12 research outputs found

    Innate Immune Recognition of Yersinia pseudotuberculosis Type III Secretion

    Get PDF
    Specialized protein translocation systems are used by many bacterial pathogens to deliver effector proteins into host cells that interfere with normal cellular functions. How the host immune system recognizes and responds to this intrusive event is not understood. To address these questions, we determined the mammalian cellular response to the virulence-associated type III secretion system (T3SS) of the human pathogen Yersinia pseudotuberculosis. We found that macrophages devoid of Toll-like receptor (TLR) signaling regulate expression of 266 genes following recognition of the Y. pseudotuberculosis T3SS. This analysis revealed two temporally distinct responses that could be separated into activation of NFκB- and type I IFN-regulated genes. Extracellular bacteria were capable of triggering these signaling events, as inhibition of bacterial uptake had no effect on the ensuing innate immune response. The cytosolic peptidoglycan sensors Nod1 and Nod2 and the inflammasome component caspase-1 were not involved in NFκB activation following recognition of the Y. pseudotuberculosis T3SS. However, caspase-1 was required for secretion of the inflammatory cytokine IL-1β in response to T3SS-positive Y. pseudotuberculosis. In order to characterize the bacterial requirements for induction of this novel TLR-, Nod1/2-, and caspase-1-independent response, we used Y. pseudotuberculosis strains lacking specific components of the T3SS. Formation of a functional T3SS pore was required, as bacteria expressing a secretion needle, but lacking the pore-forming proteins YopB or YopD, did not trigger these signaling events. However, nonspecific membrane disruption could not recapitulate the NFκB signaling triggered by Y. pseudotuberculosis expressing a functional T3SS pore. Although host cell recognition of the T3SS did not require known translocated substrates, the ensuing response could be modulated by effectors such as YopJ and YopT, as YopT amplified the response, while YopJ dampened it. Collectively, these data suggest that combined recognition of the T3SS pore and YopBD-mediated delivery of immune activating ligands into the host cytosol informs the host cell of pathogenic challenge. This leads to a unique, multifactorial response distinct from the canonical immune response to a bacterium lacking a T3SS

    Induction of Interferon-Stimulated Genes by Chlamydia pneumoniae in Fibroblasts Is Mediated by Intracellular Nucleotide-Sensing Receptors

    Get PDF
    BACKGROUND: Recognition of microorganisms by the innate immune system is mediated by pattern recognition receptors, including Toll-like receptors and cytoplasmic RIG-I-like receptors. Chlamydia, which include several human pathogenic species, are obligate intracellular gram-negative bacteria that replicate in cytoplasmic vacuoles. The infection triggers a host response contributing to both bacterial clearance and tissue damage. For instance, type I interferons (IFN)s have been demonstrated to exacerbate the course of Chlamydial lung infections in mice. METHODS/PRINCIPAL FINDINGS: Here we show that Chlamydia pneumoniae induces expression of IFN-stimulated genes (ISG)s dependent on recognition by nucleotide-sensing Toll-like receptors and RIG-I-like receptors, localized in endosomes and the cytoplasm, respectively. The ISG response was induced with a delayed kinetics, compared to virus infections, and was dependent on bacterial replication and the bacterial type III secretion system (T3SS). CONCLUSIONS/SIGNIFICANCE: Activation of the IFN response during C. pneumoniae infection is mediated by intracellular nucleotide-sensing PRRs, which operate through a mechanism dependent on the bacterial T3SS. Strategies to inhibit the chlamydial T3SS may be used to limit the detrimental effects of the type I IFN system in the host response to Chlamydia infection

    Type three secretion system-mediated escape of Burkholderia pseudomallei into the host cytosol is critical for the activation of NFκB.

    Get PDF
    BackgroundBurkholderia pseudomallei is the causative agent of melioidosis, a potentially fatal disease endemic in Southeast Asia and Northern Australia. This Gram-negative pathogen possesses numerous virulence factors including three "injection type" type three secretion systems (T3SSs). B. pseudomallei has been shown to activate NFκB in HEK293T cells in a Toll-like receptor and MyD88 independent manner that requires T3SS gene cluster 3 (T3SS3 or T3SSBsa). However, the mechanism of how T3SS3 contributes to NFκB activation is unknown.ResultsKnown T3SS3 effectors are not responsible for NFκB activation. Furthermore, T3SS3-null mutants are able to activate NFκB almost to the same extent as wildtype bacteria at late time points of infection, corresponding to delayed escape into the cytosol. NFκB activation also occurs when bacteria are delivered directly into the cytosol by photothermal nanoblade injection.ConclusionsT3SS3 does not directly activate NFκB but facilitates bacterial escape into the cytosol where the host is able to sense the presence of the pathogen through cytosolic sensors leading to NFκB activation

    Injection of Pseudomonas aeruginosa Exo Toxins into Host Cells Can Be Modulated by Host Factors at the Level of Translocon Assembly and/or Activity

    Get PDF
    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

    Activation and Inhibition of Multiple Inflammasome Pathways by the Yersinia Pestis Type Three Secretion System: A Dissertation

    Get PDF
    Host survival during plague, caused by the Gram-negative bacterium Yersinia pestis, is favored by a robust early innate immune response initiated by IL-1β and IL-18. Precursors of these cytokines are expressed downstream of TLR signaling and are then enzymatically processed into mature bioactive forms, typically by caspase-1 which is activated through a process dependent on multi-molecular structures called inflammasomes. Y. pestis evades immune detection in part by using a Type three secretion system (T3SS) to inject effector proteins (Yops) into host cells and suppress IL-1β and IL-18 production. We investigated the cooperation between two effectors, YopM and YopJ, in regulating inflammasome activation, and found that Y. pestis lacking both YopM and YopJ triggers robust caspase-1 activation and IL-1Β/IL-18 production in vitro. Furthermore, this strain is attenuated in a manner dependent upon caspase-1, IL-1β and IL-18 in vivo, yet neither effector appears essential for full virulence. We then demonstrate that YopM fails to inhibit NLRP3/NLRC4 mediated caspase-1 activation and is not a general caspase-1 inhibitor. Instead, YopM specifically prevents the activation of a Pyrin-dependent inflammasome by the Rho-GTPase inhibiting effector YopE. Mutations rendering Pyrin hyperactive are implicated in the autoinflammatory disease Familial Mediterranean Fever (FMF) in humans, and we discuss the potential significance of this disease in relation to plague. Altogether, the Y. pestis T3SS activates and inhibits several inflammasome pathways, and the fact that so many T3SS components are involved in manipulating IL-1β/IL-18 underscores the importance of these mechanisms in plague

    MODULATION OF HOST NF¿B SIGNALING PATHWAYS BY BURKHOLDERIA PSEUDOMALLEI

    Get PDF
    Ph.DDOCTOR OF PHILOSOPH

    Examination of the Antibacterial and Immunostimulatory Activity of a Wasp Venom Peptide

    Get PDF
    <p>Antimicrobial peptides (AMPs) are part of the innate immune system that is widely distributed in nature, acting as a defense mechanism against invading microorganisms. AMPs have potent antimicrobial activity against a range of microorganisms including fungi, bacteria and viruses. In view of growing multidrug resistance, AMPs are increasingly being viewed as potential therapeutic agents with a novel mechanism of action. Mastoparan is a natural, highly positively charged AMP derived from the venom of wasps. It was originally of interest based on its inherent mast cell degranulation activity. Previously, mastoparan has been shown to exhibit antimicrobial activity in vitro however these studies have been limited in scope. Here we hypothesize that mastoparan possess the capacity to be a potent broad spectrum antibacterial agent including activity against multidrug resistant bacteria. </p><p>We examined the scope of antibacterial activity exhibited by mastoparan using a variety of antimicrobial susceptibility tests and have utilized a bacterial skin infection (S. aureus) model to determine the potential of mastoparan to serve as a therapeutic agent. We tested mastoparan against 4 Gram-positive clinical isolates (e.g., S. aureus, and E. faecium), 9 Gram-negative clinical isolates (e.g., E. coli, P. aeruginosa, and B. cepacia), and 4 multidrug resistant clinical isolates (e.g., MRSA, ESBL E.coli, and ESBL K. pneumonia). These studies reveal that mastoparan exhibits broad spectrum activity against both Gram-negative (MIC: 1.9 - 125 &mug/ml) and Gram-positive (MIC: 15.6 - 125 &mug/ml) bacteria and against multidrug resistant bacteria (MIC: 7.8 - 125 &mug/ml). We also demonstrated that mastoparan disrupts the bacterial membrane, exhibits fast acting antibacterial activity and is highly effective against both multiplying and non-multiplying bacteria. Furthermore, we have shown that mastoparan demonstrates efficacy as a topical antimicrobial agent reducing lesion size by up to 79% and the amount of bacteria recovered from skin lesions by up to a 98% reduction. Based on these results we conclude that mastoparan is a highly effective antibacterial agent and is therefore a potential alternative to currently antibiotics. Mastoparan offers a promising new therapeutic option for treating bacterial infections.</p>Dissertatio

    Investigating Inflammasome Activation in Response to Legionella Pneumophila and its Application to Other Bacterial Pathogens

    Get PDF
    The mucosal surfaces of metazoan organisms provide niches for colonization by commensal microbes. However, these barrier surfaces also encounter pathogens. Therefore, sentinel immune cells must be capable of distinguishing between pathogenic and non-pathogenic organisms to tailor appropriate immune responses. Virulent microorganisms often uniquely possess mechanisms for accessing the host cell cytosol. Therefore, to detect pathogens, innate immune cells encode cytosolic receptors, which recognize conserved, pathogen-associated molecular patterns. Many mammalian cytosolic receptors activate the inflammasome, a multi-protein complex that activates the host enzyme caspase-1. Caspase-1 mediates IL-1 family cytokine release and a pro-inflammatory form of cell death, which are important for host defense. The canonical inflammasome activates caspase-1, but recent studies have shown that a related enzyme, caspase-11, contributes to inflammasome activation. However, it remains unclear if caspase-11 mediates inflammasome responses against bacteria that use virulence-associated secretion systems to deliver bacterial products into the host cytosol. Additionally, humans encode two orthologs of caspase-11, caspase-4 and caspase-5, and it is unclear if either enzyme contributes to inflammasome activation in primary macrophages. Furthermore, the bacterial ligands that trigger inflammasome activation in human cells are poorly defined. Legionella pneumophila, which causes pneumonia, uses a specialized secretion system to access the host cytosol to establish a replicative niche in both murine and human cells. Therefore, we investigated the host and bacterial requirements for inflammasome activation in response to L. pneumophila, and we interrogated if these requirements are conserved for the response against other Gram-negative bacterial pathogens. Our studies demonstrate that caspase-11 contributes to IL-1 release and cell death in response to bacterial pathogens in murine macrophages, and we find that inflammasome activation requires the presence of virulence-associated secretion systems. Using neutralizing antibodies, we show that IL-1α and IL-1β have distinct roles in pulmonary defense against L. pneumophila in vivo. Through siRNA knockdown studies, we demonstrate that human caspase-4 has a conserved role in inflammasome activation in response to multiple Gram-negative bacterial pathogens. Finally, using bacterial mutants, we show that flagellin is a trigger for inflammasome activation in human macrophages. Overall, our studies help define the mechanism by which host cells initiate defense against bacterial pathogens

    THE \u3ci\u3ePSEUDOMONAS SYRINGAE\u3c/i\u3e TYPE III SECRETION SYSTEM: THE TRANSLOCATOR PROTEINS, THEIR SECRETION, AND THE RESTRICTION OF TRANSLOCATION BY THE PLANT IMMUNE SYSTEM

    Get PDF
    Pseudomonas syringae is a Gram-negative plant pathogen whose virulence is dependent upon its type III secretion system (T3SS), a nanosyringe that facilitates translocation, or injection, of type III effector (T3E) proteins into eukaryotic cells. The primary function of P. syringae T3E proteins is suppression of plant immunity. Bacterial proteins called translocators form a translocon that forms a pore in the host plasma membrane which is traversed by T3Es. HrpK1, a putative P. syringae translocator, is a type III-secreted protein important for virulence and T3E injection, but not secretion of T3Es. Harpins are a group of proteins specific to plant pathogens that are also important for T3E translocation. P. syringae pv. tomato DC3000 has 4 harpins – HrpZ1, HrpW1, HopAK1, and HopP1. Here, HrpK1 is confirmed to be a translocator. HrpK1 had a greater impact on T3E translocation than the harpins. HrpK1 and HrpZ1 disrupted liposomes. Both proteins interacted with phosphatidic acid which interfered with T3E translocation. HrpJ, a type III-secreted protein required for HrpZ1 secretion, was also required for secretion of HrpK1, HrpW1, and HopAK1. A hrpJ mutant secreted elevated levels of the Hrp pilus protein HrpA1. HrpJ appears to control transition from Hrp pilus secretion to translocator secretion. Secretion was complemented by secretion incompetent HrpJ derivatives indicating that HrpJ controls secretion from inside the bacteria. The hrpJ mutant expressing secretion incompetent HrpJ was reduced in virulence but was complemented by HrpJ expressed inside plant cells. Additionally, transgenic Arabidopsis plants expressing HrpJ were reduced in their immune responses indicating that HrpJ can suppress plant immunity. Plants pretreated with an inducer of pathogen-associated molecular pattern-triggered immunity are unable to produce an HR. Plants, as an immune response, have evolved the ability to block T3E translocation when plant immunity has been induced prior to bacterial inoculation. This is especially true in non-host interactions whereas virulent bacteria appear to be able to attenuate injection restriction in host plants via T3E activity. Adviser: James R. Alfan
    corecore