Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Front Cell Infect Microbiol ; 12: 811407, 2022.
Article in English | MEDLINE | ID: mdl-35300377

ABSTRACT

The intracellular pathogen Chlamydia trachomatis secretes multiple early effectors into the host cell to promote invasion. A key early effector during host cell entry, Tarp (translocated actin-recruiting phosphoprotein) is comprised of multiple protein domains known to have roles in cell signaling, G-actin nucleation and F-actin bundle formation. In vitro, the actin bundles generated by Tarp are uncharacteristically flexible, however, in vivo, the biological significance of Tarp-mediated actin bundles remains unknown. We hypothesize that Tarp's ability to generate unique actin bundles, in part, facilitates chlamydial entry into epithelial cells. To study the in vivo interaction between Tarp and F-actin, we transgenically expressed Tarp in Drosophila melanogaster tissues. Tarp expressed in Drosophila is phosphorylated and forms F-actin-enriched aggregates in tissues. To gain insight into the significance of Tarp actin bundles in vivo, we utilized the well-characterized model system of mechanosensory bristle development in Drosophila melanogaster. Tarp expression in wild type flies produced curved bristles, indicating a perturbation in F-actin dynamics during bristle development. Two F-actin bundlers, Singed/Fascin and Forked/Espin, are important for normal bristle shape. Surprisingly, Tarp expression in the bristles displaced Singed/Fascin away from F-actin bundles. Tarp's competitive behavior against Fascin during F-actin bundling was confirmed in vitro. Loss of either singed or forked in flies leads to highly deformed bristles. Strikingly, Tarp partially rescued the loss of singed, reducing the severity of the bristle morphology defect. This work provides in vivo confirmation of Tarp's F-actin bundling activity and further uncovers a competitive behavior against the host bundler Singed/Fascin during bundle assembly. Also, we demonstrate the utility of Drosophila melanogaster as an in vivo cell biological platform to study bacterial effector function.


Subject(s)
Actins , Bacterial Proteins , Chlamydia trachomatis , Microfilament Proteins , Actin Cytoskeleton/metabolism , Actins/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Carrier Proteins , Chlamydia trachomatis/genetics , Chlamydia trachomatis/pathogenicity , Drosophila melanogaster , Microfilament Proteins/metabolism
2.
Biochem Biophys Res Commun ; 562: 133-138, 2021 07 12.
Article in English | MEDLINE | ID: mdl-34052658

ABSTRACT

Chlamydia trachomatis injects bacterial effector proteins into human epithelial cells to facilitate the establishment of new infections. The chlamydial type III secreted effector translocated actin recruiting phosphoprotein (Tarp) has been shown to nucleate and bundle actin filaments. It is also believed to initiate new signaling pathways via an N-terminal phosphorylation domain. A comprehensive understanding of the host pathways that are controlled by Tarp to aid in the establishment of a successful infection remains incomplete. To gain further insight into the cell signaling regulated by Tarp, we generated transgenic fruit flies engineered to express the N-terminal domain of Tarp. As many signaling pathways are conserved between flies and mammals, we hypothesized that expression of the Tarp N-domain in the fruit fly might disrupt key pathways, resulting in developmental defects. Tarp N-domain expression in the fruit fly resulted in a mechanosensory bristle duplication phenotype similar to a previously characterized fly phenotype found to be a consequence of defects in the Hippo pathway. Tarp-dependent disruption of the Hippo pathway was confirmed in a C. trachomatis tissue culture infection model. The capability of Tarp to alter Hippo pathway signaling in infected epithelial cells is a previously unrecognized pathway commandeered by chlamydia and likely contributes to the establishment of chlamydia's intracellular niche.


Subject(s)
Bacterial Proteins/metabolism , Chlamydia trachomatis/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Animals , Animals, Genetically Modified , Bacterial Proteins/chemistry , DNA-Binding Proteins/metabolism , Drosophila melanogaster/genetics , Gene Expression Regulation , HeLa Cells , Hippo Signaling Pathway , Humans , Mechanotransduction, Cellular , Muscle Proteins/metabolism , Protein Domains , RNA, Messenger/genetics , RNA, Messenger/metabolism , TEA Domain Transcription Factors , Transcription Factors/metabolism , X-Linked Inhibitor of Apoptosis Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...