1
|
Delumeau A, Quétel I, Harnais F, Sellin A, Gros O, Talarmin A, Marcelino I. Bacterial microbiota management in free-living amoebae (Heterolobosea lineage) isolated from water: The impact of amoebae identity, grazing conditions, and passage number. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 900:165816. [PMID: 37506913 DOI: 10.1016/j.scitotenv.2023.165816] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/24/2023] [Accepted: 07/24/2023] [Indexed: 07/30/2023]
Abstract
Free-living amoebae (FLA) are ubiquitous protozoa mainly found in aquatic environments. They are well-known reservoirs and vectors for the transmission of amoeba-resistant bacteria (ARB), most of which are pathogenic to humans. Yet, the natural bacterial microbiota associated with FLA remains largely unknown. Herein, we characterized the natural bacterial microbiota of different FLA species isolated from recreational waters in Guadeloupe. Monoxenic cultures of Naegleria australiensis, Naegleria sp. WTP3, Paravahlkampfia ustiana and Vahlkampfia sp. AK-2007 (Heterolobosea lineage) were cultivated under different grazing conditions, during successive passages. The whole bacterial microbiota of the waters and the amoebal cysts was characterized using 16S rRNA gene metabarcoding. The culturable subset of ARB was analyzed by mass spectrometry (MALDI-TOF MS), conventional 16S PCR, and disk diffusion method (to assess bacterial antibiotic resistance). Transmission electron microscopy was used to locate the ARB inside the amoebae. According to alpha and beta-diversity analyses, FLA bacterial microbiota were significantly different from the ones of their habitat. While Vogesella and Aquabacterium genera were detected in water, the most common ARB belonged to Pseudomonas, Bosea, and Escherichia/Shigella genera. The different FLA species showed both temporary and permanent associations with differentially bacterial taxa, suggesting host specificity. These associations depend on the number of passages and grazing conditions. Additionally, Naegleria, Vahlkampfia and Paravahlkampfia cysts were shown to naturally harbor viable bacteria of the Acinetobacter, Escherichia, Enterobacter, Pseudomonas and Microbacterium genera, all being pathogenic to humans. To our knowledge, this is the first time Paravahlkampfia and Vahlkampfia have been demonstrated as hosts of pathogenic ARB in water. Globally, the persistence of these ARB inside resistant cysts represents a potential health risk. To ensure the continued safety of recreational waters, it is crucial to (i) regularly control both the amoebae and their ARB and (ii) improve knowledge on amoebae-bacteria interactions to establish better water management protocols.
Collapse
Affiliation(s)
- Aurélie Delumeau
- Institut Pasteur de la Guadeloupe, Unité TReD-Path, Les Abymes, Guadeloupe, France
| | - Isaure Quétel
- Institut Pasteur de la Guadeloupe, Unité TReD-Path, Les Abymes, Guadeloupe, France
| | - Florian Harnais
- Institut Pasteur de la Guadeloupe, Unité TReD-Path, Les Abymes, Guadeloupe, France
| | - Arantxa Sellin
- Institut Pasteur de la Guadeloupe, Unité TReD-Path, Les Abymes, Guadeloupe, France
| | - Olivier Gros
- Institut de Systématique, Évolution, Biodiversité (ISYEB), Muséum National d'Histoire Naturelle, CNRS, Sorbonne Université, EPHE, Université des Antilles, Pointe-à-Pitre, Guadeloupe, France
| | - Antoine Talarmin
- Institut Pasteur de la Guadeloupe, Unité TReD-Path, Les Abymes, Guadeloupe, France
| | - Isabel Marcelino
- Institut Pasteur de la Guadeloupe, Unité TReD-Path, Les Abymes, Guadeloupe, France.
| |
Collapse
|
2
|
Matveeva M, Lefebvre M, Chahinian H, Yahi N, Fantini J. Host Membranes as Drivers of Virus Evolution. Viruses 2023; 15:1854. [PMID: 37766261 PMCID: PMC10535233 DOI: 10.3390/v15091854] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
The molecular mechanisms controlling the adaptation of viruses to host cells are generally poorly documented. An essential issue to resolve is whether host membranes, and especially lipid rafts, which are usually considered passive gateways for many enveloped viruses, also encode informational guidelines that could determine virus evolution. Due to their enrichment in gangliosides which confer an electronegative surface potential, lipid rafts impose a first control level favoring the selection of viruses with enhanced cationic areas, as illustrated by SARS-CoV-2 variants. Ganglioside clusters attract viral particles in a dynamic electrostatic funnel, the more cationic viruses of a viral population winning the race. However, electrostatic forces account for only a small part of the energy of raft-virus interaction, which depends mainly on the ability of viruses to form a network of hydrogen bonds with raft gangliosides. This fine tuning of virus-ganglioside interactions, which is essential to stabilize the virus on the host membrane, generates a second level of selection pressure driven by a typical induced-fit mechanism. Gangliosides play an active role in this process, wrapping around the virus spikes through a dynamic quicksand-like mechanism. Viruses are thus in an endless race for access to lipid rafts, and they are bound to evolve perpetually, combining speed (electrostatic potential) and precision (fine tuning of amino acids) under the selective pressure of the immune system. Deciphering the host membrane guidelines controlling virus evolution mechanisms may open new avenues for the design of innovative antivirals.
Collapse
Affiliation(s)
| | | | | | | | - Jacques Fantini
- Department of Biology, Faculty of Medicine, University of Aix-Marseille, INSERM UMR_S 1072, 13015 Marseille, France; (M.M.); (M.L.); (H.C.); (N.Y.)
| |
Collapse
|
3
|
Song Y, Zhang S, Ye Z, Song Y, Chen L, Tong A, He Y, Bao R. The novel type II toxin-antitoxin PacTA modulates Pseudomonas aeruginosa iron homeostasis by obstructing the DNA-binding activity of Fur. Nucleic Acids Res 2022; 50:10586-10600. [PMID: 36200834 PMCID: PMC9561280 DOI: 10.1093/nar/gkac867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 09/20/2022] [Accepted: 09/27/2022] [Indexed: 11/21/2022] Open
Abstract
Type II toxin–antitoxin (TA) systems are widely distributed in bacterial and archaeal genomes and are involved in diverse critical cellular functions such as defense against phages, biofilm formation, persistence, and virulence. GCN5-related N-acetyltransferase (GNAT) toxin, with an acetyltransferase activity-dependent mechanism of translation inhibition, represents a relatively new and expanding family of type II TA toxins. We here describe a group of GNAT-Xre TA modules widely distributed among Pseudomonas species. We investigated PacTA (one of its members encoded by PA3270/PA3269) from Pseudomonas aeruginosa and demonstrated that the PacT toxin positively regulates iron acquisition in P. aeruginosa. Notably, other than arresting translation through acetylating aminoacyl-tRNAs, PacT can directly bind to Fur, a key ferric uptake regulator, to attenuate its DNA-binding affinity and thus permit the expression of downstream iron-acquisition-related genes. We further showed that the expression of the pacTA locus is upregulated in response to iron starvation and the absence of PacT causes biofilm formation defect, thereby attenuating pathogenesis. Overall, these findings reveal a novel regulatory mechanism of GNAT toxin that controls iron-uptake-related genes and contributes to bacterial virulence.
Collapse
Affiliation(s)
- Yingjie Song
- Center of Infectious Diseases, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610093, China.,Central Laboratory, Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu 610081, China
| | - Siping Zhang
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - Zirui Ye
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - Yongyan Song
- Central Laboratory, Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu 610081, China
| | - Lin Chen
- Central Laboratory, Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu 610081, China
| | - Aiping Tong
- Center of Infectious Diseases, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610093, China
| | - Yongxing He
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - Rui Bao
- Center of Infectious Diseases, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610093, China
| |
Collapse
|
4
|
Phuengmaung P, Mekjaroen J, Saisorn W, Chatsuwan T, Somparn P, Leelahavanichkul A. Rapid Synergistic Biofilm Production of Pseudomonas and Candida on the Pulmonary Cell Surface and in Mice, a Possible Cause of Chronic Mixed Organismal Lung Lesions. Int J Mol Sci 2022; 23:ijms23169202. [PMID: 36012475 PMCID: PMC9409386 DOI: 10.3390/ijms23169202] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/12/2022] [Accepted: 08/15/2022] [Indexed: 11/16/2022] Open
Abstract
Due to the possible co-presence of Pseudomonas aeruginosa and Candida albicans (the most common nosocomial pathogens) in lungs, rapid interkingdom biofilm production is possible. As such, PA+CA produced more dominant biofilms on the pulmonary epithelial surface (NCI-H292) (confocal fluorescent extracellular matrix staining) with dominant psl upregulation, as demonstrated by polymerase chain reaction (PCR), after 8 h of experiments than PA alone. With a proteomic analysis, rhamnosyltransferase RhlB protein (Psl-associated quorum-sensing protein) was found to be among the high-abundance proteins in PA+CA than in PA biofilms, supporting psl-mediated biofilms in PA+CA on the cell surface. Additionally, PA+CA increased supernatant cytokines (IL-8 and IL-13, but not TNF-α, IL-6, and IL-10) with a similar upregulation of TLR-4, TLR-5, and TLR-9 (by PCR) compared with PA-stimulated cells. The intratracheal administration of PA+CA induced a greater severity of sepsis (serum creatinine, alanine transaminase, serum cytokines, and histology score) and prominent biofilms (fluorescent staining) with psl upregulation (PCR). In comparison with PA+CA biofilms on glass slides, PA+CA biofilms on biotic surfaces were more prominent (fluorescent staining). In conclusion, PA+CA induced Psl-predominant biofilms on the pulmonary cell surface and in mice with acute pneumonia, and these biofilms were more prominent than those induced by PA alone, highlighting the impact of Candida on rapid interkingdom biofilm production.
Collapse
Affiliation(s)
- Pornpimol Phuengmaung
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Center of Excellence on Translational Research in Inflammation and Immunology (CETRII), Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Jiradej Mekjaroen
- Center of Excellence in Systems Biology, Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Wilasinee Saisorn
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Center of Excellence on Translational Research in Inflammation and Immunology (CETRII), Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Tanittha Chatsuwan
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Poorichaya Somparn
- Center of Excellence in Systems Biology, Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Correspondence: (P.S.); (A.L.); Tel.: +66-2-256-4251 (P.S. & A.L.); Fax: +66-2-252-6920 (P.S. & A.L.)
| | - Asada Leelahavanichkul
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Center of Excellence on Translational Research in Inflammation and Immunology (CETRII), Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Correspondence: (P.S.); (A.L.); Tel.: +66-2-256-4251 (P.S. & A.L.); Fax: +66-2-252-6920 (P.S. & A.L.)
| |
Collapse
|
5
|
Adhikari BR, Sinha S, Gordon KC, Das SC. Amino Acids Improve Aerosolization and Chemical Stability of Potential Inhalable Amorphous Spray-dried Ceftazidime for Pseudomonas aeruginosa Lung Infection. Int J Pharm 2022; 621:121799. [PMID: 35525472 DOI: 10.1016/j.ijpharm.2022.121799] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 04/26/2022] [Accepted: 04/30/2022] [Indexed: 11/30/2022]
Abstract
Pseudomonas aeruginosa infection is common in cystic fibrosis as well non-cystic fibrosis bronchiectasis. The pathogen presents challenges for treatment due to its adaptive antibiotic-resistance, mainly pertaining to its biofilm-forming ability, as well as limitations associated with conventional drug delivery in achieving desired therapeutic concentration in the infection site. Hence, therapeutic approach has shifted towards the inhalation of antibiotics. Ceftazidime is a potent antibiotic against the pathogen; however, it is currently only available as a parenteral formulation. Here, spray-dryer was employed to generate inhalable high dose ceftazidime microparticles. In addition, the use of amino acids (valine, leucine, methionine, phenylalanine, and tryptophan) to improve aerosolization as well as chemical stability of amorphous ceftazidime was explored. The particles were characterized using X ray diffraction, infrared (IR) spectroscopy, calorimetry, electron microscopy, particle size analyzer, and next generation impactor. The chemical stability at 25 °C/<15% was assessed using chromatography. All co-spray dried formulations were confirmed as monophasic amorphous systems using calorimetry. In addition, principal component analysis of the IR spectra suggested potential interaction between tryptophan and ceftazidime in the co-amorphous matrix. Inclusion of amino acids improved aerosolization and chemical stability in all cases. Increase in surface asperity was clear with the use of amino acids which likely contributed to the improved aerosol performance, and potential interaction between amino acids and ceftazidime was plausibly the reason for improved chemical stability. Leucine offered the best aerosolization enhancement with a fine particle fraction of 78% and tryptophan showed stabilizing superiority by reducing chemical degradation by 51% over 10 weeks in 1:1 molar ratio. The protection against ceftazidime degradation varied with the nature of amino acids. Additionally, there was a linear relationship between degradation protection and molar mass of amino acids or percentage weight of amino acids in the formulations. None of the amino acids were successful in completely inhibiting degradation of ceftazidime in amorphous spray-dried powder to prepare a commercially viable product with desired shelf-life. All the amino acids and ceftazidime were non-toxic to A549 alveolar cell line.
Collapse
Affiliation(s)
| | - Shubhra Sinha
- School of Pharmacy, University of Otago, Dunedin 9054, New Zealand
| | - Keith C Gordon
- The Dodd-Walls Centre for Photonic and Quantum Technologies, Department of Chemistry, University of Otago, Dunedin 9016, New Zealand
| | - Shyamal C Das
- School of Pharmacy, University of Otago, Dunedin 9054, New Zealand.
| |
Collapse
|
6
|
Blazanin M, Lam WT, Vasen E, Chan BK, Turner PE. Decay and damage of therapeutic phage OMKO1 by environmental stressors. PLoS One 2022; 17:e0263887. [PMID: 35196336 PMCID: PMC8865689 DOI: 10.1371/journal.pone.0263887] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 01/25/2022] [Indexed: 01/21/2023] Open
Abstract
Antibiotic resistant bacterial pathogens are increasingly prevalent, driving the need for alternative approaches to chemical antibiotics when treating infections. One such approach is bacteriophage therapy: the use of bacteria-specific viruses that lyse (kill) their host cells. Just as the effect of environmental conditions (e.g. elevated temperature) on antibiotic efficacy is well-studied, the effect of environmental stressors on the potency of phage therapy candidates demands examination. Therapeutic phage OMKO1 infects and kills the opportunistic human pathogen Pseudomonas aeruginosa. Here, we used phage OMKO1 as a model to test how environmental stressors can lead to damage and decay of virus particles. We assessed the effects of elevated temperatures, saline concentrations, and urea concentrations. We observed that OMKO1 particles were highly tolerant to different saline concentrations, but decayed more rapidly at elevated temperatures and under high concentrations of urea. Additionally, we found that exposure to elevated temperature reduced the ability of surviving phage particles to suppress the growth of P. aeruginosa, suggesting a temperature-induced damage. Our findings demonstrate that OMKO1 is highly tolerant to a range of conditions that could be experienced inside and outside the human body, while also showing the need for careful characterization of therapeutic phages to ensure that environmental exposure does not compromise their expected potency, dosing, and pharmacokinetics.
Collapse
Affiliation(s)
- Michael Blazanin
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, United States of America
- * E-mail:
| | - Wai Tin Lam
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, United States of America
| | - Emma Vasen
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, United States of America
| | - Benjamin K. Chan
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, United States of America
| | - Paul E. Turner
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, United States of America
- Program in Microbiology, Yale School of Medicine, New Haven, CT, United States of America
| |
Collapse
|
7
|
Dzik M, Aebisher D, Olender A, Tabarkiewicz J. Evaluation of Selected Parameters of the Specific Immune Response against Pseudomonas aeruginosa Strains. Cells 2021; 11:cells11010003. [PMID: 35011563 PMCID: PMC8750466 DOI: 10.3390/cells11010003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/18/2021] [Accepted: 12/19/2021] [Indexed: 11/16/2022] Open
Abstract
The immune response to Pseudomonas aeruginosa strains could be influenced by differences in antibiotic resistance and virulence. At the present time, it is unclear which type of immune responses enables uncontrolled invasion of opportunistic pathogens. The conditional pathogenicity of Pseudomonas aeruginosa served as an inspiration to begin a study on this bacterium. The aim of this study was to gain insight into selected parameters describing immune responses with regards to the adaptable agents of this pathogen. For the analysis of the specific immune response, the potential of Pseudomonas aeruginosa to stimulate lymphocytes, including Th17 lymphocytes, dendritic cells and other components of the adaptive immune response, was examined. The highest percentage of CD83+CD1a-HLA-DR++ cells was found after stimulation with lysates of strains isolated from the patients with severe systemic infection. We found statistically significant differences in percentages of HLA-DR+ PBMCs and MFI of HLA-DR between groups of Pseudomonas aeruginosa strains isolated from the patients with different clinical courses of infection. Our results suggest that the clinical course and outcomes of Pseudomonas aeruginosa infections are not associated with impairment of the specific immune response.
Collapse
Affiliation(s)
- Michał Dzik
- Department of Clinical Immunology, Faculty of Medicine, Medical University of Lublin, 20-095 Lublin, Poland;
- Institut Asclépiade, 10410 Saint-Parres-aux-Tertres, France
| | - David Aebisher
- Centre for Innovative Research in Medical and Natural Sciences, Medical College of Rzeszów University, University of Rzeszów, 35-959 Rzeszow, Poland;
| | - Alina Olender
- Chair and Department of Medical Microbiology, Faculty of Medicine, Medical University of Lublin, 20-095 Lublin, Poland;
| | - Jacek Tabarkiewicz
- Centre for Innovative Research in Medical and Natural Sciences, Medical College of Rzeszów University, University of Rzeszów, 35-959 Rzeszow, Poland;
- Department of Human Immunology, Institute of Medical Sciences, Medical College of Rzeszów University, University of Rzeszów, 35-959 Rzeszow, Poland
- Correspondence: ; Tel.: +48-851-68-08
| |
Collapse
|
8
|
Chatterjee R, Chowdhury AR, Mukherjee D, Chakravortty D. Lipid larceny: channelizing host lipids for establishing successful pathogenesis by bacteria. Virulence 2021; 12:195-216. [PMID: 33356849 PMCID: PMC7808437 DOI: 10.1080/21505594.2020.1869441] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 12/03/2020] [Accepted: 12/17/2020] [Indexed: 12/16/2022] Open
Abstract
Lipids are complex organic compounds made up of carbon, oxygen, and hydrogen. These play a diverse and intricate role in cellular processes like membrane trafficking, protein sorting, signal transduction, and bacterial infections. Both Gram-positive bacteria (Staphylococcus sp., Listeria monocytogenes, etc.) and Gram-negative bacteria (Chlamydia sp., Salmonella sp., E. coli, etc.) can hijack the various host-lipids and utilize them structurally as well as functionally to mount a successful infection. The pathogens can deploy with various arsenals to exploit host membrane lipids and lipid-associated receptors as an attachment for toxins' landing or facilitate their entry into the host cellular niche. Bacterial species like Mycobacterium sp. can also modulate the host lipid metabolism to fetch its carbon source from the host. The sequential conversion of host membrane lipids into arachidonic acid and prostaglandin E2 due to increased activity of cPLA-2 and COX-2 upon bacterial infection creates immunosuppressive conditions and facilitates the intracellular growth and proliferation of bacteria. However, lipids' more debatable role is that they can also be a blessing in disguise. Certain host-lipids, especially sphingolipids, have been shown to play a crucial antibacterial role and help the host in combating the infections. This review shed light on the detailed role of host lipids in bacterial infections and the current understanding of the lipid in therapeutics. We have also discussed potential prospects and the need of the hour to help us cope in this race against deadly pathogens and their rapidly evolving stealthy virulence strategies.
Collapse
Affiliation(s)
- Ritika Chatterjee
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bangalore, India
| | - Atish Roy Chowdhury
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bangalore, India
| | - Debapriya Mukherjee
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bangalore, India
| | - Dipshikha Chakravortty
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bangalore, India
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore, India
| |
Collapse
|
9
|
Govender R, Amoah ID, Adegoke AA, Singh G, Kumari S, Swalaha FM, Bux F, Stenström TA. Identification, antibiotic resistance, and virulence profiling of Aeromonas and Pseudomonas species from wastewater and surface water. ENVIRONMENTAL MONITORING AND ASSESSMENT 2021; 193:294. [PMID: 33893564 DOI: 10.1007/s10661-021-09046-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 04/04/2021] [Indexed: 06/12/2023]
Abstract
Aquatic environments are hotspots for the spread of antibiotic-resistant bacteria and genes due to pollution caused mainly by anthropogenic activities. The aim of this study was to evaluate the impact of wastewater effluents, informal settlements, hospital, and veterinary clinic discharges on the occurrence, antibiotic resistance profile and virulence signatures of Aeromonas spp. and Pseudomonas spp. isolated from surface water and wastewater. High counts of Aeromonas spp. (2.5 (± 0.8) - 3.3 (± 0.4) log10 CFU mL-1) and Pseudomonas spp. (0.6 (± 1.0) - 1.8 (± 1.0) log10 CFU mL-1) were obtained. Polymerase chain reaction (PCR) and MALDI-TOF characterization identified four species of Aeromonas and five of Pseudomonas. The isolates displayed resistance to 3 or more antibiotics (71% of Aeromonas and 94% of Pseudomonas). Aeromonas spp. showed significant association with the antibiotic meropenem (χ2 = 3.993, P < 0.05). The virulence gene aer in Aeromonas was found to be positively associated with the antibiotic resistance gene blaOXA (χ2 = 6.657, P < 0.05) and the antibiotic ceftazidime (χ2 = 7.537, P < 0.05). Aeromonas recovered from both wastewater and surface water displayed high resistance to ampicillin and had higher multiple antibiotic resistance (MAR) indices close to the hospital. Pseudomonas isolates on the other hand exhibited low resistance to carbapenems but very high resistance to the third-generation cephalosporins and cefixime. The results showed that some of the Pseudomonas spp. and Aeromonas spp. isolates were extended-spectrum β-lactamase producing bacteria. In conclusion, the strong association between virulence genes and antibiotic resistance in the isolates shows the potential health risk to communities through direct and indirect exposure to the water.
Collapse
Affiliation(s)
- Reshme Govender
- Institute for Water and Wastewater Technology, Durban University of Technology, Kwa-Zulu Natal 4001, Durban, South Africa
| | - Isaac Dennis Amoah
- Institute for Water and Wastewater Technology, Durban University of Technology, Kwa-Zulu Natal 4001, Durban, South Africa
| | - Anthony Ayodeji Adegoke
- Institute for Water and Wastewater Technology, Durban University of Technology, Kwa-Zulu Natal 4001, Durban, South Africa
| | - Gulshan Singh
- Institute for Water and Wastewater Technology, Durban University of Technology, Kwa-Zulu Natal 4001, Durban, South Africa
| | - Sheena Kumari
- Institute for Water and Wastewater Technology, Durban University of Technology, Kwa-Zulu Natal 4001, Durban, South Africa.
| | - Feroz Mahomed Swalaha
- Institute for Water and Wastewater Technology, Durban University of Technology, Kwa-Zulu Natal 4001, Durban, South Africa
| | - Faizal Bux
- Institute for Water and Wastewater Technology, Durban University of Technology, Kwa-Zulu Natal 4001, Durban, South Africa
| | - Thor Axel Stenström
- Institute for Water and Wastewater Technology, Durban University of Technology, Kwa-Zulu Natal 4001, Durban, South Africa
| |
Collapse
|
10
|
Rutin-loaded liquid crystalline nanoparticles attenuate oxidative stress in bronchial epithelial cells: a PCR validation. Future Med Chem 2021; 13:543-549. [PMID: 33538615 DOI: 10.4155/fmc-2020-0297] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aim: In the present study, the inhibitory potential of rutin-loaded liquid crystalline nanoparticles (LCNs) on oxidative stress was determined in human bronchial epithelial cells (BEAS-2B) by analysing the expression levels of different antioxidant (NADPH quinine oxidoreductase-1 (NQO1); γ-glutamyl cysteine synthetase catalytic subunit (GCLC)) and pro-oxidant (NADPH oxidase (Nox)-4; Nox2B) genes. Results: Our findings revealed that the rutin-loaded LCNs inhibited the genes, namely Nox2B and Nox4, which caused oxidative stress. In addition, these nanoparticles demonstrated an upregulation in the expression of the antioxidant genes Gclc and Nqo-1 in a dose-dependent manner. Conclusion: The study indicates the promising potential of rutin-loaded LCNs as an effective treatment strategy in patients with high oxidant loads in various respiratory diseases.
Collapse
|
11
|
Balczon R, Morrow KA, Leavesley S, Francis CM, Stevens TC, Agwaramgbo E, Williams C, Stevens RP, Langham G, Voth S, Cioffi EA, Weintraub SE, Stevens T. Cystatin C regulates the cytotoxicity of infection-induced endothelial-derived β-amyloid. FEBS Open Bio 2020; 10:2464-2477. [PMID: 33030263 PMCID: PMC7609779 DOI: 10.1002/2211-5463.12997] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 08/25/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023] Open
Abstract
Infection of rat pulmonary microvascular endothelial cells with the bacterium Pseudomonas aeruginosa induces the production and release of cytotoxic oligomeric tau and beta amyloid (Aβ). Here, we characterized these cytotoxic amyloids. Cytotoxic behavior and oligomeric tau were partially resistant to digestion with proteinase K, but cytotoxicity was abolished by various denaturants including phenol, diethylpyrocarbonate (DEPC), and 1,1,1,3,3,3-hexafluoro-2-isopropanol (HFIP). Ultracentrifugation for 8 h at 150 000 g was required to remove cytotoxic activity from the supernatant. Ultracentrifugation, DEPC treatment, and immunodepletion using antibodies against Aβ also demonstrated that cytoprotective protein(s) are released from endothelial cells during P. aeruginosa infection. Mass spectrometry of endothelial cell culture media following P. aeruginosa infection allowed identification of multiple potential secreted modulators of Aβ, including cystatin C, gelsolin, and ApoJ/clusterin. Immunodepletion, co-immunoprecipitation, and ultracentrifugation determined that the cytoprotective factor released during infection of endothelial cells by P. aeruginosa is cystatin C, which appears to be in a complex with Aβ. Cytoprotective cystatin C may provide a novel therapeutic avenue for protection against the long-term consequences of infection with P. aeruginosa.
Collapse
Affiliation(s)
- Ron Balczon
- Department of Biochemistry and Molecular BiologyUniversity of South AlabamaMobileALUSA
- Center for Lung BiologyUniversity of South AlabamaMobileALUSA
| | - Kyle A. Morrow
- Department of Cell Biology and PhysiologyEdward Via College of Osteopathic MedicineMonroeLAUSA
| | - Silas Leavesley
- Center for Lung BiologyUniversity of South AlabamaMobileALUSA
- Department of Chemical and Biomedical EngineeringUniversity of South AlabamaMobileALUSA
| | - Christopher M. Francis
- Center for Lung BiologyUniversity of South AlabamaMobileALUSA
- Department of Physiology and Cell BiologyUniversity of South AlabamaMobileALUSA
| | - Trevor C. Stevens
- Center for Lung BiologyUniversity of South AlabamaMobileALUSA
- Department of Physiology and Cell BiologyUniversity of South AlabamaMobileALUSA
| | - Ezinne Agwaramgbo
- Department of Physiology and Cell BiologyUniversity of South AlabamaMobileALUSA
| | | | - Reece P. Stevens
- Center for Lung BiologyUniversity of South AlabamaMobileALUSA
- Department of Physiology and Cell BiologyUniversity of South AlabamaMobileALUSA
| | - Geri Langham
- Department of Physiology and Cell BiologyUniversity of South AlabamaMobileALUSA
| | - Sarah Voth
- Center for Lung BiologyUniversity of South AlabamaMobileALUSA
- Department of Physiology and Cell BiologyUniversity of South AlabamaMobileALUSA
| | - Eugene A. Cioffi
- Department of PharmacologyUniversity of South AlabamaMobileALUSA
| | - Susan E. Weintraub
- Department of Biochemistry and Structural Biology and Mass Spectrometry LaboratoryUniversity of Texas at San Antonio Health Sciences CenterTXUSA
| | - Troy Stevens
- Center for Lung BiologyUniversity of South AlabamaMobileALUSA
- Department of Physiology and Cell BiologyUniversity of South AlabamaMobileALUSA
| |
Collapse
|
12
|
Li M, Wei X, Li Y, Feng T, Jiang L, Zhu H, Yu X, Tang J, Chen G, Zhang J, Zhang X. PM2.5 in poultry houses synergizes with Pseudomonas aeruginosa to aggravate lung inflammation in mice through the NF-κB pathway. J Vet Sci 2020; 21:e46. [PMID: 32476320 PMCID: PMC7263920 DOI: 10.4142/jvs.2020.21.e46] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 03/08/2020] [Accepted: 03/23/2020] [Indexed: 12/25/2022] Open
Abstract
Background High concentrations of particulate matter less than 2.5 µm in diameter (PM2.5) in poultry houses is an important cause of respiratory disease in animals and humans. Pseudomonas aeruginosa is an opportunistic pathogen that can induce severe respiratory disease in animals under stress or with abnormal immune functions. When excessively high concentrations of PM2.5 in poultry houses damage the respiratory system and impair host immunity, secondary infections with P. aeruginosa can occur and produce a more intense inflammatory response, resulting in more severe lung injury. Objectives In this study, we focused on the synergistic induction of inflammatory injury in the respiratory system and the related molecular mechanisms induced by PM2.5 and P. aeruginosa in poultry houses. Methods High-throughput 16S rDNA sequence analysis was used for characterizing the bacterial diversity and relative abundance of the PM2.5 samples, and the effects of PM2.5 and P. aeruginosa stimulation on inflammation were detected by in vitro and in vivo. Results Sequencing results indicated that the PM2.5 in poultry houses contained a high abundance of potentially pathogenic genera, such as Pseudomonas (2.94%). The lung tissues of mice had more significant pathological damage when co-stimulated by PM2.5 and P. aeruginosa, and it can increase the expression levels of interleukin (IL)-6, IL-8, and tumor necrosis factor-α through nuclear factor (NF)-κB pathway in vivo and in vitro. Conclusions The results confirmed that poultry house PM2.5 in combination with P. aeruginosa could aggravate the inflammatory response and cause more severe respiratory system injuries through a process closely related to the activation of the NF-κB pathway.
Collapse
Affiliation(s)
- Meng Li
- College of Life Science, Ludong University, Yantai 264000, China.,Shandong Provincial Key Laboratory of Quality Safty Monitoring and Risk Assessment for Animal Products, Ji'nan 250022, China
| | - Xiuli Wei
- Shandong Provincial Key Laboratory of Quality Safty Monitoring and Risk Assessment for Animal Products, Ji'nan 250022, China
| | - Youzhi Li
- Shandong Provincial Key Laboratory of Quality Safty Monitoring and Risk Assessment for Animal Products, Ji'nan 250022, China
| | - Tao Feng
- Shandong Provincial Key Laboratory of Quality Safty Monitoring and Risk Assessment for Animal Products, Ji'nan 250022, China
| | - Linlin Jiang
- College of Life Science, Ludong University, Yantai 264000, China.,Yantai Key Laboratory of Animal Pathogenetic Microbiology and Immunology, Yantai 264000, China
| | - Hongwei Zhu
- College of Life Science, Ludong University, Yantai 264000, China.,Yantai Key Laboratory of Animal Pathogenetic Microbiology and Immunology, Yantai 264000, China
| | - Xin Yu
- College of Life Science, Ludong University, Yantai 264000, China.,Yantai Key Laboratory of Animal Pathogenetic Microbiology and Immunology, Yantai 264000, China
| | - Jinxiu Tang
- College of Life Science, Ludong University, Yantai 264000, China.,Yantai Key Laboratory of Animal Pathogenetic Microbiology and Immunology, Yantai 264000, China
| | - Guozhong Chen
- College of Life Science, Ludong University, Yantai 264000, China.,Yantai Key Laboratory of Animal Pathogenetic Microbiology and Immunology, Yantai 264000, China
| | - Jianlong Zhang
- College of Life Science, Ludong University, Yantai 264000, China.,Yantai Key Laboratory of Animal Pathogenetic Microbiology and Immunology, Yantai 264000, China.
| | - Xingxiao Zhang
- College of Life Science, Ludong University, Yantai 264000, China.,Yantai Key Laboratory of Animal Pathogenetic Microbiology and Immunology, Yantai 264000, China.
| |
Collapse
|
13
|
Guo J, Xiong Y, Shi C, Liu C, Li H, Qian H, Sun Z, Qin C. Characteristics of airborne bacterial communities in indoor and outdoor environments during continuous haze events in Beijing: Implications for health care. ENVIRONMENT INTERNATIONAL 2020; 139:105721. [PMID: 32305743 DOI: 10.1016/j.envint.2020.105721] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 04/02/2020] [Accepted: 04/03/2020] [Indexed: 06/11/2023]
Abstract
There is solid evidence that haze pollution threatens human health owing to the abiotic pollutants it contains. However, the characteristics of airborne bacterial communities in indoor and outdoor environments exhibiting haze occurrence are still unknown. Thus, we examined variations in both indoor and outdoor airborne bacterial communities in Beijing from December 9-27, 2016, a period which included three haze events. The outdoor airborne bacterial communities were clustered into two main groups (Groups I and II), and they shifted between two typical bacterial communities regardless of the haze event. The Chao1, Shannon, and phylogenetic diversity indexes and abundance of dominant classes changed significantly, as did airborne bacterial community type. The indoor airborne bacterial community closely tracked the outdoor bacterial community type, forming two obvious groups supported by Adonis analysis, changes in dominant classes, and bacterial diversity compared to the outdoor group. Furthermore, we found that the airborne bacterial community type could affect the morbidity of respiratory diseases. Daily pneumonia cases were significantly higher in Group I (p = 0.035), whereas daily amygdalitis cases were significantly higher in Group II (p = 0.025). Interestingly, the enriched classes in the indoor environment were quite different from those in the typical airborne bacterial community environment, except for Clostridia, which had significantly higher abundance in both indoor environments. In conclusion, we found that the two indoor and outdoor airborne bacterial community types changed independently of haze events, and the special airborne bacterial community type was closely related to the incidence of pneumonia in the heavy haze season.
Collapse
Affiliation(s)
- Jianguo Guo
- NHC Key Laboratory of Human Disease Comparative Medicine (The Institute of Laboratory Animal Sciences, CAMS&PUMC), Beijing 100021, China; Key Laboratory of Human Diseases Animal Model, State Administration of Traditional Chinese Medicine, Beijing 100021, China
| | - Yi Xiong
- Department of Food Science and Engineering, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Changhua Shi
- NHC Key Laboratory of Human Disease Comparative Medicine (The Institute of Laboratory Animal Sciences, CAMS&PUMC), Beijing 100021, China; Key Laboratory of Human Diseases Animal Model, State Administration of Traditional Chinese Medicine, Beijing 100021, China
| | - Ce Liu
- Department of Infectious Disease, Beijing Chuiyangliu Hospital, Affiliated with Tsinghua University, Beijing 100022, China
| | - Hongwei Li
- NHC Key Laboratory of Human Disease Comparative Medicine (The Institute of Laboratory Animal Sciences, CAMS&PUMC), Beijing 100021, China; Key Laboratory of Human Diseases Animal Model, State Administration of Traditional Chinese Medicine, Beijing 100021, China
| | - Hua Qian
- School of Energy and Environment, Southeast University, Nanjing 211189, China
| | - Zongke Sun
- National Institute of Environmental Health, China CDC, Beijing 100021, China
| | - Chuan Qin
- NHC Key Laboratory of Human Disease Comparative Medicine (The Institute of Laboratory Animal Sciences, CAMS&PUMC), Beijing 100021, China; Key Laboratory of Human Diseases Animal Model, State Administration of Traditional Chinese Medicine, Beijing 100021, China.
| |
Collapse
|
14
|
Danmaliki GI, Hwang PM. Solution NMR spectroscopy of membrane proteins. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183356. [PMID: 32416193 DOI: 10.1016/j.bbamem.2020.183356] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 05/08/2020] [Accepted: 05/10/2020] [Indexed: 02/06/2023]
Abstract
Integral membrane proteins (IMPs) perform unique and indispensable functions in the cell, making them attractive targets for fundamental research and drug discovery. Developments in protein production, isotope labeling, sample preparation, and pulse sequences have extended the utility of solution NMR spectroscopy for studying IMPs with multiple transmembrane segments. Here we review some recent applications of solution NMR for studying structure, dynamics, and interactions of polytopic IMPs, emphasizing strategies used to overcome common technical challenges.
Collapse
Affiliation(s)
- Gaddafi I Danmaliki
- Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Peter M Hwang
- Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada; Department of Medicine, University of Alberta, Edmonton, Alberta T6G 2H7, Canada.
| |
Collapse
|
15
|
Dickinson RS, Murphy F, Doherty C, Williams S, Mirchandani A, Willson J, Scotti JS, Preston G, Schofield CJ, Whyte MK, Walmsley SR. Pseudomonas expression of an oxygen sensing prolyl hydroxylase homologue regulates neutrophil host responses in vitro and in vivo. Wellcome Open Res 2017; 2:104. [PMID: 29387803 PMCID: PMC5701443 DOI: 10.12688/wellcomeopenres.12871.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2017] [Indexed: 12/13/2022] Open
Abstract
Background: Pseudomonas species are adapted to evade innate immune responses and can persist at sites of relative tissue hypoxia, including the mucus-plugged airways of patients with cystic fibrosis and bronchiectasis. The ability of these bacteria to directly sense and respond to changes in local oxygen availability is in part consequent upon expression of the 2-oxoglutarate oxygenase, Pseudomonas prolyl hydroxylase (PPHD), which acts on elongation factor Tu (EF-Tu), and is homologous with the human hypoxia inducible factor (HIF) prolyl hydroxylases. We report that PPHD expression regulates the neutrophil response to acute pseudomonal infection. Methods:In vitro co-culture experiments were performed with human neutrophils and PPHD-deficient and wild-type bacteria and supernatants, with viable neutrophil counts determined by flow cytometry. In vivo consequences of infection with PPHD deficient P. aeruginosa were determined in an acute pneumonia mouse model following intra-tracheal challenge. Results: Supernatants of PPHD-deficient bacterial cultures contained higher concentrations of the phenazine exotoxin pyocyanin and induced greater acceleration of neutrophil apoptosis than wild-type PAO1 supernatants in vitro. In vivo infection with PPHD mutants compared to wild-type PAO1 controls resulted in increased levels of neutrophil apoptosis and impaired control of infection, with higher numbers of P. aeruginosa recovered from the lungs of mice infected with the PPHD-deficient strain. This resulted in an overall increase in mortality in mice infected with the PPHD-deficient strain. Conclusions: Our data show that Pseudomonas expression of its prolyl hydroxylase influences the outcome of host-pathogen interactions in vitro and in vivo, demonstrating the importance of considering how both host and pathogen adaptations to hypoxia together define outcomes of infection. Given that inhibitors for the HIF prolyl hydroxylases are in late stage trials for the treatment of anaemia and that the active sites of PPHD and human HIF prolyl hydroxylases are closely related, the results are of current clinical interest.
Collapse
Affiliation(s)
- Rebecca S. Dickinson
- MRC/University of Edinburgh Centre for Inflammation Research, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Fiona Murphy
- MRC/University of Edinburgh Centre for Inflammation Research, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Catherine Doherty
- MRC/University of Edinburgh Centre for Inflammation Research, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Sam Williams
- MRC/University of Edinburgh Centre for Inflammation Research, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Ananda Mirchandani
- MRC/University of Edinburgh Centre for Inflammation Research, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Joseph Willson
- MRC/University of Edinburgh Centre for Inflammation Research, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - John S. Scotti
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Oxford, OX1 3TA, UK
| | - Gail Preston
- Department of Plant Sciences, University of Oxford, Oxford, OX1 3RB, UK
| | - Christopher J. Schofield
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Oxford, OX1 3TA, UK
| | - Moira K.B. Whyte
- MRC/University of Edinburgh Centre for Inflammation Research, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Sarah R. Walmsley
- MRC/University of Edinburgh Centre for Inflammation Research, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| |
Collapse
|
16
|
Yang CY, Lee CH, Hsieh CC, Ko WC, Lee CC. Etiology of community-onset monomicrobial bacteremic pneumonia and its clinical presentation and outcome: Klebsiella and Pseudomonas matters. J Infect Chemother 2017; 24:53-58. [PMID: 29017829 DOI: 10.1016/j.jiac.2017.08.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 08/10/2017] [Accepted: 08/31/2017] [Indexed: 10/18/2022]
Abstract
To describe the difference of the clinical features, bacteremia severity, and outcome of patient with community-onset bacteremic pneumonia between Pseudomonas, Klebsiella, and other causative microorganisms, the total 278 adults with community-onset monomicrobial bacteremic pneumonia were studied in a retrospective cohort. Klebsiella (61 patients, 21.9%) and Pseudomonas (22, 7.9%) species was the leading and the fifth common pathogen, respectively. More patients having initial presentation with critical illness (a Pitt bacteremia score ≥ 4) and a fatal comorbidity (McCabe classification) as well as a higher short- (30-day) or long-term (90-day) mortality rate was evidenced in patients infected with Klebsiella or Pseudomonas species, compared to other causative microorganisms. Compared to patients in the Klebsiella group, more frequencies of recent chemotherapy and an initial presentation of febrile neutropenia, and less proportions of diabetes mellitus were disclosed among those in the Pseudomonas group. Of importance, a significantly differential survival curve between Klebsiella or Pseudomonas species and other species during 30-day or 90-day period after bacteremia onset but a similarity of Pseudomonas and Klebsiella species was evidenced, using the Cox-regression after adjusting the independent predictors of 30-day mortality. Conclusively, of pathogens causing community-onset bacteremic pneumonia, Klebsiella and Pseudomonas species should be recognized as the highly virulent pathogens and resulted in poor short- and long-term prognoses.
Collapse
Affiliation(s)
- Chao-Yung Yang
- Department of Emergency Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan
| | - Chung-Hsun Lee
- Department of Emergency Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan; Department of Medicine, College of Medicine, National Cheng Kung University, 70403, Tainan, Taiwan
| | - Chih-Chia Hsieh
- Department of Emergency Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan
| | - Wen-Chien Ko
- Department of Medicine, College of Medicine, National Cheng Kung University, 70403, Tainan, Taiwan; Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan.
| | - Ching-Chi Lee
- Department of Emergency Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan; Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan; Division of Critical Care Medicine, Department of Internal Medicine, Madou Sin-Lau Hospital, Tainan 72152, Taiwan; Graduate Institute of Medical Sciences, College of Health Sciences, Chang Jung Christian University, Tainan 71101, Taiwan.
| |
Collapse
|
17
|
Gao L, Guo Z, Wang Y, Wang Y, Wang K, Li B, Shen L. The Two-Operon-Coded ABC Transporter Complex FpvWXYZCDEF is Required for Pseudomonas aeruginosa Growth and Virulence Under Iron-Limiting Conditions. J Membr Biol 2017; 251:91-104. [PMID: 28975384 DOI: 10.1007/s00232-017-9979-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Accepted: 08/09/2017] [Indexed: 12/12/2022]
Abstract
Iron is essential for all organisms. Bacteria have devolved sophisticated systems to maintain intracellular iron homeostasis. FpvCDEF(PA2407-2410) has been reported as an ABC transporter involved in pyoverdine-Fe uptake which does not affect growth under iron-limiting condition, when it is deleted in PAO1. In this study, we proved that fpvCDEF and fpvWXYZ(PA2403-2406) constituted an ABC transporter complex containing two operons: fpvWXYZCDE and fpvF. The operon fpvWXYZCDE was regulated by iron negatively and the single gene operon fpvF was constitutively expressed. Inactivation of any one of the components, fpvW, fpvC, fpvD, fpvE, and fpvF, led to increased expression of fpvWXYZCDE suggesting that each component of fpvWXYZCDEF could be involved in iron uptake. The ABC transporter complex encoded by fpvWXYZCDEF plays important roles in growth, oxidative stress resistance, and virulence, since the deletion of fpvWXYZCDEF resulted in defective growth, increased sensitivity to H2O2, and decreased virulence compared with PAO1(ΔfpvCDEF) and the wild type PAO1 under iron-limiting condition.
Collapse
Affiliation(s)
- Lang Gao
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Zisheng Guo
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Yue Wang
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Yulu Wang
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Kerong Wang
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Bo Li
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi'an, 710069, China.
| | - Lixin Shen
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi'an, 710069, China.
| |
Collapse
|
18
|
Rice TC, Pugh AM, Xia BT, Seitz AP, Whitacre BE, Gulbins E, Caldwell CC. Bronchoalveolar Lavage Microvesicles Protect Burn-Injured Mice from Pulmonary Infection. J Am Coll Surg 2017; 225:538-547. [PMID: 28690205 DOI: 10.1016/j.jamcollsurg.2017.06.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 06/23/2017] [Accepted: 06/23/2017] [Indexed: 12/27/2022]
Abstract
BACKGROUND Pseudomonas aeruginosa is a major cause of morbidity and mortality among burn patients, despite antibiotic therapy. There is a need to identify innate immune defenses that prevent P aeruginosa infection in injured adults in an effort to find therapeutic alternatives to antibiotics. Here, we tested our hypothesis that microvesicles (MVs) in bronchoalveolar (BAL) fluid have a role in the immunity of the lung in response to pathogens. STUDY DESIGN Microvesicles were isolated from murine BAL fluid, quantified using Nanoparticle Tracking Analysis, and injected into burn-injured mice before P aeruginosa infection. Survival was assessed and BAL bacterial loads enumerated. Neutrophil number and interleukin 6 activity were determined. Lungs were harvested and sphingosine (SPH) content analyzed via immunohistochemistry. Antimicrobial effects of MVs and SPH-enriched MVs were assessed in an in vitro assay. RESULTS Burn-injured mice have reduced BAL MV number and SPH content compared with sham. When BAL MVs from healthy mice are administered to injured mice, survival and bacterial clearance are improved robustly. We also observed that intranasal administration of MVs restores SPH levels after burn injury, MVs kill bacteria directly, and this bacterial killing is increased when the MVs are supplemented with SPH. CONCLUSIONS Using a preclinical model, BAL MVs are reduced after scald injury and BAL MV restoration to injured mice improves survival and bacterial clearance. The antimicrobial mechanisms leading to improved survival include the quantity and SPH content of BAL MVs.
Collapse
Affiliation(s)
- Teresa C Rice
- Division of Research, Department of Surgery, University of Cincinnati, Cincinnati, OH
| | - Amanda M Pugh
- Division of Research, Department of Surgery, University of Cincinnati, Cincinnati, OH
| | - Brent T Xia
- Division of Research, Department of Surgery, University of Cincinnati, Cincinnati, OH
| | - Aaron P Seitz
- Division of Research, Department of Surgery, University of Cincinnati, Cincinnati, OH
| | - Brynne E Whitacre
- Division of Research, Department of Surgery, University of Cincinnati, Cincinnati, OH
| | - Erich Gulbins
- Division of Research, Department of Surgery, University of Cincinnati, Cincinnati, OH; Department of Molecular Biology, University of Duisburg-Essen, Essen, Germany
| | - Charles C Caldwell
- Division of Research, Department of Surgery, University of Cincinnati, Cincinnati, OH.
| |
Collapse
|
19
|
Balczon R, Morrow KA, Zhou C, Edmonds B, Alexeyev M, Pittet JF, Wagener BM, Moser SA, Leavesley S, Zha X, Frank DW, Stevens T. Pseudomonas aeruginosa infection liberates transmissible, cytotoxic prion amyloids. FASEB J 2017; 31:2785-2796. [PMID: 28314768 DOI: 10.1096/fj.201601042rr] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 02/26/2017] [Indexed: 12/28/2022]
Abstract
Patients who recover from pneumonia subsequently have elevated rates of death after hospital discharge as a result of secondary organ damage, the causes of which are unknown. We used the bacterium Pseudomonas aeruginosa, a common cause of hospital-acquired pneumonia, as a model for investigating this phenomenon. We show that infection of pulmonary endothelial cells by P. aeruginosa induces production and release of a cytotoxic amyloid molecule with prion characteristics, including resistance to various nucleases and proteases. This cytotoxin was self-propagating, was neutralized by anti-amyloid Abs, and induced death of endothelial cells and neurons. Moreover, the cytotoxin induced edema in isolated lungs. Endothelial cells and isolated lungs were protected from cytotoxin-induced death by stimulation of signal transduction pathways that are linked to prion protein. Analysis of bronchoalveolar lavage fluid collected from human patients with P. aeruginosa pneumonia demonstrated cytotoxic activity, and lavage fluid contained amyloid molecules, including oligomeric τ and Aβ. Demonstration of long-lived cytotoxic agents after Pseudomonas infection may establish a molecular link to the high rates of death as a result of end-organ damage in the months after recovery from pneumonia, and modulation of signal transduction pathways that have been linked to prion protein may provide a mechanism for intervention.-Balczon, R., Morrow, K. A., Zhou, C., Edmonds, B., Alexeyev, M., Pittet, J.-F., Wagener, B. M., Moser, S. A., Leavesley, S., Zha, X., Frank, D. W., Stevens, T. Pseudomonas aeruginosa infection liberates transmissible, cytotoxic prion amyloids.
Collapse
Affiliation(s)
- Ron Balczon
- Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA; .,Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| | - K Adam Morrow
- Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA.,Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| | - Chun Zhou
- Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA.,Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| | - Bradley Edmonds
- Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| | - Mikhail Alexeyev
- Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA.,Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| | - Jean-Francois Pittet
- Department of Anesthesia and Perioperative Medicine, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, USA
| | - Brant M Wagener
- Department of Anesthesia and Perioperative Medicine, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, USA
| | - Stephen A Moser
- Department of Pathology, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, USA
| | - Silas Leavesley
- Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA.,Department of Chemical and Biomedical Engineering, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| | - Xiangming Zha
- Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA.,Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| | - Dara W Frank
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Troy Stevens
- Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA.,Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| |
Collapse
|
20
|
Capasso D, Pepe MV, Rossello J, Lepanto P, Arias P, Salzman V, Kierbel A. Elimination of Pseudomonas aeruginosa through Efferocytosis upon Binding to Apoptotic Cells. PLoS Pathog 2016; 12:e1006068. [PMID: 27977793 PMCID: PMC5158079 DOI: 10.1371/journal.ppat.1006068] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 11/15/2016] [Indexed: 02/03/2023] Open
Abstract
For opportunistic pathogens such as Pseudomonas aeruginosa, the mucosal barrier represents a formidable challenge. Infections develop only in patients with altered epithelial barriers. Here, we showed that P. aeruginosa interacts with a polarized epithelium, adhering almost exclusively at sites of multi-cellular junctions. In these sites, numerous bacteria attach to an extruded apoptotic cell or apoptotic body. This dead cell tropism is independent of the type of cell death, as P. aeruginosa also binds to necrotic cells. We further showed that P. aeruginosa is internalized through efferocytosis, a process in which surrounding epithelial cells engulf and dispose of extruded apoptotic cells. Intracellularly, along with apoptotic cell debris, P. aeruginosa inhabits an efferocytic phagosome that acquires lysosomal features, and is finally killed. We propose that elimination of P. aeruginosa through efferocytosis is part of a host defense mechanism. Our findings could be relevant for the study of cystic fibrosis, which is characterized by an exacerbated number of apoptotic cells and ineffective efferocytosis. Pseudomonas aeruginosa is an opportunistic pathogen that infects vulnerable patients, such as those with cystic fibrosis or hospitalized in intensive care units. An advance towards understanding infections caused by P. aeruginosa would be to fully elucidate the mechanisms that operate in the bacteria-epithelial barrier interplay. Here, we showed that P. aeruginosa exhibits a remarkable tropism towards dead cells. As bacteria interact with a polarized epithelium, they attach and aggregate almost exclusively on apoptotic cells extruded from the epithelium, while the rest of the surface seems reluctant to bacterial adhesion. We further showed that P. aeruginosa is internalized by epithelial cells surrounding the infected apoptotic cell through efferocytosis, a process in which apoptotic cells are engulfed and disposed of by other cells. Bacteria are eliminated intracellularly. Our findings may help to understand why contexts such as cystic fibrosis, where apoptotic cells are unusually produced and efferocytosis fails, favor P. aeruginosa colonization.
Collapse
Affiliation(s)
- Darío Capasso
- Instituto de Investigaciones Biotecnológicas Dr. Rodolfo A. Ugalde (IIB-INTECH), Universidad Nacional de San Martín, Consejo Nacional de Investigaciones Científicas y Técnicas (UNSAM-CONICET), San Martín, Buenos Aires, Argentina
| | - María Victoria Pepe
- Instituto de Investigaciones Biotecnológicas Dr. Rodolfo A. Ugalde (IIB-INTECH), Universidad Nacional de San Martín, Consejo Nacional de Investigaciones Científicas y Técnicas (UNSAM-CONICET), San Martín, Buenos Aires, Argentina
| | | | | | - Paula Arias
- Instituto de Investigaciones Biotecnológicas Dr. Rodolfo A. Ugalde (IIB-INTECH), Universidad Nacional de San Martín, Consejo Nacional de Investigaciones Científicas y Técnicas (UNSAM-CONICET), San Martín, Buenos Aires, Argentina
| | - Valentina Salzman
- Instituto de Investigaciones Biotecnológicas Dr. Rodolfo A. Ugalde (IIB-INTECH), Universidad Nacional de San Martín, Consejo Nacional de Investigaciones Científicas y Técnicas (UNSAM-CONICET), San Martín, Buenos Aires, Argentina
| | - Arlinet Kierbel
- Instituto de Investigaciones Biotecnológicas Dr. Rodolfo A. Ugalde (IIB-INTECH), Universidad Nacional de San Martín, Consejo Nacional de Investigaciones Científicas y Técnicas (UNSAM-CONICET), San Martín, Buenos Aires, Argentina
- * E-mail:
| |
Collapse
|
21
|
Khailova L, Baird CH, Rush AA, Barnes C, Wischmeyer PE. Lactobacillus rhamnosus GG treatment improves intestinal permeability and modulates inflammatory response and homeostasis of spleen and colon in experimental model of Pseudomonas aeruginosa pneumonia. Clin Nutr 2016; 36:1549-1557. [PMID: 27745813 DOI: 10.1016/j.clnu.2016.09.025] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Revised: 09/21/2016] [Accepted: 09/25/2016] [Indexed: 12/21/2022]
Abstract
BACKGROUND & AIMS Recent clinical trials and in vivo models demonstrate probiotic administration can reduce occurrence and improve outcome of pneumonia and sepsis, both major clinical challenges worldwide. Potential probiotic benefits include maintenance of gut epithelial barrier homeostasis and prevention of downstream organ dysfunction due to systemic inflammation. However, mechanism(s) of probiotic-mediated protection against pneumonia remain poorly understood. This study evaluated potential mechanistic targets in the maintenance of gut barrier homeostasis following Lactobacillus rhamnosus GG (LGG) treatment in a mouse model of pneumonia. METHODS Studies were performed in 6-8 week old FVB/N mice treated (o.g.) with or without LGG (109 CFU/ml) and intratracheally injected with Pseudomonas aeruginosa or saline. At 4, 12, and 24 h post-bacterial treatment spleen and colonic tissue were collected for analysis. RESULTS Pneumonia significantly increased intestinal permeability and gut claudin-2. LGG significantly attenuated increased gut permeability and claudin-2 following pneumonia back to sham control levels. As mucin expression is key to gut barrier homeostasis we demonstrate that LGG can enhance goblet cell expression and mucin barrier formation versus control pneumonia animals. Further as Muc2 is a key gut mucin, we show LGG corrected deficient Muc2 expression post-pneumonia. Apoptosis increased in both colon and spleen post-pneumonia, and this increase was significantly attenuated by LGG. Concomitantly, LGG corrected pneumonia-mediated loss of cell proliferation in colon and significantly enhanced cell proliferation in spleen. Finally, LGG significantly reduced pro-inflammatory cytokine gene expression in colon and spleen post-pneumonia. CONCLUSIONS These data demonstrate LGG can maintain intestinal barrier homeostasis by enhancing gut mucin expression/barrier formation, reducing apoptosis, and improving cell proliferation. This was accompanied by reduced pro-inflammatory cytokine expression in the gut and in a downstream organ (spleen). These may serve as potential mechanistic targets to explain LGG's protection against pneumonia in the clinical and in vivo setting.
Collapse
Affiliation(s)
- Ludmila Khailova
- Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Christine H Baird
- Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Aubri A Rush
- Valparaiso University, Valparaiso, IN, 46383, USA
| | | | - Paul E Wischmeyer
- Department of Anesthesiology, Duke University School of Medicine, Durham, NC, 27710, USA.
| |
Collapse
|
22
|
Banadkoki AZ, Keshavarzmehr M, Afshar Z, Aleyasin N, Fatemi MJ, Behrouz B, Hashemi FB. Protective effect of pilin protein with alum+naloxone adjuvant against acute pulmonary Pseudomonas aeruginosa infection. Biologicals 2016; 44:367-73. [PMID: 27427517 DOI: 10.1016/j.biologicals.2016.06.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 05/29/2016] [Accepted: 06/21/2016] [Indexed: 11/26/2022] Open
Abstract
Pseudomonas aeruginosa is an important opportunistic human pathogen that causes a wide variety of severe nosocomial infections. Type IV pili of P. aeruginosa are made up of polymerized pilin that aids in bacterial adhesion, biofilm formation and twitching motility. The aim of this study was to evaluate the efficacy of alum and naloxone (alum+NLX) as an adjuvant for P. aeruginosa recombinant PilA (r-PilA) as a vaccine candidate in the improvement of humoral and cellular immunity. Primary immunization with r-PilA in combination with alum+NLX followed by two booster shots was sufficient to generate robust cellular and humoral responses, which were Th1 and Th2 type responses consisting of IgG1 and IgG2a subtypes. Analysis of the cytokine response among immunized mice showed an increased production of IL-4, INF-γ and IL-17 by splenocytes upon stimulation by r-PilA. These sera were also able to reduce bacterial load in the lung tissue of challenged mice. The reduction of systemic bacterial spread resulted in increased survival rates in challenged immunized mice. In conclusion, immunization with r-PilA combined with alum+NLX evokes cellular and humoral immune responses, which play an important role in providing protection against acute P. aeruginosa lung infection among immunized mice.
Collapse
Affiliation(s)
- Abbas Zare Banadkoki
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Quality Control Department of Iran Gelatin Capsule Mfg. Co., Tehran, Iran
| | | | - Zahra Afshar
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Neda Aleyasin
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Javad Fatemi
- Burn Research Center, Hazrat Fatima Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Bahador Behrouz
- Burn Research Center, Hazrat Fatima Hospital, Iran University of Medical Sciences, Tehran, Iran; Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Farhad B Hashemi
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
23
|
Immunogenicity and protective efficacy of Pseudomonas aeruginosa type a and b flagellin vaccines in a burned mouse model. Mol Immunol 2016; 74:71-81. [DOI: 10.1016/j.molimm.2016.04.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Revised: 04/07/2016] [Accepted: 04/14/2016] [Indexed: 12/31/2022]
|
24
|
Korpi F, Hashemi FB, Irajian G, Fatemi MJ, Laghaei P, Behrouz B. Flagellin and pilin immunization against multi-drug resistant Pseudomonas aeruginosa protects mice in the burn wound sepsis model. Immunol Lett 2016; 176:8-17. [PMID: 27210422 DOI: 10.1016/j.imlet.2016.04.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 03/26/2016] [Accepted: 04/02/2016] [Indexed: 01/15/2023]
Abstract
Pseudomonas aeruginosa is a formidable pathogen and a major threat to burn patients. Antimicrobial therapy is often unsuccessful because P. aeruginosa can develop multi-drug resistance; thus, immunotherapy and vaccine can be a rational alternative. Flagella and type IV pili have been identified as important virulence factors in the colonization and pathogenesis of P. aeruginosa in burn wound infections. Immunogenicity and efficacy of mixed recombinant full-length type b flagellin (r-b-flagellin) and recombinant PilA (r-PilA) as candidate vaccines were assessed by measuring humoral and cellular responses, using an experimental burned mouse model. Primary immunization with "r-b-flagellin+r-PilA" followed by two booster shots was sufficient to generate a robust humoral response, which was predominantly a Th2 response consisting mainly of subtype IgG1 and low levels of IgG2a. Analysis of the cytokine response among immunized mice showed an increased production of IL-4, INF-γ and IL-17 by splenocytes upon stimulation by "r-b-flagellin+r-PilA". Opsonophagocytosis assays confirmed the enhanced killing of bacteria by anti "r-b-flagellin+r-PilA" immune sera. These antibodies were also able to reduce bacterial load in the site of original infection into the liver and spleen of challenged mice. The reduction of systemic bacterial spread resulted in an increased survival rate of challenged immunized mice. In conclusion, immunization with "r-b-flagellin+r-PilA" proteins provides a better protective response against P. aeruginosa infection in the burn mouse model.
Collapse
Affiliation(s)
- Fatemeh Korpi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Farhad B Hashemi
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Gholamreza Irajian
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Mohammad Javad Fatemi
- Burn Research Center, Motahari Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Parisa Laghaei
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Bahador Behrouz
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Burn Research Center, Motahari Hospital, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
25
|
Yue L, Xie Z, Li H, Pang Z, Junkins RD, Tremblay ML, Chen X, Lin TJ. Protein Tyrosine Phosphatase-1B Negatively Impacts Host Defense against Pseudomonas aeruginosa Infection. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:1234-44. [DOI: 10.1016/j.ajpath.2016.01.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 12/26/2015] [Accepted: 01/05/2016] [Indexed: 11/26/2022]
|
26
|
|
27
|
Pathophysiological changes induced by Pseudomonas aeruginosa infection are involved in MMP-12 and MMP-13 upregulation in human carcinoma epithelial cells and a pneumonia mouse model. Infect Immun 2015; 83:4791-9. [PMID: 26438797 PMCID: PMC4645383 DOI: 10.1128/iai.00619-15] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 09/18/2015] [Indexed: 12/20/2022] Open
Abstract
Pseudomonas aeruginosa infections persist in patients with cystic fibrosis (CF) and drive lung disease progression. P. aeruginosa potently activates the innate immune system mostly through the recognition of pathogen-associated molecular patterns, such as flagellin. Matrix metalloproteinases 12 and 13 (MMP-12 and MMP-13, respectively) exacerbate chronic lung infection and inflammation by promoting uncontrolled tissue rearrangements and fibrosis, yet the underlying molecular mechanisms by which this occurs remain largely unknown. In this study, we used quantitative bacteriology, histological examination, and proinflammatory cytokine levels to evaluate the effects of MMP-12 and MMP-13 on P. aeruginosa strain K-induced infection and pneumonia in H292 epithelial cells and mice, respectively. Under inflammatory stimulation, mRNA and protein expression levels of proinflammatory mediators were higher in strain K-infected mice and cells than in uninfected counterparts, in which MMP-12 and MMP-13 expression reached levels similar to those observed in epithelial cells. Moreover, we also found that the NF-κB pathway might be involved in the induction of cytokines in response to strain K infection. Taken together, these data suggest that MMP-12 and MMP-13 alter strain K infection in mice and play a role in inflammatory regulation by modulating cytokine levels.
Collapse
|
28
|
Shrestha M, Xiao Y, Robinson H, Schubot FD. Structural Analysis of the Regulatory Domain of ExsA, a Key Transcriptional Regulator of the Type Three Secretion System in Pseudomonas aeruginosa. PLoS One 2015; 10:e0136533. [PMID: 26317977 PMCID: PMC4552939 DOI: 10.1371/journal.pone.0136533] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 08/04/2015] [Indexed: 12/30/2022] Open
Abstract
Pseudomonas aeruginosa employs a type three secretion system to facilitate infections in mammalian hosts. The operons encoding genes of structural components of the secretion machinery and associated virulence factors are all under the control of the AraC-type transcriptional activator protein, ExsA. ExsA belongs to a unique subfamily of AraC-proteins that is regulated through protein-protein contacts rather than small molecule ligands. Prior to infection, ExsA is inhibited through a direct interaction with the anti-activator ExsD. To activate ExsA upon host cell contact this interaction is disrupted by the anti-antiactivator protein ExsC. Here we report the crystal structure of the regulatory domain of ExsA, which is known to mediate ExsA dimerization as well as ExsD binding. The crystal structure suggests two models for the ExsA dimer. Both models confirmed the previously shown involvement of helix α-3 in ExsA dimerization but one also suggest a role for helix α-2. These structural data are supported by the observation that a mutation in α-2 greatly diminished the ability of ExsA to activate transcription in vitro. Additional in vitro transcription studies revealed that a conserved pocket, used by AraC and the related ToxT protein for the binding of small molecule regulators, although present in ExsA is not involved in binding of ExsD.
Collapse
Affiliation(s)
- Manisha Shrestha
- Department of Biological Sciences, Virginia Polytechnic Institute & State University, Washington Street, Blacksburg, VA 24060, United States of America
| | - Yi Xiao
- Department of Biological Sciences, Virginia Polytechnic Institute & State University, Washington Street, Blacksburg, VA 24060, United States of America
| | - Howard Robinson
- Biology Department, Brookhaven National Laboratory, Upton, NY 11973–5000, United States of America
| | - Florian D. Schubot
- Department of Biological Sciences, Virginia Polytechnic Institute & State University, Washington Street, Blacksburg, VA 24060, United States of America
- * E-mail:
| |
Collapse
|
29
|
José Maschio V, Corção G, Rott MB. identification of Pseudomonas spp. as amoeba-resistant microorganisms in isolates of Acanthamoeba. Rev Inst Med Trop Sao Paulo 2015; 57:81-3. [PMID: 25651331 PMCID: PMC4325528 DOI: 10.1590/s0036-46652015000100012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 03/28/2014] [Indexed: 11/22/2022] Open
Abstract
Acanthamoeba is a “Trojan horse” of the microbial world. The aim of
this study was to identify the presence of Pseudomonas as an
amoeba-resistant microorganism in 12 isolates of Acanthamoeba. All
isolates showed the genus Pseudomonas spp. as amoeba-resistant
microorganisms. Thus, one can see that the Acanthamoeba isolates
studied are hosts of Pseudomonas.
Collapse
Affiliation(s)
- Vinicius José Maschio
- Departamento de Microbiologia, Imunologia e Parasitologia. Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Gertrudes Corção
- Departamento de Microbiologia, Imunologia e Parasitologia. Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Marilise Brittes Rott
- Departamento de Microbiologia, Imunologia e Parasitologia. Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
30
|
Discovery of bacterial fatty acid synthase type II inhibitors using a novel cellular bioluminescent reporter assay. Antimicrob Agents Chemother 2015; 59:5775-87. [PMID: 26169404 DOI: 10.1128/aac.00686-15] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 07/06/2015] [Indexed: 12/11/2022] Open
Abstract
Novel, cellular, gain-of-signal, bioluminescent reporter assays for fatty acid synthesis type II (FASII) inhibitors were constructed in an efflux-deficient strain of Pseudomonas aeruginosa and based on the discovery that FASII genes in P. aeruginosa are coordinately upregulated in response to pathway disruption. A screen of 115,000 compounds identified a series of sulfonamidobenzamide (SABA) analogs, which generated strong luminescent signals in two FASII reporter strains but not in four control reporter strains designed to respond to inhibitors of pathways other than FASII. The SABA analogs selectively inhibited lipid biosynthesis in P. aeruginosa and exhibited minimal cytotoxicity to mammalian cells (50% cytotoxic concentration [CC50] ≥ 80 μM). The most potent SABA analogs had MICs of 0.5 to 7.0 μM (0.2 to 3.0 μg/ml) against an efflux-deficient Escherichia coli (ΔtolC) strain but had no detectable MIC against efflux-proficient E. coli or against P. aeruginosa (efflux deficient or proficient). Genetic, molecular genetic, and biochemical studies revealed that SABA analogs target the enzyme (AccC) catalyzing the biotin carboxylase half-reaction of the acetyl coenzyme A (acetyl-CoA) carboxylase step in the initiation phase of FASII in E. coli and P. aeruginosa. These results validate the capability and the sensitivity of this novel bioluminescent reporter screen to identify inhibitors of E. coli and P. aeruginosa FASII.
Collapse
|
31
|
Saturni S, Contoli M, Spanevello A, Papi A. Models of Respiratory Infections: Virus-Induced Asthma Exacerbations and Beyond. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2015; 7:525-33. [PMID: 26333698 PMCID: PMC4605924 DOI: 10.4168/aair.2015.7.6.525] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 04/06/2015] [Indexed: 12/20/2022]
Abstract
Respiratory infections are one of the main health problems worldwide. They are a challenging field of study due to an intricate relationship between the pathogenicity of microbes and the host's defenses. To better understand mechanisms of respiratory infections, different models have been developed. A model is the reproduction of a disease in a system that mimics human pathophysiology. For this reason, the best models should closely resemble real-life conditions. Thus, the human model is the best. However, human models of respiratory infections have some disadvantages that limit their role. Therefore, other models, including animal, in vitro, and mathematical ones, have been developed. We will discuss advantages and limitations of available models and focus on models of viral infections as triggers of asthma exacerbations, viral infections being one of the most frequent causes of exacerbating disease. Future studies should focus on the interrelation of various models.
Collapse
Affiliation(s)
- Sara Saturni
- Section of Respiratory Medicine, University of Ferrara, Ferrara, Italy
| | - Marco Contoli
- Section of Respiratory Medicine, University of Ferrara, Ferrara, Italy
| | - Antonio Spanevello
- Department of Respiratory Diseases, Fondazione Maugeri, Tradate, University of Varese, Italy
| | - Alberto Papi
- Section of Respiratory Medicine, University of Ferrara, Ferrara, Italy.
| |
Collapse
|
32
|
Vakulskas CA, Potts AH, Babitzke P, Ahmer BMM, Romeo T. Regulation of bacterial virulence by Csr (Rsm) systems. Microbiol Mol Biol Rev 2015; 79:193-224. [PMID: 25833324 PMCID: PMC4394879 DOI: 10.1128/mmbr.00052-14] [Citation(s) in RCA: 248] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Most bacterial pathogens have the remarkable ability to flourish in the external environment and in specialized host niches. This ability requires their metabolism, physiology, and virulence factors to be responsive to changes in their surroundings. It is no surprise that the underlying genetic circuitry that supports this adaptability is multilayered and exceedingly complex. Studies over the past 2 decades have established that the CsrA/RsmA proteins, global regulators of posttranscriptional gene expression, play important roles in the expression of virulence factors of numerous proteobacterial pathogens. To accomplish these tasks, CsrA binds to the 5' untranslated and/or early coding regions of mRNAs and alters translation, mRNA turnover, and/or transcript elongation. CsrA activity is regulated by noncoding small RNAs (sRNAs) that contain multiple CsrA binding sites, which permit them to sequester multiple CsrA homodimers away from mRNA targets. Environmental cues sensed by two-component signal transduction systems and other regulatory factors govern the expression of the CsrA-binding sRNAs and, ultimately, the effects of CsrA on secretion systems, surface molecules and biofilm formation, quorum sensing, motility, pigmentation, siderophore production, and phagocytic avoidance. This review presents the workings of the Csr system, the paradigm shift that it generated for understanding posttranscriptional regulation, and its roles in virulence networks of animal and plant pathogens.
Collapse
Affiliation(s)
- Christopher A Vakulskas
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, USA
| | - Anastasia H Potts
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, USA
| | - Paul Babitzke
- Department of Biochemistry and Molecular Biology, Center for RNA Molecular Biology, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Brian M M Ahmer
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
| | - Tony Romeo
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
33
|
Velkov T, Abdul Rahim N, Zhou Q(T, Chan HK, Li J. Inhaled anti-infective chemotherapy for respiratory tract infections: successes, challenges and the road ahead. Adv Drug Deliv Rev 2015; 85:65-82. [PMID: 25446140 PMCID: PMC4429008 DOI: 10.1016/j.addr.2014.11.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 11/04/2014] [Accepted: 11/05/2014] [Indexed: 12/31/2022]
Abstract
One of the most common causes of illnesses in humans is from respiratory tract infections caused by bacterial, viral or fungal pathogens. Inhaled anti-infective drugs are crucial for the prophylaxis and treatment of respiratory tract infections. The benefit of anti-infective drug delivery via inhalation is that it affords delivery of sufficient therapeutic dosages directly to the primary site of infection, while minimizing the risks of systemic toxicity or avoiding potential suboptimal pharmacokinetics/pharmacodynamics associated with systemic drug exposure. This review provides an up-to-date treatise of approved and novel developmental inhaled anti-infective agents, with particular attention to effective strategies for their use, pulmonary pharmacokinetic properties and safety.
Collapse
|
34
|
Toval F, Guzmán-Marte A, Madriz V, Somogyi T, Rodríguez C, García F. Predominance of carbapenem-resistant Pseudomonas aeruginosa isolates carrying blaIMP and blaVIM metallo-β-lactamases in a major hospital in Costa Rica. J Med Microbiol 2014; 64:37-43. [PMID: 25355933 DOI: 10.1099/jmm.0.081802-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
This study aimed to assess the molecular basis of the resistance to carbapenems in clinical isolates of Pseudomonas aeruginosa recovered from a tertiary-level health facility in San José, Costa Rica. A total of 198 non-duplicated isolates were evaluated for their susceptibility to β-lactams, aminoglycosides and fluoroquinolones. The production of metallo-β-lactamases (MBLs), the presence of MBL encoding genes (blaIMP, blaVIM and blaGIM-1) and the occurrence of these genes within class 1 integrons were investigated. In addition, an ERIC2 PCR fingerprinting method was used to elucidate the distribution of the detected MBL genes within the strain collection. Of the 198 isolates tested, 125 (63.1 %) were categorized as carbapenem-resistant. The majority (88.8 %) of the carbapemen-resistant isolates also showed resistance to ceftazidime, cefepime, aztreonam, ticarcillin/clavulanic acid, amikacin, gentamicin, tobramycin, ciprofloxacin and gatifloxacin. Among the carbapenem-resistant isolates, 102 (81.6 %) showed MBL activity. Strikingly, both blaIMP and blaVIM genes were simultaneously detected in most (94.1 %) of the 102 MBL producers. Five carbapenem-resistant MBL producers were positive only for blaIMP genes. Almost 70 % of the isolates examined harboured the intI1 gene, accompanied by the sul1 and qacEΔ1 genes in 136 (99 %) and 122 (89 %) isolates, respectively. The majority (94.4 %) of the carbapenem-resistant isolates carried the intI1 gene, in contrast to 26 % of the carbapenem-susceptible isolates. Ninety-three out of 96 (96.9 %) isolates carrying both blaIMP and blaVIM genes also harboured the intI1, sul1 and qacEΔ1 genes. Gene cassettes from carbapenem-susceptible and MBL-negative carbapenem-resistant isolates encoded aminoglycoside-resistance enzymes (aadA2, aadA4 and aadA6) as well as orfD and qacF genes. RAPD analysis distributed 126 of the isolates in 29 clusters. Eighty of the 90 blaIMP (+) blaVIM (+) isolates were sorted into 16 different clusters, suggesting that the blaIMP and blaVIM genes detected were located within a genetic element capable of lateral transfer. Carbapenem-resistant MBL-positive isolates were recovered from almost all hospital wards and were over-represented in samples obtained from the surgical emergency and intensive care therapy units. Remarkably, three carbapenem-resistant isolates, exhibiting MBL activity and carrying both blaIMP and blaVIM genes, were recovered from outpatients. Sequence analysis of both bla genes in various isolates revealed that they correspond to the alleles blaIMP-18 and blaVIM-2. To our knowledge, this is the first report of the combination of two metallo-β-lactamases encoded by the blaIMP-18 and blaVIM-2 genes in P. aeruginosa.
Collapse
Affiliation(s)
- Francisco Toval
- Centro de Investigación en Enfermedades Tropicales, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Anel Guzmán-Marte
- Centro de Investigación en Enfermedades Tropicales, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Vivian Madriz
- Centro de Investigación en Enfermedades Tropicales, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Teresita Somogyi
- Laboratorio Clínico, Hospital Mexico, San José, Costa Rica.,Centro de Investigación en Enfermedades Tropicales, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - César Rodríguez
- Centro de Investigación en Enfermedades Tropicales, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Fernando García
- Centro de Investigación en Enfermedades Tropicales, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| |
Collapse
|
35
|
Depletion of natural killer cells increases mice susceptibility in a Pseudomonas aeruginosa pneumonia model. Crit Care Med 2014; 42:e441-50. [PMID: 24732238 DOI: 10.1097/ccm.0000000000000311] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Pseudomonas aeruginosa infection is a clinically relevant infection involved in pneumonia in ICUs. Understanding the type of immune response initiated by the host during pneumonia would help defining new strategies to interfere with the bacteria pathogenicity. In this setting, the role of natural killer cells remains controversial. We assessed the role of systemic natural killer cells in a Pseudomonas aeruginosa mouse pneumonia model. DESIGN Experimental study. SETTING Research laboratory from a university hospital. SUBJECTS RjOrl:SWISS and BALB/cJ mice (weight, 20-24 g). INTERVENTIONS Lung injuries were assessed by bacterial load, myeloperoxidase activity, endothelial permeability (pulmonary edema), immune cell infiltrate (histological analysis), proinflammatory cytokine release, and Ly6-G immunohistochemistry. Bacterial loads were assessed in the lungs and spleen. Natural killer cell number and status were assessed in spleen (flow cytometry and quantitative polymerase chain reaction). Depletion of natural killer cells was achieved through an IV anti-asialo-GM1 antibody injection. MEASUREMENTS AND MAIN RESULTS Pseudomonas aeruginosa tracheal instillation led to an acute pneumonia with a rapid decrease of bacterial load in lungs and with an increase of endothelial permeability, proinflammatory cytokines (tumor necrosis factor-α and interleukin-1β), and myeloperoxidase activity followed by Ly6-G positive cell infiltrate in lungs. Pseudomonas aeruginosa was detected in the spleen. Membrane markers of activation and maturation (CD69 and KLRG1 molecules) were increased in splenic natural killer cells during Pseudomonas aeruginosa infection. Splenic natural killer cells activated upon Pseudomonas aeruginosa infection produced interferon-γ but not interleukin-10. Ultimately, mice depleted of natural killer cells displayed an increased neutrophil numbers in the lungs and an increased mortality rate without bacterial load modifications in the lungs, indicating that mice depleted of natural killer cells were much more susceptible to infection compared with control animals. CONCLUSIONS We report for the first time that natural killer cells play a major role in the mice susceptibility toward a Pseudomonas aeruginosa-induced acute pneumonia model.
Collapse
|
36
|
Cytotoxic clinical isolates of Pseudomonas aeruginosa identified during the Steroids for Corneal Ulcers Trial show elevated resistance to fluoroquinolones. BMC Ophthalmol 2014; 14:54. [PMID: 24761794 PMCID: PMC4008435 DOI: 10.1186/1471-2415-14-54] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 04/11/2014] [Indexed: 12/18/2022] Open
Abstract
Background To determine the relationship between type three secretion genotype and fluoroquinolone resistance for P. aeruginosa strains isolated from microbial keratitis during the Steroids for Corneal Ulcers Trial (SCUT) and for two laboratory strains, PA103 and PAO1. Methods Confirmed P. aeruginosa isolates from the SCUT were divided into exoU(+) or exoU(−). The exoU(+) strains contained the gene encoding ExoU, a powerful phospholipase toxin delivered into host cells by the type three secretion system. Isolates were then assessed for susceptibility to fluoroquinolone, cephalosporin, and aminoglycoside antibiotics using disk diffusion assays. Etest was used to determine the MIC of moxifloxacin and other fluoroquinolones. Laboratory isolates in which the exoU gene was added or deleted were also tested. Results A significantly higher proportion of exoU(+) strains were resistant to ciprofloxacin (p = 0.001), gatifloxacin (p = 0.003), and ofloxacin (p = 0.002) compared to exoU(−) isolates. There was no significant difference between exoU(+) or exoU(−) negative isolates with respect to susceptibility to other antibiotics except gentamicin. Infections involving resistant exoU(+) strains trended towards worse clinical outcome. Deletion or acquisition of exoU in laboratory isolates did not affect fluoroquinolone susceptibility. Conclusions Fluoroquinolone susceptibility of P. aeruginosa isolated from the SCUT is consistent with previous studies showing elevated resistance involving exoU encoding (cytotoxic) strains, and suggest worse clinical outcome from infections involving resistant isolates. Determination of exoU expression in clinical isolates of P. aeruginosa may be helpful in directing clinical management of patients with microbial keratitis.
Collapse
|
37
|
Grossi P, Dalla Gasperina D. Treatment ofPseudomonas aeruginosainfection in critically ill patients. Expert Rev Anti Infect Ther 2014; 4:639-62. [PMID: 17009943 DOI: 10.1586/14787210.4.4.639] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Critically ill patients are on the increase in the present clinical setting. Aging of our population and increasingly aggressive medical and therapeutic interventions, including implanted foreign bodies, organ transplantation and advances in the chemotherapy of malignant diseases, have created a cohort of particularly vulnerable patients. Pseudomonas aeruginosa is one of the leading gram-negative organisms associated with nosocomial infections. This organism is frequently feared because it causes severe hospital-acquired infections, especially in immunocompromised hosts, and is often antibiotic resistant, complicating the choice of therapy. The epidemiology, microbiology, mechanisms of resistance and currently available and future treatment options for the most relevant infections caused by P. aeruginosa are reviewed.
Collapse
Affiliation(s)
- Paolo Grossi
- University of Insubria, Infectious Diseases Department, viale Borri 57, 21100 Varese, Italy.
| | | |
Collapse
|
38
|
Audia JP, Lindsey AS, Housley NA, Ochoa CR, Zhou C, Toba M, Oka M, Annamdevula NS, Fitzgerald MS, Frank DW, Alvarez DF. In the absence of effector proteins, the Pseudomonas aeruginosa type three secretion system needle tip complex contributes to lung injury and systemic inflammatory responses. PLoS One 2013; 8:e81792. [PMID: 24312357 PMCID: PMC3842252 DOI: 10.1371/journal.pone.0081792] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Accepted: 10/16/2013] [Indexed: 01/06/2023] Open
Abstract
Herein we describe a pathogenic role for the Pseudomonas aeruginosa type three secretion system (T3SS) needle tip complex protein, PcrV, in causing lung endothelial injury. We first established a model in which P. aeruginosa wild type strain PA103 caused pneumonia-induced sepsis and distal organ dysfunction. Interestingly, a PA103 derivative strain lacking its two known secreted effectors, ExoU and ExoT [denoted PA103 (ΔU/ΔT)], also caused sepsis and modest distal organ injury whereas an isogenic PA103 strain lacking the T3SS needle tip complex assembly protein [denoted PA103 (ΔPcrV)] did not. PA103 (ΔU/ΔT) infection caused neutrophil influx into the lung parenchyma, lung endothelial injury, and distal organ injury (reminiscent of sepsis). In contrast, PA103 (ΔPcrV) infection caused nominal neutrophil infiltration and lung endothelial injury, but no distal organ injury. We further examined pathogenic mechanisms of the T3SS needle tip complex using cultured rat pulmonary microvascular endothelial cells (PMVECs) and revealed a two-phase, temporal nature of infection. At 5-hours post-inoculation (early phase infection), PA103 (ΔU/ΔT) elicited PMVEC barrier disruption via perturbation of the actin cytoskeleton and did so in a cell death-independent manner. Conversely, PA103 (ΔPcrV) infection did not elicit early phase PMVEC barrier disruption. At 24-hours post-inoculation (late phase infection), PA103 (ΔU/ΔT) induced PMVEC damage and death that displayed an apoptotic component. Although PA103 (ΔPcrV) infection induced late phase PMVEC damage and death, it did so to an attenuated extent. The PA103 (ΔU/ΔT) and PA103 (ΔPcrV) mutants grew at similar rates and were able to adhere equally to PMVECs post-inoculation indicating that the observed differences in damage and barrier disruption are likely attributable to T3SS needle tip complex-mediated pathogenic differences post host cell attachment. Together, these infection data suggest that the T3SS needle tip complex and/or another undefined secreted effector(s) are important determinants of P. aeruginosa pneumonia-induced lung endothelial barrier disruption.
Collapse
Affiliation(s)
- Jonathon P. Audia
- Department of Microbiology and Immunology, University of South Alabama, Mobile, Alabama, United States of America
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States of America
- * E-mail: (JPA); (DFA)
| | - Ashley S. Lindsey
- Department of Pharmacology, University of South Alabama, Mobile, Alabama, United States of America
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States of America
| | - Nicole A. Housley
- Department of Microbiology and Immunology, University of South Alabama, Mobile, Alabama, United States of America
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States of America
| | - Courtney R. Ochoa
- Department of Pharmacology, University of South Alabama, Mobile, Alabama, United States of America
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States of America
| | - Chun Zhou
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States of America
| | - Michie Toba
- Department of Pharmacology, University of South Alabama, Mobile, Alabama, United States of America
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States of America
| | - Masahiko Oka
- Department of Pharmacology, University of South Alabama, Mobile, Alabama, United States of America
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States of America
| | - Naga S. Annamdevula
- Department of Chemical and Biomolecular Engineering, University of South Alabama, Mobile, Alabama, United States of America
| | - Meshann S. Fitzgerald
- Department of Internal Medicine, University of South Alabama, Mobile, Alabama, United States of America
- Department of Pharmacology, University of South Alabama, Mobile, Alabama, United States of America
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States of America
| | - Dara W. Frank
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Diego F. Alvarez
- Department of Internal Medicine, University of South Alabama, Mobile, Alabama, United States of America
- Department of Pharmacology, University of South Alabama, Mobile, Alabama, United States of America
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States of America
- * E-mail: (JPA); (DFA)
| |
Collapse
|
39
|
Cunha LD, Zamboni DS. Subversion of inflammasome activation and pyroptosis by pathogenic bacteria. Front Cell Infect Microbiol 2013; 3:76. [PMID: 24324933 PMCID: PMC3840304 DOI: 10.3389/fcimb.2013.00076] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 10/23/2013] [Indexed: 12/16/2022] Open
Abstract
Activation of the inflammasome occurs in response to a notably high number of pathogenic microbes and is a broad innate immune response that effectively contributes to restriction of pathogen replication and generation of adaptive immunity. Activation of these platforms leads to caspase-1- and/or caspase-11-dependent secretion of proteins, including cytokines, and induction of a specific form of cell death called pyroptosis, which directly or indirectly contribute for restriction of pathogen replication. Not surprisingly, bona fide intracellular pathogens developed strategies for manipulation of cell death to guarantee intracellular replication. In this sense, the remarkable advances in the knowledge of the inflammasome field have been accompanied by several reports characterizing the inhibition of this platform by several pathogenic bacteria. Herein, we review some processes used by pathogenic bacteria, including Yersinia spp., Pseudomonas aeruginosa, Vibrio parahaemolyticus, Chlamydia trachomatis, Francisella tularensis, Shigella flexneri, Legionella pneumophila, and Coxiella burnetii to evade the activation of the inflammasome and the induction of pyroptosis.
Collapse
Affiliation(s)
- Larissa D Cunha
- Department of Cell Biology, Ribeirão Preto Medical School, University of São Paulo (FMRP/USP) Ribeirão Preto, Brazil
| | | |
Collapse
|
40
|
The AlgZR two-component system recalibrates the RsmAYZ posttranscriptional regulatory system to inhibit expression of the Pseudomonas aeruginosa type III secretion system. J Bacteriol 2013; 196:357-66. [PMID: 24187093 DOI: 10.1128/jb.01199-13] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Pseudomonas aeruginosa causes chronic airway infections in cystic fibrosis (CF) patients. A classic feature of CF airway isolates is the mucoid phenotype. Mucoidy arises through mutation of the mucA anti-sigma factor and subsequent activation of the AlgU regulon. Inactivation of mucA also results in reduced expression of the Vfr transcription factor. Vfr regulates several important virulence factors, including a type III secretion system (T3SS). In the present study, we report that ExsA expression, the master regulator of T3SS gene expression, is further reduced in mucA mutants through a Vfr-independent mechanism involving the RsmAYZ regulatory system. RsmA is an RNA binding protein required for T3SS gene expression. Genetic experiments suggest that the AlgZR two-component system, part of the AlgU regulon, inhibits ExsA expression by increasing the expression of RsmY and RsmZ, two small noncoding RNAs that sequester RsmA from target mRNAs. Epistasis analyses revealed that increasing the concentration of free RsmA, through either rsmYZ deletion or increased RsmA expression, partially restored T3SS gene expression in the mucA mutant. Furthermore, increasing RsmA availability in combination with Vfr complementation fully restored T3SS expression. Recalibration of the RsmAYZ system by AlgZR, however, did not alter the expression of other selected RsmA-dependent targets. We account for this observation by showing that ExsA expression is more sensitive to changes in free RsmA than other members of the RsmA regulon. Together, these data indicate that recalibration of the RsmAYZ system partially accounts for reduced T3SS gene expression in mucA mutants.
Collapse
|
41
|
Balczon R, Prasain N, Ochoa C, Prater J, Zhu B, Alexeyev M, Sayner S, Frank DW, Stevens T. Pseudomonas aeruginosa exotoxin Y-mediated tau hyperphosphorylation impairs microtubule assembly in pulmonary microvascular endothelial cells. PLoS One 2013; 8:e74343. [PMID: 24023939 PMCID: PMC3762819 DOI: 10.1371/journal.pone.0074343] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 08/01/2013] [Indexed: 12/21/2022] Open
Abstract
Pseudomonas aeruginosa uses a type III secretion system to introduce the adenylyl and guanylyl cyclase exotoxin Y (ExoY) into the cytoplasm of endothelial cells. ExoY induces Tau hyperphosphorylation and insolubility, microtubule breakdown, barrier disruption and edema, although the mechanism(s) responsible for microtubule breakdown remain poorly understood. Here we investigated both microtubule behavior and centrosome activity to test the hypothesis that ExoY disrupts microtubule dynamics. Fluorescence microscopy determined that infected pulmonary microvascular endothelial cells contained fewer microtubules than control cells, and further studies demonstrated that the microtubule-associated protein Tau was hyperphosphorylated following infection and dissociated from microtubules. Disassembly/reassembly studies determined that microtubule assembly was disrupted in infected cells, with no detectable effects on either microtubule disassembly or microtubule nucleation by centrosomes. This effect of ExoY on microtubules was abolished when the cAMP-dependent kinase phosphorylation site (Ser-214) on Tau was mutated to a non-phosphorylatable form. These studies identify Tau in microvascular endothelial cells as the target of ExoY in control of microtubule architecture following pulmonary infection by Pseudomonas aeruginosa and demonstrate that phosphorylation of tau following infection decreases microtubule assembly.
Collapse
Affiliation(s)
- Ron Balczon
- Department of Cell Biology and Neuroscience, University of South Alabama, Mobile, Alabama, United States of America
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States of America
- * E-mail:
| | - Nutan Prasain
- Department of Pediatrics, University of Indiana School of Medicine, Indianapolis, Indiana, United States of America
| | - Cristhiaan Ochoa
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States of America
- Department of Pharmacology, University of South Alabama, Mobile, Alabama, United States of America
| | - Jason Prater
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States of America
- Department of Medicine, University of South Alabama, Mobile, Alabama, United States of America
| | - Bing Zhu
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States of America
- Department of Pharmacology, University of South Alabama, Mobile, Alabama, United States of America
| | - Mikhail Alexeyev
- Department of Cell Biology and Neuroscience, University of South Alabama, Mobile, Alabama, United States of America
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States of America
| | - Sarah Sayner
- Department of Cell Biology and Neuroscience, University of South Alabama, Mobile, Alabama, United States of America
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States of America
| | - Dara W. Frank
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Troy Stevens
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States of America
- Department of Pharmacology, University of South Alabama, Mobile, Alabama, United States of America
- Department of Medicine, University of South Alabama, Mobile, Alabama, United States of America
| |
Collapse
|
42
|
Dai J, Yoon SH, Sim HY, Yang YS, Oh TK, Kim JF, Hong JW. Charting microbial phenotypes in multiplex nanoliter batch bioreactors. Anal Chem 2013; 85:5892-9. [PMID: 23581968 DOI: 10.1021/ac400648z] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
High-throughput growth phenotyping is receiving great attention for establishing the genotype-phenotype map of sequenced organisms owing to the ready availability of complete genome sequences. To date, microbial growth phenotypes have been investigated mostly by the conventional method of batch cultivation using test tubes, Erlenmeyer flasks, or the recently available microwell plates. However, the current batch cultivation methods are time- and labor-intensive and often fail to consider sophisticated environmental changes. The implementation of batch cultures at the nanoliter scale has been difficult because of the quick evaporation of the culture medium inside the reactors. Here, we report a microfluidic system that allows independent cell cultures in evaporation-free multiplex nanoliter reactors under different culture conditions to assess the behavior of cells. The design allows three experimental replicates for each of eight culture environments in a single run. We demonstrate the versatility of the device by performing growth curve experiments with Escherichia coli and microbiological assays of antibiotics against the opportunistic pathogen Pseudomonas aeruginosa. Our study highlights that the microfluidic system can effectively replace the traditional batch culture methods with nanoliter volumes of bacterial cultivations, and it may be therefore promising for high-throughput growth phenotyping as well as for single-cell analyses.
Collapse
Affiliation(s)
- Jing Dai
- Materials Research and Education Center, Department of Mechanical Engineering, Auburn University, Auburn, Alabama 36849, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Adenoviral vectors have been used for a variety of vaccine applications including cancer and infectious diseases. Traditionally, Ad-based vaccines are designed to express antigens through transgene expression of a given antigen. For effective vaccine development it is often necessary to express or present multiple antigens to the immune system to elicit an optimal vaccine as observed preclinically with mosaic/polyvalent HIV vaccines or malaria vaccines. Due to the wide flexibility of Ad vectors they are an ideal platform for expressing large amounts of antigen and/or polyvalent mosaic antigens. Ad vectors that display antigens on their capsid surface can elicit a robust humoral immune response, the “antigen capsid-incorporation” strategy. The adenoviral hexon protein has been utilized to display peptides in the majority of vaccine strategies involving capsid incorporation. Based on our abilities to manipulate hexon HVR2 and HVR5, we sought to manipulate HVR1 in the context of HIV antigen display for the first time ever. More importantly, peptide incorporation within HVR1 was utilized in combination with other HVRs, thus creating multivalent vectors. To date this is the first report where dual antigens are displayed within one Ad hexon particle. These vectors utilize HVR1 as an incorporation site for a seven amino acid region of the HIV glycoprotein 41, in combination with six Histidine incorporation within HVR2 or HVR5. Our study illustrates that these multivalent antigen vectors are viable and can present HIV antigen as well as His6 within one Ad virion particle. Furthermore, mouse immunizations with these vectors demonstrate that these vectors can elicit a HIV and His6 epitope-specific humoral immune response.
Collapse
|
44
|
Bernhards RC, Marsden AE, Esher SK, Yahr TL, Schubot FD. Self-trimerization of ExsD limits inhibition of the Pseudomonas aeruginosa transcriptional activator ExsA in vitro. FEBS J 2013; 280:1084-94. [PMID: 23279839 PMCID: PMC3621117 DOI: 10.1111/febs.12103] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Revised: 10/29/2012] [Accepted: 12/29/2012] [Indexed: 11/29/2022]
Abstract
The opportunistic pathogen Pseudomonas aeruginosa ranks among the leading causes of nosocomial infection. The type III secretion system (T3SS) aids acute Pseudomonas aeruginosa infection by injecting potent cytotoxins into host cells to suppress the host's innate immune response. Expression of all T3SS-related genes is strictly dependent on the transcription factor ExsA. Consequently, ExsA and the biological processes that regulate ExsA function are of great biomedical interest. The present study focused on the ExsA-ExsC-ExsD-ExsE signaling cascade, which ties host cell contact to the upregulation of T3SS gene expression. Prior to T3SS induction, the antiactivator protein ExsD binds to ExsA and blocks ExsA-dependent transcription by interfering with ExsA dimerization and promoter interactions. Upon host cell contact, ExsD is sequestered by the T3SS chaperone ExsC, resulting in the release of ExsA and upregulation of the T3SS. Previous studies have shown that the ExsD-ExsA interactions are not freely reversible. Because independently folded ExsD and ExsA were not found to interact, it has been hypothesized that folding intermediates of the two proteins form the complex. Here, we demonstrate, for the first time, that ExsD alone is sufficient to inhibit ExsA-dependent transcription in vitro and that no other cellular factors are required. More significantly, we show that independently folded ExsD and ExsA are capable of interacting, but only at 37 °C and not at 30 °C. Guided by the crystal structure of ExsD, we designed a monomeric variant of the protein, and demonstrated that ExsD trimerization prevents ExsD from inhibiting ExsA-dependent transcription at 30 °C. We propose that this unique mechanism plays an important role in T3SS regulation.
Collapse
|
45
|
Galle M, Carpentier I, Beyaert R. Structure and function of the Type III secretion system of Pseudomonas aeruginosa. Curr Protein Pept Sci 2012; 13:831-42. [PMID: 23305368 PMCID: PMC3706959 DOI: 10.2174/138920312804871210] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Revised: 07/19/2012] [Accepted: 07/25/2012] [Indexed: 02/08/2023]
Abstract
Pseudomonas aeruginosa is a dangerous pathogen particularly because it harbors multiple virulence factors. It causes several types of infection, including dermatitis, endocarditis, and infections of the urinary tract, eye, ear, bone, joints and, of particular interest, the respiratory tract. Patients with cystic fibrosis, who are extremely susceptible to Pseudomonas infections, have a bad prognosis and high mortality. An important virulence factor of P. aeruginosa, shared with many other gram-negative bacteria, is the type III secretion system, a hollow molecular needle that transfers effector toxins directly from the bacterium into the host cell cytosol. This complex macromolecular machine works in a highly regulated manner and can manipulate the host cell in many different ways. Here we review the current knowledge of the structure of the P. aeruginosa T3SS, as well as its function and recognition by the immune system. Furthermore, we describe recent progress in the development and use of therapeutic agents targeting the T3SS.
Collapse
Affiliation(s)
- Marlies Galle
- Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, B-9052 Ghent, Belgium; the
- Department for Molecular Biomedical Research, Unit of Molecular Signal Transduction in Inflammation, VIB, Technologiepark 927, B-9052 Ghent, Belgium
| | - Isabelle Carpentier
- Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, B-9052 Ghent, Belgium; the
- Department for Molecular Biomedical Research, Unit of Molecular Signal Transduction in Inflammation, VIB, Technologiepark 927, B-9052 Ghent, Belgium
| | - Rudi Beyaert
- Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, B-9052 Ghent, Belgium; the
- Department for Molecular Biomedical Research, Unit of Molecular Signal Transduction in Inflammation, VIB, Technologiepark 927, B-9052 Ghent, Belgium
| |
Collapse
|
46
|
Freestone PP, Hirst RA, Sandrini SM, Sharaff F, Fry H, Hyman S, O'Callaghan C. Pseudomonas aeruginosa -Catecholamine Inotrope Interactions. Chest 2012; 142:1200-1210. [DOI: 10.1378/chest.11-2614] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
47
|
The Pseudomonas aeruginosa type III secretion system has an exotoxin S/T/Y independent pathogenic role during acute lung infection. PLoS One 2012; 7:e41547. [PMID: 22844497 PMCID: PMC3402384 DOI: 10.1371/journal.pone.0041547] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2011] [Accepted: 06/27/2012] [Indexed: 12/21/2022] Open
Abstract
The type III secretion system (T3SS) is a complex nanomachine of many pathogenic Gram-negative bacteria. It forms a proteinaceous channel that is inserted into the host eukaryotic cell membrane for injection of bacterial proteins that manipulate host cell signaling. However, few studies have focused on the effector-independent functions of the T3SS. Using a murine model of acute lung infection with Pseudomonas aeruginosa, an important human opportunistic pathogen, we compared the pathogenicity of mutant bacteria that lack all of the known effector toxins ( ΔSTY), with mutant bacteria that also lack the major translocator protein PopB (ΔSTY/ΔPopB) and so cannot form a functional T3SS channel in the host cell membrane. Mortality was higher among mice challenged with ΔSTY compared to mice challenged with ΔSTY/ΔPopB mutant bacteria. In addition, mice infected with ΔSTY showed decreased bacterial clearance from the lungs compared to those infected with ΔSTY/ΔPopB. Infection was in both cases associated with substantial killing of lung infiltrating macrophages. However, macrophages from ΔSTY-infected mice died by pro-inflammatory necrosis characterized by membrane permeabilization and caspase-1 mediated IL-1β production, whereas macrophages from ΔSTY/ΔPopB infected mice died by apoptosis, which is characterized by annexin V positive staining of the cell membrane and caspase-3 activation. This was confirmed in macrophages infected in vitro. These results demonstrate a T3SS effector toxin independent role for the T3SS, in particular the T3SS translocator protein PopB, in the pathogenicity of P. aeruginosa during acute lung infection.
Collapse
|
48
|
Topal H, Fulcher NB, Bitterman J, Salazar E, Buck J, Levin LR, Cann MJ, Wolfgang MC, Steegborn C. Crystal structure and regulation mechanisms of the CyaB adenylyl cyclase from the human pathogen Pseudomonas aeruginosa. J Mol Biol 2011; 416:271-86. [PMID: 22226839 DOI: 10.1016/j.jmb.2011.12.045] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Revised: 12/18/2011] [Accepted: 12/21/2011] [Indexed: 12/15/2022]
Abstract
Pseudomonas aeruginosa is an opportunistic bacterial pathogen and a major cause of healthcare-associated infections. While the organism's intrinsic and acquired resistance to most antibiotics hinders treatment of P. aeruginosa infections, the regulatory networks controlling its virulence provide novel targets for drug development. CyaB, a key regulator of P. aeruginosa virulence, belongs to the Class III adenylyl cyclase (AC) family of enzymes that synthesize the second messenger cyclic adenosine 3',5'-monophosphate. These enzymes consist of a conserved catalytic domain fused to one or more regulatory domains. We describe here the biochemical and structural characterization of CyaB and its inhibition by small molecules. We show that CyaB belongs to the Class IIIb subfamily, and like other subfamily members, its activity is stimulated by inorganic carbon. CyaB is also regulated by its N-terminal MASE2 (membrane-associated sensor 2) domain, which acts as a membrane anchor. Using a genetic screen, we identified activating mutations in CyaB. By solving the crystal structure of the CyaB catalytic domain, we rationalized the effects of these mutations and propose that CyaB employs regulatory mechanisms similar to other Class III ACs. The CyaB structure further indicates subtle differences compared to other Class III ACs in both the active site and the inhibitor binding pocket. Consistent with these differences, we observed a unique inhibition profile, including identification of a CyaB selective compound. Overall, our results reveal mechanistic details of the physiological and pharmacological regulation of CyaB and provide the basis for its exploitation as a therapeutic drug target.
Collapse
Affiliation(s)
- Hüsnü Topal
- Department of Physiological Chemistry, Ruhr-University Bochum, 44801 Bochum, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Gustke H, Kleene R, Loers G, Nehmann N, Jaehne M, Bartels KM, Jaeger KE, Schachner M, Schumacher U. Inhibition of the bacterial lectins of Pseudomonas aeruginosa with monosaccharides and peptides. Eur J Clin Microbiol Infect Dis 2011; 31:207-15. [PMID: 21604096 DOI: 10.1007/s10096-011-1295-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Accepted: 05/06/2011] [Indexed: 12/19/2022]
Abstract
Pseudomonas aeruginosa (PA) can cause infections in compromised hosts by interacting with the glycocalyx of host epithelial cells. It binds to glycostructures on mucosal surfaces via two lectins, which are carbohydrate-binding proteins, named PA-IL and PA-IIL, and blocking this interaction is, thus, an attractive anti-adhesive strategy. The aim of this study was to determine by ciliary beat frequency (CBF) analysis whether monosaccharides or peptides mimicking glycostructures represent blockers of PA lectin binding to human airway cilia. The treatment with monosaccharides and peptides alone did not change the CBF compared to controls and the tested compounds did not influence the cell morphology or survival, with the exception of peptide pOM3. PA-IL caused a decrease of the CBF within 24 h. D-galactose as well as the peptides mimicking HNK-1, polysialic acid and fucose compensated the CBF-modulating effect of PA-IL with different affinities. PA-IIL also bound to the human airway cilia in cell culture and resulted in a decrease of the CBF within 24 h. L(-)-fucose and pHNK-1 blocked the CBF-decreasing effect of PA-IIL. The HNK-1-specific glycomimetic peptide had a high affinity for binding to both PA-IL and PA-IIL, and inhibited the ciliotoxic effect of both lectins, thus, making it a strong candidate for a therapeutic anti-adhesive drug.
Collapse
Affiliation(s)
- H Gustke
- Department of Anatomy II: Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Risk factors and severity scores in hospitalized patients with community-acquired pneumonia: prediction of severity and mortality. Eur J Clin Microbiol Infect Dis 2011; 31:33-47. [PMID: 21533875 DOI: 10.1007/s10096-011-1272-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Accepted: 04/12/2011] [Indexed: 12/27/2022]
Abstract
Morbidity and mortality in patients with moderate to severe community-acquired pneumonia (CAP) is a global problem, and CAP is a leading cause of death due to infectious diseases. Prompt initiation of expanded-spectrum antimicrobials is essential for the prevention of unnecessary mortality and complications in patients, particularly in the elderly and other at-risk populations, and the treatment decisions made by practitioners have important implications for healthcare systems when hospitalization is required. Empirical antimicrobial treatment and the appropriate management of CAP patients will initially require the proper assessment of severity and patient risk for increased mortality, as well as risk factors for difficult-to-treat bacteria. This review will examine risk factors and scoring systems that may be predictive of moderate to severe CAP, which is often linked to increased risk of mortality. Understanding and recognizing potential risk factors will allow practitioners to proactively identify patients at the highest risk for severe illness or complications, thereby, guiding site-of-care decisions, as well as the choices for empiric antibiotic regimens. The decision to hospitalize a patient with CAP should include not only a clinical perspective and laboratory and radiographic findings, but also at least one objective tool of risk assessment, all in combination with sound clinical judgment.
Collapse
|