TY - JOUR A1 - Yang, Tao A1 - Heydarian, Motaharehsadat A1 - Kozjak-Pavlovic, Vera A1 - Urban, Manuela A1 - Harbottle, Richard P. A1 - Rudel, Thomas T1 - Folliculin Controls the Intracellular Survival and Trans-Epithelial Passage of Neisseria gonorrhoeae JF - Frontiers in Cellular and Infection Microbiology N2 - Neisseria gonorrhoeae, a Gram-negative obligate human pathogenic bacterium, infects human epithelial cells and causes sexually transmitted diseases. Emerging multi-antibiotic resistant gonococci and increasing numbers of infections complicate the treatment of infected patients. Here, we used an shRNA library screen and next-generation sequencing to identify factors involved in epithelial cell infection. Folliculin (FLCN), a 64 kDa protein with a tumor repressor function was identified as a novel host factor important for N. gonorrhoeae survival after uptake. We further determined that FLCN did not affect N. gonorrhoeae adherence and invasion but was essential for its survival in the cells by modulating autophagy. In addition, FLCN was also required to maintain cell to cell contacts in the epithelial layer. In an infection model with polarized cells, FLCN inhibited the polarized localization of E-cadherin and the transcytosis of gonococci across polarized epithelial cells. In conclusion, we demonstrate here the connection between FLCN and bacterial infection and in particular the role of FLCN in the intracellular survival and transcytosis of gonococci across polarized epithelial cell layers. KW - gonococcal invasion KW - folliculin KW - autophagy KW - polarized epithelium KW - polarized cell culture Y1 - 2020 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-211372 SN - 2235-2988 VL - 10 IS - 422 ER - TY - THES A1 - Yang, Tao T1 - Functional insights into the role of a bacterial virulence factor and a host factor in Neisseria gonorrhoeae infection T1 - Funktionelle Einblicke in die Rolle eines bakteriellen Virulenzfaktors und eines Wirtsfaktors bei der Infektion mit Neisseria gonorrhoeae N2 - Neisseria gonorrhoeae (GC) is a human specific pathogenic bacterium. Currently, N. gonorrhoeae developed resistance to virtually all the available antibiotics used for treatment. N. gonorrhoeae starts infection by colonizing the cell surface, followed by invasion of the host cell, intracellular persistence, transcytosis and exit into the subepithelial space. Subepithelial bacteria can reach the bloodstream and disseminate to other tissues causing systemic infections, which leads to serious conditions such as arthritis and pneumonia. A number of studies have well established the host-pathogen interactions during the initial adherence and invasion steps. However, the mechanism of intracellular survival and traversal is poorly understood so far. Hence, identification of novel bacterial virulence factors and host factors involved in the host-pathogen interaction is a crucial step in understanding disease development and uncovering novel therapeutic approaches. Besides, most of the previous studies about N. gonorrhoeae were performed in the conventional cell culture. Although they have provided insights into host-pathogen interactions, much information about the native infection microenvironment, such as cell polarization and barrier function, is still missing. This work focused on determining the function of novel bacterial virulence factor NGFG_01605 and host factor (FLCN) in gonococcal infection. NGFG_01605 was identified by Tn5 transposon library screening. It is a putative U32 protease. Unlike other proteins in this family, it is not secreted and has no ex vivo protease activity. NGFG_01605 knockout decreases gonococcal survival in the epithelial cell. 3D models based on T84 cell was developed for the bacterial transmigration assay. NGFG_01605 knockout does not affect gonococcal transmigration. The novel host factor FLCN was identified by shRNA library screening in search for factors that affected gonococcal adherence and/or internalization. We discovered that FLCN did not affect N. gonorrhoeae adherence and invasion but was essential for bacterial survival. Since programmed cell death is a host defence mechanism against intracellular pathogens, we further explored apoptosis and autophagy upon gonococcal infection and determined that FLCN did not affect apoptosis but inhibited autophagy. Moreover, we found that FLCN inhibited the expression of E-cadherin. Knockdown of E- cadherin decreased the autophagy flux and supported N. gonorrhoeae survival. Both non-polarized and polarized cells are present in the cervix, and additionally, E-cadherin represents different polarization properties on these different cells. Therefore, we established 3-D models to better understand the functions of FLCN. We discovered that FLCN was critical for N. gonorrhoeae survival in the 3-D environment as well, but not through inhibiting autophagy. Furthermore, FLCN inhibits the E-cadherin expression and disturbs its polarization in the 3-D models. Since N. gonorrhoeae can cross the epithelial cell barriers through both cell-cell junctions and transcellular migration, we further explored the roles FLCN and E-cadherin played in transmigration. FLCN delayed N. gonorrhoeae transmigration, whereas the knockdown of E-cadherin increased N. gonorrhoeae transmigration. In summary, we revealed roles of the NGFG_01605 and FLCN-E-cadherin axis play in N. gonorrhoeae infection, particularly in relation to intracellular survival and transmigration. This is also the first study that connects FLCN and human-specific pathogen infection. N2 - Neisseria gonorrhoeae (GC) ist ein humanpathogenes Bakterium. Gegen jedes kommerziell erhältliche Antibiotikum konnten bereits Resistenzen entdeckt werden. Die Infektion von Neisserien startet mit der Kolonisierung der Zelloberfläche, gefolgt von der Invasion in die Zelle, intrazellulärer Persistenz, Transzytose und Verlassen des subepithelen Raums. Subepithele Bakterien können den Blutfluss erreichen und sich somit auf andere Gewebe verbreiten und dadurch schwerwiegende Erkrankungen wie Arthritis und Lungenentzündungen verursachen. Zahlreiche Studien haben die Interaktion zwischen Wirtszelle und Pathogen zwischen Adhärenz und Invasion gut charakterisiert. Allerdings ist der Mechanismus des intrazellulären Überlebens und der Transmigration weitestgehend unbekannt. Um neue therapeutische Ansätze zu definieren ist es deshalb wichtig weitere bakterielle und zelluläre Faktoren verantwortlich für Interaktionen zwischen Wirt und Pathogen zu identifizieren. Außerdem wurden Studien über N. gonorrhoeae bisher in konventioneller Zellkultur durchgeführt. Auch wenn diese weitere Einsichten in Wirt-Pathogen Interaktionen geben, ist viel Information in der nativen Infektionsumgebung, wie Zellpolarisierung oder Barrierefunktionen, bisher nicht bekannt. Diese Arbeit fokussiert sich auf die Bestimmung eines neuen bakteriellen Virulenzfaktors, NGFG_01605, und Wirtsfaktoren (FLCN) in der Infektion von Gonokokken. NGFG_01605 wurde über Screening einer Transposonsammlung identifiziert und ist eine vermeintliche U32 Protease. Im Gegensatz zu anderen Proteinen dieser Familie, wird diese Protease nicht sekretiert und hat keine ex vivo Proteaseaktivität. Der Knockout von NGFG_01605 vermindert die Überlebensrate von Gonokokken in Epithelzellen. 3D-Modelle basierend auf T84-Zellen wurden für die Analyse der bakteriellen Transmigration entwickelt. Der Knockout von NGFG_01605 hat keinen Einfluss auf die Transmigration. Bei der Suche neuer Wirtsfaktoren, die für die Adhärenz und/oder Internalisierung wichtig sind, wurde FLCN durch ein Screening einer shRNA Sammlung entdeckt. Wir konnten zeigen, dass dieser Faktor zwar nicht für die Adhärenz oder Invasion von N. gonorrhoeae, aber für das Überleben der Bakterien in der Wirtszelle essenziell ist. Da der programmierte Zelltod ein typischer Abwehrmechanismus gegen intrazelluläre Bakterien ist, haben wir Apoptose und Autophagie weiter untersucht und konnten zeigen, dass FLCN zwar keinen Effekt auf Apoptose hat, aber Autophagie inhibiert. Außerdem konnten wir zeigen, dass FLCN die Expression von E-cadherin inhibiert. Auch der Knockdown von E-cadherin verringerte Autophagie und erhöhte das Überleben von N. gonorrhoeae. Im Gebärmutterhals sind sowohl polarisierte als auch nicht-polarisierte Zellen präsent. E-cadherin hat zudem unterschiedliche Einflüsse auf diese unterschiedlichen Zellen. Aus diesem Grund haben wir ein 3D-Modell entwickelt, um die Funktionen von FLCN besser zu verstehen. Wir entdeckten, dass FLCN zwar auch im 3D Modell wichtig für das Überleben ist, aber nicht durch Inhibition von Autophagie. Außerdem inhibiert FLCN die Expression und Verteilung von E-cadherin in 3D-Modellen. Da N. gonorrhoeae die Epithelzellbarierre durch sowohl Zell-Zell-Kontakte als auch transzelluläre Migration überwinden kann, untersuchten wir daraufhin die Rollen von FLCN und E-cadherin in der transzellulären Migration. Diese wird durch FLCN verspätet und durch Knockdown von E-cadherin erhöht. Zusammengefasst, haben wir die Rolle von NGFG_01605 und den Zusammenhang von FLCN und E-cadherin während der Infektion von N. gonorrhoeae untersucht. Unser Fokus war hierbei das intrazelluläre Überleben sowie zelluläre Transmigration. Diese Arbeit ist die Erste, die FLCN und humanpathogenspezifische Infektion miteinander in Verbindung bringt. KW - Folliculin KW - autophagy KW - polarized epithelium KW - protease KW - Gonococcal invasion KW - autophagocytose Y1 - 2021 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-208959 ER - TY - JOUR A1 - Trivanovic, Drenka A1 - Volkmann, Noah A1 - Stoeckl, Magdalena A1 - Tertel, Tobias A1 - Rudert, Maximilian A1 - Giebel, Bernd A1 - Herrmann, Marietta T1 - Enhancement of immunosuppressive activity of mesenchymal stromal cells by platelet-derived factors is accompanied by apoptotic priming JF - Stem Cell Reviews and Reports N2 - The pro-inflammatory phase of bone healing, initiated by platelet activation and eventually hematoma formation, impacts bone marrow mesenchymal stromal cells (MSCs) in unknown ways. Here, we created platelet-rich plasma (PRP) hydrogels to study how platelet-derived factors modulate functional properties of encapsulated MSCs in comparison to a non-inflammatory fibrin (FBR) hydrogel environment. MSCs were isolated from human bone marrow, while PRP was collected from pooled apheresis thrombocyte concentrates and used for hydrogel preparation. After their encapsulation in hydrogels for 72 h, retrieved MSCs were analyzed for immunomodulatory activities, apoptosis, stem cell properties, senescence, CD9\(^+\), CD63\(^+\) and CD81\(^+\) extracellular vesicle (EV) release, and metabolism-related changes. PRP-hydrogels stimulated immunosuppressive functions of MSCs, along with their upregulated susceptibility to cell death in communication with PBMCs and augmented caspase 3/7 activity. We found impaired clonal growth and cell cycle progression, and more pronounced β-galactosidase activity as well as accumulation of LC3-II-positive vacuoles in PRP-MSCs. Stimuli derived from PRP-hydrogels upregulated AKT and reduced mTOR phosphorylation in MSCs, which suggests an initiation of survival-related processes. Our results showed that PRP-hydrogels might represent a metabolically stressful environment, inducing acidification of MSCs, reducing polarization of the mitochondrial membrane and increasing lipid accumulation. These features were not detected in FBR-MSCs, which showed reduced CD63\(^+\) and CD81\(^+\) EV production and maintained clonogenicity. Our data revealed that PRP-derived hematoma components cause metabolic adaptation of MSCs followed by increased immune regulatory functions. For the first time, we showed that PRP stimuli represent a survival challenge and “apoptotic priming” that are detrimental for stem cell-like growth of MSCs and important for their therapeutic consideration. KW - hematoma KW - platelet-rich plasma KW - fibrin KW - mesenchymal stromal cells KW - immunomodulation KW - apoptosis KW - autophagy KW - senescence KW - extracellular vesicles KW - metabolism Y1 - 2023 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-324669 VL - 19 IS - 3 ER - TY - JOUR A1 - Tolay, Nazife A1 - Buchberger, Alexander T1 - Comparative profiling of stress granule clearance reveals differential contributions of the ubiquitin system JF - Life Science Alliance N2 - Stress granules (SGs) are cytoplasmic condensates containing untranslated mRNP complexes. They are induced by various proteotoxic conditions such as heat, oxidative, and osmotic stress. SGs are believed to protect mRNPs from degradation and to enable cells to rapidly resume translation when stress conditions subside. SG dynamics are controlled by various posttranslationalmodifications, but the role of the ubiquitin system has remained controversial. Here, we present a comparative analysis addressing the involvement of the ubiquitin system in SG clearance. Using high-resolution immuno-fluorescence microscopy, we found that ubiquitin associated to varying extent with SGs induced by heat, arsenite, H2O2, sorbitol, or combined puromycin and Hsp70 inhibitor treatment. SG-associated ubiquitin species included K48- and K63-linked conjugates, whereas free ubiquitin was not significantly enriched. Inhibition of the ubiquitin activating enzyme, deubiquitylating enzymes, the 26S proteasome and p97/VCP impaired the clearance of arsenite- and heat-induced SGs, whereas SGs induced by other stress conditions were little affected. Our data underline the differential involvement of the ubiquitin system in SG clearance, a process important to prevent the formation of disease-linked aberrant SGs. KW - phase transition KW - quality control KW - protein KW - inhibition KW - complexity KW - separation KW - diversity KW - autophagy KW - ALS KW - P97 Y1 - 2021 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-259810 VL - 4 IS - 5 ER - TY - JOUR A1 - Stojanović, Stevan D. A1 - Fuchs, Maximilian A1 - Fiedler, Jan A1 - Xiao, Ke A1 - Meinecke, Anna A1 - Just, Annette A1 - Pich, Andreas A1 - Thum, Thomas A1 - Kunz, Meik T1 - Comprehensive bioinformatics identifies key microRNA players in ATG7-deficient lung fibroblasts JF - International Journal of Molecular Sciences N2 - Background: Deficient autophagy has been recently implicated as a driver of pulmonary fibrosis, yet bioinformatics approaches to study this cellular process are lacking. Autophagy-related 5 and 7 (ATG5/ATG7) are critical elements of macro-autophagy. However, an alternative ATG5/ATG7-independent macro-autophagy pathway was recently discovered, its regulation being unknown. Using a bioinformatics proteome profiling analysis of ATG7-deficient human fibroblasts, we aimed to identify key microRNA (miR) regulators in autophagy. Method: We have generated ATG7-knockout MRC-5 fibroblasts and performed mass spectrometry to generate a large-scale proteomics dataset. We further quantified the interactions between various proteins combining bioinformatics molecular network reconstruction and functional enrichment analysis. The predicted key regulatory miRs were validated via quantitative polymerase chain reaction. Results: The functional enrichment analysis of the 26 deregulated proteins showed decreased cellular trafficking, increased mitophagy and senescence as the major overarching processes in ATG7-deficient lung fibroblasts. The 26 proteins reconstitute a protein interactome of 46 nodes and miR-regulated interactome of 834 nodes. The miR network shows three functional cluster modules around miR-16-5p, miR-17-5p and let-7a-5p related to multiple deregulated proteins. Confirming these results in a biological setting, serially passaged wild-type and autophagy-deficient fibroblasts displayed senescence-dependent expression profiles of miR-16-5p and miR-17-5p. Conclusions: We have developed a bioinformatics proteome profiling approach that successfully identifies biologically relevant miR regulators from a proteomics dataset of the ATG-7-deficient milieu in lung fibroblasts, and thus may be used to elucidate key molecular players in complex fibrotic pathological processes. The approach is not limited to a specific cell-type and disease, thus highlighting its high relevance in proteome and non-coding RNA research. KW - bioinformatics KW - miR KW - proteomics KW - functional network analysis KW - senescence KW - lung fibrosis KW - autophagy Y1 - 2020 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-285181 SN - 1422-0067 VL - 21 IS - 11 ER - TY - JOUR A1 - Siegmund, Daniela A1 - Wagner, Jennifer A1 - Wajant, Harald T1 - TNF receptor associated factor 2 (TRAF2) signaling in cancer JF - Cancers N2 - Tumor necrosis factor (TNF) receptor associated factor-2 (TRAF2) has been originally identified as a protein interacting with TNF receptor 2 (TNFR2) but also binds to several other receptors of the TNF receptor superfamily (TNFRSF). TRAF2, often in concert with other members of the TRAF protein family, is involved in the activation of the classical NFκB pathway and the stimulation of various mitogen-activated protein (MAP) kinase cascades by TNFRSF receptors (TNFRs), but is also required to inhibit the alternative NFκB pathway. TRAF2 has also been implicated in endoplasmic reticulum (ER) stress signaling, the regulation of autophagy, and the control of cell death programs. TRAF2 fulfills its functions by acting as a scaffold, bringing together the E3 ligase cellular inhibitor of apoptosis-1 (cIAP1) and cIAP2 with their substrates and various regulatory proteins, e.g., deubiquitinases. Furthermore, TRAF2 can act as an E3 ligase by help of its N-terminal really interesting new gene (RING) domain. The finding that TRAF2 (but also several other members of the TRAF family) interacts with the latent membrane protein 1 (LMP1) oncogene of the Epstein–Barr virus (EBV) indicated early on that TRAF2 could play a role in the oncogenesis of B-cell malignancies and EBV-associated non-keratinizing nasopharyngeal carcinoma (NPC). TRAF2 can also act as an oncogene in solid tumors, e.g., in colon cancer by promoting Wnt/β-catenin signaling. Moreover, tumor cell-expressed TRAF2 has been identified as a major factor-limiting cancer cell killing by cytotoxic T-cells after immune checkpoint blockade. However, TRAF2 can also be context-dependent as a tumor suppressor, presumably by virtue of its inhibitory effect on the alternative NFκB pathway. For example, inactivating mutations of TRAF2 have been associated with tumor development, e.g., in multiple myeloma and mantle cell lymphoma. In this review, we summarize the various TRAF2-related signaling pathways and their relevance for the oncogenic and tumor suppressive activities of TRAF2. Particularly, we discuss currently emerging concepts to target TRAF2 for therapeutic purposes. KW - apoptosis KW - autophagy KW - B-cell lymphoma KW - cellular inhibitor of apoptosis 1/2 (cIAP1/2) KW - necroptosis KW - nuclear factor ‘kappa-light-chain-enhancer’ of activated B-cells (NFκB) KW - tumor necrosis factor (TNF) KW - TNF receptor associated factor 2 (TRAF2) Y1 - 2022 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-286073 SN - 2072-6694 VL - 14 IS - 16 ER - TY - JOUR A1 - Scherzad, Agmal A1 - Meyer, Till A1 - Kleinsasser, Norbert A1 - Hackenberg, Stephan T1 - Molecular Mechanisms of Zinc Oxide Nanoparticle-Induced Genotoxicity Short Running Title: Genotoxicity of ZnO NPs JF - Materials N2 - Background: Zinc oxide nanoparticles (ZnO NPs) are among the most frequently applied nanomaterials in consumer products. Evidence exists regarding the cytotoxic effects of ZnO NPs in mammalian cells; however, knowledge about the potential genotoxicity of ZnO NPs is rare, and results presented in the current literature are inconsistent. Objectives: The aim of this review is to summarize the existing data regarding the DNA damage that ZnO NPs induce, and focus on the possible molecular mechanisms underlying genotoxic events. Methods: Electronic literature databases were systematically searched for studies that report on the genotoxicity of ZnO NPs. Results: Several methods and different endpoints demonstrate the genotoxic potential of ZnO NPs. Most publications describe in vitro assessments of the oxidative DNA damage triggered by dissoluted Zn2+ ions. Most genotoxicological investigations of ZnO NPs address acute exposure situations. Conclusion: Existing evidence indicates that ZnO NPs possibly have the potential to damage DNA. However, there is a lack of long-term exposure experiments that clarify the intracellular bioaccumulation of ZnO NPs and the possible mechanisms of DNA repair and cell survival. KW - zinc oxide nanoparticles KW - genotoxicity KW - DNA damage KW - ROS KW - autophagy Y1 - 2017 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-169948 VL - 10 IS - 12 ER - TY - THES A1 - Riedel, Alexander T1 - Untersuchungen zur endogenen MHC-Klasse-II-restringierten Präsentation nukleärer Antigene T1 - Investigation of the endogenous MHC class II-restricted presentation of nuclear antigens N2 - Die endogene Präsentation von intrazellulären Antigenen auf Major-Histokompatibilitätskomplex Klasse-II (MHC-II) -Molekülen ist von entscheidender Bedeutung für eine Reihe von immunologischen Prozessen. Die mechanistischen Grundlagen dieses Präsentationsweges sind aber noch weitgehend unverstanden. Ziel dieser Arbeit war es, einen Beitrag zum molekularen Verständnis der Abläufe zu leisten, die an der endogenen Präsentation nukleärer Antigene auf MHC-II-Molekülen beteiligt sind. Dazu sollte am Beispiel des nukleär lokalisierten Modellantigens Neomycin-Phosphotransferase II (NucNeoR) sowie des viralen Kernantigens Epstein-Barr-virus nuclear antigen 3C (EBNA3C) und entsprechender antigenspezifischer MHC-II-restringierter CD4+ T-Zellen die verantwortlichen Präsentationswege in professionell und nicht-professionell antigenpräsentierenden Zellen untersucht werden. In beiden Zellsystemen wurde NucNeoR über einen endogenen Präsentationsweg und nicht über die Freisetzung und Wiederaufnahme als exogenes Protein auf MHC-II-Molekülen präsentiert. Durch die Verwendung chemischer Inhibitoren konnte eine Beteiligung der Autophagie an der endogenen Antigenpräsentation nachgewiesen werden. Da Autophagie ausschließlich im Zytoplasma stattfindet, wurde nach möglichen Eintrittspforten für nukleäre Proteine in diesen Abbauweg gesucht. Für die Autophagie-abhängige Präsentation von NucNeoR war weder ein CRM1-vermittelter aktiver Export des Antigens aus dem Kern ins Zytoplasma, noch eine Auflösung der Kernmembran im Rahmen der Zellteilung und der dadurch bedingten Durchmischung nukleärer und zytoplasmatischer Bestandteile notwendig. Mit Hilfe eines konditionalen Antigenexpressionsystems und der Auftrennung antigenexprimierender Zellen nach Zellzyklusphasen konnte eine verstärkte Antigenpräsentation in der G1/0-Phase nachgewiesen werden, die mit fortschreitendem Zellzyklus immer mehr abnahm. Die Antigenpräsentation korrelierte dabei mit der ebenfalls im Laufe des Zellzyklus abnehmenden Transkriptions- bzw. Translationsrate des Antigens, aber nicht mit der absoluten Menge an Antigen in den Zellen. Bei abgeschalteter Antigentranskription dagegen korrelierte die Antigenpräsentation mit der MHC-II-Oberflächenexpression, die von der G1/0- bis hin zur G2/M-Phase kontinuierlich zunahm. Eine ähnliche Korrelation von Antigentranskription/ Antigentranslation und Autophagie-abhängiger Antigenpräsentation wurde auch für EBNA3C und die zytoplasmatisch lokalisierte NeoR-Variante beobachtet. Diese Ergebnisse identifizieren die Autophagie-abhängige Präsentation neusynthetisierter Proteine als den verantwortlichen molekularen Mechanismus für die endogene Präsentation der untersuchten nukleären Antigene auf MHC-II-Molekülen. Durch die Kopplung von Translation und autophagischem Abbau erlangen Proteine unabhängig von ihrer subzellulären Lokalisation Zugang zu diesem Präsentationsweg und erweitern so das Spektrum der intrazellulären Antigene, die einer CD4+ T-Zellüberwachung unterliegen. N2 - The endogenous presentation of intracellular antigens on major histocompatibility complex class II (MHC-II) molecules plays an important role in adaptive immune responses, but the underlying molecular mechanisms are not well understood. The aim of this study was to gain insight into the endogenous presentation pathways for nuclear antigens on MHC-II molecules. By using antigen-specific CD4+ T cell clones, MHC-II presentation of the nuclear antigens neomycin phosphotransferase II (NucNeoR) and Epstein-Barr virus nuclear antigen 3C (EBNA3C) was studied in professional and non-professional antigen presenting cells (APC). Both types of APC presented peptides derived from these nuclear proteins on MHC-II molecules by an endogenous presentation pathway and not by release and reuptake as exogenous protein. Inhibition of autophagy drastically reduced endogenous antigen presentation, indicating that nuclear proteins enter the MHC-II processing and loading compartment through autophagic vesicles. Because autophagocytosis occurs in the cytoplasm, potential entry routes for nuclear proteins into this pathway were investigated. Endogenous presentation of NucNeoR on MHC-II molecules by autophagy did neither involve the CRM1-mediated export of the nuclear protein into the cytoplasm, nor the redistribution of nuclear and cytoplasmic components following the dissolution of the nuclear envelope during mitosis. Conditional antigen expression and cell cycle phase separation of antigen-expressing cells revealed that antigen presentation was maximal in cells in G1/0 and then gradually decreased as cells progressed in the cell cycle. This reduction in antigen presentation correlated with a cell cycle-dependent decrease in antigen transcription/translation, but not with the total amount of antigen present in these cells. By contrast, when antigen expression was turned off, antigen presentation correlated with MHC-II surface expression, which increased from G1/0- to G2/M-phase. A similar correlation between antigen presentation and antigen transcription/translation was also observed for the antigens EBNA3C and the cytosolic variant of neomycin phosphotransferase II (NeoR). These results identify autophagocytosis of newly-synthesized proteins as the molecular mechanism mediating endogenous presentation of nuclear and cytosolic antigens on MHC-II molecules. By coupling protein translation and autophagocytosis, newly-synthesized proteins gain access to the endogenous MHC-II presentation pathway irrespective of their subcellular localisation and thereby broaden the spectrum of intracellular antigens that are presented to CD4+ T cells. KW - Autophagie KW - Antigenpräsentation KW - MHC KW - nukleäre Antigene KW - T-Zellen KW - autophagy KW - antigen presentation KW - MHC KW - nuclear antigen KW - T Lymphocytes Y1 - 2007 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-25183 ER - TY - JOUR A1 - Otto, Christoph A1 - Hahlbrock, Theresa A1 - Eich, Kilian A1 - Karaaslan, Ferdi A1 - Jürgens, Constantin A1 - Germer, Christoph-Thomas A1 - Wiegering, Armin A1 - Kämmerer, Ulrike T1 - Antiproliferative and antimetabolic effects behind the anticancer property of fermented wheat germ extract JF - BMC Complementary and Alternative Medicine N2 - Background Fermented wheat germ extract (FWGE) sold under the trade name Avemar exhibits anticancer activity in vitro and in vivo. Its mechanisms of action are divided into antiproliferative and antimetabolic effects. Its influcence on cancer cell metabolism needs further investigation. One objective of this study, therefore, was to further elucidate the antimetabolic action of FWGE. The anticancer compound 2,6-dimethoxy-1,4-benzoquinone (DMBQ) is the major bioactive compound in FWGE and is probably responsible for its anticancer activity. The second objective of this study was to compare the antiproliferative properties in vitro of FWGE and the DMBQ compound. Methods The IC\(_{50}\) values of FWGE were determined for nine human cancer cell lines after 24 h of culture. The DMBQ compound was used at a concentration of 24 μmol/l, which is equal to the molar concentration of DMBQ in FWGE. Cell viability, cell cycle, cellular redox state, glucose consumption, lactic acid production, cellular ATP levels, and the NADH/NAD\(^+\) ratio were measured. Results The mean IC\(_{50}\) value of FWGE for the nine human cancer cell lines tested was 10 mg/ml. Both FWGE (10 mg/ml) and the DMBQ compound (24 μmol/l) induced massive cell damage within 24 h after starting treatment, with changes in the cellular redox state secondary to formation of intracellular reactive oxygen species. Unlike the DMBQ compound, which was only cytotoxic, FWGE exhibited cytostatic and growth delay effects in addition to cytotoxicity. Both cytostatic and growth delay effects were linked to impaired glucose utilization which influenced the cell cycle, cellular ATP levels, and the NADH/NAD\(^+\) ratio. The growth delay effect in response to FWGE treatment led to induction of autophagy. Conclusions FWGE and the DMBQ compound both induced oxidative stress-promoted cytotoxicity. In addition, FWGE exhibited cytostatic and growth delay effects associated with impaired glucose utilization which led to autophagy, a possible previously unknown mechanism behind the influence of FWGE on cancer cell metabolism. KW - cytostatic KW - FWGE KW - benzoquinone KW - cancer cells KW - reactive oxygen species KW - autophagy KW - cytotoxicity Y1 - 2016 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-146013 VL - 16 IS - 160 ER - TY - JOUR A1 - Oelschlaegel, Diana A1 - Weiss Sadan, Tommy A1 - Salpeter, Seth A1 - Krug, Sebastian A1 - Blum, Galia A1 - Schmitz, Werner A1 - Schulze, Almut A1 - Michl, Patrick T1 - Cathepsin inhibition modulates metabolism and polarization of tumor-associated macrophages JF - Cancers N2 - Stroma-infiltrating immune cells, such as tumor-associated macrophages (TAM), play an important role in regulating tumor progression and chemoresistance. These effects are mostly conveyed by secreted mediators, among them several cathepsin proteases. In addition, increasing evidence suggests that stroma-infiltrating immune cells are able to induce profound metabolic changes within the tumor microenvironment. In this study, we aimed to characterize the impact of cathepsins in maintaining the TAM phenotype in more detail. For this purpose, we investigated the molecular effects of pharmacological cathepsin inhibition on the viability and polarization of human primary macrophages as well as its metabolic consequences. Pharmacological inhibition of cathepsins B, L, and S using a novel inhibitor, GB111-NH\(_2\), led to changes in cellular recycling processes characterized by an increased expression of autophagy- and lysosome-associated marker genes and reduced adenosine triphosphate (ATP) content. Decreased cathepsin activity in primary macrophages further led to distinct changes in fatty acid metabolites associated with increased expression of key modulators of fatty acid metabolism, such as fatty acid synthase (FASN) and acid ceramidase (ASAH1). The altered fatty acid profile was associated with an increased synthesis of the pro-inflammatory prostaglandin PGE\(_2\), which correlated with the upregulation of numerous NF\(_k\)B-dependent pro-inflammatory mediators, including interleukin-1 (IL-1), interleukin-6 (IL-6), C-C motif chemokine ligand 2 (CCL2), and tumor necrosis factor-alpha (TNFα). Our data indicate a novel link between cathepsin activity and metabolic reprogramming in macrophages, demonstrated by a profound impact on autophagy and fatty acid metabolism, which facilitates a pro-inflammatory micromilieu generally associated with enhanced tumor elimination. These results provide a strong rationale for therapeutic cathepsin inhibition to overcome the tumor-promoting effects of the immune-evasive tumor micromilieu. KW - cathepsin KW - activity-based probes KW - tumor-associated macrophage KW - autophagy KW - lysosome KW - lipid metabolism KW - inflammation Y1 - 2020 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-213040 SN - 2072-6694 VL - 12 IS - 9 ER -