1
|
Almufarriji FM, Alotaibi BS, Alamri AS, S Alkhalil S, Alwethaynani MS. Phytoconstituents from Artemisia annua medicinal plant as potent inhibitors targeting Salmonella SpvB: a molecular docking and dynamic study. J Biomol Struct Dyn 2025:1-14. [PMID: 40256876 DOI: 10.1080/07391102.2025.2492237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 04/05/2025] [Indexed: 04/22/2025]
Abstract
Salmonella, a genus with a global presence, is a leading cause of diarrheal diseases in both humans and animals. With over 2,400 distinct serotypes, most exhibiting minimal host specificity, Salmonella infection remains a significant public health issue. It poses a substantial economic burden on both developed and developing nations due to the costs associated with disease surveillance, prevention, and treatment. To address this global challenge, it is essential to explore cost-effective therapeutic interventions derived from medicinal plants. In this study, we targeted the Salmonella SpvB ATR domain for molecular docking of phytochemical compounds. A library of 392 phytochemical compounds from the Artemisia annua (Sweet wormwood) medicinal plant was utilized. In the initial screening, the top 20 phytochemical compounds were selected based on their high binding affinity toward SpvB. These 20 compounds underwent interaction analysis, revealing that two compounds, IMPHY004808 and IMPHY015047, formed crucial interactions. The IMPHY004808 compound bound at binding site residues ARG414, ARG471, LEU473, and GLU538, with residue SER501 present at the active site. Similarly, the IMPHY015047 compound formed bonds at binding site residues ARG471, ARG414, GLY472, and GLU538, while residue SER501 was present at the active site of SpvB. The trajectory analysis of 500 ns MD simulation, including deviation, fluctuation, compactness, surface area calculation, secondary structure element alterations, and hydrogen bond analysis, showed that the complexes were stable during the simulation time. Moreover, PCA with minimal vibration, FEL analysis and MMPBSA analysis strongly recommend that the complexes were stable and further validation with experimentation is needed.
Collapse
Affiliation(s)
- Fawaz M Almufarriji
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Al-Quwayiyah, Shaqra University, Riyadh, Saudi Arabia
| | - Bader S Alotaibi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Al-Quwayiyah, Shaqra University, Riyadh, Saudi Arabia
| | - Ahlam Saleh Alamri
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Al-Quwayiyah, Shaqra University, Riyadh, Saudi Arabia
| | - Samia S Alkhalil
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Al-Quwayiyah, Shaqra University, Riyadh, Saudi Arabia
| | - Maher S Alwethaynani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Al-Quwayiyah, Shaqra University, Riyadh, Saudi Arabia
| |
Collapse
|
2
|
Egan MS, O'Rourke EA, Mageswaran SK, Zuo B, Martynyuk I, Demissie T, Hunter EN, Bass AR, Chang YW, Brodsky IE, Shin S. Inflammasomes primarily restrict cytosolic Salmonella replication within human macrophages. eLife 2025; 12:RP90107. [PMID: 40162563 PMCID: PMC11957546 DOI: 10.7554/elife.90107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025] Open
Abstract
Salmonella enterica serovar Typhimurium is a facultative intracellular pathogen that utilizes its type III secretion systems (T3SSs) to inject virulence factors into host cells and colonize the host. In turn, a subset of cytosolic immune receptors respond to T3SS ligands by forming multimeric signaling complexes called inflammasomes, which activate caspases that induce interleukin-1 (IL-1) family cytokine release and an inflammatory form of cell death called pyroptosis. Human macrophages mount a multifaceted inflammasome response to Salmonella infection that ultimately restricts intracellular bacterial replication. However, how inflammasomes restrict Salmonella replication remains unknown. We find that caspase-1 is essential for mediating inflammasome responses to Salmonella and restricting bacterial replication within human macrophages, with caspase-4 contributing as well. We also demonstrate that the downstream pore-forming protein gasdermin D (GSDMD) and Ninjurin-1 (NINJ1), a mediator of terminal cell lysis, play a role in controlling Salmonella replication in human macrophages. Notably, in the absence of inflammasome responses, we observed hyperreplication of Salmonella within the cytosol of infected cells as well as increased bacterial replication within vacuoles, suggesting that inflammasomes control Salmonella replication primarily within the cytosol and also within vacuoles. These findings reveal that inflammatory caspases and pyroptotic factors mediate inflammasome responses that restrict the subcellular localization of intracellular Salmonella replication within human macrophages.
Collapse
Affiliation(s)
- Marisa S Egan
- Department of Microbiology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Emily A O'Rourke
- Department of Microbiology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Shrawan Kumar Mageswaran
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Institute of Structural Biology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Biao Zuo
- Institute of Structural Biology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Electron Microscopy Resource Laboratory, Department of Biochemistry & Biophysics, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Inna Martynyuk
- Institute of Structural Biology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Electron Microscopy Resource Laboratory, Department of Biochemistry & Biophysics, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Tabitha Demissie
- Department of Microbiology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Emma N Hunter
- Department of Microbiology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Antonia R Bass
- Department of Microbiology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Yi-Wei Chang
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Institute of Structural Biology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Igor E Brodsky
- Department of Pathobiology, University of Pennsylvania School of Veterinary MedicinePhiladelphiaUnited States
| | - Sunny Shin
- Department of Microbiology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| |
Collapse
|
3
|
Ulbrich M, Seward CH, Ivanov AI, Ward BM, Butler JS, Dziejman M. VopX, a novel Vibrio cholerae T3SS effector, modulates host actin dynamics. mBio 2025; 16:e0301824. [PMID: 39878476 PMCID: PMC11898728 DOI: 10.1128/mbio.03018-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/19/2024] [Indexed: 01/31/2025] Open
Abstract
Pathogenic Vibrio cholerae strains cause cholera using different mechanisms. O1 and O139 serogroup strains use the toxin-co-regulated pilus (TCP) and cholera toxin (CT) for intestinal colonization and to promote secretory diarrhea, while non-O1/non-O139 serogroup strains are typically non-toxigenic and use alternate virulence factors to cause a clinically similar disease. An O39 serogroup, TCP/CT-negative V. cholerae strain, named AM-19226, uses a type III secretion system (T3SS) to translocate more than 10 effector proteins into the host cell cytosol. Effectors VopF and VopM directly interact with the host actin and contribute to colonization. Our previous studies using the Saccharomyces cerevisiae model system identified VopX as a third effector that alters cytoskeletal dynamics. Herein, we used complementary approaches to translate yeast findings to a mammalian system and determined the target and mechanism of VopX activity. VopX overexpression in HeLa cells caused dramatic cell rounding. Co-culture of strain AM-19226 with polarized Caco-2/BBE monolayers increased formation of stress fibers and focal adhesions, as well as Caco-2/BBE adherence to extracellular matrix in a VopX-dependent manner. Finally, we demonstrate in vitro that VopX can act as a guanine nucleotide exchange factor for RhoA, which functions upstream of a mitogen-activated protein kinase (MAPK) signaling pathway regulating cytoskeletal dynamics. Our results suggest that VopX activity initiates a signaling cascade resulting in enhanced cell-extracellular matrix adhesion, potentially preventing detachment of host cells, and facilitating sustained bacterial colonization during infection. VopX function is therefore part of a unique pathogenic strategy employed by T3SS-positive V. cholerae, which involves multiple cytoskeletal remodeling mechanisms to support a productive infection. IMPORTANCE Despite different infection strategies, enteric pathogens commonly employ a T3SS to colonize the human host and cause disease. Effector proteins are unique to each T3SS-encoding bacterial species and generally lack conserved amino acid sequences. However, T3SS effectors from diverse pathogens target and manipulate common host cell structures and signaling proteins, such as the actin cytoskeleton and MAPK pathway components. T3SS-encoding Vibrio cholerae strains and effectors have been relatively recently identified, and the mechanisms used to mediate colonization and secretory diarrhea are poorly understood. Two V. cholerae effectors that modify the host actin cytoskeleton were shown to be important for colonization. We therefore sought to determine the target(s) and mechanism of a third actin-reorganizing effector, VopX, based on results obtained from a yeast model system. We recapitulated actin-based phenotypes in multiple mammalian model systems, leading us to identify the molecular function of the V. cholerae VopX effector protein.
Collapse
Affiliation(s)
- Megan Ulbrich
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Christopher H. Seward
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Andrei I. Ivanov
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Brian M. Ward
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - J. Scott Butler
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Michelle Dziejman
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
4
|
Egan MS, O’Rourke EA, Mageswaran SK, Zuo B, Martynyuk I, Demissie T, Hunter EN, Bass AR, Chang YW, Brodsky IE, Shin S. Inflammasomes primarily restrict cytosolic Salmonella replication within human macrophages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.17.549348. [PMID: 37503120 PMCID: PMC10370064 DOI: 10.1101/2023.07.17.549348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Salmonella enterica serovar Typhimurium is a facultative intracellular pathogen that utilizes its type III secretion systems (T3SSs) to inject virulence factors into host cells and colonize the host. In turn, a subset of cytosolic immune receptors respond to T3SS ligands by forming multimeric signaling complexes called inflammasomes, which activate caspases that induce interleukin-1 (IL-1) family cytokine release and an inflammatory form of cell death called pyroptosis. Human macrophages mount a multifaceted inflammasome response to Salmonella infection that ultimately restricts intracellular bacterial replication. However, how inflammasomes restrict Salmonella replication remains unknown. We find that caspase-1 is essential for mediating inflammasome responses to Salmonella and restricting bacterial replication within human macrophages, with caspase-4 contributing as well. We also demonstrate that the downstream pore-forming protein gasdermin D (GSDMD) and Ninjurin-1 (NINJ1), a mediator of terminal cell lysis, play a role in controlling Salmonella replication in human macrophages. Notably, in the absence of inflammasome responses, we observed hyperreplication of Salmonella within the cytosol of infected cells as well as increased bacterial replication within vacuoles, suggesting that inflammasomes control Salmonella replication primarily within the cytosol and also within vacuoles. These findings reveal that inflammatory caspases and pyroptotic factors mediate inflammasome responses that restrict the subcellular localization of intracellular Salmonella replication within human macrophages.
Collapse
Affiliation(s)
- Marisa S. Egan
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Emily A. O’Rourke
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Shrawan Kumar Mageswaran
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Biao Zuo
- Electron Microscopy Resource Laboratory, Department of Biochemistry & Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Inna Martynyuk
- Electron Microscopy Resource Laboratory, Department of Biochemistry & Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Tabitha Demissie
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Emma N. Hunter
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Antonia R. Bass
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Yi-Wei Chang
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Igor E. Brodsky
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA
| | - Sunny Shin
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
5
|
Niedzialkowska E, Runyan LA, Kudryashova E, Egelman EH, Kudryashov DS. Stabilization of F-actin by Salmonella effector SipA resembles the structural effects of inorganic phosphate and phalloidin. Structure 2024; 32:725-738.e8. [PMID: 38518780 PMCID: PMC11162321 DOI: 10.1016/j.str.2024.02.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/08/2024] [Accepted: 02/26/2024] [Indexed: 03/24/2024]
Abstract
Entry of Salmonella into host enterocytes relies on its pathogenicity island 1 effector SipA. We found that SipA binds to F-actin in a 1:2 stoichiometry with sub-nanomolar affinity. A cryo-EM reconstruction revealed that SipA's globular core binds at the groove between actin strands, whereas the extended C-terminal arm penetrates deeply into the inter-strand space, stabilizing F-actin from within. The unusually strong binding of SipA is achieved by a combination of fast association via the core and very slow dissociation dictated by the arm. Similar to Pi, BeF3, and phalloidin, SipA potently inhibited actin depolymerization by actin depolymerizing factor (ADF)/cofilin, which correlated with increased filament stiffness, supporting the hypothesis that F-actin's mechanical properties contribute to the recognition of its nucleotide state by protein partners. The remarkably strong binding to F-actin maximizes the toxin's effects at the injection site while minimizing global influence on the cytoskeleton and preventing pathogen detection by the host cell.
Collapse
Affiliation(s)
- Ewa Niedzialkowska
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22903, USA
| | - Lucas A Runyan
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | - Elena Kudryashova
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | - Edward H Egelman
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22903, USA.
| | - Dmitri S Kudryashov
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
6
|
Yang J, Barrila J, Nauman EA, Nydam SD, Yang S, Park J, Gutierrez-Jensen AD, Castro CL, Ott CM, Buss K, Steel J, Zakrajsek AD, Schuff MM, Nickerson CA. Incremental increases in physiological fluid shear progressively alter pathogenic phenotypes and gene expression in multidrug resistant Salmonella. Gut Microbes 2024; 16:2357767. [PMID: 38783686 PMCID: PMC11135960 DOI: 10.1080/19490976.2024.2357767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/15/2024] [Indexed: 05/25/2024] Open
Abstract
The ability of bacteria to sense and respond to mechanical forces has important implications for pathogens during infection, as they experience wide fluid shear fluctuations in the host. However, little is known about how mechanical forces encountered in the infected host drive microbial pathogenesis. Herein, we combined mathematical modeling with hydrodynamic bacterial culture to profile transcriptomic and pathogenesis-related phenotypes of multidrug resistant S. Typhimurium (ST313 D23580) under different fluid shear conditions relevant to its transition from the intestinal tract to the bloodstream. We report that D23580 exhibited incremental changes in transcriptomic profiles that correlated with its pathogenic phenotypes in response to these progressive increases in fluid shear. This is the first demonstration that incremental changes in fluid shear forces alter stress responses and gene expression in any ST313 strain and offers mechanistic insight into how forces encountered by bacteria during infection might impact their disease-causing ability in unexpected ways.
Collapse
Affiliation(s)
- Jiseon Yang
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ, USA
- Biodesign Center for Immunotherapy, Vaccines and Virotherapy, Arizona State University, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Jennifer Barrila
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ, USA
- Biodesign Center for Immunotherapy, Vaccines and Virotherapy, Arizona State University, Tempe, AZ, USA
| | - Eric A. Nauman
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, USA
| | - Seth D. Nydam
- Department of Animal Care & Technologies, Arizona State University, Tempe, AZ, USA
| | - Shanshan Yang
- Bioinformatics Core Facility, Bioscience, Knowledge Enterprise, Arizona State University, Tempe, AZ, USA
| | - Jin Park
- Biodesign Center for Personalized Diagnostics, Arizona State University, Tempe, AZ, USA
| | - Ami D. Gutierrez-Jensen
- Biodesign Center for Immunotherapy, Vaccines and Virotherapy, Arizona State University, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Christian L. Castro
- Biodesign Center for Immunotherapy, Vaccines and Virotherapy, Arizona State University, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
- JES Tech, Houston, TX, USA
| | - C. Mark Ott
- Biomedical Research and Environmental Sciences Division, NASA Johnson Space Center, Houston, TX, USA
| | - Kristina Buss
- Bioinformatics Core Facility, Bioscience, Knowledge Enterprise, Arizona State University, Tempe, AZ, USA
- Biodesign Center for Personalized Diagnostics, Arizona State University, Tempe, AZ, USA
| | - Jason Steel
- Bioinformatics Core Facility, Bioscience, Knowledge Enterprise, Arizona State University, Tempe, AZ, USA
- Biodesign Center for Personalized Diagnostics, Arizona State University, Tempe, AZ, USA
| | - Anne D. Zakrajsek
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, USA
| | - Mary M. Schuff
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, USA
| | - Cheryl A. Nickerson
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ, USA
- Biodesign Center for Immunotherapy, Vaccines and Virotherapy, Arizona State University, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| |
Collapse
|
7
|
Niedzialkowska E, Runyan LA, Kudryashova E, Egelman EH, Kudryashov DS. Stabilization of F-actin by Salmonella effector SipA resembles the structural effects of inorganic phosphate and phalloidin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.26.573373. [PMID: 38234808 PMCID: PMC10793455 DOI: 10.1101/2023.12.26.573373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Entry of Salmonella into host enterocytes strictly relies on its pathogenicity island 1 effector SipA. We found that SipA binds to F-actin in a unique mode in a 1:2 stoichiometry with picomolar affinity. A cryo-EM reconstruction revealed that SipA's globular core binds at the grove between actin strands, whereas the extended C-terminal arm penetrates deeply into the inter-strand space, stabilizing F-actin from within. The unusually strong binding of SipA is achieved via a combination of fast association via the core and very slow dissociation dictated by the arm. Similarly to Pi, BeF3, and phalloidin, SipA potently inhibited actin depolymerization by ADF/cofilin, which correlated with the increased filament stiffness, supporting the hypothesis that F-actin's mechanical properties contribute to the recognition of its nucleotide state by protein partners. The remarkably strong binding to F-actin maximizes the toxin's effects at the injection site while minimizing global influence on the cytoskeleton and preventing pathogen detection by the host cell.
Collapse
Affiliation(s)
- Ewa Niedzialkowska
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22903, USA
| | - Lucas A. Runyan
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | - Elena Kudryashova
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | - Edward H. Egelman
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22903, USA
| | - Dmitri S. Kudryashov
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
8
|
Waugh S, Ranasinghe A, Gomez A, Houston S, Lithgow KV, Eshghi A, Fleetwood J, Conway KME, Reynolds LA, Cameron CE. Syphilis and the host: multi-omic analysis of host cellular responses to Treponema pallidum provides novel insight into syphilis pathogenesis. Front Microbiol 2023; 14:1254342. [PMID: 37795301 PMCID: PMC10546344 DOI: 10.3389/fmicb.2023.1254342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/01/2023] [Indexed: 10/06/2023] Open
Abstract
Introduction Syphilis is a chronic, multi-stage infection caused by the extracellular bacterium Treponema pallidum ssp. pallidum. Treponema pallidum widely disseminates through the vasculature, crosses endothelial, blood-brain and placental barriers, and establishes systemic infection. Although the capacity of T. pallidum to traverse the endothelium is well-described, the response of endothelial cells to T. pallidum exposure, and the contribution of this response to treponemal traversal, is poorly understood. Methods To address this knowledge gap, we used quantitative proteomics and cytokine profiling to characterize endothelial responses to T. pallidum. Results Proteomic analyses detected altered host pathways controlling extracellular matrix organization, necroptosis and cell death, and innate immune signaling. Cytokine analyses of endothelial cells exposed to T. pallidum revealed increased secretion of interleukin (IL)-6, IL-8, and vascular endothelial growth factor (VEGF), and decreased secretion of monocyte chemoattractant protein-1 (MCP-1). Discussion This study provides insight into the molecular basis of syphilis disease symptoms and the enhanced susceptibility of individuals infected with syphilis to HIV co-infection. These investigations also enhance understanding of the host response to T. pallidum exposure and the pathogenic strategies used by T. pallidum to disseminate and persist within the host. Furthermore, our findings highlight the critical need for inclusion of appropriate controls when conducting T. pallidum-host cell interactions using in vitro- and in vivo-grown T. pallidum.
Collapse
Affiliation(s)
- Sean Waugh
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | - Akash Ranasinghe
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | - Alloysius Gomez
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | - Simon Houston
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | - Karen V. Lithgow
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | - Azad Eshghi
- University of Victoria-Genome BC Proteomics Centre, Victoria, BC, Canada
| | - Jenna Fleetwood
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | - Kate M. E. Conway
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | - Lisa A. Reynolds
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | - Caroline E. Cameron
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA, United States
| |
Collapse
|
9
|
Egan MS, Zhang J, Shin S. Human and mouse NAIP/NLRC4 inflammasome responses to bacterial infection. Curr Opin Microbiol 2023; 73:102298. [PMID: 37058933 PMCID: PMC10225321 DOI: 10.1016/j.mib.2023.102298] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 02/15/2023] [Accepted: 02/22/2023] [Indexed: 04/16/2023]
Abstract
Intracellular immune complexes known as inflammasomes sense breaches of cytosolic sanctity. Inflammasomes promote downstream proinflammatory events, including interleukin-1 (IL-1) family cytokine release and pyroptotic cell death. The nucleotide-binding leucine-rich repeat family, apoptosis inhibitory protein/nucleotide-binding leucine-rich repeat family, caspase recruitment domain (CARD) domain-containing protein 4 (NAIP/NLRC4) inflammasome is involved in a range of pathogenic and protective inflammatory processes in mammalian hosts. In particular, the NAIP/NLRC4 inflammasome responds to flagellin and components of the virulence-associated type III secretion (T3SS) apparatus in the host cytosol, thereby allowing it to be a critical mediator of host defense during bacterial infection. Notable species- and cell type-specific differences exist in NAIP/NLRC4 inflammasome responses to bacterial pathogens. With a focus on Salmonella enterica serovar Typhimurium as a model pathogen, we review differences between murine and human NAIP/NLRC4 inflammasome responses. Differences in NAIP/NLRC4 inflammasome responses across species and cell types may have arisen in part due to evolutionary pressures.
Collapse
Affiliation(s)
- Marisa S Egan
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Jenna Zhang
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Sunny Shin
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
10
|
Zhou G, Zhao Y, Ma Q, Li Q, Wang S, Shi H. Manipulation of host immune defenses by effector proteins delivered from multiple secretion systems of Salmonella and its application in vaccine research. Front Immunol 2023; 14:1152017. [PMID: 37081875 PMCID: PMC10112668 DOI: 10.3389/fimmu.2023.1152017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 03/23/2023] [Indexed: 04/07/2023] Open
Abstract
Salmonella is an important zoonotic bacterial species and hazardous for the health of human beings and livestock globally. Depending on the host, Salmonella can cause diseases ranging from gastroenteritis to life-threatening systemic infection. In this review, we discuss the effector proteins used by Salmonella to evade or manipulate four different levels of host immune defenses: commensal flora, intestinal epithelial-mucosal barrier, innate and adaptive immunity. At present, Salmonella has evolved a variety of strategies against host defense mechanisms, among which various effector proteins delivered by the secretory systems play a key role. During its passage through the digestive system, Salmonella has to face the intact intestinal epithelial barrier as well as competition with commensal flora. After invasion of host cells, Salmonella manipulates inflammatory pathways, ubiquitination and autophagy processes with the help of effector proteins. Finally, Salmonella evades the adaptive immune system by interfering the migration of dendritic cells and interacting with T and B lymphocytes. In conclusion, Salmonella can manipulate multiple aspects of host defense to promote its replication in the host.
Collapse
Affiliation(s)
- Guodong Zhou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yuying Zhao
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Qifeng Ma
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Quan Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Shifeng Wang
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - Huoying Shi
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University (JIRLAAPS), Yangzhou, China
| |
Collapse
|
11
|
Meng K, Zhu P, Shi L, Li S. Determination of the Salmonella intracellular lifestyle by the diversified interaction of Type III secretion system effectors and host GTPases. WIREs Mech Dis 2023; 15:e1587. [PMID: 36250298 DOI: 10.1002/wsbm.1587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/03/2022] [Accepted: 09/03/2022] [Indexed: 11/06/2022]
Abstract
Intracellular bacteria have developed sophisticated strategies to subvert the host endomembrane system to establish a stable replication niche. Small GTPases are critical players in regulating each step of membrane trafficking events, such as vesicle biogenesis, cargo transport, tethering, and fusion events. Salmonella is a widely studied facultative intracellular bacteria. Salmonella delivers several virulence proteins, termed effectors, to regulate GTPase dynamics and subvert host trafficking for their benefit. In this review, we summarize an updated and systematic understanding of the interactions between bacterial effectors and host GTPases in determining the intracellular lifestyle of Salmonella. This article is categorized under: Infectious Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Kun Meng
- Institute of Infection and Immunity, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Ping Zhu
- Institute of Infection and Immunity, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Liuliu Shi
- School of Basic Medical Science, Hubei University of Medicine, Shiyan, Hubei, China
| | - Shan Li
- Institute of Infection and Immunity, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China.,College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China.,College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, Hubei, China
| |
Collapse
|
12
|
The Salmonella T3SS1 effector IpaJ is regulated by ItrA and inhibits the MAPK signaling pathway. PLoS Pathog 2022; 18:e1011005. [PMID: 36477497 PMCID: PMC9728880 DOI: 10.1371/journal.ppat.1011005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 11/14/2022] [Indexed: 12/12/2022] Open
Abstract
Invasion plasmid antigen J (IpaJ) is a protein with cysteine protease activity that is present in Salmonella and Shigella species. Salmonella enterica serovar Pullorum uses IpaJ to inhibit the NF-κB pathway and the subsequent inflammatory response, resulting in bacterial survival in host macrophages. In the present study, we performed a DNA pull-down assay and EMSA and identified ItrA, a new DeoR family transcriptional regulator that could control the expression of IpaJ by directly binding to the promoter of ipaJ. The deletion of itrA inhibited the transcription of ipaJ in Salmonella. Tn-Seq revealed that two regulators of Salmonella pathogenicity island 1 (SPI-1), namely HilA and HilD, regulated the secretion of IpaJ. The deletion of hilA, hilD or SPI-1 inhibited the secretion of IpaJ in both cultured medium and Salmonella-infected cells. In contrast, the strain with the deletion of ssrB (an SPI-2 regulator-encoding gene) displayed normal IpaJ secretion, indicating that IpaJ is an effector of the SPI-1-encoded type III secretion system (T3SS1). To further demonstrate the role of IpaJ in host cells, we performed quantitative phosphoproteomics and compared the fold changes in signaling molecules in HeLa cells infected with wild-type S. Pullorum C79-13 with those in HeLa cells infected with the ipaJ-deleted strain C79-13ΔpSPI12. Both phosphoproteomics and Western blot analyses revealed that p-MEK and p-ERK molecules were increased in C79-13ΔpSPI12- and C79-13ΔpSPI12-pipaJ(C45A)-infected cells; and Co-IP assays demonstrated that IpaJ interacts with Ras to reduce its ubiquitination, indicating that IpaJ can inhibit the activation of the MAPK signaling pathway.
Collapse
|
13
|
Naseer N, Zhang J, Bauer R, Constant DA, Nice TJ, Brodsky IE, Rauch I, Shin S. Salmonella enterica Serovar Typhimurium Induces NAIP/NLRC4- and NLRP3/ASC-Independent, Caspase-4-Dependent Inflammasome Activation in Human Intestinal Epithelial Cells. Infect Immun 2022; 90:e0066321. [PMID: 35678562 PMCID: PMC9302179 DOI: 10.1128/iai.00663-21] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 05/23/2022] [Indexed: 01/09/2023] Open
Abstract
Salmonella enterica serovar Typhimurium is a Gram-negative pathogen that causes diseases ranging from gastroenteritis to systemic infection and sepsis. Salmonella uses type III secretion systems (T3SS) to inject effectors into host cells. While these effectors are necessary for bacterial invasion and intracellular survival, intracellular delivery of T3SS products also enables detection of translocated Salmonella ligands by cytosolic immune sensors. Some of these sensors form multimeric complexes called inflammasomes, which activate caspases that lead to interleukin-1 (IL-1) family cytokine release and pyroptosis. In particular, the Salmonella T3SS needle, inner rod, and flagellin proteins activate the NAIP/NLRC4 inflammasome in murine intestinal epithelial cells (IECs), which leads to restriction of bacterial replication and extrusion of infected IECs into the intestinal lumen, thereby preventing systemic dissemination of Salmonella. While these processes are quite well studied in mice, the role of the NAIP/NLRC4 inflammasome in human IECs remains unknown. Unexpectedly, we found the NAIP/NLRC4 inflammasome is dispensable for early inflammasome responses to Salmonella in both human IEC lines and enteroids. Additionally, NLRP3 and the adaptor protein ASC are not required for inflammasome activation in Caco-2 cells. Instead, we observed a necessity for caspase-4 and gasdermin D pore-forming activity in mediating inflammasome responses to Salmonella in Caco-2 cells. These findings suggest that unlike murine IECs, human IECs do not rely on NAIP/NLRC4 or NLRP3/ASC inflammasomes and instead primarily use caspase-4 to mediate inflammasome responses to Salmonella pathogenicity island 1 (SPI-1)-expressing Salmonella.
Collapse
Affiliation(s)
- Nawar Naseer
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Jenna Zhang
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Renate Bauer
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, USA
- Department of Biosciences, Paris Lodron University of Salzburg, Salzburg, Austria
| | - David A. Constant
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, USA
| | - Timothy J. Nice
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, USA
| | - Igor E. Brodsky
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, USA
| | - Isabella Rauch
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, USA
| | - Sunny Shin
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
14
|
Smith KP, Lee W, Tonelli M, Lee Y, Light SH, Cornilescu G, Chakravarthy S. Solution structure and dynamics of the mitochondrial-targeted GTPase-activating protein (GAP) VopE by an integrated NMR/SAXS approach. Protein Sci 2022; 31:e4282. [PMID: 35137487 PMCID: PMC9047041 DOI: 10.1002/pro.4282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 01/17/2022] [Accepted: 01/27/2022] [Indexed: 11/11/2022]
Abstract
The bacterial pathogen Vibrio cholerae use a type III secretion system to inject effector proteins into a host cell. Recently, a putative Toxic GTPase Activating Protein (ToxGAP) called Vibrio outer protein E (VopE) was identified as a T3SS substrate and virulence factor that affected host mitochondrial dynamics and immune response. However, biophysical and structural characterization has been absent. Here, we describe solution NMR structure of the putative GTPase-activating protein (GAP) domain (73-204) of VopE. Using size exclusion chromatography coupled with small-angle x-ray scattering and residual dipolar coupling data, we restrained the MD process to efficiently determine the overall fold and improve the quality of the output calculated structures. Comparing the structure of VopE with other ToxGAP's revealed a similar overall fold with several features unique to VopE. Specifically, the "Bulge 1," α1 helix, and noteworthy "backside linker" elements on the N-terminus are dissimilar to the other ToxGAP's. By using NMR relaxation dispersion experiments, we demonstrate that these regions undergo motions on a > 6 s-1 timescale. Based on the disposition of these mobile regions relative to the putative catalytic arginine residue, we hypothesize that the protein may undergo structural changes to bind cognate GTPases.
Collapse
Affiliation(s)
- Kyle P. Smith
- Department of Cell & Developmental BiologyNorthwestern University ChicagoIllinoisUSA
- Xilio TherapeuticsWalthamMassachusettsUSA
| | - Woonghee Lee
- Department of ChemistryUniversity of Colorado‐DenverDenverColoradoUSA
| | - Marco Tonelli
- National Magnetic Resonance Facility at Madison, Department of BiochemistryUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Yeongjoon Lee
- Department of ChemistryUniversity of Colorado‐DenverDenverColoradoUSA
| | - Samuel H. Light
- Department of MicrobiologyUniversity of ChicagoChicagoIllinoisUSA
| | - Gabriel Cornilescu
- Advanced Technology Research Facility, Frederick National Laboratory for Cancer ResearchLeidos Biomedical Research, Inc., National Cancer Institute, National Institutes of HealthFrederickMarylandUSA
| | | |
Collapse
|
15
|
Naseer N, Egan MS, Reyes Ruiz VM, Scott WP, Hunter EN, Demissie T, Rauch I, Brodsky IE, Shin S. Human NAIP/NLRC4 and NLRP3 inflammasomes detect Salmonella type III secretion system activities to restrict intracellular bacterial replication. PLoS Pathog 2022; 18:e1009718. [PMID: 35073381 PMCID: PMC8812861 DOI: 10.1371/journal.ppat.1009718] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 02/03/2022] [Accepted: 12/27/2021] [Indexed: 01/16/2023] Open
Abstract
Salmonella enterica serovar Typhimurium is a Gram-negative pathogen that uses two distinct type III secretion systems (T3SSs), termed Salmonella pathogenicity island (SPI)-1 and SPI-2, to deliver virulence factors into the host cell. The SPI-1 T3SS enables Salmonella to invade host cells, while the SPI-2 T3SS facilitates Salmonella's intracellular survival. In mice, a family of cytosolic immune sensors, including NAIP1, NAIP2, and NAIP5/6, recognizes the SPI-1 T3SS needle, inner rod, and flagellin proteins, respectively. Ligand recognition triggers assembly of the NAIP/NLRC4 inflammasome, which mediates caspase-1 activation, IL-1 family cytokine secretion, and pyroptosis of infected cells. In contrast to mice, humans encode a single NAIP that broadly recognizes all three ligands. The role of NAIP/NLRC4 or other inflammasomes during Salmonella infection of human macrophages is unclear. We find that although the NAIP/NLRC4 inflammasome is essential for detecting T3SS ligands in human macrophages, it is partially required for responses to infection, as Salmonella also activated the NLRP3 and CASP4/5 inflammasomes. Importantly, we demonstrate that combinatorial NAIP/NLRC4 and NLRP3 inflammasome activation restricts Salmonella replication in human macrophages. In contrast to SPI-1, the SPI-2 T3SS inner rod is not sensed by human or murine NAIPs, which is thought to allow Salmonella to evade host recognition and replicate intracellularly. Intriguingly, we find that human NAIP detects the SPI-2 T3SS needle protein. Critically, in the absence of both flagellin and the SPI-1 T3SS, the NAIP/NLRC4 inflammasome still controlled intracellular Salmonella burden. These findings reveal that recognition of Salmonella SPI-1 and SPI-2 T3SSs and engagement of both the NAIP/NLRC4 and NLRP3 inflammasomes control Salmonella infection in human macrophages.
Collapse
Affiliation(s)
- Nawar Naseer
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Marisa S. Egan
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Valeria M. Reyes Ruiz
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - William P. Scott
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon
| | - Emma N. Hunter
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Tabitha Demissie
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Isabella Rauch
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon
| | - Igor E. Brodsky
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania
| | - Sunny Shin
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
- * E-mail:
| |
Collapse
|
16
|
Giacomodonato MN, Sarnacki SH, Aya Castañeda MDR, Garófalo AN, Betancourt DM, Cerquetti MC, Noto Llana M. Salmonella enterica serovar Enteritidis biofilm lifestyle induces lower pathogenicity and reduces inflammatory response in a murine model compared to planktonic bacteria. Rev Argent Microbiol 2021; 54:166-174. [PMID: 34961640 DOI: 10.1016/j.ram.2021.10.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/28/2021] [Accepted: 10/04/2021] [Indexed: 12/23/2022] Open
Abstract
Salmonellaenterica serovar Enteritidis (S. Enteritidis) is the most frequent serovar involved in human salmonellosis. It has been demonstrated that about 80% of infections are related to biofilm formation. There is scant information about the pathogenicity of S. Enteritidis and its relationship to biofilm production. In this regard, this study aimed to investigate the differential host response induced by S. Enteritidis biofilm and planktonic lifestyle. To this purpose, biofilm and planktonic bacteria were inoculated to BALB/c mice and epithelial cell culture. Survival studies revealed that biofilm is less virulent than planktonic cells. Reduced signs of intestinal inflammation and lower bacterial translocation were observed in animals inoculated with Salmonella biofilm compared to the planktonic group. Results showed that Salmonella biofilm was impaired for invasion of non-phagocytic cells and induces a lower inflammatory response in vivo and in vitro compared to that of planktonic bacteria. Taken together, the outcome of Salmonella-host interaction varies depending on the bacterial lifestyle.
Collapse
Affiliation(s)
- Mónica N Giacomodonato
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPaM-UBA-CONICET), Buenos Aires, Argentina; Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Sebastián H Sarnacki
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPaM-UBA-CONICET), Buenos Aires, Argentina; Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María Del Rosario Aya Castañeda
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPaM-UBA-CONICET), Buenos Aires, Argentina; Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ailín N Garófalo
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPaM-UBA-CONICET), Buenos Aires, Argentina; Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Diana M Betancourt
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPaM-UBA-CONICET), Buenos Aires, Argentina; Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María C Cerquetti
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPaM-UBA-CONICET), Buenos Aires, Argentina; Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mariángeles Noto Llana
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPaM-UBA-CONICET), Buenos Aires, Argentina; Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
17
|
Sibinelli-Sousa S, de Araújo-Silva AL, Hespanhol JT, Bayer-Santos E. Revisiting the steps of Salmonella gut infection with a focus on antagonistic interbacterial interactions. FEBS J 2021; 289:4192-4211. [PMID: 34546626 DOI: 10.1111/febs.16211] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/12/2021] [Accepted: 09/20/2021] [Indexed: 12/20/2022]
Abstract
A commensal microbial community is established in the mammalian gut during its development, and these organisms protect the host against pathogenic invaders. The hallmark of noninvasive Salmonella gut infection is the induction of inflammation via effector proteins secreted by the type III secretion system, which modulate host responses to create a new niche in which the pathogen can overcome the colonization resistance imposed by the microbiota. Several studies have shown that endogenous microbes are important to control Salmonella infection by competing for resources. However, there is limited information about antimicrobial mechanisms used by commensals and pathogens during these in vivo disputes for niche control. This review aims to revisit the steps that Salmonella needs to overcome during gut colonization-before and after the induction of inflammation-to achieve an effective infection. We focus on a series of reported and hypothetical antagonistic interbacterial interactions in which both contact-independent and contact-dependent mechanisms might define the outcome of the infection.
Collapse
Affiliation(s)
| | | | - Julia Takuno Hespanhol
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Brazil
| | - Ethel Bayer-Santos
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Brazil
| |
Collapse
|
18
|
Armijos-Jaramillo V, Espinosa N, Vizcaíno K, Santander-Gordón D. A Novel In Silico Method for Molecular Mimicry Detection Finds a Formin with the Potential to Manipulate the Maize Cell Cytoskeleton. MOLECULAR PLANT-MICROBE INTERACTIONS : MPMI 2021; 34:815-825. [PMID: 33755496 DOI: 10.1094/mpmi-11-20-0332-r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Molecular mimicry is one of the evolutionary strategies that parasites use to manipulate the host metabolism and perform an effective infection. This phenomenon has been observed in several animal and plant pathosystems. Despite the relevance of this mechanism in pathogenesis, little is known about it in fungus-plant interactions. For that reason, we performed an in silico method to select plausible mimicry candidates for the Ustilago maydis-maize interaction. Our methodology used a tripartite sequence comparison between the parasite, the host, and nonparasitic organisms' genomes. Furthermore, we used RNA sequencing information to identify gene coexpression, and we determined subcellular localization to detect potential cases of colocalization in the imitator-imitated pairs. With these approximations, we found a putative extracellular formin in U. maydis with the potential to rearrange the host cell cytoskeleton. In parallel, we detected at least two maize genes involved in the cytoskeleton rearrangement differentially expressed under U. maydis infection; thus, this find increases the expectation for the potential mimicry role of the fungal protein. The use of several sources of data led us to develop a strict and replicable in silico methodology to detect molecular mimicry in pathosystems with enough information available. Furthermore, this is the first time that a genomewide search has been performed to detect molecular mimicry in a U. maydis-maize system. Additionally, to allow the reproducibility of this experiment and the use of this pipeline, we created a Web server called Molecular Mimicry Finder.[Formula: see text] Copyright © 2021 The Author(s). This is an open access article distributed under the CC BY-NC-ND 4.0 International license.
Collapse
Affiliation(s)
- Vinicio Armijos-Jaramillo
- Carrera de Ingeniería en Biotecnología, Facultad de Ingeniería y Ciencias Aplicadas, Universidad de Las Américas, Quito, Ecuador
- Grupo de Bio-Quimioinformática, Universidad de Las Américas, Quito, Ecuador
| | - Nicole Espinosa
- Carrera de Ingeniería en Biotecnología, Facultad de Ingeniería y Ciencias Aplicadas, Universidad de Las Américas, Quito, Ecuador
| | - Karla Vizcaíno
- Carrera de Ingeniería en Biotecnología, Facultad de Ingeniería y Ciencias Aplicadas, Universidad de Las Américas, Quito, Ecuador
| | - Daniela Santander-Gordón
- Carrera de Ingeniería en Biotecnología, Facultad de Ingeniería y Ciencias Aplicadas, Universidad de Las Américas, Quito, Ecuador
| |
Collapse
|
19
|
Plastin 3 in health and disease: a matter of balance. Cell Mol Life Sci 2021; 78:5275-5301. [PMID: 34023917 PMCID: PMC8257523 DOI: 10.1007/s00018-021-03843-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/06/2021] [Accepted: 04/20/2021] [Indexed: 02/06/2023]
Abstract
For a long time, PLS3 (plastin 3, also known as T-plastin or fimbrin) has been considered a rather inconspicuous protein, involved in F-actin-binding and -bundling. However, in recent years, a plethora of discoveries have turned PLS3 into a highly interesting protein involved in many cellular processes, signaling pathways, and diseases. PLS3 is localized on the X-chromosome, but shows sex-specific, inter-individual and tissue-specific expression variability pointing towards skewed X-inactivation. PLS3 is expressed in all solid tissues but usually not in hematopoietic cells. When escaping X-inactivation, PLS3 triggers a plethora of different types of cancers. Elevated PLS3 levels are considered a prognostic biomarker for cancer and refractory response to therapies. When it is knocked out or mutated in humans and mice, it causes osteoporosis with bone fractures; it is the only protein involved in actin dynamics responsible for osteoporosis. Instead, when PLS3 is upregulated, it acts as a highly protective SMN-independent modifier in spinal muscular atrophy (SMA). Here, it seems to counteract reduced F-actin levels by restoring impaired endocytosis and disturbed calcium homeostasis caused by reduced SMN levels. In contrast, an upregulation of PLS3 on wild-type level might cause osteoarthritis. This emphasizes that the amount of PLS3 in our cells must be precisely balanced; both too much and too little can be detrimental. Actin-dynamics, regulated by PLS3 among others, are crucial in a lot of cellular processes including endocytosis, cell migration, axonal growth, neurotransmission, translation, and others. Also, PLS3 levels influence the infection with different bacteria, mycosis, and other pathogens.
Collapse
|
20
|
Kommnick C, Hensel M. Correlative Light and Scanning Electron Microscopy to Study Interactions of Salmonella enterica with Polarized Epithelial Cell Monolayers. Methods Mol Biol 2021; 2182:103-115. [PMID: 32894490 DOI: 10.1007/978-1-0716-0791-6_10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Live cell fluorescence imaging is the method of choice to visualize dynamic cellular processes in time and space, such as adhesion to and invasion of polarized epithelial cells by Salmonella enterica sv. Typhimurium. Scanning electron microscopy provides highest resolution of surface structures of infected cells, providing ultrastructure of the apical side of host cells and infecting Salmonella. Combining both methods toward correlative light and scanning electron microscopy (CLSEM) enables new insights in adhesion and invasion mechanisms regarding dynamics over time, and high spatial resolution with precise time lines. To correlate fast live cell imaging of polarized monolayer cells with scanning electron microscopy, we developed a robust method by using gold mesh grids as convenient CLSEM carriers for standard microscopes. By this, we were able to unravel the morphology of the apical structures of monolayers of polarized epithelial cells at distinct time points during Salmonella infection.
Collapse
Affiliation(s)
- Carina Kommnick
- Abteilung Mikrobiologie, CellNanOs-Center of Cellular Nanoanalytics Osnabrück, Fachbereich Biologie/Chemie, Universität Osnabrück, Osnabrück, Germany
| | - Michael Hensel
- Abteilung Mikrobiologie, Fachbereich Biologie/Chemie, Universität Osnabrück, Osnabrück, Germany.
- CellNanOs - Center of Cellular Nanoanalytics, Universität Osnabrück, Osnabrück, Germany.
| |
Collapse
|
21
|
Sachdeva K, Sundaramurthy V. The Interplay of Host Lysosomes and Intracellular Pathogens. Front Cell Infect Microbiol 2020; 10:595502. [PMID: 33330138 PMCID: PMC7714789 DOI: 10.3389/fcimb.2020.595502] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 10/22/2020] [Indexed: 12/20/2022] Open
Abstract
Lysosomes are an integral part of the intracellular defense system against microbes. Lysosomal homeostasis in the host is adaptable and responds to conditions such as infection or nutritional deprivation. Pathogens such as Mycobacterium tuberculosis (Mtb) and Salmonella avoid lysosomal targeting by actively manipulating the host vesicular trafficking and reside in a vacuole altered from the default lysosomal trafficking. In this review, the mechanisms by which the respective pathogen containing vacuoles (PCVs) intersect with lysosomal trafficking pathways and maintain their distinctness are discussed. Despite such active inhibition of lysosomal targeting, emerging literature shows that different pathogens or pathogen derived products exhibit a global influence on the host lysosomal system. Pathogen mediated lysosomal enrichment promotes the trafficking of a sub-set of pathogens to lysosomes, indicating heterogeneity in the host-pathogen encounter. This review integrates recent advancements on the global lysosomal alterations upon infections and the host protective role of the lysosomes against these pathogens. The review also briefly discusses the heterogeneity in the lysosomal targeting of these pathogens and the possible mechanisms and consequences.
Collapse
|
22
|
Bao H, Wang S, Zhao JH, Liu SL. Salmonella secretion systems: Differential roles in pathogen-host interactions. Microbiol Res 2020; 241:126591. [PMID: 32932132 DOI: 10.1016/j.micres.2020.126591] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/24/2020] [Accepted: 08/29/2020] [Indexed: 12/26/2022]
Abstract
The bacterial genus Salmonella includes a large group of food-borne pathogens that cause a variety of gastrointestinal or systemic diseases in hosts. Salmonella use several secretion devices to inject various effectors targeting eukaryotic hosts, or bacteria. In the past few years, considerable progress has been made towards understanding the structural features and molecular mechanisms of the secretion systems of Salmonella, particularly regarding their roles in host-pathogen interactions. In this review, we summarize the current advances about the main characteristics of the Salmonella secretion systems. Clarifying the roles of the secretion systems in the process of infecting various hosts will broaden our understanding of the importance of microbial interactions in maintaining human health and will provide information for developing novel therapeutic approaches.
Collapse
Affiliation(s)
- Hongxia Bao
- Genomics Research Center, College of Pharmacy, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China.
| | - Shuang Wang
- Department of Biopharmaceutical Sciences (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Jian-Hua Zhao
- Genomics Research Center, College of Pharmacy, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Shu-Lin Liu
- Genomics Research Center, College of Pharmacy, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China; Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Canada.
| |
Collapse
|
23
|
Qin L, Zhou Z, Li Q, Zhai C, Liu L, Quilichini TD, Gao P, Kessler SA, Jaillais Y, Datla R, Peng G, Xiang D, Wei Y. Specific Recruitment of Phosphoinositide Species to the Plant-Pathogen Interfacial Membrane Underlies Arabidopsis Susceptibility to Fungal Infection. THE PLANT CELL 2020; 32:1665-1688. [PMID: 32156686 PMCID: PMC7203932 DOI: 10.1105/tpc.19.00970] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/07/2020] [Accepted: 03/09/2020] [Indexed: 05/04/2023]
Abstract
Different phosphoinositides enriched at the membranes of specific subcellular compartments within plant cells contribute to organelle identity, ensuring appropriate cellular trafficking and function. During the infection of plant cells, biotrophic pathogens such as powdery mildews enter plant cells and differentiate into haustoria. Each haustorium is enveloped by an extrahaustorial membrane (EHM) derived from the host plasma membrane. Little is known about the EHM biogenesis and identity. Here, we demonstrate that among the two plasma membrane phosphoinositides in Arabidopsis (Arabidopsis thaliana), PI(4,5)P2 is dynamically up-regulated at powdery mildew infection sites and recruited to the EHM, whereas PI4P is absent in the EHM. Lateral transport of PI(4,5)P2 into the EHM occurs through a brefeldin A-insensitive but actin-dependent trafficking pathway. Furthermore, the lower levels of PI(4,5)P2 in pip5k1 pip5k2 mutants inhibit fungal pathogen development and cause disease resistance, independent of cell death-associated defenses and involving impaired host susceptibility. Our results reveal that plant biotrophic and hemibiotrophic pathogens modulate the subcellular distribution of host phosphoinositides and recruit PI(4,5)P2 as a susceptibility factor for plant disease.
Collapse
Affiliation(s)
- Li Qin
- Department of Biology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E2, Canada
| | - Zhuqing Zhou
- Laboratory of Cell Biology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Qiang Li
- Department of Biology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E2, Canada
| | - Chun Zhai
- Saskatoon Research and Development Centre, Agriculture and Agri-Food Canada, Saskatoon, Saskatchewan S7N 0X2, Canada
| | - Lijiang Liu
- Department of Biology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E2, Canada
- Key Laboratory of Biology and Genetic Improvement of Oil Crops, Ministry of Agriculture, Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wuhan, Hubei 430062, China
| | | | - Peng Gao
- Global Institute for Food Security, University of Saskatchewan, Saskatoon, Saskatchewan S7N 0W9, Canada
| | - Sharon A Kessler
- Department of Botany and Plant Pathology, Purdue University, West Lafayette, Indiana 47907
| | - Yvon Jaillais
- Laboratoire Reproduction et Développement des Plantes, Université de Lyon, École normale supérieure de Lyon, Université Claude Bernard Lyon 1, CNRS, INRA, Lyon 69342, France
| | - Raju Datla
- Global Institute for Food Security, University of Saskatchewan, Saskatoon, Saskatchewan S7N 0W9, Canada
| | - Gary Peng
- Saskatoon Research and Development Centre, Agriculture and Agri-Food Canada, Saskatoon, Saskatchewan S7N 0X2, Canada
| | - Daoquan Xiang
- National Research Council Canada, Saskatoon, Saskatchewan S7N 0W9, Canada
| | - Yangdou Wei
- Department of Biology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E2, Canada
| |
Collapse
|
24
|
Moorthy S, Byfield FJ, Janmey PA, Klein EA. Matrix stiffness regulates endosomal escape of uropathogenic E. coli. Cell Microbiol 2020; 22:e13196. [PMID: 32083802 DOI: 10.1111/cmi.13196] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 02/11/2020] [Accepted: 02/14/2020] [Indexed: 01/05/2023]
Abstract
Uropathogenic E. coli (UPEC) infection in vivo is characterized by invasion of bladder umbrella epithelial cells followed by endosomal escape and proliferation in the cytoplasm to form intracellular bacterial communities. By contrast, UPEC infection in tissue culture models results in bacteria being trapped within Lamp1-positive endosomes where proliferation is limited. Pharmacological disruption of the actin cytoskeleton has been shown to facilitate UPEC endosomal escape in vitro and extracellular matrix stiffness is a well-characterized physiological regulator of actin dynamics; therefore, we hypothesized that substrate stiffness may play a role in UPEC endosomal escape. Using functionalized polyacrylamide substrates, we found that at physiological stiffness, UPEC escaped the endosome and proliferated rapidly in the cytoplasm of bladder epithelial cells. Dissection of the cytoskeletal signaling pathway demonstrated that inhibition of the Rho GTPase RhoB or its effector PRK1 was sufficient to increase cytoplasmic bacterial growth and that RhoB protein level was significantly reduced at physiological stiffness. Our data suggest that tissue stiffness is a critical regulator of intracellular bacterial growth. Due to the ease of doing genetic and pharmacological manipulations in cell culture, this model system may provide a useful tool for performing mechanistic studies on the intracellular life cycle of uropathogens.
Collapse
Affiliation(s)
- Sudha Moorthy
- Department of Biology, Rutgers University-Camden, Camden, New Jersey, USA
| | - Fitzroy J Byfield
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Paul A Janmey
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Physiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Eric A Klein
- Department of Biology, Rutgers University-Camden, Camden, New Jersey, USA.,Center for Computational and Integrative Biology, Rutgers University-Camden, Camden, New Jersey, USA
| |
Collapse
|
25
|
Káňová E, Tkáčová Z, Bhide K, Kulkarni A, Jiménez-Munguía I, Mertinková P, Drážovská M, Tyagi P, Bhide M. Transcriptome analysis of human brain microvascular endothelial cells response to Neisseria meningitidis and its antigen MafA using RNA-seq. Sci Rep 2019; 9:18763. [PMID: 31822804 PMCID: PMC6904618 DOI: 10.1038/s41598-019-55409-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 11/28/2019] [Indexed: 01/25/2023] Open
Abstract
Interaction of Neisseria meningitidis (NM) with human brain microvascular endothelial cells (hBMECs) initiates of multiple cellular processes, which allow bacterial translocation across the blood-brain barrier (BBB). NM is equipped with several antigens, which interacts with the host cell receptors. Recently we have shown that adhesin MafA (UniProtKB-X5EG71), relatively less studied protein, is one of those surface exposed antigens that adhere to hBMECs. The present study was designed to comprehensively map the undergoing biological processes in hBMECs challenged with NM or MafA using RNA sequencing. 708 and 726 differentially expressed genes (DEGs) were identified in hBMECs exposed to NM and MafA, respectively. Gene ontology analysis of the DEGs revealed that several biological processes, which may alter the permeability of BBB, were activated. Comparative analysis of DEGs revealed that MafA, alike NM, might provoke TLR-dependent pathway and augment cytokine response. Moreover, both MafA and NM were able to induce genes involved in cell surface modifications, endocytosis, extracellular matrix remodulation and anoikis/apoptosis. In conclusion, this study for the first time describes effect of NM on the global gene expression in hBMECs using high-throughput RNA-seq. It also presents ability of MafA to induce gene expression, which might aid NM in breaching the BBB.
Collapse
Affiliation(s)
- Evelína Káňová
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy, Komenskeho 73, 04001, Kosice, Slovakia
| | - Zuzana Tkáčová
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy, Komenskeho 73, 04001, Kosice, Slovakia
| | - Katarína Bhide
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy, Komenskeho 73, 04001, Kosice, Slovakia
| | - Amod Kulkarni
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy, Komenskeho 73, 04001, Kosice, Slovakia
| | - Irene Jiménez-Munguía
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy, Komenskeho 73, 04001, Kosice, Slovakia
| | - Patrícia Mertinková
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy, Komenskeho 73, 04001, Kosice, Slovakia
| | - Monika Drážovská
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy, Komenskeho 73, 04001, Kosice, Slovakia
| | - Punit Tyagi
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy, Komenskeho 73, 04001, Kosice, Slovakia
| | - Mangesh Bhide
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy, Komenskeho 73, 04001, Kosice, Slovakia. .,Institute of Neuroimmunology of Slovak Academy of Sciences, 84510, Bratislava, Slovakia.
| |
Collapse
|
26
|
YshB Promotes Intracellular Replication and Is Required for Salmonella Virulence. J Bacteriol 2019; 201:JB.00314-19. [PMID: 31182500 DOI: 10.1128/jb.00314-19] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 06/07/2019] [Indexed: 11/20/2022] Open
Abstract
Salmonella virulence requires the initial invasion of host cells, followed by modulation of the intracellular environment for survival and replication. In an effort to characterize the role of small RNAs in Salmonella pathogenesis, we inadvertently identified a 5-kDa protein named YshB that is involved in the intracellular survival of Salmonella We show here that yshB expression is upregulated upon entry into macrophages. When yshB expression is upregulated before bacterial entry, invasion efficiency is inhibited. Lack of YshB resulted in reduced bacterial survival within the macrophages and led to reduced virulence in a mouse model of infection.IMPORTANCE Salmonella gastroenteritis is one of the most common causes of foodborne disease, possibly affecting millions of people globally each year. Here we characterize the role of a novel small protein, YshB, in mediating Salmonella intracellular survival. This elucidation adds to the body of knowledge regarding how this bacterium achieves intracellular survival.
Collapse
|
27
|
Lou L, Zhang P, Piao R, Wang Y. Salmonella Pathogenicity Island 1 (SPI-1) and Its Complex Regulatory Network. Front Cell Infect Microbiol 2019; 9:270. [PMID: 31428589 PMCID: PMC6689963 DOI: 10.3389/fcimb.2019.00270] [Citation(s) in RCA: 206] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 07/12/2019] [Indexed: 11/30/2022] Open
Abstract
Salmonella species can infect a diverse range of birds, reptiles, and mammals, including humans. The type III protein secretion system (T3SS) encoded by Salmonella pathogenicity island 1 (SPI-1) delivers effector proteins required for intestinal invasion and the production of enteritis. The T3SS is regarded as the most important virulence factor of Salmonella. SPI-1 encodes transcription factors that regulate the expression of some virulence factors of Salmonella, while other transcription factors encoded outside SPI-1 participate in the expression of SPI-1-encoded genes. SPI-1 genes are responsible for the invasion of host cells, regulation of the host immune response, e.g., the host inflammatory response, immune cell recruitment and apoptosis, and biofilm formation. The regulatory network of SPI-1 is very complex and crucial. Here, we review the function, effectors, and regulation of SPI-1 genes and their contribution to the pathogenicity of Salmonella.
Collapse
Affiliation(s)
- Lixin Lou
- Department of Infectious Diseases, First Hospital of Jilin University, Changchun, China
| | - Peng Zhang
- Department of Infectious Diseases, First Hospital of Jilin University, Changchun, China.,Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Rongli Piao
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.,Department of Gastroenterology, First Hospital of Jilin University, Changchun, China
| | - Yang Wang
- Department of Infectious Diseases, First Hospital of Jilin University, Changchun, China.,Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
28
|
Characterization of Salmonella Typhimurium and its monophasic variant 1,4, [5],12:i:- isolated from different sources. Folia Microbiol (Praha) 2019; 64:711-718. [PMID: 30721446 DOI: 10.1007/s12223-019-00683-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 01/21/2019] [Indexed: 01/19/2023]
Abstract
In order to characterize the most commonly detected Salmonella serotypes, we tested 124 isolates of S. Typhimurium and 89 isolates of the monophasic variant of S. Typhimurium (S. 1,4, [5],12:i:-) for their antimicrobial susceptibility by means of the Kirby-Bauer disk-diffusion method, and for the detection of 19 genes (four Phage Markers (g13, Sieb, eat, g8), ten prophage-related virulence genes (gipA, gtgB, nanH, gogB, grvA, sopE, sspH1, sspH2, sodC1, gtgE), and five plasmid-borne virulence genes (spvC, pefA, mig5, rcK, srgA)) by means of PCR-based assays. A total of 213 strains were analyzed from, humans (n = 122), animals (n = 25), food (n = 46), and irrigation water (n = 20). S. Typhimurium isolates showed higher variability, in both their resistance profiles and molecular typing, than S. 1,4, [5],12:i:-. Strains from irrigation water displayed significantly higher susceptibility to antibiotics than those from the other sources. Resistance to ampicillin, streptomycin, sulfonamide, and tetracycline was the most commonly detected resistance profile (R-type), being in serovar S. 1,4, [5],12:i:-, frequently associated to resistance to other antimicrobials. Significant differences in genetic profiles in the two abovementioned Salmonella serotypes were found. None of the plasmid-borne virulence genes investigated were detected in S. 1,4, [5],12:i:- isolates, while those genes, characterized 37.9% of the S. Typhimurium strains. Differences in the prevalence of some molecular targets between the two Salmonella serotypes deserve further study. Importantly, the grvA gene was found exclusively in S. Typhimurium strains, whereas sopE, sodC, gtgB, and gipA were mainly detected, with a statistically significant difference, in S. 1,4, [5],12:i:- isolates.
Collapse
|
29
|
Chua MD, Walker BD, Jin JP, Guttman JA. Calponins Are Recruited to Actin-Rich Structures Generated by Pathogenic Escherichia coli, Listeria, and Salmonella. Anat Rec (Hoboken) 2018; 301:2103-2111. [PMID: 30312538 DOI: 10.1002/ar.23956] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 07/10/2018] [Accepted: 07/12/2018] [Indexed: 01/12/2023]
Abstract
The ingestion of enteropathogenic Escherichia coli (EPEC), Listeria monocytogenes, or Salmonella enterica serovar Typhimurium leads to their colonization of the intestinal lumen, which ultimately causes an array of ailments ranging from diarrhea to bacteremia. Once in the intestines, these microbes generate various actin-rich structures to attach, invade, or move within the host intestinal epithelial cells. Although an assortment of actin-associated proteins has been identified to varying degrees at these structures, the localization of many actin stabilizing proteins have yet to be analyzed. Here, we examined the recruitment of the actin-associated proteins, calponin 1 and 2 at EPEC pedestals, L. monocytogenes actin clouds, comet tails and listeriopods, and S. Typhimurium membrane ruffles. In other systems, calponins are known to bind to and stabilize actin filaments. In EPEC pedestals, calponin 1 was recruited uniformly throughout the structures while calponin 2 was enriched at the apical tip. During L. monocytogenes infections, calponin 1 was found through all the actin-rich structures generated by the bacteria, while calponin 2 was only present within actin-rich structures formed by L. monocytogenes near the host cell membrane. Finally, both calponins were found within S. Typhimurium-generated membrane ruffles. Taken together, we have shown that although calponin 1 is recruited to actin-rich structures formed by the three bacteria, calponin 2 is specifically recruited to only membrane-bound actin-rich structures formed by the bacteria. Thus, our findings suggest that calponin 2 is a novel marker for membrane-bound actin structures formed by pathogenic bacteria. Anat Rec, 301:2103-2111, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Michael Dominic Chua
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | | | - Jian-Ping Jin
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan
| | - Julian A Guttman
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| |
Collapse
|
30
|
Yin Y, Zhou D. Organoid and Enteroid Modeling of Salmonella Infection. Front Cell Infect Microbiol 2018; 8:102. [PMID: 29670862 PMCID: PMC5894114 DOI: 10.3389/fcimb.2018.00102] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 03/16/2018] [Indexed: 12/12/2022] Open
Abstract
Salmonella are Gram-negative rod-shaped facultative anaerobic bacteria that are comprised of over 2,000 serovars. They cause gastroenteritis (salmonellosis) with headache, abdominal pain and diarrhea clinical symptoms. Salmonellosis brings a heavy burden for the public health in both developing and developed countries. Antibiotics are usually effective in treating the infected patients with severe gastroenteritis, although antibiotic resistance is on the rise. Understanding the molecular mechanisms of Salmonella infection is vital to combat the disease. In vitro immortalized 2-D cell lines, ex vivo tissues/organs and several animal models have been successfully utilized to study Salmonella infections. Although these infection models have contributed to uncovering the molecular virulence mechanisms, some intrinsic shortcomings have limited their wider applications. Notably, cell lines only contain a single cell type, which cannot reproduce some of the hallmarks of natural infections. While ex vivo tissues/organs alleviate some of these concerns, they are more difficult to maintain, in particular for long term experiments. In addition, non-human animal models are known to reflect only part of the human disease process. Enteroids and induced intestinal organoids are emerging as effective infection models due to their closeness in mimicking the infected tissues/organs. Induced intestinal organoids are derived from iPSCs and contain mesenchymal cells whereas enteroids are derive from intestinal stem cells and are comprised of epithelial cells only. Both enteroids and induced intestinal organoids mimic the villus and crypt domains comparable to the architectures of the in vivo intestine. We review here that enteroids and induced intestinal organoids are emerging as desired infection models to study bacterial-host interactions of Salmonella.
Collapse
Affiliation(s)
- Yuebang Yin
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Daoguo Zhou
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China.,Department of Biological Sciences, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
31
|
The Potential Virulence Factors of Providencia stuartii: Motility, Adherence, and Invasion. BIOMED RESEARCH INTERNATIONAL 2018; 2018:3589135. [PMID: 29682537 PMCID: PMC5841065 DOI: 10.1155/2018/3589135] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 12/30/2017] [Accepted: 01/21/2018] [Indexed: 11/17/2022]
Abstract
Providencia stuartii is the most common Providencia species capable of causing human infections. Currently P. stuartii is involved in high incidence of urinary tract infections in catheterized patients. The ability of bacteria to swarm on semisolid (viscous) surfaces and adhere to and invade host cells determines the specificity of the disease pathogenesis and its therapy. In the present study we demonstrated morphological changes of P. stuartii NK cells during migration on the viscous medium and discussed adhesive and invasive properties utilizing the HeLa-M cell line as a host model. To visualize the interaction of P. stuartii NK bacterial cells with eukaryotic cells in vitro scanning electron and confocal microscopy were performed. We found that bacteria P. stuartii NK are able to adhere to and invade HeLa-M epithelial cells and these properties depend on the age of bacterial culture. Also, to invade the host cells the infectious dose of the bacteria is essential. The microphotographs indicate that after incubation of bacterial P. stuartii NK cells together with epithelial cells the bacterial cells both were adhered onto and invaded into the host cells.
Collapse
|
32
|
Dufresne K, Saulnier-Bellemare J, Daigle F. Functional Analysis of the Chaperone-Usher Fimbrial Gene Clusters of Salmonella enterica serovar Typhi. Front Cell Infect Microbiol 2018; 8:26. [PMID: 29473020 PMCID: PMC5809473 DOI: 10.3389/fcimb.2018.00026] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 01/19/2018] [Indexed: 01/11/2023] Open
Abstract
The human-specific pathogen Salmonella enterica serovar Typhi causes typhoid, a major public health issue in developing countries. Several aspects of its pathogenesis are still poorly understood. S. Typhi possesses 14 fimbrial gene clusters including 12 chaperone-usher fimbriae (stg, sth, bcf, fim, saf, sef, sta, stb, stc, std, ste, and tcf). These fimbriae are weakly expressed in laboratory conditions and only a few are actually characterized. In this study, expression of all S. Typhi chaperone-usher fimbriae and their potential roles in pathogenesis such as interaction with host cells, motility, or biofilm formation were assessed. All S. Typhi fimbriae were better expressed in minimal broth. Each system was overexpressed and only the fimbrial gene clusters without pseudogenes demonstrated a putative major subunits of about 17 kDa on SDS-PAGE. Six of these (Fim, Saf, Sta, Stb, Std, and Tcf) also show extracellular structure by electron microscopy. The impact of fimbrial deletion in a wild-type strain or addition of each individual fimbrial system to an S. Typhi afimbrial strain were tested for interactions with host cells, biofilm formation and motility. Several fimbriae modified bacterial interactions with human cells (THP-1 and INT-407) and biofilm formation. However, only Fim fimbriae had a deleterious effect on motility when overexpressed. Overall, chaperone-usher fimbriae seem to be an important part of the balance between the different steps (motility, adhesion, host invasion and persistence) of S. Typhi pathogenesis.
Collapse
Affiliation(s)
- Karine Dufresne
- Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, QC, Canada
| | - Julie Saulnier-Bellemare
- Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, QC, Canada
| | - France Daigle
- Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
33
|
Abstract
Salmonella enterica subspecies enterica includes several serovars infecting both humans and other animals and leading to typhoid fever or gastroenteritis. The high prevalence of associated morbidity and mortality, together with an increased emergence of multidrug-resistant strains, is a current global health issue that has prompted the development of vaccination strategies that confer protection against most serovars. Currently available systemic vaccine approaches have major limitations, including a reduced effectiveness in young children and a lack of cross-protection among different strains. Having studied host-pathogen interactions, microbiologists and immunologists argue in favor of topical gastrointestinal administration for improvement in vaccine efficacy. Here, recent advances in this field are summarized, including mechanisms of bacterial uptake at the intestinal epithelium, the assessment of protective host immunity, and improved animal models that closely mimic infection in humans. The pros and cons of existing vaccines are presented, along with recent progress made with novel formulations. Finally, new candidate antigens and their relevance in the refined design of anti-Salmonella vaccines are discussed, along with antigen vectorization strategies such as nanoparticles or secretory immunoglobulins, with a focus on potentiating mucosal vaccine efficacy.
Collapse
|
34
|
InvS Coordinates Expression of PrgH and FimZ and Is Required for Invasion of Epithelial Cells by Salmonella enterica serovar Typhimurium. J Bacteriol 2017; 199:JB.00824-16. [PMID: 28439039 DOI: 10.1128/jb.00824-16] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 04/20/2017] [Indexed: 11/20/2022] Open
Abstract
Deep sequencing has revolutionized our understanding of the bacterial RNA world and has facilitated the identification of 280 small RNAs (sRNAs) in Salmonella Despite the suspicions that sRNAs may play important roles in Salmonella pathogenesis, the functions of most sRNAs remain unknown. To advance our understanding of RNA biology in Salmonella virulence, we searched for sRNAs required for bacterial invasion into nonphagocytic cells. After screening 75 sRNAs, we discovered that the ablation of InvS caused a significant decrease of Salmonella invasion into epithelial cells. A proteomic analysis showed that InvS modulated the levels of several type III secreted Salmonella proteins. The level of PrgH, a type III secretion apparatus protein, was significantly lower in the absence of InvS, consistent with the known roles of PrgH in effector secretion and bacterial invasion. We discovered that InvS modulates fimZ expression and hence flagellar gene expression and motility. We propose that InvS coordinates the increase of PrgH and decrease in FimZ that promote efficient Salmonella invasion into nonphagocytic cells.IMPORTANCE Salmonellosis continues to be the most common foodborne infection reported by the CDC in the United States. Central to Salmonella pathogenesis is the ability to invade nonphagocytic cells and to replicate inside host cells. Invasion genes are known to be regulated by protein transcriptional networks, but little is known about the role played by small RNAs (sRNAs) in this process. We have identified a novel sRNA, InvS, that is involved in Salmonella invasion. Our result will likely provide an opportunity to better understand the fundamental question of how Salmonella regulates invasion gene expression and may inform strategies for therapeutic intervention.
Collapse
|
35
|
Riba A, Emmenlauer M, Chen A, Sigoillot F, Cong F, Dehio C, Jenkins J, Zavolan M. Explicit Modeling of siRNA-Dependent On- and Off-Target Repression Improves the Interpretation of Screening Results. Cell Syst 2017; 4:182-193.e4. [PMID: 28215525 DOI: 10.1016/j.cels.2017.01.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 12/09/2016] [Accepted: 01/13/2017] [Indexed: 12/31/2022]
Abstract
RNAi is broadly used to map gene regulatory networks, but the identification of genes that are responsible for the observed phenotypes is challenging, as small interfering RNAs (siRNAs) simultaneously downregulate the intended on targets and many partially complementary off targets. Additionally, the scarcity of publicly available control datasets hinders the development and comparative evaluation of computational methods for analyzing the data. Here, we introduce PheLiM (https://github.com/andreariba/PheLiM), a method that uses predictions of siRNA on- and off-target downregulation to infer gene-specific contributions to phenotypes. To assess the performance of PheLiM, we carried out siRNA- and CRISPR/Cas9-based genome-wide screening of two well-characterized pathways, bone morphogenetic protein (BMP) and nuclear factor κB (NF-κB), and we reanalyzed publicly available siRNA screens. We demonstrate that PheLiM has the overall highest accuracy and most reproducible results compared to other available methods. PheLiM can accommodate various methods for predicting siRNA off targets and is broadly applicable to the identification of genes underlying complex phenotypes.
Collapse
Affiliation(s)
- Andrea Riba
- Biozentrum, University of Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland
| | - Mario Emmenlauer
- Biozentrum, University of Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland
| | - Amy Chen
- Developmental & Molecular Pathways, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Frederic Sigoillot
- Developmental & Molecular Pathways, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Feng Cong
- Developmental & Molecular Pathways, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Christoph Dehio
- Biozentrum, University of Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland
| | - Jeremy Jenkins
- Developmental & Molecular Pathways, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Mihaela Zavolan
- Biozentrum, University of Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland.
| |
Collapse
|
36
|
Ingram JP, Brodsky IE, Balachandran S. Interferon-γ in Salmonella pathogenesis: New tricks for an old dog. Cytokine 2016; 98:27-32. [PMID: 27773552 DOI: 10.1016/j.cyto.2016.10.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 10/13/2016] [Accepted: 10/15/2016] [Indexed: 12/21/2022]
Abstract
Salmonella enterica is a facultative intracellular bacterium that is the leading cause of food borne illnesses in humans. The cytokine IFN-γ has well-established antibacterial properties against Salmonella and other intracellular microbes, for example its capacity to activate macrophages, promote phagocytosis, and destroy phagocytosed microbes by free radical-driven toxification of phagosomes. But IFN-γ induces the expression of hundreds of uncharacterized genes, suggesting that this cytokine deploys additional antimicrobial strategies that await discovery. Recently, one such mechanism, mediated by a family of IFN-inducible small GTPases called Guanylate Binding Proteins (GBPs) has been uncovered. GBPs were shown to facilitate the pyroptotic clearance of Salmonella from infected macrophages by rupturing the protective intracellular vacuole this microbe forms around itself. Once this protective vacuole is lost, exposed Salmonella activates pyroptosis, which destroys the infected cell. In this review, we summarize such emerging roles for IFN-γ in restricting Salmonella pathogenesis.
Collapse
Affiliation(s)
- Justin P Ingram
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA 19111, United States
| | - Igor E Brodsky
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, United States
| | - Siddharth Balachandran
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA 19111, United States.
| |
Collapse
|
37
|
Shimono M, Lu YJ, Porter K, Kvitko BH, Henty-Ridilla J, Creason A, He SY, Chang JH, Staiger CJ, Day B. The Pseudomonas syringae Type III Effector HopG1 Induces Actin Remodeling to Promote Symptom Development and Susceptibility during Infection. PLANT PHYSIOLOGY 2016; 171:2239-55. [PMID: 27217495 PMCID: PMC4936540 DOI: 10.1104/pp.16.01593] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 05/19/2016] [Indexed: 05/20/2023]
Abstract
The plant cytoskeleton underpins the function of a multitude of cellular mechanisms, including those associated with developmental- and stress-associated signaling processes. In recent years, the actin cytoskeleton has been demonstrated to play a key role in plant immune signaling, including a recent demonstration that pathogens target actin filaments to block plant defense and immunity. Herein, we quantified spatial changes in host actin filament organization after infection with Pseudomonas syringae pv. tomato DC3000 (Pst DC3000), demonstrating that the type-III effector HopG1 is required for pathogen-induced changes to actin filament architecture and host disease symptom development during infection. Using a suite of pathogen effector deletion constructs, coupled with high-resolution microscopy, we found that deletion of hopG1 from Pst DC3000 resulted in a reduction in actin bundling and a concomitant increase in the density of filament arrays in Arabidopsis, both of which correlate with host disease symptom development. As a mechanism underpinning this activity, we further show that the HopG1 effector interacts with an Arabidopsis mitochondrial-localized kinesin motor protein. Kinesin mutant plants show reduced disease symptoms after pathogen infection, which can be complemented by actin-modifying agents. In total, our results support a model in which HopG1 induces changes in the organization of the actin cytoskeleton as part of its virulence function in promoting disease symptom development.
Collapse
Affiliation(s)
- Masaki Shimono
- Department of Plant, Soil and Microbial Sciences, Michigan State University, East Lansing, Michigan 48824 (M.S., Y.-J.L., B.D.); Graduate Program in Cell and Molecular Biology, Michigan State University, East Lansing, Michigan 48824 (K.P., S.Y.H., B.D.); Department of Energy Plant Research Laboratory, Michigan State University, East Lansing, Michigan 48824 (B.H.K., S.Y.H.); Department of Biological Sciences, Purdue University, West Lafayette, Indiana 47907-2064 (J.H.-R., C.J.S.); Department of Botany and Plant Pathology, Oregon State University, Corvallis, Oregon 97331-7303 (A.C., J.H.C.);Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon 97331-7303 (A.C., J.H.C.);Howard Hughes Medical Institute and the Gordon and Betty Moore Foundation, Michigan State University, East Lansing, Michigan 48824 (S.Y.H.); Center for Genome Research and Biocomputing, Oregon State University, Corvallis, Oregon 97331-7303 (J.H.C.);Bindley Bioscience Center, Discovery Park, Purdue University, West Lafayette, Indiana 47907 (C.J.S.); and Graduate Program in Genetics, Michigan State University, East Lansing, Michigan 48824 (B.D.)
| | - Yi-Ju Lu
- Department of Plant, Soil and Microbial Sciences, Michigan State University, East Lansing, Michigan 48824 (M.S., Y.-J.L., B.D.); Graduate Program in Cell and Molecular Biology, Michigan State University, East Lansing, Michigan 48824 (K.P., S.Y.H., B.D.); Department of Energy Plant Research Laboratory, Michigan State University, East Lansing, Michigan 48824 (B.H.K., S.Y.H.); Department of Biological Sciences, Purdue University, West Lafayette, Indiana 47907-2064 (J.H.-R., C.J.S.); Department of Botany and Plant Pathology, Oregon State University, Corvallis, Oregon 97331-7303 (A.C., J.H.C.);Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon 97331-7303 (A.C., J.H.C.);Howard Hughes Medical Institute and the Gordon and Betty Moore Foundation, Michigan State University, East Lansing, Michigan 48824 (S.Y.H.); Center for Genome Research and Biocomputing, Oregon State University, Corvallis, Oregon 97331-7303 (J.H.C.);Bindley Bioscience Center, Discovery Park, Purdue University, West Lafayette, Indiana 47907 (C.J.S.); and Graduate Program in Genetics, Michigan State University, East Lansing, Michigan 48824 (B.D.)
| | - Katie Porter
- Department of Plant, Soil and Microbial Sciences, Michigan State University, East Lansing, Michigan 48824 (M.S., Y.-J.L., B.D.); Graduate Program in Cell and Molecular Biology, Michigan State University, East Lansing, Michigan 48824 (K.P., S.Y.H., B.D.); Department of Energy Plant Research Laboratory, Michigan State University, East Lansing, Michigan 48824 (B.H.K., S.Y.H.); Department of Biological Sciences, Purdue University, West Lafayette, Indiana 47907-2064 (J.H.-R., C.J.S.); Department of Botany and Plant Pathology, Oregon State University, Corvallis, Oregon 97331-7303 (A.C., J.H.C.);Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon 97331-7303 (A.C., J.H.C.);Howard Hughes Medical Institute and the Gordon and Betty Moore Foundation, Michigan State University, East Lansing, Michigan 48824 (S.Y.H.); Center for Genome Research and Biocomputing, Oregon State University, Corvallis, Oregon 97331-7303 (J.H.C.);Bindley Bioscience Center, Discovery Park, Purdue University, West Lafayette, Indiana 47907 (C.J.S.); and Graduate Program in Genetics, Michigan State University, East Lansing, Michigan 48824 (B.D.)
| | - Brian H Kvitko
- Department of Plant, Soil and Microbial Sciences, Michigan State University, East Lansing, Michigan 48824 (M.S., Y.-J.L., B.D.); Graduate Program in Cell and Molecular Biology, Michigan State University, East Lansing, Michigan 48824 (K.P., S.Y.H., B.D.); Department of Energy Plant Research Laboratory, Michigan State University, East Lansing, Michigan 48824 (B.H.K., S.Y.H.); Department of Biological Sciences, Purdue University, West Lafayette, Indiana 47907-2064 (J.H.-R., C.J.S.); Department of Botany and Plant Pathology, Oregon State University, Corvallis, Oregon 97331-7303 (A.C., J.H.C.);Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon 97331-7303 (A.C., J.H.C.);Howard Hughes Medical Institute and the Gordon and Betty Moore Foundation, Michigan State University, East Lansing, Michigan 48824 (S.Y.H.); Center for Genome Research and Biocomputing, Oregon State University, Corvallis, Oregon 97331-7303 (J.H.C.);Bindley Bioscience Center, Discovery Park, Purdue University, West Lafayette, Indiana 47907 (C.J.S.); and Graduate Program in Genetics, Michigan State University, East Lansing, Michigan 48824 (B.D.)
| | - Jessica Henty-Ridilla
- Department of Plant, Soil and Microbial Sciences, Michigan State University, East Lansing, Michigan 48824 (M.S., Y.-J.L., B.D.); Graduate Program in Cell and Molecular Biology, Michigan State University, East Lansing, Michigan 48824 (K.P., S.Y.H., B.D.); Department of Energy Plant Research Laboratory, Michigan State University, East Lansing, Michigan 48824 (B.H.K., S.Y.H.); Department of Biological Sciences, Purdue University, West Lafayette, Indiana 47907-2064 (J.H.-R., C.J.S.); Department of Botany and Plant Pathology, Oregon State University, Corvallis, Oregon 97331-7303 (A.C., J.H.C.);Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon 97331-7303 (A.C., J.H.C.);Howard Hughes Medical Institute and the Gordon and Betty Moore Foundation, Michigan State University, East Lansing, Michigan 48824 (S.Y.H.); Center for Genome Research and Biocomputing, Oregon State University, Corvallis, Oregon 97331-7303 (J.H.C.);Bindley Bioscience Center, Discovery Park, Purdue University, West Lafayette, Indiana 47907 (C.J.S.); and Graduate Program in Genetics, Michigan State University, East Lansing, Michigan 48824 (B.D.)
| | - Allison Creason
- Department of Plant, Soil and Microbial Sciences, Michigan State University, East Lansing, Michigan 48824 (M.S., Y.-J.L., B.D.); Graduate Program in Cell and Molecular Biology, Michigan State University, East Lansing, Michigan 48824 (K.P., S.Y.H., B.D.); Department of Energy Plant Research Laboratory, Michigan State University, East Lansing, Michigan 48824 (B.H.K., S.Y.H.); Department of Biological Sciences, Purdue University, West Lafayette, Indiana 47907-2064 (J.H.-R., C.J.S.); Department of Botany and Plant Pathology, Oregon State University, Corvallis, Oregon 97331-7303 (A.C., J.H.C.);Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon 97331-7303 (A.C., J.H.C.);Howard Hughes Medical Institute and the Gordon and Betty Moore Foundation, Michigan State University, East Lansing, Michigan 48824 (S.Y.H.); Center for Genome Research and Biocomputing, Oregon State University, Corvallis, Oregon 97331-7303 (J.H.C.);Bindley Bioscience Center, Discovery Park, Purdue University, West Lafayette, Indiana 47907 (C.J.S.); and Graduate Program in Genetics, Michigan State University, East Lansing, Michigan 48824 (B.D.)
| | - Sheng Yang He
- Department of Plant, Soil and Microbial Sciences, Michigan State University, East Lansing, Michigan 48824 (M.S., Y.-J.L., B.D.); Graduate Program in Cell and Molecular Biology, Michigan State University, East Lansing, Michigan 48824 (K.P., S.Y.H., B.D.); Department of Energy Plant Research Laboratory, Michigan State University, East Lansing, Michigan 48824 (B.H.K., S.Y.H.); Department of Biological Sciences, Purdue University, West Lafayette, Indiana 47907-2064 (J.H.-R., C.J.S.); Department of Botany and Plant Pathology, Oregon State University, Corvallis, Oregon 97331-7303 (A.C., J.H.C.);Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon 97331-7303 (A.C., J.H.C.);Howard Hughes Medical Institute and the Gordon and Betty Moore Foundation, Michigan State University, East Lansing, Michigan 48824 (S.Y.H.); Center for Genome Research and Biocomputing, Oregon State University, Corvallis, Oregon 97331-7303 (J.H.C.);Bindley Bioscience Center, Discovery Park, Purdue University, West Lafayette, Indiana 47907 (C.J.S.); and Graduate Program in Genetics, Michigan State University, East Lansing, Michigan 48824 (B.D.)
| | - Jeff H Chang
- Department of Plant, Soil and Microbial Sciences, Michigan State University, East Lansing, Michigan 48824 (M.S., Y.-J.L., B.D.); Graduate Program in Cell and Molecular Biology, Michigan State University, East Lansing, Michigan 48824 (K.P., S.Y.H., B.D.); Department of Energy Plant Research Laboratory, Michigan State University, East Lansing, Michigan 48824 (B.H.K., S.Y.H.); Department of Biological Sciences, Purdue University, West Lafayette, Indiana 47907-2064 (J.H.-R., C.J.S.); Department of Botany and Plant Pathology, Oregon State University, Corvallis, Oregon 97331-7303 (A.C., J.H.C.);Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon 97331-7303 (A.C., J.H.C.);Howard Hughes Medical Institute and the Gordon and Betty Moore Foundation, Michigan State University, East Lansing, Michigan 48824 (S.Y.H.); Center for Genome Research and Biocomputing, Oregon State University, Corvallis, Oregon 97331-7303 (J.H.C.);Bindley Bioscience Center, Discovery Park, Purdue University, West Lafayette, Indiana 47907 (C.J.S.); and Graduate Program in Genetics, Michigan State University, East Lansing, Michigan 48824 (B.D.)
| | - Christopher J Staiger
- Department of Plant, Soil and Microbial Sciences, Michigan State University, East Lansing, Michigan 48824 (M.S., Y.-J.L., B.D.); Graduate Program in Cell and Molecular Biology, Michigan State University, East Lansing, Michigan 48824 (K.P., S.Y.H., B.D.); Department of Energy Plant Research Laboratory, Michigan State University, East Lansing, Michigan 48824 (B.H.K., S.Y.H.); Department of Biological Sciences, Purdue University, West Lafayette, Indiana 47907-2064 (J.H.-R., C.J.S.); Department of Botany and Plant Pathology, Oregon State University, Corvallis, Oregon 97331-7303 (A.C., J.H.C.);Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon 97331-7303 (A.C., J.H.C.);Howard Hughes Medical Institute and the Gordon and Betty Moore Foundation, Michigan State University, East Lansing, Michigan 48824 (S.Y.H.); Center for Genome Research and Biocomputing, Oregon State University, Corvallis, Oregon 97331-7303 (J.H.C.);Bindley Bioscience Center, Discovery Park, Purdue University, West Lafayette, Indiana 47907 (C.J.S.); and Graduate Program in Genetics, Michigan State University, East Lansing, Michigan 48824 (B.D.)
| | - Brad Day
- Department of Plant, Soil and Microbial Sciences, Michigan State University, East Lansing, Michigan 48824 (M.S., Y.-J.L., B.D.); Graduate Program in Cell and Molecular Biology, Michigan State University, East Lansing, Michigan 48824 (K.P., S.Y.H., B.D.); Department of Energy Plant Research Laboratory, Michigan State University, East Lansing, Michigan 48824 (B.H.K., S.Y.H.); Department of Biological Sciences, Purdue University, West Lafayette, Indiana 47907-2064 (J.H.-R., C.J.S.); Department of Botany and Plant Pathology, Oregon State University, Corvallis, Oregon 97331-7303 (A.C., J.H.C.);Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon 97331-7303 (A.C., J.H.C.);Howard Hughes Medical Institute and the Gordon and Betty Moore Foundation, Michigan State University, East Lansing, Michigan 48824 (S.Y.H.); Center for Genome Research and Biocomputing, Oregon State University, Corvallis, Oregon 97331-7303 (J.H.C.);Bindley Bioscience Center, Discovery Park, Purdue University, West Lafayette, Indiana 47907 (C.J.S.); and Graduate Program in Genetics, Michigan State University, East Lansing, Michigan 48824 (B.D.)
| |
Collapse
|
38
|
Zhang R, Chang M, Zhang M, Wu Y, Qu X, Huang S. The Structurally Plastic CH2 Domain Is Linked to Distinct Functions of Fimbrins/Plastins. J Biol Chem 2016; 291:17881-96. [PMID: 27261463 DOI: 10.1074/jbc.m116.730069] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Indexed: 01/08/2023] Open
Abstract
Fimbrins/plastins have been implicated in the generation of distinct actin structures, which are linked to different cellular processes. Historically, fimbrins/plastins were mainly considered as generating tight actin bundles. Here, we demonstrate that different members of the fimbrin/plastin family have diverged biochemically during evolution to generate either tight actin bundles or loose networks with distinct biochemical and biophysical properties. Using the phylogenetically and functionally distinct Arabidopsis fimbrins FIM4 and FIM5 we found that FIM4 generates both actin bundles and cross-linked actin filaments, whereas FIM5 only generates actin bundles. The distinct functions of FIM4 and FIM5 are clearly observed at single-filament resolution. Domain swapping experiments showed that cooperation between the conformationally plastic calponin-homology domain 2 (CH2) and the N-terminal headpiece determines the function of the full-length protein. Our study suggests that the structural plasticity of fimbrins/plastins has biologically meaningful consequences, and provides novel insights into the structure-function relationship of fimbrins/plastins as well as shedding light on how cells generate distinct actin structures.
Collapse
Affiliation(s)
- Ruihui Zhang
- From the Key Laboratory of Plant Molecular Physiology, Institute of Botany, Chinese Academy of Sciences, Beijing 100093, the University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ming Chang
- the Center for Plant Biology, School of Life Sciences, Tsinghua University, Beijing 100084
| | - Meng Zhang
- From the Key Laboratory of Plant Molecular Physiology, Institute of Botany, Chinese Academy of Sciences, Beijing 100093, the University of Chinese Academy of Sciences, Beijing 100049, China
| | - Youjun Wu
- From the Key Laboratory of Plant Molecular Physiology, Institute of Botany, Chinese Academy of Sciences, Beijing 100093
| | - Xiaolu Qu
- the Center for Plant Biology, School of Life Sciences, Tsinghua University, Beijing 100084, the Tsinghua-Peking Joint Center for Life Sciences, Beijing 100084, and
| | - Shanjin Huang
- From the Key Laboratory of Plant Molecular Physiology, Institute of Botany, Chinese Academy of Sciences, Beijing 100093, the Center for Plant Biology, School of Life Sciences, Tsinghua University, Beijing 100084,
| |
Collapse
|
39
|
Miarelli M, Drumo R, Signorelli F, Marchitelli C, Pavone S, Pesciaroli M, Ruggieri J, Chirullo B, Ammendola S, Battistoni A, Alborali GL, Manuali E, Pasquali P. Salmonella Typhimurium infection primes a nutriprive mechanism in piglets. Vet Microbiol 2016; 186:117-25. [DOI: 10.1016/j.vetmic.2016.02.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 02/09/2016] [Accepted: 02/11/2016] [Indexed: 12/14/2022]
|
40
|
Ahmed HA, El-Hofy FI, Shafik SM, Abdelrahman MA, Elsaid GA. Characterization of Virulence-Associated Genes, Antimicrobial Resistance Genes, and Class 1 Integrons in Salmonella enterica serovar Typhimurium Isolates from Chicken Meat and Humans in Egypt. Foodborne Pathog Dis 2016; 13:281-8. [PMID: 26977940 DOI: 10.1089/fpd.2015.2097] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Foodborne pathogens are leading causes of illness especially in developing countries. The current study aimed to characterize virulence-associated genes and antimicrobial resistance in 30 Salmonella Typhimurium isolates of chicken and human origin at Mansoura, Egypt. The results showed that invA, avrA, mgtC, stn, and bcfC genes were identified in all the examined isolates, while 96.7% and 6.7% were positive for sopB and pef genes, respectively. The highest resistance frequencies of the isolates were to chloramphenicol and trimethoprim-sulfamethoxazole (73.3%, each), followed by streptomycin (56.7%), tetracycline and ampicillin (53.3%, each), and gentamicin (30%). However, only 2.7% of the isolates were resistant to cefotaxime and ceftriaxone each. Different resistance-associated genes, including blaTEM, aadB, aadC, aadA1, aadA2, floR, tetA(A), tetA(B), and sul1, were identified in Salmonella Typhimurium isolates with the respective frequencies of 53.3%, 6.7%, 23.3%, 46.7%, 63.3%, 73.3%, 60%, 20%, and 96.7%. None of the isolates was positive for blaSHV, blaOXA, and blaCMY genes. The results showed that the intI1 gene was detected in 24 (80%) of the examined Salmonella Typhimurium isolates. Class 1 integrons were found in 19 (79.2%) isolates that were intI1 positive. Seven integron profiles (namely: P-I to P-VII) were identified with P-V (gene cassette dfrA15, aadA2), the most prevalent profile. To the best of our knowledge, this is the first study to characterize the unusual gene cassette array dfrA12-OrfF-aadA27 from Salmonella Typhimurium isolates in Egypt.
Collapse
Affiliation(s)
- Heba A Ahmed
- 1 Department of Zoonoses, Faculty of Veterinary Medicine, Zagazig University , Zagazig, Egypt
| | - Fatma I El-Hofy
- 2 Department of Bacteriology, Immunology and Mycology, Faculty of Veterinary Medicine, Benha University , Benha, Egypt
| | - Saleh M Shafik
- 3 Department of Food Hygiene, Animal Health Research Institute , Mansoura, Egypt
| | | | - Gamilat A Elsaid
- 3 Department of Food Hygiene, Animal Health Research Institute , Mansoura, Egypt
| |
Collapse
|
41
|
MacDonald LC, O’Keefe S, Parnes MF, MacDonald H, Stretz L, Templer SJ, Wong EL, Berger BW. A Secreted Ankyrin-Repeat Protein from Clinical Stenotrophomonas maltophilia Isolates Disrupts Actin Cytoskeletal Structure. ACS Infect Dis 2016; 2:62-70. [PMID: 27622948 DOI: 10.1021/acsinfecdis.5b00103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Stenotrophomonas maltophilia is an emerging, multidrug-resistant pathogen of increasing importance for the immunocompromised, including cystic fibrosis patients. Despite its significance as an emerging pathogen, relatively little is known regarding the specific factors and mechanisms that contribute to its pathogenicity. We identify and characterize a putative ankyrin-repeat protein (Smlt3054) unique to clinical S. maltophilia isolates that binds F-actin in vitro and co-localizes with actin in transfected HEK293a cells. Smlt3054 is endogenously expressed and secreted from clinical S. maltophilia isolates, but not an environmental isolate (R551-3). The in vitro binding of Smlt3054 to F-actin resulted in a thickening of the filaments as observed by TEM. Ectopic expression of Smlt3054-GFP exhibits strong co-localization with F-actin, with distinct, retrograde F-actin waves specifically associated with Smlt3054 in individual cells as well as formation of dense, internal inclusions at the expense of retrograde F-actin waves. Collectively, our results point to an interaction between Smlt3054 and F-actin. Furthermore, as a potentially secreted protein unique to clinical S. maltophilia isolates, Smlt3054 may serve as a starting point for understanding the mechanisms by which S. maltophilia has become an emergent pathogen.
Collapse
Affiliation(s)
- Logan C. MacDonald
- Program in Bioengineering and Department
of Chemical and Biomolecular Engineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Sean O’Keefe
- Program in Bioengineering and Department
of Chemical and Biomolecular Engineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Mei-Fan Parnes
- Program in Bioengineering and Department
of Chemical and Biomolecular Engineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Hanlon MacDonald
- Program in Bioengineering and Department
of Chemical and Biomolecular Engineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Lindsey Stretz
- Program in Bioengineering and Department
of Chemical and Biomolecular Engineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Suzanne J. Templer
- Division of Infectious Disease, Lehigh Valley Health Network, 1250 South Cedar Crest Boulevard, Suite 200, Allentown, Pennsylvania 18103, United States
| | - Emily L. Wong
- Division of Infectious Disease, Lehigh Valley Health Network, 1250 South Cedar Crest Boulevard, Suite 200, Allentown, Pennsylvania 18103, United States
| | - Bryan W. Berger
- Program in Bioengineering and Department
of Chemical and Biomolecular Engineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| |
Collapse
|
42
|
Budak G, Eren Ozsoy O, Aydin Son Y, Can T, Tuncbag N. Reconstruction of the temporal signaling network in Salmonella-infected human cells. Front Microbiol 2015; 6:730. [PMID: 26257716 PMCID: PMC4507143 DOI: 10.3389/fmicb.2015.00730] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 07/03/2015] [Indexed: 12/02/2022] Open
Abstract
Salmonella enterica is a bacterial pathogen that usually infects its host through food sources. Translocation of the pathogen proteins into the host cells leads to changes in the signaling mechanism either by activating or inhibiting the host proteins. Given that the bacterial infection modifies the response network of the host, a more coherent view of the underlying biological processes and the signaling networks can be obtained by using a network modeling approach based on the reverse engineering principles. In this work, we have used a published temporal phosphoproteomic dataset of Salmonella-infected human cells and reconstructed the temporal signaling network of the human host by integrating the interactome and the phosphoproteomic dataset. We have combined two well-established network modeling frameworks, the Prize-collecting Steiner Forest (PCSF) approach and the Integer Linear Programming (ILP) based edge inference approach. The resulting network conserves the information on temporality, direction of interactions, while revealing hidden entities in the signaling, such as the SNARE binding, mTOR signaling, immune response, cytoskeleton organization, and apoptosis pathways. Targets of the Salmonella effectors in the host cells such as CDC42, RHOA, 14-3-3δ, Syntaxin family, Oxysterol-binding proteins were included in the reconstructed signaling network although they were not present in the initial phosphoproteomic data. We believe that integrated approaches, such as the one presented here, have a high potential for the identification of clinical targets in infectious diseases, especially in the Salmonella infections.
Collapse
Affiliation(s)
- Gungor Budak
- Department of Health Informatics, Graduate School of Informatics, Middle East Technical University Ankara, Turkey
| | - Oyku Eren Ozsoy
- Department of Health Informatics, Graduate School of Informatics, Middle East Technical University Ankara, Turkey
| | - Yesim Aydin Son
- Department of Health Informatics, Graduate School of Informatics, Middle East Technical University Ankara, Turkey
| | - Tolga Can
- Department of Computer Engineering, College of Engineering, Middle East Technical University Ankara, Turkey
| | - Nurcan Tuncbag
- Department of Health Informatics, Graduate School of Informatics, Middle East Technical University Ankara, Turkey
| |
Collapse
|
43
|
Ma S, Schroeder B, Sun C, Loufakis DN, Cao Z, Sriranganathan N, Lu C. Electroporation-based delivery of cell-penetrating peptide conjugates of peptide nucleic acids for antisense inhibition of intracellular bacteria. Integr Biol (Camb) 2015; 6:973-8. [PMID: 25160797 DOI: 10.1039/c4ib00172a] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Cell penetrating peptides (CPPs) have been used for a myriad of cellular delivery applications and were recently explored for delivery of antisense agents such as peptide nucleic acids (PNAs) for bacterial inhibition. Although these molecular systems (i.e. CPP-PNAs) have shown ability to inhibit growth of bacterial cultures in vitro, they show limited effectiveness in killing encapsulated intracellular bacteria in mammalian cells such as macrophages, presumably due to difficulty involved in the endosomal escape of the reagents. In this report, we show that electroporation delivery dramatically increases the bioavailability of CPP-PNAs to kill Salmonella enterica serovar Typhimurium LT2 inside macrophages. Electroporation delivers the molecules without involving endocytosis and greatly increases the antisense effect. The decrease in the average number of Salmonella per macrophage under a 1200 V cm(-1) and 5 ms pulse was a factor of 9 higher than that without electroporation (in an experiment with a multiplicity of infection of 2 : 1). Our results suggest that electroporation is an effective approach for a wide range of applications involving CPP-based delivery. The microfluidic format will allow convenient functional screening and testing of PNA-based reagents for antisense applications.
Collapse
Affiliation(s)
- Sai Ma
- School of Biomedical Engineering and Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Ali SS, Soo J, Rao C, Leung AS, Ngai DHM, Ensminger AW, Navarre WW. Silencing by H-NS potentiated the evolution of Salmonella. PLoS Pathog 2014; 10:e1004500. [PMID: 25375226 PMCID: PMC4223078 DOI: 10.1371/journal.ppat.1004500] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 10/02/2014] [Indexed: 11/17/2022] Open
Abstract
The bacterial H-NS protein silences expression from sequences with higher AT-content than the host genome and is believed to buffer the fitness consequences associated with foreign gene acquisition. Loss of H-NS results in severe growth defects in Salmonella, but the underlying reasons were unclear. An experimental evolution approach was employed to determine which secondary mutations could compensate for the loss of H-NS in Salmonella. Six independently derived S. Typhimurium hns mutant strains were serially passaged for 300 generations prior to whole genome sequencing. Growth rates of all lineages dramatically improved during the course of the experiment. Each of the hns mutant lineages acquired missense mutations in the gene encoding the H-NS paralog StpA encoding a poorly understood H-NS paralog, while 5 of the mutant lineages acquired deletions in the genes encoding the Salmonella Pathogenicity Island-1 (SPI-1) Type 3 secretion system critical to invoke inflammation. We further demonstrate that SPI-1 misregulation is a primary contributor to the decreased fitness in Salmonella hns mutants. Three of the lineages acquired additional loss of function mutations in the PhoPQ virulence regulatory system. Similarly passaged wild type Salmonella lineages did not acquire these mutations. The stpA missense mutations arose in the oligomerization domain and generated proteins that could compensate for the loss of H-NS to varying degrees. StpA variants most able to functionally substitute for H-NS displayed altered DNA binding and oligomerization properties that resembled those of H-NS. These findings indicate that H-NS was central to the evolution of the Salmonellae by buffering the negative fitness consequences caused by the secretion system that is the defining characteristic of the species.
Collapse
Affiliation(s)
- Sabrina S. Ali
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Jeremy Soo
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Chitong Rao
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Andrea S. Leung
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - David Hon-Man Ngai
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | | | | |
Collapse
|
45
|
Bachmann NL, Petty NK, Ben Zakour NL, Szubert JM, Savill J, Beatson SA. Genome analysis and CRISPR typing of Salmonella enterica serovar Virchow. BMC Genomics 2014; 15:389. [PMID: 24885207 PMCID: PMC4042001 DOI: 10.1186/1471-2164-15-389] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 05/02/2014] [Indexed: 12/11/2022] Open
Abstract
Background Salmonella enterica subsp. enterica serovar Virchow has been recognized as a significant health burden in Asia, Australia and Europe. In addition to its global distribution, S. Virchow is clinically significant due to the frequency at which it causes invasive infections and its association with outbreaks arising from food-borne transmission. Here, we examine the genome of an invasive isolate of S. Virchow SVQ1 (phage type 8) from an outbreak in southeast Queensland, Australia. In addition to identifying new potential genotyping targets that could be used for discriminating between S. Virchow strains in outbreak scenarios, we also aimed to carry out a comprehensive comparative analysis of the S. Virchow genomes. Results Genome comparisons between S. Virchow SVQ1 and S. Virchow SL491, a previously published strain, identified a high degree of genomic similarity between the two strains with fewer than 200 single nucleotide differences. Clustered Regularly Interspaced Palindromic Repeats (CRISPR) regions were identified as a highly variable region that could be used to discriminate between S. Virchow isolates. We amplified and sequenced the CRISPR regions of fifteen S. Virchow isolates collected from seven different outbreaks across Australia. We observed three allelic types of the CRISPR region from these isolates based on the presence/absence of the spacers and were able to discriminate S. Virchow phage type 8 isolates originating from different outbreaks. A comparison with 27 published Salmonella genomes found that the S. Virchow SVQ1 genome encodes 11 previously described Salmonella Pathogenicity Islands (SPI), as well as additional genomic islands including a remnant integrative conjugative element that is distinct from SPI-7. In addition, the S. Virchow genome possesses a novel prophage that encodes the Type III secretion system effector protein SopE, a key Salmonella virulence factor. The prophage shares very little similarity to the SopE prophages found in other Salmonella serovars suggesting an independent acquisition of sopE. Conclusions The availability of this genome will serve as a genome template and facilitate further studies on understanding the virulence and global distribution of the S. Virchow serovar, as well as the development of genotyping methods for outbreak investigations. Electronic supplementary material The online version of this article (doi:10.1186/1471-2164-15-389) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | - John Savill
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia.
| | | |
Collapse
|
46
|
Salmonella enterica invasion of polarized epithelial cells is a highly cooperative effort. Infect Immun 2014; 82:2657-67. [PMID: 24711567 DOI: 10.1128/iai.00023-14] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The invasion of polarized epithelial cells by Salmonella enterica requires the cooperative activity of the Salmonella pathogenicity island 1 (SPI1)-encoded type III secretion system (T3SS) and the SPI4-encoded adhesin SiiE. The invasion of polarized cells is more efficient than that of nonpolarized cells, and we observed the formation of clusters of bacteria on infected cells. Here we demonstrate that the invasion of polarized cells is a highly cooperative activity. Using a novel live-cell imaging approach, we visualized the cooperative entry of multiple bacteria into ruffles induced on the apical surfaces of polarized cells. The induction of membrane ruffles by activity of Salmonella enables otherwise noninvasive mutant strains to enter polarized host cells. Bacterial motility and chemotaxis were of lower importance for cooperativity in polarized-cell invasion. We propose that cooperative invasion is a key factor for the very efficient entry into polarized cells and a factor contributing to epithelial damage and intestinal inflammation.
Collapse
|
47
|
What a difference a Dalton makes: bacterial virulence factors modulate eukaryotic host cell signaling systems via deamidation. Microbiol Mol Biol Rev 2014; 77:527-39. [PMID: 24006474 DOI: 10.1128/mmbr.00013-13] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Pathogenic bacteria commonly deploy enzymes to promote virulence. These enzymes can modulate the functions of host cell targets. While the actions of some enzymes can be very obvious (e.g., digesting plant cell walls), others have more subtle activities. Depending on the lifestyle of the bacteria, these subtle modifications can be crucially important for pathogenesis. In particular, if bacteria rely on a living host, subtle mechanisms to alter host cellular function are likely to dominate. Several bacterial virulence factors have evolved to use enzymatic deamidation as a subtle posttranslational mechanism to modify the functions of host protein targets. Deamidation is the irreversible conversion of the amino acids glutamine and asparagine to glutamic acid and aspartic acid, respectively. Interestingly, all currently characterized bacterial deamidases affect the function of the target protein by modifying a single glutamine residue in the sequence. Deamidation of target host proteins can disrupt host signaling and downstream processes by either activating or inactivating the target. Despite the subtlety of this modification, it has been shown to cause dramatic, context-dependent effects on host cells. Several crystal structures of bacterial deamidases have been solved. All are members of the papain-like superfamily and display a cysteine-based catalytic triad. However, these proteins form distinct structural subfamilies and feature combinations of modular domains of various functions. Based on the diverse pathogens that use deamidation as a mechanism to promote virulence and the recent identification of multiple deamidases, it is clear that this enzymatic activity is emerging as an important and widespread feature in bacterial pathogenesis.
Collapse
|
48
|
Ruan HH, Li Y, Zhang XX, Liu Q, Ren H, Zhang KS, Zhao H. Identification of TRAF6 as a ubiquitin ligase engaged in the ubiquitination of SopB, a virulence effector protein secreted by Salmonella typhimurium. Biochem Biophys Res Commun 2014; 447:172-7. [PMID: 24704445 DOI: 10.1016/j.bbrc.2014.03.126] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 03/25/2014] [Indexed: 10/25/2022]
Abstract
The phosphoinositide phosphatase SopB is one of the effectors injected by Salmonella typhimurium (S.typhimurium) that diversifies its function through a ubiquitin-dependent differential localization. However, it is unclear which E3 ubiquitin ligase is responsible for ubiquitination of SopB. Based on the E1-E2-E3 trio of enzymes responsible for the ubiquitin activation and translocation to substrate proteins, we constructed an in vitro assay of SopB ubiquitination. Using this assay, we purified an E3 ubiquitin ligase, TRAF6, from the Henle-407 S100 extraction that may be responsible for the ubiquitination of SopB. To investigate the functional correlation of TRAF6, we showed that recombinant TRAF6 specifically ubiquitinates SopB in a dose-dependent manner in vitro. Upon infection, the ubiquitination of SopB was absolutely blocked by TRAF6 deletion, as shown in Traf6(-/-) mouse embryonic fibroblasts (MEFs) compared with Traf6(+/+) MEFs. However, the ectopic expression of TRAF6 in Traf6(-/-) MEFs rescued the two species of ubiquitin-conjugated SopB, which strengthens the role of TRAF6 in SopB ubiquitination. The analysis of E2 revealed that UbcH5c and not other E2 conjugating enzymes are required for TRAF6-mediated SopB ubiquitination both in vitro and in vivo. In summary, these results suggest the relevance of UbcH5c/TRAF6 in SopB during S.typhimurium infection and thereby imply that S.typhimurium has evolved a mechanism of utilizing the host's E3 ubiquitin ligase to modify and modulate the function of its effector protein in order to ensure pathogen and host cell survival.
Collapse
Affiliation(s)
- Hai-Hua Ruan
- Tianjin Key Laboratory of Food Science and Biotechnology, College of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300134, China.
| | - Ye Li
- Tianjin Key Laboratory of Food Science and Biotechnology, College of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300134, China
| | - Xi-Xuan Zhang
- Tianjin Key Laboratory of Food Science and Biotechnology, College of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300134, China
| | - Qiang Liu
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin Key Laboratory of Molecular Nuclear Medicine, Tianjin 300192, China
| | - He Ren
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Hospital, Tianjin 300071, China
| | - Kun-Sheng Zhang
- Tianjin Key Laboratory of Food Science and Biotechnology, College of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300134, China
| | - Hui Zhao
- Tianjin Key Laboratory of Food Science and Biotechnology, College of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300134, China.
| |
Collapse
|
49
|
Yukuhiro F, Miyoshi T, Noda H. Actin-mediated transovarial transmission of a yeastlike symbiont in the brown planthopper. JOURNAL OF INSECT PHYSIOLOGY 2014; 60:111-117. [PMID: 24315960 DOI: 10.1016/j.jinsphys.2013.11.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 11/25/2013] [Accepted: 11/25/2013] [Indexed: 06/02/2023]
Abstract
The brown planthopper Nilaparvata lugens (Hemiptera, Delphacidae) harbors an obligate endosymbiont called the yeastlike symbiont (YLS) in their abdominal fat body. YLS, a filamentous ascomycete belonging to the family Clavicipitaceae, does not spend any part of its life cycle outside the planthopper's body. The YLS is transferred to the next generation via transovarial transmission; it enters the epithelial plug at the posterior end of the host female's ovariole and is transferred to her offspring. In the present study, microscopic examination revealed that actin filaments play an important role in the transmission of YLS. An irregular cell protrusion on the surface of the epithelial plug facilitated the uptake of the YLS, which was then incorporated into the epithelial plug cell. Actin assembly apparently produces the protrusion and actin appears to participate in almost every stage of the process, from the entry of the YLS into the epithelial plug to its delivery to the oocyte. The epithelial plug employs a recognition system for YLS, which drastically changes the cell surface structure to enable the YLS to enter the ovariole.
Collapse
Affiliation(s)
- Fumiko Yukuhiro
- National Institute of Agrobiological Sciences, Owashi, Tsukuba, Ibaraki 305-8634, Japan
| | - Takeharu Miyoshi
- National Institute of Agrobiological Sciences, Owashi, Tsukuba, Ibaraki 305-8634, Japan
| | - Hiroaki Noda
- National Institute of Agrobiological Sciences, Owashi, Tsukuba, Ibaraki 305-8634, Japan.
| |
Collapse
|
50
|
Borges KA, Furian TQ, Borsoi A, Moraes HL, Salle CT, Nascimento VP. Detection of virulence-associated genes in Salmonella Enteritidis isolates from chicken in South of Brazil. PESQUISA VETERINARIA BRASILEIRA 2013. [DOI: 10.1590/s0100-736x2013001200004] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Salmonella spp. are considered the main agents of foodborne disease and Salmonella Enteritidis is one of the most frequently isolated serovars worldwide. The virulence of Salmonella spp. and their interaction with the host are complex processes involving virulence factors to overcome host defenses. The purpose of this study was to detect virulence genes in S. Enteritidis isolates from poultry in the South of Brazil. PCR-based assays were developed in order to detect nine genes (lpfA, agfA, sefA, invA, hilA, avrA, sopE, sivH and spvC) associated with the virulence in eighty-four isolates of S. Enteritidis isolated from poultry. The invA, hilA, sivH, sefA and avrA genes were present in 100% of the isolates; lpfA and sopE were present in 99%; agfA was present in 96%; and the spvC gene was present in 92%. It was possible to characterize the isolates with four different genetic profiles (P1, P2, P3 and P4), as it follows: P1, positive for all genes; P2, negative only for spvC; P3, negative for agfA; and P4, negative for lpfA, spvC and sopE. The most prevalent profile was P1, which was present in 88% of the isolates. Although all isolates belong to the same serovar, it was possible to observe variations in the presence of these virulence-associated genes between different isolates. The characterization of the mechanisms of virulence circulating in the population of Salmonella Enteritidis is important for a better understanding of its biology and pathogenicity. The frequency of these genes and the establishment of genetic profiles can be used to determine patterns of virulence. These patterns, associated with in vivo studies, may help develop tools to predict the ability of virulence of different strains.
Collapse
|