1
|
Kaur M, Mozaheb N, Paiva TO, Herent MF, Goormaghtigh F, Paquot A, Terrasi R, Mignolet E, Décout JL, Lorent JH, Larondelle Y, Muccioli GG, Quetin-Leclercq J, Dufrêne YF, Mingeot-Leclercq MP. Insight into the outer membrane asymmetry of P. aeruginosa and the role of MlaA in modulating the lipidic composition, mechanical, biophysical, and functional membrane properties of the cell envelope. Microbiol Spectr 2024; 12:e0148424. [PMID: 39373473 PMCID: PMC11537012 DOI: 10.1128/spectrum.01484-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 08/14/2024] [Indexed: 10/08/2024] Open
Abstract
In Gram-negative bacteria, the outer membrane (OM) is asymmetric, with lipopolysaccharides (LPS) in the outer leaflet and glycerophospholipids (GPLs) in the inner leaflet. The asymmetry is maintained by the Mla system (MlaA-MlaBCDEF), which contributes to lipid homeostasis by removing mislocalized GPLs from the outer leaflet of the OM. Here, we ascribed how Pseudomonas aeruginosa ATCC 27853 coordinately regulates pathways to provide defense against the threats posed by the deletion of mlaA. Especially, we explored (i) the effects on membrane lipid composition including LPS, GPLs, and lysophospholipids, (ii) the biophysical properties of the OM such as stiffness and fluidity, and (iii) the impact of these changes on permeability, antibiotic susceptibility, and membrane vesicles (MVs) generation. Deletion of mlaA induced an increase in total GPLs and a decrease in LPS level while also triggering alterations in lipid A structures (arabinosylation and palmitoylation), likely to be induced by a two-component system (PhoPQ-PmrAB). Altered lipid composition may serve a physiological purpose in regulating the mechanobiological and functional properties of P. aeruginosa. We demonstrated an increase in cell stiffness without alteration of turgor pressure and inner membrane (IM) fluidity in ∆mlaA. In addition, membrane vesiculation increased without any change in OM/IM permeability. An amphiphilic aminoglycoside derivative (3',6-dinonyl neamine) that targets P. aeruginosa membranes induced an opposite effect on ∆mlaA strain with a trend toward a return to the situation observed for the WT strain. Efforts dedicated to understanding the crosstalk between the OM lipid composition, and the mechanical behavior of bacterial envelope, is one needed step for designing new targets or new drugs to fight P. aeruginosa infections.IMPORTANCEPseudomonas aeruginosa is a Gram-negative bacterium responsible for severe hospital-acquired infections. The outer membrane (OM) of Gram-negative bacteria acts as an effective barrier against toxic compounds, and therefore, compromising this structure could increase sensitivity to antibiotics. The OM is asymmetric with the highly packed lipopolysaccharide monolayer at the outer leaflet and glycerophospholipids at the inner leaflet. OM asymmetry is maintained by the Mla pathway resulting in the retrograde transport of glycerophospholipids from the OM to the inner membrane. In this study, we show that deleting mlaA, the membrane component of Mla system located at the OM, affects the mechanical and functional properties of P. aeruginosa cell envelope. Our results provide insights into the role of MlaA, involved in the Mla transport pathway in P. aeruginosa.
Collapse
Affiliation(s)
- M. Kaur
- UCLouvain, Louvain Drug Research Institute, Cellular & Molecular Pharmacology, Brussels, Belgium
| | - N. Mozaheb
- UCLouvain, Louvain Drug Research Institute, Cellular & Molecular Pharmacology, Brussels, Belgium
| | - T. O. Paiva
- UCLouvain, Louvain Institute of Biomolecular Science and Technology, nanoBiophysics, Louvain-la-Neuve, Belgium
| | - M.-F. Herent
- UCLouvain, Louvain Drug Research Institute, Pharmacognosy, Brussels, Belgium
| | - F. Goormaghtigh
- UCLouvain, Louvain Drug Research Institute, Cellular & Molecular Pharmacology, Brussels, Belgium
| | - A. Paquot
- UCLouvain, Louvain Drug Research Institute, Bioanalysis and Pharmacology of Bioactive Lipids, Brussels, Belgium
| | - R. Terrasi
- UCLouvain, Louvain Drug Research Institute, Bioanalysis and Pharmacology of Bioactive Lipids, Brussels, Belgium
| | - E. Mignolet
- UCLouvain, Louvain Institute of Biomolecular Science and Technology, Biochemistry of Nutrition and Environmental Toxicology Louvain-la-Neuve, Brussels, Belgium
| | - J.-L. Décout
- Université Grenoble Alpes, CNRS, DPM, Grenoble, France
| | - J. H. Lorent
- UCLouvain, Louvain Drug Research Institute, Cellular & Molecular Pharmacology, Brussels, Belgium
| | - Y. Larondelle
- UCLouvain, Louvain Institute of Biomolecular Science and Technology, Biochemistry of Nutrition and Environmental Toxicology Louvain-la-Neuve, Brussels, Belgium
| | - G. G. Muccioli
- UCLouvain, Louvain Drug Research Institute, Bioanalysis and Pharmacology of Bioactive Lipids, Brussels, Belgium
| | - J. Quetin-Leclercq
- UCLouvain, Louvain Drug Research Institute, Pharmacognosy, Brussels, Belgium
| | - Y. F. Dufrêne
- UCLouvain, Louvain Institute of Biomolecular Science and Technology, nanoBiophysics, Louvain-la-Neuve, Belgium
| | - M.-P. Mingeot-Leclercq
- UCLouvain, Louvain Drug Research Institute, Cellular & Molecular Pharmacology, Brussels, Belgium
| |
Collapse
|
2
|
Saiman L, Waters V, LiPuma JJ, Hoffman LR, Alby K, Zhang SX, Yau YC, Downey DG, Sermet-Gaudelus I, Bouchara JP, Kidd TJ, Bell SC, Brown AW. Practical Guidance for Clinical Microbiology Laboratories: Updated guidance for processing respiratory tract samples from people with cystic fibrosis. Clin Microbiol Rev 2024; 37:e0021521. [PMID: 39158301 PMCID: PMC11391703 DOI: 10.1128/cmr.00215-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2024] Open
Abstract
SUMMARYThis guidance presents recommendations for clinical microbiology laboratories for processing respiratory samples from people with cystic fibrosis (pwCF). Appropriate processing of respiratory samples is crucial to detect bacterial and fungal pathogens, guide treatment, monitor the epidemiology of cystic fibrosis (CF) pathogens, and assess therapeutic interventions. Thanks to CF transmembrane conductance regulator modulator therapy, the health of pwCF has improved, but as a result, fewer pwCF spontaneously expectorate sputum. Thus, the collection of sputum samples has decreased, while the collection of other types of respiratory samples such as oropharyngeal and bronchoalveolar lavage samples has increased. To optimize the detection of microorganisms, including Pseudomonas aeruginosa, Staphylococcus aureus, Haemophilus influenzae, and Burkholderia cepacia complex; other less common non-lactose fermenting Gram-negative bacilli, e.g., Stenotrophomonas maltophilia, Inquilinus, Achromobacter, Ralstonia, and Pandoraea species; and yeasts and filamentous fungi, non-selective and selective culture media are recommended for all types of respiratory samples, including samples obtained from pwCF after lung transplantation. There are no consensus recommendations for laboratory practices to detect, characterize, and report small colony variants (SCVs) of S. aureus, although studies are ongoing to address the potential clinical impact of SCVs. Accurate identification of less common Gram-negative bacilli, e.g., S. maltophilia, Inquilinus, Achromobacter, Ralstonia, and Pandoraea species, as well as yeasts and filamentous fungi, is recommended to understand their epidemiology and clinical importance in pwCF. However, conventional biochemical tests and automated platforms may not accurately identify CF pathogens. MALDI-TOF MS provides excellent genus-level identification, but databases may lack representation of CF pathogens to the species-level. Thus, DNA sequence analysis should be routinely available to laboratories for selected clinical circumstances. Antimicrobial susceptibility testing (AST) is not recommended for every routine surveillance culture obtained from pwCF, although selective AST may be helpful, e.g., for unusual pathogens or exacerbations unresponsive to initial therapy. While this guidance reflects current care paradigms for pwCF, recommendations will continue to evolve as CF research expands the evidence base for laboratory practices.
Collapse
Affiliation(s)
- Lisa Saiman
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
- Department of Infection Prevention and Control, NewYork-Presbyterian Hospital, New York, New York, USA
| | - Valerie Waters
- Division of Infectious Diseases, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - John J LiPuma
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Lucas R Hoffman
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Kevin Alby
- Department of Pathology and Laboratory Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Sean X Zhang
- Division of Medical Microbiology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yvonne C Yau
- Division of Microbiology, Department of Paediatric Laboratory Medicine, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Damian G Downey
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University, Belfast, Ireland
| | | | - Jean-Philippe Bouchara
- University of Angers-University of Brest, Infections Respiratoires Fongiques, Angers, France
| | - Timothy J Kidd
- Microbiology Division, Pathology Queensland Central Laboratory, The University of Queensland, Brisbane, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Scott C Bell
- The Prince Charles Hospital, Faculty of Medicine, The University of Queensland, Brisbane, Australia
- The Translational Research Institute, Brisbane, Australia
| | - A Whitney Brown
- Cystic Fibrosis Foundation, Bethesda, Maryland, USA
- Inova Advanced Lung Disease and Transplant Program, Inova Fairfax Hospital, Falls Church, Virginia, USA
| |
Collapse
|
3
|
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
|
4
|
Valzano F, Boncompagni SR, Micieli M, Di Maggio T, Di Pilato V, Colombini L, Santoro F, Pozzi G, Rossolini GM, Pallecchi L. Activity of N-Acetylcysteine Alone and in Combination with Colistin against Pseudomonas aeruginosa Biofilms and Transcriptomic Response to N-Acetylcysteine Exposure. Microbiol Spectr 2022; 10:e0100622. [PMID: 35735984 PMCID: PMC9431628 DOI: 10.1128/spectrum.01006-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 06/03/2022] [Indexed: 11/25/2022] Open
Abstract
Chronic colonization by Pseudomonas aeruginosa is critical in cystic fibrosis (CF) and other chronic lung diseases, contributing to disease progression. Biofilm growth and a propensity to evolve multidrug resistance phenotypes drastically limit the available therapeutic options. In this perspective, there has been growing interest in evaluating combination therapies, especially for drugs that can be administered by nebulization, which allows high drug concentrations to be reached at the site of infections while limiting systemic toxicity. Here, we investigated the potential antibiofilm activity of N-acetylcysteine (NAC) alone and in combination with colistin against a panel of P. aeruginosa strains (most of which are from CF patients) and the transcriptomic response of a P. aeruginosa CF strain to NAC exposure. NAC alone (8,000 mg/L) showed a limited and strain-dependent antibiofilm activity. Nonetheless, a relevant antibiofilm synergism of NAC-colistin combinations (NAC at 8,000 mg/L plus colistin at 2 to 32 mg/L) was observed with all strains. Synergism was also confirmed with the artificial sputum medium model. RNA sequencing of NAC-exposed planktonic cultures revealed that NAC (8,000 mg/L) mainly induced (i) a Zn2+ starvation response (known to induce attenuation of P. aeruginosa virulence), (ii) downregulation of genes of the denitrification apparatus, and (iii) downregulation of flagellar biosynthesis pathway. NAC-mediated inhibition of P. aeruginosa denitrification pathway and flagellum-mediated motility were confirmed experimentally. These findings suggested that NAC-colistin combinations might contribute to the management of biofilm-associated P. aeruginosa lung infections. NAC might also have a role in reducing P. aeruginosa virulence, which could be relevant in the very early stages of lung colonization. IMPORTANCE Pseudomonas aeruginosa biofilm-related chronic lung colonization contributes to cystic fibrosis (CF) disease progression. Colistin is often a last-resort antibiotic for the treatment of such P. aeruginosa infections, and it has been increasingly used in CF, especially by nebulization. N-acetylcysteine (NAC) is a mucolytic agent with antioxidant activity, commonly administered with antibiotics for the treatment of lower respiratory tract infections. Here, we show that NAC potentiated colistin activity against in vitro biofilms models of P. aeruginosa strains, with both drugs tested at the high concentrations achievable after nebulization. In addition, we report the first transcriptomic data on the P. aeruginosa response to NAC exposure.
Collapse
Affiliation(s)
- Felice Valzano
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | | | - Maria Micieli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Tiziana Di Maggio
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Vincenzo Di Pilato
- Department of Surgical Sciences and Integrated Diagnostics, University of Genoa, Genoa, Italy
| | - Lorenzo Colombini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Francesco Santoro
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Gianni Pozzi
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Gian Maria Rossolini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Clinical Microbiology and Virology Unit, Careggi University Hospital, Florence, Italy
| | - Lucia Pallecchi
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| |
Collapse
|
5
|
Shaban TF, Alkawareek MY. Prediction of qualitative antibiofilm activity of antibiotics using supervised machine learning techniques. Comput Biol Med 2022; 140:105065. [PMID: 34839184 DOI: 10.1016/j.compbiomed.2021.105065] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 11/21/2021] [Accepted: 11/21/2021] [Indexed: 11/18/2022]
Abstract
Although biofilm-specific antibiotic susceptibility assays are available, they are time-consuming and resource-intensive, and hence they are not usually performed in clinical settings. Herein, we introduce a machine learning-based predictive modeling approach that uses routinely available and easily accessible data to qualitatively predict in vitro antibiofilm activity of antibiotics with relatively high accuracy. Three optimized models based on logistic regression, decision tree, and random forest algorithms were successfully developed in this study using data manually collected from published literature. In these models, independent variables that serve as significant predictors of antibiofilm activity are minimum inhibitory concentration, bacterial Gram type, biofilm formation method, in addition to antibiotic's mechanism of action, molecular weight, and pKa. The cross-validation method showed that the optimized models exhibit prediction accuracy of 67% ± 6.1% for the logistic regression model, 73% ± 5.8% for the decision tree model, and 74% ± 5% for the random forest model. However, the one-way ANOVA test revealed that the difference in prediction accuracy between the 3 models is not statistically significant, and hence they can be considered to have comparable performance. The presented modeling approach can serve as an alternative to the resource-intensive biofilm assays to rapidly and properly manage biofilm-associated infections, especially in resource-limited clinical settings.
Collapse
Affiliation(s)
- Taqwa F Shaban
- School of Pharmacy, The University of Jordan, Amman, Jordan
| | | |
Collapse
|
6
|
Van den Bossche S, De Broe E, Coenye T, Van Braeckel E, Crabbé A. The cystic fibrosis lung microenvironment alters antibiotic activity: causes and effects. Eur Respir Rev 2021; 30:30/161/210055. [PMID: 34526313 DOI: 10.1183/16000617.0055-2021] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/23/2021] [Indexed: 01/08/2023] Open
Abstract
Chronic airway colonisation by Pseudomonas aeruginosa, a hallmark of cystic fibrosis (CF) lung disease, is associated with increased morbidity and mortality and despite aggressive antibiotic treatment, P. aeruginosa is able to persist in CF airways. In vitro antibiotic susceptibility assays are poor predictors of antibiotic efficacy to treat respiratory tract infections in the CF patient population and the selection of the antibiotic(s) is often made on an empirical base. In the current review, we discuss the factors that are responsible for the discrepancies between antibiotic activity in vitro and clinical efficacy in vivo We describe how the CF lung microenvironment, shaped by host factors (such as iron, mucus, immune mediators and oxygen availability) and the microbiota, influences antibiotic activity and varies widely between patients. A better understanding of the CF microenvironment and population diversity may thus help improve in vitro antibiotic susceptibility testing and clinical decision making, in turn increasing the success rate of antibiotic treatment.
Collapse
Affiliation(s)
| | - Emma De Broe
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | - Tom Coenye
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | - Eva Van Braeckel
- Dept of Respiratory Medicine, Cystic Fibrosis Reference Centre, Ghent University Hospital, Ghent, Belgium.,Dept of Internal Medicine and Paediatrics, Ghent University, Ghent, Belgium
| | - Aurélie Crabbé
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| |
Collapse
|
7
|
Di Bonaventura G, Pompilio A. In Vitro Antimicrobial Susceptibility Testing of Biofilm-Growing Bacteria: Current and Emerging Methods. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1369:33-51. [PMID: 33963526 DOI: 10.1007/5584_2021_641] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The antibiotic susceptibility of bacterial pathogens is typically determined based on planktonic cells, as recommended by several international guidelines. However, most of chronic infections - such as those established in wounds, cystic fibrosis lung, and onto indwelling devices - are associated to the formation of biofilms, communities of clustered bacteria attached onto a surface, abiotic or biotic, and embedded in an extracellular matrix produced by the bacteria and complexed with molecules from the host. Sessile microorganisms show significantly increased tolerance/resistance to antibiotics compared with planktonic counterparts. Consequently, antibiotic concentrations used in standard antimicrobial susceptibility tests, although effective against planktonic bacteria in vitro, are not predictive of the concentrations required to eradicate biofilm-related infections, thus leading to treatment failure, chronicization and removal of material in patients with indwelling medical devices.Meeting the need for the in vitro evaluation of biofilm susceptibility to antibiotics, here we reviewed several methods proposed in literature highlighting their advantages and limitations to guide scientists towards an appropriate choice.
Collapse
Affiliation(s)
- Giovanni Di Bonaventura
- Department of Medical, Oral and Biotechnological Sciences, and Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy. .,Laboratory of Clinical Microbiology, Chieti, Italy.
| | - Arianna Pompilio
- Department of Medical, Oral and Biotechnological Sciences, and Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy.,Laboratory of Clinical Microbiology, Chieti, Italy
| |
Collapse
|
8
|
Akkerman-Nijland AM, Akkerman OW, Grasmeijer F, Hagedoorn P, Frijlink HW, Rottier BL, Koppelman GH, Touw DJ. The pharmacokinetics of antibiotics in cystic fibrosis. Expert Opin Drug Metab Toxicol 2020; 17:53-68. [PMID: 33213220 DOI: 10.1080/17425255.2021.1836157] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Dosing of antibiotics in people with cystic fibrosis (CF) is challenging, due to altered pharmacokinetics, difficulty of lung tissue penetration, and increasing presence of antimicrobial resistance. AREAS COVERED The purpose of this work is to critically review original data as well as previous reviews and guidelines on pharmacokinetics of systemic and inhaled antibiotics in CF, with the aim to propose strategies for optimization of antibacterial therapy in both children and adults with CF. EXPERT OPINION For systemic antibiotics, absorption is comparable in CF patients and non-CF controls. The volume of distribution (Vd) of most antibiotics is similar between people with CF with normal body composition and healthy individuals. However, there are a few exceptions, like cefotiam and tobramycin. Many antibiotic class-dependent changes in drug metabolism and excretion are reported, with an increased total body clearance for ß-lactam antibiotics, aminoglycosides, fluoroquinolones, and trimethoprim. We, therefore, recommend following class-specific guidelines for CF, mostly resulting in higher dosages per kg bodyweight in CF compared to non-CF controls. Higher local antibiotic concentrations in the airways can be obtained by inhalation therapy, with which eradication of bacteria may be achieved while minimizing systemic exposure and risk of toxicity.
Collapse
Affiliation(s)
- Anne M Akkerman-Nijland
- Department of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen , Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen , Groningen, The Netherlands
| | - Onno W Akkerman
- Department of Pulmonary Diseases and Tuberculosis, University of Groningen, University Medical Center Groningen , Groningen, The Netherlands
| | - Floris Grasmeijer
- Department of Pharmacy, PureIMS B.V , Roden, The Netherlands.,Department of Pharmaceutical Technology and Biopharmacy, University of Groningen , Groningen, The Netherlands
| | - Paul Hagedoorn
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen , Groningen, The Netherlands
| | - Henderik W Frijlink
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen , Groningen, The Netherlands
| | - Bart L Rottier
- Department of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen , Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen , Groningen, The Netherlands
| | - Gerard H Koppelman
- Department of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen , Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen , Groningen, The Netherlands
| | - Daniel J Touw
- Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen , Groningen, The Netherlands.,Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen , Groningen, The Netherlands
| |
Collapse
|
9
|
Activity of Antibiotics against Pseudomonas aeruginosa in an In Vitro Model of Biofilms in the Context of Cystic Fibrosis: Influence of the Culture Medium. Antimicrob Agents Chemother 2020; 64:AAC.02204-19. [PMID: 32015047 DOI: 10.1128/aac.02204-19] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 01/24/2020] [Indexed: 12/28/2022] Open
Abstract
Pseudomonas aeruginosa is a major cause of respiratory biofilm-related infections in patients with cystic fibrosis. We developed an in vitro pharmacodynamic model to study the activity of antipseudomonal antibiotics against PAO1 biofilms grown in artificial sputum medium with agar [ASM(+)] versus that against biofilms grown in Trypticase soy broth supplemented with glucose and NaCl (TGN). We measured bacterial counts, metabolic activity (fluorescein diacetate [FDA] hydrolysis), and biomass (crystal violet absorbance). Biofilms grew slower in ASM(+) than in TGN but reached the same CFU counts and metabolic activity in both media and a slightly higher biomass after 48 h in ASM(+) than in TGN. The concentration-response curves of the antibiotics after 24 h of incubation with mature biofilms showed maximal effects ranging from a 3 (ciprofloxacin)- to a 1.5 (ceftazidime, meropenem)-log10-CFU decrease, with tobramycin and colistin showing intermediate values. These maximal reductions in the numbers of CFU were similar in both media for ciprofloxacin and β-lactams but lower in ASM(+) than in TGN for tobramycin and colistin; they were reached at concentrations lower than the human maximum concentration in plasma for ciprofloxacin and β-lactams only. The reductions in metabolic activity and in biomass were low in both media. Small-colony variants were selected by tobramycin in ASM(+) and by ciprofloxacin in both media. The model was then successfully applied to 4 isolates from patients with cystic fibrosis. These biofilms showed CFU counts similar to those of PAO1 biofilms in ASM(+) but a higher biomass than PAO1 biofilms in ASM(+) and moderate differences in their susceptibility to antibiotics from that of PAO1 biofilms grown in this medium. This model proved useful to establish the pharmacodynamic profile of drugs against P. aeruginosa biofilms in the context of cystic fibrosis.
Collapse
|
10
|
Hengzhuang W, Green K, Pressler T, Skov M, Katzenstein TL, Wu X, Høiby N. Optimization of colistin dosing regimen for cystic fibrosis patients with chronic Pseudomonas aeruginosa biofilm lung infections. Pediatr Pulmonol 2019; 54:575-580. [PMID: 30803159 DOI: 10.1002/ppul.24269] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 07/17/2018] [Indexed: 11/08/2022]
Abstract
OBJECTIVE The present study was performed to explore dosing regimens of colistin in patients of cystic fibrosis (CF) with Pseudomonas aeruginosa chronic biofilm lung infection. METHODS Ten CF patients were involved. One dose colistimethate sodium (CMS) of 6 MIU (million international units) and 9 MIU were administered by intravenous infusion over 45 and 90 min. Venous blood was collected at different time points after the infusion of CMS. Pharmacokinetic parameters of colistin were calculated. Minimum inhibitory concentration for planktonic P. aeruginosa, minimum biofilm inhibitory concentration and minimum biofilm eradication concentration of P. aeruginosa were determined. Monte Carlo simulation was performed to determine the clinical probability of target attainment of different dosing regimens of colistin in CF patients. RESULTS For 90 min (6 MIU), 45 min (6 MIU), and 45 min (9 MIU) intravenous infusion of colistin, Cmax was 8.9 ± 1.8, 15 ± 5.5, and 31.7 ± 5.3 μg/mL, respectively; Tmax was 1.2 ± 0.4, 0.7 ± 0.2, and 0.8 ± 0.2 h, respectively; AUCtot were 31 ± 3.8, 34 ± 10, and 135 ± 31mg · h/L, respectively; t1/2 was 2.1 ± 0.4, 2 ± 0.3, and 3.3 ± 0.4 h, respectively. MBIC and MBEC of colistin on biofilms at 24 h period treatment were 16-128 μg/mL for non-mucoid and mucoid biofilms of P. aeruginosa. For 90 min (6 MIU), 45 min (6 MIU) and 45 min iv infusion (9 MIU) with one dose colistin, PTA was 49.8%, 53.8%, 99.4% for planktonic infection, and 11.3%, 14.6%, 65.3%, respectively for biofilm infection. CONCLUSIONS colistin treatment using 45 min iv infusion is better than 90 min iv infusion in this study. Colistin dosage of 9 MIU is better than 6 MIU on both planktonic and biofilm infections of P. aeruginosa in this study.
Collapse
Affiliation(s)
- Wang Hengzhuang
- Department of Clinical Microbiology, Rigshospitalet, University of Copenhagen, Denmark.,Department of Immunology and Microbiology, Costerton Biofilm Center, University of Copenhagen, Denmark
| | - Kent Green
- The Cystic Fibrosis Center, Rigshospitalet, University of Copenhagen, Denmark
| | - Tacjana Pressler
- The Cystic Fibrosis Center, Rigshospitalet, University of Copenhagen, Denmark
| | - Marianne Skov
- The Cystic Fibrosis Center, Rigshospitalet, University of Copenhagen, Denmark
| | - Terese L Katzenstein
- The Cystic Fibrosis Center, Rigshospitalet, University of Copenhagen, Denmark.,Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Xiaojie Wu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China
| | - Niels Høiby
- Department of Clinical Microbiology, Rigshospitalet, University of Copenhagen, Denmark.,Department of Immunology and Microbiology, Costerton Biofilm Center, University of Copenhagen, Denmark
| |
Collapse
|
11
|
Grassi L, Batoni G, Ostyn L, Rigole P, Van den Bossche S, Rinaldi AC, Maisetta G, Esin S, Coenye T, Crabbé A. The Antimicrobial Peptide lin-SB056-1 and Its Dendrimeric Derivative Prevent Pseudomonas aeruginosa Biofilm Formation in Physiologically Relevant Models of Chronic Infections. Front Microbiol 2019; 10:198. [PMID: 30800115 PMCID: PMC6376900 DOI: 10.3389/fmicb.2019.00198] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 01/24/2019] [Indexed: 12/11/2022] Open
Abstract
Antimicrobial peptides (AMPs) are promising templates for the development of novel antibiofilm drugs. Despite the large number of studies on screening and optimization of AMPs, only a few of these evaluated the antibiofilm activity in physiologically relevant model systems. Potent in vitro activity of AMPs often does not translate into in vivo effectiveness due to the interference of the host microenvironment with peptide stability/availability. Hence, mimicking the complex environment found in biofilm-associated infections is essential to predict the clinical potential of novel AMP-based antimicrobials. In the present study, we examined the antibiofilm activity of the semi-synthetic peptide lin-SB056-1 and its dendrimeric derivative (lin-SB056-1)2-K against Pseudomonas aeruginosa in an in vivo-like three-dimensional (3-D) lung epithelial cell model and an in vitro wound model (consisting of an artificial dermis and blood components at physiological levels). Although moderately active when tested alone, lin-SB056-1 was effective in reducing P. aeruginosa biofilm formation in association with 3-D lung epithelial cells in combination with the chelating agent EDTA. The dimeric derivative (lin-SB056-1)2-K demonstrated an enhanced biofilm-inhibitory activity as compared to both lin-SB056-1 and the lin-SB056-1/EDTA combination, reducing the number of biofilm-associated bacteria up to 3-Log units at concentrations causing less than 20% cell death. Biofilm inhibition by (lin-SB056-1)2-K was reported both for the reference strain PAO1 and cystic fibrosis lung isolates of P. aeruginosa. In addition, using fluorescence microscopy, a significant decrease in biofilm-like structures associated with 3-D cells was observed after peptide exposure. Interestingly, effectiveness of (lin-SB056-1)2-K was also demonstrated in the wound model with a reduction of up to 1-Log unit in biofilm formation by P. aeruginosa PAO1 and wound isolates. Overall, combination treatment and peptide dendrimerization emerged as promising strategies to improve the efficacy of AMPs, especially under challenging host-mimicking conditions. Furthermore, the results of the present study underlined the importance of evaluating the biological properties of novel AMPs in in vivo-like model systems representative of specific infectious sites in order to make a more realistic prediction of their therapeutic success, and avoid the inclusion of unpromising peptides in animal studies and clinical trials.
Collapse
Affiliation(s)
- Lucia Grassi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Giovanna Batoni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Lisa Ostyn
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | - Petra Rigole
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | | | - Andrea C Rinaldi
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Giuseppantonio Maisetta
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Semih Esin
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Tom Coenye
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | - Aurélie Crabbé
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| |
Collapse
|
12
|
Pompilio A, Geminiani C, Bosco D, Rana R, Aceto A, Bucciarelli T, Scotti L, Di Bonaventura G. Electrochemically Synthesized Silver Nanoparticles Are Active Against Planktonic and Biofilm Cells of Pseudomonas aeruginosa and Other Cystic Fibrosis-Associated Bacterial Pathogens. Front Microbiol 2018; 9:1349. [PMID: 30026732 PMCID: PMC6041389 DOI: 10.3389/fmicb.2018.01349] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 06/04/2018] [Indexed: 02/05/2023] Open
Abstract
A novel, electrochemically synthesized, silver nanoparticles (AgNPs) formulation was evaluated in vitro against Pseudomonas aeruginosa, Burkholderia cepacia, Stenotrophomonas maltophilia, and Staphylococcus aureus strains from cystic fibrosis (CF) patients. AgNPs were particularly active against P. aeruginosa and B. cepacia planktonic cells (median MIC: 1.06 and 2.12 μg/ml, respectively) by a rapid, bactericidal and concentration-dependent effect. AgNPs showed to be particularly effective against P. aeruginosa and S. aureus biofilm causing a viability reduction ranging from 50% (1×MIC) to >99.9% (4×MIC). Electron microscopy showed that AgNPs deconstruct extracellular matrix of P. aeruginosa biofilm, and accumulate at the cell surface causing cell death secondary to membrane damage. Compared to Tobramycin, AgNPs showed comparable, or even better, activity against planktonic and biofilm P. aeruginosa cells. AgNPs at concentrations effective against B. cepacia and P. aeruginosa were not toxic to G. mellonella larvae. Our silver-based formulation might be an alternative to antibiotics in CF patients. Further in vitro and in vivo studies are warranted to confirm this therapeutic potential.
Collapse
Affiliation(s)
- Arianna Pompilio
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy.,Center of Excellence on Aging and Translational Medicine (CeSI-MeT), Chieti, Italy
| | - Cristina Geminiani
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy.,Center of Excellence on Aging and Translational Medicine (CeSI-MeT), Chieti, Italy
| | - Domenico Bosco
- Department of Biomorphological Science, Molecular Genetic Institute, Italian National Research Council, Chieti, Italy
| | - Rosalba Rana
- Department of Medicine and Science of Aging, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Antonio Aceto
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Tonino Bucciarelli
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Luca Scotti
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Giovanni Di Bonaventura
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy.,Center of Excellence on Aging and Translational Medicine (CeSI-MeT), Chieti, Italy
| |
Collapse
|
13
|
Antimicrobial molecules in the lung: formulation challenges and future directions for innovation. Future Med Chem 2018; 10:575-604. [PMID: 29473765 DOI: 10.4155/fmc-2017-0162] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Inhaled antimicrobials have been extremely beneficial in treating respiratory infections, particularly chronic infections in a lung with cystic fibrosis. The pulmonary delivery of antibiotics has been demonstrated to improve treatment efficacy, reduce systemic side effects and, critically, reduce drug exposure to commensal bacteria compared with systemic administration, reducing selective pressure for antimicrobial resistance. This review will explore the specific challenges of pulmonary delivery of a number of differing antimicrobial molecules, and the formulation and technological approaches that have been used to overcome these difficulties. It will also explore the future challenges being faced in the development of inhaled products and respiratory infection treatment, and identify future directions of innovation, with a particular focus on respiratory infections caused by multiple drug-resistant pathogens.
Collapse
|
14
|
Kolouchová I, Maťátková O, Paldrychová M, Kodeš Z, Kvasničková E, Sigler K, Čejková A, Šmidrkal J, Demnerová K, Masák J. Resveratrol, pterostilbene, and baicalein: plant-derived anti-biofilm agents. Folia Microbiol (Praha) 2017; 63:261-272. [DOI: 10.1007/s12223-017-0549-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 09/11/2017] [Indexed: 01/09/2023]
|
15
|
Tangso KJ, C D da Cunha PH, Spicer P, Li J, Boyd BJ. Antimicrobial Activity from Colistin-Heparin Lamellar-Phase Complexes for the Coating of Biomedical Devices. ACS APPLIED MATERIALS & INTERFACES 2016; 8:31321-31329. [PMID: 27750410 DOI: 10.1021/acsami.6b10027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Infections arising in hospitalized patients, particularly those who have undergone surgery and are reliant on receiving treatment through biomedical devices, continue to be a rising concern. It is well-known that aqueous mixtures of oppositely charged surfactant and polymer molecules can self-assemble to form liquid crystalline structures, primarily via electrostatically driven interactions that have demonstrated great potential as tailored-release nanomaterials. Colistin is a re-emerging antibiotic used against multidrug-resistant Gram-negative bacteria. Its amphiphilic structure allows it to form micellar aggregates in solution. Thus, the aim of this study was to determine whether structured complexes form between colistin and negatively charged biopolymers, such as the highly sulfated anticoagulant, heparin. Cross-polarized light microscopy and synchrotron small-angle X-ray scattering were employed to visualize the appearance of birefringent structures and identify liquid crystalline structures, respectively, formed across the interface between solutions of colistin and heparin. A lamellar phase with a lattice parameter of ∼40 Å was formed upon contact between the oppositely charged solutions of colistin and heparin. In addition, in vitro release studies showed a slow release of colistin from the lamellar-phase gel complexes into the bulk media, and disk diffusion bioassays revealed antimicrobial activity against Pseudomonas aeruginosa. This system provides a novel, cost-effective, and simple approach to reducing the risk of infections by potentially applying the formulation as a coating for biomedical implants or tubing.
Collapse
Affiliation(s)
| | - Paulo Henrique C D da Cunha
- Universidade Estadual de Londrina , Rodovia Celso Garcia Cid, Pr 455 Km 380, Campus Universitário, Londrina, Paraná, Brazil
| | - Patrick Spicer
- School of Chemical Engineering, University of New South Wales , Sydney, NSW 2052, Australia
| | | | | |
Collapse
|
16
|
Bos AC, Passé KM, Mouton JW, Janssens HM, Tiddens HAWM. The fate of inhaled antibiotics after deposition in cystic fibrosis: How to get drug to the bug? J Cyst Fibros 2016; 16:13-23. [PMID: 28254026 DOI: 10.1016/j.jcf.2016.10.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 09/26/2016] [Accepted: 10/01/2016] [Indexed: 10/20/2022]
Abstract
BACKGROUND Chronic airway infections in patients with cystic fibrosis (CF) are most often treated with inhaled antibiotics of which deposition patterns have been extensively studied. However, the journey of aerosol particles does not end after deposition within the bronchial tree. OBJECTIVES To review how local conditions affect the clinical efficacy of antibiotic aerosol particles after deposition in the airways of patients with CF. METHODS Electronic databases were searched from inception to September 2015. Original studies describing the effect of CF sputum or bacterial factors on antibiotic efficacy and formulations to increase efficacy were included. RESULTS 35 articles were included which mostly described in vitro studies and mainly investigated aminoglycosides. After deposition, diffusion through the mucus layer was reduced for aminoglycosides, β-lactam antibiotics and fluoroquinolones. Within CF mucus, low oxygen tension adversely affected aminoglycosides, β-lactam antibiotics, and chloramphenicol; and molecules inactivated aminoglycosides but not β-lactam antibiotics. Finally, the alginate layer surrounding Pseudomonas aeruginosa was an important factor in the resistance against all antibiotics. CONCLUSIONS After deposition in the airways, the local efficacy of inhaled antibiotics can be reduced by molecules within CF mucus and the alginate layer surrounding P. aeruginosa.
Collapse
Affiliation(s)
- Aukje C Bos
- Department of Paediatric Pulmonology and Allergology, Erasmus Medical Centre (MC) - Sophia Children's Hospital, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands.
| | - Kimberly M Passé
- Department of Paediatric Pulmonology and Allergology, Erasmus Medical Centre (MC) - Sophia Children's Hospital, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands.
| | - Johan W Mouton
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands.
| | - Hettie M Janssens
- Department of Paediatric Pulmonology and Allergology, Erasmus Medical Centre (MC) - Sophia Children's Hospital, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands.
| | - Harm A W M Tiddens
- Department of Paediatric Pulmonology and Allergology, Erasmus Medical Centre (MC) - Sophia Children's Hospital, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands.
| |
Collapse
|
17
|
Pompilio A, Crocetta V, Verginelli F, Di Bonaventura G. In vitro activity of levofloxacin against planktonic and biofilm Stenotrophomonas maltophilia lifestyles under conditions relevant to pulmonary infection in cystic fibrosis, and relationship with SmeDEF multidrug efflux pump expression. FEMS Microbiol Lett 2016; 363:fnw145. [PMID: 27242375 DOI: 10.1093/femsle/fnw145] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/26/2016] [Indexed: 11/14/2022] Open
Abstract
The activity of levofloxacin against planktonic and biofilm Stenotrophomonas maltophilia cells and the role played by the multidrug efflux pump SmeDEF were evaluated under conditions relevant to the cystic fibrosis (CF) lung. MIC, MBC and MBEC of levofloxacin were assessed, against five CF strains, under 'standard' (CLSI-recommended) and 'CF-like' (pH 6.8, 5% CO2, in a synthetic CF sputum) conditions. Levofloxacin was tested against biofilms at concentrations (10, 50 and 100 μg mL(-1)) corresponding to achievable serum levels and sputum levels by aerosolisation. smeD expression was evaluated, under both conditions, in planktonic and biofilm cells by RT-PCR. The bactericidal effect of levofloxacin was decreased, in three out of five strains tested, under 'CF-like' conditions (MBC: 2-4 vs 8-16 μg mL(-1), under 'standard' and 'CF-like' conditions, respectively). Biofilm was intrinsically resistant to levofloxacin, regardless of conditions tested (MBECs ≥ 100 μg mL(-1) for all strains). Only under 'CF-like' conditions, smeD expression increased during planktonic-to-biofilm transition, and in biofilm cells compared to stationary planktonic cells. Our findings confirmed that S. maltophilia biofilm is intrinsically resistant to therapeutic concentrations of levofloxacin. Under conditions relevant to CF, smeD overexpression could contribute to levofloxacin resistance. Further studies are warranted to define the clinical relevance of our findings.
Collapse
Affiliation(s)
- Arianna Pompilio
- Department of Medical, Oral and Biotechnological Sciences, 'G. d'Annunzio' University of Chieti-Pescara, Chieti, Via dei Vestini 31, 66100 Italy Laboratory of Clinical Microbiology, Center of Excellence on Aging and Translational Medicine (CeSI-MeT), 'G. d'Annunzio' University of Chieti-Pescara, Via Luigi Polacchi, 66100 Chieti, Italy
| | - Valentina Crocetta
- Department of Medical, Oral and Biotechnological Sciences, 'G. d'Annunzio' University of Chieti-Pescara, Chieti, Via dei Vestini 31, 66100 Italy Laboratory of Clinical Microbiology, Center of Excellence on Aging and Translational Medicine (CeSI-MeT), 'G. d'Annunzio' University of Chieti-Pescara, Via Luigi Polacchi, 66100 Chieti, Italy
| | - Fabio Verginelli
- Laboratory of Clinical Microbiology, Center of Excellence on Aging and Translational Medicine (CeSI-MeT), 'G. d'Annunzio' University of Chieti-Pescara, Via Luigi Polacchi, 66100 Chieti, Italy Department of Pharmacy, 'G. d'Annunzio' University of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| | - Giovanni Di Bonaventura
- Department of Medical, Oral and Biotechnological Sciences, 'G. d'Annunzio' University of Chieti-Pescara, Chieti, Via dei Vestini 31, 66100 Italy Laboratory of Clinical Microbiology, Center of Excellence on Aging and Translational Medicine (CeSI-MeT), 'G. d'Annunzio' University of Chieti-Pescara, Via Luigi Polacchi, 66100 Chieti, Italy
| |
Collapse
|
18
|
A Novel Computerized Cell Count Algorithm for Biofilm Analysis. PLoS One 2016; 11:e0154937. [PMID: 27149069 PMCID: PMC4858220 DOI: 10.1371/journal.pone.0154937] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 04/21/2016] [Indexed: 01/01/2023] Open
Abstract
Biofilms are the preferred sessile and matrix-embedded life form of most microorganisms on surfaces. In the medical field, biofilms are a frequent cause of treatment failure because they protect the bacteria from antibiotics and immune cells. Antibiotics are selected according to the minimal inhibitory concentration (MIC) based on the planktonic form of bacteria. Determination of the minimal biofilm eradicating concentration (MBEC), which can be up to 1,000-fold greater than the MIC, is not currently conducted as routine diagnostic testing, primarily because of the methodical hurdles of available biofilm assessing protocols that are time- and cost-consuming. Comparative analysis of biofilms is also limited as most quantitative methods such as crystal violet staining are indirect and highly imprecise. In this paper, we present a novel algorithm for assessing biofilm resistance to antibiotics that overcomes several of the limitations of alternative methods. This algorithm aims for a computer-based analysis of confocal microscope 3D images of biofilms after live/dead stains providing various biofilm parameters such as numbers of viable and dead cells and their vertical distributions within the biofilm, or biofilm thickness. The performance of this algorithm was evaluated using computer-simulated 2D and 3D images of coccal and rodent cells varying different parameters such as cell density, shading or cell size. Finally, genuine biofilms that were untreated or treated with nitroxoline or colistin were analyzed and the results were compared with quantitative microbiological standard methods. This novel algorithm allows a direct, fast and reproducible analysis of biofilms after live/dead staining. It performed well in biofilms of moderate cell densities in a 2D set-up however the 3D analysis remains still imperfect and difficult to evaluate. Nevertheless, this is a first try to develop an easy but conclusive tool that eventually might be implemented into routine diagnostics to determine the MBEC and to improve outcomes of patients with biofilm-associated infections.
Collapse
|
19
|
Kolpen M, Appeldorff CF, Brandt S, Mousavi N, Kragh KN, Aydogan S, Uppal HA, Bjarnsholt T, Ciofu O, Høiby N, Jensen PØ. Increased bactericidal activity of colistin on Pseudomonas aeruginosa biofilms in anaerobic conditions. Pathog Dis 2015; 74:ftv086. [PMID: 26458402 PMCID: PMC4655427 DOI: 10.1093/femspd/ftv086] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2015] [Indexed: 12/19/2022] Open
Abstract
Tolerance towards antibiotics of Pseudomonas aeruginosa biofilms is recognized as a major cause of therapeutic failure of chronic lung infection in cystic fibrosis (CF) patients. This lung infection is characterized by antibiotic-tolerant biofilms in mucus with zones of O2 depletion mainly due to polymorphonuclear leukocytic activity. In contrast to the main types of bactericidal antibiotics, it has not been possible to establish an association between the bactericidal effects of colistin and the production of detectable levels of OH ˙ on several strains of planktonic P. aeruginosa. Therefore, we propose that production of OH ˙ may not contribute significantly to the bactericidal activity of colistin on P. aeruginosa biofilm. Thus, we investigated the effect of colistin treatment on biofilm of wild-type PAO1, a catalase-deficient mutant (ΔkatA) and a colistin-resistant CF isolate cultured in microtiter plates in normoxic- or anoxic atmosphere with 1 mM nitrate. The killing of bacteria during colistin treatment was measured by CFU counts, and the OH⋅ formation was measured by 3(')-(p-hydroxylphenyl fluorescein) fluorescein (HPF) fluorescence. Validation of the assay was done by hydrogen peroxide treatment. OH⋅ formation was undetectable in aerobic PAO1 biofilms during 3 h of colistin treatment. Interestingly, we demonstrate increased susceptibility of P. aeruginosa biofilms towards colistin during anaerobic conditions. In fact, the maximum enhancement of killing by anaerobic conditions exceeded 2 logs using 4 mg L(-1) of colistin compared to killing at aerobic conditions.
Collapse
Affiliation(s)
- Mette Kolpen
- Department of Clinical Microbiology, Rigshospitalet, 2100 Copenhagen, Denmark Department of Immunology and Microbiology, UC-CARE, Faculty of Health Sciences University of Copenhagen, 2200 Copenhagen, Denmark
| | | | - Sarah Brandt
- Department of Clinical Microbiology, Rigshospitalet, 2100 Copenhagen, Denmark
| | - Nabi Mousavi
- Department of Clinical Microbiology, Rigshospitalet, 2100 Copenhagen, Denmark
| | - Kasper N Kragh
- Department of Immunology and Microbiology, UC-CARE, Faculty of Health Sciences University of Copenhagen, 2200 Copenhagen, Denmark
| | - Sevtap Aydogan
- Department of Clinical Microbiology, Rigshospitalet, 2100 Copenhagen, Denmark
| | - Haleema A Uppal
- Department of Clinical Microbiology, Rigshospitalet, 2100 Copenhagen, Denmark
| | - Thomas Bjarnsholt
- Department of Clinical Microbiology, Rigshospitalet, 2100 Copenhagen, Denmark Department of Immunology and Microbiology, UC-CARE, Faculty of Health Sciences University of Copenhagen, 2200 Copenhagen, Denmark
| | - Oana Ciofu
- Department of Immunology and Microbiology, UC-CARE, Faculty of Health Sciences University of Copenhagen, 2200 Copenhagen, Denmark
| | - Niels Høiby
- Department of Clinical Microbiology, Rigshospitalet, 2100 Copenhagen, Denmark Department of Immunology and Microbiology, UC-CARE, Faculty of Health Sciences University of Copenhagen, 2200 Copenhagen, Denmark
| | - Peter Ø Jensen
- Department of Clinical Microbiology, Rigshospitalet, 2100 Copenhagen, Denmark
| |
Collapse
|