801
|
Tang L, Liang X, Moore R, Dong TG. Commentary: The icmF3 Locus is Involved in Multiple Adaptation- and Virulence-related Characteristics in Pseudomonas aeruginosa PAO1. Front Cell Infect Microbiol 2015; 5:83. [PMID: 26636045 PMCID: PMC4649053 DOI: 10.3389/fcimb.2015.00083] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 11/03/2015] [Indexed: 11/25/2022] Open
Affiliation(s)
- Le Tang
- Ecosystem and Public Health, University of Calgary Calgary, AB, Canada
| | - Xiaoye Liang
- Ecosystem and Public Health, University of Calgary Calgary, AB, Canada
| | - Richard Moore
- Ecosystem and Public Health, University of Calgary Calgary, AB, Canada
| | - Tao G Dong
- Ecosystem and Public Health, University of Calgary Calgary, AB, Canada
| |
Collapse
|
802
|
Abstract
Antimicrobial-resistant bacteria pose a serious threat in the clinic. This is particularly true for opportunistic pathogens that possess high intrinsic resistance. Though many studies have focused on understanding the acquisition of bacterial resistance upon exposure to antimicrobials, the mechanisms controlling intrinsic resistance are not well understood. In this study, we subjected the model opportunistic superbug Pseudomonas aeruginosa to 14 antimicrobials under highly controlled conditions and assessed its response using expression- and fitness-based genomic approaches. Our results reveal that gene expression changes and mutant fitness in response to sub-MIC antimicrobials do not correlate on a genomewide scale, indicating that gene expression is not a good predictor of fitness determinants. In general, fewer fitness determinants were identified for antiseptics and disinfectants than for antibiotics. Analysis of gene expression and fitness data together allowed the prediction of antagonistic interactions between antimicrobials and insight into the molecular mechanisms controlling these interactions. Infections involving multidrug-resistant pathogens are difficult to treat because the therapeutic options are limited. These infections impose a significant financial burden on infected patients and on health care systems. Despite years of antimicrobial resistance research, we lack a comprehensive understanding of the intrinsic mechanisms controlling antimicrobial resistance. This work uses two fine-scale genomic approaches to identify genetic loci important for antimicrobial resistance of the opportunistic pathogen Pseudomonas aeruginosa. Our results reveal that antibiotics have more resistance determinants than antiseptics/disinfectants and that gene expression upon exposure to antimicrobials is not a good predictor of these resistance determinants. In addition, we show that when used together, genomewide gene expression and fitness profiling can provide mechanistic insights into multidrug resistance mechanisms.
Collapse
|
803
|
Bai F, Ho Lim C, Jia J, Santostefano K, Simmons C, Kasahara H, Wu W, Terada N, Jin S. Directed Differentiation of Embryonic Stem Cells Into Cardiomyocytes by Bacterial Injection of Defined Transcription Factors. Sci Rep 2015; 5:15014. [PMID: 26449528 PMCID: PMC4598736 DOI: 10.1038/srep15014] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 09/15/2015] [Indexed: 02/06/2023] Open
Abstract
Forced expression of defined transcriptional factors has been well documented as an effective method for cellular reprogramming or directed differentiation. However, transgene expression is not amenable for therapeutic application due to potential insertional mutagenesis. Here, we have developed a bacterial type III secretion system (T3SS)-based protein delivery tool and shown its application in directing pluripotent stem cell differentiation by a controlled delivery of transcription factors relevant to early heart development. By fusing to an N-terminal secretion sequence for T3SS-dependent injection, three transcriptional factors, namely Gata4, Mef2c, and Tbx5 (abbreviated as GMT), were translocated into murine embryonic stem cells (ESCs), where the proteins are effectively targeted to the nucleus with an average intracellular half-life of 5.5 hours. Exogenous GMT protein injection activated the cardiac program, and multiple rounds of GMT protein delivery significantly improved the efficiency of ESC differentiation into cardiomyocytes. Combination of T3SS-mediated GMT delivery and Activin A treatment showed an additive effect, resulting in on average 60% of the ESCs differentiated into cardiomyocytes. ESC derived cardiomyocytes displayed spontaneous rhythmic contractile movement as well as normal hormonal responses. This work serves as a foundation for the bacterial delivery of multiple transcription factors to direct cell fate without jeopardizing genomic integrity.
Collapse
Affiliation(s)
- Fang Bai
- State Key Laboratory of Medicinal Chemical Biology and Colleges of Pharmacy and Life Sciences, Nankai University, Tianjin, China.,Department of Molecular Genetics and Microbiology College of Medicine, University of Florida, Gainesville, FL 32610
| | - Chae Ho Lim
- Department of Pathology College of Medicine, University of Florida, Gainesville, FL 32610
| | - Jingyue Jia
- State Key Laboratory of Medicinal Chemical Biology and Colleges of Pharmacy and Life Sciences, Nankai University, Tianjin, China.,Department of Molecular Genetics and Microbiology College of Medicine, University of Florida, Gainesville, FL 32610
| | - Katherine Santostefano
- Department of Pathology College of Medicine, University of Florida, Gainesville, FL 32610
| | - Chelsey Simmons
- Department of Mechanical &Aerospace Engineering College of Engineering, University of Florida, Gainesville, FL 32611
| | - Hideko Kasahara
- Department of Physiology and Functional Genomics, University of Florida, College of Medicine, Gainesville, Florida, United States of America
| | - Weihui Wu
- State Key Laboratory of Medicinal Chemical Biology and Colleges of Pharmacy and Life Sciences, Nankai University, Tianjin, China
| | - Naohiro Terada
- Department of Pathology College of Medicine, University of Florida, Gainesville, FL 32610
| | - Shouguang Jin
- State Key Laboratory of Medicinal Chemical Biology and Colleges of Pharmacy and Life Sciences, Nankai University, Tianjin, China.,Department of Molecular Genetics and Microbiology College of Medicine, University of Florida, Gainesville, FL 32610
| |
Collapse
|
804
|
Dupin L, Zuttion F, Géhin T, Meyer A, Phaner-Goutorbe M, Vasseur JJ, Souteyrand E, Morvan F, Chevolot Y. Effects of the Surface Densities of Glycoclusters on the Determination of Their IC50andKdValue Determination by Using a Microarray. Chembiochem 2015; 16:2329-36. [DOI: 10.1002/cbic.201500371] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Indexed: 01/21/2023]
Affiliation(s)
- Lucie Dupin
- Institut des Nanotechnologies de Lyon (INL); UMR 5270 CNRS; Ecole Centrale de Lyon; Université de Lyon; 36 Avenue Guy de Collongue 69134 Ecully France
| | - Francesca Zuttion
- Institut des Nanotechnologies de Lyon (INL); UMR 5270 CNRS; Ecole Centrale de Lyon; Université de Lyon; 36 Avenue Guy de Collongue 69134 Ecully France
| | - Thomas Géhin
- Institut des Nanotechnologies de Lyon (INL); UMR 5270 CNRS; Ecole Centrale de Lyon; Université de Lyon; 36 Avenue Guy de Collongue 69134 Ecully France
| | - Albert Meyer
- Institut des Biomolécules Max Mousseron (IBMM); UMR 5247 CNRS; UM; ENSCM; Université de Montpellier; Place E. Bataillon CC1704 34095 Montpellier Cedex 5 France
| | - Magali Phaner-Goutorbe
- Institut des Nanotechnologies de Lyon (INL); UMR 5270 CNRS; Ecole Centrale de Lyon; Université de Lyon; 36 Avenue Guy de Collongue 69134 Ecully France
| | - Jean-Jacques Vasseur
- Institut des Biomolécules Max Mousseron (IBMM); UMR 5247 CNRS; UM; ENSCM; Université de Montpellier; Place E. Bataillon CC1704 34095 Montpellier Cedex 5 France
| | - Eliane Souteyrand
- Institut des Nanotechnologies de Lyon (INL); UMR 5270 CNRS; Ecole Centrale de Lyon; Université de Lyon; 36 Avenue Guy de Collongue 69134 Ecully France
| | - François Morvan
- Institut des Biomolécules Max Mousseron (IBMM); UMR 5247 CNRS; UM; ENSCM; Université de Montpellier; Place E. Bataillon CC1704 34095 Montpellier Cedex 5 France
| | - Yann Chevolot
- Institut des Nanotechnologies de Lyon (INL); UMR 5270 CNRS; Ecole Centrale de Lyon; Université de Lyon; 36 Avenue Guy de Collongue 69134 Ecully France
| |
Collapse
|
805
|
Klapiszewski Ł, Rzemieniecki T, Krawczyk M, Malina D, Norman M, Zdarta J, Majchrzak I, Dobrowolska A, Czaczyk K, Jesionowski T. Kraft lignin/silica–AgNPs as a functional material with antibacterial activity. Colloids Surf B Biointerfaces 2015. [DOI: 10.1016/j.colsurfb.2015.06.056] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
806
|
Savale SA, Pol CS, Khare R, Verma N, Gaikwad S, Mandal B, Behera BC. Radical scavenging, prolyl endopeptidase inhibitory, and antimicrobial potential of a cultured Himalayan lichen Cetrelia olivetorum. PHARMACEUTICAL BIOLOGY 2015; 54:692-700. [PMID: 26429132 DOI: 10.3109/13880209.2015.1072567] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
CONTEXT Lichens are source of natural bioactive compounds which are traditionally used to cure a variety of ailments. OBJECTIVE The objective of this study is to assess free radical scavenging, prolyl endopeptidase inhibitory (PEPI), and antimicrobial potential of a high altitude lichen species Cetrelia olivetorum (Nyl.) W. L. Culb. & C. F. Culb (Parmeliaceae). MATERIALS AND METHODS Lichen C. olivetorum has been cultured in vitro, and optimized culture conditions were implemented in bioreactor to obtain high quantity of biomass for the study of radical scavenging, PEPI, and antimicrobial activities. Radical scavenging activity of methanol extract of Cetrelia olivetorum (MECO) was tested at 100 µg/mL, PEPI activity at 25 and 50 µg/mL, and antimicrobial activity at 5, 25, 50, and 100 µg/mL conc. All the biological activities of natural thallus extract and its derived culture extract were evaluated spectrophotometrically. RESULTS Murashige and Skoog medium supplemented with 3% glucose and 100 ppb indole-3-butyric acid (IBA) supported biomass growth at flask level and yielded 5.095 g biomass in bioreactor. MECO of both the cultured and the natural lichen exhibited half inhibiting concentration (IC50) for radical scavenging activities in the range of 50-60 µg/mL, whereas the IC50 value of standard antioxidants was found to be in the range of 12-29 µg/mL. The IC50 value of lichen extract for PEPI activity was 144-288 µg/mL, whereas the IC50 value of standard prolyl endopeptidase inhibitor, Z-pro-prolinal, was 57.73 µg/mL. As far as the antimicrobial activity of MECO is concerned, minimum inhibitory concentration (MIC) value of lichen extracts against tested microorganisms was obtained in the range of 50-104 µg/mL and found to be more effective than commercially available standard erythromycin. DISCUSSION Murashige and Skoog medium containing IBA was found to be suitable for maximum biomass production of C. olivetorum under bioreactor conditions. The cultured lichen biomass extract also showed antioxidant, PEPI, and antimicrobial potential. CONCLUSION The present study indicates therapeutic potential of Himalayan lichen C. olivetorum against neurodegenerative diseases owing to its radical scavenging, PEPI, and antimicrobial activities. Further, the result encourages its commercial exploitation through mass culture for production of its bioactive components and their use in pharmaceutical and nutraceutical industries.
Collapse
Affiliation(s)
| | | | - Roshni Khare
- a Agharkar Research Institute , Pune , Maharashtra , India
| | - Neeraj Verma
- a Agharkar Research Institute , Pune , Maharashtra , India
| | | | - Bapi Mandal
- a Agharkar Research Institute , Pune , Maharashtra , India
| | | |
Collapse
|
807
|
Freschi L, Jeukens J, Kukavica-Ibrulj I, Boyle B, Dupont MJ, Laroche J, Larose S, Maaroufi H, Fothergill JL, Moore M, Winsor GL, Aaron SD, Barbeau J, Bell SC, Burns JL, Camara M, Cantin A, Charette SJ, Dewar K, Déziel É, Grimwood K, Hancock REW, Harrison JJ, Heeb S, Jelsbak L, Jia B, Kenna DT, Kidd TJ, Klockgether J, Lam JS, Lamont IL, Lewenza S, Loman N, Malouin F, Manos J, McArthur AG, McKeown J, Milot J, Naghra H, Nguyen D, Pereira SK, Perron GG, Pirnay JP, Rainey PB, Rousseau S, Santos PM, Stephenson A, Taylor V, Turton JF, Waglechner N, Williams P, Thrane SW, Wright GD, Brinkman FSL, Tucker NP, Tümmler B, Winstanley C, Levesque RC. Clinical utilization of genomics data produced by the international Pseudomonas aeruginosa consortium. Front Microbiol 2015; 6:1036. [PMID: 26483767 PMCID: PMC4586430 DOI: 10.3389/fmicb.2015.01036] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 09/11/2015] [Indexed: 11/24/2022] Open
Abstract
The International Pseudomonas aeruginosa Consortium is sequencing over 1000 genomes and building an analysis pipeline for the study of Pseudomonas genome evolution, antibiotic resistance and virulence genes. Metadata, including genomic and phenotypic data for each isolate of the collection, are available through the International Pseudomonas Consortium Database (http://ipcd.ibis.ulaval.ca/). Here, we present our strategy and the results that emerged from the analysis of the first 389 genomes. With as yet unmatched resolution, our results confirm that P. aeruginosa strains can be divided into three major groups that are further divided into subgroups, some not previously reported in the literature. We also provide the first snapshot of P. aeruginosa strain diversity with respect to antibiotic resistance. Our approach will allow us to draw potential links between environmental strains and those implicated in human and animal infections, understand how patients become infected and how the infection evolves over time as well as identify prognostic markers for better evidence-based decisions on patient care.
Collapse
Affiliation(s)
- Luca Freschi
- Institute for Integrative and Systems Biology, Université Laval Quebec, QC, Canada
| | - Julie Jeukens
- Institute for Integrative and Systems Biology, Université Laval Quebec, QC, Canada
| | | | - Brian Boyle
- Institute for Integrative and Systems Biology, Université Laval Quebec, QC, Canada
| | - Marie-Josée Dupont
- Institute for Integrative and Systems Biology, Université Laval Quebec, QC, Canada
| | - Jérôme Laroche
- Institute for Integrative and Systems Biology, Université Laval Quebec, QC, Canada
| | - Stéphane Larose
- Institute for Integrative and Systems Biology, Université Laval Quebec, QC, Canada
| | - Halim Maaroufi
- Institute for Integrative and Systems Biology, Université Laval Quebec, QC, Canada
| | - Joanne L Fothergill
- Institute of Infection and Global Health, University of Liverpool Liverpool, UK
| | - Matthew Moore
- Institute of Infection and Global Health, University of Liverpool Liverpool, UK
| | - Geoffrey L Winsor
- Department of Molecular Biology and Biochemistry, Simon Fraser University Vancouver, BC, Canada
| | - Shawn D Aaron
- Ottawa Hospital Research Institute Ottawa, ON, Canada
| | - Jean Barbeau
- Faculté de Médecine Dentaire, Université de Montréal Montréal, QC, Canada
| | - Scott C Bell
- QIMR Berghofer Medical Research Institute Brisbane, QLD, Australia
| | - Jane L Burns
- Seattle Children's Research Institute, University of Washington School of Medicine Seattle, WA, USA
| | - Miguel Camara
- School of Life Sciences, University of Nottingham Nottingham, UK
| | - André Cantin
- Département de Médecine, Université de Sherbrooke Sherbrooke, QC, Canada
| | - Steve J Charette
- Institute for Integrative and Systems Biology, Université Laval Quebec, QC, Canada ; Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec Quebec, QC, Canada ; Département de Biochimie, de Microbiologie et de Bio-informatique, Faculté des Sciences et de Génie, Université Laval Quebec, QC, Canada
| | - Ken Dewar
- Department of Human Genetics, McGill University Montreal, QC, Canada
| | - Éric Déziel
- INRS Institut Armand Frappier Laval, QC, Canada
| | - Keith Grimwood
- School of Medicine, Griffith University Gold Coast, QLD, Australia
| | - Robert E W Hancock
- Department of Microbiology and Immunology, University of British Columbia Vancouver, BC, Canada
| | - Joe J Harrison
- Biological Sciences, University of Calgary Calgary, AB, Canada
| | - Stephan Heeb
- School of Life Sciences, University of Nottingham Nottingham, UK
| | - Lars Jelsbak
- Department of Systems Biology, Technical University of Denmark Lyngby, Denmark
| | - Baofeng Jia
- M.G. DeGroote Institute for Infectious Disease Research, McMaster University Hamilton, ON, Canada
| | - Dervla T Kenna
- Antimicrobial Resistance and Healthcare Associated Infections Reference Unit, Public Health England London, UK
| | - Timothy J Kidd
- Child Health Research Centre, The University of Queensland Brisbane, QLD, Australia ; Centre for Infection and Immunity, Queen's University Belfast Belfast, UK
| | - Jens Klockgether
- Klinische Forschergruppe, Medizinische Hochschule Hannover, Germany
| | - Joseph S Lam
- Department of Molecular and Cellular Biology, University of Guelph Guelph, ON, Canada
| | - Iain L Lamont
- Department of Biochemistry, University of Otago Dunedin, New Zealand
| | - Shawn Lewenza
- Biological Sciences, University of Calgary Calgary, AB, Canada
| | - Nick Loman
- Institute for Microbiology and Infection, University of Birmingham Birmingham, UK
| | - François Malouin
- Département de Médecine, Université de Sherbrooke Sherbrooke, QC, Canada
| | - Jim Manos
- Department of Infectious Diseases and Immunology, The University of Sydney Sydney, NSW, Australia
| | - Andrew G McArthur
- M.G. DeGroote Institute for Infectious Disease Research, McMaster University Hamilton, ON, Canada
| | - Josie McKeown
- School of Life Sciences, University of Nottingham Nottingham, UK
| | - Julie Milot
- Department of Pneumology, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval Quebec, QC, Canada
| | - Hardeep Naghra
- School of Life Sciences, University of Nottingham Nottingham, UK
| | - Dao Nguyen
- Department of Human Genetics, McGill University Montreal, QC, Canada ; Department of Microbiology and Immunology and Department of Experimental Medicine, McGill University Montreal, QC, Canada
| | - Sheldon K Pereira
- M.G. DeGroote Institute for Infectious Disease Research, McMaster University Hamilton, ON, Canada
| | - Gabriel G Perron
- Department of Biology, Bard College, Annandale-On-Hudson NY, USA
| | - Jean-Paul Pirnay
- Laboratory for Molecular and Cellular Technology, Queen Astrid Military Hospital Brussels, Belgium
| | - Paul B Rainey
- New Zealand Institute for Advanced Study, Massey University Albany, New Zealand ; Max Planck Institute for Evolutionary Biology Plön, Germany
| | - Simon Rousseau
- Department of Human Genetics, McGill University Montreal, QC, Canada
| | - Pedro M Santos
- Department of Biology, University of Minho Braga, Portugal
| | | | - Véronique Taylor
- Department of Molecular and Cellular Biology, University of Guelph Guelph, ON, Canada
| | - Jane F Turton
- Antimicrobial Resistance and Healthcare Associated Infections Reference Unit, Public Health England London, UK
| | - Nicholas Waglechner
- M.G. DeGroote Institute for Infectious Disease Research, McMaster University Hamilton, ON, Canada
| | - Paul Williams
- School of Life Sciences, University of Nottingham Nottingham, UK
| | - Sandra W Thrane
- Department of Systems Biology, Technical University of Denmark Lyngby, Denmark
| | - Gerard D Wright
- M.G. DeGroote Institute for Infectious Disease Research, McMaster University Hamilton, ON, Canada
| | - Fiona S L Brinkman
- Department of Molecular Biology and Biochemistry, Simon Fraser University Vancouver, BC, Canada
| | - Nicholas P Tucker
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde Glasgow, UK
| | - Burkhard Tümmler
- Klinische Forschergruppe, Medizinische Hochschule Hannover, Germany
| | - Craig Winstanley
- Institute of Infection and Global Health, University of Liverpool Liverpool, UK
| | - Roger C Levesque
- Institute for Integrative and Systems Biology, Université Laval Quebec, QC, Canada
| |
Collapse
|
808
|
Barber MF, Elde NC. Buried Treasure: Evolutionary Perspectives on Microbial Iron Piracy. Trends Genet 2015; 31:627-636. [PMID: 26431675 PMCID: PMC4639441 DOI: 10.1016/j.tig.2015.09.001] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 08/18/2015] [Accepted: 09/04/2015] [Indexed: 12/14/2022]
Abstract
Host–pathogen interactions provide valuable systems for the study of evolutionary genetics and natural selection. The sequestration of essential iron has emerged as a crucial innate defense system termed nutritional immunity, leading pathogens to evolve mechanisms of ‘iron piracy’ to scavenge this metal from host proteins. This battle for iron carries numerous consequences not only for host–pathogen evolution but also microbial community interactions. Here we highlight recent and potential future areas of investigation on the evolutionary implications of microbial iron piracy in relation to molecular arms races, host range, competition, and virulence. Applying evolutionary genetic approaches to the study of microbial iron acquisition could also provide new inroads for understanding and combating infectious disease. The battle between microbes and their hosts for nutrient iron is emerging as a new front of evolutionary genetic conflict. Molecular arms races can emerge between host iron-binding proteins and microbial ‘iron piracy’ factors that steal this nutrient for growth. Such rapid evolution may also contribute to the host range of pathogenic microbes. Iron acquisition plays an important role in evolutionary interactions between microbes, both in the environment and within the host. Competition for iron can prevent infection by pathogens, while genetic changes in iron acquisition systems can enhance microbial virulence. Evolutionary conflicts for nutrient iron are revealing potential new genetic mechanisms of disease resistance as well as avenues for therapeutic development.
Collapse
Affiliation(s)
- Matthew F Barber
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT 84112, USA.
| | - Nels C Elde
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| |
Collapse
|
809
|
Viganor L, Galdino ACM, Nunes APF, Santos KRN, Branquinha MH, Devereux M, Kellett A, McCann M, Santos ALS. Anti-Pseudomonas aeruginosa activity of 1,10-phenanthroline-based drugs against both planktonic- and biofilm-growing cells. J Antimicrob Chemother 2015; 71:128-34. [PMID: 26416778 DOI: 10.1093/jac/dkv292] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 08/21/2015] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVES The beneficial antimicrobial properties of 1,10-phenanthroline (phen)-based drugs, together with the imperative need to develop new chemotherapeutic options for prevention/treatment of infections caused by MDR Gram-negative bacteria, led us to evaluate the effects of phen, 1,10-phenanthroline-5,6-dione (phendione), [Ag(phendione)2]ClO4 and [Cu(phendione)3](ClO4)2·4H2O on planktonic- and biofilm-growing Pseudomonas aeruginosa. METHODS Thirty-two non-duplicated Brazilian clinical isolates of P. aeruginosa with distinct genetic backgrounds were used in all experiments. The effect of test compounds on planktonic bacterial proliferation was determined as recommended by CLSI protocol. The effect on biofilm formation was evaluated by crystal violet incorporation (biomass determination) and XTT (viability assay). Mature biofilm disorganization was evidenced by staining with crystal violet. RESULTS Phen-based compounds presented anti-P. aeruginosa activity, but with different potencies concerning the geometric mean MIC: [Cu(phendione)3](2+) (7.76 μM) > [Ag(phendione)2](+) (14.05 μM) > phendione (31.15 μM) > phen (579.28 μM). MICs of each compound were similar irrespective of whether the P. aeruginosa isolates were susceptible or resistant to classical antimicrobials (ceftazidime, meropenem and imipenem). The pretreatment of bacteria with phen, phendione and phendione's metal derivatives at 0.5 × MIC value inhibited biofilm formation, particularly the use of [Cu(phendione)3](2+) and [Ag(phendione)2](+), which significantly reduced both biomass (48% and 44%, respectively) and viability (78% and 77%, respectively). The compounds studied also disrupted mature biofilm in a dose-dependent manner, especially [Ag(phendione)2](+) and [Cu(phendione)3](2+) (IC50, 9.39 and 10.16 μM, respectively). CONCLUSIONS Coordination of phendione to Ag(+) and Cu(2+) represents a new promising group of anti-infective agents, which revealed a potent anti-P. aeruginosa action against both planktonic- and biofilm-growing cells.
Collapse
Affiliation(s)
- Livia Viganor
- General Microbiology Department, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil The Inorganic Pharmaceutical and Biomimetic Research Centre, Focas Research Institute, Dublin Institute of Technology, Dublin, Ireland
| | - Anna Clara M Galdino
- General Microbiology Department, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil Biochemistry Post-Graduation Programme, Chemistry Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Paula F Nunes
- Pathology Department and Infection Diseases Post-Graduation Programme, Federal University of Espírito Santo, Espírito Santo, Brazil
| | - Kátia R N Santos
- Medical Microbiology Department, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marta H Branquinha
- General Microbiology Department, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Michael Devereux
- The Inorganic Pharmaceutical and Biomimetic Research Centre, Focas Research Institute, Dublin Institute of Technology, Dublin, Ireland
| | - Andrew Kellett
- School of Chemical Sciences and the National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Malachy McCann
- Chemistry Department, Maynooth University, National University of Ireland, Maynooth, Ireland
| | - André L S Santos
- General Microbiology Department, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil The Inorganic Pharmaceutical and Biomimetic Research Centre, Focas Research Institute, Dublin Institute of Technology, Dublin, Ireland
| |
Collapse
|
810
|
Hosseinkhan N, Zarrineh P, Rokni-Zadeh H, Ashouri MR, Masoudi-Nejad A. Co-expressional conservation in virulence and stress related genes of three Gammaproteobacterial species: Escherichia coli, Salmonella enterica and Pseudomonas aeruginosa. MOLECULAR BIOSYSTEMS 2015; 11:3137-48. [PMID: 26387845 DOI: 10.1039/c5mb00353a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Gene co-expression analysis is one of the main aspects of systems biology that uses high-throughput gene expression data. In the present study we applied cross-species co-expressional analysis on a module of biofilm and stress response associated genes. We addressed different kinds of stresses in three most intensively studied members of Gammaproteobacteria including Escherichia coli K12, Pseudomonas aeruginosa PAO1 and Salmonella enterica for which large sets of gene expression data are available. Our aim was to evaluate the presence of common stress response strategies adopted by these microorganisms that may be assigned to the other members of Gammaproteobacteria. Results of functional annotation analysis revealed distinct categories among co-expressed genes, most of which concerned biological processes associated with virulence and stress response. Transcriptional regulatory analysis of genes present in co-expressed modules showed that the global stress sigma factor, RpoS, besides several local transcription factors accounts for the observed co-expressional response, and that several cases of feed-forward loops exist between global regulators, local transcription factors and their targets. Our results lend partial support to our underlying assumption of the conservation of core biological processes and regulatory interactions among these related Gammaproteobacteria members. This has led to the implementation of transferring gene function annotations from well-studied Gammaproteobacterial species to less-characterized members. These findings can shed light on the discovery of new drug targets capable of controlling severe infections caused by these groups of bacteria.
Collapse
Affiliation(s)
- Nazanin Hosseinkhan
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| | | | | | | | | |
Collapse
|
811
|
Bactericidal Effect of Tomatidine-Tobramycin Combination against Methicillin-Resistant Staphylococcus aureus and Pseudomonas aeruginosa Is Enhanced by Interspecific Small-Molecule Interactions. Antimicrob Agents Chemother 2015; 59:7458-64. [PMID: 26392496 PMCID: PMC4649251 DOI: 10.1128/aac.01711-15] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 09/12/2015] [Indexed: 11/20/2022] Open
Abstract
This study investigated the antibacterial activity of the plant alkaloid tomatidine (TO) against Staphylococcus aureus grown in the presence of Pseudomonas aeruginosa. Since the P. aeruginosa exoproduct 4-hydroxy-2-heptylquinoline-N-oxide (HQNO) is known to cause a respiratory deficiency in S. aureus and respiratory-deficient S. aureus are known to be hypersensitive to TO, we assessed kill kinetics of TO (8 μg/ml) against S. aureus in coculture with P. aeruginosa. Kill kinetics were also assessed using P. aeruginosa mutants deficient in the production of different exoproducts and quorum sensing-related compounds. After 24 h in coculture, TO increased the killing of S. aureus by 3.4 log10 CFU/ml in comparison to that observed in a coculture without TO. The effect of TO was abolished when S. aureus was in coculture with the lasRrhlR, pqsA, pqsL, or lasA mutant of P. aeruginosa. The bactericidal effect of TO against S. aureus in coculture with the pqsL mutant was restored by supplemental HQNO. In an S. aureus monoculture, the combination of HQNO and TO was bacteriostatic, indicating that the pqsL mutant produced an additional factor required for the bactericidal effect. The bactericidal activity of TO was also observed against a tobramycin-resistant methicillin-resistant S. aureus (MRSA) in coculture with P. aeruginosa, and the addition of tobramycin significantly suppressed the growth of both microorganisms. TO shows a strong bactericidal effect against S. aureus when cocultured with P. aeruginosa. The combination of TO and tobramycin may represent a new treatment approach for cystic fibrosis patients frequently cocolonized by MRSA and P. aeruginosa.
Collapse
|
812
|
Michalska M, Wolf P. Pseudomonas Exotoxin A: optimized by evolution for effective killing. Front Microbiol 2015; 6:963. [PMID: 26441897 PMCID: PMC4584936 DOI: 10.3389/fmicb.2015.00963] [Citation(s) in RCA: 161] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 08/31/2015] [Indexed: 11/13/2022] Open
Abstract
Pseudomonas Exotoxin A (PE) is the most toxic virulence factor of the pathogenic bacterium Pseudomonas aeruginosa. This review describes current knowledge about the intoxication pathways of PE. Moreover, PE represents a remarkable example for pathoadaptive evolution, how bacterial molecules have been structurally and functionally optimized under evolutionary pressure to effectively impair and kill their host cells.
Collapse
Affiliation(s)
- Marta Michalska
- Department of Urology, Medical Center, University of Freiburg Freiburg, Germany
| | - Philipp Wolf
- Department of Urology, Medical Center, University of Freiburg Freiburg, Germany
| |
Collapse
|
813
|
Couto N, Schooling SR, Dutcher JR, Barber J. Proteome Profiles of Outer Membrane Vesicles and Extracellular Matrix of Pseudomonas aeruginosa Biofilms. J Proteome Res 2015; 14:4207-22. [PMID: 26303878 DOI: 10.1021/acs.jproteome.5b00312] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In the present work, two different proteomic platforms, gel-based and gel-free, were used to map the matrix and outer membrane vesicle exoproteomes of Pseudomonas aeruginosa PAO1 biofilms. These two proteomic strategies allowed us a confident identification of 207 and 327 proteins from enriched outer membrane vesicles and whole matrix isolated from biofilms. Because of the physicochemical characteristics of these subproteomes, the two strategies showed complementarity, and thus, the most comprehensive analysis of P. aeruginosa exoproteome to date was achieved. Under our conditions, outer membrane vesicles contribute approximately 20% of the whole matrix proteome, demonstrating that membrane vesicles are an important component of the matrix. The proteomic profiles were analyzed in terms of their biological context, namely, a biofilm. Accordingly relevant metabolic processes involved in cellular adaptation to the biofilm lifestyle as well as those related to P. aeruginosa virulence capabilities were a key feature of the analyses. The diversity of the matrix proteome corroborates the idea of high heterogeneity within the biofilm; cells can display different levels of metabolism and can adapt to local microenvironments making this proteomic analysis challenging. In addition to analyzing our own primary data, we extend the analysis to published data by other groups in order to deepen our understanding of the complexity inherent within biofilm populations.
Collapse
Affiliation(s)
- Narciso Couto
- Michael Barber Centre for Mass Spectrometry, Manchester Institute for Biotechnology, University of Manchester , Princess Road, Manchester, M1 7DN, U.K
| | - Sarah R Schooling
- Department of Molecular and Cellular Biology, College of Biological Science, University of Guelph , Guelph, ON N1G 2W1, Canada.,Department of Physics, University of Guelph , Guelph, ON N1G 2W1, Canada
| | - John R Dutcher
- Department of Physics, University of Guelph , Guelph, ON N1G 2W1, Canada
| | - Jill Barber
- Michael Barber Centre for Mass Spectrometry, Manchester Institute for Biotechnology, University of Manchester , Princess Road, Manchester, M1 7DN, U.K.,Manchester Pharmacy School, University of Manchester , Stopford Building, Oxford Road, Manchester, M13 9PT, U.K
| |
Collapse
|
814
|
Haney EF, Mansour SC, Hilchie AL, de la Fuente-Núñez C, Hancock REW. High throughput screening methods for assessing antibiofilm and immunomodulatory activities of synthetic peptides. Peptides 2015; 71:276-85. [PMID: 25836992 PMCID: PMC4581888 DOI: 10.1016/j.peptides.2015.03.015] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 03/17/2015] [Accepted: 03/19/2015] [Indexed: 12/31/2022]
Abstract
The recent observation that certain cationic peptides possess potent antibiofilm activity demonstrated that small peptides could be used to treat biofilm-associated infections. Other so-called innate defense regulator peptides possess potent immunomodulatory properties such as leukocyte recruitment and suppression of harmful inflammation. A peptide that directly targets biofilm cells while favorably modulating the immune response would be particularly advantageous for treating serious skin infections caused by Staphylococcus aureus. In the present work, using SPOT-synthesized peptide arrays on cellulose membranes, we outline a strategy for systematically assessing the antibiofilm activity of hundreds of IDR-1002 (VQRWLIVWRIRK-NH2) and IDR-HH2 (VQLRIRVAVIRA-NH2) peptide variants against MRSA biofilms. In addition, the ability of these peptides to stimulate production of a monocyte chemoattractant protein (MCP-1) and suppress LPS-induced interleukin (IL)-1β production in human peripheral blood mononuclear cells (PBMCs) was evaluated. These results informed the synthesis of second-generation peptides resulting in a new peptide, IDR-2009 (KWRLLIRWRIQK-NH2), with enhanced MCP-1 stimulatory activity, favorable IL-1β suppression characteristics and strong antibiofilm activity against MRSA and Pseudomonas aeruginosa biofilms. This work provides a proof-of-concept that multiple peptide activities can be optimized simultaneously to generate novel sequences that possess a variety of biological properties.
Collapse
Affiliation(s)
- Evan F Haney
- Center for Microbial Diseases and Immunity Research, Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sarah C Mansour
- Center for Microbial Diseases and Immunity Research, Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ashley L Hilchie
- Center for Microbial Diseases and Immunity Research, Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - César de la Fuente-Núñez
- Center for Microbial Diseases and Immunity Research, Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Robert E W Hancock
- Center for Microbial Diseases and Immunity Research, Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
815
|
Lorè NI, Iraqi FA, Bragonzi A. Host genetic diversity influences the severity of Pseudomonas aeruginosa pneumonia in the Collaborative Cross mice. BMC Genet 2015; 16:106. [PMID: 26310945 PMCID: PMC4551369 DOI: 10.1186/s12863-015-0260-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 08/03/2015] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Pseudomonas aeruginosa is one of the top three causes of opportunistic infections in humans. Patients with a compromised immune system, due to immunosuppressive therapies or underlying diseases such as cancer, AIDS or the hereditary disease cystic fibrosis, are at risk of developing P. aeruginosa infection. However, clinical evidence indicates extremely variable outcomes of P. aeruginosa infections in individuals at risk, suggesting that host multi-complex genetic traits may influence the severity of this opportunistic infection. Here, we have used an innovative experimental model to dissect whether host genetic background, such as those found in the outbred population, could influence the risk of morbidity and mortality to P. aeruginosa pneumonia. RESULTS A highly genetically-diverse mouse resource population, Collaborative Cross (CC) mice, was infected with a clinical strain of P. aeruginosa and subsequently monitored for mortality, mean survival time, and morbidity, change in body weight for seven days post infection. Disease phenotypes ranged from complete resistance and recovery of body weight to lethal disease. Initial variables, including body weight, age and gender, have limited influence on P. aeruginosa outcome, emphasizing the role of host genetic background in defining the risk of morbidity and mortality. When broad-sense heritability of phenotypic traits was evaluated, it confirmed the influence of genetic profile rather than environmental factors among the CC lines during P. aeruginosa infection. CONCLUSION This innovative model system can potentially reproduce the variables responses of disease severity observed in humans during P. aeruginosa pneumonia. Our results demonstrated that a widely-marked differential response to P. aeruginosa airway infection in term of morbidity and mortality, is mainly affected by host genetic factors, as multiple genetic loci or polymorphic variations.
Collapse
Affiliation(s)
- Nicola Ivan Lorè
- Infection and Cystic Fibrosis Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS - San Raffaele Scientific Institute, Milan, Italy.
| | - Fuad A Iraqi
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, 69978, Tel Aviv, Israel.
| | - Alessandra Bragonzi
- Infection and Cystic Fibrosis Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS - San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
816
|
Oliver A, Mulet X, López-Causapé C, Juan C. The increasing threat of Pseudomonas aeruginosa high-risk clones. Drug Resist Updat 2015; 21-22:41-59. [PMID: 26304792 DOI: 10.1016/j.drup.2015.08.002] [Citation(s) in RCA: 438] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Accepted: 08/04/2015] [Indexed: 01/01/2023]
Abstract
The increasing prevalence of chronic and hospital-acquired infections produced by multidrug-resistant (MDR) or extensively drug-resistant (XDR) Pseudomonas aeruginosa strains is associated with significant morbidity and mortality. This growing threat results from the extraordinary capacity of this pathogen for developing resistance through chromosomal mutations and from the increasing prevalence of transferable resistance determinants, particularly those encoding carbapenemases or extended-spectrum β-lactamases (ESBLs). P. aeruginosa has a nonclonal epidemic population structure, composed of a limited number of widespread clones which are selected from a background of a large quantity of rare and unrelated genotypes that are recombining at high frequency. Indeed, recent concerning reports have provided evidence of the existence of MDR/XDR global clones, denominated high-risk clones, disseminated in hospitals worldwide; ST235, ST111, and ST175 are likely those more widespread. Noteworthy, the vast majority of infections by MDR, and specially XDR, strains are produced by these and few other clones worldwide. Moreover, the association of high-risk clones, particularly ST235, with transferable resistance is overwhelming; nearly 100 different horizontally-acquired resistance elements and up to 39 different acquired β-lactamases have been reported so far among ST235 isolates. Likewise, MDR internationally-disseminated epidemic strains, such as the Liverpool Epidemic Strain (LES, ST146), have been noted as well among cystic fibrosis patients. Here we review the population structure, epidemiology, antimicrobial resistance mechanisms and virulence of the P. aeruginosa high-risk clones. The phenotypic and genetic factors potentially driving the success of high-risk clones, the aspects related to their detection in the clinical microbiology laboratory and the implications for infection control and public health are also discussed.
Collapse
Affiliation(s)
- Antonio Oliver
- Servicio de Microbiología and Unidad de Investigación, Hospital Universitario Son Espases, Instituto de Investigación Sanitaria de Palma (IdISPa), Ctra. Valldemossa 79, 07010 Palma de Mallorca, Spain.
| | - Xavier Mulet
- Servicio de Microbiología and Unidad de Investigación, Hospital Universitario Son Espases, Instituto de Investigación Sanitaria de Palma (IdISPa), Ctra. Valldemossa 79, 07010 Palma de Mallorca, Spain
| | - Carla López-Causapé
- Servicio de Microbiología and Unidad de Investigación, Hospital Universitario Son Espases, Instituto de Investigación Sanitaria de Palma (IdISPa), Ctra. Valldemossa 79, 07010 Palma de Mallorca, Spain
| | - Carlos Juan
- Servicio de Microbiología and Unidad de Investigación, Hospital Universitario Son Espases, Instituto de Investigación Sanitaria de Palma (IdISPa), Ctra. Valldemossa 79, 07010 Palma de Mallorca, Spain
| |
Collapse
|
817
|
Mittal R, Lisi CV, Gerring R, Mittal J, Mathee K, Narasimhan G, Azad RK, Yao Q, Grati M, Yan D, Eshraghi AA, Angeli SI, Telischi FF, Liu XZ. Current concepts in the pathogenesis and treatment of chronic suppurative otitis media. J Med Microbiol 2015; 64:1103-1116. [PMID: 26248613 DOI: 10.1099/jmm.0.000155] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Otitis media (OM) is an inflammation of the middle ear associated with infection. Despite appropriate therapy, acute OM (AOM) can progress to chronic suppurative OM (CSOM) associated with ear drum perforation and purulent discharge. The effusion prevents the middle ear ossicles from properly relaying sound vibrations from the ear drum to the oval window of the inner ear, causing conductive hearing loss. In addition, the inflammatory mediators generated during CSOM can penetrate into the inner ear through the round window. This can cause the loss of hair cells in the cochlea, leading to sensorineural hearing loss. Pseudomonas aeruginosa and Staphylococcus aureus are the most predominant pathogens that cause CSOM. Although the pathogenesis of AOM is well studied, very limited research is available in relation to CSOM. With the emergence of antibiotic resistance as well as the ototoxicity of antibiotics and the potential risks of surgery, there is an urgent need to develop effective therapeutic strategies against CSOM. This warrants understanding the role of host immunity in CSOM and how the bacteria evade these potent immune responses. Understanding the molecular mechanisms leading to CSOM will help in designing novel treatment modalities against the disease and hence preventing the hearing loss.
Collapse
Affiliation(s)
- Rahul Mittal
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Christopher V Lisi
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Robert Gerring
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jeenu Mittal
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Kalai Mathee
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Giri Narasimhan
- Bioinformatics Research Group (BioRG), School of Computing and Information Sciences, Florida International University, Miami, FL, USA
| | - Rajeev K Azad
- Department of Biological Sciences and Mathematics, University of North Texas, Denton, TX, USA
| | - Qi Yao
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - M'hamed Grati
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Denise Yan
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Adrien A Eshraghi
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Simon I Angeli
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Fred F Telischi
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Xue-Zhong Liu
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
818
|
Chen J, Gao X, Hong L, Ma L, Li Y. Expression, purification and functional characterization of a novel 3α-hydroxysteroid dehydrogenase from Pseudomonas aeruginosa. Protein Expr Purif 2015; 115:102-8. [PMID: 26193374 DOI: 10.1016/j.pep.2015.07.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Revised: 06/27/2015] [Accepted: 07/15/2015] [Indexed: 12/17/2022]
Abstract
3α-Hydroxysteroid dehydrogenase (3α-HSD) catalyzes the oxidation of the 3-hydroxyl group of steroids. The enzymatic conversion is a critical step in the enzymatic assay of urinary sulfated bile acids (SBAs), which is a valuable diagnosis index of hepatobiliary diseases. However, the source of 3α-HSD for clinical applications is limited. In this study, an open reading frame (ORF) encoding a novel 3α-HSD was successfully cloned from Pseudomonas aeruginosa and expressed in Escherichia coli BL21 (DE3). The recombinant protein was purified by immobilized metal ion affinity chromatography. Enzyme characterization studies revealed that the protein has 3α-HSD activity and the Km value for sodium cholate is 1.06 mmol L(-1). More than 60% relative enzyme activity was observed in a wide range of pH and temperature, with an optimum pH at 8.0 and an optimum temperature at 30°C. The enzyme's good thermostability under 40°C would be favorable in clinical applications. Ion interference experiments indicated that Zn(2+) was an activating cofactor which increased the enzyme activity 1.75-fold. With the favorable characteristics mentioned above, the new 3α-HSD is a promising enzyme for clinical applications. More importantly, the present work is the first report on a 3α-HSD from P. aeruginosa.
Collapse
Affiliation(s)
- Jianmin Chen
- West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu 610041, China; School of Basic Medical Sciences, Chengdu Medical College, Chengdu 610500, China.
| | - Xiufeng Gao
- West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu 610041, China.
| | - Lin Hong
- West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Liting Ma
- West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Yongsheng Li
- School of Chemical Engineering, Sichuan University, Chengdu 610065, China.
| |
Collapse
|
819
|
Antimicrobial Activity of Fosfomycin-Tobramycin Combination against Pseudomonas aeruginosa Isolates Assessed by Time-Kill Assays and Mutant Prevention Concentrations. Antimicrob Agents Chemother 2015. [PMID: 26195514 DOI: 10.1128/aac.00822-15] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The antibacterial activity of fosfomycin-tobramycin combination was studied by time-kill assay in eight Pseudomonas aeruginosa clinical isolates belonging to the fosfomycin wild-type population (MIC = 64 μg/ml) but with different tobramycin susceptibilities (MIC range, 1 to 64 μg/ml). The mutant prevention concentration (MPC) and mutant selection window (MSW) were determined in five of these strains (tobramycin MIC range, 1 to 64 μg/ml) in aerobic and anaerobic conditions simulating environments that are present in biofilm-mediated infections. Fosfomycin-tobramycin was synergistic and bactericidal for the isolates with mutations in the mexZ repressor gene, with a tobramycin MIC of 4 μg/ml. This effect was not observed in strains displaying tobramycin MICs of 1 to 2 μg/ml due to the strong bactericidal effect of tobramycin alone. Fosfomycin presented higher MPC values (range, 2,048 to >2,048 μg/ml) in aerobic and anaerobic conditions than did tobramycin (range, 16 to 256 μg/ml). Interestingly, the association rendered narrow or even null MSWs in the two conditions. However, for isolates with high-level tobramycin resistance that harbored aminoglycoside nucleotidyltransferases, time-kill assays showed no synergy, with wide MSWs in the two environments. glpT gene mutations responsible for fosfomycin resistance in P. aeruginosa were determined in fosfomycin-susceptible wild-type strains and mutant derivatives recovered from MPC studies. All mutant derivatives had changes in the GlpT amino acid sequence, which resulted in a truncated permease responsible for fosfomycin resistance. These results suggest that fosfomycin-tobramycin can be an alternative for infections due to P. aeruginosa since it has demonstrated synergistic and bactericidal activity in susceptible isolates and those with low-level tobramycin resistance. It also prevents the emergence of resistant mutants in either aerobic or anaerobic environments.
Collapse
|
820
|
Visualizing and quantifying Pseudomonas aeruginosa infection in the hindbrain ventricle of zebrafish using confocal laser scanning microscopy. J Microbiol Methods 2015; 117:85-94. [PMID: 26188283 DOI: 10.1016/j.mimet.2015.07.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 07/08/2015] [Accepted: 07/09/2015] [Indexed: 02/06/2023]
Abstract
Pseudomonas aeruginosa colonizes surfaces using a stepwise process that involves several phases, including attachment, production of exopolysaccharides, formation of microcolonies and the eventual development of biofilms. This process has been extensively characterized in vitro using both light and electron microscopic techniques. However, our ability to visualize this process in situ at the site of infection has been limited by the nature of the vertebrate models available. The optically clear zebrafish (Danio rerio) is an emerging model well suited for imaging bacterial infections. In this study, we infected the hindbrain ventricle of 54 h post-fertilization zebrafish with P. aeruginosa PAO1 and visualized and quantified microcolony formation using confocal laser scanning microscopy and image analyses. In comparison to wildtype PAO1, infection with a P. aeruginosa mutant deficient in the ability to produce the exopolysaccharide Psl caused less zebrafish mortality and fewer, smaller microcolonies per zebrafish at both 18 h and 29 h post-infection. The work presented here demonstrates reproducible in situ visualization and quantification methods for determining the extent of P. aeruginosa infection in a vertebrate model. We demonstrate how this model system can be manipulated to understand the effect of virulence factors on pathogenicity. Furthermore, this model can be adapted to study biofilm formation in situ, thereby extending our understanding of how bacterial persistence leads to chronic infections.
Collapse
|
821
|
Li Y, Bai F, Xia H, Zhuang L, Xu H, Jin Y, Zhang X, Bai Y, Qiao M. A novel regulator PA5022 (aefA) is involved in swimming motility, biofilm formation and elastase activity of Pseudomonas aeruginosa. Microbiol Res 2015; 176:14-20. [DOI: 10.1016/j.micres.2015.04.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 03/27/2015] [Accepted: 04/01/2015] [Indexed: 10/23/2022]
|
822
|
Abstract
Blood group antigens represent polymorphic traits inherited among individuals and populations. At present, there are 34 recognized human blood groups and hundreds of individual blood group antigens and alleles. Differences in blood group antigen expression can increase or decrease host susceptibility to many infections. Blood groups can play a direct role in infection by serving as receptors and/or coreceptors for microorganisms, parasites, and viruses. In addition, many blood group antigens facilitate intracellular uptake, signal transduction, or adhesion through the organization of membrane microdomains. Several blood groups can modify the innate immune response to infection. Several distinct phenotypes associated with increased host resistance to malaria are overrepresented in populations living in areas where malaria is endemic, as a result of evolutionary pressures. Microorganisms can also stimulate antibodies against blood group antigens, including ABO, T, and Kell. Finally, there is a symbiotic relationship between blood group expression and maturation of the gastrointestinal microbiome.
Collapse
Affiliation(s)
- Laura Cooling
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
823
|
The Frog Skin-Derived Antimicrobial Peptide Esculentin-1a(1-21)NH2 Promotes the Migration of Human HaCaT Keratinocytes in an EGF Receptor-Dependent Manner: A Novel Promoter of Human Skin Wound Healing? PLoS One 2015; 10:e0128663. [PMID: 26068861 PMCID: PMC4466536 DOI: 10.1371/journal.pone.0128663] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Accepted: 04/29/2015] [Indexed: 02/06/2023] Open
Abstract
One of the many functions of skin is to protect the organism against a wide range of pathogens. Antimicrobial peptides (AMPs) produced by the skin epithelium provide an effective chemical shield against microbial pathogens. However, whereas antibacterial/antifungal activities of AMPs have been extensively characterized, much less is known regarding their wound healing-modulatory properties. By using an in vitro re-epithelialisation assay employing special cell-culture inserts, we detected that a derivative of the frog-skin AMP esculentin-1a, named esculentin-1a(1-21)NH2, significantly stimulates migration of immortalized human keratinocytes (HaCaT cells) over a wide range of peptide concentrations (0.025–4 μM), and this notably more efficiently than human cathelicidin (LL-37). This activity is preserved in primary human epidermal keratinocytes. By using appropriate inhibitors and an enzyme-linked immunosorbent assay we found that the peptide-induced cell migration involves activation of the epidermal growth factor receptor and STAT3 protein. These results suggest that esculentin-1a(1-21)NH2 now deserves to be tested in standard wound healing assays as a novel candidate promoter of skin re-epithelialisation. The established ability of esculentin-1a(1-21)NH2 to kill microbes without harming mammalian cells, namely its high anti-Pseudomonal activity, makes this AMP a particularly attractive candidate wound healing promoter, especially in the management of chronic, often Pseudomonas-infected, skin ulcers.
Collapse
|
824
|
Jia J, Bai F, Jin Y, Santostefano KE, Ha UH, Wu D, Wu W, Terada N, Jin S. Efficient Gene Editing in Pluripotent Stem Cells by Bacterial Injection of Transcription Activator-Like Effector Nuclease Proteins. Stem Cells Transl Med 2015; 4:913-26. [PMID: 26062981 DOI: 10.5966/sctm.2015-0030] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Accepted: 04/27/2015] [Indexed: 11/16/2022] Open
Abstract
The type III secretion system (T3SS) of Pseudomonas aeruginosa is a powerful tool for direct protein delivery into mammalian cells and has successfully been used to deliver various exogenous proteins into mammalian cells. In the present study, transcription activator-like effector nuclease (TALEN) proteins have been efficiently delivered using the P. aeruginosa T3SS into mouse embryonic stem cells (mESCs), human ESCs (hESCs), and human induced pluripotent stem cells (hiPSCs) for genome editing. This bacterial delivery system offers an alternative method of TALEN delivery that is highly efficient in cleavage of the chromosomal target and presumably safer by avoiding plasmid DNA introduction. We combined the method of bacterial T3SS-mediated TALEN protein injection and transfection of an oligonucleotide template to effectively generate precise genetic modifications in the stem cells. Initially, we efficiently edited a single-base in the gfp gene of a mESC line to silence green fluorescent protein (GFP) production. The resulting GFP-negative mESC was cloned from a single cell and subsequently mutated back to a GFP-positive mESC line. Using the same approach, the gfp gene was also effectively knocked out in hESCs. In addition, a defined single-base edition was effectively introduced into the X-chromosome-linked HPRT1 gene in hiPSCs, generating an in vitro model of Lesch-Nyhan syndrome. T3SS-mediated TALEN protein delivery provides a highly efficient alternative for introducing precise gene editing within pluripotent stem cells for the purpose of disease genotype-phenotype relationship studies and cellular replacement therapies.
Collapse
Affiliation(s)
- Jingyue Jia
- State Key Laboratory of Medical and Chemical Biology, College of Life Sciences, Nankai University, Tianjin, People's Republic of China; Department of Molecular Genetics and Microbiology and Department of Pathology, University of Florida, Gainesville, Florida, USA; Department of Biotechnology and Bioinformatics, Korea University, Sejong, Republic of Korea; Key Laboratory of Regenerative Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China
| | - Fang Bai
- State Key Laboratory of Medical and Chemical Biology, College of Life Sciences, Nankai University, Tianjin, People's Republic of China; Department of Molecular Genetics and Microbiology and Department of Pathology, University of Florida, Gainesville, Florida, USA; Department of Biotechnology and Bioinformatics, Korea University, Sejong, Republic of Korea; Key Laboratory of Regenerative Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China
| | - Yongxin Jin
- State Key Laboratory of Medical and Chemical Biology, College of Life Sciences, Nankai University, Tianjin, People's Republic of China; Department of Molecular Genetics and Microbiology and Department of Pathology, University of Florida, Gainesville, Florida, USA; Department of Biotechnology and Bioinformatics, Korea University, Sejong, Republic of Korea; Key Laboratory of Regenerative Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China
| | - Katherine E Santostefano
- State Key Laboratory of Medical and Chemical Biology, College of Life Sciences, Nankai University, Tianjin, People's Republic of China; Department of Molecular Genetics and Microbiology and Department of Pathology, University of Florida, Gainesville, Florida, USA; Department of Biotechnology and Bioinformatics, Korea University, Sejong, Republic of Korea; Key Laboratory of Regenerative Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China
| | - Un-Hwan Ha
- State Key Laboratory of Medical and Chemical Biology, College of Life Sciences, Nankai University, Tianjin, People's Republic of China; Department of Molecular Genetics and Microbiology and Department of Pathology, University of Florida, Gainesville, Florida, USA; Department of Biotechnology and Bioinformatics, Korea University, Sejong, Republic of Korea; Key Laboratory of Regenerative Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China
| | - Donghai Wu
- State Key Laboratory of Medical and Chemical Biology, College of Life Sciences, Nankai University, Tianjin, People's Republic of China; Department of Molecular Genetics and Microbiology and Department of Pathology, University of Florida, Gainesville, Florida, USA; Department of Biotechnology and Bioinformatics, Korea University, Sejong, Republic of Korea; Key Laboratory of Regenerative Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China
| | - Weihui Wu
- State Key Laboratory of Medical and Chemical Biology, College of Life Sciences, Nankai University, Tianjin, People's Republic of China; Department of Molecular Genetics and Microbiology and Department of Pathology, University of Florida, Gainesville, Florida, USA; Department of Biotechnology and Bioinformatics, Korea University, Sejong, Republic of Korea; Key Laboratory of Regenerative Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China
| | - Naohiro Terada
- State Key Laboratory of Medical and Chemical Biology, College of Life Sciences, Nankai University, Tianjin, People's Republic of China; Department of Molecular Genetics and Microbiology and Department of Pathology, University of Florida, Gainesville, Florida, USA; Department of Biotechnology and Bioinformatics, Korea University, Sejong, Republic of Korea; Key Laboratory of Regenerative Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China
| | - Shouguang Jin
- State Key Laboratory of Medical and Chemical Biology, College of Life Sciences, Nankai University, Tianjin, People's Republic of China; Department of Molecular Genetics and Microbiology and Department of Pathology, University of Florida, Gainesville, Florida, USA; Department of Biotechnology and Bioinformatics, Korea University, Sejong, Republic of Korea; Key Laboratory of Regenerative Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China
| |
Collapse
|
825
|
Steele AD, Knouse KW, Keohane CE, Wuest WM. Total synthesis and biological investigation of (-)-promysalin. J Am Chem Soc 2015; 137:7314-7. [PMID: 26024439 DOI: 10.1021/jacs.5b04767] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Compounds that specifically target pathogenic bacteria are greatly needed, and identifying the method by which they act would provide new avenues of treatment. Herein we report the concise, high-yielding total synthesis (eight steps, 35% yield) of promysalin, a natural product that displays antivirulence phenotypes against pathogenic bacteria. Guided by bioinformatics, four diastereomers were synthesized, and the relative and absolute stereochemistries were confirmed by spectral and biological analysis. Finally, we show for the first time that promysalin displays two antivirulence phenotypes: the dispersion of mature biofilms and the inhibition of pyoverdine production, hinting at a unique pathogenic-specific mechanism of action.
Collapse
Affiliation(s)
- Andrew D Steele
- Department of Chemistry, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Kyle W Knouse
- Department of Chemistry, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Colleen E Keohane
- Department of Chemistry, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - William M Wuest
- Department of Chemistry, Temple University, Philadelphia, Pennsylvania 19122, United States
| |
Collapse
|
826
|
Abstract
Iron-sulfur clusters act as important cofactors for a number of transcriptional regulators in bacteria, including many mammalian pathogens. The sensitivity of iron-sulfur clusters to iron availability, oxygen tension, and reactive oxygen and nitrogen species enables bacteria to use such regulators to adapt their gene expression profiles rapidly in response to changing environmental conditions. In this review, we discuss how the [4Fe-4S] or [2Fe-2S] cluster-containing regulators FNR, Wbl, aconitase, IscR, NsrR, SoxR, and AirSR contribute to bacterial pathogenesis through control of both metabolism and classical virulence factors. In addition, we briefly review mammalian iron homeostasis as well as oxidative/nitrosative stress to provide context for understanding the function of bacterial iron-sulfur cluster sensors in different niches within the host.
Collapse
Affiliation(s)
- Halie K Miller
- Department of Microbiology and Environmental Toxicology, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA.
| | | |
Collapse
|
827
|
Danis-Wlodarczyk K, Olszak T, Arabski M, Wasik S, Majkowska-Skrobek G, Augustyniak D, Gula G, Briers Y, Jang HB, Vandenheuvel D, Duda KA, Lavigne R, Drulis-Kawa Z. Characterization of the Newly Isolated Lytic Bacteriophages KTN6 and KT28 and Their Efficacy against Pseudomonas aeruginosa Biofilm. PLoS One 2015; 10:e0127603. [PMID: 25996839 PMCID: PMC4440721 DOI: 10.1371/journal.pone.0127603] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 04/16/2015] [Indexed: 11/18/2022] Open
Abstract
We here describe two novel lytic phages, KT28 and KTN6, infecting Pseudomonas aeruginosa, isolated from a sewage sample from an irrigated field near Wroclaw, in Poland. Both viruses show characteristic features of Pbunalikevirus genus within the Myoviridae family with respect to shape and size of head/tail, as well as LPS host receptor recognition. Genome analysis confirmed the similarity to other PB1-related phages, ranging between 48 and 96%. Pseudomonas phage KT28 has a genome size of 66,381 bp and KTN6 of 65,994 bp. The latent period, burst size, stability and host range was determined for both viruses under standard laboratory conditions. Biofilm eradication efficacy was tested on peg-lid plate assay and PET membrane surface. Significant reduction of colony forming units was observed (70-90%) in 24 h to 72 h old Pseudomonas aeruginosa PAO1 biofilm cultures for both phages. Furthermore, a pyocyanin and pyoverdin reduction tests reveal that tested phages lowers the amount of both secreted dyes in 48-72 h old biofilms. Diffusion and goniometry experiments revealed the increase of diffusion rate through the biofilm matrix after phage application. These characteristics indicate these phages could be used to prevent Pseudomonas aeruginosa infections and biofilm formation. It was also shown, that PB1-related phage treatment of biofilm caused the emergence of stable phage-resistant mutants growing as small colony variants.
Collapse
Affiliation(s)
- Katarzyna Danis-Wlodarczyk
- Department of Pathogen Biology and Immunology, Institute of Genetics and Microbiology, University of Wroclaw, Wroclaw, Poland
- Division of Gene Technology, Catholic University of Leuven, Leuven, Belgium
| | - Tomasz Olszak
- Department of Pathogen Biology and Immunology, Institute of Genetics and Microbiology, University of Wroclaw, Wroclaw, Poland
| | - Michal Arabski
- Department of Microbiology, Institute of Biology, The Jan Kochanowski University in Kielce, Kielce, Poland
| | - Slawomir Wasik
- Department of Molecular Physics, Institute of Physics, The Jan Kochanowski University in Kielce, Kielce, Poland
| | - Grazyna Majkowska-Skrobek
- Department of Pathogen Biology and Immunology, Institute of Genetics and Microbiology, University of Wroclaw, Wroclaw, Poland
| | - Daria Augustyniak
- Department of Pathogen Biology and Immunology, Institute of Genetics and Microbiology, University of Wroclaw, Wroclaw, Poland
| | - Grzegorz Gula
- Department of Pathogen Biology and Immunology, Institute of Genetics and Microbiology, University of Wroclaw, Wroclaw, Poland
| | - Yves Briers
- Division of Gene Technology, Catholic University of Leuven, Leuven, Belgium
| | - Ho Bin Jang
- Division of Gene Technology, Catholic University of Leuven, Leuven, Belgium
| | | | - Katarzyna Anna Duda
- Division of Structural Biochemistry, Research Center Borstel, Leibniz-Center for Medicine and Biosciences, Borstel, Germany
| | - Rob Lavigne
- Division of Gene Technology, Catholic University of Leuven, Leuven, Belgium
| | - Zuzanna Drulis-Kawa
- Department of Pathogen Biology and Immunology, Institute of Genetics and Microbiology, University of Wroclaw, Wroclaw, Poland
- * E-mail:
| |
Collapse
|
828
|
Sharma K, Bhattacharyya D. Immunoglobulin isotype isolated from human placental extract does not interfere in complement-mediated bacterial opsonization within the wound milieu. FEBS Open Bio 2015; 5:369-77. [PMID: 25984442 PMCID: PMC4431336 DOI: 10.1016/j.fob.2015.04.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 04/08/2015] [Accepted: 04/10/2015] [Indexed: 11/29/2022] Open
Abstract
Human placental extract has many applications as a wound healer. Immunoglobulin G is a key glycoprotein present in human placental extract. Placental IgG (25.2 ± 3.97 μg/ml) did not exert anti-complementary effects.
The wound healing potency of an aqueous extract of placenta can be evaluated through the presence of numerous regulatory components. The presence of glycans was detected by thin layer chromatography and fluorophore-assisted carbohydrate electrophoresis. Mass spectrometric analysis revealed the existence of multiple fragments of immunoglobulin G (IgG). IgG was present in the extract at a concentration of 25.2 ± 3.97 μg/ml. IgG possesses anti-complementary activity by diverting the complement activation from target surface. Thus, effect of placental IgG on complement–bacteria interaction was investigated through classical and alternative pathway and the preparation was ascertained to be safe with respect to their interference in the process of bacterial opsonization.
Collapse
Key Words
- ANTS, 8-aminonaphthalene-1,3,6-trisulfonate
- ATP, adenosine triphosphate
- BCIP, 5-Bromo 4-Chloro 3′ indolylphosphate
- BHI, Brain–Heart Infusion
- BSA, bovine serum albumin
- CNBr, cyanogens bromide
- Complement
- EDTA, ethylenediamine tetra acetic acid
- EGTA, ethylene glycol tetra acetic acid
- G6PDH, glucose-6-phosphate dehydrogenase
- Human placental extract
- IgG, immunoglobulin G
- Immunoglobulin
- NAD+, nicotinamide adenine dinucleotide
- NBT, nitro-blue tetrazolium chloride
- PNGase F, peptide N-glycosidase F
- Pseudomonas aeruginosa wound physiology
Collapse
Affiliation(s)
- Kanika Sharma
- Division of Structural Biology and Bioinformatics, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C.Mullick Road, Jadavpur, Kolkata 7000032, India
| | - Debasish Bhattacharyya
- Division of Structural Biology and Bioinformatics, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C.Mullick Road, Jadavpur, Kolkata 7000032, India
| |
Collapse
|
829
|
Grosso-Becera MV, Servín-González L, Soberón-Chávez G. RNA structures are involved in the thermoregulation of bacterial virulence-associated traits. Trends Microbiol 2015; 23:509-18. [PMID: 25999019 DOI: 10.1016/j.tim.2015.04.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 04/01/2015] [Accepted: 04/16/2015] [Indexed: 11/25/2022]
Abstract
Pathogenic bacteria are exposed to temperature changes during colonization of the human body and during exposure to environmental conditions. Virulence-associated traits are mainly expressed by pathogenic bacteria at 37°C. We review different cases of post-transcriptional regulation of virulence-associated proteins through RNA structures (called RNA thermometers or RNATs) that modulate the translation of mRNAs. The analysis of RNATs in pathogenic bacteria has started to produce a comprehensive picture of the structures involved, and of the genes regulated by this mechanism. However, we are still not able to predict the functionality of putative RNATs predicted by bioinformatics methods, and there is not a global approach to measure the effect of these RNA structures in gene regulation during bacterial infections.
Collapse
Affiliation(s)
- María Victoria Grosso-Becera
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones, Biomédicas, Universidad Nacional Autónoma de México, Tercer Circuito Escolar, Apartado Postal 70228, DF, México
| | - Luis Servín-González
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones, Biomédicas, Universidad Nacional Autónoma de México, Tercer Circuito Escolar, Apartado Postal 70228, DF, México
| | - Gloria Soberón-Chávez
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones, Biomédicas, Universidad Nacional Autónoma de México, Tercer Circuito Escolar, Apartado Postal 70228, DF, México.
| |
Collapse
|
830
|
Dubern JF, Cigana C, De Simone M, Lazenby J, Juhas M, Schwager S, Bianconi I, Döring G, Eberl L, Williams P, Bragonzi A, Cámara M. Integrated whole-genome screening for Pseudomonas aeruginosa virulence genes using multiple disease models reveals that pathogenicity is host specific. Environ Microbiol 2015; 17:4379-93. [PMID: 25845292 PMCID: PMC4676916 DOI: 10.1111/1462-2920.12863] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 03/19/2015] [Accepted: 03/31/2015] [Indexed: 11/28/2022]
Abstract
Pseudomonas aeruginosa is a multi-host opportunistic pathogen causing a wide range of diseases because of the armoury of virulence factors it produces, and it is difficult to eradicate because of its intrinsic resistance to antibiotics. Using an integrated whole-genome approach, we searched for P. aeruginosa virulence genes with multi-host relevance. We constructed a random library of 57 360 Tn5 mutants in P. aeruginosa PAO1-L and screened it in vitro for those showing pleiotropic effects in virulence phenotypes (reduced swarming, exo-protease and pyocyanin production). A set of these pleiotropic mutants were assayed for reduced toxicity in Drosophila melanogaster, Caenorhabditis elegans, human cell lines and mice. Surprisingly, the screening revealed that the virulence of the majority of P. aeruginosa mutants varied between disease models, suggesting that virulence is dependent on the disease model used and hence the host environment. Genomic analysis revealed that these virulence-related genes encoded proteins from almost all functional classes, which were conserved among P. aeruginosa strains. Thus, we provide strong evidence that although P. aeruginosa is capable of infecting a wide range of hosts, many of its virulence determinants are host specific. These findings have important implication when searching for novel anti-virulence targets to develop new treatments against P. aeruginosa.
Collapse
Affiliation(s)
- Jean-Frédéric Dubern
- School of Life Sciences, Centre for Biomolecular Sciences, University of Nottingham, Nottingham, UK
| | - Cristina Cigana
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS, San Raffaele Scientific Institute, Milano, Italy
| | - Maura De Simone
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS, San Raffaele Scientific Institute, Milano, Italy
| | - James Lazenby
- School of Life Sciences, Centre for Biomolecular Sciences, University of Nottingham, Nottingham, UK
| | - Mario Juhas
- Department of Microbiology, Institute of Plant Biology, University of Zürich, Zürich, Switzerland
| | - Stephan Schwager
- Department of Microbiology, Institute of Plant Biology, University of Zürich, Zürich, Switzerland
| | - Irene Bianconi
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS, San Raffaele Scientific Institute, Milano, Italy
| | - Gerd Döring
- Institute of Medical Microbiology and Hygiene, University of Tübingen, Tübingen, Germany
| | - Leo Eberl
- Department of Microbiology, Institute of Plant Biology, University of Zürich, Zürich, Switzerland
| | - Paul Williams
- School of Life Sciences, Centre for Biomolecular Sciences, University of Nottingham, Nottingham, UK
| | - Alessandra Bragonzi
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS, San Raffaele Scientific Institute, Milano, Italy
| | - Miguel Cámara
- School of Life Sciences, Centre for Biomolecular Sciences, University of Nottingham, Nottingham, UK
| |
Collapse
|
831
|
Drees SL, Fetzner S. PqsE of Pseudomonas aeruginosa Acts as Pathway-Specific Thioesterase in the Biosynthesis of Alkylquinolone Signaling Molecules. ACTA ACUST UNITED AC 2015; 22:611-8. [PMID: 25960261 DOI: 10.1016/j.chembiol.2015.04.012] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 04/02/2015] [Accepted: 04/07/2015] [Indexed: 01/18/2023]
Abstract
Pseudomonas aeruginosa uses the alkylquinolones PQS (2-heptyl-3-hydroxy-4(1H)-quinolone) and HHQ (2-heptyl-4(1H)-quinolone) as quorum-sensing signal molecules, controlling the expression of many virulence genes as a function of cell population density. The biosynthesis of HHQ is generally accepted to require the pqsABCD gene products. We now reconstitute the biosynthetic pathway in vitro, and demonstrate that in addition to PqsABCD, PqsE has a role in HHQ synthesis. PqsE acts as thioesterase, hydrolyzing the biosynthetic intermediate 2-aminobenzoylacetyl-coenzyme A to form 2-aminobenzoylacetate, the precursor of HHQ and 2-aminoacetophenone. The role of PqsE can be taken over to some extent by the broad-specificity thioesterase TesB, explaining why the pqsE deletion mutant of P. aeruginosa still synthesizes HHQ. Interestingly, the pqsE mutant produces increased levels of 2,4-dihydroxyquinoline, resulting from intramolecular cyclization of 2-aminobenzoylacetyl-coenzyme A. Overall, our data suggest that PqsE promotes the efficiency of alkylquinolone signal molecule biosynthesis in P. aeruginosa and balances the levels of secondary metabolites deriving from the alkylquinolone biosynthetic pathway.
Collapse
Affiliation(s)
- Steffen Lorenz Drees
- Institute of Molecular Microbiology and Biotechnology, University of Münster, Corrensstrasse 3, 48149 Münster, Germany
| | - Susanne Fetzner
- Institute of Molecular Microbiology and Biotechnology, University of Münster, Corrensstrasse 3, 48149 Münster, Germany.
| |
Collapse
|
832
|
Marafino JN, Gallagher TM, Barragan J, Volkers BL, LaDow JE, Bonifer K, Fitzgerald G, Floyd JL, McKenna K, Minahan NT, Walsh B, Seifert K, Caran KL. Colloidal and antibacterial properties of novel triple-headed, double-tailed amphiphiles: exploring structure-activity relationships and synergistic mixtures. Bioorg Med Chem 2015; 23:3566-73. [PMID: 25936261 DOI: 10.1016/j.bmc.2015.04.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 04/01/2015] [Accepted: 04/09/2015] [Indexed: 01/03/2023]
Abstract
Two novel series of tris-cationic, tripled-headed, double-tailed amphiphiles were synthesized and the effects of tail length and head group composition on the critical aggregation concentration (CAC), thermodynamic parameters, and minimum inhibitory concentration (MIC) against six bacterial strains were investigated. Synergistic antibacterial combinations of these amphiphiles were also identified. Amphiphiles in this study are composed of a benzene core with three benzylic ammonium bromide groups, two of which have alkyl chains, each 8-16 carbons in length. The third head group is a trimethylammonium or pyridinium. Log of critical aggregation concentration (log[CAC]) and heat of aggregation (ΔHagg) were both inversely proportional to the length of the linear hydrocarbon chains. Antibacterial activity increases with tail length until an optimal tail length of 12 carbons per chain, above which, activity decreased. The derivatives with two 12 carbon chains had the best antibacterial activity, killing all tested strains at concentrations of 1-2μM for Gram-positive and 4-16μM for Gram-negative bacteria. The identity of the third head group (trimethylammonium or pyridinium) had minimal effect on colloidal and antibacterial activity. The antibacterial activity of several binary combinations of amphiphiles from this study was higher than activity of individual amphiphiles, indicating that these combinations are synergistic. These amphiphiles show promise as novel antibacterial agents that could be used in a variety of applications.
Collapse
Affiliation(s)
- John N Marafino
- James Madison University, Department of Biology, 951 Carrier Drive, MSC 7801, Harrisonburg, VA 22807, USA; James Madison University, Department of Chemistry and Biochemistry, 901 Carrier Drive, MSC 4501, Harrisonburg, VA 22807, USA
| | - Tara M Gallagher
- James Madison University, Department of Biology, 951 Carrier Drive, MSC 7801, Harrisonburg, VA 22807, USA
| | - Jhosdyn Barragan
- James Madison University, Department of Chemistry and Biochemistry, 901 Carrier Drive, MSC 4501, Harrisonburg, VA 22807, USA
| | - Brandi L Volkers
- James Madison University, Department of Biology, 951 Carrier Drive, MSC 7801, Harrisonburg, VA 22807, USA
| | - Jade E LaDow
- James Madison University, Department of Biology, 951 Carrier Drive, MSC 7801, Harrisonburg, VA 22807, USA
| | - Kyle Bonifer
- James Madison University, Department of Biology, 951 Carrier Drive, MSC 7801, Harrisonburg, VA 22807, USA
| | - Gabriel Fitzgerald
- James Madison University, Department of Chemistry and Biochemistry, 901 Carrier Drive, MSC 4501, Harrisonburg, VA 22807, USA
| | - Jason L Floyd
- James Madison University, Department of Biology, 951 Carrier Drive, MSC 7801, Harrisonburg, VA 22807, USA
| | - Kristin McKenna
- James Madison University, Department of Chemistry and Biochemistry, 901 Carrier Drive, MSC 4501, Harrisonburg, VA 22807, USA
| | - Nicholas T Minahan
- James Madison University, Department of Biology, 951 Carrier Drive, MSC 7801, Harrisonburg, VA 22807, USA
| | - Brenna Walsh
- James Madison University, Department of Chemistry and Biochemistry, 901 Carrier Drive, MSC 4501, Harrisonburg, VA 22807, USA
| | - Kyle Seifert
- James Madison University, Department of Biology, 951 Carrier Drive, MSC 7801, Harrisonburg, VA 22807, USA.
| | - Kevin L Caran
- James Madison University, Department of Chemistry and Biochemistry, 901 Carrier Drive, MSC 4501, Harrisonburg, VA 22807, USA.
| |
Collapse
|
833
|
Miller LC, O'Loughlin CT, Zhang Z, Siryaporn A, Silpe JE, Bassler BL, Semmelhack MF. Development of potent inhibitors of pyocyanin production in Pseudomonas aeruginosa. J Med Chem 2015; 58:1298-306. [PMID: 25597392 DOI: 10.1021/jm5015082] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The development of new approaches for the treatment of antimicrobial-resistant infections is an urgent public health priority. The Pseudomonas aeruginosa pathogen, in particular, is a leading source of infection in hospital settings, with few available treatment options. In the context of an effort to develop antivirulence strategies to combat bacterial infection, we identified a series of highly effective small molecules that inhibit the production of pyocyanin, a redox-active virulence factor produced by P. aeruginosa. Interestingly, these new antagonists appear to suppress P. aeruginosa virulence factor production through a pathway that is independent of LasR and RhlR.
Collapse
Affiliation(s)
- Laura C Miller
- Department of Chemistry and ‡Department of Molecular Biology, Princeton University , Washington Road, Princeton, New Jersey 08544, United States
| | | | | | | | | | | | | |
Collapse
|
834
|
Morita Y, Tomida J, Kawamura Y. Efflux-mediated fluoroquinolone resistance in the multidrug-resistant Pseudomonas aeruginosa clinical isolate PA7: identification of a novel MexS variant involved in upregulation of the mexEF-oprN multidrug efflux operon. Front Microbiol 2015; 6:8. [PMID: 25653649 PMCID: PMC4301020 DOI: 10.3389/fmicb.2015.00008] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 01/05/2015] [Indexed: 11/24/2022] Open
Abstract
The emergence of multidrug-resistant Pseudomonas aeruginosa has become a serious problem in medical settings. P. aeruginosa clinical isolate PA7 is resistant to fluoroquinolones, aminoglycosides, and most β-lactams but not imipenem. In this study, enhanced efflux-mediated fluoroquinolone resistance of PA7 was shown to reflect increased expression of two resistance nodulation cell division (RND) -type multidrug efflux operons, mexEF-oprN and mexXY-oprA. Such a clinical isolate has rarely been reported because MexEF-OprN-overproducing mutants often increase susceptibility to aminoglycosides apparently owing to impairment of the MexXY system. A mutant of PA7 lacking three RND-type multidrug efflux operons (mexAB-oprM, mexEF-oprN, and mexXY-oprA) was susceptible to all anti-pseudomonas agents we tested, supporting an idea that these RND-type multidrug efflux transporters are molecular targets to overcome multidrug resistance in P. aeruginosa. mexEF-oprN-upregulation in P. aeruginosa PA7 was shown due to a MexS variant harboring the Valine-155 amino acid residue. This is the first genetic evidence shown that a MexS variant causes mexEF-oprN-upregulation in P. aeruginosa clinical isolates.
Collapse
Affiliation(s)
- Yuji Morita
- Department of Microbiology, School of Pharmacy, Aichi Gakuin University Nagoya, Japan
| | - Junko Tomida
- Department of Microbiology, School of Pharmacy, Aichi Gakuin University Nagoya, Japan
| | - Yoshiaki Kawamura
- Department of Microbiology, School of Pharmacy, Aichi Gakuin University Nagoya, Japan
| |
Collapse
|
835
|
Chavshin AR, Oshaghi MA, Vatandoost H, Yakhchali B, Zarenejad F, Terenius O. Malpighian tubules are important determinants of Pseudomonas transstadial transmission and longtime persistence in Anopheles stephensi. Parasit Vectors 2015; 8:36. [PMID: 25604581 PMCID: PMC4307889 DOI: 10.1186/s13071-015-0635-6] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 01/02/2015] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Pseudomonas is a genus of bacteria commonly found in investigations of gut microbes in malaria mosquitoes. Among those mosquitoes is the dominating malaria vector in Asia, Anopheles stephensi, where Pseudomonas is a prevailing bacterium and natural inhabitant of its breeding places. In order to explore the reason for finding Pseudomonas so frequently, an investigation of its localization and transstadial properties was undertaken. METHODS A Pseudomonas isolate from An. stephensi was transformed successfully with an endogenous plasmid modified to express green fluorescent protein (GFP). Subsequently, the Pseudomonas-GFP was added to the laboratory larval breeding place of An. stephensi and taken up by the larvae. After 24 hours, the larvae were cleaned and moved to a bath with double-distilled water. Also, female adults were fed sugar solution containing Pseudomonas-GFP. The Pseudomonas-GFP was traced in the alimentary canal of larvae, pupae and adults. RESULTS Fluorescent microscopy and PCR assays showed that the Pseudomonas bacteria underwent transstadial transmission from larvae to pupae and then to adults. In blood-fed female mosquitoes, the bacteria increased in numbers and remained in the mosquito body for at least three weeks after eclosion. In addition to the midgut, the Malpighian tubules of both larvae and adult mosquitoes were colonized by the bacteria. Also Pseudomonas-GFP that was distributed through sugar solution was able to colonize the Malpighian tubules of adult females. CONCLUSIONS Colonization of the Malpighian tubules by Pseudomonas bacteria seems to be important for the transstadial passage from larvae to adult and presumably for the longevity of the bacteria in the adult mosquito. The existence of an entry point in the larval stage, and the long duration in the female gut, opens up for a possible use of Pseudomonas in mosquito paratransgenesis.
Collapse
Affiliation(s)
- Ali Reza Chavshin
- Social Determinants of Health, Research Center, Urmia University of Medical Sciences (UMSU), Urmia, Iran.
- Department of Medical Entomology and Vector Control, School of Public Health, Urmia University of Medical Sciences (UMSU), Urmia, Iran.
| | - Mohammad Ali Oshaghi
- Department of Medical Entomology and Vector Control, School of Public Health, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| | - Hasan Vatandoost
- Department of Medical Entomology and Vector Control, School of Public Health, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
- Institute for Environmental Research (IER), Tehran, Iran.
| | - Bagher Yakhchali
- Department of Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran.
| | - Fahimeh Zarenejad
- Department of Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran.
| | - Olle Terenius
- Department of Ecology, Swedish University of Agricultural Sciences (SLU), Uppsala, Sweden.
| |
Collapse
|
836
|
Abstract
Bacterial biofilms infect 2-4% of medical devices upon implantation, resulting in multiple surgeries and increased recovery time due to the very great increase in antibiotic resistance in the biofilm phenotype. This work investigates the feasibility of thermal mitigation of biofilms at physiologically accessible temperatures. Pseudomonas aeruginosa biofilms were cultured to high bacterial density (1.7 × 10(9) CFU cm(-2)) and subjected to thermal shocks ranging from 50°C to 80°C for durations of 1-30 min. The decrease in viable bacteria was closely correlated with an Arrhenius temperature dependence and Weibull-style time dependence, demonstrating up to six orders of magnitude reduction in bacterial load. The bacterial load for films with more conventional initial bacterial densities dropped below quantifiable levels, indicating thermal mitigation as a viable approach to biofilm control.
Collapse
Affiliation(s)
- Ann O’Toole
- Department of Chemical and Biochemical Engineering 4133 Seamans Center for the Engineering Arts & Sciences University of Iowa Iowa City, IA 52242, U.S.A
| | - Erica B. Ricker
- Department of Chemical and Biochemical Engineering 4133 Seamans Center for the Engineering Arts & Sciences University of Iowa Iowa City, IA 52242, U.S.A
| | - Eric Nuxoll
- Department of Chemical and Biochemical Engineering 4133 Seamans Center for the Engineering Arts & Sciences University of Iowa Iowa City, IA 52242, U.S.A
| |
Collapse
|
837
|
Dudnik A, Dudler R. Virulence determinants of Pseudomonas syringae strains isolated from grasses in the context of a small type III effector repertoire. BMC Microbiol 2014; 14:304. [PMID: 25472590 PMCID: PMC4262972 DOI: 10.1186/s12866-014-0304-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 11/20/2014] [Indexed: 11/10/2022] Open
Abstract
Background Pseudomonas syringae is pathogenic to a large number of plant species. For host colonization and disease progression, strains of this bacterium utilize an array of type III-secreted effectors and other virulence factors, including small secreted molecules such as syringolin A, a peptide derivative that inhibits the eukaryotic proteasome. In strains colonizing dicotyledonous plants, the compound was demonstrated to suppress the salicylic-acid-dependent defense pathway. Here, we analyze virulence factors of three strains colonizing wheat (Triticum aestivum): P. syringae pathovar syringae (Psy) strains B64 and SM, as well as P. syringae BRIP34876. These strains have a relatively small repertoire of only seven to eleven type III secreted effectors (T3Es) and differ in their capacity to produce syringolin A. The aim of this study was to analyze the contribution of various known virulence factors in the context of a small T3E repertoire. Results We demonstrate that syringolin A production enhances disease symptom development upon direct infiltration of strains into wheat leaves. However, it is not universally required for colonization, as Psy SM, which lacks syringolin biosynthesis genes, reaches cell densities comparable to syringolin A producer P. syringae BRIP34876. Next, we show that despite the small set of T3E-encoding genes, the type III secretion system remains the key pathogenicity determinant in these strains, and that phenotypic effects of deleting T3E-coding genes become apparent only when multiple effectors are removed. Conclusions Whereas production of syringolin A is not required for successful colonization of wheat leaves by P. syringae strains, its production results in increased lesion formation. Despite the small number of known T3Es encoded by the analyzed strains, the type III secretion system is essential for endophytic growth of these strains. Electronic supplementary material The online version of this article (doi:10.1186/s12866-014-0304-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alexey Dudnik
- Institute of Plant Biology, University of Zurich, Zollikerstrasse 107, Zurich, 8008, Switzerland. .,Present address: Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kogle Allé 4, Hørsholm, 2970, Denmark.
| | - Robert Dudler
- Institute of Plant Biology, University of Zurich, Zollikerstrasse 107, Zurich, 8008, Switzerland.
| |
Collapse
|
838
|
Grimwood K, Kyd JM, Owen SJ, Massa HM, Cripps AW. Vaccination against respiratory Pseudomonas aeruginosa infection. Hum Vaccin Immunother 2014; 11:14-20. [PMID: 25483510 PMCID: PMC4514401 DOI: 10.4161/hv.34296] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Respiratory infections caused by Pseudomonas aeruginosa are a major clinical problem globally, particularly for patients with chronic pulmonary disorders, such as those with cystic fibrosis (CF), non-CF bronchiectasis (nCFB) and severe chronic obstructive pulmonary disease (COPD). In addition, critically ill and immunocompromised patients are also at significant risk of P. aeruginosa infection. For almost half a century, research efforts have focused toward development of a vaccine against infections caused by P. aeruginosa, but a licensed vaccine is not yet available. Significant advances in identifying potential vaccine antigens have been made. Immunisations via both the mucosal and systemic routes have been trialled in animal models and their effectiveness in clearing acute infections demonstrated. The challenge for translation of this research to human applications remains, since P. aeruginosa infections in the human respiratory tract can present both as an acute or chronic infection. In addition, immunisation prior to infection may not be possible for many patients with CF, nCFB or COPD. Therefore, development of a therapeutic vaccine provides an alternative approach for treatment of chronic infection. Preliminary animal and human studies suggest that mucosal immunisation may be effective as a therapeutic vaccine against P. aeruginosa respiratory infections. Nevertheless, more research is needed to improve our understanding of the basic biology of P. aeruginosa and the mechanisms needed to upregulate the induction of host immune pathways to prevent infection. Recognition of variability in the host immune responses for a range of patient health conditions at risk from P. aeruginosa infection is also required to support development of a successful vaccine delivery strategy and vaccine. Activation of mucosal immune responses may provide improved efficacy of vaccination for P. aeruginosa during both acute exacerbations and chronic infection.
Collapse
Affiliation(s)
- Keith Grimwood
- a School of Medicine; Griffith University; Gold Coast, Queensland Australia
| | | | | | | | | |
Collapse
|
839
|
Draft Genome Assembly of Pseudomonas aeruginosa Quality Control Reference Strain Boston 41501. GENOME ANNOUNCEMENTS 2014; 2:2/5/e00960-14. [PMID: 25278526 PMCID: PMC4183870 DOI: 10.1128/genomea.00960-14] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
We present the scaffolded genome assembly of Pseudomonas aeruginosa Boston 41501, now publicly available in GenBank (JOVK00000000) in 10 contigs placed into a single scaffold. The 6.82-Mbp genome contains 66.1% G+C content and 6,295 coding sequences, including type 4 pilus and type 3 secretion system production genes.
Collapse
|
840
|
Liposomal antibiotic formulations for targeting the lungs in the treatment of Pseudomonas aeruginosa. Ther Deliv 2014; 5:409-27. [PMID: 24856168 DOI: 10.4155/tde.14.13] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Pseudomonas aeruginosa is a Gram-negative bacterium that causes serious lung infections in cystic fibrosis, non-cystic fibrosis bronchiectasis, immunocompromised, and mechanically ventilated patients. The arsenal of conventional antipseudomonal antibiotic drugs include the extended-spectrum penicillins, cephalosporins, carbapenems, monobactams, polymyxins, fluoroquinolones, and aminoglycosides but their toxicity and/or increasing antibiotic resistance are of particular concern. Improvement of existing therapies against Pseudomonas aeruginosa infections involves the use of liposomes - artificial phospholipid vesicles that are biocompatible, biodegradable, and nontoxic and able to entrap and carry hydrophilic, hydrophobic, and amphiphilic molecules to the site of action. The goal of developing liposomal antibiotic formulations is to improve their therapeutic efficacy by reducing drug toxicity and/or by enhancing the delivery and retention of antibiotics at the site of infection. The focus of this review is to appraise the current progress of the development and application of liposomal antibiotic delivery systems for the treatment pulmonary infections caused by P. aeruginosa.
Collapse
|
841
|
De Simone M, Spagnuolo L, Lorè NI, Rossi G, Cigana C, De Fino I, Iraqi FA, Bragonzi A. Host genetic background influences the response to the opportunistic Pseudomonas aeruginosa infection altering cell-mediated immunity and bacterial replication. PLoS One 2014; 9:e106873. [PMID: 25268734 PMCID: PMC4182038 DOI: 10.1371/journal.pone.0106873] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 08/02/2014] [Indexed: 12/22/2022] Open
Abstract
Pseudomonas aeruginosa is a common cause of healthcare-associated infections including pneumonia, bloodstream, urinary tract, and surgical site infections. The clinical outcome of P. aeruginosa infections may be extremely variable among individuals at risk and patients affected by cystic fibrosis. However, risk factors for P. aeruginosa infection remain largely unknown. To identify and track the host factors influencing P. aeruginosa lung infections, inbred immunocompetent mouse strains were screened in a pneumonia model system. A/J, BALB/cJ, BALB/cAnNCrl, BALB/cByJ, C3H/HeOuJ, C57BL/6J, C57BL/6NCrl, DBA/2J, and 129S2/SvPasCRL mice were infected with P. aeruginosa clinical strain and monitored for body weight and mortality up to seven days. The most deviant survival phenotypes were observed for A/J, 129S2/SvPasCRL and DBA/2J showing high susceptibility while BALB/cAnNCrl and C3H/HeOuJ showing more resistance to P. aeruginosa infection. Next, one of the most susceptible and resistant mouse strains were characterized for their deviant clinical and immunological phenotype by scoring bacterial count, cell-mediated immunity, cytokines and chemokines profile and lung pathology in an early time course. Susceptible A/J mice showed significantly higher bacterial burden, higher cytokines and chemokines levels but lower leukocyte recruitment, particularly neutrophils, when compared to C3H/HeOuJ resistant mice. Pathologic scores showed lower inflammatory severity, reduced intraluminal and interstitial inflammation extent, bronchial and parenchymal involvement and diminished alveolar damage in the lungs of A/J when compared to C3H/HeOuJ. Our findings indicate that during an early phase of infection a prompt inflammatory response in the airways set the conditions for a non-permissive environment to P. aeruginosa replication and lock the spread to other organs. Host gene(s) may have a role in the reduction of cell-mediated immunity playing a critical role in the control of P. aeruginosa infection. These results now provide a basis for mapping genomic regions underlying host susceptibility to P. aeruginosa infection.
Collapse
Affiliation(s)
- Maura De Simone
- Infection and Cystic Fibrosis Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Lorenza Spagnuolo
- Infection and Cystic Fibrosis Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Nicola Ivan Lorè
- Infection and Cystic Fibrosis Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Giacomo Rossi
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Cristina Cigana
- Infection and Cystic Fibrosis Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Ida De Fino
- Infection and Cystic Fibrosis Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Fuad A. Iraqi
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Alessandra Bragonzi
- Infection and Cystic Fibrosis Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy
| |
Collapse
|
842
|
Mendis N, Lin YR, Faucher SP. Comparison of virulence properties of Pseudomonas aeruginosa exposed to water and grown in rich broth. Can J Microbiol 2014; 60:777-81. [PMID: 25352257 DOI: 10.1139/cjm-2014-0519] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen that can infect susceptible patients suffering from cystic fibrosis, immunosuppression, and severe burns. Nosocomial- and community-acquired infection is likely due to contact with water sources contaminated with P. aeruginosa. Most of what is known about the virulence properties of P. aeruginosa was derived from studies using fairly rich broths, which do not represent conditions found in water, such as low nutrient concentrations. Here, we compare biofilm production, invasion of epithelial cells, cytotoxicity, and pyocyanin production of P. aeruginosa in water with P. aeruginosa grown in rich broth. Since tap water is variable, we used a defined water medium, Fraquil, to ensure reproducibility of the results. We found that P. aeruginosa does not readily form biofilm in Fraquil. Pseudomonas aeruginosa is equally able to attach to and invade epithelial cells but is more cytotoxic after incubation in water for 30 days than when it is grown in rich broth. Moreover, P. aeruginosa produces less pyocyanin when exposed to water. Our results show that P. aeruginosa seems to have different properties when exposed to water than when grown in rich broth.
Collapse
Affiliation(s)
- Nilmini Mendis
- Department of Natural Resource Sciences, Faculty of Agricultural and Environmental Sciences, Macdonald Campus, McGill University, 21 111 Lakeshore Road, Ste-Anne-de-Bellevue, QC H9X 3V9, Canada
| | | | | |
Collapse
|
843
|
Van den Bossche A, Ceyssens PJ, De Smet J, Hendrix H, Bellon H, Leimer N, Wagemans J, Delattre AS, Cenens W, Aertsen A, Landuyt B, Minakhin L, Severinov K, Noben JP, Lavigne R. Systematic identification of hypothetical bacteriophage proteins targeting key protein complexes of Pseudomonas aeruginosa. J Proteome Res 2014; 13:4446-56. [PMID: 25185497 DOI: 10.1021/pr500796n] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Addressing the functionality of predicted genes remains an enormous challenge in the postgenomic era. A prime example of genes lacking functional assignments are the poorly conserved, early expressed genes of lytic bacteriophages, whose products are involved in the subversion of the host metabolism. In this study, we focused on the composition of important macromolecular complexes of Pseudomonas aeruginosa involved in transcription, DNA replication, fatty acid biosynthesis, RNA regulation, energy metabolism, and cell division during infection with members of seven distinct clades of lytic phages. Using affinity purifications of these host protein complexes coupled to mass spectrometric analyses, 37 host complex-associated phage proteins could be identified. Importantly, eight of these show an inhibitory effect on bacterial growth upon episomal expression, suggesting that these phage proteins are potentially involved in hijacking the host complexes. Using complementary protein-protein interaction assays, we further mapped the inhibitory interaction of gp12 of phage 14-1 to the α subunit of the RNA polymerase. Together, our data demonstrate the powerful use of interactomics to unravel the biological role of hypothetical phage proteins, which constitute an enormous untapped source of novel antibacterial proteins. (Data are available via ProteomeXchange with identifier PXD001199.).
Collapse
|
844
|
Chavshin AR, Oshaghi MA, Vatandoost H, Pourmand MR, Raeisi A, Terenius O. Isolation and identification of culturable bacteria from wild Anopheles culicifacies, a first step in a paratransgenesis approach. Parasit Vectors 2014; 7:419. [PMID: 25189316 PMCID: PMC4261757 DOI: 10.1186/1756-3305-7-419] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 08/17/2014] [Indexed: 11/10/2022] Open
Abstract
Background Due to the effect of midgut bacteria on proliferation of parasites and their potential as paratransgenesis tools, their identification in malaria vector mosquitoes is important. Anopheles culicifacies s.l. is one of the main malaria vectors in Asia; however, its midgut microbiota remains un-studied. This work was primarily designed to isolate potential candidates for use in a paratransgenesis approach, but also to give a picture of the midgut microbiota of wild-caught An. culicifacies larvae and adults from the southeast corner of Iran, which has the highest malaria endemicity in the country. Methods A total of 68 larvae and 34 adult females (newly eclosed and older) from three different biotopes in Iran were analyzed for their midgut microflora. The mosquitoes had their midgut bacterial contents plated on three different culture media (brain heart agar, nutrient agar and blood agar) yielding 57 bacterial isolates. The 16S rRNA genes of the isolates were sequence analyzed for species designation, which then was confirmed by biochemical analysis. Results A total of twelve bacterial genera were identified: Acinetobacter, Aeromonas, Bacillus, Chryseobacterium, Delftia, Exiguobacterium, Kurthia, Microbacterium, Pseudomonas, Staphylococcus, Thorsellia and Variovorax. In older females, only Gram-negative bacteria were found, whereas larvae and newly-eclosed adults also harbored Gram-positive bacteria. The diversity of isolates also varied between sampling sites and mosquito stages, with the largest number of genera found in the Anguri district and in larvae, respectively. Pseudomonas was the most common genus retrieved from all sampling sites, and in both larvae and adults, suggesting a potential transstadial passage of these bacteria. Interestingly, identical 16S sequences of Pseudomonas were found in mosquitoes originating from different habitats at least 45 km apart, which could suggest that these bacteria have been adapted to the mosquitoes. Conclusions The study of vector mosquito microbiota has recently gathered increased interest because of the potential influence on vector competence. By adding data from a hitherto uncharacterized malaria mosquito, a better picture of gut flora in vector mosquitoes was obtained. Furthermore, some species of the predominant genus Pseudomonas will be evaluated for the selection of a paratransgenesis candidate.
Collapse
Affiliation(s)
| | - Mohammad Ali Oshaghi
- Department of Medical Entomology and Vector Control, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| | | | | | | | | |
Collapse
|
845
|
Ahmed GF, Elkhatib WF, Noreddin AM. Inhibition of Pseudomonas aeruginosa PAO1 adhesion to and invasion of A549 lung epithelial cells by natural extracts. J Infect Public Health 2014; 7:436-44. [PMID: 24894307 DOI: 10.1016/j.jiph.2014.01.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Revised: 09/23/2013] [Accepted: 01/25/2014] [Indexed: 11/26/2022] Open
Abstract
Pseudomonas aeruginosa colonizes the lungs in cystic fibrosis (CF) and mechanically ventilated patients by binding to the cellular receptors on the surface of the lung epithelium. Studies have shown that blocking this interaction could be achieved with sub-minimum inhibitory concentrations of antibiotics such as ciprofloxacin. The development of bacterial resistance is a probable drawback of such an intervention. The use of natural extracts to interfere with bacterial adhesion and invasion has recently gained substantial attention and is hypothesized to inhibit bacterial binding and consequently prevent or reduce pathogenicity. This study used an A549 lung epithelial cell infection model, and the results revealed that a combination of aqueous cranberry extract with ciprofloxacin could completely prevent the adhesion and invasion of P. aeruginosa PAO1 compared to the untreated control. All of the natural extracts (cranberry, dextran, and soybean extracts) and ciprofloxacin showed a significant reduction (P<0.0001) in P. aeruginosa PAO1 adhesion to and invasion of lung epithelial cells relative to the control. The cranberry, dextran, and soybean extracts could substantially increase the anti-adhesion and anti-invasion effects of ciprofloxacin to the averages of 100% (P<0.0001), 80% (P<0.0001), and 60% (P<0.0001), respectively. Those extracts might result in a lower rate of the development of bacterial resistance; they are relatively safe and inexpensive agents, and utilizing such extracts, alone or in combination with ciprofloxacin, as potential anti-adhesion and anti-invasion remedies, could be valuable in preventing or reducing P. aeruginosa lung infections.
Collapse
Affiliation(s)
- Ghada F Ahmed
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, 717 Delaware St. SE, Minneapolis, MN 55414, USA
| | - Walid F Elkhatib
- Department of Microbiology & Immunology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt; Department of Pharmacy Practice, School of Pharmacy, Hampton University, Hampton, VA 23668, USA.
| | - Ayman M Noreddin
- Department of Pharmacy Practice, School of Pharmacy, Hampton University, Hampton, VA 23668, USA
| |
Collapse
|
846
|
Grimwood K, Bell SC, Chang AB. Antimicrobial treatment of non-cystic fibrosis bronchiectasis. Expert Rev Anti Infect Ther 2014; 12:1277-96. [PMID: 25156239 DOI: 10.1586/14787210.2014.952282] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Bronchiectasis unrelated to cystic fibrosis is characterized by chronic wet or productive cough, recurrent exacerbations and irreversible bronchial dilatation. After antibiotics and vaccines became available and living standards in affluent countries improved, its resulting reduced prevalence meant bronchiectasis was considered an 'orphan disease'. This perception has changed recently with increasing use of CT scans to diagnose bronchiectasis, including in those with severe chronic obstructive pulmonary disease or 'difficult to control' asthma, and adds to its already known importance in non-affluent countries and disadvantaged Indigenous communities. Following years of neglect, there is renewed interest in identifying the pathogenetic mechanisms of bronchiectasis, including the role of infection, and conducting clinical trials. This is providing much needed evidence to guide antimicrobial therapy, which has relied previously upon extrapolating treatments used in cystic fibrosis and chronic obstructive pulmonary disease. While many knowledge gaps and management challenges remain, the future is improving for patients with bronchiectasis.
Collapse
Affiliation(s)
- Keith Grimwood
- Griffith Health Institute, Griffith University, Gold Coast, QLD 4222, Australia
| | | | | |
Collapse
|
847
|
Lo Sciuto A, Fernández-Piñar R, Bertuccini L, Iosi F, Superti F, Imperi F. The periplasmic protein TolB as a potential drug target in Pseudomonas aeruginosa. PLoS One 2014; 9:e103784. [PMID: 25093328 PMCID: PMC4122361 DOI: 10.1371/journal.pone.0103784] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 07/05/2014] [Indexed: 01/09/2023] Open
Abstract
The Gram-negative bacterium Pseudomonas aeruginosa is one of the most dreaded pathogens in the hospital setting, and represents a prototype of multi-drug resistant "superbug" for which effective therapeutic options are very limited. The identification and characterization of new cellular functions that are essential for P. aeruginosa viability and/or virulence could drive the development of anti-Pseudomonas compounds with novel mechanisms of action. In this study we investigated whether TolB, the periplasmic component of the Tol-Pal trans-envelope protein complex of Gram-negative bacteria, represents a potential drug target in P. aeruginosa. By combining conditional mutagenesis with the analysis of specific pathogenicity-related phenotypes, we demonstrated that TolB is essential for P. aeruginosa growth, both in laboratory and clinical strains, and that TolB-depleted P. aeruginosa cells are strongly defective in cell-envelope integrity, resistance to human serum and several antibiotics, as well as in the ability to cause infection and persist in an insect model of P. aeruginosa infection. The essentiality of TolB for P. aeruginosa growth, resistance and pathogenicity highlights the potential of TolB as a novel molecular target for anti-P. aeruginosa drug discovery.
Collapse
Affiliation(s)
- Alessandra Lo Sciuto
- Department of Biology and Biotechnology “Charles Darwin”, Sapienza University of Rome, Rome, Italy
| | - Regina Fernández-Piñar
- Department of Biology and Biotechnology “Charles Darwin”, Sapienza University of Rome, Rome, Italy
| | - Lucia Bertuccini
- Ultrastructural Infectious Pathology Section, Department of Technology and Health, National Institute of Health, Rome, Italy
| | - Francesca Iosi
- Ultrastructural Infectious Pathology Section, Department of Technology and Health, National Institute of Health, Rome, Italy
| | - Fabiana Superti
- Ultrastructural Infectious Pathology Section, Department of Technology and Health, National Institute of Health, Rome, Italy
| | - Francesco Imperi
- Department of Biology and Biotechnology “Charles Darwin”, Sapienza University of Rome, Rome, Italy
- Pasteur Institute-Cenci Bolognetti Foundation, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
848
|
Simard M, Hill LA, Underhill CM, Keller BO, Villanueva I, Hancock REW, Hammond GL. Pseudomonas aeruginosa elastase disrupts the cortisol-binding activity of corticosteroid-binding globulin. Endocrinology 2014; 155:2900-8. [PMID: 24848868 PMCID: PMC4098004 DOI: 10.1210/en.2014-1055] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The serine protease inhibitor (SERPIN) family member corticosteroid-binding globulin (CBG) is the main carrier of glucocorticoids in plasma. Human CBG mediates the targeted release of cortisol at sites of inflammation through cleavage of its reactive center loop (RCL) by neutrophil elastase. The RCLs of SERPIN family members are targeted by diverse endogenous and exogenous proteases, including several bacterial proteases. We tested different bacteria for their ability to secrete proteases that disrupt CBG cortisol-binding activity, and characterized the responsible protease and site of CBG cleavage. Serum CBG integrity was assessed by Western blotting and cortisol-binding capacity assay. Effects of time, pH, temperature, and protease inhibitors were tested. Proteolytically active proteins from bacterial media were purified by fast protein liquid chromatography, and the active protease and CBG cleavage sites were identified by mass spectrometry. Among the bacteria tested, medium from Pseudomonas aeruginosa actively disrupted the cortisol-binding activity of CBG. This proteolytic activity was inhibited by zinc chelators and occurred most efficiently at pH 7 and elevated physiological temperature (ie, 41°C). Mass spectrometric analysis of a semi-purified fraction of P. aeruginosa media identified the virulence factor LasB as the responsible protease, and this was confirmed by assaying media from LasB-deficient P. aeruginosa. This metalloprotease cleaves the CBG RCL at a major site, distinct from that targeted by neutrophil elastase. Our results suggest that humoral responses to P. aeruginosa infection are influenced by this pathogen's ability to secrete a protease that promotes the release of the anti-inflammatory steroid, cortisol, from its plasma transport protein.
Collapse
Affiliation(s)
- Marc Simard
- Department of Cellular and Physiological Sciences (M.S., L.A.H., C.M.U., G.L.H.), University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, British Columbia, Canada, V6T 1Z3; Department of Pathology and Laboratory Medicine (B.O.K.), University of British Columbia, Child and Family Research Institute, 950 W 28th Ave, Vancouver, British Columbia, Canada, V5Z 4H4; Department of Microbiology and Immunology (I.V.), University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, British Columbia, Canada, V6T 1Z3; and Department of Microbiology and Immunology (R.E.W.H.), University of British Columbia, Centre for Microbial Diseases and Immunity Research, 2259 Lower Mall Research Station, Vancouver, British Columbia, Canada, V6T 1Z4
| | | | | | | | | | | | | |
Collapse
|
849
|
Scannapieco FA, Shay K. Oral health disparities in older adults: oral bacteria, inflammation, and aspiration pneumonia. Dent Clin North Am 2014; 58:771-82. [PMID: 25201541 DOI: 10.1016/j.cden.2014.06.005] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Poor oral hygiene has been suggested to be a risk factor for aspiration pneumonia in the institutionalized and disabled elderly. Control of oral biofilm formation in these populations reduces the numbers of potential respiratory pathogens in the oral secretions, which in turn reduces the risk for pneumonia. Together with other preventive measures, improved oral hygiene helps to control lower respiratory infections in frail elderly hospital and nursing home patients.
Collapse
Affiliation(s)
- Frank A Scannapieco
- Department of Oral Biology, School of Dental Medicine, University at Buffalo - The State University of New York, Foster Hall, Buffalo, NY 14214, USA.
| | - Kenneth Shay
- Geriatrics and Extended Care Services (10P4G), US Department of Veterans Affairs, PO Box 134002, Ann Arbor, MI 48113-4002, USA
| |
Collapse
|
850
|
Silva LV, Galdino ACM, Nunes APF, dos Santos KRN, Moreira BM, Cacci LC, Sodré CL, Ziccardi M, Branquinha MH, Santos ALS. Virulence attributes in Brazilian clinical isolates of Pseudomonas aeruginosa. Int J Med Microbiol 2014; 304:990-1000. [PMID: 25127423 DOI: 10.1016/j.ijmm.2014.07.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 06/17/2014] [Accepted: 07/15/2014] [Indexed: 11/25/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic human pathogen responsible for causing a huge variety of acute and chronic infections with significant levels of morbidity and mortality. Its success as a pathogen comes from its genetic/metabolic plasticity, intrinsic/acquired antimicrobial resistance, capacity to form biofilm and expression of numerous virulence factors. Herein, we have analyzed the genetic variability, antimicrobial susceptibility as well as the production of metallo-β-lactamases (MBLs) and virulence attributes (elastase, pyocyanin and biofilm) in 96 strains of P. aeruginosa isolated from different anatomical sites of patients attended at Brazilian hospitals. Our results revealed a great genetic variability, in which 86 distinct RAPD types (89.6% of polymorphisms) were detected. Regarding the susceptibility profile, 48 strains (50%) were resistant to the antimicrobials, as follows: 22.92% to the three tested antibiotics, 12.5% to both imipenem and meropenem, 11.46% to ceftazidime only, 2.08% to imipenem only and 1.04% to both ceftazidime and meropenem. Out of the 34 clinical strains of P. aeruginosa resistant to both imipenem and meropenem, 25 (73.53%) were MBL producers by phenotypic method while 12 (35.29%) were PCR positive for the MBL gene SPM-1. All P. aeruginosa strains produced pyocyanin, elastase and biofilm, although in different levels. Some associations were demonstrated among the susceptibility and/or production of these virulence traits with the anatomical site of strain isolation. For instance, almost all strains isolated from urine (85.71%) were resistant to the three antibiotics, while the vast majority of strains isolated from rectum (95%) and mouth (66.67%) were susceptible to all tested antibiotics. Urine isolates produced the highest pyocyanin concentration (20.15±5.65 μg/ml), while strains isolated from pleural secretion and mouth produced elevated elastase activity (1441.43±303.08 FAU) and biofilm formation (OD590 0.676±0.32), respectively. Also, MBL-positive strains produced robust biofilm compared to MBL-negative strains. Collectively, the production of site-dependent virulence factors can be highlighted as potential therapeutic targets for the treatment of infections caused by heterogeneous and resistant strains of P. aeruginosa.
Collapse
Affiliation(s)
- Lívia V Silva
- Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Anna Clara M Galdino
- Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Programa de Pós-Graduação em Bioquímica, Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Paula F Nunes
- Departamento de Patologia e Programa de Pós-Graduação em Doenças Infecciosas, Universidade Federal do Espírito Santo, Espírito Santo, Brazil
| | - Kátia R N dos Santos
- Departamento de Microbiologia Médica, Instituto de Microbiologia Paulo de Góes, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Beatriz M Moreira
- Departamento de Microbiologia Médica, Instituto de Microbiologia Paulo de Góes, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana C Cacci
- Programa de Pós-Graduação em Doenças Infecciosas e Parasitárias, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cátia L Sodré
- Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil
| | - Mariangela Ziccardi
- Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Marta H Branquinha
- Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - André L S Santos
- Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Programa de Pós-Graduação em Bioquímica, Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|