1
|
Meirelles LA, Vayena E, Debache A, Schmidt E, Rossy T, Distler T, Hatzimanikatis V, Persat A. Pseudomonas aeruginosa faces a fitness trade-off between mucosal colonization and antibiotic tolerance during airway infection. Nat Microbiol 2024; 9:3284-3303. [PMID: 39455898 DOI: 10.1038/s41564-024-01842-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024]
Abstract
Pseudomonas aeruginosa frequently causes antibiotic-recalcitrant pneumonia, but the mechanisms driving its adaptation during human infections remain unclear. To reveal the selective pressures and adaptation strategies at the mucosal surface, here we investigated P. aeruginosa growth and antibiotic tolerance in tissue-engineered airways by transposon insertion sequencing (Tn-seq). Metabolic modelling based on Tn-seq data revealed the nutritional requirements for P. aeruginosa growth, highlighting reliance on glucose and lactate and varying requirements for amino acid biosynthesis. Tn-seq also revealed selection against biofilm formation during mucosal growth in the absence of antibiotics. Live imaging in engineered organoids showed that biofilm-dwelling cells remained sessile while colonizing the mucosal surface, limiting nutrient foraging and reduced growth. Conversely, biofilm formation increased antibiotic tolerance at the mucosal surface. Moreover, mutants with exacerbated biofilm phenotypes protected less tolerant but more cytotoxic strains, contributing to phenotypic heterogeneity. P. aeruginosa must therefore navigate conflicting physical and biological selective pressures to establish chronic infections.
Collapse
Affiliation(s)
- Lucas A Meirelles
- Global Health Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Evangelia Vayena
- Laboratory of Computational Systems Biotechnology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Auriane Debache
- Global Health Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Eric Schmidt
- Global Health Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Tamara Rossy
- Global Health Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Tania Distler
- Global Health Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Vassily Hatzimanikatis
- Laboratory of Computational Systems Biotechnology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Alexandre Persat
- Global Health Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
2
|
Eklöf J, Alispahic IA, Armbruster K, Lapperre TS, Browatzki A, Overgaard RH, Harboe ZB, Janner J, Moberg M, Ulrik CS, Andreassen HF, Weinreich UM, Kjærgaard JL, Villadsen J, Fenlev CS, Jensen TT, Christensen CW, Bangsborg J, Ostergaard C, Ghathian KSA, Jordan A, Klausen TW, Nielsen TL, Wilcke T, Seersholm N, Sivapalan P, Jensen JUS. Systemic antibiotics for Pseudomonas aeruginosa infection in outpatients with non-hospitalised exacerbations of pre-existing lung diseases: a randomised clinical trial. Respir Res 2024; 25:236. [PMID: 38844921 PMCID: PMC11157704 DOI: 10.1186/s12931-024-02860-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 05/29/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND The effect of dual systemic antibiotic therapy against Pseudomonas aeruginosa in patients with pre-existing lung disease is unknown. To assess whether dual systemic antibiotics against P. aeruginosa in outpatients with COPD, non-cystic fibrosis (non-CF) bronchiectasis, or asthma can improve outcomes. METHODS Multicenter, randomised, open-label trial conducted at seven respiratory outpatient clinics in Denmark. Outpatients with COPD, non-CF bronchiectasis, or asthma with a current P. aeruginosa-positive lower respiratory tract culture (clinical routine samples obtained based on symptoms of exacerbation not requiring hospitalisation), regardless of prior P. aeruginosa-status, no current need for hospitalisation, and at least two moderate or one hospitalisation-requiring exacerbation within the last year were eligible. Patients were assigned 1:1 to 14 days of dual systemic anti-pseudomonal antibiotics or no antibiotic treatment. Primary outcome was time to prednisolone or antibiotic-requiring exacerbation or death from day 20 to day 365. RESULTS The trial was stopped prematurely based in lack of recruitment during the COVID-19 pandemic, this decision was endorsed by the Data and Safety Monitoring Board. Forty-nine outpatients were included in the study. There was a reduction in risk of the primary outcome in the antibiotic group compared to the control group (HR 0.51 (95%CI 0.27-0.96), p = 0.037). The incidence of admissions with exacerbation within one year was 1.1 (95%CI 0.6-1.7) in the dual antibiotic group vs. 2.9 (95%CI 1.3-4.5) in the control group, p = 0.037. CONCLUSIONS Use of dual systemic antibiotics for 14 days against P. aeruginosa in outpatients with chronic lung diseases and no judged need for hospitalisation, improved clinical outcomes markedly. The main limitation was the premature closure of the trial. TRIAL REGISTRATION ClinicalTrials.gov, NCT03262142, registration date 2017-08-25.
Collapse
Affiliation(s)
- Josefin Eklöf
- Department of Internal Medicine, Herlev Gentofte University Hospital, Section of Respiratory Medicine, Copenhagen University Hospital, Herlev Gentofte, Hellerup, Denmark.
| | - Imane Achir Alispahic
- Department of Internal Medicine, Herlev Gentofte University Hospital, Section of Respiratory Medicine, Copenhagen University Hospital, Herlev Gentofte, Hellerup, Denmark
| | - Karin Armbruster
- Department of Internal Medicine, Herlev Gentofte University Hospital, Section of Respiratory Medicine, Copenhagen University Hospital, Herlev Gentofte, Hellerup, Denmark
| | - Therese Sophie Lapperre
- Department of Respiratory Medicine and Infectious Diseases, Copenhagen University Hospital, Bispebjerg Frederiksberg, Denmark
- Department of Respiratory Medicine, Antwerp University Hospital, Antwerp, Belgium
- Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Antwerp, Belgium
| | - Andrea Browatzki
- Department of Respiratory Medicine and Infectious Diseases, Copenhagen University Hospital, North Zealand, Denmark
| | - Rikke Holmen Overgaard
- Department of Respiratory Medicine and Infectious Diseases, Copenhagen University Hospital, North Zealand, Denmark
| | - Zitta Barrella Harboe
- Department of Respiratory Medicine and Infectious Diseases, Copenhagen University Hospital, North Zealand, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Julie Janner
- Department of Respiratory Medicine, Copenhagen University Hospital, Hvidovre, Denmark
| | - Mia Moberg
- Department of Respiratory Medicine, Copenhagen University Hospital, Hvidovre, Denmark
| | | | - Helle Frost Andreassen
- Department of Respiratory Medicine and Infectious Diseases, Copenhagen University Hospital, Bispebjerg Frederiksberg, Denmark
| | - Ulla Møller Weinreich
- Department of Respiratory Medicine, Aalborg University Hospital and Department of Clinical Medicine, Aalborg, Denmark
| | - Jakob Lyngby Kjærgaard
- Department of Internal Medicine, Herlev Gentofte University Hospital, Section of Respiratory Medicine, Copenhagen University Hospital, Herlev Gentofte, Hellerup, Denmark
| | - Jenny Villadsen
- Department of Internal Medicine, Herlev Gentofte University Hospital, Section of Respiratory Medicine, Copenhagen University Hospital, Herlev Gentofte, Hellerup, Denmark
| | - Camilla Sund Fenlev
- Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Antwerp, Belgium
| | | | | | - Jette Bangsborg
- Department of Clinical Microbiology, Copenhagen University Hospital, Herlev, Denmark
| | - Christian Ostergaard
- Department of Clinical Microbiology, Copenhagen University Hospital, Hvidovre, Denmark
| | | | - Alexander Jordan
- Department of Internal Medicine, Herlev Gentofte University Hospital, Section of Respiratory Medicine, Copenhagen University Hospital, Herlev Gentofte, Hellerup, Denmark
| | - Tobias Wirenfeldt Klausen
- Department of Internal Medicine, Herlev Gentofte University Hospital, Section of Respiratory Medicine, Copenhagen University Hospital, Herlev Gentofte, Hellerup, Denmark
| | - Thyge Lynghøj Nielsen
- Department of Respiratory Medicine and Infectious Diseases, Copenhagen University Hospital, North Zealand, Denmark
| | - Torgny Wilcke
- Department of Internal Medicine, Herlev Gentofte University Hospital, Section of Respiratory Medicine, Copenhagen University Hospital, Herlev Gentofte, Hellerup, Denmark
| | - Niels Seersholm
- Department of Internal Medicine, Herlev Gentofte University Hospital, Section of Respiratory Medicine, Copenhagen University Hospital, Herlev Gentofte, Hellerup, Denmark
| | - Pradeesh Sivapalan
- Department of Internal Medicine, Herlev Gentofte University Hospital, Section of Respiratory Medicine, Copenhagen University Hospital, Herlev Gentofte, Hellerup, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Jens-Ulrik Stæhr Jensen
- Department of Internal Medicine, Herlev Gentofte University Hospital, Section of Respiratory Medicine, Copenhagen University Hospital, Herlev Gentofte, Hellerup, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
3
|
A truncated mutation of MucA in Pseudomonas aeruginosa from a bronchiectasis patient affects T3SS expression and inflammasome activation. Acta Biochim Biophys Sin (Shanghai) 2022; 54:1740-1747. [PMID: 36604139 PMCID: PMC9828237 DOI: 10.3724/abbs.2022169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen that causes chronic airway infection in bronchiectasis patients and is closely associated with poor prognosis. Strains isolated from chronically infected patients typically have a mucoid phenotype due to the overproduction of alginate. In this study, we isolate a P. aeruginosa strain from the sputum of a patient with bronchiectasis and find that a truncated mutation occurred in mucA, which is named mucA117. mucA117 causes the strain to transform into a mucoid phenotype, downregulates the expression of T3SS and inflammasome ligands such as fliC and allows it to avoid inflammasome activation. The truncated mutation of the MucA protein may help P. aeruginosa escape clearance by the immune system, enabling long-term colonization.
Collapse
|
4
|
Filamentous Pseudomonas Phage Pf4 in the Context of Therapy-Inducibility, Infectivity, Lysogenic Conversion, and Potential Application. Viruses 2022; 14:v14061261. [PMID: 35746731 PMCID: PMC9228429 DOI: 10.3390/v14061261] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 05/29/2022] [Accepted: 05/30/2022] [Indexed: 02/04/2023] Open
Abstract
More than 20% of all Pseudomonas aeruginosa are infected with Pf4-related filamentous phage and although their role in virulence of P. aeruginosa strain PAO1 is well documented, its properties related to therapy are not elucidated in detail. The aim of this study was to determine how phage and antibiotic therapy induce Pf4, whether the released virions can infect other strains and how the phage influences the phenotype of new hosts. The subinhibitory concentrations of ciprofloxacin and mitomycin C increased Pf4 production for more than 50% during the first and sixth hour of exposure, respectively, while mutants appearing after infection with obligatory lytic phage at low MOI produced Pf4 more than four times after 12–24 h of treatment. This indicates that production of Pf4 is enhanced during therapy with these agents. The released virions can infect new P. aeruginosa strains, as confirmed for models UCBPP-PA14 (PA14) and LESB58, existing both episomally and in a form of a prophage, as confirmed by PCR, RFLP, and sequencing. The differences in properties of Pf4-infected, and uninfected PA14 and LESB58 strains were obvious, as infection with Pf4 significantly decreased cell autoaggregation, pyoverdine, and pyocyanin production, while significantly increased swimming motility and biofilm production in both strains. In addition, in strain PA14, Pf4 increased cell surface hydrophobicity and small colony variants’ appearance, but also decreased twitching and swarming motility. This indicates that released Pf4 during therapy can infect new strains and cause lysogenic conversion. The infection with Pf4 increased LESB58 sensitivity to ciprofloxacin, gentamicin, ceftazidime, tetracycline, and streptomycin, and PA14 to ciprofloxacin and ceftazidime. Moreover, the Pf4-infected LESB58 was re-sensitized to ceftazidime and tetracycline, with changes from resistant to intermediate resistant and sensitive, respectively. The obtained results open a new field in phage therapy—treatment with selected filamentous phages in order to re-sensitize pathogenic bacteria to certain antibiotics. However, this approach should be considered with precautions, taking into account potential lysogenic conversion.
Collapse
|
5
|
Madden DE, Olagoke O, Baird T, Neill J, Ramsay KA, Fraser TA, Bell SC, Sarovich DS, Price EP. Express Yourself: Quantitative Real-Time PCR Assays for Rapid Chromosomal Antimicrobial Resistance Detection in Pseudomonas aeruginosa. Antimicrob Agents Chemother 2022; 66:e0020422. [PMID: 35467369 PMCID: PMC9112894 DOI: 10.1128/aac.00204-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 03/31/2022] [Indexed: 01/03/2023] Open
Abstract
The rise of antimicrobial-resistant (AMR) bacteria is a global health emergency. One critical facet of tackling this epidemic is more rapid AMR diagnosis in serious multidrug-resistant pathogens like Pseudomonas aeruginosa. Here, we designed and then validated two multiplex quantitative real-time PCR (qPCR) assays to simultaneously detect differential expression of the resistance-nodulation-division efflux pumps MexAB-OprM, MexCD-OprJ, MexEF-OprN, and MexXY-OprM, the AmpC β-lactamase, and the porin OprD, which are commonly associated with chromosomally encoded AMR. Next, qPCRs were tested on 15 sputa from 11 participants with P. aeruginosa respiratory infections to determine AMR profiles in vivo. We confirmed multiplex qPCR testing feasibility directly on sputa, representing a key advancement in in vivo AMR diagnosis. Notably, comparison of sputa with their derived isolates grown in Luria-Bertani broth (±2.5% NaCl) or a 5-antibiotic cocktail showed marked expression differences, illustrating the difficulty in replicating in vivo expression profiles in vitro. Cystic fibrosis sputa showed significantly reduced mexE and mexY expression compared with chronic obstructive pulmonary disease sputa, despite harboring fluoroquinolone- and aminoglycoside-resistant strains, indicating that these loci do not contribute to AMR in vivo. oprD was also significantly downregulated in cystic fibrosis sputa, even in the absence of contemporaneous carbapenem use, suggesting a common adaptive trait in chronic infections that may affect carbapenem efficacy. Sputum ampC expression was highest in participants receiving carbapenems (6.7 to 15×), some of whom were simultaneously receiving cephalosporins, the latter of which would be rendered ineffective by the upregulated ampC. Our qPCR assays provide valuable insights into the P. aeruginosa resistome, and their use on clinical specimens will permit timely treatment alterations that will improve patient outcomes and antimicrobial stewardship measures.
Collapse
Affiliation(s)
- Danielle E. Madden
- Centre for Bioinnovation, University of the Sunshine Coast, Sippy Downs, Queensland, Australia
- Sunshine Coast Health Institute, Birtinya, Queensland, Australia
| | - Olusola Olagoke
- Centre for Bioinnovation, University of the Sunshine Coast, Sippy Downs, Queensland, Australia
- Sunshine Coast Health Institute, Birtinya, Queensland, Australia
| | - Timothy Baird
- Sunshine Coast Health Institute, Birtinya, Queensland, Australia
- Respiratory Department, Sunshine Coast University Hospital, Birtinya, Queensland, Australia
| | - Jane Neill
- Sunshine Coast Health Institute, Birtinya, Queensland, Australia
- Respiratory Department, Sunshine Coast University Hospital, Birtinya, Queensland, Australia
| | - Kay A. Ramsay
- Child Health Research Centre, The University of Queensland, South Brisbane, Queensland, Australia
| | - Tamieka A. Fraser
- Centre for Bioinnovation, University of the Sunshine Coast, Sippy Downs, Queensland, Australia
- Sunshine Coast Health Institute, Birtinya, Queensland, Australia
| | - Scott C. Bell
- Child Health Research Centre, The University of Queensland, South Brisbane, Queensland, Australia
- Adult Cystic Fibrosis Centre, The Prince Charles Hospital, Chermside, Queensland, Australia
- Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Derek S. Sarovich
- Centre for Bioinnovation, University of the Sunshine Coast, Sippy Downs, Queensland, Australia
- Sunshine Coast Health Institute, Birtinya, Queensland, Australia
| | - Erin P. Price
- Centre for Bioinnovation, University of the Sunshine Coast, Sippy Downs, Queensland, Australia
- Sunshine Coast Health Institute, Birtinya, Queensland, Australia
| |
Collapse
|
6
|
Chang RYK, Li M, Chow MY, Ke WR, Tai W, Chan HK. A dual action of D-amino acids on anti-biofilm activity and moisture-protection of inhalable ciprofloxacin powders. Eur J Pharm Biopharm 2022; 173:132-140. [DOI: 10.1016/j.ejpb.2022.03.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 02/16/2022] [Accepted: 03/08/2022] [Indexed: 01/03/2023]
|
7
|
Persistence and genetic adaptation of Pseudomonas aeruginosa in patients with chronic obstructive pulmonary disease. Clin Microbiol Infect 2022; 28:990-995. [DOI: 10.1016/j.cmi.2022.01.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 12/26/2022]
|
8
|
Wang Y, Chang RYK, Britton WJ, Chan HK. Advances in the development of antimicrobial peptides and proteins for inhaled therapy. Adv Drug Deliv Rev 2022; 180:114066. [PMID: 34813794 DOI: 10.1016/j.addr.2021.114066] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/18/2021] [Accepted: 11/18/2021] [Indexed: 12/13/2022]
Abstract
Antimicrobial peptides and proteins (APPs) are becoming increasingly important in targeting multidrug-resistant (MDR) bacteria. APPs is a rapidly emerging area with novel molecules being produced and further optimised to enhance antimicrobial efficacy, while overcoming issues associated with biologics such as potential toxicity and low bioavailability resulting from short half-life. Inhalation delivery of these agents can be an effective treatment of respiratory infections owing to the high local drug concentration in the lungs with lower exposure to systemic circulation hence reducing systemic toxicity. This review describes the recent studies on inhaled APPs, including in vitro and in vivo antimicrobial activities, toxicity assessments, and formulation strategies whenever available. The review also includes studies on combination of APPs with other antimicrobial agents to achieve enhanced synergistic antimicrobial effect. Since different APPs have different biological and chemical stabilities, a targeted formulation strategy should be considered for developing stable and inhalable antimicrobial peptides and proteins. These strategies include the use of sodium chloride to reduce electrostatic interaction between APP and extracellular DNA in sputum, the use of D-enantiomers or dendrimers to minimise protease-mediated degradation and or the use of prodrugs to reduce toxicity. Although great effort has been put towards optimising the biological functions of APPs, studies assessing biological stability in inhalable aerosols are scarce, particularly for novel molecules. As such, formulation and manufacture of inhalable liquid and powder formulations of APPs are underexplored, yet they are crucial areas of research for clinical translation.
Collapse
|
9
|
Abstract
Microbes are hardly seen as planktonic species and are most commonly found as biofilm communities in cases of chronic infections. Biofilms are regarded as a biological condition, where a large group of microorganisms gets adhered to a biotic or abiotic surface. In this context, Pseudomonas aeruginosa, a Gram-negative nosocomial pathogen is the main causative organism responsible for life-threatening and persistent infections in individuals affected with cystic fibrosis and other lung ailments. The bacteria can form a strong biofilm structure when it adheres to a surface suitable for the development of a biofilm matrix. These bacterial biofilms pose higher natural resistance to conventional antibiotic therapy due to their multiple tolerance mechanisms. This prevailing condition has led to an increasing rate of treatment failures associated with P. aeruginosa biofilm infections. A better understanding of the effect of a diverse group of antibiotics on established biofilms would be necessary to avoid inappropriate treatment strategies. Hence, the search for other alternative strategies as effective biofilm treatment options has become a growing area of research. The current review aims to give an overview of the mechanisms governing biofilm formation and the different strategies employed so far in the control of biofilm infections caused by P. aeruginosa. Moreover, this review can also help researchers to search for new antibiofilm agents to tackle the effect of biofilm infections that are currently imprudent to conventional antibiotics.
Collapse
|
10
|
Mayeux G, Gayet L, Liguori L, Odier M, Martin DK, Cortès S, Schaack B, Lenormand JL. Cell-free expression of the outer membrane protein OprF of Pseudomonas aeruginosa for vaccine purposes. Life Sci Alliance 2021; 4:4/6/e202000958. [PMID: 33972378 PMCID: PMC8127326 DOI: 10.26508/lsa.202000958] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 04/22/2021] [Accepted: 04/26/2021] [Indexed: 11/24/2022] Open
Abstract
Production of recombinant proteoliposomes containing OprF from P. aeruginosa promotes the active open conformation of the porin exposing native epitopes. These OprF proteoliposomes were used as vaccines to protect mice against a P. aeruginosa acute pulmonary infection model. Pseudomonas aeruginosa is the second-leading cause of nosocomial infections and pneumonia in hospitals. Because of its extraordinary capacity for developing resistance to antibiotics, treating infections by Pseudomonas is becoming a challenge, lengthening hospital stays, and increasing medical costs and mortality. The outer membrane protein OprF is a well-conserved and immunogenic porin playing an important role in quorum sensing and in biofilm formation. Here, we used a bacterial cell-free expression system to reconstitute OprF under its native forms in liposomes and we demonstrated that the resulting OprF proteoliposomes can be used as a fully functional recombinant vaccine against P. aeruginosa. Remarkably, we showed that our system promotes the folding of OprF into its active open oligomerized state as well as the formation of mega-pores. Our approach thus represents an easy and efficient way for producing bacterial membrane antigens exposing native epitopes for vaccine purposes.
Collapse
Affiliation(s)
- Géraldine Mayeux
- TheREx and Synabi, University Grenoble Alpes, CNRS, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble Institut Polytechnique (INP), Translational Innovation in Medicine and Complexity (TIMC), Grenoble, France
| | - Landry Gayet
- TheREx and Synabi, University Grenoble Alpes, CNRS, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble Institut Polytechnique (INP), Translational Innovation in Medicine and Complexity (TIMC), Grenoble, France
| | - Lavinia Liguori
- TheREx and Synabi, University Grenoble Alpes, CNRS, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble Institut Polytechnique (INP), Translational Innovation in Medicine and Complexity (TIMC), Grenoble, France.,Maison Familiale Rurale Moirans, Moirans, France
| | - Marine Odier
- TheREx and Synabi, University Grenoble Alpes, CNRS, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble Institut Polytechnique (INP), Translational Innovation in Medicine and Complexity (TIMC), Grenoble, France.,Catalent Pharma Solutions, Eberbach, Germany
| | - Donald K Martin
- TheREx and Synabi, University Grenoble Alpes, CNRS, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble Institut Polytechnique (INP), Translational Innovation in Medicine and Complexity (TIMC), Grenoble, France
| | | | - Béatrice Schaack
- TheREx and Synabi, University Grenoble Alpes, CNRS, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble Institut Polytechnique (INP), Translational Innovation in Medicine and Complexity (TIMC), Grenoble, France.,University Grenoble Alpes, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), CNRS, Institut de Biologie Structurale (IBS), Grenoble, France
| | - Jean-Luc Lenormand
- TheREx and Synabi, University Grenoble Alpes, CNRS, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble Institut Polytechnique (INP), Translational Innovation in Medicine and Complexity (TIMC), Grenoble, France
| |
Collapse
|
11
|
Wang T, Du X, Ji L, Han Y, Dang J, Wen J, Wang Y, Pu Q, Wu M, Liang H. Pseudomonas aeruginosa T6SS-mediated molybdate transport contributes to bacterial competition during anaerobiosis. Cell Rep 2021; 35:108957. [PMID: 33852869 DOI: 10.1016/j.celrep.2021.108957] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 01/06/2021] [Accepted: 03/16/2021] [Indexed: 12/20/2022] Open
Abstract
Type VI secretion system (T6SS) is widely distributed in Gram-negative bacteria and functions as a versatile protein export machinery that translocates effectors into eukaryotic or prokaryotic target cells. Growing evidence indicates that T6SS can deliver several effectors to promote bacterial survival in harmful environments through metal ion acquisition. Here, we report that the Pseudomonas aeruginosa H2-T6SS mediates molybdate (MoO42-) acquisition by secretion of a molybdate-binding protein, ModA. The expression of H2-T6SS genes is activated by the master regulator Anr and anaerobiosis. We also identified a ModA-binding protein, IcmP, an insulin-cleaving metalloproteinase outer membrane protein. The T6SS-ModA-IcmP system provides P. aeruginosa with a growth advantage in bacterial competition under anaerobic conditions and plays an important role in bacterial virulence. Overall, this study clarifies the role of T6SS in secretion of an anion-binding protein, emphasizing the fundamental importance of this bacterium using T6SS-mediated molybdate uptake to adapt to complex environmental conditions.
Collapse
Affiliation(s)
- Tietao Wang
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, ShaanXi 710069, China
| | - Xiao Du
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, ShaanXi 710069, China
| | - Linxuan Ji
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, ShaanXi 710069, China
| | - Yuying Han
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, ShaanXi 710069, China
| | - Jing Dang
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, ShaanXi 710069, China
| | - Jing Wen
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, ShaanXi 710069, China
| | - Yarong Wang
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, ShaanXi 710069, China
| | - Qinqin Pu
- Department of Basic Science, School of Medicine and Health Science, University of North Dakota, Grand Forks, ND 58203, USA
| | - Min Wu
- Department of Basic Science, School of Medicine and Health Science, University of North Dakota, Grand Forks, ND 58203, USA
| | - Haihua Liang
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, ShaanXi 710069, China.
| |
Collapse
|
12
|
Troudi A, Douafer H, Bolla JM, Klibi N, Brunel JM. Antibiotic Adjuvants to Rescue Pseudomonas aeruginosa from Tetracycline Antibiotics Resistance. ACTA ACUST UNITED AC 2021. [DOI: 10.2174/2211352518999200629164624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Introduction:
An attractive antibiotic-adjuvant strategy consisting of the design and
synthesis of polyaminoisoprenyl molecules able to restore the antibiotic activity of tetracycline antibiotics
against resistant Pseudomonas aeruginosa bacterial strains has been developed.
Methods:
These chemo-sensitizers are readily prepared from geraniol and farnesol in an efficient
two steps synthesis with good to moderate yields varying from 38 to 64% and leading to a significant
decrease in antibiotic resistance.
Results:
Thus, the influence of the nature of the tetracycline antibiotic used as well as the structure
of the polyaminoisoprenyl derivatives involved in the outcome of the antibiotic-adjuvant combination
against P. aeruginosa resistance to tetracyclines were investigated.
Conclusion:
Additionally, our data suggested that their mechanism of action is closely associated
with the increase of the outer-membrane permeability.
Collapse
Affiliation(s)
- Azza Troudi
- Aix Marseille Univ, INSERM, SSA, MCT, 13385 Marseille, France
| | - Hana Douafer
- Aix Marseille Univ, INSERM, SSA, MCT, 13385 Marseille, France
| | | | - Naouel Klibi
- Laboratory of Microorganisms and Active Biomolecules, Department of Biology, Faculty of Sciences of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Jean M. Brunel
- Aix Marseille Univ, INSERM, SSA, MCT, 13385 Marseille, France
| |
Collapse
|
13
|
Khan M, Husain FM, Zia Q, Ahmad E, Jamal A, Alaidarous M, Banawas S, Alam MM, Alshehri BA, Jameel M, Alam P, Ahamed MI, Ansari AH, Ahmad I. Anti-quorum Sensing and Anti-biofilm Activity of Zinc Oxide Nanospikes. ACS OMEGA 2020; 5:32203-32215. [PMID: 33376858 PMCID: PMC7758897 DOI: 10.1021/acsomega.0c03634] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 11/04/2020] [Indexed: 05/25/2023]
Abstract
This study evaluates the impact of two separate incubation periods (4 and 6 weeks) on the morphology of sol-gel-fabricated ZnO nanospikes (ZNs), that is, ZN1 and ZN2, respectively. We further analyzed the inhibitory effects of ZN1 and ZN2 on quorum sensing (QS) and biofilm formation in Pseudomonas aeruginosa (PAO1) and Chromobacterium violaceum (strains 12472 and CVO26). The size of the synthesized ZNs was in the range of 40-130 nm, and finer nanoparticles were synthesized after an incubation period of 6 weeks. Treatment with ZNs decreased the production of violacein in the pathogen without affecting the bacterial growth, which indicated that ZNs inhibited the QS signaling regulated by N-acyl homoserine lactone. ZN2 had a higher inhibitory action on the virulence factor productivity than ZN1. Furthermore, ZN2-treated cells displayed a substantial decrease in azocasein-degrading protease activity (80%), elastase activity (83%), and pyocyanin production (85%) relative to untreated P. aeruginosa PAO1 cells. Treatment with ZN2 decreased swarming motility and exopolysaccharide production by 89 and 85%, respectively. ZN2 was effective against both the las & pqs systems of P. aeruginosa and exhibited broad-spectrum activity. Additionally, ZN2 was more efficient in inhibiting the biofilm formation at the attachment stage than ZN1. These findings revealed that in P. aeruginosa, ZN2 demonstrated inhibitory effects on QS as well as on the development of biofilms. Thus, ZN2 can be potentially used to treat drug-resistant P. aeruginosa infections.
Collapse
Affiliation(s)
- Mohd.
Farhan Khan
- Nano
Solver Lab, Department of Mechanical Engineering, Z. H. College of
Engineering & Technology, Aligarh Muslim
University, Aligarh 202002, India
- Department
of Science, Gagan College of Management
and Technology, Aligarh 202002, India
| | - Fohad Mabood Husain
- Department
of Food Science and Nutrition, College of Food and Agriculture Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| | - Qamar Zia
- Health
and Basic Science Research Centre, Majmaah
University, Majmaah 11952, Saudi Arabia
- Department
of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah 11952, Saudi Arabia
| | - Ejaz Ahmad
- Interdisciplinary
Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, India
| | - Azfar Jamal
- Health
and Basic Science Research Centre, Majmaah
University, Majmaah 11952, Saudi Arabia
- Department
of Biology, College of Science, Majmaah
University, Majmaah 11952, Saudi Arabia
| | - Mohammed Alaidarous
- Health
and Basic Science Research Centre, Majmaah
University, Majmaah 11952, Saudi Arabia
- Department
of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah 11952, Saudi Arabia
| | - Saeed Banawas
- Health
and Basic Science Research Centre, Majmaah
University, Majmaah 11952, Saudi Arabia
- Department
of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah 11952, Saudi Arabia
- Department
of Biomedical Sciences, Oregon State University, Corvallis, Oregon 97331, United States
| | - Md. Manzar Alam
- Regional
Research Institute of Unani Medicine (Under CCRUM, Ministry of AYUSH), Patna 800008, India
| | - Bader A. Alshehri
- Health
and Basic Science Research Centre, Majmaah
University, Majmaah 11952, Saudi Arabia
| | - Mohd. Jameel
- Department
of Zoology, Faculty of Life Sciences, Aligarh
Muslim University, Aligarh 202002, India
| | - Pravej Alam
- Department of Biology, Prince Sattam bin
Abdulaziz Univrsity, Alkharj 11942, Kingdom of Saudi Arabia
| | - Mohd Imran Ahamed
- Department of Chemistry, Aligarh
Muslim
University, Aligarh 202002, India
| | - Akhter H. Ansari
- Nano
Solver Lab, Department of Mechanical Engineering, Z. H. College of
Engineering & Technology, Aligarh Muslim
University, Aligarh 202002, India
| | - Iqbal Ahmad
- Department of
Agricultural Microbiology, Faculty of Agricultural
Sciences, Aligarh Muslim University, Aligarh 202002, India
| |
Collapse
|
14
|
Riquelme SA, Prince A. Airway immunometabolites fuel Pseudomonas aeruginosa infection. Respir Res 2020; 21:326. [PMID: 33302964 PMCID: PMC7731785 DOI: 10.1186/s12931-020-01591-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 11/30/2020] [Indexed: 02/07/2023] Open
Abstract
Pulmonary infections are associated with a brisk inflammatory reaction to bacterial surface components. Lipopolysaccharides (LPS) trigger macrophage activation and release of mitochondrial metabolites that control the intensity of the immune response. Whereas succinate induces oxidative stress (ROS), HIF1α stabilization, glycolysis and IL-1β release, itaconate suppresses inflammation by inhibiting succinate oxidation, glycolytic flux and promoting anti-oxidant Nrf2-HO-1 functions. P. aeruginosa is a major pathogen associated with acute and chronic lung infection. Although both secreted toxins, LPS and proteases are key factors to establish acute P. aeruginosa pneumonia, lack of these components in chronic P. aeruginosa isolates suggest these organisms exploit other mechanisms to adapt and persist in the lung. Upon inhalation, P. aeruginosa strains trigger airway macrophage reprograming and bacterial variants obtained from acutely and chronically infected subjects exhibit metabolic adaptation consistent with succinate and itaconate assimilation; namely, high expression of extracellular polysaccharides (EPS), reduced lptD-LPS function, increased glyoxylate shunt (GS) activity and substantial biofilm production. In this review we discuss recent findings illustrating how P. aeruginosa induces and adapts to macrophage metabolites in the human lung, and that catabolism of succinate and itaconate contribute to their formidable abilities to tolerate oxidative stress, phagocytosis and immune clearance.
Collapse
Affiliation(s)
| | - Alice Prince
- Department of Pediatrics, Columbia University, New York, NY, 10032, USA.
| |
Collapse
|
15
|
Exploring the Therapeutic Efficacy of Zingerone Nanoparticles in Treating Biofilm-Associated Pyelonephritis Caused by Pseudomonas aeruginosa in the Murine Model. Inflammation 2020; 43:2344-2356. [DOI: 10.1007/s10753-020-01304-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
16
|
Low-Avidity Autoantibodies against Bactericidal/Permeability-Increasing Protein Occur in Gram-Negative and Gram-Positive Bacteremia. Infect Immun 2020; 88:IAI.00444-20. [PMID: 32747603 PMCID: PMC7504969 DOI: 10.1128/iai.00444-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 07/24/2020] [Indexed: 12/14/2022] Open
Abstract
Antibody autoreactivity against bactericidal/permeability-increasing protein (BPI) is strongly associated with Pseudomonas aeruginosa infection in cystic fibrosis (CF), non-CF bronchiectasis (BE), and chronic obstructive pulmonary disease (COPD). We examined the pathogen-specific nature of this autoreactivity by examining antibodies to BPI in bacteremia patients. Antibodies to BPI and bacterial antigens were measured in sera by ELISA from five patient cohorts (n = 214). Antibody autoreactivity against bactericidal/permeability-increasing protein (BPI) is strongly associated with Pseudomonas aeruginosa infection in cystic fibrosis (CF), non-CF bronchiectasis (BE), and chronic obstructive pulmonary disease (COPD). We examined the pathogen-specific nature of this autoreactivity by examining antibodies to BPI in bacteremia patients. Antibodies to BPI and bacterial antigens were measured in sera by ELISA from five patient cohorts (n = 214). Antibody avidity was investigated. Bacteremic patient sera (n = 32) exhibited IgG antibody autoreactivity against BPI in 64.7% and 46.7% of patients with positive blood cultures for P. aeruginosa and Escherichia coli, respectively. Autoantibody titers correlated with IgG responses to bacterial extracts and lipopolysaccharide (LPS). A prospective cohort of bacteremic patient sera exhibited anti-BPI IgG responses in 23/154 (14.9%) patients with autoreactivity present at the time of positive blood cultures in patients with Gram-negative and Gram-positive bacteria, including 8/60 (13.3%) patients with Staphylococcus aureus. Chronic tissue infection with S. aureus was associated with BPI antibody autoreactivity in 2/15 patients (13.3%). Previously, we demonstrated that BPI autoreactivity in CF patient sera exhibits high avidity. Here, a similar pattern was seen in BE patient sera. In contrast, sera from patients with bacteremia exhibited low avidity. These data indicate that low-avidity IgG responses to BPI can arise acutely in response to bacteremia and that this association is not limited to P. aeruginosa. This is to be contrasted with chronic respiratory infection with P. aeruginosa, suggesting that either the chronicity or the site of infection selects for the generation of high-avidity responses, with biologic consequences for airway immunity.
Collapse
|
17
|
Douafer H, Andrieu V, Wafo E, Brunel JM. Characterization of a new aerosol antibiotic/adjuvant combination for the treatment of P. aeruginosa lung infections. Int J Pharm 2020; 586:119548. [DOI: 10.1016/j.ijpharm.2020.119548] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/10/2020] [Accepted: 06/12/2020] [Indexed: 12/27/2022]
|
18
|
The Concerted Action of Two B3-Like Prophage Genes Excludes Superinfecting Bacteriophages by Blocking DNA Entry into Pseudomonas aeruginosa. J Virol 2020; 94:JVI.00953-20. [PMID: 32461312 DOI: 10.1128/jvi.00953-20] [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: 05/15/2020] [Accepted: 05/15/2020] [Indexed: 12/15/2022] Open
Abstract
In this study, we describe seven vegetative phage genomes homologous to the historic phage B3 that infect Pseudomonas aeruginosa Like other phage groups, the B3-like group contains conserved (core) and variable (accessory) open reading frames (ORFs) grouped at fixed regions in their genomes; however, in either case, many ORFs remain without assigned functions. We constructed lysogens of the seven B3-like phages in strain Ps33 of P. aeruginosa, a novel clinical isolate, and assayed the exclusion phenotype against a variety of temperate and virulent superinfecting phages. In addition to the classic exclusion conferred by the phage immunity repressor, the phenotype observed in B3-like lysogens suggested the presence of other exclusion genes. We set out to identify the genes responsible for this exclusion phenotype. Phage Ps56 was chosen as the study subject since it excluded numerous temperate and virulent phages. Restriction of the Ps56 genome, cloning of several fragments, and resection of the fragments that retained the exclusion phenotype allowed us to identify two core ORFs, so far without any assigned function, as responsible for a type of exclusion. Neither gene expressed separately from plasmids showed activity, but the concurrent expression of both ORFs is needed for exclusion. Our data suggest that phage adsorption occurs but that phage genome translocation to the host's cytoplasm is defective. To our knowledge, this is the first report on this type of exclusion mediated by a prophage in P. aeruginosa IMPORTANCE Pseudomonas aeruginosa is a Gram-negative bacterium frequently isolated from infected immunocompromised patients, and the strains are resistant to a broad spectrum of antibiotics. Recently, the use of phages has been proposed as an alternative therapy against multidrug-resistant bacteria. However, this approach may present various hurdles. This work addresses the problem that pathogenic bacteria may be lysogenized by phages carrying genes encoding resistance against secondary infections, such as those used in phage therapy. Discovering phage genes that exclude superinfecting phages not only assigns novel functions to orphan genes in databases but also provides insight into selection of the proper phages for use in phage therapy.
Collapse
|
19
|
Risk Factors of Multidrug-Resistant Bacteria in Lower Respiratory Tract Infections: A Systematic Review and Meta-Analysis. CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY 2020; 2020:7268519. [PMID: 32670442 PMCID: PMC7345606 DOI: 10.1155/2020/7268519] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/13/2020] [Accepted: 05/26/2020] [Indexed: 12/13/2022]
Abstract
Background Multidrug-resistant (MDR) bacteria are the main cause of lower respiratory tract infections (LRTIs) with high mortality. The purpose of this study is to identify the risk factors associated with MDR by performing a systematic review and meta-analysis. Methods PubMed, EMBASE (via Ovid), and Cochrane Library were systematically searched for studies on the risk factors for MDR bacteria in LRTIs as of November 30, 2019. Literature screening, data abstraction, and quality assessment of the eligible studies were performed independently by two researchers. Results A total of 3,607 articles were retrieved, of which 21 articles representing 20 cohort studies published in English were included after title/abstract and full-text screening. Among the 21 articles involving 7,650 patients and 1,360 MDR organisms, ten reported the risk factors for MDR Gram-positive bacteria (GPB) and Gram-negative bacteria (GNB), ten for MDR GNB, and one for MDR GPB. The meta-analysis results suggested that prior antibiotic treatment, inappropriate antibiotic therapy, chronic lung disease, chronic liver disease and cerebral disease, prior MDR and PA infection/colonization, recent hospitalization, longer hospitalization stay, endotracheal tracheostomy and mechanical ventilation, tube feeding, nursing home residence, and higher disease severity score were independent risk factors for MDR bacteria. Conclusions This review identified fourteen clinical factors that might increase the risk of MDR bacteria in patients with LRTIs. Clinicians could take into account these factors when selecting antibiotics for patients and determine whether coverage for MDR bacteria is required. More well-designed studies are needed to confirm the various risk factors for MDR bacteria in the future.
Collapse
|
20
|
Sen-Kilic E, Blackwood CB, Boehm DT, Witt WT, Malkowski AC, Bevere JR, Wong TY, Hall JM, Bradford SD, Varney ME, Damron FH, Barbier M. Intranasal Peptide-Based FpvA-KLH Conjugate Vaccine Protects Mice From Pseudomonas aeruginosa Acute Murine Pneumonia. Front Immunol 2019; 10:2497. [PMID: 31708925 PMCID: PMC6819369 DOI: 10.3389/fimmu.2019.02497] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 10/07/2019] [Indexed: 12/12/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen causing acute and chronic respiratory infections associated with morbidity and mortality, especially in patients with cystic fibrosis. Vaccination against P. aeruginosa before colonization may be a solution against these infections and improve the quality of life of at-risk patients. To develop a vaccine against P. aeruginosa, we formulated a novel peptide-based P. aeruginosa subunit vaccine based on the extracellular regions of one of its major siderophore receptors, FpvA. We evaluated the effectiveness and immunogenicity of the FpvA peptides conjugated to keyhole limpet hemocyanin (KLH) with the adjuvant curdlan in a murine vaccination and challenge model. Immunization with the FpvA-KLH vaccine decreased the bacterial burden and lung edema after P. aeruginosa challenge. Vaccination with FpvA-KLH lead to antigen-specific IgG and IgM antibodies in sera, and IgA antibodies in lung supernatant. FpvA-KLH immunized mice had an increase in recruitment of CD11b+ dendritic cells as well as resident memory CD4+ T cells in the lungs compared to non-vaccinated challenged mice. Splenocytes isolated from vaccinated animals showed that the FpvA-KLH vaccine with the adjuvant curdlan induces antigen-specific IL-17 production and leads to a Th17 type of immune response. These results indicate that the intranasal FpvA-KLH conjugate vaccine can elicit both mucosal and systemic immune responses. These observations suggest that the intranasal peptide-based FpvA-KLH conjugate vaccine with curdlan is a potential vaccine candidate against P. aeruginosa pneumonia.
Collapse
Affiliation(s)
- Emel Sen-Kilic
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States.,Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Catherine B Blackwood
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States.,Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Dylan T Boehm
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States.,Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - William T Witt
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States.,Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Aaron C Malkowski
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States.,Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Justin R Bevere
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States.,Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Ting Y Wong
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States.,Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Jesse M Hall
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States.,Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Shelby D Bradford
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States.,Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Melinda E Varney
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States.,Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Fredrick Heath Damron
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States.,Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Mariette Barbier
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States.,Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, United States
| |
Collapse
|
21
|
Clark ST, Guttman DS, Hwang DM. Diversification of Pseudomonas aeruginosa within the cystic fibrosis lung and its effects on antibiotic resistance. FEMS Microbiol Lett 2019; 365:4834010. [PMID: 29401362 DOI: 10.1093/femsle/fny026] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 01/30/2018] [Indexed: 12/13/2022] Open
Abstract
The evolution and diversification of bacterial pathogens within human hosts represent potential barriers to the diagnosis and treatment of life-threatening infections. Tremendous genetic and phenotypic diversity is characteristic of host adaptation in strains of Pseudomonas aeruginosa that infect the airways of individuals with chronic lung diseases and prove to be extremely difficult to eradicate. In this MiniReview, we examine recent advances in understanding within-host diversity and antimicrobial resistance in P. aeruginosa populations from the lower airways of individuals with the fatal genetic disease cystic fibrosis and the potential impacts that this diversity may have on detecting and interpreting antimicrobial susceptibility within these populations.
Collapse
Affiliation(s)
- Shawn T Clark
- Toronto General Hospital Research Institute, University Health Network, 101 College Street, PMCRT - MaRS Centre, Toronto, Ontario M5G 1L7, Canada
| | - David S Guttman
- Department of Cell & Systems Biology, University of Toronto, 25 Harbord Street, Toronto, Ontario M5S 3G5, Canada.,Centre for the Analysis of Genome Evolution & Function, University of Toronto, 25 Willcocks Street, Toronto, Ontario M5S 3B2, Canada
| | - David M Hwang
- Toronto General Hospital Research Institute, University Health Network, 101 College Street, PMCRT - MaRS Centre, Toronto, Ontario M5G 1L7, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
22
|
Carreira AC, Santos TC, Lone MA, Zupančič E, Lloyd-Evans E, de Almeida RFM, Hornemann T, Silva LC. Mammalian sphingoid bases: Biophysical, physiological and pathological properties. Prog Lipid Res 2019:100995. [PMID: 31445071 DOI: 10.1016/j.plipres.2019.100995] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 05/17/2019] [Accepted: 05/20/2019] [Indexed: 12/19/2022]
Abstract
Sphingoid bases encompass a group of long chain amino alcohols which form the essential structure of sphingolipids. Over the last years, these amphiphilic molecules were moving more and more into the focus of biomedical research due to their role as bioactive molecules. In fact, free sphingoid bases interact with specific receptors and target molecules and have been associated with numerous biological and physiological processes. In addition, they can modulate the biophysical properties of biological membranes. Several human diseases are related to pathological changes in the structure and metabolism of sphingoid bases. Yet, the mechanisms underlying their biological and pathophysiological actions remain elusive. Within this review, we aimed to summarize the current knowledge on the biochemical and biophysical properties of the most common sphingoid bases and to discuss their importance in health and disease.
Collapse
Affiliation(s)
- A C Carreira
- iMed.ULisboa - Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; Centro de Química e Bioquímica (CQB) e Centro de Química Estrutural (CQE), Faculdade de Ciências, Universidade de Lisboa, Ed. C8, Campo Grande, 1749-016 Lisboa, Portugal; Sir Martin Evans Building, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - T C Santos
- iMed.ULisboa - Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; Centro de Química-Física Molecular - Institute of Nanoscience and Nanotechnology (CQFM-IN) and IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Institute for Clinical Chemistry, University Hospital Zurich, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| | - M A Lone
- Institute for Clinical Chemistry, University Hospital Zurich, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| | - E Zupančič
- iMed.ULisboa - Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - E Lloyd-Evans
- Sir Martin Evans Building, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - R F M de Almeida
- Centro de Química e Bioquímica (CQB) e Centro de Química Estrutural (CQE), Faculdade de Ciências, Universidade de Lisboa, Ed. C8, Campo Grande, 1749-016 Lisboa, Portugal
| | - T Hornemann
- Institute for Clinical Chemistry, University Hospital Zurich, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| | - L C Silva
- iMed.ULisboa - Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; Centro de Química-Física Molecular - Institute of Nanoscience and Nanotechnology (CQFM-IN) and IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
23
|
Skopelja-Gardner S, Theprungsirikul J, Lewis KA, Hammond JH, Carlson KM, Hazlett HF, Nymon A, Nguyen D, Berwin BL, Hogan DA, Rigby WFC. Regulation of Pseudomonas aeruginosa-Mediated Neutrophil Extracellular Traps. Front Immunol 2019; 10:1670. [PMID: 31379861 PMCID: PMC6657737 DOI: 10.3389/fimmu.2019.01670] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 07/04/2019] [Indexed: 12/13/2022] Open
Abstract
Pseudomonas aeruginosa is the most prevalent opportunistic pathogen in the airways of cystic fibrosis (CF) patients. The pulmonary disorder is characterized by recurrent microbial infections and an exaggerated host inflammatory immune response led primarily by influx of neutrophils. Under these conditions, chronic colonization with P. aeruginosa is associated with diminished pulmonary function and increased morbidity and mortality. P. aeruginosa has a wide array of genetic mechanisms that facilitate its persistent colonization of the airway despite extensive innate host immune responses. Loss of function mutations in the quorum sensing regulatory gene lasR have been shown to confer survival advantage and a more pathogenic character to P. aeruginosa in CF patients. However, the strategies used by LasR-deficient P. aeruginosa to modulate neutrophil-mediated bactericidal functions are unknown. We sought to understand the role of LasR in P. aeruginosa-mediated neutrophil extracellular trap (NET) formation, an important anti-microbial mechanism deployed by neutrophils, the first-line responder in the infected airway. We observe mechanistic and phenotypic differences between NETs triggered by LasR-sufficient and LasR-deficient P. aeruginosa strains. We uncover that LasR-deficient P. aeruginosa strains fail to induce robust NET formation in both human and murine neutrophils, independently of bacterial motility or LPS expression. LasR does not mediate NET release via downstream quorum sensing signaling pathways but rather via transcriptional regulation of virulence factors, including, but not restricted to, LasB elastase and LasA protease. Finally, our studies uncover the differential requirements for NADPH oxidase in NET formation triggered by different P. aeruginosa strains.
Collapse
Affiliation(s)
- Sladjana Skopelja-Gardner
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Jomkuan Theprungsirikul
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Kimberley A Lewis
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - John H Hammond
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Kyrsten M Carlson
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Haley F Hazlett
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Amanda Nymon
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Dao Nguyen
- Department of Microbiology and Immunology, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Brent L Berwin
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Deborah A Hogan
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - William F C Rigby
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States.,Division of Rheumatology, Department of Medicine, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| |
Collapse
|
24
|
Theprungsirikul J, Skopelja-Gardner S, Meagher RE, Clancy JP, Zemanick ET, Ashare A, Rigby WFC. Dissociation of systemic and mucosal autoimmunity in cystic fibrosis. J Cyst Fibros 2019; 19:196-202. [PMID: 31262645 PMCID: PMC10377741 DOI: 10.1016/j.jcf.2019.06.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 05/07/2019] [Accepted: 06/12/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND Pseudomonas aeruginosa accounts for ~80% of cystic fibrosis (CF) airway infection. It shows a remarkable correlation with presence of autoantibody to bactericidal/permeability-increasing protein (BPI), which is not understood. In this study, we sought to better understand the characteristics of systemic and mucosal autoimmunity and their relation to humoral immunity to P. aeruginosa. METHODS Antibody titers and isotypes to BPI and P. aeruginosa were characterized in sera and bronchoalveolar lavage (BAL) of adult and pediatric CF patients (n = 131), by ELISA and/or immunoblot. RESULTS Serum BPI autoantibodies were common (~43%) in adult while rare (≪5%) in pediatric (≤18 yrs) CF patients. Serum BPI IgG autoantibodies were of high avidity and strongly correlated with anti-P. aeruginosa IgG responses. A parallel relationship was observed with IgA, but not IgG, responses in adult and pediatric CF patient in the BAL. Thus, BAL IgA anti-BPI antibodies were independent of age and correlated with the presence of BPI cleavage in BAL. CONCLUSIONS IgG and IgA autoreactivity to BPI in CF patients was demonstrated in serum and BAL, respectively, and correlated with the isotype of the antibody response to P. aeruginosa. The co-occurrence of anti-BPI and anti-P. aeruginosa IgA in the BAL, but not serum, of pediatric CF patients suggests that BPI tolerance is broken in the P. aeruginosa-infected airway and that serologic IgG autoantibodies are later induced, potentially through a separate pathway. The relationship between P. aeruginosa, BPI cleavage, and IgA autoantibodies in the BAL suggests a role for cryptic epitope generation in the breaking of tolerance.
Collapse
Affiliation(s)
- J Theprungsirikul
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - S Skopelja-Gardner
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - R E Meagher
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - J P Clancy
- Division of Pulmonary Medicine, Department of Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - E T Zemanick
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Children's Hospital Colorado, Aurora, CO, USA
| | - A Ashare
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA; Division of Pulmonology, Department of Medicine, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - W F C Rigby
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA; Division of Rheumatology, Department of Medicine, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA.
| |
Collapse
|
25
|
Panmanee W, Su S, Schurr MJ, Lau GW, Zhu X, Ren Z, McDaniel CT, Lu LJ, Ohman DE, Muruve DA, Panos RJ, Yu HD, Thompson TB, Tseng BS, Hassett DJ. The anti-sigma factor MucA of Pseudomonas aeruginosa: Dramatic differences of a mucA22 vs. a ΔmucA mutant in anaerobic acidified nitrite sensitivity of planktonic and biofilm bacteria in vitro and during chronic murine lung infection. PLoS One 2019; 14:e0216401. [PMID: 31158231 PMCID: PMC6546240 DOI: 10.1371/journal.pone.0216401] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 04/20/2019] [Indexed: 11/29/2022] Open
Abstract
Mucoid mucA22 Pseudomonas aeruginosa (PA) is an opportunistic lung pathogen of cystic fibrosis (CF) and chronic obstructive pulmonary disease (COPD) patients that is highly sensitive to acidified nitrite (A-NO2-). In this study, we first screened PA mutant strains for sensitivity or resistance to 20 mM A-NO2- under anaerobic conditions that represent the chronic stages of the aforementioned diseases. Mutants found to be sensitive to A-NO2- included PA0964 (pmpR, PQS biosynthesis), PA4455 (probable ABC transporter permease), katA (major catalase, KatA) and rhlR (quorum sensing regulator). In contrast, mutants lacking PA0450 (a putative phosphate transporter) and PA1505 (moaA2) were A-NO2- resistant. However, we were puzzled when we discovered that mucA22 mutant bacteria, a frequently isolated mucA allele in CF and to a lesser extent COPD, were more sensitive to A-NO2- than a truncated ΔmucA deletion (Δ157–194) mutant in planktonic and biofilm culture, as well as during a chronic murine lung infection. Subsequent transcriptional profiling of anaerobic, A-NO2--treated bacteria revealed restoration of near wild-type transcript levels of protective NO2- and nitric oxide (NO) reductase (nirS and norCB, respectively) in the ΔmucA mutant in contrast to extremely low levels in the A-NO2--sensitive mucA22 mutant. Proteins that were S-nitrosylated by NO derived from A-NO2- reduction in the sensitive mucA22 strain were those involved in anaerobic respiration (NirQ, NirS), pyruvate fermentation (UspK), global gene regulation (Vfr), the TCA cycle (succinate dehydrogenase, SdhB) and several double mutants were even more sensitive to A-NO2-. Bioinformatic-based data point to future studies designed to elucidate potential cellular binding partners for MucA and MucA22. Given that A-NO2- is a potentially viable treatment strategy to combat PA and other infections, this study offers novel developments as to how clinicians might better treat problematic PA infections in COPD and CF airway diseases.
Collapse
Affiliation(s)
- Warunya Panmanee
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH United States of America
| | - Shengchang Su
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH United States of America
| | - Michael J. Schurr
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO United States of America
| | - Gee W. Lau
- College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL United States of America
| | - Xiaoting Zhu
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH United States of America
| | - Zhaowei Ren
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH United States of America
| | - Cameron T. McDaniel
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH United States of America
| | - Long J. Lu
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH United States of America
| | - Dennis E. Ohman
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, Richmond, VA United States of America
- McGuire Veterans Affairs Medical Center, Richmond, VA United States of America
| | - Daniel A. Muruve
- Department of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Ralph J. Panos
- Department of Medicine, Cincinnati Veterans Affairs Medical Center, Cincinnati, OH United States of America
- Pulmonary, Critical Care, and Sleep Division, Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH United States of America
| | - Hongwei D. Yu
- Department of Biochemistry and Microbiology, Marshall University, Huntington, WV United States of America
| | - Thomas B. Thompson
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH United States of America
| | - Boo Shan Tseng
- Department of Life Sciences, University of Nevada-Las Vegas, Las Vegas, NV United States of America
| | - Daniel J. Hassett
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH United States of America
- * E-mail:
| |
Collapse
|
26
|
Carreira AC, Santos TC, Lone MA, Zupančič E, Lloyd-Evans E, de Almeida RFM, Hornemann T, Silva LC. Mammalian sphingoid bases: Biophysical, physiological and pathological properties. Prog Lipid Res 2019; 75:100988. [PMID: 31132366 DOI: 10.1016/j.plipres.2019.100988] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 05/17/2019] [Accepted: 05/21/2019] [Indexed: 12/11/2022]
Abstract
Sphingoid bases encompass a group of long chain amino alcohols which form the essential structure of sphingolipids. Over the last years, these amphiphilic molecules were moving more and more into the focus of biomedical research due to their role as bioactive molecules. In fact, free sphingoid bases interact with specific receptors and target molecules, and have been associated with numerous biological and physiological processes. In addition, they can modulate the biophysical properties of biological membranes. Several human diseases are related to pathological changes in the structure and metabolism of sphingoid bases. Yet, the mechanisms underlying their biological and pathophysiological actions remain elusive. Within this review, we aimed to summarize the current knowledge on the biochemical and biophysical properties of the most common sphingoid bases and to discuss their importance in health and disease.
Collapse
Affiliation(s)
- A C Carreira
- iMed.ULisboa - Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, Lisboa 1649-003, Portugal; Centro de Química e Bioquímica (CQB) e Centro de Química Estrutural (CQE), Faculdade de Ciências, Universidade de Lisboa, Ed. C8, Campo Grande, Lisboa 1749-016, Portugal; Sir Martin Evans Building, School of Biosciences, Cardiff University, Cardiff, UK
| | - T C Santos
- iMed.ULisboa - Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, Lisboa 1649-003, Portugal; Centro de Química-Física Molecular - Institute of Nanoscience and Nanotechnology (CQFM-IN), IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Institute for Clinical Chemistry, University Hospital Zurich, Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| | - M A Lone
- Institute for Clinical Chemistry, University Hospital Zurich, Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| | - E Zupančič
- iMed.ULisboa - Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, Lisboa 1649-003, Portugal
| | - E Lloyd-Evans
- Sir Martin Evans Building, School of Biosciences, Cardiff University, Cardiff, UK
| | - R F M de Almeida
- Centro de Química e Bioquímica (CQB) e Centro de Química Estrutural (CQE), Faculdade de Ciências, Universidade de Lisboa, Ed. C8, Campo Grande, Lisboa 1749-016, Portugal
| | - T Hornemann
- Institute for Clinical Chemistry, University Hospital Zurich, Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| | - L C Silva
- iMed.ULisboa - Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, Lisboa 1649-003, Portugal; Centro de Química-Física Molecular - Institute of Nanoscience and Nanotechnology (CQFM-IN), IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
27
|
Hill AT, Sullivan AL, Chalmers JD, De Soyza A, Elborn SJ, Floto AR, Grillo L, Gruffydd-Jones K, Harvey A, Haworth CS, Hiscocks E, Hurst JR, Johnson C, Kelleher PW, Bedi P, Payne K, Saleh H, Screaton NJ, Smith M, Tunney M, Whitters D, Wilson R, Loebinger MR. British Thoracic Society Guideline for bronchiectasis in adults. Thorax 2019; 74:1-69. [PMID: 30545985 DOI: 10.1136/thoraxjnl-2018-212463] [Citation(s) in RCA: 284] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Adam T Hill
- Respiratory Medicine, Royal Infirmary of Edinburgh and University of Edinburgh, Edinburgh, UK
| | - Anita L Sullivan
- Department of Respiratory Medicine, University Hospitals Birmingham NHS Foundation Trust (Queen Elizabeth Hospital), Birmingham, UK
| | - James D Chalmers
- Scottish Centre for Respiratory Research, University of Dundee, Ninewells Hospital, Dundee, UK
| | - Anthony De Soyza
- Institute of Cellular Medicine, NIHR Biomedical Research Centre for Aging and Freeman Hospital Adult Bronchiectasis service, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle, UK
| | - Stuart J Elborn
- Royal Brompton Hospital and Imperial College London, and Queens University Belfast
| | - Andres R Floto
- Department of Medicine, University of Cambridge, Cambridge UK.,Cambridge Centre for Lung Infection, Royal Papworth Hospital, Cambridge UK
| | | | | | - Alex Harvey
- Department of Clinical Sciences, Brunel University London, London, UK
| | - Charles S Haworth
- Cambridge Centre for Lung Infection, Royal Papworth Hospital, Cambridge UK
| | | | - John R Hurst
- UCL Respiratory, University College London, London, UK
| | | | - Peter W Kelleher
- Centre for Immunology and Vaccinology, Chelsea &Westminster Hospital Campus, Department of Medicine, Imperial College London.,Host Defence Unit, Department of Respiratory Medicine, Royal Brompton Hospital and Harefield NHS Foundation Trust, London.,Chest & Allergy Clinic St Mary's Hospital, Imperial College Healthcare NHS Trust
| | - Pallavi Bedi
- University of Edinburgh MRC Centre for Inflammation Research, Edinburgh, UK
| | | | | | | | - Maeve Smith
- University of Alberta, Edmonton, Alberta, Canada
| | - Michael Tunney
- School of Pharmacy, Queens University Belfast, Belfast, UK
| | | | - Robert Wilson
- Host Defence Unit, Department of Respiratory Medicine, Royal Brompton Hospital and Harefield NHS Foundation Trust, London
| | - Michael R Loebinger
- Host Defence Unit, Department of Respiratory Medicine, Royal Brompton Hospital and Harefield NHS Foundation Trust, London
| |
Collapse
|
28
|
Stockwell RE, Chin M, Johnson GR, Wood ME, Sherrard LJ, Ballard E, O'Rourke P, Ramsay KA, Kidd TJ, Jabbour N, Thomson RM, Knibbs LD, Morawska L, Bell SC. Transmission of bacteria in bronchiectasis and chronic obstructive pulmonary disease: Low burden of cough aerosols. Respirology 2019; 24:980-987. [PMID: 30919511 DOI: 10.1111/resp.13544] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 03/04/2019] [Accepted: 03/11/2019] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND OBJECTIVE Aerosol transmission of Pseudomonas aeruginosa has been suggested as a possible mode of respiratory infection spread in patients with cystic fibrosis (CF); however, whether this occurs in other suppurative lung diseases is unknown. Therefore, we aimed to determine if (i) patients with bronchiectasis (unrelated to CF) or chronic obstructive pulmonary disease (COPD) can aerosolize P. aeruginosa during coughing and (ii) if genetically indistinguishable (shared) P. aeruginosa strains are present in these disease cohorts. METHODS People with bronchiectasis or COPD and P. aeruginosa respiratory infection were recruited for two studies. Aerosol study: Participants (n = 20) underwent cough testing using validated cough rigs to determine the survival of P. aeruginosa aerosols in the air over distance and duration. Genotyping study: P. aeruginosa sputum isolates (n = 95) were genotyped using the iPLEX20SNP platform, with a subset subjected to the enterobacterial repetitive intergenic consensus polymerase chain reaction (ERIC-PCR) assay to ascertain their genetic relatedness. RESULTS Aerosol study: Overall, 7 of 20 (35%) participants released P. aeruginosa cough aerosols during at least one of the cough aerosol tests. These cough aerosols remained viable for 4 m from the source and for 15 min after coughing. The mean total aerosol count of P. aeruginosa at 2 m was two colony-forming units. Typing study: No shared P. aeruginosa strains were identified. CONCLUSION Low viable count of P. aeruginosa cough aerosols and a lack of shared P. aeruginosa strains observed suggest that aerosol transmission of P. aeruginosa is an unlikely mode of respiratory infection spread in patients with bronchiectasis and COPD.
Collapse
Affiliation(s)
- Rebecca E Stockwell
- Lung Bacteria Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Melanie Chin
- Department of Thoracic Medicine, The Prince Charles Hospital, Brisbane, QLD, Australia.,Division of Respirology, The University of Ottawa, Ottawa, ON, Canada.,Clinical Epidemiology, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Graham R Johnson
- International Laboratory for Air Quality and Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Michelle E Wood
- Lung Bacteria Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.,Department of Thoracic Medicine, The Prince Charles Hospital, Brisbane, QLD, Australia
| | | | - Emma Ballard
- Statistical Support Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Peter O'Rourke
- Statistical Support Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Kay A Ramsay
- Lung Bacteria Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.,Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Timothy J Kidd
- Lung Bacteria Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Nassib Jabbour
- International Laboratory for Air Quality and Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Rachel M Thomson
- Department of Thoracic Medicine, The Prince Charles Hospital, Brisbane, QLD, Australia.,Gallipoli Medical Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Luke D Knibbs
- School of Public Health, The University of Queensland, Brisbane, QLD, Australia
| | - Lidia Morawska
- International Laboratory for Air Quality and Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Scott C Bell
- Lung Bacteria Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.,Department of Thoracic Medicine, The Prince Charles Hospital, Brisbane, QLD, Australia
| |
Collapse
|
29
|
Bloomfield SJ, Midwinter AC, Biggs PJ, French NP, Marshall JC, Hayman DTS, Carter PE, Thornley C, Yap R, Benschop J. Long-term Colonization by Campylobacter jejuni Within a Human Host: Evolution, Antimicrobial Resistance, and Adaptation. J Infect Dis 2019; 217:103-111. [PMID: 29099940 DOI: 10.1093/infdis/jix561] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 10/25/2017] [Indexed: 01/24/2023] Open
Abstract
Background Campylobacteriosis is inflammation of the gastrointestinal tract as a result of Campylobacter infection. Most campylobacteriosis cases are acute and self-limiting, with Campylobacter excretion ceasing a few weeks after symptoms cease. We identified a patient with fecal specimens positive for Campylobacter jejuni (ST45) intermittently during a 10-year period. Methods Sixteen Campylobacter isolates were collected from the patient during 2006-2016. The isolates' genomes were sequenced to determine their relatedness, and their antimicrobial susceptibility patterns and motility were measured to determine the effects of antibiotic therapy and long-term excretion on the Campylobacter population. Results Phylogenetic analyses estimated that the isolates shared a date of common ancestor between 1998 and 2006, coinciding with the onset of symptoms for the patient. Genomic analysis identified selection for changes in motility, and antimicrobial susceptibility testing suggested that the Campylobacter population developed resistance to several antibiotics coinciding with periods of antibiotic therapy. Conclusions The patient was consistently colonized with organisms from a Campylobacter population that adapted to the internal environment of the patient. Genomic and phylogenetic analyses can give insight into a patient's infection history and the effect of antimicrobial treatment on Campylobacter populations in this unusual situation of long-term colonization of an individual.
Collapse
Affiliation(s)
- Samuel J Bloomfield
- mEpiLab, Hopkirk Research Institute, Massey University, Palmerston North, New Zealand
| | - Anne C Midwinter
- mEpiLab, Hopkirk Research Institute, Massey University, Palmerston North, New Zealand
| | - Patrick J Biggs
- mEpiLab, Hopkirk Research Institute, Massey University, Palmerston North, New Zealand
| | - Nigel P French
- mEpiLab, Hopkirk Research Institute, Massey University, Palmerston North, New Zealand.,New Zealand Food Safety Science and Research Centre, Hopkirk ResearchInstitute, Massey University, Palmerston North, New Zealand
| | - Jonathan C Marshall
- mEpiLab, Hopkirk Research Institute, Massey University, Palmerston North, New Zealand
| | - David T S Hayman
- mEpiLab, Hopkirk Research Institute, Massey University, Palmerston North, New Zealand
| | | | - Craig Thornley
- Regional Public Health, Lower Hutt Hospital, Lower Hutt, New Zealand
| | - Rudyard Yap
- Palmerston North Hospital, Palmerston North, New Zealand
| | - Jackie Benschop
- mEpiLab, Hopkirk Research Institute, Massey University, Palmerston North, New Zealand
| |
Collapse
|
30
|
Ahmed SAKS, Rudden M, Smyth TJ, Dooley JSG, Marchant R, Banat IM. Natural quorum sensing inhibitors effectively downregulate gene expression of Pseudomonas aeruginosa virulence factors. Appl Microbiol Biotechnol 2019; 103:3521-3535. [PMID: 30852658 PMCID: PMC6449319 DOI: 10.1007/s00253-019-09618-0] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 01/02/2019] [Accepted: 01/04/2019] [Indexed: 12/02/2022]
Abstract
At present, anti-virulence drugs are being considered as potential therapeutic alternatives and/or adjuvants to currently failing antibiotics. These drugs do not kill bacteria but inhibit virulence factors essential for establishing infection and pathogenesis through targeting non-essential metabolic pathways reducing the selective pressure to develop resistance. We investigated the effect of naturally isolated plant compounds on the repression of the quorum sensing (QS) system which is linked to virulence/pathogenicity in Pseudomonas aeruginosa. Our results show that trans-cinnamaldehyde (CA) and salicylic acid (SA) significantly inhibit expression of QS regulatory and virulence genes in P. aeruginosa PAO1 at sub-inhibitory levels without any bactericidal effect. CA effectively downregulated both the las and rhl QS systems with lasI and lasR levels inhibited by 13- and 7-fold respectively compared to 3- and 2-fold reductions with SA treatment, during the stationary growth phase. The QS inhibitors (QSI) also reduced the production of extracellular virulence factors with CA reducing protease, elastase and pyocyanin by 65%, 22% and 32%, respectively. The QSIs significantly reduced biofilm formation and concomitantly with repressed rhamnolipid gene expression, only trace amount of extracellular rhamnolipids were detected. The QSIs did not completely inhibit virulence factor expression and production but their administration significantly lowered the virulence phenotypes at both the transcriptional and extracellular levels. This study shows the significant inhibitory effect of natural plant-derived compounds on the repression of QS systems in P. aeruginosa.
Collapse
Affiliation(s)
- Syed A K S Ahmed
- School of Biomedical Sciences, Ulster University, Coleraine, BT52 1SA, UK
| | - Michelle Rudden
- Department of Biology, University of York, Wentworth, York, YO10 5DD, UK
| | - Thomas J Smyth
- School of Science, Institute of Technology Sligo, Sligo, Ireland
| | - James S G Dooley
- School of Biomedical Sciences, Ulster University, Coleraine, BT52 1SA, UK.
| | - Roger Marchant
- School of Biomedical Sciences, Ulster University, Coleraine, BT52 1SA, UK
| | - Ibrahim M Banat
- School of Biomedical Sciences, Ulster University, Coleraine, BT52 1SA, UK
| |
Collapse
|
31
|
Degrading of the Pseudomonas Aeruginosa Biofilm by Extracellular Levanase SacC from Bacillus subtilis. BIONANOSCIENCE 2019. [DOI: 10.1007/s12668-018-0581-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
32
|
Skopelja-Gardner S, Theprungsirikul J, Meagher RE, Beliveau CM, Bradley KE, Avery M, Henkle E, Siegel S, Gifford AH, Winthrop KL, Rigby WFC. Autoimmunity to bactericidal/permeability-increasing protein in bronchiectasis exhibits a requirement for Pseudomonas aeruginosa IgG response. Eur Respir J 2019; 53:13993003.01891-2018. [PMID: 30385530 DOI: 10.1183/13993003.01891-2018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 10/19/2018] [Indexed: 11/05/2022]
Affiliation(s)
| | - Jomkuan Theprungsirikul
- Dept of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Rachel E Meagher
- Dept of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Cathleen M Beliveau
- Dept of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Katherine E Bradley
- Dept of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Meade Avery
- Dept of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Emily Henkle
- Center for Infectious Disease Studies, OHSU-PSU School of Public Health, Portland, OR, USA
| | - Sarah Siegel
- Center for Infectious Disease Studies, OHSU-PSU School of Public Health, Portland, OR, USA
| | - Alex H Gifford
- Division of Pulmonology, Dept of Medicine, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA.,The Dartmouth Institute for Health Policy and Clinical Practice, Lebanon, NH, USA
| | - Kevin L Winthrop
- Center for Infectious Disease Studies, OHSU-PSU School of Public Health, Portland, OR, USA
| | - William F C Rigby
- Dept of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA.,Division of Rheumatology, Dept of Medicine, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| |
Collapse
|
33
|
Melittin Inhibition and Eradication Activity for Resistant Polymicrobial Biofilm Isolated from a Dairy Industry after Disinfection. Int J Microbiol 2019; 2019:4012394. [PMID: 30766602 PMCID: PMC6350607 DOI: 10.1155/2019/4012394] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 12/10/2018] [Indexed: 12/18/2022] Open
Abstract
The emerging concern about the increase of antibiotic resistance and associated biofilm has encouraged scientists to look for alternative antibiotics such as antimicrobial peptides (AMPs). This study evaluated the ability of melittin to act as an antibacterial biofilm inhibitor and biofilm remover considering isolates from dairy industry. Minimum inhibitory concentrations (MICs), minimum bactericidal concentrations (MBCs), minimum biofilm inhibitory concentrations (MBICs), and biofilm removal activities were studied in polymicrobial biofilms produced from isolates. MIC and MBC were set at 1–3 µg/mL and 25–50 µg/mL for Gram-positive and Gram-negative bacteria, respectively. Results demonstrated a good MBIC reaching 85% inhibition ability and a good activity and better penetration in deeper layers against the mixed preformed biofilm, thereby increasing its activity against all isolates also at the lowest tested concentrations. Melittin showed interesting characteristics suggesting its potential to act as an antimicrobial agent for polymicrobial biofilm from dairy industry even in environmental isolates.
Collapse
|
34
|
Sallman RS, Hussein SS, Ali MR. ERIC- PCR Typing, RAPD-PCR Fingerprinting and Quorum Sensing Gene Analysis of Pseudomonas aeruginosa Isolated from Different Clinical Sources. AL-MUSTANSIRIYAH JOURNAL OF SCIENCE 2018. [DOI: 10.23851/mjs.v29i2.345] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Recently, Pseudomonas aeruginosa infections proportions have increased significantly. Molecular typing and virulence analysis are good techniques, which can lead us to know P. aeruginosa infections. P. aeruginosa isolates were identified by using molecular method (16S rDNA gene) via PCR technique for accurate identification. The highest percent 41.26% of P. aeruginosa bacteria was found in the burn infections followed by 28.57% in wound swabs, 17.46% in ear discharge and lowest percentage were obtained from sputum samples. All isolates classified into six groups (A-F) according to classes of antibiotics. Of the 63 bacterial isolates, 100% were resistant to carbencillin, whereas 31.74% were resistant to ticarcillin and all isolates susceptible to imipenem. In addition all of clinical isolates indicated multidrug resistant (MDR) patterns, the highest rate of MDR was observed with pattern C these isolates were able to resist (9-12) antibiotics. All isolates were typed genotypically by using two methods of amplification, ERIC and RAPD-PCR. The results of the ERIC-PCR typing of P. aeruginosa bacteria that 96.82% showed amplification bands ERIC-PCR also revealed 17 groups of genotypes (A-R) and 4 unique isolates. The results of RAPD-PCR fingerprint revealed 12 groups of genotypes (A-M) of 40–90% similarity according to coefficient values and 4 unique isolates, except 7.93% were untypeble. QS genes (lasI, lasR, rhlI, rhlR), screen showed all isolates 100% were positive for one or more QS genes, in the other hand 82.53% carrying lasI, lasR, rhlI, and rhlR, while the 15.87% carrying lasI, rhlI, and rhlR and 1.58% carrying lasI, lasR, and rhlR genes. ERIC genotyping significantly correlated resistance patterns but not with virulence control QS genes. RAPD genotyping significantly correlated with source of infection, resistance patterns and virulence control QS genes. These results can help initial diagnosis MDR P. aeruginosa outbreaks associated with specific genotyping patterns.
Collapse
|
35
|
Risk factors for mortality in patients with Pseudomonas aeruginosa pneumonia: Clinical impact of mucA gene mutation. Respir Med 2018; 140:27-31. [PMID: 29957276 DOI: 10.1016/j.rmed.2018.05.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 05/18/2018] [Accepted: 05/19/2018] [Indexed: 11/20/2022]
Abstract
OBJECTIVES Pseudomonas aeruginosa frequently colonizes the lungs of chronic obstructive pulmonary disease (COPD) patients. Mucoid conversion is a hallmark of chronic P. aeruginosa infection, which is mediated by mucA gene mutations. The aim of this study is to identify predictive factors for mortality and the influence of mucA gene mutation in COPD patients with P. aeruginosa pneumonia. METHODS This study assessed 75 COPD patients with P. aeruginosa pneumonia at two university hospitals. The clinical and laboratory data were collected, and the P. aeruginosa isolates analyzed for the presence of mucA gene mutations. RESULTS MucA gene mutation of P. aeruginosa was an independent predictor of mortality (odds ratio [OR] 10.43, 95% confidence interval [CI]: 1.53-70.90, p = 0.017). In addition, the APACHE II score and C-reactive protein/Albumin (CA) ratio were independent predictive factors for mortality (OR 1.25, 95% CI: 1.07-1.46, p = 0.004; and OR 1.06, 95% CI: 1.02-1.10, p = 0.003, respectively). The optimal cutoff value of CA ratio for the greatest sensitivity and specificity was calculated as 31.27 (sensitivity, 85.7%; specificity, 80.3%). CONCLUSIONS CA ratio and mucA gene mutation of P. aeruginosa could be used as predictors to identify poor prognosis in COPD patients with P. aeruginosa pneumonia.
Collapse
|
36
|
Bruguera-Avila N, Marin A, Garcia-Olive I, Radua J, Prat C, Gil M, Ruiz-Manzano J. Effectiveness of treatment with nebulized colistin in patients with COPD. Int J Chron Obstruct Pulmon Dis 2017; 12:2909-2915. [PMID: 29042767 PMCID: PMC5634377 DOI: 10.2147/copd.s138428] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Objectives To analyze whether the introduction of nebulized colistin in patients with chronic obstructive pulmonary disease (COPD) and infection with Pseudomonas aeruginosa (PA) is associated with a decrease of the number and duration of severe exacerbations. Materials and methods Thirty six patients with COPD and infection with PA treated with nebulized colistin attending a day hospital during a 5-year (January 2010–December 2014) period were prospectively included. Repeated-measures t-tests were used to assess whether the introduction of colistin was associated with changes in the number of exacerbations or the length of the hospitalizations, comparing for each patient the year prior to the introduction of colistin with the year after. Results After the introduction of colistin, the number of admissions decreased from 2.0 to 0.9 per individual year (P=0.0007), and hospitalizations were shorter (23.3 vs 10.9 days, P=0.00005). These results persisted when patients with and without bronchiectasis or with and without persistence of Pseudomonas were separately analyzed. No pre–post differences were detected in the number of exacerbations not requiring admission. Conclusion Nebulized colistin seems associated with a strong decrease in the number and duration of hospitalizations due to exacerbation in patients with COPD and infection with PA. Clinical trials with a larger number of patients are needed in order to confirm these results.
Collapse
Affiliation(s)
- Nuria Bruguera-Avila
- Department of Medicine, Hospital Sant Jaume de Calella, Calella, Barcelona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona, Bellatera, Spain.,Department of Pulmonary Medicine, Hospital Universitari Germans Trias i Pujol, Carretera del Canyet sn, Badalona, Barcelona, Spain
| | - Alicia Marin
- Department of Pulmonary Medicine, Hospital Universitari Germans Trias i Pujol, Carretera del Canyet sn, Badalona, Barcelona, Spain.,Ciber de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain.,Fundació Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Carretera del Canyet sn, Badalona, Barcelona, Spain
| | - Ignasi Garcia-Olive
- Department of Pulmonary Medicine, Hospital Universitari Germans Trias i Pujol, Carretera del Canyet sn, Badalona, Barcelona, Spain.,Ciber de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain.,Fundació Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Carretera del Canyet sn, Badalona, Barcelona, Spain
| | - Joaquim Radua
- Department of Statistics, FIDMAG Germanes Hospitalaries Research Unit, Sant Boi de Llobregat, Barcelona, Spain.,CiberSam - Ciber de Salud Mental, Madrid, Spain.,Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Cristina Prat
- Ciber de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain.,Department of Microbiology, Hospital Universitari Germans Trias i Pujol, Carretera del Canyet sn, Badalona, Barcelona, Spain.,Department of Genetics and Microbiology of Universitat Autònoma de Barcelona, Bellatera, Spain
| | - Montserrat Gil
- Department of Pulmonary Medicine, Hospital Universitari Germans Trias i Pujol, Carretera del Canyet sn, Badalona, Barcelona, Spain
| | - Juan Ruiz-Manzano
- Department of Medicine, Universitat Autònoma de Barcelona, Bellatera, Spain.,Department of Pulmonary Medicine, Hospital Universitari Germans Trias i Pujol, Carretera del Canyet sn, Badalona, Barcelona, Spain.,Ciber de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain.,Fundació Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Carretera del Canyet sn, Badalona, Barcelona, Spain
| |
Collapse
|
37
|
Aditi, Shariff M, Chhabra SK, Rahman MU. Similar virulence properties of infection and colonization associated Pseudomonas aeruginosa. J Med Microbiol 2017; 66:1489-1498. [PMID: 28893354 DOI: 10.1099/jmm.0.000569] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
PURPOSE Pseudomonas aeruginosa is one of the agents that are commonly implicated in nosocomial infections. However, it is also present as a commensal in various body sites of healthy persons, making the diagnosis of infection by culture difficult. A number of virulence factors expressed by the organism have been implicated in its pathogenicity. We undertook this study to identify the host and organism factors associated with infection. METHODOLOGY Pathogenic, colonizing and environmental isolates were tested for apr, lasB, the T3SS effector exoenzymes (exoS, exoT, exoU and exoY) and toxA genes, biofilm production and antimicrobial susceptibility. The isolates were further typed by RAPD. RESULTS Eighty-seven isolates from 61 patients, including 11 environmental isolates, were obtained. None of the virulence factors were found to be significantly associated with infection, and nor was the antimicrobial susceptibility. The presence of the exoU gene and infection by MDR strains correlated significantly with the duration of hospital stay. Positivity for exoS and exoU genes was found to be strongly correlated with multi-drug resistance. exoU positivity correlated strongly with fluoroquinolone resistance. Sinks in the ward and intensive care unit were found to be a niche for XDR P. aeruginosa. Eighty-five isolates were typeable using the ERIC2 primer, showing 71 distinct RAPD patterns with >15 % difference in UPGMA-generated dice coefficients. CONCLUSIONS exoU positivity is associated with severe disease, as evidenced by the longer duration of hospital stay of these patients. However, the presence of virulence factors or multi-drug resistance in the cultured strain should not prompt the administration of anti-pseudomonal chemotherapy.
Collapse
Affiliation(s)
- Aditi
- Department of Microbiology, Vallabhbhai Patel Chest Institute, Delhi, India
| | - Malini Shariff
- Department of Microbiology, Vallabhbhai Patel Chest Institute, Delhi, India
| | - Sunil K Chhabra
- Department of Cardio-respiratory Physiology, Vallabhbhai Patel Chest Institute, Delhi, India
| | | |
Collapse
|
38
|
A Low-Molecular-Weight Alginate Oligosaccharide Disrupts Pseudomonal Microcolony Formation and Enhances Antibiotic Effectiveness. Antimicrob Agents Chemother 2017. [PMID: 28630204 DOI: 10.1128/aac.00762-17] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
In chronic respiratory disease, the formation of dense, 3-dimensional "microcolonies" by Pseudomonas aeruginosa within the airway plays an important role in contributing to resistance to treatment. An in vitro biofilm model of pseudomonal microcolony formation using artificial-sputum (AS) medium was established to study the effects of low-molecular-weight alginate oligomers (OligoG CF-5/20) on pseudomonal growth, microcolony formation, and the efficacy of colistin. The studies employed clinical cystic fibrosis (CF) isolates (n = 3) and reference nonmucoid and mucoid multidrug-resistant (MDR) CF isolates (n = 7). Bacterial growth and biofilm development and disruption were studied using cell viability assays and image analysis with scanning electron and confocal laser scanning microscopy. Pseudomonal growth in AS medium was associated with increased ATP production (P < 0.05) and the formation (at 48 h) of discrete (>10-μm) microcolonies. In conventional growth medium, colistin retained an ability to inhibit growth of planktonic bacteria, although the MIC was increased (0.1 to 0.4 μg/ml) in AS medium compared to Mueller-Hinton (MH) medium. In contrast, in an established-biofilm model in AS medium, the efficacy of colistin was decreased. OligoG CF-5/20 (≥2%) treatment, however, induced dose-dependent biofilm disruption (P < 0.05) and led to colistin retaining its antimicrobial activity (P < 0.05). While circular dichroism indicated that OligoG CF-5/20 did not change the orientation of the alginate carboxyl groups, mass spectrometry demonstrated that the oligomers induced dose-dependent (>0.2%; P < 0.05) reductions in pseudomonal quorum-sensing signaling. These findings reinforce the potential clinical significance of microcolony formation in the CF lung and highlight a novel approach to treat MDR pseudomonal infections.
Collapse
|
39
|
Selective phenylalanine to proline substitution for improved antimicrobial and anticancer activities of peptides designed on phenylalanine heptad repeat. Acta Biomater 2017; 57:170-186. [PMID: 28483698 DOI: 10.1016/j.actbio.2017.05.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 04/19/2017] [Accepted: 05/04/2017] [Indexed: 01/09/2023]
Abstract
Introducing cell-selectivity in antimicrobial peptides (AMPs) without compromising the antimicrobial and anti-endotoxin properties is a crucial step towards the development of new antimicrobial agents. A peptide designed on phenylalanine heptad repeat possesses significant cytotoxicity along with desired antimicrobial and anti-endotoxin properties. Amino acid substitutions at 'a' and/or 'd' positions of heptad repeats of AMPs could alter their helical structure in mammalian membrane-mimetic environments and cytotoxicity towards mammalian cells. Since proline is a helix breaker, effects of selective proline substitution(s) at 'a' and/or 'd' positions of a 15-residue peptide designed on phenylalanine heptad repeat (FR-15) were investigated. Proline-substituted FR-15 variants were highly selective toward bacteria and fungi over hRBCs and murine 3T3 cells and also retained their antibacterial activities at high salt, serum and elevated temperatures. These non-cytotoxic variants also inhibited LPS-induced production of pro-inflammatory cytokines/chemokines in human monocytes, THP-1, RAW 264.7 and in BALB/c mice. The two non-cytotoxic variants (FR8P and FR11P) showed potent anti-cancer activity against highly metastatic human breast cancer cell line MDA-MB-231 with IC50 values less than 10μM. At sub-IC50 concentrations, FR8P and FR11P also showed anti-migratory and anti-invasive effects against MDA-MB-231 cells. FR8P and FR11P induced cellular apoptosis by triggering intrinsic apoptotic pathway through depolarization of mitochondrial membrane potential and activation of caspases. Overall the results demonstrated the utilization of selective phenylalanine to proline substitution in a heptad repeat of phenylalanine residues for the design of cell-selective, broad-spectrum AMPs with significant anti-cancer properties. STATEMENT OF SIGNIFICANCE We have demonstrated a methodology to design cell-selective potent antimicrobial and anti-endotoxin peptides by utilizing phenylalanine zipper as a template and replacement of phenylalanine residue(s) from "a" and/or "d" position(s) with proline residue(s) produced non-cytotoxic AMPs with improved antibacterial properties against the drug-resistant strains of bacteria. The work showed that the 'a' and 'd' positions of the phenylalanine heptad repeat could be replaced by an appropriate amino acid to control cytotoxicity of the peptide without compromising its potency in antimicrobial and anti-endotoxin properties. The direct bacterial membrane targeting mechanism of proline substituted analogs of parent peptide makes difficult for bacteria to grow resistance against them. The peptides designed could be lead molecules in the area of sepsis as they possess significant anti-LPS activities for in vitro and in vivo. Interestingly since cancer cells and bacterial cell membranes possess the structural resemblances, the cancer cells are also targets for these peptides making them lead molecules in this field. However, unlike in bacteria where the peptides showed membrane permeabilization property to lyse them, the peptides induced apoptosis in MDA-MB-231 breast cancer cells to inhibit their proliferation and growth. The results are significant because it reveals that "a" and "d" positions of a phenylalanine zipper can be utilized as switches to design cell-selective, antimicrobial, anti-endotoxin and anticancer peptides.
Collapse
|
40
|
Margaroli C, Tirouvanziam R. Neutrophil plasticity enables the development of pathological microenvironments: implications for cystic fibrosis airway disease. Mol Cell Pediatr 2016; 3:38. [PMID: 27868161 PMCID: PMC5136534 DOI: 10.1186/s40348-016-0066-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 11/04/2016] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION The pathological course of several chronic inflammatory diseases, including cystic fibrosis, chronic obstructive pulmonary disease, and rheumatoid arthritis, features an aberrant innate immune response dominated by neutrophils. In cystic fibrosis, neutrophil burden and activity of neutrophil elastase in the extracellular fluid have been identified as strong predictors of lung disease severity. REVIEW Although neutrophils are generally considered to be rigid, pre-programmed effector leukocytes, recent studies suggest extensive plasticity in how neutrophil functions unfold upon recruitment to peripheral tissues, and how they choose their ultimate fate. Indeed, upon migration to cystic fibrosis airways, neutrophils display dysregulated lifespan, metabolic activation, and altered effector and regulatory functions, consistent with profound adaptation and phenotypic reprogramming. Licensed by signals present in cystic fibrosis airway microenvironment to survive and develop these novel functions, neutrophils orchestrate, in partnership with the epithelium and with the resident microbiota, the evolution of a pathological microenvironment. This microenvironment is defined by altered proteolytic, redox, and metabolic balance and the presence of stable luminal structures in which neutrophils and microbes coexist. CONCLUSIONS The elucidation of molecular mechanisms driving neutrophil plasticity in vivo will open new treatment opportunities designed to modulate, rather than block, the crucial adaptive functions fulfilled by neutrophils. This review aims to outline emerging mechanisms of neutrophil plasticity and their participation in the building of pathological microenvironments in the context of cystic fibrosis and other diseases with similar features.
Collapse
Affiliation(s)
- Camilla Margaroli
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Center for CF and Airways Disease Research, Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA
- Emory + Children's Center, 2015 Uppergate Dr NE, Rm 344, Atlanta, GA, 30322-1014, USA
| | - Rabindra Tirouvanziam
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Center for CF and Airways Disease Research, Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA.
- Emory + Children's Center, 2015 Uppergate Dr NE, Rm 344, Atlanta, GA, 30322-1014, USA.
| |
Collapse
|
41
|
A Novel RNase 3/ECP Peptide for Pseudomonas aeruginosa Biofilm Eradication That Combines Antimicrobial, Lipopolysaccharide Binding, and Cell-Agglutinating Activities. Antimicrob Agents Chemother 2016; 60:6313-25. [PMID: 27527084 DOI: 10.1128/aac.00830-16] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 07/22/2016] [Indexed: 12/19/2022] Open
Abstract
Eradication of established biofilm communities of pathogenic Gram-negative species is one of the pending challenges for the development of new antimicrobial agents. In particular, Pseudomonas aeruginosa is one of the main dreaded nosocomial species, with a tendency to form organized microbial communities that offer an enhanced resistance to conventional antibiotics. We describe here an engineered antimicrobial peptide (AMP) which combines bactericidal activity with a high bacterial cell agglutination and lipopolysaccharide (LPS) affinity. The RN3(5-17P22-36) peptide is a 30-mer derived from the eosinophil cationic protein (ECP), a host defense RNase secreted by eosinophils upon infection, with a wide spectrum of antipathogen activity. The protein displays high biofilm eradication activity that is not dependent on its RNase catalytic activity, as evaluated by using an active site-defective mutant. On the other hand, the peptide encompasses both the LPS-binding and aggregation-prone regions from the parental protein, which provide the appropriate structural features for the peptide's attachment to the bacterial exopolysaccharide layer and further improved removal of established biofilms. Moreover, the peptide's high cationicity and amphipathicity promote the cell membrane destabilization action. The results are also compared side by side with other reported AMPs effective against either planktonic and/or biofilm forms of Pseudomonas aeruginosa strain PAO1. The ECP and its derived peptide are unique in combining high bactericidal potency and cell agglutination activity, achieving effective biofilm eradication at a low micromolar range. We conclude that the designed RN3(5-17P22-36) peptide is a promising lead candidate against Gram-negative biofilms.
Collapse
|
42
|
McDaniel C, Su S, Panmanee W, Lau GW, Browne T, Cox K, Paul AT, Ko SHB, Mortensen JE, Lam JS, Muruve DA, Hassett DJ. A Putative ABC Transporter Permease Is Necessary for Resistance to Acidified Nitrite and EDTA in Pseudomonas aeruginosa under Aerobic and Anaerobic Planktonic and Biofilm Conditions. Front Microbiol 2016; 7:291. [PMID: 27064218 PMCID: PMC4817314 DOI: 10.3389/fmicb.2016.00291] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 02/23/2016] [Indexed: 11/18/2022] Open
Abstract
Pseudomonas aeruginosa (PA) is an important airway pathogen of cystic fibrosis and chronic obstructive disease patients. Multiply drug resistant PA is becoming increasing prevalent and new strategies are needed to combat such insidious organisms. We have previously shown that a mucoid, mucA22 mutant PA is exquisitely sensitive to acidified nitrite (A-NO2−, pH 6.5) at concentrations that are well tolerated in humans. Here, we used a transposon mutagenesis approach to identify PA mutants that are hypersensitive to A-NO2−. Among greater than 10,000 mutants screened, we focused on PA4455, in which the transposon was found to disrupt the production of a putative cytoplasmic membrane-spanning ABC transporter permease. The PA4455 mutant was not only highly sensitive to A-NO2−, but also the membrane perturbing agent, EDTA and the antibiotics doxycycline, tigecycline, colistin, and chloramphenicol, respectively. Treatment of bacteria with A-NO2− plus EDTA, however, had the most dramatic and synergistic effect, with virtually all bacteria killed by 10 mM A-NO2−, and EDTA (1 mM, aerobic, anaerobic). Most importantly, the PA4455 mutant was also sensitive to A-NO2− in biofilms. A-NO2− sensitivity and an anaerobic growth defect was also noted in two mutants (rmlC and wbpM) that are defective in B-band LPS synthesis, potentially indicating a membrane defect in the PA4455 mutant. Finally, this study describes a gene, PA4455, that when mutated, allows for dramatic sensitivity to the potential therapeutic agent, A-NO2− as well as EDTA. Furthermore, the synergy between the two compounds could offer future benefits against antibiotic resistant PA strains.
Collapse
Affiliation(s)
- Cameron McDaniel
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine Cincinnati, OH, USA
| | - Shengchang Su
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine Cincinnati, OH, USA
| | - Warunya Panmanee
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine Cincinnati, OH, USA
| | - Gee W Lau
- College of Veterinary Medicine, University of Illinois at Urbana-Champaign Urbana, IL, USA
| | - Tristan Browne
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine Cincinnati, OH, USA
| | - Kevin Cox
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine Cincinnati, OH, USA
| | - Andrew T Paul
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine Cincinnati, OH, USA
| | - Seung-Hyun B Ko
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine Cincinnati, OH, USA
| | - Joel E Mortensen
- Diagnostic and Infectious Diseases Testing Laboratory, Cincinnati Children's Hospital Medical Center Cincinnati, OH, USA
| | - Joseph S Lam
- Department of Molecular and Cellular Biology, University of Guelph Guelph, ON, Canada
| | - Daniel A Muruve
- Department of Medicine, University of Calgary Calgary, AB, Canada
| | - Daniel J Hassett
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of MedicineCincinnati, OH, USA; Department of Research Services, Cincinnati Veteran's Association Medical CenterCincinnati, OH, USA
| |
Collapse
|
43
|
Du Q, Jin J, Liu X, Sun Y. Bronchiectasis as a Comorbidity of Chronic Obstructive Pulmonary Disease: A Systematic Review and Meta-Analysis. PLoS One 2016; 11:e0150532. [PMID: 26978269 PMCID: PMC4792534 DOI: 10.1371/journal.pone.0150532] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 02/15/2016] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Bronchiectasis revealed by chest computed tomography in COPD patients and its comorbid effect on prognosis have not been addressed by large-sized studies. Understanding the presence of bronchiectasis in COPD is important for future intervention and preventing disease progression. METHODS Observational studies were identified from electronic literature searches in Cochrane library, PubMed, ScienceDirect databases, American Thoracic Society and European Respiratory Society meeting abstracts. A systematic review and meta-analysis of studies was performed to summarize the factors associated with bronchiectasis in COPD patients. Primary outcomes included the risks for exacerbation frequency, isolation of a potentially pathogenic microorganism, severe airway obstruction and mortality. Odds ratios (ORs) were pooled by random effects models. RESULTS Fourteen observational studies were eligible for the study. Compared with COPD without bronchiectasis, comorbid bronchiectasis in COPD increased the risk of exacerbation (1.97, 95% CI, 1.29-3.00), isolation of a potentially pathogenic microorganism (4.11, 95%CI, 2.16-7.82), severe airway obstruction (1.31, 95% CI, 1.09-1.58) and mortality (1.96, 95% CI, 1.04-3.70). CONCLUSIONS The presence of bronchiectasis in patients with COPD was associated with exacerbation frequency, isolation of a potentially pathogenic microorganism, severe airway obstruction and mortality.
Collapse
Affiliation(s)
- Qingxia Du
- Department of Respiratory Medicine and Department of Emergency, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Jianmin Jin
- Department of Respiratory Medicine and Department of Emergency, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Xiaofang Liu
- Department of Respiratory Medicine and Department of Emergency, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Yongchang Sun
- Department of Respiratory Medicine and Department of Emergency, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191 China
| |
Collapse
|
44
|
Tatke G, Kumari H, Silva-Herzog E, Ramirez L, Mathee K. Pseudomonas aeruginosa MifS-MifR Two-Component System Is Specific for α-Ketoglutarate Utilization. PLoS One 2015; 10:e0129629. [PMID: 26114434 PMCID: PMC4482717 DOI: 10.1371/journal.pone.0129629] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 05/10/2015] [Indexed: 12/31/2022] Open
Abstract
Pseudomonas aeruginosa is a Gram-negative, metabolically versatile opportunistic pathogen that elaborates a multitude of virulence factors, and is extraordinarily resistant to a gamut of clinically significant antibiotics. This ability, in part, is mediated by two-component regulatory systems (TCS) that play a crucial role in modulating virulence mechanisms and metabolism. MifS (PA5512) and MifR (PA5511) form one such TCS implicated in biofilm formation. MifS is a sensor kinase whereas MifR belongs to the NtrC superfamily of transcriptional regulators that interact with RpoN (σ54). In this study we demonstrate that the mifS and mifR genes form a two-gene operon. The close proximity of mifSR operon to poxB (PA5514) encoding a ß-lactamase hinted at the role of MifSR TCS in regulating antibiotic resistance. To better understand this TCS, clean in-frame deletions were made in P. aeruginosa PAO1 creating PAO∆mifS, PAO∆mifR and PAO∆mifSR. The loss of mifSR had no effect on the antibiotic resistance profile. Phenotypic microarray (BioLOG) analyses of PAO∆mifS and PAO∆mifR revealed that these mutants were unable to utilize C5-dicarboxylate α-ketoglutarate (α-KG), a key tricarboxylic acid cycle intermediate. This finding was confirmed using growth analyses, and the defect can be rescued by mifR or mifSR expressed in trans. These mifSR mutants were able to utilize all the other TCA cycle intermediates (citrate, succinate, fumarate, oxaloacetate or malate) and sugars (glucose or sucrose) except α-KG as the sole carbon source. We confirmed that the mifSR mutants have functional dehydrogenase complex suggesting a possible defect in α-KG transport. The inability of the mutants to utilize α-KG was rescued by expressing PA5530, encoding C5-dicarboxylate transporter, under a regulatable promoter. In addition, we demonstrate that besides MifSR and PA5530, α-KG utilization requires functional RpoN. These data clearly suggests that P. aeruginosa MifSR TCS is involved in sensing α-KG and regulating its transport and subsequent metabolism.
Collapse
Affiliation(s)
- Gorakh Tatke
- Department of Biological Sciences, College of Arts & Sciences, Florida International University, Miami, Florida, United States of America
- Department of Molecular Microbiology and Infectious Diseases, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, United States of America
| | - Hansi Kumari
- Department of Molecular Microbiology and Infectious Diseases, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, United States of America
| | - Eugenia Silva-Herzog
- Department of Molecular Microbiology and Infectious Diseases, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, United States of America
| | - Lourdes Ramirez
- Department of Biological Sciences, College of Arts & Sciences, Florida International University, Miami, Florida, United States of America
| | - Kalai Mathee
- Department of Molecular Microbiology and Infectious Diseases, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, United States of America
- * E-mail:
| |
Collapse
|
45
|
Pyrosequencing analysis of microbial community and food-borne bacteria on restaurant cutting boards collected in Seri Kembangan, Malaysia, and their correlation with grades of food premises. Int J Food Microbiol 2015; 200:57-65. [PMID: 25679309 DOI: 10.1016/j.ijfoodmicro.2015.01.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 01/28/2015] [Accepted: 01/30/2015] [Indexed: 01/10/2023]
Abstract
This study adopts the pyrosequencing technique to identify bacteria present on 26 kitchen cutting boards collected from different grades of food premises around Seri Kembangan, a city in Malaysia. Pyrosequencing generated 452,401 of total reads of OTUs with an average of 1.4×10(7) bacterial cells/cm(2). Proteobacteria, Firmicutes and Bacteroides were identified as the most abundant phyla in the samples. Taxonomic richness was generally high with >1000 operational taxonomic units (OTUs) observed across all samples. The highest appearance frequencies (100%) were OTUs closely related to Enterobacter sp., Enterobacter aerogenes, Pseudomonas sp. and Pseudomonas putida. Several OTUs were identified most closely related to known food-borne pathogens, including Bacillus cereus, Cronobacter sakazaki, Cronobacter turisensis, Escherichia coli, E. coli O157:H7, Hafnia alvei, Kurthia gibsonii, Salmonella bongori, Salmonella enterica, Salmonella paratyphi, Salmonella tyhpi, Salmonella typhimurium and Yersinia enterocolitica ranging from 0.005% to 0.68% relative abundance. The condition and grade of the food premises on a three point cleanliness scale did not correlate with the bacterial abundance and type. Regardless of the status and grades, all food premises have the same likelihood to introduce food-borne bacteria from cutting boards to their foods and must always prioritize the correct food handling procedure in order to avoid unwanted outbreak of food-borne illnesses.
Collapse
|
46
|
Dosler S, Karaaslan E. Inhibition and destruction of Pseudomonas aeruginosa biofilms by antibiotics and antimicrobial peptides. Peptides 2014; 62:32-7. [PMID: 25285879 DOI: 10.1016/j.peptides.2014.09.021] [Citation(s) in RCA: 162] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 09/24/2014] [Accepted: 09/24/2014] [Indexed: 11/25/2022]
Abstract
Pseudomonas aeruginosa is one of the major nosocomial pathogen that can causes a wide variety of acute and chronic infections P. aeruginosa is a dreaded bacteria not just because of the high intrinsic and acquired antibiotic resistance rates but also the biofilm formation and production of multiple virulence factors. We investigated the in vitro activities of antibiotics (ceftazidime, tobramycin, ciprofloxacin, doripenem, piperacillin and colistin) and antimicrobial cationic peptides (AMPs; LL-37, CAMA: cecropin(1-7)-melittin A(2-9) amide, melittin, defensin and magainin-II) alone or in combination against biofilms of laboratory strain ATCC 27853 and 4 clinical strains of P. aeruginosa. The minimum inhibitory concentrations (MIC), minimum bactericidal concentration (MBC) and minimum biofilm eradication concentrations (MBEC) were determined by microbroth dilution technique. The MBEC values of antibiotics and AMPs were 80->5120 and 640->640mg/L, respectively. When combined with the LL-37 or CAMA at 1/10× MBEC, the MBEC values of antibiotics that active against biofilms, were decreased up to 8-fold. All of the antibiotics, and AMPs were able to inhibit the attachment of bacteria at the 1/10× MIC and biofilm formation at 1× or 1/10× MIC concentrations. Time killing curve studies showed 3-log10 killing against biofilms in 24h with almost all studied antibiotics and AMPs. Synergism were seen in most of the studied combinations especially CAMA/LL-37+ciprofloxacin against at least one or two strains' biofilms. Since biofilms are not affected the antibiotics at therapeutic concentrations, using a combination of antimicrobial agents including AMPs, or inhibition of biofilm formation by blocking the attachment of bacteria to surfaces might be alternative methods to fight with biofilm associated infections.
Collapse
Affiliation(s)
- Sibel Dosler
- University of Istanbul, Faculty of Pharmacy, Department of Pharmaceutical Microbiology, 34116 Beyazit, Istanbul, Turkiye.
| | - Elif Karaaslan
- University of Bezmi Alem, Faculty of Medicine, Department of Medical Microbiology, 34116 Fatih, Istanbul, Turkiye.
| |
Collapse
|
47
|
Gallego M, Pomares X, Espasa M, Castañer E, Solé M, Suárez D, Monsó E, Montón C. Pseudomonas aeruginosa isolates in severe chronic obstructive pulmonary disease: characterization and risk factors. BMC Pulm Med 2014; 14:103. [PMID: 24964956 PMCID: PMC4094400 DOI: 10.1186/1471-2466-14-103] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 06/19/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Patients with severe chronic obstructive pulmonary disease (COPD) are at increased risk of infection by P. aeruginosa. The specific role of bronchiectasis in both infection and chronic colonization by this microorganism in COPD, however, remains ill defined.To evaluate the prevalence and risk factors for P. aeruginosa recovery from sputum in outpatients with severe COPD, characterizing P. aeruginosa isolates by pulsed-field gel electrophoresis (PFGE) and focusing on the influence of bronchiectasis on chronic colonization in these patients. METHODS A case-cohort study of 118 patients with severe COPD attended at a Respiratory Day Unit for an acute infectious exacerbation and followed up over one year. High-resolution CT scans were performed during stability for bronchiectasis assessment and sputum cultures were obtained during exacerbation and stability in all patients. P. aeruginosa isolates were genotyped by PFGE. Determinants of the recovery of P. aeruginosa in sputum and chronic colonization by this microorganism were assessed by multivariate analysis. RESULTS P. aeruginosa was isolated from 41 of the 118 patients studied (34.7%). Five of these 41 patients (12.2%) with P. aeruginosa recovery fulfilled criteria for chronic colonization. In the multivariate analysis, the extent of bronchiectasis (OR 9.8, 95% CI: 1.7 to 54.8) and the number of antibiotic courses (OR 1.7, 95% CI: 1.1 to 2.5) were independently associated with an increased risk of P. aeruginosa isolation. Chronic colonization was unrelated to the presence of bronchiectasis (p=0.75). In patients with chronic colonization the isolates of P. aeruginosa retrieved corresponded to the same clones during the follow-up, and most of the multidrug resistant isolates (19/21) were harbored by these patients. CONCLUSIONS The main risk factors for P. aeruginosa isolation in severe COPD were the extent of bronchiectasis and exposure to antibiotics. Over 10% of these patients fulfilled criteria for chronic colonization by P. aeruginosa and showed clonal persistence, independently of the presence of bronchiectasis.
Collapse
Affiliation(s)
- Miguel Gallego
- Department of Respiratory Medicine, Hospital de Sabadell, Institut Universitari Parc Taulí-UAB, Sabadell, Spain
- Universitat Autonoma de Barcelona, Esfera UAB, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, CIBERES, Bunyola, Spain
| | - Xavier Pomares
- Department of Respiratory Medicine, Hospital de Sabadell, Institut Universitari Parc Taulí-UAB, Sabadell, Spain
| | - Mateu Espasa
- Department of Microbiology, SDI UDIAT, Institut Universitari Parc Taulí-UAB, Sabadell, Spain
| | - Eva Castañer
- Department of Radiology, SDI UDIAT, Institut Universitari Parc Taulí-UAB
| | - Mar Solé
- Barcelona Centre for International Health Research (CRESIB), Department of Clinical Microbiology, Hospital Clínic-Fundació Clínic per la Recerca Biomèdica, University of Barcelona, Barcelona, Spain
| | - David Suárez
- Epidemiology and Assessment Unit, Fundació Parc Taulí, Universitat Autònoma de Barcelona, Sabadell, Spain
| | - Eduard Monsó
- Department of Respiratory Medicine, Hospital de Sabadell, Institut Universitari Parc Taulí-UAB, Sabadell, Spain
- Universitat Autonoma de Barcelona, Esfera UAB, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, CIBERES, Bunyola, Spain
| | - Concepción Montón
- Department of Respiratory Medicine, Hospital de Sabadell, Institut Universitari Parc Taulí-UAB, Sabadell, Spain
| |
Collapse
|
48
|
Qin X. Chronic pulmonary pseudomonal infection in patients with cystic fibrosis: A model for early phase symbiotic evolution. Crit Rev Microbiol 2014; 42:144-57. [PMID: 24766052 DOI: 10.3109/1040841x.2014.907235] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Gain of "antimicrobial resistance" and "adaptive virulence" has been the dominant view of Pseudomonas aeruginosa (Pa) in cystic fibrosis (CF) in the progressively damaged host airway over the course of this chronic infection. However, the pathogenic effects of CF airway-adapted Pa strains are notably reduced. We propose that CF Pa and other bacterial cohabitants undergo host adaptation which resembles the changes found in bacterial symbionts in animal hosts. Development of clonally selected and intraspecific isogenic Pa strains which display divergent colony morphology, growth rate, auxotrophy, and antibiotic susceptibility in vitro suggests an adaptive sequence of infective exploitation-parasitism-symbiotic evolution driven by host defenses. Most importantly, the emergence of CF pseudomonal auxotrophy is frequently associated with a few specific amino acids. The selective retention or loss of specific amino acid biosynthesis in CF-adapted Pa reflects bacterium-host symbiosis and coevolution during chronic infection, not nutrient availability. This principle also argues against the long-standing concept of dietary availability leading to evolution of essential amino acid requirements in humans. A novel model of pseudomonal adaptation through multicellular bacterial syntrophy is proposed to explain early events in bacterial gene decay and decreased (not increased) virulence due to symbiotic response to host defense.
Collapse
Affiliation(s)
- Xuan Qin
- a Microbiology Laboratory, Seattle Children's Hospital , and.,b Department of Laboratory Medicine , University of Washington , School of Medicine Seattle , Washington , USA
| |
Collapse
|
49
|
Su S, Panmanee W, Wilson JJ, Mahtani HK, Li Q, VanderWielen BD, Makris TM, Rogers M, McDaniel C, Lipscomb JD, Irvin RT, Schurr MJ, Lancaster JR, Kovall RA, Hassett DJ. Catalase (KatA) plays a role in protection against anaerobic nitric oxide in Pseudomonas aeruginosa. PLoS One 2014; 9:e91813. [PMID: 24663218 PMCID: PMC3963858 DOI: 10.1371/journal.pone.0091813] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 02/14/2014] [Indexed: 12/30/2022] Open
Abstract
Pseudomonas aeruginosa (PA) is a common bacterial pathogen, responsible for a high incidence of nosocomial and respiratory infections. KatA is the major catalase of PA that detoxifies hydrogen peroxide (H2O2), a reactive oxygen intermediate generated during aerobic respiration. Paradoxically, PA displays elevated KatA activity under anaerobic growth conditions where the substrate of KatA, H2O2, is not produced. The aim of the present study is to elucidate the mechanism underlying this phenomenon and define the role of KatA in PA during anaerobiosis using genetic, biochemical and biophysical approaches. We demonstrated that anaerobic wild-type PAO1 cells yielded higher levels of katA transcription and expression than aerobic cells, whereas a nitrite reductase mutant ΔnirS produced ∼50% the KatA activity of PAO1, suggesting that a basal NO level was required for the increased KatA activity. We also found that transcription of the katA gene was controlled, in part, by the master anaerobic regulator, ANR. A ΔkatA mutant and a mucoid mucA22 ΔkatA bacteria demonstrated increased sensitivity to acidified nitrite (an NO generator) in anaerobic planktonic and biofilm cultures. EPR spectra of anaerobic bacteria showed that levels of dinitrosyl iron complexes (DNIC), indicators of NO stress, were increased significantly in the ΔkatA mutant, and dramatically in a ΔnorCB mutant compared to basal levels of DNIC in PAO1 and ΔnirS mutant. Expression of KatA dramatically reduced the DNIC levels in ΔnorCB mutant. We further revealed direct NO-KatA interactions in vitro using EPR, optical spectroscopy and X-ray crystallography. KatA has a 5-coordinate high spin ferric heme that binds NO without prior reduction of the heme iron (Kd ∼6 μM). Collectively, we conclude that KatA is expressed to protect PA against NO generated during anaerobic respiration. We proposed that such protective effects of KatA may involve buffering of free NO when potentially toxic concentrations of NO are approached.
Collapse
Affiliation(s)
- Shengchang Su
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Warunya Panmanee
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Jeffrey J. Wilson
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Harry K. Mahtani
- Departments of Anesthesiology, Cell, Developmental and Integrative Biology, and Environmental Health Sciences, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Qian Li
- Departments of Anesthesiology, Cell, Developmental and Integrative Biology, and Environmental Health Sciences, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Bradley D. VanderWielen
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Thomas M. Makris
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Melanie Rogers
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Cameron McDaniel
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - John D. Lipscomb
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Randall T. Irvin
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Michael J. Schurr
- Department of Microbiology, University of Colorado, Aurora, Colorado, United States of America
| | - Jack R. Lancaster
- Departments of Anesthesiology, Cell, Developmental and Integrative Biology, and Environmental Health Sciences, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Rhett A. Kovall
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Daniel J. Hassett
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, Cincinnati, Ohio, United States of America
- * E-mail:
| |
Collapse
|
50
|
Sharma G, Rao S, Bansal A, Dang S, Gupta S, Gabrani R. Pseudomonas aeruginosa biofilm: Potential therapeutic targets. Biologicals 2014; 42:1-7. [DOI: 10.1016/j.biologicals.2013.11.001] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 11/07/2013] [Indexed: 10/26/2022] Open
|