12 research outputs found

    Listeria monocytogenes Internalin B Activates Junctional Endocytosis to Accelerate Intestinal Invasion

    Get PDF
    Listeria monocytogenes (Lm) uses InlA to invade the tips of the intestinal villi, a location at which cell extrusion generates a transient defect in epithelial polarity that exposes the receptor for InlA, E-cadherin, on the cell surface. As the dying cell is removed from the epithelium, the surrounding cells reorganize to form a multicellular junction (MCJ) that Lm exploits to find its basolateral receptor and invade. By examining individual infected villi using 3D-confocal imaging, we uncovered a novel role for the second major invasin, InlB, during invasion of the intestine. We infected mice intragastrically with isogenic strains of Lm that express or lack InlB and that have a modified InlA capable of binding murine E-cadherin and found that Lm lacking InlB invade the same number of villi but have decreased numbers of bacteria within each infected villus tip. We studied the mechanism of InlB action at the MCJs of polarized MDCK monolayers and find that InlB does not act as an adhesin, but instead accelerates bacterial internalization after attachment. InlB locally activates its receptor, c-Met, and increases endocytosis of junctional components, including E-cadherin. We show that MCJs are naturally more endocytic than other sites of the apical membrane, that endocytosis and Lm invasion of MCJs depends on functional dynamin, and that c-Met activation by soluble InlB or hepatocyte growth factor (HGF) increases MCJ endocytosis. Also, in vivo, InlB applied through the intestinal lumen increases endocytosis at the villus tips. Our findings demonstrate a two-step mechanism of synergy between Lm's invasins: InlA provides the specificity of Lm adhesion to MCJs at the villus tips and InlB locally activates c-Met to accelerate junctional endocytosis and bacterial invasion of the intestine

    The role of the SD100A domain in IFNAR1 in type I interferon signaling

    No full text
    grantor: University of TorontoThe Type I interferon (IFN) system plays a prevalent role in regulating antiviral and growth inhibitory activities and in modulating the immune system. Study of the signaling pathways associated with these features of the IFN system has led to the observation that IFNs act in a species specific manner and that Type I IFNs are able to signal differentially using the same receptor complex. Furthermore, IFN subtypes require different extracellular receptor domains to elicit signaling. To assess the contribution of the SD100A domain of the IFNAR1 chain to Type I IFN binding and signaling, a chimaeric cDNA construct in which the SD100A domain of the murine chain was replaced with the human equivalent was constructed. This HMM/M IFNAR1 was introduced by transfection into IFNAR1 deficient mouse embryonic fibroblasts and stable transfectants were established for study. Protein extracts from murine IFN-Ã4 treated HMM/M expressing clones and wild-type IFNAR1 (MMM/M) expressing clones show similar Stat activation. Antiviral and antiproliferative responses were also comparable in the MMM/M and HMM/M clones. These results suggest that the IFNAR1 SD100A domain does not contribute to Type I IFN binding and signaling in a species specific manner in the context of murine IFN-Ã4.M.Sc

    In situ proteolysis, crystallization and preliminary X-ray diffraction analysis of a VHH that binds listeria internalin B

    No full text
    The variable region of camelid heavy-chain antibodies produces the smallest known antibody fragment with antigen-binding capability (a V<inf>H</inf>H). The V<inf>H</inf>H R303 binds internalin B (InlB), a virulence factor expressed by the pathogen Listeria monocytogenes. InlB is critical for initiation of Listeria infection, as it binds a receptor (c-Met) on epithelial cells, triggering the entry of bacteria into host cells. InlB is surface-exposed and is required for virulence, hence a V<inf>H</inf>H targeting InlB has potential applications for pathogen detection or therapeutic intervention. Here, the expression, purification, crystallization and X-ray diffraction of R303 are reported. Crystals of R303 were obtained following in situ proteolysis with trypsin. Gel filtration and SDS-PAGE revealed that trypsin removed the C-terminal tag region of R303, facilitating crystal formation. Crystals of R303 diffracted to 1.3\uc5 resolution and belonged to the monoclinic space group P21, with unit-cell parameters a = 46.4, b = 31.2, c = 74.8\uc5, \u3b2 = 93.8\ub0. The crystals exhibited a Matthews coefficient of 1.95\uc53Da-1 with two molecules in the asymmetric unit.Peer reviewed: YesNRC publication: Ye

    Characterization of high affinity anti-Internalin B VHH antibody fragments isolated from naturally and artificially immunized repertoires

    No full text
    The need for rapid and easy technologies for the detection of food-borne and environmental pathogens is essential for safeguarding the health of populations. Furthermore, distribution of tainted food and water can have consequences which can affect whole economies. Antibodies and antibody fragments have been historically used in detection platforms due to their antigen specificity and robust physicochemical properties. In this study, we report the isolation and characterization of antibody fragments from the heavy chain antibody repertoire (VHH) of Camelidae which bind with specificity and high affinity to the Listeria monocytogenes invasin, Internalin B (InlB). To the best of our knowledge, this is the first report of anti-InlB VHHs from camelids. These anti-InlB VHHs were not cross-reactive to the structurally related Listeria invasin Internalin A (InlA) and are potential reagents to be used in the development of detection and medical technologies.Peer reviewed: YesNRC publication: Ye
    corecore