1
|
Miner MV, Rauch I. Why put yourself on a pedestal? The pathogenic role of the A/E pedestal. Infect Immun 2024; 92:e0048923. [PMID: 38591884 PMCID: PMC11384751 DOI: 10.1128/iai.00489-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024] Open
Abstract
Certain Escherichia coli (E. coli) strains are attaching and effacing (A/E) lesion pathogens that primarily infect intestinal epithelial cells. They cause actin restructuring and polymerization within the host cell to create an actin-rich protrusion below the site of adherence, termed the pedestal. Although there is clarity on the pathways initiating pedestal formation, the underlying purpose(s) of the pedestal remains ambiguous. The conservation of pedestal-forming activity across multiple pathogens and redundancy in formation pathways indicate a pathogenic advantage. However, few decisive conclusions have been drawn, given that the results vary between model systems. Some research argues that the pedestal increases the colonization capability of the bacterium. These studies utilize A/E pathogens specifically deficient in pedestal formation to evaluate adhesion and intestinal colonization following infection. There have been many proposed mechanisms for the colonization benefit conferred by the pedestal. One suggested benefit is that the pedestal allows for direct cytosolic anchoring through incorporation of the established host cortical actin, causing a stable link between the pathogen and cell structure. The pedestal may confer enhanced motility, as enteropathogenic E. coli (EPEC) and enterohemorrhagic E. coli (EHEC) are better able to migrate on the surface of host cells and infect neighboring cells in the presence of the pedestal. Additionally, some research suggests that the pedestal improves effector delivery. This review will investigate the purpose of pedestal formation using evidence from recent literature and will critically evaluate the methodology and model systems. Most importantly, we will contextualize the proposed functions to reconcile potential synergistic effects.
Collapse
Affiliation(s)
- M. V. Miner
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, USA
| | - I. Rauch
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
2
|
Kirchenwitz M, Halfen J, von Peinen K, Prettin S, Kollasser J, Zur Lage S, Blankenfeldt W, Brakebusch C, Rottner K, Steffen A, Stradal TEB. RhoB promotes Salmonella survival by regulating autophagy. Eur J Cell Biol 2023; 102:151358. [PMID: 37703749 DOI: 10.1016/j.ejcb.2023.151358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 07/05/2023] [Accepted: 08/29/2023] [Indexed: 09/15/2023] Open
Abstract
Salmonella enterica serovar Typhimurium manipulates cellular Rho GTPases for host cell invasion by effector protein translocation via the Type III Secretion System (T3SS). The two Guanine nucleotide exchange (GEF) mimicking factors SopE and -E2 and the inositol phosphate phosphatase (PiPase) SopB activate the Rho GTPases Rac1, Cdc42 and RhoA, thereby mediating bacterial invasion. S. Typhimurium lacking these three effector proteins are largely invasion-defective. Type III secretion is crucial for both early and later phases of the intracellular life of S. Typhimurium. Here we investigated whether and how the small GTPase RhoB, known to localize on endomembrane vesicles and at the invasion site of S. Typhimurium, contributes to bacterial invasion and to subsequent steps relevant for S. Typhimurium lifestyle. We show that RhoB is significantly upregulated within hours of Salmonella infection. This effect depends on the presence of the bacterial effector SopB, but does not require its phosphatase activity. Our data reveal that SopB and RhoB bind to each other, and that RhoB localizes on early phagosomes of intracellular S. Typhimurium. Whereas both SopB and RhoB promote intracellular survival of Salmonella, RhoB is specifically required for Salmonella-induced upregulation of autophagy. Finally, in the absence of RhoB, vacuolar escape and cytosolic hyper-replication of S. Typhimurium is diminished. Our findings thus uncover a role for RhoB in Salmonella-induced autophagy, which supports intracellular survival of the bacterium and is promoted through a positive feedback loop by the Salmonella effector SopB.
Collapse
Affiliation(s)
- Marco Kirchenwitz
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Jessica Halfen
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Kristin von Peinen
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Silvia Prettin
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Jana Kollasser
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Susanne Zur Lage
- Department Structure and Function of Proteins, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Wulf Blankenfeldt
- Department Structure and Function of Proteins, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Cord Brakebusch
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Klemens Rottner
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany; Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstrasse 7, 38106 Braunschweig, Germany
| | - Anika Steffen
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Theresia E B Stradal
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany.
| |
Collapse
|
3
|
Fasciano AC, Dasanayake GS, Estes MK, Zachos NC, Breault DT, Isberg RR, Tan S, Mecsas J. Yersinia pseudotuberculosis YopE prevents uptake by M cells and instigates M cell extrusion in human ileal enteroid-derived monolayers. Gut Microbes 2022; 13:1988390. [PMID: 34793276 PMCID: PMC8604394 DOI: 10.1080/19490976.2021.1988390] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Many pathogens use M cells to access the underlying Peyer's patches and spread to systemic sites via the lymph as demonstrated by ligated loop murine intestinal models. However, the study of interactions between M cells and microbial pathogens has stalled due to the lack of cell culture systems. To overcome this obstacle, we use human ileal enteroid-derived monolayers containing five intestinal cell types including M cells to study the interactions between the enteric pathogen, Yersinia pseudotuberculosis (Yptb), and M cells. The Yptb type three secretion system (T3SS) effector Yops inhibit host defenses including phagocytosis and are critical for colonization of the intestine and Peyer's patches. Therefore, it is not understood how Yptb traverses through M cells to breach the epithelium. By growing Yptb under two physiological conditions that mimic the early infectious stage (low T3SS-expression) or host-adapted stage (high T3SS-expression), we found that large numbers of Yptb specifically associated with M cells, recapitulating murine studies. Transcytosis through M cells was significantly higher by Yptb expressing low levels of T3SS, because YopE and YopH prevented Yptb uptake. YopE also caused M cells to extrude from the epithelium without inducing cell-death or disrupting monolayer integrity. Sequential infection with early infectious stage Yptb reduced host-adapted Yptb association with M cells. These data underscore the strength of enteroids as a model by discovering that Yops impede M cell function, indicating that early infectious stage Yptb more effectively penetrates M cells while the host may defend against M cell penetration of host-adapted Yptb.
Collapse
Affiliation(s)
- Alyssa C. Fasciano
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, USA
| | - Gaya S. Dasanayake
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, USA
| | - Mary K. Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, USA
| | - Nicholas C. Zachos
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - David T. Breault
- Division of Endocrinology, Boston Children’s Hospital, Department of Pediatrics, Harvard Medical School, Boston, USA
| | - Ralph R. Isberg
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, USA,Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, USA
| | - Shumin Tan
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, USA
| | - Joan Mecsas
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, USA,Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, USA,CONTACT Joan Mecsas Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, USA
| |
Collapse
|
4
|
Rahmatelahi H, El-Matbouli M, Menanteau-Ledouble S. Delivering the pain: an overview of the type III secretion system with special consideration for aquatic pathogens. Vet Res 2021; 52:146. [PMID: 34924019 PMCID: PMC8684695 DOI: 10.1186/s13567-021-01015-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 11/08/2021] [Indexed: 11/10/2022] Open
Abstract
Gram-negative bacteria are known to subvert eukaryotic cell physiological mechanisms using a wide array of virulence factors, among which the type three-secretion system (T3SS) is often one of the most important. The T3SS constitutes a needle-like apparatus that the bacterium uses to inject a diverse set of effector proteins directly into the cytoplasm of the host cells where they can hamper the host cellular machinery for a variety of purposes. While the structure of the T3SS is somewhat conserved and well described, effector proteins are much more diverse and specific for each pathogen. The T3SS can remodel the cytoskeleton integrity to promote intracellular invasion, as well as silence specific eukaryotic cell signals, notably to hinder or elude the immune response and cause apoptosis. This is also the case in aquatic bacterial pathogens where the T3SS can often play a central role in the establishment of disease, although it remains understudied in several species of important fish pathogens, notably in Yersinia ruckeri. In the present review, we summarise what is known of the T3SS, with a special focus on aquatic pathogens and suggest some possible avenues for research including the potential to target the T3SS for the development of new anti-virulence drugs.
Collapse
Affiliation(s)
- Hadis Rahmatelahi
- Clinical Division of Fish Medicine, University of Veterinary Medicine, 1210, Vienna, Austria
| | - Mansour El-Matbouli
- Clinical Division of Fish Medicine, University of Veterinary Medicine, 1210, Vienna, Austria
| | - Simon Menanteau-Ledouble
- Clinical Division of Fish Medicine, University of Veterinary Medicine, 1210, Vienna, Austria.
- Department of Chemistry and Bioscience, Aalborg University, Fredrik Bajers Vej 7H, 9220, Aalborg Ø, Denmark.
| |
Collapse
|
5
|
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: 1.0] [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
|
6
|
The type 3 secretion system requires actin polymerization to open translocon pores. PLoS Pathog 2021; 17:e1009932. [PMID: 34499700 PMCID: PMC8454972 DOI: 10.1371/journal.ppat.1009932] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/21/2021] [Accepted: 08/31/2021] [Indexed: 11/19/2022] Open
Abstract
Many bacterial pathogens require a type 3 secretion system (T3SS) to establish a niche. Host contact activates bacterial T3SS assembly of a translocon pore in the host plasma membrane. Following pore formation, the T3SS docks onto the translocon pore. Docking establishes a continuous passage that enables the translocation of virulence proteins, effectors, into the host cytosol. Here we investigate the contribution of actin polymerization to T3SS-mediated translocation. Using the T3SS model organism Shigella flexneri, we show that actin polymerization is required for assembling the translocon pore in an open conformation, thereby enabling effector translocation. Opening of the pore channel is associated with a conformational change to the pore, which is dependent upon actin polymerization and a coiled-coil domain in the pore protein IpaC. Analysis of an IpaC mutant that is defective in ruffle formation shows that actin polymerization-dependent pore opening is distinct from the previously described actin polymerization-dependent ruffles that are required for bacterial internalization. Moreover, actin polymerization is not required for other pore functions, including docking or pore protein insertion into the plasma membrane. Thus, activation of the T3SS is a multilayered process in which host signals are sensed by the translocon pore leading to the activation of effector translocation.
Collapse
|
7
|
Armentrout EI, Kundracik EC, Rietsch A. Cell-type-specific hypertranslocation of effectors by the Pseudomonas aeruginosa type III secretion system. Mol Microbiol 2020; 115:305-319. [PMID: 33012037 DOI: 10.1111/mmi.14617] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 09/22/2020] [Indexed: 12/23/2022]
Abstract
Many Gram-negative pathogens use a type III secretion system (T3SS) to promote disease by injecting effector proteins into host cells. Common to many T3SSs is that injection of effector proteins is feedback inhibited. The mechanism of feedback inhibition and its role in pathogenesis are unclear. In the case of P. aeruginosa, the effector protein ExoS is central to limiting effector injection. ExoS is bifunctional, with an amino-terminal RhoGAP and a carboxy-terminal ADP-ribosyltransferase domain. We demonstrate that both domains are required to fully feedback inhibit effector injection. The RhoGAP-, but not the ADP-ribosyltransferase domain of the related effector protein ExoT also participates. Feedback inhibition does not involve translocator insertion nor pore-formation. Instead, feedback inhibition is due, in part, to a loss of the activating trigger for effector injection, and likely also decreased translocon stability. Surprisingly, feedback inhibition is abrogated in phagocytic cells. The lack of feedback inhibition in these cells requires phagocytic uptake of the bacteria, but cannot be explained through acidification of the phagosome or calcium limitation. Given that phagocytes are crucial for controlling P. aeruginosa infections, our data suggest that feedback inhibition allows P. aeruginosa to direct its effector arsenal against the cell types most damaging to its survival.
Collapse
Affiliation(s)
- Erin I Armentrout
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, OH, USA
| | - Emma C Kundracik
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, OH, USA
| | - Arne Rietsch
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
8
|
Malik HS, Bliska JB. The pyrin inflammasome and the Yersinia effector interaction. Immunol Rev 2020; 297:96-107. [PMID: 32721043 DOI: 10.1111/imr.12907] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 07/08/2020] [Indexed: 12/11/2022]
Abstract
Pyrin is a cytosolic pattern-recognition receptor that normally functions as a guard to trigger capase-1 inflammasome assembly in response to bacterial toxins and effectors that inactivate RhoA. The MEFV gene encoding human pyrin is preferentially expressed in phagocytes. Key domains in pyrin include a pyrin domain (PYD), a linker region, and a B30.2 domain. Binding of ASC to pyrin by a PYD-PYD interaction triggers inflammasome assembly. Pyrin is held in an inactive conformation by negative regulation mechanisms to avoid premature inflammasome assembly. One mechanism of negative regulation involves phosphorylation of the linker by PRK kinase which in turn is positively regulated by active RhoA. The B30.2 domain also negatively regulates pyrin. Gain of function mutations in MEFV responsible for the autoinflammatory disease Familial Mediterranean Fever (FMF) map to exon 10 encoding the B30.2 domain. Insights into pyrin regulation have come from studies of several Yersinia effectors, which are injected into phagocytes and interact with the RhoA-PRK-pyrin axis during infection. Two effectors, YopE and YopT, inactivate RhoA to disrupt phagocytic signaling. To counteract an effector-triggered immune response, a third effector, YopM, binds to and inhibits pyrin by hijacking PRK and RSK and directing linker phosphorylation. Inhibition of pyrin by YopM is required for virulence of Yersinia pestis, the agent of plague. Recent results from infection studies with human phagocytes and mice producing pyrin B30.2 FMF variants show that gain of function MEFV mutations bypass inhibition by YopM. Population genetic data suggest that MEFV mutations were selected for in individuals of Mediterranean decent during historic plague pandemics. This review discusses current concepts of pyrin regulation and its interaction with Yersinia effectors.
Collapse
Affiliation(s)
- Haleema S Malik
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - James B Bliska
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| |
Collapse
|
9
|
Krukonis ES, Thomson JJ. Complement evasion mechanisms of the systemic pathogens Yersiniae and Salmonellae. FEBS Lett 2020; 594:2598-2620. [DOI: 10.1002/1873-3468.13771] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 03/04/2020] [Accepted: 03/05/2020] [Indexed: 01/12/2023]
Affiliation(s)
- Eric S. Krukonis
- Division of Integrated Biomedical Sciences University of Detroit Mercy School of Dentistry Detroit MI USA
| | - Joshua J. Thomson
- Division of Integrated Biomedical Sciences University of Detroit Mercy School of Dentistry Detroit MI USA
| |
Collapse
|
10
|
Abstract
The human and animal pathogens Yersinia pestis, which causes bubonic and pneumonic plague, and Yersinia pseudotuberculosis and Yersinia enterocolitica, which cause gastroenteritis, share a type 3 secretion system which injects effector proteins, Yops, into host cells. This system is critical for virulence of all three pathogens in tissue infection. Neutrophils are rapidly recruited to infected sites and all three pathogens frequently interact with and inject Yops into these cells during tissue infection. Host receptors, serum factors, and bacterial adhesins appear to collaborate to promote neutrophil- Yersinia interactions in tissues. The ability of neutrophils to control infection is mixed depending on the stage of infection and points to the efficiency of Yops and other bacterial factors to mitigate bactericidal effects of neutrophils. Yersinia in close proximity to neutrophils has higher levels of expression from yop promoters, and neutrophils in close proximity to Yersinia express higher levels of pro-survival genes than migrating neutrophils. In infected tissues, YopM increases neutrophil survival and YopH targets a SKAP2/SLP-76 signal transduction pathway. Yet the full impact of these and other Yops and other Yersinia factors on neutrophils in infected tissues has yet to be understood.
Collapse
Affiliation(s)
- Joan Mecsas
- Department of Molecular Biology and Microbiology, 136 Harrison Ave, Tufts University School of Medicine, Boston, MA, 02111, USA
| |
Collapse
|
11
|
Bohn E, Sonnabend M, Klein K, Autenrieth IB. Bacterial adhesion and host cell factors leading to effector protein injection by type III secretion system. Int J Med Microbiol 2019; 309:344-350. [DOI: 10.1016/j.ijmm.2019.05.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 05/02/2019] [Accepted: 05/31/2019] [Indexed: 01/14/2023] Open
|
12
|
Nauth T, Huschka F, Schweizer M, Bosse JB, Diepold A, Failla AV, Steffen A, Stradal TEB, Wolters M, Aepfelbacher M. Visualization of translocons in Yersinia type III protein secretion machines during host cell infection. PLoS Pathog 2018; 14:e1007527. [PMID: 30586431 PMCID: PMC6324820 DOI: 10.1371/journal.ppat.1007527] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 01/08/2019] [Accepted: 12/14/2018] [Indexed: 12/13/2022] Open
Abstract
Type III secretion systems (T3SSs) are essential virulence factors of numerous bacterial pathogens. Upon host cell contact the T3SS machinery—also named injectisome—assembles a pore complex/translocon within host cell membranes that serves as an entry gate for the bacterial effectors. Whether and how translocons are physically connected to injectisome needles, whether their phenotype is related to the level of effector translocation and which target cell factors trigger their formation have remained unclear. We employed the superresolution fluorescence microscopy techniques Stimulated Emission Depletion (STED) and Structured Illumination Microscopy (SIM) as well as immunogold electron microscopy to visualize Y. enterocolitica translocons during infection of different target cell types. Thereby we were able to resolve translocon and needle complex proteins within the same injectisomes and demonstrate that these fully assembled injectisomes are generated in a prevacuole, a PI(4,5)P2 enriched host cell compartment inaccessible to large extracellular proteins like antibodies. Furthermore, the operable translocons were produced by the yersiniae to a much larger degree in macrophages (up to 25% of bacteria) than in HeLa cells (2% of bacteria). However, when the Rho GTPase Rac1 was activated in the HeLa cells, uptake of the yersiniae into the prevacuole, translocon formation and effector translocation were strongly enhanced reaching the same levels as in macrophages. Our findings indicate that operable T3SS translocons can be visualized as part of fully assembled injectisomes with superresolution fluorescence microscopy techniques. By using this technology, we provide novel information about the spatiotemporal organization of T3SS translocons and their regulation by host cell factors. Many human, animal and plant pathogenic bacteria employ a molecular machine termed injectisome to inject their toxins into host cells. Because injectisomes are crucial for these bacteria’s infectious potential they have been considered as targets for antiinfective drugs. Injectisomes are highly similar between the different bacterial pathogens and most of their overall structure is well established at the molecular level. However, only little information is available for a central part of the injectisome named the translocon. This pore-like assembly integrates into host cell membranes and thereby serves as an entry gate for the bacterial toxins. We used state of the art fluorescence microscopy to watch translocons of the diarrheagenic pathogen Yersinia enterocolitica during infection of human host cells. Thereby we could for the first time—with fluorescence microscopy—visualize translocons connected to other parts of the injectisome. Furthermore, because translocons mark functional injectisomes we could obtain evidence that injectisomes only become active for secretion of translocators when the bacteria are almost completely enclosed by host cells. These findings provide a novel view on the organization and regulation of bacterial translocons and may thus open up new strategies to block the function of infectious bacteria.
Collapse
Affiliation(s)
- Theresa Nauth
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Franziska Huschka
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Michaela Schweizer
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Jens B. Bosse
- Heinrich-Pette-Institute (HPI), Leibniz-Institute for Experimental Virology, Hamburg, Germany
| | - Andreas Diepold
- Department of Ecophysiology, Max-Planck-Institute for Terrestrial Microbiology, Marburg, Germany
| | - Antonio Virgilio Failla
- UKE Microscopy Imaging Facility, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Anika Steffen
- Department of Cell Biology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Theresia E. B. Stradal
- Department of Cell Biology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Manuel Wolters
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Martin Aepfelbacher
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
- * E-mail:
| |
Collapse
|
13
|
Büttner D. Behind the lines-actions of bacterial type III effector proteins in plant cells. FEMS Microbiol Rev 2018; 40:894-937. [PMID: 28201715 PMCID: PMC5091034 DOI: 10.1093/femsre/fuw026] [Citation(s) in RCA: 193] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 03/31/2016] [Accepted: 07/03/2016] [Indexed: 01/30/2023] Open
Abstract
Pathogenicity of most Gram-negative plant-pathogenic bacteria depends on the type III secretion (T3S) system, which translocates bacterial effector proteins into plant cells. Type III effectors modulate plant cellular pathways to the benefit of the pathogen and promote bacterial multiplication. One major virulence function of type III effectors is the suppression of plant innate immunity, which is triggered upon recognition of pathogen-derived molecular patterns by plant receptor proteins. Type III effectors also interfere with additional plant cellular processes including proteasome-dependent protein degradation, phytohormone signaling, the formation of the cytoskeleton, vesicle transport and gene expression. This review summarizes our current knowledge on the molecular functions of type III effector proteins with known plant target molecules. Furthermore, plant defense strategies for the detection of effector protein activities or effector-triggered alterations in plant targets are discussed.
Collapse
Affiliation(s)
- Daniela Büttner
- Genetics Department, Institute of Biology, Martin-Luther University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
14
|
Duncan MC, Herrera NG, Johnson KS, Engel JN, Auerbuch V. Bacterial internalization is required to trigger NIK-dependent NF-κB activation in response to the bacterial type three secretion system. PLoS One 2017; 12:e0171406. [PMID: 28166267 PMCID: PMC5293232 DOI: 10.1371/journal.pone.0171406] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 01/20/2017] [Indexed: 01/11/2023] Open
Abstract
Infection of human cells with Yersinia pseudotuberculosis expressing a functional type III secretion system (T3SS) leads to activation of host NF-κB. We show that the Yersinia T3SS activates distinct NF-κB pathways dependent upon bacterial subcellular localization. We found that wildtype Yersinia able to remain extracellular triggered NF-κB activation independently of the non-canonical NF-κB kinase NIK in HEK293T cells. In contrast, Yersinia lacking the actin-targeting effectors YopEHO, which become internalized into host cells, induce a NIK-dependent response and nuclear entry of the non-canonical NF-κB subunit p52. Blocking actin polymerization and uptake of effector mutant bacteria using cytochalasin D shifted the host NF-κB response from NIK-independent to primarily NIK-dependent. We observed similar results using Pseudomonas aeruginosa, which expresses a related T3SS and the actin-targeting effector ExoT. As the NF-κB response of HEK293T cells to effectorless Yersinia has been used both as a screening tool for chemical inhibitors of the T3SS and for bacterial forward genetic screens, a better understanding of this response is important for tool optimization and interpretation.
Collapse
Affiliation(s)
- Miles C. Duncan
- Department of Microbiology and Environmental Toxicology, University of California Santa Cruz, Santa Cruz, California, United States of America
| | - Natalia G. Herrera
- Department of Microbiology and Environmental Toxicology, University of California Santa Cruz, Santa Cruz, California, United States of America
| | - Kevin S. Johnson
- Department of Microbiology and Environmental Toxicology, University of California Santa Cruz, Santa Cruz, California, United States of America
| | - Joanne N. Engel
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, California, United States of America
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Victoria Auerbuch
- Department of Microbiology and Environmental Toxicology, University of California Santa Cruz, Santa Cruz, California, United States of America
| |
Collapse
|
15
|
Yersinia enterocolitica YopH-Deficient Strain Activates Neutrophil Recruitment to Peyer's Patches and Promotes Clearance of the Virulent Strain. Infect Immun 2016; 84:3172-3181. [PMID: 27550935 DOI: 10.1128/iai.00568-16] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 08/18/2016] [Indexed: 01/06/2023] Open
Abstract
Yersinia enterocolitica evades the immune response by injecting Yersinia outer proteins (Yops) into the cytosol of host cells. YopH is a tyrosine phosphatase critical for Yersinia virulence. However, the mucosal immune mechanisms subverted by YopH during in vivo orogastric infection with Y. enterocolitica remain elusive. The results of this study revealed neutrophil recruitment to Peyer's patches (PP) after infection with a YopH-deficient mutant strain (Y. enterocolitica ΔyopH). While the Y. enterocolitica wild-type (WT) strain in PP induced the major neutrophil chemoattractant CXCL1 mRNA and protein levels, infection with the Y. enterocolitica ΔyopH mutant strain exhibited a higher expression of the CXCL1 receptor, CXCR2, in blood neutrophils, leading to efficient neutrophil recruitment to the PP. In contrast, migration of neutrophils into PP was impaired upon infection with Y. enterocolitica WT strain. In vitro infection of blood neutrophils revealed the involvement of YopH in CXCR2 expression. Depletion of neutrophils during Y. enterocolitica ΔyopH infection raised the bacterial load in PP. Moreover, the clearance of WT Y. enterocolitica was improved when an equal mixture of Y. enterocolitica WT and Y. enterocolitica ΔyopH strains was used in infecting the mice. This study indicates that Y. enterocolitica prevents early neutrophil recruitment in the intestine and that the effector protein YopH plays an important role in the immune evasion mechanism. The findings highlight the potential use of the Y. enterocolitica YopH-deficient strain as an oral vaccine carrier.
Collapse
|
16
|
|
17
|
Wolters M, Zobiak B, Nauth T, Aepfelbacher M. Analysis of Yersinia enterocolitica Effector Translocation into Host Cells Using Beta-lactamase Effector Fusions. J Vis Exp 2015. [PMID: 26484613 DOI: 10.3791/53115] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Many gram-negative bacteria including pathogenic Yersinia spp. employ type III secretion systems to translocate effector proteins into eukaryotic target cells. Inside the host cell the effector proteins manipulate cellular functions to the benefit of the bacteria. To better understand the control of type III secretion during host cell interaction, sensitive and accurate assays to measure translocation are required. We here describe the application of an assay based on the fusion of a Yersinia enterocolitica effector protein fragment (Yersinia outer protein; YopE) with TEM-1 beta-lactamase for quantitative analysis of translocation. The assay relies on cleavage of a cell permeant FRET dye (CCF4/AM) by translocated beta-lactamase fusion. After cleavage of the cephalosporin core of CCF4 by the beta-lactamase, FRET from coumarin to fluorescein is disrupted and excitation of the coumarin moiety leads to blue fluorescence emission. Different applications of this method have been described in the literature highlighting its versatility. The method allows for analysis of translocation in vitro and also in in vivo, e.g., in a mouse model. Detection of the fluorescence signals can be performed using plate readers, FACS analysis or fluorescence microscopy. In the setup described here, in vitro translocation of effector fusions into HeLa cells by different Yersinia mutants is monitored by laser scanning microscopy. Recording intracellular conversion of the FRET reporter by the beta-lactamase effector fusion in real-time provides robust quantitative results. We here show exemplary data, demonstrating increased translocation by a Y. enterocolitica YopE mutant compared to the wild type strain.
Collapse
Affiliation(s)
- Manuel Wolters
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf;
| | - Bernd Zobiak
- UKE Microscopy Imaging Facility, University Medical Center Hamburg-Eppendorf
| | - Theresa Nauth
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf
| | - Martin Aepfelbacher
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf
| |
Collapse
|
18
|
Asrat S, Davis KM, Isberg RR. Modulation of the host innate immune and inflammatory response by translocated bacterial proteins. Cell Microbiol 2015; 17:785-795. [PMID: 25850689 DOI: 10.1111/cmi.12445] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 03/30/2015] [Accepted: 03/31/2015] [Indexed: 12/15/2022]
Abstract
Bacterial secretion systems play a central role in interfering with host inflammatory responses to promote replication in tissue sites. Many intracellular bacteria utilize secretion systems to promote their uptake and survival within host cells. An intracellular niche can help bacteria avoid killing by phagocytic cells, and may limit host sensing of bacterial components. Secretion systems can also play an important role in limiting host sensing of bacteria by translocating proteins that disrupt host immune signalling pathways. Extracellular bacteria, on the other hand, utilize secretion systems to prevent uptake by host cells and maintain an extracellular niche. Secretion systems, in this case, limit sensing and inflammatory signalling which can occur as bacteria replicate and release bacterial products in the extracellular space. In this review, we will cover the common mechanisms used by intracellular and extracellular bacteria to modulate innate immune and inflammatory signalling pathways, with a focus on translocated proteins of the type III and type IV secretion systems.
Collapse
Affiliation(s)
- Seblewongel Asrat
- Howard Hughes Medical Institute, Tufts University School of Medicine,150 Harrison Ave., Boston, MA 02111, USA.,Department of Molecular Biology and Microbiology, Tufts University School of Medicine,150 Harrison Ave., Boston, MA 02111, USA.,Graduate Program in Molecular Microbiology, Sackler School of Graduate Biomedical Science, Tufts University School of Medicine,150 Harrison Ave., Boston, MA 02111, USA
| | - Kimberly M Davis
- Howard Hughes Medical Institute, Tufts University School of Medicine,150 Harrison Ave., Boston, MA 02111, USA.,Department of Molecular Biology and Microbiology, Tufts University School of Medicine,150 Harrison Ave., Boston, MA 02111, USA
| | - Ralph R Isberg
- Howard Hughes Medical Institute, Tufts University School of Medicine,150 Harrison Ave., Boston, MA 02111, USA.,Department of Molecular Biology and Microbiology, Tufts University School of Medicine,150 Harrison Ave., Boston, MA 02111, USA
| |
Collapse
|
19
|
Solomon R, Zhang W, McCrann G, Bliska JB, Viboud GI. Random mutagenesis identifies a C-terminal region of YopD important for Yersinia type III secretion function. PLoS One 2015; 10:e0120471. [PMID: 25807250 PMCID: PMC4433470 DOI: 10.1371/journal.pone.0120471] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 01/22/2015] [Indexed: 11/19/2022] Open
Abstract
A common virulence mechanism among bacterial pathogens is the use of specialized secretion systems that deliver virulence proteins through a translocation channel inserted in the host cell membrane. During Yersinia infection, the host recognizes the type III secretion system mounting a pro-inflammatory response. However, soon after they are translocated, the effectors efficiently counteract that response. In this study we sought to identify YopD residues responsible for type III secretion system function. Through random mutagenesis, we identified eight Y. pseudotuberculosis yopD mutants with single amino acid changes affecting various type III secretion functions. Three severely defective mutants had substitutions in residues encompassing a 35 amino acid region (residues 168-203) located between the transmembrane domain and the C-terminal putative coiled-coil region of YopD. These mutations did not affect regulation of the low calcium response or YopB-YopD interaction but markedly inhibited MAPK and NFκB. [corrected] activation. When some of these mutations were introduced into the native yopD gene, defects in effector translocation and pore formation were also observed. We conclude that this newly identified region is important for YopD translocon function. The role of this domain in vivo remains elusive, as amino acid substitutions in that region did not significantly affect virulence of Y. pseudotuberculosis in orogastrically-infected mice.
Collapse
Affiliation(s)
- Rebecca Solomon
- Clinical Laboratory Science, School of Health, Technology and Management, Stony Brook University, Stony Brook, New York, United States of America
- Department of Molecular Genetics and Microbiology, Center for Infectious Diseases, School of Medicine, Stony Brook University, Stony Brook, New York, United States of America
| | - Weibing Zhang
- Clinical Laboratory Science, School of Health, Technology and Management, Stony Brook University, Stony Brook, New York, United States of America
- Department of Molecular Genetics and Microbiology, Center for Infectious Diseases, School of Medicine, Stony Brook University, Stony Brook, New York, United States of America
| | - Grace McCrann
- Clinical Laboratory Science, School of Health, Technology and Management, Stony Brook University, Stony Brook, New York, United States of America
| | - James B. Bliska
- Department of Molecular Genetics and Microbiology, Center for Infectious Diseases, School of Medicine, Stony Brook University, Stony Brook, New York, United States of America
| | - Gloria I. Viboud
- Clinical Laboratory Science, School of Health, Technology and Management, Stony Brook University, Stony Brook, New York, United States of America
- Department of Molecular Genetics and Microbiology, Center for Infectious Diseases, School of Medicine, Stony Brook University, Stony Brook, New York, United States of America
- * E-mail:
| |
Collapse
|
20
|
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: 2.5] [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
|
21
|
Adams W, Morgan J, Kwuan L, Auerbuch V. Yersinia pseudotuberculosis YopD mutants that genetically separate effector protein translocation from host membrane disruption. Mol Microbiol 2015; 96:764-78. [PMID: 25684661 DOI: 10.1111/mmi.12970] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2015] [Indexed: 12/20/2022]
Abstract
The Yersinia type III secretion system (T3SS) translocates Yop effector proteins into host cells to manipulate immune defenses such as phagocytosis and reactive oxygen species (ROS) production. The T3SS translocator proteins YopB and YopD form pores in host membranes, facilitating Yop translocation. While the YopD amino and carboxy termini participate in pore formation, the role of the YopD central region between amino acids 150-227 remains unknown. We assessed the contribution of this region by generating Y. pseudotuberculosis yopD(Δ150-170) and yopD(Δ207-227) mutants and analyzing their T3SS functions. These strains exhibited wild-type levels of Yop secretion in vitro and enabled robust pore formation in macrophages. However, the yopDΔ150-170 and yopD(Δ207-227) mutants were defective in Yop translocation into CHO cells and splenocyte-derived neutrophils and macrophages. These data suggest that YopD-mediated host membrane disruption and effector Yop translocation are genetically separable activities requiring distinct protein domains. Importantly, the yopD(Δ150-170) and yopD(Δ207-227) mutants were defective in Yop-mediated inhibition of macrophage cell death and ROS production in neutrophil-like cells, and were attenuated in disseminated Yersinia infection. Therefore, the ability of the YopD central region to facilitate optimal effector protein delivery into phagocytes, and therefore robust effector Yop function, is important for Yersinia virulence.
Collapse
Affiliation(s)
- Walter Adams
- Department of Microbiology and Environmental Toxicology, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Jessica Morgan
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Laura Kwuan
- Department of Microbiology and Environmental Toxicology, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Victoria Auerbuch
- Department of Microbiology and Environmental Toxicology, University of California Santa Cruz, Santa Cruz, CA, USA
| |
Collapse
|
22
|
Identification of mammalian proteins that collaborate with type III secretion system function: involvement of a chemokine receptor in supporting translocon activity. mBio 2015; 6:e02023-14. [PMID: 25691588 PMCID: PMC4337563 DOI: 10.1128/mbio.02023-14] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The type III secretion system (T3SS) is a highly conserved protein delivery system found in multiple Gram-negative pathogens, including Yersinia pseudotuberculosis. Most studies of Yersinia species type III intoxication of host cells have focused on the bacterial determinants that promote assembly and function of the secretion system. In this study, we performed a pooled RNA interference (RNAi) screen to identify mammalian host proteins required for the cytotoxic effects associated with the Yersinia translocated substrate YopE, a GTPase-activating protein (GAP) that inactivates the small Rho GTPases. Cell populations were positively selected for short hairpin RNAs (shRNAs) that interfere with YopE activity using a combination of fluorescence resonance energy transfer (FRET) and flow cytometry, and the degree of enrichment was determined by deep sequencing. Analysis of the candidates identified by the enrichment process revealed that many were important for the initial step of Y. pseudotuberculosis T3SS function, YopB/D pore formation. These candidates included shRNA that depleted downstream effectors of RhoA signaling, coated pit formation, and receptors involved in cell signaling, including the chemokine receptor CCR5 (chemokine [C-C motif] receptor 5). Depletion of CCR5 in 293T cells yielded a defect in YopB/D pore formation and effector translocation, while both phenotypes could be complemented by overexpression of CCR5 protein. Yop effector translocation was also decreased in isolated primary phagocytic cells from a Ccr5−/− knockout mouse. We postulate that CCR5 acts to promote translocation by modulating cytoskeletal activities necessary for proper assembly of the YopB/D translocation pore. Overall, this study presents a new approach to investigating the contribution of the host cell to T3SS in Y. pseudotuberculosis. Many Gram-negative bacteria require type III secretion systems (T3SS) for host survival, making these highly specialized secretion systems good targets for antimicrobial agents. After the bacterium binds to host cells, T3SS deposit proteins into the cytosol of host cells through a needle-like appendage and a protein translocon channel. Translocation of proteins via this system is highly regulated, and the contribution of the host cell in promoting assembly and insertion of the channel into the plasma membrane, folding of the bacterial proteins, and trafficking of these substrates are all poorly characterized events. In this study, we identified host cell proteins important for activity of YopE, a Yersinia pseudotuberculosis T3SS-delivered protein. The results demonstrate that insertion and assembly of the translocon are complex processes, requiring a variety of membrane trafficking and cytoskeletal processes, as well as a surprising role for cell surface signaling molecules in supporting proper function.
Collapse
|
23
|
Wang X, Parashar K, Sitaram A, Bliska JB. The GAP activity of type III effector YopE triggers killing of Yersinia in macrophages. PLoS Pathog 2014; 10:e1004346. [PMID: 25165815 PMCID: PMC4148447 DOI: 10.1371/journal.ppat.1004346] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 07/17/2014] [Indexed: 12/28/2022] Open
Abstract
The mammalian immune system has the ability to discriminate between pathogens and innocuous microbes by detecting conserved molecular patterns. In addition to conserved microbial patterns, the mammalian immune system may recognize distinct pathogen-induced processes through a mechanism which is poorly understood. Previous studies have shown that a type III secretion system (T3SS) in Yersinia pseudotuberculosis leads to decreased survival of this bacterium in primary murine macrophages by unknown mechanisms. Here, we use colony forming unit assays and fluorescence microscopy to investigate how the T3SS triggers killing of Yersinia in macrophages. We present evidence that Yersinia outer protein E (YopE) delivered by the T3SS triggers intracellular killing response against Yersinia. YopE mimics eukaryotic GTPase activating proteins (GAPs) and inactivates Rho GTPases in host cells. Unlike wild-type YopE, catalytically dead YopER144A is impaired in restricting Yersinia intracellular survival, highlighting that the GAP activity of YopE is detected as a danger signal. Additionally, a second translocated effector, YopT, counteracts the YopE triggered killing effect by decreasing the translocation level of YopE and possibly by competing for the same pool of Rho GTPase targets. Moreover, inactivation of Rho GTPases by Clostridium difficile Toxin B mimics the effect of YopE and promotes increased killing of Yersinia in macrophages. Using a Rac inhibitor NSC23766 and a Rho inhibitor TAT-C3, we show that macrophages restrict Yersinia intracellular survival in response to Rac1 inhibition, but not Rho inhibition. In summary, our findings reveal that primary macrophages sense manipulation of Rho GTPases by Yersinia YopE and actively counteract pathogenic infection by restricting intracellular bacterial survival. Our results uncover a new mode of innate immune recognition in response to pathogenic infection. The type III secretion system (T3SS) is a macromolecular protein export pathway found in gram-negative bacteria. It delivers bacterial toxins into eukaryotic cells to promote pathogenic infection. T3SSs and the bacterial toxins delivered are critical arsenals for many bacterial pathogens of clinical significance, such as Yersinia, Salmonella and Shigella. On the other hand, the mammalian immune system may recognize the T3SS as a danger signal to signify pathogenic infection, and to stimulate appropriate defense against pathogens. Here, we show that the innate immune system recognizes the activity of YopE delivered by the Yersinia T3SS. Modulation of host Rho GTPases by YopE elicits a defensive response, which results in killing of bacteria in host cells. Inhibition of host Rho GTPases by Clostridium difficile Toxin B, another bacterial toxin, mimics the YopE-triggered killing effect. Our study demonstrates that host cells sense manipulation of Rho GTPases by bacterial toxins as a surveillance mechanism, revealing new insights into innate immune recognition of pathogenic infections.
Collapse
Affiliation(s)
- Xiaoying Wang
- Department of Molecular Genetics and Microbiology, Center for Infectious Diseases, Stony Brook University, Stony Brook, New York, United States of America
| | - Kaustubh Parashar
- Department of Molecular Genetics and Microbiology, Center for Infectious Diseases, Stony Brook University, Stony Brook, New York, United States of America
| | - Ananya Sitaram
- Department of Molecular Genetics and Microbiology, Center for Infectious Diseases, Stony Brook University, Stony Brook, New York, United States of America
| | - James B. Bliska
- Department of Molecular Genetics and Microbiology, Center for Infectious Diseases, Stony Brook University, Stony Brook, New York, United States of America
- * E-mail:
| |
Collapse
|
24
|
Thinwa J, Segovia JA, Bose S, Dube PH. Integrin-mediated first signal for inflammasome activation in intestinal epithelial cells. THE JOURNAL OF IMMUNOLOGY 2014; 193:1373-82. [PMID: 24965773 DOI: 10.4049/jimmunol.1400145] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
How intestinal epithelial cells (IECs) recognize pathogens and activate inflammasomes at intestinal surfaces is poorly understood. We hypothesized that IECs use integrin receptors to recognize pathogens and initiate inflammation within the intestinal tract. We find that IECs infected with Yersinia enterocolitica, an enteric pathogen, use β1 integrins as pathogen recognition receptors detecting the bacterial adhesin invasin (Inv). The Inv-integrin interaction provides the first signal for NLRP3 inflammasome activation with the type three secretion system translocon providing the second signal for inflammasome activation, resulting in release of IL-18. During infection, Yersinia employs two virulence factors, YopE and YopH, to counteract Inv-mediated integrin-dependent inflammasome activation. Furthermore, NLRP3 inflammasome activation in epithelial cells requires components of the focal adhesion complex signaling pathway, focal adhesion kinase, and rac1. The binding of Inv to β1 integrins rapidly induces IL-18 mRNA expression, suggesting integrins provide a first signal for NLRP3 inflammasome activation. These data suggest integrins function as pathogen recognition receptors on IECs to rapidly induce inflammasome-derived IL-18-mediated responses.
Collapse
Affiliation(s)
- Josephine Thinwa
- Department of Microbiology and Immunology, University of Texas Health Science Center, San Antonio, TX 78229; and
| | - Jesus A Segovia
- Department of Microbiology and Immunology, University of Texas Health Science Center, San Antonio, TX 78229; and Center for Airway Inflammation Research, University of Texas Health Science Center, San Antonio, TX 78229
| | - Santanu Bose
- Department of Microbiology and Immunology, University of Texas Health Science Center, San Antonio, TX 78229; and Center for Airway Inflammation Research, University of Texas Health Science Center, San Antonio, TX 78229
| | - Peter H Dube
- Department of Microbiology and Immunology, University of Texas Health Science Center, San Antonio, TX 78229; and Center for Airway Inflammation Research, University of Texas Health Science Center, San Antonio, TX 78229
| |
Collapse
|
25
|
Actin pedestal formation by enterohemorrhagic Escherichia coli enhances bacterial host cell attachment and concomitant type III translocation. Infect Immun 2014; 82:3713-22. [PMID: 24958711 DOI: 10.1128/iai.01523-13] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Attachment of enterohemorrhagic Escherichia coli (EHEC) to intestinal epithelial cells is critical for colonization and is associated with localized actin assembly beneath bound bacteria. The formation of these actin "pedestals" is dependent on the translocation of effectors into mammalian cells via a type III secretion system (T3SS). Tir, an effector required for pedestal formation, localizes in the host cell plasma membrane and promotes attachment of bacteria to mammalian cells by binding to the EHEC outer surface protein Intimin. Actin pedestal formation has been shown to foster intestinal colonization by EHEC in some animal models, but the mechanisms responsible for this remain undefined. Investigation of the role of Tir-mediated actin assembly promoting host cell binding is complicated by other, potentially redundant EHEC-encoded binding pathways, so we utilized cell binding assays that specifically detect binding mediated by Tir-Intimin interaction. We also assessed the role of Tir-mediated actin assembly in two-step assays that temporally segregated initial translocation of Tir from subsequent Tir-Intimin interaction, thereby permitting the distinction of effects on translocation from effects on cell attachment. In these experimental systems, we compromised Tir-mediated actin assembly by chemically inhibiting actin assembly or by infecting mammalian cells with EHEC mutants that translocate Tir but are specifically defective in Tir-mediated pedestal formation. We found that an inability of Tir to promote actin assembly resulted in a significant and striking decrease in bacterial binding mediated by Tir and Intimin. Bacterial mutants defective for pedestal formation translocated type III effectors to mammalian cells with reduced efficiency, but the decrease in translocation could be entirely accounted for by the decrease in host cell attachment.
Collapse
|
26
|
Direct neutralization of type III effector translocation by the variable region of a monoclonal antibody to Yersinia pestis LcrV. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2014; 21:667-73. [PMID: 24599533 DOI: 10.1128/cvi.00013-14] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Plague is an acute infection caused by the Gram-negative bacterium Yersinia pestis. Antibodies that are protective against plague target LcrV, an essential virulence protein and component of a type III secretion system of Y. pestis. Secreted LcrV localizes to the tips of type III needles on the bacterial surface, and its function is necessary for the translocation of Yersinia outer proteins (Yops) into the cytosol of host cells infected by Y. pestis. Translocated Yops counteract macrophage functions, for example, by inhibiting phagocytosis (YopE) or inducing cytotoxicity (YopJ). Although LcrV is the best-characterized protective antigen of Y. pestis, the mechanism of protection by anti-LcrV antibodies is not fully understood. Antibodies bind to LcrV at needle tips, neutralize Yop translocation, and promote opsonophagocytosis of Y. pestis by macrophages in vitro. However, it is not clear if anti-LcrV antibodies neutralize Yop translocation directly or if they do so indirectly, by promoting opsonophagocytosis. To determine if the protective IgG1 monoclonal antibody (MAb) 7.3 is directly neutralizing, an IgG2a subclass variant, a deglycosylated variant, F(ab')2, and Fab were tested for the ability to inhibit the translocation of Yops into Y. pestis-infected macrophages in vitro. Macrophage cytotoxicity and cellular fractionation assays show that the Fc of MAb 7.3 is not required for the neutralization of YopJ or YopE translocation. In addition, the use of Fc receptor-deficient macrophages, and the use of cytochalasin D to inhibit actin polymerization, confirmed that opsonophagocytosis is not required for MAb 7.3 to neutralize translocation. These data indicate that the binding of the variable region of MAb 7.3 to LcrV is sufficient to directly neutralize Yop translocation.
Collapse
|
27
|
Schweer J, Kulkarni D, Kochut A, Pezoldt J, Pisano F, Pils MC, Genth H, Huehn J, Dersch P. The cytotoxic necrotizing factor of Yersinia pseudotuberculosis (CNFY) enhances inflammation and Yop delivery during infection by activation of Rho GTPases. PLoS Pathog 2013; 9:e1003746. [PMID: 24244167 PMCID: PMC3820761 DOI: 10.1371/journal.ppat.1003746] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 09/20/2013] [Indexed: 12/19/2022] Open
Abstract
Some isolates of Yersinia pseudotuberculosis produce the cytotoxic necrotizing factor (CNFY), but the functional consequences of this toxin for host-pathogen interactions during the infection are unknown. In the present study we show that CNFY has a strong influence on virulence. We demonstrate that the CNFY toxin is thermo-regulated and highly expressed in all colonized lymphatic tissues and organs of orally infected mice. Most strikingly, we found that a cnfY knock-out variant of a naturally toxin-expressing Y. pseudotuberculosis isolate is strongly impaired in its ability to disseminate into the mesenteric lymph nodes, liver and spleen, and has fully lost its lethality. The CNFY toxin contributes significantly to the induction of acute inflammatory responses and to the formation of necrotic areas in infected tissues. The analysis of the host immune response demonstrated that presence of CNFY leads to a strong reduction of professional phagocytes and natural killer cells in particular in the spleen, whereas loss of the toxin allows efficient tissue infiltration of these immune cells and rapid killing of the pathogen. Addition of purified CNFY triggers formation of actin-rich membrane ruffles and filopodia, which correlates with the activation of the Rho GTPases, RhoA, Rac1 and Cdc42. The analysis of type III effector delivery into epithelial and immune cells in vitro and during the course of the infection further demonstrated that CNFY enhances the Yop translocation process and supports a role for the toxin in the suppression of the antibacterial host response. In summary, we highlight the importance of CNFY for pathogenicity by showing that this toxin modulates inflammatory responses, protects the bacteria from attacks of innate immune effectors and enhances the severity of a Yersinia infection. Various toxins and effector proteins of bacterial pathogens have been found to manipulate eukaryotic cell machineries to promote persistence and proliferation within their hosts. Many of these virulence factors target small Rho GTPases, but their role in pathogenesis is often unknown. Here, we addressed the expression and functional consequences of the CNFY toxin found in some isolates of Y. pseudotuberculosis. We found that CNFY besides modulating the cell cytoskeleton by activation of the GTPases RhoA, Rac1 and Cdc42, contributes to increased inflammation and tissue damage. Moreover, CNFY increases the ability of Yersinia to prevent the attack of the immune system, by enhancing the delivery of antiphagocytic and cytotoxic effectors into professional phagocytes. Our findings provide the first insights into the multi-functional action and severe consequences of the CNFY toxin on the inflammatory response and disease-associated tissue damage during the natural course of the infection.
Collapse
Affiliation(s)
- Janina Schweer
- Department of Molecular Infection Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Devesha Kulkarni
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Annika Kochut
- Department of Molecular Infection Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Joern Pezoldt
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Fabio Pisano
- Department of Molecular Infection Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Marina C. Pils
- Mouse Pathology, Animal Experimental Unit, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Harald Genth
- Institute for Toxicology, Medical School Hannover, Hannover, Germany
| | - Jochen Huehn
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Petra Dersch
- Department of Molecular Infection Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- * E-mail:
| |
Collapse
|
28
|
Paczosa MK, Fisher ML, Maldonado-Arocho FJ, Mecsas J. Yersinia pseudotuberculosis uses Ail and YadA to circumvent neutrophils by directing Yop translocation during lung infection. Cell Microbiol 2013; 16:247-68. [PMID: 24119087 DOI: 10.1111/cmi.12219] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 09/03/2013] [Accepted: 09/19/2013] [Indexed: 12/14/2022]
Abstract
A Yersinia pseudotuberculosis (Yptb) murine model of lung infection was previously developed using the serotype III IP2666NdeI strain, which robustly colonized lungs but only sporadically disseminated to the spleen and liver. We demonstrate here that a serotype Ib Yptb strain, IP32953, colonizes the lungs at higher levels and disseminates more efficiently to the spleen and liver compared with IP2666NdeI . The role of adhesins was investigated during IP32953 lung infection by constructing isogenic Δail, Δinv, ΔpsaE and ΔyadA mutants. An IP32953ΔailΔyadA mutant initially colonized but failed to persist in the lungs and disseminate to the spleen and liver. Yptb expressing these adhesins selectively bound to and targeted neutrophils for translocation of Yops. This selective targeting was critical for virulence because persistence of the ΔailΔyadA mutant was restored following intranasal infection of neutropenic mice. Furthermore, Ail and YadA prevented killing by complement-mediated mechanisms during dissemination to and/or growth in the spleen and liver, but not in the lungs. Combined, these results demonstratethat Ail and YadA are critical, redundant virulence factors during lung infection, because they thwart neutrophils by directing Yop-translocation specifically into these cells.
Collapse
Affiliation(s)
- Michelle K Paczosa
- Graduate Program in Immunology, MERGE-ID Track, Sackler School of Biomedical Sciences, Tufts University, Boston, MA, USA
| | | | | | | |
Collapse
|
29
|
Bliska JB, Wang X, Viboud GI, Brodsky IE. Modulation of innate immune responses by Yersinia type III secretion system translocators and effectors. Cell Microbiol 2013; 15:1622-31. [PMID: 23834311 DOI: 10.1111/cmi.12164] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Revised: 06/21/2013] [Accepted: 07/01/2013] [Indexed: 12/13/2022]
Abstract
The innate immune system of mammals responds to microbial infection through detection of conserved molecular determinants called 'pathogen-associated molecular patterns' (PAMPs). Pathogens use virulence factors to counteract PAMP-directed responses. The innate immune system can in turn recognize signals generated by virulence factors, allowing for a heightened response to dangerous pathogens. Many Gram-negative bacterial pathogens encode type III secretion systems (T3SSs) that translocate effector proteins, subvert PAMP-directed responses and are critical for infection. A plasmid-encoded T3SS in the human-pathogenic Yersinia species translocates seven effectors into infected host cells. Delivery of effectors by the T3SS requires plasma membrane insertion of two translocators, which are thought to form a channel called a translocon. Studies of the Yersinia T3SS have provided key advances in our understanding of how innate immune responses are generated by perturbations in plasma membrane and other signals that result from translocon insertion. Additionally, studies in this system revealed that effectors function to inhibit innateimmune responses resulting from insertion of translocons into plasma membrane. Here, we review these advances with the goal of providing insight into how a T3SS can activate and inhibit innate immune responses, allowing a virulent pathogen to bypass host defences.
Collapse
Affiliation(s)
- James B Bliska
- Center for Infectious Diseases and Department of Molecular Genetics and Microbiology, School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | | | | | | |
Collapse
|
30
|
Shrestha N, Boucher J, Bahnan W, Clark ES, Rosqvist R, Fields KA, Khan WN, Schesser K. The host-encoded Heme Regulated Inhibitor (HRI) facilitates virulence-associated activities of bacterial pathogens. PLoS One 2013; 8:e68754. [PMID: 23874749 PMCID: PMC3707855 DOI: 10.1371/journal.pone.0068754] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 05/31/2013] [Indexed: 11/18/2022] Open
Abstract
Here we show that cells lacking the heme-regulated inhibitor (HRI) are highly resistant to infection by bacterial pathogens. By examining the infection process in wild-type and HRI null cells, we found that HRI is required for pathogens to execute their virulence-associated cellular activities. Specifically, unlike wild-type cells, HRI null cells infected with the gram-negative bacterial pathogen Yersinia are essentially impervious to the cytoskeleton-damaging effects of the Yop virulence factors. This effect is due to reduced functioning of the Yersinia type 3 secretion (T3S) system which injects virulence factors directly into the host cell cytosol. Reduced T3S activity is also observed in HRI null cells infected with the bacterial pathogen Chlamydia which results in a dramatic reduction in its intracellular proliferation. We go on to show that a HRI-mediated process plays a central role in the cellular infection cycle of the Gram-positive pathogen Listeria. For this pathogen, HRI is required for the post-invasion trafficking of the bacterium to the infected host cytosol. Thus by depriving Listeria of its intracellular niche, there is a highly reduced proliferation of Listeria in HRI null cells. We provide evidence that these infection-associated functions of HRI (an eIF2α kinase) are independent of its activity as a regulator of protein synthesis. This is the first report of a host factor whose absence interferes with the function of T3S secretion and cytosolic access by pathogens and makes HRI an excellent target for inhibitors due to its broad virulence-associated activities.
Collapse
Affiliation(s)
- Niraj Shrestha
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Justin Boucher
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Wael Bahnan
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Emily S. Clark
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Roland Rosqvist
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Kenneth A. Fields
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Wasif N. Khan
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Kurt Schesser
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- * E-mail:
| |
Collapse
|
31
|
Wolters M, Boyle EC, Lardong K, Trülzsch K, Steffen A, Rottner K, Ruckdeschel K, Aepfelbacher M. Cytotoxic necrotizing factor-Y boosts Yersinia effector translocation by activating Rac protein. J Biol Chem 2013; 288:23543-53. [PMID: 23803609 DOI: 10.1074/jbc.m112.448662] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Pathogenic Yersinia spp. translocate the effectors YopT, YopE, and YopO/YpkA into target cells to inactivate Rho family GTP-binding proteins and block immune responses. Some Yersinia spp. also secrete the Rho protein activator cytotoxic necrotizing factor-Y (CNF-Y), but it has been unclear how the bacteria may benefit from Rho protein activation. We show here that CNF-Y increases Yop translocation in Yersinia enterocolitica-infected cells up to 5-fold. CNF-Y strongly activated RhoA and also delayed in time Rac1 and Cdc42, but when individually expressed, constitutively active mutants of Rac1, but not of RhoA, increased Yop translocation. Consistently, knock-out or knockdown of Rac1 but not of RhoA, -B, or -C inhibited Yersinia effector translocation in CNF-Y-treated and control cells. Activation or knockdown of Cdc42 also affected Yop translocation but much less efficiently than Rac. The increase in Yop translocation induced by CNF-Y was essentially independent of the presence of YopE, YopT, or YopO in the infecting Yersinia strain, indicating that none of the Yops reported to inhibit translocation could reverse the CNF-Y effect. In summary, the CNF-Y activity of Yersinia strongly enhances Yop translocation through activation of Rac.
Collapse
Affiliation(s)
- Manuel Wolters
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Maldonado-Arocho FJ, Green C, Fisher ML, Paczosa MK, Mecsas J. Adhesins and host serum factors drive Yop translocation by yersinia into professional phagocytes during animal infection. PLoS Pathog 2013; 9:e1003415. [PMID: 23818844 PMCID: PMC3688556 DOI: 10.1371/journal.ppat.1003415] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 04/29/2013] [Indexed: 11/18/2022] Open
Abstract
Yersinia delivers Yops into numerous types of cultured cells, but predominantly into professional phagocytes and B cells during animal infection. The basis for this cellular tropism during animal infection is not understood. This work demonstrates that efficient and specific Yop translocation into phagocytes by Yersinia pseudotuberculosis (Yptb) is a multi-factorial process requiring several adhesins and host complement. When WT Yptb or a multiple adhesin mutant strain, ΔailΔinvΔyadA, colonized tissues to comparable levels, ΔailΔinvΔyadA translocated Yops into significantly fewer cells, demonstrating that these adhesins are critical for translocation into high numbers of cells. However, phagocytes were still selectively targeted for translocation, indicating that other bacterial and/or host factors contribute to this function. Complement depletion showed that complement-restricted infection by ΔailΔinvΔyadA but not WT, indicating that adhesins disarm complement in mice either by prevention of opsonophagocytosis or by suppressing production of pro-inflammatory cytokines. Furthermore, in the absence of the three adhesins and complement, the spectrum of cells targeted for translocation was significantly altered, indicating that Yersinia adhesins and complement direct Yop translocation into neutrophils during animal infection. In summary, these findings demonstrate that in infected tissues, Yersinia uses adhesins both to disarm complement-dependent killing and to efficiently translocate Yops into phagocytes. Many bacterial pathogens use a needle-like structure to deliver proteins into host cells to cause disease. Yersinia species use one such structure, called a type III secretion system, to deliver a set of 6–7 proteins, called Yops, into host cells. These Yops act to dismantle host defenses and establish infection. Bacterial adhesins and host factors have been suggested to promote proper delivery of Yops into specific mammalian cells. We identify three Yersinia pseudotuberculosis adhesins that significantly contribute to bacterial survival and efficient Yop delivery into host cells during animal infection. We also demonstrate that host serum factors in combination with Yersinia adhesins contribute to the number of cells that are injected with Yops and to the specific cell types targeted for injection. Our study illustrates that bacterial adhesins and host factors contribute to efficient delivery of effector proteins into targeted host cells during infection.
Collapse
Affiliation(s)
| | - Carlos Green
- Sackler School of Biomedical Sciences, Boston, Massachusetts, United States of America
| | - Michael L. Fisher
- Sackler School of Biomedical Sciences, Boston, Massachusetts, United States of America
| | - Michelle K. Paczosa
- Sackler School of Biomedical Sciences, Boston, Massachusetts, United States of America
| | - Joan Mecsas
- Tufts University School of Medicine, Boston, Massachusetts, United States of America
- Sackler School of Biomedical Sciences, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
33
|
Dewoody RS, Merritt PM, Marketon MM. Regulation of the Yersinia type III secretion system: traffic control. Front Cell Infect Microbiol 2013; 3:4. [PMID: 23390616 PMCID: PMC3565153 DOI: 10.3389/fcimb.2013.00004] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Accepted: 01/16/2013] [Indexed: 01/03/2023] Open
Abstract
Yersinia species, as well as many other Gram-negative pathogens, use a type III secretion system (T3SS) to translocate effector proteins from the bacterial cytoplasm to the host cytosol. This T3SS resembles a molecular syringe, with a needle-like shaft connected to a basal body structure, which spans the inner and outer bacterial membranes. The basal body of the injectisome shares a high degree of homology with the bacterial flagellum. Extending from the T3SS basal body is the needle, which is a polymer of a single protein, YscF. The distal end of the needle serves as a platform for the assembly of a tip complex composed of LcrV. Though never directly observed, prevailing models assume that LcrV assists in the insertion of the pore-forming proteins YopB and YopD into the host cell membrane. This completes a bridge between the bacterium and host cell to provide a continuous channel through which effectors are delivered. Significant effort has gone into understanding how the T3SS is assembled, how its substrates are recognized and how substrate delivery is controlled. Arguably the latter topic is the least understood; however, recent advances have provided new insight, and therefore, this review will focus primarily on summarizing the current state of knowledge regarding the control of substrate delivery by the T3SS. Specifically, we will discuss the roles of YopK, as well as YopN and YopE, which have long been linked to regulation of translocation. We also propose models whereby the YopK regulator communicates with the basal body of the T3SS to control translocation.
Collapse
|
34
|
Dewoody R, Merritt PM, Marketon MM. YopK controls both rate and fidelity of Yop translocation. Mol Microbiol 2013; 87:301-17. [PMID: 23205707 PMCID: PMC3545096 DOI: 10.1111/mmi.12099] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2012] [Indexed: 11/27/2022]
Abstract
Yersinia pestis, the causative agent of plague, utilizes a type III secretion system (T3SS) to intoxicate host cells. The injection of T3SS substrates must be carefully controlled, and dysregulation leads to altered infection kinetics and early clearance of Y. pestis. While the sequence of events leading up to cell contact and initiation of translocation has received much attention, the regulatory events that take place after effector translocation is less understood. Here we show that the regulator YopK is required to maintain fidelity of substrate specificity, in addition to controlling translocation rate. YopK was found to interact with YopD within targeted cells during Y. pestis infection, suggesting that YopK's regulatory mechanism involves a direct interaction with the translocation pore. In addition, we identified a single amino acid in YopK that is essential for translocation rate regulation but is dispensable for maintaining fidelity of translocation. Furthermore, we found that expression of YopK within host cells was sufficient to downregulate translocation rate, but it did not affect translocation fidelity. Together, our data support a model in which YopK is a bifunctional protein whose activities are genetically and spatially distinct such that fidelity control occurs within bacteria and rate control occurs within host cells.
Collapse
|
35
|
Protein export according to schedule: architecture, assembly, and regulation of type III secretion systems from plant- and animal-pathogenic bacteria. Microbiol Mol Biol Rev 2012; 76:262-310. [PMID: 22688814 DOI: 10.1128/mmbr.05017-11] [Citation(s) in RCA: 312] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Flagellar and translocation-associated type III secretion (T3S) systems are present in most gram-negative plant- and animal-pathogenic bacteria and are often essential for bacterial motility or pathogenicity. The architectures of the complex membrane-spanning secretion apparatuses of both systems are similar, but they are associated with different extracellular appendages, including the flagellar hook and filament or the needle/pilus structures of translocation-associated T3S systems. The needle/pilus is connected to a bacterial translocon that is inserted into the host plasma membrane and mediates the transkingdom transport of bacterial effector proteins into eukaryotic cells. During the last 3 to 5 years, significant progress has been made in the characterization of membrane-associated core components and extracellular structures of T3S systems. Furthermore, transcriptional and posttranscriptional regulators that control T3S gene expression and substrate specificity have been described. Given the architecture of the T3S system, it is assumed that extracellular components of the secretion apparatus are secreted prior to effector proteins, suggesting that there is a hierarchy in T3S. The aim of this review is to summarize our current knowledge of T3S system components and associated control proteins from both plant- and animal-pathogenic bacteria.
Collapse
|
36
|
López-Gómez A, Cano V, Moranta D, Morey P, García Del Portillo F, Bengoechea JA, Garmendia J. Host cell kinases, α5 and β1 integrins, and Rac1 signalling on the microtubule cytoskeleton are important for non-typable Haemophilus influenzae invasion of respiratory epithelial cells. MICROBIOLOGY-SGM 2012; 158:2384-2398. [PMID: 22723286 DOI: 10.1099/mic.0.059972-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Non-typable Haemophilus influenzae (NTHi) is a common commensal of the human nasopharynx, but causes opportunistic infection when the respiratory tract is compromised by infection or disease. The ability of NTHi to invade epithelial cells has been described, but the underlying molecular mechanisms are poorly characterized. We previously determined that NTHi promotes phosphorylation of the serine-threonine kinase Akt in A549 human lung epithelial cells, and that Akt phosphorylation and NTHi cell invasion are prevented by inhibition of phosphoinositide 3-kinase (PI3K). Because PI3K-Akt signalling is associated with several host cell networks, the purpose of the current study was to identify eukaryotic molecules important for NTHi epithelial invasion. We found that inhibition of Akt activity reduced NTHi internalization; differently, bacterial entry was increased by phospholipase Cγ1 inhibition but was not affected by protein kinase inhibition. We also found that α5 and β1 integrins, and the tyrosine kinases focal adhesion kinase and Src, are important for NTHi A549 cell invasion. NTHi internalization was shown to be favoured by activation of Rac1 guanosine triphosphatase (GTPase), together with the guanine nucleotide exchange factor Vav2 and the effector Pak1. Also, Pak1 might be associated with inactivation of the microtubule destabilizing agent Op18/stathmin, to facilitate microtubule polymerization and NTHi entry. Conversely, inhibition of RhoA GTPase and its effector ROCK increased the number of internalized bacteria. Src and Rac1 were found to be important for NTHi-triggered Akt phosphorylation. An increase in host cyclic AMP reduced bacterial entry, which was linked to protein kinase A. These findings suggest that NTHi finely manipulates host signalling molecules to invade respiratory epithelial cells.
Collapse
Affiliation(s)
- Antonio López-Gómez
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Bunyola, Spain.,Laboratory Microbial Pathogenesis, Fundación Investigación Sanitaria Illes Balears, Bunyola, Spain
| | - Victoria Cano
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Bunyola, Spain.,Laboratory Microbial Pathogenesis, Fundación Investigación Sanitaria Illes Balears, Bunyola, Spain
| | - David Moranta
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Bunyola, Spain.,Laboratory Microbial Pathogenesis, Fundación Investigación Sanitaria Illes Balears, Bunyola, Spain
| | - Pau Morey
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Bunyola, Spain.,Laboratory Microbial Pathogenesis, Fundación Investigación Sanitaria Illes Balears, Bunyola, Spain
| | | | - José Antonio Bengoechea
- Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Bunyola, Spain.,Laboratory Microbial Pathogenesis, Fundación Investigación Sanitaria Illes Balears, Bunyola, Spain
| | - Junkal Garmendia
- Instituto de Agrobiotecnología, CSIC-Universidad Pública de Navarra-Gobierno de Navarra, Mutilva, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Bunyola, Spain.,Laboratory Microbial Pathogenesis, Fundación Investigación Sanitaria Illes Balears, Bunyola, Spain
| |
Collapse
|
37
|
Examining the role of actin-plasma membrane association in Pseudomonas aeruginosa infection and type III secretion translocation in migratory T24 epithelial cells. Infect Immun 2012; 80:3049-64. [PMID: 22689823 DOI: 10.1128/iai.00231-12] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The opportunistic pathogen Pseudomonas aeruginosa targets wounded epithelial barriers, but the cellular alteration that increases susceptibility to P. aeruginosa infection remains unclear. This study examined how cell migration contributes to the establishment of P. aeruginosa infections using (i) highly migratory T24 epithelial cells as a cell culture model, (ii) mutations in the type III secretion (T3S) effector ExoS to manipulate P. aeruginosa infection, and (iii) high-resolution immunofluorescent microscopy to monitor ExoS translocation. ExoS includes both GTPase-activating (GAP) and ADP-ribosyltransferase (ADPRT) activities, and P. aeruginosa cells expressing wild-type ExoS preferentially bound to the leading edge of T24 cells, where ExoS altered leading-edge architecture and actin anchoring in conjunction with interrupting T3S translocation. Inactivation of ExoS GAP activity allowed P. aeruginosa to be internalized and secrete ExoS within T24 cells, but as with wild-type ExoS, translocation was limited in association with disruption of actin anchoring. Inactivation of ExoS ADPRT activity resulted in significantly enhanced T3S translocation by P. aeruginosa cells that remained extracellular and in conjunction with maintenance of actin-plasma membrane association. Infection with P. aeruginosa expressing ExoS lacking both GAP and ADPRT activities resulted in the highest level of T3S translocation, and this occurred in conjunction with the entry and alignment of P. aeruginosa and ExoS along actin filaments. Collectively, in using ExoS mutants to modulate and visualize T3S translocation, we were able to (i) confirm effector secretion by internalized P. aeruginosa, (ii) differentiate the mechanisms underlying the effects of ExoS GAP and ADPRT activities on P. aeruginosa internalization and T3S translocation, (iii) confirm that ExoS ADPRT activity targeted a cellular substrate that interrupted T3S translocation, (iv) visualize the ability of P. aeruginosa and ExoS to align with actin filaments, and (v) demonstrate an association between actin anchoring at the leading edge of T24 cells and the establishment of P. aeruginosa infection. Our studies also highlight the contribution of ExoS to the opportunistic nature of P. aeruginosa infection through its ability to exert cytotoxic effects that interrupt T3S translocation and P. aeruginosa internalization, which in turn limit the P. aeruginosa infectious process.
Collapse
|
38
|
Tsang TM, Annis DS, Kronshage M, Fenno JT, Usselman LD, Mosher DF, Krukonis ES. Ail protein binds ninth type III fibronectin repeat (9FNIII) within central 120-kDa region of fibronectin to facilitate cell binding by Yersinia pestis. J Biol Chem 2012; 287:16759-67. [PMID: 22447929 DOI: 10.1074/jbc.m112.358978] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The Yersinia pestis adhesin molecule Ail interacts with the extracellular matrix protein fibronectin (Fn) on host cells to facilitate efficient delivery of cytotoxic Yop proteins, a process essential for plague virulence. A number of bacterial pathogens are known to bind to the N-terminal region of Fn, comprising type I Fn (FNI) repeats. Using proteolytically generated Fn fragments and purified recombinant Fn fragments, we demonstrated that Ail binds the centrally located 120-kDa fragment containing type III Fn (FNIII) repeats. A panel of monoclonal antibodies (mAbs) that recognize specific epitopes within the 120-kDa fragment demonstrated that mAb binding to (9)FNIII blocks Ail-mediated bacterial binding to Fn. Epitopes of three mAbs that blocked Ail binding to Fn were mapped to a similar face of (9)FNIII. Antibodies directed against (9)FNIII also inhibited Ail-dependent cell binding activity, thus demonstrating the biological relevance of this Ail binding region on Fn. Bacteria expressing Ail on their surface could also bind a minimal fragment of Fn containing repeats (9-10)FNIII, and this binding was blocked by a mAb specific for (9)FNIII. These data demonstrate that Ail binds to (9)FNIII of Fn and presents Fn to host cells to facilitate cell binding and delivery of Yops (cytotoxins of Y. pestis), a novel interaction, distinct from other bacterial Fn-binding proteins.
Collapse
Affiliation(s)
- Tiffany M Tsang
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
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: 1.8] [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
|
40
|
Peters KN, Anderson DM. Modulation of host cell death pathways by Yersinia species and the type III effector YopK. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 954:229-36. [PMID: 22782768 DOI: 10.1007/978-1-4614-3561-7_29] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Kristen N Peters
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
| | | |
Collapse
|
41
|
A protective epitope in type III effector YopE is a major CD8 T cell antigen during primary infection with Yersinia pseudotuberculosis. Infect Immun 2011; 80:206-14. [PMID: 22064714 DOI: 10.1128/iai.05971-11] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Virulence in human-pathogenic Yersinia species is associated with a plasmid-encoded type III secretion system that translocates a set of Yop effector proteins into host cells. One effector, YopE, functions as a Rho GTPase-activating protein (GAP). In addition to acting as a virulence factor, YopE can function as a protective antigen. C57BL/6 mice infected with attenuated Yersinia pestis generate a dominant H2-Kb-restricted CD8 T cell response to an epitope in the N-terminal domain of YopE (YopE69-77), and intranasal vaccination with the YopE69-77 peptide and the mucosal adjuvant cholera toxin (CT) elicits CD8 T cells that are protective against lethal pulmonary challenge with Y. pestis. Because YopE69-77 is conserved in many Yersinia strains, we sought to determine if YopE is a protective antigen for Yersinia pseudotuberculosis and if primary infection with this enteric pathogen elicits a CD8 T cell response to this epitope. Intranasal immunization with the YopE69-77 peptide and CT elicited a CD8 T cell response that was protective against lethal intragastric Y. pseudotuberculosis challenge. The YopE69-77 epitope was a major antigen (∼30% of splenic CD8 T cells were specific for this peptide at the peak of the response) during primary infection with Y. pseudotuberculosis, as shown by flow cytometry tetramer staining. Results of infections with Y. pseudotuberculosis expressing catalytically inactive YopE demonstrated that GAP activity is dispensable for a CD8 T cell response to YopE69-77. Determining the features of YopE that are important for this response will lead to a better understanding of how protective CD8 T cell immunity is generated against Yersinia and other pathogens with type III secretion systems.
Collapse
|
42
|
Galindo CL, Rosenzweig JA, Kirtley ML, Chopra AK. Pathogenesis of Y. enterocolitica and Y. pseudotuberculosis in Human Yersiniosis. J Pathog 2011; 2011:182051. [PMID: 22567322 PMCID: PMC3335670 DOI: 10.4061/2011/182051] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Revised: 06/27/2011] [Accepted: 07/01/2011] [Indexed: 12/15/2022] Open
Abstract
Yersiniosis is a food-borne illness that has become more prevalent in recent years due to human transmission via the fecal-oral route and prevalence in farm animals. Yersiniosis is primarily caused by Yersinia enterocolitica and less frequently by Yersinia pseudotuberculosis. Infection is usually characterized by a self-limiting acute infection beginning in the intestine and spreading to the mesenteric lymph nodes. However, more serious infections and chronic conditions can also occur, particularly in immunocompromised individuals. Y. enterocolitica and Y. pseudotuberculosis are both heterogeneous organisms that vary considerably in their degrees of pathogenicity, although some generalizations can be ascribed to pathogenic variants. Adhesion molecules and a type III secretion system are critical for the establishment and progression of infection. Additionally, host innate and adaptive immune responses are both required for yersiniae clearance. Despite the ubiquity of enteric Yersinia species and their association as important causes of food poisoning world-wide, few national enteric pathogen surveillance programs include the yersiniae as notifiable pathogens. Moreover, no standard exists whereby identification and reporting systems can be effectively compared and global trends developed. This review discusses yersinial virulence factors, mechanisms of infection, and host responses in addition to the current state of surveillance, detection, and prevention of yersiniosis.
Collapse
Affiliation(s)
- Cristi L Galindo
- Department of Microbiology & Immunology, Sealy Center for Vaccine Development, Institute of Human Infections & Immunity, and the Galveston National Laboratory, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-1070, USA
| | | | | | | |
Collapse
|
43
|
Insight into bacterial virulence mechanisms against host immune response via the Yersinia pestis-human protein-protein interaction network. Infect Immun 2011; 79:4413-24. [PMID: 21911467 DOI: 10.1128/iai.05622-11] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
A Yersinia pestis-human protein interaction network is reported here to improve our understanding of its pathogenesis. Up to 204 interactions between 66 Y. pestis bait proteins and 109 human proteins were identified by yeast two-hybrid assay and then combined with 23 previously published interactions to construct a protein-protein interaction network. Topological analysis of the interaction network revealed that human proteins targeted by Y. pestis were significantly enriched in the proteins that are central in the human protein-protein interaction network. Analysis of this network showed that signaling pathways important for host immune responses were preferentially targeted by Y. pestis, including the pathways involved in focal adhesion, regulation of cytoskeleton, leukocyte transendoepithelial migration, and Toll-like receptor (TLR) and mitogen-activated protein kinase (MAPK) signaling. Cellular pathways targeted by Y. pestis are highly relevant to its pathogenesis. Interactions with host proteins involved in focal adhesion and cytoskeketon regulation pathways could account for resistance of Y. pestis to phagocytosis. Interference with TLR and MAPK signaling pathways by Y. pestis reflects common characteristics of pathogen-host interaction that bacterial pathogens have evolved to evade host innate immune response by interacting with proteins in those signaling pathways. Interestingly, a large portion of human proteins interacting with Y. pestis (16/109) also interacted with viral proteins (Epstein-Barr virus [EBV] and hepatitis C virus [HCV]), suggesting that viral and bacterial pathogens attack common cellular functions to facilitate infections. In addition, we identified vasodilator-stimulated phosphoprotein (VASP) as a novel interaction partner of YpkA and showed that YpkA could inhibit in vitro actin assembly mediated by VASP.
Collapse
|
44
|
Abstract
Invasion of non-phagocytic cells by a number of bacterial pathogens involves the subversion of the actin cytoskeletal remodelling machinery to produce actin-rich cell surface projections designed to engulf the bacteria. The signalling that occurs to induce these actin-rich structures has considerable overlap among a diverse group of bacteria. The molecular organization within these structures act in concert to internalize the invading pathogen. This dynamic process could be subdivided into three acts - actin recruitment, engulfment, and finally, actin disassembly/internalization. This review will present the current state of knowledge of the molecular processes involved in each stage of bacterial invasion, and provide a perspective that highlights the temporal and spatial control of actin remodelling that occurs during bacterial invasion.
Collapse
Affiliation(s)
- Rey Carabeo
- Centre for Molecular Microbiology and Infection, Division of Cell and Molecular Biology, Imperial College London, London, UK.
| |
Collapse
|
45
|
Wölke S, Ackermann N, Heesemann J. The Yersinia enterocolitica type 3 secretion system (T3SS) as toolbox for studying the cell biological effects of bacterial Rho GTPase modulating T3SS effector proteins. Cell Microbiol 2011; 13:1339-57. [PMID: 21718421 DOI: 10.1111/j.1462-5822.2011.01623.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: 01/08/2023]
Abstract
The bacterial effector proteins IpgB(1) and IpgB(2) of Shigella and Map of Escherichia coli activate the Rho GTPases Rac1, RhoA and Cdc42, respectively, whereas YopE and YopT of Yersinia inhibit these Rho family GTPases. We established a Yersinia toolbox which allows to study the cellular effects of these effectors in different combinations in the context of Yersinia type 3 secretion system (Ysc)-T3SS-mediated injection into HeLa cells. For this purpose hybrid proteins were constructed by fusion of YopE with the effector protein of interest. As expected, injected hybrid proteins induced membrane ruffles and Yersinia uptake for IpgB(1) , stress fibres for IpgB(2) and microspikes for Map. By co-infection experiments we could demonstrate (i) IpgB(2) -mediated and ROCK-dependent inhibition of IpgB(1) -mediated Rac1 effects, (ii) YopT-mediated suppression of IpgB(1) -induced Yersinia invasion and (iii) failure of YopE-mediated suppression of IpgB(1) -induced Yersinia invasion, presumably due to preferential inhibition of RhoG by YopE GAP function. By infecting polarized MDCK cells we could demonstrate that Map or IpgB(1) but not IpgB(2) affects cell monolayer integrity. In summary, the Yersinia toolbox is suitable to study cellular effects of effector proteins of diverse bacterial species separately or in combination in the context of bacterial T3SS-mediated injection.
Collapse
Affiliation(s)
- Stefan Wölke
- Max von Pettenkofer Institut, LMU Munich, Pettenkofer Straße 9A, 80336 Munich, Germany
| | | | | |
Collapse
|
46
|
Dewoody R, Merritt PM, Houppert AS, Marketon MM. YopK regulates the Yersinia pestis type III secretion system from within host cells. Mol Microbiol 2011; 79:1445-61. [PMID: 21205017 PMCID: PMC3210821 DOI: 10.1111/j.1365-2958.2011.07534.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The pathogenic Yersinia species share a conserved type III secretion system, which delivers cytotoxic effectors known as Yops into target mammalian cells. In all three species, YopK (also called YopQ) plays an important role in regulating this process. In cell culture infections, yopK mutants inject higher levels of Yops, leading to increase cytotoxicity; however, in vivo the same mutants are highly attenuated. In this work, we investigate the mechanism behind this paradox. Using a β-lactamase reporter assay to directly measure the effect of YopK on translocation, we demonstrated that YopK controls the rate of Yop injection. Furthermore, we find that YopK cannot regulate effector Yop translocation from within the bacterial cytosol. YopE is also injected into host cells and was previously shown to contribute to regulation of the injectisome. In this work we show that YopK and YopE work at different steps to regulate Yop injection, with YopK functioning independently of YopE. Finally, by expressing YopK within tissue culture cells, we confirm that YopK regulates translocation from inside the host cell, and we show that cells pre-loaded with YopK are resistant to Yop injection. These results suggest a novel role for YopK in controlling the Yersinia type III secretion system.
Collapse
Affiliation(s)
- Rebecca Dewoody
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Peter M. Merritt
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | | | | |
Collapse
|
47
|
Aepfelbacher M, Roppenser B, Hentschke M, Ruckdeschel K. Activity modulation of the bacterial Rho GAP YopE: an inspiration for the investigation of mammalian Rho GAPs. Eur J Cell Biol 2011; 90:951-4. [PMID: 21255863 DOI: 10.1016/j.ejcb.2010.12.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2010] [Revised: 12/15/2010] [Accepted: 12/15/2010] [Indexed: 12/16/2022] Open
Abstract
The Yersinia enterocolitica Rho GTPase Activating Protein (Rho GAP) YopE belongs to a group of bacterial virulence factors that is translocated into infected target cells by a type three secretion system. Structurally and biochemically YopE resembles eukaryotic Rho GAPs which control various cellular functions by modulating the activity of Rho GTP binding proteins. Here we summarise the published information on cellular effects, Rho protein substrates, compartmentalisation and turnover of YopE. A fascinating picture evolves of how this virulence factor integrates in host cellular regulatory mechanisms to fine tune bacterial pathogenicity.
Collapse
Affiliation(s)
- Martin Aepfelbacher
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany.
| | | | | | | |
Collapse
|
48
|
Day B, Henty JL, Porter KJ, Staiger CJ. The pathogen-actin connection: a platform for defense signaling in plants. ANNUAL REVIEW OF PHYTOPATHOLOGY 2011; 49:483-506. [PMID: 21495845 DOI: 10.1146/annurev-phyto-072910-095426] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
The cytoskeleton, a dynamic network of cytoplasmic polymers, plays a central role in numerous fundamental processes, such as development, reproduction, and cellular responses to biotic and abiotic stimuli. As a platform for innate immune responses in mammalian cells, the actin cytoskeleton is a central component in the organization and activation of host defenses, including signaling and cellular repair. In plants, our understanding of the genetic and biochemical responses in both pathogen and host that are required for virulence and resistance has grown enormously. Additional advances in live-cell imaging of cytoskeletal dynamics have markedly altered our view of actin turnover in plants. In this review, we outline current knowledge of host resistance following pathogen perception, both in terms of the genetic interactions that mediate defense signaling, as well as the biochemical and cellular processes that are required for defense signaling.
Collapse
Affiliation(s)
- Brad Day
- Department of Plant Pathology, Michigan State University, East Lansing, Michigan 48824-1311, USA.
| | | | | | | |
Collapse
|
49
|
Destabilization of YopE by the ubiquitin-proteasome pathway fine-tunes Yop delivery into host cells and facilitates systemic spread of Yersinia enterocolitica in host lymphoid tissue. Infect Immun 2010; 79:1166-75. [PMID: 21149597 DOI: 10.1128/iai.00694-10] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pathogenic Yersinia species inject a panel of Yop virulence proteins by type III protein secretion into host cells to modulate cellular defense responses. This enables the survival and dissemination of the bacteria in the host lymphoid tissue. We have previously shown that YopE of the Y. enterocolitica serogroup O8 is degraded in the host cell through the ubiquitin-proteasome pathway. YopE normally manipulates rearrangements of the actin cytoskeleton and triggers phagocytosis resistance. To shed light into the physiological role of YopE inactivation, we mutagenized the lysine polyubiquitin acceptor sites of YopE in the Y. enterocolitica serogroup O8 virulence plasmid. The resulting mutant strain escaped polyubiquitination and degradation of YopE and displayed increased intracellular YopE levels, which was accompanied by a pronounced cytotoxic effect on infected cells. Despite its intensified activity on cultured cells, the Yersinia mutant with stabilized YopE showed reduced dissemination into liver and spleen following enteral infection of mice. Furthermore, the accumulation of degradation-resistant YopE was accompanied by the diminished delivery of YopP and YopH into cultured, Yersinia-infected cells. A role of YopE in the regulation of Yop translocation has already been described. Our results imply that the inactivation of YopE by the proteasome could be a tool to ensure intermediate intracellular YopE levels, which may effectuate optimized Yop injection into host cells. In this regard, Y. enterocolitica O8 appears to exploit the host ubiquitin proteasome system to destabilize YopE and to fine-tune the activities of the Yop virulence arsenal on the infected host organism.
Collapse
|
50
|
Vingadassalom D, Campellone KG, Brady MJ, Skehan B, Battle SE, Robbins D, Kapoor A, Hecht G, Snapper SB, Leong JM. Enterohemorrhagic E. coli requires N-WASP for efficient type III translocation but not for EspFU-mediated actin pedestal formation. PLoS Pathog 2010; 6:e1001056. [PMID: 20808845 PMCID: PMC2924363 DOI: 10.1371/journal.ppat.1001056] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2009] [Accepted: 07/21/2010] [Indexed: 12/19/2022] Open
Abstract
Upon infection of mammalian cells, enterohemorrhagic E. coli (EHEC) O157:H7 utilizes a type III secretion system to translocate the effectors Tir and EspFU (aka TccP) that trigger the formation of F-actin-rich ‘pedestals’ beneath bound bacteria. EspFU is localized to the plasma membrane by Tir and binds the nucleation-promoting factor N-WASP, which in turn activates the Arp2/3 actin assembly complex. Although N-WASP has been shown to be required for EHEC pedestal formation, the precise steps in the process that it influences have not been determined. We found that N-WASP and actin assembly promote EHEC-mediated translocation of Tir and EspFU into mammalian host cells. When we utilized the related pathogen enteropathogenic E. coli to enhance type III translocation of EHEC Tir and EspFU, we found surprisingly that actin pedestals were generated on N-WASP-deficient cells. Similar to pedestal formation on wild type cells, Tir and EspFU were the only bacterial effectors required for pedestal formation, and the EspFU sequences required to interact with N-WASP were found to also be essential to stimulate this alternate actin assembly pathway. In the absence of N-WASP, the Arp2/3 complex was both recruited to sites of bacterial attachment and required for actin assembly. Our results indicate that actin assembly facilitates type III translocation, and reveal that EspFU, presumably by recruiting an alternate host factor that can signal to the Arp2/3 complex, exhibits remarkable versatility in its strategies for stimulating actin polymerization. The food-borne pathogen enterohemorrhagic E. coli (EHEC) O157:H7 can cause severe diarrhoea and life-threatening systemic illnesses. During infection, EHEC attaches to cells lining the human intestine and injects Tir and EspFU, two bacterial molecules that alter the host cell actin cytoskeleton and stimulate the formation of “pedestals” just beneath bound bacteria. Pedestal formation promotes colonization during the later stages of infection. N-WASP, a host protein known to regulate actin assembly in mammalian cells, was previously shown to be manipulated by Tir and EspFU to stimulate actin assembly, and to be required for EHEC to generate actin pedestals. Surprisingly, we show here that N-WASP promotes the efficient delivery of Tir and EspFU into mammalian cells, and that when we utilized a related E. coli to enhance type III delivery of Tir and EspFU, actin pedestals assembled even in its absence. Thus, EHEC stimulates at least two pathways of actin assembly to generate pedestals, one mediated by N-WASP and one by an unidentified alternate factor. This flexibility likely reflects an important function of pedestal formation by EHEC, and study of the underlying mechanisms may provide new insights into the pathogenesis of infection as well as the regulation of the actin cytoskeleton of mammalian cells.
Collapse
Affiliation(s)
- Didier Vingadassalom
- Department of Molecular Genetics and Microbiology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Kenneth G. Campellone
- Department of Molecular Genetics and Microbiology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Michael J. Brady
- Department of Molecular Genetics and Microbiology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Brian Skehan
- Department of Molecular Genetics and Microbiology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Scott E. Battle
- Section of Digestive Diseases and Nutrition, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Douglas Robbins
- Department of Molecular Genetics and Microbiology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Archana Kapoor
- Department of Medicine and Immunology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Gail Hecht
- Section of Digestive Diseases and Nutrition, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Scott B. Snapper
- Department of Medicine and Immunology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - John M. Leong
- Department of Molecular Genetics and Microbiology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|