1
|
Jedel E, Schator D, Kumar NG, Sullivan AB, Rietsch A, Evans DJ, Fleiszig SMJ. The Pseudomonas aeruginosa T3SS can contribute to traversal of an in situ epithelial multilayer independently of the T3SS needle. mBio 2025; 16:e0026625. [PMID: 40084853 PMCID: PMC11980567 DOI: 10.1128/mbio.00266-25] [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: 01/23/2025] [Accepted: 02/11/2025] [Indexed: 03/16/2025] Open
Abstract
Multilayered epithelia lining our tissue surfaces normally resist traversal by opportunistic bacteria. Previously, we developed a strategy to experimentally perturb this resistance in situ in the corneas of mouse eyes and used it to show that traversal of a multilayered epithelium by Pseudomonas aeruginosa requires ExsA, the transcriptional activator of its type 3 secretion system (T3SS). Here, we developed a novel strategy for quantitatively localizing individual traversing bacteria within the in situ multilayered corneal epithelium and explored the contributions of T3SS components. The results showed that T3SS translocon and T3SS effector mutants had reduced epithelial traversal efficiency. Surprisingly, a ΔpscC mutant unable to assemble the T3SS needle traversed as efficiently as wild-type P. aeruginosa, while a ΔexsD mutant "constitutively on" for T3SS expression was traversal defective. The dispensability of the T3SS needle for effector-mediated traversal was confirmed using a mutant lacking the T3SS operon except for the effector genes (ΔpscU-L mutant). That mutant reacquired the ability to traverse if complemented with rhamnose-inducible exsA, but not if the effector genes were also deleted (ΔpscU-LΔexoSTY). Western immunoblot confirmed ExoS in culture supernatants of rhamnose-induced exsA-complemented ΔpscU-L mutants lacking all T3SS needle protein genes. Together, these results show that epithelial traversal by P. aeruginosa can involve T3SS effectors and translocon proteins independently of the T3SS needle previously thought essential for T3SS function. This advances our understanding of P. aeruginosa pathogenesis and has relevance to the development of therapeutics targeting the T3SS system.IMPORTANCEWhile the capacity to cross an epithelial barrier can be a critical step in bacterial pathogenesis, our understanding of the mechanisms involved is derived largely from cell culture experimentation. The latter is due to the practical limitations of in vivo/in situ models and the challenge of visualizing individual bacteria in the context of host tissue. Here, factors used by P. aeruginosa to traverse an epithelial multilayer in situ were studied by (i) leveraging the transparent properties and superficial location of the cornea, (ii) using our established method for enabling bacterial traversal susceptibility, and (iii) developing a novel strategy for accurate and quantitative localization of individual traversing bacteria in situ. Outcomes showed that T3SS translocon and T3SS effector proteins synergistically contribute to epithelial traversal efficiency independently of the T3SS needle. These findings challenge the assumption that the T3SS needle is essential for T3SS effectors or translocon proteins to contribute to bacterial pathogenesis.
Collapse
Affiliation(s)
- Eric Jedel
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, California, USA
- Graduate Program in Infectious Diseases and Immunity, University of California, Berkeley, California, USA
| | - Daniel Schator
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, California, USA
| | - Naren G. Kumar
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, California, USA
| | - Aaron B. Sullivan
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, California, USA
- Department of Biology, San Francisco State University, San Francisco, CA, USA
| | - Arne Rietsch
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, Ohio, USA
| | - David J. Evans
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, California, USA
- College of Pharmacy, Touro University California, Vallejo, California, USA
| | - Suzanne M. J. Fleiszig
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, California, USA
- Graduate Program in Infectious Diseases and Immunity, University of California, Berkeley, California, USA
- Graduate Groups in Vision Science and Microbiology, University of California, Berkeley, California, USA
| |
Collapse
|
2
|
Kumar NG, Grosser MR, Wan S, Schator D, Ahn E, Jedel E, Nieto V, Evans DJ, Fleiszig SMJ. Contact Lens Wear Alters Transcriptional Responses to Pseudomonas aeruginosa in Both the Corneal Epithelium and the Bacteria. Invest Ophthalmol Vis Sci 2025; 66:31. [PMID: 39932472 PMCID: PMC11817980 DOI: 10.1167/iovs.66.2.31] [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: 12/05/2024] [Accepted: 01/07/2025] [Indexed: 02/14/2025] Open
Abstract
Purpose Healthy corneas resist colonization by virtually all microbes, yet contact lens wear can predispose the cornea to sight-threatening infection with Pseudomonas aeruginosa. Here, we explored how lens wear changes corneal epithelium transcriptional responses to P. aeruginosa and its impact on bacterial gene expression. Methods Male and female C57BL/6J mice were fitted with a contact lens on one eye for 24 hours. After lens removal, corneas were immediately challenged for 4 hours with P. aeruginosa. A separate group of naïve mice was similarly challenged with bacteria. Bacteria-challenged eyes were compared to uninoculated naïve controls, as was lens wear alone. Total RNA sequencing determined corneal epithelium and bacterial gene expression. Results Prior lens wear profoundly altered the corneal response to P. aeruginosa, including upregulated pattern recognition receptors (tlr3, nod1); downregulated lectin pathway of complement activation (masp1); amplified upregulation of tcf7, gpr55, ifi205, and wfdc2 (immune defense); and further suppression of efemp1 (corneal stromal integrity). Without lens wear, P. aeruginosa upregulated mitochondrial and ubiquinone metabolism genes. Lens wear alone upregulated axl, grn, tcf7, and gpr55 (immune defense) and downregulated Ca2+-dependent genes necab1, snx31, and npr3. P. aeruginosa exposure to prior lens wearing versus naïve corneas upregulated bacterial genes of virulence (popD), its regulation (rsmY, PA1226), and antimicrobial resistance (arnB, oprR). Conclusions Prior lens wear impacts corneal epithelium gene expression, altering its responses to P. aeruginosa and how P. aeruginosa responds to it favoring virulence, survival, and adaptation. Impacted genes and associated networks provide avenues for research to better understand infection pathogenesis.
Collapse
Affiliation(s)
- Naren Gajenthra Kumar
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, California, United States
| | - Melinda R Grosser
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, California, United States
| | - Stephanie Wan
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, California, United States
| | - Daniel Schator
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, California, United States
| | - Eugene Ahn
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, California, United States
| | - Eric Jedel
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, California, United States
- Graduate Program in Infectious Diseases and Immunity, University of California, Berkeley, California, United States
| | - Vincent Nieto
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, California, United States
| | - David J Evans
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, California, United States
- College of Pharmacy, Touro University California, Vallejo, California, United States
| | - Suzanne M J Fleiszig
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, California, United States
- Graduate Groups in Vision Science, Microbiology and Infectious Disease and Immunity, University of California, Berkeley, California, United States
| |
Collapse
|
3
|
Jedel E, Schator D, Kumar NG, Sullivan AB, Rietsch A, Evans DJ, Fleiszig SMJ. The Pseudomonas aeruginosa T3SS can contribute to traversal of an in situ epithelial multilayer independently of the T3SS needle. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.28.635263. [PMID: 39975055 PMCID: PMC11838347 DOI: 10.1101/2025.01.28.635263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Multilayered epithelia lining our tissue surfaces normally resist traversal by opportunistic bacteria. Previously, we developed a strategy to experimentally perturbate this resistance in situ in the corneas of mouse eyes and used it to show that traversal of a multilayered epithelium by Pseudomonas aeruginosa requires ExsA, the transcriptional activator of its type 3 secretion system (T3SS). Here, we developed a novel strategy for quantitively localizing individual traversing bacteria within the in situ multilayered corneal epithelium and explored contributions of T3SS components. The results showed that T3SS translocon and T3SS effector mutants had reduced epithelial traversal efficiency. Surprisingly, a ΔpscC mutant unable to assemble the T3SS needle traversed as efficiently as wild-type P. aeruginosa, while a ΔexsD mutant 'constitutively on' for T3SS expression was traversal defective. Dispensability of the T3SS needle for effector-mediated traversal was confirmed using a mutant lacking the T3SS operon except the effector genes (ΔpscU-L mutant). That mutant reacquired the ability to traverse if complemented with rhamnose-inducible exsA, but not if the effector genes were also deleted (ΔpscU-LΔexoSTY). Western immunoblot confirmed ExoS in culture supernatants of rhamnose-induced exsA-complemented ΔpscU-L mutants lacking all T3SS needle protein genes. Together, these results show that epithelial traversal by P. aeruginosa can involve T3SS effectors and translocon proteins independently of the T3SS needle previously thought essential for T3SS function. This advances our understanding of P. aeruginosa pathogenesis and has relevance to development of therapeutics targeting the T3SS system.
Collapse
Affiliation(s)
- Eric Jedel
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, CA USA
- Graduate Program in Infectious Diseases and Immunity, University of California, Berkeley, CA USA
| | - Daniel Schator
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, CA USA
| | - Naren G. Kumar
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, CA USA
| | - Aaron B. Sullivan
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, CA USA
| | - Arne Rietsch
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, OH USA
| | - David J. Evans
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, CA USA
- College of Pharmacy, Touro University California, Vallejo, CA USA
| | - Suzanne M. J. Fleiszig
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, CA USA
- Graduate Program in Infectious Diseases and Immunity, University of California, Berkeley, CA USA
- Graduate Groups in Vision Science and Microbiology, University of California, Berkeley, CA USA
| |
Collapse
|
4
|
El Husseini N, Carter JA, Lee VT. Urinary tract infections and catheter-associated urinary tract infections caused by Pseudomonas aeruginosa. Microbiol Mol Biol Rev 2024; 88:e0006622. [PMID: 39431861 DOI: 10.1128/mmbr.00066-22] [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: 10/22/2024] Open
Abstract
SUMMARYUrinary tract infection (UTI) is one of the most common infections in otherwise healthy individuals. UTI is also common in healthcare settings where patients often require urinary catheters to alleviate urinary retention. The placement of a urinary catheter often leads to catheter-associated urinary tract infection (CAUTI) caused by a broad range of opportunistic pathogens, commonly referred to as ESKAPE (Enterococcus, Staphylococcus, Klebsiella, Acinetobacter, Pseudomonas, and Enterobacter) pathogens. Our understanding of CAUTI is complicated by the differences in pathogens, in initial microbial load, changes that occur due to the duration of catheterization, and the relationship between infection (colonization) and disease symptoms. To advance our understanding of CAUTI, we reviewed UTI and CAUTI caused by Pseudomonas aeruginosa which is unique in that it is not commonly found associated with human microbiomes. For this reason, the ability of P. aeruginosa to cause UTI and CAUTI requires the introduction of the bacteria to the bladder from catheterization. Once in the host, the virulence factors used by P. aeruginosa in these infections remain an area of ongoing research. In this review, we will discuss studies that focus on P. aeruginosa UTI and CAUTI to better understand the infection dynamics and outcome in clinical settings, virulence factors associated with P. aeruginosa isolated from the urinary tract, and animal studies to test which bacterial factors are required for this infection. Understanding how P. aeruginosa can cause UTI and CAUTI can provide an understanding of how these infections initiate and progress and may provide possible strategies to limit these infections.
Collapse
Affiliation(s)
- Nour El Husseini
- Department of Cell Biology and Molecular Genetics, University of Maryland at College Park, College Park, Maryland, USA
| | - Jared A Carter
- Department of Cell Biology and Molecular Genetics, University of Maryland at College Park, College Park, Maryland, USA
| | - Vincent T Lee
- Department of Cell Biology and Molecular Genetics, University of Maryland at College Park, College Park, Maryland, USA
| |
Collapse
|
5
|
Kumar NG, Grosser MR, Wan S, Schator D, Ahn E, Jedel E, Nieto V, Evans DJ, Fleiszig SMJ. Contact Lens Wear Alters Transcriptional Responses to Pseudomonas aeruginosa in Both the Corneal Epithelium and the Bacteria. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.03.626720. [PMID: 39677621 PMCID: PMC11643048 DOI: 10.1101/2024.12.03.626720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Purpose Healthy corneas resist colonization by virtually all microbes yet contact lens wear can predispose the cornea to sight-threatening infection with Pseudomonas aeruginosa. Here, we explored how lens wear changes corneal epithelium transcriptional responses to P. aeruginosa and its impact on bacterial gene expression. Methods Male and female C57BL/6J mice were fitted with a contact lens on one eye for 24 h. After lens removal, corneas were immediately challenged for 4 h with P. aeruginosa. A separate group of naïve mice were similarly challenged with bacteria. Bacteria-challenged eyes were compared to uninoculated naive controls as was lens wear alone. Total RNA-sequencing determined corneal epithelium and bacterial gene expression. Results Prior lens wear profoundly altered the corneal response to P. aeruginosa, including: upregulated pattern-recognition receptors (tlr3, nod1), downregulated lectin pathway of complement activation (masp1), amplified upregulation of tcf7, gpr55, ifi205, wfdc2 (immune defense) and further suppression of efemp1 (corneal stromal integrity). Without lens wear, P. aeruginosa upregulated mitochondrial and ubiquinone metabolism genes. Lens wear alone upregulated axl, grn, tcf7, gpr55 (immune defense) and downregulated Ca2+-dependent genes necab1, snx31 and npr3. P. aeruginosa exposure to prior lens wearing vs. naïve corneas upregulated bacterial genes of virulence (popD), its regulation (rsmY, PA1226) and antimicrobial resistance (arnB, oprR). Conclusion Prior lens wear impacts corneal epithelium gene expression altering its responses to P. aeruginosa and how P. aeruginosa responds to it favoring virulence, survival and adaptation. Impacted genes and associated networks provide avenues for research to better understand infection pathogenesis.
Collapse
Affiliation(s)
- Naren G. Kumar
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, CA USA
| | - Melinda R Grosser
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, CA USA
| | - Stephanie Wan
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, CA USA
| | - Daniel Schator
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, CA USA
| | - Eugene Ahn
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, CA USA
| | - Eric Jedel
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, CA USA
- Graduate Program in Infectious Diseases and Immunity, University of California, Berkeley, CA USA
| | - Vincent Nieto
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, CA USA
| | - David J. Evans
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, CA USA
- College of Pharmacy, Touro University California, Vallejo, CA USA
| | - Suzanne M. J. Fleiszig
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, CA USA
- Graduate Groups in Vision Science and Microbiology, University of California, Berkeley, CA USA
| |
Collapse
|
6
|
Lê-Bury P, Echenique-Rivera H, Pizarro-Cerdá J, Dussurget O. Determinants of bacterial survival and proliferation in blood. FEMS Microbiol Rev 2024; 48:fuae013. [PMID: 38734892 PMCID: PMC11163986 DOI: 10.1093/femsre/fuae013] [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: 11/06/2023] [Revised: 04/29/2024] [Accepted: 05/10/2024] [Indexed: 05/13/2024] Open
Abstract
Bloodstream infection is a major public health concern associated with high mortality and high healthcare costs worldwide. Bacteremia can trigger fatal sepsis whose prevention, diagnosis, and management have been recognized as a global health priority by the World Health Organization. Additionally, infection control is increasingly threatened by antimicrobial resistance, which is the focus of global action plans in the framework of a One Health response. In-depth knowledge of the infection process is needed to develop efficient preventive and therapeutic measures. The pathogenesis of bloodstream infection is a dynamic process resulting from the invasion of the vascular system by bacteria, which finely regulate their metabolic pathways and virulence factors to overcome the blood immune defenses and proliferate. In this review, we highlight our current understanding of determinants of bacterial survival and proliferation in the bloodstream and discuss their interactions with the molecular and cellular components of blood.
Collapse
Affiliation(s)
- Pierre Lê-Bury
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Yersinia Research Unit, 28 rue du Dr Roux, 75015 Paris, France
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Autoimmune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), 18 route du Panorama, 92260 Fontenay-aux-Roses, France
| | - Hebert Echenique-Rivera
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Yersinia Research Unit, 28 rue du Dr Roux, 75015 Paris, France
| | - Javier Pizarro-Cerdá
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Yersinia Research Unit, 28 rue du Dr Roux, 75015 Paris, France
- Institut Pasteur, Université Paris Cité, Yersinia National Reference Laboratory, WHO Collaborating Research & Reference Centre for Plague FRA-146, 28 rue du Dr Roux, 75015 Paris, France
| | - Olivier Dussurget
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Yersinia Research Unit, 28 rue du Dr Roux, 75015 Paris, France
| |
Collapse
|
7
|
Zhang H, Yang J, Cheng J, Zeng J, Ma X, Lin J. PQS and pyochelin in Pseudomonas aeruginosa share inner membrane transporters to mediate iron uptake. Microbiol Spectr 2024; 12:e0325623. [PMID: 38171001 PMCID: PMC10846271 DOI: 10.1128/spectrum.03256-23] [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: 08/31/2023] [Accepted: 11/29/2023] [Indexed: 01/05/2024] Open
Abstract
Bacteria absorb different forms of iron through various channels to meet their needs. Our previous studies have shown that TseF, a type VI secretion system effector for Fe uptake, facilitates the delivery of outer membrane vesicle-associated Pseudomonas quinolone signal (PQS)-Fe3+ to bacterial cells by a process involving the Fe(III) pyochelin receptor FptA and the porin OprF. However, the form in which the PQS-Fe3+ complex enters the periplasm and how it is moved into the cytoplasm remain unclear. Here, we first demonstrate that the PQS-Fe3+ complex enters the cell directly through FptA or OprF. Next, we show that inner membrane transporters such as FptX, PchHI, and FepBCDG are not only necessary for Pseudomonas aeruginosa to absorb PQS-Fe3+ and pyochelin (PCH)-Fe3+ but are also necessary for the virulence of P. aeruginosa toward Galleria mellonella larvae. Furthermore, we suggest that the function of PQS-Fe3+ (but not PQS)-mediated quorum-sensing regulation is dependent on FptX, PchHI, and FepBCDG. Additionally, the findings indicate that unlike FptX, neither FepBCDG nor PchHI play roles in the autoregulatory loop involving PchR, but further deletion of fepBCDG and pchHI can reverse the inactive PchR phenotype caused by fptX deletion and reactivate the expression of the PCH pathway genes under iron-limited conditions. Finally, this work identifies the interaction between FptX, PchHI, and FepBCDG, indicating that a larger complex could be formed to mediate the uptake of PQS-Fe3+ and PCH-Fe3+. These results pave the way for a better understanding of the PQS and PCH iron absorption pathways and provide future directions for research on tackling P. aeruginosa infections.IMPORTANCEPseudomonas aeruginosa has evolved a number of strategies to acquire the iron it needs from its host, with the most common being the synthesis, secretion, and uptake of siderophores such as pyoverdine, pyochelin, and the quorum-sensing signaling molecule Pseudomonas quinolone signal (PQS). However, despite intensive studies of the siderophore uptake pathways of P. aeruginosa, our understanding of how siderophores transport iron across the inner membrane into the cytoplasm is still incomplete. Herein, we reveal that PQS and pyochelin in P. aeruginosa share inner membrane transporters such as FptX, PchHI, and FepBCDG to mediate iron uptake. Meanwhile, PQS and pyochelin-mediated signaling operate to a large extent via these inner membrane transporters. Our study revealed the existence of shared uptake pathways between PQS and pyochelin, which could lead us to reexamine the role of these two molecules in the iron uptake and virulence of P. aeruginosa.
Collapse
Affiliation(s)
- Heng Zhang
- Shaanxi Key Laboratory of Chinese Jujube, College of Life Sciences, Yan’an University, Yan’an, China
| | - Jianshe Yang
- Shaanxi Key Laboratory of Chinese Jujube, College of Life Sciences, Yan’an University, Yan’an, China
| | - Juanli Cheng
- Shaanxi Key Laboratory of Chinese Jujube, College of Life Sciences, Yan’an University, Yan’an, China
| | - Jing Zeng
- Shaanxi Key Laboratory of Chinese Jujube, College of Life Sciences, Yan’an University, Yan’an, China
| | - Xin Ma
- Shaanxi Key Laboratory of Chinese Jujube, College of Life Sciences, Yan’an University, Yan’an, China
| | - Jinshui Lin
- Shaanxi Key Laboratory of Chinese Jujube, College of Life Sciences, Yan’an University, Yan’an, China
| |
Collapse
|
8
|
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
|
9
|
Song K, Chen L, Suo N, Kong X, Li J, Wang T, Song L, Cheng M, Guo X, Huang Z, Huang Z, Yang Y, Tian X, Choo SW. Whole-transcriptome analysis reveals mechanisms underlying antibacterial activity and biofilm inhibition by a malic acid combination (MAC) in Pseudomonas aeruginosa. PeerJ 2023; 11:e16476. [PMID: 38084141 PMCID: PMC10710775 DOI: 10.7717/peerj.16476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 10/26/2023] [Indexed: 12/18/2023] Open
Abstract
Background Pseudomonas aeruginosa is a highly prevalent bacterial species known for its ability to cause various infections and its remarkable adaptability and biofilm-forming capabilities. In earlier work, we conducted research involving the screening of 33 metabolites obtained from a commercial source against two prevalent bacterial strains, Escherichia coli and Staphylococcus aureus. Through screening assays, we discovered a novel malic acid combination (MAC) consisting of malic acid, citric acid, glycine, and hippuric acid, which displayed significant inhibitory effects. However, the precise underlying mechanism and the potential impact of the MAC on bacterial biofilm formation remain unknown and warrant further investigation. Methods To determine the antibacterial effectiveness of the MAC against Pseudomonas aeruginosa, we conducted minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) assays. Transmission electron microscopy (TEM) and scanning electron microscopy (SEM) techniques were employed to observe bacterial morphology and biofilm formation. We further performed a biofilm inhibition assay to assess the effect of the MAC on biofilm formation. Whole-transcriptome sequencing and bioinformatics analysis were employed to elucidate the antibacterial mechanism of the MAC. Additionally, the expression levels of differentially expressed genes were validated using the real-time PCR approach. Results Our findings demonstrated the antibacterial activity of the MAC against P. aeruginosa. SEM analysis revealed that the MAC can induce morphological changes in bacterial cells. The biofilm assay showed that the MAC could reduce biofilm formation. Whole-transcriptome analysis revealed 1093 differentially expressed genes consisting of 659 upregulated genes and 434 downregulated genes, in response to the MAC treatment. Mechanistically, the MAC inhibited P. aeruginosa growth by targeting metabolic processes, secretion system, signal transduction, and cell membrane functions, thereby potentially compromising the survival of this human pathogen. This study provides valuable insights into the antibacterial and antibiofilm activities of the MAC, a synergistic and cost-effective malic acid combination, which holds promise as a potential therapeutic drug cocktail for treating human infectious diseases in the future.
Collapse
Affiliation(s)
- Kunping Song
- Wenzhou-Kean University, College of Science, Mathematics and Technology, Wenzhou, Zhejiang, China
| | - Li Chen
- Universiti Malaya, Institute of Biological Sciences, Faculty of Science, Kuala Lumpur, Kuala Lumpur, Malaysia
| | - Nanhua Suo
- Wenzhou-Kean University, College of Science, Mathematics and Technology, Wenzhou, Zhejiang, China
| | - Xinyi Kong
- Wenzhou-Kean University, College of Science, Mathematics and Technology, Wenzhou, Zhejiang, China
| | - Juexi Li
- Wenzhou-Kean University, College of Science, Mathematics and Technology, Wenzhou, Zhejiang, China
| | - Tianyu Wang
- Wenzhou-Kean University, College of Science, Mathematics and Technology, Wenzhou, Zhejiang, China
| | - Lanni Song
- Wenzhou-Kean University, Wenzhou Municipal Key Laboratory for Applied Biomedical and Biopharmaceutical Informatics, Wenzhou, Zhejiang, China
| | - Mengwei Cheng
- Wenzhou-Kean University, Wenzhou Municipal Key Laboratory for Applied Biomedical and Biopharmaceutical Informatics, Wenzhou, Zhejiang, China
| | - Xindian Guo
- Wenzhou No.2 Foreign Language School, Wenzhou, Zhejiang, China
| | - Zhenghe Huang
- Wenzhou No.2 Foreign Language School, Wenzhou, Zhejiang, China
| | - Zichen Huang
- Wenzhou No.2 Foreign Language School, Wenzhou, Zhejiang, China
| | - Yixin Yang
- Wenzhou-Kean University, College of Science, Mathematics and Technology, Wenzhou, Zhejiang, China
- Wenzhou-Kean University, Wenzhou Municipal Key Laboratory for Applied Biomedical and Biopharmaceutical Informatics, Wenzhou, Zhejiang, China
- Wenzhou-Kean University, Zhejiang Bioinformatics International Science and Technology Cooperation Center, Wenzhou, Zhejiang, China
| | - Xuechen Tian
- Wenzhou-Kean University, Wenzhou Municipal Key Laboratory for Applied Biomedical and Biopharmaceutical Informatics, Wenzhou, Zhejiang, China
- Wenzhou-Kean University, Zhejiang Bioinformatics International Science and Technology Cooperation Center, Wenzhou, Zhejiang, China
| | - Siew Woh Choo
- Wenzhou-Kean University, College of Science, Mathematics and Technology, Wenzhou, Zhejiang, China
- Wenzhou-Kean University, Wenzhou Municipal Key Laboratory for Applied Biomedical and Biopharmaceutical Informatics, Wenzhou, Zhejiang, China
- Wenzhou-Kean University, Zhejiang Bioinformatics International Science and Technology Cooperation Center, Wenzhou, Zhejiang, China
| |
Collapse
|
10
|
Kroken AR, Klein KA, Mitchell PS, Nieto V, Jedel EJ, Evans DJ, Fleiszig SMJ. Intracellular replication of Pseudomonas aeruginosa in epithelial cells requires suppression of the caspase-4 inflammasome. mSphere 2023; 8:e0035123. [PMID: 37589460 PMCID: PMC10597407 DOI: 10.1128/msphere.00351-23] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 06/30/2023] [Indexed: 08/18/2023] Open
Abstract
Pathogenesis of Pseudomonas aeruginosa infections can include bacterial survival inside epithelial cells. Previously, we showed that this involves multiple roles played by the type three secretion system (T3SS), and specifically the effector ExoS. This includes ExoS-dependent inhibition of a lytic host cell response that subsequently enables intracellular replication. Here, we studied the underlying cell death response to intracellular P. aeruginosa, comparing wild-type to T3SS mutants varying in capacity to induce cell death and that localize to different intracellular compartments. Results showed that corneal epithelial cell death induced by intracellular P. aeruginosa lacking the T3SS, which remains in vacuoles, correlated with the activation of nuclear factor-κB as measured by p65 relocalization and tumor necrosis factor alpha transcription and secretion. Deletion of caspase-4 through CRISPR-Cas9 mutagenesis delayed cell death caused by these intracellular T3SS mutants. Caspase-4 deletion also countered more rapid cell death caused by T3SS effector-null mutants still expressing the T3SS apparatus that traffic to the host cell cytoplasm, and in doing so rescued intracellular replication normally dependent on ExoS. While HeLa cells lacked a lytic death response to T3SS mutants, it was found to be enabled by interferon gamma treatment. Together, these results show that epithelial cells can activate the noncanonical inflammasome pathway to limit proliferation of intracellular P. aeruginosa, not fully dependent on bacterially driven vacuole escape. Since ExoS inhibits the lytic response, the data implicate targeting of caspase-4, an intracellular pattern recognition receptor, as another contributor to the role of ExoS in the intracellular lifestyle of P. aeruginosa. IMPORTANCE Pseudomonas aeruginosa can exhibit an intracellular lifestyle within epithelial cells in vivo and in vitro. The type three secretion system (T3SS) effector ExoS contributes via multiple mechanisms, including extending the life of invaded host cells. Here, we aimed to understand the underlying cell death inhibited by ExoS when P. aeruginosa is intracellular. Results showed that intracellular P. aeruginosa lacking T3SS effectors could elicit rapid cell lysis via the noncanonical inflammasome pathway. Caspase-4 contributed to cell lysis even when the intracellular bacteria lacked the entire T33S and were consequently unable to escape vacuoles, representing a naturally occurring subpopulation during wild-type infection. Together, the data show the caspase-4 inflammasome as an epithelial cell defense against intracellular P. aeruginosa, and implicate its targeting as another mechanism by which ExoS preserves the host cell replicative niche.
Collapse
Affiliation(s)
- Abby R. Kroken
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, Illinois, USA
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, California, USA
| | - Keith A. Klein
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, Illinois, USA
| | - Patrick S. Mitchell
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Vincent Nieto
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, California, USA
| | - Eric J. Jedel
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, California, USA
| | - David J. Evans
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, California, USA
- College of Pharmacy, Touro University California, Vallejo, California, USA
| | - Suzanne M. J. Fleiszig
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, California, USA
- Graduate Groups in Vision Sciences, Microbiology, and Infectious Diseases & Immunity, University of California, Berkeley, California, USA
| |
Collapse
|
11
|
Mekonnen SA, El Husseini N, Turdiev A, Carter JA, Belew AT, El-Sayed NM, Lee VT. Catheter-associated urinary tract infection by Pseudomonas aeruginosa progresses through acute and chronic phases of infection. Proc Natl Acad Sci U S A 2022; 119:e2209383119. [PMID: 36469780 PMCID: PMC9897465 DOI: 10.1073/pnas.2209383119] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 11/08/2022] [Indexed: 12/12/2022] Open
Abstract
Healthcare-associated infections are major causes of complications that lead to extended hospital stays and significant medical costs. The use of medical devices, including catheters, increases the risk of bacterial colonization and infection through the presence of a foreign surface. Two outcomes are observed for catheterized patients: catheter-associated asymptomatic bacteriuria and catheter-associated urinary tract infection (CAUTI). However, the relationship between these two events remains unclear. To understand this relationship, we studied a murine model of Pseudomonas aeruginosa CAUTI. In this model, we also observe two outcomes in infected animals: acute symptoms that is associated with CAUTI and chronic colonization that is associated with asymptomatic bacteriuria. The timing of the acute outcome takes place in the first week of infection, whereas chronic colonization occurs in the second week of infection. We further showed that mutants lacking genes encoding type III secretion system (T3SS), T3SS effector proteins, T3SS injection pore, or T3SS transcriptional activation all fail to cause acute symptoms of CAUTI. Nonetheless, all mutants defective for T3SS colonized the catheter and bladders at levels similar to the parental strain. In contrast, through induction of the T3SS master regulator ExsA, all infected animals showed acute phenotypes with bacteremia. Our results demonstrated that the acute symptoms, which are analogous to CAUTI, and chronic colonization, which is analogous to asymptomatic bacteriuria, are independent events that require distinct bacterial virulence factors. Experimental delineation of asymptomatic bacteriuria and CAUTI informs different strategies for the treatment and intervention of device-associated infections.
Collapse
Affiliation(s)
- Solomon A. Mekonnen
- Department of Cell Biology and Molecular Genetics, University of Maryland at College Park, College Park, MD20742
| | - Nour El Husseini
- Department of Cell Biology and Molecular Genetics, University of Maryland at College Park, College Park, MD20742
| | - Asan Turdiev
- Department of Cell Biology and Molecular Genetics, University of Maryland at College Park, College Park, MD20742
| | - Jared A. Carter
- Department of Cell Biology and Molecular Genetics, University of Maryland at College Park, College Park, MD20742
| | - Ashton Trey Belew
- Department of Cell Biology and Molecular Genetics, University of Maryland at College Park, College Park, MD20742
| | - Najib M. El-Sayed
- Department of Cell Biology and Molecular Genetics, University of Maryland at College Park, College Park, MD20742
- Center for Bioinformatics and Computational Biology, University of Maryland at College Park, College Park, MD20742
| | - Vincent T. Lee
- Department of Cell Biology and Molecular Genetics, University of Maryland at College Park, College Park, MD20742
| |
Collapse
|
12
|
Rudra B, Duncan L, Shah AJ, Shah HN, Gupta RS. Phylogenomic and comparative genomic studies robustly demarcate two distinct clades of Pseudomonas aeruginosa strains: proposal to transfer the strains from an outlier clade to a novel species Pseudomonas paraeruginosa sp. nov. Int J Syst Evol Microbiol 2022; 72. [PMID: 36355412 DOI: 10.1099/ijsem.0.005542] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The strains of
Pseudomonas aeruginosa
exhibit considerable differences in their genotypic and pathogenic properties. To clarify their evolutionary/taxonomic relationships, comprehensive phylogenomic and comparative genomic studies were conducted on the genome sequences of 212
P
.
aeruginosa
strains covering their genetic diversity. In a phylogenomic tree based on 118 conserved proteins, the analysed strains formed two distinct clades. One of these clades, Clade-1, encompassing >70 % of the strains including the type strain DSM 50071T, represents the species P. aeruginosa sensu stricto. Clade-2, referred to in earlier work as the outlier group, with NCTC 13628T as its type strain, constitutes a novel species level lineage. The average nucleotide identity, average amino acid identity and digital DNA–DNA hybridization values between the strains from Clade-1 and Clade-2 are in the range of 93.4–93.7, 95.1–95.3 and 52–53 %, respectively. The 16S rRNA gene of
P. aeruginosa
DSM 50071T also shows 98.3 % similarity to that of NCTC 13628T. These values are lower than the suggested cut-off values for species distinction, indicating that the Clade-2 strains (NCTC 13628T) constitute a new species. We also report the identification of 12 conserved signature indels in different proteins and 24 conserved signature proteins that are exclusively found in either Clade-1 or Clade-2, providing a reliable means for distinguishing these clades. Additionally, in contrast to swimming motility, twitching motility is only present in Clade-1 strains. Based on earlier work, the strains from these two clades also differ in their pathogenic mechanisms (presence/absence of Type III secretion system), production of biosurfactants, phenazines and siderophores, and several other genomic characteristics. Based on the evidence from different studies, we propose that the Clade-2 strains constitute a novel species for which the name Pseudomonas paraeruginosa is proposed. The type strain is NCTC 13628T (=PA7T=ATCC 9027T). The description of
Pseudomonas aeruginosa
is also emended to include information for different molecular markers specific for this species.
Collapse
Affiliation(s)
- Bashudev Rudra
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton L8N 3Z5, Canada
| | - Louise Duncan
- School of Cancer and Pharmaceutical Sciences, King's College London, London SE1 9NH, UK
| | - Ajit J Shah
- Department of Natural Sciences, Middlesex University, London NW4 4BT, UK
| | - Haroun N Shah
- Department of Natural Sciences, Middlesex University, London NW4 4BT, UK
| | - Radhey S Gupta
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton L8N 3Z5, Canada
| |
Collapse
|
13
|
Sangeet S, Pawar S, Nawani N, Junnarkar M, Gaikwad S. Computational approach to attenuate virulence of Pseudomonas aeruginosa through bioinspired silver nanoparticles. 3 Biotech 2022; 12:317. [PMID: 36276439 PMCID: PMC9547761 DOI: 10.1007/s13205-022-03367-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 09/17/2022] [Indexed: 11/24/2022] Open
Abstract
In this study we aim to investigate the computational docking approach of biofabricated silver nanoparticles against P. aeruginosa virulent exoenzymes, such as ExoS and ExoY. Therefore, the synthesis and characterization of biofabricated silver nanoparticles using Piper betle leaves (Pb-AgNPs) were carried out. The surface topology and functional group attachment on the surface of Pb-AgNPs were analyzed using UV-visible spectroscopy, Scanning Electron Microscopy, Fourier Transformed Infrared Spectroscopy (FTIR), and X-Ray Diffraction. The FTIR analysis revealed that the synthesized silver nanoparticles were capped with P. betle phytochemicals importantly Eugenol and Hydroxychavicol. These are the major bioactive compounds present in P. betle leaves; therefore, computational docking of Eugenol-conjugated AgNPs (PbEu-AgNPs) and Hydroxychavicol-conjugated AgNPs (PbHy-AgNPs) against ExoS and ExoY was performed. The active residues of PbEu-AgNPs and PbHy-AgNPs interacted with the active site of ExoS and ExoY exoenzymes. Biofabricated AgNP-mediated inhibition of these virulent exoenzymes blocked the adverse effect of P. aeruginosa on the host cell. The computational analysis provides new approach into the design of biofabricated AgNPs as promising anti-infective nanomedicine agents. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-022-03367-0.
Collapse
Affiliation(s)
- Satyam Sangeet
- Microbial Diversity Research Center, Dr. D.Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune, Maharashtra India
- Present Address: Department of Chemical Sciences, Indian Institute of Science Education and Research (IISER), Kolkata, 741246 India
| | - Sarika Pawar
- Microbial Diversity Research Center, Dr. D.Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune, Maharashtra India
| | - Neelu Nawani
- Microbial Diversity Research Center, Dr. D.Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune, Maharashtra India
| | - Manisha Junnarkar
- Microbial Diversity Research Center, Dr. D.Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune, Maharashtra India
| | - Swapnil Gaikwad
- Microbial Diversity Research Center, Dr. D.Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune, Maharashtra India
| |
Collapse
|
14
|
Holban AM, Gregoire CM, Gestal MC. Conquering the host: Bordetella spp. and Pseudomonas aeruginosa molecular regulators in lung infection. Front Microbiol 2022; 13:983149. [PMID: 36225372 PMCID: PMC9549215 DOI: 10.3389/fmicb.2022.983149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/17/2022] [Indexed: 11/27/2022] Open
Abstract
When bacteria sense cues from the host environment, stress responses are activated. Two component systems, sigma factors, small RNAs, ppGpp stringent response, and chaperones start coordinate the expression of virulence factors or immunomodulators to allow bacteria to respond. Although, some of these are well studied, such as the two-component systems, the contribution of other regulators, such as sigma factors or ppGpp, is increasingly gaining attention. Pseudomonas aeruginosa is the gold standard pathogen for studying the molecular mechanisms to sense and respond to environmental cues. Bordetella spp., on the other hand, is a microbial model for studying host-pathogen interactions at the molecular level. These two pathogens have the ability to colonize the lungs of patients with chronic diseases, suggesting that they have the potential to share a niche and interact. However, the molecular networks that facilitate adaptation of Bordetella spp. to cues are unclear. Here, we offer a side-by-side comparison of what is known about these diverse molecular mechanisms that bacteria utilize to counteract host immune responses, while highlighting the relatively unexplored interactions between them.
Collapse
Affiliation(s)
- Alina M. Holban
- Research Institute of the University of Bucharest (ICUB), Bucharest, Romania
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Courtney M. Gregoire
- Department of Microbiology and Immunology, Louisiana State University Health Science Center, Shreveport, LA, United States
| | - Monica C. Gestal
- Department of Microbiology and Immunology, Louisiana State University Health Science Center, Shreveport, LA, United States
- *Correspondence: Monica C. Gestal, ;
| |
Collapse
|
15
|
Huber P. ExlA: A New Contributor to Pseudomonas aeruginosa Virulence. Front Cell Infect Microbiol 2022; 12:929150. [PMID: 35811671 PMCID: PMC9260685 DOI: 10.3389/fcimb.2022.929150] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 05/19/2022] [Indexed: 11/17/2022] Open
Abstract
ExlA (also called exolysin) is a recently discovered virulence factor secreted by a subset of Pseudomonas aeruginosa strains in which a type 3 secretion system is lacking. exlA-positive strains were identified worldwide in the clinic, causing several types of infectious diseases, and were detected in various locations in the environment. ExlA possesses pore-forming activity and is cytolytic for most human cell types. It belongs to a class of poorly characterized bacterial toxins, sharing a similar protein domain organization and a common secretion pathway. This review summarizes the recent findings regarding ExlA synthesis, its secretion pathway, and its toxic behavior for host cells.
Collapse
|
16
|
Veetilvalappil VV, Manuel A, Aranjani JM, Tawale R, Koteshwara A. Pathogenic arsenal of Pseudomonas aeruginosa: an update on virulence factors. Future Microbiol 2022; 17:465-481. [PMID: 35289684 DOI: 10.2217/fmb-2021-0158] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The emergence of Pseudomonas aeruginosa as a potential threat in persistent infections can be attributed to the plethora of virulence factors expressed by it. This review discusses the various virulence factors that help this pathogen to establish an infection and regulatory systems controlling these virulence factors. Cell-associated virulence factors such as flagella, type IV pili and non-pilus adhesins have been reviewed. Extracellular virulence factors have also been explained. Quorum-sensing systems present in P. aeruginosa play a cardinal role in regulating the expression of virulence factors. The identification of novel virulence factors in hypervirulent strains indicate that the expression of virulence is dynamic and constantly evolving. An understanding of this is critical for the better clinical management of infections.
Collapse
Affiliation(s)
- Vimal V Veetilvalappil
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Atulya Manuel
- Central Frozen Semen Production and Training Institute, Bengaluru, Karnataka, 560088, India
| | - Jesil M Aranjani
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Roshan Tawale
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Ananthamurthy Koteshwara
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| |
Collapse
|
17
|
Ahangarzadeh M, Ghorbanpour Najafabadi M, Peyghan R, Houshmand H, Sharif Rohani M, Soltani M. Detection and distribution of virulence genes in Aeromonas hydrophila isolates causing infection in cultured carps. VETERINARY RESEARCH FORUM : AN INTERNATIONAL QUARTERLY JOURNAL 2022; 13:55-60. [PMID: 35601789 PMCID: PMC9094588 DOI: 10.30466/vrf.2020.115998.2761] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 06/01/2020] [Indexed: 11/01/2022]
Abstract
Aeromonas hydrophila is a bacterium associated with many diseases and disorders such as fin rot, skin ulcers and lethal hemorrhagic septicemia in fish. It bears several virulence factors including type III secretion system (T3SS), aerolysin, cytolytic enterotoxin and enzymes (e.g., hemolysins, lipase) that seem to play an important role in its pathogenesis. Detection of virulence markers by polymerase chain reaction (PCR) is a key procedure in defining the patho-genic ability of pathogenic bacteria and preparing a vaccine for its treatment. In this sense, this study was aimed to determine the frequency of virulence genes in isolates obtained from infected cultured carps in Khuzestan province. Out of 200 moribund carps with septicemic symptoms, 125 isolates were belonged to the motile aeromonads and 59 isolates were identified as A. hydrophila by biochemical methods. Finally, using PCR analysis, 31 isolates were identified as A. hydrophila. Five virulence genes were detected in these isolates including hemolysin, aerolysin, cytolytic enterotoxin and T3SS (aopB and ascV) by specific primers. Results showed that 23 (74.19%), 18 (58.06%), 16 (51.61%), 13 (41.63%) and 10 (32.25%) isolates possessed cytolytic enterotoxin, hemolysin, aerolysin, and T3SS genes, respectively. The results of the present study showed that among 31 isolates, only five isolates had all of dominant virulence genes. Thirteen other isolates had genotypes including hlyA +, aerA + , and act + . The remaining isolates had at least one virulence gene. This study showed that determination of the virulence genes by PCR can be a reliable method to identify a potential pathogenic Aeromonad strain.
Collapse
Affiliation(s)
- Mina Ahangarzadeh
- South of Iran Aquaculture Research Institute, Iranian Fisheries Science Research Institute, Agricultural Research Education and Extension Organization (AREEO), Ahvaz, Iran; ,Correspondence Mina Ahangarzadeh. DVM, PhD, South of Iran Aquaculture Research Institute, Iranian Fisheries Science Research Institute, Agricultural Research Education and Extension Organization (AREEO), Ahvaz, Iran. E-mail:
| | | | - Rahim Peyghan
- Department of Aquatic Animal Health, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran;
| | - Hossein Houshmand
- South of Iran Aquaculture Research Institute, Iranian Fisheries Science Research Institute, Agricultural Research Education and Extension Organization (AREEO), Ahvaz, Iran;
| | - Mostafa Sharif Rohani
- Department of Fish diseases, Iranian Fisheries Research Science Institute (IFSRI), Agriculture Research, Education and Extension Organization (AREEO), Tehran, Iran;
| | - Mehdi Soltani
- Department of Aquatic Animal Health, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran; ,Centre for Sustainable Aquatic Ecosystems, Harry Butler Institute, Murdoch University, Perth, Australia.
| |
Collapse
|
18
|
Kroken AR, Gajenthra Kumar N, Yahr TL, Smith BE, Nieto V, Horneman H, Evans DJ, Fleiszig SMJ. Exotoxin S secreted by internalized Pseudomonas aeruginosa delays lytic host cell death. PLoS Pathog 2022; 18:e1010306. [PMID: 35130333 PMCID: PMC8853526 DOI: 10.1371/journal.ppat.1010306] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 02/17/2022] [Accepted: 01/25/2022] [Indexed: 12/22/2022] Open
Abstract
The Pseudomonas aeruginosa toxin ExoS, secreted by the type III secretion system (T3SS), supports intracellular persistence via its ADP-ribosyltransferase (ADPr) activity. For epithelial cells, this involves inhibiting vacuole acidification, promoting vacuolar escape, countering autophagy, and niche construction in the cytoplasm and within plasma membrane blebs. Paradoxically, ExoS and other P. aeruginosa T3SS effectors can also have antiphagocytic and cytotoxic activities. Here, we sought to reconcile these apparently contradictory activities of ExoS by studying the relationships between intracellular persistence and host epithelial cell death. Methods involved quantitative imaging and the use of antibiotics that vary in host cell membrane permeability to selectively kill intracellular and extracellular populations after invasion. Results showed that intracellular P. aeruginosa mutants lacking T3SS effector toxins could kill (permeabilize) cells when extracellular bacteria were eliminated. Surprisingly, wild-type strain PAO1 (encoding ExoS, ExoT and ExoY) caused cell death more slowly, the time extended from 5.2 to 9.5 h for corneal epithelial cells and from 10.2 to 13.0 h for HeLa cells. Use of specific mutants/complementation and controls for initial invasion showed that ExoS ADPr activity delayed cell death. Triggering T3SS expression only after bacteria invaded cells using rhamnose-induction in T3SS mutants rescued the ExoS-dependent intracellular phenotype, showing that injected effectors from extracellular bacteria were not required. The ADPr activity of ExoS was further found to support internalization by countering the antiphagocytic activity of both the ExoS and ExoT RhoGAP domains. Together, these results show two additional roles for ExoS ADPr activity in supporting the intracellular lifestyle of P. aeruginosa; suppression of host cell death to preserve a replicative niche and inhibition of T3SS effector antiphagocytic activities to allow invasion. These findings add to the growing body of evidence that ExoS-encoding (invasive) P. aeruginosa strains can be facultative intracellular pathogens, and that intracellularly secreted T3SS effectors contribute to pathogenesis. While the ADPr domain of the T3SS effector ExoS plays multiple roles in the intracellular lifestyle of P. aeruginosa, ExoS can also be cytotoxic and/or antiphagocytic. Here, we show that when P. aeruginosa enters the cytosol of epithelial cells, cell death is triggered independently of T3SS effector toxins, but ExoS ADPr activity delays this to enable continued intracellular survival and replication. Using rhamnose induction to express the T3SS only after invasion restored this ExoS-dependent phenotype, showing that intracellularly secreted effectors can enable intracellular pathogenesis. ExoS ADPr activity also countered antiphagocytic activity of ExoS and ExoT RhoGAP domains. These results show two additional roles for ExoS ADPr activity in promoting internalization of P. aeruginosa and protecting the intracellular niche, continuing to challenge the notions that P. aeruginosa is exclusively an extracellular pathogen, that it needs to inject T3SS effectors across plasma membranes, and that ExoS is necessarily cytotoxic to host cells.
Collapse
Affiliation(s)
- Abby R. Kroken
- School of Optometry, University of California, Berkeley, Berkeley, California, United States of America
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, Illinois, United States of America
| | - Naren Gajenthra Kumar
- School of Optometry, University of California, Berkeley, Berkeley, California, United States of America
| | - Timothy L. Yahr
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, United States of America
| | - Benjamin E. Smith
- Vision Science Program, University of California, Berkeley, Berkeley, California, United States of America
| | - Vincent Nieto
- School of Optometry, University of California, Berkeley, Berkeley, California, United States of America
| | - Hart Horneman
- School of Optometry, University of California, Berkeley, Berkeley, California, United States of America
| | - David J. Evans
- School of Optometry, University of California, Berkeley, Berkeley, California, United States of America
- College of Pharmacy, Touro University California, Vallejo, California, United States of America
| | - Suzanne M. J. Fleiszig
- School of Optometry, University of California, Berkeley, Berkeley, California, United States of America
- Vision Science Program, University of California, Berkeley, Berkeley, California, United States of America
- Graduate Groups in Microbiology, and Infectious Diseases & Immunity, University of California, Berkeley, Berkeley, California, United States of America
- * E-mail:
| |
Collapse
|
19
|
de Oliveira AL, Barbieri NL, Newman DM, Young MM, Nolan LK, Logue CM. Characterizing the Type 6 Secretion System (T6SS) and its role in the virulence of avian pathogenic Escherichia coli strain APECO18. PeerJ 2021; 9:e12631. [PMID: 35003930 PMCID: PMC8686734 DOI: 10.7717/peerj.12631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 11/22/2021] [Indexed: 11/22/2022] Open
Abstract
Avian pathogenic E. coli is the causative agent of extra-intestinal infections in birds known as colibacillosis, which can manifest as localized or systemic infections. The disease affects all stages of poultry production, resulting in economic losses that occur due to morbidity, carcass condemnation and increased mortality of the birds. APEC strains have a diverse virulence trait repertoire, which includes virulence factors involved in adherence to and invasion of the host cells, serum resistance factors, and toxins. However, the pathogenesis of APEC infections remains to be fully elucidated. The Type 6 secretion (T6SS) system has recently gained attention due to its role in the infection process and protection of bacteria from host defenses in human and animal pathogens. Previous work has shown that T6SS components are involved in the adherence to and invasion of host cells, as well as in the formation of biofilm, and intramacrophage bacterial replication. Here, we analyzed the frequency of T6SS genes hcp, impK, evpB, vasK and icmF in a collection of APEC strains and their potential role in virulence-associated phenotypes of APECO18. The T6SS genes were found to be significantly more prevalent in APEC than in fecal E. coli isolates from healthy birds. Expression of T6SS genes was analyzed in culture media and upon contact with host cells. Mutants were generated for hcp, impK, evpB, and icmF and characterized for their impact on virulence-associated phenotypes, including adherence to and invasion of host model cells, and resistance to predation by Dictyostelium discoideum. Deletion of the aforementioned genes did not significantly affect adherence and invasion capabilities of APECO18. Deletion of hcp reduced resistance of APECO18 to predation by D. discoideum, suggesting that T6SS is involved in the virulence of APECO18.
Collapse
Affiliation(s)
- Aline L. de Oliveira
- Department of Population Health, University of Georgia, Athens, GA, United States of America
- Department of Microbiology, University of Georgia, Athens, GA, United States of America
| | - Nicolle L. Barbieri
- Department of Population Health, University of Georgia, Athens, GA, United States of America
| | - Darby M. Newman
- Department of Population Health, University of Georgia, Athens, GA, United States of America
| | - Meaghan M. Young
- Department of Population Health, University of Georgia, Athens, GA, United States of America
| | - Lisa K. Nolan
- Department of Infectious Diseases, University of Georgia, Athens, GA, United States of America
| | - Catherine M. Logue
- Department of Population Health, University of Georgia, Athens, GA, United States of America
| |
Collapse
|
20
|
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: 1.5] [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
|
21
|
Silistre H, Raoux-Barbot D, Mancinelli F, Sangouard F, Dupin A, Belyy A, Deruelle V, Renault L, Ladant D, Touqui L, Mechold U. Prevalence of ExoY Activity in Pseudomonas aeruginosa Reference Panel Strains and Impact on Cytotoxicity in Epithelial Cells. Front Microbiol 2021; 12:666097. [PMID: 34675890 PMCID: PMC8524455 DOI: 10.3389/fmicb.2021.666097] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 07/30/2021] [Indexed: 11/13/2022] Open
Abstract
ExoY is among the effectors that are injected by the type III secretion system (T3SS) of Pseudomonas aeruginosa into host cells. Inside eukaryotic cells, ExoY interacts with F-actin, which stimulates its potent nucleotidyl cyclase activity to produce cyclic nucleotide monophosphates (cNMPs). ExoY has broad substrate specificity with GTP as a preferential substrate in vitro. How ExoY contributes to the virulence of P. aeruginosa remains largely unknown. Here, we examined the prevalence of active ExoY among strains from the international P. aeruginosa reference panel, a collection of strains that includes environmental and clinical isolates, commonly used laboratory strains, and sequential clonal isolates from cystic fibrosis (CF) patients and thus represents the large diversity of this bacterial species. The ability to secrete active ExoY was determined by measuring the F-actin stimulated guanylate cyclase (GC) activity in bacterial culture supernatants. We found an overall ExoY activity prevalence of about 60% among the 40 examined strains with no significant difference between CF and non-CF isolates. In parallel, we used cellular infection models of human lung epithelial cells to compare the cytotoxic effects of isogenic reference strains expressing active ExoY or lacking the exoY gene. We found that P. aeruginosa strains lacking ExoY were in fact more cytotoxic to the epithelial cells than those secreting active ExoY. This suggests that under certain conditions, ExoY might partly alleviate the cytotoxic effects of other virulence factors of P. aeruginosa.
Collapse
Affiliation(s)
- Hazel Silistre
- Unité de Biochimie des Interactions Macromoléculaires, Département de Biologie Structurale et Chimie, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Dorothée Raoux-Barbot
- Unité de Biochimie des Interactions Macromoléculaires, Département de Biologie Structurale et Chimie, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Federica Mancinelli
- Unité de Biochimie des Interactions Macromoléculaires, Département de Biologie Structurale et Chimie, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Flora Sangouard
- Unité de Biochimie des Interactions Macromoléculaires, Département de Biologie Structurale et Chimie, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Alice Dupin
- Unité de Biochimie des Interactions Macromoléculaires, Département de Biologie Structurale et Chimie, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Alexander Belyy
- Unité de Biochimie des Interactions Macromoléculaires, Département de Biologie Structurale et Chimie, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Vincent Deruelle
- Unité de Biochimie des Interactions Macromoléculaires, Département de Biologie Structurale et Chimie, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Louis Renault
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Daniel Ladant
- Unité de Biochimie des Interactions Macromoléculaires, Département de Biologie Structurale et Chimie, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Lhousseine Touqui
- Mucoviscidose: Physiopathologie et Phénogénomique, Centre de Recherche Saint-Antoine (CRSA), INSERM UMR S 938, Sorbonne Université, Paris, France.,Mucoviscidose et Bronchopathies Chroniques, Département Santé Globale, Institute Pasteur, Paris, France
| | - Undine Mechold
- Unité de Biochimie des Interactions Macromoléculaires, Département de Biologie Structurale et Chimie, Institut Pasteur, CNRS UMR 3528, Paris, France
| |
Collapse
|
22
|
Pont S, Blanc-Potard AB. Zebrafish Embryo Infection Model to Investigate Pseudomonas aeruginosa Interaction With Innate Immunity and Validate New Therapeutics. Front Cell Infect Microbiol 2021; 11:745851. [PMID: 34660345 PMCID: PMC8515127 DOI: 10.3389/fcimb.2021.745851] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 09/08/2021] [Indexed: 12/26/2022] Open
Abstract
The opportunistic human pathogen Pseudomonas aeruginosa is responsible for a variety of acute infections and is a major cause of mortality in chronically infected patients with cystic fibrosis (CF). Considering the intrinsic and acquired resistance of P. aeruginosa to currently used antibiotics, new therapeutic strategies against this pathogen are urgently needed. Whereas virulence factors of P. aeruginosa are well characterized, the interplay between P. aeruginosa and the innate immune response during infection remains unclear. Zebrafish embryo is now firmly established as a potent vertebrate model for the study of infectious human diseases, due to strong similarities of its innate immune system with that of humans and the unprecedented possibilities of non-invasive real-time imaging. This model has been successfully developed to investigate the contribution of bacterial and host factors involved in P. aeruginosa pathogenesis, as well as rapidly assess the efficacy of anti-Pseudomonas molecules. Importantly, zebrafish embryo appears as the state-of-the-art model to address in vivo the contribution of innate immunity in the outcome of P. aeruginosa infection. Of interest, is the finding that the zebrafish encodes a CFTR channel closely related to human CFTR, which allowed to develop a model to address P. aeruginosa pathogenesis, innate immune response, and treatment evaluation in a CF context.
Collapse
Affiliation(s)
- Stéphane Pont
- Laboratory of Pathogen-Host Interactions (LPHI), Université Montpellier, Montpellier, France.,CNRS, UMR5235, Montpellier, France
| | - Anne-Béatrice Blanc-Potard
- Laboratory of Pathogen-Host Interactions (LPHI), Université Montpellier, Montpellier, France.,CNRS, UMR5235, Montpellier, France
| |
Collapse
|
23
|
Wen J, Xuan B, Liu Y, Wang L, He L, Meng X, Zhou T, Wang Y. Updating the NLRC4 Inflammasome: from Bacterial Infections to Autoimmunity and Cancer. Front Immunol 2021; 12:702527. [PMID: 34276697 PMCID: PMC8283967 DOI: 10.3389/fimmu.2021.702527] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/17/2021] [Indexed: 01/07/2023] Open
Abstract
Inflammasomes comprise a family of cytosolic multi-protein complexes that modulate the activation of cysteine-aspartate-specific protease 1 (caspase-1) and promote the maturation and secretion of interleukin (IL)-1β and IL-18, leading to an inflammatory response. Different types of inflammasomes are defined by their sensor protein which recognizes pathogenic ligands and then directs inflammasome assembly. Although the specific molecular mechanisms underlying the activation of most inflammasomes are still unclear, NLRC4 inflammasomes have emerged as multifaceted agents of the innate immune response, playing important roles in immune defense against a variety of pathogens. Other studies have also expanded the scope of NLRC4 inflammasomes to include a range of inherited human autoimmune diseases as well as proposed roles in cancer. In this review article, we provide an updated overview of NLRC4 inflammasomes, describing their composition, activation mechanisms and roles in both microbial infections and other disease conditions.
Collapse
Affiliation(s)
- Jiexia Wen
- Department of Central Laboratory, First Hospital of Qinhuangdao, Hebei Medical University, Qinhuangdao, China
| | - Bin Xuan
- Department of General Surgery, First Hospital of Qinhuangdao, Hebei Medical University, Qinhuangdao, China
| | - Yang Liu
- Department of General Surgery, First Hospital of Qinhuangdao, Hebei Medical University, Qinhuangdao, China
| | - Liwei Wang
- Department of General Surgery, First Hospital of Qinhuangdao, Hebei Medical University, Qinhuangdao, China
| | - Li He
- Department of General Surgery, First Hospital of Qinhuangdao, Hebei Medical University, Qinhuangdao, China
| | - Xiangcai Meng
- Department of General Surgery, First Hospital of Qinhuangdao, Hebei Medical University, Qinhuangdao, China
| | - Tao Zhou
- Department of General Surgery, First Hospital of Qinhuangdao, Hebei Medical University, Qinhuangdao, China
| | - Yimin Wang
- Department of Central Laboratory, First Hospital of Qinhuangdao, Hebei Medical University, Qinhuangdao, China.,Department of General Surgery, First Hospital of Qinhuangdao, Hebei Medical University, Qinhuangdao, China
| |
Collapse
|
24
|
Morin CD, Déziel E, Gauthier J, Levesque RC, Lau GW. An Organ System-Based Synopsis of Pseudomonas aeruginosa Virulence. Virulence 2021; 12:1469-1507. [PMID: 34180343 PMCID: PMC8237970 DOI: 10.1080/21505594.2021.1926408] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Driven in part by its metabolic versatility, high intrinsic antibiotic resistance, and a large repertoire of virulence factors, Pseudomonas aeruginosa is expertly adapted to thrive in a wide variety of environments, and in the process, making it a notorious opportunistic pathogen. Apart from the extensively studied chronic infection in the lungs of people with cystic fibrosis (CF), P. aeruginosa also causes multiple serious infections encompassing essentially all organs of the human body, among others, lung infection in patients with chronic obstructive pulmonary disease, primary ciliary dyskinesia and ventilator-associated pneumonia; bacteremia and sepsis; soft tissue infection in burns, open wounds and postsurgery patients; urinary tract infection; diabetic foot ulcers; chronic suppurative otitis media and otitis externa; and keratitis associated with extended contact lens use. Although well characterized in the context of CF, pathogenic processes mediated by various P. aeruginosa virulence factors in other organ systems remain poorly understood. In this review, we use an organ system-based approach to provide a synopsis of disease mechanisms exerted by P. aeruginosa virulence determinants that contribute to its success as a versatile pathogen.
Collapse
Affiliation(s)
- Charles D Morin
- Centre Armand-Frappier Santé Biotechnologie, Institut National De La Recherche Scientifique (INRS), Laval, Quebec, Canada
| | - Eric Déziel
- Centre Armand-Frappier Santé Biotechnologie, Institut National De La Recherche Scientifique (INRS), Laval, Quebec, Canada
| | - Jeff Gauthier
- Département De Microbiologie-infectiologie Et Immunologie, Institut De Biologie Intégrative Et Des Systèmes (IBIS), Université Laval, Québec City, Quebec, Canada
| | - Roger C Levesque
- Département De Microbiologie-infectiologie Et Immunologie, Institut De Biologie Intégrative Et Des Systèmes (IBIS), Université Laval, Québec City, Quebec, Canada
| | - Gee W Lau
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL, US
| |
Collapse
|
25
|
Gharieb R, Saad M, Khedr M, El Gohary A, Ibrahim H. Occurrence, virulence, carbapenem resistance, susceptibility to disinfectants and public health hazard of Pseudomonas aeruginosa isolated from animals, humans and environment in intensive farms. J Appl Microbiol 2021; 132:256-267. [PMID: 34171153 DOI: 10.1111/jam.15191] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/17/2021] [Accepted: 06/22/2021] [Indexed: 01/19/2023]
Abstract
AIMS This work aimed to determine the occurrence, virulence, antibiogram, carbapenem resistance genes and susceptibility to disinfectants of Pseudomonas aeruginosa isolated from animals, environment and workers in intensive farms. METHODS AND RESULTS A total of 610 samples from intensive beef cattle and sheep farms in Kafr El Sheikh Governorate, Egypt were screened for the presence of P. aeruginosa using bacteriological assays. The isolates were characterized by PCR and tested for susceptibility to antibiotics using disk diffusion method and disinfectants by quantitative suspension test. In all, 60 P. aeruginosa isolates were recovered in this study and all isolates harboured at least one of the virulence genes tested. Human P. aeruginosa isolates were highly resistant to cephalosporins, fluroquinolones, aminoglycosides, carbapenems and penicillins+β-lactamase inhibitors than non-human isolates. Colistin resistance was higher in non-human than human P. aeruginosa isolates, whereas low resistance to aztreonam was observed in non-human and human isolates. Carbapenem-resistant P. aeruginosa (CRPA) strains were recovered from workers (56.5%), sheep (8.3%) and cattle (8.3%). All CRPA harboured at least one of the carbapenem resistance genes tested and most of them showed multidrug resistance (MDR) or extensive drug resistance (XDR) phenotypes. Glutaraldehyde 1% and hydrogen peroxide 3% eliminated P. aeruginosa completely in the absence and presence of organic matter within short contact time compared with other disinfectants. CONCLUSIONS This study reported the occurrence of CRPA in animals and workers in intensive farms. Glutaraldehyde and hydrogen peroxide were the most effective disinfectants against P. aeruginosa. SIGNIFICANCE AND IMPACT OF THE STUDY The occurrence of CRPA in intensive livestock farms is a serious challenge that threatens animal and human health and increases the risk of P. aeruginosa infection in the community. Therefore, it is vital to control the spread of CRPA by banning or restricting the use of antibiotics and applying proper cleaning and disinfection protocols in livestock farms.
Collapse
Affiliation(s)
- Rasha Gharieb
- Department of Zoonoses, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Mai Saad
- Department of Veterinary Public Health, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Mariam Khedr
- Department of Veterinary Public Health, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | | | | |
Collapse
|
26
|
Rao L, De La Rosa I, Xu Y, Sha Y, Bhattacharya A, Holtzman MJ, Gilbert BE, Eissa NT. Pseudomonas aeruginosa survives in epithelia by ExoS-mediated inhibition of autophagy and mTOR. EMBO Rep 2021; 22:e50613. [PMID: 33345425 PMCID: PMC7857434 DOI: 10.15252/embr.202050613] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 10/29/2020] [Accepted: 11/12/2020] [Indexed: 12/12/2022] Open
Abstract
One major factor that contributes to the virulence of Pseudomonas aeruginosa is its ability to reside and replicate unchallenged inside airway epithelial cells. The mechanism by which P. aeruginosa escapes destruction by intracellular host defense mechanisms, such as autophagy, is not known. Here, we show that the type III secretion system effector protein ExoS facilitates P. aeruginosa survival in airway epithelial cells by inhibiting autophagy in host cells. Autophagy inhibition is independent of mTOR activity, as the latter is also inhibited by ExoS, albeit by a different mechanism. Deficiency of the critical autophagy gene Atg7 in airway epithelial cells, both in vitro and in mouse models, greatly enhances the survival of ExoS-deficient P. aeruginosa but does not affect the survival of ExoS-containing bacteria. The inhibitory effect of ExoS on autophagy and mTOR depends on the activity of its ADP-ribosyltransferase domain. Inhibition of mTOR is caused by ExoS-mediated ADP ribosylation of RAS, whereas autophagy inhibition is due to the suppression of autophagic Vps34 kinase activity.
Collapse
Affiliation(s)
- Lang Rao
- Department of MedicineBaylor College of MedicineHoustonTXUSA
- Veterans Administration Long Beach Health Care System and University of California at IrvineIrvineCAUSA
- Southern California Institute for Research and EducationLong BeachCAUSA
| | | | - Yi Xu
- Department of MedicineBaylor College of MedicineHoustonTXUSA
| | - Youbao Sha
- Department of MedicineBaylor College of MedicineHoustonTXUSA
| | | | - Michael J Holtzman
- Department of Internal MedicineWashington University School of MedicineSt. LouisMOUSA
| | - Brian E Gilbert
- Department of Molecular Virology and MicrobiologyBaylor College of MedicineHoustonTXUSA
| | - N Tony Eissa
- Department of MedicineBaylor College of MedicineHoustonTXUSA
- Veterans Administration Long Beach Health Care System and University of California at IrvineIrvineCAUSA
| |
Collapse
|
27
|
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: 6] [Impact Index Per Article: 1.2] [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
|
28
|
Multifunctional Monoclonal Antibody Targeting Pseudomonas aeruginosa Keratitis in Mice. Vaccines (Basel) 2020; 8:vaccines8040638. [PMID: 33147726 PMCID: PMC7712430 DOI: 10.3390/vaccines8040638] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/24/2020] [Accepted: 10/29/2020] [Indexed: 01/13/2023] Open
Abstract
A worrisome trend in the study and treatment of infectious disease noted in recent years is the increase in multidrug resistant strains of bacteria concurrent with a scarcity of new antimicrobial agents to counteract this rise. This is particularly true amongst bacteria within the Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species (ESKAPE) designation. P. aeruginosa is one of the most common causes of bacterial keratitis. Therefore, it is of vital importance to characterize new antimicrobial agents with anti-Pseudomonal activity for use with the ocular surface. MEDI3902 is a multifunctional antibody that targets the P. aeruginosa persistence factor Psl exopolysaccharide, and the type 3 secretion protein PcrV. We initially assessed this antibody for ocular surface toxicity. The antimicrobial activity of the antibody was then tested by treating mice with established P. aeruginosa keratitis with both topical and intravenous treatment modalities. MEDI3902, was shown to be non-toxic to the ocular surface of mice when given topically. It was also effective compared to the control antibody at preventing P. aeruginosa keratitis with a one-time treatment at the time of infection. Both topical and intravenous administration of MEDI3902 has been proved significant in treating established keratitis infections as well, speeding the resolution of infection significantly more than that of the control IgG. We report the first use of a topical immunotherapeutic multifunctional agent targeting Psl and type 3 secretion on the ocular surface as an antimicrobial agent. While MEDI3902 has been shown to prevent Pseudomonas biofilm formation in keratitis models when given prophylactically intravitally, we show that MEDI3902 has the capability to also treat an active infection when given intravenously to mice with Pseudomonas keratitis. Our data indicate antibodies are well tolerated and nontoxic on the ocular surface. They reduce infection in mice treated concurrently at inoculation and reduced the signs of cornea pathology in mice with established infection. Taken together, these data indicate treatment with monoclonal antibodies directed against Psl and PcrV may be clinically effective in the treatment of P. aeruginosa keratitis and suggest that the design of further antibodies to be an additional tool in the treatment of bacterial keratitis.
Collapse
|
29
|
Bouillot S, Pont S, Gallet B, Moriscot C, Deruelle V, Attrée I, Huber P. Inflammasome activation by Pseudomonas aeruginosa's ExlA pore-forming toxin is detrimental for the host. Cell Microbiol 2020; 22:e13251. [PMID: 32779854 DOI: 10.1111/cmi.13251] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 07/24/2020] [Accepted: 07/30/2020] [Indexed: 02/06/2023]
Abstract
During acute Pseudomonas aeruginosa infection, the inflammatory response is essential for bacterial clearance. Neutrophil recruitment can be initiated following the assembly of an inflammasome within sentinel macrophages, leading to activation of caspase-1, which in turn triggers macrophage pyroptosis and IL-1β/IL-18 maturation. Inflammasome formation can be induced by a number of bacterial determinants, including Type III secretion systems (T3SSs) or pore-forming toxins, or, alternatively, by lipopolysaccharide (LPS) via caspase-11 activation. Surprisingly, previous studies indicated that a T3SS-induced inflammasome increased pathogenicity in mouse models of P. aeruginosa infection. Here, we investigated the immune reaction of mice infected with a T3SS-negative P. aeruginosa strain (IHMA879472). Virulence of this strain relies on ExlA, a secreted pore-forming toxin. IHMA879472 promoted massive neutrophil infiltration in infected lungs, owing to efficient priming of toll-like receptors, and thus enhanced the expression of inflammatory proteins including pro-IL-1β and TNF-α. However, mature-IL-1β and IL-18 were undetectable in wild-type mice, suggesting that ExlA failed to effectively activate caspase-1. Nevertheless, caspase-1/11 deficiency improved survival following infection with IHMA879472, as previously described for T3SS+ bacteria. We conclude that the detrimental effect associated with the ExlA-induced inflammasome is probably not due to hyperinflammation, rather it stems from another inflammasome-dependent process.
Collapse
Affiliation(s)
- Stéphanie Bouillot
- Unité de Biologie Cellulaire et Infection, CEA, INSERM, CNRS, Université Grenoble-Alpes, Grenoble, France
| | - Stéphane Pont
- Unité de Biologie Cellulaire et Infection, CEA, INSERM, CNRS, Université Grenoble-Alpes, Grenoble, France
| | - Benoit Gallet
- Institut de Biologie Structurale, CEA, CNRS, Université Grenoble-Alpes, Grenoble, France
| | - Christine Moriscot
- Institut de Biologie Structurale, CEA, CNRS, Université Grenoble-Alpes, Grenoble, France
| | - Vincent Deruelle
- Unité de Biologie Cellulaire et Infection, CEA, INSERM, CNRS, Université Grenoble-Alpes, Grenoble, France
| | - Ina Attrée
- Unité de Biologie Cellulaire et Infection, CEA, INSERM, CNRS, Université Grenoble-Alpes, Grenoble, France
| | - Philippe Huber
- Unité de Biologie Cellulaire et Infection, CEA, INSERM, CNRS, Université Grenoble-Alpes, Grenoble, France
| |
Collapse
|
30
|
Andrade WA, Zamboni DS. NLRC4 biology in immunity and inflammation. J Leukoc Biol 2020; 108:1117-1127. [PMID: 32531834 DOI: 10.1002/jlb.3mr0420-573r] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/29/2020] [Accepted: 05/05/2020] [Indexed: 12/14/2022] Open
Abstract
Inflammasomes are cytosolic multiprotein complexes that sense microbial infections or host cell damage, triggering cytokine production and a proinflammatory form of cell death, called pyroptosis. Whereas pyroptosis and cytokine production may often promote host resistance to infections, uncontrolled inflammasome activation leads to autoinflammatory diseases in humans. Among the multiple inflammasomes described, the neuronal apoptosis inhibitory protein/nucleotide-binding domain leucine-rich repeat-containing protein family caspase activation and recruitment domain-containing protein 4 (NLRC4) inflammasome emerged as a critical component for the restriction of bacterial infections. Accordingly, our understanding of this inflammasome advanced remarkably over the last 10 yr, expanding our knowledge about ligand-receptor interaction; cryo-EM structure; and downstream effectors and substrates, such as gasdermin-D, caspase-1, caspase-8, and caspase-7. In this review, we discuss recent advances on the biology of the NLRC4 inflammasome, in terms of structure and activation mechanisms, importance in bacterial and nonbacterial diseases, and the identification of NLRC4 gain-of-function mutations leading to NLRC4-associated autoinflammatory diseases in humans.
Collapse
Affiliation(s)
- Warrison A Andrade
- Department of Cell Biology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Ribeirão Preto, São Paulo, Brazil
| | - Dario S Zamboni
- Department of Cell Biology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
31
|
Chatterjee P, Sass G, Swietnicki W, Stevens DA. Review of Potential Pseudomonas Weaponry, Relevant to the Pseudomonas-Aspergillus Interplay, for the Mycology Community. J Fungi (Basel) 2020; 6:jof6020081. [PMID: 32517271 PMCID: PMC7345761 DOI: 10.3390/jof6020081] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/03/2020] [Accepted: 06/03/2020] [Indexed: 12/15/2022] Open
Abstract
Pseudomonas aeruginosa is one of the most prominent opportunistic bacteria in airways of cystic fibrosis patients and in immunocompromised patients. These bacteria share the same polymicrobial niche with other microbes, such as the opportunistic fungus Aspergillus fumigatus. Their inter-kingdom interactions and diverse exchange of secreted metabolites are responsible for how they both fare in competition for ecological niches. The outcomes of their contests likely determine persistent damage and degeneration of lung function. With a myriad of virulence factors and metabolites of promising antifungal activity, P. aeruginosa products or their derivatives may prove useful in prophylaxis and therapy against A. fumigatus. Quorum sensing underlies the primary virulence strategy of P. aeruginosa, which serves as cell–cell communication and ultimately leads to the production of multiple virulence factors. Understanding the quorum-sensing-related pathogenic mechanisms of P. aeruginosa is a first step for understanding intermicrobial competition. In this review, we provide a basic overview of some of the central virulence factors of P. aeruginosa that are regulated by quorum-sensing response pathways and briefly discuss the hitherto known antifungal properties of these virulence factors. This review also addresses the role of the bacterial secretion machinery regarding virulence factor secretion and maintenance of cell–cell communication.
Collapse
Affiliation(s)
- Paulami Chatterjee
- California Institute for Medical Research, San Jose, CA 95128, USA; (P.C.); (G.S.)
| | - Gabriele Sass
- California Institute for Medical Research, San Jose, CA 95128, USA; (P.C.); (G.S.)
| | - Wieslaw Swietnicki
- Department of Immunology of Infectious Diseases, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 50-114 Wroclaw, Poland;
| | - David A. Stevens
- California Institute for Medical Research, San Jose, CA 95128, USA; (P.C.); (G.S.)
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Correspondence: ; Tel.: +1-408-998-4554
| |
Collapse
|
32
|
Fleiszig SMJ, Kroken AR, Nieto V, Grosser MR, Wan SJ, Metruccio MME, Evans DJ. Contact lens-related corneal infection: Intrinsic resistance and its compromise. Prog Retin Eye Res 2019; 76:100804. [PMID: 31756497 DOI: 10.1016/j.preteyeres.2019.100804] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 11/05/2019] [Accepted: 11/12/2019] [Indexed: 12/20/2022]
Abstract
Contact lenses represent a widely utilized form of vision correction with more than 140 million wearers worldwide. Although generally well-tolerated, contact lenses can cause corneal infection (microbial keratitis), with an approximate annualized incidence ranging from ~2 to ~20 cases per 10,000 wearers, and sometimes resulting in permanent vision loss. Research suggests that the pathogenesis of contact lens-associated microbial keratitis is complex and multifactorial, likely requiring multiple conspiring factors that compromise the intrinsic resistance of a healthy cornea to infection. Here, we outline our perspective of the mechanisms by which contact lens wear sometimes renders the cornea susceptible to infection, focusing primarily on our own research efforts during the past three decades. This has included studies of host factors underlying the constitutive barrier function of the healthy cornea, its response to bacterial challenge when intrinsic resistance is not compromised, pathogen virulence mechanisms, and the effects of contact lens wear that alter the outcome of host-microbe interactions. For almost all of this work, we have utilized the bacterium Pseudomonas aeruginosa because it is the leading cause of lens-related microbial keratitis. While not yet common among corneal isolates, clinical isolates of P. aeruginosa have emerged that are resistant to virtually all currently available antibiotics, leading the United States CDC (Centers for Disease Control) to add P. aeruginosa to its list of most serious threats. Compounding this concern, the development of advanced contact lenses for biosensing and augmented reality, together with the escalating incidence of myopia, could portent an epidemic of vision-threatening corneal infections in the future. Thankfully, technological advances in genomics, proteomics, metabolomics and imaging combined with emerging models of contact lens-associated P. aeruginosa infection hold promise for solving the problem - and possibly life-threatening infections impacting other tissues.
Collapse
Affiliation(s)
- Suzanne M J Fleiszig
- School of Optometry, University of California, Berkeley, CA, USA; Graduate Group in Vision Science, University of California, Berkeley, CA, USA; Graduate Groups in Microbiology and Infectious Diseases & Immunity, University of California, Berkeley, CA, USA.
| | - Abby R Kroken
- School of Optometry, University of California, Berkeley, CA, USA
| | - Vincent Nieto
- School of Optometry, University of California, Berkeley, CA, USA
| | | | - Stephanie J Wan
- Graduate Group in Vision Science, University of California, Berkeley, CA, USA
| | | | - David J Evans
- School of Optometry, University of California, Berkeley, CA, USA; College of Pharmacy, Touro University California, Vallejo, CA, USA
| |
Collapse
|
33
|
Hritonenko V, Metruccio M, Evans D, Fleiszig S. Epithelial cell lysates induce ExoS expression and secretion by Pseudomonas aeruginosa. FEMS Microbiol Lett 2019. [PMID: 29518189 DOI: 10.1093/femsle/fny053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The type three secretion system (T3SS) is important for the intracellular survival of Pseudomonas aeruginosa. Known T3SS inducers include low Ca2+, serum or host cell contact. Here, we used corneal epithelial cell lysates to test if host cytosolic factors could also induce the T3SS. Invasive P. aeruginosa strain PAO1 was exposed to cell lysates for 16 h, and expression of T3SS effectors determined by q-PCR and Western immunoblot. Lysate exposure reduced PAO1 growth (∼5-fold) versus trypticase soy broth (TSB), but also resulted in appearance of a protein in culture supernatants, but not bacterial cell pellets, which reacted with antibody raised against ExoS. T3SS-inducing media (TSBi) caused the expression and secretion of ExoS and ExoT. Heat-treated lysates induced the protein; 1:3 diluted lysates did not. The protein that bound anti-ExoS antibody was found in supernatants of lysate-exposed exoT mutants, but not exoS or pscC mutants, suggesting a secreted form of ExoS, albeit slightly larger than that induced by TSBi. Lysate-exposed strain PAK expressed the same protein. Lysates caused PAO1 exoS and exoT gene expression, but only ∼20% and ∼6% of TSBi, respectively. T3SS induction by epithelial cell lysates could help explain T3SS expression by internalized P. aeruginosa.
Collapse
Affiliation(s)
| | - Matteo Metruccio
- School of Optometry, University of California, Berkeley, CA 94720-2020, USA
| | - David Evans
- School of Optometry, University of California, Berkeley, CA 94720-2020, USA.,College of Pharmacy, Touro University California, Vallejo, CA 94592-2020, USA
| | - Suzanne Fleiszig
- School of Optometry, University of California, Berkeley, CA 94720-2020, USA.,Graduate Groups in Vision Science, Microbiology, and Infectious Diseases & Immunity, University of California, Berkeley, CA 94720-2020, USA
| |
Collapse
|
34
|
Garai P, Berry L, Moussouni M, Bleves S, Blanc-Potard AB. Killing from the inside: Intracellular role of T3SS in the fate of Pseudomonas aeruginosa within macrophages revealed by mgtC and oprF mutants. PLoS Pathog 2019; 15:e1007812. [PMID: 31220187 PMCID: PMC6586356 DOI: 10.1371/journal.ppat.1007812] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 05/02/2019] [Indexed: 12/19/2022] Open
Abstract
While considered solely an extracellular pathogen, increasing evidence indicates that Pseudomonas aeruginosa encounters intracellular environment in diverse mammalian cell types, including macrophages. In the present study, we have deciphered the intramacrophage fate of wild-type P. aeruginosa PAO1 strain by live and electron microscopy. P. aeruginosa first resided in phagosomal vacuoles and subsequently could be detected in the cytoplasm, indicating phagosomal escape of the pathogen, a finding also supported by vacuolar rupture assay. The intracellular bacteria could eventually induce cell lysis, both in a macrophage cell line and primary human macrophages. Two bacterial factors, MgtC and OprF, recently identified to be important for survival of P. aeruginosa in macrophages, were found to be involved in bacterial escape from the phagosome as well as in cell lysis caused by intracellular bacteria. Strikingly, type III secretion system (T3SS) genes of P. aeruginosa were down-regulated within macrophages in both mgtC and oprF mutants. Concordantly, cyclic di-GMP (c-di-GMP) level was increased in both mutants, providing a clue for negative regulation of T3SS inside macrophages. Consistent with the phenotypes and gene expression pattern of mgtC and oprF mutants, a T3SS mutant (ΔpscN) exhibited defect in phagosomal escape and macrophage lysis driven by internalized bacteria. Importantly, these effects appeared to be largely dependent on the ExoS effector, in contrast with the known T3SS-dependent, but ExoS independent, cytotoxicity caused by extracellular P. aeruginosa towards macrophages. Moreover, this macrophage damage caused by intracellular P. aeruginosa was found to be dependent on GTPase Activating Protein (GAP) domain of ExoS. Hence, our work highlights T3SS and ExoS, whose expression is modulated by MgtC and OprF, as key players in the intramacrophage life of P. aeruginosa which allow internalized bacteria to lyse macrophages. The ability of professional phagocytes to ingest and kill microorganisms is central to host defense and Pseudomonas aeruginosa has developed mechanisms to avoid being killed by phagocytes. While considered an extracellular pathogen, P. aeruginosa has been reported to be engulfed by macrophages in animal models. Here, we visualized the fate of P. aeruginosa within cultured macrophages, revealing macrophage lysis driven by intracellular P. aeruginosa. Two bacterial factors, MgtC and OprF, recently discovered to be involved in the intramacrophage survival of P. aeruginosa, appeared to play a role in this cytotoxicity caused by intracellular bacteria. We provided evidence that type III secretion system (T3SS) gene expression is lowered intracellularly in mgtC and oprF mutants. We further showed that intramacrophage P. aeruginosa uses its T3SS, specifically the ExoS effector, to promote phagosomal escape and cell lysis. We thus describe a transient intramacrophage stage of P. aeruginosa that could contribute to bacterial dissemination.
Collapse
Affiliation(s)
- Preeti Garai
- Laboratoire de Dynamique des Interactions Membranaires Normales et Pathologiques, Université de Montpellier, CNRS-UMR5235, Montpellier, France
| | - Laurence Berry
- Laboratoire de Dynamique des Interactions Membranaires Normales et Pathologiques, Université de Montpellier, CNRS-UMR5235, Montpellier, France
| | - Malika Moussouni
- Laboratoire de Dynamique des Interactions Membranaires Normales et Pathologiques, Université de Montpellier, CNRS-UMR5235, Montpellier, France
| | - Sophie Bleves
- LISM, Institut de Microbiologie de la Méditerranée, CNRS & Aix-Marseille Univ, Marseille, France
| | - Anne-Béatrice Blanc-Potard
- Laboratoire de Dynamique des Interactions Membranaires Normales et Pathologiques, Université de Montpellier, CNRS-UMR5235, Montpellier, France
- * E-mail:
| |
Collapse
|
35
|
Fitting Pieces into the Puzzle of Pseudomonas aeruginosa Type III Secretion System Gene Expression. J Bacteriol 2019; 201:JB.00209-19. [PMID: 31010903 DOI: 10.1128/jb.00209-19] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Type III secretion systems (T3SS) are widely distributed in Gram-negative microorganisms and critical for host-pathogen and host-symbiont interactions with plants and animals. Central features of the T3SS are a highly conserved set of secretion and translocation genes and contact dependence wherein host-pathogen interactions trigger effector protein delivery and serve as an inducing signal for T3SS gene expression. In addition to these conserved features, there are pathogen-specific properties that include a unique repertoire of effector genes and mechanisms to control T3SS gene expression. The Pseudomonas aeruginosa T3SS serves as a model system to understand transcriptional and posttranscriptional mechanisms involved in the control of T3SS gene expression. The central regulatory feature is a partner-switching system that controls the DNA-binding activity of ExsA, the primary regulator of T3SS gene expression. Superimposed upon the partner-switching mechanism are cyclic AMP and cyclic di-GMP signaling systems, two-component systems, global regulators, and RNA-binding proteins that have positive and negative effects on ExsA transcription and/or synthesis. In the present review, we discuss advances in our understanding of how these regulatory systems orchestrate the activation of T3SS gene expression in the context of acute infections and repression of the T3SS as P. aeruginosa adapts to and colonizes the cystic fibrosis airways.
Collapse
|
36
|
Kong W, Dong M, Yan R, Liang Q, Zhang H, Luo W, Zhang Y, Liang H, Duan K. A Unique ATPase, ArtR (PA4595), Represses the Type III Secretion System in Pseudomonas aeruginosa. Front Microbiol 2019; 10:560. [PMID: 30949153 PMCID: PMC6437102 DOI: 10.3389/fmicb.2019.00560] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 03/05/2019] [Indexed: 11/13/2022] Open
Abstract
Pseudomonas aeruginosa is an important human pathogen which uses the type III secretion system (T3SS) as a primary virulence factor to establish infections in humans. The results presented in this report revealed that the ATP-binding protein PA4595 (named ArtR, a Regulator that is an ATP-activated Repressor of T3SS) represses T3SS expression in P. aeruginosa. The expression of T3SS genes, including exoS, exoY, exoT, exsCEBA, and exsD-pscB-L, increased significantly when artR was knockout. The effect of ArtR on ExsA is at the transcriptional level, not at the translational level. The regulatory role and cytoplasm localization of ArtR suggest it belongs to the REG sub-family of ATP-binding cassette (ABC) family. Purified GST-tagged ArtR showed ATPase activity in vitro. The conserved aspartate residues in the dual Walker B motifs prove to be essential for the regulatory function of ArtR. The regulation of T3SS by ArtR is unique, which does not involve the known GacS/A-RsmY/Z-RsmA-ExsA pathway or Vfr. This is the first REG subfamily of ATP-binding cassette that is reported to regulate T3SS genes in bacteria. The results specify a novel player in the regulatory networks of T3SS in P. aeruginosa.
Collapse
Affiliation(s)
- Weina Kong
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi'an, China.,Department of Oral Biology and Department of Medical Microbiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Mengmeng Dong
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi'an, China
| | - Rong Yan
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi'an, China
| | - Qingqing Liang
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi'an, China
| | - Huiqun Zhang
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi'an, China
| | - Wei Luo
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi'an, China
| | - Yani Zhang
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi'an, China
| | - Haihua Liang
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi'an, China
| | - Kangmin Duan
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi'an, China.,Department of Oral Biology and Department of Medical Microbiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
37
|
The Role of Pseudomonas aeruginosa ExoY in an Acute Mouse Lung Infection Model. Toxins (Basel) 2018; 10:toxins10050185. [PMID: 29734720 PMCID: PMC5983241 DOI: 10.3390/toxins10050185] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 04/20/2018] [Accepted: 05/02/2018] [Indexed: 12/31/2022] Open
Abstract
The effector protein Exotoxin Y (ExoY) produced by Pseudomonas aeruginosa is injected via the type III secretion system (T3SS) into host cells. ExoY acts as nucleotidyl cyclase promoting the intracellular accumulation of cyclic nucleotides. To what extent nucleotidyl cyclase activity contributes to the pathogenicity of ExoY and which mechanisms participate in the manifestation of lung infection is still unclear. Here, we used an acute airway infection model in mice to address the role of ExoY in lung infection. In infected lungs, a dose-dependent phenotype of infection with bacteria-expressing ExoY was mirrored by haemorrhage, formation of interstitial oedema in alveolar septa, and infiltration of the perivascular space with erythrocytes and neutrophilic granulocytes. Analyses of the infection process on the cellular and organismal level comparing infections with Pseudomonas aeruginosa mutants expressing either nucleotidyl cyclase-active or -inactive ExoY revealed differential cytokine secretion, increased prevalence of apoptosis, and a break of lung barrier integrity in mice infected with cyclase-active ExoY. Notably, of all measured cyclic nucleotides, only the increase of cyclic UMP in infected mouse lungs coincides temporally with the observed early pathologic changes. In summary, our results suggest that the nucleotidyl cyclase activity of ExoY can contribute to P. aeruginosa acute pathogenicity.
Collapse
|
38
|
The Impact of ExoS on Pseudomonas aeruginosa Internalization by Epithelial Cells Is Independent of fleQ and Correlates with Bistability of Type Three Secretion System Gene Expression. mBio 2018; 9:mBio.00668-18. [PMID: 29717012 PMCID: PMC5930308 DOI: 10.1128/mbio.00668-18] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Pseudomonas aeruginosa is internalized into multiple types of epithelial cell in vitro and in vivo and yet is often regarded as an exclusively extracellular pathogen. Paradoxically, ExoS, a type three secretion system (T3SS) effector, has antiphagocytic activities but is required for intracellular survival of P. aeruginosa and its occupation of bleb niches in epithelial cells. Here, we addressed mechanisms for this dichotomy using invasive (ExoS-expressing) P. aeruginosa and corresponding effector-null isogenic T3SS mutants, effector-null mutants of cytotoxic P. aeruginosa with and without ExoS transformation, antibiotic exclusion assays, and imaging using a T3SS-GFP reporter. Except for effector-null PA103, all strains were internalized while encoding ExoS. Intracellular bacteria showed T3SS activation that continued in replicating daughter cells. Correcting the fleQ mutation in effector-null PA103 promoted internalization by >10-fold with or without ExoS. Conversely, mutating fleQ in PAO1 reduced internalization by >10-fold, also with or without ExoS. Effector-null PA103 remained less well internalized than PAO1 matched for fleQ status, but only with ExoS expression, suggesting additional differences between these strains. Quantifying T3SS activation using GFP fluorescence and quantitative reverse transcription-PCR (qRT-PCR) showed that T3SS expression was hyperinducible for strain PA103ΔexoUT versus other isolates and was unrelated to fleQ status. These findings support the principle that P. aeruginosa is not exclusively an extracellular pathogen, with internalization influenced by the relative proportions of T3SS-positive and T3SS-negative bacteria in the population during host cell interaction. These data also challenge current thinking about T3SS effector delivery into host cells and suggest that T3SS bistability is an important consideration in studying P. aeruginosa pathogenesis. P. aeruginosa is often referred to as an extracellular pathogen, despite its demonstrated capacity to invade and survive within host cells. Fueling the confusion, P. aeruginosa encodes T3SS effectors with anti-internalization activity that, paradoxically, play critical roles in intracellular survival. Here, we sought to address why ExoS does not prevent internalization of the P. aeruginosa strains that natively encode it. Results showed that ExoS exerted unusually strong anti-internalization activity under conditions of expression in the effector-null background of strain PA103, often used to study T3SS effector activity. Inhibition of internalization was associated with T3SS hyperinducibility and ExoS delivery. PA103 fleQ mutation, preventing flagellar assembly, further reduced internalization but did so independently of ExoS. The results revealed intracellular T3SS expression by all strains and suggested that T3SS bistability influences P. aeruginosa internalization. These findings reconcile controversies in the literature surrounding P. aeruginosa internalization and support the principle that P. aeruginosa is not exclusively an extracellular pathogen.
Collapse
|
39
|
LaCourse KD, Peterson SB, Kulasekara HD, Radey MC, Kim J, Mougous JD. Conditional toxicity and synergy drive diversity among antibacterial effectors. Nat Microbiol 2018; 3:440-446. [PMID: 29459733 PMCID: PMC5876133 DOI: 10.1038/s41564-018-0113-y] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 01/16/2018] [Indexed: 12/19/2022]
Abstract
Bacteria in polymicrobial habitats contend with a persistent barrage of competitors, often under rapidly changing environmental conditions1. The direct antagonism of competitor cells is thus an important bacterial survival strategy2. Towards this end, many bacterial species employ an arsenal of antimicrobial effectors with multiple activities; however, the benefits conferred by the simultaneous deployment of diverse toxins are unknown. Here we show that the multiple effectors delivered to competitor bacteria by the type VI secretion system (T6SS) of Pseudomonas aeruginosa display conditional efficacy and act synergistically. One of these effectors, Tse4, is most active in high salinity environments and synergizes with effectors that degrade the cell wall or inactivate intracellular electron carriers. We find Tse4 synergizes with these disparate mechanisms by forming pores that disrupt the ΔΨ component of the proton motive force. Our results provide evidence that the concomitant delivery of a cocktail of effectors serves as a bet-hedging strategy to promote bacterial competitiveness in the face of unpredictable and variable environmental conditions.
Collapse
Affiliation(s)
| | - S Brook Peterson
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | | | - Matthew C Radey
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Jungyun Kim
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Joseph D Mougous
- Department of Microbiology, University of Washington, Seattle, WA, USA. .,Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA.
| |
Collapse
|
40
|
Dortet L, Lombardi C, Cretin F, Dessen A, Filloux A. Pore-forming activity of the Pseudomonas aeruginosa type III secretion system translocon alters the host epigenome. Nat Microbiol 2018; 3:378-386. [PMID: 29403015 DOI: 10.1038/s41564-018-0109-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 12/21/2017] [Indexed: 12/13/2022]
Abstract
Recent studies highlight that bacterial pathogens can reprogram target cells by influencing epigenetic factors. The type III secretion system (T3SS) is a bacterial nanomachine that resembles a syringe on the bacterial surface. The T3SS 'needle' delivers translocon proteins into eukaryotic cell membranes, subsequently allowing injection of bacterial effectors into the cytosol. Here we show that Pseudomonas aeruginosa induces early T3SS-dependent dephosphorylation and deacetylation of histone H3 in eukaryotic cells. This is not triggered by any of the P. aeruginosa T3SS effectors, but results from the insertion of the PopB-PopD translocon into the membrane. This suggests that the P. aeruginosa translocon is a genuine T3SS effector acting as a pore-forming toxin. We visualized the translocon plugged into the host cell membrane after the bacterium has left the site of contact, and demonstrate that subsequent ion exchange through this pore is responsible for histone H3 modifications and host cell subversion.
Collapse
Affiliation(s)
- Laurent Dortet
- MRC Centre for Molecular Microbiology and Infection, Department of Life Sciences, Imperial College London, London, UK.,EA7361 'Structure, dynamic, function and expression of broad spectrum β-lactamases', Faculty of Medicine, Paris-Sud University, LabEx Lermit, Le Kremlin-Bicêtre, France
| | - Charlotte Lombardi
- Institut de Biologie Structurale (IBS), University Grenoble-Alpes, CEA, CNRS, Bacterial Pathogenesis Group, Grenoble, France
| | - François Cretin
- University Grenoble-Alpes, Bacterial Pathogenesis and Cellular Responses, CNRS-ERL5261, U1036_S, INSERM, Biosciences and Biotechnology Institute of Grenoble, CEA-Grenoble, Grenoble, France
| | - Andréa Dessen
- Institut de Biologie Structurale (IBS), University Grenoble-Alpes, CEA, CNRS, Bacterial Pathogenesis Group, Grenoble, France.,Brazilian Biosciences National Laboratory (LNBio), CNPEM, São Paulo, Brazil
| | - Alain Filloux
- MRC Centre for Molecular Microbiology and Infection, Department of Life Sciences, Imperial College London, London, UK.
| |
Collapse
|
41
|
Garcia M, Morello E, Garnier J, Barrault C, Garnier M, Burucoa C, Lecron JC, Si-Tahar M, Bernard FX, Bodet C. Pseudomonas aeruginosa flagellum is critical for invasion, cutaneous persistence and induction of inflammatory response of skin epidermis. Virulence 2018; 9:1163-1175. [PMID: 30070169 PMCID: PMC6086312 DOI: 10.1080/21505594.2018.1480830] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 05/21/2018] [Indexed: 12/22/2022] Open
Abstract
Pseudomonas aeruginosa, an opportunistic pathogen involved in skin and lung diseases, possesses numerous virulence factors, including type 2 and 3 secretion systems (T2SS and T3SS) and its flagellum, whose functions remain poorly known during cutaneous infection. Using isogenic mutants deleted from genes encoding each or all of these three virulence factors, we investigated their role in induction of inflammatory response and in tissue invasiveness in human primary keratinocytes and reconstructed epidermis. Our results showed that flagellum, but not T2SS and T3SS, is involved in induction of a large panel of cytokine, chemokine, and antimicrobial peptide (AMP) mRNA in the infected keratinocytes. Chemokine secretion and AMP tissular production were also dependent on the presence of the bacterial flagellum. This pro-inflammatory effect was significantly reduced in keratinocytes infected in presence of anti-toll-like receptor 5 (TLR5) neutralizing antibody. Bacterial invasion of human epidermis and persistence in a mouse model of sub-cutaneous infection were dependent on the P. aeruginosa flagellum. We demonstrated that flagellum constitutes the main virulence factor of P. aeruginosa involved not only in early induction of the epidermis inflammatory response but also in bacterial invasion and cutaneous persistence. P. aeruginosa is mainly sensed by TLR5 during the early innate immune response of human primary keratinocytes.
Collapse
Affiliation(s)
- Magali Garcia
-
Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université de Poitiers
, Poitiers, France
-
Laboratoire de Virologie et Mycobactériologie, CHU de Poitiers
, Poitiers, France
| | - Eric Morello
-
Centre d'Etude des Pathologies Respiratoires, INSERM UMR 1100, Université de Tours
, Tours, France
| | | | | | - Martine Garnier
-
Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université de Poitiers
, Poitiers, France
| | - Christophe Burucoa
-
Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université de Poitiers
, Poitiers, France
-
Laboratoire de Bactériologie et Hygiène, CHU de Poitiers
, Poitiers, France
| | - Jean-Claude Lecron
-
Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université de Poitiers
, Poitiers, France
-
Laboratoire d’Immunologie et Inflammation, CHU de Poitiers
, Poitiers, France
| | - Mustapha Si-Tahar
-
Centre d'Etude des Pathologies Respiratoires, INSERM UMR 1100, Université de Tours
, Tours, France
| | | | - Charles Bodet
-
Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université de Poitiers
, Poitiers, France
| |
Collapse
|
42
|
Takata I, Yamagishi Y, Mikamo H. Association of the exoU genotype with a multidrug non-susceptible phenotype and mRNA expressions of resistance genes in Pseudomonas aeruginosa. J Infect Chemother 2017; 24:45-52. [PMID: 29107652 DOI: 10.1016/j.jiac.2017.08.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 08/10/2017] [Accepted: 08/31/2017] [Indexed: 12/28/2022]
Abstract
The increased prevalence of the virulence factor exoU + genotype among multidrug-resistant Pseudomonas aeruginosa has been previously reported. However, the genes that are related to the multidrug resistance of the exoU + genotype strain have not been analyzed and remain to be elucidated. The objective of this study was to analyze the correlations between virulence factors and resistance genes. The exoU + genotype was frequently found in carbapenem and fluoroquinolone non-susceptible strains. The imp carbapenemase genotype, the quinolone-resistance-determining region mutation in GyrA and ParC and the defective mutation in OprD were not frequently found in the exoU + genotype and carbapenem and fluoroquinolone non-susceptible strains. On the other hand, mexY and ampC mRNA overexpressing strains were more frequently found in the exoU + genotype and carbapenem and fluoroquinolone non-susceptible strains. Moreover, sequence type 235, a high risk clone of multidrug-resistant P. aeruginosa, was prevalent among the exoU + genotype and carbapenem and fluoroquinolone non-susceptible strains. ExoU is highly virulent protein, and the overexpression of efflux pumps and AmpC β-lactamase induce a multidrug-resistant phenotype. Therefore, the increased prevalence of P. aeruginosa strains with an exoU + genotype and the overexpression of efflux pumps and AmpC β-lactamase are likely to make P. aeruginosa infections difficult to treat. An understanding of the prevalence of both the exoU + genotype and the mRNA overexpression of resistance genes may help to select empirical therapy for the treatment of nosocomial infections caused by P. aeruginosa.
Collapse
Affiliation(s)
- Iichiro Takata
- Department of Clinical Infectious Diseases, Aichi Medical University Graduate School of Medicine, Aichi, Japan
| | - Yuka Yamagishi
- Department of Clinical Infectious Diseases, Aichi Medical University Graduate School of Medicine, Aichi, Japan
| | - Hiroshige Mikamo
- Department of Clinical Infectious Diseases, Aichi Medical University Graduate School of Medicine, Aichi, Japan.
| |
Collapse
|
43
|
Pseudomonas aeruginosa Magnesium Transporter MgtE Inhibits Type III Secretion System Gene Expression by Stimulating rsmYZ Transcription. J Bacteriol 2017; 199:JB.00268-17. [PMID: 28847924 DOI: 10.1128/jb.00268-17] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 08/23/2017] [Indexed: 11/20/2022] Open
Abstract
Pseudomonas aeruginosa causes numerous acute and chronic opportunistic infections in humans. One of its most formidable weapons is a type III secretion system (T3SS), which injects powerful toxins directly into host cells. The toxins lead to cell dysfunction and, ultimately, cell death. Identification of regulatory pathways that control T3SS gene expression may lead to the discovery of novel therapeutics to treat P. aeruginosa infections. In a previous study, we found that expression of the magnesium transporter gene mgtE inhibits T3SS gene transcription. MgtE-dependent inhibition appeared to interfere with the synthesis or function of the master T3SS transcriptional activator ExsA, although the exact mechanism was unclear. We now demonstrate that mgtE expression acts through the GacAS two-component system to activate rsmY and rsmZ transcription. This event ultimately leads to inhibition of exsA translation. This inhibitory effect is specific to exsA as translation of other genes in the exsCEBA operon is not inhibited by mgtE Moreover, our data reveal that MgtE acts solely through this pathway to regulate T3SS gene transcription. Our study reveals an important mechanism that may allow P. aeruginosa to fine-tune T3SS activity in response to certain environmental stimuli.IMPORTANCE The type III secretion system (T3SS) is a critical virulence factor utilized by numerous Gram-negative bacteria, including Pseudomonas aeruginosa, to intoxicate and kill host cells. Elucidating T3SS regulatory mechanisms may uncover targets for novel anti-P. aeruginosa therapeutics and provide deeper understanding of bacterial pathogenesis. We previously found that the magnesium transporter MgtE inhibits T3SS gene transcription in P. aeruginosa In this study, we describe the mechanism of MgtE-dependent inhibition of the T3SS. Our report also illustrates how MgtE might respond to environmental cues, such as magnesium levels, to fine-tune T3SS gene expression.
Collapse
|
44
|
Vourc'h M, Roquilly A, Broquet A, David G, Hulin P, Jacqueline C, Caillon J, Retiere C, Asehnoune K. Exoenzyme T Plays a Pivotal Role in the IFN-γ Production after Pseudomonas Challenge in IL-12 Primed Natural Killer Cells. Front Immunol 2017; 8:1283. [PMID: 29067027 PMCID: PMC5641345 DOI: 10.3389/fimmu.2017.01283] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 09/25/2017] [Indexed: 11/22/2022] Open
Abstract
Pseudomonas aeruginosa (PA) expresses the type III secretion system (T3SS) and effector exoenzymes that interfere with intracellular pathways. Natural killer (NK) cells play a key role in antibacterial immunity and their activation is highly dependent on IL-12 produced by myeloid cells. We studied PA and NK cell interactions and the role of IL-12 using human peripheral blood mononuclear cells, sorted human NK cells, and a human NK cell line (NK92). We used a wild-type (WT) strain of PA (PAO1) or isogenic PA-deleted strains to delineate the role of T3SS and exoenzymes. Our hypotheses were tested in vivo in a PA-pneumonia mouse model. Human NK cells or NK92 cell line produced low levels of IFN-γ in response to PA without IL-12 stimulation, whereas PA significantly increased IFN-γ after IL-12 priming. The modulation of IFN-γ production by PA required bacteria-to-cell contact. Among T3SS effectors, exoenzyme T (ExoT) upregulates IFN-γ production and control ERK activation. In vivo, ExoT also increases IFN-γ levels and the percentage of IFN-γ+ NK cells in lungs during PA pneumonia, confirming in vitro data. In conclusion, our results suggest that T3SS could modulate the production of IFN-γ by NK cells after PA infection through ERK activation.
Collapse
Affiliation(s)
- Mickael Vourc'h
- Laboratoire UPRES EA3826 «Thérapeutiques cliniques et expérimentales des infections», IRS2 - Nantes Biotech, Université de Nantes, Nantes, France.,Intensive Care Unit, Anesthesia and Critical Care Department, Hôtel Dieu, University Hospital of Nantes, Nantes, France
| | - Antoine Roquilly
- Laboratoire UPRES EA3826 «Thérapeutiques cliniques et expérimentales des infections», IRS2 - Nantes Biotech, Université de Nantes, Nantes, France.,Intensive Care Unit, Anesthesia and Critical Care Department, Hôtel Dieu, University Hospital of Nantes, Nantes, France
| | - Alexis Broquet
- Laboratoire UPRES EA3826 «Thérapeutiques cliniques et expérimentales des infections», IRS2 - Nantes Biotech, Université de Nantes, Nantes, France
| | - Gaelle David
- Etablissement Français du Sang, Nantes, France.,CRCINA, INSERM U1232, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Philippe Hulin
- MicroPICell, Cell and Tissue Imaging Core, UMS Inserm 016/CNRS 3356/FED 4203, Villejuif, France
| | - Cedric Jacqueline
- Laboratoire UPRES EA3826 «Thérapeutiques cliniques et expérimentales des infections», IRS2 - Nantes Biotech, Université de Nantes, Nantes, France
| | - Jocelyne Caillon
- Laboratoire UPRES EA3826 «Thérapeutiques cliniques et expérimentales des infections», IRS2 - Nantes Biotech, Université de Nantes, Nantes, France
| | - Christelle Retiere
- Etablissement Français du Sang, Nantes, France.,CRCINA, INSERM U1232, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Karim Asehnoune
- Laboratoire UPRES EA3826 «Thérapeutiques cliniques et expérimentales des infections», IRS2 - Nantes Biotech, Université de Nantes, Nantes, France.,Intensive Care Unit, Anesthesia and Critical Care Department, Hôtel Dieu, University Hospital of Nantes, Nantes, France
| |
Collapse
|
45
|
Cole BJ, Feltcher ME, Waters RJ, Wetmore KM, Mucyn TS, Ryan EM, Wang G, Ul-Hasan S, McDonald M, Yoshikuni Y, Malmstrom RR, Deutschbauer AM, Dangl JL, Visel A. Genome-wide identification of bacterial plant colonization genes. PLoS Biol 2017; 15:e2002860. [PMID: 28938018 PMCID: PMC5627942 DOI: 10.1371/journal.pbio.2002860] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 10/04/2017] [Accepted: 09/05/2017] [Indexed: 01/09/2023] Open
Abstract
Diverse soil-resident bacteria can contribute to plant growth and health, but the molecular mechanisms enabling them to effectively colonize their plant hosts remain poorly understood. We used randomly barcoded transposon mutagenesis sequencing (RB-TnSeq) in Pseudomonas simiae, a model root-colonizing bacterium, to establish a genome-wide map of bacterial genes required for colonization of the Arabidopsis thaliana root system. We identified 115 genes (2% of all P. simiae genes) with functions that are required for maximal competitive colonization of the root system. Among the genes we identified were some with obvious colonization-related roles in motility and carbon metabolism, as well as 44 other genes that had no or vague functional predictions. Independent validation assays of individual genes confirmed colonization functions for 20 of 22 (91%) cases tested. To further characterize genes identified by our screen, we compared the functional contributions of P. simiae genes to growth in 90 distinct in vitro conditions by RB-TnSeq, highlighting specific metabolic functions associated with root colonization genes. Our analysis of bacterial genes by sequence-driven saturation mutagenesis revealed a genome-wide map of the genetic determinants of plant root colonization and offers a starting point for targeted improvement of the colonization capabilities of plant-beneficial microbes. Plants fix carbon to create an abundance of sugars and amino acids, thus providing an enticing environment for microorganisms that reside in soil. Once these microorganisms have colonized the root environment, they can dramatically influence plant growth and development. We set out to identify a comprehensive set of microbial genes that control or influence root colonization, using a genome-wide transposon mutagenesis approach (randomly barcoded transposon sequencing [RB-TnSeq]). By using this method, we identified several hundred genes that, when mutated, affect the ability of the bacterium P. simiae to competitively colonize the root system of the model plant A. thaliana. These included many genes purported to be involved in carbohydrate metabolism, cell wall biosynthesis, and motility, underscoring the notion that sugar metabolism, defense, and motility are all key features of a root-colonizing microbe. We also identified several amino acid transport and metabolism genes with mutations that confer a fitness advantage in root colonization. Lastly, we identified several genes with no known function that significantly alter root colonization ability when mutated. These findings suggest novel engineering strategies to improve biological product development, and will facilitate the mechanistic exploration of the root colonization process.
Collapse
Affiliation(s)
- Benjamin J. Cole
- US Department of Energy Joint Genome Institute, Walnut Creek, California, United States of America
| | - Meghan E. Feltcher
- Department of Biology, Department of Microbiology and Immunology, Curriculum in Genetics and Molecular Biology, Howard Hughes Medical Institute, Carolina Center for Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Robert J. Waters
- US Department of Energy Joint Genome Institute, Walnut Creek, California, United States of America
| | - Kelly M. Wetmore
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Tatiana S. Mucyn
- Department of Biology, Department of Microbiology and Immunology, Curriculum in Genetics and Molecular Biology, Howard Hughes Medical Institute, Carolina Center for Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Elizabeth M. Ryan
- US Department of Energy Joint Genome Institute, Walnut Creek, California, United States of America
| | - Gaoyan Wang
- US Department of Energy Joint Genome Institute, Walnut Creek, California, United States of America
| | - Sabah Ul-Hasan
- US Department of Energy Joint Genome Institute, Walnut Creek, California, United States of America
- School of Natural Sciences, University of California Merced, Merced, California, United States of America
| | - Meredith McDonald
- Department of Biology, Department of Microbiology and Immunology, Curriculum in Genetics and Molecular Biology, Howard Hughes Medical Institute, Carolina Center for Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Yasuo Yoshikuni
- US Department of Energy Joint Genome Institute, Walnut Creek, California, United States of America
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Rex R. Malmstrom
- US Department of Energy Joint Genome Institute, Walnut Creek, California, United States of America
| | - Adam M. Deutschbauer
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Jeffery L. Dangl
- Department of Biology, Department of Microbiology and Immunology, Curriculum in Genetics and Molecular Biology, Howard Hughes Medical Institute, Carolina Center for Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- * E-mail: (JLD); (AV)
| | - Axel Visel
- US Department of Energy Joint Genome Institute, Walnut Creek, California, United States of America
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
- School of Natural Sciences, University of California Merced, Merced, California, United States of America
- * E-mail: (JLD); (AV)
| |
Collapse
|
46
|
Bacterial Nucleotidyl Cyclase Inhibits the Host Innate Immune Response by Suppressing TAK1 Activation. Infect Immun 2017; 85:IAI.00239-17. [PMID: 28652310 DOI: 10.1128/iai.00239-17] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 06/13/2017] [Indexed: 12/22/2022] Open
Abstract
Exoenzyme Y (ExoY) is a type III secretion system effector found in 90% of the Pseudomonas aeruginosa isolates. Although it is known that ExoY is a soluble nucleotidyl cyclase that increases the cytoplasmic levels of nucleoside 3',5'-cyclic monophosphates (cNMPs) to mediate endothelial Tau phosphorylation and permeability, its functional role in the innate immune response is still poorly understood. Transforming growth factor β-activated kinase 1 (TAK1) is critical for mediating Toll-like receptor (TLR) signaling and subsequent activation of NF-κB and AP-1, which are transcriptional activators of innate immunity. Here, we report that ExoY inhibits proinflammatory cytokine production through suppressing the activation of TAK1 as well as downstream NF-κB and mitogen-activated protein (MAP) kinases. Mice infected with ExoY-deficient P. aeruginosa had higher levels of tumor necrosis factor (TNF) and interleukin-6 (IL-6), more neutrophil recruitment, and a lower bacterial load in lung tissue than mice infected with wild-type P. aeruginosa Taken together, our findings identify a previously unknown mechanism by which P. aeruginosa ExoY inhibits the host innate immune response.
Collapse
|
47
|
Pseudomonas aeruginosa Exolysin promotes bacterial growth in lungs, alveolar damage and bacterial dissemination. Sci Rep 2017; 7:2120. [PMID: 28522850 PMCID: PMC5437091 DOI: 10.1038/s41598-017-02349-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 04/10/2017] [Indexed: 11/18/2022] Open
Abstract
Exolysin (ExlA) is a recently-identified pore-forming toxin secreted by a subset of Pseudomonas aeruginosa strains identified worldwide and devoid of Type III secretion system (T3SS), a major virulence factor. Here, we characterized at the ultrastructural level the lesions caused by an ExlA-secreting strain, CLJ1, in mouse infected lungs. CLJ1 induced necrotic lesions in pneumocytes and endothelial cells, resulting in alveolo-vascular barrier breakdown. Ectopic expression of ExlA in an exlA-negative strain induced similar tissue injuries. In addition, ExlA conferred on bacteria the capacity to proliferate in lungs and to disseminate in secondary organs, similar to bacteria possessing a functional T3SS. CLJ1 did not promote a strong neutrophil infiltration in the alveoli, owing to the weak pro-inflammatory cytokine reaction engendered by the strain. However, CLJ1 was rapidly eliminated from the blood in a bacteremia model, suggesting that it can be promptly phagocytosed by immune cells. Together, our study ascribes to ExlA-secreting bacteria the capacity to proliferate in the lung and to damage pulmonary tissues, thereby promoting metastatic infections, in absence of substantial immune response exacerbation.
Collapse
|
48
|
Thanabalasuriar A, Surewaard BG, Willson ME, Neupane AS, Stover CK, Warrener P, Wilson G, Keller AE, Sellman BR, DiGiandomenico A, Kubes P. Bispecific antibody targets multiple Pseudomonas aeruginosa evasion mechanisms in the lung vasculature. J Clin Invest 2017; 127:2249-2261. [PMID: 28463232 DOI: 10.1172/jci89652] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 03/02/2017] [Indexed: 12/19/2022] Open
Abstract
Pseudomonas aeruginosa is a major cause of severe infections that lead to bacteremia and high patient mortality. P. aeruginosa has evolved numerous evasion and subversion mechanisms that work in concert to overcome immune recognition and effector functions in hospitalized and immunosuppressed individuals. Here, we have used multilaser spinning-disk intravital microscopy to monitor the blood-borne stage in a murine bacteremic model of P. aeruginosa infection. P. aeruginosa adhered avidly to lung vasculature, where patrolling neutrophils and other immune cells were virtually blind to the pathogen's presence. This cloaking phenomenon was attributed to expression of Psl exopolysaccharide. Although an anti-Psl mAb activated complement and enhanced neutrophil recognition of P. aeruginosa, neutrophil-mediated clearance of the pathogen was suboptimal owing to a second subversion mechanism, namely the type 3 secretion (T3S) injectisome. Indeed, T3S prevented phagosome acidification and resisted killing inside these compartments. Antibody-mediated inhibition of the T3S protein PcrV did not enhance bacterial phagocytosis but did enhance killing of the few bacteria ingested by neutrophils. A bispecific mAb targeting both Psl and PcrV enhanced neutrophil uptake of P. aeruginosa and also greatly increased inhibition of T3S function, allowing for phagosome acidification and bacterial killing. These data highlight the need to block multiple evasion and subversion mechanisms in tandem to kill P. aeruginosa.
Collapse
Affiliation(s)
- Ajitha Thanabalasuriar
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Bas Gj Surewaard
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Michelle E Willson
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Arpan S Neupane
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | | | | | | | | | | | | | - Paul Kubes
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
49
|
Pseudomonas aeruginosa Pore-Forming Exolysin and Type IV Pili Cooperate To Induce Host Cell Lysis. mBio 2017; 8:mBio.02250-16. [PMID: 28119472 PMCID: PMC5263249 DOI: 10.1128/mbio.02250-16] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Clinical strains of Pseudomonas aeruginosa lacking the type III secretion system genes employ a toxin, exolysin (ExlA), for host cell membrane disruption. Here, we demonstrated that ExlA export requires a predicted outer membrane protein, ExlB, showing that ExlA and ExlB define a new active two-partner secretion (TPS) system of P. aeruginosa In addition to the TPS signals, ExlA harbors several distinct domains, which include one hemagglutinin domain, five arginine-glycine-aspartic acid (RGD) motifs, and a C-terminal region lacking any identifiable sequence motifs. However, this C-terminal region is important for the toxic activity, since its deletion abolishes host cell lysis. Using lipid vesicles and eukaryotic cells, including red blood cells, we demonstrated that ExlA has a pore-forming activity which precedes cell membrane disruption of nucleated cells. Finally, we developed a high-throughput cell-based live-dead assay and used it to screen a transposon mutant library of an ExlA-producing P. aeruginosa clinical strain for bacterial factors required for ExlA-mediated toxicity. The screen resulted in the identification of proteins involved in the formation of type IV pili as being required for ExlA to exert its cytotoxic activity by promoting close contact between bacteria and the host cell. These findings represent the first example of cooperation between a pore-forming toxin of the TPS family and surface appendages in host cell intoxication. IMPORTANCE The course and outcome of acute, toxigenic infections by Pseudomonas aeruginosa clinical isolates rely on the deployment of one of two virulence strategies: delivery of effectors by the well-known type III secretion system or the cytolytic activity of the recently identified two-partner secreted toxin, exolysin. Here, we characterize several features of the mammalian cell intoxication process mediated by exolysin. We found that exolysin requires the outer membrane protein ExlB for export into extracellular medium. Using in vitro recombinant protein and ex vivo assays, we demonstrated a pore-forming activity of exolysin. A cellular cytotoxicity screen of a transposon mutant library, made in an exolysin-producing clinical strain, identified type IV pili as bacterial appendages required for exolysin toxic function. This work deciphers molecular mechanisms underlying the activity of novel virulence factors used by P. aeruginosa clinical strains lacking the type III secretion system, including a requirement for the toxin-producing bacteria to be attached to the targeted cell to induce cytolysis, and defines new targets for developing antivirulence strategies.
Collapse
|
50
|
Kaiser SJ, Mutters NT, DeRosa A, Ewers C, Frank U, Günther F. Determinants for persistence of Pseudomonas aeruginosa in hospitals: interplay between resistance, virulence and biofilm formation. Eur J Clin Microbiol Infect Dis 2016; 36:243-253. [PMID: 27734161 DOI: 10.1007/s10096-016-2792-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 09/14/2016] [Indexed: 12/27/2022]
Abstract
Pseudomonas aeruginosa (Pa) is one of the major bacterial pathogens causing nosocomial infections. During the past few decades, multidrug-resistant (MDR) and extensively drug-resistant (XDR) lineages of Pa have emerged in hospital settings with increasing numbers. However, it remains unclear which determinants of Pa facilitated this spread. A total of 211 clinical XDR and 38 susceptible clinical Pa isolates (nonXDR), as well as 47 environmental isolates (EI), were collected at the Heidelberg University Hospital. We used RAPD PCR to identify genetic clusters. Carriage of carbapenamases (CPM) and virulence genes were analyzed by PCR, biofilm formation capacity was assessed, in vitro fitness was evaluated using competitive growth assays, and interaction with the host's immune system was analyzed using serum killing and neutrophil killing assays. XDR isolates showed significantly elevated biofilm formation (p < 0.05) and higher competitive fitness compared to nonXDR and EI isolates. Thirty percent (62/205) of the XDR isolates carried a CPM. Similarities in distribution of virulence factors, as well as biofilm formation properties, between CPM+ Pa isolates and EI and between CPM- and nonXDR isolates were detected. Molecular typing revealed two distinct genetic clusters within the XDR population, which were characterized by even higher biofilm formation. In contrast, XDR isolates were more susceptible to the immune response than nonXDR isolates. Our study provides evidence that the ability to form biofilms is an outstanding determinant for persistence and endemic spread of Pa in the hospital setting.
Collapse
Affiliation(s)
- S J Kaiser
- Department of Infectious Diseases, Heidelberg University Hospital, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany
| | - N T Mutters
- Department of Infectious Diseases, Heidelberg University Hospital, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany
| | - A DeRosa
- Department of Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - C Ewers
- Institute of Hygiene and Infectious Diseases of Animals, Justus Liebig University Giessen, Giessen, Germany
| | - U Frank
- Department of Infectious Diseases, Heidelberg University Hospital, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany
| | - F Günther
- Department of Infectious Diseases, Heidelberg University Hospital, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany.
| |
Collapse
|