A Translocated Effector Required for Bartonella Dissemination from Derma to Blood Safeguards Migratory Host Cells from Damage by Co-translocated Effectors

Numerous bacterial pathogens secrete multiple effectors to modulate host cellular functions. These effectors may interfere with each other to efficiently control the infection process. Bartonellae are Gram-negative, facultative intracellular bacteria using a VirB type IV secretion system to transloc...

Full description

Bibliographic Details
Main Authors: Okujava, Rusudan (Author), Guye, Patrick (Contributor), Lu, Yun-Yueh (Author), Mistl, Claudia (Author), Polus, Florine (Author), Vayssier-Taussat, Muriel (Author), Halin, Cornelia (Author), Rolink, Antonius G. (Author), Dehio, Christoph (Author)
Other Authors: Massachusetts Institute of Technology. Department of Biological Engineering (Contributor)
Format: Article
Language:English
Published: Public Library of Science, 2014-09-12T15:28:46Z.
Subjects:
Online Access:Get fulltext
Description
Summary:Numerous bacterial pathogens secrete multiple effectors to modulate host cellular functions. These effectors may interfere with each other to efficiently control the infection process. Bartonellae are Gram-negative, facultative intracellular bacteria using a VirB type IV secretion system to translocate a cocktail of Bartonella effector proteins (Beps) into host cells. Based on in vitro infection models we demonstrate here that BepE protects infected migratory cells from injurious effects triggered by BepC and is required for in vivo dissemination of bacteria from the dermal site of inoculation to blood. Human endothelial cells (HUVECs) infected with a ΔbepE mutant of B. henselae (Bhe) displayed a cell fragmentation phenotype resulting from Bep-dependent disturbance of rear edge detachment during migration. A ΔbepCE mutant did not show cell fragmentation, indicating that BepC is critical for triggering this deleterious phenotype. Complementation of ΔbepE with BepE[subscript Bhe] or its homologues from other Bartonella species abolished cell fragmentation. This cyto-protective activity is confined to the C-terminal Bartonella intracellular delivery (BID) domain of BepE[subscript Bhe] (BID2.E[subscript Bhe]). Ectopic expression of BID2.E[subscript Bhe] impeded the disruption of actin stress fibers by Rho Inhibitor 1, indicating that BepE restores normal cell migration via the RhoA signaling pathway, a major regulator of rear edge retraction. An intradermal (i.d.) model for B. tribocorum (Btr) infection in the rat reservoir host mimicking the natural route of infection by blood sucking arthropods allowed demonstrating a vital role for BepE in bacterial dissemination from derma to blood. While the Btr mutant ΔbepDE was abacteremic following i.d. inoculation, complementation with BepE[subscript Btr], BepE[subscript Bhe] or BIDs.E[subscript Bhe] restored bacteremia. Given that we observed a similar protective effect of BepE[subscript Bhe] on infected bone marrow-derived dendritic cells migrating through a monolayer of lymphatic endothelial cells we propose that infected dermal dendritic cells may be involved in disseminating Bartonella towards the blood stream in a BepE-dependent manner.