1
|
Junya O, Jumpei F, Kinoshita M, Sudo K, Kawaguchi K, Inoue K, Naito Y, Moriyama K, Nakamura T, Iwano H, Sawa T. Effects of the combination of anti-PcrV antibody and bacteriophage therapy in a mouse model of Pseudomonas aeruginosa pneumonia. Microbiol Spectr 2024:e0178124. [PMID: 39440986 DOI: 10.1128/spectrum.01781-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 09/02/2024] [Indexed: 10/25/2024] Open
Abstract
Acute lung injury caused by Pseudomonas aeruginosa is attributed to the translocation of cytotoxin into pulmonary epithelial cells via the P. aeruginosa type III secretion system. This virulence can be blocked with a specific antibody against PcrV in this secretion system. However, because anti-PcrV antibodies do not have bactericidal activity, the treatment of bacteria depends on the phagocytic system of the host. In this study, we investigated the therapeutic effect of combination therapy with an anti-PcrV antibody and bactericidal bacteriophages on acute lung injury and subsequent death in mice compared with a single treatment. After the mice intratracheally received a lethal dose of the cytotoxic P. aeruginosa strain, a second instillation was performed with saline, anti-PcrV IgG, bacteriophages, or a mixture of anti-PcrV and bacteriophages. The survival rates 24 h after infection were as follows: 7.1% in the saline group, 26.7% in the anti-PcrV group, 41.2% in the phage group, and 66.7% in the anti-PcrV + phage group (P < 0.001 vs saline-treated group). The activity of surviving mice in the anti-PcrV + phage group was significantly greater than that in the saline group. The lung weight in the anti-PcrV + phage group was significantly lower than that in the anti-PcrV group. In conclusion, combination therapy with an anti-PcrV antibody and a bacteriophage reduces acute lung injury and suggests improved survival compared with each treatment alone. This combination therapy, which does not rely on conventional antibiotics, could constitute a new strategy for treating multidrug-resistant P. aeruginosa infections.IMPORTANCECombination therapy with either bacteriophages alone or in combination with anti-PcrV antibodies in a mouse model of Pseudomonas aeruginosa pneumonia may reduce the acute lung injury and improve survival. This combination therapy, which does not rely on conventional antibiotics, may be a new strategy to treat multidrug-resistant Pseudomonas aeruginosa infections.
Collapse
Affiliation(s)
- Ohara Junya
- Department of Anesthesiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Fujiki Jumpei
- Laboratory of Veterinary Biochemistry, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido, Japan
| | - Mao Kinoshita
- Department of Anesthesiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazuki Sudo
- Department of Anesthesiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Ken Kawaguchi
- Department of Anesthesiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Keita Inoue
- Division of Critical Care Medicine, University Hospital, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoshifumi Naito
- Department of Anesthesiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kiyoshi Moriyama
- Department of Anesthesiology, School of Medicine, Kyorin University, Mitaka, Japan
| | - Tomohiro Nakamura
- Laboratory of Veterinary Biochemistry, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido, Japan
| | - Hidetomo Iwano
- Laboratory of Veterinary Biochemistry, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido, Japan
| | - Teiji Sawa
- Department of Anesthesiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
2
|
Numata S, Hara T, Izawa M, Okuno Y, Sato Y, Yamane S, Maki H, Sato T, Yamano Y. Novel humanized anti-PcrV monoclonal antibody COT-143 protects mice from lethal Pseudomonas aeruginosa infection via inhibition of toxin translocation by the type III secretion system. Antimicrob Agents Chemother 2024; 68:e0069424. [PMID: 39269189 PMCID: PMC11459929 DOI: 10.1128/aac.00694-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 08/08/2024] [Indexed: 09/15/2024] Open
Abstract
Treatment of Pseudomonas aeruginosa infection is challenging due to its intrinsic and acquired antibiotic resistance. As the number of current therapeutic options for P. aeruginosa infections is limited, developing novel treatments against the pathogen is an urgent clinical priority. The suppression of virulence of P. aeruginosa could be a new therapeutic option, and the type III secretion system (T3SS), which enables the bacteria to translocate various kinds of toxins into host cells and inhibits cellular functions, is considered as one possible target. In this report, we examined T3SS inhibition by COT-143/INFEX702, a humanized monoclonal antibody against PcrV, T3SS component, and present the crystal structure of the antibody-PcrV complex. COT-143 inhibited T3SS-dependent cytotoxicity and protected mice from the mortality caused by P. aeruginosa infection. The inhibition of cytotoxicity coincided with inhibition of translocon formation in a host cell membrane, which is necessary for T3SS intoxication. COT-143 protected murine neutrophils and facilitated phagocytosis of P. aeruginosa. These results suggest that COT-143 facilitates P. aeruginosa clearance by protecting neutrophil via inhibition of T3SS-dependent toxin translocation. This is the first report to show that an anti-PcrV antibody directly interferes with translocon formation to inhibit intoxication of host cells.
Collapse
Affiliation(s)
- Shunsuke Numata
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd., Toyonaka, Japan
| | - Takafumi Hara
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd., Toyonaka, Japan
| | - Masaaki Izawa
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd., Toyonaka, Japan
| | - Yosuke Okuno
- Shionogi TechnoAdvance Research & Co., Ltd., Toyonaka, Japan
| | - Yasuhiko Sato
- Business Development Department, Shionogi & Co., Ltd., Toyonaka, Japan
| | - Shoji Yamane
- Shionogi TechnoAdvance Research & Co., Ltd., Toyonaka, Japan
| | - Hideki Maki
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd., Toyonaka, Japan
| | - Takafumi Sato
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd., Toyonaka, Japan
| | - Yoshinori Yamano
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd., Toyonaka, Japan
| |
Collapse
|
3
|
Murugan N, Krishnamoorthy R, Khan JM, Gatasheh MK, Malathi J, Madhavan HNR, Ramalingam G, Jayaramana S. Unveiling the ocular battlefield: Insights into Pseudomonas aeruginosa virulence factors and their implications for multidrug resistance. Int J Biol Macromol 2024; 267:131677. [PMID: 38641280 DOI: 10.1016/j.ijbiomac.2024.131677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/13/2024] [Accepted: 04/16/2024] [Indexed: 04/21/2024]
Abstract
The research investigates the virulence factors of Pseudomonas aeruginosa (P. aeruginosa), a pathogen known for its ability to cause human infections by releasing various exoenzymes and virulence factors. Particularly relevant in ocular infections, where tissue degeneration can occur, even after bacterial growth has ceased due to the potential role of secreted proteins/enzymes. Clinical isolates of P. aeruginosa, both ocular (146) and non-ocular (54), were examined to determine the frequency and mechanism of virulence factors. Phenotypic characterization revealed the production of alginate, biofilm, phospholipase C, and alkaline protease, while genotypic testing using internal uniplex PCR identified the presence of Exo U, S, T, Y, and LasB genes. Results showed a significant prevalence of Exo U and Y genes in ocular isolates, a finding unique to Indian studies. Additionally, the study noted that ocular isolates often contained all four secretomes, suggesting a potential link between these factors and ocular infections. These findings contribute to understanding the pathogenesis of P. aeruginosa infections, particularly in ocular contexts, and highlights the importance of comprehensive virulence factor analysis in clinical settings.
Collapse
Affiliation(s)
- Nandagopal Murugan
- Department of Microbiology, L & T Microbiology Research Centre, Vision Research Foundation, Sankara Nethralaya, Chennai-6000 06, India; Valluvar Rosalind Diagnostic & Research Lab, Tiruvotriyur, Chennai-600019, India
| | - Rajapandiyan Krishnamoorthy
- Department of Food Science and Nutrition, College of Food and Agriculture Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| | - Javed Masood Khan
- Department of Food Science and Nutrition, College of Food and Agriculture Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mansour K Gatasheh
- Department of Biochemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Jambulingam Malathi
- Department of Microbiology, L & T Microbiology Research Centre, Vision Research Foundation, Sankara Nethralaya, Chennai-6000 06, India; Valluvar Rosalind Diagnostic & Research Lab, Tiruvotriyur, Chennai-600019, India
| | - Hajib Narahari Rao Madhavan
- Department of Microbiology, L & T Microbiology Research Centre, Vision Research Foundation, Sankara Nethralaya, Chennai-6000 06, India; Valluvar Rosalind Diagnostic & Research Lab, Tiruvotriyur, Chennai-600019, India
| | - Gopinath Ramalingam
- Department of Microbiology, Government Theni Medical College and Hospital, Theni, Tamil Nadu-625512, India
| | - Selvaraj Jayaramana
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai-600077, India.
| |
Collapse
|
4
|
Dirr L, Cleeves S, Ramón Roth I, Li L, Fiebig T, Ve T, Häussler S, Braun A, von Itzstein M, Führing JI. Tetramerization is essential for the enzymatic function of the Pseudomonas aeruginosa virulence factor UDP-glucose pyrophosphorylase. mBio 2024; 15:e0211423. [PMID: 38470050 PMCID: PMC11005391 DOI: 10.1128/mbio.02114-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 02/20/2024] [Indexed: 03/13/2024] Open
Abstract
Multidrug-resistant bacteria such as the opportunistic pathogen Pseudomonas aeruginosa, which causes life-threatening infections especially in immunocompromised individuals and cystic fibrosis patients, pose an increasing threat to public health. In the search for new treatment options, P. aeruginosa uridine diphosphate-glucose pyrophosphorylase (PaUGP) has been proposed as a novel drug target because it is required for the biosynthesis of important virulence factors and linked to pathogenicity in animal models. Here, we show that UGP-deficient P. aeruginosa exhibits severely reduced virulence against human lung tissue and cells, emphasizing the enzyme's suitability as a drug target. To establish a basis for the development of selective PaUGP inhibitors, we solved the product-bound crystal structure of tetrameric PaUGP and conducted a comprehensive structure-function analysis, identifying key residues at two different molecular interfaces that are essential for tetramer integrity and catalytic activity and demonstrating that tetramerization is pivotal for PaUGP function. Importantly, we show that part of the PaUGP oligomerization interface is uniquely conserved across bacterial UGPs but does not exist in the human enzyme, therefore representing an allosteric site that may be targeted to selectively inhibit bacterial UGPs.IMPORTANCEInfections with the opportunistic bacterial pathogen Pseudomonas aeruginosa are becoming increasingly difficult to treat due to multidrug resistance. Here, we show that the enzyme uridine diphosphate-glucose pyrophosphorylase (UGP) is involved in P. aeruginosa virulence toward human lung tissue and cells, making it a potential target for the development of new antibacterial drugs. Our exploration of P. aeruginosa (Pa)UGP structure-function relationships reveals that the activity of PaUGP depends on the formation of a tetrameric enzyme complex. We found that a molecular interface involved in tetramer formation is conserved in all bacterial UGPs but not in the human enzyme, and therefore hypothesize that it provides an ideal point of attack to selectively inhibit bacterial UGPs and exploit them as drug targets.
Collapse
Affiliation(s)
- Larissa Dirr
- Institute for Glycomics, Gold Coast Campus, Griffith University, Gold Coast, Queensland, Australia
| | - Sven Cleeves
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Member of Fraunhofer International Consortium for Anti-Infective Research (iCAIR), Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), German Center for Lung Research (DZL), Hannover, Germany
| | - Isabel Ramón Roth
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Linghui Li
- Institute for Glycomics, Gold Coast Campus, Griffith University, Gold Coast, Queensland, Australia
| | - Timm Fiebig
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Thomas Ve
- Institute for Glycomics, Gold Coast Campus, Griffith University, Gold Coast, Queensland, Australia
| | - Susanne Häussler
- Department of Molecular Bacteriology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Institute for Molecular Bacteriology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany
- Department of Clinical Microbiology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Armin Braun
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Member of Fraunhofer International Consortium for Anti-Infective Research (iCAIR), Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), German Center for Lung Research (DZL), Hannover, Germany
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Mark von Itzstein
- Institute for Glycomics, Gold Coast Campus, Griffith University, Gold Coast, Queensland, Australia
| | - Jana I. Führing
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| |
Collapse
|
5
|
Gies SL, Tessmer MH, Frank DW, Feix JB. Site-directed spin label EPR studies of the structure and membrane interactions of the bacterial phospholipase ExoU. APPLIED MAGNETIC RESONANCE 2024; 55:279-295. [PMID: 39175603 PMCID: PMC11340903 DOI: 10.1007/s00723-023-01620-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 08/24/2024]
Abstract
Site-directed spin labeling (SDSL) has been invaluable in the analysis of protein structure and dynamics, and has been particularly useful in the study of membrane proteins. ExoU, an important virulence factor in Pseudomonas aeruginosa infections, is a bacterial phospholipase A2 that functions at the membrane - aqueous interface. Using SDSL methodology developed in the Hubbell lab, we find that the region surrounding the catalytic site of ExoU is buried within the tertiary structure of the protein in the soluble, apoenzyme state, but shows a significant increase in dynamics upon membrane binding and activation by ubiquitin. Continuous wave (CW) power saturation EPR studies show that the conserved serine hydrolase motif of ExoU localizes to the membrane surface in the active, holoenzyme state. SDSL studies on the C-terminal four-helix bundle (4HB) domain of ExoU similarly show a co-operative effect of ubiquitin binding and membrane association. CW power saturation studies of the 4HB domain indicate that two interhelical loops intercalate into the lipid bilayer upon formation of the holoenzyme state, anchoring ExoU at the membrane surface. Together these studies establish the orientation and localization of ExoU and the membrane surface, and illustrate the power of SDSL as applied to peripheral membrane proteins.
Collapse
Affiliation(s)
- Samantha L. Gies
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
- Current address: Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, 63104, USA
| | - Maxx H. Tessmer
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
- Current address: Department of Chemistry, University of Washington, Seattle, WA, 98195, USA
| | - Dara W. Frank
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Jimmy B. Feix
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| |
Collapse
|
6
|
Guo H, Geddes EJ, Opperman TJ, Heuck AP. Cell-Based Assay to Determine Type 3 Secretion System Translocon Assembly in Pseudomonas aeruginosa Using Split Luciferase. ACS Infect Dis 2023; 9:2652-2664. [PMID: 37978950 DOI: 10.1021/acsinfecdis.3c00482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
Multi-drug-resistant Pseudomonas aeruginosa poses a serious threat to hospitalized patients. This organism expresses an arsenal of virulence factors that enables it to readily establish infections and disseminate in the host. The Type 3 secretion system (T3SS) and its associated effectors play a crucial role in the pathogenesis of P. aeruginosa, making them attractive targets for the development of novel therapeutic agents. The T3SS translocon, composed of PopD and PopB, is an essential component of the T3SS secretion apparatus. In the properly assembled translocon, the N-terminus of PopD protrudes into the cytoplasm of the target mammalian cell, which can be exploited as a molecular indicator of functional translocon assembly. In this article, we describe a novel whole-cell-based assay that employs the split NanoLuc luciferase detection system to provide a readout for translocon assembly. The assay demonstrates a favorable signal/noise ratio (13.6) and robustness (Z' = 0.67), making it highly suitable for high-throughput screening of small-molecule inhibitors targeting T3SS translocon assembly.
Collapse
Affiliation(s)
- Hanling Guo
- Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Emily J Geddes
- Microbiotix, Inc., Worcester, Massachusetts 01605, United States
| | | | - Alejandro P Heuck
- Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, Massachusetts 01003, United States
| |
Collapse
|
7
|
Di Pilato V, Willison E, Marchese A. The microbiology and pathogenesis of nonfermenting Gram-negative infections. Curr Opin Infect Dis 2023; 36:537-544. [PMID: 37732777 PMCID: PMC10624403 DOI: 10.1097/qco.0000000000000969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
PURPOSE OF REVIEW This review provides an overview of most recent evidence about pathogenesis traits and virulence factors contributing to successful colonization or infection by P. aeruginosa , A. baumannii , S. maltophilia and B. cepacia complex, among the most clinically relevant nonfermenting Gram-negative bacteria (NFGNB). RECENT FINDINGS The growing clinical importance of NFGNB as important opportunistic pathogens causing difficult-to-treat infections in a fragile patients' population in stressed by numerous studies. Identification of novel virulence factors and deciphering of their mechanisms of action have greatly furthered our understanding of NFGNB pathogenesis, revealing that each pathogen-specific armamentarium of virulence factors (adhesins, motility, capsule, biofilm, lipopolysaccharide, exotoxins, exoenzymes, secretion systems, siderophores) can be likely responsible for the difference in the pathophysiology even in the context of a similar infection site. Emerging evidence of the immunomodulatory effect of some virulence factors is also acknowledged. SUMMARY NFGNB continue to be a serious global problem as cause of life-threatening opportunistic infections, owing to a highly heterogeneous content of virulence factors and their extensive number of intrinsic resistance mechanisms. Further efforts in development of novel effective antimicrobials and of alternative strategies targeting key virulence factors are warranted.
Collapse
Affiliation(s)
- Vincenzo Di Pilato
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa
| | - Edward Willison
- Microbiology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Anna Marchese
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa
- Microbiology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
8
|
Olaniran OB, Donia A, Adeleke OE, Bokhari H. Prevalence of Type III Secretion System (T3SS) and Biofilm Development in Genetically Heterogeneous Clinical Isolates of Pseudomonas aeruginosa from Nigeria. Curr Microbiol 2023; 80:349. [PMID: 37733140 DOI: 10.1007/s00284-023-03467-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 09/01/2023] [Indexed: 09/22/2023]
Abstract
Pseudomonas aeruginosa infection in seriously ill patients is a major concern due to its ability to form biofilm and secrete effector toxins. There is little information on the prevalence of T3SS effector toxins and biofilm production in clinical isolates of P. aeruginosa from Nigeria. The goal of this study is to evaluate the prevalence of T3SS toxins and biofilm production among isolates from selected tertiary hospitals in Nigeria. This study examined 430 clinical isolates from our previous work, comprising 181 MDR (multidrug-resistant) and 249 non-MDR isolates. Biofilm production and type III secretion toxins were determined using colorimetric microtiter plate assay and polymerase chain reaction, respectively. Carbapenem-resistant isolates were typed using REP-PCR and BOX-PCR. Biofilm production was detected in 386/430 (89.8%) of the isolates. Out of 386 biofilm producers, 167 (43.3%) were multidrug-resistant isolates. PCR identified four T3SS virulence types among 430 isolates, including 78 (18.1%) exoU+/exoS- isolates, 343 (79.8%) exoU-/exoS + isolates, 5 (1.2%) exoU+/exoS + isolates, and 4 (0.9%) exoU-/exoS- isolates. Both REP- and BOX-PCR consist of eight clusters. On the REP-PCR dendrogram, ExoU+/ExoS- isolates majorly occupied cluster IV. Clusters IV, VII, and VIII consist of isolates from wounds on BOX-PCR dendrogram. There was a positive association between strong biofilm production and multidrug resistance in our P. aeruginosa isolates. This study identified multidrug-resistant, biofilm-producing P. aeruginosa strains that secrete cytotoxic effectors which are significant virulence factors in P. aeruginosa. This poses a severe risk to our healthcare system and highlights the importance of continuous surveillance to prevent infectious disease outbreaks.
Collapse
Affiliation(s)
- Oluwatoyin B Olaniran
- Department of Pharmaceutical Microbiology, Olabisi Onabanjo University, Ago-Iwoye, Nigeria
- Department of Biosciences, Faculty of Science, COMSATS University Islamabad, Islamabad, Pakistan
| | - Ahmed Donia
- Institute for Water and Wastewater Technology, Durban University of Technology, Durban, South Africa
- Department of Biosciences, Faculty of Science, COMSATS University Islamabad, Islamabad, Pakistan
| | - Olufemi E Adeleke
- Department of Pharmaceutical Microbiology, University of Ibadan, Ibadan, Nigeria
| | - Habib Bokhari
- Department of Biosciences, Faculty of Science, COMSATS University Islamabad, Islamabad, Pakistan.
- Kohsar University Murree, Murree, Pakistan.
| |
Collapse
|
9
|
Sánchez-Jiménez A, Llamas MA, Marcos-Torres FJ. Transcriptional Regulators Controlling Virulence in Pseudomonas aeruginosa. Int J Mol Sci 2023; 24:11895. [PMID: 37569271 PMCID: PMC10418997 DOI: 10.3390/ijms241511895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 07/21/2023] [Accepted: 07/22/2023] [Indexed: 08/13/2023] Open
Abstract
Pseudomonas aeruginosa is a pathogen capable of colonizing virtually every human tissue. The host colonization competence and versatility of this pathogen are powered by a wide array of virulence factors necessary in different steps of the infection process. This includes factors involved in bacterial motility and attachment, biofilm formation, the production and secretion of extracellular invasive enzymes and exotoxins, the production of toxic secondary metabolites, and the acquisition of iron. Expression of these virulence factors during infection is tightly regulated, which allows their production only when they are needed. This process optimizes host colonization and virulence. In this work, we review the intricate network of transcriptional regulators that control the expression of virulence factors in P. aeruginosa, including one- and two-component systems and σ factors. Because inhibition of virulence holds promise as a target for new antimicrobials, blocking the regulators that trigger the production of virulence determinants in P. aeruginosa is a promising strategy to fight this clinically relevant pathogen.
Collapse
Affiliation(s)
| | - María A. Llamas
- Department of Biotechnology and Environmental Protection, Estación Experimental del Zaidín-Consejo Superior de Investigaciones Científicas, 18008 Granada, Spain;
| | - Francisco Javier Marcos-Torres
- Department of Biotechnology and Environmental Protection, Estación Experimental del Zaidín-Consejo Superior de Investigaciones Científicas, 18008 Granada, Spain;
| |
Collapse
|
10
|
Impact of fluoroquinolones and aminoglycosides on P. aeruginosa virulence factor production and cytotoxicity. Biochem J 2022; 479:2511-2527. [PMID: 36504127 DOI: 10.1042/bcj20220527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/06/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022]
Abstract
The opportunistic pathogen Pseudomonas aeruginosa is one of leading causes of disability and mortality worldwide and the world health organisation has listed it with the highest priority for the need of new antimicrobial therapies. P. aeruginosa strains responsible for the poorest clinical outcomes express either ExoS or ExoU, which are injected into target host cells via the type III secretion system (T3SS). ExoS is a bifunctional cytotoxin that promotes intracellular survival of invasive P. aeruginosa by preventing targeting of the bacteria to acidified intracellular compartments. ExoU is a phospholipase which causes destruction of host cell plasma membranes, leading to acute tissue damage and bacterial dissemination. Fluoroquinolones are usually employed as a first line of therapy as they have been shown to be more active against P. aeruginosa in vitrothan other antimicrobial classes. Their overuse over the past decade, however, has resulted in the emergence of antibiotic resistance. In certain clinical situations, aminoglycosides have been shown to be more effective then fluoroquinolones, despite their reduced potency towards P. aeruginosa in vitro. In this study, we evaluated the effects of fluoroquinolones (moxifloxacin and ciprofloxacin) and aminoglycosides (tobramycin and gentamycin) on T3SS expression and toxicity, in corneal epithelial cell infection models. We discovered that tobramycin disrupted T3SS expression and reduced both ExoS and ExoU mediated cytotoxicity, protecting infected HCE-t cells at concentrations below the minimal inhibitory concentration (MIC). The fluoroquinolones moxifloxacin and ciprofloxacin, however, up-regulated the T3SS and did not inhibit and may have increased the cytotoxic effects of ExoS and ExoU.
Collapse
|
11
|
Lear L, Hesse E, Buckling A, Vos M. Copper selects for siderophore-mediated virulence in Pseudomonas aeruginosa. BMC Microbiol 2022; 22:303. [PMID: 36510131 PMCID: PMC9745993 DOI: 10.1186/s12866-022-02720-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 09/30/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Iron is essential for almost all bacterial pathogens and consequently it is actively withheld by their hosts. However, the production of extracellular siderophores enables iron sequestration by pathogens, increasing their virulence. Another function of siderophores is extracellular detoxification of non-ferrous metals. Here, we experimentally link the detoxification and virulence roles of siderophores by testing whether the opportunistic pathogen Pseudomonas aeruginosa displays greater virulence after exposure to copper. To do this, we incubated P. aeruginosa under different environmentally relevant copper regimes for either two or twelve days. Subsequent growth in a copper-free environment removed phenotypic effects, before we quantified pyoverdine production (the primary siderophore produced by P. aeruginosa), and virulence using the Galleria mellonella infection model. RESULTS Copper selected for increased pyoverdine production, which was positively correlated with virulence. This effect increased with time, such that populations incubated with high copper for twelve days were the most virulent. Replication of the experiment with a non-pyoverdine producing strain of P. aeruginosa demonstrated that pyoverdine production was largely responsible for the change in virulence. CONCLUSIONS We here show a direct link between metal stress and bacterial virulence, highlighting another dimension of the detrimental effects of metal pollution on human health.
Collapse
Affiliation(s)
- Luke Lear
- grid.8391.30000 0004 1936 8024European Centre for Environment and Human Health, University of Exeter Medical School, Penryn, Cornwall, TR10 9FE UK
| | - Elze Hesse
- grid.8391.30000 0004 1936 8024College of Life and Environmental Science, University of Exeter, Penryn, Cornwall, TR10 9FE UK
| | - Angus Buckling
- grid.8391.30000 0004 1936 8024College of Life and Environmental Science, University of Exeter, Penryn, Cornwall, TR10 9FE UK
| | - Michiel Vos
- grid.8391.30000 0004 1936 8024European Centre for Environment and Human Health, University of Exeter Medical School, Penryn, Cornwall, TR10 9FE UK
| |
Collapse
|
12
|
Elnagar RM, Elshaer M, Osama Shouman O, Sabry El-Kazzaz S. Type III Secretion System (Exoenzymes) as a Virulence Determinant in Pseudomonas aeruginosa Isolated from Burn Patients in Mansoura University Hospitals, Egypt. IRANIAN JOURNAL OF MEDICAL MICROBIOLOGY 2022. [DOI: 10.30699/ijmm.16.6.520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
13
|
Zhang Y, Wang L, Chen L, Zhu P, Huang N, Chen T, Chen L, Wang Z, Liao W, Cao J, Zhou T. Novel Insight of Transcription Factor PtrA on Pathogenicity and Carbapenems Resistance in Pseudomonas aeruginosa. Infect Drug Resist 2022; 15:4213-4227. [PMID: 35959145 PMCID: PMC9359796 DOI: 10.2147/idr.s371597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 07/23/2022] [Indexed: 11/23/2022] Open
Affiliation(s)
- Ying Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, People’s Republic of China
| | - Lingbo Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, People’s Republic of China
| | - Liqiong Chen
- Department of Medical Laboratory Science, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Peiwu Zhu
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, People’s Republic of China
| | - Na Huang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, People’s Republic of China
| | - Tao Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, People’s Republic of China
| | - Lijiang Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, People’s Republic of China
| | - Zhongyong Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, People’s Republic of China
| | - Wenli Liao
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, People’s Republic of China
| | - Jianming Cao
- Department of Medical Laboratory Science, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, People’s Republic of China
- Jianming Cao, Department of Medical Laboratory Science, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, 325035, People’s Republic of China, Tel +86-577-88069595, Email
| | - Tieli Zhou
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, People’s Republic of China
- Correspondence: Tieli Zhou, Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, 325035, People’s Republic of China, Tel +86-577-8668-9885, Email
| |
Collapse
|
14
|
Liao C, Huang X, Wang Q, Yao D, Lu W. Virulence Factors of Pseudomonas Aeruginosa and Antivirulence Strategies to Combat Its Drug Resistance. Front Cell Infect Microbiol 2022; 12:926758. [PMID: 35873152 PMCID: PMC9299443 DOI: 10.3389/fcimb.2022.926758] [Citation(s) in RCA: 93] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 06/09/2022] [Indexed: 11/24/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen causing nosocomial infections in severely ill and immunocompromised patients. Ubiquitously disseminated in the environment, especially in hospitals, it has become a major threat to human health due to the constant emergence of drug-resistant strains. Multiple resistance mechanisms are exploited by P. aeruginosa, which usually result in chronic infections difficult to eradicate. Diverse virulence factors responsible for bacterial adhesion and colonization, host immune suppression, and immune escape, play important roles in the pathogenic process of P. aeruginosa. As such, antivirulence treatment that aims at reducing virulence while sparing the bacterium for its eventual elimination by the immune system, or combination therapies, has significant advantages over traditional antibiotic therapy, as the former imposes minimal selective pressure on P. aeruginosa, thus less likely to induce drug resistance. In this review, we will discuss the virulence factors of P. aeruginosa, their pathogenic roles, and recent advances in antivirulence drug discovery for the treatment of P. aeruginosa infections.
Collapse
Affiliation(s)
- Chongbing Liao
- Key Laboratory of Medical Molecular Virology (Ministry of Education (MOE)/National Health Commission (NHC)/Chinese Academy of Medical Sciences (CAMS)), School of Basic Medical Science, Fudan University, Shanghai, China
| | - Xin Huang
- Key Laboratory of Medical Molecular Virology (Ministry of Education (MOE)/National Health Commission (NHC)/Chinese Academy of Medical Sciences (CAMS)), School of Basic Medical Science, Fudan University, Shanghai, China
| | - Qingxia Wang
- Key Laboratory of Medical Molecular Virology (Ministry of Education (MOE)/National Health Commission (NHC)/Chinese Academy of Medical Sciences (CAMS)), School of Basic Medical Science, Fudan University, Shanghai, China
| | - Dan Yao
- Key Laboratory of Medical Molecular Virology (Ministry of Education (MOE)/National Health Commission (NHC)/Chinese Academy of Medical Sciences (CAMS)), School of Basic Medical Science, Fudan University, Shanghai, China
| | - Wuyuan Lu
- Key Laboratory of Medical Molecular Virology (Ministry of Education (MOE)/National Health Commission (NHC)/Chinese Academy of Medical Sciences (CAMS)), School of Basic Medical Science, Fudan University, Shanghai, China
- Shanghai Institute of Infectious Disease and Biosecurity, School of Public Health, Fudan University, Shanghai, China
| |
Collapse
|
15
|
Constantino-Teles P, Jouault A, Touqui L, Saliba AM. Role of Host and Bacterial Lipids in Pseudomonas aeruginosa Respiratory Infections. Front Immunol 2022; 13:931027. [PMID: 35860265 PMCID: PMC9289105 DOI: 10.3389/fimmu.2022.931027] [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: 04/29/2022] [Accepted: 06/07/2022] [Indexed: 11/13/2022] Open
Abstract
The opportunistic pathogen Pseudomonas aeruginosa is one of the most common agents of respiratory infections and has been associated with high morbidity and mortality rates. The ability of P. aeruginosa to cause severe respiratory infections results from the coordinated action of a variety of virulence factors that promote bacterial persistence in the lungs. Several of these P. aeruginosa virulence mechanisms are mediated by bacterial lipids, mainly lipopolysaccharide, rhamnolipid, and outer membrane vesicles. Other mechanisms arise from the activity of P. aeruginosa enzymes, particularly ExoU, phospholipase C, and lipoxygenase A, which modulate host lipid signaling pathways. Moreover, host phospholipases, such as cPLA2α and sPLA2, are also activated during the infectious process and play important roles in P. aeruginosa pathogenesis. These mechanisms affect key points of the P. aeruginosa-host interaction, such as: i) biofilm formation that contributes to bacterial colonization and survival, ii) invasion of tissue barriers that allows bacterial dissemination, iii) modulation of inflammatory responses, and iv) escape from host defenses. In this mini-review, we present the lipid-based mechanism that interferes with the establishment of P. aeruginosa in the lungs and discuss how bacterial and host lipids can impact the outcome of P. aeruginosa respiratory infections.
Collapse
Affiliation(s)
- Pamella Constantino-Teles
- Department of Microbiology, Immunology and Parasitology, Faculty of Medical Sciences, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Albane Jouault
- Sorbonne Université, Centre de Recherche Saint-Antoine, Inserm, Institut Pasteur, Mucoviscidose et Bronchopathies Chroniques, Département Santé Globale, Paris, France
| | - Lhousseine Touqui
- Sorbonne Université, Centre de Recherche Saint-Antoine, Inserm, Institut Pasteur, Mucoviscidose et Bronchopathies Chroniques, Département Santé Globale, Paris, France
| | - Alessandra Mattos Saliba
- Department of Microbiology, Immunology and Parasitology, Faculty of Medical Sciences, Rio de Janeiro State University, Rio de Janeiro, Brazil
- *Correspondence: Alessandra Mattos Saliba,
| |
Collapse
|
16
|
Oberbach A, Schlichting N, Hagl C, Lehmann S, Kullnick Y, Friedrich M, Köhl U, Horn F, Kumbhari V, Löffler B, Schmidt F, Joskowiak D, Born F, Saha S, Bagaev E. Four decades of experience of prosthetic valve endocarditis reflect a high variety of diverse pathogens. Cardiovasc Res 2022; 119:410-428. [PMID: 35420122 DOI: 10.1093/cvr/cvac055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/04/2022] [Accepted: 03/23/2022] [Indexed: 01/18/2023] Open
Abstract
Prosthetic valve endocarditis (PVE) remains a serious condition with a high mortality rate. Precise identification of the PVE-associated pathogen/s and their virulence is essential for successful therapy, and patient survival. The commonly described PVE-associated pathogens are staphylococci, streptococci and enterococci, with Staphylococcus aureus being the most frequently diagnosed species. Furthermore, multi-drug resistance pathogens are increasing in prevalence, and continue to pose new challenges mandating a personalized approach. Blood cultures in combination with echocardiography are the most common methods to diagnose PVE, often being the only indication, it exists. In many cases, the diagnostic strategy recommended in the clinical guidelines does not identify the precise microbial agent and to frequently, false negative blood cultures are reported. Despite the fact that blood culture findings are not always a good indicator of the actual PVE agent in the valve tissue, only a minority of re-operated prostheses are subjected to microbiological diagnostic evaluation. In this review, we focus on the diversity and the complete spectrum of PVE-associated bacterial, fungal and viral pathogens in blood, and prosthetic heart valve, their possible virulence potential, and their challenges in making a microbial diagnosis. We are curious to understand if the unacceptable high mortality of PVE is associated with the high number of negative microbial findings in connection with a possible PVE. Herein, we discuss the possibilities and limits of the diagnostic methods conventionally used and make recommendations for enhanced pathogen identification. We also show possible virulence factors of the most common PVE-associated pathogens and their clinical effects. Based on blood culture, molecular biological diagnostics, and specific valve examination, better derivations for the antibiotic therapy as well as possible preventive intervention can be established in the future.
Collapse
Affiliation(s)
- Andreas Oberbach
- Department of Cardiac Surgery, Ludwig Maximilian University, Munich, Germany.,Fraunhofer Institute for Cell Therapy and Immunology, Department of Diagnostics, Leipzig, Germany
| | - Nadine Schlichting
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Diagnostics, Leipzig, Germany.,Institute of Clinical Immunology, University of Leipzig, Leipzig, Germany
| | - Christian Hagl
- Department of Cardiac Surgery, Ludwig Maximilian University, Munich, Germany.,Munich Heart Alliance, Partner Site German Centre for Cardiovascular Disease (DZHK), Munich, Germany
| | - Stefanie Lehmann
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Diagnostics, Leipzig, Germany.,Institute of Clinical Immunology, University of Leipzig, Leipzig, Germany
| | - Yvonne Kullnick
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Diagnostics, Leipzig, Germany.,Institute of Clinical Immunology, University of Leipzig, Leipzig, Germany
| | - Maik Friedrich
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Diagnostics, Leipzig, Germany.,Institute of Clinical Immunology, University of Leipzig, Leipzig, Germany
| | - Ulrike Köhl
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Diagnostics, Leipzig, Germany.,Institute of Clinical Immunology, University of Leipzig, Leipzig, Germany
| | - Friedemann Horn
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Diagnostics, Leipzig, Germany.,Institute of Clinical Immunology, University of Leipzig, Leipzig, Germany
| | - Vivek Kumbhari
- Department of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic, Florida, USA
| | - Bettina Löffler
- Institute of Medical Microbiology, Jena University Hospital, Jena, Germany
| | - Frank Schmidt
- Proteomics Core, Weill Cornell Medical Centre Qatar, Doha, Qatar
| | - Dominik Joskowiak
- Department of Cardiac Surgery, Ludwig Maximilian University, Munich, Germany
| | - Frank Born
- Department of Cardiac Surgery, Ludwig Maximilian University, Munich, Germany
| | - Shekhar Saha
- Department of Cardiac Surgery, Ludwig Maximilian University, Munich, Germany
| | - Erik Bagaev
- Department of Cardiac Surgery, Ludwig Maximilian University, Munich, Germany
| |
Collapse
|
17
|
Zhao H, Clevenger AL, Coburn PS, Callegan MC, Rybenkov V. Condensins are essential for Pseudomonas aeruginosa corneal virulence through their control of lifestyle and virulence programs. Mol Microbiol 2022; 117:937-957. [PMID: 35072315 PMCID: PMC9512581 DOI: 10.1111/mmi.14883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 01/20/2022] [Accepted: 01/20/2022] [Indexed: 12/01/2022]
Abstract
Pseudomonas aeruginosa is a significant opportunistic pathogen responsible for numerous human infections. Its high pathogenicity resides in a diverse array of virulence factors and an ability to adapt to hostile environments. We report that these factors are tied to the activity of condensins, SMC and MksBEF, which primarily function in structural chromosome maintenance. This study revealed that both proteins are required for P. aeruginosa virulence during corneal infection. The reduction in virulence was traced to broad changes in gene expression. Transcriptional signatures of smc and mksB mutants were largely dissimilar and non-additive, with the double mutant displaying a distinct gene expression profile. Affected regulons included those responsible for lifestyle control, primary metabolism, surface adhesion and biofilm growth, iron and sulfur assimilation, and numerous virulence factors, including type 3 and type 6 secretion systems. The in vitro phenotypes of condensin mutants mirrored their transcriptional profiles and included impaired production and secretion of multiple virulence factors, growth deficiencies under nutrient limiting conditions, and altered c-di-GMP signaling. Notably, c-di-GMP mediated some but not all transcriptional responses of the mutants. Thus, condensins are integrated into the control of multiple genetic programs related to epigenetic and virulent behavior of P. aeruginosa.
Collapse
Affiliation(s)
- Hang Zhao
- Department of Chemistry and Biochemistry, University of Oklahoma, 101 Stephenson Parkway, Norman, OK 73019, USA
| | - April L. Clevenger
- Department of Chemistry and Biochemistry, University of Oklahoma, 101 Stephenson Parkway, Norman, OK 73019, USA
| | - Phillip S. Coburn
- Department of Ophthalmology, the University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd., PA-418, Oklahoma City, OK73104, USA
| | - Michelle C. Callegan
- Department of Ophthalmology, the University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd., PA-418, Oklahoma City, OK73104, USA
| | - Valentin Rybenkov
- Department of Chemistry and Biochemistry, University of Oklahoma, 101 Stephenson Parkway, Norman, OK 73019, USA
| |
Collapse
|
18
|
Pliska N. Epidemiology of traumatic injuries and associated infectious complications in the Republic of Kazakhstan. J Med Life 2022; 15:509-514. [PMID: 35646172 PMCID: PMC9126446 DOI: 10.25122/jml-2021-0377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/08/2021] [Indexed: 11/15/2022] Open
Abstract
Traumatism is one of the most important contemporary medical and social issues for most countries worldwide. Since the 20th century, the urgency of traumatism has been increasing. There was an increase in fatal traumatism, including non-fatal cases resulting in permanent disability or temporary disability. This study aimed to investigate the epidemiology of injuries in the Republic of Kazakhstan and identify the statistical patterns of surgical treatment. Furthermore, this study aimed to identify the incidence of infectious complications in patients who received trauma and orthopedic care, their structure and dynamics, and compare this data with the literature. From 2017 to 2019, there were more upper and lower extremity injuries in the Republic of Kazakhstan in the age group of 15-17 years, which corresponds to Russian statistics. Of the 10 injuries, one in three undergoes surgical intervention. In two large cities, Nur-Sultan and Almaty, surgical interventions are performed more often than in other regions. The most frequent infectious complication associated with traumatism is osteomyelitis, with the most causative species being staphylococci.
Collapse
Affiliation(s)
- Natalya Pliska
- Immunobacteriological Laboratory, National Scientific Center of Traumatology and Orthopedics, Nur-Sultan, Republic of Kazakhstan,Corresponding Author: Natalya Pliska, Immunobacteriological Laboratory, National Scientific Center of Traumatology and Orthopedics, Nur-Sultan, Republic of Kazakhstan. E-mail:
| |
Collapse
|
19
|
Hardy KS, Tuckey AN, Housley NA, Andrews J, Patel M, Al-Mehdi AB, Barrington RA, Cassel SL, Sutterwala FS, Audia JP. The Pseudomonas aeruginosa Type III Secretion System Exoenzyme Effector ExoU Induces Mitochondrial Damage in a Murine Bone Marrow-Derived Macrophage Infection Model. Infect Immun 2022; 90:e0047021. [PMID: 35130452 PMCID: PMC8929383 DOI: 10.1128/iai.00470-21] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 01/18/2022] [Indexed: 11/20/2022] Open
Abstract
Pseudomonas aeruginosa is a Gram-negative, opportunistic pathogen that causes nosocomial pneumonia, urinary tract infections, and bacteremia. A hallmark of P. aeruginosa pathogenesis is disruption of host cell function by the type III secretion system (T3SS) and its cognate exoenzyme effectors. The T3SS effector ExoU is phospholipase A2 (PLA2) that targets the host cell plasmalemmal membrane to induce cytolysis and is an important virulence factor that mediates immune avoidance. In addition, ExoU has been shown to subvert the host inflammatory response in a noncytolytic manner. In primary bone marrow-derived macrophages (BMDMs), P. aeruginosa infection is sensed by the nucleotide-binding domain containing leucine-rich repeats-like receptor 4 (NLRC4) inflammasome, which triggers caspase-1 activation and inflammation. ExoU transiently inhibits NLRC4 inflammasome-mediated activation of caspase-1 and its downstream target, interleukin 1β (IL-1β), to suppress activation of inflammation. In the present study, we sought to identify additional noncytolytic virulence functions for ExoU and discovered an unexpected association between ExoU, host mitochondria, and NLRC4. We show that infection of BMDMs with P. aeruginosa strains expressing ExoU elicited mitochondrial oxidative stress. In addition, mitochondria and mitochondrion-associated membrane fractions enriched from infected cells exhibited evidence of autophagy activation, indicative of damage. The observation that ExoU elicited mitochondrial stress and damage suggested that ExoU may also associate with mitochondria during infection. Indeed, ExoU phospholipase A2 enzymatic activity was present in enriched mitochondria and mitochondrion-associated membrane fractions isolated from P. aeruginosa-infected BMDMs. Intriguingly, enriched mitochondria and mitochondrion-associated membrane fractions isolated from infected Nlrc4 homozygous knockout BMDMs displayed significantly lower levels of ExoU enzyme activity, suggesting that NLRC4 plays a role in the ExoU-mitochondrion association. These observations prompted us to assay enriched mitochondria and mitochondrion-associated membrane fractions for NLRC4, caspase-1, and IL-1β. NLRC4 and pro-caspase-1 were detected in enriched mitochondria and mitochondrion-associated membrane fractions isolated from noninfected BMDMs, and active caspase-1 and active IL-1β were detected in response to P. aeruginosa infection. Interestingly, ExoU inhibited mitochondrion-associated caspase-1 and IL-1β activation. The implications of ExoU-mediated effects on mitochondria and the NLRC4 inflammasome during P. aeruginosa infection are discussed.
Collapse
Affiliation(s)
- Kierra S. Hardy
- Department of Microbiology and Immunology, University of South Alabama College of Medicine, Mobile, Alabama, USA
- Center for Lung Biology, University of South Alabama College of Medicine, Mobile, Alabama, USA
| | - Amanda N. Tuckey
- Department of Microbiology and Immunology, University of South Alabama College of Medicine, Mobile, Alabama, USA
- Center for Lung Biology, University of South Alabama College of Medicine, Mobile, Alabama, USA
| | - Nicole A. Housley
- Department of Microbiology and Immunology, University of South Alabama College of Medicine, Mobile, Alabama, USA
- Center for Lung Biology, University of South Alabama College of Medicine, Mobile, Alabama, USA
| | - Joel Andrews
- Mitchell Cancer Institute, University of South Alabama College of Medicine, Mobile, Alabama, USA
| | - Mita Patel
- Department of Pharmcology, University of South Alabama College of Medicine, Mobile, Alabama, USA
| | - Abu-Bakr Al-Mehdi
- Department of Pharmcology, University of South Alabama College of Medicine, Mobile, Alabama, USA
| | - Robert A. Barrington
- Department of Microbiology and Immunology, University of South Alabama College of Medicine, Mobile, Alabama, USA
- Center for Lung Biology, University of South Alabama College of Medicine, Mobile, Alabama, USA
| | - Suzanne L. Cassel
- Women’s Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Fayyaz S. Sutterwala
- Women’s Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Jonathon P. Audia
- Department of Microbiology and Immunology, University of South Alabama College of Medicine, Mobile, Alabama, USA
- Center for Lung Biology, University of South Alabama College of Medicine, Mobile, Alabama, USA
| |
Collapse
|
20
|
Naito Y, Kato H, Zhou L, Sugita S, He H, Zheng J, Hao Q, Sawa T, Lee JW. Therapeutic Effects of Hyaluronic Acid Against Cytotoxic Extracellular Vesicles Released During Pseudomonas Aeruginosa Pneumonia. Shock 2022; 57:408-416. [PMID: 34387224 PMCID: PMC8840981 DOI: 10.1097/shk.0000000000001846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT Extracellular vesicles (EVs) have now been recognized as important mediators of cellular communication during injury and repair. We previously found that plasma EVs isolated from ex vivo perfused human lungs injured with Escherichia coli bacterial pneumonia were inflammatory, and exogenous administration of high molecular weight (HMW) hyaluronic acid (HA) as therapy bound to these EVs, decreasing inflammation and injury. In the current study, we studied the role of EVs released during severe Pseudomonas aeruginosa (PA) pneumonia in mice and determined whether intravenous administration of exogenous HMW HA would have therapeutic effects against the bacterial pneumonia. EVs were collected from the bronchoalveolar lavage fluid (BALF) of mice infected with PA103 by ultracentrifugation and analyzed by NanoSight and flow cytometry. In a cytotoxicity assay, administration of EVs released from infected mice (I-EVs) decreased the viability of A549 cells compared to EV isolated from sham control mice (C-EVs). Either exogenous HMW HA or an anti-CD44 antibody, when co-incubated with I-EVs, significantly improved the viability of the A549 cells. In mice with PA103 pneumonia, administration of HMW HA improved pulmonary edema and bacterial count in the lungs and decreased TNF-α and caspase-3 levels in the supernatant of lung homogenates. In conclusion, EVs isolated from BALF of mice with P. aeruginosa pneumonia were cytotoxic and inflammatory, and intravenous HMW HA administration was protective against P. aeruginosa pneumonia.
Collapse
Affiliation(s)
- Yoshifumi Naito
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, Japan
| | - Hideya Kato
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, Japan
| | - Li Zhou
- Department of Anesthesiology, University of California San Francisco, San Francisco, California
| | - Shinji Sugita
- Department of Anesthesiology, University of California San Francisco, San Francisco, California
| | - Hongli He
- Department of Anesthesiology, University of California San Francisco, San Francisco, California
| | - Justin Zheng
- Department of Anesthesiology, University of California San Francisco, San Francisco, California
| | - Qi Hao
- Department of Anesthesiology, University of California San Francisco, San Francisco, California
| | - Teiji Sawa
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, Japan
| | - Jae-Woo Lee
- Department of Anesthesiology, University of California San Francisco, San Francisco, California
| |
Collapse
|
21
|
Hardy KS, Tuckey AN, Renema P, Patel M, Al-Mehdi AB, Spadafora D, Schlumpf CA, Barrington RA, Alexeyev MF, Stevens T, Pittet JF, Wagener BM, Simmons JD, Alvarez DF, Audia JP. ExoU Induces Lung Endothelial Cell Damage and Activates Pro-Inflammatory Caspase-1 during Pseudomonas aeruginosa Infection. Toxins (Basel) 2022; 14:toxins14020152. [PMID: 35202178 PMCID: PMC8878379 DOI: 10.3390/toxins14020152] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/07/2022] [Accepted: 02/11/2022] [Indexed: 12/24/2022] Open
Abstract
The Gram-negative, opportunistic pathogen Pseudomonas aeruginosa utilizes a type III secretion system to inject exoenzyme effectors into a target host cell. Of the four best-studied exoenzymes, ExoU causes rapid cell damage and death. ExoU is a phospholipase A2 (PLA2) that hydrolyses host cell membranes, and P. aeruginosa strains expressing ExoU are associated with poor outcomes in critically ill patients with pneumonia. While the effects of ExoU on lung epithelial and immune cells are well studied, a role for ExoU in disrupting lung endothelial cell function has only recently emerged. Lung endothelial cells maintain a barrier to fluid and protein flux into tissue and airspaces and regulate inflammation. Herein, we describe a pulmonary microvascular endothelial cell (PMVEC) culture infection model to examine the effects of ExoU. Using characterized P. aeruginosa strains and primary clinical isolates, we show that strains expressing ExoU disrupt PMVEC barrier function by causing substantial PMVEC damage and lysis, in a PLA2-dependent manner. In addition, we show that strains expressing ExoU activate the pro-inflammatory caspase-1, in a PLA2-dependent manner. Considering the important roles for mitochondria and oxidative stress in regulating inflammatory responses, we next examined the effects of ExoU on reactive oxygen species production. Infection of PMVECs with P. aeruginosa strains expressing ExoU triggered a robust oxidative stress compared to strains expressing other exoenzyme effectors. We also provide evidence that, intriguingly, ExoU PLA2 activity was detectable in mitochondria and mitochondria-associated membrane fractions isolated from P. aeruginosa-infected PMVECs. Interestingly, ExoU-mediated activation of caspase-1 was partially inhibited by reactive oxygen species scavengers. Together, these data suggest ExoU exerts pleiotropic effects on PMVEC function during P. aeruginosa infection that may inhibit endothelial barrier and inflammatory functions.
Collapse
Affiliation(s)
- Kierra S. Hardy
- Department of Microbiology and Immunology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA; (K.S.H.); (A.N.T.); (C.A.S.); (R.A.B.)
- Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA; (P.R.); (M.P.); (A.-B.A.-M.); (M.F.A.); (T.S.); (J.D.S.); (D.F.A.)
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Amanda N. Tuckey
- Department of Microbiology and Immunology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA; (K.S.H.); (A.N.T.); (C.A.S.); (R.A.B.)
- Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA; (P.R.); (M.P.); (A.-B.A.-M.); (M.F.A.); (T.S.); (J.D.S.); (D.F.A.)
| | - Phoibe Renema
- Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA; (P.R.); (M.P.); (A.-B.A.-M.); (M.F.A.); (T.S.); (J.D.S.); (D.F.A.)
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA
- Department of Biomedical Sciences, College of Allied Health, University of South Alabama Mobile, Mobile, AL 36688, USA
| | - Mita Patel
- Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA; (P.R.); (M.P.); (A.-B.A.-M.); (M.F.A.); (T.S.); (J.D.S.); (D.F.A.)
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA
| | - Abu-Bakr Al-Mehdi
- Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA; (P.R.); (M.P.); (A.-B.A.-M.); (M.F.A.); (T.S.); (J.D.S.); (D.F.A.)
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA
| | - Domenico Spadafora
- Flow Cytometry Core Lab, College of Medicine, University of South Alabama, Mobile, AL 36688, USA;
| | - Cody A. Schlumpf
- Department of Microbiology and Immunology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA; (K.S.H.); (A.N.T.); (C.A.S.); (R.A.B.)
| | - Robert A. Barrington
- Department of Microbiology and Immunology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA; (K.S.H.); (A.N.T.); (C.A.S.); (R.A.B.)
- Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA; (P.R.); (M.P.); (A.-B.A.-M.); (M.F.A.); (T.S.); (J.D.S.); (D.F.A.)
- Flow Cytometry Core Lab, College of Medicine, University of South Alabama, Mobile, AL 36688, USA;
| | - Mikhail F. Alexeyev
- Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA; (P.R.); (M.P.); (A.-B.A.-M.); (M.F.A.); (T.S.); (J.D.S.); (D.F.A.)
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA
| | - Troy Stevens
- Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA; (P.R.); (M.P.); (A.-B.A.-M.); (M.F.A.); (T.S.); (J.D.S.); (D.F.A.)
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA
| | - Jean-Francois Pittet
- Department of Anesthesiology and Perioperative Medicine, Birmingham School of Medicine, University of Alabama, Birmingham, AL 35294, USA; (J.-F.P.); (B.M.W.)
| | - Brant M. Wagener
- Department of Anesthesiology and Perioperative Medicine, Birmingham School of Medicine, University of Alabama, Birmingham, AL 35294, USA; (J.-F.P.); (B.M.W.)
| | - Jon D. Simmons
- Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA; (P.R.); (M.P.); (A.-B.A.-M.); (M.F.A.); (T.S.); (J.D.S.); (D.F.A.)
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA
- Department of Surgery, College of Medicine, University of South Alabama, Mobile, AL 36688, USA
| | - Diego F. Alvarez
- Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA; (P.R.); (M.P.); (A.-B.A.-M.); (M.F.A.); (T.S.); (J.D.S.); (D.F.A.)
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA
- Department of Physiology and Pharmacology, College of Osteopathic Medicine, Sam Houston State University, Conroe, TX 77304, USA
| | - Jonathon P. Audia
- Department of Microbiology and Immunology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA; (K.S.H.); (A.N.T.); (C.A.S.); (R.A.B.)
- Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA; (P.R.); (M.P.); (A.-B.A.-M.); (M.F.A.); (T.S.); (J.D.S.); (D.F.A.)
- Correspondence:
| |
Collapse
|
22
|
Potential Therapeutic Targets for Combination Antibody Therapy against Pseudomonas aeruginosa Infections. Antibiotics (Basel) 2021; 10:antibiotics10121530. [PMID: 34943742 PMCID: PMC8698887 DOI: 10.3390/antibiotics10121530] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/03/2021] [Accepted: 12/09/2021] [Indexed: 12/12/2022] Open
Abstract
Despite advances in antimicrobial therapy and even the advent of some effective vaccines, Pseudomonas aeruginosa (P. aeruginosa) remains a significant cause of infectious disease, primarily due to antibiotic resistance. Although P. aeruginosa is commonly treatable with readily available therapeutics, these therapies are not always efficacious, particularly for certain classes of patients (e.g., cystic fibrosis (CF)) and for drug-resistant strains. Multi-drug resistant P. aeruginosa infections are listed on both the CDC’s and WHO’s list of serious worldwide threats. This increasing emergence of drug resistance and prevalence of P. aeruginosa highlights the need to identify new therapeutic strategies. Combinations of monoclonal antibodies against different targets and epitopes have demonstrated synergistic efficacy with each other as well as in combination with antimicrobial agents typically used to treat these infections. Such a strategy has reduced the ability of infectious agents to develop resistance. This manuscript details the development of potential therapeutic targets for polyclonal antibody therapies to combat the emergence of multidrug-resistant P. aeruginosa infections. In particular, potential drug targets for combinational immunotherapy against P. aeruginosa are identified to combat current and future drug resistance.
Collapse
|
23
|
Hardy KS, Tessmer MH, Frank DW, Audia JP. Perspectives on the Pseudomonas aeruginosa Type III Secretion System Effector ExoU and Its Subversion of the Host Innate Immune Response to Infection. Toxins (Basel) 2021; 13:880. [PMID: 34941717 PMCID: PMC8708460 DOI: 10.3390/toxins13120880] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/19/2021] [Accepted: 12/04/2021] [Indexed: 12/02/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic, Gram-negative pathogen and an important cause of hospital acquired infections, especially in immunocompromised patients. Highly virulent P. aeruginosa strains use a type III secretion system (T3SS) to inject exoenzyme effectors directly into the cytoplasm of a target host cell. P. aeruginosa strains that express the T3SS effector, ExoU, associate with adverse outcomes in critically ill patients with pneumonia, owing to the ability of ExoU to rapidly damage host cell membranes and subvert the innate immune response to infection. Herein, we review the structure, function, regulation, and virulence characteristics of the T3SS effector ExoU, a highly cytotoxic phospholipase A2 enzyme.
Collapse
Affiliation(s)
- Kierra S. Hardy
- Department of Microbiology and Immunology, University of South Alabama College of Medicine, Mobile, AL 36608, USA;
- Center for Lung Biology, University of South Alabama College of Medicine, Mobile, AL 36608, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Maxx H. Tessmer
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA;
| | - Dara W. Frank
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Jonathon P. Audia
- Department of Microbiology and Immunology, University of South Alabama College of Medicine, Mobile, AL 36608, USA;
- Center for Lung Biology, University of South Alabama College of Medicine, Mobile, AL 36608, USA
| |
Collapse
|
24
|
Su R, Zhang Y, Zhang J, Wang H, Luo Y, Chan HF, Tao Y, Chen Z, Li M. Nanomedicine to advance the treatment of bacteria-induced acute lung injury. J Mater Chem B 2021; 9:9100-9115. [PMID: 34672317 DOI: 10.1039/d1tb01770e] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Bacteria-induced acute lung injury (ALI) is associated with a high mortality rate due to the lack of an effective treatment. Patients often rely on supportive care such as low tidal volume ventilation to alleviate the symptoms. Nanomedicine has recently received much attention owing to its premium benefits of delivering drugs in a sustainable and controllable manner while minimizing the potential side effects. It can effectively improve the prognosis of bacteria-induced ALI through targeted delivery of drugs, regulation of multiple inflammatory pathways, and combating antibiotic resistance. Hence, in this review, we first discuss the pathogenesis of ALI and its potential therapeutics. In particular, the state-of-the-art nanomedicines for the treatment of bacteria-induced ALI are highlighted, including their administration routes, in vivo distribution, and clearance. Furthermore, the available bacteria-induced ALI animal models are also summarized. In the end, future perspectives of nanomedicine for ALI treatment are proposed.
Collapse
Affiliation(s)
- Ruonan Su
- Center for Nanomedicine, Department of Pediatrics and Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China. .,Laboratory of Biomaterials and Translational Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Yu Zhang
- Department of Biological and Environmental Engineering, Cornell University, Ithaca 14853, USA
| | - Jiabin Zhang
- Center for Nanomedicine, Department of Pediatrics and Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Haixia Wang
- Center for Nanomedicine, Department of Pediatrics and Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China. .,Laboratory of Biomaterials and Translational Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Yun Luo
- Department of Urology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China
| | - Hon Fai Chan
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Science, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Yu Tao
- Center for Nanomedicine, Department of Pediatrics and Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China. .,Laboratory of Biomaterials and Translational Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Zhuanggui Chen
- Center for Nanomedicine, Department of Pediatrics and Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Mingqiang Li
- Center for Nanomedicine, Department of Pediatrics and Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China. .,Laboratory of Biomaterials and Translational Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.,Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, China
| |
Collapse
|
25
|
Belyy A, Merino F, Mechold U, Raunser S. Mechanism of actin-dependent activation of nucleotidyl cyclase toxins from bacterial human pathogens. Nat Commun 2021; 12:6628. [PMID: 34785651 PMCID: PMC8595890 DOI: 10.1038/s41467-021-26889-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 10/25/2021] [Indexed: 12/19/2022] Open
Abstract
Bacterial human pathogens secrete initially inactive nucleotidyl cyclases that become potent enzymes by binding to actin inside eukaryotic host cells. The underlying molecular mechanism of this activation is, however, unclear. Here, we report structures of ExoY from Pseudomonas aeruginosa and Vibrio vulnificus bound to their corresponding activators F-actin and profilin-G-actin. The structures reveal that in contrast to the apo-state, two flexible regions become ordered and interact strongly with actin. The specific stabilization of these regions results in an allosteric stabilization of the nucleotide binding pocket and thereby to an activation of the enzyme. Differences in the sequence and conformation of the actin-binding regions are responsible for the selective binding to either F- or G-actin. Other nucleotidyl cyclase toxins that bind to calmodulin rather than actin undergo a similar disordered-to-ordered transition during activation, suggesting that the allosteric activation-by-stabilization mechanism of ExoY is conserved in these enzymes, albeit the different activator.
Collapse
Affiliation(s)
- Alexander Belyy
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Otto-Hahn-Str. 11, 44227, Dortmund, Germany
| | - Felipe Merino
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Otto-Hahn-Str. 11, 44227, Dortmund, Germany
- Department of Protein Evolution, Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076, Tübingen, Germany
| | - Undine Mechold
- Unité de Biochimie des Interactions Macromoléculaires, Département de Biologie Structurale et Chimie, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Stefan Raunser
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Otto-Hahn-Str. 11, 44227, Dortmund, Germany.
| |
Collapse
|
26
|
The Epidemiology and Pathogenesis and Treatment of Pseudomonas aeruginosa Infections: An Update. Drugs 2021; 81:2117-2131. [PMID: 34743315 PMCID: PMC8572145 DOI: 10.1007/s40265-021-01635-6] [Citation(s) in RCA: 215] [Impact Index Per Article: 71.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2021] [Indexed: 12/20/2022]
Abstract
Pseudomonas aeruginosa is a Gram-negative bacterial pathogen that is a common cause of nosocomial infections, particularly pneumonia, infection in immunocompromised hosts, and in those with structural lung disease such as cystic fibrosis. Epidemiological studies have
identified increasing trends of antimicrobial resistance, including multi-drug resistant (MDR) isolates in recent years. P. aeruginosa has several virulence mechanisms that increase its ability to cause severe infections, such as secreted toxins, quorum sensing and biofilm formation. Management of P. aeruginosa infections focuses on prevention when possible, obtaining cultures, and prompt initiation of antimicrobial therapy, occasionally with combination therapy depending on the clinical scenario to ensure activity against P. aeruginosa. Newer anti-pseudomonal antibiotics are available and are increasingly being used in the management of MDR P. aeruginosa.
Collapse
|
27
|
Song Y, Zhang S, Luo G, Shen Y, Li C, Zhu Y, Huang Q, Mou X, Tang X, Liu T, Wu S, Tong A, He Y, Bao R. Type II Antitoxin HigA Is a Key Virulence Regulator in Pseudomonas aeruginosa. ACS Infect Dis 2021; 7:2930-2940. [PMID: 34554722 DOI: 10.1021/acsinfecdis.1c00401] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Bacterial type II toxin-antitoxin (TA) systems are abundant genetic elements and are involved in a diverse array of physiological processes. These systems encode an antitoxin protein that directly binds and effectively neutralizes the protein toxin. Recent studies have highlighted the key roles of type II TA modules in bacterial virulence and pathogenesis, but the underlying mechanisms remain unclear. Here, we investigated the antitoxin HigA in Pseudomonas aeruginosa infection. Proteomic analysis of the higA deletion strain revealed an enhanced expression of pathogenic proteins. We further verified that HigA negatively controlled T3SS and T6SS expression by directly interacting with the promoter regions of the regulators amrZ and exsA, respectively. In other words, the reversal of HigA-mediated transcriptional inhibition on stress stimulation could induce virulence genes. These findings confirm the crucial roles of the type II antitoxin in bacterial infection, which highlights the potential of the HigBA TA system as an antibacterial treatment target.
Collapse
Affiliation(s)
- Yingjie Song
- Center of Infectious Diseases, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610093, China
| | - Siping Zhang
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou 730000, P.R. China
| | - Guihua Luo
- Center of Infectious Diseases, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610093, China
| | - Yalin Shen
- Center of Infectious Diseases, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610093, China
| | - Changcheng Li
- Center of Infectious Diseases, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610093, China
| | - Yibo Zhu
- Center of Infectious Diseases, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610093, China
| | - Qin Huang
- Center of Infectious Diseases, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610093, China
| | - Xingyu Mou
- Center of Infectious Diseases, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610093, China
| | - Xinyue Tang
- Center of Infectious Diseases, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610093, China
| | - Tonggen Liu
- Center of Infectious Diseases, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610093, China
| | - Siying Wu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610044, China
| | - Aiping Tong
- Center of Infectious Diseases, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610093, China
| | - Yongxing He
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou 730000, P.R. China
| | - Rui Bao
- Center of Infectious Diseases, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610093, China
| |
Collapse
|
28
|
Rocha LS, Silva BPD, Correia TML, Silva RPD, Meireles DDA, Pereira R, Netto LES, Meotti FC, Queiroz RF. Peroxiredoxin AhpC1 protects Pseudomonas aeruginosa against the inflammatory oxidative burst and confers virulence. Redox Biol 2021; 46:102075. [PMID: 34315109 PMCID: PMC8327333 DOI: 10.1016/j.redox.2021.102075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/13/2021] [Accepted: 07/17/2021] [Indexed: 11/16/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic bacterium in patients with cystic fibrosis and hospital acquired infections. It presents a plethora of virulence factors and antioxidant enzymes that help to subvert the immune system. In this study, we identified the 2-Cys peroxiredoxin, alkyl-hydroperoxide reductase C1 (AhpC1), as a relevant scavenger of oxidants generated during inflammatory oxidative burst and a mechanism of P. aeruginosa (PA14) escaping from killing. Deletion of AhpC1 led to a higher sensitivity to hypochlorous acid (HOCl, IC50 3.2 ± 0.3 versus 19.1 ± 0.2 μM), hydrogen peroxide (IC50 91.2 ± 0.3 versus 496.5 ± 6.4 μM) and the organic peroxide urate hydroperoxide. ΔahpC1 strain was more sensitive to the killing by isolated neutrophils and less virulent in a mice model of infection. All mice intranasally instilled with ΔahpC1 survived as long as they were monitored (15 days), whereas 100% wild-type and ΔahpC1 complemented with ahpC1 gene (ΔahpC1 attB:ahpC1) died within 3 days. A significantly lower number of colonies was detected in the lung and spleen of ΔahpC1-infected mice. Total leucocytes, neutrophils, myeloperoxidase activity, pro-inflammatory cytokines, nitrite production and lipid peroxidation were much lower in lungs or bronchoalveolar liquid of mice infected with ΔahpC1. Purified AhpC neutralized the inflammatory organic peroxide, urate hydroperoxide, at a rate constant of 2.3 ± 0.1 × 106 M-1s-1, and only the ΔahpC1 strain was sensitive to this oxidant. Incubation of neutrophils with uric acid, the urate hydroperoxide precursor, impaired neutrophil killing of wild-type but improved the killing of ΔahpC1. Hyperuricemic mice presented higher levels of serum cytokines and succumbed much faster to PA14 infection when compared to normouricemic mice. In summary, ΔahpC1 PA14 presented a lower virulence, which was attributed to a poorer ability to neutralize the oxidants generated by inflammatory oxidative burst, leading to a more efficient killing by the host. The enzyme is particularly relevant in detoxifying the newly reported inflammatory organic peroxide, urate hydroperoxide.
Collapse
Affiliation(s)
- Leonardo Silva Rocha
- Programa Multicêntrico de Pós-graduação em Bioquímica e Biologia Molecular, Universidade Estadual do Sudoeste da Bahia, Brazil
| | | | - Thiago M L Correia
- Programa Multicêntrico de Pós-graduação Multicêntrico em Ciências Fisiológicas, Instituto Multidisciplinar de Saúde, Universidade Federal da Bahia, Brazil
| | | | - Diogo de Abreu Meireles
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, Brazil
| | - Rafael Pereira
- Programa Multicêntrico de Pós-graduação em Bioquímica e Biologia Molecular, Universidade Estadual do Sudoeste da Bahia, Brazil; Programa Multicêntrico de Pós-graduação Multicêntrico em Ciências Fisiológicas, Instituto Multidisciplinar de Saúde, Universidade Federal da Bahia, Brazil; Departamento de Ciências Biológicas, Universidade Estadual do Sudoeste da Bahia, Brazil
| | - Luis Eduardo Soares Netto
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, Brazil
| | - Flavia Carla Meotti
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Brazil.
| | - Raphael Ferreira Queiroz
- Programa Multicêntrico de Pós-graduação em Bioquímica e Biologia Molecular, Universidade Estadual do Sudoeste da Bahia, Brazil; Departamento de Ciências Naturais, Universidade Estadual do Sudoeste da Bahia, Brazil.
| |
Collapse
|
29
|
A Primed Subpopulation of Bacteria Enables Rapid Expression of the Type 3 Secretion System in Pseudomonas aeruginosa. mBio 2021; 12:e0083121. [PMID: 34154400 PMCID: PMC8262847 DOI: 10.1128/mbio.00831-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Type 3 secretion systems (T3SS) are complex nanomachines that span the cell envelope and play a central role in the biology of Gram-negative pathogens and symbionts. In Pseudomonas aeruginosa, T3SS expression is strongly associated with human disease severity and with mortality in murine acute pneumonia models. Uniform exposure of isogenic cells to T3SS-activating signal results in heterogeneous expression of this critical virulence trait. To understand the function of such diversity, we measured the production of the T3SS master regulator ExsA and the expression of T3SS genes using fluorescent reporters. We found that heterogeneous expression of ExsA in the absence of activating signal generates a "primed" subpopulation of cells that can rapidly induce T3SS gene expression in response to signal. T3SS expression is accompanied by a reproductive trade-off as measured by increased division time of T3SS-expressing cells. Although T3SS-primed cells are a minority of the population, they compose the majority of T3SS-expressing cells for several hours following activation. The primed state therefore allows P. aeruginosa to maximize reproductive fitness while maintaining the capacity to quickly express the T3SS. As T3SS effectors can serve as shared public goods for nonproducing cells, this division of labor benefits the population as a whole. IMPORTANCE The expression of specific virulence traits is strongly associated with Pseudomonas aeruginosa's success in establishing acute infections but is thought to carry a cost for bacteria. Producing multiprotein secretion systems or motility organelles is metabolically expensive and can target a cell for recognition by innate immune system receptors that recognize structural components of the type 3 secretion system (T3SS) or flagellum. These acute virulence factors are also negatively selected when P. aeruginosa establishes chronic infections in the lung. We demonstrate a regulatory mechanism by which only a minority subpopulation of genetically identical P. aeruginosa cells is "primed" to respond to signals that turn on T3SS expression. This phenotypic heterogeneity allows the population to maximize the benefit of rapid T3SS effector production while maintaining a rapidly growing and nonexpressing reservoir of cells that perpetuates this genotype within the population.
Collapse
|
30
|
A pipeline to evaluate inhibitors of the Pseudomonas aeruginosa exotoxin U. Biochem J 2021; 478:647-668. [PMID: 33459338 PMCID: PMC7886320 DOI: 10.1042/bcj20200780] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 01/15/2021] [Accepted: 01/18/2021] [Indexed: 01/07/2023]
Abstract
Pseudomonas aeruginosa has recently been highlighted by the World Health Organisation (WHO) as a major threat with high priority for the development of new therapies. In severe P. aeruginosa infections, the phospholipase activity of the type 3 secretion system toxin, ExoU, induces lysis of target host cells and results in the poorest clinical outcomes. We have developed an integrated pipeline to evaluate small molecule inhibitors of ExoU in vitro and in cultured cell models, including a disease-relevant corneal epithelial (HCE-T) scratch and infection model using florescence microscopy and cell viability assays. Compounds Pseudolipasin A, compound A and compound B were effective in vitro inhibitors of ExoU and mitigated P. aeruginosa ExoU-dependent cytotoxicity after infection of HCE-T cells at concentrations as low as 0.5 µM. Addition of the antimicrobial moxifloxacin controlled bacterial load, allowing these assays to be extended from 6 h to 24 h. P. aeruginosa remained cytotoxic to HCE-T cells with moxifloxacin, present at the minimal inhibitory concentration for 24 h, but, when used in combination with either Pseudolipasin A, compound A or compound B, a greater amount of viable cells and scratch healing were observed. Thus, our pipeline provides evidence that ExoU inhibitors could be used in combination with certain antimicrobials as a novel means to treat infections due to ExoU producing P. aeruginosa, as well as the means to identify more potent ExoU inhibitors for future therapeutics.
Collapse
|
31
|
Pseudomonas aeruginosa Ventilator-Associated Pneumonia Rabbit Model for Preclinical Drug Development. Antimicrob Agents Chemother 2021; 65:e0272420. [PMID: 33972247 DOI: 10.1128/aac.02724-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Development and validation of large animal models of Pseudomonas aeruginosa ventilator-associated pneumonia are needed for testing new drug candidates in a manner that mimics how they will be used clinically. We developed a new model in which rabbits were ventilated with low tidal volume and challenged with P. aeruginosa to recapitulate hallmark clinical features of acute respiratory distress syndrome (ARDS): acute lung injury and inflammation, progressive decrease in arterial oxygen partial pressure to fractional inspired oxygen PaO2:FiO2, leukopenia, neutropenia, thrombocytopenia, hyperlactatemia, severe hypotension, bacterial dissemination from lung to other organs, multiorgan dysfunction, and ultimately death. We evaluated the predictive power of this rabbit model for antibiotic efficacy testing by determining whether a humanized dosing regimen of meropenem, a potent antipseudomonal β-lactam antibiotic, when administered with or without intensive care unit (ICU)-supportive care (fluid challenge and norepinephrine), could halt or reverse natural disease progression. Our humanized meropenem dosing regimen produced a plasma concentration-time profile in the rabbit model similar to those reported in patients with ventilator-associated bacterial pneumonia. In this rabbit model, treatment with humanized meropenem and ICU-supportive care achieved the highest level of survival, halted the worsening of ARDS biomarkers, and reversed lethal hypotension, although treatment with humanized meropenem alone also conferred some protection compared to treatment with placebo (saline) alone or placebo plus ICU-supportive care. In conclusion, this rabbit model could help predict whether an antibiotic will be efficacious for the treatment of human ventilator-associated pneumonia.
Collapse
|
32
|
Gupte A, Jyot J, Ravi M, Ramphal R. High pyocyanin production and non-motility of Pseudomonas aeruginosa isolates are correlated with septic shock or death in bacteremic patients. PLoS One 2021; 16:e0253259. [PMID: 34115807 PMCID: PMC8195364 DOI: 10.1371/journal.pone.0253259] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 06/01/2021] [Indexed: 12/26/2022] Open
Abstract
Studies of the outcome of Pseudomonas aeruginosa bacteremia (Pab) have focused mainly on antibiotic appropriateness. However, P. aeruginosa possesses many virulence factors whose roles in outcomes have not been examined in humans, except for the type III secretion system (T3SS) toxins. The purpose of this study was to examine the role of virulence factors other than the T3SS toxins. Bacterial isolates were collected from 75 patients who suffered from Pa blood stream infections. Host factors such as neutropenia, immunosuppression, comorbidities, time to effective antibiotics, source of bacteremia, and presence of multidrug resistant (MDR) isolate were studied. The isolates were analyzed for the presence of toxin genes, proteolytic activity, swimming and twitching motility, and pyocyanin production. The data were analyzed to ascertain which virulence factors correlated with poor outcomes defined as septic shock or death (SS) within 7 days. Septic shock or death occurred in 25/75 patients. Univariate analysis identified age as a host factor that exerted a significant effect on these outcomes. Ineffective antibiotics administered during the first 24 hours of treatment or MDR P. aeruginosa did not influence the frequency of SS, nor did the presence of lasB, exoA, exoS exoU, plcH genes and proteolytic activity. However, 6/8 patients infected with non-motile isolates, developed SS, p = 0.014 and 5/6 isolates that produced large amounts of pyocyanin (>18ug/ml), were associated with SS, p = 0.014. Multivariate analysis indicated that the odds ratio (OR) for development of SS with a non-motile isolate was 6.8, with a 95% confidence interval (CI) (1.37, 51.5), p = 0.030 and with high pyocyanin producing isolates, an OR of 16.9, 95% CI = (2.27, 360), p = .017. This study evaluating the role of microbial factors that significantly effect outcomes following Pa bloodstream infection suggests that P. aeruginosa strains showing high pyocyanin production and the lack of motility independently increase the risk of SS.
Collapse
Affiliation(s)
- Asmita Gupte
- Division of Infectious Diseases and Global Medicine, Department of Medicine, University of Florida, Gainesville, Florida, United States of America
- * E-mail: (AG); (RR)
| | - Jeevan Jyot
- Division of Infectious Diseases and Global Medicine, Department of Medicine, University of Florida, Gainesville, Florida, United States of America
- Department of Pharmaceutical Outcomes & Policy, College of Pharmacy, University of Florida, Gainesville, Florida, United States of America
| | - Malleswari Ravi
- Division of Infectious Diseases and Global Medicine, Department of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Reuben Ramphal
- Division of Infectious Diseases and Global Medicine, Department of Medicine, University of Florida, Gainesville, Florida, United States of America
- * E-mail: (AG); (RR)
| |
Collapse
|
33
|
Horna G, Ruiz J. Type 3 secretion system as an anti-Pseudomonal target. Microb Pathog 2021; 155:104907. [PMID: 33930424 DOI: 10.1016/j.micpath.2021.104907] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 04/17/2021] [Accepted: 04/19/2021] [Indexed: 10/21/2022]
Abstract
Type 3 secretion systems (T3SSs) are a series of mechanisms involved in bacterial pathogenesis. While Pseudomonas aeruginosa only possess one T3SS, it plays a key role in the virulence of P. aeruginosa virulence. This finding suggests that T3SS impairment may be an alternative for antimicrobial agents, allowing P. aeruginosa infections to be directly combated avoiding antimicrobial pressure on this and other microorganisms. To date, different approaches have been proposed, including T3SS inhibition, vaccination strategies, development of anti-T3SS antibodies and gene silencing.
Collapse
Affiliation(s)
- Gertrudis Horna
- Universidad Catolica Los Angeles de Chimbote, Instituto de Investigación, Chimbote, Peru
| | - Joaquim Ruiz
- Laboratorio de Microbiología Molecular y Genómica Bacteriana, Universidad Científica del Sur, Lima, Peru.
| |
Collapse
|
34
|
Laborda P, Alcalde-Rico M, Chini A, Martínez JL, Hernando-Amado S. Discovery of inhibitors of Pseudomonas aeruginosa virulence through the search for natural-like compounds with a dual role as inducers and substrates of efflux pumps. Environ Microbiol 2021; 23:7396-7411. [PMID: 33818002 DOI: 10.1111/1462-2920.15511] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/25/2021] [Accepted: 04/03/2021] [Indexed: 12/26/2022]
Abstract
Multidrug efflux pumps are ancient elements encoded in every genome, from bacteria to humans. In bacteria, in addition to antibiotics, efflux pumps extrude a wide range of substrates, including quorum sensing signals, bacterial metabolites, or plant-produced compounds. This indicates that their original functions may differ from their recently acquired role in the extrusion of antibiotics during human infection. Concerning plant-produced compounds, some of them are substrates and inducers of the same efflux pump, suggesting a coordinated plant/bacteria coevolution. Herein we analyse the ability of 1243 compounds from a Natural Product-Like library to induce the expression of P. aeruginosa mexCD-oprJ or mexAB-oprM efflux pumps' encoding genes. We further characterized natural-like compounds that do not trigger antibiotic resistance in P. aeruginosa and that act as virulence inhibitors, choosing those that were not only inducers but substrates of the same efflux pump. Four compounds impair swarming motility, exotoxin secretion through the Type 3 Secretion System (T3SS) and the ability to kill Caenorhabditis elegans, which might be explained by the downregulation of genes encoding flagellum and T3SS. Our results emphasize the possibility of discovering new anti-virulence drugs by screening natural or natural-like libraries for compounds that behave as both, inducers and substrates of efflux pumps.
Collapse
Affiliation(s)
- Pablo Laborda
- Centro Nacional de Biotecnología, CSIC, Darwin 3, Madrid, 28049, Spain
| | - Manuel Alcalde-Rico
- Centro Nacional de Biotecnología, CSIC, Darwin 3, Madrid, 28049, Spain.,Grupo de Resistencia Antimicrobiana en Bacterias Patógenas y Ambientales (GRABPA), Instituto de Biología, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile.,Millennium Nucleus for Collaborative Research on Bacterial Resistance (MICROB-R), Valparaíso, Chile
| | - Andrea Chini
- Centro Nacional de Biotecnología, CSIC, Darwin 3, Madrid, 28049, Spain
| | - José L Martínez
- Centro Nacional de Biotecnología, CSIC, Darwin 3, Madrid, 28049, Spain
| | | |
Collapse
|
35
|
Horna G, Ruiz J. Type 3 secretion system of Pseudomonas aeruginosa. Microbiol Res 2021; 246:126719. [PMID: 33582609 DOI: 10.1016/j.micres.2021.126719] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 01/19/2021] [Accepted: 01/26/2021] [Indexed: 12/27/2022]
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen, mainly affecting severe patients, such as those in intensive care units (ICUs). High levels of antibiotic resistance and a long battery of virulence factors characterise this pathogen. Among virulence factors, the T3SS (Type 3 Secretion Systems) are especially relevant, being one of the most important virulence factors in P. aeruginosa. T3SS are a complex "molecular syringe" able to inject different effectors in host cells, subverting cell machinery influencing immune responses, and increasing bacterial survival rates. While T3SS have been largely studied and the molecular structure and main effector functions have been established, a series of questions and further points remain to be clarified or established. The key role of T3SS in P. aeruginosa virulence has resulted in the search for T3SS-targeting molecules able to impair their functions and subsequently improve patient outcomes. This review aims to summarise the most relevant features of the P. aeruginosa T3SS.
Collapse
Affiliation(s)
- Gertrudis Horna
- Universidad Catolica Los Angeles de Chimbote, Instituto de Investigación, Chimbote, Peru.
| | - Joaquim Ruiz
- Laboratorio de Microbiología Molecular y Genómica Bacteriana, Universidad Científica del Sur, Panamericana Sur, Km 19, Lima, Peru.
| |
Collapse
|
36
|
Identification of Small Molecules Blocking the Pseudomonas aeruginosa type III Secretion System Protein PcrV. Biomolecules 2021; 11:biom11010055. [PMID: 33406810 PMCID: PMC7824769 DOI: 10.3390/biom11010055] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/21/2020] [Accepted: 12/29/2020] [Indexed: 01/09/2023] Open
Abstract
Pseudomonas aeruginosa is an opportunistic bacterial pathogen that employs its type III secretion system (T3SS) during the acute phase of infection to translocate cytotoxins into the host cell cytoplasm to evade the immune system. The PcrV protein is located at the tip of the T3SS, facilitates the integration of pore-forming proteins into the eukaryotic cell membrane, and is required for translocation of cytotoxins into the host cell. In this study, we used surface plasmon resonance screening to identify small molecule binders of PcrV. A follow-up structure-activity relationship analysis resulted in PcrV binders that protect macrophages in a P. aeruginosa cell-based infection assay. Treatment of P. aeruginosa infections is challenging due to acquired, intrinsic, and adaptive resistance in addition to a broad arsenal of virulence systems such as the T3SS. Virulence blocking molecules targeting PcrV constitute valuable starting points for development of next generation antibacterials to treat infections caused by P. aeruginosa.
Collapse
|
37
|
Papagiannitsis CC, Verra A, Galani V, Xitsas S, Bitar I, Hrabak J, Petinaki E. Unravelling the Features of Success of VIM-Producing ST111 and ST235 Pseudomonas aeruginosa in a Greek Hospital. Microorganisms 2020; 8:E1884. [PMID: 33260774 PMCID: PMC7761518 DOI: 10.3390/microorganisms8121884] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 11/19/2020] [Accepted: 11/26/2020] [Indexed: 01/04/2023] Open
Abstract
The objective of this study was to analyze the characteristics that contribute to the successful dissemination of VIM-producing Pseudomonas aeruginosa (P. aeruginosa), belonging to ST111 and ST235, in a Greek hospital. A total of 120 non-repetitive P. aeruginosa, which had meropenem minimal inhibitory concentrations (MICs) greater than 2 mg/L, were studied. VIM-encoding genes were amplified and sequenced within their integrons. Isolates were typed by multilocus sequence typing (MLST). Six VIM-producers, representative of different integron structures and sequence types (STs), were completely sequenced using Illumina platform. Sixty-one P. aeruginosa were confirmed to produce VIM-type carbapenemases. ST111 dominated (n = 34) among VIM-producers, while 15 VIM-producers belonged to ST235. The blaVIM-like genes were located in three integron types, including In59, In595 and In1760, which were integrated into P. aeruginosa chromosomes. Whole genome sequencing (WGS) data demonstrated that ST111 and ST235 MBL producers carried several resistance and virulence genes. Additionally, the presence of type I-C and type I-E clustered regularly interspaced short palindromic repeats (CRISPR)/Cas locus was observed in ST235 and ST395 isolates, respectively. In conclusion, our findings confirmed the clonal spread of ST111 P. aeruginosa, carrying the VIM-2-encoding integron In59, in the University Hospital of Larissa (UHL). In addition, they highlighted the important role of high-risk clones, ST111 and ST235, in the successful dissemination and establishment into hospital settings of clinically important pathogens carrying resistance determinants.
Collapse
Affiliation(s)
- Costas C. Papagiannitsis
- Department of Microbiology, University Hospital of Larissa, 41110 Larissa, Greece; (A.V.); (V.G.); (S.X.); (E.P.)
| | - Aggeliki Verra
- Department of Microbiology, University Hospital of Larissa, 41110 Larissa, Greece; (A.V.); (V.G.); (S.X.); (E.P.)
| | - Vasiliki Galani
- Department of Microbiology, University Hospital of Larissa, 41110 Larissa, Greece; (A.V.); (V.G.); (S.X.); (E.P.)
| | - Stelios Xitsas
- Department of Microbiology, University Hospital of Larissa, 41110 Larissa, Greece; (A.V.); (V.G.); (S.X.); (E.P.)
| | - Ibrahim Bitar
- Biomedical Center, Faculty of Medicine, Charles University, 32300 Pilsen, Czech Republic; (I.B.); (J.H.)
| | - Jaroslav Hrabak
- Biomedical Center, Faculty of Medicine, Charles University, 32300 Pilsen, Czech Republic; (I.B.); (J.H.)
| | - Efthymia Petinaki
- Department of Microbiology, University Hospital of Larissa, 41110 Larissa, Greece; (A.V.); (V.G.); (S.X.); (E.P.)
| |
Collapse
|
38
|
Armentrout EI, Kundracik EC, Rietsch A. Cell-type-specific hypertranslocation of effectors by the Pseudomonas aeruginosa type III secretion system. Mol Microbiol 2020; 115:305-319. [PMID: 33012037 DOI: 10.1111/mmi.14617] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 09/22/2020] [Indexed: 12/23/2022]
Abstract
Many Gram-negative pathogens use a type III secretion system (T3SS) to promote disease by injecting effector proteins into host cells. Common to many T3SSs is that injection of effector proteins is feedback inhibited. The mechanism of feedback inhibition and its role in pathogenesis are unclear. In the case of P. aeruginosa, the effector protein ExoS is central to limiting effector injection. ExoS is bifunctional, with an amino-terminal RhoGAP and a carboxy-terminal ADP-ribosyltransferase domain. We demonstrate that both domains are required to fully feedback inhibit effector injection. The RhoGAP-, but not the ADP-ribosyltransferase domain of the related effector protein ExoT also participates. Feedback inhibition does not involve translocator insertion nor pore-formation. Instead, feedback inhibition is due, in part, to a loss of the activating trigger for effector injection, and likely also decreased translocon stability. Surprisingly, feedback inhibition is abrogated in phagocytic cells. The lack of feedback inhibition in these cells requires phagocytic uptake of the bacteria, but cannot be explained through acidification of the phagosome or calcium limitation. Given that phagocytes are crucial for controlling P. aeruginosa infections, our data suggest that feedback inhibition allows P. aeruginosa to direct its effector arsenal against the cell types most damaging to its survival.
Collapse
Affiliation(s)
- Erin I Armentrout
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, OH, USA
| | - Emma C Kundracik
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, OH, USA
| | - Arne Rietsch
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
39
|
Wagener BM, Anjum N, Evans C, Brandon A, Honavar J, Creighton J, Traber MG, Stuart RL, Stevens T, Pittet JF. α-Tocopherol Attenuates the Severity of Pseudomonas aeruginosa-induced Pneumonia. Am J Respir Cell Mol Biol 2020; 63:234-243. [PMID: 32243761 DOI: 10.1165/rcmb.2019-0185oc] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Pseudomonas aeruginosa is a lethal pathogen that causes high mortality and morbidity in immunocompromised and critically ill patients. The type III secretion system (T3SS) of P. aeruginosa mediates many of the adverse effects of infection with this pathogen, including increased lung permeability in a Toll-like receptor 4/RhoA/PAI-1 (plasminogen activator inhibitor-1)-dependent manner. α-Tocopherol has antiinflammatory properties that may make it a useful adjunct in treatment of this moribund infection. We measured transendothelial and transepithelial resistance, RhoA and PAI-1 activation, stress fiber formation, P. aeruginosa T3SS exoenzyme (ExoY) intoxication into host cells, and survival in a murine model of pneumonia in the presence of P. aeruginosa and pretreatment with α-tocopherol. We found that α-tocopherol alleviated P. aeruginosa-mediated alveolar endothelial and epithelial paracellular permeability by inhibiting RhoA, in part, via PAI-1 activation, and increased survival in a mouse model of P. aeruginosa pneumonia. Furthermore, we found that α-tocopherol decreased the activation of RhoA and PAI-1 by blocking the injection of T3SS exoenzymes into alveolar epithelial cells. P. aeruginosa is becoming increasingly antibiotic resistant. We provide evidence that α-tocopherol could be a useful therapeutic agent for individuals who are susceptible to infection with P. aeruginosa, such as those who are immunocompromised or critically ill.
Collapse
Affiliation(s)
- Brant M Wagener
- Department of Anesthesiology and Perioperative Medicine.,Center for Free Radical Biology, and
| | - Naseem Anjum
- Department of Anesthesiology and Perioperative Medicine
| | - Cilina Evans
- Department of Anesthesiology and Perioperative Medicine
| | | | | | | | - Maret G Traber
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon
| | | | - Troy Stevens
- Department of Pharmacology and Medicine and the Center for Lung Biology, University of South Alabama, Mobile, Alabama
| | - Jean-Francois Pittet
- Department of Anesthesiology and Perioperative Medicine.,Center for Lung Injury and Repair, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
40
|
Carruthers NJ, McClellan SA, Somayajulu M, Pitchaikannu A, Bessert D, Peng X, Huitsing K, Stemmer PM, Hazlett LD. Effects of Glycyrrhizin on Multi-Drug Resistant Pseudomonas aeruginosa. Pathogens 2020; 9:pathogens9090766. [PMID: 32962036 PMCID: PMC7557769 DOI: 10.3390/pathogens9090766] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 09/16/2020] [Indexed: 02/06/2023] Open
Abstract
The effects of glycyrrhizin (GLY) on multi-drug resistant (MDR) systemic (MDR9) vs. ocular (B1045) Pseudomonas aeruginosa clinical isolates were determined. Proteomes of each isolate with/without GLY treatment were profiled using liquid chromatography mass spectrometry (LC-MS/MS). The effect of GLY on adherence of MDR isolates to immortalized human (HCET) and mouse (MCEC) corneal epithelial cells, and biofilm and dispersal was tested. Both isolates were treated with GLY (0.25 minimum inhibitory concentration (MIC), 10 mg/mL for MDR9 and 3.75 mg/mL for B1045) and subjected to proteomic analysis. MDR9 had a greater response to GLY (51% of identified proteins affected vs. <1% in B1045). In MDR9 vs. controls, GLY decreased the abundance of proteins for: antibiotic resistance, biofilm formation, and type III secretion. Further, antibiotic resistance and type III secretion proteins had higher control abundances in MDR9 vs. B1045. GLY (5 and 10 mg/mL) significantly reduced binding of both isolates to MCEC, and B1045 to HCET. MDR9 binding to HCET was only reduced at 10 mg/mL GLY. GLY (5 and 10 mg/mL) enhanced dispersal for both isolates, at early (6.5 h) but not later times (24–72 h). This study provides evidence that GLY has a greater effect on the proteome of MDR9 vs. B1045, yet it was equally effective at disrupting adherence and early biofilm dispersal.
Collapse
Affiliation(s)
- Nicholas J. Carruthers
- Institute of Environmental Health Sciences, Wayne State University School of Medicine, 540 E. Canfield Avenue, Detroit, MI 48201, USA; (N.J.C.); (P.M.S.)
| | - Sharon A. McClellan
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, School of Medicine, Detroit, MI 48201, USA; (S.A.M.); (M.S.); (A.P.); (D.B.); (K.H.)
| | - Mallika Somayajulu
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, School of Medicine, Detroit, MI 48201, USA; (S.A.M.); (M.S.); (A.P.); (D.B.); (K.H.)
| | - Ahalya Pitchaikannu
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, School of Medicine, Detroit, MI 48201, USA; (S.A.M.); (M.S.); (A.P.); (D.B.); (K.H.)
| | - Denise Bessert
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, School of Medicine, Detroit, MI 48201, USA; (S.A.M.); (M.S.); (A.P.); (D.B.); (K.H.)
| | - Xudong Peng
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao 266071, China;
| | - Kylie Huitsing
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, School of Medicine, Detroit, MI 48201, USA; (S.A.M.); (M.S.); (A.P.); (D.B.); (K.H.)
| | - Paul M. Stemmer
- Institute of Environmental Health Sciences, Wayne State University School of Medicine, 540 E. Canfield Avenue, Detroit, MI 48201, USA; (N.J.C.); (P.M.S.)
| | - Linda D. Hazlett
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, School of Medicine, Detroit, MI 48201, USA; (S.A.M.); (M.S.); (A.P.); (D.B.); (K.H.)
- Correspondence: ; Tel.: +1-313-577-1079; Fax: +1-313-577-3125
| |
Collapse
|
41
|
Moir DT, Bowlin NO, Berube BJ, Yabut J, Mills DM, Nguyen GT, Aron ZD, Williams JD, Mecsas J, Hauser AR, Bowlin TL. A Structure-Function-Inhibition Analysis of the Pseudomonas aeruginosa Type III Secretion Needle Protein PscF. J Bacteriol 2020; 202:e00055-20. [PMID: 32601072 PMCID: PMC7925083 DOI: 10.1128/jb.00055-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 06/19/2020] [Indexed: 01/10/2023] Open
Abstract
The Pseudomonas aeruginosa type III secretion system (T3SS) needle comprised of multiple PscF subunits is essential for the translocation of effector toxins into human cells, facilitating the establishment and dissemination of infection. Mutations in the pscF gene provide resistance to the phenoxyacetamide (PhA) series of T3SS inhibitory chemical probes. To better understand PscF functions and interactions with PhA, alleles of pscF with 71 single mutations altering 49 of the 85 residues of the encoded protein were evaluated for their effects on T3SS phenotypes. Of these, 37% eliminated and 63% maintained secretion, with representatives of both evenly distributed across the entire protein. Mutations in 14 codons conferred a degree of PhA resistance without eliminating secretion, and all but one were in the alpha-helical C-terminal 25% of PscF. PhA-resistant mutants exhibited no cross-resistance to two T3SS inhibitors with different chemical scaffolds. Two mutations caused constitutive T3SS secretion. The pscF allele at its native locus, whether wild type (WT), constitutive, or PhA resistant, was dominant over other pscF alleles expressed from nonnative loci and promoters, but mixed phenotypes were observed in chromosomal ΔpscF strains with both WT and mutant alleles at nonnative loci. Some PhA-resistant mutants exhibited reduced translocation efficiency that was improved in a PhA dose-dependent manner, suggesting that PhA can bind to those resistant needles. In summary, these results are consistent with a direct interaction between PhA inhibitors and the T3SS needle, suggest a mechanism of blocking conformational changes, and demonstrate that PscF affects T3SS regulation, as well as carrying out secretion and translocation.IMPORTANCEP. aeruginosa effector toxin translocation into host innate immune cells is critical for the establishment and dissemination of P. aeruginosa infections. The medical need for new anti-P. aeruginosa agents is evident by the fact that P. aeruginosa ventilator-associated pneumonia is associated with a high mortality rate (40 to 69%) and recurs in >30% of patients, even with standard-of-care antibiotic therapy. The results described here confirm roles for the PscF needle in T3SS secretion and translocation and suggest that it affects regulation, possibly by interaction with T3SS regulatory proteins. The results also support a model of direct interaction of the needle with PhA and suggest that, with further development, members of the PhA series may prove useful as drugs for P. aeruginosa infection.
Collapse
Affiliation(s)
| | | | - Bryan J Berube
- Department of Microbiology and Immunology, Northwestern University, Chicago, Illinois, USA
- Department of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Jaden Yabut
- Microbiotix, Inc., Worcester, Massachusetts, USA
| | | | - Giang T Nguyen
- Tufts Graduate School in Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, USA
| | | | | | - Joan Mecsas
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Alan R Hauser
- Department of Microbiology and Immunology, Northwestern University, Chicago, Illinois, USA
| | | |
Collapse
|
42
|
Mirzaei R, Mohammadzadeh R, Sholeh M, Karampoor S, Abdi M, Dogan E, Moghadam MS, Kazemi S, Jalalifar S, Dalir A, Yousefimashouf R, Mirzaei E, Khodavirdipour A, Alikhani MY. The importance of intracellular bacterial biofilm in infectious diseases. Microb Pathog 2020; 147:104393. [PMID: 32711113 DOI: 10.1016/j.micpath.2020.104393] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 07/07/2020] [Accepted: 07/08/2020] [Indexed: 12/17/2022]
Abstract
Various bacterial species, previously known as extracellular pathogens, can reside inside different host cells by adapting to intracellular modes by forming microbial aggregates with similar characteristics to bacterial biofilms. Additionally, bacterial invasion of human cells leads to failure in antibiotic therapy, as most conventional anti-bacterial agents cannot reach intracellular biofilm in normal concentrations. Various studies have shown that bacteria such as uropathogenic Escherichia coli, Pseudomonas aeruginosa, Borrelia burgdorferi,Moraxella catarrhalis, non-typeable Haemophilus influenzae, Streptococcus pneumonia, and group A Streptococci produce biofilm-like structures within the host cells. For the first time in this review, we will describe and discuss the new information about intracellular bacterial biofilm formation and its importance in bacterial infectious diseases.
Collapse
Affiliation(s)
- Rasoul Mirzaei
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rokhsareh Mohammadzadeh
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Sholeh
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sajad Karampoor
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Milad Abdi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Student Research Committee, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Eyup Dogan
- Department of Basic Biotechnology, Biotechnology Institute, Ankara, Turkey
| | - Mohammad Shokri Moghadam
- Department of Microbiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Sima Kazemi
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Saba Jalalifar
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amine Dalir
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Rasoul Yousefimashouf
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ebrahim Mirzaei
- Department of Medical Genetics and Molecular Biology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amir Khodavirdipour
- Division of Humann Genetics, Department of Anatomy, St. John's Hospital, Bangalore, India
| | - Mohammad Yousef Alikhani
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
43
|
Warncke P, Fink S, Wiegand C, Hipler UC, Fischer D. A shell-less hen's egg test as infection model to determine the biocompatibility and antimicrobial efficacy of drugs and drug formulations against Pseudomonas aeruginosa. Int J Pharm 2020; 585:119557. [PMID: 32565284 DOI: 10.1016/j.ijpharm.2020.119557] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 06/12/2020] [Accepted: 06/14/2020] [Indexed: 12/20/2022]
Abstract
A shell-less hen's egg based infection test with Pseudomonas aeruginosa was established to investigate the antimicrobial efficacy of drugs and drug formulations close to the in vivo situation. The test system using preincubated fertilized chicken eggs transferred in petri dishes was optimized with respect to the controlled local application of liquid materials and bacteria as well as the bacterial cultivation conditions. The applicability of the ex ovo infection model was confirmed with antimicrobial susceptibility tests using tobramycin, ciprofloxacin and meropenem. The validity of the ex ovo data was demonstrated by correlation with in vitro data of the CellTiter®-Blue and the microplate laser nephelometry assay. Real-time imaging of the progress of infection and the efficacy of the treatment could be realized by the MolecuLight i:X™ technique. Furthermore, in a proof-of-concept efficacy, biocompatibility and even the presence of irritants were determined side-by-side using commercial ophthalmics. In conclusion, this egg based infection model could bridge the gap between in vitro and in vivo models for the evaluation of antimicrobial susceptibility to reduce animal tests according to the 3R concept.
Collapse
Affiliation(s)
- Paul Warncke
- Pharmaceutical Technology and Biopharmacy, Friedrich-Schiller-University Jena, Lessingstraße 8, 07743 Jena, Germany
| | - Sarah Fink
- Department of Dermatology, University Medical Center Jena, Erfurter Str 35, 07740 Jena, Germany
| | - Cornelia Wiegand
- Department of Dermatology, University Medical Center Jena, Erfurter Str 35, 07740 Jena, Germany
| | - Uta-Christina Hipler
- Department of Dermatology, University Medical Center Jena, Erfurter Str 35, 07740 Jena, Germany
| | - Dagmar Fischer
- Pharmaceutical Technology and Biopharmacy, Friedrich-Schiller-University Jena, Lessingstraße 8, 07743 Jena, Germany; Jena Center for Soft Matter (JCSM), Friedrich-Schiller-University Jena, Humboldtstraße 10, 07743 Jena, Germany.
| |
Collapse
|
44
|
Lu Z, Meng S, Chang W, Fan S, Xie J, Guo F, Yang Y, Qiu H, Liu L. Mesenchymal stem cells activate Notch signaling to induce regulatory dendritic cells in LPS-induced acute lung injury. J Transl Med 2020; 18:241. [PMID: 32546185 PMCID: PMC7298963 DOI: 10.1186/s12967-020-02410-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 06/06/2020] [Indexed: 12/12/2022] Open
Abstract
Background Mesenchymal stem cells (MSCs) have been shown to alleviate acute lung injury (ALI) and induce the production of regulatory dendritic cells (DCregs), but the potential link between these two cell types remains unclear. The goal of this study was to investigate the effect and mechanism of MSC-induced regulatory dendritic cells in ALI mice. Material/methods In vivo experiments, C57BL/6 wild-type male mice were sacrificed at different times after intratracheal injection of LPS to observe changes in lung DC maturation and pathological damage. MSCs, DCregs or/and carboxyfluorescein diacetate succinimidyl ester (CFSE)-labeled DCs were administered to the mice by tail vein, and flow cytometry was performed to measure the phenotype of lung DCs and T cells. Lung injury was estimated by the lung wet weight/body weight ratio and histopathological analysis. In vitro, Western blotting or flow cytometry was used to detect the expression of Notch ligand or receptor in MSCs or DCs after coculture or LPS stimulation. Finally, in vivo and in vitro, we used the Notch signaling inhibitor DAPT to verify the effect of the Notch pathway on MSC-induced DCregs and their pulmonary protection. Results We showed significant accumulation and maturation of lung DCs 2 h after intratracheal injection of LPS, which were positively correlated with the lung pathological injury score. MSC treatment alleviated ALI lung injury, accompanied by a decrease in the number and maturity of classical DCs in the lungs. CFSE-labeled DCs migrated to the lungs of ALI mice more than those of the normal group, and the elimination of CFSE-labeled DCs in the blood was slower. MSCs inhibited the migration of CFSE-labeled DCs to the lung and promoted their elimination in the blood. DCregs, which are obtained by contact coculture of mDCs with MSCs, expressed reduced levels of MHCII, CD86, CD40 and increased levels of PD-L1, and had a reduced ability to stimulate lymphocyte proliferation and activation (expression of CD44 and CD69). mDCs expressing Notch2 significantly increased after coculture with MSCs or rhJagged1, and MSCs expressed more Jagged1 after LPS stimulation. After stimulation of mDCs with recombinant Jagged1, DCs with low expression of MHCII, CD86 and CD40 were also induced, and the effects of both rhJagged1 and MSCs on DCs were blocked by the Notch inhibitor DAPT. Intra-airway DAPT reversed the inhibitory effect of mesenchymal stem cells on DC recruitment to the lungs and its maturation. Conclusions Our results suggested that the recruitment and maturation of lung DCs is an important process in early ALI, MSCs attenuate LPS-induced ALI by inducing the production of DCregs by activating Notch signaling.
Collapse
Affiliation(s)
- Zhonghua Lu
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjia Bridge, Hunan Road, Gu Lou District, Nanjing, 210009, China
| | - Shanshan Meng
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjia Bridge, Hunan Road, Gu Lou District, Nanjing, 210009, China
| | - Wei Chang
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjia Bridge, Hunan Road, Gu Lou District, Nanjing, 210009, China
| | - Shanwen Fan
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjia Bridge, Hunan Road, Gu Lou District, Nanjing, 210009, China
| | - Jianfeng Xie
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjia Bridge, Hunan Road, Gu Lou District, Nanjing, 210009, China
| | - Fengmei Guo
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjia Bridge, Hunan Road, Gu Lou District, Nanjing, 210009, China
| | - Yi Yang
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjia Bridge, Hunan Road, Gu Lou District, Nanjing, 210009, China
| | - Haibo Qiu
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjia Bridge, Hunan Road, Gu Lou District, Nanjing, 210009, China
| | - Ling Liu
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjia Bridge, Hunan Road, Gu Lou District, Nanjing, 210009, China.
| |
Collapse
|
45
|
Exoenzyme Y Contributes to End-Organ Dysfunction Caused by Pseudomonas aeruginosa Pneumonia in Critically Ill Patients: An Exploratory Study. Toxins (Basel) 2020; 12:toxins12060369. [PMID: 32512716 PMCID: PMC7354586 DOI: 10.3390/toxins12060369] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 05/31/2020] [Accepted: 06/01/2020] [Indexed: 12/19/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen that causes pneumonia in immunocompromised and intensive care unit (ICU) patients. During host infection, P. aeruginosa upregulates the type III secretion system (T3SS), which is used to intoxicate host cells with exoenzyme (Exo) virulence factors. Of the four known Exo virulence factors (U, S, T and Y), ExoU has been shown in prior studies to associate with high mortality rates. Preclinical studies have shown that ExoY is an important edema factor in lung infection caused by P. aeruginosa, although its importance in clinical isolates of P. aeruginosa is unknown. We hypothesized that expression of ExoY would be highly prevalent in clinical isolates and would significantly contribute to patient morbidity secondary to P. aeruginosa pneumonia. A single-center, prospective observational study was conducted at the University of Alabama at Birmingham Hospital. Mechanically ventilated ICU patients with a bronchoalveolar lavage fluid culture positive for P. aeruginosa were included. Enrolled patients were followed from ICU admission to discharge and clinical P. aeruginosa isolates were genotyped for the presence of exoenzyme genes. Ninety-nine patients were enrolled in the study. ExoY was present in 93% of P. aeruginosa clinical isolates. Moreover, ExoY alone (ExoY+/ExoU−) was present in 75% of P. aeruginosa isolates, compared to 2% ExoU alone (ExoY−/ExoU+). We found that bacteria isolated from human samples expressed active ExoY and ExoU, and the presence of ExoY in clinical isolates was associated with end-organ dysfunction. This is the first study we are aware of that demonstrates that ExoY is important in clinical outcomes secondary to nosocomial pneumonia.
Collapse
|
46
|
Karna SLR, Nguyen JQ, Evani SJ, Qian LW, Chen P, Abercrombie JJ, Sebastian EA, Fourcaudot AB, Leung KP. T3SS and alginate biosynthesis of Pseudomonas aeruginosa impair healing of infected rabbit wounds. Microb Pathog 2020; 147:104254. [PMID: 32416139 DOI: 10.1016/j.micpath.2020.104254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 05/05/2020] [Accepted: 05/08/2020] [Indexed: 10/24/2022]
Abstract
Pseudomonas aeruginosa (a Gram-negative bacterium) is an opportunistic pathogen found in many infected wounds and is known to impair healing. To test the hypothesis that knocking out P. aeruginosa genes that are overexpressed during wound infection can cripple a pathogen's ability to impair healing, we assessed two pathways: the Type III secretion system (T3SS) and alginate biosynthesis. We generated single- and double-mutant strains of ExsA (T3SS activator), AlgD (GDP- mannose 6-dehydrogenase of alginate biosynthesis) and their complemented strains and evaluated their pathogenicity in a rabbit ear full-thickness excision-wound infection model. Wounds were inoculated with different strains (wild type, mutants, and complementary strains) at 106 CFU/wound on post-wounding day 3. After 24 h, 5 days and 9 days post-infection, wounds were harvested for measuring bacterial counts (viable and total) and wound healing (epithelial gap). On day 9 post-infection, the viable counts of the double mutant, (exsA/algD)‾ were 100-fold lower than the counts of the wild type (PAO1), single mutants, or the complement double-mutant, (exsA/algD)‾/+. Also, when compared to wounds infected with wild type or control strains, wounds infected with the double-knockout mutant was less inhibitory to wound healing (p < 0.05). Additionally, the double mutant showed greater susceptibility to macrophage phagocytosis in vitro than all other strains (p < 0.001). In conclusion, compared to single gene knockouts, double knockout of virulence genes in T3SS pathway and alginate biosynthesis pathway is more effective in reducing P. aeruginosa pathogenicity and its ability to impair wound healing. This study highlights the necessity of a dual-targeted anti-virulence strategy to improve healing outcomes of P. aeruginosa-infected wounds.
Collapse
Affiliation(s)
- S L Rajasekhar Karna
- Division of Combat Wound Repair, US Army Institute of Surgical Research, JBSA Fort Sam Houston, TX, USA
| | - Jesse Q Nguyen
- Division of Combat Wound Repair, US Army Institute of Surgical Research, JBSA Fort Sam Houston, TX, USA
| | - Shankar Jaikishan Evani
- Division of Combat Wound Repair, US Army Institute of Surgical Research, JBSA Fort Sam Houston, TX, USA
| | - Li-Wu Qian
- Division of Combat Wound Repair, US Army Institute of Surgical Research, JBSA Fort Sam Houston, TX, USA
| | - Ping Chen
- Division of Combat Wound Repair, US Army Institute of Surgical Research, JBSA Fort Sam Houston, TX, USA
| | - Johnathan J Abercrombie
- Division of Combat Wound Repair, US Army Institute of Surgical Research, JBSA Fort Sam Houston, TX, USA
| | - Eliza A Sebastian
- Division of Combat Wound Repair, US Army Institute of Surgical Research, JBSA Fort Sam Houston, TX, USA
| | - Andrea B Fourcaudot
- Division of Combat Wound Repair, US Army Institute of Surgical Research, JBSA Fort Sam Houston, TX, USA
| | - Kai P Leung
- Division of Combat Wound Repair, US Army Institute of Surgical Research, JBSA Fort Sam Houston, TX, USA.
| |
Collapse
|
47
|
Zhang L, Chen W, Jiang Q, Fei Z, Xiao M. Genome analysis of plant growth-promoting rhizobacterium Pseudomonas chlororaphis subsp. aurantiaca JD37 and insights from comparasion of genomics with three Pseudomonas strains. Microbiol Res 2020; 237:126483. [PMID: 32402945 DOI: 10.1016/j.micres.2020.126483] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 11/19/2022]
Abstract
Pseudomonas chlororaphis subsp. aurantiaca strain JD37 is a plant growth-promoting rhizobacterium (PGPR), which has important biotechnological features such as plant growth promotion, rhizosphere colonization and biocontrol activities. In present study, the genome sequence of JD37 was obtained and comparative genomic analysis were performed to explore unique features of the JD37 genome and its relationship with other Pseudomonas PGPR: P. chlororaphis PA23, P. protegens Pf-5 and P. aeruginosa M18. JD37 possessed a single circular chromosome of 6,702,062 bp in length with an average GC content of 62.75 %. No plasmid was detected in JD37. A total of 5003 functional proteins of JD37 were predicted according to the clusters of orthologous groups (COGs) database. The JD37 genome consisted of various genes involved in plant growth promotion, biocontrol activities and defense responses. Genes involved in the rhizosphere colonization and motility were also found in the genome of JD37, suggesting the common plant growth-promoting traits in PGPR. The identified resistance genes (e.g. those related to metal resistance, antibiotics, and osmotic and temperature-shock) and secondary metabolite biosynthesis revealed the pathways for metabolites it produced. Data presented in present study further provided valuable information on its molecular genetics and adaptive capacity in the rhizosphere niche.
Collapse
Affiliation(s)
- Lei Zhang
- Development Center of Plant Germplasm Resources, College of Life Sciences, Shanghai Normal University, Shanghai, 200234, China
| | - Wenbo Chen
- Boyce Thompson Institute, Cornell University, Ithaca, NY, 14853, USA
| | - Qiuyue Jiang
- Development Center of Plant Germplasm Resources, College of Life Sciences, Shanghai Normal University, Shanghai, 200234, China; Department of Microbiology and Microbial Engineering, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Zhangjun Fei
- Boyce Thompson Institute, Cornell University, Ithaca, NY, 14853, USA
| | - Ming Xiao
- Development Center of Plant Germplasm Resources, College of Life Sciences, Shanghai Normal University, Shanghai, 200234, China.
| |
Collapse
|
48
|
Riquelme SA, Wong Fok Lung T, Prince A. Pulmonary Pathogens Adapt to Immune Signaling Metabolites in the Airway. Front Immunol 2020; 11:385. [PMID: 32231665 PMCID: PMC7082326 DOI: 10.3389/fimmu.2020.00385] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 02/18/2020] [Indexed: 12/17/2022] Open
Abstract
A limited number of pulmonary pathogens are able to evade normal mucosal defenses to establish acute infection and then adapt to cause chronic pneumonias. Pathogens, such as Pseudomonas aeruginosa or Staphylococcus aureus, are typically associated with infection in patients with underlying pulmonary disease or damage, such as cystic fibrosis (CF) or chronic obstructive pulmonary disease (COPD). To establish infection, bacteria express a well-defined set of so-called virulence factors that facilitate colonization and activate an immune response, gene products that have been identified in murine models. Less well-understood are the adaptive changes that occur over time in vivo, enabling the organisms to evade innate and adaptive immune clearance mechanisms. These colonizers proliferate, generating a population sufficient to provide selection for mutants, such as small colony variants and mucoid variants, that are optimized for long term infection. Such host-adapted strains have evolved in response to selective pressure such as antibiotics and the recruitment of phagocytes at sites of infection and their release of signaling metabolites (e.g., succinate). These metabolites can potentially function as substrates for bacterial growth and but also generate oxidant stress. Whole genome sequencing and quantified expression of selected genes have helped to explain how P. aeruginosa and S. aureus adapt to the presence of these metabolites over the course of in vivo infection. The serial isolation of clonally related strains from patients with cystic fibrosis has provided the opportunity to identify bacterial metabolic pathways that are altered under this immune pressure, such as the anti-oxidant glyoxylate and pentose phosphate pathways, routes contributing to the generation of biofilms. These metabolic pathways and biofilm itself enable the organisms to dissipate oxidant stress, while providing protection from phagocytosis. Stimulation of host immune signaling metabolites by these pathogens drives bacterial adaptation and promotes their persistence in the airways. The inherent metabolic flexibility of P. aeruginosa and S. aureus is a major factor in their success as pulmonary pathogens.
Collapse
Affiliation(s)
- Sebastián A Riquelme
- Department of Pediatrics, Columbia University Medical Center, New York, NY, United States
| | - Tania Wong Fok Lung
- Department of Pediatrics, Columbia University Medical Center, New York, NY, United States
| | - Alice Prince
- Department of Pediatrics, Columbia University Medical Center, New York, NY, United States
| |
Collapse
|
49
|
Hassuna NA, Mandour SA, Mohamed ES. Virulence Constitution of Multi-Drug-Resistant Pseudomonas aeruginosa in Upper Egypt. Infect Drug Resist 2020; 13:587-595. [PMID: 32110069 PMCID: PMC7036984 DOI: 10.2147/idr.s233694] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 02/05/2020] [Indexed: 11/23/2022] Open
Abstract
Purpose Ventilator-associated pneumonia caused by Pseudomonas aeruginosa (P. aeruginosa) is a major health-care problem. In this study, we explored the epidemiology of virulence determinants among multi-drug-resistant (MDR) clinical P. aeruginosa isolates from hospitalized patients with ventilator-associated pneumonia in intensive care units in Upper Egypt. Patients and Methods MDR P. aeruginosa isolates were screened for the presence of eight virulence factors and typed by ERIC-PCR. Results A total of 39 clinical MDR isolates were selected out of 173 isolated P. aeruginosa showing a combination of adhesion and cytotoxicity virulence patterns, with the detection of aprA, exoU, exoS, lasB, algD, toxA in 74.3%, 58.9%, 46.1%, 41.2%, 30.7%, 20.5% of the isolates, respectively. The MDR isolates were grouped into 13 different virulence profiles according to the pattern of virulence gene distribution. exoU genotype was more predominant among the P. aeruginosa isolates with more than 48% of the isolates harboring this gene alone, 7% harboring both exoU and exoS and 43.5% harboring exoS gene. An intermediate degree of diversity was detected by ERIC-PCR typing where the isolates were clustered in 7 major groups, indicating possible cross-infection within the hospital. Conclusion Our results highlight the increased frequency of virulent P. aeruginosa isolates with a shift to the more virulent cytotoxic exoU genotype. Further hospital infection-control measures are mandatory to control the hospital cross-transmission of these highly virulent isolates. This study could vastly be a help to develop efficient treatment policies against P. aeruginosa induced ventilator-associated pneumonia.
Collapse
Affiliation(s)
- Noha A Hassuna
- Medical Microbiology and Immunology, Faculty of Medicine, Minia University, Minia, Egypt
| | - Sahar A Mandour
- Microbiology and Immunology Department, Faculty of Pharmacy, Deraya University, Minia, Egypt
| | - Ebtisam Samir Mohamed
- Medical Microbiology and Immunology, Faculty of Medicine, Minia University, Minia, Egypt
| |
Collapse
|
50
|
Sawa T, Momiyama K, Mihara T, Kainuma A, Kinoshita M, Moriyama K. Molecular epidemiology of clinically high-risk Pseudomonas aeruginosa strains: Practical overview. Microbiol Immunol 2020; 64:331-344. [PMID: 31965613 DOI: 10.1111/1348-0421.12776] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 01/07/2020] [Accepted: 01/20/2020] [Indexed: 12/13/2022]
Abstract
In recent years, numerous outbreaks of multidrug-resistant Pseudomonas aeruginosa have been reported across the world. Once an outbreak occurs, besides routinely testing isolates for susceptibility to antimicrobials, it is required to check their virulence genotypes and clonality profiles. Replacing pulsed-field gel electrophoresis DNA fingerprinting are faster, easier-to-use, and less expensive polymerase chain reaction (PCR)-based methods for characterizing hospital isolates. P. aeruginosa possesses a mosaic genome structure and a highly conserved core genome displaying low sequence diversity and a highly variable accessory genome that communicates with other Pseudomonas species via horizontal gene transfer. Multiple-locus variable-number tandem-repeat analysis and multilocus sequence typing methods allow for phylogenetic analysis of isolates by PCR amplification of target genes with the support of Internet-based services. The target genes located in the core genome regions usually contain low-frequency mutations, allowing the resulting phylogenetic trees to infer evolutionary processes. The multiplex PCR-based open reading frame typing (POT) method, integron PCR, and exoenzyme genotyping can determine a genotype by PCR amplifying a specific insertion gene in the accessory genome region using a single or a multiple primer set. Thus, analyzing P. aeruginosa isolates for their clonality, virulence factors, and resistance characteristics is achievable by combining the clonality evaluation of the core genome based on multiple-locus targeting methods with other methods that can identify specific virulence and antimicrobial genes. Software packages such as eBURST, R, and Dendroscope, which are powerful tools for phylogenetic analyses, enable researchers and clinicians to visualize clonality associations in clinical isolates.
Collapse
Affiliation(s)
- Teiji Sawa
- Department of Anesthesiology, School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kyoko Momiyama
- School of Pharmacy, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Toshihito Mihara
- Department of Anesthesiology, School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Atsushi Kainuma
- Department of Anesthesiology, School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Mao Kinoshita
- Department of Anesthesiology, School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kiyoshi Moriyama
- Department of Anesthesiology, School of Medicine, Kyorin University, Tokyo, Japan
| |
Collapse
|