TY - THES A1 - Endres, Leo Maximilian T1 - Development of multicellular \(in\) \(vitro\) models of the meningeal blood-CSF barrier to study \(Neisseria\) \(meningitidis\) infection T1 - Entwicklung multizellulärer \(in\) \(vitro\) Modelle der meningealen Blut-Liquor Schranke zur Untersuchung der \(Neisseria\) \(meningitidis\) Infektion N2 - Neisseria meningitidis (the meningococcus) is one of the major causes of bacterial meningitis, a life-threatening inflammation of the meninges. Traversal of the meningeal blood-cerebrospinal fluid barrier (mBCSFB), which is composed of highly specialized brain endothelial cells (BECs), and subsequent interaction with leptomeningeal cells (LMCs) are critical for disease progression. Due to the human-exclusive tropism of N. meningitidis, research on this complex host-pathogen interaction is mostly limited to in vitro studies. Previous studies have primarily used peripheral or immortalized BECs alone, which do not retain relevant barrier phenotypes in culture. To study meningococcal interaction with the mBCSFB in a physiologically more accurate context, BEC-LMC co-culture models were developed in this project using BEC-like cells derived from induced pluripotent stem cells (iBECs) or hCMEC/D3 cells in combination with LMCs derived from tumor biopsies. Distinct BEC and LMC layers as well as characteristic expression of cellular markers were observed using transmission electron microscopy (TEM) and immunofluorescence staining. Clear junctional expression of brain endothelial tight and adherens junction proteins was detected in the iBEC layer. LMC co-culture increased iBEC barrier tightness and stability over a period of seven days, as determined by sodium fluorescein (NaF) permeability and transendothelial electrical resistance (TEER). Infection experiments demonstrated comparable meningococcal adhesion and invasion of the BEC layer in all models tested, consistent with previously published data. While only few bacteria crossed the iBEC-LMC barrier initially, transmigration rates increased substantially over 24 hours, despite constant high TEER. After 24 hours of infection, deterioration of the barrier properties was observed including loss of TEER and altered expression of tight and adherens junction components. Reduced mRNA levels of ZO-1, claudin-5, and VE-cadherin were detected in BECs from all models. qPCR and siRNA knockdown data suggested that transcriptional downregulation of these genes was potentially but not solely mediated by Snail1. Immunofluorescence staining showed reduced junctional coverage of occludin, indicating N. meningitidis-induced post-transcriptional modulation of this protein, as previous studies have suggested. Together, these results suggest a potential combination of transcellular and paracellular meningococcal traversal of the mBCSFB, with the more accessible paracellular route becoming available upon barrier disruption after prolonged N. meningitidis infection. Finally, N. meningitidis induced cellular expression of pro-inflammatory cytokines and chemokines such as IL-8 in all mBCSFB models. Overall, the work described in this thesis highlights the usefulness of advanced in vitro models of the mBCSFB that mimic native physiology and exhibit relevant barrier properties to study infection with meningeal pathogens such as N. meningitidis. N2 - Neisseria meningitidis (der Meningokokkus) ist einer der Hauptursachen bakterieller Meningitis, einer lebensbedrohlichen Entzündung der Hirnhäute. Entscheidend für das für das Voranschreiten der Krankheit ist die Fähigkeit des Erregers, die meningeale Blut-Liquor-Schranke (mBCSFB), bestehend aus spezialisierten Hirnendothelzellen (BECs) und leptomeningealen Zellen (LMCs), zu überwinden und in den submeningealen Raum einzudringen. Da es sich bei N. meningitidis um ein rein humanes Pathogen handelt, beschränkt sich die Erforschung dieser speziellen Interaktion primär auf die Verwendung von in vitro Modellen. Bisher wurden hierfür hauptsächlich periphere oder immortalisierte BECs verwendet, welchen jedoch wichtige Barriere-Eigenschaften fehlen. Um die Interaktion von N. meningitidis mit der mBCSFB in einem physiologisch relevanteren Umfeld zu untersuchen, wurden in dieser Arbeit neuartige BEC-LMC Kokulturmodelle entwickelt. Dabei wurden sowohl BEC-ähnliche Zellen, die aus induzierten pluripotenten Stammzellen generiert wurden (iBECs), als auch hCMEC/D3 Zellen verwendet und zusammen mit LMCs aus Tumorbiopsien kultiviert. Mittels Transmissions-Elektronenmikroskopie und Immunfluoreszenzfärbung konnten die unterschiedlichen Zellschichten und deren Expression charakteristischer zellulärer Marker dargestellt werden. Durchgängige Expression von wichtigen Bestandteilen Barriere-formender Zellverbindungen, sogenannter Tight und Adherens Junctions, wurde in der iBEC-Schicht beobachtet. Die Integrität der zellulären Barriere wurde mittels transendothelialer elektrischer Resistenz (TEER) und Permeabilität gegenüber Natrium-Fluorescein (NaF) bestimmt. Erhöhte TEER-Werte und verringerte NaF-Permeabilität, gemessen über einen Zeitraum von sieben Tagen, zeigten eine durch die Kokultur mit LMCs ausgelöste Steigerung der Dichtigkeit und Stabilität der iBEC-Barriere. Infektionsexperimente mit N. meningitidis zeigten in allen Modellen vergleichbare bakterielle Adhäsion und Invasion der BEC-Schicht. Bakterielle Transmigration durch die gesamten Zellbarriere war im iBEC-LMC Modell kurz nach Infektion nur in geringem Maße detektierbar, nahm jedoch innerhalb von 24 Stunden deutlich zu. Interessanterweise wurde bis zu 24 Stunden nach Infektion noch eine hohe Integrität der Barriere gemessen, welche allerdings im weiteren Verlauf verloren ging. Neben signifikantem TEER-Verlust wurde eine verringerte Expression der Tight und Adherens Junction Proteine ZO-1, claudin-5, und VE-cadherin mittels qPCR festgestellt. qPCR und siRNA Knockdown Experimente deuteten darauf hin, dass dies möglicherweise, aber nicht ausschließlich, auf den Transkriptionsfaktor Snail1 zurückzuführen war. Zusätzlich zu den beobachteten Effekten auf die zelluläre Transkription von Tight Junction Genen, zeigten Immunfluoreszenzfärbungen eine verringerte Expression von Occludin an den Zell-Zell-Verbindungen, was auf eine post-translationale Modulation schließen lässt. Zusammen deuten die Ergebnisse dieser Infektionsstudien auf eine mögliche Kombination aus trans- und parazellulärer bakterieller Transmigration der mBCSFB hin. Zuletzt wurden in dieser Arbeit noch die Immunaktivierung von BECs nach N. meningitidis Infektion in den neuen BEC-LMC Kokulturmodellen untersucht. Hierbei wurde eine erhöhte Expression von Zytokinen, insbesondere Interleukin-8, beobachtet. Insgesamt konnten in dieser Arbeit neue, fortschrittlicher in vitro Modelle der mBCSFB entwickelt werden, welche die humane Physiologie besser widerspiegeln und daher für Infektionsstudien mit Meningitis-verursachenden Erregern wie N. meningitidis von besonderem Nutzen sind. KW - Bakterielle Hirnhautentzündung KW - Blut-Liquor-Schranke KW - Induzierte pluripotente Stammzelle KW - Neisseria meningitidis KW - In-vitro-Kultur KW - Brain endothelial cells KW - Leptomeningeal cells KW - Hirnendothelzellen KW - Leptomeningealzellen Y1 - 2024 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-346216 ER - TY - THES A1 - Münstermann, Marcel T1 - The roles of the anaphylatoxin receptors during invasive disease as well as mucosal colonization caused by \(Neisseria\) \(meningitidis\) T1 - Die Rolle der Anaphylatoxinrezeptoren während invasiver Infektion sowie mukosaler Kolonisation verursacht durch \(Neisseria\) \(meningitidis\) N2 - The human specific gram-negative bacterium Neisseria meningitidis (Nme, meningococci) is a common colonizer of the upper respiratory tract. Upon becoming invasive, Nme can cause meningitis and life-threatening sepsis. The most important immune defense mechanism in invasive meningococcal disease (IMD) is the complement mediated killing of bacteria. The complement cascade is activated through different pathogen associated patterns and finally leads to the lysis of the bacteria by the membrane attack complex. In addition to the direct bacterial killing, the complement system is also an important player in different inflammatory processes. A hallmark of IMD is an overreaction of the immune system and the release of the potent anaphylatoxins C3a and C5a by the complement system is an important factor hereby. There are three anaphylatoxin receptors (ATRs), the C3aR, the C5aR1 and the C5aR2, capable of detecting these anaphylatoxins. It has already been shown that blocking the ATR C5aR1 strongly benefitted the outcome of IMD in a murine sepsis model. However, the roles of ATRs C3aR and C5aR2 in IMD are still unclear. This work aims to analyze the role of these ATRs in meningococcal sepsis and to identify possible underlying mechanisms. Furthermore, a possible involvement of the complement system, the ATRs and the type II CRISPR/Cas system on nasopharyngeal colonization is analyzed. In vivo depletion experiments showed that without neutrophils or monocytes/macrophages the complement system alone was not able to clear a low dose Nme infection, which highlights the importance of cellular components in IMD. Analyzing the role of the ATRs in knock-out mice with high dose Nme infections, revealed that the lack of C5aR2, like the lack of C5aR1, was beneficial for the outcome of meningococcal induced sepsis. In contrast, the lack of C3aR in knock-out mice was detrimental. The positive outcome associated with the C5aRs could be reproduced by using an antagonist against both C5aRs or an antagonist specifically against C5aR1 in WT mice. These findings are giving hope to future therapeutic applications. Next, a possible contribution of neutrophils to this positive outcome was analyzed. Absence of C5aR1 led to a decrease of degranulation by neutrophils in a murine whole blood model, while the other ATRs showed no effect. Neutrophil analysis in human whole blood, on the other hand, revealed a reduced oxidative burst and IL-8 secretion upon inhibition of all three ATRs. A functional difference between the C5aRs and the C3aR in neutrophils was observed in phagocytosis, which was reduced upon C3aR inhibition, but was unaltered with C5aR1 or C5aR2 inhibition. Possible underlying mechanisms in the phosphorylation of ERK1/2 were analyzed in bone marrow derived macrophages isolated from ATR knock-out mice. The later phosphorylation of ERK1/2 in macrophages without C5aR1 or C5aR2 expression might explain, why blocking the C5aRs is beneficial for the outcome of IMD in mice. In contrast to these findings, the colonization of the nasopharynx in huCEACAM 1 expressing mice by Nme did not seem to depend on the Complement system factors C3 and C5 nor the ATRs. Additionally, no difference in the colonization could be observed in this model using Nme mutants lacking different parts of the type 2 CRISPR/Cas system. Conclusively, this work highlights the importance of the complement system, the ATRs and the cellular components in IMD. Contrariwise, these factors did not play a role in the analyzed nasopharyngeal infection model. The beneficial effects of C5aR1 and C5aR2 lack/inhibition in IMD might have medicinal applications, which could support the standard therapies of IMD in the future. N2 - Das human spezifische pathogene Gram-negative Bakterium Neisseria meningitidis (Nme, Meningokokken) ist ein Kommensale angesiedelt im Nasopharynx. Bei invasiver Erkrankung können Nme Meningitis oder eine lebensbedrohliche Sepsis verursachen. Die wichtigste Verteidigung des Immunsystems in invasiver Meningokokken-Erkrankung (IMD) ist die Abtötung von Bakterien durch das Komplementsystem. Die Komplementkaskade wird durch verschiedene pathogenassoziierte Muster in Gang gesetzt und resultiert in dem Aufbau des Membranangriffskomplex, welcher die Bakterien schließlich lysiert. Darüber hinaus spielt das Komplementsystem auch eine wichtige Rolle in verschiedenen inflammatorischen Prozessen im Körper. Ein charakteristisches Merkmal von IMD ist eine übermäßige Reaktion des Immunsystems und dabei ist die Freisetzung der Anaphylatoxine C3a und C5a, durch das Komplementsystem, ein wichtiger Faktor. Es gibt drei Anaphylatoxin Rezeptoren (ATR), den C3aR, den C5aR1 und den C5aR2, welche die jeweiligen Anaphylatoxine erkennen. In murinen Modellen wurde bereits gezeigt, dass die Inhibition des C5aR1 einen positiven Einfluss auf den Verlauf von IMD hat. Im Kontrast dazu sind die Rollen der ATRs C3aR und C5aR2 in IMD weiter unklar. Diese Arbeit hat als Ziel, die Rolle der ATRs in Meningokokken induzierter Sepsis zu untersuchen und mögliche zugrundeliegende Mechanismen zu finden. Des Weiteren soll ein möglicher Einfluss des Komplementsystems, der ATRs und des Typ II CRISPR/Cas Systems auf die Kolonisation durch Nme im Nasopharynx untersuchen werden. In vivo Depletions-Versuche zeigten, dass ohne Neutrophile oder Monozyten/Makrophagen das Komplementsystem allein nicht in der Lage war eine Nme-Infektion mit einer niedrigen Infektionsdosis zu beseitigen. Dies zeigt die Wichtigkeit von Immunzellen neben dem Komplementsystem in IMD. Experimente mit hohen Nme-Dosen in ATR knock-out Mäusen zeigten, dass die fehlende Expression von C5aR2, wie die von C5aR1, sich positiv auf den Ausgang von IMD auswirkte. Im Gegensatz dazu, verschlimmerte das Fehlen des C3aR Rezeptors den Ausgang der IMD. Die positive Wirkung in den C5aR knock-out Mäusen, konnte auch mit der Gabe von einem gegen beide C5aRs oder einem spezifisch gegen C5aR1 gerichteten Antagonisten in WT Mäusen beobachtet werden. Diese Ergebnisse geben Hoffnung auf eine mögliche zukünftige therapeutische Applikation. Als nächstes wurde eine mögliche Beteiligung von Neutrophilen an dem positiven Ausgang von IMD in Abhängigkeit von den ATRs untersucht. Eine fehlende C5aR1 Expression führte zu einer verminderten Degranulation durch Neutrophile in dem verwendeten murinen Vollblutmodel, wohingegen die fehlende Expression der anderen ATRs keinen Effekt zeigte. Im Gegensatz dazu, zeigten Versuche mit humanem Vollblut einen verminderten Oxidativen Burst sowie eine verminderte Ausschüttung von IL-8 bei der Blockade von allen drei ATRs. Ein Unterschied zwischen den C5aRs und dem C3aR zeigte sich hingegen in der Phagozytose, welche mit C3aR Inhibierung reduziert war, aber unverändert nach der Inhibierung von C5aR1 oder C5aR2 blieb. Mögliche zugrundeliegende Mechanismen in der Phosphorylation von ERK1/2 wurden anschließend in Knochenmark-gereiften Makrophagen von ATR knock-out Mäusen untersucht. Ohne C5aR1 oder C5aR2 Expression wurde eine verzögerte Phosphorylierung von ERK1/2 in den Makrophagen beobachtet, was erklären könnte warum die Blockade von C5aRs den Ausgang von Meningokokken induzierter Sepsis in Mäusen positiv beeinflusst. Im Gegensatz zu diesen Ergebnissen wurde die Kolonisation des Nasopharynx durch Nme in huCEACAM-1 exprimierenden Mäusen, weder durch die Komplementfaktoren C3 und C5 noch durch die ATRs beeinflusst. Zusätzlich konnte auch kein Unterschied in der Besiedelung des Nasopharynx durch Nme-Mutanten, die verschiedene Mutationen des Typ 2 CRISPR/Cas Systems besaßen, beobachtet werden. Diese Arbeit zeigt die Wichtigkeit des Komplementsystems, der ATRs und der Immunzellen in IMD. Zusätzlich zeigt diese Arbeit, dass das Komplementsystem und die ATRs jedoch keine Auswirkungen auf die Kolonisation des Nasopharynx in Mäusen haben. Die äußerst positive Auswirkung auf IMD, wenn C5aR1 und C5aR2 nicht gebildet oder blockiert werden, könnte medizinisch von Bedeutung sein und eventuell in der Zukunft die Standarttherapie bei IMD unterstützen. KW - Anaphylatoxine KW - Komplement C3a KW - Komplement C5a KW - Neisseria meningitidis KW - Komplement KW - anaphylatoxin receptors KW - invasive meningococcal diseases KW - Anaphylatoxinrezeptoren Y1 - 2022 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-269759 ER - TY - JOUR A1 - Peters, Simon A1 - Fohmann, Ingo A1 - Rudel, Thomas A1 - Schubert-Unkmeir, Alexandra T1 - A Comprehensive Review on the Interplay between Neisseria spp. and Host Sphingolipid Metabolites JF - Cells N2 - Sphingolipids represent a class of structural related lipids involved in membrane biology and various cellular processes including cell growth, apoptosis, inflammation and migration. Over the past decade, sphingolipids have become the focus of intensive studies regarding their involvement in infectious diseases. Pathogens can manipulate the sphingolipid metabolism resulting in cell membrane reorganization and receptor recruitment to facilitate their entry. They may recruit specific host sphingolipid metabolites to establish a favorable niche for intracellular survival and proliferation. In contrast, some sphingolipid metabolites can also act as a first line defense against bacteria based on their antimicrobial activity. In this review, we will focus on the strategies employed by pathogenic Neisseria spp. to modulate the sphingolipid metabolism and hijack the sphingolipid balance in the host to promote cellular colonization, invasion and intracellular survival. Novel techniques and innovative approaches will be highlighted that allow imaging of sphingolipid derivatives in the host cell as well as in the pathogen. KW - sphingolipids KW - host–pathogen interaction KW - Neisseria meningitidis KW - Neisseria gonorrhoeae Y1 - 2021 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-250203 SN - 2073-4409 VL - 10 IS - 11 ER - TY - THES A1 - Krüger, Sören T1 - Unterschiedliche Einflüsse von Komplement auf Reaktionen neutrophiler Granulozyten auf die Infektion mit \(Neisseria\) \(meningitidis\) T1 - Differential influences of complement on neutrophil responses to \(Neisseria\) \(meningitidis\) infection N2 - The gram-negative diplococcus Neisseria meningitidis (Nme) is a frequent human-specific, commensal bacterium of the upper respiratory tract. Under certain conditions especially in infants, meningococci can translocate into the bloodstream and cause invasive meningococcal disease (IMD) manifesting as meningitis or sepsis or a combination of both. IMD is feared for its rapid progression and high fatality rate if it remains untreated. IMD affects up to one million people annually causing substantial morbidity and mortality worldwide. It is well-established that the complement system is an important protective factor in meningococcal disease through opsonization of bacteria with C3b and the lytic activity of the membrane attack complex although the inflammatory C5a/C5aR1 axis can aggravate IMD. The role of neutrophil granulocytes in meningococcal infection is less clear despite their abundant recruitment throughout the course of disease. This study aimed to characterize neutrophil responses to Nme in vitro and the influence of complement on these responses. In infection assays with whole blood and isolated PMNs, effective binding, internalization and killing of Nme by neutrophils was demonstrated. A significant complement-dependence of neutrophil phagocytosis and oxidative burst was observed. The opsonizing and lytic pathway of the complement cascade were found to be most relevant for these responses since blockade of C3 using inhibitor Compstatin Cp20 reduced phagocytosis and oxidative burst significantly more than the blockade of the inflammatory branch with C5aR1-antagonist PMX53. Opsonization with specific antibodies could not replicate the effect of complement activation indicating that engagement of neutrophil complement receptors, particularly complement receptor 3, is involved. Other neutrophil effector functions such as degranulation and IL-8 release were activated in a complement-independent manner implying activation by other inflammatory signals. Considering existing evidence on the overall protective effect of PMNs, further studies investigating the contribution of each neutrophil effector function to infection survival in vivo are required. Ideally, this should be studied in a murine meningitis or sepsis model in the context of complement activation. N2 - Der Gram-negative Diplokokkus Neisseria meningitidis (Nme) ist ein weit verbreitetes, human-spezifisches, kommensales Bakterium des oberen Respirationstraktes. Unter bestimmten Bedingungen, insbesondere bei Kleinkindern, können Meningokokken in die Blutbahn translozieren und eine invasive Meningokokkenerkrankung (IMD) auslösen, die sich als Meningitis, Sepsis oder eine Kombination beider Krankheitsbilder manifestiert. IMD werden aufgrund ihrer raschen Progression und ohne Behandlung hohen Letalität gefürchtet. IMD betreffen jährlich bis zu einer Million Menschen weltweit mit substanzieller Morbidität und Mortalität. Es ist bekannt, dass das Komplement-system bei IMD als protektiver Faktor durch die Opsonisierung von Nme mit C3b und Lyse mittels Membranangriffskomplex wirkt, obwohl die inflammatorische C5a/C5aR1-Achse die Erkrankung aggravieren kann. Die Rolle neutrophiler Granulozyten (PMNs) bei IMD bleibt hingegen kontrovers, obwohl eine starke Rekrutierung dieser Zellen stattfindet. Diese Studie charakterisiert die Reaktionen neutrophiler Granulozyten auf Nme in vitro und deren Modulation durch Komplement. In Infektionsassays mit Vollblut und isolierten PMNs konnte die Bindung, Internalisierung und Elimination von Nme durch PMNs demonstriert werden. Oxidativer burst und Phagozytose zeigten dabei eine ausgeprägte Abhängigkeit von Komplement. Die opsonisierende und lytische Wirkung von Komplement erwiesen sich dabei als relevant für diese Funktionen, da die Blockade von C3 durch den Inhibitor Compstatin Cp20 Phagozytose und oxidativen Burst signifikant stärker reduzierte als die Blockade der Inflammation durch C5a mittels C5aR1-Antagonist PMX53. Die Opsonisierung mit spezifischen Antikörpern konnte den Effekt der Komplementaktivierung nicht replizieren, was auf eine Signalvermittlung durch Komplementrezeptoren insbesondere CR3 hindeutet. Andere Effektorfunktionen wie Degranulation und IL-8 Freisetzung waren komplementunabhängig, was auf deren Initiierung durch andere inflammatorische Signale hindeutet. In Anbetracht bestehender Evidenz für einen insgesamt protektiven Effekt von PMN sind weitere Studien nötig, die den Effekt der einzelnen Effektorfunktionen auf das Outcome in vivo untersuchen. Besonders geeignet wäre dafür ein murines Sepsis oder Meningitis-Modell. KW - Neisseria meningitidis KW - Neutrophiler Granulozyt KW - Komplement KW - Complement system KW - Neutrophil granulocyte Y1 - 2021 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-249697 ER - TY - JOUR A1 - Bauriedl, Saskia A1 - Gerovac, Milan A1 - Heidrich, Nadja A1 - Bischler, Thorsten A1 - Barquist, Lars A1 - Vogel, Jörg A1 - Schoen, Christoph T1 - The minimal meningococcal ProQ protein has an intrinsic capacity for structure-based global RNA recognition JF - Nature Communications N2 - FinO-domain proteins are a widespread family of bacterial RNA-binding proteins with regulatory functions. Their target spectrum ranges from a single RNA pair, in the case of plasmid-encoded FinO, to global RNA regulons, as with enterobacterial ProQ. To assess whether the FinO domain itself is intrinsically selective or promiscuous, we determine in vivo targets of Neisseria meningitidis, which consists of solely a FinO domain. UV-CLIP-seq identifies associations with 16 small non-coding sRNAs and 166 mRNAs. Meningococcal ProQ predominantly binds to highly structured regions and generally acts to stabilize its RNA targets. Loss of ProQ alters transcript levels of >250 genes, demonstrating that this minimal ProQ protein impacts gene expression globally. Phenotypic analyses indicate that ProQ promotes oxidative stress resistance and DNA damage repair. We conclude that FinO domain proteins recognize some abundant type of RNA shape and evolve RNA binding selectivity through acquisition of additional regions that constrain target recognition. FinO-domain proteins are bacterial RNA-binding proteins with a wide range of target specificities. Here, the authors employ UV CLIP-seq and show that minimal ProQ protein of Neisseria meningitidis binds to various small non-coding RNAs and mRNAs involved in virulence. KW - Neisseria meningitidis KW - natural transformation KW - dual function KW - FinO family KW - HFQ KW - chaperone KW - transcriptome KW - regulator KW - sequence KW - in vivo Y1 - 2020 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-230040 VL - 11 ER - TY - THES A1 - Peters, Simon T1 - The impact of sphingolipids on \(Neisseria\) \(meningitidis\) and their role in meningococcal pathogenicity T1 - Einfluss von Sphingolipiden auf \(Neisseria\) \(meningitidis\) und deren Bedeutung für die Pathogenität N2 - The obligate human pathogen Neisseria meningitidis is a major cause of sepsis and meningitis worldwide. It affects mainly toddlers and infants and is responsible for thousands of deaths each year. In this study, different aspects of the importance of sphingolipids in meningococcal pathogenicity were investigated. In a first step, the acid sphingomyelinase (ASM), which degrades membrane sphingomyelin to ceramide, was studied in the context of meningococcal infection. A requirement for ASM surface activity is its translocation from the lysosomal compartment to the cell surface, a process that is currently poorly understood. This study used various approaches, including classical invasion and adherence assays, flow cytometry, and classical and super resolution immunofluorescence microscopy (dSTORM). The results showed that the live, highly piliated N. meningitidis strain 8013/12 induced calcium-dependent ASM translocation in human brain microvascular endothelial cells (HBMEC). Furthermore, it promoted the formation of ceramide-rich platforms (CRPs). In addition, ASM translocation and CRP formation were observed after treating the cells with pili-enriched fractions derived from the same strain. The importance for N. meningitidis to utilize this pathway was shown by the inhibition of the calcium-dependent ASM translocation, which greatly decreased the number of invasive bacteria. I also investigated the importance of the glycosphingolipids GM1 and Gb3. The results showed that GM1, but not Gb3, plays an important role in the ability of N. meningitidis to invade HBMEC. By combining dSTORM imaging and microbiological approaches, we demonstrated that GM1 accumulated prolifically around bacteria during the infection, and that this interaction seemed essential for meningococcal invasion. Sphingolipids are not only known for their beneficial effect on pathogens. Sphingoid bases, including sphingosine, are known for their antimicrobial activity. In the last part of this study, a novel correlative light and electron microscopy approach was established in the combination with click chemistry to precisely localize azido-functionalized sphingolipids in N. meningitidis. The result showed a distinct concentration-dependent localization in either the outer membrane (low concentration) or accumulated in the cytosol (high concentration). This pattern was confirmed by mass spectrometry on separated membrane fractions. Our data provide a first insight into the underlying mechanism of antimicrobial sphingolipids. N2 - Der obligate Humanpathogen Neisseria meningitidis ist weltweit einer der Hauptursachen für Sepsis und Meningitis. Er befällt vor allem Kleinkinder und Säuglinge und ist jedes Jahr für Tausende von Todesfällen verantwortlich. In dieser Studie wurden verschiedene Aspekte der Bedeutung von Sphingolipiden bei der Pathogenität von Meningokokken untersucht. In einem ersten Schritt wurde die saure Sphingomyelinase (ASM), die Membran-Sphingomyelin zu Ceramid abbaut, im Zusammenhang mit einer Meningokokken-Infektion untersucht. Eine Voraussetzung für die Oberflächenaktivität der ASM ist ihre Translokation vom lysosomalen Kompartiment auf die Zelloberfläche, ein Prozess, der derzeit noch wenig verstanden wird. In dieser Studie wurden verschiedene Ansätze verwendet, darunter klassische Invasions- und Adhärenztests, Durchflusszytometrie sowie klassische und superauflösende Immunfluoreszenzmikroskopie (dSTORM). Die Ergebnisse zeigten, dass der lebende, hochpiliatisierte N. meningitidis Stamm 8013/12 eine kalziumabhängige ASM-Translokation in mikrovaskulären Endothelzellen des menschlichen Gehirns (HBMEC) induzierte. Des Weiteren förderte er die Bildung Ceramid-reicher Plattformen (CRPs). Zusätzlich wurden ASM-Translokation und CRP-Bildung beobachtet, nachdem die Zellen mit pili-angereicherten Fraktionen desselben Stammes behandelt worden waren. Die Bedeutung für N. meningitidis in der Pathogenese zeigte sich durch die Hemmung der Calcium-abhängigen ASM-Translokation, wodurch die Zahl der invasiven Bakterien stark reduziert wurde. Ich untersuchte auch die Bedeutung der Glykosphingolipide GM1 und Gb3. Die Ergebnisse zeigten, dass GM1, aber nicht Gb3, eine wichtige Rolle bei der Fähigkeit von N. meningitidis spielt, in Gehirnendothelzellen einzudringen. Durch die Kombination von dSTORM-Bildgebung und mikrobiologischen Ansätzen konnten wir zeigen, dass sich GM1 während der Infektion vermehrt um die Bakterien herum anreicherte und dass diese Interaktion für die Invasion von Meningokokken essenziell ist. Sphingolipide sind nicht nur für ihre positive Wirkung auf Krankheitserreger bekannt. Sphingoidbasen, einschließlich Sphingosin, sind zusätzlich für ihre antimikrobielle Aktivität bekannt. Im letzten Teil dieser Studie wurde ein neuartiger korrelativer licht- und elektronenmikroskopischer Ansatz in der Kombination mit Click-Chemie etabliert, um azidofunktionalisierte Sphingolipide in N. meningitidis genau zu lokalisieren. Das Ergebnis zeigte eine deutliche konzentrationsabhängige Lokalisation entweder in der äußeren Membran (niedrige Konzentration) oder akkumuliert im Zytosol (hohe Konzentration). Dieses Muster konnte durch einen Massenspektrometrischen Ansatz bestätigt werden. Hierfür wurde eine Separation der inneren und äußeren Membran, nach Behandlung mit der niedrigen Konzentration, etabliert. Die verschiedenen Membranfraktionen wurden anschließend auf ihren Gehalt an funktionalisierten Sphingolipiden hin untersucht und bestätigten die lokalisierung in der äußeren Membran. Unsere Daten geben einen ersten Einblick in den zugrundeliegenden Mechanismus der antimikrobiellen Sphingolipide. KW - Neisseria meningitidis KW - Sphingolipide KW - Infektion KW - Pathogenität KW - host-pathogen interaction KW - antimicrobial Y1 - 2021 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-226233 ER - TY - THES A1 - Bauriedl, Saskia Corinna T1 - The influence of riboregulation on fitness and virulence in Neisseria meningitidis T1 - Der Einfluss der Riboregulation auf Fitness und Virulenz von Neisseria meningitidis N2 - Neisseria meningitidis (N. meningitidis) is a human commensal that occasionally causes life-threatening infections such as bacterial meningitis and septicemia. Despite experi-mental evidence that the expression of small non-coding RNAs (sRNAs) as well as the RNA chaperone Hfq affect meningococcal physiology, the impact of RNA-based regula-tion (riboregulation) on fitness and virulence in N. meningitidis is only poorly understood. Therefore, this study addressed these issues using a combination of high-throughput tech-nologies. A differential RNA-sequencing (dRNA-seq) approach was applied to produce a single-nucleotide resolution map of the primary transcriptome of N. meningitidis strain 8013. The dRNA-seq analysis predicted 1,625 transcriptional start sites including 65 putative sRNAs, of which 20 were further validated by northern blot analysis. By Hfq RNA im-munopreci-pitation sequencing a large Hfq-centered post-transcriptional regulatory net-work comprising 23 sRNAs and 401 potential mRNA targets was identified. Rifampicin stability assays demonstrated that Hfq binding confers enhanced stability on its associat-ed sRNAs. Based on these data, the interactions of two paralogous sRNAs and their cog-nate target mRNA prpB were validated in vivo as well as in vitro. Both sRNAs directly repress prpB encoding a methylisocitrate lyse which was previously shown to be involved in meningococcal colonization of the human nasopharynx. Besides the well-described RNA chaperone Hfq, FinO-domain proteins have recently been recognized as a widespread family of RNA-binding proteins (RBPs) with regulatory roles in diverse bacteria. They display an intriguing bandwidth of target sites, ranging from a single RNA pair as recognized by plasmid-encoded FinO to the global RNA regu-lons of enterobacterial ProQ proteins. To better understand the intrinsic targeting mode of this RBP family, in vivo targets of the minimal ProQ protein of N. meningitidis were de-termined. In vivo UV crosslinking with RNA deep sequencing (UV-CLIP) identified as-sociations of ProQ with 16 sRNAs and 166 mRNAs encoding a variety of biological functions and thus revealed ProQ as another global RBP in meningococci. It could be shown that meningococcal ProQ predominantly binds to highly structured RNA regions including DNA uptake sequences (DUS) and rho-independent transcription terminators and stabilizes many of its RNA targets as proved by rifampicin stability experiments. As expected from the large suite of ProQ-bound RNAs, proQ deletion globally affects both gene and protein expression in N. meningitidis, changing the expression levels of at least 244 mRNAs and 80 proteins. Phenotypic analyses suggested that ProQ promotes oxida-tive stress tolerance and UV damage repair capacity, both of which are required for full virulence of N. meningitidis. Together, this work uncovers the co-existence of two major post-transcriptional regulons, one governed by ProQ, the other by Hfq, in N. meningitidis. It further highlights the role of these distinct RBPs and its associated sRNAs to bacterial virulence and indicates that riboregulation is likely to contribute to the way how meningococci adapt to different host niches. N2 - Neisseria meningitidis (N. meningitidis) ist ein kommensal lebendes Bakterium, welches unter nicht vollständig geklärten Bedingungen auch lebensbedrohliche Infektionen im Menschen wie bakterielle Meningitis und Sepsis verursachen kann. Obwohl experimentell nachgewiesen wurde, dass die Expression kleiner, nicht kodierender RNAs (sRNAs) so-wie des RNA-Chaperons Hfq in Meningokokken physiologisch relevant ist, blieb der Ein-fluss der RNA-basierten Genregulation (Riboregulation) auf die Fitness und Virulenz von N. meningitidis bisher unvollständig verstanden. Daher befasste sich diese Studie durch Kombination verschiedener Hochdurchsatz-Technologien mit dieser Fragestellung. Es wurde differentielle RNA-Sequenzierung (dRNA-seq) angewendet, um das primäre Transkriptom des N. meningitidis Stamms 8013 möglichst genau zu kartieren. Die durch-geführte dRNA-seq-Analyse detektierte 1.625 Transkriptionsstartstellen (TSS) einschließ-lich 65 potentieller sRNAs. Durch Anwendung von Northern-Blot-Analysen konnten an-schließend 20 sRNAs experimentell validiert werden. Darüber hinaus wurde durch Ko-Immunopräzipitation mit Hfq (RIP-seq) ein großes, Hfq-zentriertes, post- transkripti-onelles regulatorisches Netzwerk identifiziert, welches 23 sRNAs und 401 mRNAs um-fasst. Rifampicin-Stabilitätsversuche zeigten, dass durch Hfq-Bindung die Stabilität die-ser sRNAs erhöht wird. Basierend auf diesen Daten konnte die Interaktion zwischen zweier Hfq-gebundener paraloger sRNAs und der prpB mRNA sowohl in vivo als auch in vitro bestätigt werden. Beide sRNAs reprimieren die Translation des PrpB-Genes, wel-ches für eine Methylisocitratlyase kodiert und wahrscheinlich die Kolonisation des menschlichen Nasopharynxs durch Meningokokken begünstigt. Neben dem ausführlich charakterisierten RNA-Chaperon Hfq wurden Proteine mit FinO-Domäne kürzlich als eine neue Familie von RNA-bindenden Proteinen (RBPs) mit regula-torischen Funktionen in verschiedenen Bakterien identifiziert. Sie weisen eine große Bandbreite regulierter Gene auf: Während das Plasmid-kodierte FinO-Protein nur ein ein-zelnes RNA-Paar bindet, stellt das enterobakterielle ProQ-Protein ein globales RBP dar. Um die Wirkungsweise dieser RBP-Familie besser zu verstehen, wurde in vivo untersucht, wie viele RNAs mit dem minimalen ProQ-Protein in N. meningitidis assoziiert sind. Durch Kombination von UV-Crosslinken mit RNA-Sequenzierung (UV-CLIP) konnte die Bin-dung von 16 sRNAs und 166 biologisch diverser mRNAs mit ProQ identifiziert werden, welches daher ebenfalls ein globales RBP in Meningokokken darstellt. Es konnte gezeigt werden, dass ProQ vorwiegend RNA-Regionen mit ausgeprägter Sekundärstruktur bin-det, darunter DNA-Aufnahmesequenzen (DUS) und Rho-unabhängige Transkriptions-terminatoren. Die ProQ-Bindung führt dabei häufig zur Stabilisation der RNAs, was durch Rifampicin-Stabilitätsexperimente nachgewiesen wurde. Wie aufgrund der großen Zahl ProQ-gebundener RNAs zu erwarten, beeinflusste die Deletion des ProQ Proteins die zelluläre Expression von mindestens 244 mRNAs und 80 Proteinen. Phänotypische Analysen deuten darauf hin, dass ProQ sowohl die Toleranz gegenüber oxidativem Stress als auch die Reparatur von DNA-Schäden reguliert, die beide für die vollständige Viru-lenz von N. meningitidis von Bedeutung sind. Zusammenfassend beschreibt diese Arbeit die Koexistenz von zwei großen posttranskrip-tionellen Regulons in N. meningitidis, von denen eines von ProQ und das andere von Hfq kontrolliert wird. Im Rahmen dieser Arbeit wurde die Rolle beider RBPs und ihrer assozi-ierten sRNAs für die bakterielle Virulenz verdeutlicht und hervorgehoben, dass Riboregu-lation sehr wahrscheinlich dazu beiträgt, wie sich Meningokokken an verschiedene Wirts-nischen anpassen. KW - Neisseria meningitidis KW - Small non-messenger RNS KW - Hfq KW - ProQ KW - Non-coding RNA KW - High throughput screening Y1 - 2020 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-192978 ER - TY - JOUR A1 - Taha, Muhamed-Kheir A1 - Claus, Heike A1 - Lappann, Martin A1 - Veyrier, Frédéric J. A1 - Otto, Andreas A1 - Becher, Dörte A1 - Deghmane, Ala-Eddine A1 - Frosch, Matthias A1 - Hellenbrand, Wiebke A1 - Hong, Eva A1 - du Châtelet, Isabelle Parent A1 - Prior, Karola A1 - Harmsen, Dag A1 - Vogel, Ulrich T1 - Evolutionary Events Associated with an Outbreak of Meningococcal Disease in Men Who Have Sex with Men JF - PLoS ONE N2 - Meningococci spread via respiratory droplets, whereas the closely related gonococci are transmitted sexually. Several outbreaks of invasive meningococcal disease have been reported in Europe and the United States among men who have sex with men (MSM). We recently identified an outbreak of serogroup C meningococcal disease among MSM in Germany and France. In this study, genomic and proteomic techniques were used to analyze the outbreak isolates. In addition, genetically identical urethritis isolates were recovered from France and Germany and included in the analysis. Genome sequencing revealed that the isolates from the outbreak among MSM and from urethritis cases belonged to a clade within clonal complex 11. Proteome analysis showed they expressed nitrite reductase, enabling anaerobic growth as previously described for gonococci. Invasive isolates from MSM, but not urethritis isolates, further expressed functional human factor H binding protein associated with enhanced survival in a newly developed transgenic mouse model expressing human factor H, a complement regulatory protein. In conclusion, our data suggest that urethritis and outbreak isolates followed a joint adaptation route including adaption to the urogenital tract. KW - nitrites KW - genome sequencing KW - men who have sex with men KW - meningococcal disease KW - Neisseria meningitidis KW - Neisseria gonorrhoeae KW - mammalian genomics KW - mouse models Y1 - 2016 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-179870 VL - 11 IS - 5 ER - TY - JOUR A1 - Drayß, Maria A1 - Claus, Heike A1 - Hubert, Kerstin A1 - Thiel, Katrin A1 - Berger, Anja A1 - Sing, Andreas A1 - van der Linden, Mark A1 - Vogel, Ulrich A1 - Lâm, Thiên-Trí T1 - Asymptomatic carriage of Neisseria meningitidis, Haemophilus influenzae, Streptococcus pneumoniae, Group A Streptococcus and Staphylococcus aureus among adults aged 65 years and older JF - PLoS ONE N2 - Objective The aim of this study was to determine the prevalence of Neisseria meningitidis, Haemophilus influenzae, Streptococcus pneumoniae, group A Streptococcus (GAS), and Staphylococcus aureus in asymptomatic elderly people and to unravel risk factors leading to colonization. Methods A multi-centre cross-sectional study was conducted including 677 asymptomatic adults aged 65 years or more, living at home or in nursing homes. Study areas were Greater Aachen (North-Rhine-Westphalia) and Wuerzburg (Bavaria), both regions with medium to high population density. Nasal and oropharyngeal swabs as well as questionnaires were collected from October 2012 to May 2013. Statistical analysis included multiple logistic regression models. Results The carriage rate was 1.9% ([95%CI: 1.0–3.3%]; 13/677) for H. influenzae, 0.3% ([95%CI: 0–1.1%]; 2/677) for N. meningitidis and 0% ([95% CI: 0–0.5%]; 0/677) for S. pneumoniae and GAS. Staphylococcus aureus was harboured by 28.5% of the individuals ([95% CI: 25.1–32.1%]; 193/677) and 0.7% ([95% CI: 0.2–1.7%]; 5/677) were positive for methicillin-resistant S. aureus. Among elderly community-dwellers colonization with S. aureus was significantly associated with higher educational level (adjusted OR: 1.905 [95% CI: 1.248–2.908]; p = 0.003). Among nursing home residents colonization was associated with being married (adjusted OR: 3.367 [1.502–7.546]; p = 0.003). Conclusion The prevalence of N. meningitidis, H. influenzae, S. pneumoniae and GAS was low among older people in Germany. The S. aureus rate was expectedly high, while MRSA was found in less than 1% of the individuals. KW - Geriatric care KW - Geriatrics KW - Elderly KW - Staphylococcus aureus KW - Nursing homes KW - Haemophilus influenzae KW - Neisseria meningitidis KW - Methicillin-resistant Staphylococcus aureus Y1 - 2019 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-201042 VL - 14 IS - 2 ER - TY - JOUR A1 - Schlegel, Jan A1 - Peters, Simon A1 - Doose, Sören A1 - Schubert-Unkmeir, Alexandra A1 - Sauer, Markus T1 - Super-resolution microscopy reveals local accumulation of plasma membrane gangliosides at Neisseria meningitidis Invasion Sites JF - Frontiers in Cell and Developmental Biology N2 - Neisseria meningitidis (meningococcus) is a Gram-negative bacterium responsible for epidemic meningitis and sepsis worldwide. A critical step in the development of meningitis is the interaction of bacteria with cells forming the blood-cerebrospinal fluid barrier, which requires tight adhesion of the pathogen to highly specialized brain endothelial cells. Two endothelial receptors, CD147 and the β2-adrenergic receptor, have been found to be sequentially recruited by meningococci involving the interaction with type IV pilus. Despite the identification of cellular key players in bacterial adhesion the detailed mechanism of invasion is still poorly understood. Here, we investigated cellular dynamics and mobility of the type IV pilus receptor CD147 upon treatment with pili enriched fractions and specific antibodies directed against two extracellular Ig-like domains in living human brain microvascular endothelial cells. Modulation of CD147 mobility after ligand binding revealed by single-molecule tracking experiments demonstrates receptor activation and indicates plasma membrane rearrangements. Exploiting the binding of Shiga (STxB) and Cholera toxin B (CTxB) subunits to the two native plasma membrane sphingolipids globotriaosylceramide (Gb3) and raft-associated monosialotetrahexosylganglioside GM1, respectively, we investigated their involvement in bacterial invasion by super-resolution microscopy. Structured illumination microscopy (SIM) and direct stochastic optical reconstruction microscopy (dSTORM) unraveled accumulation and coating of meningococci with GM1 upon cellular uptake. Blocking of CTxB binding sites did not impair bacterial adhesion but dramatically reduced bacterial invasion efficiency. In addition, cell cycle arrest in G1 phase induced by serum starvation led to an overall increase of GM1 molecules in the plasma membrane and consequently also in bacterial invasion efficiency. Our results will help to understand downstream signaling events after initial type IV pilus-host cell interactions and thus have general impact on the development of new therapeutics targeting key molecules involved in infection. KW - Neisseria meningitidis KW - sphingolipids KW - gangliosides and lipid rafts KW - super-resolution microscopy KW - single-molecule tracking Y1 - 2019 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-201639 VL - 7 IS - 194 ER - TY - JOUR A1 - Gomes, Sara F. Martins A1 - Westermann, Alexander J. A1 - Sauerwein, Till A1 - Hertlein, Tobias A1 - Förstner, Konrad U. A1 - Ohlsen, Knut A1 - Metzger, Marco A1 - Shusta, Eric V. A1 - Kim, Brandon J. A1 - Appelt-Menzel, Antje A1 - Schubert-Unkmeir, Alexandra T1 - Induced pluripotent stem cell-derived brain endothelial cells as a cellular model to study Neisseria meningitidis infection JF - Frontiers in Microbiology N2 - Meningococcal meningitis is a severe central nervous system infection that occurs when Neisseria meningitidis (Nm) penetrates brain endothelial cells (BECs) of the meningeal blood-cerebrospinal fluid barrier. As a human-specific pathogen, in vivo models are greatly limited and pose a significant challenge. In vitro cell models have been developed, however, most lack critical BEC phenotypes limiting their usefulness. Human BECs generated from induced pluripotent stem cells (iPSCs) retain BEC properties and offer the prospect of modeling the human-specific Nm interaction with BECs. Here, we exploit iPSC-BECs as a novel cellular model to study Nm host-pathogen interactions, and provide an overview of host responses to Nm infection. Using iPSC-BECs, we first confirmed that multiple Nm strains and mutants follow similar phenotypes to previously described models. The recruitment of the recently published pilus adhesin receptor CD147 underneath meningococcal microcolonies could be verified in iPSC-BECs. Nm was also observed to significantly increase the expression of pro-inflammatory and neutrophil-specific chemokines IL6, CXCL1, CXCL2, CXCL8, and CCL20, and the secretion of IFN-γ and RANTES. For the first time, we directly observe that Nm disrupts the three tight junction proteins ZO-1, Occludin, and Claudin-5, which become frayed and/or discontinuous in BECs upon Nm challenge. In accordance with tight junction loss, a sharp loss in trans-endothelial electrical resistance, and an increase in sodium fluorescein permeability and in bacterial transmigration, was observed. Finally, we established RNA-Seq of sorted, infected iPSC-BECs, providing expression data of Nm-responsive host genes. Altogether, this model provides novel insights into Nm pathogenesis, including an impact of Nm on barrier properties and tight junction complexes, and suggests that the paracellular route may contribute to Nm traversal of BECs. KW - Neisseria meningitidis KW - meningococcus KW - bacteria KW - stem cells KW - blood-cerebrospinal fluid barrier KW - blood-brain barrier KW - brain endothelial cells Y1 - 2019 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-201562 VL - 10 IS - 1181 ER - TY - THES A1 - Hagmann, Hanns Antony T1 - The impact of the CRISPR/Cas system on the interaction of Neisseria meningitidis with human host cells T1 - Der Einfluss des CRISPR/Cas-Systems auf die Interaktion von Neisseria meningitidis mit menschlichen Wirtszellen N2 - Neisseria meningitidis, a commensal β-proteobacterium residing exclusively in the human nasopharynx, is a leading cause of sepsis and epidemic meningitis worldwide. While comparative genome analysis was able to define hyperinvasive lineages that are responsible for most of the cases of invasive meningococcal disease (IMD), the genetic basis of their virulence remains unclear. Recent studies demonstrate that the type II C CRISPR/Cas system of meningococci is associated with carriage and less invasive lineages. CRISPR/Cas, an adaptive defence system against foreign DNA, was shown to be involved in gene regulation in Francisella novicida. This study shows that knockout strains of N. meningitidis lacking the Cas9 protein are impaired in the adhesion to human nasopharyngeal cells in a strain-dependant manner, which constitutes a central step in the pathogenesis of IMD. Consequently, this study indicates that the meningococcal CRISPR/Cas system fulfils functions beyond the defence of foreign DNA and is involved in the regulation of meningococcal virulence. N2 - Neisseria meningitidis, ein ß-Proteobakterium, welches als Kommensale ausschließlich den humanen Nasopharynx besiedelt, ist ein weltweit führender Verursacher von Sepsis und epidemischer Meningitis. Auch wenn mittels vergleichender Genomanalysen hyperinvasive Stämme definiert werden konnten, welche für die meisten Fälle von invasiven Meningokokkenerkrankungen verantwortlich sind, bleibt die genetische Grundlage ihrer Virulenz ungeklärt. In vorangegangenen Studien konnte gezeigt werden, dass das Typ II-C CRISPR/Cas-System der Meningokokken assoziiert ist mit Trägerstämmen. CRISPR/Cas ist ein adaptives Verteidigungssystem der Bakterien gegen fremde DNA, das darüber hinaus Aufgaben in der Genregulation von Francisella novicida erfüllt. Diese Arbeit zeigt, dass knockout Stämme von N. meningitidis, denen das Cas9-Protein fehlt, in Abhängigkeit von ihrem genetischen Hintergrund die Fähigkeit verlieren an Zellen des menschlichen Nasopharynx zu adhärieren. Die Adhäsion an den Wirtszellen stellt einen zentralen Schritt in der Pathogenese der invasiven Meningokokkenerkrankungen dar. Die Ergebnisse dieser Arbeit deuten darauf hin, dass das CRISPR/Cas-System in Meningokokken neben seiner Funktion als bakterielles Immunsystem an der Regulation der bakteriellen Virulenz beteiligt sein könnte. KW - CRISPR/Cas-Methode KW - Neisseria meningitidis KW - Bacteria-Host Cell Interaction KW - Regulation of Gene Expression KW - Cas9 KW - Type II-C CRISPR/Cas KW - Adhesion and Invasion Y1 - 2020 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-199490 ER - TY - JOUR A1 - Heidrich, Nadja A1 - Bauriedl, Saskia A1 - Barquist, Lars A1 - Li, Lei A1 - Schoen, Christoph A1 - Vogel, Jörg T1 - The primary transcriptome of Neisseria meningitidis and its interaction with the RNA chaperone Hfq JF - Nucleic Acids Research N2 - Neisseria meningitidis is a human commensal that can also cause life-threatening meningitis and septicemia. Despite growing evidence for RNA-based regulation in meningococci, their transcriptome structure and output of regulatory small RNAs (sRNAs) are incompletely understood. Using dRNA-seq, we have mapped at single-nucleotide resolution the primary transcriptome of N. meningitidis strain 8013. Annotation of 1625 transcriptional start sites defines transcription units for most protein-coding genes but also reveals a paucity of classical σ70-type promoters, suggesting the existence of activators that compensate for the lack of −35 consensus sequences in N. meningitidis. The transcriptome maps also reveal 65 candidate sRNAs, a third of which were validated by northern blot analysis. Immunoprecipitation with the RNA chaperone Hfq drafts an unexpectedly large post-transcriptional regulatory network in this organism, comprising 23 sRNAs and hundreds of potential mRNA targets. Based on this data, using a newly developed gfp reporter system we validate an Hfq-dependent mRNA repression of the putative colonization factor PrpB by the two trans-acting sRNAs RcoF1/2. Our genome-wide RNA compendium will allow for a better understanding of meningococcal transcriptome organization and riboregulation with implications for colonization of the human nasopharynx. KW - RNA KW - Neisseria meningitidis KW - dRNA-seq KW - transcriptome KW - RNA chaperone Hfq Y1 - 2017 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-170828 VL - 45 IS - 10 ER - TY - THES A1 - Gomes, Sara Ferreira Martins T1 - Induced Pluripotent Stem Cell-derived Brain Endothelial Cells as a Cellular Model to Study Neisseria meningitidis Infection T1 - Induziert pluripotente Stammzellen-basierte Hirnendothelzellen als zelluläres Modell zur Untersuchung der Infektion mit Neisseria meningitidis N2 - Bacterial meningitis occurs when blood-borne bacteria are able to penetrate highly specialized brain endothelial cells (BECs) and gain access to the meninges. Neisseria meningitidis (Nm) is a human-exclusive pathogen for which suitable in vitro models are severely lacking. Until recently, modeling BEC-Nm interactions has been almost exclusively limited to immortalized human cells that lack proper BEC phenotypes. Specifically, these in vitro models lack barrier properties, and continuous tight junctions. Alternatively, humanized mice have been used, but these must rely on known interactions and have limited translatability. This motivates the need to establish novel human-based in vitro BEC models that have barrier phenotypes to research Nm-BEC interactions. Recently, a human induced pluripotent stem cell (iPSC) model of BECs has been developed that possesses superior BEC phenotypes and closely mimics the in vivo blood vessels present at the blood-meningeal barrier. Here, iPSC-BECs were tested as a novel cellular model to study Nm-host pathogen interactions, with focus on host responses to Nm infection. Two wild type strains and three mutant strains of Nm were used to confirm that these followed similar phenotypes to previously described models. Importantly, the recruitment of the recently published pilus adhesin receptor CD147 underneath meningococcal microcolonies could be verified in iPSC-BECs. Nm was also observed to significantly increase the expression of pro-inflammatory and neutrophil-specific chemokines IL6, CXCL1, CXCL2, CXCL8, and CCL20, at distinct time points of infection, and the secretion of IFN γ and RANTES by iPSC-BECs. Nm was directly observed to disrupt tight junction proteins ZO-1, Occludin, and Claudin-5 at late time points of infection, which became frayed and/or discontinuous upon infection. This destruction is preceded by, and might be dependent on, SNAI1 activation (a transcriptional repressor of tight junction proteins). In accordance with tight junction loss, a sharp loss in trans-endothelial electrical resistance, and an increase in sodium fluorescein permeability was observed at late infection time points. Notably, bacterial transmigration correlated with junctional disruption, indicating that the paracellular route contributes for bacterial crossing of BECs. Finally, RNA-Sequencing (RNA-Seq) of sorted, infected iPSC-BECs was established through the use of fluorescence-activated cell sorting (FACS) techniques following infection. This allowed the detection of expression data of Nm-responsive host genes not previously described thus far to play a role during meningitidis. In conclusion, here the utility of iPSC-BECs in vitro to study Nm infection could be demonstrated. This is the first BEC in vitro model to express all major BEC tight junctions and to display high barrier potential. Altogether, here this model provides novel insights into Nm pathogenesis, including an impact of Nm on barrier properties and tight junction complexes and suggests that the paracellular route contributes to Nm traversal of BECs. N2 - Eine bakterielle Meningitis tritt auf, wenn durch Blut übertragene Bakterien hochspezialisierte Hirnendothelzellen (BEC) durchdringen und Zugang zu den Meningen erhalten. Neisseria meningitidis (Nm) ist ein human-exklusiver Erreger, für dessen Untersuchung es an geeigneten In-vitro-Modellen mangelt. Bis vor kurzem war die Modellierung von BEC-Nm-Wechselwirkungen fast ausschließlich auf immortalisierte humane Zellen beschränkt, denen wichtige BEC-Phänotypen fehlen. Besonders hervorzuheben sind das Fehlen physiologischer Barriereeigenschaften durch unkontinuierliche dichte Zell-Zell-Verbindungen. Als alternative Modellorganismen können humanisierte Mäuse verwendet werden, die sich jedoch auf bekannte Wirt-Erreger-Wechselwirkungen stützen und durch Speziesunterschiede eine eingeschränkte Übersetzbarkeit aufweisen. Dies begründet die Notwendigkeit, neuartige humane In-vitro-BEC-Modelle zu etablieren, die physiologische Barrierephänotypen aufweisen, um Nm-BEC-Wechselwirkungen zu untersuchen. Kürzlich wurde ein humanes Modell entwickelt, welches auf aus induziert pluripotenten Stammzellen (iPSCs) abgeleiteten humanen BECs basiert und sich durch einen physiologischen Blut-Hirn-Schranken-Phänotyp auszeichnet. Die iPSC-BECs wurden in dieser Arbeit als neuartiges zelluläres Modell getestet, um Nm-Wirt-Pathogen-Wechselwirkungen zu untersuchen, wobei der Schwerpunkt auf Wirtsreaktionen auf Nm-Infektionen lag. Zwei Wildtypstämme und drei Mutantenstämme von Nm wurden verwendet, um zu bestätigen, dass diese ähnlichen Phänotypen wie in zuvor beschriebenen Modellen folgten. Hervorzuheben ist, dass die Rekrutierung des kürzlich veröffentlichten Pilus-Adhäsin-Rezeptors CD147 unter Meningokokken-Mikrokolonien in iPSC-BECs verifiziert werden konnte. Es wurde auch beobachtet, dass Nm die Expression der entzündungsfördernden und neutrophilen spezifischen Chemokine IL6, CXCL1, CXCL2, CXCL8 und CCL20 zu bestimmten Zeitpunkten der Infektion sowie die Sekretion von IFN-γ und RANTES durch iPSC-BECs signifikant erhöht. Es wurde zudem beobachtet, dass Nm die Tight Junction-Proteine ZO-1, Occludin und Claudin-5 zu späten Zeitpunkten der Infektion zerstört, verursacht durch die Infektion wurde ein ausgefranster und/oder diskontinuierlicher Tight Junction-Phänotyp beobachtet. Dieser Zerstörung geht die SNAI1-Aktivierung (ein Transkriptionsrepressor für Tight Junction-Proteine) voraus und könnte von ihr abhängig sein. In Übereinstimmung mit dem Verlust der Tight Junctions wurde zu späten Infektionszeitpunkten ein starker Verlust des transendothelialen elektrischen Widerstands und eine Zunahme der Natriumfluoreszein-Permeabilität beobachtet. Bemerkenswerterweise korrelierte die bakterielle Transmigration mit dem Verlust der Tight Junctions, was darauf hinweist, dass der parazelluläre Weg zur bakteriellen Überwindung von BECs eine entscheidende Rolle spielt. Schließlich wurde die RNA-Sequencing (RNA-Seq) von sortierten, infizierten iPSC-BECs durch die Verwendung von fluoreszenzaktivierten Zellsortiertechniken (FACS) nach der Infektion durchgeführt. Dies ermöglichte erstmalig den Nachweis von Expressionsdaten von Nm-responsiven Wirtsgenen, welche bei der Meningitidis eine Rolle zu spielen scheinen. Zusammenfassend konnte im Rahmen der vorliegenden Arbeit der Nutzen von iPSC-BECs In-Vitro-Modellen zur Untersuchung von Nm-Infektionen gezeigt werden. Dies ist das erste BEC-In-vitro-Modell, das alle wichtigen BEC-Tight Junctions exprimiert und ein hohes Barrierepotential aufweist. Insgesamt liefert das eingesetzte Modell neue Einblicke in die Nm-Pathogenese, einschließlich der Beeinflussung der Barriereeigenschaften und der Tight Junction-Komplexe durch Nm, und gibt erste Hinweise darauf, dass die parazelluläre Route zum Nm-Übertritt von BEC-Barrieren eine entscheidende Rolle spielt. KW - Neisseria meningitidis KW - endothelial cells KW - blood brain barrier KW - blood cerebrospinal fluid barrier KW - cellular model KW - Neisseria meningitidis KW - endothelial cells Y1 - 2019 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-188550 ER - TY - JOUR A1 - Strobel, Lea A1 - Johswich, Kay O. T1 - Anticoagulants impact on innate immune responses and bacterial survival in whole blood models of Neisseria meningitidis infection JF - Scientific Reports N2 - Neisseria meningitidis (meningococcus) causes invasive diseases such as meningitis or septicaemia. Ex vivo infection of human whole blood is a valuable tool to study meningococcal virulence factors and the host innate immune responses. In order to consider effects of cellular mediators, the coagulation cascade must be inhibited to avoid clotting. There is considerable variation in the anticoagulants used among studies of N. meningitidis whole blood infections, featuring citrate, heparin or derivatives of hirudin, a polypeptide from leech saliva. Here, we compare the influence of these three different anticoagulants, and additionally Mg/EGTA, on host innate immune responses as well as on viability of N. meningitidis strains isolated from healthy carriers and disease cases, reflecting different sequence types and capsule phenotypes. We found that the anticoagulants significantly impact on cellular responses and, strain-dependently, also on bacterial survival. Hirudin does not inhibit complement and is therefore superior over the other anticoagulants; indeed hirudin-plasma most closely reflects the characteristics of serum during N. meningitidis infection. We further demonstrate the impact of heparin on complement activation on N. meningitidis and its consequences on meningococcal survival in immune sera, which appears to be independent of the heparin binding antigens Opc and NHBA. KW - infection KW - pathogens KW - Neisseria meningitidis KW - anticoagulants Y1 - 2018 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-176226 VL - 8 IS - 10225 ER - TY - JOUR A1 - Herrmann, Johannes A1 - Muenstermann, Marcel A1 - Strobel, Lea A1 - Schubert-Unkmeir, Alexandra A1 - Woodruff, Trent M. A1 - Gray-Owen, Scott D. A1 - Klos, Andreas A1 - Johswich, Kay O. T1 - Complement C5a receptor 1 exacerbates the pathophysiology of N. meningitidis sepsis and is a potential target for disease treatment JF - mBio N2 - Sepsis caused by Neisseria meningitidis (meningococcus) is a rapidly progressing, life-threatening disease. Because its initial symptoms are rather unspecific, medical attention is often sought too late, i.e., when the systemic inflammatory response is already unleashed. This in turn limits the success of antibiotic treatment. The complement system is generally accepted as the most important innate immune determinant against invasive meningococcal disease since it protects the host through the bactericidal membrane attack complex. However, complement activation concomitantly liberates the C5a peptide, and it remains unclear whether this potent anaphylatoxin contributes to protection and/or drives the rapidly progressing immunopathogenesis associated with meningococcal disease. Here, we dissected the specific contribution of C5a receptor 1 (C5aR1), the canonical receptor for C5a, using a mouse model of meningococcal sepsis. Mice lacking C3 or C5 displayed susceptibility that was enhanced by >1,000-fold or 100-fold, respectively, consistent with the contribution of these components to protection. In clear contrast, C5ar1\(^{-/-}\) mice resisted invasive meningococcal infection and cleared N. meningitidis more rapidly than wild-type (WT) animals. This favorable outcome stemmed from an ameliorated inflammatory cytokine response to N. meningitidis in C5ar1\(^{-/-}\) mice in both in vivo and ex vivo whole-blood infections. In addition, inhibition of C5aR1 signaling without interference with the complement bactericidal activity reduced the inflammatory response also in human whole blood. Enticingly, pharmacologic C5aR1 blockade enhanced mouse survival and lowered meningococcal burden even when the treatment was administered after sepsis induction. Together, our findings demonstrate that C5aR1 drives the pathophysiology associated with meningococcal sepsis and provides a promising target for adjunctive therapy. Importance: The devastating consequences of N. meningitidis sepsis arise due to the rapidly arising and self-propagating inflammatory response that mobilizes antibacterial defenses but also drives the immunopathology associated with meningococcemia. The complement cascade provides innate broad-spectrum protection against infection by directly damaging the envelope of pathogenic microbes through the membrane attack complex and triggers an inflammatory response via the C5a peptide and its receptor C5aR1 aimed at mobilizing cellular effectors of immunity. Here, we consider the potential of separating the bactericidal activities of the complement cascade from its immune activating function to improve outcome of N. meningitidis sepsis. Our findings demonstrate that the specific genetic or pharmacological disruption of C5aR1 rapidly ameliorates disease by suppressing the pathogenic inflammatory response and, surprisingly, allows faster clearance of the bacterial infection. This outcome provides a clear demonstration of the therapeutic benefit of the use of C5aR1-specific inhibitors to improve the outcome of invasive meningococcal disease. KW - C5aR1 KW - whole-blood model KW - Neisseria meningitidis KW - anaphylatoxins KW - complement system KW - inflammation KW - invasive disease KW - mouse model KW - neutrophils KW - sepsis Y1 - 2018 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-175792 VL - 9 IS - 1 ER - TY - JOUR A1 - Klughammer, Johanna A1 - Dittrich, Marcus A1 - Blom, Jochen A1 - Mitesser, Vera A1 - Vogel, Ulrich A1 - Frosch, Matthias A1 - Goesmann, Alexander A1 - Müller, Tobias A1 - Schoen, Christoph T1 - Comparative genome sequencing reveals within-host genetic changes in Neisseria meningitidis during invasive disease JF - PLoS ONE N2 - Some members of the physiological human microbiome occasionally cause life-threatening disease even in immunocompetent individuals. A prime example of such a commensal pathogen is Neisseria meningitidis, which normally resides in the human nasopharynx but is also a leading cause of sepsis and epidemic meningitis. Using N. meningitidis as model organism, we tested the hypothesis that virulence of commensal pathogens is a consequence of within host evolution and selection of invasive variants due to mutations at contingency genes, a mechanism called phase variation. In line with the hypothesis that phase variation evolved as an adaptation to colonize diverse hosts, computational comparisons of all 27 to date completely sequenced and annotated meningococcal genomes retrieved from public databases showed that contingency genes are indeed enriched for genes involved in host interactions. To assess within-host genetic changes in meningococci, we further used ultra-deep whole-genome sequencing of throat-blood strain pairs isolated from four patients suffering from invasive meningococcal disease. We detected up to three mutations per strain pair, affecting predominantly contingency genes involved in type IV pilus biogenesis. However, there was not a single (set) of mutation(s) that could invariably be found in all four pairs of strains. Phenotypic assays further showed that these genetic changes were generally not associated with increased serum resistance, higher fitness in human blood ex vivo or differences in the interaction with human epithelial and endothelial cells in vitro. In conclusion, we hypothesize that virulence of meningococci results from accidental emergence of invasive variants during carriage and without within host evolution of invasive phenotypes during disease progression in vivo. KW - blood KW - comparative genomics KW - throat KW - genetic loci KW - Neisseria meningitidis KW - genomic libraries KW - genome sequencing KW - sequence assembly tools Y1 - 2017 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-159547 VL - 12 IS - 1 ER - TY - THES A1 - Hagmann [geb. Kischkies], Laura Violetta T1 - Stringent response regulation and its impact on ex vivo survival in the commensal pathogen \(Neisseria\) \(meningitidis\) T1 - Regulation der stringenten Kontrolle und ihre Auswirkungen auf das ex vivo Überleben des kommensalen Erregers \(Neisseria\) \(meningitidis\) N2 - Neisseria meningitidis is a commensal bacterium which sometimes causes serious disease in humans. Recent studies in numerous human pathogenic bacteria have shown that the stringent response contributes to bacterial virulence. Therefore, this study analyzed the regulation of the stringent response in meningococci and in particular of RelA as well as its contribution to ex vivo fitness in a strain- and condition- dependent manner by using the carriage strain α522 and the hyperinvasive strain MC58 in different in vitro and ex vivo conditions. Growth experiments revealed that both wild-type strains were almost indistinguishable in their ex vivo phenotypes. However, quantitative real time PCR (qRT-PCR) found differences in the gene expression of relA between both strains. Furthermore, in contrast to the MC58 RelA mutant strain α522 deficient in RelA was unable to survive in human whole blood, although both strains showed the same ex vivo phenotypes in saliva and cerebrospinal fluid. Moreover, strain α522 was depended on a short non-coding AT-rich repeat element (ATRrelA) in the promoter region of relA to survive in human blood. Furthermore, cell culture experiments with human epithelial cells revealed that in both strains the deletion of relA resulted in a significantly decreased invasion rate while not significantly affecting adhesion. In order to better understand the conditional lethality of the relA deletion, computational and experimental analyses were carried out to unravel differences in amino acid biosynthetic pathways between both strains. Whereas strain MC58 is able to synthesize all 20 amino acids, strain α522 has an auxotrophy for cysteine and glutamine. In addition, the in vitro growth experiments found that RelA is required for growth in the absence of external amino acids in both strains. Furthermore, the mutant strain MC58 harboring an ATRrelA in its relA promoter region showed improved growth in minimal medium supplemented with L-cysteine and/or L-glutamine compared to the wild-type strain. Contrary, in strain α522 no differences between the wild-type and the ATRrelA deletion mutant were observed. Together this indicates that ATRrelA interferes with the complex regulatory interplay between the stringent response pathway and L-cysteine as well as L-glutamine metabolism. It further suggests that meningococcal virulence is linked to relA in a strain- and condition- depended manner. In conclusion, this work highlighted the role of the stringent response and of non-coding regulatory elements for bacterial virulence and indicates that virulence might be related to the way how meningococci accomplish growth within the host environments. N2 - Neisseria meningitidis ist ein kommensal lebendes, fakultativ pathogenes Bakterium, welches unter nicht vollständig verstandenen Umständen lebensbedrohliche Krankheitsbilder bei Menschen verursacht. Aktuelle Studien haben gezeigt, dass die stringente Antwort einen Einfluss auf die bakterielle Virulenz haben kann. Aus diesem Grund untersucht diese Arbeit die Regulation der stringenten Antwort, insbesondere die Rolle von RelA, sowie den Einfluss der stringenten Antwort auf die Ex-vivo-Fitness der Meningokokken. Die Ergebnisse wurden für den Trägerstamm α522 und den hyperinvasiven Stamm MC58 erhoben und miteinander verglichen. Wachstumsexperimente zeigten, dass sich beide Wildtyp-Stämme in ihren Ex-vivo-Phänotypen nicht unterscheiden. Jedoch wurden mittels quantitativer Echtzeit-PCR (qRT-PCR) Unterschiede zwischen beiden Stämmen in der Genexpression von relA gefunden. Zudem war die α522 relA Mutante im Gegensatz zu der MC58 relA Mutante nicht in der Lage, in menschlichem Vollblut zu überleben. Allerdings zeigten in Saliva und Liquor beide Stämme den gleichen Phänotyp. Außerdem war der Trägerstamm auf eine kurze, nicht-codierende AT-reiche Region (ATRrelA) in der Promotorregion von relA angewiesen, um im menschlichen Blut überleben zu können. Darüber hinaus zeigten Zellkulturexperimente mit humanen Epithelzellen, dass die Deletion relA die Invasionsrate in beiden Stämmen signifikant verringerte, obwohl die Adhäsionsrate durch die Deletion unbeeinflusst blieb. Um besser verstehen zu können, weshalb die Deletion von relA unter bestimmten Bedingungen letal ist, wurden mit In-silico- und experimentellen Analysen nach Unterschieden in den Aminosäurebiosynthesewegen beider Stämme gesucht. Es zeigte sich, dass Stamm MC58 in der Lage ist alle 20 Aminosäuren zu synthetisieren, während Stamm α522 eine Auxotrophie für Cystein und Glutamin aufweist. Ferner zeigten die In-vitro-Wachstumsversuche, dass RelA bei Aminosäuremangel essentiell für beide Stämme ist. Darüber hinaus zeigte eine MC58 Mutante mit einer ATRrelA –Kopie in der relA Promotorregion ein im Vergleich zum Wildtyp-Stamm verbessertes Wachstum in mit L-Cystein und/oder L-Glutamin angereichertem Minimalmedium. Gegensätzlich dazu zeigte der Stamm α522 keine Unterschiede im Wachstum zwischen dem Wildtyp und einer ATRrelA Deletions-Mutante. Dies deutet darauf hin, dass ATRrelA an dem komplexen regulatorischen Zusammenspiel der stringenten Antwort und dem L-Cystein- beziehungsweise dem L-Glutamin-Metabolismus beteiligt ist. Es lässt sich vermuten, dass RelA zu der Virulenz von Meningokokken in einer stamm- und umgebungsspezifischen Weise beiträgt. Abschließend hebt diese Arbeit die Rolle von kleinen regulatorischen Elementen für die bakterielle Virulenz hervor und postuliert, dass die Virulenz der Meningokokken auf ihrer Fähigkeit basiert, sich der durch den Wirt gegebenen Umgebung anzupassen. KW - Neisseria meningitidis KW - Stringente Kontrolle KW - Virulenzfaktor KW - Genregulation KW - Transposon KW - Stringent response KW - RelA KW - MITE KW - Stringente Antwort Y1 - 2016 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-144352 ER - TY - JOUR A1 - Schoen, Christoph A1 - Kischkies, Laura A1 - Elias, Johannes A1 - Ampattu, Biju Joseph T1 - Metabolism and virulence in Neisseria meningitidis N2 - A longstanding question in infection biology addresses the genetic basis for invasive behavior in commensal pathogens. A prime example for such a pathogen is Neisseria meningitidis. On the one hand it is a harmless commensal bacterium exquisitely adapted to humans, and on the other hand it sometimes behaves like a ferocious pathogen causing potentially lethal disease such as sepsis and acute bacterial meningitis. Despite the lack of a classical repertoire of virulence genes in N. meningitidis separating commensal from invasive strains, molecular epidemiology suggests that carriage and invasive strains belong to genetically distinct populations. In recent years, it has become increasingly clear that metabolic adaptation enables meningococci to exploit host resources, supporting the concept of nutritional virulence as a crucial determinant of invasive capability. Here, we discuss the contribution of core metabolic pathways in the context of colonization and invasion with special emphasis on results from genome-wide surveys. The metabolism of lactate, the oxidative stress response, and, in particular, glutathione metabolism as well as the denitrification pathway provide examples of how meningococcal metabolism is intimately linked to pathogenesis. We further discuss evidence from genome-wide approaches regarding potential metabolic differences between strains from hyperinvasive and carriage lineages and present new data assessing in vitro growth differences of strains from these two populations. We hypothesize that strains from carriage and hyperinvasive lineages differ in the expression of regulatory genes involved particularly in stress responses and amino acid metabolism under infection conditions. KW - Neisseria meningitidis KW - virulence KW - pathometabolism KW - oxidative stress KW - glutathione KW - γ-glutamyl cycle KW - glutamate dehydrogenase KW - nitrite respiration Y1 - 2014 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-113118 ER - TY - JOUR A1 - Schubert-Unkmeir, Alexandra A1 - Schneider-Schaulies, Sibylle A1 - Gulbins, Erich A1 - Hebling, Sabrina A1 - Simonis, Alexander T1 - Differential Activation of Acid Sphingomyelinase and Ceramide Release Determines Invasiveness of Neisseria meningitidis into Brain Endothelial Cells N2 - The interaction with brain endothelial cells is central to the pathogenicity of Neisseria meningitidis infections. Here, we show that N. meningitidis causes transient activation of acid sphingomyelinase (ASM) followed by ceramide release in brain endothelial cells. In response to N. meningitidis infection, ASM and ceramide are displayed at the outer leaflet of the cell membrane and condense into large membrane platforms which also concentrate the ErbB2 receptor. The outer membrane protein Opc and phosphatidylcholine-specific phospholipase C that is activated upon binding of the pathogen to heparan sulfate proteoglycans, are required for N. meningitidis-mediated ASM activation. Pharmacologic or genetic ablation of ASM abrogated meningococcal internalization without affecting bacterial adherence. In accordance, the restricted invasiveness of a defined set of pathogenic isolates of the ST-11/ST-8 clonal complex into brain endothelial cells directly correlated with their restricted ability to induce ASM and ceramide release. In conclusion, ASM activation and ceramide release are essential for internalization of Opc-expressing meningococci into brain endothelial cells, and this segregates with invasiveness of N. meningitidis strains. Author Summary Neisseria meningitidis, an obligate human pathogen, is a causative agent of septicemia and meningitis worldwide. Meningococcal infection manifests in a variety of forms, including meningitis, meningococcemia with meningitis or meningococcemia without obvious meningitis. The interaction of N. meningitidis with human cells lining the blood vessels of the blood-cerebrospinal fluid barrier is a prerequisite for the development of meningitis. As a major pathogenicity factor, the meningococcal outer membrane protein Opc enhances bacterial entry into brain endothelial cells, however, mechanisms underlying trapping of receptors and signaling molecules following this interaction remained elusive. We now show that Opc-expressing meningococci activate acid sphingomyelinase (ASM) in brain endothelial cells, which hydrolyses sphingomyelin to cause ceramide release and formation of extended ceramide-enriched membrane platforms wherein ErbB2, an important receptor involved in bacterial uptake, clusters. Mechanistically, ASM activation relied on binding of N. meningitidis to its attachment receptor, HSPG, followed by activation of PC-PLC. Meningococcal isolates of the ST-11 clonal complex, which are reported to be more likely to cause severe sepsis, but rarely meningitis, barely invaded brain endothelial cells and revealed a highly restricted ability to induce ASM and ceramide release. Thus, our results unravel a differential activation of the ASM/ceramide system by the species N. meningitidis determining its invasiveness into brain endothelial cells. KW - small interfering RNAs KW - Neisseria meningitidis KW - bacterial pathogens KW - endothelial cells KW - meningococcal disease KW - flow cytometry KW - cell staining KW - Escherichia coli infections Y1 - 2014 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-113031 ER -