101
|
Sousa SA, Seixas AM, Mandal M, Rodríguez-Ortega MJ, Leitão JH. Characterization of the Burkholderia cenocepacia J2315 Surface-Exposed Immunoproteome. Vaccines (Basel) 2020; 8:vaccines8030509. [PMID: 32899969 PMCID: PMC7565204 DOI: 10.3390/vaccines8030509] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/27/2020] [Accepted: 09/03/2020] [Indexed: 11/16/2022] Open
Abstract
Infections by the Burkholderia cepacia complex (Bcc) remain seriously life threatening to cystic fibrosis (CF) patients, and no effective eradication is available. A vaccine to protect patients against Bcc infections is a highly attractive therapeutic option, but none is available. A strategy combining the bioinformatics identification of putative surface-exposed proteins with an experimental approach encompassing the “shaving” of surface-exposed proteins with trypsin followed by peptide identification by liquid chromatography and mass spectrometry is here reported. The methodology allowed the bioinformatics identification of 263 potentially surface-exposed proteins, 16 of them also experimentally identified by the “shaving” approach. Of the proteins identified, 143 have a high probability of containing B-cell epitopes that are surface-exposed. The immunogenicity of three of these proteins was demonstrated using serum samples from Bcc-infected CF patients and Western blotting, validating the usefulness of this methodology in identifying potentially immunogenic surface-exposed proteins that might be used for the development of Bcc-protective vaccines.
Collapse
Affiliation(s)
- Sílvia A. Sousa
- iBB–Institute for Bioengineering and Biosciences, 1049-001 Lisbon, Portugal; (A.M.M.S.); (M.M.)
- Correspondence: (S.A.S.); (J.H.L.); Tel.: +351-2184-19986 (S.A.S.); +351-2184-17688 (J.H.L.)
| | - António M.M. Seixas
- iBB–Institute for Bioengineering and Biosciences, 1049-001 Lisbon, Portugal; (A.M.M.S.); (M.M.)
| | - Manoj Mandal
- iBB–Institute for Bioengineering and Biosciences, 1049-001 Lisbon, Portugal; (A.M.M.S.); (M.M.)
| | | | - Jorge H. Leitão
- iBB–Institute for Bioengineering and Biosciences, 1049-001 Lisbon, Portugal; (A.M.M.S.); (M.M.)
- Correspondence: (S.A.S.); (J.H.L.); Tel.: +351-2184-19986 (S.A.S.); +351-2184-17688 (J.H.L.)
| |
Collapse
|
102
|
Silva E, Monteiro R, Grainha T, Alves D, Pereira MO, Sousa AM. Fostering Innovation in the Treatment of Chronic Polymicrobial Cystic Fibrosis-Associated Infections Exploring Aspartic Acid and Succinic Acid as Ciprofloxacin Adjuvants. Front Cell Infect Microbiol 2020; 10:441. [PMID: 32974221 PMCID: PMC7481398 DOI: 10.3389/fcimb.2020.00441] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 07/17/2020] [Indexed: 11/23/2022] Open
Abstract
Cystic fibrosis (CF) disease provokes the accumulation of thick and viscous sputum in the lungs, favoring the development of chronic and polymicrobial infections. Pseudomonas aeruginosa is the main bacterium responsible for these chronic infections, and much of the difficulty involved in eradicating it is due to biofilm formation. However, this could be mitigated using adjuvant compounds that help or potentiate the antibiotic action. Therefore, the main goal of this study was to search for substances that function as adjuvants and also as biofilm-controlling compounds, preventing or dismantling P. aeruginosa biofilms formed in an in vitro CF airway environment. Dual combinations of compounds with subinhibitory (1 and 2 mg/L) and inhibitory concentrations (4 mg/L) of ciprofloxacin were tested to inhibit the bacterial growth and biofilm formation (prophylactic approach) and to eradicate 24-h-old P. aeruginosa populations, including planktonic cells and biofilms (treatment approach). Our results revealed that aspartic acid (Asp) and succinic acid (Suc) restored ciprofloxacin action against P. aeruginosa. Suc combined with 2 mg/L of ciprofloxacin (Suc-Cip) was able to eradicate bacteria, and Asp combined with 4 mg/L of ciprofloxacin (Asp–Cip) seemed to eradicate the whole 24-h-old populations, including planktonic cells and biofilms. Based on biomass depletion data, we noted that Asp induced cell death and Suc seemed somehow to block or reduce the expression of ciprofloxacin resistance. As far as we know, this kind of action had not been reported up till now. The presence of Staphylococcus aureus and Burkholderia cenocepacia did not affect the efficacy of the Asp–Cip and Suc–Cip therapies against P. aeruginosa and, also important, P. aeruginosa depletion from polymicrobial communities did not create a window of opportunity for these species to thrive. Rather the contrary, Asp and Suc also improved ciprofloxacin action against B. cenocepacia. Further studies on the cytotoxicity using lung epithelial cells indicated toxicity of Suc–Cip caused by the Suc. In conclusion, we provided evidences that Asp and Suc could be potential ciprofloxacin adjuvants to eradicate P. aeruginosa living within polymicrobial communities. Asp–Cip and Suc–Cip could be promising therapeutic options to cope with CF treatment failures.
Collapse
Affiliation(s)
- Eduarda Silva
- CEB - Centre of Biological Engineering, LIBRO - Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Braga, Portugal
| | - Rosana Monteiro
- CEB - Centre of Biological Engineering, LIBRO - Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Braga, Portugal
| | - Tânia Grainha
- CEB - Centre of Biological Engineering, LIBRO - Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Braga, Portugal
| | - Diana Alves
- CEB - Centre of Biological Engineering, LIBRO - Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Braga, Portugal
| | - Maria Olivia Pereira
- CEB - Centre of Biological Engineering, LIBRO - Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Braga, Portugal
| | - Ana Margarida Sousa
- CEB - Centre of Biological Engineering, LIBRO - Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Braga, Portugal
| |
Collapse
|
103
|
Bogdanovski K, Chau T, Robinson CJ, MacDonald SD, Peterson AM, Mashek CM, Wallin WA, Rimkus M, Montgomery F, Lucas da Silva J, Gupta S, Ghaffari A, Zelazny AM, Olivier KN. Antibacterial activity of high-dose nitric oxide against pulmonary Mycobacterium abscessus disease. Access Microbiol 2020; 2:acmi000154. [PMID: 33195983 PMCID: PMC7656188 DOI: 10.1099/acmi.0.000154] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 06/15/2020] [Indexed: 12/27/2022] Open
Abstract
Introduction Mycobacterium abscessus is an emerging pulmonary pathogen with limited treatment options. Nitric oxide (NO) demonstrates antibacterial activity against various bacterial species, including mycobacteria. In this study, we evaluated the effect of adjunctive inhaled NO therapy, using a novel NO generator, in a CF patient with pulmonary M. abscessus disease, and examined heterogeneity of response to NO in vitro. Methods In the compassionate-use treatment, a 24-year-old CF patient with pulmonary M. abscessus was treated with two courses of adjunctive intermittent NO, first at 160 p.p.m. for 21 days and subsequently by escalating the dose up to 240 p.p.m. for 8 days. Methemoglobin, pulmonary function, 6 min walk distance (6MWD), qualify of life and sputum microbiology were assessed. In vitro susceptibility tests were performed against patient's isolate and comparison clinical isolates and quantified by Hill's slopes calculated from time-kill curves. Results M. abscessus lung infection eradication was not achieved, but improvements in selected qualify of life domains, lung function and 6MWD were observed during the study. Inhaled NO was well tolerated at 160 p.p.m. Dosing at 240 p.p.m. was stopped due to adverse symptoms, although methemoglobin levels remained within safety thresholds. In vitro susceptibility tests showed a dose-dependent NO effect on M. abscessus susceptibility and significant heterogeneity in response between M. abscessus clinical isolates. The patient's isolate was found to be the least susceptible strain in vitro. Conclusion These results demonstrate heterogeneity in M. abscessus susceptibility to NO and suggest that longer treatment regimens could be required to see the reduction or eradication of more resistant pulmonary strains.
Collapse
Affiliation(s)
- Kristijan Bogdanovski
- Laboratory of Chronic Airway Infection, Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Trisha Chau
- Laboratory of Chronic Airway Infection, Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Chevalia J Robinson
- Laboratory of Chronic Airway Infection, Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sandra D MacDonald
- Laboratory of Chronic Airway Infection, Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ann M Peterson
- Nursing Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Christine M Mashek
- Nursing Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Windy A Wallin
- Critical Care Therapy Section, Clinical Center, National Institutes of Health, Bethesda, USA
| | | | | | - Joas Lucas da Silva
- Laboratory of Chronic Airway Infection, Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Shashank Gupta
- Laboratory of Chronic Airway Infection, Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Adrian M Zelazny
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Kenneth N Olivier
- Laboratory of Chronic Airway Infection, Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
104
|
Kublanovskaya A, Baulina O, Chekanov K, Lobakova E. The microalga Haematococcus lacustris (Chlorophyceae) forms natural biofilms in supralittoral White Sea coastal rock ponds. PLANTA 2020; 252:37. [PMID: 32778946 DOI: 10.1007/s00425-020-03438-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 07/29/2020] [Indexed: 06/11/2023]
Abstract
Haematococcus lacustris inhabits supralittoral rock ponds and forms, under natural conditions, biofilms including layered cyanobacterial and fermentative microbial mats. Dry mats, formed under extremely stressful conditions, contained only haematocysts. Under favorable growth conditions, modeled for dry biofilms in vitro, microalgal free-living stages were detected. Haematococcus lacustris is the microalga known for its high potential to survive under a wide range of unfavorable conditions, particularly in the supralittoral temporal rock ponds of the White Sea. Previously, we described microbial communities containing H. lacustris in this region. In many cases, they were organized into systems exhibiting complex three-dimensional structure similar to that of natural biofilms. In this study, for the first time, we clarify structural description and provide microscopic evidence that these communities of H. lacustris and bacteria are assembled into the true biofilms. There are (1) simple single layer biofilms on the surface of rocks and macrophytic algae, (2) floccules (or flocs) not attached to a surface, (3) as well as stratified (layered) biofilms, wet, and dehydrated in nature. Being involved into primary organic production, H. lacustris and cyanobacteria are located exclusively in the upper layers of stratified biofilms, where they are capable to absorb sufficient for photosynthesis amount of light. The presence of acidic polysaccharides in the extracellular matrix revealed by specific staining with ruthenium red in the H. lacustris-containing microbial communities is a biochemical evidence of biofilm formation. Meanwhile, the presence of bacterial L-form is an ultrastructural confirmation of that fact. Under favorable conditions, modeled in vitro, H. lacustris from the dry microbial mats moves to the free-living states represented by vegetative palmelloid cells and motile zoospores. Owing to the fact that inside biofilms cells of microorganisms exist under stable conditions, we consider the biofilm formation as an additional mechanism that contributes to the survival of H. lacustris in the supralittoral zone of the White Sea.
Collapse
Affiliation(s)
- Anna Kublanovskaya
- Faculty of Biology, Lomonosov Moscow State University, 1-12 Leninskie Gory, Moscow, 119192, Russia.
| | - Olga Baulina
- Faculty of Biology, Lomonosov Moscow State University, 1-12 Leninskie Gory, Moscow, 119192, Russia
| | - Konstantin Chekanov
- Faculty of Biology, Lomonosov Moscow State University, 1-12 Leninskie Gory, Moscow, 119192, Russia
- Centre for Humanities Research and Technology, National Research Nuclear University MEPhI, 31 Kashirskoye highway, Moscow, 115522, Russia
| | - Elena Lobakova
- Faculty of Biology, Lomonosov Moscow State University, 1-12 Leninskie Gory, Moscow, 119192, Russia
| |
Collapse
|
105
|
Rouillard KR, Markovetz MR, Bacudio LG, Hill DB, Schoenfisch MH. Pseudomonas aeruginosa Biofilm Eradication via Nitric Oxide-Releasing Cyclodextrins. ACS Infect Dis 2020; 6:1940-1950. [PMID: 32510928 DOI: 10.1021/acsinfecdis.0c00246] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Pseudomonas aeruginosa is the main contributor to the morbidity and mortality of cystic fibrosis (CF) patients. Chronic respiratory infections are rarely eradicated due to protection from CF mucus and the biofilm matrix. The composition of the biofilm matrix determines its viscoelastic properties and affects antibiotic efficacy. Nitric oxide (NO) can both disrupt the physical structure of the biofilm and eradicate interior colonies. The effects of a CF-like growth environment on P. aeruginosa biofilm susceptibility to NO were investigated using parallel plate macrorheology and particle tracking microrheology. Biofilms grown in the presence of mucins and DNA contained greater concentrations of DNA in the matrix and exhibited concomitantly larger viscoelastic moduli compared to those grown in tryptic soy broth. Greater viscoelastic moduli correlated with increased tolerance to tobramycin and colistin. Remarkably, NO-releasing cyclodextrins eradicated all biofilms at the same concentration. The capacity of NO-releasing cyclodextrins to eradicate P. aeruginosa biofilms irrespective of matrix composition suggests that NO-based therapies may be superior antibiofilm treatments compared to conventional antibiotics.
Collapse
Affiliation(s)
- Kaitlyn R. Rouillard
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Matthew R. Markovetz
- Marsico Lung Institute/CF Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Lawrence G. Bacudio
- Marsico Lung Institute/CF Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - David B. Hill
- Marsico Lung Institute/CF Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Department of Physics and Astronomy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Mark H. Schoenfisch
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Vast Therapeutics, Durham, North Carolina 27703, United States
- UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
106
|
Lo Sciuto A, Cervoni M, Stefanelli R, Mancone C, Imperi F. Effect of lipid A aminoarabinosylation on Pseudomonas aeruginosa colistin resistance and fitness. Int J Antimicrob Agents 2020; 55:105957. [PMID: 32278012 DOI: 10.1016/j.ijantimicag.2020.105957] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 03/10/2020] [Accepted: 03/20/2020] [Indexed: 01/06/2023]
Abstract
Colistin represents the last-line treatment option against many multidrug-resistant Gram-negative pathogens. Several lines of evidence indicate that aminoarabinosylation of the lipid A moiety of lipopolysaccharide (LPS) is an essential step for the development of colistin resistance in Pseudomonas aeruginosa. However, whether it is sufficient to confer resistance in this bacterium remains unclear. The aim of this work was to investigate the specific contribution of lipid A aminoarabinosylation to colistin resistance in P. aeruginosa and evaluate the effect of this resistance mechanism on bacterial fitness. Recombinant strains constitutively expressing the enzymes for lipid A aminoarabinosylation were generated in a small collection of reference and clinical isolates and verified by quantitative reverse transcription polymerase chain reaction (qRT-PCR), lipid A extraction and mass spectrometry. The effect of aminoarabinosylated lipid A on colistin resistance was found to be strain- and culture condition-dependent. Higher levels of resistance were generally obtained in the presence of divalent cations, which appear to be important for aminoarabinosylation-mediated colistin resistance. High colistin resistance was also observed for most strains in human serum and in artificial sputum medium, which should partly mimic growth conditions during infection. The results of growth, biofilm, cell envelope integrity and Galleria mellonella infection assays indicate that lipid A aminoarabinosylation does not cause relevant fitness costs in P. aeruginosa.
Collapse
Affiliation(s)
| | | | - Roberta Stefanelli
- Department of Science, Roma Tre University, Rome, Italy; Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Carmine Mancone
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | | |
Collapse
|
107
|
Dench J, Hinz A, Aris‐Brosou S, Kassen R. Identifying the drivers of computationally detected correlated evolution among sites under antibiotic selection. Evol Appl 2020; 13:781-793. [PMID: 32211067 PMCID: PMC7086105 DOI: 10.1111/eva.12900] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 10/02/2019] [Accepted: 11/14/2019] [Indexed: 11/29/2022] Open
Abstract
The ultimate causes of correlated evolution among sites in a genome remain difficult to tease apart. To address this problem directly, we performed a high-throughput search for correlated evolution among sites associated with resistance to a fluoroquinolone antibiotic using whole-genome data from clinical strains of Pseudomonas aeruginosa, before validating our computational predictions experimentally. We show that for at least two sites, this correlation is underlain by epistasis. Our analysis also revealed eight additional pairs of synonymous substitutions displaying correlated evolution underlain by physical linkage, rather than selection associated with antibiotic resistance. Our results provide direct evidence that both epistasis and physical linkage among sites can drive the correlated evolution identified by high-throughput computational tools. In other words, the observation of correlated evolution is not by itself sufficient evidence to guarantee that the sites in question are epistatic; such a claim requires additional evidence, ideally coming from direct estimates of epistasis, based on experimental evidence.
Collapse
Affiliation(s)
- Jonathan Dench
- Department of BiologyUniversity of OttawaOttawaOntarioCanada
| | - Aaron Hinz
- Department of BiologyUniversity of OttawaOttawaOntarioCanada
| | - Stéphane Aris‐Brosou
- Department of BiologyUniversity of OttawaOttawaOntarioCanada
- Department of Mathematics and StatisticsUniversity of OttawaOttawaOntarioCanada
| | - Rees Kassen
- Department of BiologyUniversity of OttawaOttawaOntarioCanada
| |
Collapse
|
108
|
Activity of Antibiotics against Pseudomonas aeruginosa in an In Vitro Model of Biofilms in the Context of Cystic Fibrosis: Influence of the Culture Medium. Antimicrob Agents Chemother 2020; 64:AAC.02204-19. [PMID: 32015047 DOI: 10.1128/aac.02204-19] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 01/24/2020] [Indexed: 12/28/2022] Open
Abstract
Pseudomonas aeruginosa is a major cause of respiratory biofilm-related infections in patients with cystic fibrosis. We developed an in vitro pharmacodynamic model to study the activity of antipseudomonal antibiotics against PAO1 biofilms grown in artificial sputum medium with agar [ASM(+)] versus that against biofilms grown in Trypticase soy broth supplemented with glucose and NaCl (TGN). We measured bacterial counts, metabolic activity (fluorescein diacetate [FDA] hydrolysis), and biomass (crystal violet absorbance). Biofilms grew slower in ASM(+) than in TGN but reached the same CFU counts and metabolic activity in both media and a slightly higher biomass after 48 h in ASM(+) than in TGN. The concentration-response curves of the antibiotics after 24 h of incubation with mature biofilms showed maximal effects ranging from a 3 (ciprofloxacin)- to a 1.5 (ceftazidime, meropenem)-log10-CFU decrease, with tobramycin and colistin showing intermediate values. These maximal reductions in the numbers of CFU were similar in both media for ciprofloxacin and β-lactams but lower in ASM(+) than in TGN for tobramycin and colistin; they were reached at concentrations lower than the human maximum concentration in plasma for ciprofloxacin and β-lactams only. The reductions in metabolic activity and in biomass were low in both media. Small-colony variants were selected by tobramycin in ASM(+) and by ciprofloxacin in both media. The model was then successfully applied to 4 isolates from patients with cystic fibrosis. These biofilms showed CFU counts similar to those of PAO1 biofilms in ASM(+) but a higher biomass than PAO1 biofilms in ASM(+) and moderate differences in their susceptibility to antibiotics from that of PAO1 biofilms grown in this medium. This model proved useful to establish the pharmacodynamic profile of drugs against P. aeruginosa biofilms in the context of cystic fibrosis.
Collapse
|
109
|
Effects of Lysozyme, Proteinase K, and Cephalosporins on Biofilm Formation by Clinical Isolates of Pseudomonas aeruginosa. Interdiscip Perspect Infect Dis 2020; 2020:6156720. [PMID: 32089678 PMCID: PMC7031717 DOI: 10.1155/2020/6156720] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/01/2020] [Accepted: 01/10/2020] [Indexed: 02/07/2023] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen that can form biofilms, which confer resistance to immune clearance and antibacterial treatment. Therefore, effective strategies to prevent biofilm formation are warranted. Here, 103 P. aeruginosa clinical isolates were quantitatively screened for biofilm formation ability via the tissue culture plate method. The effects of lysozyme (hydrolytic enzyme) and proteinase K (protease) on biofilm formation were evaluated at different concentrations. Lysozyme (30 μg/mL), but not proteinase K, significantly inhibited biofilm formation (19% inhibition). Treatment of 24-hour-old biofilms of P. aeruginosa isolates with 50 times the minimum inhibitory concentrations (MICs) of ceftazidime and cefepime significantly decreased the biofilm mass by 32.8% and 44%, respectively. Moreover, the exposure of 24-hour-old biofilms of P. aeruginosa isolates to lysozyme (30 μg/mL) and 50 times MICs of ceftazidime or cefepime resulted in a significant reduction in biofilm mass as compared with the exposure to lysozyme or either antibacterial agent alone. The best antibiofilm effect (49.3%) was observed with the combination of lysozyme (30 μg/mL) and 50 times MIC of cefepime. The promising antibiofilm activity observed after treatment with 50 times MIC of ceftazidime or cefepime alone or in combination with lysozyme (30 μg/mL) is indicative of a novel strategy to eradicate pseudomonal biofilms in intravascular devices and contact lenses.
Collapse
|
110
|
Treffon J, Chaves-Moreno D, Niemann S, Pieper DH, Vogl T, Roth J, Kahl BC. Importance of superoxide dismutases A and M for protection of Staphylococcus aureus in the oxidative stressful environment of cystic fibrosis airways. Cell Microbiol 2020; 22:e13158. [PMID: 31895486 DOI: 10.1111/cmi.13158] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 12/18/2019] [Accepted: 12/20/2019] [Indexed: 12/18/2022]
Abstract
Staphylococcus aureus is one of the earliest pathogens that persists the airways of cystic fibrosis (CF) patients and contributes to increased inflammation and decreased lung function. In contrast to other staphylococci, S. aureus possesses two superoxide dismutases (SODs), SodA and SodM, with SodM being unique to S. aureus. Both SODs arm S. aureus for its fight against oxidative stress, a by-product of inflammatory reactions. Despite complex investigations, it is still unclear if both enzymes are crucial for the special pathogenicity of S. aureus. To investigate the role of both SODs during staphylococcal persistence in CF airways, we analysed survival and gene expression of S. aureus CF isolates and laboratory strains in different CF-related in vitro and ex vivo settings. Bacteria located in inflammatory and oxidised CF sputum transcribed high levels of sodA and sodM. Especially expression values of sodM were remarkably higher in CF sputum than in bacterial in vitro cultures. Interestingly, also S. aureus located in airway epithelial cells expressed elevated transcript numbers of both SODs, indicating that S. aureus is exposed to oxidative stress at various sites within CF airways. Both enzymes promoted survival of S. aureus during polymorphonuclear leukocyte killing and seem to act compensatory, thereby giving evidence that the interwoven interaction of SodA and SodM contributes to S. aureus virulence and facilitates S. aureus persistence within CF airways.
Collapse
Affiliation(s)
- Janina Treffon
- Institute of Medical Microbiology, University Hospital Münster, Münster, Germany
| | - Diego Chaves-Moreno
- Microbial Interactions and Processes Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Silke Niemann
- Institute of Medical Microbiology, University Hospital Münster, Münster, Germany
| | - Dietmar Helmut Pieper
- Microbial Interactions and Processes Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Thomas Vogl
- Institute of Immunology, University Hospital Münster, Münster, Germany
| | - Johannes Roth
- Institute of Immunology, University Hospital Münster, Münster, Germany
| | - Barbara C Kahl
- Institute of Medical Microbiology, University Hospital Münster, Münster, Germany
| |
Collapse
|
111
|
Britt NS, Hazlett DS, Horvat RT, Liesman RM, Steed ME. Activity of pulmonary vancomycin exposures versus planktonic and biofilm isolates of methicillin-resistant Staphylococcus aureus from cystic fibrosis sputum. Int J Antimicrob Agents 2020; 55:105898. [PMID: 31931147 DOI: 10.1016/j.ijantimicag.2020.105898] [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: 08/09/2019] [Revised: 11/30/2019] [Accepted: 01/04/2020] [Indexed: 11/30/2022]
Abstract
Vancomycin is commonly used to treat methicillin-resistant Staphylococcus aureus (MRSA) infections in patients with cystic fibrosis (CF) lung disease. However, there are limited data to support the in vitro activity of this agent against MRSA isolated from CF sputum. The primary objective of this study was to evaluate the activity of vancomycin at pulmonary concentrations (intravenous and inhaled) against four clinical MRSA CF sputum isolates in planktonic and biofilm time-kill (TK) experiments. Vancomycin minimum inhibitory concentrations (MICs) were determined for these isolates at standard inoculum (SI) (~106 CFU/mL) and high inoculum (HI) (~108 CFU/mL) as well as in biofilms cultivated using physiological medium representing the microenvironment of the CF lung. Vancomycin concentrations of 10, 25, 100 and 275 µg/mL were evaluated in TK experiments against planktonic MRSA at varying inocula and versus biofilm MRSA. Vancomycin MICs increased from 0.5 µg/mL when tested at SI to 8-16 µg/mL at HI. Vancomycin MICs were further increased to 16-32 µg/mL in biofilm studies. In TK experiments, vancomycin displayed bactericidal activity (≥3 log10 killing at 24 h) against 1/4 and 0/4 planktonic MRSA isolates at SI and HI, respectively, whereas vancomycin was bactericidal against 0/4 isolates against MRSA biofilms. Based on these findings, vancomycin monotherapy appears unlikely to eradicate MRSA from the respiratory tract of patients with CF, even at high concentrations similar to those observed with inhaled therapy. Novel vancomycin formulations with enhanced biofilm penetration or combination therapy with other potentially synergistic agents should be explored.
Collapse
Affiliation(s)
- Nicholas S Britt
- Department of Pharmacy Practice, University of Kansas School of Pharmacy, Lawrence, Kansas, USA; Department of Internal Medicine, Division of Infectious Diseases, University of Kansas School of Medicine, Kansas City, Kansas, USA.
| | - Daniel S Hazlett
- Department of Pharmacy Practice, University of Kansas School of Pharmacy, Lawrence, Kansas, USA
| | - Rebecca T Horvat
- Department of Pathology and Laboratory Medicine, University of Kansas School of Medicine, Kansas City, Kansas, USA
| | - Rachael M Liesman
- Department of Pathology and Laboratory Medicine, University of Kansas School of Medicine, Kansas City, Kansas, USA
| | - Molly E Steed
- Department of Pharmacy Practice, University of Kansas School of Pharmacy, Lawrence, Kansas, USA
| |
Collapse
|
112
|
Thibault D, Jensen PA, Wood S, Qabar C, Clark S, Shainheit MG, Isberg RR, van Opijnen T. Droplet Tn-Seq combines microfluidics with Tn-Seq for identifying complex single-cell phenotypes. Nat Commun 2019; 10:5729. [PMID: 31844066 PMCID: PMC6914776 DOI: 10.1038/s41467-019-13719-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 11/18/2019] [Indexed: 02/07/2023] Open
Abstract
While Tn-Seq is a powerful tool to determine genome-wide bacterial fitness in high-throughput, culturing transposon-mutant libraries in pools can mask community or other complex single-cell phenotypes. Droplet Tn-Seq (dTn-Seq) solves this problem by microfluidics facilitated encapsulation of individual transposon mutants into growth medium-in-oil droplets, thereby enabling isolated growth, free from the influence of the population. Here we describe and validate microfluidic chip design, production, encapsulation, and dTn-Seq sample preparation. We determine that 1-3% of mutants in Streptococcus pneumoniae have a different fitness when grown in isolation and show how dTn-Seq can help identify leads for gene function, including those involved in hyper-competence, processing of alpha-1-acid glycoprotein, sensitivity against the human leukocyte elastase and microcolony formation. Additionally, we show dTn-Seq compatibility with microscopy, FACS and investigations of bacterial cell-to-cell and bacteria-host cell interactions. dTn-Seq reduces costs and retains the advantages of Tn-Seq, while expanding the method's original applicability.
Collapse
Affiliation(s)
- Derek Thibault
- Biology Department, Boston College, Chestnut Hill, MA, 02467, USA
| | - Paul A Jensen
- Biology Department, Boston College, Chestnut Hill, MA, 02467, USA
- Department of Bioengineering and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Stephen Wood
- Biology Department, Boston College, Chestnut Hill, MA, 02467, USA
| | - Christine Qabar
- Department of Biological Sciences, Towson University, Towson, MD, 21252, USA
| | - Stacie Clark
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, 02111, USA
| | - Mara G Shainheit
- Department of Biological Sciences, Towson University, Towson, MD, 21252, USA
| | - Ralph R Isberg
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, 02111, USA
| | - Tim van Opijnen
- Biology Department, Boston College, Chestnut Hill, MA, 02467, USA.
| |
Collapse
|
113
|
Millette G, Langlois JP, Brouillette E, Frost EH, Cantin AM, Malouin F. Despite Antagonism in vitro, Pseudomonas aeruginosa Enhances Staphylococcus aureus Colonization in a Murine Lung Infection Model. Front Microbiol 2019; 10:2880. [PMID: 31921058 PMCID: PMC6923662 DOI: 10.3389/fmicb.2019.02880] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 11/29/2019] [Indexed: 11/13/2022] Open
Abstract
Staphylococcus aureus and Pseudomonas aeruginosa are prevalent lung pathogens in cystic fibrosis (CF). Whereas co-infection worsens the clinical outcome, prototypical strains are usually antagonistic in vitro. We sought to resolve the discrepancy between these in vitro and in vivo observations. In vitro, growth kinetics for co-cultures of co-isolates from CF patients showed that not all P. aeruginosa strains affected S. aureus viability. On solid media, S. aureus slow-growing colonies were visualized around some P. aeruginosa strains whether or not S. aureus viability was reduced in liquid co-cultures. The S. aureus-P. aeruginosa interactions were then characterized in a mouse lung infection model. Lung homogenates were plated on selective media allowing colony counts of either bacterium. Overall, 35 P. aeruginosa and 10 S. aureus strains (clinical, reference, and mutant strains), for a total of 200 co-infections, were evaluated. We observed that S. aureus colonization of lung tissues was promoted by P. aeruginosa and even by strains showing antagonism in vitro. Promotion was proportional to the extent of P. aeruginosa colonization, but no correlation was found with the degree of myeloperoxidase quantification (as marker of inflammation) or with specific virulence-associated factors using known mutant strains of S. aureus and P. aeruginosa. On the other hand, P. aeruginosa significantly increased the expression of two possible cell receptors for S. aureus, i.e., ICAM-1 and ITGA-5 (marker for integrin α5β1) in lung tissue, while mono-infections by S. aureus did not. This study provides insights on polymicrobial interactions that may influence the progression of CF-associated pulmonary infections.
Collapse
Affiliation(s)
- Guillaume Millette
- Centre d'Étude et de Valorisation de la Diversité Microbienne, Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Jean-Philippe Langlois
- Centre d'Étude et de Valorisation de la Diversité Microbienne, Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Eric Brouillette
- Centre d'Étude et de Valorisation de la Diversité Microbienne, Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Eric H Frost
- Département de Microbiologie et d'Infectiologie, Faculté de Médecine et de Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - André M Cantin
- Service de Pneumologie, Département de Médecine, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - François Malouin
- Centre d'Étude et de Valorisation de la Diversité Microbienne, Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
114
|
Hall JR, Maloney SE, Jin H, Taylor JB, Schoenfisch MH. Nitric oxide diffusion through cystic fibrosis-relevant media and lung tissue. RSC Adv 2019; 9:40176-40183. [PMID: 32655858 PMCID: PMC7351356 DOI: 10.1039/c9ra07367a] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
A simplified diffusion cell methodology was employed to measure the diffusion coefficient of nitric oxide (NO) through phosphate buffered saline (PBS) and artificial sputum medium (ASM)—an in vitro analog for airway mucus. Diffusion through the proteinaceous ASM yielded a significantly lower diffusion coefficient compared to PBS, which is attributed to both the physical obstruction by the mucin mesh and reactive nature of NO radicals towards the biological compounds in ASM. To further confirm that ASM was restricting NO from diffusing freely, a macromolecular propylamine-modified cyclodextrin donor (CD-PA) was employed to release the NO more slowly. The NO diffusion characteristics in ASM via the NO donor were also slower relative to PBS. As NO is likely to interact with lung cells after passing through the mucus barrier, the diffusion of both NO and the CD-PA macromolecular NO donor through differentiated lung tissue was investigated with and without an ASM layer. Comparison of NO diffusion through the three diffusion barriers indicated that the lung tissue significantly impeded NO penetration over the course of the experiment compared to PBS and ASM. In fact, the diffusion of CD-PA through the lung tissue was hindered until after the release of its NO payload, potentially due to the increased net charge of the NO donor structure. Of importance, the viability of the tissue was not influenced by the NO-releasing CD-PA at bactericidal concentrations. Nitric oxide diffusion monitored through artificial sputum medium using an adaptable diffusion cell and released from donor through human lung tissue.![]()
Collapse
Affiliation(s)
- Jackson R Hall
- Department of Chemistry, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599
| | - Sara E Maloney
- Department of Chemistry, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599
| | - Haibao Jin
- Department of Chemistry, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599
| | - James B Taylor
- Department of Chemistry, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599
| | - Mark H Schoenfisch
- Department of Chemistry, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599
| |
Collapse
|
115
|
O'Brien TJ, Welch M. Recapitulation of polymicrobial communities associated with cystic fibrosis airway infections: a perspective. Future Microbiol 2019; 14:1437-1450. [PMID: 31778075 DOI: 10.2217/fmb-2019-0200] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The airways of persons with cystic fibrosis are prone to infection by a diverse and dynamic polymicrobial consortium. Currently, no models exist that permit recapitulation of this consortium within the laboratory. Such microbial ecosystems likely have a network of interspecies interactions, serving to modulate metabolic pathways and impact upon disease severity. The contribution of less abundant/fastidious microbial species on this cross-talk has often been neglected due to lack of experimental tractability. Here, we critically assess the existing models for studying polymicrobial infections. Particular attention is paid to 3Rs-compliant in vitro and in silico infection models, offering significant advantages over mammalian infection models. We outline why these models will likely become the 'go to' approaches when recapitulating polymicrobial cystic fibrosis infection.
Collapse
Affiliation(s)
- Thomas J O'Brien
- Department of Biochemistry, University of Cambridge, Cambridge, CB2 1QW, UK
| | - Martin Welch
- Department of Biochemistry, University of Cambridge, Cambridge, CB2 1QW, UK
| |
Collapse
|
116
|
Hassan AA, Vitorino MV, Robalo T, Rodrigues MS, Sá-Correia I. Variation of Burkholderia cenocepacia cell wall morphology and mechanical properties during cystic fibrosis lung infection, assessed by atomic force microscopy. Sci Rep 2019; 9:16118. [PMID: 31695169 PMCID: PMC6834607 DOI: 10.1038/s41598-019-52604-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 10/21/2019] [Indexed: 12/13/2022] Open
Abstract
The influence that Burkholderia cenocepacia adaptive evolution during long-term infection in cystic fibrosis (CF) patients has on cell wall morphology and mechanical properties is poorly understood despite their crucial role in cell physiology, persistent infection and pathogenesis. Cell wall morphology and physical properties of three B. cenocepacia isolates collected from a CF patient over a period of 3.5 years were compared using atomic force microscopy (AFM). These serial clonal variants include the first isolate retrieved from the patient and two late isolates obtained after three years of infection and before the patient's death with cepacia syndrome. A consistent and progressive decrease of cell height and a cell shape evolution during infection, from the typical rods to morphology closer to cocci, were observed. The images of cells grown in biofilms showed an identical cell size reduction pattern. Additionally, the apparent elasticity modulus significantly decreases from the early isolate to the last clonal variant retrieved from the patient but the intermediary highly antibiotic resistant clonal isolate showed the highest elasticity values. Concerning the adhesion of bacteria surface to the AFM tip, the first isolate was found to adhere better than the late isolates whose lipopolysaccharide (LPS) structure loss the O-antigen (OAg) during CF infection. The OAg is known to influence Gram-negative bacteria adhesion and be an important factor in B. cenocepacia adaptation to chronic infection. Results reinforce the concept of the occurrence of phenotypic heterogeneity and adaptive evolution, also at the level of cell size, form, envelope topography and physical properties during long-term infection.
Collapse
Affiliation(s)
- A Amir Hassan
- iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, 1049-001, Portugal
- Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, 1049-001, Portugal
| | - Miguel V Vitorino
- BioISI - Biosystems and Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, 1749-016, Lisboa, Portugal
- Departamento de Física, Faculdade de Ciências, Universidade de Lisboa, 1749-016, Lisboa, Portugal
| | - Tiago Robalo
- BioISI - Biosystems and Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, 1749-016, Lisboa, Portugal
| | - Mário S Rodrigues
- BioISI - Biosystems and Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, 1749-016, Lisboa, Portugal.
- Departamento de Física, Faculdade de Ciências, Universidade de Lisboa, 1749-016, Lisboa, Portugal.
| | - Isabel Sá-Correia
- iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, 1049-001, Portugal.
- Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, 1049-001, Portugal.
| |
Collapse
|
117
|
Flynn S, Reen FJ, O'Gara F. Exposure to Bile Leads to the Emergence of Adaptive Signaling Variants in the Opportunistic Pathogen Pseudomonas aeruginosa. Front Microbiol 2019; 10:2013. [PMID: 31555243 PMCID: PMC6727882 DOI: 10.3389/fmicb.2019.02013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 08/16/2019] [Indexed: 12/18/2022] Open
Abstract
The chronic colonization of the respiratory tract by the opportunistic pathogen Pseudomonas aeruginosa is the primary cause of morbidity and mortality in cystic fibrosis (CF) patients. P. aeruginosa has been shown to undergo extensive genomic adaptation facilitating its persistence within the CF lung allowing it to evade the host immune response and outcompete co-colonizing residents of the lung microbiota. However, whilst several studies have described the various mutations that frequently arise in clinical isolates of P. aeruginosa, the environmental factors governing the emergence of these genetic variants is less well characterized. Gastro-oesophageal reflux has recently emerged as a major co-morbidity in CF and is often associated with the presence of bile acids in the lungs most likely by (micro) aspiration. In order to investigate whether bile may select for genetic variants, P. aeruginosa was experimentally evolved in artificial sputum medium, a synthetic media resembling environmental conditions found within the CF lung. Pigmented derivatives of P. aeruginosa emerged exclusively in the presence of bile. Genome sequencing analysis identified single nucleotide polymorphisms (SNPs) in quorum sensing (lasR) and both the pyocyanin (phzS) and pyomelanin (hmgA) biosynthetic pathways. Phenotypic analysis revealed an altered bile response when compared to the ancestral P. aeruginosa progenitor strain. While the recovered pigmented derivatives retained the bile mediated suppression of swarming motility and enhanced antibiotic tolerance, the biofilm, and redox responses to bile were abolished in the adapted mutants. Though loss of pseudomonas quinolone signal (PQS) production in the pigmented isolates was not linked to the altered biofilm response, the loss of redox repression could be explained by defective alkyl-quinolone (AQ) production in the presence of bile. Collectively, these findings suggest that the adaptive variants of P. aeruginosa that arise following long term bile exposure enables the emergence of ecologically competitive sub-populations. Altered pigmentation and AQ signaling may contribute to an enhancement in fitness facilitating population survival within a bile positive environment.
Collapse
Affiliation(s)
- Stephanie Flynn
- BIOMERIT Research Centre, School of Microbiology, University College Cork - National University of Ireland, Cork, Ireland
| | - F Jerry Reen
- School of Microbiology, University College Cork - National University of Ireland, Cork, Ireland
| | - Fergal O'Gara
- BIOMERIT Research Centre, School of Microbiology, University College Cork - National University of Ireland, Cork, Ireland.,Telethon Kids Institute, Perth, WA, Australia.,School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
| |
Collapse
|
118
|
Sousa SA, Soares-Castro P, Seixas AMM, Feliciano JR, Balugas B, Barreto C, Pereira L, Santos PM, Leitão JH. New insights into the immunoproteome of B. cenocepacia J2315 using serum samples from cystic fibrosis patients. N Biotechnol 2019; 54:62-70. [PMID: 31465856 DOI: 10.1016/j.nbt.2019.08.006] [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: 07/19/2018] [Revised: 07/09/2019] [Accepted: 08/24/2019] [Indexed: 11/26/2022]
Abstract
Bacteria of the Burkholderia cepacia complex (Bcc) are ubiquitous multidrug resistant organisms and opportunistic pathogens capable of causing life threatening lung infections among cystic fibrosis (CF) patients. No effective therapies are currently available to eradicate Bcc bacteria from CF patients, as these organisms are inherently resistant to the majority of clinically available antimicrobials. An immunoproteomics approach was used to identify Bcc proteins that stimulate the humoral immune response of the CF host, using bacterial cells grown under conditions mimicking the CF lung environment and serum samples from CF patients with a clinical record of Bcc infection. 24 proteins of the Bcc strain B. cenocepacia J2315 were identified as immunoreactive, 19 here reported as immunogenic for the first time. Ten proteins were predicted as extracytoplasmic, 9 of them being conserved in Bcc genomes. The immunogenic Bcc extracytoplasmic proteins are potential targets for development of novel therapeutic strategies and diagnostic tools to protect patients against the onset of chronic Bcc lung infections.
Collapse
Affiliation(s)
- Sílvia A Sousa
- iBB-Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisbon, Portugal.
| | - Pedro Soares-Castro
- CBMA-Centre of Molecular and Environmental Biology, Department of Biology, University of Minho, Campus de Gualtar, Braga, Portugal
| | - António M M Seixas
- iBB-Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisbon, Portugal
| | - Joana R Feliciano
- iBB-Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisbon, Portugal
| | - Bernardo Balugas
- iBB-Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisbon, Portugal
| | - Celeste Barreto
- Cystic Fibrosis Center, Department of Paediatrics, Hospital de Santa Maria, Centro Hospitalar Lisboa Norte, Lisbon, Portugal
| | - Luísa Pereira
- Cystic Fibrosis Center, Department of Paediatrics, Hospital de Santa Maria, Centro Hospitalar Lisboa Norte, Lisbon, Portugal
| | - Pedro M Santos
- CBMA-Centre of Molecular and Environmental Biology, Department of Biology, University of Minho, Campus de Gualtar, Braga, Portugal
| | - Jorge H Leitão
- iBB-Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisbon, Portugal.
| |
Collapse
|
119
|
Ahonen MJR, Dorrier JM, Schoenfisch MH. Antibiofilm Efficacy of Nitric Oxide-Releasing Alginates against Cystic Fibrosis Bacterial Pathogens. ACS Infect Dis 2019; 5:1327-1335. [PMID: 31136714 PMCID: PMC6773255 DOI: 10.1021/acsinfecdis.9b00016] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Colonization of the lungs by biofilm-forming pathogens is a major cause of mortality in cystic fibrosis (CF). In CF patients, these pathogens are difficult to treat due to the additional protection provided by both the biofilm exopolysaccharide matrix and thick, viscous mucus. The antibiofilm efficacy of nitric oxide (NO)-releasing alginates was evaluated against Pseudomonas aeruginosa, Burkholderia cepacia, Staphylococcus aureus, and methicillin-resistant S. aureus biofilms in both aerobic and anaerobic environments. Varying the amine precursor grafted onto alginate oligosaccharides imparted tunable NO storage (∼0.1-0.3 μmol/mg) and release kinetics (∼4-40 min half-lives) in the artificial sputum media used for biofilm testing. The NO-releasing alginates were highly antibacterial against the four CF-relevant pathogens, achieving a 5-log reduction in biofilm viability after 24 h of treatment, with biocidal efficacy dependent on NO-release kinetics. Aerobic biofilms required greater starting NO doses to achieve killing relative to the anaerobic biofilms. Relative to tobramycin (the minimum concentration of antibacterial agent required to achieve a 5-log reduction in viability after 24 h, MBEC24h, of ≥2000 μg/mL) and vancomycin (MBEC24h ≥ 1000 μg/mL), the NO-releasing alginates proved to be more effective (NO dose ≤ 520 μg/mL) regardless of growth conditions.
Collapse
Affiliation(s)
- Mona Jasmine R. Ahonen
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, CB 3290, NC 27599, United States
| | - Jamie M. Dorrier
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, CB 3290, NC 27599, United States
| | - Mark H. Schoenfisch
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, CB 3290, NC 27599, United States
| |
Collapse
|
120
|
Rozenbaum R, Andrén OCJ, van der Mei HC, Woudstra W, Busscher HJ, Malkoch M, Sharma PK. Penetration and Accumulation of Dendrons with Different Peripheral Composition in Pseudomonas aeruginosa Biofilms. NANO LETTERS 2019; 19:4327-4333. [PMID: 31142116 PMCID: PMC6628176 DOI: 10.1021/acs.nanolett.9b00838] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/28/2019] [Indexed: 06/09/2023]
Abstract
Multidrug resistant bacterial infections threaten to become the number one cause of death by the year 2050. Development of antimicrobial dendritic polymers is considered promising as an alternative infection control strategy. For antimicrobial dendritic polymers to effectively kill bacteria residing in infectious biofilms, they have to penetrate and accumulate deep into biofilms. Biofilms are often recalcitrant to antimicrobial penetration and accumulation. Therefore, this work aims to determine the role of compact dendrons with different peripheral composition in their penetration into Pseudomonas aeruginosa biofilms. Red fluorescently labeled dendrons with pH-responsive NH3+ peripheral groups initially penetrated faster from a buffer suspension at pH 7.0 into the acidic environment of P. aeruginosa biofilms than dendrons with OH or COO- groups at their periphery. In addition, dendrons with NH3+ peripheral groups accumulated near the top of the biofilm due to electrostatic double-layer attraction with negatively charged biofilm components. However, accumulation of dendrons with OH and COO- peripheral groups was more evenly distributed across the depth of the biofilms than NH3+ composed dendrons and exceeded accumulation of NH3+ composed dendrons after 10 min of exposure. Unlike dendrons with NH3+ groups at their periphery, dendrons with OH or COO- peripheral groups, lacking strong electrostatic double-layer attraction with biofilm components, were largely washed-out during exposure to PBS without dendrons. Thus, penetration and accumulation of dendrons into biofilms is controlled by their peripheral composition through electrostatic double-layer interactions, which is an important finding for the further development of new antimicrobial or antimicrobial-carrying dendritic polymers.
Collapse
Affiliation(s)
- René
T. Rozenbaum
- Department
of Biomedical Engineering, University of
Groningen and University Medical Center Groningen, P.O. Box 196, 9700 AD, Groningen, The Netherlands
| | - Oliver C. J. Andrén
- Department
of Fibre and Polymer Technology, KTH Royal
Institute of Technology, 10044 Stockholm, Sweden
| | - Henny C. van der Mei
- Department
of Biomedical Engineering, University of
Groningen and University Medical Center Groningen, P.O. Box 196, 9700 AD, Groningen, The Netherlands
| | - Willem Woudstra
- Department
of Biomedical Engineering, University of
Groningen and University Medical Center Groningen, P.O. Box 196, 9700 AD, Groningen, The Netherlands
| | - Henk J. Busscher
- Department
of Biomedical Engineering, University of
Groningen and University Medical Center Groningen, P.O. Box 196, 9700 AD, Groningen, The Netherlands
| | - Michael Malkoch
- Department
of Fibre and Polymer Technology, KTH Royal
Institute of Technology, 10044 Stockholm, Sweden
| | - Prashant K. Sharma
- Department
of Biomedical Engineering, University of
Groningen and University Medical Center Groningen, P.O. Box 196, 9700 AD, Groningen, The Netherlands
| |
Collapse
|
121
|
Suryaletha K, Narendrakumar L, John J, Radhakrishnan MP, George S, Thomas S. Decoding the proteomic changes involved in the biofilm formation of Enterococcus faecalis SK460 to elucidate potential biofilm determinants. BMC Microbiol 2019; 19:146. [PMID: 31253082 PMCID: PMC6599329 DOI: 10.1186/s12866-019-1527-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 06/20/2019] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Enterococcus faecalis is a major clinically relevant nosocomial bacterial pathogen frequently isolated from polymicrobial infections. The biofilm forming ability of E. faecalis attributes a key role in its virulence and drug resistance. Biofilm cells are phenotypically and metabolically different from their planktonic counterparts and many aspects involved in E. faecalis biofilm formation are yet to be elucidated. The strain E. faecalis SK460 used in the present study is esp (Enterococcal surface protein) and fsr (two-component signal transduction system) negative non-gelatinase producing strong biofilm former isolated from a chronic diabetic foot ulcer patient. We executed a label-free quantitative proteomic approach to elucidate the differential protein expression pattern at planktonic and biofilm stages of SK460 to come up with potential determinants associated with Enterococcal biofilm formation. RESULTS The Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses of proteomic data revealed that biofilm cells expressed higher levels of proteins which are associated with glycolysis, amino acid biosynthesis, biosynthesis of secondary metabolites, microbial metabolism in diverse environments and stress response factors. Besides these basic survival pathways, LuxS-mediated quorum sensing, arginine metabolism, rhamnose biosynthesis, pheromone and adhesion associated proteins were found to be upregulated during the biofilm transit from planktonic stages. The selected subsets were validated by quantitative real-time PCR. In silico functional interaction analysis revealed that the genes involved in upregulated pathways pose a close molecular interaction thereby coordinating the regulatory network to thrive as a biofilm community. CONCLUSIONS The present study describes the first report of the quantitative proteome analysis of an esp and fsr negative non gelatinase producing E. faecalis. Proteome analysis evidenced enhanced expression of glycolytic pathways, stress response factors, LuxS quorum signaling system, rhamnopolysaccharide synthesis and pheromone associated proteins in biofilm phenotype. We also pointed out the relevance of LuxS quorum sensing and pheromone associated proteins in the biofilm development of E. faecalis which lacks the Fsr quorum signaling system. These validated biofilm determinants can act as potential inhibiting targets in Enterococcal infections.
Collapse
Affiliation(s)
- Karthika Suryaletha
- Cholera and Biofilm Research Laboratory, Pathogen Biology, Rajiv Gandhi Centre for Biotechnology, (National Institute under the Department of Biotechnology, Government of India), Trivandrum, Kerala, 695014, India
| | - Lekshmi Narendrakumar
- Cholera and Biofilm Research Laboratory, Pathogen Biology, Rajiv Gandhi Centre for Biotechnology, (National Institute under the Department of Biotechnology, Government of India), Trivandrum, Kerala, 695014, India
| | - Joby John
- Department of Surgery, Government Medical College Hospital, Trivandrum, Kerala, 695011, India
| | - Megha Periyappilly Radhakrishnan
- Cholera and Biofilm Research Laboratory, Pathogen Biology, Rajiv Gandhi Centre for Biotechnology, (National Institute under the Department of Biotechnology, Government of India), Trivandrum, Kerala, 695014, India
| | - Sanil George
- Interdisciplinary Biology, Rajiv Gandhi Centre for Biotechnology, Trivandrum, Kerala, 695014, India
| | - Sabu Thomas
- Cholera and Biofilm Research Laboratory, Pathogen Biology, Rajiv Gandhi Centre for Biotechnology, (National Institute under the Department of Biotechnology, Government of India), Trivandrum, Kerala, 695014, India.
| |
Collapse
|
122
|
Mycobacterium abscessus Cells Have Altered Antibiotic Tolerance and Surface Glycolipids in Artificial Cystic Fibrosis Sputum Medium. Antimicrob Agents Chemother 2019; 63:AAC.02488-18. [PMID: 31010859 DOI: 10.1128/aac.02488-18] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 04/15/2019] [Indexed: 11/20/2022] Open
Abstract
Mycobacterium abscessus is a biofilm-forming, multidrug-resistant nontuberculous mycobacterial (NTM) pathogen increasingly found in cystic fibrosis patients. Antibiotic treatment for these infections is often unsuccessful, partly due to M. abscessus's high intrinsic antibiotic resistance. It is not clear whether antibiotic tolerance caused by biofilm formation also contributes to poor treatment outcomes. We studied the surface glycolipids and antibiotic tolerance of M. abscessus biofilms grown in artificial cystic fibrosis sputum (ACFS) medium to determine how they are affected by nutrient conditions that mimic infection. We found that M. abscessus displays more of the virulence lipid trehalose dimycolate when grown in ACFS than when grown in standard lab medium. In ACFS medium, biofilm-associated cells were more antibiotic tolerant than planktonic cells in the same well. This contrasts with standard lab media, where both biofilm and planktonic cells are highly antibiotic tolerant. These results indicate that M. abscessus cell physiology in biofilms depends on environmental factors and that nutrient conditions found within cystic fibrosis infections could contribute to both increased virulence and antibiotic tolerance.
Collapse
|
123
|
Activity of Antibiotics against Staphylococcus aureus in an In Vitro Model of Biofilms in the Context of Cystic Fibrosis: Influence of the Culture Medium. Antimicrob Agents Chemother 2019; 63:AAC.00602-19. [PMID: 31036685 DOI: 10.1128/aac.00602-19] [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: 03/21/2019] [Accepted: 04/20/2019] [Indexed: 01/27/2023] Open
Abstract
Staphylococcus aureus is a highly prevalent pathogen in the respiratory tract of young patients with cystic fibrosis (CF) and causes biofilm-related infections. Here, we set up an in vitro model of a biofilm grown in Trypticase soy broth supplemented with glucose and NaCl (TGN) or in artificial sputum medium (ASM) and used it to evaluate on a pharmacodynamic basis the activity of antibiotics used in CF patients and active on staphylococci (meropenem, vancomycin, azithromycin, linezolid, rifampin, ciprofloxacin, tobramycin). Rheological studies showed that ASM was more elastic than viscous, as was also observed for sputa from CF patients, with elastic and viscous moduli being, respectively, similar to and slightly lower than those of CF sputa. Biofilms formed by methicillin-sensitive S. aureus strain ATCC 25923 and methicillin-resistant S. aureus strain ATCC 33591 reached maturity after 24 h, with biomass (measured by crystal violet staining) and metabolic activity (assessed by following resazurin metabolization) being lower in ASM than in TGN and viability (assessed by bacterial counts) being similar in both media. Full concentration-response curves of antibiotics obtained after 24 h of incubation of biofilms showed that all antibiotics were drastically less potent and less efficient in ASM than in TGN toward viability, metabolic activity, and biomass. Tobramycin selected for small-colony variants, specifically in biofilms grown in ASM; the auxotrophism of these variants could not be established. These data highlight the major influence exerted by the culture medium on S. aureus responsiveness to antibiotics in biofilms. The use of ASM may help to determine effective drug concentrations or to evaluate new therapeutic options against biofilms in CF patients.
Collapse
|
124
|
Role of Viscoelasticity in Bacterial Killing by Antimicrobials in Differently Grown Pseudomonas aeruginosa Biofilms. Antimicrob Agents Chemother 2019; 63:AAC.01972-18. [PMID: 30745390 DOI: 10.1128/aac.01972-18] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 12/28/2018] [Indexed: 01/04/2023] Open
Abstract
Pseudomonas aeruginosa colonizes the sputum of most adult cystic fibrosis patients, forming difficult-to-eradicate biofilms in which bacteria are protected in their self-produced extracellular polymeric substance (EPS) matrices. EPS provide biofilms with viscoelastic properties, causing time-dependent relaxation after stress-induced deformation, according to multiple characteristic time constants. These time constants reflect different biofilm (matrix) components. Since the viscoelasticity of biofilms has been related to antimicrobial penetration but not yet bacterial killing, this study aims to relate killing of P. aeruginosa, in its biofilm mode of growth, by three antimicrobials to biofilm viscoelasticity. P. aeruginosa biofilms were grown for 18 h in a constant-depth film fermenter, with mucin-containing artificial sputum medium (ASM+), artificial sputum medium without mucin (ASM-), or Luria-Bertani (LB) broth; this yielded 100-μm-thick biofilms that differed in their amounts of matrix environmental DNA (eDNA) and polysaccharides. Low-load compression testing, followed by three-element Maxwell analyses, showed that the fastest relaxation component, associated with unbound water, was most important in LB-medium-grown biofilms. Slower components due to water with dissolved polysaccharides, insoluble polysaccharides, and eDNA were most important in the relaxation of ASM+-grown biofilms. ASM--grown biofilms showed intermediate stress relaxation. P. aeruginosa in LB-medium-grown biofilms was killed most by exposure to tobramycin, colistin, or an antimicrobial peptide, while ASM+ provided the most protective matrix, with less water and most insoluble polysaccharides and eDNA. In conclusion, stress relaxation of P. aeruginosa biofilms grown in different media revealed differences in matrix composition that, within the constraints of the antimicrobials and growth media applied, correlated with the matrix protection offered against different antimicrobials.
Collapse
|
125
|
Tovi N, Frenk S, Hadar Y, Minz D. Host Specificity and Spatial Distribution Preference of Three Pseudomonas Isolates. Front Microbiol 2019; 9:3263. [PMID: 30687261 PMCID: PMC6335278 DOI: 10.3389/fmicb.2018.03263] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 12/14/2018] [Indexed: 12/17/2022] Open
Abstract
Plant hosts recruit and maintain a distinct root-associated microbiota based on host and bacterium traits. However, past studies disregarded microbial strain-host specificity and spatial micro-heterogeneity of the root compartment. Using genetic manipulation, confocal laser scanning microscopy, real-time quantitative PCR, and genome sequencing we characterized the colonization patterns of three Pseudomonas spp. isolates native to wheat roots, on the micro-scale. Namely, isolates P. fluorescens NT0133, P. stutzeri NT124, and P. stutzeri NT128. All three isolates preferentially colonized wheat over cucumber roots that served as control for host specificity. Furthermore, not only had the isolates strong host specificity but each isolate had a distinct spatial distribution on the root, all within a few millimeters. Isolate P. stutzeri-NT0124 preferentially colonized root tips, whereas P. fluorescens-NT0133 showed a preference for zones distant from the tip. In contrast, isolate P. stutzeri-NT0128 had no preference for a specific niche on the root. While all isolates maintained genetic potential for motility and biofilm formation their phenotype varied significantly and corresponded to their niche preference. These results demonstrate the importance of spatial colonization patterns, governed by both niche and bacterial characteristics which will have great importance in future attempts to manipulate the plant microbiome by constructing synthetic microbial consortia.
Collapse
Affiliation(s)
- Nesli Tovi
- Department of Soil, Water, and Environmental Sciences, Agricultural Research Organization–Volcani Center, Rishon LeZion, Israel
- Department of Plant Pathology and Microbiology, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Sammy Frenk
- Australian Centre for Ecogenomics, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Yitzhak Hadar
- Department of Plant Pathology and Microbiology, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Dror Minz
- Department of Soil, Water, and Environmental Sciences, Agricultural Research Organization–Volcani Center, Rishon LeZion, Israel
| |
Collapse
|
126
|
Screening for Growth-Inhibitory ORFans in Pseudomonas aeruginosa-Infecting Bacteriophages. Methods Mol Biol 2019; 1898:147-162. [PMID: 30570730 DOI: 10.1007/978-1-4939-8940-9_12] [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] [Indexed: 01/14/2023]
Abstract
Like all viruses, bacteriophages heavily depend on their host's physiology for reproduction. Therefore, phages have evolved numerous proteins that influence the host metabolism to facilitate the infection process. Some of these proteins strongly perturb the host cell, ultimately leading to cell death. These growth-inhibitory phage proteins presumably target key metabolic processes, which may provide a basis for innovative phage-derived antibacterials. Unfortunately, most of these proteins are the so-called ORFans, since they have no known function or sequence homology to any other gene. We here describe a screening method for the identification of growth-inhibitory ORFans of bacteriophages infecting gram-negative bacteria (e.g., Pseudomonas aeruginosa), using the pUC18-mini-Tn7T-Lac vector system, which allows for stable single-copy integration of the phage ORFans in the Pseudomonas genome under the control of an IPTG-inducible promoter. Furthermore, we describe a method to examine the effect of the phage proteins in different hosts, using different vector copy numbers. Finally, we explain how to investigate the effect of ORFan expression on the host morphology using time-lapse microscopy.
Collapse
|
127
|
Sousa AM, Monteiro R, Pereira MO. Unveiling the early events of Pseudomonas aeruginosa adaptation in cystic fibrosis airway environment using a long-term in vitro maintenance. Int J Med Microbiol 2018; 308:1053-1064. [PMID: 30377031 DOI: 10.1016/j.ijmm.2018.10.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 09/16/2018] [Accepted: 10/10/2018] [Indexed: 01/22/2023] Open
Abstract
Pseudomonas aeruginosa chronic infections are the major cause of high morbidity and mortality in cystic fibrosis (CF) patients due to the use of sophisticated mechanisms of adaptation, including clonal diversification into specialized CF-adapted phenotypes. In contrast to chronic infections, very little is known about what occurs after CF lungs colonization and at early infection stages. This study aims to investigate the early events of P. aeruginosa adaptation to CF environment, in particular, to inspect the occurrence of clonal diversification at early stages of infection development and its impact on antibiotherapy effectiveness. To mimic CF early infections, three P. aeruginosa strains were long-term grown in artificial sputum (ASM) over 10 days and phenotypic diversity verified through colony morphology characterization. Biofilm sub- and inhibitory concentrations of ciprofloxacin were applied to non- and diversified populations to evaluate antibiotic effectiveness on P. aeruginosa eradication. Our results demonstrated that clonal diversification might occur after ASM colonization and growth. However, this phenotypic diversification did not compromise ciprofloxacin efficacy in P. aeruginosa eradication since a biofilm minimal inhibitory dosage would be applied. The expected absence of mutators in P. aeruginosa populations led us to speculate that clonal diversification in the absence of ciprofloxacin treatments could be driven by niche specialization. Yet, biofilm sub-inhibitory concentrations of ciprofloxacin seemed to overlap niche specialization as "fitter" variants emerged, such as mucoid, small colony and pinpoint variants, known to be highly resistant to antibiotics. The pathogenic potential of all emergent colony morphotypes-associated bacteria, distinct from the wild-morphotypes, revealed that P. aeruginosa evolved to a non-swimming phenotype. Impaired swimming motility seemed to be one of the first evolutionary steps of P. aeruginosa in CF lungs that could pave the way for further adaptation steps including biofilm formation and progress to chronic infection. Based on our findings, impaired swimming motility seemed to be a candidate to disease marker of P. aeruginosa infection development. Despite our in vitro CF model represents a step forward towards in vivo scenario simulation and provided valuable insights about the early events, more and distinct P. aeruginosa strains should be studied to strengthen our results.
Collapse
Affiliation(s)
- Ana Margarida Sousa
- CEB - Centre of Biological Engineering, LIBRO - Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Rosana Monteiro
- CEB - Centre of Biological Engineering, LIBRO - Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Maria Olívia Pereira
- CEB - Centre of Biological Engineering, LIBRO - Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.
| |
Collapse
|
128
|
Pompilio A, Geminiani C, Mantini P, Siriwardena TN, Di Bonaventura I, Reymond JL, Di Bonaventura G. Peptide dendrimers as "lead compounds" for the treatment of chronic lung infections by Pseudomonas aeruginosa in cystic fibrosis patients: in vitro and in vivo studies. Infect Drug Resist 2018; 11:1767-1782. [PMID: 30349334 PMCID: PMC6188189 DOI: 10.2147/idr.s168868] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Aim In the present work, the potential of the D-enantiomeric dendrimers dG3KL and dTNS18 was evaluated in relation to tobramycin (Tob), for the development of novel antibacterials to treat Pseudomonas aeruginosa chronic lung infections in patients with cystic fibrosis. Results The activity of dendrimers against planktonic P. aeruginosa cells was less than Tob against three of the four strains tested (median minimum inhibitory concentration [MIC] 8 vs 1 µg/mL, respectively), but 32-fold higher against the PaPh32 strain isolated at posttransplantation stage. Results from comparative minimum bactericidal concentration/MIC evaluation and time-kill assay suggested a bactericidal mechanism for all test agents. Subinhibitory concentrations of both dendrimers and Tob significantly affected biofilm formation by all strains in a dose-dependent manner, although the PaPh26 strain, isolated during the chronic stage of infection, was particularly susceptible to dendrimers. The activity of dendrimers against preformed P. aeruginosa biofilm was generally comparable to Tob, considering both dispersion and viability of biofilm. Particularly, exposure to the test agent at 10 × MIC caused significant biofilm death (>90%, even to eradication), though with strain-specific differences. Single administration of dendrimers or Tob at 10 × MIC was not toxic in Galleria mellonella wax-moth larvae over 96 hours. However, contrarily to Tob, dendrimers were not protective against systemic infection caused by P. aeruginosa in G. mellonella. Kinetics of P. aeruginosa growth in hemolymph showed that bacterial load increased over time in the presence of dendrimers. Conclusion Overall, our findings indicated that dG3KL and dTNS18 peptide dendrimers show in vitro activity comparable to Tob against both P. aeruginosa planktonic and biofilm cells at concentrations not toxic in vivo. Further studies are warranted to explore different dosages and to increase the bioavailability of the peptides to solve the lack of protective effect observed in G. mellonella larvae.
Collapse
Affiliation(s)
- Arianna Pompilio
- Department of Medical, Oral, and Biotechnological Sciences, G d'Annunzio University of Chieti-Pescara, Chieti 66100, Italy, .,Center of Excellence on Aging and Translational Medicine, G d'Annunzio University of Chieti-Pescara, Chieti, Italy,
| | - Cristina Geminiani
- Department of Medical, Oral, and Biotechnological Sciences, G d'Annunzio University of Chieti-Pescara, Chieti 66100, Italy, .,Center of Excellence on Aging and Translational Medicine, G d'Annunzio University of Chieti-Pescara, Chieti, Italy,
| | - Paolo Mantini
- Department of Medical, Oral, and Biotechnological Sciences, G d'Annunzio University of Chieti-Pescara, Chieti 66100, Italy, .,Center of Excellence on Aging and Translational Medicine, G d'Annunzio University of Chieti-Pescara, Chieti, Italy,
| | | | - Ivan Di Bonaventura
- Department of Chemistry and Biochemistry, University of Bern, Bern, Switzerland
| | - Jean Louis Reymond
- Department of Chemistry and Biochemistry, University of Bern, Bern, Switzerland
| | - Giovanni Di Bonaventura
- Department of Medical, Oral, and Biotechnological Sciences, G d'Annunzio University of Chieti-Pescara, Chieti 66100, Italy, .,Center of Excellence on Aging and Translational Medicine, G d'Annunzio University of Chieti-Pescara, Chieti, Italy,
| |
Collapse
|
129
|
Bjarnsholt T, Buhlin K, Dufrêne YF, Gomelsky M, Moroni A, Ramstedt M, Rumbaugh KP, Schulte T, Sun L, Åkerlund B, Römling U. Biofilm formation - what we can learn from recent developments. J Intern Med 2018; 284:332-345. [PMID: 29856510 PMCID: PMC6927207 DOI: 10.1111/joim.12782] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Although biofilms have been observed early in the history of microbial research, their impact has only recently been fully recognized. Biofilm infections, which contribute to up to 80% of human microbial infections, are associated with common human disorders, such as diabetes mellitus and poor dental hygiene, but also with medical implants. The associated chronic infections such as wound infections, dental caries and periodontitis significantly enhance morbidity, affect quality of life and can aid development of follow-up diseases such as cancer. Biofilm infections remain challenging to treat and antibiotic monotherapy is often insufficient, although some rediscovered traditional compounds have shown surprising efficiency. Innovative anti-biofilm strategies include application of anti-biofilm small molecules, intrinsic or external stimulation of production of reactive molecules, utilization of materials with antimicrobial properties and dispersion of biofilms by digestion of the extracellular matrix, also in combination with physical biofilm breakdown. Although basic principles of biofilm formation have been deciphered, the molecular understanding of the formation and structural organization of various types of biofilms has just begun to emerge. Basic studies of biofilm physiology have also resulted in an unexpected discovery of cyclic dinucleotide second messengers that are involved in interkingdom crosstalk via specific mammalian receptors. These findings even open up new venues for exploring novel anti-biofilm strategies.
Collapse
Affiliation(s)
- T Bjarnsholt
- Department of Immunology and Microbiology, Costerton Biofilm Centre, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Microbiology, Copenhagen University Hospital, Copenhagen, Denmark
| | - K Buhlin
- Department of Dental Medicine, Division of Oral Facial Diagnostics and Surgery, Karolinska Institutet, Huddinge, Sweden
| | - Y F Dufrêne
- Institute of Life Sciences, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - M Gomelsky
- Department of Molecular Biology, University of Wyoming, Laramie, WY, USA
| | - A Moroni
- Department of Biology and CNR-Istituto di Biofisica, Università degli Studi di Milano, Milano, Italy
| | - M Ramstedt
- Department of Chemistry, Umeå University, Umeå, Sweden
| | - K P Rumbaugh
- Departments of Surgery & Immunology & Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - T Schulte
- Department of Medicine Solna, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - L Sun
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - B Åkerlund
- Department of Medicine Huddinge, Unit of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - U Römling
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
130
|
Quinn RA, Comstock W, Zhang T, Morton JT, da Silva R, Tran A, Aksenov A, Nothias LF, Wangpraseurt D, Melnik AV, Ackermann G, Conrad D, Klapper I, Knight R, Dorrestein PC. Niche partitioning of a pathogenic microbiome driven by chemical gradients. SCIENCE ADVANCES 2018; 4:eaau1908. [PMID: 30263961 PMCID: PMC6157970 DOI: 10.1126/sciadv.aau1908] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 08/10/2018] [Indexed: 05/25/2023]
Abstract
Environmental microbial communities are stratified by chemical gradients that shape the structure and function of these systems. Similar chemical gradients exist in the human body, but how they influence these microbial systems is more poorly understood. Understanding these effects can be particularly important for dysbiotic shifts in microbiome structure that are often associated with disease. We show that pH and oxygen strongly partition the microbial community from a diseased human lung into two mutually exclusive communities of pathogens and anaerobes. Antimicrobial treatment disrupted this chemical partitioning, causing complex death, survival, and resistance outcomes that were highly dependent on the individual microorganism and on community stratification. These effects were mathematically modeled, enabling a predictive understanding of this complex polymicrobial system. Harnessing the power of these chemical gradients could be a drug-free method of shaping microbial communities in the human body from undesirable dysbiotic states.
Collapse
Affiliation(s)
- Robert A. Quinn
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California at San Diego, La Jolla, CA 92093, USA
- Center for Microbiome Innovation, University of California at San Diego, La Jolla, CA 92093, USA
| | - William Comstock
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California at San Diego, La Jolla, CA 92093, USA
| | - Tianyu Zhang
- Department of Mathematical Sciences, Montana State University, Bozeman, MT 59717, USA
| | - James T. Morton
- Department of Computer Science and Engineering, University of California at San Diego, La Jolla, CA 92093, USA
| | - Ricardo da Silva
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California at San Diego, La Jolla, CA 92093, USA
| | - Alda Tran
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California at San Diego, La Jolla, CA 92093, USA
| | - Alexander Aksenov
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California at San Diego, La Jolla, CA 92093, USA
- Center for Microbiome Innovation, University of California at San Diego, La Jolla, CA 92093, USA
| | - Louis-Felix Nothias
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California at San Diego, La Jolla, CA 92093, USA
| | - Daniel Wangpraseurt
- Department of Chemistry, University of Cambridge, Cambridge, UK
- Scripps Institution of Oceanography, University of California at San Diego, La Jolla, CA 92093, USA
| | - Alexey V. Melnik
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California at San Diego, La Jolla, CA 92093, USA
| | - Gail Ackermann
- Department of Pediatrics, University of California at San Diego, La Jolla, CA 92093, USA
| | - Douglas Conrad
- Department of Medicine, University of California at San Diego, La Jolla, CA 92093, USA
| | - Isaac Klapper
- Department of Mathematics, Temple University, Philadelphia, PA 19122, USA
| | - Rob Knight
- Center for Microbiome Innovation, University of California at San Diego, La Jolla, CA 92093, USA
- Department of Computer Science and Engineering, University of California at San Diego, La Jolla, CA 92093, USA
- Department of Pediatrics, University of California at San Diego, La Jolla, CA 92093, USA
| | - Pieter C. Dorrestein
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California at San Diego, La Jolla, CA 92093, USA
- Center for Microbiome Innovation, University of California at San Diego, La Jolla, CA 92093, USA
- Department of Pediatrics, University of California at San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
131
|
Assessment of in vivo versus in vitro biofilm formation of clinical methicillin-resistant Staphylococcus aureus isolates from endotracheal tubes. Sci Rep 2018; 8:11906. [PMID: 30093624 PMCID: PMC6085380 DOI: 10.1038/s41598-018-30494-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 07/31/2018] [Indexed: 12/31/2022] Open
Abstract
Our aim was to demonstrate that biofilm formation in a clinical strain of methicillin-resistant Staphylococcus aureus (MRSA) can be enhanced by environment exposure in an endotracheal tube (ETT) and to determine how it is affected by systemic treatment and atmospheric conditions. Second, we aimed to assess biofilm production dynamics after extubation. We prospectively analyzed 70 ETT samples obtained from pigs randomized to be untreated (controls, n = 20), or treated with vancomycin (n = 32) or linezolid (n = 18). A clinical MRSA strain (MRSA-in) was inoculated in pigs to create a pneumonia model, before treating with antibiotics. Tracheally intubated pigs with MRSA severe pneumonia, were mechanically ventilated for 69 ± 16 hours. All MRSA isolates retrieved from ETTs (ETT-MRSA) were tested for their in vitro biofilm production by microtiter plate assay. In vitro biofilm production of MRSA isolates was sequentially studied over the next 8 days post-extubation to assess biofilm capability dynamics over time. All experiments were performed under ambient air (O2) or ambient air supplemented with 5% CO2. We collected 52 ETT-MRSA isolates (placebo N = 19, linezolid N = 11, and vancomycin N = 22) that were clonally identical to the MRSA-in. Among the ETT-MRSA isolates, biofilm production more than doubled after extubation in 40% and 50% under 5% CO2 and O2, respectively. Systemic antibiotic treatment during intubation did not affect this outcome. Under both atmospheric conditions, biofilm production for MRSA-in was at least doubled for 9 ETT-MRSA isolates, and assessment of these showed that biofilm production decreased progressively over a 4-day period after extubation. In conclusion, a weak biofilm producer MRSA strain significantly enhances its biofilm production within an ETT, but it is influenced by the ETT environment rather than by the systemic treatment used during intubation or by the atmospheric conditions used for bacterial growth.
Collapse
|
132
|
Kennedy JA, Baron VO, Hammond RJH, Sloan DJ, Gillespie SH. Centrifugation and decontamination procedures selectively impair recovery of important populations in Mycobacterium smegmatis. Tuberculosis (Edinb) 2018; 112:79-82. [PMID: 30205972 DOI: 10.1016/j.tube.2018.07.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 07/28/2018] [Accepted: 07/31/2018] [Indexed: 10/28/2022]
Abstract
Diagnosis and treatment monitoring of patients with tuberculosis (TB) requires detection of all viable mycobacteria in clinical samples. Quantitation of Mycobacterium tuberculosis (Mtb) in sputum is commonly performed by culture after sample decontamination to prevent overgrowth by contaminant organisms. Exponentially growing cultures have cells that predominately lack non-polar lipid bodies whereas stationary cultures have a predominance of cells with non-polar lipid bodies. This may reflect rapidly growing 'active' and non-replicating 'persister' sub-populations respectively in sputum from TB patients. We investigated the effect of decontamination on culture-based quantitation of exponential and stationary phase cultures of Mycobacterium smegmatis in an artificial sputum model. Exponentially growing populations were between 89 and 50 times more susceptible to decontamination than stationary phase cultures when quantified by most probable number and colony forming units. These findings suggest that decontamination selectively eliminates the 'active' population. This may impair diagnostic sensitivity, treatment monitoring, and compromise clinical trials designed to identify new antibiotic combinations with activity against all mycobacterial cell states.
Collapse
Affiliation(s)
- John A Kennedy
- School of Medicine, University of St Andrews, Fife, United Kingdom
| | - Vincent O Baron
- School of Medicine, University of St Andrews, Fife, United Kingdom
| | | | - Derek J Sloan
- School of Medicine, University of St Andrews, Fife, United Kingdom
| | | |
Collapse
|
133
|
Price EP, Viberg LT, Kidd TJ, Bell SC, Currie BJ, Sarovich DS. Transcriptomic analysis of longitudinal Burkholderia pseudomallei infecting the cystic fibrosis lung. Microb Genom 2018; 4. [PMID: 29989529 PMCID: PMC6159556 DOI: 10.1099/mgen.0.000194] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The melioidosis bacterium, Burkholderia pseudomallei, is increasingly being recognised as a pathogen in patients with cystic fibrosis (CF). We have recently catalogued genome-wide variation of paired, isogenic B. pseudomallei isolates from seven Australasian CF cases, which were collected between 4 and 55 months apart. Here, we extend this investigation by documenting the transcriptomic changes in B. pseudomallei in five cases. Following growth in an artificial CF sputum medium, four of the five paired isolates exhibited significant differential gene expression (DE) that affected between 32 and 792 genes. The greatest number of DE events was observed between the strains from patient CF9, consistent with the hypermutator status of the latter strain, which is deficient in the DNA mismatch repair protein MutS. Two patient isolates harboured duplications that concomitantly increased expression of the β-lactamase-encoding gene penA, and a 35 kb deletion in another abolished expression of 29 genes. Convergent expression profiles in the chronically-adapted isolates identified two significantly downregulated and 17 significantly upregulated loci, including the resistance-nodulation-division (RND) efflux pump BpeEF-OprC, the quorum-sensing hhqABCDE operon, and a cyanide- and pyocyanin-insensitive cytochrome bd quinol oxidase. These convergent pathoadaptations lead to increased expression of pathways that may suppress competing bacterial and fungal pathogens, and that enhance survival in oxygen-restricted environments, the latter of which may render conventional antibiotics less effective in vivo. Treating chronically adapted B. pseudomallei infections with antibiotics designed to target anaerobic infections, such as the nitroimidazole class of antibiotics, may significantly improve pathogen eradication attempts by exploiting this Achilles heel.
Collapse
Affiliation(s)
- Erin P Price
- 1Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, Sippy Downs, QLD, Australia.,2Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| | - Linda T Viberg
- 2Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| | - Timothy J Kidd
- 3Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.,4School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Scott C Bell
- 3Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.,5QIMR Berghofer Medical Research Institute, Herston, QLD, Australia.,6Department of Thoracic Medicine, The Prince Charles Hospital, Chermside, QLD, Australia
| | - Bart J Currie
- 2Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia.,7Department of Infectious Diseases and Northern Territory Medical Program, Royal Darwin Hospital, Darwin, Northern Territory, Australia
| | - Derek S Sarovich
- 1Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, Sippy Downs, QLD, Australia.,2Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| |
Collapse
|
134
|
Bricio-Moreno L, Sheridan VH, Goodhead I, Armstrong S, Wong JKL, Waters EM, Sarsby J, Panagiotou S, Dunn J, Chakraborty A, Fang Y, Griswold KE, Winstanley C, Fothergill JL, Kadioglu A, Neill DR. Evolutionary trade-offs associated with loss of PmrB function in host-adapted Pseudomonas aeruginosa. Nat Commun 2018; 9:2635. [PMID: 29980663 PMCID: PMC6035264 DOI: 10.1038/s41467-018-04996-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 06/06/2018] [Indexed: 12/22/2022] Open
Abstract
Pseudomonas aeruginosa colonises the upper airway of cystic fibrosis (CF) patients, providing a reservoir of host-adapted genotypes that subsequently establish chronic lung infection. We previously experimentally-evolved P. aeruginosa in a murine model of respiratory tract infection and observed early-acquired mutations in pmrB, encoding the sensor kinase of a two-component system that promoted establishment and persistence of infection. Here, using proteomics, we show downregulation of proteins involved in LPS biosynthesis, antimicrobial resistance and phenazine production in pmrB mutants, and upregulation of proteins involved in adherence, lysozyme resistance and inhibition of the chloride ion channel CFTR, relative to wild-type strain LESB65. Accordingly, pmrB mutants are susceptible to antibiotic treatment but show enhanced adherence to airway epithelial cells, resistance to lysozyme treatment, and downregulate host CFTR expression. We propose that P. aeruginosa pmrB mutations in CF patients are subject to an evolutionary trade-off, leading to enhanced colonisation potential, CFTR inhibition, and resistance to host defences, but also to increased susceptibility to antibiotics.
Collapse
Affiliation(s)
- Laura Bricio-Moreno
- Institute of Infection and Global Health, University of Liverpool, Liverpool, L69 7BE, UK
| | - Victoria H Sheridan
- Institute of Infection and Global Health, University of Liverpool, Liverpool, L69 7BE, UK
| | - Ian Goodhead
- School of Environment and Life Sciences, University of Salford, Salford, M5 4WT, UK
| | - Stuart Armstrong
- Institute of Infection and Global Health, University of Liverpool, Liverpool, L69 7BE, UK
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, University of Liverpool, Liverpool, L69 3GL, UK
| | - Janet K L Wong
- Institute of Infection and Global Health, University of Liverpool, Liverpool, L69 7BE, UK
| | - Elaine M Waters
- Institute of Infection and Global Health, University of Liverpool, Liverpool, L69 7BE, UK
- Department of Microbiology, School of Natural Science, National University of Ireland, Galway, H91 TK33, Ireland
| | - Joscelyn Sarsby
- Institute of Integrative Biology, University of Liverpool, Liverpool, L69 7ZB, UK
| | - Stavros Panagiotou
- Institute of Infection and Global Health, University of Liverpool, Liverpool, L69 7BE, UK
| | - James Dunn
- Institute of Infection and Global Health, University of Liverpool, Liverpool, L69 7BE, UK
| | - Adrita Chakraborty
- Institute of Infection and Global Health, University of Liverpool, Liverpool, L69 7BE, UK
| | - Yongliang Fang
- Thayer School of Engineering, Dartmouth, Hanover, NH, 03755, USA
| | - Karl E Griswold
- Thayer School of Engineering, Dartmouth, Hanover, NH, 03755, USA
| | - Craig Winstanley
- Institute of Infection and Global Health, University of Liverpool, Liverpool, L69 7BE, UK
| | - Joanne L Fothergill
- Institute of Infection and Global Health, University of Liverpool, Liverpool, L69 7BE, UK.
| | - Aras Kadioglu
- Institute of Infection and Global Health, University of Liverpool, Liverpool, L69 7BE, UK
| | - Daniel R Neill
- Institute of Infection and Global Health, University of Liverpool, Liverpool, L69 7BE, UK.
| |
Collapse
|
135
|
Müller L, Murgia X, Siebenbürger L, Börger C, Schwarzkopf K, Sewald K, Häussler S, Braun A, Lehr CM, Hittinger M, Wronski S. Human airway mucus alters susceptibility of Pseudomonas aeruginosa biofilms to tobramycin, but not colistin. J Antimicrob Chemother 2018; 73:2762-2769. [DOI: 10.1093/jac/dky241] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 05/26/2018] [Indexed: 02/06/2023] Open
Affiliation(s)
- Laura Müller
- Fraunhofer Institute for Toxicology and Experimental Medicine (Fraunhofer ITEM), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research (DZL), Member of the REBIRTH Cluster of Excellence, Nikolai-Fuchs-Straße 1, Hannover, Germany
| | - Xabier Murgia
- Helmholtz Institute for Pharmaceutical Research (HIPS), Helmholtz Centre for Infection Research, Universitätscampus E8.1, Saarbrücken, Germany
- Korea Institute of Science and Technology, KIST Europe, Campus E7.1, Saarbrücken, Germany
| | | | | | - Konrad Schwarzkopf
- Department of Anaesthesia and Intensive Care, Klinikum Saarbrücken, Winterberg 1, Saarbrücken, Germany
| | - Katherina Sewald
- Fraunhofer Institute for Toxicology and Experimental Medicine (Fraunhofer ITEM), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research (DZL), Member of the REBIRTH Cluster of Excellence, Nikolai-Fuchs-Straße 1, Hannover, Germany
| | - Susanne Häussler
- Helmholtz Institute for Infection Research, Inhoffenstraße 7, Braunschweig, Germany
- TWINCORE, Centre for Experimental and Clinical Infection Research, Feodor-Lynen-Straße 7, Hannover, Germany
| | - Armin Braun
- Fraunhofer Institute for Toxicology and Experimental Medicine (Fraunhofer ITEM), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research (DZL), Member of the REBIRTH Cluster of Excellence, Nikolai-Fuchs-Straße 1, Hannover, Germany
| | - Claus-Michael Lehr
- Helmholtz Institute for Pharmaceutical Research (HIPS), Helmholtz Centre for Infection Research, Universitätscampus E8.1, Saarbrücken, Germany
- PharmBioTec GmbH, Science Park 1, Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Campus, Saarbrücken, Germany
| | | | - Sabine Wronski
- Fraunhofer Institute for Toxicology and Experimental Medicine (Fraunhofer ITEM), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research (DZL), Member of the REBIRTH Cluster of Excellence, Nikolai-Fuchs-Straße 1, Hannover, Germany
| |
Collapse
|
136
|
Gagné-Thivierge C, Barbeau J, Levesque RC, Charette SJ. A new approach to study attached biofilms and floating communities from Pseudomonas aeruginosa strains of various origins reveals diverse effects of divalent ions. FEMS Microbiol Lett 2018; 365:5044545. [DOI: 10.1093/femsle/fny155] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 06/24/2018] [Indexed: 12/31/2022] Open
Affiliation(s)
- Cynthia Gagné-Thivierge
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Quebec City, Quebec G1V 0A6, Canada
- Département de biochimie, de microbiologie et de bio-informatique, Faculté des Sciences et de Génie, Université Laval, Quebec City, Quebec G1V 0A6, Canada
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Quebec City, Quebec G1V 4G5, Canada
| | - Jean Barbeau
- Faculté de Médecine Dentaire, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Roger C Levesque
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Quebec City, Quebec G1V 0A6, Canada
- Département de microbiologie, infectiologie et immunologie, Faculté de Médecine, Université Laval, Quebec city, Quebec G1V 0A6, Canada
| | - Steve J Charette
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Quebec City, Quebec G1V 0A6, Canada
- Département de biochimie, de microbiologie et de bio-informatique, Faculté des Sciences et de Génie, Université Laval, Quebec City, Quebec G1V 0A6, Canada
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Quebec City, Quebec G1V 4G5, Canada
| |
Collapse
|
137
|
A phosphatidic acid-binding protein is important for lipid homeostasis and adaptation to anaerobic biofilm conditions in Pseudomonas aeruginosa. Biochem J 2018; 475:1885-1907. [DOI: 10.1042/bcj20180257] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 04/27/2018] [Accepted: 05/01/2018] [Indexed: 01/22/2023]
Abstract
A quantitative Pseudomonas aeruginosa proteomics approach revealed increased abundance of the so-far uncharacterized protein PA3911 in anaerobic biofilms grown under conditions of the cystic fibrosis lung. Physiological relevance of ORF PA3911 was demonstrated, inter alia, using phenotype microarray experiments. The mutant strain showed increased susceptibility in the presence of antimicrobials (minocycline, nafcillin, oxacillin, chloramphenicol and thiamphenicol), enhanced twitching motility and significantly impaired biofilm formation. PA3911 is a soluble, cytoplasmic protein in P. aeruginosa. In protein–lipid overlay experiments, purified PA3911 bound specifically to phosphatidic acid (PA), the central hub of phospholipid metabolism. Structure-guided site-directed mutagenesis was used to explore the proposed ligand-binding cavity of PA3911. Protein variants of Leu56, Leu58, Val69 and Leu114 were shown to impair PA interaction. A comparative shotgun lipidomics approach demonstrated a multifaceted response of P. aeruginosa to anaerobic conditions at the lipid head group and fatty acid level. Lipid homeostasis in the PA3911 mutant strain was imbalanced with respect to lysophosphatidylcholine, phosphatidylcholine and diacylglycerol under anaerobic and/or aerobic conditions. The impact of the newly identified PA-binding protein on lipid homeostasis and the related macroscopic phenotypes of P. aeruginosa are discussed.
Collapse
|
138
|
Daly SM, Sturge CR, Marshall-Batty KR, Felder-Scott CF, Jain R, Geller BL, Greenberg DE. Antisense Inhibitors Retain Activity in Pulmonary Models of Burkholderia Infection. ACS Infect Dis 2018; 4:806-814. [PMID: 29461800 DOI: 10.1021/acsinfecdis.7b00235] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The Burkholderia cepacia complex is a group of Gram-negative bacteria that are opportunistic pathogens in immunocompromised individuals, such as those with cystic fibrosis (CF) or chronic granulomatous disease (CGD). Burkholderia are intrinsically resistant to many antibiotics and the lack of antibiotic development necessitates novel therapeutics. Peptide-conjugated phosphorodiamidate morpholino oligomers are antisense molecules that inhibit bacterial mRNA translation. Targeting of PPMOs to the gene acpP, which is essential for membrane synthesis, lead to defects in the membrane and ultimately bactericidal activity. Exploration of additional PPMO sequences identified the ATG and Shine-Dalgarno sites as the most efficacious for targeting acpP. The CF lung is a complex microenvironment, but PPMO inhibition was still efficacious in an artificial model of CF sputum. PPMOs had low toxicity in human CF cells at doses that were antibacterial. PPMOs also reduced the bacterial burden in the lungs of immunocompromised CyBB mice, a model of CGD. Finally, the use of multiple PPMOs was efficacious in inhibiting the growth of both Burkholderia and Pseudomonas in an in vitro model of coinfection. Due to the intrinsic resistance of Burkholderia to traditional antibiotics, PPMOs represent a novel and viable approach to the treatment of Burkholderia infections.
Collapse
Affiliation(s)
- Seth M. Daly
- Department of Internal Medicine, UT Southwestern, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Carolyn R. Sturge
- Department of Internal Medicine, UT Southwestern, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Kimberly R. Marshall-Batty
- Department of Internal Medicine, UT Southwestern, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Christina F. Felder-Scott
- Department of Internal Medicine, UT Southwestern, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Raksha Jain
- Department of Internal Medicine, UT Southwestern, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
- Department of Microbiology, UT Southwestern, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Bruce L. Geller
- Department of Microbiology, Oregon State University, 226 Nash Hall, Corvallis, Oregon 97331, United States
| | - David E. Greenberg
- Department of Internal Medicine, UT Southwestern, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
- Department of Microbiology, UT Southwestern, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| |
Collapse
|
139
|
Schneider-Futschik EK, Paulin OKA, Hoyer D, Roberts KD, Ziogas J, Baker MA, Karas J, Li J, Velkov T. Sputum Active Polymyxin Lipopeptides: Activity against Cystic Fibrosis Pseudomonas aeruginosa Isolates and Their Interactions with Sputum Biomolecules. ACS Infect Dis 2018; 4:646-655. [PMID: 29566483 PMCID: PMC5952261 DOI: 10.1021/acsinfecdis.7b00238] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
![]()
The
mucoid biofilm mode of growth of Pseudomonas aeruginosa (P. aeruginosa) in the lungs of cystic fibrosis
patients makes eradication of infections with antibiotic therapy very
difficult. The lipopeptide antibiotics polymyxin B and colistin are
currently the last-resort therapies for infections caused by multidrug-resistant P. aeruginosa. In the present study, we investigated
the antibacterial activity of a series of polymyxin lipopeptides (polymyxin
B, colistin, FADDI-003, octapeptin A3, and polymyxin A2) against a panel of polymyxin-susceptible and polymyxin-resistant P. aeruginosa cystic fibrosis isolates grown under
planktonic or biofilm conditions in artificial sputum and their interactions
with sputum component biomolecules. In sputum media under planktonic
conditions, the lipopeptides FADDI-003 and octapeptin A3 displayed very promising activity against the polymyxin-resistant
isolate FADDI-PA066 (polymyxin B minimum inhibitory concentration
(MIC) = 32 mg/L), while retaining their activity against the polymyxin-sensitive
strains FADDI-PA021 (polymyxin B MIC = 1 mg/L) and FADDI-PA020 (polymyxin
B MIC = 2 mg/L). Polymyxin A2 was only effective against
the polymyxin-sensitive isolates. However, under biofilm growth conditions,
the hydrophobic lipopeptide FADDI-003 was inactive compared to the
more hydrophilic lipopeptides, octapeptin A3, polymyxin
A2, polymyxin B, and colistin. Transmission electron micrographs
revealed octapeptin A3 caused reduction in the cell numbers
in biofilm as well as biofilm disruption/“antibiofilm”
activity. We therefore assessed the interactions of the lipopeptides
with the component sputum biomolecules, mucin, deoxyribonucleic acid
(DNA), surfactant, F-actin, lipopolysaccharide, and phospholipids.
We observed the general trend that sputum biomolecules reduce lipopeptide
antibacterial activity. Collectively, our data suggests that, in the
airways, lipopeptide binding to component sputum biomolecules may
reduce antibacterial efficacy and is dependent on the physicochemical
properties of the lipopeptide.
Collapse
Affiliation(s)
- Elena K. Schneider-Futschik
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Department of Pharmacology and Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Olivia K. A. Paulin
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Daniel Hoyer
- Department of Pharmacology and Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria 3010, Australia
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, Victoria 3052, Australia
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Kade D. Roberts
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - James Ziogas
- Department of Pharmacology and Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Mark A. Baker
- Priority Research Centre in Reproductive Science, School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales 2308, Australia
| | - John Karas
- Department of Pharmacology and Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Jian Li
- Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, Victoria 3800, Australia
| | - Tony Velkov
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Department of Pharmacology and Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
140
|
Gill EE, Chan LS, Winsor GL, Dobson N, Lo R, Ho Sui SJ, Dhillon BK, Taylor PK, Shrestha R, Spencer C, Hancock REW, Unrau PJ, Brinkman FSL. High-throughput detection of RNA processing in bacteria. BMC Genomics 2018; 19:223. [PMID: 29587634 PMCID: PMC5870498 DOI: 10.1186/s12864-018-4538-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 02/12/2018] [Indexed: 01/19/2023] Open
Abstract
Background Understanding the RNA processing of an organism’s transcriptome is an essential but challenging step in understanding its biology. Here we investigate with unprecedented detail the transcriptome of Pseudomonas aeruginosa PAO1, a medically important and innately multi-drug resistant bacterium. We systematically mapped RNA cleavage and dephosphorylation sites that result in 5′-monophosphate terminated RNA (pRNA) using monophosphate RNA-Seq (pRNA-Seq). Transcriptional start sites (TSS) were also mapped using differential RNA-Seq (dRNA-Seq) and both datasets were compared to conventional RNA-Seq performed in a variety of growth conditions. Results The pRNA-Seq library revealed known tRNA, rRNA and transfer-messenger RNA (tmRNA) processing sites, together with previously uncharacterized RNA cleavage events that were found disproportionately near the 5′ ends of transcripts associated with basic bacterial functions such as oxidative phosphorylation and purine metabolism. The majority (97%) of the processed mRNAs were cleaved at precise codon positions within defined sequence motifs indicative of distinct endonucleolytic activities. The most abundant of these motifs corresponded closely to an E. coli RNase E site previously established in vitro. Using the dRNA-Seq library, we performed an operon analysis and predicted 3159 potential TSS. A correlation analysis uncovered 105 antiparallel pairs of TSS that were separated by 18 bp from each other and were centered on single palindromic TAT(A/T)ATA motifs (likely − 10 promoter elements), suggesting that, consistent with previous in vitro experimentation, these sites can initiate transcription bi-directionally and may thus provide a novel form of transcriptional regulation. TSS and RNA-Seq analysis allowed us to confirm expression of small non-coding RNAs (ncRNAs), many of which are differentially expressed in swarming and biofilm formation conditions. Conclusions This study uses pRNA-Seq, a method that provides a genome-wide survey of RNA processing, to study the bacterium Pseudomonas aeruginosa and discover extensive transcript processing not previously appreciated. We have also gained novel insight into RNA maturation and turnover as well as a potential novel form of transcription regulation. NOTE: All sequence data has been submitted to the NCBI sequence read archive. Accession numbers are as follows: [NCBI sequence read archive: SRX156386, SRX157659, SRX157660, SRX157661, SRX157683 and SRX158075]. The sequence data is viewable using Jbrowse on www.pseudomonas.com. Electronic supplementary material The online version of this article (10.1186/s12864-018-4538-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Erin E Gill
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - Luisa S Chan
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - Geoffrey L Winsor
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - Neil Dobson
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - Raymond Lo
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - Shannan J Ho Sui
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - Bhavjinder K Dhillon
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - Patrick K Taylor
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Raunak Shrestha
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - Cory Spencer
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - Robert E W Hancock
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Peter J Unrau
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada.
| | - Fiona S L Brinkman
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada.
| |
Collapse
|
141
|
Bahamondez-Canas TF, Zhang H, Tewes F, Leal J, Smyth HDC. PEGylation of Tobramycin Improves Mucus Penetration and Antimicrobial Activity against Pseudomonas aeruginosa Biofilms in Vitro. Mol Pharm 2018. [PMID: 29514003 DOI: 10.1021/acs.molpharmaceut.8b00011] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Pseudomonas aeruginosa is the predominant pathogen in the persistent lung infections of cystic fibrosis (CF) patients among other diseases. One of the mechanisms of resistance of P. aeruginosa infections is the formation and presence of biofilms. Previously, we demonstrated that PEGylated-tobramycin (Tob-PEG) had superior antimicrobial activity against P. aeruginosa biofilms compared to tobramycin (Tob). The goal of this study was to optimize the method of PEGylation of Tob and assess its activity in an in vitro CF-like mucus barrier biofilm model. Tob was PEGylated using three separate chemical conjugation methods and analyzed by 1H NMR. A comparison of the Tob-PEG products from the different conjugation methods showed significant differences in the reduction of biofilm proliferation after 24 h of treatment. In the CF-like mucus barrier model, Tob-PEG was significantly better than Tob in reducing P. aeruginosa proliferation after only 5 h of treatment ( p < 0.01). Finally, Tob-PEG caused a reduction in the number of surviving P. aeruginosa biofilm colonies higher than that of Tob ( p < 0.0001). We demonstrate the significantly improved antimicrobial activity of Tob-PEG against P. aeruginosa biofilms compared to Tob using two PEGylation methods. Tob-PEG had better in vitro activity compared to that of Tob against P. aeruginosa biofilms growing in a CF-like mucus barrier model.
Collapse
Affiliation(s)
- Tania F Bahamondez-Canas
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy , The University of Texas at Austin , Austin , Texas 78712 , United States
| | - Hairui Zhang
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy , The University of Texas at Austin , Austin , Texas 78712 , United States
| | - Frederic Tewes
- INSERM, U1070, UFR de Médecine Pharmacie , Université de Poitiers , 86073 Poitiers Cedex 9 , France
| | - Jasmim Leal
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy , The University of Texas at Austin , Austin , Texas 78712 , United States
| | - Hugh D C Smyth
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy , The University of Texas at Austin , Austin , Texas 78712 , United States.,Center for Infectious Disease , The University of Texas at Austin , Austin , Texas 78712 , United States
| |
Collapse
|
142
|
Antimicrobial Activity of Ibuprofen against Cystic Fibrosis-Associated Gram-Negative Pathogens. Antimicrob Agents Chemother 2018; 62:AAC.01574-17. [PMID: 29311081 PMCID: PMC5826130 DOI: 10.1128/aac.01574-17] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 12/14/2017] [Indexed: 12/13/2022] Open
Abstract
Clinical trials have demonstrated the benefits of ibuprofen therapy in cystic fibrosis (CF) patients, an effect that is currently attributed to ibuprofen's anti-inflammatory properties. Yet, a few previous reports demonstrated an antimicrobial activity of ibuprofen as well, although none investigated its direct effects on the pathogens found in the CF lung, which is the focus of this work. Determination of ibuprofen's in vitro antimicrobial activity against Pseudomonas aeruginosa and Burkholderia species strains through measurements of the endpoint number of CFU and growth kinetics showed that ibuprofen reduced the growth rate and bacterial burden of the tested strains in a dose-dependent fashion. In an in vitroPseudomonas biofilm model, a reduction in the rate of biomass accumulation over 8 h of growth with ibuprofen treatment was observed. Next, an acute Pseudomonas pneumonia model was used to test this antimicrobial activity after the oral delivery of ibuprofen. Following intranasal inoculation, ibuprofen-treated mice exhibited lower CFU counts and improved survival compared with the control animals. Preliminary biodistribution studies performed after the delivery of ibuprofen to mice by aerosol demonstrated a rapid accumulation of ibuprofen in serum and minimum retention in lung tissue and bronchoalveolar lavage fluid. Therefore, ibuprofen-encapsulated polymeric nanoparticles (Ibu-NPs) were formulated to improve the pharmacokinetic profile. Ibu-NPs formulated for aerosol delivery inhibited the growth of P. aeruginosa in vitro and may provide a convenient dosing method. These results provide an additional explanation for the previously observed therapeutic effects of ibuprofen in CF patients and further strengthen the argument for its use by these patients.
Collapse
|
143
|
Abstract
Pseudomonas aeruginosa, a Gram-negative bacterium, is characterized by its versatility that enables persistent survival under adverse conditions. It can grow on diverse energy sources and readily acquire resistance to antimicrobial agents. As an opportunistic human pathogen, it also causes chronic infections inside the anaerobic mucus airways of cystic fibrosis patients. As a strict respirer, P. aeruginosa can grow by anaerobic nitrate ( [Formula: see text] ) respiration. Nitric oxide (NO) produced as an intermediate during anaerobic respiration exerts many important effects on the biological characteristics of P. aeruginosa. This review provides information regarding (i) how P. aeruginosa grows by anaerobic respiration, (ii) mechanisms by which NO is produced under such growth, and (iii) bacterial adaptation to NO. We also review the clinical relevance of NO in the fitness of P. aeruginosa and the use of NO as a potential therapeutic for treating P. aeruginosa infection.
Collapse
|
144
|
Falcone M, Ferrara S, Rossi E, Johansen HK, Molin S, Bertoni G. The Small RNA ErsA of Pseudomonas aeruginosa Contributes to Biofilm Development and Motility through Post-transcriptional Modulation of AmrZ. Front Microbiol 2018; 9:238. [PMID: 29497413 PMCID: PMC5819304 DOI: 10.3389/fmicb.2018.00238] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 01/31/2018] [Indexed: 11/13/2022] Open
Abstract
The small RNA ErsA of Pseudomonas aeruginosa was previously suggested to be involved in biofilm formation via negative post-transcriptional regulation of the algC gene that encodes the virulence-associated enzyme AlgC, which provides sugar precursors for the synthesis of several polysaccharides. In this study, we show that a knock-out ersA mutant strain forms a flat and uniform biofilm, not characterized by mushroom-multicellular structures typical of a mature biofilm. Conversely, the knock-out mutant strain showed enhanced swarming and twitching motilities. To assess the influence of ErsA on the P. aeruginosa transcriptome, we performed RNA-seq experiments comparing the knock-out mutant with the wild-type. More than 160 genes were found differentially expressed in the knock-out mutant. Parts of these genes, important for biofilm formation and motility regulation, are known to belong also to the AmrZ transcriptional regulator regulon. Here, we show that ErsA binds in vitro and positively regulates amrZ mRNA at post-transcriptional level in vivo suggesting an interesting contribution of the ErsA-amrZ mRNA interaction in biofilm development at several regulatory levels.
Collapse
Affiliation(s)
- Marilena Falcone
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milan, Italy
| | - Silvia Ferrara
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milan, Italy
| | - Elio Rossi
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
| | - Helle K Johansen
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Søren Molin
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Giovanni Bertoni
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
145
|
Abstract
Microbial biofilms can colonize medical devices and human tissues, and their role in microbial pathogenesis is now well established. Not only are biofilms ubiquitous in natural and human-made environments, but they are also estimated to be associated with approximately two-thirds of nosocomial infections. This multicellular aggregated form of microbial growth confers a remarkable resistance to killing by antimicrobials and host defenses, leading biofilms to cause a wide range of subacute or chronic infections that are difficult to eradicate. We have gained tremendous knowledge on the molecular, genetic, microbiological, and biophysical processes involved in biofilm formation. These insights now shape our understanding, diagnosis, and management of many infectious diseases and direct the development of novel antimicrobial therapies that target biofilms. Bacterial and fungal biofilms play an important role in a range of diseases in pulmonary and critical care medicine, most importantly catheter-associated infections, ventilator-associated pneumonia, chronic Pseudomonas aeruginosa infections in cystic fibrosis lung disease, and Aspergillus fumigatus pulmonary infections.
Collapse
|
146
|
Ohneck EJ, Arivett BA, Fiester SE, Wood CR, Metz ML, Simeone GM, Actis LA. Mucin acts as a nutrient source and a signal for the differential expression of genes coding for cellular processes and virulence factors in Acinetobacter baumannii. PLoS One 2018; 13:e0190599. [PMID: 29309434 PMCID: PMC5757984 DOI: 10.1371/journal.pone.0190599] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 12/18/2017] [Indexed: 12/27/2022] Open
Abstract
The capacity of Acinetobacter baumannii to persist and cause infections depends on its interaction with abiotic and biotic surfaces, including those found on medical devices and host mucosal surfaces. However, the extracellular stimuli affecting these interactions are poorly understood. Based on our previous observations, we hypothesized that mucin, a glycoprotein secreted by lung epithelial cells, particularly during respiratory infections, significantly alters A. baumannii's physiology and its interaction with the surrounding environment. Biofilm, virulence and growth assays showed that mucin enhances the interaction of A. baumannii ATCC 19606T with abiotic and biotic surfaces and its cytolytic activity against epithelial cells while serving as a nutrient source. The global effect of mucin on the physiology and virulence of this pathogen is supported by RNA-Seq data showing that its presence in a low nutrient medium results in the differential transcription of 427 predicted protein-coding genes. The reduced expression of ion acquisition genes and the increased transcription of genes coding for energy production together with the detection of mucin degradation indicate that this host glycoprotein is a nutrient source. The increased expression of genes coding for adherence and biofilm biogenesis on abiotic and biotic surfaces, the degradation of phenylacetic acid and the production of an active type VI secretion system further supports the role mucin plays in virulence. Taken together, our observations indicate that A. baumannii recognizes mucin as an environmental signal, which triggers a response cascade that allows this pathogen to acquire critical nutrients and promotes host-pathogen interactions that play a role in the pathogenesis of bacterial infections.
Collapse
Affiliation(s)
- Emily J. Ohneck
- Department of Microbiology, Miami University, Oxford, OH, United States of America
| | - Brock A. Arivett
- Department of Microbiology, Miami University, Oxford, OH, United States of America
| | - Steven E. Fiester
- Department of Microbiology, Miami University, Oxford, OH, United States of America
| | - Cecily R. Wood
- Department of Microbiology, Miami University, Oxford, OH, United States of America
| | - Maeva L. Metz
- Department of Microbiology, Miami University, Oxford, OH, United States of America
| | - Gabriella M. Simeone
- Department of Microbiology, Miami University, Oxford, OH, United States of America
| | - Luis A. Actis
- Department of Microbiology, Miami University, Oxford, OH, United States of America
| |
Collapse
|
147
|
Lawal O, Muhamadali H, Ahmed WM, White IR, Nijsen TME, Goodacre R, Fowler SJ. Headspace volatile organic compounds from bacteria implicated in ventilator-associated pneumonia analysed by TD-GC/MS. J Breath Res 2018; 12:026002. [PMID: 28947683 DOI: 10.1088/1752-7163/aa8efc] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Ventilator-associated pneumonia (VAP) is a healthcare-acquired infection arising from the invasion of the lower respiratory tract by opportunistic pathogens in ventilated patients. The current method of diagnosis requires the culture of an airway sample such as bronchoalveolar lavage, which is invasive to obtain and may take up to seven days to identify a causal pathogen, or indeed rule out infection. While awaiting results, patients are administered empirical antibiotics; risks of this approach include lack of effect on the causal pathogen, contribution to the development of antibiotic resistance and downstream effects such as increased length of intensive care stay, cost, morbidity and mortality. Specific biomarkers which could identify causal pathogens in a timely manner are needed as they would allow judicious use of the most appropriate antimicrobial therapy. Volatile organic compound (VOC) analysis in exhaled breath is proposed as an alternative due to its non-invasive nature and its potential to provide rapid diagnosis at the patient's bedside. VOCs in exhaled breath originate from exogenous, endogenous, as well as microbial sources. To identify potential markers, VAP-associated pathogens Escherichia coli, Klebsiella pneumoniae, Pseudomonas aeruginosa, and Staphylococcus aureus were cultured in both artificial sputum medium and nutrient broth, and their headspaces were sampled and analysed for VOCs. Previously reported volatile markers were identified in this study, including indole and 1-undecene, alongside compounds that are novel to this investigation, cyclopentanone and 1-hexanol. We further investigated media components (substrates) to identify those that are essential for indole and cyclopentanone production, with potential implications for understanding microbial metabolism in the lung.
Collapse
Affiliation(s)
- Oluwasola Lawal
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
- Philips Research, Royal Philips B.V., Eindhoven, The Netherlands
- School of Chemistry, Manchester Institute of Biotechnology, The University of Manchester, Manchester, United Kingdom
| | - Howbeer Muhamadali
- School of Chemistry, Manchester Institute of Biotechnology, The University of Manchester, Manchester, United Kingdom
| | - Waqar M Ahmed
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Iain R White
- School of Chemistry, Manchester Institute of Biotechnology, The University of Manchester, Manchester, United Kingdom
| | | | - Royston Goodacre
- School of Chemistry, Manchester Institute of Biotechnology, The University of Manchester, Manchester, United Kingdom
| | - Stephen J Fowler
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
- Manchester Academic Health Science Centre, The University of Manchester and University Hospital of South Manchester NHS Foundation Trust, Manchester, United Kingdom
| |
Collapse
|
148
|
SLC6A14 Is a Genetic Modifier of Cystic Fibrosis That Regulates Pseudomonas aeruginosa Attachment to Human Bronchial Epithelial Cells. mBio 2017; 8:mBio.02073-17. [PMID: 29259090 PMCID: PMC5736915 DOI: 10.1128/mbio.02073-17] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Cystic fibrosis (CF) is caused by mutations in the CFTR gene and is associated with progressive and ultimately fatal infectious lung disease. There can be considerable variability in disease severity among individuals with the same CFTR mutations, and recent genome-wide association studies have identified secondary genetic factors that contribute to this. One of these modifier genes is SLC6A14, which encodes an amino acid transporter. Importantly, variants of this gene have been associated with age at first acquisition of Pseudomonas aeruginosa In this study, we aimed to determine the function of SLC6A14 in airway epithelia and how it might affect colonization by P. aeruginosa We show that SLC6A14 is expressed in respiratory epithelial cells and transports l-arginine out of the airway surface liquid (ASL). Exposure of airway epithelia to flagellin from P. aeruginosa led to upregulation of SLC6A14 expression and increased SLC6A14-dependent uptake of l-arginine from the ASL. In support of the hypothesis that l-arginine affects P. aeruginosa attachment, we showed that l-arginine supplementation promoted P. aeruginosa attachment to an abiotic surface in a dose-dependent manner. In a coculture model, we found that inhibition of SLC6A14-dependent l-arginine transport enhanced P. aeruginosa attachment. In Slc6a14-/y (knockout) mice, P. aeruginosa attachment to lung tissue was also significantly enhanced. Together, these findings suggest that SLC6A14 activity plays a role in the modification of the initial stages of airway infection by altering the level of l-arginine in the ASL, which in turn affects the attachment of P. aeruginosaIMPORTANCE CF patients with shared CFTR gene mutations show significant variability in their clinical presentation of infectious lung disease. Genome-wide association studies have been used to identify secondary genetic factors that may explain the variable susceptibility to infection by opportunistic pathogens, including P. aeruginosa, the leading cause of pathogen-induced lung damage in nonpediatric CF patients. Once identified and characterized, these secondary genetic modifiers may allow for the development of personalized medicine for patients and ultimately the extension of life. In this study, we interrogated the biological role of one of these modifiers, SLC6A14, and showed that it contributes to host defense by depleting extracellular arginine (an attachment-promoting metabolite for P. aeruginosa) from the airway surface liquid.
Collapse
|
149
|
Bourdillon KA, Delury CP, Cullen BM. Biofilms and delayed healing - an in vitro evaluation of silver- and iodine-containing dressings and their effect on bacterial and human cells. Int Wound J 2017; 14:1066-1075. [PMID: 28503756 PMCID: PMC7949739 DOI: 10.1111/iwj.12761] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 03/30/2017] [Accepted: 03/31/2017] [Indexed: 01/18/2023] Open
Abstract
This study investigated whether there are differences in the ability of wound dressings to modulate certain factors known to affect wound healing. A selection of antimicrobial dressings (AQUACEL® Ag Extra™ , AQUACEL® Ag+ Extra™ , IODOFLEX™ , ACTICOAT™ 7 and PROMOGRAN PRISMA™ matrix) were tested for their effect on both bacterial bioburden and human dermal fibroblasts. Some dressings underwent further evaluation for activity against Pseudomonas aeruginosa biofilms using a colony-drip flow reactor model. The ability of in vitro biofilms to produce proteases, and the effect of PROMOGRAN PRISMA matrix on such proteases, was also investigated. All antimicrobial dressings tested reduced vegetative bacterial load; however, only PROMOGRAN PRISMA matrix was able to significantly reduce biofilm populations (P = 0·01). Additionally, PROMOGRAN PRISMA matrix was the only dressing that did not inhibit dermal fibroblast growth. All other dressings were detrimental to cell viability. In vitro biofilms of Pseudomonas aeruginosa were demonstrated as being capable of releasing bacterial proteases into their surroundings, and incubation with PROMOGRAN PRISMA matrix led to a 77% reduction in activity of such proteases (P = 0·002). The unique ability of PROMOGRAN PRISMA matrix to reduce in vitro vegetative bacteria, biofilm bacteria and bacterial proteases while still allowing dermal fibroblast proliferation may help rebalance the wound environment and reduce the occurrence of infection.
Collapse
Affiliation(s)
- Katie A Bourdillon
- Research & Development DepartmentSystagenix, An Acelity CompanySkiptonUK
| | - Craig P Delury
- Research & Development DepartmentSystagenix, An Acelity CompanySkiptonUK
| | - Breda M Cullen
- Research & Development DepartmentSystagenix, An Acelity CompanySkiptonUK
| |
Collapse
|
150
|
Chevalier S, Bouffartigues E, Bodilis J, Maillot O, Lesouhaitier O, Feuilloley MGJ, Orange N, Dufour A, Cornelis P. Structure, function and regulation of Pseudomonas aeruginosa porins. FEMS Microbiol Rev 2017; 41:698-722. [PMID: 28981745 DOI: 10.1093/femsre/fux020] [Citation(s) in RCA: 236] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 04/24/2017] [Indexed: 12/11/2022] Open
Abstract
Pseudomonas aeruginosa is a Gram-negative bacterium belonging to the γ-proteobacteria. Like other members of the Pseudomonas genus, it is known for its metabolic versatility and its ability to colonize a wide range of ecological niches, such as rhizosphere, water environments and animal hosts, including humans where it can cause severe infections. Another particularity of P. aeruginosa is its high intrinsic resistance to antiseptics and antibiotics, which is partly due to its low outer membrane permeability. In contrast to Enterobacteria, pseudomonads do not possess general diffusion porins in their outer membrane, but rather express specific channel proteins for the uptake of different nutrients. The major outer membrane 'porin', OprF, has been extensively investigated, and displays structural, adhesion and signaling functions while its role in the diffusion of nutrients is still under discussion. Other porins include OprB and OprB2 for the diffusion of glucose, the two small outer membrane proteins OprG and OprH, and the two porins involved in phosphate/pyrophosphate uptake, OprP and OprO. The remaining nineteen porins belong to the so-called OprD (Occ) family, which is further split into two subfamilies termed OccD (8 members) and OccK (11 members). In the past years, a large amount of information concerning the structure, function and regulation of these porins has been published, justifying why an updated review is timely.
Collapse
Affiliation(s)
- Sylvie Chevalier
- Laboratory of Microbiology Signals and Microenvironment LMSM EA 4312, University of Rouen, Normandy University, 27000 Evreux, France
| | - Emeline Bouffartigues
- Laboratory of Microbiology Signals and Microenvironment LMSM EA 4312, University of Rouen, Normandy University, 27000 Evreux, France
| | - Josselin Bodilis
- Laboratory of Microbiology Signals and Microenvironment LMSM EA 4312, University of Rouen, Normandy University, 27000 Evreux, France
| | - Olivier Maillot
- Laboratory of Microbiology Signals and Microenvironment LMSM EA 4312, University of Rouen, Normandy University, 27000 Evreux, France
| | - Olivier Lesouhaitier
- Laboratory of Microbiology Signals and Microenvironment LMSM EA 4312, University of Rouen, Normandy University, 27000 Evreux, France
| | - Marc G J Feuilloley
- Laboratory of Microbiology Signals and Microenvironment LMSM EA 4312, University of Rouen, Normandy University, 27000 Evreux, France
| | - Nicole Orange
- Laboratory of Microbiology Signals and Microenvironment LMSM EA 4312, University of Rouen, Normandy University, 27000 Evreux, France
| | - Alain Dufour
- IUEM, Laboratoire de Biotechnologie et Chimie Marines EA 3884, Université de Bretagne-Sud (UEB), 56321 Lorient, France
| | - Pierre Cornelis
- Laboratory of Microbiology Signals and Microenvironment LMSM EA 4312, University of Rouen, Normandy University, 27000 Evreux, France
| |
Collapse
|