1
|
Yu H, Xu Y, Imani S, Zhao Z, Ullah S, Wang Q. Navigating ESKAPE Pathogens: Considerations and Caveats for Animal Infection Models Development. ACS Infect Dis 2024; 10:2336-2355. [PMID: 38866389 PMCID: PMC11249778 DOI: 10.1021/acsinfecdis.4c00007] [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: 01/04/2024] [Revised: 05/19/2024] [Accepted: 05/29/2024] [Indexed: 06/14/2024]
Abstract
The misuse of antibiotics has led to the global spread of drug-resistant bacteria, especially multi-drug-resistant (MDR) ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species). These opportunistic bacteria pose a significant threat, in particular within hospitals, where they cause nosocomial infections, leading to substantial morbidity and mortality. To comprehensively explore ESKAPE pathogenesis, virulence, host immune response, diagnostics, and therapeutics, researchers increasingly rely on necessitate suitable animal infection models. However, no single model can fully replicate all aspects of infectious diseases. Notably when studying opportunistic pathogens in immunocompetent hosts, rapid clearance by the host immune system can limit the expression of characteristic disease symptoms. In this study, we examine the critical role of animal infection models in understanding ESKAPE pathogens, addressing limitations and research gaps. We discuss applications and highlight key considerations for effective models. Thoughtful decisions on disease replication, parameter monitoring, and data collection are crucial for model reliability. By meticulously replicating human diseases and addressing limitations, researchers maximize the potential of animal infection models. This aids in targeted therapeutic development, bridges knowledge gaps, and helps combat MDR ESKAPE pathogens, safeguarding public health.
Collapse
Affiliation(s)
- Haojie Yu
- Key
Laboratory of Artificial Organs and Computational Medicine in Zhejiang
Province, Key Laboratory of Pollution Exposure and Health Intervention
of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, Zhejiang China
- Stomatology
Hospital, School of Stomatology, Zhejiang University School of Medicine,
Zhejiang Provincial Clinical Research Center for Oral Diseases, Key
Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Yongchang Xu
- Key
Laboratory of Aging and Cancer Biology of Zhejiang Province, School
of Basic Medical Sciences, Hangzhou Normal
University, Hangzhou 311121, China
| | - Saber Imani
- Shulan
International Medical College, Zhejiang
Shuren University, Hangzhou 310015, Zhejiang China
| | - Zhuo Zhao
- Department
of Computer Science and Engineering, University
of Notre Dame, Notre
Dame, Indiana 46556, United States
| | - Saif Ullah
- Department
of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87131, United States
| | - Qingjing Wang
- Key
Laboratory of Artificial Organs and Computational Medicine in Zhejiang
Province, Key Laboratory of Pollution Exposure and Health Intervention
of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, Zhejiang China
| |
Collapse
|
2
|
Plotniece A, Sobolev A, Supuran CT, Carta F, Björkling F, Franzyk H, Yli-Kauhaluoma J, Augustyns K, Cos P, De Vooght L, Govaerts M, Aizawa J, Tammela P, Žalubovskis R. Selected strategies to fight pathogenic bacteria. J Enzyme Inhib Med Chem 2023; 38:2155816. [PMID: 36629427 PMCID: PMC9848314 DOI: 10.1080/14756366.2022.2155816] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/02/2022] [Accepted: 12/02/2022] [Indexed: 01/12/2023] Open
Abstract
Natural products and analogues are a source of antibacterial drug discovery. Considering drug resistance levels emerging for antibiotics, identification of bacterial metalloenzymes and the synthesis of selective inhibitors are interesting for antibacterial agent development. Peptide nucleic acids are attractive antisense and antigene agents representing a novel strategy to target pathogens due to their unique mechanism of action. Antisense inhibition and development of antisense peptide nucleic acids is a new approach to antibacterial agents. Due to the increased resistance of biofilms to antibiotics, alternative therapeutic options are necessary. To develop antimicrobial strategies, optimised in vitro and in vivo models are needed. In vivo models to study biofilm-related respiratory infections, device-related infections: ventilator-associated pneumonia, tissue-related infections: chronic infection models based on alginate or agar beads, methods to battle biofilm-related infections are discussed. Drug delivery in case of antibacterials often is a serious issue therefore this review includes overview of drug delivery nanosystems.
Collapse
Affiliation(s)
- Aiva Plotniece
- Latvian Institute of Organic Synthesis, Riga, Latvia
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Riga Stradiņš University, Riga, Latvia
| | | | - Claudiu T. Supuran
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Firenze, Italy
| | - Fabrizio Carta
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Firenze, Italy
| | - Fredrik Björkling
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, Center for Peptide-Based Antibiotics, University of Copenhagen, Copenhagen East, Denmark
| | - Henrik Franzyk
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, Center for Peptide-Based Antibiotics, University of Copenhagen, Copenhagen East, Denmark
| | - Jari Yli-Kauhaluoma
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, Drug Research Program, University of Helsinki, Helsinki, Finland
| | - Koen Augustyns
- Infla-Med, Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Laboratory of Medicinal Chemistry, University of Antwerp, Antwerp, Belgium
| | - Paul Cos
- Department of Pharmaceutical Sciences, Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| | - Linda De Vooght
- Department of Pharmaceutical Sciences, Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| | - Matthias Govaerts
- Department of Pharmaceutical Sciences, Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| | - Juliana Aizawa
- Department of Pharmaceutical Sciences, Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| | - Päivi Tammela
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, Drug Research Program, University of Helsinki, Helsinki, Finland
| | - Raivis Žalubovskis
- Latvian Institute of Organic Synthesis, Riga, Latvia
- Faculty of Materials Science and Applied Chemistry, Institute of Technology of Organic Chemistry, Riga Technical University, Riga, Latvia
| |
Collapse
|
3
|
Tewes F, Lamy B, Laroche J, Lamarche I, Marchand S. PK-PD Evaluation of Inhaled Microparticles loaded with Ciprofloxacin-Copper complex in a Rat Model of Chronic Pseudomonas aeruginosa Lung Infection. Int J Pharm X 2023; 5:100178. [PMID: 36970713 PMCID: PMC10033950 DOI: 10.1016/j.ijpx.2023.100178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/04/2023] [Accepted: 03/06/2023] [Indexed: 03/29/2023] Open
Abstract
The potential gain in efficacy of pulmonary administration over IV administration of some antibiotics such as ciprofloxacin (CIP) may be limited by the short residence time of the drug at the site of infection after nebulization. Complexation of CIP with copper reduced its apparent permeability in vitro through a Calu-3 cell monolayer and greatly increased its pulmonary residence time after aerosolisation in healthy rats. Chronic P. aeruginosa lung infections in cystic fibrosis patients result in airway and alveolar inflammation that may increase the permeability of inhaled antibiotics and alter their fate in the lung after inhalation compared to what was seen in healthy conditions. The objective of this study was to compare the pharmacokinetics and efficacy of CIP-Cu2+ complex-loaded microparticles administered by pulmonary route with a CIP solution administered by IV to model rats with chronic lung infection. After a single pulmonary administration of microparticles loaded with CIP-Cu2+ complex, pulmonary exposure to CIP was increased 2077-fold compared to IV administration of CIP solution. This single lung administration significantly reduced the lung burden of P. aeruginosa expressed as CFU/lung measured 24 h after administration by 10-fold while IV administration of the same dose of CIP was ineffective compared to the untreated control. This better efficacy of inhaled microparticles loaded with CIP-Cu2+ complex compared with CIP solution can be attributed to the higher pulmonary exposure to CIP obtained with inhaled CIP-Cu2+ complex-loaded microparticles than that obtained with IV solution.
Collapse
Affiliation(s)
- Frederic Tewes
- Université de Poitiers, INSERM U1070, Poitiers, France
- Corresponding author.
| | - Barbara Lamy
- Université de Poitiers, INSERM U1070, Poitiers, France
| | - Julian Laroche
- CHU de Poitiers, laboratoire de Toxicologie et de Pharmacocinetique, Poitiers, France
| | | | - Sandrine Marchand
- Université de Poitiers, INSERM U1070, Poitiers, France
- CHU de Poitiers, laboratoire de Toxicologie et de Pharmacocinetique, Poitiers, France
| |
Collapse
|
4
|
Rodgers AM, Lindsay J, Monahan A, Dubois AV, Faniyi AA, Plant BJ, Mall MA, Ekkelenkamp MB, Elborn S, Ingram RJ. Biologically Relevant Murine Models of Chronic Pseudomonas aeruginosa Respiratory Infection. Pathogens 2023; 12:1053. [PMID: 37624013 PMCID: PMC10458525 DOI: 10.3390/pathogens12081053] [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: 06/03/2023] [Revised: 08/09/2023] [Accepted: 08/14/2023] [Indexed: 08/26/2023] Open
Abstract
Pseudomonas aeruginosa (P. aeruginosa) is an opportunistic pathogen and the leading cause of infection in patients with cystic fibrosis (CF). The ability of P. aeruginosa to evade host responses and develop into chronic infection causes significant morbidity and mortality. Several mouse models have been developed to study chronic respiratory infections induced by P. aeruginosa, with the bead agar model being the most widely used. However, this model has several limitations, including the requirement for surgical procedures and high mortality rates. Herein, we describe novel and adapted biologically relevant models of chronic lung infection caused by P. aeruginosa. Three methods are described: a clinical isolate infection model, utilising isolates obtained from patients with CF; an incomplete antibiotic clearance model, leading to bacterial bounce-back; and the establishment of chronic infection; and an adapted water bottle chronic infection model. These models circumvent the requirement for a surgical procedure and, importantly, can be induced with clinical isolates of P. aeruginosa and in wild-type mice. We also demonstrate successful induction of chronic infection in the transgenic βENaC murine model of CF. We envisage that the models described will facilitate the investigations of host and microbial factors, and the efficacy of novel antimicrobials, during chronic P. aeruginosa respiratory infections.
Collapse
Affiliation(s)
- Aoife M. Rodgers
- Wellcome-Wolfson Institute of Experimental Medicine, Queen’s University Belfast, Belfast BT7 1NN, UK (S.E.)
| | - Jaime Lindsay
- Wellcome-Wolfson Institute of Experimental Medicine, Queen’s University Belfast, Belfast BT7 1NN, UK (S.E.)
| | - Avril Monahan
- Wellcome-Wolfson Institute of Experimental Medicine, Queen’s University Belfast, Belfast BT7 1NN, UK (S.E.)
| | - Alice V. Dubois
- Wellcome-Wolfson Institute of Experimental Medicine, Queen’s University Belfast, Belfast BT7 1NN, UK (S.E.)
| | - Aduragbemi A. Faniyi
- Wellcome-Wolfson Institute of Experimental Medicine, Queen’s University Belfast, Belfast BT7 1NN, UK (S.E.)
| | - Barry J. Plant
- Cork Centre for Cystic Fibrosis (3CF), Cork University Hospital, University College Cork, T12 E8YV Cork, Ireland
- The HRB Funded Clinical Research Facility, University College Cork, T12 E8YV Cork, Ireland
| | - Marcus A. Mall
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité—University of Medicine Berlin, 10117 Berlin, Germany
- German Center for Lung Research (DZL), 10117 Berlin, Germany
- Berlin Institute of Health at Charité—University of Medicine Berlin, 10117 Berlin, Germany
| | - Miquel B. Ekkelenkamp
- Department of Medical Microbiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Stuart Elborn
- Wellcome-Wolfson Institute of Experimental Medicine, Queen’s University Belfast, Belfast BT7 1NN, UK (S.E.)
| | - Rebecca J. Ingram
- Wellcome-Wolfson Institute of Experimental Medicine, Queen’s University Belfast, Belfast BT7 1NN, UK (S.E.)
| |
Collapse
|
5
|
Verstraete L, Aizawa J, Govaerts M, De Vooght L, Lavigne R, Michiels J, Van den Bergh B, Cos P. In Vitro Persistence Level Reflects In Vivo Antibiotic Survival of Natural Pseudomonas aeruginosa Isolates in a Murine Lung Infection Model. Microbiol Spectr 2023; 11:e0497022. [PMID: 37140371 PMCID: PMC10269860 DOI: 10.1128/spectrum.04970-22] [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: 12/05/2022] [Accepted: 04/11/2023] [Indexed: 05/05/2023] Open
Abstract
Clinicians are increasingly confronted with the limitations of antibiotics to clear bacterial infections in patients. It has long been assumed that only antibiotic resistance plays a pivotal role in this phenomenon. Indeed, the worldwide emergence of antibiotic resistance is considered one of the major health threats of the 21st century. However, the presence of persister cells also has a significant influence on treatment outcomes. These antibiotic-tolerant cells are present in every bacterial population and are the result of the phenotypic switching of normal, antibiotic-sensitive cells. Persister cells complicate current antibiotic therapies and contribute to the development of resistance. In the past, extensive research has been performed to investigate persistence in laboratory settings; however, antibiotic tolerance under conditions that mimic the clinical setting remain poorly understood. In this study, we optimized a mouse model for lung infections with the opportunistic pathogen Pseudomonas aeruginosa. In this model, mice are intratracheally infected with P. aeruginosa embedded in seaweed alginate beads and subsequently treated with tobramycin via nasal droplets. A diverse panel of 18 P. aeruginosa strains originating from environmental, human, and animal clinical sources was selected to assess survival in the animal model. Survival levels were positively correlated with the survival levels determined via time-kill assays, a common method to study persistence in the laboratory. We showed that survival levels are comparable and thus that the classical persister assays are indicative of antibiotic tolerance in a clinical setting. The optimized animal model also enables us to test potential antipersister therapies and study persistence in relevant settings. IMPORTANCE The importance of targeting persister cells in antibiotic therapies is becoming more evident, as these antibiotic-tolerant cells underlie relapsing infections and resistance development. Here, we studied persistence in a clinically relevant pathogen, Pseudomonas aeruginosa. It is one of the six ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, P. aeruginosa, and Enterobacter spp.), which are considered major health threats. P. aeruginosa is mostly known to cause chronic lung infections in cystic fibrosis patients. We mimicked these lung infections in a mouse model to study persistence under more clinical conditions. It was shown that the survival levels of natural P. aeruginosa isolates in this model are positively correlated with the survival levels measured in classical persistence assays in vitro. These results not only validate the use of our current techniques to study persistence but also open opportunities to study new persistence mechanisms or evaluate new antipersister strategies in vivo.
Collapse
Affiliation(s)
- Laure Verstraete
- Centre of Microbial and Plant Genetics, KU Leuven, Leuven, Belgium
- Center for Microbiology, Flanders Institute for Biotechnology, Leuven, Belgium
| | - Juliana Aizawa
- Laboratory for Microbiology, Parasitology and Hygiene, University of Antwerp, Antwerp, Belgium
| | - Matthias Govaerts
- Laboratory for Microbiology, Parasitology and Hygiene, University of Antwerp, Antwerp, Belgium
| | - Linda De Vooght
- Laboratory for Microbiology, Parasitology and Hygiene, University of Antwerp, Antwerp, Belgium
| | - Rob Lavigne
- Laboratory of Gene Technology, KU Leuven, Leuven, Belgium
| | - Jan Michiels
- Centre of Microbial and Plant Genetics, KU Leuven, Leuven, Belgium
- Center for Microbiology, Flanders Institute for Biotechnology, Leuven, Belgium
| | - Bram Van den Bergh
- Centre of Microbial and Plant Genetics, KU Leuven, Leuven, Belgium
- Center for Microbiology, Flanders Institute for Biotechnology, Leuven, Belgium
| | - Paul Cos
- Laboratory for Microbiology, Parasitology and Hygiene, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
6
|
Xia JY, Zeng YF, Wu XJ, Xu F. Short-term ex vivo tissue culture models help study human lung infectionsA review. Medicine (Baltimore) 2023; 102:e32589. [PMID: 36607848 PMCID: PMC9829290 DOI: 10.1097/md.0000000000032589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Most studies on human lung infection have been performed using animal models, formalin or other fixed tissues, and in vitro cultures of established cell lines. However, the experimental data and results obtained from these studies may not completely represent the complicated molecular events that take place in intact human lung tissue in vivo. The newly developed ex vivo short-term tissue culture model can mimic the in vivo microenvironment of humans and allow investigations of different cell types that closely interact with each other in intact human lung tissues. Therefore, this kind of model may be a promising tool for future studies of different human lung infections, owing to its special advantages in providing more realistic events that occur in vivo. In this review, we have summarized the preliminary applications of this novel short-term ex vivo tissue culture model, with a particular emphasis on its applications in some common human lung infections.
Collapse
Affiliation(s)
- Jing-Yan Xia
- Department of Radiation Oncology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
| | - Yi-Fei Zeng
- Department of Infectious Diseases, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
| | - Xue-Jie Wu
- Department of Infectious Diseases, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
| | - Feng Xu
- Department of Infectious Diseases, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, China
- * Correspondence: Feng Xu, Department of Infectious Diseases, The Second Affiliated Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, Zhejiang 310009, PR China (e-mail: )
| |
Collapse
|
7
|
Secli V, Di Biagio C, Martini A, Michetti E, Pacello F, Ammendola S, Battistoni A. Localized Infections with P. aeruginosa Strains Defective in Zinc Uptake Reveal That Zebrafish Embryos Recapitulate Nutritional Immunity Responses of Higher Eukaryotes. Int J Mol Sci 2023; 24:ijms24020944. [PMID: 36674459 PMCID: PMC9862628 DOI: 10.3390/ijms24020944] [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: 11/09/2022] [Revised: 12/14/2022] [Accepted: 12/23/2022] [Indexed: 01/06/2023] Open
Abstract
The innate immune responses of mammals to microbial infections include strategies based on manipulating the local concentration of metals such as iron (Fe) and zinc (Zn), commonly described as nutritional immunity. To evaluate whether these strategies are also present in zebrafish embryos, we have conducted a series of heart cavity-localized infection experiments with Pseudomonas aeruginosa strains characterized by a different ability to acquire Zn. We have found that, 48 h after infection, the bacterial strains lacking critical components of the Zn importers ZnuABC and ZrmABCD have a reduced colonization capacity compared to the wild-type strain. This observation, together with the finding of a high level of expression of Zur-regulated genes, suggests the existence of antimicrobial mechanisms based on Zn sequestration. However, we have observed that strains lacking such Zn importers have a selective advantage over the wild-type strain in the early stages of infection. Analysis of the expression of the gene that encodes for a Zn efflux pump has revealed that at short times after infection, P. aeruginosa is exposed to high concentrations of Zn. At the same time, zebrafish respond to the infection by activating the expression of the Zn transporters Slc30a1 and Slc30a4, whose mammalian homologs mediate a redistribution of Zn in phagocytes aimed at intoxicating bacteria with a metal excess. These observations indicate that teleosts share similar nutritional immunity mechanisms with higher vertebrates, and confirm the usefulness of the zebrafish model for studying host-pathogen interactions.
Collapse
Affiliation(s)
- Valerio Secli
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Claudia Di Biagio
- Laboratory of Experimental Ecology and Aquaculture, Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Arianna Martini
- Laboratory of Experimental Ecology and Aquaculture, Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
- Council for Agricultural Research and Economics, Research, Centre for Animal Production and Aquaculture, Via Salaria 31, 00015 Monterotondo, Italy
| | - Emma Michetti
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Francesca Pacello
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Serena Ammendola
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Andrea Battistoni
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy
- Correspondence:
| |
Collapse
|
8
|
Lindgren NR, McDaniel MS, Novak L, Swords WE. Acute polymicrobial airway infections: analysis in cystic fibrosis mice. MICROBIOLOGY (READING, ENGLAND) 2023; 169:001290. [PMID: 36748431 PMCID: PMC9993112 DOI: 10.1099/mic.0.001290] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Cystic fibrosis (CF) is a genetic disorder affecting epithelial ion transport, which among other impacts results in defective mucociliary clearance and innate defenses in the respiratory tract. Consequently, people with CF experience lifelong infections of the respiratory mucosa that are chronic and polymicrobial in nature. Young children with CF are initially colonized by opportunists like nontypeable Haemophilus influenzae (NTHi), which normally resides within the microbiome of the nasopharynx and upper airways and can also cause infections of the respiratory mucosa that include bronchitis and otitis media. NTHi is typically supplanted by other microbes as patients age; for example, people with CF are often chronically infected with mucoid strains of Pseudomonas aeruginosa, which prior work in our laboratory has shown to promote colonization and persistence by other opportunists that include Stenotrophomonas maltophilia. Our previous work has shown that polymicrobial infection impacts host colonization and persistence of incoming microbes via diverse mechanisms that include priming of host immunity that can promote microbial clearance, and cooperativity within polymicrobial biofilms, which can promote persistence. In infection studies with BALB/c Cftrtm1UNC mice, results showed, as previously observed for WT BALB/c mice, preceding infection with NTHi decreased colonization and persistence by P. aeruginosa. Likewise, polymicrobial infection of BALB/c Cftrtm1UNC and C57BL/6 Cftrtm1UncTg(FABPhCFTR)1Jaw/J mice showed correlation between S. maltophilia and P. aeruginosa, with increased bacterial colonization and lung pathology. Based on these results, we conclude that our previous observations regarding polymicrobial infections with CF opportunists in WT mice are also validated using CF mice.
Collapse
Affiliation(s)
- Natalie R Lindgren
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama, Birmingham, USA.,Gregory Fleming James Center for Cystic Fibrosis Research, University of Alabama, Birmingham, Birmingham, USA
| | - Melissa S McDaniel
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama, Birmingham, USA.,Gregory Fleming James Center for Cystic Fibrosis Research, University of Alabama, Birmingham, Birmingham, USA
| | - Lea Novak
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, USA
| | - W Edward Swords
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama, Birmingham, USA.,Gregory Fleming James Center for Cystic Fibrosis Research, University of Alabama, Birmingham, Birmingham, USA
| |
Collapse
|
9
|
Grubb BR, Livraghi-Butrico A. Animal models of cystic fibrosis in the era of highly effective modulator therapies. Curr Opin Pharmacol 2022; 64:102235. [DOI: 10.1016/j.coph.2022.102235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 12/17/2022]
|
10
|
Gangoda L, Schenk RL, Best SA, Nedeva C, Louis C, D’Silva DB, Fairfax K, Jarnicki AG, Puthalakath H, Sutherland KD, Strasser A, Herold MJ. Absence of pro-survival A1 has no impact on inflammatory cell survival in vivo during acute lung inflammation and peritonitis. Cell Death Differ 2022; 29:96-104. [PMID: 34304242 PMCID: PMC8738744 DOI: 10.1038/s41418-021-00839-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 02/07/2023] Open
Abstract
Inflammation is a natural defence mechanism of the body to protect against pathogens. It is induced by immune cells, such as macrophages and neutrophils, which are rapidly recruited to the site of infection, mediating host defence. The processes for eliminating inflammatory cells after pathogen clearance are critical in preventing sustained inflammation, which can instigate diverse pathologies. During chronic inflammation, the excessive and uncontrollable activity of the immune system can cause extensive tissue damage. New therapies aimed at preventing this over-activity of the immune system could have major clinical benefits. Here, we investigated the role of the pro-survival Bcl-2 family member A1 in the survival of inflammatory cells under normal and inflammatory conditions using murine models of lung and peritoneal inflammation. Despite the robust upregulation of A1 protein levels in wild-type cells upon induction of inflammation, the survival of inflammatory cells was not impacted in A1-deficient mice compared to wild-type controls. These findings indicate that A1 does not play a major role in immune cell homoeostasis during inflammation and therefore does not constitute an attractive therapeutic target for such morbidities.
Collapse
Affiliation(s)
- Lahiru Gangoda
- grid.1042.7The Walter and Eliza Hall Institute of Medical Research (WEHI), Melbourne, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, University of Melbourne, Melbourne, VIC Australia ,grid.1018.80000 0001 2342 0938La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC Australia
| | - Robyn L. Schenk
- grid.1042.7The Walter and Eliza Hall Institute of Medical Research (WEHI), Melbourne, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, University of Melbourne, Melbourne, VIC Australia
| | - Sarah A. Best
- grid.1042.7The Walter and Eliza Hall Institute of Medical Research (WEHI), Melbourne, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, University of Melbourne, Melbourne, VIC Australia
| | - Christina Nedeva
- grid.1018.80000 0001 2342 0938La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC Australia
| | - Cynthia Louis
- grid.1042.7The Walter and Eliza Hall Institute of Medical Research (WEHI), Melbourne, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, University of Melbourne, Melbourne, VIC Australia
| | - Damian B. D’Silva
- grid.1042.7The Walter and Eliza Hall Institute of Medical Research (WEHI), Melbourne, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, University of Melbourne, Melbourne, VIC Australia
| | - Kirsten Fairfax
- grid.1042.7The Walter and Eliza Hall Institute of Medical Research (WEHI), Melbourne, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, University of Melbourne, Melbourne, VIC Australia ,grid.1009.80000 0004 1936 826XMenzies Institute for Medical Research, University of Tasmania, Hobart, TAS Australia
| | - Andrew G. Jarnicki
- grid.1008.90000 0001 2179 088XDepartment of Biochemistry and Pharmacology, University of Melbourne, Parkville, VIC Australia
| | - Hamsa Puthalakath
- grid.1018.80000 0001 2342 0938La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC Australia
| | - Kate D. Sutherland
- grid.1042.7The Walter and Eliza Hall Institute of Medical Research (WEHI), Melbourne, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, University of Melbourne, Melbourne, VIC Australia
| | - Andreas Strasser
- grid.1042.7The Walter and Eliza Hall Institute of Medical Research (WEHI), Melbourne, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, University of Melbourne, Melbourne, VIC Australia
| | - Marco J. Herold
- grid.1042.7The Walter and Eliza Hall Institute of Medical Research (WEHI), Melbourne, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, University of Melbourne, Melbourne, VIC Australia
| |
Collapse
|
11
|
Aljalamdeh R, Price R, Jones MD, Bolhuis A. The effect of particle size of inhaled tobramycin dry powder on the eradication of Pseudomonas aeruginosa biofilms. Eur J Pharm Sci 2021; 158:105680. [PMID: 33346008 DOI: 10.1016/j.ejps.2020.105680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/26/2020] [Accepted: 12/14/2020] [Indexed: 10/22/2022]
Abstract
Pseudomonas aeruginosa is the predominant opportunistic bacterium that causes chronic respiratory infections in cystic fibrosis (CF) patients. This bacterium can form biofilms, which are structured communities of cells encased within a self-produced matrix. Such biofilms have a high level of resistance to multiple classes of antibiotics. A widely used treatment of P. aeruginosa lung infections in CF patients is tobramycin dry powder inhalation. The behaviour of particles in the lung has been well studied, and dry powder inhalers are optimised for optimal dispersion of the drug into different zones of the lung. However, one question that has not been addressed is whether the size of an antibiotic particle influences the antibiofilm activity against P. aeruginosa. We investigated this by fractionating tobramycin particles using a Next Generation Impactor (NGI). The fractions obtained were then tested in an in vitro model on P. aeruginosa biofilms. The results indicate that the antibiofilm activity of tobramycin dry powder inhaler can indeed be influenced by the particle size. Against P. aeruginosa biofilms of two clinical isolates, smaller tobramycin particles (aerodynamic diameter <2.82 µm) showed better efficacy by approximately 20% as compared to larger tobramycin particles (aerodynamic diameter <11.7 µm) However, this effect was only observed when biofilms were treated for 3 hours, whereas there was no difference after treatment for 24 hours. This suggests that in our model the rate of dissolution of larger particles limits the effectiveness of tobramycin over a 3-hour time period, which is relevant as this is equivalent to the time in which most tobramycin is cleared from the lung.
Collapse
Affiliation(s)
- Reham Aljalamdeh
- Department of Pharmacy and Pharmacology, University of Bath, Bath BA2 7AY, United Kingdom
| | - Robert Price
- Department of Pharmacy and Pharmacology, University of Bath, Bath BA2 7AY, United Kingdom
| | - Matthew D Jones
- Department of Pharmacy and Pharmacology, University of Bath, Bath BA2 7AY, United Kingdom
| | - Albert Bolhuis
- Department of Pharmacy and Pharmacology, University of Bath, Bath BA2 7AY, United Kingdom.
| |
Collapse
|
12
|
Otero Fernandez M, Thomas RJ, Oswin H, Haddrell AE, Reid JP. Transformative Approach To Investigate the Microphysical Factors Influencing Airborne Transmission of Pathogens. Appl Environ Microbiol 2020; 86:e01543-20. [PMID: 32978136 PMCID: PMC7657628 DOI: 10.1128/aem.01543-20] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 09/17/2020] [Indexed: 01/06/2023] Open
Abstract
Emerging outbreaks of airborne pathogenic infections worldwide, such as the current severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic, have raised the need to understand parameters affecting the airborne survival of microbes in order to develop measures for effective infection control. We report a novel experimental strategy, TAMBAS (tandem approach for microphysical and biological assessment of airborne microorganism survival), to explore the synergistic interactions between the physicochemical and biological processes that impact airborne microbe survival in aerosol droplets. This innovative approach provides a unique and detailed understanding of the processes taking place from aerosol droplet generation through to equilibration and viability decay in the local environment, elucidating decay mechanisms not previously described. The impact of evaporation kinetics, solute hygroscopicity and concentration, particle morphology, and equilibrium particle size on airborne survival are reported, using Escherichia coli MRE162 as a benchmark system. For this system, we report that (i) particle crystallization does not directly impact microbe longevity, (ii) bacteria act as crystallization nuclei during droplet drying and equilibration, and (iii) the kinetics of size and compositional change appear to have a larger effect on microbe longevity than the equilibrium solute concentration.IMPORTANCE A transformative approach to identify the physicochemical processes that impact the biological decay rates of bacteria in aerosol droplets is described. It is shown that the evaporation process and changes in the phase and morphology of the aerosol particle during evaporation impact microorganism viability. The equilibrium droplet size was found to affect airborne bacterial viability. Furthermore, the presence of Escherichia coli MRE162 in a droplet does not affect aerosol growth/evaporation but influences the dynamic behavior of the aerosol by processing the culture medium prior to aerosolization, affecting the hygroscopicity of the culture medium; this highlights the importance of the inorganic and organic chemical composition within the aerosolized droplets that impact hygroscopicity. Bacteria also act as crystallization nuclei. The novel approach and data have implications for increased mechanistic understanding of aerosol survival and infectivity in bioaerosol studies spanning the medical, veterinary, farming, and agricultural fields, including the role of microorganisms in atmospheric processing and cloud formation.
Collapse
Affiliation(s)
| | - Richard J Thomas
- Defence Science Technology Laboratory (DSTL), Porton Down, Salisbury, United Kingdom
| | - Henry Oswin
- School of Chemistry, University of Bristol, Bristol, United Kingdom
| | - Allen E Haddrell
- School of Chemistry, University of Bristol, Bristol, United Kingdom
| | - Jonathan P Reid
- School of Chemistry, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
13
|
Perault AI, Chandler CE, Rasko DA, Ernst RK, Wolfgang MC, Cotter PA. Host Adaptation Predisposes Pseudomonas aeruginosa to Type VI Secretion System-Mediated Predation by the Burkholderia cepacia Complex. Cell Host Microbe 2020; 28:534-547.e3. [PMID: 32755549 PMCID: PMC7554260 DOI: 10.1016/j.chom.2020.06.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/05/2020] [Accepted: 06/24/2020] [Indexed: 12/14/2022]
Abstract
Pseudomonas aeruginosa and Burkholderia cepacia complex (Bcc) species are opportunistic lung pathogens of cystic fibrosis (CF) patients. While P. aeruginosa can initiate long-term infections in younger CF patients, Bcc infections only arise in teenagers and adults. Both P. aeruginosa and Bcc use type VI secretion systems (T6SSs) to mediate interbacterial competition. Here, we show P. aeruginosa isolates from teenage and adult CF patients, but not those from young CF patients, are outcompeted by the epidemic Bcc isolate Burkholderia cenocepacia strain AU1054 in a T6SS-dependent manner. The genomes of susceptible P. aeruginosa isolates harbor T6SS-abrogating mutations, the repair of which, in some cases, rendered the isolates resistant. Moreover, seven of eight Bcc strains outcompeted P. aeruginosa strains isolated from the same patients. Our findings suggest certain mutations that arise as P. aeruginosa adapts to the CF lung abrogate T6SS activity, making P. aeruginosa and its human host susceptible to potentially fatal Bcc superinfection.
Collapse
Affiliation(s)
- Andrew I Perault
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Courtney E Chandler
- Department of Microbial Pathogenesis, University of Maryland, Baltimore, Baltimore, MD 21201, USA
| | - David A Rasko
- Institute for Genome Sciences, University of Maryland, Baltimore, Baltimore, MD 21201, USA; Department of Microbiology and Immunology, University of Maryland, Baltimore, Baltimore, MD 21201, USA
| | - Robert K Ernst
- Department of Microbial Pathogenesis, University of Maryland, Baltimore, Baltimore, MD 21201, USA
| | - Matthew C Wolfgang
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Marsio Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Peggy A Cotter
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
14
|
Sou T, Bergström CAS. Contemporary Formulation Development for Inhaled Pharmaceuticals. J Pharm Sci 2020; 110:66-86. [PMID: 32916138 DOI: 10.1016/j.xphs.2020.09.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/02/2020] [Accepted: 09/03/2020] [Indexed: 12/22/2022]
Abstract
Pulmonary delivery has gained increased interests over the past few decades. For respiratory conditions, targeted drug delivery directly to the site of action can achieve a high local concentration for efficacy with reduced systemic exposure and adverse effects. For systemic conditions, the unique physiology of the lung evolutionarily designed for rapid gaseous exchange presents an entry route for systemic drug delivery. Although the development of inhaled formulations has come a long way over the last few decades, many aspects of it remain to be elucidated. In particular, a reliable and well-understood method for in vitro-in vivo correlations remains to be established. With the rapid and ongoing advancement of technology, there is much potential to better utilise computational methods including different types of modelling and simulation approaches to support inhaled formulation development. This review intends to provide an introduction on some fundamental concepts in pulmonary drug delivery and inhaled formulation development followed by discussions on some challenges and opportunities in the translation of inhaled pharmaceuticals from preclinical studies to clinical development. The review concludes with some recent advancements in modelling and simulation approaches that could play an increasingly important role in modern formulation development of inhaled pharmaceuticals.
Collapse
Affiliation(s)
- Tomás Sou
- Drug Delivery, Department of Pharmacy, Uppsala University, Uppsala, Sweden; Pharmacometrics, Department of Pharmacy, Uppsala University, Uppsala, Sweden.
| | - Christel A S Bergström
- Drug Delivery, Department of Pharmacy, Uppsala University, Uppsala, Sweden; The Swedish Drug Delivery Center, Department of Pharmacy, Uppsala University, Uppsala, Sweden
| |
Collapse
|
15
|
Tewes F, Bahamondez-Canas TF, Moraga-Espinoza D, Smyth HDC, Watts AB. In vivo efficacy of a dry powder formulation of ciprofloxacin-copper complex in a chronic lung infection model of bioluminescent Pseudomonas aeruginosa. Eur J Pharm Biopharm 2020; 152:210-217. [PMID: 32442738 DOI: 10.1016/j.ejpb.2020.05.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 05/01/2020] [Accepted: 05/15/2020] [Indexed: 12/21/2022]
Abstract
A significant limitation of locally delivered treatments for chronic pulmonary infections is often the short residence time within the airways. Ciprofloxacin (CIP), for example, undergoes rapid absorption from the airway lumen. Previously, we demonstrated that the complexation of CIP with copper (CIP-Cu) reduces its apparent epithelial permeability and pulmonary absorption rate without affecting antimicrobial activity against Pseudomonas aeruginosa grown planktonically or as biofilms. This study aimed to evaluate the in vivo efficacy of CIP-Cu, prepared as a dry powder, in a chronic lung infection model. The powders were prepared by jet milling (CIP-HCl) and by spray drying (CIP-Cu). A bioluminescent strain of P. aeruginosa (PAO1::p16Slux) was used to prepare bacteria-loaded agar beads that were inoculated intratracheally to rats. The dynamics of the infection were monitored using luminometry. The bacteria/beads ratio was optimized to allow the highest luminescence signal and animal survival for 8 days. The efficacy of the treatment was evaluated by luminometry in addition to the end-point (Day 8) where colony counting was performed after lung harvesting. Luminescent P. aeruginosa entrapped in agar beads were useful to monitor the spatial development of the chronic lung infection in rats. The rats were treated with the dry powders in a nose-only inhalation exposure system (NOIES). CIP-Cu and CIP-HCl powders showed similar aerodynamic properties and comparable CIP lung deposition. However, treatment with CIP-Cu significantly (p < 0.01) reduced by 4-log the number of CFU of P. aeruginosa per lung in the chronic infection model, whereas CIP-HCl effect was not different from the untreated control group.
Collapse
Affiliation(s)
- Frédéric Tewes
- INSERM U1070, UFR de Médecine Pharmacie, Université de Poitiers, 1 rue Georges Bonnet, TSA 51106, 86073 Poitiers Cedex 9, France; College of Pharmacy, The University of Texas at Austin, 2409 West University Avenue, PHR 4.214, Austin, TX 78712, USA.
| | - Tania F Bahamondez-Canas
- College of Pharmacy, The University of Texas at Austin, 2409 West University Avenue, PHR 4.214, Austin, TX 78712, USA; Escuela de Farmacia, Universidad de Valparaiso, Gran Bretaña 1093, Playa Ancha, Valparaíso, Chile; Centro de Investigación Farmacopea, Universidad de Valparaíso, Santa Marta 183, Playa Ancha, Valparaíso, Chile
| | - Daniel Moraga-Espinoza
- College of Pharmacy, The University of Texas at Austin, 2409 West University Avenue, PHR 4.214, Austin, TX 78712, USA; Escuela de Farmacia, Universidad de Valparaiso, Gran Bretaña 1093, Playa Ancha, Valparaíso, Chile; Centro de Investigación Farmacopea, Universidad de Valparaíso, Santa Marta 183, Playa Ancha, Valparaíso, Chile
| | - Hugh D C Smyth
- College of Pharmacy, The University of Texas at Austin, 2409 West University Avenue, PHR 4.214, Austin, TX 78712, USA
| | - Alan B Watts
- College of Pharmacy, The University of Texas at Austin, 2409 West University Avenue, PHR 4.214, Austin, TX 78712, USA
| |
Collapse
|
16
|
Brillault J, Tewes F. Control of the Lung Residence Time of Highly Permeable Molecules after Nebulization: Example of the Fluoroquinolones. Pharmaceutics 2020; 12:pharmaceutics12040387. [PMID: 32340298 PMCID: PMC7238242 DOI: 10.3390/pharmaceutics12040387] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/05/2020] [Accepted: 04/07/2020] [Indexed: 12/29/2022] Open
Abstract
Pulmonary drug delivery is a promising strategy to treat lung infectious disease as it allows for a high local drug concentration and low systemic side effects. This is particularly true for low-permeability drugs, such as tobramycin or colistin, that penetrate the lung at a low rate after systemic administration and greatly benefit from lung administration in terms of the local drug concentration. However, for relatively high-permeable drugs, such as fluoroquinolones (FQs), the rate of absorption is so high that the pulmonary administration has no therapeutic advantage compared to systemic or oral administration. Formulation strategies have thus been developed to decrease the absorption rate and increase FQs’ residence time in the lung after inhalation. In the present review, some of these strategies, which generally consist of either decreasing the lung epithelium permeability or decreasing the release rate of FQs into the epithelial lining fluid after lung deposition, are presented in regards to their clinical aspects.
Collapse
Affiliation(s)
- Julien Brillault
- INSERM U-1070, Pôle Biologie Santé, 86000 Poitiers, France
- UFR Médecine-Pharmacie, Université de Poitiers, 86073 Poitiers, France
- Correspondence: (J.B.); (F.T.)
| | - Frédéric Tewes
- INSERM U-1070, Pôle Biologie Santé, 86000 Poitiers, France
- UFR Médecine-Pharmacie, Université de Poitiers, 86073 Poitiers, France
- Correspondence: (J.B.); (F.T.)
| |
Collapse
|
17
|
Sécher T, Dalonneau E, Ferreira M, Parent C, Azzopardi N, Paintaud G, Si-Tahar M, Heuzé-Vourc'h N. In a murine model of acute lung infection, airway administration of a therapeutic antibody confers greater protection than parenteral administration. J Control Release 2019; 303:24-33. [PMID: 30981816 DOI: 10.1016/j.jconrel.2019.04.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 03/06/2019] [Accepted: 04/03/2019] [Indexed: 11/28/2022]
Abstract
Due to growing antibiotic resistance, pneumonia caused by Pseudomonas aeruginosa is a major threat to human health and is driving the development of novel anti-infectious agents. Preventively or curatively administered pathogen-specific therapeutic antibodies (Abs) have several advantages, including a low level of toxicity and a unique pharmacological profile. At present, most Abs against respiratory infections are administered parenterally; this may not be optimal for therapeutics that have to reach the lungs to be effective. Although the airways constitute a logical delivery route for biologics designed to treat respiratory diseases, there are few scientific data on the advantages or disadvantages of this route in the context of pneumonia treatment. The objective of the present study was to evaluate the efficacy and fate of an anti-P. aeruginosa Ab targeting pcrV (mAb166) as a function of the administration route during pneumonia. The airway-administered mAb166 displayed a favorable pharmacokinetic profile during the acute phase of the infection, and was associated with greater protection (relative to other delivery routes) of infected animals. Airway administration was associated with lower levels of lung inflammation, greater bacterial clearance, and recruitment of neutrophils in the airways. In conclusion, the present study is the first to have compared the pharmacokinetics and efficacy of an anti-infectious Ab administered by different routes in an animal model of pneumonia. Our findings suggest that local delivery to the airways is associated with a more potent anti-bacterial response (relative to parenteral administration), and thus open up new perspectives for the prevention and treatment of pneumonia with Abs.
Collapse
Affiliation(s)
- Thomas Sécher
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France; Université de Tours, F-37032 Tours, France
| | - Emilie Dalonneau
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France; Université de Tours, F-37032 Tours, France
| | - Marion Ferreira
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France; Université de Tours, F-37032 Tours, France; CHRU de Tours, Département de Pneumologie et d'exploration respiratoire fonctionnelle, F-37032 Tours, France
| | - Christelle Parent
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France; Université de Tours, F-37032 Tours, France
| | | | - Gilles Paintaud
- Université de Tours, GICC, PATCH Team, F-37032 Tours, France; CHRU de Tours, Laboratoire de Pharmacologie-Toxicologie, F-37032 Tours, France
| | - Mustapha Si-Tahar
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France; Université de Tours, F-37032 Tours, France
| | - Nathalie Heuzé-Vourc'h
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France; Université de Tours, F-37032 Tours, France.
| |
Collapse
|
18
|
Derbali RM, Aoun V, Moussa G, Frei G, Tehrani SF, Del’Orto JC, Hildgen P, Roullin VG, Chain JL. Tailored Nanocarriers for the Pulmonary Delivery of Levofloxacin against Pseudomonas aeruginosa: A Comparative Study. Mol Pharm 2019; 16:1906-1916. [DOI: 10.1021/acs.molpharmaceut.8b01256] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
19
|
Kumar SS, Tandberg JI, Penesyan A, Elbourne LDH, Suarez-Bosche N, Don E, Skadberg E, Fenaroli F, Cole N, Winther-Larsen HC, Paulsen IT. Dual Transcriptomics of Host-Pathogen Interaction of Cystic Fibrosis Isolate Pseudomonas aeruginosa PASS1 With Zebrafish. Front Cell Infect Microbiol 2018; 8:406. [PMID: 30524971 PMCID: PMC6262203 DOI: 10.3389/fcimb.2018.00406] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 10/29/2018] [Indexed: 01/09/2023] Open
Abstract
Pseudomonas aeruginosa is a significant cause of mortality in patients with cystic fibrosis (CF). To explore the interaction of the CF isolate P. aeruginosa PASS1 with the innate immune response, we have used Danio rerio (zebrafish) as an infection model. Confocal laser scanning microscopy (CLSM) enabled visualization of direct interactions between zebrafish macrophages and P. aeruginosa PASS1. Dual RNA-sequencing of host-pathogen was undertaken to profile RNA expression simultaneously in the pathogen and the host during P. aeruginosa infection. Following establishment of infection in zebrafish embryos with PASS1, 3 days post infection (dpi), there were 6739 genes found to be significantly differentially expressed in zebrafish and 176 genes in PASS1. A range of virulence genes were upregulated in PASS1, including genes encoding pyoverdine biosynthesis, flagellin, non-hemolytic phospholipase C, proteases, superoxide dismutase and fimbrial subunits. Additionally, iron and phosphate acquisition genes were upregulated in PASS1 cells in the zebrafish. Transcriptional changes in the host immune response genes highlighted phagocytosis as a key response mechanism to PASS1 infection. Transcriptional regulators of neutrophil and macrophage phagocytosis were upregulated alongside transcriptional regulators governing response to tissue injury, infection, and inflammation. The zebrafish host showed significant downregulation of the ribosomal RNAs and other genes involved in translation, suggesting that protein translation in the host is affected by PASS1 infection.
Collapse
Affiliation(s)
- Sheemal S Kumar
- Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
| | - Julia I Tandberg
- Department of Pharmaceutical Biosciences, Centre of Integrative Microbial Evolution, School of Pharmacy, University of Oslo, Oslo, Norway
| | - Anahit Penesyan
- Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
| | - Liam D H Elbourne
- Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
| | - Nadia Suarez-Bosche
- Microscopy Unit, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
| | - Emily Don
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Eline Skadberg
- Department of Pharmaceutical Biosciences, Centre of Integrative Microbial Evolution, School of Pharmacy, University of Oslo, Oslo, Norway
| | - Federico Fenaroli
- Department of Biosciences, The Faculty of Mathematic and Natural Sciences, University of Oslo, Oslo, Norway
| | - Nicholas Cole
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Hanne Cecilie Winther-Larsen
- Department of Pharmaceutical Biosciences, Centre of Integrative Microbial Evolution, School of Pharmacy, University of Oslo, Oslo, Norway
| | - Ian T Paulsen
- Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
20
|
In vitro and ex vivo systems at the forefront of infection modeling and drug discovery. Biomaterials 2018; 198:228-249. [PMID: 30384974 PMCID: PMC7172914 DOI: 10.1016/j.biomaterials.2018.10.030] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 10/05/2018] [Accepted: 10/23/2018] [Indexed: 12/11/2022]
Abstract
Bacterial infections and antibiotic resistant bacteria have become a growing problem over the past decade. As a result, the Centers for Disease Control predict more deaths resulting from microorganisms than all cancers combined by 2050. Currently, many traditional models used to study bacterial infections fail to precisely replicate the in vivo bacterial environment. These models often fail to incorporate fluid flow, bio-mechanical cues, intercellular interactions, host-bacteria interactions, and even the simple inclusion of relevant physiological proteins in culture media. As a result of these inadequate models, there is often a poor correlation between in vitro and in vivo assays, limiting therapeutic potential. Thus, the urgency to establish in vitro and ex vivo systems to investigate the mechanisms underlying bacterial infections and to discover new-age therapeutics against bacterial infections is dire. In this review, we present an update of current in vitro and ex vivo models that are comprehensively changing the landscape of traditional microbiology assays. Further, we provide a comparative analysis of previous research on various established organ-disease models. Lastly, we provide insight on future techniques that may more accurately test new formulations to meet the growing demand of antibiotic resistant bacterial infections.
Collapse
|
21
|
Gagné-Thivierge C, Kukavica-Ibrulj I, Filion G, Dekimpe V, Tan SGE, Vincent AT, Déziel É, Levesque RC, Charette SJ. A multi-host approach to identify a transposon mutant of Pseudomonas aeruginosa LESB58 lacking full virulence. BMC Res Notes 2018; 11:198. [PMID: 29580289 PMCID: PMC5870910 DOI: 10.1186/s13104-018-3308-7] [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: 12/08/2017] [Accepted: 03/20/2018] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE Pseudomonas aeruginosa is an opportunistic bacterial pathogen well known to cause chronic lung infections in individuals with cystic fibrosis (CF). Some strains adapted to this particular niche show distinct phenotypes, such as biofilm hyperproduction. It is necessary to study CF clinical P. aeruginosa isolates, such as Liverpool Epidemic Strains (LES), to acquire a better understanding of the key genes essential for in vivo maintenance and the major virulence mechanisms involved in CF lung infections. Previously, a library of 9216 mutants of the LESB58 strain were generated by signature-tagged mutagenesis (STM) and screened in the rat model of chronic lung infection, allowing the identification of 163 STM mutants showing defects in in vivo maintenance. RESULTS In the present study, these 163 mutants were successively screened in two additional surrogate host models (the amoeba and the fruit fly). The STM PALES_11731 mutant was the unique non-virulent in the three hosts. A competitive index study in rat lungs confirmed that the mutant was 20-fold less virulent than the wild-type strain. This study demonstrated the pertinence to use a multi-host approach to study the genetic determinants of P. aeruginosa strains infecting CF patients.
Collapse
Affiliation(s)
- Cynthia Gagné-Thivierge
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Quebec City, QC, Canada.,Département de biochimie, de microbiologie et de bio-informatique, Faculté des sciences et de génie, Université Laval, Quebec City, QC, Canada.,Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec City, QC, Canada
| | - Irena Kukavica-Ibrulj
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Quebec City, QC, Canada.,Département de microbiologie, infectiologie et immunologie, Faculté de Médecine, Université Laval, Quebec City, QC, Canada
| | - Geneviève Filion
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Quebec City, QC, Canada.,Département de biochimie, de microbiologie et de bio-informatique, Faculté des sciences et de génie, Université Laval, Quebec City, QC, Canada.,Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec City, QC, Canada
| | | | - Sok Gheck E Tan
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Quebec City, QC, Canada.,Département de biochimie, de microbiologie et de bio-informatique, Faculté des sciences et de génie, Université Laval, Quebec City, QC, Canada.,Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec City, QC, Canada
| | - Antony T Vincent
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Quebec City, QC, Canada.,Département de biochimie, de microbiologie et de bio-informatique, Faculté des sciences et de génie, Université Laval, Quebec City, QC, Canada.,Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec City, QC, Canada
| | - Éric Déziel
- INRS-Institut Armand Frappier, Laval, QC, Canada
| | - Roger C Levesque
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Quebec City, QC, Canada.,Département de microbiologie, infectiologie et immunologie, Faculté de Médecine, Université Laval, Quebec City, QC, Canada
| | - Steve J Charette
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Quebec City, QC, Canada. .,Département de biochimie, de microbiologie et de bio-informatique, Faculté des sciences et de génie, Université Laval, Quebec City, QC, Canada. .,Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec City, QC, Canada.
| |
Collapse
|
22
|
Maura D, Bandyopadhaya A, Rahme LG. Animal Models for Pseudomonas aeruginosa Quorum Sensing Studies. Methods Mol Biol 2018; 1673:227-241. [PMID: 29130177 DOI: 10.1007/978-1-4939-7309-5_18] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Quorum sensing (QS) systems play global regulatory roles in bacterial virulence. They synchronize the expression of multiple virulence factors and they control and modulate bacterial antibiotic tolerance systems and host defense mechanisms. Therefore, it is important to obtain knowledge about QS modes of action and to test putative therapeutics that may interrupt QS actions in the context of infections. This chapter describes methods to study bacterial pathogenesis in murine acute and persistent/relapsing infection models, using the Gram-negative bacterial pathogen Pseudomonas aeruginosa as an example. These infection models can be used to probe bacterial virulence functions and in mechanistic studies, as well as for the assessment of the therapeutic potential of antibacterials, including anti-virulence agents.
Collapse
Affiliation(s)
- Damien Maura
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA.,Shriners Hospitals for Children Boston, Boston, MA, USA
| | - Arunava Bandyopadhaya
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA.,Shriners Hospitals for Children Boston, Boston, MA, USA
| | - Laurence G Rahme
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA. .,Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA. .,Shriners Hospitals for Children Boston, Boston, MA, USA.
| |
Collapse
|
23
|
Wuerth KC, Falsafi R, Hancock REW. Synthetic host defense peptide IDR-1002 reduces inflammation in Pseudomonas aeruginosa lung infection. PLoS One 2017; 12:e0187565. [PMID: 29107983 PMCID: PMC5673212 DOI: 10.1371/journal.pone.0187565] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 10/23/2017] [Indexed: 11/18/2022] Open
Abstract
Pseudomonas aeruginosa is a frequent cause of lung infections, particularly in chronic infections in cystic fibrosis patients. However, treatment is challenging due to P. aeruginosa evasion of the host immune system and the rise of antibiotic resistant strains. Host defense peptides (HDPs) and synthetic derivatives called innate defense regulators (IDRs) have shown promise in several infection models as an alternative to antibiotic treatment. Here we tested peptide IDR-1002 against P. aeruginosa in vitro and in vivo. Treatment of bronchial epithelial cells and macrophages with IDR-1002 or in combination with live P. aeruginosa or its LPS led to the reduction of agonist-induced cytokines and chemokines and limited cell killing by live P. aeruginosa. In an in vivo model using P. aeruginosa combined with alginate to mimic a chronic model, IDR-1002 did not reduce the bacterial burden in the lungs, but IDR-1002 mice showed a significant decrease in IL-6 in the lungs and in gross pathology of infection, while histology revealed that IDR-1002 treated mice had reduced alveolar macrophage infiltration around the site of infection and reduced inflammation. Overall, these results indicate that IDR-1002 has promise for combating P. aeruginosa lung infections and their resulting inflammation.
Collapse
Affiliation(s)
- Kelli C. Wuerth
- Centre for Microbial Diseases and Immunity Research, Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Reza Falsafi
- Centre for Microbial Diseases and Immunity Research, Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Robert E. W. Hancock
- Centre for Microbial Diseases and Immunity Research, Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
- * E-mail:
| |
Collapse
|
24
|
Susceptibility of airways to Pseudomonas aeruginosa infection: mouse neuraminidase model. MONATSHEFTE FUR CHEMIE 2017. [DOI: 10.1007/s00706-017-2035-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
25
|
Gomes IB, Meireles A, Gonçalves AL, Goeres DM, Sjollema J, Simões LC, Simões M. Standardized reactors for the study of medical biofilms: a review of the principles and latest modifications. Crit Rev Biotechnol 2017; 38:657-670. [DOI: 10.1080/07388551.2017.1380601] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Inês B. Gomes
- LEPABE – Laboratório de Engenharia de Processos, Ambiente, Biotecnologia e Energia, Faculdade de Engenharia da Universidade do Porto, Porto, Portugal
| | - Ana Meireles
- LEPABE – Laboratório de Engenharia de Processos, Ambiente, Biotecnologia e Energia, Faculdade de Engenharia da Universidade do Porto, Porto, Portugal
| | - Ana L. Gonçalves
- LEPABE – Laboratório de Engenharia de Processos, Ambiente, Biotecnologia e Energia, Faculdade de Engenharia da Universidade do Porto, Porto, Portugal
| | - Darla M. Goeres
- Center for Biofilm Engineering, Montana State University, Bozeman, MT, USA
| | - Jelmer Sjollema
- University of Groningen, University Medical Center Groningen, Department of Biomedical Engineering, Groningen, The Netherlands
| | - Lúcia C. Simões
- LEPABE – Laboratório de Engenharia de Processos, Ambiente, Biotecnologia e Energia, Faculdade de Engenharia da Universidade do Porto, Porto, Portugal
| | - Manuel Simões
- LEPABE – Laboratório de Engenharia de Processos, Ambiente, Biotecnologia e Energia, Faculdade de Engenharia da Universidade do Porto, Porto, Portugal
| |
Collapse
|
26
|
Haddrell AE, Thomas RJ. Aerobiology: Experimental Considerations, Observations, and Future Tools. Appl Environ Microbiol 2017; 83:e00809-17. [PMID: 28667111 PMCID: PMC5561278 DOI: 10.1128/aem.00809-17] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Understanding airborne survival and decay of microorganisms is important for a range of public health and biodefense applications, including epidemiological and risk analysis modeling. Techniques for experimental aerosol generation, retention in the aerosol phase, and sampling require careful consideration and understanding so that they are representative of the conditions the bioaerosol would experience in the environment. This review explores the current understanding of atmospheric transport in relation to advances and limitations of aerosol generation, maintenance in the aerosol phase, and sampling techniques. Potential tools for the future are examined at the interface between atmospheric chemistry, aerosol physics, and molecular microbiology where the heterogeneity and variability of aerosols can be explored at the single-droplet and single-microorganism levels within a bioaerosol. The review highlights the importance of method comparison and validation in bioaerosol research and the benefits that the application of novel techniques could bring to increasing the understanding of aerobiological phenomena in diverse research fields, particularly during the progression of atmospheric transport, where complex interdependent physicochemical and biological processes occur within bioaerosol particles.
Collapse
Affiliation(s)
- Allen E Haddrell
- School of Chemistry, University of Bristol, Bristol, United Kingdom
| | - Richard J Thomas
- Defence Science and Technology Laboratory, Porton Down, Salisbury, Wiltshire, United Kingdom
| |
Collapse
|
27
|
Experimental Pseudomonas aeruginosa mediated rhino sinusitis in mink. Int J Pediatr Otorhinolaryngol 2017; 96:156-163. [PMID: 28302328 DOI: 10.1016/j.ijporl.2016.12.037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 12/25/2016] [Accepted: 12/26/2016] [Indexed: 12/13/2022]
Abstract
OBJECTIVES The nasal and sinus cavities in children may serve as reservoirs for microorganisms that cause recurrent and chronic lung infections. This study evaluates whether the mink can be used as an animal model for studying Pseudomonas aeruginosa mediated rhino-sinusitis since there is no suitable traditional animal model for this disease. METHODS Nasal tissue samples from infected and control mink were fixed in formalin, demineralized, and embedded in paraffin. A histological examination of sections from the infected animals revealed disintegration of the respiratory epithelium lining the nasal turbinates and swelling and edema of the submucosa. The expression of mucins and sialylated glycans was examined using immunohistochemistry. RESULTS MUC1, MUC2 and MUC5AC were upregulated in the inoculated animals as a much stronger staining was present in the respiratory epithelium in the infected animals compared to the controls. The goblet cells in the nasal epithelium from the infected mink showed high affinity to the Maackia amurensis lectin and anti-asialo GM1 indicating a high concentration of α2-3 sialic acid respectively βGalNAc1-4Galβ containing glycans in these mucin producing cells. The nasal cavity in the infected mink shows features of carbohydrate expression comparable to what has been described in the respiratory system after Pseudomonas aeruginosa infection in humans. CONCLUSION It is suggested that the mink is suitable for studying Pseudomonas aeruginosa mediated rhino-sinusitis.
Collapse
|
28
|
Alternative strategies for the study and treatment of clinical bacterial biofilms. Emerg Top Life Sci 2017; 1:41-53. [PMID: 33525815 DOI: 10.1042/etls20160020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 02/24/2017] [Accepted: 02/28/2017] [Indexed: 11/17/2022]
Abstract
Biofilms represent an adaptive lifestyle where microbes grow as structured aggregates in many different environments, e.g. on body surfaces and medical devices. They are a profound threat in medical (and industrial) settings and cause two-thirds of all infections. Biofilm bacteria are especially recalcitrant to common antibiotic treatments, demonstrating adaptive multidrug resistance. For this reason, novel methods to eradicate or prevent biofilm infections are greatly needed. Recent advances have been made in exploring alternative strategies that affect biofilm lifestyle, inhibit biofilm formation, degrade biofilm components and/or cause dispersal. As such, naturally derived compounds, molecules that interfere with bacterial signaling systems, anti-biofilm peptides and phages show great promise. Their implementation as either stand-alone drugs or complementary therapies has the potential to eradicate resilient biofilm infections. Additionally, altering the surface properties of indwelling medical devices through bioengineering approaches has been examined as a method for preventing biofilm formation. There is also a need for improving current biofilm detection methods since in vitro methods often do not accurately measure live bacteria in biofilms or mimic in vivo conditions. We propose that the design and development of novel compounds will be enabled by the improvement and use of appropriate in vitro and in vivo models.
Collapse
|
29
|
Bielen K, 's Jongers B, Malhotra-Kumar S, Jorens PG, Goossens H, Kumar-Singh S. Animal models of hospital-acquired pneumonia: current practices and future perspectives. ANNALS OF TRANSLATIONAL MEDICINE 2017; 5:132. [PMID: 28462212 DOI: 10.21037/atm.2017.03.72] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Lower respiratory tract infections are amongst the leading causes of mortality and morbidity worldwide. Especially in hospital settings and more particularly in critically ill ventilated patients, nosocomial pneumonia is one of the most serious infectious complications frequently caused by opportunistic pathogens. Pseudomonas aeruginosa is one of the most important causes of ventilator-associated pneumonia as well as the major cause of chronic pneumonia in cystic fibrosis patients. Animal models of pneumonia allow us to investigate distinct types of pneumonia at various disease stages, studies that are not possible in patients. Different animal models of pneumonia such as one-hit acute pneumonia models, ventilator-associated pneumonia models and biofilm pneumonia models associated with cystic fibrosis have been extensively studied and have considerably aided our understanding of disease pathogenesis and testing and developing new treatment strategies. The present review aims to guide investigators in choosing appropriate animal pneumonia models by describing and comparing the relevant characteristics of each model using P. aeruginosa as a model etiology for hospital-acquired pneumonia. Key to establishing and studying these animal models of infection are well-defined end-points that allow precise monitoring and characterization of disease development that could ultimately aid in translating these findings to patient populations in order to guide therapy. In this respect, and discussed here, is the development of humanized animal models of bacterial pneumonia that could offer unique advantages to study bacterial virulence factor expression and host cytokine production for translational purposes.
Collapse
Affiliation(s)
- Kenny Bielen
- Molecular Pathology Group, Faculty of Medicine and Health Sciences, Laboratory of Cell Biology and Histology, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium.,Laboratory of Medical Microbiology - Vaccine and Infectious Disease Institute, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium
| | - Bart 's Jongers
- Molecular Pathology Group, Faculty of Medicine and Health Sciences, Laboratory of Cell Biology and Histology, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium.,Laboratory of Medical Microbiology - Vaccine and Infectious Disease Institute, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium
| | - Surbhi Malhotra-Kumar
- Laboratory of Medical Microbiology - Vaccine and Infectious Disease Institute, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium
| | - Philippe G Jorens
- Department of Critical Care Medicine, Antwerp University Hospital and University of Antwerp, LEMP, Wilrijkstraat 10, B-2650 Edegem, Belgium
| | - Herman Goossens
- Laboratory of Medical Microbiology - Vaccine and Infectious Disease Institute, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium
| | - Samir Kumar-Singh
- Molecular Pathology Group, Faculty of Medicine and Health Sciences, Laboratory of Cell Biology and Histology, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium.,Laboratory of Medical Microbiology - Vaccine and Infectious Disease Institute, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium
| |
Collapse
|
30
|
New Mouse Model for Chronic Infections by Gram-Negative Bacteria Enabling the Study of Anti-Infective Efficacy and Host-Microbe Interactions. mBio 2017; 8:mBio.00140-17. [PMID: 28246361 PMCID: PMC5347345 DOI: 10.1128/mbio.00140-17] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Only a few, relatively cumbersome animal models enable long-term Gram-negative bacterial infections that mimic human situations, where untreated infections can last for weeks. Here, we describe a simple murine cutaneous abscess model that enables chronic or progressive infections, depending on the subcutaneously injected bacterial strain. In this model, Pseudomonas aeruginosa cystic fibrosis epidemic isolate LESB58 caused localized high-density skin and soft tissue infections and necrotic skin lesions for up to 10 days but did not disseminate in either CD-1 or C57BL/6 mice. The model was adapted for use with four major Gram-negative nosocomial pathogens, Acinetobacter baumannii, Klebsiella pneumoniae, Enterobacter cloacae, and Escherichia coli. This model enabled noninvasive imaging and tracking of lux-tagged bacteria, the influx of activated neutrophils, and production of reactive oxygen-nitrogen species at the infection site. Screening antimicrobials against high-density infections showed that local but not intravenous administration of gentamicin, ciprofloxacin, and meropenem significantly but incompletely reduced bacterial counts and superficial tissue dermonecrosis. Bacterial RNA isolated from the abscess tissue revealed that Pseudomonas genes involved in iron uptake, toxin production, surface lipopolysaccharide regulation, adherence, and lipase production were highly upregulated whereas phenazine production and expression of global activator gacA were downregulated. The model was validated for studying virulence using mutants of more-virulent P. aeruginosa strain PA14. Thus, mutants defective in flagella or motility, type III secretion, or siderophore biosynthesis were noninvasive and suppressed dermal necrosis in mice, while a strain with a mutation in the bfiS gene encoding a sensor kinase showed enhanced invasiveness and mortality in mice compared to controls infected with wild-type P. aeruginosa PA14. More than two-thirds of hospital infections are chronic or high-density biofilm infections and difficult to treat due to adaptive, multidrug resistance. Unfortunately, current models of chronic infection are technically challenging and difficult to track without sacrificing animals. Here we describe a model of chronic subcutaneous infection and abscess formation by medically important nosocomial Gram-negative pathogens that is simple and can be used for tracking infections by imaging, examining pathology and immune responses, testing antimicrobial treatments suitable for high-density bacterial infections, and studying virulence. We propose that this mouse model can be a game changer for modeling hard-to-treat Gram-negative bacterial chronic and skin infections.
Collapse
|
31
|
A murine model of early Pseudomonas aeruginosa lung disease with transition to chronic infection. Sci Rep 2016; 6:35838. [PMID: 27804985 PMCID: PMC5090221 DOI: 10.1038/srep35838] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 09/12/2016] [Indexed: 12/02/2022] Open
Abstract
Pseudomonas aeruginosa (PA) remains an important pathogen in patients with cystic fibrosis (CF) lung disease as well as non-CF bronchiectasis and chronic obstructive airways disease. Initial infections are cleared but chronic infection with mucoid strains ensues in the majority of CF patients and specific interventions to prevent this critical infection transition are lacking. The PA bead model has been widely used to study pulmonary P.aeruginosa infection but has limitations in animal husbandry and in accurately mimicking human disease. We have developed an adapted agar bead murine model using a clinical mucoid strain that demonstrates the key features of transition from transitory to chronic airways infection. Infected animals show very limited acute morbidity and mortality, but undergo infection-related weight loss and neutrophilic inflammation, development of anti-pseudomonal antibodies, variable bacterial clearance, endobronchial infection and microbial adaptation with PA small colony variants. We anticipate this model will allow research into the host and microbial factors governing this critical period in Pseudomonas aeruginosa pulmonary pathogenesis when transition to chronicity is occurring.
Collapse
|
32
|
Lorenz A, Pawar V, Häussler S, Weiss S. Insights into host-pathogen interactions from state-of-the-art animal models of respiratory Pseudomonas aeruginosa infections. FEBS Lett 2016; 590:3941-3959. [PMID: 27730639 DOI: 10.1002/1873-3468.12454] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Revised: 10/04/2016] [Accepted: 10/07/2016] [Indexed: 12/29/2022]
Abstract
Pseudomonas aeruginosa is an important opportunistic pathogen that can cause acute respiratory infections in immunocompetent patients or chronic infections in immunocompromised individuals and in patients with cystic fibrosis. When acquiring the chronic infection state, bacteria are encapsulated within biofilm structures enabling them to withstand diverse environmental assaults, including immune reactions and antimicrobial therapy. Understanding the molecular interactions within the bacteria, as well as with the host or other bacteria, is essential for developing innovative treatment strategies. Such knowledge might be accumulated in vitro. However, it is ultimately necessary to confirm these findings in vivo. In the present Review, we describe state-of-the-art in vivo models that allow studying P. aeruginosa infections in molecular detail. The portrayed mammalian models exclusively focus on respiratory infections. The data obtained by alternative animal models which lack lung tissue, often provide molecular insights that are easily transferable to mammals. Importantly, these surrogate in vivo systems reveal complex molecular interactions of P. aeruginosa with the host. Herein, we also provide a critical assessment of the advantages and disadvantages of such models.
Collapse
Affiliation(s)
- Anne Lorenz
- Institute for Molecular Bacteriology, Center of Clinical and Experimental Infection Research, TWINCORE GmbH, A Joint Venture of the Hannover Medical School and the Helmholtz Center for Infection Research, Germany
| | - Vinay Pawar
- Department of Molecular Bacteriology, Helmholtz Center for Infection Research, Braunschweig, Germany.,Department of Molecular Immunology, Helmholtz Center for Infection Research, Braunschweig, Germany.,Institute of Immunology, Medical School Hannover, Germany
| | - Susanne Häussler
- Institute for Molecular Bacteriology, Center of Clinical and Experimental Infection Research, TWINCORE GmbH, A Joint Venture of the Hannover Medical School and the Helmholtz Center for Infection Research, Germany.,Department of Molecular Bacteriology, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Siegfried Weiss
- Department of Molecular Immunology, Helmholtz Center for Infection Research, Braunschweig, Germany.,Institute of Immunology, Medical School Hannover, Germany
| |
Collapse
|
33
|
Harrison F, Diggle SP. An ex vivo lung model to study bronchioles infected with Pseudomonas aeruginosa biofilms. Microbiology (Reading) 2016; 162:1755-1760. [DOI: 10.1099/mic.0.000352] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Freya Harrison
- School of Life Sciences, University of Warwick, Coventry, UK
- Centre for Biomolecular Sciences, School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Stephen P. Diggle
- Centre for Biomolecular Sciences, School of Life Sciences, University of Nottingham, Nottingham, UK
| |
Collapse
|
34
|
The fermentation product 2,3-butanediol alters P. aeruginosa clearance, cytokine response and the lung microbiome. ISME JOURNAL 2016; 10:2978-2983. [PMID: 27177192 DOI: 10.1038/ismej.2016.76] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 03/31/2016] [Accepted: 04/04/2016] [Indexed: 12/12/2022]
Abstract
Diseases that favor colonization of the respiratory tract with Pseudomonas aeruginosa are characterized by an altered airway microbiome. Virulence of P. aeruginosa respiratory tract infection is likely influenced by interactions with other lung microbiota or their products. The bacterial fermentation product 2,3-butanediol enhances virulence and biofilm formation of P. aeruginosa in vitro. This study assessed the effects of 2,3-butanediol on P. aeruginosa persistence, inflammatory response, and the lung microbiome in vivo. Here, P. aeruginosa grown in the presence of 2,3-butanediol and encapsulated in agar beads persisted longer in the murine respiratory tract, induced enhanced TNF-α and IL-6 responses and resulted in increased colonization in the lung tissue by environmental microbes. These results led to the following hypothesis that now needs to be tested with a larger study: fermentation products from the lung microbiota not only have a role in P. aeruginosa virulence and abundance, but also on the increased colonization of the respiratory tract with environmental microbes, resulting in dynamic shifts in microbiota diversity and disease susceptibility.
Collapse
|
35
|
Roberts AE, Kragh KN, Bjarnsholt T, Diggle SP. The Limitations of In Vitro Experimentation in Understanding Biofilms and Chronic Infection. J Mol Biol 2015; 427:3646-61. [DOI: 10.1016/j.jmb.2015.09.002] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 08/12/2015] [Accepted: 09/01/2015] [Indexed: 11/28/2022]
|
36
|
Pathophysiological changes induced by Pseudomonas aeruginosa infection are involved in MMP-12 and MMP-13 upregulation in human carcinoma epithelial cells and a pneumonia mouse model. Infect Immun 2015; 83:4791-9. [PMID: 26438797 PMCID: PMC4645383 DOI: 10.1128/iai.00619-15] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 09/18/2015] [Indexed: 12/20/2022] Open
Abstract
Pseudomonas aeruginosa infections persist in patients with cystic fibrosis (CF) and drive lung disease progression. P. aeruginosa potently activates the innate immune system mostly through the recognition of pathogen-associated molecular patterns, such as flagellin. Matrix metalloproteinases 12 and 13 (MMP-12 and MMP-13, respectively) exacerbate chronic lung infection and inflammation by promoting uncontrolled tissue rearrangements and fibrosis, yet the underlying molecular mechanisms by which this occurs remain largely unknown. In this study, we used quantitative bacteriology, histological examination, and proinflammatory cytokine levels to evaluate the effects of MMP-12 and MMP-13 on P. aeruginosa strain K-induced infection and pneumonia in H292 epithelial cells and mice, respectively. Under inflammatory stimulation, mRNA and protein expression levels of proinflammatory mediators were higher in strain K-infected mice and cells than in uninfected counterparts, in which MMP-12 and MMP-13 expression reached levels similar to those observed in epithelial cells. Moreover, we also found that the NF-κB pathway might be involved in the induction of cytokines in response to strain K infection. Taken together, these data suggest that MMP-12 and MMP-13 alter strain K infection in mice and play a role in inflammatory regulation by modulating cytokine levels.
Collapse
|
37
|
Pseudomonas aeruginosa adaptation in the nasopharyngeal reservoir leads to migration and persistence in the lungs. Nat Commun 2014; 5:4780. [PMID: 25179232 DOI: 10.1038/ncomms5780] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 07/23/2014] [Indexed: 12/27/2022] Open
Abstract
Chronic bacterial infections are a key feature of a variety of lung conditions. The opportunistic bacterium, Pseudomonas aeruginosa, is extremely skilled at both colonizing and persisting in the airways of patients with lung damage. It has been suggested that the upper airways (including the paranasal sinuses and nasopharynx) play an important role as a silent reservoir of bacteria. Over time, P. aeruginosa can adapt to its niche, leading to increased resistance in the face of the immune system and intense therapy regimes. Here we describe a mouse inhalation model of P. aeruginosa chronic infection that can be studied for at least 28 days. We present evidence for adaptation in vivo, in terms of genotype and phenotype including antibiotic resistance. Our data suggest that there is persistence in the upper respiratory tract and that this is key in the establishment of lung infection. This model provides a unique platform for studying evolutionary dynamics and therapeutics.
Collapse
|
38
|
Development of an ex vivo porcine lung model for studying growth, virulence, and signaling of Pseudomonas aeruginosa. Infect Immun 2014; 82:3312-23. [PMID: 24866798 PMCID: PMC4136229 DOI: 10.1128/iai.01554-14] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Research into chronic infection by bacterial pathogens, such as Pseudomonas aeruginosa, uses various in vitro and live host models. While these have increased our understanding of pathogen growth, virulence, and evolution, each model has certain limitations. In vitro models cannot recapitulate the complex spatial structure of host organs, while experiments on live hosts are limited in terms of sample size and infection duration for ethical reasons; live mammal models also require specialized facilities which are costly to run. To address this, we have developed an ex vivo pig lung (EVPL) model for quantifying Pseudomonas aeruginosa growth, quorum sensing (QS), virulence factor production, and tissue damage in an environment that mimics a chronically infected cystic fibrosis (CF) lung. In a first test of our model, we show that lasR mutants, which do not respond to 3-oxo-C12-homoserine lactone (HSL)-mediated QS, exhibit reduced virulence factor production in EVPL. We also show that lasR mutants grow as well as or better than a corresponding wild-type strain in EVPL. lasR mutants frequently and repeatedly arise during chronic CF lung infection, but the evolutionary forces governing their appearance and spread are not clear. Our data are not consistent with the hypothesis that lasR mutants act as social “cheats” in the lung; rather, our results support the hypothesis that lasR mutants are more adapted to the lung environment. More generally, this model will facilitate improved studies of microbial disease, especially studies of how cells of the same and different species interact in polymicrobial infections in a spatially structured environment.
Collapse
|
39
|
Titz A. Carbohydrate-Based Anti-Virulence Compounds Against Chronic Pseudomonas aeruginosa Infections with a Focus on Small Molecules. TOPICS IN MEDICINAL CHEMISTRY 2014. [DOI: 10.1007/7355_2014_44] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
40
|
Kukavica-Ibrulj I, Facchini M, Cigana C, Levesque RC, Bragonzi A. Assessing Pseudomonas aeruginosa virulence and the host response using murine models of acute and chronic lung infection. Methods Mol Biol 2014; 1149:757-71. [PMID: 24818948 DOI: 10.1007/978-1-4939-0473-0_58] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Murine models of acute and chronic lung infection have been used in studying Pseudomonas aeruginosa for assessing in vivo behavior and for monitoring of the host response. These models provide an important resource for studies of the initiation and maintenance of bacterial infection, identify bacterial genes essential for in vivo maintenance and for the development and testing of new therapies. The rat has been used extensively as a model of chronic lung infection, whereas the mouse has been a model of acute and chronic infection. Intratracheal administration of planktonic bacterial cells in the mouse provides a model of acute pneumonia. Bacteria enmeshed in agar beads can be used in the rat and mouse to reproduce the lung pathology of cystic fibrosis patients with advanced chronic pulmonary disease. Here, we describe the methods to assess virulence of P. aeruginosa using prototype and clinical strains in the Sprague-Dawley rat and the C57BL/6NCrlBR mouse by monitoring several measurable read-outs including weight loss, mortality, in vivo growth curves, the competitive index of infectivity, and the inflammatory response.
Collapse
Affiliation(s)
- Irena Kukavica-Ibrulj
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, 1030 av. de la médecine, Québec, QC, Canada, G1V 0A6
| | | | | | | | | |
Collapse
|
41
|
Requirement of the Pseudomonas aeruginosa CbrA sensor kinase for full virulence in a murine acute lung infection model. Infect Immun 2013; 82:1256-67. [PMID: 24379284 DOI: 10.1128/iai.01527-13] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen that is a major cause of respiratory tract and other nosocomial infections. The sensor kinase CbrA is a central regulator of carbon and nitrogen metabolism and in vitro also regulates virulence-related processes in P. aeruginosa. Here, we investigated the role of CbrA in two murine models of infection. In both peritoneal infections in leukopenic mice and lung infection models, the cbrA mutant was less virulent since substantially larger numbers of cbrA mutant bacteria were required to cause the same level of infection as wild-type or complemented bacteria. In contrast, in the chronic rat lung model the cbrA mutant grew and persisted as well as the wild type, indicating that the decrease of in vivo virulence of the cbrA mutant did not result from growth deficiencies on particular carbon substrates observed in vitro. In addition, a mutant in the cognate response regulator CbrB showed no defect in virulence in the peritoneal infection model, ruling out the involvement of certain alterations of virulence properties in the cbrA mutant including defective swarming motility, increased biofilm formation, and cytotoxicity, since these alterations are controlled through CbrB. Further investigations indicated that the mutant was more susceptible to uptake by phagocytes in vitro, resulting in greater overall bacterial killing. Consistent with the virulence defect, it took a smaller number of Dictyostelium discoideum amoebae to kill the cbrA mutant than to kill the wild type. Transcriptional analysis of the cbrA mutant during D. discoideum infection led to the conclusion that CbrA played an important role in the iron metabolism, protection of P. aeruginosa against oxidative stress, and the regulation of certain virulence factors.
Collapse
|
42
|
Choosing an appropriate infection model to study quorum sensing inhibition in Pseudomonas infections. Int J Mol Sci 2013; 14:19309-40. [PMID: 24065108 PMCID: PMC3794835 DOI: 10.3390/ijms140919309] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 09/13/2013] [Accepted: 09/17/2013] [Indexed: 02/07/2023] Open
Abstract
Bacteria, although considered for decades to be antisocial organisms whose sole purpose is to find nutrients and multiply are, in fact, highly communicative organisms. Referred to as quorum sensing, cell-to-cell communication mechanisms have been adopted by bacteria in order to co-ordinate their gene expression. By behaving as a community rather than as individuals, bacteria can simultaneously switch on their virulence factor production and establish successful infections in eukaryotes. Understanding pathogen-host interactions requires the use of infection models. As the use of rodents is limited, for ethical considerations and the high costs associated with their use, alternative models based on invertebrates have been developed. Invertebrate models have the benefits of low handling costs, limited space requirements and rapid generation of results. This review presents examples of such models available for studying the pathogenicity of the Gram-negative bacterium Pseudomonas aeruginosa. Quorum sensing interference, known as quorum quenching, suggests a promising disease-control strategy since quorum-quenching mechanisms appear to play important roles in microbe-microbe and host-pathogen interactions. Examples of natural and synthetic quorum sensing inhibitors and their potential as antimicrobials in Pseudomonas-related infections are discussed in the second part of this review.
Collapse
|
43
|
Starkey MR, Jarnicki AG, Essilfie AT, Gellatly SL, Kim RY, Brown AC, Foster PS, Horvat JC, Hansbro PM. Murine models of infectious exacerbations of airway inflammation. Curr Opin Pharmacol 2013; 13:337-44. [PMID: 23566696 DOI: 10.1016/j.coph.2013.03.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Revised: 03/08/2013] [Accepted: 03/14/2013] [Indexed: 12/24/2022]
Abstract
Airway inflammation underpins the pathogenesis of the major human chronic respiratory diseases. It is now well recognized that respiratory infections with bacteria and viruses are important in the induction, progression and exacerbation of these diseases. There are no effective therapies that prevent or reverse these events. The development and use of mouse models are proving valuable in understanding the role of infection in disease pathogenesis. They have recently been used to show that infections in early life alter immune responses and lung structure to increase asthma severity, and alter immune responses in later life to induce steroid resistance. Infection following smoke exposure or in experimental chronic obstructive pulmonary disease exacerbates inflammation and remodeling, and worsens cystic fibrosis. Further exploration of these models will facilitate the identification of new therapeutic approaches and the testing of new preventions and treatments.
Collapse
Affiliation(s)
- Malcolm Ronald Starkey
- Centre for Asthma and Respiratory Disease, The Hunter Medical Research Institute and The University of Newcastle, Newcastle, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Galle M, Carpentier I, Beyaert R. Structure and function of the Type III secretion system of Pseudomonas aeruginosa. Curr Protein Pept Sci 2012; 13:831-42. [PMID: 23305368 PMCID: PMC3706959 DOI: 10.2174/138920312804871210] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Revised: 07/19/2012] [Accepted: 07/25/2012] [Indexed: 02/08/2023]
Abstract
Pseudomonas aeruginosa is a dangerous pathogen particularly because it harbors multiple virulence factors. It causes several types of infection, including dermatitis, endocarditis, and infections of the urinary tract, eye, ear, bone, joints and, of particular interest, the respiratory tract. Patients with cystic fibrosis, who are extremely susceptible to Pseudomonas infections, have a bad prognosis and high mortality. An important virulence factor of P. aeruginosa, shared with many other gram-negative bacteria, is the type III secretion system, a hollow molecular needle that transfers effector toxins directly from the bacterium into the host cell cytosol. This complex macromolecular machine works in a highly regulated manner and can manipulate the host cell in many different ways. Here we review the current knowledge of the structure of the P. aeruginosa T3SS, as well as its function and recognition by the immune system. Furthermore, we describe recent progress in the development and use of therapeutic agents targeting the T3SS.
Collapse
Affiliation(s)
- Marlies Galle
- Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, B-9052 Ghent, Belgium; the
- Department for Molecular Biomedical Research, Unit of Molecular Signal Transduction in Inflammation, VIB, Technologiepark 927, B-9052 Ghent, Belgium
| | - Isabelle Carpentier
- Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, B-9052 Ghent, Belgium; the
- Department for Molecular Biomedical Research, Unit of Molecular Signal Transduction in Inflammation, VIB, Technologiepark 927, B-9052 Ghent, Belgium
| | - Rudi Beyaert
- Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, B-9052 Ghent, Belgium; the
- Department for Molecular Biomedical Research, Unit of Molecular Signal Transduction in Inflammation, VIB, Technologiepark 927, B-9052 Ghent, Belgium
| |
Collapse
|
45
|
Efficacy of liposomal bismuth-ethanedithiol-loaded tobramycin after intratracheal administration in rats with pulmonary Pseudomonas aeruginosa infection. Antimicrob Agents Chemother 2012; 57:569-78. [PMID: 23147741 DOI: 10.1128/aac.01634-12] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
We sought to investigate alterations in quorum-sensing signal molecule N-acyl homoserine lactone secretion and in the release of Pseudomonas aeruginosa virulence factors, as well as the in vivo antimicrobial activity of bismuth-ethanedithiol incorporated into a liposome-loaded tobramycin formulation (LipoBiEDT-TOB) administered to rats chronically infected with P. aeruginosa. The quorum-sensing signal molecule N-acyl homoserine lactone was monitored by using a biosensor organism. P. aeruginosa virulence factors were assessed spectrophotometrically. An agar beads model of chronic Pseudomonas lung infection in rats was used to evaluate the efficacy of the liposomal formulation in the reduction of bacterial count. The levels of active tobramycin in the lungs and the kidneys were evaluated by microbiological assay. LipoBiEDT-TOB was effective in disrupting both quorum-sensing signal molecules N-3-oxo-dodeccanoylhomoserine lactone and N-butanoylhomoserine lactone, as well as significantly (P < 0.05) reducing lipase, chitinase, and protease production. At 24 h after 3 treatments, the CFU counts in lungs of animals treated with LipoBiEDT-TOB were of 3 log(10) CFU/lung, comparated to 7.4 and 4.7 log(10) CFU/lung, respectively, in untreated lungs and in lungs treated with free antibiotic. The antibiotic concentration after the last dose of LipoBiEDT-TOB was 25.1 μg/lung, while no tobramycin was detected in the kidneys. As for the free antibiotic, we found 6.5 μg/kidney but could not detect any tobramycin in the lungs. Taken together, LipoBiEDT-TOB reduced the production of quorum-sensing molecules and virulence factors and could highly improve the management of chronic pulmonary infection in cystic fibrosis patients.
Collapse
|
46
|
Skolimowski M, Weiss Nielsen M, Abeille F, Skafte-Pedersen P, Sabourin D, Fercher A, Papkovsky D, Molin S, Taboryski R, Sternberg C, Dufva M, Geschke O, Emnéus J. Modular microfluidic system as a model of cystic fibrosis airways. BIOMICROFLUIDICS 2012; 6:34109. [PMID: 23908680 PMCID: PMC3423306 DOI: 10.1063/1.4742911] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Accepted: 07/24/2012] [Indexed: 05/13/2023]
Abstract
A modular microfluidic airways model system that can simulate the changes in oxygen tension in different compartments of the cystic fibrosis (CF) airways was designed, developed, and tested. The fully reconfigurable system composed of modules with different functionalities: multichannel peristaltic pumps, bubble traps, gas exchange chip, and cell culture chambers. We have successfully applied this system for studying the antibiotic therapy of Pseudomonas aeruginosa, the bacteria mainly responsible for morbidity and mortality in cystic fibrosis, in different oxygen environments. Furthermore, we have mimicked the bacterial reinoculation of the aerobic compartments (lower respiratory tract) from the anaerobic compartments (cystic fibrosis sinuses) following an antibiotic treatment. This effect is hypothesised as the one on the main reasons for recurrent lung infections in cystic fibrosis patients.
Collapse
Affiliation(s)
- M Skolimowski
- Department of Micro- and Nanotechnology, Technical University of Denmark, Ørsted Plads, Building 345B, Kgs. Lyngby DK-2800, Denmark ; Department of Systems Biology, Technical University of Denmark, Matematiktorvet, Building 301, Kgs. Lyngby DK-2800, Denmark
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Nowakowska E, Schulz T, Molenda N, Schillers H, Prehm P. Recovery of ΔF508-CFTR function by analogs of hyaluronan disaccharide. J Cell Biochem 2012; 113:156-64. [PMID: 21882224 DOI: 10.1002/jcb.23339] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
We recently discovered that hyaluronan was exported from fibroblasts by MRP5 and from epithelial cells by cystic fibrosis (CF) transmembrane conductance regulator (CFTR) that was known as a chloride channel. On this basis we developed membrane permeable analogs of hyaluronan disaccharide as new class of compounds to modify their efflux. We found substances that activated hyaluronan export from human breast cancer cells. The most active compound 2-(2-acetamido-3,5-dihydroxyphenoxy)-5-aminobenzoic acid (Hylout4) was tested for its influence on the activity of epithelial cells. It activated the ion efflux by normal and defective ΔF508-CFTR. It also enhanced the plasma membrane concentration of the ΔF508-CFTR protein and reduced the transepithelial resistance of epithelial cells. In human trials of healthy persons, it caused an opening of CFTR in the nasal epithelium. Thus compound Hylout4 is a corrector that recovered ΔF508-CFTR from intracellular degradation and activated its export function.
Collapse
Affiliation(s)
- Ewa Nowakowska
- Muenster University Hospital, Institute of Physiological Chemistry and Pathobiochemistry, Waldeyerstr. 15, D-48129 Muenster, Germany
| | | | | | | | | |
Collapse
|
48
|
Gould NS, Min E, Martin RJ, Day BJ. CFTR is the primary known apical glutathione transporter involved in cigarette smoke-induced adaptive responses in the lung. Free Radic Biol Med 2012; 52:1201-6. [PMID: 22266045 PMCID: PMC3920665 DOI: 10.1016/j.freeradbiomed.2012.01.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 12/14/2011] [Accepted: 01/04/2012] [Indexed: 10/14/2022]
Abstract
One of the most abundant antioxidants in the lung is glutathione (GSH), a low-molecular-weight thiol, which functions to attenuate both oxidative stress and inflammation. GSH is concentrated in the epithelial lining fluid (ELF) of the lung and can be elevated in response to the increased oxidant burden from cigarette smoke (CS). However, the transporter(s) responsible for the increase in ELF GSH with cigarette smoke is not known. Three candidate apical GSH transporters in the lung are CFTR, BCRP, and MRP2, but their potential roles in ELF GSH transport in response to CS have not been investigated. In vitro, the inhibition of CFTR, BCRP, or MRP2 resulted in decreased GSH efflux in response to cigarette smoke extract. In vivo, mice deficient in CFTR, BCRP, or MRP2 were exposed to either air or acute CS. CFTR-deficient mice had reduced basal and CS-induced GSH in the ELF, whereas BCRP or MRP2 deficiency had no effect on ELF GSH basal or CS-exposed levels. Furthermore, BCRP or MRP2 deficiency had little effect on lung tissue GSH. These data indicate that CFTR is predominantly involved in maintaining basal ELF GSH and increasing ELF GSH in response to CS.
Collapse
Affiliation(s)
- Neal S. Gould
- Department of Medicine, National Jewish Health, Denver, CO
- Department of Pharmaceutical Sciences, University of Colorado Denver, Aurora, CO
| | - Elysia Min
- Department of Medicine, National Jewish Health, Denver, CO
| | - Richard J. Martin
- Department of Medicine, National Jewish Health, Denver, CO
- Department of Medicine, University of Colorado Denver, Aurora, CO
| | - Brian J. Day
- Department of Medicine, National Jewish Health, Denver, CO
- Department of Pharmaceutical Sciences, University of Colorado Denver, Aurora, CO
- Department of Medicine, University of Colorado Denver, Aurora, CO
- Department of Immunology, University of Colorado Denver, Aurora, CO
| |
Collapse
|
49
|
N-acetylcysteine enhances cystic fibrosis sputum penetration and airway gene transfer by highly compacted DNA nanoparticles. Mol Ther 2011; 19:1981-9. [PMID: 21829177 DOI: 10.1038/mt.2011.160] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
For effective airway gene therapy of cystic fibrosis (CF), inhaled gene carriers must first penetrate the hyperviscoelastic sputum covering the epithelium. Whether clinically studied gene carriers can penetrate CF sputum remains unknown. Here, we measured the diffusion of a clinically tested nonviral gene carrier, composed of poly-l-lysine conjugated with a 10 kDa polyethylene glycol segment (CK(30)PEG(10k)). We found that CK(30)PEG(10k)/DNA nanoparticles were trapped in CF sputum. To improve gene carrier diffusion across sputum, we tested adjuvant regimens consisting of N-acetylcysteine (NAC), recombinant human DNase (rhDNase) or NAC together with rhDNase. While rhDNase alone did not enhance gene carrier diffusion, NAC and NAC + rhDNase increased average effective diffusivities by 6-fold and 13-fold, respectively, leading to markedly greater fractions of gene carriers that may penetrate sputum layers. We further tested the adjuvant effects of NAC in the airways of mice with Pseudomonas aeruginosa lipopolysaccharide (LPS)-induced mucus hypersecretion. Intranasal dosing of NAC prior to CK(30)PEG(10k)/DNA nanoparticles enhanced gene expression by up to ~12-fold compared to saline control, reaching levels observed in the lungs of mice without LPS challenge. Our findings suggest that a promising synthetic nanoparticle gene carrier may transfer genes substantially more effectively to lungs of CF patients if administered following adjuvant mucolytic therapy with NAC or NAC + rhDNase.
Collapse
|
50
|
Sibley CD, Surette MG. The polymicrobial nature of airway infections in cystic fibrosis: Cangene Gold Medal Lecture. Can J Microbiol 2011; 57:69-77. [PMID: 21326348 DOI: 10.1139/w10-105] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Microbial communities characterize the airways of cystic fibrosis (CF) patients. Members of these diverse and dynamic communities can be thought of as pathogens, benign commensals, or synergens--organisms not considered pathogens in the traditional sense but with the capacity to alter the pathogenesis of the community through microbe-microbe or polymicrobe-host interactions. Very few bacterial pathogens have been implicated as clinically relevant in CF; however, the CF airway microbiome can be a reservoir of previously unrecognized but clinically relevant organisms. A combination of culture-dependent and culture-independent approaches provides a more comprehensive perspective of CF microbiology than either approach alone. Here we review these concepts, highlight the future challenges for CF microbiology, and discuss the implications for the management of CF airway infections. We suggest that the success of treatment interventions for chronic CF lung disease will rely on the context of the microbes within microbial communities. The microbiology of CF airways may serve as a model to investigate the emergent properties of other clinically relevant microbial communities in the human body.
Collapse
Affiliation(s)
- Christopher D Sibley
- Department of Microbiology and Infectious Diseases, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada.
| | | |
Collapse
|