@phdthesis{Kuehlkamp2001, author = {K{\"u}hlkamp, Thomas}, title = {Der plasmamembran assoziierte Transportregulator RS1 bindet Ubiquitin und gelangt in den Zellkern}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-1179507}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2001}, abstract = {Die vorliegende Arbeit liefert wichtige Erkenntnisse {\"u}ber die subzellul{\"a}re Verteilung und die Funktion des RS1-Proteins vom Schwein (pRS1), einem Regulator von Plasmamembran-transportern. Das gr{\"u}n fluoreszierende Protein (GFP) wurde mit pRS1 fusioniert und in LLC-PK1 Zellen exprimiert. Das GFP-pRS1 Fusionsprodukt (96 kD) konnte an der Plasmamembran, im Zytosol und im Zellkern entdeckt werden. Bei GFP-Fusion mit trunkierten pRS1-Proteinen zeigte sich, dass der C-Terminus die Kernlokalisierung beeinflusst. Dagegen wurde die Kernlokalisierung durch eine Trunkierung des N-Terminus nicht gest{\"o}rt. Im C-Terminus des pRS1 konnte von AS 579 bis 616 eine Ubiquitin associated domain (UBA) identifiziert werden, die auch in den anderen bisher bekannten RS1-Proteinen aus Mensch, Kaninchen und Maus konserviert vorliegt. Eine Ubiquitin-Affinit{\"a}tschromatographie zeigte, dass das pRS1-Protein Ubiquitin auf nicht kovalente Weise bindet. Nach der Trunkierung der UBA-Dom{\"a}ne war keine Wechselwirkung des pRS1-Proteins mit Ubiquitin mehr feststellbar. Ein konserviertes Di-Leucin-Endozytose-Motiv (pRS1 AS 366/67) deutet eine Funktion des pRS1-Proteins bei der Internalisierung von Plasmamembranproteinen an. Deshalb wurde das Endozytoseverhalten von pRS1 {\"u}berexprimierenden LLC-PK1 Zellen untersucht, wobei sich zeigte, dass diese Zellen eine deutlich h{\"o}here Aufnahme des Endozytosefarbstoffes RH 414 aufwiesen als Zellen, die pRS1 nicht {\"u}berexprimierten. Die in dieser Arbeit gesammelten Daten zum RS1-Protein wurden zusammen mit fr{\"u}her erhobenen Ergebnissen zum RS1-Protein im Rahmen eines Modells zusammengefasst. In diesem hypothetischen Modell wird angenommen, dass RS1 ein Adapterprotein ist, welches die ubiquitinabh{\"a}ngige Endozytose von Plasmamembrantransportern vermittelt und als Signalmolek{\"u}l in den Zellkern gelangen kann, wo es an der Transcriptionsrepression des SGLT1 beteiligt ist.}, subject = {Ubiquitin}, language = {de} } @phdthesis{Hartung2006, author = {Hartung, Anke}, title = {Localization of BMP receptors in distinct plasma membrane domains and its impact on BMP signaling}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-18360}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2006}, abstract = {Endocytosis of growth factor receptors plays an important role in the activation and propagation as well as the attenuation of signaling pathways. Its malfunctioning can cause several pathologies, e.g. by controlling the level of receptors at the cell surface. BMPs are members of the TGF-ß superfamily and are involved in the regulation of proliferation, differentiation, chemotaxis and apoptosis. BMP signaling is initiated at two types of transmembrane serine/threonine kinases, BRI and BRII. BMP receptor activation occurs upon ligand binding to preformed complexes (PFCs) or BMP2-induced signaling complexes (BISCs) composed of BRI and BRII. Binding of BMP2 to PFCs results in activation of the Smad pathway, whereas BISCs initiate the activation of Smad-independent pathways via p38 resulting in the induction of Alkaline phosphatase (ALP). BMP receptor endocytosis has not been extensively studied and the potential role of localization to different regions of the plasma membrane in determining the signaling pathways activated by PFCs and BISCs was not explored so far. In the present work, the localization of BMP receptors in distinct membrane domains and the consequential impact on BMP signaling were investigated. By separating detergent-resistant membranes (DRMs) from cell lysates and subsequent gradient ultracentrifugation, it could be demonstrated that BRI and BRII cofractionate with cav-1, the marker protein of caveolae. Moreover, both receptor types interacted with cav-1 and showed a partially colocalization with cav-1 at the plasma membrane. Although these results point to a caveolar localization, BMP receptors cofractionated also with DRMs in cells exhibiting no caveolae, suggesting an additional non-caveolar raft localization. Beyond that, BRII could also be localized to clathrin-coated pits (CCPs) by means of immuno-electronmicroscopy studies. The second part of this thesis demonstrated that both membrane regions influence BMP signaling in distinct ways. Smad1/5 was shown to be phosphorylated independently of endocytic events at the cell surface. On the one hand, disruption of DRM regions by cholesterol depletion inhibited specifically BMP2-mediated ALP production, while Smad signaling was unaffected. On the other hand, inhibition of clathrin-mediated endocytosis by specific inhibitors affected BMP2-induced Smad signaling as well as the induction of ALP, suggesting that both Smad-dependent and Smad-independent signaling pathways are required for BMP2 induced ALP production. These findings propose an important regulatory impact of different endocytic routes and membrane regions on BMP signaling as well as that a distinct membrane localization of BMP receptors account for specific signaling properties initiated at PFCs or BISCs.}, subject = {Knochen-Morphogenese-Proteine}, language = {en} } @phdthesis{Subbarayal2015, author = {Subbarayal, Prema}, title = {The role of human Ephrin receptor tyrosine kinase A2 (EphA2) in Chlamydia trachomatis infection}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-114778}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2015}, abstract = {Chlamydia trachomatis (Ctr), an obligate intracellular gram negative human pathogen, causes sexually transmitted diseases and acquired blindness in developing countries. The infectious elementary bodies (EB) of Ctr involved in adherence and invasion processes are critical for chlamydial infectivity and subsequent pathogenesis which requires cooperative interaction of several host cell factors. Few receptors have been known for this early event, yet the molecular mechanism of these receptors involvement throughout Ctr infection is not known. Chlamydial inclusion membrane serves as a signaling platform that coordinates Chlamydia-host cell interaction which encouraged me to look for host cell factors that associates with the inclusion membrane, using proteome analysis. The role of these factors in chlamydial replication was analyzed by RNA interference (RNAi) (in collaboration with AG Thomas Meyer). Interestingly, EphrinA2 receptor (EphA2), a cell surface tyrosine kinase receptor, implicated in many cancers, was identified as one of the potential candidates. Due to the presence of EphA2 in the Ctr inclusion proteome data, I investigated the role of EphA2 in Ctr infection. EphA2 was identified as a direct interacting receptor for adherence and entry of C. trachomatis. Pre-incubation of Ctr-EB with recombinant human EphA2, knockdown of EphA2 by siRNA, pretreatment of cells with anti-EphA2 antibodies or the tyrosine kinase inhibitor dasatinib significantly reduced Ctr infection. This marked reduction of Ctr infection was seen with both epithelial and endothelial cells used in this study. Ctr activates EphA2 upon infection and invades the cell together with the activated EphA2 receptor that interacts and activates PI3K survival signal, promoting chlamydial replication. EphA2 upregulation during infection is associated with Ctr inclusion membrane inside the cell and are prevented being translocated to the cell surface. Ephrins are natural ligands for Ephrin receptors that repress the activation of the PI3K/Akt pathway in a process called reverse signaling. Purified Ephrin-A1, a ligand of EphA2, strongly interferes with chlamydial infection and normal development, supporting the central role of these receptors in Chlamydia infection. Overexpression of full length EphA2, but not the mutant form lacking the intracellular cytoplasmic domain, enhanced PI3K activation and Ctr infection. Ctr infection induces EphA2 upregulation and is mediated by activation of ERK signaling pathway. Interfering with EphA2 upregulation sensitizes Ctr-infected cells to apoptosis induced by tumor necrosis factor-alpha (TNF-α) suggesting the importance of intracellular EphA2 signaling. Collectively, these results revealed the first Ephrin receptor "EphA2" that functions in promoting chlamydial infection. In addition, the engagement of a cell surface receptor at the inclusion membrane is a new mechanism how Chlamydia subverts the host cell and induces apoptosis resistance. By applying the natural ligand Ephrin-A1 and targeting EphA2 offers a promising new approach to interfere with Chlamydia infection. Thus, the work provides the evidence for a host cell surface tyrosine kinase receptor that is exploited for invasion as well as for receptor-mediated intracellular signaling to facilitate the chlamydial replication.}, subject = {Chlamydia trachomatis}, language = {en} }