute of Infectious Disease Research, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Eric D Brown
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada; Michael G. DeGroote Institute of Infectious Disease Research, McMaster University, Hamilton, Ontario L8N 3Z5, Canada.
| |
Collapse
|
10
|
Monteiro R, Silva E, Pereira MO, Sousa AM. Mechanistic Insights into Succinic Acid as an Adjuvant for Ciprofloxacin in Treating Pseudomonas aeruginosa Growing Within Cystic Fibrosis Airway Mucus. Microorganisms 2024; 12:2538. [PMID: 39770741 PMCID: PMC11678660 DOI: 10.3390/microorganisms12122538] [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: 10/31/2024] [Revised: 11/27/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025] Open
Abstract
Pseudomonas aeruginosa is a major cause of chronic respiratory infections in patients with cystic fibrosis (CF), with biofilm formation contributing to its persistence and antibiotic resistance. This study aimed to gain insights into the mechanistic action of succinic acid as a ciprofloxacin adjuvant against clinically relevant CF isolates, including small colony variants and mucoid strains, and a ciprofloxacin-resistant strain grown within CF dense mucus. Time-kill assays in artificial CF mucus, along with planktonic and surface-attached biofilm experiments, were used to assess the activity of succinic acid alone and in combination with sublethal ciprofloxacin concentrations. Succinic acid demonstrated an adjuvant effect of ciprofloxacin against P. aeruginosa grown within CF mucus at pH levels below pKa1 during the early bacterial growth stages. In examining planktonic growth and biofilms under these conditions, we found that succinic acid demonstrated strong antibacterial and antibiofilm properties. Conversely, succinic acid activity decreased at later growth stages, though it enhanced the ciprofloxacin effect, especially against mucoid biofilms. Moreover, we noted that, in dense CF mucus, succinic acid activity was attenuated compared to a non-CF environment, indicating diffusion challenges. These findings underscore the potential of succinic acid as a therapeutic adjuvant for improving antibiotic treatment outcomes and overcoming biofilm-associated resistance in CF.
Collapse
Affiliation(s)
- Rosana Monteiro
- Centre of Biological Engineering, LIBRO—Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; (R.M.); (E.S.); (M.O.P.)
| | - Eduarda Silva
- Centre of Biological Engineering, LIBRO—Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; (R.M.); (E.S.); (M.O.P.)
| | - Maria Olivia Pereira
- Centre of Biological Engineering, LIBRO—Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; (R.M.); (E.S.); (M.O.P.)
- LABBELS—Associate Laboratory, Braga/Guimarães, Portugal
| | - Ana Margarida Sousa
- Centre of Biological Engineering, LIBRO—Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; (R.M.); (E.S.); (M.O.P.)
- LABBELS—Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
11
|
Neetu, Ramya TNC. A comparative study of the efficacy of alginate lyases in the presence of metal ions elevated in the cystic fibrosis lung milieu. Biochem Biophys Rep 2024; 40:101821. [PMID: 39286289 PMCID: PMC11404220 DOI: 10.1016/j.bbrep.2024.101821] [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: 07/16/2024] [Revised: 08/23/2024] [Accepted: 08/29/2024] [Indexed: 09/19/2024] Open
Abstract
Pseudomonas aeruginosa, a common cause of morbidity in cystic fibrosis, chronically infects the patient's lungs by forming an alginate-containing biofilm. Alginate lyases are polysaccharide lyases that lyse alginate and are, therefore, potential biofilm-disruptive agents. However, cystic fibrosis sputum contains high levels of metals such as iron and zinc. The efficacy of alginate lyases under these conditions of elevated metal concentrations has not been categorically determined. Here, we have assessed the enzyme activity of two exolytic and five endolytic alginate lyases in the presence of metal ions (Fe2+, Zn2+, Mn2+, Mg2+, Ca2+, Ni2+, Cu2+) elevated in the cystic fibrosis lung milieu. Several of these alginate lyases exhibited increased activity in the presence of Ca2+, and the polysaccharide lyase family 7 members studied here exhibited decreased activity in the presence of Zn2+. The enzyme activity of the PL7 alginate lyases from Cellulophaga algicola (CaAly/CaFLDAly) and Vibrio sp. (VspAlyVI) was not affected in the presence of a mix of all the above-mentioned metal ions at the elevated concentrations found in the cystic fibrosis lung milieu. Specific alginate lyases might, therefore, retain the ability to degrade the alginate-containing Pseudomonas biofilm in the presence of metal ions elevated in the cystic fibrosis lung milieu.
Collapse
Affiliation(s)
- Neetu
- CSIR- Institute of Microbial Technology, Sector 39-A, Chandigarh, 160036, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - T N C Ramya
- CSIR- Institute of Microbial Technology, Sector 39-A, Chandigarh, 160036, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| |
Collapse
|
12
|
Duong K, Moss E, Reichhardt C. Solid-state NMR compositional analysis of sputum from people with cystic fibrosis. SOLID STATE NUCLEAR MAGNETIC RESONANCE 2024; 134:101975. [PMID: 39489104 DOI: 10.1016/j.ssnmr.2024.101975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/12/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
People with the genetic disease cystic fibrosis (CF) often have chronic airway infections and produce airway secretions called sputum. A better understanding of sputum composition is desired in order to track changes in response to CF therapeutics and to improve laboratory models for the study of CF airway infections. The glycosylated protein mucin is a primary component. Along with extracellular DNA, mucin gives rise to the high viscoelasticity of sputum, which inhibits airway clearance and is thought to promote chronic airway infections in people with CF. Past studies of sputum composition identified additional biomolecular components of sputum including other proteins, both glycosylated and not glycosylated, free amino acids, and lipids. Typically, studies of sputum, as well as other complex biological materials, have focused on soluble or isolated components. Solid-state NMR is not limited to the study of soluble components. Instead, it can provide molecular-level information about insoluble biological samples. Additionally, solid-state NMR can provide information about sample composition without requiring any processing of the sample, eliminating the possibility of misestimating certain components due to insolubility or potential sample loss in isolation steps. In this study, we used both 13C and 31P CPMAS to investigate the total composition of sputum samples obtained from six people with CF. We compared these spectra to those of commercially available mucin, DNA, and phospholipid samples. Lastly, we performed complementary biochemical analyses to identify specific proteins present in the sputum samples. Overall, our findings provide insight into the composition of unprocessed sputum samples from people with CF, which can be used as a benchmark for future investigations of CF and infections in the airways of people with CF. Further, this study provides opportunities to expand the solid-state NMR approach to include dynamic nuclear polarization (DNP) to obtain high-resolution information of sputum and similar biological samples that are not feasible to isotopically enrich.
Collapse
Affiliation(s)
- Kathy Duong
- Department of Chemistry, Washington University, St. Louis, MO, 63130, United States
| | - Evan Moss
- Department of Chemistry, Washington University, St. Louis, MO, 63130, United States
| | - Courtney Reichhardt
- Department of Chemistry, Washington University, St. Louis, MO, 63130, United States.
| |
Collapse
|
13
|
Baker EJ, Allcott G, Cox JAG. Polymicrobial infection in cystic fibrosis and future perspectives for improving Mycobacterium abscessus drug discovery. NPJ ANTIMICROBIALS AND RESISTANCE 2024; 2:38. [PMID: 39843836 PMCID: PMC11721438 DOI: 10.1038/s44259-024-00060-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 11/03/2024] [Indexed: 01/24/2025]
Abstract
Polymicrobial communities inhabit the cystic fibrosis (CF) airway, whereby microbial interactions can occur. One prominent CF pathogen is Mycobacterium abscessus, whose treatment is largely unsuccessful. This creates a need to discover novel antimicrobial agents to treat M. abscessus, however the methods used within antibiotic discovery are typically monomicrobial. This review will discuss this pathogen whilst considering the CF polymicrobial environment, to highlight future perspectives to improve M. abscessus drug discovery.
Collapse
Affiliation(s)
- Emily J Baker
- College of Health and Life Sciences, Aston University, Aston Triangle, Birmingham, B4 7ET, UK
| | - Gemma Allcott
- College of Health and Life Sciences, Aston University, Aston Triangle, Birmingham, B4 7ET, UK
| | - Jonathan A G Cox
- College of Health and Life Sciences, Aston University, Aston Triangle, Birmingham, B4 7ET, UK.
| |
Collapse
|
14
|
Baker EJ, Allcott G, Molloy A, Cox JAG. Cystic fibrosis sputum media induces an overall loss of antibiotic susceptibility in Mycobacterium abscessus. NPJ ANTIMICROBIALS AND RESISTANCE 2024; 2:34. [PMID: 39843503 PMCID: PMC11721417 DOI: 10.1038/s44259-024-00054-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 10/03/2024] [Indexed: 01/24/2025]
Abstract
Mycobacterium abscessus complex (MABSC) comprises a group of environmental microorganisms, which are a concerning cause of opportunistic respiratory infections in patients with cystic fibrosis or bronchiectasis. Only 45.6% of MABSC treatments are successful, and therefore this is a need to discover new antimicrobials that can treat these pathogens. However, the transferability of outcomes to the clinic is flawed by an inability to accurately represent the lung environment within the laboratory. Herein, we apply two preestablished formulations of sputum media (ACFS and SCFM1) to MABSC antibiotic susceptibility testing. Using conventional broth microdilution, we have observed strain and antibiotic dependent alterations in antimicrobial sensitivity in each sputum media compared standard laboratory media (7H9), with an overall reduction in susceptibility within the physiologically relevant conditions. We provide a timely contribution to the field of M. abscessus antibiotic discovery by emphasising the need for improved physiological relevance.
Collapse
Affiliation(s)
- Emily J Baker
- College of Health and Life Sciences, Aston University, Aston Triangle, Birmingham, B4 7ET, UK
| | - Gemma Allcott
- College of Health and Life Sciences, Aston University, Aston Triangle, Birmingham, B4 7ET, UK
| | - Antonia Molloy
- College of Health and Life Sciences, Aston University, Aston Triangle, Birmingham, B4 7ET, UK
| | - Jonathan A G Cox
- College of Health and Life Sciences, Aston University, Aston Triangle, Birmingham, B4 7ET, UK.
| |
Collapse
|
15
|
Shukla A, Rodriguez S, Brennan-Krohn T. Activity of antibiotics against Burkholderia cepacia complex in artificial sputum medium. J Antimicrob Chemother 2024; 79:2867-2876. [PMID: 39224940 PMCID: PMC11531831 DOI: 10.1093/jac/dkae299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Burkholderia cepacia complex (Bcc) is a collection of intrinsically drug-resistant Gram-negative bacteria that cause life-threatening disease in people with cystic fibrosis (CF). Standard antimicrobial susceptibility testing methods have poor predictive value for clinical outcomes in Bcc infections, probably due in part to differences between in vitro testing conditions and the environment in which Bcc grow in the lungs of people with CF. OBJECTIVES To compare the activity of commonly used antibiotics under standard in vitro testing conditions with activity in conditions mimicking those found in vivo. METHODS Two Bcc strains were grown alone and with six different antibiotics (minocycline, ceftazidime, meropenem, tobramycin, levofloxacin, trimethoprim-sulfamethoxazole) in two different media: standard cation-adjusted Mueller-Hinton broth and an artificial sputum medium designed to simulate the environment in the lungs of people with CF through addition of components including mucin, free DNA and amino acids. Two different starting conditions were used for time-kill assays: a standard ∼5 × 106 cfu/mL inoculum, and a high-density inoculum in which bacteria were grown for 72 hours before addition of antibiotics. Growth detection was performed by colony enumeration and by detection of resazurin reduction. RESULTS There were major discrepancies between standard susceptibility results and activity in our models. Some antibiotics, including ceftazidime, showed minimal activity in all time-kill assays despite low minimal inhibitory concentrations, while others, notably tobramycin, were more active in high-density growth conditions than in standard time-kill assays. CONCLUSIONS This work underscores the urgent need to develop more clinically relevant susceptibility testing approaches for Bcc.
Collapse
Affiliation(s)
- Anusha Shukla
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Shade Rodriguez
- Pathobiology Graduate Program, Brown University, Providence, RI, USA
| | - Thea Brennan-Krohn
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
16
|
Krzyżek P. What Is a Biofilm? Lessons Learned from Interactions with Immune Cells. Int J Mol Sci 2024; 25:11684. [PMID: 39519236 PMCID: PMC11546875 DOI: 10.3390/ijms252111684] [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: 10/14/2024] [Revised: 10/27/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Biofilms are unique, multicellular life forms that challenge our understanding of the microbial functioning. The last decades of research on biofilms have allowed us to better understand their importance in the context of both health and various pathologies in the human body, although many knowledge gaps hindering their correct comprehension still exist. Biofilms are classically described as mushroom-shaped structures attached to the substrate; however, an increasing body of evidence shows that their morphology in clinical conditions may differ significantly from that classically presented. Although this may result partly from the unique physicochemical conditions within the host, the interaction between microbes and immune cells during development of a biofilm should not be underestimated. The current Opinion confronts the classical view on biofilms with the latest scientific research describing the vitality of interactions with immune cells as a modulator of the biofilm phenotype and behavior in clinical conditions.
Collapse
Affiliation(s)
- Paweł Krzyżek
- Department of Microbiology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland
| |
Collapse
|
17
|
Awad R, Marchand S, Couet W, Nasser M, Buyck JM, Tewes F. Assessment of inhaled cationic antibiotics in an in vitro model of Pseudomonas aeruginosa lung biofilm. Microb Pathog 2024; 197:107020. [PMID: 39419459 DOI: 10.1016/j.micpath.2024.107020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 10/04/2024] [Accepted: 10/14/2024] [Indexed: 10/19/2024]
Abstract
OBJECTIVES This study aimed to evaluate the efficacy of inhaled cationic antibiotics, including tobramycin (TOB) and colistin (CST), using an in vitro alginate bead model that simulates Pseudomonas aeruginosa lung biofilms. METHODS Bioluminescent P. aeruginosa were encapsulated within alginate beads and dispersed in either Mueller-Hinton broth (MHB) or artificial sputum medium (ASM). The impact of bead size and culture medium on TOB and CST efficacy was assessed by monitoring bioluminescence kinetics, followed by colony-forming unit (CFU/mL) measurements. Antibiotic efficacy was quantified using a Hill inhibitory model to analyze variations in CFU/mL in response to TOB and CST concentrations. RESULTS The TOB EC50 was found to be 8-fold higher when P. aeruginosa was encapsulated in larger beads (1200 μm) compared to smaller beads (60 μm). TOB efficacy further decreased twofold when larger beads were dispersed in ASM. In contrast, CST demonstrated superior efficacy, being four times more potent than TOB, with corresponding EC50 values of 20.5 ± 2.8 times MIC and 78.4 ± 10.2 times MIC, respectively. No change in MICs was observed for either antibiotic, even after exposing bacteria to 200 times the MIC. CONCLUSIONS This P. aeruginosa biofilm model highlights how alginate and mucus modulated the efficacy of TOB and CST, and suggested the superior efficacy of CST in eradicating pulmonary biofilms.
Collapse
Affiliation(s)
- Rana Awad
- Université de Poitiers, PHAR2, Inserm U1070, Poitiers, France
| | - Sandrine Marchand
- Université de Poitiers, PHAR2, Inserm U1070, Poitiers, France; CHU de Poitiers, Laboratoire de Toxicologie et de Pharmacocinétique, Poitiers, France
| | - William Couet
- Université de Poitiers, PHAR2, Inserm U1070, Poitiers, France; CHU de Poitiers, Laboratoire de Toxicologie et de Pharmacocinétique, Poitiers, France
| | | | - Julien M Buyck
- Université de Poitiers, PHAR2, Inserm U1070, Poitiers, France
| | - Frédéric Tewes
- Université de Poitiers, PHAR2, Inserm U1070, Poitiers, France.
| |
Collapse
|
18
|
O’Brien C, Spencer S, Jafari N, Huang AJ, Scott AJ, Cheng Z, Leung BM. Modeling Cystic Fibrosis Chronic Infection Using Engineered Mucus-like Hydrogels. ACS Biomater Sci Eng 2024; 10:6558-6568. [PMID: 39297972 PMCID: PMC11483100 DOI: 10.1021/acsbiomaterials.4c01271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/10/2024] [Accepted: 09/12/2024] [Indexed: 10/15/2024]
Abstract
The airway mucus of patients with cystic fibrosis has altered properties, which create a microenvironment primed for chronic infections that are difficult to treat. These complex polymicrobial airway infections and corresponding mammalian-microbe interactions are challenging to model in vitro. Here, we report the development of mucus-like hydrogels with varied compositions and viscoelastic properties reflecting differences between healthy and cystic fibrosis airway mucus. Models of cystic fibrosis and healthy airway microenvironments were created by combining the hydrogels with relevant pathogens, human bronchial epithelial cells, and an antibiotic. Notably, pathogen antibiotic resistance was not solely dependent on the altered properties of the mucus-like hydrogels but was also influenced by culture conditions including microbe species, monomicrobial or polymicrobial culture, and the presence of epithelial cells. Additionally, the cystic fibrosis airway model showed the ability to mimic features characteristic of chronic cystic fibrosis airway infections including sustained polymicrobial growth and increased antibiotic tolerance.
Collapse
Affiliation(s)
- Courtney
L. O’Brien
- School
of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Sarah Spencer
- School
of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Naeimeh Jafari
- Department
of Applied Oral Science, Faculty of Dentistry, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Andy J. Huang
- School
of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Alison J. Scott
- Department
of Process Engineering and Applied Science, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Zhenyu Cheng
- Department
of Microbiology and Immunology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Brendan M. Leung
- School
of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
- Department
of Applied Oral Science, Faculty of Dentistry, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
- Department
of Microbiology and Immunology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
- Department
of Pathology, Faculty of Medicine, Dalhousie
University, Halifax, Nova Scotia B3H 4R2, Canada
| |
Collapse
|
19
|
Sass G, Kethineni S, Stevens DA. Anti-Fungal ( Aspergillus fumigatus) Activity of Pseudomonas aeruginosa in Cystic Fibrosis Synthetic Sputum. Pathogens 2024; 13:875. [PMID: 39452746 PMCID: PMC11510391 DOI: 10.3390/pathogens13100875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 10/26/2024] Open
Abstract
Aspergillus fumigatus (Af) and Pseudomonas aeruginosa (Pa) are pathogens inhabiting the lungs of persons with cystic fibrosis (CF), or immune-compromised patients, causing or aggravating disease. We previously investigated their microbial interaction as well as susceptibility to anti-fungal drugs using RPMI medium (contains undetectable iron concentrations), as is standard for susceptibility testing. Here we investigated microbial interaction in synthetic sputum medium (SSPM), a complex mixture designed to mimic the milieu in CF lungs. SSPM contains Fe2+. Pa laboratory strain PA14 or PA14 siderophore mutant planktonic culture filtrate, prepared in RPMI or SSPM, were compared for inhibition of Af biofilm formation. SSPM enhanced bacterial and fungal growth and the production of the Pa molecules pyoverdine, phenazines, and rhamnolipids. Af was more susceptible to these molecules in SSPM (with the exception of pyoverdine). SSPM interfered with fungal susceptibility to pyoverdine. Studies with the mutant helped to reveal that the reduced anti-fungal activity of pyoverdine in SSPM appears to be compensated by higher production of other anti-fungal molecules, e.g., rhamnolipids, phenazines, and PQS, and higher Af sensitivity to these molecules. In summary, SSPM better defines Pa-Af intermicrobial competition in the milieu of CF lungs.
Collapse
Affiliation(s)
- Gabriele Sass
- California Institute for Medical Research, San Jose, CA 95128, USA; (G.S.); (S.K.)
| | - Satya Kethineni
- California Institute for Medical Research, San Jose, CA 95128, USA; (G.S.); (S.K.)
| | - David A. Stevens
- California Institute for Medical Research, San Jose, CA 95128, USA; (G.S.); (S.K.)
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
20
|
Zheng X, Gomez-Rivas EJ, Lamont SI, Daneshjoo K, Shieh A, Wozniak DJ, Parsek MR. The surface interface and swimming motility influence surface-sensing responses in Pseudomonas aeruginosa. Proc Natl Acad Sci U S A 2024; 121:e2411981121. [PMID: 39284057 PMCID: PMC11441478 DOI: 10.1073/pnas.2411981121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 08/01/2024] [Indexed: 10/02/2024] Open
Abstract
Bacterial biofilms have been implicated in several chronic infections. After initial attachment, a critical first step in biofilm formation is a cell inducing a surface-sensing response. In the Gram-negative opportunistic pathogen Pseudomonas aeruginosa, two second messengers, cyclic diguanylate monophosphate (c-di-GMP) and cyclic adenosine monophosphate (cAMP), are produced by different surface-sensing mechanisms. However, given the disparate cellular behaviors regulated by these second messengers, how newly attached cells coordinate these pathways remains unclear. Some of the uncertainty relates to studies using different strains, experimental systems, and usually focusing on a single second messenger. In this study, we developed a tricolor reporter system to simultaneously gauge c-di-GMP and cAMP levels in single cells. Using PAO1, we show that c-di-GMP and cAMP are selectively activated in two commonly used experimental systems to study surface sensing. By further examining the conditions that differentiate a c-di-GMP or cAMP response, we demonstrate that an agarose-air interface activates cAMP signaling through type IV pili and the Pil-Chp system. However, a liquid-agarose interface favors the activation of c-di-GMP signaling. This response is dependent on flagellar motility and correlated with higher swimming speed. Collectively, this work indicates that c-di-GMP and cAMP signaling responses are dependent on the surface context.
Collapse
Affiliation(s)
- Xuhui Zheng
- Department of Microbiology, University of Washington, Seattle, WA
| | | | - Sabrina I. Lamont
- Departments of Microbial Infection and Immunity, Microbiology, The Ohio State University, Columbus, OH
| | | | - Angeli Shieh
- Department of Microbiology, University of Washington, Seattle, WA
| | - Daniel J. Wozniak
- Departments of Microbial Infection and Immunity, Microbiology, The Ohio State University, Columbus, OH
| | | |
Collapse
|
21
|
Walsh D, Parmenter C, Bakker SE, Lithgow T, Traven A, Harrison F. A new model of endotracheal tube biofilm identifies combinations of matrix-degrading enzymes and antimicrobials able to eradicate biofilms of pathogens that cause ventilator-associated pneumonia. MICROBIOLOGY (READING, ENGLAND) 2024; 170:001480. [PMID: 39088248 PMCID: PMC11541551 DOI: 10.1099/mic.0.001480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 07/04/2024] [Indexed: 08/02/2024]
Abstract
Ventilator-associated pneumonia is defined as pneumonia that develops in a patient who has been on mechanical ventilation for more than 48 hours through an endotracheal tube. It is caused by biofilm formation on the indwelling tube, which introduces pathogenic microbes such as Pseudomonas aeruginosa, Klebsiella pneumoniae and Candida albicans into the patient's lower airways. Currently, there is a lack of accurate in vitro models of ventilator-associated pneumonia development. This greatly limits our understanding of how the in-host environment alters pathogen physiology and the efficacy of ventilator-associated pneumonia prevention or treatment strategies. Here, we showcase a reproducible model that simulates the biofilm formation of these pathogens in a host-mimicking environment and demonstrate that the biofilm matrix produced differs from that observed in standard laboratory growth medium. In our model, pathogens are grown on endotracheal tube segments in the presence of a novel synthetic ventilated airway mucus medium that simulates the in-host environment. Matrix-degrading enzymes and cryo-scanning electron microscopy were employed to characterize the system in terms of biofilm matrix composition and structure, as compared to standard laboratory growth medium. As seen in patients, the biofilms of ventilator-associated pneumonia pathogens in our model either required very high concentrations of antimicrobials for eradication or could not be eradicated. However, combining matrix-degrading enzymes with antimicrobials greatly improved the biofilm eradication of all pathogens. Our in vitro endotracheal tube model informs on fundamental microbiology in the ventilator-associated pneumonia context and has broad applicability as a screening platform for antibiofilm measures including the use of matrix-degrading enzymes as antimicrobial adjuvants.
Collapse
Affiliation(s)
- Dean Walsh
- School of Life Sciences, University of Warwick, Coventry, UK
| | - Chris Parmenter
- Nanoscale and Microscale Research Centre, University of Nottingham, Nottingham, UK
| | | | - Trevor Lithgow
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
- Center To Impact AMR, Monash University, Clayton 3800, Victoria, Australia
| | - Ana Traven
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
- Center To Impact AMR, Monash University, Clayton 3800, Victoria, Australia
| | - Freya Harrison
- School of Life Sciences, University of Warwick, Coventry, UK
| |
Collapse
|
22
|
Di Bonaventura G, Lupetti V, Pompilio A. Impact of Growth Conditions on High-Throughput Identification of Repurposing Drugs for Pseudomonas aeruginosa Cystic Fibrosis Lung Infections. Antibiotics (Basel) 2024; 13:642. [PMID: 39061324 PMCID: PMC11273527 DOI: 10.3390/antibiotics13070642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/26/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Pseudomonas aeruginosa lung infections in cystic fibrosis (CF) patients represent a therapeutic challenge due to antibiotic resistance. Repurposing existing drugs is a promising approach for identifying new antimicrobials. A crucial factor in successful drug repurposing is using assay conditions that mirror the site of infection. Here, the impact of growth conditions on the anti-P. aeruginosa activity of a library of 3386 compounds was evaluated. To this, after 24 h exposure, the survival rate of CF P. aeruginosa RP73 planktonic cells was assessed spectrophotometrically under "CF-like" (artificial CF sputum, pH 6.8, 5% CO2) and enriched (Tryptone Soya Broth, pH 7.2, and aerobiosis) conditions. Among non-antibiotic compounds (n = 3127), 13.4% were active regardless of growth conditions, although only 3.2% had comparable activity; 4% and 6.2% were more active under CF-like or enriched conditions, respectively. Interestingly, 22.1% and 26.6% were active exclusively under CF-like and enriched conditions, respectively. Notably, 7 and 12 hits caused 100% killing under CF-like and enriched conditions, respectively. Among antibiotics (n = 234), 42.3% were active under both conditions, although only 18.4% showed comparable activity; 9.4% and 14.5% were more active under CF-like and enriched conditions, respectively. Interestingly, 23% and 16.6% were active exclusively under CF-like and enriched conditions, respectively. Sulphonamides showed higher activity under CF-like conditions, whereas tetracyclines, fluoroquinolones, and macrolides were more effective under enriched settings. Our findings indicated that growth conditions significantly affect the anti-P. aeruginosa activity of antibiotics and non-antibiotic drugs. Consequently, repurposing studies and susceptibility tests should be performed under physicochemical conditions that the pathogen tackles at the site of infection.
Collapse
Affiliation(s)
- Giovanni Di Bonaventura
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (V.L.); (A.P.)
- Center for Advanced Studies and Technology, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Veronica Lupetti
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (V.L.); (A.P.)
- Center for Advanced Studies and Technology, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Arianna Pompilio
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (V.L.); (A.P.)
- Center for Advanced Studies and Technology, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| |
Collapse
|
23
|
Bianchi M, Esin S, Kaya E, Batoni G, Maisetta G. Anti-Persisters Activity of Lacticaseibacillus rhamnosus Culture Filtrates against Pseudomonas aeruginosa in Artificial Sputum Medium. Int J Mol Sci 2024; 25:7113. [PMID: 39000222 PMCID: PMC11241021 DOI: 10.3390/ijms25137113] [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: 05/01/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Persisters are antibiotic-tolerant bacteria, playing a role in the recalcitrance and relapse of many bacterial infections, including P. aeruginosa pulmonary infections in Cystic Fibrosis (CF) patients. Among novel antimicrobial strategies, the use of probiotics and their products is emerging as a particularly promising approach. The aim of this study was to evaluate the anti-persisters activity of culture filtrate supernatants of Lacticaseibacillus rhamnosus (LRM-CFS) against P. aeruginosa in artificial sputum medium (ASM), which resembles the CF lung environment. Planktonic persisters of two clinical strains of P. aeruginosa (PaCF1 and PaCF4) were obtained following two different procedures: (i) exposing stationary-phase cultures to cyanide m-chlorophenylhydrazone (CCCP) in LB medium; (ii) incubating stationary-phase cultures with high doses of tobramycin (128-fold MIC) in ASM. In addition, persisters from biofilm were obtained by exposing 48 h old biofilm of P. aeruginosa to 128 x MIC of ciprofloxacin. LRM-CFS at dilutions of 1:6 and 1:4 resulted in being bactericidal in ASM against both PaCF1 and PaCF4 persisters obtained after CCCP or tobramycin treatment. Moreover, LRM-CFS at dilution 1:4 caused a reduction of antibiotic-tolerant bacteria in the biofilm of both P. aeruginosa strains. Overall, LRM-CFS represents a promising adjuvant therapeutic strategy against P. aeruginosa recalcitrant infections in CF patients.
Collapse
Affiliation(s)
- Marta Bianchi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via S. Zeno 37, 56123 Pisa, Italy
| | - Semih Esin
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via S. Zeno 37, 56123 Pisa, Italy
| | - Esingül Kaya
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via S. Zeno 37, 56123 Pisa, Italy
| | - Giovanna Batoni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via S. Zeno 37, 56123 Pisa, Italy
| | - Giuseppantonio Maisetta
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via S. Zeno 37, 56123 Pisa, Italy
| |
Collapse
|
24
|
Greenwald MA, Meinig SL, Plott LM, Roca C, Higgs MG, Vitko NP, Markovetz MR, Rouillard KR, Carpenter J, Kesimer M, Hill DB, Schisler JC, Wolfgang MC. Mucus polymer concentration and in vivo adaptation converge to define the antibiotic response of Pseudomonas aeruginosa during chronic lung infection. mBio 2024; 15:e0345123. [PMID: 38651896 PMCID: PMC11237767 DOI: 10.1128/mbio.03451-23] [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: 12/18/2023] [Accepted: 03/26/2024] [Indexed: 04/25/2024] Open
Abstract
The airway milieu of individuals with muco-obstructive airway diseases (MADs) is defined by the accumulation of dehydrated mucus due to hyperabsorption of airway surface liquid and defective mucociliary clearance. Pathological mucus becomes progressively more viscous with age and disease severity due to the concentration and overproduction of mucin and accumulation of host-derived extracellular DNA (eDNA). Respiratory mucus of MADs provides a niche for recurrent and persistent colonization by respiratory pathogens, including Pseudomonas aeruginosa, which is responsible for the majority of morbidity and mortality in MADs. Despite high concentration inhaled antibiotic therapies and the absence of antibiotic resistance, antipseudomonal treatment failure in MADs remains a significant clinical challenge. Understanding the drivers of antibiotic tolerance is essential for developing more effective treatments that eradicate persistent infections. The complex and dynamic environment of diseased airways makes it difficult to model antibiotic efficacy in vitro. We aimed to understand how mucin and eDNA concentrations, the two dominant polymers in respiratory mucus, alter the antibiotic tolerance of P. aeruginosa. Our results demonstrate that polymer concentration and molecular weight affect P. aeruginosa survival post antibiotic challenge. Polymer-driven antibiotic tolerance was not explicitly associated with reduced antibiotic diffusion. Lastly, we established a robust and standardized in vitro model for recapitulating the ex vivo antibiotic tolerance of P. aeruginosa observed in expectorated sputum across age, underlying MAD etiology, and disease severity, which revealed the inherent variability in intrinsic antibiotic tolerance of host-evolved P. aeruginosa populations. IMPORTANCE Antibiotic treatment failure in Pseudomonas aeruginosa chronic lung infections is associated with increased morbidity and mortality, illustrating the clinical challenge of bacterial infection control. Understanding the underlying infection environment, as well as the host and bacterial factors driving antibiotic tolerance and the ability to accurately recapitulate these factors in vitro, is crucial for improving antibiotic treatment outcomes. Here, we demonstrate that increasing concentration and molecular weight of mucin and host eDNA drive increased antibiotic tolerance to tobramycin. Through systematic testing and modeling, we identified a biologically relevant in vitro condition that recapitulates antibiotic tolerance observed in ex vivo treated sputum. Ultimately, this study revealed a dominant effect of in vivo evolved bacterial populations in defining inter-subject ex vivo antibiotic tolerance and establishes a robust and translatable in vitro model for therapeutic development.
Collapse
Affiliation(s)
- Matthew A Greenwald
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, USA
- Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Suzanne L Meinig
- Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Lucas M Plott
- Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Cristian Roca
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, USA
- Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Matthew G Higgs
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, USA
- Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Nicholas P Vitko
- Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Matthew R Markovetz
- Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Kaitlyn R Rouillard
- Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Jerome Carpenter
- Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Mehmet Kesimer
- Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, USA
| | - David B Hill
- Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, USA
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Jonathan C Schisler
- Department of Pharmacology, The University of North Carolina, Chapel Hill, North Carolina, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Matthew C Wolfgang
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, USA
- Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
25
|
Lewis JM, Jebeli L, Coulon PML, Lay CE, Scott NE. Glycoproteomic and proteomic analysis of Burkholderia cenocepacia reveals glycosylation events within FliF and MotB are dispensable for motility. Microbiol Spectr 2024; 12:e0034624. [PMID: 38709084 PMCID: PMC11237607 DOI: 10.1128/spectrum.00346-24] [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: 02/10/2024] [Accepted: 04/16/2024] [Indexed: 05/07/2024] Open
Abstract
Across the Burkholderia genus O-linked protein glycosylation is highly conserved. While the inhibition of glycosylation has been shown to be detrimental for virulence in Burkholderia cepacia complex species, such as Burkholderia cenocepacia, little is known about how specific glycosylation sites impact protein functionality. Within this study, we sought to improve our understanding of the breadth, dynamics, and requirement for glycosylation across the B. cenocepacia O-glycoproteome. Assessing the B. cenocepacia glycoproteome across different culture media using complementary glycoproteomic approaches, we increase the known glycoproteome to 141 glycoproteins. Leveraging this repertoire of glycoproteins, we quantitively assessed the glycoproteome of B. cenocepacia using Data-Independent Acquisition (DIA) revealing the B. cenocepacia glycoproteome is largely stable across conditions with most glycoproteins constitutively expressed. Examination of how the absence of glycosylation impacts the glycoproteome reveals that the protein abundance of only five glycoproteins (BCAL1086, BCAL2974, BCAL0525, BCAM0505, and BCAL0127) are altered by the loss of glycosylation. Assessing ΔfliF (ΔBCAL0525), ΔmotB (ΔBCAL0127), and ΔBCAM0505 strains, we demonstrate the loss of FliF, and to a lesser extent MotB, mirror the proteomic effects observed in the absence of glycosylation in ΔpglL. While both MotB and FliF are essential for motility, we find loss of glycosylation sites in MotB or FliF does not impact motility supporting these sites are dispensable for function. Combined this work broadens our understanding of the B. cenocepacia glycoproteome supporting that the loss of glycoproteins in the absence of glycosylation is not an indicator of the requirement for glycosylation for protein function. IMPORTANCE Burkholderia cenocepacia is an opportunistic pathogen of concern within the Cystic Fibrosis community. Despite a greater appreciation of the unique physiology of B. cenocepacia gained over the last 20 years a complete understanding of the proteome and especially the O-glycoproteome, is lacking. In this study, we utilize systems biology approaches to expand the known B. cenocepacia glycoproteome as well as track the dynamics of glycoproteins across growth phases, culturing media and in response to the loss of glycosylation. We show that the glycoproteome of B. cenocepacia is largely stable across conditions and that the loss of glycosylation only impacts five glycoproteins including the motility associated proteins FliF and MotB. Examination of MotB and FliF shows, while these proteins are essential for motility, glycosylation is dispensable. Combined this work supports that B. cenocepacia glycosylation can be dispensable for protein function and may influence protein properties beyond stability.
Collapse
Affiliation(s)
- Jessica M Lewis
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Leila Jebeli
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Pauline M L Coulon
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Catrina E Lay
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Nichollas E Scott
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| |
Collapse
|
26
|
Sousa AM, Pereira MO. Challenges with drug efficacy prediction of in vitro models of biofilms infecting cystic fibrosis airway. Expert Opin Drug Discov 2024; 19:635-638. [PMID: 38712907 DOI: 10.1080/17460441.2024.2350567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 04/29/2024] [Indexed: 05/08/2024]
Affiliation(s)
- Ana Margarida Sousa
- CEB - Centre of Biological Engineering, University of Minho, Braga, Portugal
- LABBELS - Associate Laboratory, Braga, Portugal
| | - Maria Olívia Pereira
- CEB - Centre of Biological Engineering, University of Minho, Braga, Portugal
- LABBELS - Associate Laboratory, Braga, Portugal
| |
Collapse
|
27
|
Pompilio A, Kaya E, Lupetti V, Catelli E, Bianchi M, Maisetta G, Esin S, Di Bonaventura G, Batoni G. Cell-free supernatants from Lactobacillus strains exert antibacterial, antibiofilm, and antivirulence activity against Pseudomonas aeruginosa from cystic fibrosis patients. Microbes Infect 2024; 26:105301. [PMID: 38237656 DOI: 10.1016/j.micinf.2024.105301] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 01/10/2024] [Accepted: 01/15/2024] [Indexed: 01/26/2024]
Abstract
Chronic lung infections caused by Pseudomonas aeruginosa play a significant role in the mortality and morbidity of cystic fibrosis (CF) patients. The widespread bacterial resistance to conventional antimicrobials demands identifying new strategies to complement or replace current antibiotic therapies. In this study, we evaluated the antibacterial, antibiofilm, and antivirulence properties of cell-free supernatants (CFS) from several Lactobacillus probiotic strains against P. aeruginosa isolated from the sputum of CF patients. A strong and fast antibacterial activity of CFS from different strains of lactobacilli was observed at acidic pH towards P. aeruginosa, both in planktonic and biofilm mode of growth, in conditions mimicking CF lung. Interestingly, although when adjusted at pH 6.0, CFS lost most of their antibacterial potential, they retained some antivirulence activity towards P. aeruginosa, largely dependent on the dose, exposure time, and the Lactobacillus-P. aeruginosa strain combination. In vivo testing in the invertebrate Galleria mellonella model disclosed the lack of toxicity of acidic CFS and their ability to prevent P. aeruginosa infection. For the first time, the results revealed lactobacilli postbiotic activities in the context of the pulmonary environment, pointing to innovative postbiotics' uses in anti-infective therapy.
Collapse
Affiliation(s)
- Arianna Pompilio
- Department of Medical, Oral, and Biotechnological Sciences, G. d'Annunzio University of Chieti-Pescara, Via dei Vestini, 31, 66100 Chieti, Italy; Center for Advanced Studies and Technology, G. d'Annunzio University of Chieti-Pescara, Via L. Polacchi 11, 66100 Chieti, Italy
| | - Esingül Kaya
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via S. Zeno 37, 56123 Pisa, Italy
| | - Veronica Lupetti
- Department of Medical, Oral, and Biotechnological Sciences, G. d'Annunzio University of Chieti-Pescara, Via dei Vestini, 31, 66100 Chieti, Italy; Center for Advanced Studies and Technology, G. d'Annunzio University of Chieti-Pescara, Via L. Polacchi 11, 66100 Chieti, Italy
| | - Elisa Catelli
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via S. Zeno 37, 56123 Pisa, Italy
| | - Marta Bianchi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via S. Zeno 37, 56123 Pisa, Italy
| | - Giuseppantonio Maisetta
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via S. Zeno 37, 56123 Pisa, Italy
| | - Semih Esin
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via S. Zeno 37, 56123 Pisa, Italy
| | - Giovanni Di Bonaventura
- Department of Medical, Oral, and Biotechnological Sciences, G. d'Annunzio University of Chieti-Pescara, Via dei Vestini, 31, 66100 Chieti, Italy; Center for Advanced Studies and Technology, G. d'Annunzio University of Chieti-Pescara, Via L. Polacchi 11, 66100 Chieti, Italy.
| | - Giovanna Batoni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via S. Zeno 37, 56123 Pisa, Italy.
| |
Collapse
|
28
|
Seixas AMM, Silva C, Marques JMM, Mateus P, Rodríguez-Ortega MJ, Feliciano JR, Leitão JH, Sousa SA. Surface-Exposed Protein Moieties of Burkholderia cenocepacia J2315 in Microaerophilic and Aerobic Conditions. Vaccines (Basel) 2024; 12:398. [PMID: 38675780 PMCID: PMC11054960 DOI: 10.3390/vaccines12040398] [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: 02/23/2024] [Revised: 03/18/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024] Open
Abstract
Burkholderia cepacia complex infections remain life-threatening to cystic fibrosis patients, and due to the limited eradication efficiency of current treatments, novel antimicrobial therapies are urgently needed. Surface proteins are among the best targets to develop new therapeutic strategies since they are exposed to the host's immune system. A surface-shaving approach was performed using Burkholderia cenocepacia J2315 to quantitatively compare the relative abundance of surface-exposed proteins (SEPs) expressed by the bacterium when grown under aerobic and microaerophilic conditions. After trypsin incubation of live bacteria and identification of resulting peptides by liquid chromatography coupled with mass spectrometry, a total of 461 proteins with ≥2 unique peptides were identified. Bioinformatics analyses revealed a total of 53 proteins predicted as localized at the outer membrane (OM) or extracellularly (E). Additionally, 37 proteins were predicted as moonlight proteins with OM or E secondary localization. B-cell linear epitope bioinformatics analysis of the proteins predicted to be OM and E-localized revealed 71 SEP moieties with predicted immunogenic epitopes. The protegenicity higher scores of proteins BCAM2761, BCAS0104, BCAL0151, and BCAL0849 point out these proteins as the best antigens for vaccine development. Additionally, 10 of the OM proteins also presented a high probability of playing important roles in adhesion to host cells, making them potential targets for passive immunotherapeutic approaches. The immunoreactivity of three of the OM proteins identified was experimentally demonstrated using serum samples from cystic fibrosis patients, validating our strategy for identifying immunoreactive moieties from surface-exposed proteins of potential interest for future immunotherapies development.
Collapse
Affiliation(s)
- António M. M. Seixas
- Department of Bioengineering, IBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; (A.M.M.S.); (J.M.M.M.); (P.M.); (J.R.F.)
- Associate Laboratory, i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Carolina Silva
- Department of Bioengineering, IBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; (A.M.M.S.); (J.M.M.M.); (P.M.); (J.R.F.)
- Associate Laboratory, i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Joana M. M. Marques
- Department of Bioengineering, IBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; (A.M.M.S.); (J.M.M.M.); (P.M.); (J.R.F.)
- Associate Laboratory, i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Patrícia Mateus
- Department of Bioengineering, IBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; (A.M.M.S.); (J.M.M.M.); (P.M.); (J.R.F.)
- Associate Laboratory, i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Manuel J. Rodríguez-Ortega
- Departamento de Bioquímica y Biología Molecular, Universidad de Córdoba, Campus de Excelencia Internacional CeiA3, 14071 Córdoba, Spain;
| | - Joana R. Feliciano
- Department of Bioengineering, IBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; (A.M.M.S.); (J.M.M.M.); (P.M.); (J.R.F.)
- Associate Laboratory, i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Jorge H. Leitão
- Department of Bioengineering, IBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; (A.M.M.S.); (J.M.M.M.); (P.M.); (J.R.F.)
- Associate Laboratory, i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Sílvia A. Sousa
- Department of Bioengineering, IBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; (A.M.M.S.); (J.M.M.M.); (P.M.); (J.R.F.)
- Associate Laboratory, i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| |
Collapse
|
29
|
Palekyte A, Morkowska A, Billington O, Morris-Jones S, Millard J, Marakalala MJ, Owolabi O, Sambou B, Zumla A, Sutherland JS, McHugh TD, Honeyborne I. Acetic Acid Enables Molecular Enumeration of Mycobacterium tuberculosis from Sputum and Eliminates the Need for a Biosafety Level 3 Laboratory. Clin Chem 2024; 70:642-652. [PMID: 38479728 DOI: 10.1093/clinchem/hvae013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 12/08/2023] [Indexed: 04/04/2024]
Abstract
BACKGROUND Improved monitoring of Mycobacterium tuberculosis response to treatment is urgently required. We previously developed the molecular bacterial load assay (MBLA), but it is challenging to integrate into the clinical diagnostic laboratory due to a labor-intensive protocol required at biosafety level 3 (BSL-3). A modified assay was needed. METHODS The rapid enumeration and diagnostic for tuberculosis (READ-TB) assay was developed. Acetic acid was tested and compared to 4 M guanidine thiocyanate to be simultaneously bactericidal and preserve mycobacterial RNA. The extraction was based on silica column technology and incorporated low-cost reagents: 3 M sodium acetate and ethanol for the RNA extraction to replace phenol-chloroform. READ-TB was fully validated and compared directly to the MBLA using sputa collected from individuals with tuberculosis. RESULTS Acetic acid was bactericidal to M. tuberculosis with no significant loss in 16S rRNA or an unprotected mRNA fragment when sputum was stored in acetic acid at 25°C for 2 weeks or -20°C for 1 year. This novel use of acetic acid allows processing of sputum for READ-TB at biosafety level 2 (BSL-2) on sample receipt. READ-TB is semiautomated and rapid. READ-TB correlated with the MBLA when 85 human sputum samples were directly compared (R2 = 0.74). CONCLUSIONS READ-TB is an improved version of the MBLA and is available to be adopted by clinical microbiology laboratories as a tool for tuberculosis treatment monitoring. READ-TB will have a particular impact in low- and middle-income countries (LMICs) for laboratories with no BSL-3 laboratory and for clinical trials testing new combinations of anti-tuberculosis drugs.
Collapse
Affiliation(s)
- Ana Palekyte
- Centre for Clinical Microbiology, Division of Infection and Immunity, University College London, London, United Kingdom
| | - Anna Morkowska
- Imperial College London, Infection and Immunity, Department of Microbiology, Charing Cross Hospital, London, United Kingdom
| | | | - Stephen Morris-Jones
- Department of Clinical Microbiology, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - James Millard
- Liverpool Centre for Global Health Research, University of Liverpool, Liverpool, United Kingdom
- Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
- Africa Health Research Institute, Durban, South Africa
| | | | - Olumuyiwa Owolabi
- Vaccines and Immunity, MRC Unit The Gambia at The London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | - Basil Sambou
- Vaccines and Immunity, MRC Unit The Gambia at The London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | - Alimuddin Zumla
- Centre for Clinical Microbiology, Division of Infection and Immunity, University College London, London, United Kingdom
- National Institute of Health and Research Biomedical Research Centre, UCL Hospitals National Health Service Foundation Trust, London, United Kingdom
| | - Jayne S Sutherland
- Vaccines and Immunity, MRC Unit The Gambia at The London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | - Timothy D McHugh
- Centre for Clinical Microbiology, Division of Infection and Immunity, University College London, London, United Kingdom
| | - Isobella Honeyborne
- Centre for Clinical Microbiology, Division of Infection and Immunity, University College London, London, United Kingdom
| |
Collapse
|
30
|
Walsh D, Bevan J, Harrison F. How Does Airway Surface Liquid Composition Vary in Different Pulmonary Diseases, and How Can We Use This Knowledge to Model Microbial Infections? Microorganisms 2024; 12:732. [PMID: 38674677 PMCID: PMC11052052 DOI: 10.3390/microorganisms12040732] [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: 03/11/2024] [Revised: 03/26/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
Growth environment greatly alters many facets of pathogen physiology, including pathogenesis and antimicrobial tolerance. The importance of host-mimicking environments for attaining an accurate picture of pathogen behaviour is widely recognised. Whilst this recognition has translated into the extensive development of artificial cystic fibrosis (CF) sputum medium, attempts to mimic the growth environment in other respiratory disease states have been completely neglected. The composition of the airway surface liquid (ASL) in different pulmonary diseases is far less well characterised than CF sputum, making it very difficult for researchers to model these infection environments. In this review, we discuss the components of human ASL, how different lung pathologies affect ASL composition, and how different pathogens interact with these components. This will provide researchers interested in mimicking different respiratory environments with the information necessary to design a host-mimicking medium, allowing for better understanding of how to treat pathogens causing infection in these environments.
Collapse
Affiliation(s)
- Dean Walsh
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK (F.H.)
| | | | | |
Collapse
|
31
|
Gnaien M, Maufrais C, Rebai Y, Kallel A, Ma L, Hamouda S, Khalsi F, Meftah K, Smaoui H, Khemiri M, Hadj Fredj S, Bachellier-Bassi S, Najjar I, Messaoud T, Boussetta K, Kallel K, Mardassi H, d’Enfert C, Bougnoux ME, Znaidi S. A gain-of-function mutation in zinc cluster transcription factor Rob1 drives Candida albicans adaptive growth in the cystic fibrosis lung environment. PLoS Pathog 2024; 20:e1012154. [PMID: 38603707 PMCID: PMC11037546 DOI: 10.1371/journal.ppat.1012154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 04/23/2024] [Accepted: 03/26/2024] [Indexed: 04/13/2024] Open
Abstract
Candida albicans chronically colonizes the respiratory tract of patients with Cystic Fibrosis (CF). It competes with CF-associated pathogens (e.g. Pseudomonas aeruginosa) and contributes to disease severity. We hypothesize that C. albicans undergoes specific adaptation mechanisms that explain its persistence in the CF lung environment. To identify the underlying genetic and phenotypic determinants, we serially recovered 146 C. albicans clinical isolates over a period of 30 months from the sputum of 25 antifungal-naive CF patients. Multilocus sequence typing analyses revealed that most patients were individually colonized with genetically close strains, facilitating comparative analyses between serial isolates. We strikingly observed differential ability to filament and form monospecies and dual-species biofilms with P. aeruginosa among 18 serial isolates sharing the same diploid sequence type, recovered within one year from a pediatric patient. Whole genome sequencing revealed that their genomes were highly heterozygous and similar to each other, displaying a highly clonal subpopulation structure. Data mining identified 34 non-synonymous heterozygous SNPs in 19 open reading frames differentiating the hyperfilamentous and strong biofilm-former strains from the remaining isolates. Among these, we detected a glycine-to-glutamate substitution at position 299 (G299E) in the deduced amino acid sequence of the zinc cluster transcription factor ROB1 (ROB1G299E), encoding a major regulator of filamentous growth and biofilm formation. Introduction of the G299E heterozygous mutation in a co-isolated weak biofilm-former CF strain was sufficient to confer hyperfilamentous growth, increased expression of hyphal-specific genes, increased monospecies biofilm formation and increased survival in dual-species biofilms formed with P. aeruginosa, indicating that ROB1G299E is a gain-of-function mutation. Disruption of ROB1 in a hyperfilamentous isolate carrying the ROB1G299E allele abolished hyperfilamentation and biofilm formation. Our study links a single heterozygous mutation to the ability of C. albicans to better survive during the interaction with other CF-associated microbes and illuminates how adaptive traits emerge in microbial pathogens to persistently colonize and/or infect the CF-patient airways.
Collapse
Affiliation(s)
- Mayssa Gnaien
- Institut Pasteur de Tunis, University of Tunis El Manar, Laboratoire de Microbiologie Moléculaire, Vaccinologie et Développement Biotechnologique (LR16IPT01), Tunis, Tunisia
| | - Corinne Maufrais
- Institut Pasteur, Université Paris Cité, INRAE USC2019A, Département Mycologie, Unité Biologie et Pathogénicité Fongiques, Paris, France
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, F-75015 Paris, France
| | - Yasmine Rebai
- Institut Pasteur de Tunis, University of Tunis El Manar, Laboratoire de Microbiologie Moléculaire, Vaccinologie et Développement Biotechnologique (LR16IPT01), Tunis, Tunisia
| | - Aicha Kallel
- Institut Pasteur de Tunis, University of Tunis El Manar, Laboratoire de Microbiologie Moléculaire, Vaccinologie et Développement Biotechnologique (LR16IPT01), Tunis, Tunisia
- Hôpital La Rabta, Laboratoire de Parasitologie et de Mycologie, UR17SP03, Tunis, Tunisia
| | - Laurence Ma
- Institut Pasteur, Université Paris Cité, Biomics core facility, Centre de Ressources et Recherche Technologique (C2RT), Paris, France
| | - Samia Hamouda
- Hôpital d’Enfants Béchir Hamza de Tunis, Tunis, Tunisia
| | - Fatma Khalsi
- Hôpital d’Enfants Béchir Hamza de Tunis, Tunis, Tunisia
| | | | - Hanen Smaoui
- Hôpital d’Enfants Béchir Hamza de Tunis, Tunis, Tunisia
| | - Monia Khemiri
- Hôpital d’Enfants Béchir Hamza de Tunis, Tunis, Tunisia
| | | | - Sophie Bachellier-Bassi
- Institut Pasteur, Université Paris Cité, INRAE USC2019A, Département Mycologie, Unité Biologie et Pathogénicité Fongiques, Paris, France
| | - Imène Najjar
- Institut Pasteur, Université Paris Cité, Biomics core facility, Centre de Ressources et Recherche Technologique (C2RT), Paris, France
| | | | | | - Kalthoum Kallel
- Hôpital La Rabta, Laboratoire de Parasitologie et de Mycologie, UR17SP03, Tunis, Tunisia
| | - Helmi Mardassi
- Institut Pasteur de Tunis, University of Tunis El Manar, Laboratoire de Microbiologie Moléculaire, Vaccinologie et Développement Biotechnologique (LR16IPT01), Tunis, Tunisia
| | - Christophe d’Enfert
- Institut Pasteur, Université Paris Cité, INRAE USC2019A, Département Mycologie, Unité Biologie et Pathogénicité Fongiques, Paris, France
| | - Marie-Elisabeth Bougnoux
- Institut Pasteur, Université Paris Cité, INRAE USC2019A, Département Mycologie, Unité Biologie et Pathogénicité Fongiques, Paris, France
| | - Sadri Znaidi
- Institut Pasteur de Tunis, University of Tunis El Manar, Laboratoire de Microbiologie Moléculaire, Vaccinologie et Développement Biotechnologique (LR16IPT01), Tunis, Tunisia
- Institut Pasteur, Université Paris Cité, INRAE USC2019A, Département Mycologie, Unité Biologie et Pathogénicité Fongiques, Paris, France
| |
Collapse
|
32
|
Hibbert T, Krpetic Z, Latimer J, Leighton H, McHugh R, Pottenger S, Wragg C, James CE. Antimicrobials: An update on new strategies to diversify treatment for bacterial infections. Adv Microb Physiol 2024; 84:135-241. [PMID: 38821632 DOI: 10.1016/bs.ampbs.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2024]
Abstract
Ninety-five years after Fleming's discovery of penicillin, a bounty of antibiotic compounds have been discovered, modified, or synthesised. Diversification of target sites, improved stability and altered activity spectra have enabled continued antibiotic efficacy, but overwhelming reliance and misuse has fuelled the global spread of antimicrobial resistance (AMR). An estimated 1.27 million deaths were attributable to antibiotic resistant bacteria in 2019, representing a major threat to modern medicine. Although antibiotics remain at the heart of strategies for treatment and control of bacterial diseases, the threat of AMR has reached catastrophic proportions urgently calling for fresh innovation. The last decade has been peppered with ground-breaking developments in genome sequencing, high throughput screening technologies and machine learning. These advances have opened new doors for bioprospecting for novel antimicrobials. They have also enabled more thorough exploration of complex and polymicrobial infections and interactions with the healthy microbiome. Using models of infection that more closely resemble the infection state in vivo, we are now beginning to measure the impacts of antimicrobial therapy on host/microbiota/pathogen interactions. However new approaches are needed for developing and standardising appropriate methods to measure efficacy of novel antimicrobial combinations in these contexts. A battery of promising new antimicrobials is now in various stages of development including co-administered inhibitors, phages, nanoparticles, immunotherapy, anti-biofilm and anti-virulence agents. These novel therapeutics need multidisciplinary collaboration and new ways of thinking to bring them into large scale clinical use.
Collapse
Affiliation(s)
- Tegan Hibbert
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, UK
| | - Zeljka Krpetic
- School of Science, Engineering, and Environment, University of Salford, Salford, UK
| | - Joe Latimer
- School of Science, Engineering, and Environment, University of Salford, Salford, UK
| | - Hollie Leighton
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, UK
| | - Rebecca McHugh
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Sian Pottenger
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, UK
| | - Charlotte Wragg
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, UK
| | - Chloë E James
- School of Science, Engineering, and Environment, University of Salford, Salford, UK.
| |
Collapse
|
33
|
Tabassum N, Khan F, Jeong GJ, Oh D, Kim YM. Antibiofilm and antivirulence activities of laminarin-gold nanoparticles in standard and host-mimicking media. Appl Microbiol Biotechnol 2024; 108:203. [PMID: 38349556 PMCID: PMC10864539 DOI: 10.1007/s00253-024-13050-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 01/02/2024] [Accepted: 02/05/2024] [Indexed: 02/15/2024]
Abstract
The rapidly rising antimicrobial resistance (AMR) in pathogenic bacteria has become one of the most serious public health challenges, with a high death rate. Most pathogenic bacteria have been recognized as a source of AMR and a primary barrier to antimicrobial treatment failure due to the development of biofilms and the production of virulence factors. In this work, nanotechnology was employed as a substitute method to control the formation of biofilms and attenuate virulence features in Pseudomonas aeruginosa and Staphylococcus aureus. We synthesized biocompatible gold nanoparticles from marine-derived laminarin as potential biofilm and virulence treatments. Laminarin-gold nanoparticles (Lam-AuNPs) have been identified as spherical, 49.84 ± 7.32 nm in size and - 26.49 ± 1.29 mV zeta potential. The MIC value of Lam-AuNPs against several drug-resistant microbial pathogens varied from 2 to 1024 μg/mL in both standard and host-mimicking media. Sub-MIC values of Lam-AuNPs were reported to effectively reduce the production of P. aeruginosa and S. aureus biofilms in both standard and host-mimicking growth media. Furthermore, the sub-MIC of Lam-AuNPs strongly reduced hemolysis, pyocyanin, pyoverdine, protease, and several forms of flagellar and pili-mediated motility in P. aeruginosa. Lam-AuNPs also inhibited S. aureus hemolysis and the production of amyloid fibrils. The Lam-AuNPs strongly dispersed the preformed mature biofilm of these pathogens in a dose-dependent manner. The Lam-AuNPs would be considered an alternative antibiofilm and antivirulence agent to control P. aeruginosa and S. aureus infections. KEY POINTS: • Lam-AuNPs were biosynthesized to control biofilm and virulence. • Lam-AuNPs show effective biofilm inhibition in standard and host-mimicking media. • Lam-AuNPs suppress various virulence factors of P. aeruginosa and S. aureus.
Collapse
Affiliation(s)
- Nazia Tabassum
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Fazlurrahman Khan
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea.
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea.
- Institute of Fisheries Sciences, Pukyong National University, Busan, 48513, Republic of Korea.
| | - Geum-Jae Jeong
- Department of Food Science and Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Dokyung Oh
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea
- Department of Food Science and Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Young-Mog Kim
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea.
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea.
- Department of Food Science and Technology, Pukyong National University, Busan, 48513, Republic of Korea.
| |
Collapse
|
34
|
Ruhluel D, Fisher L, Barton TE, Leighton H, Kumar S, Amores Morillo P, O’Brien S, Fothergill JL, Neill DR. Secondary messenger signalling influences Pseudomonas aeruginosa adaptation to sinus and lung environments. THE ISME JOURNAL 2024; 18:wrae065. [PMID: 38647527 PMCID: PMC11102083 DOI: 10.1093/ismejo/wrae065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 03/08/2024] [Accepted: 04/18/2024] [Indexed: 04/25/2024]
Abstract
Pseudomonas aeruginosa is a cause of chronic respiratory tract infections in people with cystic fibrosis (CF), non-CF bronchiectasis, and chronic obstructive pulmonary disease. Prolonged infection allows the accumulation of mutations and horizontal gene transfer, increasing the likelihood of adaptive phenotypic traits. Adaptation is proposed to arise first in bacterial populations colonizing upper airway environments. Here, we model this process using an experimental evolution approach. Pseudomonas aeruginosa PAO1, which is not airway adapted, was serially passaged, separately, in media chemically reflective of upper or lower airway environments. To explore whether the CF environment selects for unique traits, we separately passaged PAO1 in airway-mimicking media with or without CF-specific factors. Our findings demonstrated that all airway environments-sinus and lungs, under CF and non-CF conditions-selected for loss of twitching motility, increased resistance to multiple antibiotic classes, and a hyper-biofilm phenotype. These traits conferred increased airway colonization potential in an in vivo model. CF-like conditions exerted stronger selective pressures, leading to emergence of more pronounced phenotypes. Loss of twitching was associated with mutations in type IV pili genes. Type IV pili mediate surface attachment, twitching, and induction of cAMP signalling. We additionally identified multiple evolutionary routes to increased biofilm formation involving regulation of cyclic-di-GMP signalling. These included the loss of function mutations in bifA and dipA phosphodiesterase genes and activating mutations in the siaA phosphatase. These data highlight that airway environments select for traits associated with sessile lifestyles and suggest upper airway niches support emergence of phenotypes that promote establishment of lung infection.
Collapse
Affiliation(s)
- Dilem Ruhluel
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Ronald Ross Building, 8 West Derby Street, Liverpool, United Kingdom
| | - Lewis Fisher
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Thomas E Barton
- Division of Molecular Microbiology, University of Dundee, Dow Street, Dundee, United Kingdom
| | - Hollie Leighton
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Ronald Ross Building, 8 West Derby Street, Liverpool, United Kingdom
| | - Sumit Kumar
- Division of Molecular Microbiology, University of Dundee, Dow Street, Dundee, United Kingdom
| | - Paula Amores Morillo
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Ronald Ross Building, 8 West Derby Street, Liverpool, United Kingdom
| | - Siobhan O’Brien
- Department of Microbiology, Moyne Institute of Preventive Medicine, Trinity College, Dublin, 2, Ireland
| | - Joanne L Fothergill
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Ronald Ross Building, 8 West Derby Street, Liverpool, United Kingdom
| | - Daniel R Neill
- Division of Molecular Microbiology, University of Dundee, Dow Street, Dundee, United Kingdom
| |
Collapse
|
35
|
Bottery MJ, Johansen HK, Pitchford JW, Friman VP. Co-occurring microflora and mucin drive Pseudomonas aeruginosa diversification and pathoadaptation. ISME COMMUNICATIONS 2024; 4:ycae043. [PMID: 38707844 PMCID: PMC11067959 DOI: 10.1093/ismeco/ycae043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 03/25/2024] [Accepted: 03/25/2024] [Indexed: 05/07/2024]
Abstract
While several environmental factors contribute to the evolutionary diversification of the pathogenic bacterium Pseudomonas aeruginosa during cystic fibrosis lung infections, relatively little is known about the impact of the surrounding microbiota. By using in vitro experimental evolution, we show that the presence of Stenotrophomonas maltophilia, Staphylococcus aureus, or them both, prevent the evolution of loss of virulence, which repeatedly occurs in the absence of these species due to mutations in regulators of the Pseudomonas Quinolone Signal quorum sensing system, vqsM and pqsR. Moreover, the strength of the effect of co-occurring species is attenuated through changes in the physical environment by the addition of mucin, resulting in selection for phenotypes resembling those evolved in the absence of the co-occurring species. Together, our findings show that variation in mucosal environment and the surrounding polymicrobial environment can determine the evolutionary trajectory of P. aeruginosa, partly explaining its diversification and pathoadaptation from acute to chronic phenotype during cystic fibrosis lung infections.
Collapse
Affiliation(s)
- Michael J Bottery
- Division of Evolution Infection and Genomics, School of Biological Sciences, University of Manchester, Manchester M13 9PL, United Kingdom
| | - Helle Krogh Johansen
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen 9301, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Jon W Pitchford
- Department of Biology, University of York, Wentworth Way, York YO10 5DD, United Kingdom
- Department of Mathematics, University of York, Heslington, York YO10 5DD, United Kingdom
| | - Ville-Petri Friman
- Department of Biology, University of York, Wentworth Way, York YO10 5DD, United Kingdom
- Department of Microbiology, University of Helsinki, Helsinki 00014, Finland
| |
Collapse
|
36
|
Greenwald MA, Meinig SL, Plott LM, Roca C, Higgs MG, Vitko NP, Markovetz MR, Rouillard KR, Carpenter J, Kesimer M, Hill DB, Schisler JC, Wolfgang MC. Mucus polymer concentration and in vivo adaptation converge to define the antibiotic response of Pseudomonas aeruginosa during chronic lung infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.20.572620. [PMID: 38187602 PMCID: PMC10769284 DOI: 10.1101/2023.12.20.572620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
The airway milieu of individuals with muco-obstructive airway diseases (MADs) is defined by the accumulation of dehydrated mucus due to hyperabsorption of airway surface liquid and defective mucociliary clearance. Pathological mucus becomes progressively more viscous with age and disease severity due to the concentration and overproduction of mucin and accumulation of host-derived extracellular DNA (eDNA). Respiratory mucus of MADs provides a niche for recurrent and persistent colonization by respiratory pathogens, including Pseudomonas aeruginosa , which is responsible for the majority of morbidity and mortality in MADs. Despite high concentration inhaled antibiotic therapies and the absence of antibiotic resistance, antipseudomonal treatment failure in MADs remains a significant clinical challenge. Understanding the drivers of antibiotic recalcitrance is essential for developing more effective treatments that eradicate persistent infections. The complex and dynamic environment of diseased airways makes it difficult to model antibiotic efficacy in vitro . We aimed to understand how mucin and eDNA concentrations, the two dominant polymers in respiratory mucus, alter the antibiotic tolerance of P. aeruginosa . Our results demonstrate that polymer concentration and molecular weight affect P. aeruginosa survival post antibiotic challenge. Polymer-driven antibiotic tolerance was not explicitly associated with reduced antibiotic diffusion. Lastly, we established a robust and standardized in vitro model for recapitulating the ex vivo antibiotic tolerance of P. aeruginosa observed in expectorated sputum across age, underlying MAD etiology, and disease severity, which revealed the inherent variability in intrinsic antibiotic tolerance of host-evolved P. aeruginosa populations. Importance Antibiotic treatment failure in Pseudomonas aeruginosa chronic lung infections is associated with increased morbidity and mortality, illustrating the clinical challenge of bacterial infection control. Understanding the underlying infection environment, as well as the host and bacterial factors driving antibiotic tolerance and the ability to accurately recapitulate these factors in vitro , is crucial for improving antibiotic treatment outcomes. Here, we demonstrate that increasing concentration and molecular weight of mucin and host eDNA drive increased antibiotic tolerance to tobramycin. Through systematic testing and modeling, we identified a biologically relevant in vitro condition that recapitulates antibiotic tolerance observed in ex vivo treated sputum. Ultimately, this study revealed a dominant effect of in vivo evolved bacterial populations in defining inter-subject ex vivo antibiotic tolerance and establishes a robust and translatable in vitro model for therapeutic development.
Collapse
|
37
|
Youf R, Ghanem R, Nasir A, Lemercier G, Montier T, Le Gall T. Impact of mucus and biofilm on antimicrobial photodynamic therapy: Evaluation using Ruthenium(II) complexes. Biofilm 2023; 5:100113. [PMID: 37396462 PMCID: PMC10313506 DOI: 10.1016/j.bioflm.2023.100113] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/17/2023] [Accepted: 03/19/2023] [Indexed: 07/04/2023] Open
Abstract
The biofilm lifestyle of bacterial pathogens is a hallmark of chronic lung infections such as in cystic fibrosis (CF) patients. Bacterial adaptation to the complex conditions in CF-affected lungs and repeated antibiotherapies lead to increasingly tolerant and hard-to-treat biofilms. In the context of growing antimicrobial resistance and restricted therapeutic options, antimicrobial photodynamic therapy (aPDT) shows great promise as an alternative to conventional antimicrobial modalities. Typically, aPDT consists in irradiating a non-toxic photosensitizer (PS) to generate reactive oxygen species (ROS), which kill pathogens in the surrounding environment. In a previous study, we reported that some ruthenium (II) complexes ([Ru(II)]) can mediate potent photodynamic inactivation (PDI) against planktonic cultures of Pseudomonas aeruginosa and Staphylococcus aureus clinical isolates. In the present work, [Ru(II)] were further assayed to evaluate their ability to photo-inactivate such bacteria under more complex experimental conditions better recapitulating the microenvironment in lung infected airways. Bacterial PDI was tentatively correlated with the properties of [Ru(II)] in biofilms, in mucus, and following diffusion across the latter. Altogether, the results obtained demonstrate the negative impacting role of mucus and biofilm components on [Ru(II)]-mediated PDT, following different possible mechanisms of action. Technical limitations were also identified that may be overcome, making this report a pilot for other similar studies. In conclusion, [Ru(II)] may be subjected to specific chemical engineering and/or drug formulation to adapt their properties to the harsh micro-environmental conditions of the infected respiratory tract.
Collapse
Affiliation(s)
- Raphaëlle Youf
- Inserm, Univ Brest, EFS, UMR 1078, GGB, F-29200, Brest, France
| | - Rosy Ghanem
- Inserm, Univ Brest, EFS, UMR 1078, GGB, F-29200, Brest, France
- CHU de Brest, Service de Génétique Médicale et de Biologie de la Reproduction, 29200, Brest, France
| | - Adeel Nasir
- Inserm, Univ Brest, EFS, UMR 1078, GGB, F-29200, Brest, France
| | - Gilles Lemercier
- Université de Reims Champagne-Ardenne, UMR CNRS 7312, BP 1039, CEDEX 2, 51687, Reims, France
| | - Tristan Montier
- Inserm, Univ Brest, EFS, UMR 1078, GGB, F-29200, Brest, France
- CHU de Brest, Service de Génétique Médicale et de Biologie de la Reproduction, 29200, Brest, France
- CHU de Brest, Centre de Référence des Maladies Rares Maladies Neuromusculaires, 29200, Brest, France
| | - Tony Le Gall
- Inserm, Univ Brest, EFS, UMR 1078, GGB, F-29200, Brest, France
| |
Collapse
|
38
|
Tabassum N, Jeong GJ, Jo DM, Khan F, Kim YM. Treatment of Staphylococcus aureus and Candida albicans polymicrobial biofilms by phloroglucinol-gold nanoparticles. Microb Pathog 2023; 185:106416. [PMID: 37866550 DOI: 10.1016/j.micpath.2023.106416] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/15/2023] [Accepted: 10/19/2023] [Indexed: 10/24/2023]
Abstract
The co-isolation of Staphylococcus aureus and Candida albicans from host tissues and organs and their in vitro and in vivo interaction studies suggest a synergistic relationship in forming polymicrobial biofilms. In particular, during polymicrobial biofilm formation, S. aureus becomes coated in the extracellular matrix secreted by C. albicans, leading to enhanced resistance to antibiotics. Accordingly, understanding the interactions between S. aureus and C. albicans in polymicrobial biofilms is of utmost importance in establishing treatment strategies for polymicrobial infections. As an alternate technique, nanoparticles were used in this investigation to suppress polymicrobial biofilm. The current study aims to manufacture gold nanoparticles (AuNPs) using phloroglucinol (PG), a natural chemical, and test their inhibitory capabilities against S. aureus and C. albicans biofilms in standard and host-mimicking media (like saliva and sputum). PG-AuNPs have a spherical form with an average size of 46.71 ± 6.40 nm. The minimum inhibitory concentration (MIC) values differed when PG-AuNPs were evaluated in the standard and host-mimicking artificial media. The MIC of PG-AuNPs against S. aureus and C. albicans was 2048 μg/mL in both the standard and artificial sputum media. However, the MIC in saliva was only 128 μg/mL. The initial stage polymicrobial biofilm of S. aureus and C. albicans was dramatically decreased at the sub-MIC of PG-AuNPs in both standard and host-mimicking media. S. aureus and C. albicans mature polymicrobial biofilms were more effectively eliminated by MIC and sub-MIC of PG-AuNPs. This study indicates that PG-AuNPs have the ability to limit the formation of polymicrobial biofilms caused by bacterial and fungal diseases.
Collapse
Affiliation(s)
- Nazia Tabassum
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Geum-Jae Jeong
- Department of Food Science and Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Du-Min Jo
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea; Department of Food Science and Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Fazlurrahman Khan
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea; Institute of Fisheries Sciences, Pukyong National University, Busan, 48513, Republic of Korea.
| | - Young-Mog Kim
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea; Department of Food Science and Technology, Pukyong National University, Busan, 48513, Republic of Korea.
| |
Collapse
|
39
|
Sun N, Yang AP, Wang SM, Zhu GL, Liu J, Wang TY, Wang ZJ, Qi BW, Liu XY, Lv SX, Li MH, Fu Q. Mechanism of synergistic remediation of soil phenanthrene contamination in paddy fields by rice-crab coculture and bioaugmentation with Pseudomonas sp. ENVIRONMENT INTERNATIONAL 2023; 182:108315. [PMID: 37963424 DOI: 10.1016/j.envint.2023.108315] [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: 05/30/2023] [Revised: 09/30/2023] [Accepted: 11/07/2023] [Indexed: 11/16/2023]
Abstract
Polycyclic aromatic hydrocarbons (PAHs) are persistent and harmful pollutants with high priority concern in agricultural fields. This work constructed a rice-crab coculture and bioaugmentation (RCM) system to remediate phenanthrene (a model PAH) contamination in rice fields. The results showed that RCM had a higher remediation performance of phenanthrene in rice paddy compared with rice cultivation alone, microbial addition alone, and crab-rice coculture, reaching a remediation efficiency of 88.92 % in 42 d. The concentration of phenanthrene in the rice plants decreased to 6.58 mg/kg, and its bioconcentration effect was efficiently inhibited in the RCM system. In addition, some low molecular weight organic acids of rice root increased by 12.87 %∼73.87 %, and some amino acids increased by 140 %∼1150 % in RCM. Bioturbation of crabs improves soil aeration structure and microbial migration, and adding Pseudomonas promoted the proliferation of some plant growth-promoting rhizobacteria (PGPRs), which facilitated the degradation of phenanthrene. This coupling rice-crab coculture with bioaugmentation had favorable effects on soil enzyme activity, microbial community structure, and PAH degradation genes in paddy fields, enhancing the removal of and resistance to PAH contamination in paddy fields and providing new strategies for achieving a balance between production and remediation in contaminated paddy fields.
Collapse
Affiliation(s)
- Nan Sun
- School of Water Conservancy and Civil Engineering, Northeast Agricultural University, Harbin 150030, China; Key Laboratory of Efficient Use of Agricultural Water Resources, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Northeast Agricultural University, Harbin 150030, China; Northeast Agricultural University/Heilongjiang Academy of Environmental Science Joint Postdoctoral Mobile Station, Harbin 150030, China
| | - An-Pei Yang
- School of Water Conservancy and Civil Engineering, Northeast Agricultural University, Harbin 150030, China; Research Center for Ecological Agriculture and Soil-Water Environment Restoration, Northeast Agricultural University, Harbin 150030, China
| | - Si-Ming Wang
- School of Water Conservancy and Civil Engineering, Northeast Agricultural University, Harbin 150030, China; Research Center for Ecological Agriculture and Soil-Water Environment Restoration, Northeast Agricultural University, Harbin 150030, China
| | - Guang-Lei Zhu
- School of Water Conservancy and Civil Engineering, Northeast Agricultural University, Harbin 150030, China; Research Center for Ecological Agriculture and Soil-Water Environment Restoration, Northeast Agricultural University, Harbin 150030, China
| | - Jin Liu
- School of Water Conservancy and Civil Engineering, Northeast Agricultural University, Harbin 150030, China; Research Center for Ecological Agriculture and Soil-Water Environment Restoration, Northeast Agricultural University, Harbin 150030, China
| | - Tian-Yi Wang
- School of Water Conservancy and Civil Engineering, Northeast Agricultural University, Harbin 150030, China; Research Center for Ecological Agriculture and Soil-Water Environment Restoration, Northeast Agricultural University, Harbin 150030, China
| | - Zi-Jian Wang
- School of Water Conservancy and Civil Engineering, Northeast Agricultural University, Harbin 150030, China; Research Center for Ecological Agriculture and Soil-Water Environment Restoration, Northeast Agricultural University, Harbin 150030, China
| | - Bo-Wei Qi
- School of Water Conservancy and Civil Engineering, Northeast Agricultural University, Harbin 150030, China; Research Center for Ecological Agriculture and Soil-Water Environment Restoration, Northeast Agricultural University, Harbin 150030, China
| | - Xin-Ying Liu
- School of Water Conservancy and Civil Engineering, Northeast Agricultural University, Harbin 150030, China; Research Center for Ecological Agriculture and Soil-Water Environment Restoration, Northeast Agricultural University, Harbin 150030, China
| | - Shao-Xuan Lv
- School of Water Conservancy and Civil Engineering, Northeast Agricultural University, Harbin 150030, China; Research Center for Ecological Agriculture and Soil-Water Environment Restoration, Northeast Agricultural University, Harbin 150030, China
| | - Ming-Hang Li
- School of Water Conservancy and Civil Engineering, Northeast Agricultural University, Harbin 150030, China; Research Center for Ecological Agriculture and Soil-Water Environment Restoration, Northeast Agricultural University, Harbin 150030, China
| | - Qiang Fu
- School of Water Conservancy and Civil Engineering, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
40
|
Vasiljevs S, Gupta A, Baines D. Effect of glucose on growth and co-culture of Staphylococcus aureus and Pseudomonas aeruginosa in artificial sputum medium. Heliyon 2023; 9:e21469. [PMID: 37908712 PMCID: PMC10613906 DOI: 10.1016/j.heliyon.2023.e21469] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 10/05/2023] [Accepted: 10/21/2023] [Indexed: 11/02/2023] Open
Abstract
People with cystic fibrosis-related diabetes (CFRD) suffer from chronic infections with Staphylococcus aureus and/or Pseudomonas aeruginosa. In people with CFRD, the concentration of glucose in the airway surface liquid (ASL) was shown to be elevated from 0.4 to 4 mM. The effect of glucose on bacterial growth/interactions in ASL is not well understood and here we studied the relationship between these lung pathogens in artificial sputum medium (ASM), an environment similar to ASL in vivo. S. aureus exhibited more rapid adaptation to growth in ASM than P. aeruginosa. Supplementation of ASM with glucose significantly increased the growth of S. aureus (p < 0.01, n = 5) and P. aeruginosa (p < 0.001, n = 3). ASM conditioned by the presence of S. aureus promoted growth of P. aeruginosa with less lag time compared with non-conditioned ASM, or conditioned medium that had been heated to 121 °C. Stable co-culture of S. aureus and P. aeruginosa could be established in a 50:50 mix of ASM and S. aureus-conditioned supernatant. These data indicate that glucose, in a nutrient depleted environment, can promote the growth of S. aureus and P. aeruginosa. In addition, heat labile factors present in S. aureus pre-conditioned ASM promoted the growth of P. aeruginosa. We suggest that the use of ASM allows investigation of the effects of nutrients such as glucose on common lung pathogens. ASM could be further used to understand the relationship between S. aureus and P. aeruginosa in a co-culture scenario. Our model of stable co-culture could be extrapolated to include other common lung pathogens and could be used to better understand disease progression in vitro.
Collapse
Affiliation(s)
- Stanislavs Vasiljevs
- Institute for Infection and Immunity, St George's University of London, Cranmer Terrace, Tooting, London, SW17 0RE, UK
| | - Arya Gupta
- School of Health, Leeds Beckett University, Leeds, LS1 3HE, UK
| | - Deborah Baines
- Institute for Infection and Immunity, St George's University of London, Cranmer Terrace, Tooting, London, SW17 0RE, UK
| |
Collapse
|
41
|
Carullo G, Di Bonaventura G, Rossi S, Lupetti V, Tudino V, Brogi S, Butini S, Campiani G, Gemma S, Pompilio A. Development of Quinazolinone Derivatives as Modulators of Virulence Factors of Pseudomonas aeruginosa Cystic Fibrosis Strains. Molecules 2023; 28:6535. [PMID: 37764311 PMCID: PMC10536951 DOI: 10.3390/molecules28186535] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 09/04/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
Pseudomonas aeruginosa (PA), one of the ESKAPE pathogens, is an opportunistic Gram-negative bacterium responsible for nosocomial infections in humans but also for infections in patients affected by AIDS, cancer, or cystic fibrosis (CF). Treatment of PA infections in CF patients is a global healthcare problem due to the ability of PA to gain antibiotic tolerance through biofilm formation. Anti-virulence compounds represent a promising approach as adjuvant therapy, which could reduce or eliminate the pathogenicity of PA without impacting its growth. Pyocyanin is one of the virulence factors whose production is modulated by the Pseudomonas quinolone signal (PQS) through its receptor PqsR. Different PqsR modulators have been synthesized over the years, highlighting this new powerful therapeutic strategy. Based on the promising structure of quinazolin-4(3H)-one, we developed compounds 7a-d, 8a,b, 9, 10, and 11a-f able to reduce biofilm formation and the production of virulence factors (pyocyanin and pyoverdine) at 50 µM in two PA strains responsible for CF acute and chronic infections. The developed compounds did not reduce the cell viability of IB3-1 bronchial CF cells, and computational studies confirmed the potential ability of novel compounds to act as potential Pqs system modulators.
Collapse
Affiliation(s)
- Gabriele Carullo
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (G.C.); (S.R.); (V.T.); (S.B.); (G.C.)
| | - Giovanni Di Bonaventura
- Department of Medical, Oral and Biotechnological Sciences, School of Medicine and Health Sciences “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy; (G.D.B.); (V.L.); (A.P.)
| | - Sara Rossi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (G.C.); (S.R.); (V.T.); (S.B.); (G.C.)
| | - Veronica Lupetti
- Department of Medical, Oral and Biotechnological Sciences, School of Medicine and Health Sciences “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy; (G.D.B.); (V.L.); (A.P.)
| | - Valeria Tudino
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (G.C.); (S.R.); (V.T.); (S.B.); (G.C.)
| | - Simone Brogi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy;
| | - Stefania Butini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (G.C.); (S.R.); (V.T.); (S.B.); (G.C.)
| | - Giuseppe Campiani
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (G.C.); (S.R.); (V.T.); (S.B.); (G.C.)
| | - Sandra Gemma
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (G.C.); (S.R.); (V.T.); (S.B.); (G.C.)
| | - Arianna Pompilio
- Department of Medical, Oral and Biotechnological Sciences, School of Medicine and Health Sciences “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy; (G.D.B.); (V.L.); (A.P.)
| |
Collapse
|
42
|
Di Bonaventura G, Lupetti V, Di Giulio A, Muzzi M, Piccirilli A, Cariani L, Pompilio A. Repurposing High-Throughput Screening Identifies Unconventional Drugs with Antibacterial and Antibiofilm Activities against Pseudomonas aeruginosa under Experimental Conditions Relevant to Cystic Fibrosis. Microbiol Spectr 2023; 11:e0035223. [PMID: 37306577 PMCID: PMC10433973 DOI: 10.1128/spectrum.00352-23] [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: 01/22/2023] [Accepted: 05/13/2023] [Indexed: 06/13/2023] Open
Abstract
Pseudomonas aeruginosa is the most common pathogen infecting cystic fibrosis (CF) lungs, causing acute and chronic infections. Intrinsic and acquired antibiotic resistance allow P. aeruginosa to colonize and persist despite antibiotic treatment, making new therapeutic approaches necessary. Combining high-throughput screening and drug repurposing is an effective way to develop new therapeutic uses for drugs. This study screened a drug library of 3,386 drugs, mostly FDA approved, to identify antimicrobials against P. aeruginosa under physicochemical conditions relevant to CF-infected lungs. Based on the antibacterial activity, assessed spectrophotometrically against the prototype RP73 strain and 10 other CF virulent strains, and the toxic potential evaluated toward CF IB3-1 bronchial epithelial cells, five potential hits were selected for further analysis: the anti-inflammatory and antioxidant ebselen, the anticancer drugs tirapazamine, carmofur, and 5-fluorouracil, and the antifungal tavaborole. A time-kill assay showed that ebselen has the potential to cause rapid and dose-dependent bactericidal activity. The antibiofilm activity was evaluated by viable cell count and crystal violet assays, revealing carmofur and 5-fluorouracil as the most active drugs in preventing biofilm formation regardless of the concentration. In contrast, tirapazamine and tavaborole were the only drugs actively dispersing preformed biofilms. Tavaborole was the most active drug against CF pathogens other than P. aeruginosa, especially against Burkholderia cepacia and Acinetobacter baumannii, while carmofur, ebselen, and tirapazamine were particularly active against Staphylococcus aureus and B. cepacia. Electron microscopy and propidium iodide uptake assay revealed that ebselen, carmofur, and tirapazamine significantly damage cell membranes, with leakage and cytoplasm loss, by increasing membrane permeability. IMPORTANCE Antibiotic resistance makes it urgent to design new strategies for treating pulmonary infections in CF patients. The repurposing approach accelerates drug discovery and development, as the drugs' general pharmacological, pharmacokinetic, and toxicological properties are already well known. In the present study, for the first time, a high-throughput compound library screening was performed under experimental conditions relevant to CF-infected lungs. Among 3,386 drugs screened, the clinically used drugs from outside infection treatment ebselen, tirapazamine, carmofur, 5-fluorouracil, and tavaborole showed, although to different extents, anti-P. aeruginosa activity against planktonic and biofilm cells and broad-spectrum activity against other CF pathogens at concentrations not toxic to bronchial epithelial cells. The mode-of-action studies revealed ebselen, carmofur, and tirapazamine targeted the cell membrane, increasing its permeability with subsequent cell lysis. These drugs are strong candidates for repurposing for treating CF lung P. aeruginosa infections.
Collapse
Affiliation(s)
- Giovanni Di Bonaventura
- Department of Medical, Oral, and Biotechnological Sciences, G. d’Annunzio University of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology, G. d’Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Veronica Lupetti
- Department of Medical, Oral, and Biotechnological Sciences, G. d’Annunzio University of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology, G. d’Annunzio University of Chieti-Pescara, Chieti, Italy
| | | | | | - Alessandra Piccirilli
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L’Aquila, Italy
| | - Lisa Cariani
- Microbiology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Arianna Pompilio
- Department of Medical, Oral, and Biotechnological Sciences, G. d’Annunzio University of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology, G. d’Annunzio University of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
43
|
Manner C, Dias Teixeira R, Saha D, Kaczmarczyk A, Zemp R, Wyss F, Jaeger T, Laventie BJ, Boyer S, Malone JG, Qvortrup K, Andersen JB, Givskov M, Tolker-Nielsen T, Hiller S, Drescher K, Jenal U. A genetic switch controls Pseudomonas aeruginosa surface colonization. Nat Microbiol 2023; 8:1520-1533. [PMID: 37291227 DOI: 10.1038/s41564-023-01403-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 05/05/2023] [Indexed: 06/10/2023]
Abstract
Efficient colonization of mucosal surfaces is essential for opportunistic pathogens like Pseudomonas aeruginosa, but how bacteria collectively and individually adapt to optimize adherence, virulence and dispersal is largely unclear. Here we identified a stochastic genetic switch, hecR-hecE, which is expressed bimodally and generates functionally distinct bacterial subpopulations to balance P. aeruginosa growth and dispersal on surfaces. HecE inhibits the phosphodiesterase BifA and stimulates the diguanylate cyclase WspR to increase c-di-GMP second messenger levels and promote surface colonization in a subpopulation of cells; low-level HecE-expressing cells disperse. The fraction of HecE+ cells is tuned by different stress factors and determines the balance between biofilm formation and long-range cell dispersal of surface-grown communities. We also demonstrate that the HecE pathway represents a druggable target to effectively counter P. aeruginosa surface colonization. Exposing such binary states opens up new ways to control mucosal infections by a major human pathogen.
Collapse
Affiliation(s)
| | | | - Dibya Saha
- Biozentrum, University of Basel, Basel, Switzerland
| | | | | | - Fabian Wyss
- Biozentrum, University of Basel, Basel, Switzerland
| | - Tina Jaeger
- Biozentrum, University of Basel, Basel, Switzerland
- Department Biomedizin, University of Basel, Basel, Switzerland
| | | | - Sebastien Boyer
- sciCORE, Centre for Scientific Computing, University of Basel, Basel, Switzerland
| | - Jacob G Malone
- Biozentrum, University of Basel, Basel, Switzerland
- Department of Molecular Microbiology, John Innes Centre, Norwich, UK
| | - Katrine Qvortrup
- Department of Chemistry, Technical University of Denmark, Lyngby, Denmark
| | - Jens Bo Andersen
- Costerton Biofilm Center, University of Copenhagen, Copenhagen, Denmark
| | - Michael Givskov
- Costerton Biofilm Center, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | - Urs Jenal
- Biozentrum, University of Basel, Basel, Switzerland.
| |
Collapse
|
44
|
Batoni G, Catelli E, Kaya E, Pompilio A, Bianchi M, Ghelardi E, Di Bonaventura G, Esin S, Maisetta G. Antibacterial and Antibiofilm Effects of Lactobacilli Strains against Clinical Isolates of Pseudomonas aeruginosa under Conditions Relevant to Cystic Fibrosis. Antibiotics (Basel) 2023; 12:1158. [PMID: 37508254 PMCID: PMC10376640 DOI: 10.3390/antibiotics12071158] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
Therapy of lung infections sustained by Pseudomonas aeruginosa in cystic fibrosis (CF) patients is challenging due to the presence of a sticky mucus in the airways and the ability of the bacterium to form biofilm, which exhibits increased antibiotic tolerance. A lung-directed bacteriotherapy through the airway administration of probiotics could represent an alternative approach to probiotic diet supplementation to improve the benefits and clinical outcomes of this kind of intervention in CF patients. This study aims to evaluate the ability of probiotic strains to grow in artificial sputum medium (ASM), mimicking the CF lung microenvironment, and to affect the planktonic and biofilm growth of CF clinical strains of P. aeruginosa in the same conditions. The results demonstrate that Lacticaseibacillus rhamnosus and Lactiplantibacillus plantarum (LP) can grow in ASM. LP inhibited the planktonic growth of P. aeruginosa, while both lactobacilli reduced the pre-formed biofilm of P. aeruginosa. Interestingly, LP was demonstrated to reduce the amount of polysaccharides in the extracellular matrix of P. aeruginosa biofilms and to potentiate the antibiofilm effects of tobramycin. Overall, the results indicated that LP is a promising candidate as an adjuvant in the antimicrobial therapy of P. aeruginosa infections in CF patients.
Collapse
Affiliation(s)
- Giovanna Batoni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via S. Zeno 37, 56123 Pisa, Italy
| | - Elisa Catelli
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via S. Zeno 37, 56123 Pisa, Italy
| | - Esingül Kaya
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via S. Zeno 37, 56123 Pisa, Italy
| | - Arianna Pompilio
- Department of Medical, Oral, and Biotechnological Sciences, G. d'Annunzio University of Chieti-Pescara, Via dei Vestini, 31, 66100 Chieti, Italy
- Center for Advanced Studies and Technology, G. d'Annunzio University of Chieti-Pescara, Via L. Polacchi 11, 66100 Chieti, Italy
| | - Marta Bianchi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via S. Zeno 37, 56123 Pisa, Italy
| | - Emilia Ghelardi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via S. Zeno 37, 56123 Pisa, Italy
| | - Giovanni Di Bonaventura
- Department of Medical, Oral, and Biotechnological Sciences, G. d'Annunzio University of Chieti-Pescara, Via dei Vestini, 31, 66100 Chieti, Italy
- Center for Advanced Studies and Technology, G. d'Annunzio University of Chieti-Pescara, Via L. Polacchi 11, 66100 Chieti, Italy
| | - Semih Esin
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via S. Zeno 37, 56123 Pisa, Italy
| | - Giuseppantonio Maisetta
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via S. Zeno 37, 56123 Pisa, Italy
| |
Collapse
|
45
|
Craddock VD, Steere EL, Harman H, Britt NS. Activity of Delafloxacin and Comparator Fluoroquinolones against Multidrug-Resistant Pseudomonas aeruginosa in an In Vitro Cystic Fibrosis Sputum Model. Antibiotics (Basel) 2023; 12:1078. [PMID: 37370396 DOI: 10.3390/antibiotics12061078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Delafloxacin (DLX) is a recently approved fluoroquinolone with broad activity against common cystic fibrosis (CF) pathogens, including multidrug-resistant Pseudomonas aeruginosa (MDR-Psa). Delafloxacin has been previously shown to have excellent lung and biofilm penetration and enhanced activity at lower pH environments, such as those that would be observed in the CF lung. We analyzed six Psa strains isolated from CF sputum and compared DLX to ciprofloxacin (CPX) and levofloxacin (LVX). Minimum inhibitory concentrations (MICs) were determined for DLX using standard culture media (pH 7.3) and artificial sputum media (ASM), a physiologic media recapitulating the CF lung microenvironment (pH 6.9). Delafloxacin activity was further compared to CPX and LVX in an in vitro CF sputum time-kill model at physiologically relevant drug concentrations (Cmax, Cmed, Cmin). Delafloxacin exhibited 2- to 4-fold MIC reductions in ASM, which corresponded with significant improvements in bacterial killing in the CF sputum time-kill model between DLX and LVX at Cmed (p = 0.033) and Cmin (p = 0.004). Compared to CPX, DLX demonstrated significantly greater killing at Cmin (p = 0.024). Overall, DLX demonstrated favorable in vitro activity compared to alternative fluoroquinolones against MDR-Psa. Delafloxacin may be considered as an option against MDR-Psa pulmonary infections in CF.
Collapse
Affiliation(s)
- Vaughn D Craddock
- Department of Pharmacy Practice, University of Kansas School of Pharmacy, Lawrence, KS 66047, USA
| | - Evan L Steere
- Department of Population Health, University of Kansas School of Medicine, Kansas City, KS 66160, USA
| | - Hannah Harman
- Department of Pharmacy Practice, University of Kansas School of Pharmacy, Lawrence, KS 66047, USA
| | - Nicholas S Britt
- Department of Pharmacy Practice, University of Kansas School of Pharmacy, Lawrence, KS 66047, USA
- Department of Internal Medicine, University of Kansas School of Medicine, Kansas City, KS 66160, USA
| |
Collapse
|
46
|
Dou Q, Zhu Y, Li C, Bian Z, Song H, Zhang R, Wang Y, Zhang X, Wang Y. 4F-Indole Enhances the Susceptibility of Pseudomonas aeruginosa to Aminoglycoside Antibiotics. Microbiol Spectr 2023; 11:e0451922. [PMID: 36975825 PMCID: PMC10100892 DOI: 10.1128/spectrum.04519-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 03/06/2023] [Indexed: 03/29/2023] Open
Abstract
Infections caused by multidrug-resistant bacteria are becoming increasingly serious. The aminoglycoside antibiotics have been widely used to treat severe Gram-negative bacterial infections. Here, we reported that a class of small molecules, namely, halogenated indoles, can resensitize Pseudomonas aeruginosa PAO1 to aminoglycoside antibiotics such as gentamicin, kanamycin, tobramycin, amikacin, neomycin, ribosomalin sulfate, and cisomicin. We selected 4F-indole as a representative of halogenated indoles to investigate its mechanism and found that the two-component system (TCS) PmrA/PmrB inhibited the expression of multidrug efflux pump MexXY-OprM, allowing kanamycin to act intracellularly. Moreover, 4F-indole inhibited the biosynthesis of several virulence factors, such as pyocyanin, type III secretion system (T3SS), and type VI secretion system (T6SS) exported effectors, and reduced the swimming and twitching motility by suppressing the expression of flagella and type IV pili. This study suggests that the combination of 4F-indole and kanamycin can be more effective against P. aeruginosa PAO1 and affect its multiple physiological activities, providing a novel insight into the reactivation of aminoglycoside antibiotics. IMPORTANCE Infections caused by Pseudomonas aeruginosa have become a major public health crisis. Its resistance to existing antibiotics causes clinical infections that are hard to cure. In this study, we found that halogenated indoles in combination with aminoglycoside antibiotics could be more effective than antibiotics alone against P. aeruginosa PAO1 and preliminarily revealed the mechanism of the 4F-indole-induced regulatory effect. Moreover, the regulatory effect of 4F-indole on different physiological behaviors of P. aeruginosa PAO1 was analyzed by combined transcriptomics and metabolomics. We explain that 4F-indole has potential as a novel antibiotic adjuvant, thus slowing down the further development of bacterial resistance.
Collapse
Affiliation(s)
- Qin Dou
- College of Marine Life Sciences, and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, China
| | - Yuxiang Zhu
- College of Marine Life Sciences, and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, China
| | - Chunhui Li
- College of Marine Life Sciences, and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, China
| | - Zeran Bian
- College of Marine Life Sciences, and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, China
| | - Huihui Song
- College of Marine Life Sciences, and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, China
| | - Ruizhen Zhang
- College of Marine Life Sciences, and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, China
| | - Yingsong Wang
- College of Marine Life Sciences, and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, China
| | - Xile Zhang
- College of Marine Life Sciences, and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, China
| | - Yan Wang
- College of Marine Life Sciences, and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, China
| |
Collapse
|
47
|
De Bleeckere A, Van den Bossche S, De Sutter PJ, Beirens T, Crabbé A, Coenye T. High throughput determination of the biofilm prevention concentration for Pseudomonas aeruginosa biofilms using a synthetic cystic fibrosis sputum medium. Biofilm 2023; 5:100106. [PMID: 36845825 PMCID: PMC9945637 DOI: 10.1016/j.bioflm.2023.100106] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 01/17/2023] [Accepted: 02/04/2023] [Indexed: 02/09/2023] Open
Abstract
The presence of Pseudomonas aeruginosa biofilms in cystic fibrosis (CF) patients suffering from chronic lung infections contributes to the failure of antimicrobial therapy. Conventionally, the minimal inhibitory concentration (MIC) is determined to assess the antimicrobial susceptibility of a pathogen, however this parameter fails to predict success in treating biofilm-associated infections. In the present study we developed a high throughput method to determine the antimicrobial concentration required to prevent P. aeruginosa biofilm formation, using a synthetic cystic fibrosis sputum medium (SCFM2). Biofilms were grown in SCFM2 for 24 h in the presence of antibiotics (tobramycin, ciprofloxacin or colistin), whereafter biofilms were disrupted and a resazurin staining was used to quantify the number of surviving metabolically active cells. In parallel, the content of all wells was plated to determine the number of colony forming units (CFU). Biofilm preventing concentrations (BPCs) were compared to MICs and minimal bactericidal concentrations (MBCs) determined according to EUCAST guidelines. Correlations between the resazurin-derived fluorescence and CFU counts were assessed with Kendall's Tau Rank tests. A significant correlation between fluorescence and CFU counts was observed for 9 out of 10 strains investigated, suggesting the fluorometric assay is a reliable alternative to plating for most P. aeruginosa isolates to determine biofilm susceptibility in relevant conditions. For all isolates a clear difference between MICs and BPCs of all three antibiotics was observed, with the BPCs being consistently higher than the MICs. Additionally, the extent of this difference appeared to be antibiotic-dependent. Our findings suggest that this high throughput assay could be a valuable addition to evaluate the antimicrobial susceptibility in P. aeruginosa biofilms in the context of CF.
Collapse
Affiliation(s)
- Amber De Bleeckere
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | | | - Pieter-Jan De Sutter
- Laboratory of Medical Biochemistry and Clinical Analysis, Ghent University, Ghent, Belgium
| | - Tine Beirens
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | - Aurélie Crabbé
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | - Tom Coenye
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium,Corresponding author.
| |
Collapse
|
48
|
Jean-Pierre V, Boudet A, Sorlin P, Menetrey Q, Chiron R, Lavigne JP, Marchandin H. Biofilm Formation by Staphylococcus aureus in the Specific Context of Cystic Fibrosis. Int J Mol Sci 2022; 24:ijms24010597. [PMID: 36614040 PMCID: PMC9820612 DOI: 10.3390/ijms24010597] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022] Open
Abstract
Staphylococcus aureus is a major human pathogen whose characteristics support its success in various clinical settings including Cystic Fibrosis (CF). In CF, S. aureus is indeed the most commonly identified opportunistic pathogen in children and the overall population. S. aureus colonization/infection, either by methicillin-susceptible or methicillin-resistant strains, will become chronic in about one third of CF patients. The persistence of S. aureus in CF patients' lungs, despite various eradication strategies, is favored by several traits in both host and pathogen. Among the latter, living in biofilm is a highly protective way to survive despite deleterious environmental conditions, and is a common characteristic shared by the main pathogens identified in CF. This is why CF has earned the status of a biofilm-associated disease for several years now. Biofilm formation by S. aureus, and the molecular mechanisms governing and regulating it, have been extensively studied but have received less attention in the specific context of CF lungs. Here, we review the current knowledge on S. aureus biofilm in this very context, i.e., the importance, study methods, molecular data published on mono- and multi-species biofilm and anti-biofilm strategies. This focus on studies including clinical isolates from CF patients shows that they are still under-represented in the literature compared with studies based on reference strains, and underlines the need for such studies. Indeed, CF clinical strains display specific characteristics that may not be extrapolated from results obtained on laboratory strains.
Collapse
Affiliation(s)
- Vincent Jean-Pierre
- HSM—HydroSciences Montpellier, Université de Montpellier, CNRS, IRD, Service de Microbiologie et Hygiène Hospitalière, CHU Nîmes, 34093 Montpellier, France
| | - Agathe Boudet
- VBIC—Virulence Bactérienne et Infections Chroniques, Université de Montpellier, INSERM U1047, Service de Microbiologie et Hygiène Hospitalière, CHU Nîmes, 30900 Nîmes, France
| | - Pauline Sorlin
- HSM—HydroSciences Montpellier, Université de Montpellier, CNRS, IRD, 34093 Montpellier, France
| | - Quentin Menetrey
- INFINITE—Institute for Translational Research in Inflammation, Université de Lille, INSERM U1286, CHU Lille, 59000 Lille, France
| | - Raphaël Chiron
- HSM—HydroSciences Montpellier, Université de Montpellier, CNRS, IRD, Centre de Ressources et de Compétences de la Mucoviscidose, CHU Montpellier, 34295 Montpellier, France
| | - Jean-Philippe Lavigne
- VBIC—Virulence Bactérienne et Infections Chroniques, Université de Montpellier, INSERM U1047, Service de Microbiologie et Hygiène Hospitalière, CHU Nîmes, 30900 Nîmes, France
| | - Hélène Marchandin
- HSM—HydroSciences Montpellier, Université de Montpellier, CNRS, IRD, Service de Microbiologie et Hygiène Hospitalière, CHU Nîmes, 34093 Montpellier, France
- Correspondence:
| |
Collapse
|
49
|
Choudhary MI, Römling U, Nadeem F, Bilal HM, Zafar M, Jahan H, ur-Rahman A. Innovative Strategies to Overcome Antimicrobial Resistance and Tolerance. Microorganisms 2022; 11:microorganisms11010016. [PMID: 36677308 PMCID: PMC9863313 DOI: 10.3390/microorganisms11010016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/15/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Antimicrobial resistance and tolerance are natural phenomena that arose due to evolutionary adaptation of microorganisms against various xenobiotic agents. These adaptation mechanisms make the current treatment options challenging as it is increasingly difficult to treat a broad range of infections, associated biofilm formation, intracellular and host adapted microbes, as well as persister cells and microbes in protected niches. Therefore, novel strategies are needed to identify the most promising drug targets to overcome the existing hurdles in the treatment of infectious diseases. Furthermore, discovery of novel drug candidates is also much needed, as few novel antimicrobial drugs have been introduced in the last two decades. In this review, we focus on the strategies that may help in the development of innovative small molecules which can interfere with microbial resistance mechanisms. We also highlight the recent advances in optimization of growth media which mimic host conditions and genome scale molecular analyses of microbial response against antimicrobial agents. Furthermore, we discuss the identification of antibiofilm molecules and their mechanisms of action in the light of the distinct physiology and metabolism of biofilm cells. This review thus provides the most recent advances in host mimicking growth media for effective drug discovery and development of antimicrobial and antibiofilm agents.
Collapse
Affiliation(s)
- M. Iqbal Choudhary
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Ute Römling
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 65 Stockholm, Sweden
- Correspondence: (U.R.); (H.J.); Tel.: +46-8-5248-7319 (U.R.); +92-21-111-232-292 (ext. 301) (H.J.)
| | - Faiza Nadeem
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Hafiz Muhammad Bilal
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Munirah Zafar
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Humera Jahan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
- Correspondence: (U.R.); (H.J.); Tel.: +46-8-5248-7319 (U.R.); +92-21-111-232-292 (ext. 301) (H.J.)
| | - Atta ur-Rahman
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| |
Collapse
|
50
|
Barton TE, Frost F, Fothergill JL, Neill DR. Challenges and opportunities in the development of novel antimicrobial therapeutics for cystic fibrosis. J Med Microbiol 2022; 71. [PMID: 36748497 DOI: 10.1099/jmm.0.001643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Chronic respiratory infection is the primary driver of mortality in individuals with cystic fibrosis (CF). Existing drug screening models utilised in preclinical antimicrobial development are unable to mimic the complex CF respiratory environment. Consequently, antimicrobials showing promising activity in preclinical models often fail to translate through to clinical efficacy in people with CF. Model systems used in CF anti-infective drug discovery and development range from antimicrobial susceptibility testing in nutrient broth, through to 2D and 3D in vitro tissue culture systems and in vivo models. No single model fully recapitulates every key aspect of the CF lung. To improve the outcomes of people with CF (PwCF) it is necessary to develop a set of preclinical models that collectively recapitulate the CF respiratory environment to a high degree of accuracy. Models must be validated for their ability to mimic aspects of the CF lung and associated lung infection, through evaluation of biomarkers that can also be assessed following treatment in the clinic. This will give preclinical models greater predictive power for identification of antimicrobials with clinical efficacy. The landscape of CF is changing, with the advent of modulator therapies that correct the function of the CFTR protein, while antivirulence drugs and phage therapy are emerging alternative treatments to chronic infection. This review discusses the challenges faced in current antimicrobial development pipelines, including the advantages and disadvantages of current preclinical models and the impact of emerging treatments.
Collapse
Affiliation(s)
- Thomas E Barton
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Ronald Ross Building, 8 West Derby Street, Liverpool, L69 7BE, UK
| | - Frederick Frost
- Adult Cystic Fibrosis Centre, Liverpool Heart & Chest Hospital NHS Foundation Trust, Liverpool, UK.,Liverpool Centre for Cardiovascular Sciences, University of Liverpool, Liverpool, UK
| | - Joanne L Fothergill
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Ronald Ross Building, 8 West Derby Street, Liverpool, L69 7BE, UK
| | - Daniel R Neill
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Ronald Ross Building, 8 West Derby Street, Liverpool, L69 7BE, UK
| |
Collapse
|