1
|
Li X, Ormsby MJ, Fallata G, Meikle LM, Walker D, Xu D, Wall DM. PF-431396 hydrate inhibition of kinase phosphorylation during adherent-invasive Escherichia coli infection inhibits intra-macrophage replication and inflammatory cytokine release. MICROBIOLOGY (READING, ENGLAND) 2023; 169. [PMID: 37311220 DOI: 10.1099/mic.0.001337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Adherent-invasive Escherichia coli (AIEC) have been implicated in the aetiology of Crohn's disease (CD). They are characterized by an ability to adhere to and invade intestinal epithelial cells, and to replicate intracellularly in macrophages resulting in inflammation. Proline-rich tyrosine kinase 2 (PYK2) has previously been identified as a risk locus for inflammatory bowel disease and a regulator of intestinal inflammation. It is overexpressed in patients with colorectal cancer, a major long-term complication of CD. Here we show that Pyk2 levels are significantly increased during AIEC infection of murine macrophages while the inhibitor PF-431396 hydrate, which blocks Pyk2 activation, significantly decreased intramacrophage AIEC numbers. Imaging flow cytometry indicated that Pyk2 inhibition blocked intramacrophage replication of AIEC with no change in the overall number of infected cells, but a significant reduction in bacterial burden per cell. This reduction in intracellular bacteria resulted in a 20-fold decrease in tumour necrosis factor α secretion by cells post-AIEC infection. These data demonstrate a key role for Pyk2 in modulating AIEC intracellular replication and associated inflammation and may provide a new avenue for future therapeutic intervention in CD.
Collapse
Affiliation(s)
- Xiang Li
- School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, Sir Graeme Davies Building, University of Glasgow, Glasgow G12 8TA, UK
| | - Michael J Ormsby
- School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, Sir Graeme Davies Building, University of Glasgow, Glasgow G12 8TA, UK
- Present address: Biological and Environmental Sciences, Faculty of Natural Science, University of Stirling, Stirling, FK49 4LA, UK
| | - Ghaith Fallata
- School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, Sir Graeme Davies Building, University of Glasgow, Glasgow G12 8TA, UK
- Department of Basic Science, College of Science and Health Professions, King Saud bin Abdulaziz University for Health Sciences, Jeddah 22384, Saudi Arabia
| | - Lynsey M Meikle
- School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, Sir Graeme Davies Building, University of Glasgow, Glasgow G12 8TA, UK
| | - Daniel Walker
- Strathclyde Institute for Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK
| | - Damo Xu
- School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, Sir Graeme Davies Building, University of Glasgow, Glasgow G12 8TA, UK
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen Key Laboratory of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, PR China
| | - Daniel M Wall
- School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, Sir Graeme Davies Building, University of Glasgow, Glasgow G12 8TA, UK
| |
Collapse
|
2
|
Mechanical Forces Govern Interactions of Host Cells with Intracellular Bacterial Pathogens. Microbiol Mol Biol Rev 2022; 86:e0009420. [PMID: 35285720 PMCID: PMC9199418 DOI: 10.1128/mmbr.00094-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
To combat infectious diseases, it is important to understand how host cells interact with bacterial pathogens. Signals conveyed from pathogen to host, and vice versa, may be either chemical or mechanical. While the molecular and biochemical basis of host-pathogen interactions has been extensively explored, relatively less is known about mechanical signals and responses in the context of those interactions. Nevertheless, a wide variety of bacterial pathogens appear to have developed mechanisms to alter the cellular biomechanics of their hosts in order to promote their survival and dissemination, and in turn many host responses to infection rely on mechanical alterations in host cells and tissues to limit the spread of infection. In this review, we present recent findings on how mechanical forces generated by host cells can promote or obstruct the dissemination of intracellular bacterial pathogens. In addition, we discuss how in vivo extracellular mechanical signals influence interactions between host cells and intracellular bacterial pathogens. Examples of such signals include shear stresses caused by fluid flow over the surface of cells and variable stiffness of the extracellular matrix on which cells are anchored. We highlight bioengineering-inspired tools and techniques that can be used to measure host cell mechanics during infection. These allow for the interrogation of how mechanical signals can modulate infection alongside biochemical signals. We hope that this review will inspire the microbiology community to embrace those tools in future studies so that host cell biomechanics can be more readily explored in the context of infection studies.
Collapse
|
3
|
Role of the Yersinia pseudotuberculosis Virulence Plasmid in Pathogen-Phagocyte Interactions in Mesenteric Lymph Nodes. EcoSal Plus 2021; 9:eESP00142021. [PMID: 34910573 DOI: 10.1128/ecosalplus.esp-0014-2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Yersinia pseudotuberculosis is an Enterobacteriaceae family member that is commonly transmitted by the fecal-oral route to cause infections. From the small intestine, Y. pseudotuberculosis can invade through Peyer's patches and lymph vessels to infect the mesenteric lymph nodes (MLNs). Infection of MLNs by Y. pseudotuberculosis results in the clinical presentation of mesenteric lymphadenitis. MLNs are important for immune responses to intestinal pathogens and microbiota in addition to their clinical relevance to Y. pseudotuberculosis infections. A characteristic of Y. pseudotuberculosis infection in MLNs is the formation of pyogranulomas. Pyogranulomas are composed of neutrophils, inflammatory monocytes, and lymphocytes surrounding extracellular microcolonies of Y. pseudotuberculosis. Key elements of the complex pathogen-host interaction in MLNs have been identified using mouse infection models. Y. pseudotuberculosis requires the virulence plasmid pYV to induce the formation of pyogranulomas in MLNs. The YadA adhesin and the Ysc-Yop type III secretion system (T3SS) are encoded on pYV. YadA mediates bacterial binding to host receptors, which engages the T3SS to preferentially translocate seven Yop effectors into phagocytes. The effectors promote pathogenesis by blocking innate immune defenses such as superoxide production, degranulation, and inflammasome activation, resulting in survival and growth of Y. pseudotuberculosis. On the other hand, certain effectors can trigger immune defenses in phagocytes. For example, YopJ triggers activation of caspase-8 and an apoptotic cell death response in monocytes within pyogranulomas that limits dissemination of Y. pseudotuberculosis from MLNs to the bloodstream. YopE can be processed as an antigen by phagocytes in MLNs, resulting in T and B cell responses to Y. pseudotuberculosis. Immune responses to Y. pseudotuberculosis in MLNs can also be detrimental to the host in the form of chronic lymphadenopathy. This review focuses on interactions between Y. pseudotuberculosis and phagocytes mediated by pYV that concurrently promote pathogenesis and host defense in MLNs. We propose that MLN pyogranulomas are immunological arenas in which opposing pYV-driven forces determine the outcome of infection in favor of the pathogen or host.
Collapse
|
4
|
Krachler AM, Sirisaengtaksin N, Monteith P, Paine CET, Coates CJ, Lim J. Defective phagocyte association during infection of Galleria mellonella with Yersinia pseudotuberculosis is detrimental to both insect host and microbe. Virulence 2021; 12:638-653. [PMID: 33550901 PMCID: PMC7889024 DOI: 10.1080/21505594.2021.1878672] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 11/03/2022] Open
Abstract
Adhesins facilitate bacterial colonization and invasion of host tissues and are considered virulence factors, but their impact on immune-mediated damage as a driver of pathogenesis remains unclear. Yersinia pseudotuberculosis encodes for a multivalent adhesion molecule (MAM), a mammalian cell entry (MCE) family protein and adhesin. MAMs are widespread in Gram-negative bacteria and enable enteric bacteria to colonize epithelial tissues. Their role in bacterial interactions with the host innate immune system and contribution to pathogenicity remains unclear. Here, we investigated howY. pseudotuberculosis MAM contributes to pathogenesis during infection of the Galleria mellonella insect model. We show that Y. pseudotuberculosis MAM is required for efficient bacterial binding and uptake by hemocytes, the host phagocytes. Y. pseudotuberculosis interactions with insect and mammalian phagocytes are determined by bacterial and host factors. Loss of MAM, and deficient microbe-phagocyte interaction, increased pathogenesis in G. mellonella. Diminished phagocyte association also led to increased bacterial clearance. Furthermore, Y. pseudotuberculosis that failed to engage phagocytes hyperactivated humoral immune responses, most notably melanin production. Despite clearing the pathogen, excessive melanization also increased phagocyte death and host mortality. Our findings provide a basis for further studies investigating how microbe- and host-factors integrate to drive pathogenesis in a tractable experimental system.
Collapse
Affiliation(s)
- Anne Marie Krachler
- Department of Microbiology and Molecular Genetics, University of Texas McGovern Medical School at Houston, Houston, TX, USA
| | - Natalie Sirisaengtaksin
- Department of Microbiology and Molecular Genetics, University of Texas McGovern Medical School at Houston, Houston, TX, USA
| | - Pauline Monteith
- Biological and Environmental Sciences, University of Stirling, Stirling, UK
| | - C. E. Timothy Paine
- School of Environmental and Rural Sciences, University of New England, Armidale, Australia
| | - Christopher J. Coates
- Department of Biosciences, College of Science, Swansea University, Swansea, Wales UK
| | - Jenson Lim
- Biological and Environmental Sciences, University of Stirling, Stirling, UK
| |
Collapse
|
5
|
Kaur D, Mukhopadhaya A. Outer membrane protein OmpV mediates Salmonella enterica serovar typhimurium adhesion to intestinal epithelial cells via fibronectin and α1β1 integrin. Cell Microbiol 2020; 22:e13172. [PMID: 32017350 DOI: 10.1111/cmi.13172] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 01/19/2020] [Accepted: 01/20/2020] [Indexed: 12/11/2022]
Abstract
Salmonella typhimurium is an invasive Gram-negative enteric bacterium, which causes salmonellosis, a type of gastroenteritis in humans and typhoid-like symptoms in mice. Upon entering through the contaminated food and water, S. typhimurium adheres, colonises, and invades intestinal epithelial cells (IECs) of the small intestine. In this study, we have shown that upon deletion of the outer membrane protein OmpV, there is a significant decrease in adherence of S. typhimurium to the IECs, indicating that OmpV is an important adhesin of S. typhimurium. Further, our study showed that OmpV binds to the extracellular matrix component fibronectin and signals through α1β1 integrin receptor on the IECs and OmpV-mediated activation of α1β1, resulting in the activation of focal adhesion kinase and F-actin modulation. Actin modulation is crucial for bacterial invasion. To the best of our knowledge, this is the first report of an adhesin mediated its effect through integrin in S. typhimurium. Further, we have observed a decrease in pathogenicity in terms of increased LD50 dose, lesser bacterial numbers in stool, and less colonisation of bacteria in different organs of mice infected with Δompv mutant compared with the wild-type bacteria, thus confirming the crucial role of OmpV in the pathogenesis of S. typhimurium.
Collapse
Affiliation(s)
- Deepinder Kaur
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Sahibzada Ajit Singh Nagar, India
| | - Arunika Mukhopadhaya
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Sahibzada Ajit Singh Nagar, India
| |
Collapse
|
6
|
Proline-Rich Protein Tyrosine Kinase 2 in Inflammation and Cancer. Cancers (Basel) 2018; 10:cancers10050139. [PMID: 29738483 PMCID: PMC5977112 DOI: 10.3390/cancers10050139] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 04/27/2018] [Accepted: 04/27/2018] [Indexed: 01/13/2023] Open
Abstract
Focal adhesion kinase (FAK) and its homologous FAK-related proline-rich tyrosine kinase 2 (Pyk2) contain the same domain, exhibit high sequence homology and are defined as a distinct family of non-receptor tyrosine kinases. This group of kinases plays critical roles in cytoskeletal dynamics and cell adhesion by regulating survival and growth signaling. This review summarizes the physiological and pathological functions of Pyk2 in inflammation and cancers. In particular, overexpression of Pyk2 in cancerous tissues is correlated with poor outcomes. Pyk2 stimulates multiple oncogenic signaling pathways, such as Wnt/β-catenin, PI3K/Akt, MAPK/ERK, and TGF-β/EGFR/VEGF, and facilitates carcinogenesis, migration, invasion, epithelial⁻mesenchymal transition and metastasis. Therefore, Pyk2 is a high-value therapeutic target and has clinical significance.
Collapse
|
7
|
Kim J, Fukuto HS, Brown DA, Bliska JB, London E. Effects of host cell sterol composition upon internalization of Yersinia pseudotuberculosis and clustered β1 integrin. J Biol Chem 2017; 293:1466-1479. [PMID: 29197826 DOI: 10.1074/jbc.m117.811224] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 11/29/2017] [Indexed: 11/06/2022] Open
Abstract
Yersinia pseudotuberculosis is a foodborne pathogenic bacterium that causes acute gastrointestinal illness, but its mechanisms of infection are incompletely described. We examined how host cell sterol composition affected Y. pseudotuberculosis uptake. To do this, we depleted or substituted cholesterol in human MDA-MB-231 epithelial cells with various alternative sterols. Decreasing host cell cholesterol significantly reduced pathogen internalization. When host cell cholesterol was substituted with various sterols, only desmosterol and 7-dehydrocholesterol supported internalization. This specificity was not due to sterol dependence of bacterial attachment to host cells, which was similar with all sterols studied. Because a key step in Y. pseudotuberculosis internalization is interaction of the bacterial adhesins invasin and YadA with host cell β1 integrin, we compared the sterol dependence of wildtype Y. pseudotuberculosis internalization with that of Δinv, ΔyadA, and ΔinvΔyadA mutant strains. YadA deletion decreased bacterial adherence to host cells, whereas invasin deletion had no effect. Nevertheless, host cell sterol substitution had a similar effect on internalization of these bacterial deletion strains as on the wildtype bacteria. The ΔinvΔyadA double mutant adhered least to cells and so was not significantly internalized. The sterol structure dependence of Y. pseudotuberculosis internalization differed from that of endocytosis, as monitored using antibody-clustered β1 integrin and previous studies on other proteins, which had a more permissive sterol dependence. This study suggests that agents could be designed to interfere with internalization of Yersinia without disturbing endocytosis.
Collapse
Affiliation(s)
- JiHyun Kim
- From the Departments of Biochemistry and Cell Biology and
| | - Hana S Fukuto
- Molecular Genetics and Microbiology and.,Center for Infectious Diseases, Stony Brook University, Stony Brook, New York 11794
| | | | - James B Bliska
- Molecular Genetics and Microbiology and.,Center for Infectious Diseases, Stony Brook University, Stony Brook, New York 11794
| | - Erwin London
- From the Departments of Biochemistry and Cell Biology and
| |
Collapse
|
8
|
Keller B, Mühlenkamp M, Deuschle E, Siegfried A, Mössner S, Schade J, Griesinger T, Katava N, Braunsdorf C, Fehrenbacher B, Jiménez‐Soto LF, Schaller M, Haas R, Genth H, Retta SF, Meyer H, Böttcher RT, Zent R, Schütz M, Autenrieth IB, Bohn E. Yersinia enterocolitica
exploits different pathways to accomplish adhesion and toxin injection into host cells. Cell Microbiol 2015; 17:1179-204. [DOI: 10.1111/cmi.12429] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Revised: 02/05/2015] [Accepted: 02/06/2015] [Indexed: 12/12/2022]
Affiliation(s)
- Birgit Keller
- Interfakultäres Institut für Mikrobiologie und Infektionsmedizin Eberhard Karls Universität Tübingen Germany
| | - Melanie Mühlenkamp
- Interfakultäres Institut für Mikrobiologie und Infektionsmedizin Eberhard Karls Universität Tübingen Germany
| | - Eva Deuschle
- Interfakultäres Institut für Mikrobiologie und Infektionsmedizin Eberhard Karls Universität Tübingen Germany
| | - Alexandra Siegfried
- Interfakultäres Institut für Mikrobiologie und Infektionsmedizin Eberhard Karls Universität Tübingen Germany
| | - Sara Mössner
- Interfakultäres Institut für Mikrobiologie und Infektionsmedizin Eberhard Karls Universität Tübingen Germany
| | - Jessica Schade
- Interfakultäres Institut für Mikrobiologie und Infektionsmedizin Eberhard Karls Universität Tübingen Germany
| | - Tanja Griesinger
- Interfakultäres Institut für Mikrobiologie und Infektionsmedizin Eberhard Karls Universität Tübingen Germany
| | - Nenad Katava
- Interfakultäres Institut für Mikrobiologie und Infektionsmedizin Eberhard Karls Universität Tübingen Germany
| | | | | | | | - Martin Schaller
- Department of Dermatology Eberhard Karls University Tübingen Germany
| | - Rainer Haas
- Max von Pettenkofer‐Institut Ludwig‐Maximilians University Munich Germany
| | - Harald Genth
- Institute of Toxicology Medical School Hannover Hannover Germany
| | - Saverio F. Retta
- Department of Clinical and Biological Sciences University of Torino Orbassano Italy
| | - Hannelore Meyer
- Max Planck Institut für Biochemie Martinsried Germany
- Institut für Medizinische Mikrobiologie, Immunologie und Hygiene Technische Universität München Germany
| | | | - Roy Zent
- Department of Medicine (Division of Nephrology) Vanderbilt University Medical Center Nashville TN USA
| | - Monika Schütz
- Interfakultäres Institut für Mikrobiologie und Infektionsmedizin Eberhard Karls Universität Tübingen Germany
- Department of Medicine (Division of Nephrology) Vanderbilt University Medical Center Nashville TN USA
| | - Ingo B. Autenrieth
- Interfakultäres Institut für Mikrobiologie und Infektionsmedizin Eberhard Karls Universität Tübingen Germany
- German Centre of Infection Research (DZIF) Partner Site Tübingen Germany
| | - Erwin Bohn
- Interfakultäres Institut für Mikrobiologie und Infektionsmedizin Eberhard Karls Universität Tübingen Germany
| |
Collapse
|
9
|
Gillenius E, Urban CF. The adhesive protein invasin of Yersinia pseudotuberculosis induces neutrophil extracellular traps via β1 integrins. Microbes Infect 2015; 17:327-36. [PMID: 25576025 DOI: 10.1016/j.micinf.2014.12.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 12/23/2014] [Accepted: 12/29/2014] [Indexed: 11/15/2022]
Abstract
Yersinia pseudotuberculosis adhesive protein invasin is crucial for the bacteria to cross the intestine epithelium by binding to β1 integrins on M-cells and gaining access to the underlying tissues. After the crossing invasin can bind to β1 integrins on other cell surfaces, however effector proteins delivered by the type III secretion system Y. pseudotuberculosis efficiently inhibit potential immune responses induced by this interaction. Here, we use mutant Y. pseudotuberculosis strains lacking the type III secretion system and additionally invasin-expressing Escherichia coli to analyze neutrophil responses towards invasin. Our data reveals that invasin induces production of reactive oxygen species and release of chromatin into the extracellular milieu, which we confirmed to be neutrophil extracellular traps by immunofluorescence microscopy. This was mediated through β1 integrins and was dependent on both the production of reactive oxygen species and signaling through phosphoinositide 3-kinase. We therefore have gained insight into a potential role of integrins in inflammation and infection clearance that has not previously been described, suggesting that targeting of β1 integrins could be utilized as an adjunctive therapy against yersiniosis.
Collapse
Affiliation(s)
- Erik Gillenius
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden; Laboratory for Molecular Infection Medicine, Sweden (MIMS) and Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
| | - Constantin F Urban
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden; Laboratory for Molecular Infection Medicine, Sweden (MIMS) and Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden.
| |
Collapse
|
10
|
Contribution of Crk adaptor proteins to host cell and bacteria interactions. BIOMED RESEARCH INTERNATIONAL 2014; 2014:372901. [PMID: 25506591 PMCID: PMC4260429 DOI: 10.1155/2014/372901] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 09/14/2014] [Indexed: 12/27/2022]
Abstract
The Crk adaptor family of proteins comprises the alternatively spliced CrkI and CrkII isoforms, as well as the paralog Crk-like (CrkL) protein, which is encoded by a different gene. Initially thought to be involved in signaling during apoptosis and cell adhesion, this ubiquitously expressed family of proteins is now known to play essential roles in integrating signals from a wide range of stimuli. In this review, we describe the structure and function of the different Crk proteins. We then focus on the emerging roles of Crk adaptors during Enterobacteriaceae pathogenesis, with special emphasis on the important human pathogens Salmonella, Shigella, Yersinia, and enteropathogenic Escherichia coli. Throughout, we remark on opportunities for future research into this intriguing family of proteins.
Collapse
|
11
|
Koch I, Dach K, Heesemann J, Hoffmann R. Yersinia enterocolitica inactivates NK cells. Int J Med Microbiol 2013; 303:433-42. [PMID: 23810728 DOI: 10.1016/j.ijmm.2013.05.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 05/07/2013] [Accepted: 05/20/2013] [Indexed: 12/16/2022] Open
Abstract
Natural Killer (NK) cells serve as an important source of proinflammatory cytokines early during infection. Hypothesizing that Yersinia enterocolitica might interact with and inactivate NK cells, we examined NK cell-Y. enterocolitica interactions in vitro and in vivo. Y. enterocolitica adheres to NK cells in an Invasin dependent manner and inhibits NK cell cytotoxicity and IFN-γ production induced by IL-12+IL-18 or IL-12 alone. YopP, an acetyltransferase known to inhibit MAPK and NFκB signaling, suppresses IL-12 and IL-12+IL-18 mediated IFN-γ production in NK cells by inhibiting phosphorylation of Tyk2 and STAT4 in addition to MAPK. YopP inhibits induction of all genes whose expression is induced by IL-12+IL-18 in NK cells. Y. enterocolitica-mediated adherence to and inactivation of NK cells also occurs after infection in vivo. Thus, we present the first report of a bacterial pathogen inactivating NK cells, and report interaction with Tyk2-STAT4 signaling as a novel function of YopP.
Collapse
Affiliation(s)
- Isabel Koch
- Ludwig Maximilians University, Max von Pettenkofer Institut, Department of Bacteriology, 80336 Munich, Germany
| | | | | | | |
Collapse
|
12
|
Integrins and small GTPases as modulators of phagocytosis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 302:321-54. [PMID: 23351714 DOI: 10.1016/b978-0-12-407699-0.00006-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Phagocytosis is the mechanism whereby cells engulf large particles. This process has long been recognized as a critical component of the innate immune response, which constitutes the organism's defense against microorganisms. In addition, phagocytic internalization of apoptotic cells or cell fragments plays important roles in tissue homeostasis and remodeling. Phagocytosis requires target interactions with receptors on the plasma membrane of the phagocytic cell. Integrins have been identified as important mediators of particle clearance, in addition to their well-established roles in cell adhesion, migration and mechanotransduction. Indeed, these ubiquitously expressed proteins impart phagocytic capacity to epithelial, endothelial and mesenchymal cell types. The importance of integrins in particle internalization is emphasized by the ability of microbial and viral pathogens to exploit their signaling pathways to invade host cells, and by the wide variety of disorders that arise from abnormalities in integrin-dependent phagocytic uptake.
Collapse
|
13
|
Abstract
Microbial adhesion is generally a complex process, involving multiple adhesins on a single microbe and their respective target receptors on host cells. In some situations, various adhesins of a microbe may co-operate in an apparently hierarchical and sequential manner whereby the first adhesive event triggers the target cell to express receptors for additional microbial adhesins. In other instances, adhesins may act in concert leading to high avidity interactions, often a prelude to cellular invasion and tissue penetration. Mechanisms used to target the host include both lectin-like interactions and protein-protein interactions; the latter are often highly specific for the host or a tissue within the host. This reflective chapter aims to offer a point of view on microbial adhesion by presenting some experiences and thoughts especially related to respiratory pathogens and explore if there can be any future hope of controlling bacterial infections via preventing adhesion or invasion stages of microbial pathogenesis.
Collapse
|
14
|
Roles of chaperone/usher pathways of Yersinia pestis in a murine model of plague and adhesion to host cells. Infect Immun 2012; 80:3490-500. [PMID: 22851745 DOI: 10.1128/iai.00434-12] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Yersinia pestis and many other Gram-negative pathogenic bacteria use the chaperone/usher (CU) pathway to assemble virulence-associated surface fibers termed pili or fimbriae. Y. pestis has two well-characterized CU pathways: the caf genes coding for the F1 capsule and the psa genes coding for the pH 6 antigen. The Y. pestis genome contains additional CU pathways that are capable of assembling pilus fibers, but the roles of these pathways in the pathogenesis of plague are not understood. We constructed deletion mutations in the usher genes for six of the additional Y. pestis CU pathways. The wild-type (WT) and usher deletion strains were compared in the murine bubonic (subcutaneous) and pneumonic (intranasal) plague infection models. Y. pestis strains containing deletions in CU pathways y0348-0352, y1858-1862, and y1869-1873 were attenuated for virulence compared to the WT strain by the intranasal, but not subcutaneous, routes of infection, suggesting specific roles for these pathways during pneumonic plague. We examined binding of the Y. pestis WT and usher deletion strains to A549 human lung epithelial cells, HEp-2 human cervical epithelial cells, and primary human and murine macrophages. Y. pestis CU pathways y0348-0352 and y1858-1862 were found to contribute to adhesion to all host cells tested, whereas pathway y1869-1873 was specific for binding to macrophages. The correlation between the virulence attenuation and host cell binding phenotypes of the usher deletion mutants identifies three of the additional CU pathways of Y. pestis as mediating interactions with host cells that are important for the pathogenesis of plague.
Collapse
|
15
|
Verove J, Bernarde C, Bohn YST, Boulay F, Rabiet MJ, Attree I, Cretin F. Injection of Pseudomonas aeruginosa Exo toxins into host cells can be modulated by host factors at the level of translocon assembly and/or activity. PLoS One 2012; 7:e30488. [PMID: 22299042 PMCID: PMC3267729 DOI: 10.1371/journal.pone.0030488] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2011] [Accepted: 12/20/2011] [Indexed: 01/03/2023] Open
Abstract
Pseudomonas aeruginosa type III secretion apparatus exports and translocates four exotoxins into the cytoplasm of the host cell. The translocation requires two hydrophobic bacterial proteins, PopB and PopD, that are found associated with host cell membranes following infection. In this work we examined the influence of host cell elements on exotoxin translocation efficiency. We developed a quantitative flow cytometry based assay of translocation that used protein fusions between either ExoS or ExoY and the ß-lactamase reporter enzyme. In parallel, association of translocon proteins with host plasma membranes was evaluated by immunodetection of PopB/D following sucrose gradient fractionation of membranes. A pro-myelocytic cell line (HL-60) and a pro-monocytic cell line (U937) were found resistant to toxin injection even though PopB/D associated with host cell plasma membranes. Differentiation of these cells to either macrophage- or neutrophil-like cell lines resulted in injection-sensitive phenotype without significantly changing the level of membrane-inserted translocon proteins. As previous in vitro studies have indicated that the lysis of liposomes by PopB and PopD requires both cholesterol and phosphatidyl-serine, we first examined the role of cholesterol in translocation efficiency. Treatment of sensitive HL-60 cells with methyl-ß-cyclodextrine, a cholesterol-depleting agent, resulted in a diminished injection of ExoS-Bla. Moreover, the PopB translocator was found in the membrane fraction, obtained from sucrose-gradient purifications, containing the lipid-raft marker flotillin. Examination of components of signalling pathways influencing the toxin injection was further assayed through a pharmacological approach. A systematic detection of translocon proteins within host membranes showed that, in addition to membrane composition, some general signalling pathways involved in actin polymerization may be critical for the formation of a functional pore. In conclusion, we provide new insights in regulation of translocation process and suggest possible cross-talks between eukaryotic cell and the pathogen at the level of exotoxin translocation.
Collapse
Affiliation(s)
- Julien Verove
- INSERM, U1036, Biology of Cancer and Infection, Grenoble, France
- CNRS, ERL 5261, Bacterial Pathogenesis and Cellular Responses, Grenoble, France
- Université Joseph Fourier-Grenoble I, Grenoble, France
- CEA, DSV/iRTSV, Grenoble, France
| | - Cédric Bernarde
- INSERM, U1036, Biology of Cancer and Infection, Grenoble, France
- CNRS, ERL 5261, Bacterial Pathogenesis and Cellular Responses, Grenoble, France
- Université Joseph Fourier-Grenoble I, Grenoble, France
- CEA, DSV/iRTSV, Grenoble, France
| | - Yu-Sing Tammy Bohn
- INSERM, U1036, Biology of Cancer and Infection, Grenoble, France
- CNRS, ERL 5261, Bacterial Pathogenesis and Cellular Responses, Grenoble, France
- Université Joseph Fourier-Grenoble I, Grenoble, France
- CEA, DSV/iRTSV, Grenoble, France
| | - François Boulay
- INSERM, U1036, Biology of Cancer and Infection, Grenoble, France
- CNRS, ERL 5261, Bacterial Pathogenesis and Cellular Responses, Grenoble, France
- Université Joseph Fourier-Grenoble I, Grenoble, France
- CEA, DSV/iRTSV, Grenoble, France
| | - Marie-Josèphe Rabiet
- INSERM, U1036, Biology of Cancer and Infection, Grenoble, France
- CNRS, ERL 5261, Bacterial Pathogenesis and Cellular Responses, Grenoble, France
- Université Joseph Fourier-Grenoble I, Grenoble, France
- CEA, DSV/iRTSV, Grenoble, France
| | - Ina Attree
- INSERM, U1036, Biology of Cancer and Infection, Grenoble, France
- CNRS, ERL 5261, Bacterial Pathogenesis and Cellular Responses, Grenoble, France
- Université Joseph Fourier-Grenoble I, Grenoble, France
- CEA, DSV/iRTSV, Grenoble, France
| | - François Cretin
- INSERM, U1036, Biology of Cancer and Infection, Grenoble, France
- CNRS, ERL 5261, Bacterial Pathogenesis and Cellular Responses, Grenoble, France
- Université Joseph Fourier-Grenoble I, Grenoble, France
- CEA, DSV/iRTSV, Grenoble, France
- * E-mail:
| |
Collapse
|
16
|
Trček J, Oellerich MF, Niedung K, Ebel F, Freund S, Trülzsch K. Gut proteases target Yersinia invasin in vivo. BMC Res Notes 2011; 4:129. [PMID: 21501502 PMCID: PMC3094372 DOI: 10.1186/1756-0500-4-129] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Accepted: 04/18/2011] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Yersinia enterocolitica is a common cause of food borne gastrointestinal disease. After oral uptake, yersiniae invade Peyer's patches of the distal ileum. This is accomplished by the binding of the Yersinia invasin to β1 integrins on the apical surface of M cells which overlie follicle associated lymphoid tissue. The gut represents a barrier that severely limits yersiniae from reaching deeper tissues such as Peyer's patches. We wondered if gut protease attack on invasion factors could contribute to the low number of yersiniae invading Peyer's patches. FINDINGS Here we show that invasin is rapidly degraded in vivo by gut proteases in the mouse infection model. In vivo proteolytic degradation is due to proteolysis by several gut proteases such as trypsin, α-chymotrypsin, pancreatic elastase, and pepsin. Protease treated yersiniae are shown to be less invasive in a cell culture model. YadA, another surface adhesin is cleaved by similar concentrations of gut proteases but Myf was not cleaved, showing that not all surface proteins are equally susceptible to degradation by gut proteases. CONCLUSIONS We demonstrate that gut proteases target important Yersinia virulence factors such as invasin and YadA in vivo. Since invasin is completely degraded within 2-3 h after reaching the small intestine of mice, it is no longer available to mediate invasion of Peyer's patches.
Collapse
Affiliation(s)
- Janja Trček
- Max von Pettenkofer Institut für Hygiene und Medizinische Mikrobiologie, Ludwig Maximilians Universität München, Germany.
| | | | | | | | | | | |
Collapse
|
17
|
Trcek J, Berschl K, Trülzsch K. In vivo analysis of Yersinia enterocolitica infection using luxCDABE. FEMS Microbiol Lett 2010; 307:201-6. [PMID: 20636977 DOI: 10.1111/j.1574-6968.2010.01983.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Yersiniae expressing an L-arabinose-inducible luxCDABE reporter were used to analyze the colonization of mice. Infection of live mice was followed over a period of 6 days. These experiments revealed frequent colonization of cervical lymph nodes after oral, but not intravenous infection. Furthermore, the well-known colonization of the small intestine, Peyer's patches (PPs) of the ileum, the cecal lymph follicle, mesenteric lymph nodes, liver, and spleen was easily detectable. Removal of the intestinal tract of mice revealed that the number of abscessed PPs and other tissues can be easily quantified. Experiments with an invasin mutant expressing luxCDABE revealed a significantly reduced number of abscessed PPs, cecal lymph follicles, and lymph nodes in yersiniae lacking invasin.
Collapse
Affiliation(s)
- Janja Trcek
- Max von Pettenkofer-Institut für Hygiene und Medizinische Mikrobiologie, Ludwig Maximilans Universität, Munich, Germany
| | | | | |
Collapse
|
18
|
Reis RSD, Horn F. Enteropathogenic Escherichia coli, Samonella, Shigella and Yersinia: cellular aspects of host-bacteria interactions in enteric diseases. Gut Pathog 2010; 2:8. [PMID: 20649986 PMCID: PMC2921366 DOI: 10.1186/1757-4749-2-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Accepted: 07/22/2010] [Indexed: 12/18/2022] Open
Abstract
A successful infection of the human intestine by enteropathogenic bacteria depends on the ability of bacteria to attach and colonize the intestinal epithelium and, in some cases, to invade the host cell, survive intracellularly and disseminate from cell to cell. To accomplish these processes bacteria have evolved an arsenal of molecules that are mostly secreted by dedicated type III secretion systems, and that interact with the host, subverting normal cellular functions. Here we overview the most important molecular strategies developed by enteropathogenic Escherichia coli, Salmonella enterica, Shigella flexneri, and Yersinia enterocolitica to cause enteric infections. Despite having evolved different effectors, these four microorganisms share common host cellular targets.
Collapse
Affiliation(s)
- Roberta Souza Dos Reis
- Departamento de Biofísica, Universidade Federal do Rio Grande do Sul, P,O, Box 15005, 91501-970, Porto Alegre, Brazil.
| | | |
Collapse
|
19
|
Deuretzbacher A, Czymmeck N, Reimer R, Trülzsch K, Gaus K, Hohenberg H, Heesemann J, Aepfelbacher M, Ruckdeschel K. Beta1 integrin-dependent engulfment of Yersinia enterocolitica by macrophages is coupled to the activation of autophagy and suppressed by type III protein secretion. THE JOURNAL OF IMMUNOLOGY 2009; 183:5847-60. [PMID: 19812190 DOI: 10.4049/jimmunol.0804242] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Autophagy is a central lysosomal degradation process that is essential for the maintenance of cellular homeostasis. Autophagy has furthermore emerged as integral part of the host immune response. Autophagic processes promote the separation and degradation of intracellular microorganisms which contributes to the development of innate and adaptive immunity. Some pathogenic microbes have therefore evolved mechanisms to evade or impede autophagy. We analyzed the effects of the enteropathogenic bacterium Yersinia enterocolitica on autophagy in macrophages. Yersiniae use a number of defined adhesins and secreted proteins to manipulate host immune responses. Our results showed that Y. enterocolitica defective in type III protein secretion efficiently activated autophagy in macrophages. Autophagy was mediated by the Yersinia adhesins invasin and YadA and particularly depended on the engagement of beta(1) integrin receptors. Several autophagy-related events followed beta(1) integrin-mediated engulfment of the bacteria including the formation of autophagosomes, processing of the marker protein LC3, redistribution of GFP-LC3 to bacteria-containing vacuoles, and the segregation of intracellular bacteria by autophagosomal compartments. These results provide direct evidence for the linkage of beta(1) integrin-mediated phagocytosis and autophagy induction. Multiple microbes signal through integrin receptors, and our results suggest a general principle by which the sensing of an extracellular microbe triggers autophagy. Owing to the importance of autophagy as host defense response, wild-type Y. enterocolitica suppressed autophagy by mobilizing type III protein secretion. The subversion of autophagy may be part of the Y. enterocolitica virulence strategy that supports bacterial survival when beta(1) integrin-dependent internalization and autophagy activation by macrophages are deleterious for the pathogen.
Collapse
Affiliation(s)
- Anne Deuretzbacher
- Institute for Medical Microbiology, Virology, and Hygiene, University Medical Center Eppendorf, Hamburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Uliczka F, Kornprobst T, Eitel J, Schneider D, Dersch P. Cell invasion of Yersinia pseudotuberculosis by invasin and YadA requires protein kinase C, phospholipase C-gamma1 and Akt kinase. Cell Microbiol 2009; 11:1782-801. [PMID: 19681907 DOI: 10.1111/j.1462-5822.2009.01371.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The outer membrane proteins YadA and invasin of Yersinia pseudotuberculosis promote invasion into mammalian cells through beta(1)-integrins and trigger the production of interleukin (IL)-8. FAK, c-Src and the PI3 kinase were previously found to be important for both YadA- and invasin-promoted uptake. Here, we demonstrate that two different downstream effectors of PI3 kinase, Akt and phospholipase Cgamma1 are required for efficient cell invasion. Inhibition of Akt or phospholipase C-gamma (PLC-gamma)1 by pharmaceutical agents as well as reduced expression of the isoforms Akt1 and Akt2, and of PLC-gamma1 by RNA interference decreased entry of YadA- and Inv-expressing bacteria significantly. In addition, we report that the conventional protein kinases C (PKC)alpha and -beta, positioned downstream of PLC-gamma1, are activated upon Inv- or YadA-promoted cell entry. They colocalize with intracellular bacteria and their depletion by siRNA treatment also resulted in a strong reduction of cell entry. In contrast, neither Akt nor PLC-gamma1, and the PKCs are essential for YadA- and Inv-mediated IL-8 synthesis and release. We conclude that YadA and invasin of Y. pseudotuberculosis both trigger similar signal transduction pathways during integrin-mediated phagocytosis into epithelial cells, which lead to the activation of Akt, PLC-gamma1, PKCalpha and -beta downstream of PI3 kinase, separate from the MAPK-dependent pathway that triggers IL-8 production.
Collapse
Affiliation(s)
- Frank Uliczka
- Institut für Mikrobiologie, Technische Universität Braunschweig, 38106 Braunschweig, Germany
| | | | | | | | | |
Collapse
|
21
|
Velasquez Almonacid LA, Tafuri S, Dipineto L, Matteoli G, Fiorillo E, Della Morte R, Fioretti A, Menna LF, Staiano N. Role of connexin-43 hemichannels in the pathogenesis of Yersinia enterocolitica. Vet J 2008; 182:452-7. [PMID: 18824377 DOI: 10.1016/j.tvjl.2008.08.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2008] [Revised: 07/10/2008] [Accepted: 08/07/2008] [Indexed: 01/09/2023]
Abstract
Connexin (Cx) channels are sites of cytoplasmic communication between contacting cells. Evidence indicates that the opening of hemichannels occurs under both physiological and pathological conditions. In this paper, the involvement of Cx-43 hemichannels is demonstrated in the pathogenesis of Yersinia. Parental HeLa cells and transfected HeLa cells stably expressing Cx-43 (HCx43) were infected with Yersiniaenterocolitica, and bacterial uptake was measured by the colony-forming unit method. Bacterial uptake was higher in HCx43 cells than in parental cells and was inhibited by the Cx channel blocker, 18-alpha-glycyrrhetinic acid (AGA). The inhibitory effect of AGA was more pronounced on the Y. enterocolitica uptake by HCx43 cells than by parental cells. The ability of HCx43 cells to incorporate the permeable fluorescent tracer Lucifer Yellow (LY) was assessed. Dye incorporation was inhibited by AGA, whereas Y. enterocolitica infection of HCx43 cells increased LY incorporation. Western blotting analysis demonstrated that Y. enterocolitica infection of HCx43 cells induced tyrosine phosphorylation of Cx-43, thus supporting a critical role for Cx-43 in the strategies exploited by bacterial pathogens to invade non-phagocytic cells.
Collapse
Affiliation(s)
- L A Velasquez Almonacid
- Dipartimento di Patologia e Sanità Animale, Università di Napoli Federico II, via F. Delpino 1, 80137 Napoli, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Unusual, virulence plasmid-dependent growth behavior of Yersinia enterocolitica in three-dimensional collagen gels. J Bacteriol 2008; 190:4111-20. [PMID: 18408033 DOI: 10.1128/jb.00156-08] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
As a first approach to establishing a three-dimensional culture infection model, we studied the growth behavior of the extracellular pathogen Yersinia enterocolitica in three-dimensional collagen gels (3D-CoG). Surprisingly, we observed that plasmidless Y. enterocolitica was motile in the 3D-CoG in contrast to its growth in traditional motility agar at 37 degrees C. Motility at 37 degrees C was abrogated in the presence of the virulence plasmid pYV or the exclusive expression of the pYV-located Yersinia adhesion gene yadA. YadA-producing yersiniae formed densely packed (dp) microcolonies, whereas pYVDelta yadA-carrying yersiniae formed loosely packed microcolonies at 37 degrees C in 3D-CoG. Furthermore, we demonstrated that the packing density of the microcolonies was dependent on the head domain of YadA. Moreover, dp microcolony formation did not depend on the capacity of YadA to bind to collagen fibers, as demonstrated by the use of yersiniae producing collagen nonbinding YadA. By using a yopE-gfp reporter, we demonstrated Ca(2+)-dependent expression of this pYV-localized virulence gene by yersiniae in 3D-CoG. In conclusion, this study revealed unique plasmid-dependent growth behavior of yersiniae in a three-dimensional matrix environment that resembles the behavior of yersiniae (e.g., formation of microcolonies) in infected mouse tissue. Thus, this 3D-CoG model may be a first step to a more complex level of in vitro infection models that mimic living tissue, enabling us to study the dynamics of pathogen-host cell interactions.
Collapse
|
23
|
Murphy BS, Wulff CR, Garvy BA, Straley SC. Yersinia pestis YadC: a novel vaccine candidate against plague. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 603:400-14. [PMID: 17966436 DOI: 10.1007/978-0-387-72124-8_37] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
Current subunit vaccines provide partial protection against pneumonic plague if the infecting Y. pestis strain is encapsulated (F1+). Here we describe YadC, a novel Y. pestis outer membrane protein that provides partial protection against a F1(-) Y. pestis strain. Swiss-Webster mice were immunized subcutaneously with glutathione S-transferase (GST) or His6-tagged (HT) purified fusion proteins (GST-YadC137-409 or HT-LcrV) or buffer emulsified with Alhydrogel. Intravenous challenge with 1 x 10(4) F1(-) Deltapgm Y. pestis CO99-3015 revealed no protection for those mice immunized with GST-Alhydrogel alone, full protection for HT-LcrV-immunized mice, and partial protection for GST-YadC137-409-immunized mice. Similarly, C57BL/6 mice were immunized with GST-YadC137-409, HT-LcrV, or GST all with Alhydrogel adjuvant. After intranasal challenge with 3 x 10(3) F1(-) Y. pestis CO99-3015, 87% of GST-YadC137-409-immunized mice survived pneumonic plague. This is compared to the GST control group (0 surviving mice) and the LcrV-immunized group where 50% survived the challenge. This protection was correlated with a predominantly IgG1 response in LcrV-immunized mice and an IgG1/IgG3 antibody response in YadC-immunized mice. Additionally, we report the cytokine response from HT-LcrV- and GST-YadC137-409-stimulated peripherally derived macrophages. YadC-stimulated cells demonstrated a predominant pro-inflammatory cytokine production. This mixed Thl/Th2 response suggests that YadC's protection may involve a different adaptive immune response than the LcrV protein that currently is part of plague vaccines.
Collapse
Affiliation(s)
- Brian S Murphy
- Department of Internal Medicine, University of Kentucky, Lexington, USA.
| | | | | | | |
Collapse
|
24
|
A conserved glycine residue of trimeric autotransporter domains plays a key role in Yersinia adhesin A autotransport. J Bacteriol 2007; 189:9011-9. [PMID: 17921300 DOI: 10.1128/jb.00985-07] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The Yersinia adhesin A (YadA) is a trimeric autotransporter adhesin of enteric yersiniae. It consists of three major domains: a head mediating adherence to host cells, a stalk involved in serum resistance, and an anchor that forms a membrane pore and is responsible for the autotransport function. The anchor contains a glycine residue, nearly invariant throughout trimeric autotransporter adhesins, that faces the pore lumen. To address the role of this glycine, we replaced it with polar amino acids of increasing side chain size and expressed wild-type and mutant YadA in Escherichia coli. The mutations did not impair the YadA-mediated adhesion to collagen and to host cells or the host cell cytokine production, but they decreased the expression levels and stability of YadA trimers with increasing side chain size. Likewise, autoagglutination and resistance to serum were decreased in these mutants. We found that the periplasmic protease DegP is involved in the degradation of YadA and that in an E. coli degP deletion strain, mutant versions of YadA were expressed almost to wild-type levels. We conclude that the conserved glycine residue affects both the export and the stability of YadA and consequently some of its putative functions in pathogenesis.
Collapse
|
25
|
Fälker S, Schilling J, Schmidt MA, Heusipp G. Overproduction of DNA adenine methyltransferase alters motility, invasion, and the lipopolysaccharide O-antigen composition of Yersinia enterocolitica. Infect Immun 2007; 75:4990-7. [PMID: 17682042 PMCID: PMC2044514 DOI: 10.1128/iai.00457-07] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
DNA adenine methyltransferase (Dam) not only regulates basic cellular functions but also interferes with the proper expression of virulence factors in various pathogens. We showed previously that for the human pathogen Yersinia enterocolitica, overproduction of Dam results in increased invasion of epithelial cells. Since invasion and motility are coordinately regulated in Y. enterocolitica, we analyzed the motility of a Dam-overproducing (Dam(OP)) strain and found it to be highly motile. In Dam(OP) strains, the operon encoding the master regulator of flagellum biosynthesis, flhDC, is upregulated. We show that the increased invasion is not due to enhanced expression of known and putative Y. enterocolitica invasion and adhesion factors, such as Inv, YadA, Ail, Myf fibrils, Pil, or Flp pili. However, overproduction of Dam no longer results in increased invasion for an inv mutant strain, indicating that Inv is necessary for increased invasion after overproduction of Dam. Since we show that overproduction of Dam results in an increased amount of rough lipopolysaccharide (LPS) molecules lacking O-antigen side chains, this implies that reduced steric hindrance by LPS might contribute to increased invasion by a Y. enterocolitica Dam(OP) strain. Our data add an important new aspect to the various virulence-associated phenotypes influenced by DNA methylation in Y. enterocolitica and indicate that Dam targets regulatory processes modulating the composition and function of the bacterial surface.
Collapse
Affiliation(s)
- Stefan Fälker
- Institut für Infektiologie, Zentrum für Molekularbiologie der Entzündung, Westfälische Wilhelms-Universität Münster, 48149 Münster, Germany
| | | | | | | |
Collapse
|
26
|
Tobback E, Decostere A, Hermans K, Haesebrouck F, Chiers K. Yersinia ruckeri infections in salmonid fish. JOURNAL OF FISH DISEASES 2007; 30:257-68. [PMID: 17501736 DOI: 10.1111/j.1365-2761.2007.00816.x] [Citation(s) in RCA: 140] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Yersinia ruckeri is the causative agent of yersiniosis or enteric redmouth disease leading to significant economic losses in salmonid aquaculture worldwide. Infection may result in a septicaemic condition with haemorrhages on the body surface and in the internal organs. Despite the significance of the disease, very little information is available on the pathogenesis, hampering the development of preventive measures to efficiently combat this bacterial agent. This review discusses the agent and the disease it causes. The possibility of the presence of similar virulence markers and/or pathogenic mechanisms between the Yersinia species which elicit disease in humans and Y. ruckeri is also examined.
Collapse
Affiliation(s)
- E Tobback
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.
| | | | | | | | | |
Collapse
|
27
|
Wong KW, Mohammadi S, Isberg RR. Disruption of RhoGDI and RhoA regulation by a Rac1 specificity switch mutant. J Biol Chem 2006; 281:40379-88. [PMID: 17074770 DOI: 10.1074/jbc.m605387200] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Rho family GTPases are important regulators of the actin cytoskeleton. Activation of these proteins can be promoted by guanine nucleotide exchange factors containing Dbl and Pleckstrin homology domains resulting in membrane insertion of a Rho family member, whereas the inactive GDP-bound form is sequestered primarily in the cytoplasm, bound to the guanosine dissociation inhibitor RhoGDI. Dominant interfering variants of Rac1, but not Cdc42, inhibit beta1 integrin-promoted uptake of Yersinia pseudotuberculosis. Unexpectedly, we found that the Rac1(W56F) guanine nucleotide exchange factors specificity switch mutant blocked invasin-promoted uptake as well as Cdc42-dependent uptake of enteropathogenic Escherichia coli. Fluorescence resonance energy transfer experiments demonstrated that Rac1(W56F) retained the ability to be loaded with GTP, bind a downstream effector, and interact with RhoGDI. Mutational analyses of intragenic suppressors and coexpression studies demonstrated that binding of the Rac1(W56F) mutant to RhoGDI appeared to play a role in the inhibition of uptake. As RhoGDI inhibits RhoA, overactivation of RhoA may account for the uptake interference caused by Rac1(W56F). Consistent with this model, a dominant interfering form of RhoA restored significant uptake in the presence of the Rac1(W56F) mutant but had no effect on another interfering Rac1 form. Furthermore, the cellular GTP-RhoA level was elevated by the presence of Rac1(W56F) mutant protein. These data are consistent with the proposition that Rac1(W56F) blocks invasin-promoted uptake by preventing RhoGDI from inactivating RhoA. We conclude that RhoGDI allows cross-talk between Rho family members that promote potentially antagonistic processes, and disruption of this cross-talk can interfere with invasin-promoted uptake.
Collapse
Affiliation(s)
- Ka-Wing Wong
- Howard Hughes Medical Institute, Department of Molecular Biology and Microbiology, Tufts University Medical School, Boston, Massachusetts 02111, USA
| | | | | |
Collapse
|
28
|
Owen KA, Thomas KS, Bouton AH. The differential expression of Yersinia pseudotuberculosis adhesins determines the requirement for FAK and/or Pyk2 during bacterial phagocytosis by macrophages. Cell Microbiol 2006; 9:596-609. [PMID: 16987330 DOI: 10.1111/j.1462-5822.2006.00811.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Phagocytosis of Yersinia pseudotuberculosis by macrophages is initiated by interactions between host cell integrin receptors and the bacterial adhesins, invasin and YadA. Two non-receptor protein tyrosine kinases, FAK and Pyk2, have been implicated in this process. In this study, we investigated the mechanisms of activation and functional requirements for these kinases during phagocytosis. A panel of Yersinia strains that differentially express invasin and YadA were used to infect cells in which FAK and/or Pyk2 expression was reduced by RNA interference. Bacterial strains that simultaneously express invasin and YadA activated FAK and Pyk2 signalling pathways that perform non-redundant functions required for Yersinia internalization. In contrast, FAK activation was found to be sufficient for phagocytosis of bacteria expressing invasin alone, and Pyk2 activation was sufficient when YadA was expressed in the absence of invasin. Based on these data, we suggest that the activation states of FAK and Pyk2, as well as the subsequent signalling events that lead to phagocytosis, are differentially regulated through the unique mechanisms of integrin engagement utilized by invasin and YadA. These findings lend insight into the molecular events that control bacterial phagocytosis as well as other integrin-based processes such as cell adhesion and migration.
Collapse
Affiliation(s)
- Katherine A Owen
- Department of Microbiology, Box 800734, University of Virginia Health System, Charlottesville, VA 22908-0734, USA
| | | | | |
Collapse
|
29
|
Hudson KJ, Bouton AH. Yersinia pseudotuberculosis adhesins regulate tissue-specific colonization and immune cell localization in a mouse model of systemic infection. Infect Immun 2006; 74:6487-90. [PMID: 16920785 PMCID: PMC1695483 DOI: 10.1128/iai.00718-06] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Yersinia pseudotuberculosis mutants deficient for the adhesins invasin and/or YadA were injected intravenously into BALB/c mice. Invasin expression inhibited colonization of the liver and spleen. YadA decreased liver colonization but promoted growth within the lung. The persistence of leukocytes within liver microabscesses correlated with enhanced colonization and lack of adhesin expression.
Collapse
Affiliation(s)
- Krischan J Hudson
- Department of Microbiology, University of Virginia Health System, Box 800734, 1300 Jefferson Park Ave., Charlottesville, VA 22908-0734, USA
| | | |
Collapse
|
30
|
Heesemann J, Sing A, Trülzsch K. Yersinia's stratagem: targeting innate and adaptive immune defense. Curr Opin Microbiol 2006; 9:55-61. [PMID: 16413818 DOI: 10.1016/j.mib.2005.10.018] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2005] [Accepted: 10/23/2005] [Indexed: 10/25/2022]
Abstract
In contrast to Salmonella and Shigella, enteropathogenic Yersinia species are extracellular multiplying Gram-negative bacteria. This life style requires a sophisticated anti-host strategy, which is implemented by the Yersinia virulence plasmid. This plasmid encodes the type 3 secretion system (injectisome), at least six microinjected anti-host effector proteins, a trimeric coiled coil outer membrane protein (Yersinia adhesin) with cell adhesin and protective functions against complement and defensins, and the released V antigen, which has Toll-like receptor 2 agonist activity.
Collapse
Affiliation(s)
- Jürgen Heesemann
- Max von Pettenkofer-Institut, Pettenkoferstrasse 9 a, D-80336 Muenchen, Germany.
| | | | | |
Collapse
|