1
|
Tribin FE, Lieux C, Maestre-Mesa J, Durkee H, Krishna K, Chou B, Neag E, Tóthová JD, Martinez JD, Flynn HW, Parel JM, Miller D, Amescua G. Clinical Features and Treatment Outcomes of Carbapenem-Resistant Pseudomonas aeruginosa Keratitis. JAMA Ophthalmol 2024; 142:407-415. [PMID: 38512246 PMCID: PMC10958388 DOI: 10.1001/jamaophthalmol.2024.0259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 12/27/2023] [Indexed: 03/22/2024]
Abstract
Importance Evaluation of the microbiological diagnostic profile of multidrug-resistant Pseudomonas aeruginosa keratitis and potential management with rose bengal-photodynamic antimicrobial therapy (RB-PDAT) is important. Objective To document the disease progression of carbapenemase-resistant P aeruginosa keratitis after an artificial tear contamination outbreak. Design, Setting, and Participants This retrospective observation case series included 9 patients 40 years or older who presented at Bascom Palmer Eye Institute and had positive test results for multidrug-resistant P aeruginosa keratitis between January 1, 2022, and October 31, 2023. Main Outcomes and Measures Evaluation of type III secretion phenotype, carbapenemase-resistance genes blaGES and blaVIM susceptibility to antibiotics, and in vitro and in vivo outcomes of RB-PDAT against multidrug-resistant P aeruginosa keratitis. Results Among the 9 patients included in the analysis (5 women and 4 men; mean [SD] age, 73.4 [14.0] years), all samples tested positive for exoU and carbapenemase-resistant blaVIM and blaGES genes. Additionally, isolates were resistant to carbapenems as indicated by minimum inhibitory concentration testing. In vitro efficacy of RB-PDAT indicated its potential application for treating recalcitrant cases. These cases highlight the rapid progression and challenging management of multidrug-resistant P aeruginosa. Two patients were treated with RB-PDAT as an adjuvant to antibiotic therapy and had improved visual outcomes. Conclusions and Relevance This case series highlights the concerning progression in resistance and virulence of P aeruginosa and emphasizes the need to explore alternative therapies like RB-PDAT that have broad coverage and no known antibiotic resistance. The findings support further investigation into the potential effects of RB-PDAT for other multidrug-resistant microbes.
Collapse
Affiliation(s)
- Felipe Echeverri Tribin
- Ophthalmic Biophysics Center, Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, Florida
| | - Caroline Lieux
- Anne Bates Leach Eye Center, Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, Florida
| | - Jorge Maestre-Mesa
- Ocular Microbiology Laboratory, Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, Florida
| | - Heather Durkee
- Ophthalmic Biophysics Center, Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, Florida
| | - Katherine Krishna
- Ophthalmic Biophysics Center, Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, Florida
| | - Brandon Chou
- Ophthalmic Biophysics Center, Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, Florida
| | - Emily Neag
- Anne Bates Leach Eye Center, Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, Florida
| | - Jana D’Amato Tóthová
- Ophthalmic Biophysics Center, Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, Florida
| | - Jaime D. Martinez
- Ophthalmic Biophysics Center, Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, Florida
| | - Harry W. Flynn
- Anne Bates Leach Eye Center, Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, Florida
- Ocular Microbiology Laboratory, Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, Florida
| | - Jean Marie Parel
- Ophthalmic Biophysics Center, Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, Florida
- Anne Bates Leach Eye Center, Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, Florida
- Ocular Microbiology Laboratory, Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, Florida
| | - Darlene Miller
- Ophthalmic Biophysics Center, Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, Florida
- Anne Bates Leach Eye Center, Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, Florida
- Ocular Microbiology Laboratory, Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, Florida
| | - Guillermo Amescua
- Ophthalmic Biophysics Center, Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, Florida
- Anne Bates Leach Eye Center, Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, Florida
- Ocular Microbiology Laboratory, Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
2
|
Zelikman S, Dudkevich R, Korenfeld-Tzemach H, Shmidov E, Levi-Ferber M, Shoshani S, Ben-Aroya S, Henis-Korenblit S, Banin E. PemB, a type III secretion effector in Pseudomonas aeruginosa, affects Caenorhabditis elegans life span. Heliyon 2024; 10:e29751. [PMID: 38681583 PMCID: PMC11053225 DOI: 10.1016/j.heliyon.2024.e29751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 04/08/2024] [Accepted: 04/15/2024] [Indexed: 05/01/2024] Open
Abstract
Pseudomonas aeruginosa is one of the leading nosocomial opportunistic pathogens causing acute and chronic infections. Among its main virulent factors is the Type III secretion system (T3SS) which enhances disease severity by delivering effectors to the host in a highly regulated manner. Despite its importance for virulence, only six T3SS-dependent effectors have been discovered so far. Previously, we identified two new potential effectors using a machine-learning algorithm approach. Here we demonstrate that one of these effectors, PemB, is indeed virulent. Using a live Caenorhabditis elegans infection model, we demonstrate this effector damages the integrity of the intestine barrier leading to the death of the host. Implementing a high-throughput assay using Saccharomyces cerevisiae, we identified several candidate proteins that interact with PemB. One of them, EFT1, has an ortholog in C. elegans (eef-2) and is also an essential gene and a well-known target utilized by different pathogens to induce toxicity to the worm. Accordingly, we found that by silencing the eef-2 gene in C. elegans, PemB could no longer induce its toxic effect. The current study further uncovers the complex machinery assisting P. aeruginosa virulence and may provide novel insight how to manage infection associated with this hard-to-treat pathogen.
Collapse
Affiliation(s)
- Shira Zelikman
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Max and Anna Webb, 5290002, Ramat Gan, Israel
- The Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb, 5290002, Ramat Gan, Israel
| | - Reut Dudkevich
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Max and Anna Webb, 5290002, Ramat Gan, Israel
| | - Hadar Korenfeld-Tzemach
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Max and Anna Webb, 5290002, Ramat Gan, Israel
- The Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb, 5290002, Ramat Gan, Israel
| | - Esther Shmidov
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Max and Anna Webb, 5290002, Ramat Gan, Israel
- The Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb, 5290002, Ramat Gan, Israel
| | - Mor Levi-Ferber
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Max and Anna Webb, 5290002, Ramat Gan, Israel
| | - Sivan Shoshani
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Max and Anna Webb, 5290002, Ramat Gan, Israel
- The Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb, 5290002, Ramat Gan, Israel
| | - Shay Ben-Aroya
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Max and Anna Webb, 5290002, Ramat Gan, Israel
- The Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb, 5290002, Ramat Gan, Israel
| | - Sivan Henis-Korenblit
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Max and Anna Webb, 5290002, Ramat Gan, Israel
| | - Ehud Banin
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Max and Anna Webb, 5290002, Ramat Gan, Israel
- The Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb, 5290002, Ramat Gan, Israel
| |
Collapse
|
3
|
Touré H, Herrmann JL, Szuplewski S, Girard-Misguich F. Drosophila melanogaster as an organism model for studying cystic fibrosis and its major associated microbial infections. Infect Immun 2023; 91:e0024023. [PMID: 37847031 PMCID: PMC10652941 DOI: 10.1128/iai.00240-23] [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] [Indexed: 10/18/2023] Open
Abstract
Cystic fibrosis (CF) is a human genetic disease caused by mutations in the cystic fibrosis transmembrane conductance regulator gene that encodes a chloride channel. The most severe clinical manifestation is associated with chronic pulmonary infections by pathogenic and opportunistic microbes. Drosophila melanogaster has become the invertebrate model of choice for modeling microbial infections and studying the induced innate immune response. Here, we review its contribution to the understanding of infections with six major pathogens associated with CF (Staphylococcus aureus, Pseudomonas aeruginosa, Burkholderia cepacia, Mycobacterium abscessus, Streptococcus pneumoniae, and Aspergillus fumigatus) together with the perspectives opened by the recent availability of two CF models in this model organism.
Collapse
Affiliation(s)
- Hamadoun Touré
- Université Paris-Saclay, UVSQ, INSERM, Infection et Inflammation, Montigny-le-Bretonneux, France
| | - Jean-Louis Herrmann
- Université Paris-Saclay, UVSQ, INSERM, Infection et Inflammation, Montigny-le-Bretonneux, France
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Ile-de-France Ouest, GHU Paris-Saclay, Hôpital Raymond Poincaré, Garches, France
| | - Sébastien Szuplewski
- Université Paris-Saclay, UVSQ, Laboratoire de Génétique et Biologie Cellulaire, Montigny-le-Bretonneux, France
| | - Fabienne Girard-Misguich
- Université Paris-Saclay, UVSQ, INSERM, Infection et Inflammation, Montigny-le-Bretonneux, France
| |
Collapse
|
4
|
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: 6] [Impact Index Per Article: 3.0] [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
|
5
|
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. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.13.528260. [PMID: 36824932 PMCID: PMC9948977 DOI: 10.1101/2023.02.13.528260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
Pathogenesis of Pseudomonas aeruginosa infections can include bacterial survival inside epithelial cells. Previously, we showed 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 activation of NF-κB as measured by p65 relocalization and TNFα 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 TSSS 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.
Collapse
Affiliation(s)
- Abby R Kroken
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL USA
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, CA USA
| | - Keith A Klein
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL USA
| | | | - Vincent Nieto
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, CA USA
| | - Eric J Jedel
- 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 Sciences, Microbiology, and Infectious Diseases & Immunity, University of California, Berkeley, CA USA
| |
Collapse
|
6
|
Wood SJ, Goldufsky JW, Seu MY, Dorafshar AH, Shafikhani SH. Pseudomonas aeruginosa Cytotoxins: Mechanisms of Cytotoxicity and Impact on Inflammatory Responses. Cells 2023; 12:cells12010195. [PMID: 36611990 PMCID: PMC9818787 DOI: 10.3390/cells12010195] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/01/2022] [Accepted: 12/01/2022] [Indexed: 01/05/2023] Open
Abstract
Pseudomonas aeruginosa is one of the most virulent opportunistic Gram-negative bacterial pathogens in humans. It causes many acute and chronic infections with morbidity and mortality rates as high as 40%. P. aeruginosa owes its pathogenic versatility to a large arsenal of cell-associated and secreted virulence factors which enable this pathogen to colonize various niches within hosts and protect it from host innate immune defenses. Induction of cytotoxicity in target host cells is a major virulence strategy for P. aeruginosa during the course of infection. P. aeruginosa has invested heavily in this strategy, as manifested by a plethora of cytotoxins that can induce various forms of cell death in target host cells. In this review, we provide an in-depth review of P. aeruginosa cytotoxins based on their mechanisms of cytotoxicity and the possible consequences of their cytotoxicity on host immune responses.
Collapse
Affiliation(s)
- Stephen J. Wood
- Department of Medicine, Division of Hematology, Oncology and Cell Therapy, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Rush University Medical Center, Chicago, IL 60612, USA
| | - Josef W. Goldufsky
- Department of Medicine, Division of Hematology, Oncology and Cell Therapy, Rush University Medical Center, Chicago, IL 60612, USA
| | - Michelle Y. Seu
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Rush University Medical Center, Chicago, IL 60612, USA
| | - Amir H. Dorafshar
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Rush University Medical Center, Chicago, IL 60612, USA
| | - Sasha H. Shafikhani
- Department of Medicine, Division of Hematology, Oncology and Cell Therapy, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL 60612, USA
- Cancer Center, Rush University Medical Center, Chicago, IL 60612, USA
- Correspondence:
| |
Collapse
|
7
|
Qin S, Xiao W, Zhou C, Pu Q, Deng X, Lan L, Liang H, Song X, Wu M. Pseudomonas aeruginosa: pathogenesis, virulence factors, antibiotic resistance, interaction with host, technology advances and emerging therapeutics. Signal Transduct Target Ther 2022; 7:199. [PMID: 35752612 PMCID: PMC9233671 DOI: 10.1038/s41392-022-01056-1] [Citation(s) in RCA: 368] [Impact Index Per Article: 122.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 06/04/2022] [Accepted: 06/08/2022] [Indexed: 02/05/2023] Open
Abstract
Pseudomonas aeruginosa (P. aeruginosa) is a Gram-negative opportunistic pathogen that infects patients with cystic fibrosis, burn wounds, immunodeficiency, chronic obstructive pulmonary disorder (COPD), cancer, and severe infection requiring ventilation, such as COVID-19. P. aeruginosa is also a widely-used model bacterium for all biological areas. In addition to continued, intense efforts in understanding bacterial pathogenesis of P. aeruginosa including virulence factors (LPS, quorum sensing, two-component systems, 6 type secretion systems, outer membrane vesicles (OMVs), CRISPR-Cas and their regulation), rapid progress has been made in further studying host-pathogen interaction, particularly host immune networks involving autophagy, inflammasome, non-coding RNAs, cGAS, etc. Furthermore, numerous technologic advances, such as bioinformatics, metabolomics, scRNA-seq, nanoparticles, drug screening, and phage therapy, have been used to improve our understanding of P. aeruginosa pathogenesis and host defense. Nevertheless, much remains to be uncovered about interactions between P. aeruginosa and host immune responses, including mechanisms of drug resistance by known or unannotated bacterial virulence factors as well as mammalian cell signaling pathways. The widespread use of antibiotics and the slow development of effective antimicrobials present daunting challenges and necessitate new theoretical and practical platforms to screen and develop mechanism-tested novel drugs to treat intractable infections, especially those caused by multi-drug resistance strains. Benefited from has advancing in research tools and technology, dissecting this pathogen's feature has entered into molecular and mechanistic details as well as dynamic and holistic views. Herein, we comprehensively review the progress and discuss the current status of P. aeruginosa biophysical traits, behaviors, virulence factors, invasive regulators, and host defense patterns against its infection, which point out new directions for future investigation and add to the design of novel and/or alternative therapeutics to combat this clinically significant pathogen.
Collapse
Affiliation(s)
- Shugang Qin
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Wen Xiao
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Chuanmin Zhou
- State Key Laboratory of Virology, School of Public Health, Wuhan University, Wuhan, 430071, P.R. China
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58203, USA
| | - Qinqin Pu
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58203, USA
| | - Xin Deng
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, People's Republic of China
| | - Lefu Lan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Haihua Liang
- College of Life Sciences, Northwest University, Xi'an, ShaanXi, 710069, China
| | - Xiangrong Song
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Min Wu
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58203, USA.
| |
Collapse
|
8
|
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: 7] [Impact Index Per Article: 2.3] [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
|
9
|
Fujiki Y, Mato N, Watanabe S, Shibano T, Tonai K, Takahashi K, Saito T, Okuyama A, Takigami A, Bando M, Suzuki T, Cui L, Hagiwara K. Virulent Pseudomonas aeruginosa pneumonia in an immunocompetent adult associated with a home whirlpool bath: A case report. Respir Med Case Rep 2022; 38:101673. [PMID: 35694132 PMCID: PMC9178473 DOI: 10.1016/j.rmcr.2022.101673] [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: 01/31/2022] [Revised: 04/05/2022] [Accepted: 05/17/2022] [Indexed: 11/29/2022] Open
Abstract
We present a case of life-threatening pneumonia caused by Pseudomonas aeruginosa (P. aeruginosa) in a healthy 67-year-old man. Rapid disseminated infection resulted in the right hemorrhagic pneumonia and bacteremia. Antimicrobial therapy had limited effects, radical pneumonectomy eventually resolved the prolonged infection. Concurrently, we explored the environmental factors responsible for fulminant P. aeruginosa infection. Multi-locus sequence typing demonstrated that P. aeruginosa isolated from the patient was identical to that collected from home whirlpool bath by the common virulent factor gene. Massive inhalation of contaminated aerosol and pathogen virulence may have synergistically contributed to the severity in this case.
Collapse
Affiliation(s)
- Yasumaro Fujiki
- Division of Pulmonary Medicine, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Naoko Mato
- Division of Pulmonary Medicine, Department of Medicine, Jichi Medical University, Tochigi, Japan
- Corresponding author. 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan.
| | - Shinya Watanabe
- Division of Bacteriology, School of Medicine, Jichi Medical University, Tochigi, Japan
| | - Tomoki Shibano
- Division of General Thoracic Surgery, Department of Surgery, Jichi Medical University, Tochigi, Japan
| | - Ken Tonai
- Division of Intensive Care, Department of Anesthesiology & Intensive Care Medicine, Jichi Medical University, Tochigi, Japan
| | - Kento Takahashi
- Division of Pulmonary Medicine, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Tatsuya Saito
- Division of Pulmonary Medicine, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Akiko Okuyama
- Division of Pulmonary Medicine, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Ayako Takigami
- Division of Pulmonary Medicine, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Masashi Bando
- Division of Pulmonary Medicine, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Takuji Suzuki
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Longzhu Cui
- Division of Bacteriology, School of Medicine, Jichi Medical University, Tochigi, Japan
| | - Koichi Hagiwara
- Division of Pulmonary Medicine, Department of Medicine, Jichi Medical University, Tochigi, Japan
| |
Collapse
|
10
|
Pseudomonas aeruginosa adaptation and evolution in patients with cystic fibrosis. Nat Rev Microbiol 2021; 19:331-342. [PMID: 33214718 DOI: 10.1038/s41579-020-00477-5] [Citation(s) in RCA: 228] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2020] [Indexed: 01/29/2023]
Abstract
Intense genome sequencing of Pseudomonas aeruginosa isolates from cystic fibrosis (CF) airways has shown inefficient eradication of the infecting bacteria, as well as previously undocumented patient-to-patient transmission of adapted clones. However, genome sequencing has limited potential as a predictor of chronic infection and of the adaptive state during infection, and thus there is increasing interest in linking phenotypic traits to the genome sequences. Phenotypic information ranges from genome-wide transcriptomic analysis of patient samples to determination of more specific traits associated with metabolic changes, stress responses, antibiotic resistance and tolerance, biofilm formation and slow growth. Environmental conditions in the CF lung shape both genetic and phenotypic changes of P. aeruginosa during infection. In this Review, we discuss the adaptive and evolutionary trajectories that lead to early diversification and late convergence, which enable P. aeruginosa to succeed in this niche, and we point out how knowledge of these biological features may be used to guide diagnosis and therapy.
Collapse
|
11
|
Horna G, Ruiz J. Type 3 secretion system of Pseudomonas aeruginosa. Microbiol Res 2021; 246:126719. [PMID: 33582609 DOI: 10.1016/j.micres.2021.126719] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 01/19/2021] [Accepted: 01/26/2021] [Indexed: 12/27/2022]
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen, mainly affecting severe patients, such as those in intensive care units (ICUs). High levels of antibiotic resistance and a long battery of virulence factors characterise this pathogen. Among virulence factors, the T3SS (Type 3 Secretion Systems) are especially relevant, being one of the most important virulence factors in P. aeruginosa. T3SS are a complex "molecular syringe" able to inject different effectors in host cells, subverting cell machinery influencing immune responses, and increasing bacterial survival rates. While T3SS have been largely studied and the molecular structure and main effector functions have been established, a series of questions and further points remain to be clarified or established. The key role of T3SS in P. aeruginosa virulence has resulted in the search for T3SS-targeting molecules able to impair their functions and subsequently improve patient outcomes. This review aims to summarise the most relevant features of the P. aeruginosa T3SS.
Collapse
Affiliation(s)
- Gertrudis Horna
- Universidad Catolica Los Angeles de Chimbote, Instituto de Investigación, Chimbote, Peru.
| | - Joaquim Ruiz
- Laboratorio de Microbiología Molecular y Genómica Bacteriana, Universidad Científica del Sur, Panamericana Sur, Km 19, Lima, Peru.
| |
Collapse
|
12
|
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: 9] [Impact Index Per Article: 2.3] [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
|
13
|
Kennedy SM, Deshpande P, Gallagher AG, Horsburgh MJ, Allison HE, Kaye SB, Wellings DA, Williams RL. Antimicrobial Activity of Poly-epsilon-lysine Peptide Hydrogels Against Pseudomonas aeruginosa. Invest Ophthalmol Vis Sci 2021; 61:18. [PMID: 32776141 PMCID: PMC7441358 DOI: 10.1167/iovs.61.10.18] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Purpose To determine the antimicrobial activity of poly-epsilon-lysine (pɛK) functionalization of hydrogels against Pseudomonas aeruginosa. Methods Antimicrobial activities of pɛK and pɛK+ hydrogels were tested against both keratitis and a laboratory strain of Paeruginosa at a range of inocula sizes, over 4 and 24 hours. The number of viable CFU on pɛK and pɛK+ hydrogels or commercial contact lenses (CL) was investigated. Ex vivo porcine corneas were inoculated with Paeruginosa PAO1 (103 CFU) and incubated with pɛK+ hydrogels or commercial hydrogel CL for 24 hours and the effects of infection determined. Results PɛK+ hydrogels showed log reductions in viable CFU compared with pɛK hydrogels for all Paeruginosa strains, depending on inocula sizes and incubation time. After 24 hours pɛK+ hydrogels showed >5 and >7.5 log reduction in CFU compared with commercial hydrogel CL at 103 and 106 CFU, respectively. In an ex vivo porcine corneal infection model, pɛK+ hydrogels led to a significant decrease in viable PAO1 CFU and histologic analysis indicated a decreased infiltration of PAO1 into the stroma. Conclusions PɛK+ hydrogels demonstrated enhanced antimicrobial activity versus nonfunctionalized pɛK hydrogels against clinically relevant Paeruginosa strains. PɛK+ hydrogels have the potential to be used as a bandage CL with innate antimicrobial characteristics to minimize the risk of microbial keratitis.
Collapse
Affiliation(s)
- Stephnie M Kennedy
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Pallavi Deshpande
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | | | - Malcolm J Horsburgh
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Heather E Allison
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Stephen B Kaye
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | | | - Rachel L Williams
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
14
|
Sheremet AB, Nesterenko LN, Zigangirova NA. The Type Three Secretion System of Pseudomonas aeruginosa as a Target for Development of Antivirulence Drugs. MOLECULAR GENETICS, MICROBIOLOGY AND VIROLOGY 2020. [DOI: 10.3103/s0891416820010073] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
15
|
Lakshmanan D, Harikrishnan A, Jyoti K, Idul Ali M, Jeevaratnam K. A compound isolated from Alpinia officinarum Hance. inhibits swarming motility of Pseudomonas aeruginosa and down regulates virulence genes. J Appl Microbiol 2020; 128:1355-1365. [PMID: 31869477 DOI: 10.1111/jam.14563] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 12/18/2019] [Accepted: 12/18/2019] [Indexed: 12/16/2022]
Abstract
AIM The study was aimed at purifying the active principle from Alpinia officinarum rhizomes responsible for inhibition of swarming motility of Pseudomonas aeruginosa and analysing the mechanism of action. METHODS AND RESULTS The active compound from methanol extract of A. officinarum was purified by silica gel column chromatography followed by elution from Amberlite resin. The compound 1-(3,5-dihydroxyphenyl)-2-(methylamino)ethan-1-one, inhibited swarming motility at 12·5 µg ml-1 . This inhibition was independent of rhamnolipid production. Real-time PCR analysis showed significant down-regulation of virulence-associated genes including T3SS exoS, exoT and flagella master regulator fleQ. CONCLUSIONS The compound from A. officinarum inhibited swarming motility and significantly down-regulated the expression of type III secretory system effector genes exoS and exoT and flagellar master regulator fleQ genes. SIGNIFICANCE AND IMPACT OF THE STUDY The study identifies a potent swarming inhibitory compound from the common medicinal plant A. officinarum and reinstates the potential of plant-derived compounds in tackling virulence properties of pathogenic bacteria.
Collapse
Affiliation(s)
- D Lakshmanan
- Department of Biochemistry and Molecular Biology, Pondicherry University, Kalapet, Pondicherry, India
| | - A Harikrishnan
- Department of Chemistry, Pondicherry University, Kalapet, Pondicherry, India
| | - K Jyoti
- Department of Biochemistry and Molecular Biology, Pondicherry University, Kalapet, Pondicherry, India
| | - M Idul Ali
- Department of Biochemistry and Molecular Biology, Pondicherry University, Kalapet, Pondicherry, India
| | - K Jeevaratnam
- Department of Biochemistry and Molecular Biology, Pondicherry University, Kalapet, Pondicherry, India
| |
Collapse
|
16
|
Luo L, Matthews JD, Robinson BS, Jones RM. Vibrio parahaemolyticus VopA Is a Potent Inhibitor of Cell Migration and Apoptosis in the Intestinal Epithelium of Drosophila melanogaster. Infect Immun 2019; 87:e00669-18. [PMID: 30617204 PMCID: PMC6386545 DOI: 10.1128/iai.00669-18] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 12/22/2018] [Indexed: 11/20/2022] Open
Abstract
Animal models have played a key role in providing an understanding of the mechanisms that govern the pathophysiology of intestinal diseases. To expand on the repertoire of organisms available to study enteric diseases, we report on the use of the Drosophila melanogaster model to identify a novel function of an effector protein secreted by Vibrio parahaemolyticus, which is an enteric pathogen found in contaminated seafood. During pathogenesis, V. parahaemolyticus secretes effector proteins that usurp the host's innate immune signaling pathways, thus allowing the bacterium to evade detection by the innate immune system. One secreted effector protein, VopA, has potent inhibitory effects on mitogen-activated protein kinase (MAPK) signaling pathways via the acetylation of critical residues within the catalytic loops of mitogen-activated protein kinase kinases (MAPKKs). Using the Drosophila model and cultured mammalian cells, we show that VopA also has potent modulating activity on focal adhesion complex (FAC) proteins, where VopA markedly reduced the levels of focal adhesion kinase (FAK) phosphorylation at Ser910, whereas the phosphorylation levels of FAK at Tyr397 and Tyr861 were markedly increased. Cultured cells expressing VopA were also impaired in their ability to migrate and repopulate areas subjected to a scratch wound. Consistently, expression of VopA in Drosophila midgut enterocytes disrupted the normal enterocyte arrangement. Finally, VopA inhibited apoptosis in both Drosophila tissues and mammalian cultured cells. Together, our data show that VopA can alter normal intestinal homeostatic processes to facilitate opportunities for V. parahaemolyticus to prolong infection within the host.
Collapse
Affiliation(s)
- Liping Luo
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jason D Matthews
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Brian S Robinson
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Rheinallt M Jones
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
17
|
Kaminski A, Gupta KH, Goldufsky JW, Lee HW, Gupta V, Shafikhani SH. Pseudomonas aeruginosa ExoS Induces Intrinsic Apoptosis in Target Host Cells in a Manner That is Dependent on its GAP Domain Activity. Sci Rep 2018; 8:14047. [PMID: 30232373 PMCID: PMC6145893 DOI: 10.1038/s41598-018-32491-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 09/03/2018] [Indexed: 11/08/2022] Open
Abstract
Pseudomonas aeruginosa is a Gram-negative opportunistic pathogen that causes serious infections in immunocompromised individuals and cystic fibrosis patients. ExoS and ExoT are two homologous bifunctional Type III Secretion System (T3SS) virulence factors that induce apoptosis in target host cells. They possess a GTPase Activating Protein (GAP) domain at their N-termini, which share ~76% homology, and an ADP-ribosyltransferase (ADPRT) domain at their C-termini, which target non-overlapping substrates. Both the GAP and the ADPRT domains contribute to ExoT's cytotoxicity in target epithelial cells, whereas, ExoS-induced apoptosis is reported to be primarily due to its ADPRT domain. In this report, we demonstrate that ExoS/GAP domain is both necessary and sufficient to induce mitochondrial apoptosis. Our data demonstrate that intoxication with ExoS/GAP domain leads to enrichment of Bax and Bim into the mitochondrial outer-membrane, disruption of mitochondrial membrane and release of and cytochrome c into the cytosol, which activates initiator caspase-9 and effector caspase-3, that executes cellular death. We posit that the contribution of the GAP domain in ExoS-induced apoptosis was overlooked in prior studies due to its slower kinetics of cytotoxicity as compared to ADPRT. Our data clarify the field and reveal a novel virulence function for ExoS/GAP as an inducer of apoptosis.
Collapse
Affiliation(s)
- Amber Kaminski
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, USA
| | - Kajal H Gupta
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, USA
| | - Josef W Goldufsky
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, USA
| | - Ha Won Lee
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Vineet Gupta
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Sasha H Shafikhani
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA.
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, USA.
- Cancer Center, Rush University Medical Center, Chicago, IL, USA.
| |
Collapse
|
18
|
Belmadi N, Wu Y, Touqui L. Immuno-modulatory functions of the type-3 secretion system and impacts on the pulmonary host defense: A role for ExoS of Pseudomonas aeruginosa in cystic fibrosis. Toxicon 2018; 143:68-73. [PMID: 29339019 DOI: 10.1016/j.toxicon.2018.01.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 01/08/2018] [Accepted: 01/09/2018] [Indexed: 12/27/2022]
Abstract
Number of previous reviews had described the structures and the various functions of the exotoxins produced by the type-3 secretion system of Pseudomonas aeruginosa and their roles in the interactions of this bacterium with host cells. In this review, we summarize some relevant data of literature on ExoS, an exotoxin from the type-3 secretion system of P. aeruginosa, with a particular focus on the role of this toxin in the airways innate response of the host to infection by this bacterium, and its implication in the elimination of Staphylococcus aureus from the airways of patients with cystic fibrosis.
Collapse
Affiliation(s)
- Nawal Belmadi
- Mucoviscidose et Bronchopathies Chroniques, Unité Mixte Institut Pasteur/Paris V, Faculté de Médecine Cochin, Paris, France
| | - Yongzheng Wu
- Unité de Biologie cellulaire de l'infection microbienne, CNRS, UMR 3691, Institut Pasteur, Paris, France
| | - Lhousseine Touqui
- Mucoviscidose et Bronchopathies Chroniques, Unité Mixte Institut Pasteur/Paris V, Faculté de Médecine Cochin, Paris, France.
| |
Collapse
|
19
|
Saleeb M, Sundin C, Aglar Ö, Pinto AF, Ebrahimi M, Forsberg Å, Schüler H, Elofsson M. Structure-activity relationships for inhibitors of Pseudomonas aeruginosa exoenzyme S ADP-ribosyltransferase activity. Eur J Med Chem 2017; 143:568-576. [PMID: 29207339 DOI: 10.1016/j.ejmech.2017.11.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 11/15/2017] [Accepted: 11/17/2017] [Indexed: 12/18/2022]
Abstract
During infection, the Gram-negative opportunistic pathogen Pseudomonas aeruginosa employs its type III secretion system to translocate the toxin exoenzyme S (ExoS) into the eukaryotic host cell cytoplasm. ExoS is an essential in vivo virulence factor that enables P. aeruginosa to avoid phagocytosis and eventually kill the host cell. ExoS elicits its pathogenicity mainly via ADP-ribosyltransferase (ADPRT) activity. We recently identified a new class of ExoS ADPRT inhibitors with in vitro IC50 of around 20 μM in an enzymatic assay using a recombinant ExoS ADPRT domain. Herein, we report structure-activity relationships of this compound class by comparing a total of 51 compounds based on a thieno [2,3-d]pyrimidin-4(3H)-one and 4-oxo-3,4-dihydroquinazoline scaffolds. Improved inhibitors with in vitro IC50 values of 6 μM were identified. Importantly, we demonstrated that the most potent inhibitors block ADPRT activity of native full-length ExoS secreted by viable P. aeruginosa with an IC50 value of 1.3 μM in an enzymatic assay. This compound class holds promise as starting point for development of novel antibacterial agents.
Collapse
Affiliation(s)
- Michael Saleeb
- Department of Chemistry, Umeå University, 90187, Umeå, Sweden
| | | | - Öznur Aglar
- Department of Chemistry, Umeå University, 90187, Umeå, Sweden
| | - Ana Filipa Pinto
- Department of Medicinal Biochemistry and Biophysics, Karolinska Institute, 17177, Stockholm, Sweden
| | - Mahsa Ebrahimi
- Department of Medicinal Biochemistry and Biophysics, Karolinska Institute, 17177, Stockholm, Sweden
| | - Åke Forsberg
- Department of Molecular Biology, Umeå University, 90187, Umeå, Sweden
| | - Herwig Schüler
- Department of Medicinal Biochemistry and Biophysics, Karolinska Institute, 17177, Stockholm, Sweden
| | - Mikael Elofsson
- Department of Chemistry, Umeå University, 90187, Umeå, Sweden.
| |
Collapse
|
20
|
Morales-Espinosa R, Delgado G, Espinosa LF, Isselo D, Méndez JL, Rodriguez C, Miranda G, Cravioto A. Fingerprint Analysis and Identification of Strains ST309 as a Potential High Risk Clone in a Pseudomonas aeruginosa Population Isolated from Children with Bacteremia in Mexico City. Front Microbiol 2017; 8:313. [PMID: 28298909 PMCID: PMC5331068 DOI: 10.3389/fmicb.2017.00313] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 02/15/2017] [Indexed: 11/13/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen and is associated with nosocomial infections. Its ability to thrive in a broad range of environments is due to a large and diverse genome of which its accessory genome is part. The objective of this study was to characterize P. aeruginosa strains isolated from children who developed bacteremia, using pulse-field gel electrophoresis, and in terms of its genomic islands, virulence genes, multilocus sequence type, and antimicrobial susceptibility. Our results showed that P. aeruginosa strains presented the seven virulence genes: toxA, lasB, lecA, algR, plcH, phzA1, and toxR, a type IV pilin alleles (TFP) group I or II. Additionally, we detected a novel pilin and accessory gene, expanding the number of TFP alleles to group VI. All strains presented the PAPI-2 Island and the majority were exoU+ and exoS+ genotype. Ten percent of the strains were multi-drug resistant phenotype, 18% extensively drug-resistant, 68% moderately resistant and only 3% were susceptible to all the antimicrobial tested. The most prevalent acquired β-Lactamase was KPC. We identified a group of ST309 strains, as a potential high risk clone. Our finding also showed that the strains isolated from patients with bacteremia have important virulence factors involved in colonization and dissemination as: a TFP group I or II; the presence of the exoU gene within the PAPI-2 island and the presence of the exoS gene.
Collapse
Affiliation(s)
- Rosario Morales-Espinosa
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México Mexico City, Mexico
| | - Gabriela Delgado
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México Mexico City, Mexico
| | - Luis F Espinosa
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México Mexico City, Mexico
| | - Dassaev Isselo
- Servicio de Pediatría, Hospital Regional 36 San Alejandro, IMSS Puebla, Mexico
| | - José L Méndez
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México Mexico City, Mexico
| | - Cristina Rodriguez
- Laboratorio de Bacteriología, Facultad de Veterinaria y Zootecnia, Universidad Nacional Autónoma de México Mexico City, Mexico
| | - Guadalupe Miranda
- Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Unidad de Investigación en Epidemiología Hospitalaria Mexico City, Mexico
| | | |
Collapse
|
21
|
Chatterjee M, Anju C, Biswas L, Anil Kumar V, Gopi Mohan C, Biswas R. Antibiotic resistance in Pseudomonas aeruginosa and alternative therapeutic options. Int J Med Microbiol 2016; 306:48-58. [DOI: 10.1016/j.ijmm.2015.11.004] [Citation(s) in RCA: 157] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 11/18/2015] [Accepted: 11/26/2015] [Indexed: 01/05/2023] Open
|
22
|
A Novel Insight into Dehydroleucodine Mediated Attenuation of Pseudomonas aeruginosa Virulence Mechanism. BIOMED RESEARCH INTERNATIONAL 2015; 2015:216097. [PMID: 26640783 PMCID: PMC4658400 DOI: 10.1155/2015/216097] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Revised: 09/09/2015] [Accepted: 09/30/2015] [Indexed: 11/17/2022]
Abstract
Increasing resistance of Pseudomonas aeruginosa (P. aeruginosa) to conventional treatments demands the search for novel therapeutic strategies. In this study, the antimicrobial activity of dehydroleucodine (DhL), a sesquiterpene lactone obtained from Artemisia (A.) douglasiana, was screened against several pathogenic virulence effectors of P. aeruginosa. In vitro, minimum inhibitory concentration of DhL was determined against P. aeruginosa strains PAO1, PA103, PA14, and multidrug resistant clinical strain, CDN118. Results showed that DhL was active against each strain where PAO1 and PA103 showed higher susceptibility (MIC 0.48 mg/mL) as compared to PA14 (MIC 0.96 mg/mL) and CDN118 (MIC 0.98 mg/mL). Also, when PAO1 strain was grown in the presence of DhL (MIC50, 0.12 mg/mL), a delay in the generation time was noticed along with significant inhibition of secretory protease and elastase activities, interruption in biofilm attachment phase in a stationary culture, and a significant decline in Type III effector ExoS. At MIC50, DhL treatment increased the sensitivity of P. aeruginosa towards potent antibiotics. Furthermore, treatment of P. aeruginosa with DhL prevented toxin-induced apoptosis in macrophages. These observations suggest that DhL activity was at the bacterial transcriptional level. Hence, antimicrobial activity of DhL may serve as leads in the development of new anti-Pseudomonas pharmaceuticals.
Collapse
|
23
|
Han X, Wang L, Li W, Li B, Yang Y, Yan H, Qu L, Chen Y. Use of green fluorescent protein to monitor Lactobacillus plantarum in the gastrointestinal tract of goats. Braz J Microbiol 2015; 46:849-54. [PMID: 26413069 PMCID: PMC4568869 DOI: 10.1590/s1517-838246320140556] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Accepted: 02/02/2015] [Indexed: 11/28/2022] Open
Abstract
The experiment aimed to specifically monitor the passage of lactobacilli in
vivo after oral administration. The green fluorescent protein (GFP) gene
was cloned downstream from the constitutive p32 promoter from L. lactis
subsp. cremoris Wg2. The recombinant expression vector, pLEM415-gfp-p32,
was electroporated into Lactobacillus plantarum (L.
plantarum) isolated from goat. Green fluorescent protein (GFP)
was successfully expressed in L. plantarum. After 2 h
post-administration, transformed Lactobacillus could be detectable
in all luminal contents. In the rumen, bacteria concentration initially decreased,
reached the minimum at 42 h post-oral administration and then increased. However,
this concentration decreased constantly in the duodenum. This result indicated that
L. plantarum could colonize in the rumen but not in the
duodenum.
Collapse
Affiliation(s)
- Xufeng Han
- College of Animal Science and Technology, Northwest A&F University, People's Republic of China, China
| | - Lei Wang
- College of Animal Science and Technology, Northwest A&F University, People's Republic of China, China
| | - Wei Li
- College of Animal Science and Technology, Northwest A&F University, People's Republic of China, China
| | - Bibo Li
- College of Animal Science and Technology, Northwest A&F University, People's Republic of China, China
| | - Yuxin Yang
- College of Animal Science and Technology, Northwest A&F University, People's Republic of China, China
| | - Hailong Yan
- College of Life Science, Yulin University, People's Republic of China, China
| | - Lei Qu
- College of Life Science, Yulin University, People's Republic of China, China
| | - Yulin Chen
- College of Animal Science and Technology, Northwest A&F University, People's Republic of China, China
| |
Collapse
|
24
|
The Role of ExoS in Dissemination of Pseudomonas aeruginosa during Pneumonia. PLoS Pathog 2015; 11:e1004945. [PMID: 26090668 PMCID: PMC4474835 DOI: 10.1371/journal.ppat.1004945] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 05/11/2015] [Indexed: 01/11/2023] Open
Abstract
Hospital-acquired pneumonia is associated with high rates of morbidity and mortality, and dissemination to the bloodstream is a recognized risk factor for particularly poor outcomes. Yet the mechanism by which bacteria in the lungs gain access to the bloodstream remains poorly understood. In this study, we used a mouse model of Pseudomonas aeruginosa pneumonia to examine this mechanism. P. aeruginosa uses a type III secretion system to deliver effector proteins such as ExoS directly into the cytosol of eukaryotic cells. ExoS, a bi-functional GTPase activating protein (GAP) and ADP-ribosyltransferase (ADPRT), inhibits phagocytosis during pneumonia but has also been linked to a higher incidence of dissemination to the bloodstream. We used a novel imaging methodology to identify ExoS intoxicated cells during pneumonia and found that ExoS is injected into not only leukocytes but also epithelial cells. Phagocytic cells, primarily neutrophils, were targeted for injection with ExoS early during infection, but type I pneumocytes became increasingly injected at later time points. Interestingly, injection of these pneumocytes did not occur randomly but rather in discrete regions, which we designate ““fields of cell injection” (FOCI). These FOCI increased in size as the infection progressed and contained dead type I pneumocytes. Both of these phenotypes were attenuated in infections caused by bacteria secreting ADPRT-deficient ExoS, indicating that FOCI growth and type I pneumocyte death were dependent on the ADPRT activity of ExoS. During the course of infection, increased FOCI size was associated with enhanced disruption of the pulmonary-vascular barrier and increased bacterial dissemination into the blood, both of which were also dependent on the ADPRT activity of ExoS. We conclude that the ADPRT activity of ExoS acts upon type I pneumocytes to disrupt the pulmonary-vascular barrier during P. aeruginosa pneumonia, leading to bacterial dissemination. Dissemination to the bloodstream is a poor prognostic sign in patients with hospital-acquired pneumonia, yet the mechanism by which this occurs is poorly understood. To begin to address this issue, we have used a mouse model of P. aeruginosa pneumonia to study the mechanism by which the type-III-secreted effector protein ExoS enhances bacterial dissemination. We show that intoxication of type I pneumocytes by ExoS leads to cell death and disruption of the pulmonary-vascular barrier, allowing bacterial dissemination into the bloodstream. These effects required the ADP-ribosyltransferase activity of ExoS, as strains secreting an ExoS variant lacking this activity demonstrated reduced type I pneumocytes death and pulmonary-vascular breakdown. This study indicates that inhibitors of the ADP-ribosyltransferase activity of ExoS could serve as novel therapeutics for the prevention of bacteremic pneumonia.
Collapse
|
25
|
Ranjani J, Pushpanathan M, Mahesh A, Niraimathi M, Gunasekaran P, Rajendhran J. Pseudomonas aeruginosa PAO1 induces distinct cell death mechanisms in H9C2 cells and its differentiated form. J Basic Microbiol 2015; 55:1191-202. [PMID: 26011149 DOI: 10.1002/jobm.201500037] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 05/02/2015] [Indexed: 12/23/2022]
Abstract
Bacterial infections in myocardium may lead to the myocardial damage, which may progress to dilated cardiomyopathy and cardiac arrest. Pseudomonas aeruginosa has been reported to cause myocarditis and other systemic infections especially in immunocompromised patients. To understand the cellular responses during the establishment of infection in myocardium, we challenged differentiated H9C2 cells with P. aeruginosa PAO1. We also did comparison studies with infected undifferentiated form of H9C2 cells. Invasion studies revealed that PAO1 can invade both forms of cells and is able to survive and replicate within the host. Internalization of PAO1 was confirmed by live cell imaging and flow cytometry analysis. Though invasion of the pathogen triggered an increased ROS production in the host cells at earlier post-infection periods, it was decreased at later post-infection periods. Invasion of PAO1 induced cell death through apoptosis in differentiated H9C2 cells. Significant decrease in cell size, formation of polarized mitochondria, and nuclear fragmentation were observed in the infected differentiated cells. On the contrary, cell death preceded by multinucleation was observed in infected undifferentiated H9C2 cells. Morphological markers such as multinuclei and micro nuclei were observed. Cell cycle arrest in G2/M phase corroborates that the undifferentiated H9C2 cells experienced cell death preceded by multinucleation.
Collapse
Affiliation(s)
- Jothi Ranjani
- Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, Tamil Nadu, India
| | - Muthuirulan Pushpanathan
- Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, Tamil Nadu, India
| | - Ayyavu Mahesh
- IPLS Program, School of Biological Sciences, Madurai Kamaraj University, Madurai, Tamil Nadu, India
| | - Marimuthu Niraimathi
- IPLS Program, School of Biological Sciences, Madurai Kamaraj University, Madurai, Tamil Nadu, India
| | - Paramasamy Gunasekaran
- Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, Tamil Nadu, India.,Thiruvalluvar University, Vellore, Tamil Nadu, India
| | - Jeyaprakash Rajendhran
- Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, Tamil Nadu, India
| |
Collapse
|
26
|
Pseudomonas aeruginosa-induced bleb-niche formation in epithelial cells is independent of actinomyosin contraction and enhanced by loss of cystic fibrosis transmembrane-conductance regulator osmoregulatory function. mBio 2015; 6:e02533. [PMID: 25714715 PMCID: PMC4358002 DOI: 10.1128/mbio.02533-14] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The opportunistic pathogen Pseudomonas aeruginosa can infect almost any site in the body but most often targets epithelial cell-lined tissues such as the airways, skin, and the cornea of the eye. A common predisposing factor is cystic fibrosis (CF), caused by defects in the cystic fibrosis transmembrane-conductance regulator (CFTR). Previously, we showed that when P. aeruginosa enters epithelial cells it replicates intracellularly and occupies plasma membrane blebs. This phenotype is dependent on the type 3 secretion system (T3SS) effector ExoS, shown by others to induce host cell apoptosis. Here, we examined mechanisms for P. aeruginosa-induced bleb formation, focusing on its relationship to apoptosis and the CFTR. The data showed that P. aeruginosa-induced blebbing in epithelial cells is independent of actin contraction and is inhibited by hyperosmotic media (400 to 600 mOsM), distinguishing bacterially induced blebs from apoptotic blebs. Cells with defective CFTR displayed enhanced bleb formation upon infection, as demonstrated using bronchial epithelial cells from a patient with cystic fibrosis and a CFTR inhibitor, CFTR(Inh)-172. The defect was found to be correctable either by incubation in hyperosmotic media or by complementation with CFTR (pGFP-CFTR), suggesting that the osmoregulatory function of CFTR counters P. aeruginosa-induced bleb-niche formation. Accordingly, and despite their reduced capacity for bacterial internalization, CFTR-deficient cells showed greater bacterial occupation of blebs and enhanced intracellular replication. Together, these data suggest that P. aeruginosa bleb niches are distinct from apoptotic blebs, are driven by osmotic forces countered by CFTR, and could provide a novel mechanism for bacterial persistence in the host. Pseudomonas aeruginosa is an opportunistic pathogen problematic in hospitalized patients and those with cystic fibrosis (CF). Previously, we showed that P. aeruginosa can enter epithelial cells and replicate within them and traffics to the membrane blebs that it induces. This “bleb-niche” formation requires ExoS, previously shown to cause apoptosis. Here, we show that the driving force for bleb-niche formation is osmotic pressure, differentiating P. aeruginosa-induced blebs from apoptotic blebs. Either CFTR inhibition or CFTR mutation (as seen in people with CF) causes P. aeruginosa to make more bleb niches and provides an osmotic driving force for blebbing. CFTR inhibition also enhances bacterial occupation of blebs and intracellular replication. Since CFTR is targeted for removal from the plasma membrane when P. aeruginosa invades a healthy cell, these findings could relate to pathogenesis in both CF and healthy patient populations.
Collapse
|
27
|
The ADP-ribosyltransferase domain of the effector protein ExoS inhibits phagocytosis of Pseudomonas aeruginosa during pneumonia. mBio 2014; 5:e01080-14. [PMID: 24917597 PMCID: PMC4056551 DOI: 10.1128/mbio.01080-14] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Pseudomonas aeruginosa is a Gram-negative pathogen commonly associated with nosocomial infections such as hospital-acquired pneumonia. It uses a type III secretion system to deliver effector proteins directly into the cytosol of host cells. Type III secretion in P. aeruginosa has been linked to severe disease and worse clinical outcomes in animal and human studies. The majority of P. aeruginosa strains secrete ExoS, a bifunctional toxin with GTPase-activating protein and ADP-ribosyltransferase activities. Numerous in vitro studies have investigated the targets and cellular effects of ExoS, linking both its enzymatic activities with inhibition of bacterial internalization. However, little is known about how this toxin facilitates the progression of infection in vivo. In this study, we used a mouse model to investigate the role of ExoS in inhibiting phagocytosis during pneumonia. We first confirmed previous findings that the ADP-ribosyltransferase activity of ExoS, but not the GTPase-activating protein activity, was responsible for bacterial persistence and decreased host survival in this model. We then used two distinct assays to demonstrate that ExoS inhibited phagocytosis during pneumonia. In contrast to the findings of several in vitro studies, this in vivo inhibition was also dependent on the ADP-ribosyltransferase activity, but not the GTPase-activating protein activity, of ExoS. These results demonstrate for the first time the antiphagocytic function of ExoS in the context of an actual infection and indicate that blocking the ADP-ribosyltransferase activity of ExoS may have potential therapeutic benefit. Pseudomonas aeruginosa is a major cause of hospital-acquired infections. To cause severe disease, this bacterium uses a type III secretion system that delivers four effector proteins, ExoS, ExoT, ExoU, and ExoY, into host cells. The majority of P. aeruginosa strains secrete ExoS, a bifunctional toxin with GTPase-activating protein and ADP-ribosyltransferase activities. In cell culture models, both enzymatic activities have been associated with decreased bacterial internalization. However, our study is the first to examine a role for ExoS in blocking phagocytosis in an animal model. We report that ExoS does inhibit phagocytosis during pneumonia. The ADP-ribosyltransferase activity, but not the GTPase-activating protein activity, of ExoS is necessary for this effect. Our findings highlight the ability of P. aeruginosa to manipulate the inflammatory response during pneumonia to facilitate bacterial survival.
Collapse
|
28
|
Neeld D, Jin Y, Bichsel C, Jia J, Guo J, Bai F, Wu W, Ha UH, Terada N, Jin S. Pseudomonas aeruginosa injects NDK into host cells through a type III secretion system. MICROBIOLOGY-SGM 2014; 160:1417-1426. [PMID: 24699069 DOI: 10.1099/mic.0.078139-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Pseudomonas aeruginosa is a Gram-negative opportunistic human pathogen possessing a type III secretion system (T3SS) which injects toxic effector proteins into mammalian host cells. In previous studies, P. aeruginosa strains lacking all of the known type III effectors were shown to cause cytotoxicity upon prolonged infection time. In this study, we report the identification of a new cytotoxin, nucleoside diphosphate kinase (NDK), which is injected into eukaryotic cells in a T3SS-dependent manner. Injection of NDK is inhibited by the presence of previously known effectors of the T3SS, with an effectorless strain injecting the highest amount, suggesting active competition with the known T3SS effectors. NDK is shown to cause a cytotoxic response when expressed in eukaryotic cells, and P. aeruginosa strains harbouring NDK also show a greater toxicity than strains lacking it. Interestingly, the cytotoxic effect of intracellular NDK is independent of its kinase activity. In previous studies, NDK was shown to be secreted into culture supernatants via a type I secretion system and cause cytotoxicity in a kinase-dependent manner. Therefore, the current study highlights an alternative route of NDK secretion as well as two different cytotoxic mechanisms of NDK, depending on the extra- or intra-cellular location of the protein.
Collapse
Affiliation(s)
- Dennis Neeld
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL, USA
| | - Yongxin Jin
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, PR China
| | - Candace Bichsel
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL, USA
| | - Jinghua Jia
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL, USA
| | - Jianhui Guo
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, PR China
| | - Fang Bai
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, PR China
| | - Weihui Wu
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, PR China
| | - Un-Hwan Ha
- Department of Biotechnology and Bioinformatics, Korea University, Sejong, Republic of Korea
| | - Naohiro Terada
- Department of Pathology, University of Florida, Gainesville, FL, USA
| | - Shouguang Jin
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL, USA
| |
Collapse
|
29
|
Okuda J, Hanabusa A, Gotoh N. ExoS of Pseudomonas aeruginosa binds to a human KIF7 to induce cytotoxicity in cultured human bronchial epithelial cells. J Infect Chemother 2014; 20:121-7. [DOI: 10.1016/j.jiac.2013.09.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 09/09/2013] [Accepted: 09/13/2013] [Indexed: 12/13/2022]
|
30
|
Elsen S, Collin-Faure V, Gidrol X, Lemercier C. The opportunistic pathogen Pseudomonas aeruginosa activates the DNA double-strand break signaling and repair pathway in infected cells. Cell Mol Life Sci 2013; 70:4385-97. [PMID: 23760206 PMCID: PMC11113669 DOI: 10.1007/s00018-013-1392-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 05/24/2013] [Accepted: 05/29/2013] [Indexed: 11/30/2022]
Abstract
Highly hazardous DNA double-strand breaks can be induced in eukaryotic cells by a number of agents including pathogenic bacterial strains. We have investigated the genotoxic potential of Pseudomonas aeruginosa, an opportunistic pathogen causing devastating nosocomial infections in cystic fibrosis or immunocompromised patients. Our data revealed that infection of immune or epithelial cells by P. aeruginosa triggered DNA strand breaks and phosphorylation of histone H2AX (γH2AX), a marker of DNA double-strand breaks. Moreover, it induced formation of discrete nuclear repair foci similar to gamma-irradiation-induced foci, and containing γH2AX and 53BP1, an adaptor protein mediating the DNA-damage response pathway. Gene deletion, mutagenesis, and complementation in P. aeruginosa identified ExoS bacterial toxin as the major factor involved in γH2AX induction. Chemical inhibition of several kinases known to phosphorylate H2AX demonstrated that Ataxia Telangiectasia Mutated (ATM) was the principal kinase in P. aeruginosa-induced H2AX phosphorylation. Finally, infection led to ATM kinase activation by an auto-phosphorylation mechanism. Together, these data show for the first time that infection by P. aeruginosa activates the DNA double-strand break repair machinery of the host cells. This novel information sheds new light on the consequences of P. aeruginosa infection in mammalian cells. As pathogenic Escherichia coli or carcinogenic Helicobacter pylori can alter genome integrity through DNA double-strand breaks, leading to chromosomal instability and eventually cancer, our findings highlight possible new routes for further investigations of P. aeruginosa in cancer biology and they identify ATM as a potential target molecule for drug design.
Collapse
Affiliation(s)
- Sylvie Elsen
- CEA, DSV, iRTSV-BCI, INSERM, UMR-S 1036, Biologie Du Cancer Et de L’Infection, CNRS, ERL 5261, Bacterial Pathogenesis and Cellular Responses, UJF-Grenoble 1, Grenoble, France
| | | | - Xavier Gidrol
- CEA, DSV, iRTSV-BGE, INSERM, Unit 1038, Biologie à Grande Echelle, UJF-Grenoble 1, Grenoble, France
| | - Claudie Lemercier
- CEA, DSV, iRTSV-BGE, INSERM, Unit 1038, Biologie à Grande Echelle, UJF-Grenoble 1, Grenoble, France
- INSERM Unit 1038, CEA, DSV, iRTSV-BGE, 17 Rue Des Martyrs, 38054 Grenoble Cedex 9, France
| |
Collapse
|
31
|
A universal primer multiplex PCR method for typing of toxinogenic Pseudomonas aeruginosa. Appl Microbiol Biotechnol 2012; 95:1579-87. [PMID: 22923133 DOI: 10.1007/s00253-012-4277-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Revised: 06/08/2012] [Accepted: 06/30/2012] [Indexed: 10/28/2022]
Abstract
Pseudomonas aeruginosa is a well-known opportunistic pathogen that can cause acute nosocomial necrotizing pneumonia and genetic disorder cystic fibrosis of lung patients. Pathogenic interactions between P. aeruginosa and hosts are often guided by the secreted virulence determinants that interact with specific host targets. Exotoxin A, pyocyanin, elastase, and type III secretion system are the most significant virulence determinants and cause great concern. However, P. aeruginosa in various environments has high genotypic diversity, leading to deficiency of exotoxin genes for some P. aeruginosa strains. In current study, a universal primer-multiplex PCR method (UP-MPCR) was employed for the detection of five significant enterotoxin genes (toxA, phzM, lasB, ExoU, and ExoS) and one internal control gene ecfX in P. aeruginosa. Owing to the application of universal primer (UP), different targeted products have identical amplified efficiency and the sensitivity of multiplex PCR is improved. In addition, the complexity of multiplex PCR system is reduced and the compatibility of primers in a reaction is greatly increased. This UP-MPCR method can detect the presence of five P. aeruginosa enterotoxin genes in a single assay more rapidly and sensitively than conventional methods. In 214 drinking water and environmental isolates, the ExoU, ExoS, phzM, toxA, and lasB genes were detected in 20 (9 %), 180 (84 %), 179 (84 %), 196 (92 %), and 171 (80 %) isolates, respectively.
Collapse
|
32
|
Van der Velden J, Janssen-Heininger YMW, Mandalapu S, Scheller EV, Kolls JK, Alcorn JF. Differential requirement for c-Jun N-terminal kinase 1 in lung inflammation and host defense. PLoS One 2012; 7:e34638. [PMID: 22514650 PMCID: PMC3326034 DOI: 10.1371/journal.pone.0034638] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Accepted: 03/02/2012] [Indexed: 01/07/2023] Open
Abstract
The c-Jun N-terminal kinase (JNK) - 1 pathway has been implicated in the cellular response to stress in many tissues and models. JNK1 is known to play a role in a variety of signaling cascades, including those involved in lung disease pathogenesis. Recently, a role for JNK1 signaling in immune cell function has emerged. The goal of the present study was to determine the role of JNK1 in host defense against both bacterial and viral pneumonia, as well as the impact of JNK1 signaling on IL-17 mediated immunity. Wild type (WT) and JNK1 -/- mice were challenged with Escherichia coli, Staphylococcus aureus, or Influenza A. In addition, WT and JNK1 -/- mice and epithelial cells were stimulated with IL-17A. The impact of JNK1 deletion on pathogen clearance, inflammation, and histopathology was assessed. JNK1 was required for clearance of E. coli, inflammatory cell recruitment, and cytokine production. Interestingly, JNK1 deletion had only a small impact on the host response to S. aureus. JNK1 -/- mice had decreased Influenza A burden in viral pneumonia, yet displayed worsened morbidity. Finally, JNK1 was required for IL-17A mediated induction of inflammatory cytokines and antimicrobial peptides both in epithelial cells and the lung. These data identify JNK1 as an important signaling molecule in host defense and demonstrate a pathogen specific role in disease. Manipulation of the JNK1 pathway may represent a novel therapeutic target in pneumonia.
Collapse
Affiliation(s)
- Jos Van der Velden
- Department of Pathology, University of Vermont, Burlington, Vermont, United States of America
| | | | - Sivanarayna Mandalapu
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania, United States of America
| | - Erich V. Scheller
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania, United States of America
| | - Jay K. Kolls
- RK Mellon Foundation Institute, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania, United States of America
| | - John F. Alcorn
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
33
|
Jones RM, Luo L, Moberg KH. Aeromonas salmonicida-secreted protein AopP is a potent inducer of apoptosis in a mammalian and a Drosophila model. Cell Microbiol 2011; 14:274-85. [PMID: 22040305 DOI: 10.1111/j.1462-5822.2011.01717.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Some pathogens are able to establish themselves within the host because they have evolved mechanisms to disrupt host innate immunity. For example, a number of pathogens secrete preformed effector proteins via type III secretion apparatuses that influence innate immune or apoptotic signalling pathways. One group of effector proteins that usurp innate immune signalling is the YopJ-like family of bacterial effector proteins, which includes AopP from Aeromonas salmonicida. Aeromonas species are known to cause gastrointestinal disease in humans, and are associated mainly with subcutaneous wound infections and septicaemia in other metazoans, particularly fish. AopP has been reported to have inhibitory activity against the NF-κB pathway in cultured cells, although the pathological outcomes of AopP activity have not been examined. Here, we show that AopP has potent pro-apoptotic activity when expressed in cultured mammalian macrophage or epithelial cells, or when ectopically expressed in Drosophila melanogaster haemocytes or imaginal disk epithelial cells. Furthermore, apoptosis was significantly elevated upon concurrent AopP expression and TNF-α cellular stimulation. Together, our results demonstrate how the specificity of a YopJ-like protein towards signalling pathways directly governs cellular pathological outcome in disease.
Collapse
Affiliation(s)
- Rheinallt M Jones
- Departments of Pathology and Laboratory Medicine Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | | | | |
Collapse
|
34
|
Use of GFP to trace the colonization of Lactococcus lactis WH-C1 in the gastrointestinal tract of mice. J Microbiol Methods 2011; 86:390-2. [PMID: 21704659 DOI: 10.1016/j.mimet.2011.06.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Revised: 06/08/2011] [Accepted: 06/10/2011] [Indexed: 01/30/2023]
Abstract
A new expression vector for Lactococcus was constructed using nisI as a selection marker and GFP as a reporter protein to explore the colonization characteristics in vivo of Lactococcus lactis WH-C1. By high expression of GFP, it was shown WH-C1 could pass through the stomach and survive in the gastrointestinal tract.
Collapse
|
35
|
The ADP-ribosylation domain of Pseudomonas aeruginosa ExoS is required for membrane bleb niche formation and bacterial survival within epithelial cells. Infect Immun 2010; 78:4500-10. [PMID: 20732998 DOI: 10.1128/iai.00417-10] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Pseudomonas aeruginosa can establish a niche within the plasma membrane of epithelial cells (bleb niches) within which bacteria can survive, replicate, and swim at speeds detectable by real-time phase-contrast imaging. This novel virulence strategy is dependent on the bacterial type three secretion system (T3SS), since mutants lacking the T3SS needle or known T3SS effectors localize to perinuclear vacuoles and fail to replicate. Here, we determined which of the three effectors (ExoS, ExoT, or ExoY) were required for bleb niche formation and intracellular replication. PAO1 strains with mutations in exoS, exoT, exoY, or combinations thereof were compared to wild-type and complemented strains. P. aeruginosa exoS mutants, but not exoT or exoY mutants, lost the capacity for bleb niche formation and intracellular replication. Complementation with exoS rescued both phenotypes, either in the background of an exoS mutant or in a mutant lacking all three known effectors. Complementation with activity domain mutants of exoS revealed that the ADP-ribosyltransferase (ADP-r) activity of ExoS, but not the Rho-GAP activity nor the membrane localization domain (MLD) of ExoS, was required to elicit this phenotype. Membrane bleb niches that contained P. aeruginosa also bound annexin V-enhanced green fluorescent protein (EGFP), a marker of early apoptosis. These data show that P. aeruginosa bleb niches and intracellular survival involve ExoS ADP-r activity and implicate a connection between bleb niche formation and the known role(s) of ExoS-mediated apoptosis and/or Rab GTPase inactivation.
Collapse
|
36
|
Hawdon NA, Aval PS, Barnes RJ, Gravelle SK, Rosengren J, Khan S, Ciofu O, Johansen HK, Høiby N, Ulanova M. Cellular responses of A549 alveolar epithelial cells to serially collected Pseudomonas aeruginosa from cystic fibrosis patients at different stages of pulmonary infection. ACTA ACUST UNITED AC 2010; 59:207-20. [PMID: 20528926 DOI: 10.1111/j.1574-695x.2010.00693.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Pseudomonas aeruginosa is the major cause of chronic pulmonary disease in cystic fibrosis (CF) patients. During chronic infection, P. aeruginosa lose certain virulence factors, transform into a mucoid phenotype, and develop antibiotic resistance. We hypothesized that these genetic and phenotypic alterations of P. aeruginosa affect the airway epithelial responses. A549 cells were infected with 27 well-characterized isolates of P. aeruginosa from CF patients obtained during longitudinal observation, or with P. aeruginosa mutant strains lacking flagella, pili, lipopolysaccharide, or pyocyanin. Pseudomonas aeruginosa isolates from the early stages of the infection exhibited high adherence to A549 cells, were readily internalized, and able to induce reactive oxygen species (ROS) production, apoptosis of infected cells, and the release of granulocyte macrophage colony-stimulating factor. Late P. aeruginosa isolates collected from patients with chronic lung infection were shown to have reduced adherence to and internalization into A549 cells compared with bacteria from patients with intermittent P. aeruginosa colonization, and induced lower production of ROS and apoptosis, but caused high proinflammatory cytokine and adhesion molecule expression. Our findings suggest that despite the loss of virulence factors during the adaptation process in the CF lung by late P. aeruginosa strains, they retain high proinflammatory abilities that likely contribute to the disease pathogenesis.
Collapse
|
37
|
Cui Y, Li T, Zhang D, Han J. Expression of Ezrin and phosphorylated Ezrin (pEzrin) in pancreatic ductal adenocarcinoma. Cancer Invest 2010; 28:242-7. [PMID: 20158339 DOI: 10.3109/07357900903124498] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
It has been suggested that ezrin activation plays a key role in the regulation of cancer metastasis. In this study, we immunohistochemically investigated the expression patterns of total ezrin and its two phosphorylated forms, pEzrin(- Thr567) and pEzrin(- Tyr353), in 66 samples of invasive pancreatic carcinomas and 11 samples of normal pancreas tissues. Positive expressions of ezrin and pEzrin(- Thr567) were detected in most PDAC tissues, significantly higher than that of pEzrin(- Tyr353). Furthermore, overexpression of pEzrin(- Tyr353) in pancreatic cancers was associated with positive lymph node metastasis, less differentiation, pAkt overexpression, and shorter survival times. pEzrin(- Tyr353) may be a potent prognosis predictor for pancreatic cancer.
Collapse
Affiliation(s)
- Yazhou Cui
- Shandong Medicinal Biotechnology Center, Shandong Academy of Medical Sciences, Key Laboratory of Ministry of Health for Biotech-Drug, Key Laboratory for Modern Medicine and Technology of Shandong Province, Jinan, China
| | | | | | | |
Collapse
|
38
|
Hare NJ, Cordwell SJ. Proteomics of bacterial pathogens: Pseudomonas aeruginosa infections in cystic fibrosis - a case study. Proteomics Clin Appl 2010; 4:228-48. [PMID: 21137046 DOI: 10.1002/prca.200900144] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2009] [Revised: 09/09/2009] [Accepted: 09/30/2009] [Indexed: 12/20/2022]
Abstract
Technology development in the high throughput sciences of genomics, transcriptomics and proteomics, has been driven by bacteriological research. These organisms are excellent models for testing new methodology due to their comparatively small genome size, the relative ease of culturing large amounts of material, and the inherent biomedical, environmental and biotechnological interest in their underlying biology. Techniques developed in prokaryotes have since become applicable to higher organisms and human disease, opening vast research opportunities for understanding complex molecular processes. Pseudomonas aeruginosa is an excellent example of a microbe with fascinating properties suitable for stretching the boundaries of technology, and with underlying biology that remains poorly understood. P. aeruginosa is an opportunistic pathogen in humans and contains one of the largest genetic capabilities for a single-celled organism (approximately 5500 genes), which allows it to encode a wide variety of surface-associated and secreted virulence factors, as well as adapt to harsh environments, forming resistance to an array of antibacterial agents. While it is a major threat as a nosocomial pathogen, and particularly in the immunocompromised, it is also the most significant cause of mortality in patients suffering from the genetic disorder, cystic fibrosis. This review examines the role of proteomics in gaining a better understanding of the molecular basis of P. aeruginosa infection and persistence in the lungs of cystic fibrosis patients.
Collapse
Affiliation(s)
- Nathan J Hare
- School of Molecular and Microbial Biosciences, The University of Sydney, Sydney, Australia
| | | |
Collapse
|
39
|
Cao Z, Shi L, Li Y, Wang J, Wang D, Wang G, Sun B, Mu L, Yang M, Li H. Pseudomonas aeruginosa: mannose sensitive hemagglutinin inhibits the growth of human hepatocarcinoma cells via mannose-mediated apoptosis. Dig Dis Sci 2009; 54:2118-27. [PMID: 19052865 DOI: 10.1007/s10620-008-0603-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2008] [Accepted: 10/24/2008] [Indexed: 12/17/2022]
Abstract
A vaccine derived from the outer membrane proteins of the Gram-negative bacterium Pseudomonas aeruginosa has been shown to have immune modulatory properties. An inactivated mutant strain of P. aeruginosa with mannose sensitive hemagglutinin fimbria (PA-MSHA) has been used for adjuvant therapy for malignant cancer. In this study, the growth of human hepatocellular carcinoma Hep G2 and BEL-7402 cells is inhibited by PA-MSHA, but not by mannose-cleaved PA-MSHA. PA-MSHA-treated cells arrested in the S phase of the cell cycle and underwent apoptosis. We hypothesize that apoptosis induced by treatment of Hep G2 and BEL-7402 cells with PA-MSHA is mediated by the mannose residues of PA-MSHA and is propagated through the extrinsic apoptosis pathway directly through caspase-8. These data provide mechanistic details for the potential application of PA-MSHA-based treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Zhenyuan Cao
- Department of Intervention, The First Affiliated Clinic College of Harbin Medical University, Harbin, Heilongjiang, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Chung JW, Piao ZH, Yoon SR, Kim MS, Jeong M, Lee SH, Min JK, Kim JW, Cho YH, Kim JC, Ahn JK, Kim KE, Choi I. Pseudomonas aeruginosa eliminates natural killer cells via phagocytosis-induced apoptosis. PLoS Pathog 2009; 5:e1000561. [PMID: 19714221 PMCID: PMC2726936 DOI: 10.1371/journal.ppat.1000561] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2009] [Accepted: 07/31/2009] [Indexed: 12/19/2022] Open
Abstract
Pseudomonas aeruginosa (PA) is an opportunistic pathogen that causes the relapse of illness in immunocompromised patients, leading to prolonged hospitalization, increased medical expense, and death. In this report, we show that PA invades natural killer (NK) cells and induces phagocytosis-induced cell death (PICD) of lymphocytes. In vivo tumor metastasis was augmented by PA infection, with a significant reduction in NK cell number. Adoptive transfer of NK cells mitigated PA-induced metastasis. Internalization of PA into NK cells was observed by transmission electron microscopy. In addition, PA invaded NK cells via phosphoinositide 3-kinase (PI3K) activation, and the phagocytic event led to caspase 9-dependent apoptosis of NK cells. PA-mediated NK cell apoptosis was dependent on activation of mitogen-activated protein (MAP) kinase and the generation of reactive oxygen species (ROS). These data suggest that the phagocytosis of PA by NK cells is a critical event that affects the relapse of diseases in immunocompromised patients, such as those with cancer, and provides important insights into the interactions between PA and NK cells.
Collapse
Affiliation(s)
- Jin Woong Chung
- Cell Therapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong, Daejeon, Republic of Korea
- Department of Biological Science, Dong-A University, Busan, Republic of Korea
| | - Zheng-Hao Piao
- Cell Therapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong, Daejeon, Republic of Korea
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon, Republic of Korea
| | - Suk Ran Yoon
- Cell Therapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong, Daejeon, Republic of Korea
| | - Mi Sun Kim
- Cell Therapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong, Daejeon, Republic of Korea
| | - Mira Jeong
- Cell Therapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong, Daejeon, Republic of Korea
- Department of Functional Genomics, University of Science and Technology, Daejeon, Republic of Korea
| | - Suk Hyung Lee
- Cell Therapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong, Daejeon, Republic of Korea
- Department of Functional Genomics, University of Science and Technology, Daejeon, Republic of Korea
| | - Jeong Ki Min
- Antibody Therapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Jae Wha Kim
- Genome Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong, Daejeon, Republic of Korea
| | - You-Hee Cho
- Department of Life Science, College of Natural Science, Sogang University, Seoul Republic of Korea
| | - Jin Chul Kim
- Department of Microbiology, School of Bioscience and Biotechnology, Chungnam National University, Daejeon, Republic of Korea
| | - Jeong Keun Ahn
- Department of Microbiology, School of Bioscience and Biotechnology, Chungnam National University, Daejeon, Republic of Korea
| | - Kyoon Eon Kim
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon, Republic of Korea
| | - Inpyo Choi
- Cell Therapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong, Daejeon, Republic of Korea
- Department of Functional Genomics, University of Science and Technology, Daejeon, Republic of Korea
- * E-mail:
| |
Collapse
|
41
|
Jones RM, Wu H, Wentworth C, Luo L, Collier-Hyams L, Neish AS. Salmonella AvrA Coordinates Suppression of Host Immune and Apoptotic Defenses via JNK Pathway Blockade. Cell Host Microbe 2008; 3:233-44. [PMID: 18407067 DOI: 10.1016/j.chom.2008.02.016] [Citation(s) in RCA: 210] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2007] [Revised: 02/05/2008] [Accepted: 02/28/2008] [Indexed: 12/11/2022]
Abstract
Salmonellae are bacterial pathogens that have evolved sophisticated strategies to evade host immune defenses. These strategies include the secretion of effector proteins into mammalian cells so as to subvert innate immune and apoptotic signaling pathways, thereby allowing Salmonella to avoid elimination. Here, we show that the secreted Salmonella typhimurium effector protein AvrA possesses acetyltransferase activity toward specific mitogen-activated protein kinase kinases (MAPKKs) and potently inhibits c-Jun N-terminal kinase (JNK) and NF-kappaB signaling pathways in both transgenic Drosophila and murine models. Furthermore, we show that AvrA dampens the proapoptotic innate immune response to Salmonella at the mouse intestinal mucosa. This activity is consistent with the natural history of Salmonella in mammalian hosts, where the bacteria elicit transient inflammation but do not destroy epithelial cells. Our findings suggest that targeting JNK signaling to dampen apoptosis may be a conserved strategy for intracellular pathogens.
Collapse
Affiliation(s)
- Rheinallt M Jones
- Epithelial Pathobiology Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | | | |
Collapse
|
42
|
Angus AA, Lee AA, Augustin DK, Lee EJ, Evans DJ, Fleiszig SMJ. Pseudomonas aeruginosa induces membrane blebs in epithelial cells, which are utilized as a niche for intracellular replication and motility. Infect Immun 2008; 76:1992-2001. [PMID: 18316391 PMCID: PMC2346716 DOI: 10.1128/iai.01221-07] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2007] [Revised: 10/10/2007] [Accepted: 02/14/2008] [Indexed: 11/20/2022] Open
Abstract
Pseudomonas aeruginosa is known to invade epithelial cells during infection and in vitro. However, little is known of bacterial or epithelial factors modulating P. aeruginosa intracellular survival or replication after invasion, except that it requires a complete lipopolysaccharide core. In this study, real-time video microscopy revealed that invasive P. aeruginosa isolates induced the formation of membrane blebs in multiple epithelial cell types and that these were then exploited for intracellular replication and rapid real-time motility. Further studies revealed that the type three secretion system (T3SS) of P. aeruginosa was required for blebbing. Mutants lacking either the entire T3SS or specific T3SS components were instead localized to intracellular perinuclear vacuoles. Most T3SS mutants that trafficked to perinuclear vacuoles gradually lost intracellular viability, and vacuoles containing those bacteria were labeled by the late endosomal marker lysosome-associated marker protein 3 (LAMP-3). Interestingly, mutants deficient only in the T3SS translocon structure survived and replicated within the vacuoles that did not label with LAMP-3. Taken together, these data suggest two novel roles of the P. aeruginosa T3SS in enabling bacterial intracellular survival: translocon-dependent formation of membrane blebs, which form a host cell niche for bacterial growth and motility, and effector-dependent bacterial survival and replication within intracellular perinuclear vacuoles.
Collapse
Affiliation(s)
- Annette A Angus
- School of Optometry, University of California, Berkeley, CA 94720, USA
| | | | | | | | | | | |
Collapse
|
43
|
Chai WS, Zhu XM, Li SH, Fan JX, Chen BY. Role of Bcl-2 family members in caspase-3/9-dependent apoptosis during Pseudomonas aeruginosa infection in U937 cells. Apoptosis 2008; 13:833-43. [DOI: 10.1007/s10495-008-0197-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
44
|
Coiras M, Camafeita E, López-Huertas MR, Calvo E, López JA, Alcamí J. Application of proteomics technology for analyzing the interactions between host cells and intracellular infectious agents. Proteomics 2008; 8:852-73. [PMID: 18297655 PMCID: PMC7167661 DOI: 10.1002/pmic.200700664] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Host–pathogen interactions involve protein expression changes within both the host and the pathogen. An understanding of the nature of these interactions provides insight into metabolic processes and critical regulatory events of the host cell as well as into the mechanisms of pathogenesis by infectious microorganisms. Pathogen exposure induces changes in host proteins at many functional levels including cell signaling pathways, protein degradation, cytokines and growth factor production, phagocytosis, apoptosis, and cytoskeletal rearrangement. Since proteins are responsible for the cell biological functions, pathogens have evolved to manipulate the host cell proteome to achieve optimal replication. Intracellular pathogens can also change their proteome to adapt to the host cell and escape from immune surveillance, or can incorporate cellular proteins to invade other cells. Given that the interactions of intracellular infectious agents with host cells are mainly at the protein level, proteomics is the most suitable tool for investigating these interactions. Proteomics is the systematic analysis of proteins, particularly their interactions, modifications, localization and functions, that permits the study of the association between pathogens with their host cells as well as complex interactions such as the host–vector–pathogen interplay. A review on the most relevant proteomic applications used in the study of host–pathogen interactions is presented.
Collapse
Affiliation(s)
- Mayte Coiras
- Unidad de Inmunopatología del SIDA, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | | | | | | | | | | |
Collapse
|
45
|
Shafikhani SH, Morales C, Engel J. The Pseudomonas aeruginosa type III secreted toxin ExoT is necessary and sufficient to induce apoptosis in epithelial cells. Cell Microbiol 2008; 10:994-1007. [PMID: 18053004 PMCID: PMC10952005 DOI: 10.1111/j.1462-5822.2007.01102.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Type III secreted (T3SS) effectors are important virulence factors in acute infections caused by Pseudomonas aeruginosa. PA103, a well-studied human lung isolate, encodes and secretes two effectors, ExoU and ExoT. ExoU is a potent cytotoxin that causes necrotic cell death. In addition, PA103 can induce cell death in macrophages in an ExoU-independent but T3SS-dependent manner. We now demonstrate that ExoT is both necessary and sufficient to cause apoptosis in HeLa cells and that it activates the mitochondrial/cytochrome c-dependent apoptotic pathway. We further show that ExoT induction of cell death is primarily dependent on its ADP ribosyltransferase domain activity. Our data also indicate that the T3SS apparatus can cause necrotic cell death, which is effectively blocked by ExoT, suggesting that P. aeruginosa may have evolved strategies to prevent T3SS-induced necrosis.
Collapse
Affiliation(s)
- Sasha H. Shafikhani
- Department of Medicine, University of California, San Francisco, CA 94143, USA
- Department of Microbiology and Immunology, University of California, San Francisco, CA 94143, USA
| | - Christina Morales
- Department of Medicine, University of California, San Francisco, CA 94143, USA
- Department of Microbiology and Immunology, University of California, San Francisco, CA 94143, USA
| | - Joanne Engel
- Department of Medicine, University of California, San Francisco, CA 94143, USA
- Department of Microbiology and Immunology, University of California, San Francisco, CA 94143, USA
- Program in Microbial Pathogenesis and Host Defense, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
46
|
Arnoldo A, Curak J, Kittanakom S, Chevelev I, Lee VT, Sahebol-Amri M, Koscik B, Ljuma L, Roy PJ, Bedalov A, Giaever G, Nislow C, Merrill RA, Lory S, Stagljar I. Identification of small molecule inhibitors of Pseudomonas aeruginosa exoenzyme S using a yeast phenotypic screen. PLoS Genet 2008; 4:e1000005. [PMID: 18454192 PMCID: PMC2265467 DOI: 10.1371/journal.pgen.1000005] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2007] [Accepted: 01/17/2008] [Indexed: 11/19/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic human pathogen that is a key factor in the mortality of cystic fibrosis patients, and infection represents an increased threat for human health worldwide. Because resistance of Pseudomonas aeruginosa to antibiotics is increasing, new inhibitors of pharmacologically validated targets of this bacterium are needed. Here we demonstrate that a cell-based yeast phenotypic assay, combined with a large-scale inhibitor screen, identified small molecule inhibitors that can suppress the toxicity caused by heterologous expression of selected Pseudomonas aeruginosa ORFs. We identified the first small molecule inhibitor of Exoenzyme S (ExoS), a toxin involved in Type III secretion. We show that this inhibitor, exosin, modulates ExoS ADP-ribosyltransferase activity in vitro, suggesting the inhibition is direct. Moreover, exosin and two of its analogues display a significant protective effect against Pseudomonas infection in vivo. Furthermore, because the assay was performed in yeast, we were able to demonstrate that several yeast homologues of the known human ExoS targets are likely ADP-ribosylated by the toxin. For example, using an in vitro enzymatic assay, we demonstrate that yeast Ras2p is directly modified by ExoS. Lastly, by surveying a collection of yeast deletion mutants, we identified Bmh1p, a yeast homologue of the human FAS, as an ExoS cofactor, revealing that portions of the bacterial toxin mode of action are conserved from yeast to human. Taken together, our integrated cell-based, chemical-genetic approach demonstrates that such screens can augment traditional drug screening approaches and facilitate the discovery of new compounds against a broad range of human pathogens.
Collapse
Affiliation(s)
- Anthony Arnoldo
- Terrence Donnelly Centre for Cellular and Biomolecular Research, Department of Biochemistry, University of Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Ontario, Canada
| | - Jasna Curak
- Terrence Donnelly Centre for Cellular and Biomolecular Research, Department of Biochemistry, University of Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Ontario, Canada
| | - Saranya Kittanakom
- Terrence Donnelly Centre for Cellular and Biomolecular Research, Department of Biochemistry, University of Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Ontario, Canada
| | - Igor Chevelev
- Terrence Donnelly Centre for Cellular and Biomolecular Research, Department of Biochemistry, University of Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Ontario, Canada
| | - Vincent T. Lee
- Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Mehdi Sahebol-Amri
- Terrence Donnelly Centre for Cellular and Biomolecular Research, Department of Biochemistry, University of Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Ontario, Canada
| | - Becky Koscik
- Terrence Donnelly Centre for Cellular and Biomolecular Research, Department of Biochemistry, University of Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Ontario, Canada
| | - Lana Ljuma
- Terrence Donnelly Centre for Cellular and Biomolecular Research, Department of Biochemistry, University of Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Ontario, Canada
| | - Peter J. Roy
- Terrence Donnelly Centre for Cellular and Biomolecular Research, Department of Biochemistry, University of Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Ontario, Canada
| | - Antonio Bedalov
- Clinical Research and Human Biology Divisions, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Guri Giaever
- Terrence Donnelly Centre for Cellular and Biomolecular Research, Department of Biochemistry, University of Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Ontario, Canada
- Department of Pharmaceutical Sciences, University of Toronto, Ontario, Canada
| | - Corey Nislow
- Terrence Donnelly Centre for Cellular and Biomolecular Research, Department of Biochemistry, University of Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Ontario, Canada
- Department of Pharmaceutical Sciences, University of Toronto, Ontario, Canada
| | - Rod A. Merrill
- Department of Molecular and Cellular Biology, University of Guelph, Ontario, Canada
| | - Stephen Lory
- Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Igor Stagljar
- Terrence Donnelly Centre for Cellular and Biomolecular Research, Department of Biochemistry, University of Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
47
|
Abstract
Bacterial pathogens utilize toxins to modify or kill host cells. The bacterial ADP-ribosyltransferases are a family of protein toxins that covalently transfer the ADP-ribose portion of NAD to host proteins. Each bacterial ADP-ribosyltransferase toxin modifies a specific host protein(s) that yields a unique pathology. These toxins possess the capacity to enter a host cell or to use a bacterial Type III apparatus for delivery into the host cell. Advances in our understanding of bacterial toxin action parallel the development of biophysical and structural biology as well as our understanding of the mammalian cell. Bacterial toxins have been utilized as vaccines, as tools to dissect host cell physiology, and more recently for the development of novel therapies to treat human disease.
Collapse
Affiliation(s)
- Qing Deng
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA.
| | | |
Collapse
|
48
|
Evans DJ, McNamara NA, Fleiszig SMJ. Life at the front: dissecting bacterial-host interactions at the ocular surface. Ocul Surf 2007; 5:213-27. [PMID: 17660895 DOI: 10.1016/s1542-0124(12)70612-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The ocular surface usually looks quiet, presenting a general impression of biological inactivity. Yet, the ability of the cornea to maintain health while continually exposed to environmental insults, and in the relative absence of immune strategies afforded by other body sites, reflects its complexity. Because it is critical for transparency and, therefore, our survival, the fine structure of the cornea has likely provided the driving force for the evolution of what appears to be a truly remarkable system. While several molecules are now known to participate, we are only beginning to obtain the knowledge to fully explain the mechanisms involved in corneal resistance to infection. Full explanation will require a better understanding of the interplay between microbes and various components of the ocular surface, and of the critical factors determining health as the usual outcome. To understand infectious disease, we need to consider how the scenario changes in conditions associated with susceptibility. What we learn in the process could yield a wealth of potential therapies for a wide variety of diseases of the eye and of other sites.
Collapse
Affiliation(s)
- David J Evans
- School of Optometry, University of California, Berkeley, California 94720-2020, USA
| | | | | |
Collapse
|
49
|
Sha J, Wang SF, Suarez G, Sierra JC, Fadl AA, Erova TE, Foltz SM, Khajanchi BK, Silver A, Graf J, Schein CH, Chopra AK. Further characterization of a type III secretion system (T3SS) and of a new effector protein from a clinical isolate of Aeromonas hydrophila--part I. Microb Pathog 2007; 43:127-46. [PMID: 17644303 DOI: 10.1016/j.micpath.2007.05.002] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A type III secretion system (T3SS)-associated cytotoxin, AexT, with ADP-ribosyltransferase activity and homology to Pseudomonas aeruginosa bifuncational toxins ExoT/S, was recently identified from a fish pathogen Aeromonas salmonicida. In this study, we reported the molecular characterization of an aexT-like toxin gene (designated as aexU) from a diarrheal isolate SSU of A. hydrophila. The aexU gene was 1539bp in length and encoded a protein of 512 amino acid (aa) residues. The NH(2)-terminus of AexU (aa residues 1-231) exhibited a 67% homology with the NH(2)-terminus of AexT from A. salmonicida. Importantly, its COOH-terminus (aa residues 232-512) had no homology with any known functional proteins in the database; however, the full-length AexU retained ADP-ribosyltransferase activity. The expression and subsequent secretion of AexU was T3SS dependent, as inactivation of the ascV gene that codes for an inner-membrane component of the T3SS channel from the wild-type (WT) bacterium, blocked translocation of AexU in HT-29 human colonic epithelial cells. We provided evidence that inactivation of acrV and axsE genes (homologs of lcrV and exsE in Yersinia species and P. aeruginosa, respectively) from A. hydrophila SSU, altered expression and/or secretion of AexU. We deleted an aexU gene from the WT, as well as from the DeltaaopB mutant, of A. hydrophila, generating a single knockout (DeltaaexU) and a double knockout mutant, DeltaaopB/DeltaaexU. Increased phagocytosis was observed in RAW264.7 murine macrophages infected with the DeltaaopB/DeltaaexU mutant, as compared to macrophages when infected with the parental DeltaaopB strain. Further, mice infected with the DeltaaexU mutant had a 60% survival rate, compared to animals infected with the WT or the DeltaaexU-complemented strain that caused 90-100% of the animals to die at a 2-3 LD(50s) dose. Immunization of mice with the recombinant AexU protected them from subsequent lethal challenge dose by the WT bacterium. Finally, we detected specific anti-AexU antibodies in the sera of mice that survived challenge by the WT bacterium, which may indicate that AexU plays an important role in the pathogenesis of Aeromonas infections.
Collapse
Affiliation(s)
- Jian Sha
- Department of Microbiology , The University of Texas Medical Branch, 301 University Blvd., UTMB Galveston, TX 77555-1070, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Yang H, Shan Z, Kim J, Wu W, Lian W, Zeng L, Xing L, Jin S. Regulatory role of PopN and its interacting partners in type III secretion of Pseudomonas aeruginosa. J Bacteriol 2007; 189:2599-609. [PMID: 17237176 PMCID: PMC1855783 DOI: 10.1128/jb.01680-06] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The type III secretion system (T3SS) of Pseudomonas aeruginosa plays a significant role in pathogenesis. We have previously identified type III secretion factor (TSF), which is required for effective secretion of the type III effector molecules, in addition to the low calcium signal. TSF includes many low-affinity high-capacity calcium binding proteins, such as serum albumin and casein. A search for the TSF binding targets on the bacterial outer membrane resulted in identification of PopN, a component of the T3SS that is readily detectable on the bacterial cell surface. PopN specifically interacts with Pcr1, and both popN and pcr1 mutants have a constitutive type III secretion phenotype, suggesting that the two proteins form a complex that functions as a T3SS repressor. Further analysis of the popN operon genes resulted in identification of protein-protein interactions between Pcr1 and Pcr4 and between Pcr4 and Pcr3, as well as between PopN and Pcr2 in the presence of PscB. Unlike popN and pcr1 mutants, pcr3 and pcr4 mutants are totally defective in type III secretion, while a pcr2 mutant exhibits reduced type III secretion. Interestingly, PopN, Pcr1, Pcr2, and Pcr4 are all secreted in a type III secretion machinery-dependent manner, while Pcr3 is not. These findings imply that these components have important regulatory roles in controlling type III secretion.
Collapse
Affiliation(s)
- Hongjing Yang
- Department of Molecular Genetics and Microbiology, P.O. Box 100266, University of Florida, Gainesville, FL 32610-0266, USA
| | | | | | | | | | | | | | | |
Collapse
|