@phdthesis{Pfeuffer2000, author = {Pfeuffer, Thilo}, title = {Interaktion des Proteins ActA von Listeria monocytogenes mit dem Wirtszellprotein LaXp180}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-1859}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2000}, abstract = {Listeria monocytogenes, ein fakultativ intrazellul{\"a}rer Krankheitserreger, besitzt die F{\"a}higkeit, Wirtszellen zu penetrieren, sich in ihnen zu vermehren, sich intrazellul{\"a}r zu bewegen und auch benachbarte Zellen direkt zu infizieren. Die intrazellul{\"a}re Fortbewegung erfolgt durch Polymerisation von zellul{\"a}rem Aktin, wodurch charakteristische Aktinschweife an einem Pol der Bakterien entstehen. Der einzige bakterielle Faktor, der f{\"u}r die Aktinpolymerisation notwendig ist, ist das Oberfl{\"a}chenprotein ActA. ActA allein ist aber nicht in der Lage, Aktin zu polymerisieren, sondern kann dies nur in Assoziation mit Proteinen der Wirtszelle. Die einzigen bisher bekannten Wirtszellproteine, die direkt mit ActA interagieren, sind das Phosphoprotein VASP und der Arp2/3-Komplex. VASP bindet an den zentralen prolinreichen Bereich von ActA und beschleunigt durch die Rekrutierung von Profilin den Prozeß der Aktinpolymerisation. Der Arp2/3-Komplex interagiert mit dem N-terminalen Bereich von ActA und initiiert die eigentliche Aktin-Polymerisation. Um weitere eukaryotische, mit ActA interagierende Proteine (AIPs) zu isolieren, wurde {\"u}ber einen "Yeast Two-Hybrid"-Test mit ActA als K{\"o}der eine embryonale Maus-cDNA-Genbank getestet. Dabei wurden drei verschiedene AIPs identifiziert, von denen eines identisch mit dem humanen Protein LaXp180 (auch "CC1" genannt) ist. LaXp180 ist ein 180 kDa Protein mit {\"u}ber 50 theoretischen Phosphorylierungsstellen in der N-terminalen H{\"a}lfte, w{\"a}hrend die C-terminale H{\"a}lfte "coiled-coil"-Strukturen ausbilden kann. Dar{\"u}berhinaus enth{\"a}lt LaXp180 eine Kern-Lokalisations-Sequenz und ein Leucin-Zipper-Motiv. Die Bindung von LaXp180 an ActA wurde in vitro unter Verwendung von rekombinantem His6-Tag-LaXp180 und rekombinantem ActA best{\"a}tigt, da rekombinantes ActA nur an einer Ni-Agarose-S{\"a}ule gebunden wurde, wenn diese vorher mit His6-Tag-LaXp180 beladen war. {\"U}ber RT-PCR konnte zum ersten Mal die Expression LaXp180-spezifischer mRNA in verschiedenen S{\"a}ugerzellen nachgewiesen und mit einem polyklonalen anti-LaXp180-Serum durch Immunopr{\"a}zipitation erstmals ein 194 kDa großes Protein in S{\"a}ugerzellextrakten detektiert werden. Die intrazellul{\"a}re Lokalisation von LaXp180 wurde {\"u}ber Immunfluoreszenzmikroskopie untersucht. Immunfluoreszenzf{\"a}rbungen von Fibroblasten mit dem anti-LaXp180-Serum zeigten eine starke F{\"a}rbung der Zellkerne und definierter Bereiche direkt neben den Kernen, w{\"a}hrend das restliche Zytoplasma schwach gef{\"a}rbt war. {\"U}ber Immunfluoreszenzmikroskopie mit dem anti-LaXp180-Serum an mit L. monocytogenes infizierten Zellen konnte gezeigt werden, daß LaXp180 mit der Oberfl{\"a}che vieler, aber nicht aller intrazellul{\"a}rer, ActA-exprimierender Listerien kolokalisiert. Dagegen wurde nie eine Kolokalisation mit intrazellul{\"a}ren, aber ActA-defizienten Mutanten beobachtet. Dar{\"u}berhinaus ist LaXp180 asymmetrisch auf der Bakterienoberfl{\"a}che verteilt und schließt sich gegenseitig mit der F-Aktin-Polymerisation aus. LaXp180 ist ein putativer Bindungspartner von Stathmin, einem 19 kDa Phosphoprotein, das die Mikrotubuli-Dynamik reguliert. {\"U}ber Immunfluoreszenz konnte gezeigt werden, daß auch Stathmin mit intrazellul{\"a}ren, ActA-exprimierenden L. monocytogenes kolokalisiert.}, subject = {Listeria monocytogenes}, language = {de} } @phdthesis{Winkler2015, author = {Winkler, Ann-Cathrin Nicole}, title = {Identification of human host cell factors involved in \(Staphylococcus\) \(aureus\) 6850 infection}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-114300}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2015}, abstract = {Staphylococcus aureus is both a human commensal and a pathogen. 20\%-30\% of all individuals are permanently or occasionally carriers of S. aureus without any symptoms. In contrast to this, S. aureus can cause life-threatening diseases e.g. endocarditis, osteomyelitis or sepsis. Here, the increase in antibiotic resistances makes it more and more difficult to treat these infections and hence the number of fatalities rises constantly. Since the pharmaceutical industry has no fundamentally new antibiotics in their pipeline, it is essential to better understand the interplay between S. aureus and the human host cell in order to find new, innovative treatment options. In this study, a RNA interference based whole genome pool screen was performed to identify human proteins, which play a role during S. aureus infections. Since 1,600 invasion and 2,271 cell death linked factors were enriched at least 2 fold, the big challenge was to filter out the important ones. Here, a STRING pathway analysis proved to be the best option. Subsequently, the identified hits were validated with the help of inhibitors and a second, individualised small interfering RNA-based screen. In the course of this work two important steps were identified, that are critical for host cell death: the first is bacterial invasion, the second phagosomal escape. The second step is obligatory for intracellular bacterial replication and subsequent host cell death. Invasion in turn is determining for all following events. Accordingly, the effect of the identified factors towards these two crucial steps was determined. Under screening conditions, escape was indirectly measured via intracellular replication. Three inhibitors (JNKII, Methyl-beta-cyclodeytrin, 9-Phenantrol) could be identified for the invasion process. In addition, siRNAs targeted against 16 different genes (including CAPN2, CAPN4 and PIK3CG), could significantly reduce bacterial invasion. Seven siRNAs (FPR2, CAPN4, JUN, LYN, HRAS, AKT1, ITGAM) were able to inhibit intracellular replication significantly. Further studies showed that the IP3 receptor inhibitor 2-APB, the calpain inhibitor calpeptin and the proteasome inhibitor MG-132 are able to prevent phagosomal escape and as a consequence intracellular replication and host cell death. In this context the role of calpains, calcium, the proteasome and the mitochondrial membrane potential was further investigated in cell culture. Here, an antagonistic behaviour of calpain 1 and 2 during bacterial invasion was observed. Intracellular calcium signalling plays a major role, since its inhibition protects host cells from death. Beside this, the loss of mitochondrial membrane potential is characteristic for S. aureus infection but not responsible for host cell death. The reduction of membrane potential can be significantly diminished by the inhibition of the mitochondrial Na+/Ca2+ exchanger. All together, this work shows that human host cells massively contribute to different steps in S. aureus infection rather than being simply killed by bacterial pore-forming toxins. Various individual host cell factors were identified, which contribute either to invasion or to phagosomal escape and therefore to S. aureus induced cytotoxicity. Finally, several inhibitors of S. aureus infection were identified. One of them, 2-APB, was already tested in a sepsis mouse model and reduced bacterial load of kidneys. Thus, this study shows valuable evidence for novel treatment options against S. aureus infections, based on the manipulation of host cell signalling cascades.}, subject = {Staphylococcus aureus}, language = {en} } @phdthesis{Stelzner2020, author = {Stelzner, Kathrin}, title = {Identification of factors involved in Staphylococcus aureus- induced host cell death}, doi = {10.25972/OPUS-18899}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-188991}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2020}, abstract = {Staphylococcus aureus is a Gram-positive commensal bacterium, that asymptomatically colonizes human skin and mucosal surfaces. Upon opportune conditions, such as immunodeficiency or breached barriers of the host, it can cause a plethora of infections ranging from local, superficial infections to life-threatening diseases. Despite being regarded as an extracellular pathogen, S. aureus can invade and survive within non-phagocytic and phagocytic cells. Eventually, the pathogen escapes from the host cell resulting in killing of the host cell, which is associated with tissue destruction and spread of infection. However, the exact molecular mechanisms underlying S. aureus-induced host cell death remain to be elucidated. In the present work, a genome-wide haploid genetic screen was performed to identify host cell genes crucial for S. aureus intracellular cytotoxicity. A mutant library of the haploid cell line HAP1 was infected with the pathogen and cells surviving the infection were selected. Twelve genes were identified, which were significantly enriched when compared to an infection with a non-cytotoxic S. aureus strain. Additionally, characteristics of regulated cell death pathways and the role of Ca2+ signaling in S. aureus-infected cells were investigated. Live cell imaging of Ca2+ reporter cell lines was used to analyze single cells. S. aureus-induced host cell death exhibited morphological features of apoptosis and activation of caspases was detected. Cellular H2O2 levels were elevated during S. aureus intracellular infection. Further, intracellular S. aureus provoked cytosolic Ca2+ overload in epithelial cells. This resulted from Ca2+ release from endoplasmic reticulum and Ca2+ influx via the plasma membrane and led to mitochondrial Ca2+ overload. The final step of S. aureus-induced cell death was plasma membrane permeabilization, a typical feature of necrotic cell death. In order to identify bacterial virulence factors implicated in S. aureus-induced host cell killing, the cytotoxicity of selected mutants was investigated. Intracellular S. aureus employs the bacterial cysteine protease staphopain A to activate an apoptosis-like cell death characterized by cell contraction and membrane bleb formation. Phagosomal escape represents a prerequisite staphopain A-induced cell death, whereas bacterial intracellular replication is dispensable. Moreover, staphopain A contributed to efficient colonization of the lung in a murine pneumonia model. In conclusion, this work identified at least two independent cell death pathways activated by intracellular S. aureus. While initially staphopain A mediates S. aureus-induced host cell killing, cytosolic Ca2+-overload follows later and leads to the final demise of the host cell.}, subject = {Staphylococcus aureus}, language = {en} }