@phdthesis{Adelfinger2012, author = {Adelfinger, Marion}, title = {Pr{\"a}klinische Verwendung verschiedener onkolytischer Vaccinia-Viren zur Therapie von Human- und Hundetumoren}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-70688}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2012}, abstract = {Nach Einsch{\"a}tzung der Weltgesundheitsorganisation WHO wird Krebs im Jahr 2013 die weltweit h{\"a}ufigste Todesursache bei Menschen und Haustieren sein. Diese Situation erfordert die Entwicklung neuer therapeutischer Ans{\"a}tze. Hauptziel einer Tumortherapie ist es, sowohl den Prim{\"a}rtumor als auch die Metastasen m{\"o}glichst vollst{\"a}ndig zu entfernen. Dabei wird nach Methoden gesucht, die im Gegensatz zu den meisten gegenw{\"a}rtigen therapeutischen Eins{\"a}tzen, wie der chirurgischen Entfernung b{\"o}sartiger Neubildungen, Chemotherapie und Strahlentherapie, selektiv die b{\"o}sartigen Zellen erkennen und zerst{\"o}ren k{\"o}nnen. Eine faszinierende M{\"o}glichkeit in dieser Hinsicht ist die Verwendung von onkolytischen Viren, die die F{\"a}higkeit besitzen, sich selektiv sowohl in Prim{\"a}rtumoren als auch in Metastasen anzusiedeln und die Krebszellen dort zu zerst{\"o}ren. Das Konzept, dass Viren n{\"u}tzlich f{\"u}r die Bek{\"a}mpfung von Krebs sein k{\"o}nnten, ist nicht neu. Allerdings konnte erst in den letzten Jahren durch zahlreiche Studien best{\"a}tigt werden, dass verschiedene Viren in der Lage sind, eine signifikante Antitumorwirkung in vivo auszu{\"u}ben. Zu den erfolgversprechenden onkolytischen Viren z{\"a}hlen insbesondere Adenovirus, Herpes simplex Virus, Reovirus und Vaccinia-Virus, die sich bereits in Phase III der klinischen Studien befinden oder kurz davor sind. Die therapeutische Nutzung von tumorspezifischen onkolytischen Viren beim Menschen hat bereits begonnen. Im Rahmen der vorliegenden Doktorarbeit wurden verschiedene Aspekte der Wirkungsweise von Vaccinia-Virus-St{\"a}mmen bei der Therapie verschiedener Tumore aus Mensch und Hund im Xenotransplantat-Mausmodell bearbeitet: die Onkolyse der Krebszellen und Inhibition des Tumorwachstums sowie die Effekte der Virusinfektion auf das Tumormikromilieu und die Mitwirkung des angeborenen Immunsystems bei der Virotherapie. Das Tumormikromilieu (Stroma) setzt sich aus einer Vielzahl verschiedener Zellen und Komponenten der extrazellul{\"a}ren Matrix zusammen. Die Krebszellen bilden unter anderem mit Endothelzellen des Blut- und Lymphsystems und verschiedenen Immunzellen eine komplexe Organ-{\"a}hnliche Struktur. Weitere wichtige Bestandteile des Stromas sind Wachstumsfaktoren, Chemokine und Zytokine und die Tumorvaskulatur. Diese ist durch zahlreiche strukturelle und funktionelle Abnormalit{\"a}ten charakterisiert, wodurch die Effektivit{\"a}t von Strahlen- und Chemotherapie herabgesetzt wird. Weiterhin ist das Tumormikromilieu durch seine {\"A}hnlichkeit mit einer chronischen Entz{\"u}ndungsreaktion gekennzeichnet und wirkt immunsupprimierend auf rekrutierte Leukozyten, die wiederum die Inflammation verst{\"a}rken und die Angiogenese und das Tumorwachstum weiter f{\"o}rdern. Aufgrund dieser vielen Komponenten ist die Zusammensetzung jedes Tumors einzigartig, weswegen Standardtherapien h{\"a}ufig nicht zu einer Heilung f{\"u}hren. Die Wirkung der Viren bei der Virotherapie beruht vermutlich auf 4 Mechanismen, die einzeln oder in Kombination auftreten k{\"o}nnen: die direkte Onkolyse der Krebszellen, die Zerst{\"o}rung des Tumorblutgef{\"a}ßsystems, die Aktivierung des Immunsystems des Wirts und die Suppression der microRNA-Expression des Wirtes. Zus{\"a}tzlich kann die Expression therapeutischer Gene die onkolytische Wirkung verst{\"a}rken. Zum Nachweis der Onkolyse der Krebszellen und Inhibition des Tumorwachstums wurde zuerst das Virus GLV-1h68 in einem autologen humanen Melanomzellpaar, 888-MEL und 1936-MEL, eingesetzt. Das GLV-1h68-Virus wurde auf Basis des Wildtyp Vaccinia-Virus LIVP durch die Insertion von 3 Expressionskassetten in den drei Genloci F14.5L, J2R und A56 genetisch konstruiert. 888-MEL, eine zu einem fr{\"u}hen Zeitpunkt der Krebserkrankung aus einer Metastase isolierte Zelllinie, zeigt nach Infektion mit GLV-1h68 im Mausmodell Tumornekrose („Responder"), w{\"a}hrend 1936-MEL aus einer sp{\"a}ten Metastasierungsphase kaum mit Onkolyse auf eine Virusinfektion reagiert („Poor-Responder"). Die onkolytische Wirkung konnte mittels Durchflusszytometrie in Tumoren beider Zelllinien zu einem fr{\"u}hen Zeitpunkt nach Virusinfektion nachgewiesen werden. In 888-MEL-Tumoren wurde hierbei eine große Zahl infizierter und toter Zellen nach Virusinfektion gefunden. Gleichzeitig wurde eine hohe Zahl an Immunzellen detektiert, die nach Virusinfektion reduziert war. In den schw{\"a}cher reagierenden 1936-MEL-Tumoren konnte eine Onkolyse bei Infektion mit h{\"o}herer Virusmenge und zu einem fr{\"u}heren Zeitpunkt demonstriert werden, wodurch mehr Zellen infiziert wurden. Zus{\"a}tzlich wurde eine Steigerung der nur in geringer Zahl vorhandenen Immunzellen nachgewiesen. Trotz des unterschiedlichen Tumormikromilieus konnte somit ein onkolytischer Effekt in beiden Tumormodellen erzielt werden. ...}, subject = {Vaccinia-Virus}, language = {de} } @article{AdelfingerBesslerCeciletal.2015, author = {Adelfinger, Marion and Bessler, Simon and Cecil, Alexander and Langbein-Laugwitz, Johanna and Frentzen, Alexa and Gentschev, Ivaylo and Szalay, Aladar A.}, title = {Preclinical Testing Oncolytic Vaccinia Virus Strain GLV-5b451 Expressing an Anti-VEGF Single-Chain Antibody for Canine Cancer Therapy}, series = {Viruses}, volume = {7}, journal = {Viruses}, doi = {10.3390/v7072811}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-125705}, pages = {4075-4092}, year = {2015}, abstract = {Virotherapy on the basis of oncolytic vaccinia virus (VACV) strains is a novel approach for canine cancer therapy. Here we describe, for the first time, the characterization and the use of VACV strain GLV-5b451 expressing the anti-vascular endothelial growth factor (VEGF) single-chain antibody (scAb) GLAF-2 as therapeutic agent against different canine cancers. Cell culture data demonstrated that GLV-5b451 efficiently infected and destroyed all four tested canine cancer cell lines including: mammary carcinoma (MTH52c), mammary adenoma (ZMTH3), prostate carcinoma (CT1258), and soft tissue sarcoma (STSA-1). The GLV-5b451 virus-mediated production of GLAF-2 antibody was observed in all four cancer cell lines. In addition, this antibody specifically recognized canine VEGF. Finally, in canine soft tissue sarcoma (CSTS) xenografted mice, a single systemic administration of GLV-5b451 was found to be safe and led to anti-tumor effects resulting in the significant reduction and substantial long-term inhibition of tumor growth. A CD31-based immuno-staining showed significantly decreased neo-angiogenesis in GLV-5b451-treated tumors compared to the controls. In summary, these findings indicate that GLV-5b451 has potential for use as a therapeutic agent in the treatment of CSTS.}, language = {en} } @phdthesis{Amelingmeier2022, author = {Amelingmeier, Florian}, title = {Identifizierung und Untersuchung TOP-mRNA - bindender Faktoren}, doi = {10.25972/OPUS-28923}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-289231}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2022}, abstract = {Im Zellkern eukaryotischer Zellen werden Gene in mRNAs transkribiert, welche umfangreich prozessiert und aus dem Zellkern exportiert werden. Im Zytoplasma erfolgt die Translation der mRNAs in Proteine, ein Prozess, welcher viel Energie ben{\"o}tigt und daher mittels vielf{\"a}ltiger Mechanismen streng reguliert wird. Ein Beispiel hierf{\"u}r stellt die Klasse der TOP-mRNAs dar, eine RNA-Spezies, welche haupts{\"a}chlich Transkripte von Genen umfasst, die selbst in die Translation involviert sind. Die prominentesten Vertreter dieser Klasse sind die Proteine der kleinen und großen ribosomalen Untereinheiten. TOP-mRNAs zeichnen sich durch ein gemeinsames Sequenz-Motiv am Anfang Ihrer 5'-UTR aus, welches aus einem Pyrimidinstrang besteht und unmittelbar nach dem Cap mit einem Cytosin beginnt. Dieses allen TOP-RNAs gemeinsame Motiv erm{\"o}glicht die zeitgleiche Translationskontrolle dieser RNA-Klasse. So kann die Translation der TOP-mRNAs unter Stressbedingungen wie z.B. N{\"a}hrstoffmangel koordiniert inhibiert werden, wodurch Energie eingespart wird. Bereits lange wird nach einem Regulator gesucht, der an dieses TOP-Motiv bindet und die koordinierte Regulation erm{\"o}glicht. Man kann sich hier einen Inhibitor oder auch einen Aktivator vorstellen. Verschiedene Proteine wurden bereits in Erw{\"a}gung gezogen. In dieser Arbeit wurde das Protein TIAR mittels Massenspektrometrie als TOP-interagierender Faktor identifiziert und dessen Bindungseigenschaften mit dem TOP-Motiv durch Shift Assays untersucht. Hierbei konnten Minimalkonstrukte verschiedener Organismen sowie RNA-TOP - Sequenzen identifiziert werden, welche sich f{\"u}r Strukturanalysen eignen w{\"u}rden. Als weiterer TOP-interagierender Faktor wurde {\"u}ber verschiedene sequenzielle Reinigungsschritte das Protein 14-3-3ε identifiziert. Weiterhin wurden die TOP-Motiv-bindenden Proteine LARP1 und LARP7 auf Ihre Bindungseigenschaften mit Ihren Zielsequenzen untersucht. W{\"a}hrend gezeigt werden konnte, dass LARP1 einen inhibierenden Einfluss auf TOP-RNAs hat, wurde in weiteren Shift-Assays die Bindungseigenschaften von LARP7 mit 7SK untersucht, wobei ebenfalls ein minimales LARP7-Konstrukt sowie 7SK-Konstrukte f{\"u}r Strukturanalysen identifiziert werden konnten. Weiterhin konnte gezeigt werden, dass verschiedene Substanzen wie tRNA und Arginin einen starken Einfluss auf die LARP7-7SK - Interaktion aus{\"u}ben, welcher in weiteren Studien ber{\"u}cksichtigt werden sollte.}, subject = {Proteinbiosynthese}, language = {de} } @article{AsciertoWorschechYuetal.2011, author = {Ascierto, Maria Libera and Worschech, Andrea and Yu, Zhiya and Adams, Sharon and Reinboth, Jennifer and Chen, Nanhai G and Pos, Zoltan and Roychoudhuri, Rahul and Di Pasquale, Giovanni and Bedognetti, Davide and Uccellini, Lorenzo and Rossano, Fabio and Ascierto, Paolo A and Stroncek, David F and Restifo, Nicholas P and Wang, Ena and Szalay, Aladar A and Marincola, Francesco M}, title = {Permissivity of the NCI-60 cancer cell lines to oncolytic Vaccinia Virus GLV-1h68}, series = {BMC Cancer}, volume = {11}, journal = {BMC Cancer}, number = {451}, doi = {10.1186/1471-2407-11-451}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-141503}, pages = {1-14}, year = {2011}, abstract = {Background: Oncolytic viral therapy represents an alternative therapeutic strategy for the treatment of cancer. We previously described GLV-1h68, a modified Vaccinia Virus with exclusive tropism for tumor cells, and we observed a cell line-specific relationship between the ability of GLV-1h68 to replicate in vitro and its ability to colonize and eliminate tumor in vivo. Methods: In the current study we surveyed the in vitro permissivity to GLV-1h68 replication of the NCI-60 panel of cell lines. Selected cell lines were also tested for permissivity to another Vaccinia Virus and a vesicular stomatitis virus (VSV) strain. In order to identify correlates of permissity to viral infection, we measured transcriptional profiles of the cell lines prior infection. Results: We observed highly heterogeneous permissivity to VACV infection amongst the cell lines. The heterogeneity of permissivity was independent of tissue with the exception of B cell derivation. Cell lines were also tested for permissivity to another Vaccinia Virus and a vesicular stomatitis virus (VSV) strain and a significant correlation was found suggesting a common permissive phenotype. While no clear transcriptional pattern could be identified as predictor of permissivity to infection, some associations were observed suggesting multifactorial basis permissivity to viral infection. Conclusions: Our findings have implications for the design of oncolytic therapies for cancer and offer insights into the nature of permissivity of tumor cells to viral infection.}, language = {en} } @phdthesis{Auth2005, author = {Auth, Tanja}, title = {Funktionelle Analyse der Interaktion und Lokalisation von Replikationsfaktoren und replikationsrelevanten Proteinen in Mauszellen}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-13082}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2005}, abstract = {Zeil dieser Arbeit war die Identifikation von Proteinen, die mit Bestandteilen des f{\"u}r die DNA-Replikation essentiellen pr{\"a}replikativen Komplexes in der Maus wechselwirken. Hierbei konnten Interkationen des Heterochromatin Proteins 1a mit den Replikationsfaktoren ORC1, ORC2 und CDC6 sowohl in Two Hybrid-Studien als auch in Immunpr{\"a}zipitationen gezeigt werden. Dar{\"u}berhinaus konnten signifikante Kolokalisationen dieser Proteine mit HP1a an heterochromatischen Regionen in murinen NIH3T3-Zellen nachgewiesen werden. Ebenfalls konnte hier erstmals eine Lokalisation von HP1a am Centrosom demonstriert werden. Ein siRNA-vermittelter Knock Down von HP1a zeigte jedoch keinen direkten Einfluß auf die Replikation. Es konnte hingegen gezeigt werden, daß ein Knock Down von HP1a in signifikatnen Defekten in der Cytokinese und einer deutlich verlangsamten Zellproliferation resultiert. So konnten h{\"a}ufig multinukle{\"a}re Zellen und eine Arretierung in der G1-Phase beobachtet werden. Weiterhin wurde der Einfluß der Phosphorylierung von HP1a durch die Casein Kinase II mithilfe von Phosphorylierungsmutanten untersucht. Im Gegensatz zu Drosophila-Zellen zeigten sich in murinen Zellen jedoch keine Auswirkungen dieser Mutationen auf die Lokalisation von HP1a an Heterochromatin. Wieterhin konnten Interaktionen des Replikationsinhibitors Geminin mit den Replikationsproteinen ORC1, ORC2 und CDC7 sowohl im Two Hybrid-System als auch in Immunpr{\"a}zipitationen gezeigt werden, die unterschiedliche Zellzyklusabh{\"a}ngigkeiten aufwiesen. In murinen NIH3T3-Zellen zeigte eine Knock Down von Geminin jedoch im Gegensatz zu anderen Zellinien keinen Einfluß auf die Replikation. In weiteren Teilen dieser Arbeit konnten Interaktionen des Retinoblastoma Proteins mit ORC2 und MCM7 sowohl in Two Hybrid- als auch in Immunpr{\"a}zipitations-Experimenten gezeigt werden. Dar{\"u}berhinaus wies Pescadillo Interaktionen mit den Replikationsproteinen ORC2, ORC6, MCM2, MCM3, MCM6 und CDC45 im Two Hybrid-System und Interaktionen mit MCM2 und MCM3 in Biolumineszenz-Resonanzenergietransfer-Experimenten auf. Eine Kolokalisation von Pescadillo und ORC6 in den Nukleoli l{\"a}ßt auf eine Funktion beider Proteinen bei der Ribosomen Biogenese schließen. Es konnten ebenfalls Interaktionen der Untereinheit E1 des humanen Papillomavirus Subtyp 11 mit den Replikationsfaktoren ORC2,3,4,5,6, MCM2, MCM3, MCM6, CDC6, CDC7, CDT1, HP1a, Rb und Pescadillo im Two Hybrid-System beobachtet werden.}, subject = {Maus}, language = {de} } @phdthesis{Bayer2005, author = {Bayer, Tanja}, title = {Toxicity and biotransformation of 1,1,1,3,3-Pentafluoropropane, 3,3,3-Trifluoropropionic acid and 1,1,1,3-Tetrachloropropane}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-15731}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2005}, abstract = {The biotransformation of 1,1,1,3,3-pentafluoropropane was investigated in rats and in in vitro systems. First, the metabolites were identified in vivo using GC/MS and 19F NMR analysis. The main metabolite was identified as trifluoroacetic acid, the minor metabolite as 3,3,3-trifluoropropionic acid and as a cleavage product, inorganic fluoride was found. As the in vitro system, liver microsomes from rat and human samples and rat liver homogenates were used. Trifluoroacetic acid and 3,3,3 trifluoropropionic acid were confirmed in vitro as metabolic intermediates, following biotransformation of 1,1,1,3,3-pentafluoropropane by the cytochrome P-450-system. Studies, designed for clarifying the cardiotoxicity of 1,1,1,3,3-pentafluoropropane were driven by the hypothesis that 3,3,3-trifluoropropionic acid is the toxic agent. This was based on the lethal toxicity, which was observed in previous in vivo experiments. In addition, the point of its structural similarity to toxic agents as for example monofluoroacetic acid or of possible metabolic intermediates like difluoroacrylic acid with known toxicity were considered to support this assumption. However, trifluoroacetic acid was neglected as the sought-after toxic agent because of its different toxic effects, known from literature. Investigations on the biotransformation of 3,3,3-trifluoropropionic acid were performed and resulted in no metabolic activity and in poor elimination of 3,3,3-trifluoropropionic acid in vivo. The histopathological effects on the heart, which were observed in the 90-day oral toxicity study of 1,1,1,3,3-pentafluoropropane in rats, namely mononuclear inflammatory cell infiltrations and degenerated myocardial fibers, were not observed after a 28 day repeated exposure of up to 10 mg/kg b.w. of 3,3,3-trifluoropropionic acid. However, a single high dose of 3,3,3-trifluoropropionic acid lead to severe toxicological effects. The difference in the observed toxic effects after a single and repeated administration may be due to adaptive mechanisms in rats. The toxicological effects included clinical signs like ataxia, coma and cramps. The conditions of the rats suggested possible inhibition of the energy supply to the organism. Furthermore, the interference of 3,3,3-trifluoropropionic acid in the functionality of the organism was investigated. Experiments were performed in vitro in rat liver and heart mitochondria to investigate effects on the mitochondrial ß-oxidation. However, the transformation of the substrate [U14C] palmitic acid in the ß oxidation pathway was not inhibited by 3,3,3-trifluoropropionic acid. In addition, no cytotoxicity of 3,3,3 trifluoropropionic acid was observed in the cell culture systems. The main effect after a single dose of 3,3,3-trifluoropropionic acid was seen in clinical pathology and metabonomic analysis. The decrease in blood glucose is considered to have the most far-reaching consequences for the toxicity of 3,3,3-trifluoropropionic acid. If considering this change as the primary effect after a single dose, secondary effects, for example, the above-mentioned clinical signs could be explained. In addition, the observed high level of ketone bodies might have been responsible for life-threatening possible ketoacidosis. In general, ketoacidosis occurs after an imbalance between glycolysis, lipolysis, TCA cycle activity and respiratory function. Based on the results, ß-oxidation of fatty acids was not affected, and due to the decrease in glucose levels and the high levels of acetyl CoA, glycolysis was considered not to be impaired. Increased amounts of acetyl CoA might be a result of insufficient activity of the TCA cycle. However, the inhibition of the TCA cycle can be based on the impairment of specific enzymes and/or on the involvement of messenger substrates like insulin. Supporting the first mentioned aspect are decreased levels of TCA cycle intermediates, like \&\#945;-ketoglutarate or citrate, as seen in 1H-NMR spectra of urine. However, the second aspect would explain the drop in blood glucose with the impairment of glucose transporters or the impairment of the insulin balance. If a single dose of 3,3,3-trifluoropropionic acid had stimulated the insulin release, glycolysis would be activated, and high amounts of acetyl CoA would be produced. In case of impaired use by the TCA cycle, levels of ketone bodies would be increased. Experiments were designed to characterize the direct effect of 3,3,3-trifluoropropionic acid on rat insulinoma-derived INS-1 cells as possible increase in insulin release. Further investigations are necessary to answer in which step of the metabolic pathway 3,3,3-trifluoropropionic acid interferes or finally which specific enzyme is inhibited or activated by 3,3,3-trifluoropropionic acid, leading to the drop in blood glucose and finally in lethal toxicity.}, subject = {Fluorkohlenwasserstoffe}, language = {en} } @phdthesis{Bedenk2018, author = {Bedenk, Kristina}, title = {Biochemische und strukturelle Charakterisierung der Genexpressionsmaschinerie des Vaccinia Virus}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-135538}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2018}, abstract = {Die Familie der Pockenviren zeichnet sich durch ein komplexes DNA Genom aus und hat großes medizinisches Potential. Am eindrucksvollsten ist dies f{\"u}r das Vaccinia-Virus (VACV) belegt, welches nicht nur als Pocken-Impfstoff eingesetzt wird, sondern auch als onkolytisches Virus in der Tumorbiologie. VACV hat einen außergew{\"o}hnlichen Replikationszyklus, welcher ausschließlich im Zytoplasma der Wirtszelle stattfindet. Somit ist die gesamte virale Genexpressionsmaschinerie v{\"o}llig unabh{\"a}ngig von kernvermittelten Reaktionen des Wirts und somit auch aus Sicht der Grundlagenforschung von gr{\"o}ßtem Interesse. Die Schl{\"u}sselkomponente der viralen Genexpression ist die makromolekulare DNA-abh{\"a}ngige RNA Polymerase (vvRPO), deren Untereinheiten allesamt Virus-kodiert sind. Zwar wurden in den letzten Jahren Protokolle zur biochemischen und funktionellen Charakterisierung der vvRPO etabliert, ein detailliertes Wissen {\"u}ber deren Zusammenlagerung in vivo und die r{\"a}umlichen und zeitlichen Interaktionen mit den Transkriptions- bzw. Prozessierungsfaktoren sind aber weitgehend unbekannt. Diese Arbeit umfasst Untersuchungen zur strukturellen und funktionellen Charakterisierung der vvRPO und seiner assoziierten Faktoren. Grundlage hierf{\"u}r war die Etablierung eines Reinigungsprotokolls mithilfe eines neu konstruierten rekombinanten VACV (GLV-1h439). Diese Strategie erlaubte es hoch-molekulare native vvRPO Komplexe zu isolieren. Ein transkriptions-inaktiver Komplex (Komplex I) mit einer kalkulierten Masse von 575 kDa bestand aus den acht Untereinheiten des vvRPO Holoenzyms und den Polymerase-assoziierten Faktoren RAP94 und D6. Ein zweiter, transkriptionell aktiver Komplex (Komplex II) mit einer Masse von 803 kDa enthielt, neben dem Holoenzym der vvRPO, noch weitere Faktoren, die prim{\"a}r die Erkennung der DNA-Matrize und die Prozessierung der naszierenden RNA vermitteln. Hierbei handelt es sich um RAP94, das virale Capping Enzym bestehend aus den zwei Untereinheiten D1 und D12, A7 und dem Terminationsfaktor NPH I. Interessanterweise enthielt dieser Komplex zus{\"a}tzlich mit E11 eine bislang unbekannte weitere Protein-Komponente, sowie tRNAGln und tRNAArg. Der isolierte Kompelx II ist daher ein Ribonukleoprotein (RNP). Die Verf{\"u}gbarkeit von hoch-reinen vvRPO Komplexen erlaubte es erstmals deren strukturelle Architektur zu untersuchen. Hierf{\"u}r wurden drei experimentelle Ans{\"a}tze, die klassische R{\"o}ntgenstrukturanalyse, die Kryo-Elektronenmikroskopie (Kryo-EM) und Quervernetzungssstudien miteinander kombiniert. Die Strukturen der Komplexe I und II haben eine Aufl{\"o}sung von 11-12 {\AA}, wobei auff{\"a}llig war, dass beide eine markante strukturelle {\"A}hnlichkeit zur eukaryotischen RNA Polymerase II aufwiesen. Dar{\"u}ber hinaus gelang es zus{\"a}tzliche Bereiche im Komplex II zu definieren, welche die Polymerase-assoziierten Prozessierungsfaktoren beherbergen. Zudem konnte die atomare Struktur von E11, mittels R{\"o}ntgenstrukturanalyse bei einer Aufl{\"o}sung von 1,9 {\AA}, gel{\"o}st werden. Das E11 Protein besitzt ein neuartiges Faltungsmuster und weist einen intensiven Dimerisierungskontakt auf, welcher sich {\"u}ber vier ß-Faltbl{\"a}tter ausbildet. Die im Rahmen dieser Arbeit erhaltenen Daten legen die Grundlage f{\"u}r ein detailliertes Verst{\"a}ndnis der r{\"a}umlichen Organisation der viralen Transkriptonsmaschinerie. Dar{\"u}ber hinaus werden sie funktionelle Studien erm{\"o}glichen, welche die Rolle der einzelnen Proteine, sowie der tRNAs bei der mRNA Synthese kl{\"a}ren helfen.}, subject = {Vaccinia-Virus}, language = {de} } @article{BenhalevyGuptaDananetal.2017, author = {Benhalevy, Daniel and Gupta, Sanjay K. and Danan, Charles H. and Ghosal, Suman and Sun, Hong-Wei and Kazemeier, Hinke G. and Paeschke, Katrin and Hafner, Markus and Juranek, Stefan A.}, title = {The Human CCHC-type Zinc Finger Nucleic Acid-Binding Protein Binds G-Rich Elements in Target mRNA Coding Sequences and Promotes Translation}, series = {Cell Reports}, volume = {18}, journal = {Cell Reports}, number = {12}, doi = {10.1016/j.celrep.2017.02.080}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-171122}, pages = {2979-2990}, year = {2017}, abstract = {The CCHC-type zinc finger nucleic acid-binding protein (CNBP/ZNF9) is conserved in eukaryotes and is essential for embryonic development in mammals. It has been implicated in transcriptional, as well as post-transcriptional, gene regulation; however, its nucleic acid ligands and molecular function remain elusive. Here, we use multiple systems-wide approaches to identify CNBP targets and function. We used photoactivatable ribonucleoside-enhanced crosslinking and immunoprecipitation (PAR-CLIP) to identify 8,420 CNBP binding sites on 4,178 mRNAs. CNBP preferentially bound G-rich elements in the target mRNA coding sequences, most of which were previously found to form G-quadruplex and other stable structures in vitro. Functional analyses, including RNA sequencing, ribosome profiling, and quantitative mass spectrometry, revealed that CNBP binding did not influence target mRNA abundance but rather increased their translational efficiency. Considering that CNBP binding prevented G-quadruplex structure formation in vitro, we hypothesize that CNBP is supporting translation by resolving stable structures on mRNAs.}, language = {en} } @phdthesis{Benz2007, author = {Benz, Peter Michael}, title = {Cytoskeleton assembly at endothelial cell-cell contacts is regulated by Alpha-II-spectrin/vasp complexes}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-23802}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2007}, abstract = {Directed cortical actin assembly is the driving force for intercellular adhesion. Vasodilator-stimulated phosphoprotein (VASP) participates in actin-fiber formation and VASP activity is regulated by phosphorylations. We screened for endothelial cell proteins, which bind to VASP dependent on its phosphorylation status. Differential proteomics identified \&\#945;II-spectrin as novel VASP-interacting protein. \&\#945;II-spectrin binds to the triple GP5-motif in VASP via its SH3 domain. cAMP-dependent protein kinase-mediated VASP phosphorylation at Ser157 inhibits \&\#945;II-spectrin/VASP complex formation. VASP becomes dephosphorylated upon formation of cell-cell contacts and in confluent but not in sparse endothelial cells \&\#945;II-spectrin colocalizes with non-phosphorylated VASP at cell-cell junctions. Ectopic expression of the \&\#945;II-spectrin SH3 domain fused to claudin-5 translocates VASP to cell-cell contacts and is sufficient to initiate the formation of cortical actin cytoskeletons. \&\#945;II-spectrin SH3 domain overexpression stabilizes cell-cell contacts and decreases endothelial permeability. Conversely, permeability of VASP-deficient endothelial cells is elevated. In a skin edema model, microvascular leakage is increased in VASP-deficient over wild-type mice. We propose that \&\#945;II-spectrin/VASP complexes regulate cortical actin cytoskeleton assembly with implications for formation of endothelial cell-cell contacts and regulation of vascular permeability.}, language = {en} } @phdthesis{Brand2005, author = {Brand, Normen}, title = {Lokalisation, Regulation und Interaktionen muriner DNA-Replikationsproteine}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-14057}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2005}, abstract = {Die DNA-Replikation ist ein entscheidendes Ereignis im eukaryontischen Zellzyklus, das die exakte Duplizierung des Genoms gew{\"a}hrleistet und das geordnete Zusammenspiel einer Vielzahl von Proteinen erfordert. Um diese enorme logistische Herausforderung zu bewerkstelligen ist die DNA-Replikation in mehrere Schritte organisiert, die Initiationsprozesse, Elongation und DNA-Reparatur umfassen. Der Initiationsschritt ist gekennzeichnet durch die Chromatin-Assoziation des hexameren ORC (origin recognition complex), der kontrovers diskutierte DNA-Sequenzen als Origins erkennt und bindet sowie als Landeplattform f{\"u}r weitere Proteinkomponenten dient. Der MCM-Komplex aus den sechs Untereinheiten Mcm2 7 komplettiert in Abh{\"a}ngigkeit von Cdc6 und Cdt1 den pr{\"a}-replikativen Komplex (pre-RC) und wird vermutlich nach der Initiation vom Origin entfernt, um als DNA-Helikase f{\"u}r die Entwindung der DNA-Doppelhelix zu sorgen. Dies erm{\"o}glicht den Proteinen der Elongations-Maschinerie DNA an mikroskopisch sichtbaren Orten, die als Replikationsfoci bezeichnet werden, korrekt zu synthetisieren. PCNA (proliferating cell nuclear antigen) ist eine Hauptkomponente der Replikationsfoci und fungiert als Ringklemme, die die DNA-Polymerasen und weitere Replikationsfaktoren an die DNA bindet. Im Rahmen dieser Arbeit wurde die Verteilung von ORC- MCM- und PCNA-Proteinen in murinen L-Fibroblasten durch Dual-Color-Immunfluoreszenz- (IF-) Studien untersucht. Dabei konnte gezeigt werden, dass die Proteine des ORC, des MCM-Komplexes und der Elongations-Maschinerie Positionen f{\"u}r drei verschiedene mechanistische Teil-Prozesse markieren, die an der DNA-Replikation beteiligt sind und an distinkten und r{\"a}umlich getrennten Orten stattfinden: Initiation, Helikase-Aktivit{\"a}t und Elongation. IF-Studien weisen außerdem darauf hin, dass die Acetylierung von Histonen im Zusammenhang mit der Auswahl der Origins steht. Die Assemblierung des pre-RC steht unter der Kontrolle mehrerer Protein-Kinasen. Um zu untersuchen, ob Protein-Komponenten des pre-RC auch vom Hauptregulator von mitotischen Ereignissen, der POLO-like kinase1 (Plk1), phosphoryliert werden, wurden in vitro-Kinase-Assays mit Wildtyp-Plk1 bzw. der Kinase-defizienten Mutante Plk1 (K82M) als Negativ-Kontrolle und potentiellen Targetproteinen durchgef{\"u}hrt. Orc2, Cdc7 und Cdc45 konnten als in vitro-Substrate f{\"u}r die Plk1-Kinase identifiziert werden. Diese Proteine sind außerdem in der Mitose an den Centrosomen, Cdc7 und Cdc45 an den Mikrotubuli und Orc2 und Cdc45 am Midbody lokalisiert. Diese mitotischen Lokalisations-Muster korrelieren mit denen von Plk1. Die Aufkl{\"a}rung von Protein-Protein-Interaktionen ist f{\"u}r das Verst{\"a}ndnis der Vorg{\"a}nge bei der DNA-Replikation essentiell. Mit der BRET (Biolumineszenz-Resonanzenergie-Transfer)-Technik konnten direkte Interaktionen zwischen Orc2 \& Orc3, Orc2 \& Orc4, Orc2 \& Orc5, Orc4 \& Orc6, Plk1 \& Orc2 und Plk1 \& Dbf4 gezeigt werden. Zus{\"a}tzlich wurden die Auswirkungen von Histon-Hyperacetylierung und der Depletion von Cyclin-abh{\"a}ngigen Kinasen (CDKs) auf die Interaktion zwischen Orc2 und Orc3 untersucht. Orc2 und Orc3 sind sowohl endogen als auch {\"u}berexprimiert im Zellkern und im Cytoplasma lokalisiert. Um herauszufinden, ob die Kernlokalisation von Orc3 Voraussetzung f{\"u}r die Interaktion zwischen Orc2 und Orc3 ist, wurde ein putatives Kernlokalisationssignal (NLS) in der aminoterminalen Region von Orc3 in einem EGFP-ORC3-Fusionsplasmid deletiert. Die Expression dieser Mutante resultierte in L-Fibroblasten und HEK293T-Zellen in ausschließlich cytoplasmatischer Lokalisation. BRET-Assays, bei denen ORC2-Rluc und die NLS-defiziente EGFP-ORC3-Mutante eingesetzt wurden, lieferten ein BRET ratio, das ununterscheidbar von dem mit Wildtyp EGFP-ORC3 erhaltenen Signal war. Diese Ergebnisse weisen darauf hin, dass die Interaktion zwischen Orc2 und Orc3 nicht auf den Zellkern beschr{\"a}nkt ist. Mit der erst k{\"u}rzlich entwickelten BiFC- (bimolecular fluorescence complementation) Technik konnte sowohl die cytoplasmatische als auch die nukle{\"a}re Lokalisation der Interaktion zwischen Orc2 und Orc3 gezeigt werden. FLIP- (fluorescence loss in photobleaching-) Studien mit BiFC-positiven Zellen, die eine ausschließlich nukle{\"a}re Lokalisation der Interaktion zwischen Orc2 und Orc3 aufwiesen, zeigten eine verringerte Mobilit{\"a}t des bin{\"a}ren Komplexes Orc2/Orc3 (t ½ = 10 s) im Vergleich zu EGFP-Fusionsproteinen von Orc2 (t ½ = 8 s) und Orc3 (t ½ = 6 s) auf. Dies deutet darauf hin, dass die Assoziation mit dem Bindungspartner zu einer erh{\"o}hten Chromatin-Bindung von Orc2 und Orc3 f{\"u}hrt. Zus{\"a}tzlich wurden die Auswirkungen von Punktmutationen auf die subzellul{\"a}re Lokalisation und die intranukle{\"a}re Dynamik des in Replikationsfoci lokalisierten Cdc6-EGFP-Fusionsproteins untersucht und die Mobilit{\"a}t von promyelocytic leukaemia nuclear bodies (PML NBs) und der darin enthaltenen Proteinkomponenten analysiert.}, subject = {Maus}, language = {de} } @phdthesis{Brosi2021, author = {Brosi, Cornelia}, title = {Functional characterization of the TTF complex and its role in neurodevelopmental disorders}, doi = {10.25972/OPUS-15778}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-157783}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2021}, abstract = {The eukaryotic gene expression requires extensive regulations to enable the homeostasis of the cell and to allow dynamic responses due to external stimuli. Although many regulatory mechanisms involve the transcription as the first step of the gene expression, intensive regulation occurs also in the post-transcriptional mRNA metabolism. Thereby, the particular composition of the mRNPs plays a central role as the components associated with the mRNA form a specific "mRNP code" which determines the fate of the mRNA. Many proteins which are involved in this regulation and the mRNA metabolism are affected in diseases and especially neurological disorders often result from an aberrant mRNP code which leads to changes in the regulation and expression of mRNPs. The focus of this work was on a trimeric protein complex which is termed TTF complex based on its subunits TDRD3, TOP3β and FMRP. Biochemical investigations revealed that the three components of the TTF complex are nucleo-cytosolic shuttle proteins which localize in the cytoplasm at the steady-state, associate with mRNPs and are presumably connected to the translation. Upon cellular stress conditions, the TTF components concentrate in stress granules. Thus, the TTF complex is part of the mRNP code, however its target RNAs and function are still completely unknown. Since the loss of functional FMRP results in the fragile X syndrome and TOP3β is associated with schizophrenia and intellectual disability, the TTF complex connects these phenotypically related neuro-psychiatric disorders with each other on a molecular level. Therefore, the aim of this work was to biochemically characterize the TTF complex and to define its function in the mRNA metabolism. In this work, evidence was provided that TDRD3 acts as the central unit of the TTF complex and directly binds to FMRP as well as to TOP3β. Thereby, the interaction of TDRD3 and TOP3β is very stable, whereas FMRP is a dynamic component. Interestingly, the TTF complex is not bound directly to mRNA, but is recruited via the exon junction complex (EJC) to mRNPs. This interaction is mediated by a specific binding motif of TDRD3, the EBM. Upon biochemical and biological investigations, it was possible to identify the interactome of the TTF complex and to define the role in the mRNA metabolism. The data revealed that the TTF complex is mainly associated with "early" mRNPs and is probably involved in the pioneer round of translation. Furthermore, TOP3β was found to bind directly to the ribosome and thus, establishes a connection between the EJC and the translation machinery. A reduction of the TTF components resulted in selective changes in the proteome in cultured cells, whereby individual protein subsets seem to be regulated rather than the global protein expression. Moreover, the enzymatic analysis of TOP3β indicated that TOP3β is a type IA topoisomerase which can catalytically attack not only DNA but also RNA. This aspect is particularly interesting with regard to the connection between early mRNPs and the translation which has been revealed in this work. The data obtained in this work suggest that the TTF complex plays a role in regulating the metabolism of an early mRNP subset possibly in the course of the pioneer round of translation. Until now, the link between an RNA topoisomerase and the mRNA metabolism is thereby unique and thus provides a completely new perspective on the steps in the post-transcriptional gene expression and its regulation.}, subject = {Messenger-RNP}, language = {en} } @article{BuchbergerBoehm2013, author = {Buchberger, Alexander and B{\"o}hm, Stephanie}, title = {The Budding Yeast Cdc48Shp1 Complex Promotes Cell Cycle Progression by Positive Regulation of Protein Phosphatase 1 (Glc7)}, series = {PLoS One}, journal = {PLoS One}, doi = {10.1371/journal.pone.0056486}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-96073}, year = {2013}, abstract = {The conserved, ubiquitin-selective AAA ATPase Cdc48 regulates numerous cellular processes including protein quality control, DNA repair and the cell cycle. Cdc48 function is tightly controlled by a multitude of cofactors mediating substrate specificity and processing. The UBX domain protein Shp1 is a bona fide substrate-recruiting cofactor of Cdc48 in the budding yeast S. cerevisiae. Even though Shp1 has been proposed to be a positive regulator of Glc7, the catalytic subunit of protein phosphatase 1 in S. cerevisiae, its cellular functions in complex with Cdc48 remain largely unknown. Here we show that deletion of the SHP1 gene results in severe growth defects and a cell cycle delay at the metaphase to anaphase transition caused by reduced Glc7 activity. Using an engineered Cdc48 binding-deficient variant of Shp1, we establish the Cdc48Shp1 complex as a critical regulator of mitotic Glc7 activity. We demonstrate that shp1 mutants possess a perturbed balance of Glc7 phosphatase and Ipl1 (Aurora B) kinase activities and show that hyper-phosphorylation of the kinetochore protein Dam1, a key mitotic substrate of Glc7 and Ipl1, is a critical defect in shp1. We also show for the first time a physical interaction between Glc7 and Shp1 in vivo. Whereas loss of Shp1 does not significantly affect Glc7 protein levels or localization, it causes reduced binding of the activator protein Glc8 to Glc7. Our data suggest that the Cdc48Shp1 complex controls Glc7 activity by regulating its interaction with Glc8 and possibly further regulatory subunits.}, language = {en} } @phdthesis{Buckel2012, author = {Buckel, Lisa}, title = {Evaluating the combination of oncolytic vaccinia virus and ionizing radiation in therapy of preclinical glioma models}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-85309}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2012}, abstract = {Glioblastoma multiforme (GBM) represents the most aggressive form of malignant brain tumors and remains a therapeutically challenge. Intense research in the field has lead to the testing of oncolytic viruses to improve tumor control. Currently, a variety of different oncolytic viruses are being evaluated for their ability to be used in anti-cancer therapy and a few have entered clinical trials. Vaccinia virus, is one of the viruses being studied. GLV-1h68, an oncolytic vaccinia virus engineered by Genelux Corporation, was constructed by insertion of three gene cassettes, RUC-GFP fusion, β-galactosidase and β- glucuronidase into the genome of the LIVP strain. Since focal tumor radiotherapy is a mainstay for cancer treatment, including glioma therapy, it is of clinical relevance to assess how systemically administered oncolytic vaccinia virus could be combined with targeted ionizing radiation for therapeutic gain. In this work we show how focal ionizing radiation (IR) can be combined with multiple systemically delivered oncolytic vaccinia virus strains in murine models of human U-87 glioma. After initial experiments which confirmed that ionizing radiation does not damage viral DNA or alter viral tropism, animal studies were carried out to analyze the interaction of vaccinia virus and ionizing radiation in the in vivo setting. We found that irradiation of the tumor target, prior to systemic administration of oncolytic vaccinia virus GLV-1h68, increased viral replication within the U-87 xenografts as measured by viral reporter gene expression and viral titers. Importantly, while GLV-1h68 alone had minimal effect on U-87 tumor growth delay, IR enhanced GLV-1h68 replication, which translated to increased tumor growth delay and mouse survival in subcutaneous and orthotopic U-87 glioma murine models compared to monotherapy with IR or GLV-1h68. The ability of IR to enhance vaccinia replication was not restricted to the multi-mutated GLV-1h68, but was also seen with the less attenuated oncolytic vaccinia, LIVP 1.1.1. We have demonstrated that in animals treated with combination of ionizing radiation and LIVP 1.1.1 a strong pro-inflammatory tissue response was induced. When IR was given in a more clinically relevant fractionated scheme, we found oncolytic vaccinia virus replication also increased. This indicates that vaccinia virus could be incorporated into either larger hypo-fraction or more conventionally fractionated radiotherapy schemes. The ability of focal IR to mediate selective replication of systemically injected oncolytic vaccinia was demonstrated in a bilateral glioma model. In mice with bilateral U-87 tumors in both hindlimbs, systemically administered oncolytic vaccinia replicated preferentially in the focally irradiated tumor compared to the shielded non- irradiated tumor in the same mouse We demonstrated that tumor control could be further improved when fractionated focal ionizing radiation was combined with a vaccinia virus caring an anti-angiogenic payload targeting vascular endothelial growth factor (VEGF). Our studies showed that following ionizing radiation expression of VEGF is upregulated in U-87 glioma cells in culture. We further showed a concentration dependent increase in radioresistance of human endothelial cells in presence of VEGF. Interestingly, we found effects of vascular endothelial growth factor on endothelial cells were reversible by adding purified GLAF-1 to the cells. GLAF-1 is a single- chain antibody targeting human and murine VEGF and is expressed by oncolytic vaccinia virus GLV-109. In U-87 glioma xenograft murine models the combination of fractionated ionizing radiation with GLV-1h164, a vaccinia virus also targeting VEGF, resulted in the best volumetric tumor response and a drastic decrease in vascular endothelial growth factor. Histological analysis of embedded tumor sections 14 days after viral administration confirmed that blocking VEGF translated into a decrease in vessel number to 30\% of vessel number found in control tumors in animals treated with GLV-164 and fractionated IR which was lower than for all other treatment groups. Our experiments with GLV-1h164 and fractionated radiotherapy have shown that in addition to ionizing radiation and viral induced tumor cell destruction we were able to effectively target the tumor vasculature. This was achieved by enhanced viral replication translating in increased levels of GLAF-2 disrupting tumor vessels as well as the radiosensitization of tumor vasculature to IR by blocking VEGF. Our preclinical results have important clinical implications of how focal radiotherapy can be combined with systemic oncolytic viral administration for highly aggressive, locally advanced tumors with the potential, by using a vaccinia virus targeting human vascular endothelial growth factor, to further increase tumor radiation sensitivity by engaging the vascular component in addition to cancer cells.}, subject = {Gliom}, language = {en} } @article{CecilGentschevAdelfingeretal.2019, author = {Cecil, Alexander and Gentschev, Ivaylo and Adelfinger, Marion and Dandekar, Thomas and Szalay, Aladar A.}, title = {Vaccinia virus injected human tumors: oncolytic virus efficiency predicted by antigen profiling analysis fitted boolean models}, series = {Bioengineered}, volume = {10}, journal = {Bioengineered}, number = {1}, doi = {10.1080/21655979.2019.1622220}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-200507}, pages = {190-196}, year = {2019}, abstract = {Virotherapy on the basis of oncolytic vaccinia virus (VACV) strains is a promising approach for cancer therapy. Recently, we showed that the oncolytic vaccinia virus GLV-1h68 has a therapeutic potential in treating human prostate and hepatocellular carcinomas in xenografted mice. In this study, we describe the use of dynamic boolean modeling for tumor growth prediction of vaccinia virus-injected human tumors. Antigen profiling data of vaccinia virus GLV-1h68-injected human xenografted mice were obtained, analyzed and used to calculate differences in the tumor growth signaling network by tumor type and gender. Our model combines networks for apoptosis, MAPK, p53, WNT, Hedgehog, the T-killer cell mediated cell death, Interferon and Interleukin signaling networks. The in silico findings conform very well with in vivo findings of tumor growth. Similar to a previously published analysis of vaccinia virus-injected canine tumors, we were able to confirm the suitability of our boolean modeling for prediction of human tumor growth after virus infection in the current study as well. In summary, these findings indicate that our boolean models could be a useful tool for testing of the efficacy of VACV-mediated cancer therapy already before its use in human patients.}, language = {en} } @phdthesis{Chari2009, author = {Chari, Ashwin}, title = {The Reaction Mechanism of Cellular U snRNP Assembly}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-40804}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2009}, abstract = {Macromolecular complexes, also termed molecular machines, facilitate a large spectrum of biological reactions and tasks crucial to the survival of cells. These complexes are composed of either protein only, or proteins bound to nucleic acids (DNA or RNA). Prominent examples for each class are the proteosome, the nucleosome and the ribosome. How such units are assembled within the context of a living cell is a central question in molecular biology. Earlier studies had indicated that even very large complexes such as ribosomes could be reconstituted from purified constituents in vitro. The structural information required for the formation of macromolecular complexes, hence, lies within the subunits itself and, thus, allow for self- assembly. However, increasing evidence suggests that in vivo many macromolecular complexes do not form spontaneously but require assisting factors ("assembly chaperones") for their maturation. In this thesis the assembly of RNA-protein (RNP) complexes has been studied by a combination of biochemical and structural approaches. A resourceful model system to study this process is the biogenesis pathway of the uridine-rich small nuclear ribonucleoproteins (U snRNPs) of the spliceosome. This molecular machine catalyzes pre-mRNA splicing, i.e. the removal of non-coding introns and the joining of coding exons to functional mRNA. The composition and architecture of U snRNPs is well defined, also, the nucleo-cytoplasmic transport events enabling the formation of these particles in vivo have been analyzed in some detail. Furthermore, recent studies suggest that the formation of U snRNPs in vivo is mediated by an elaborate assembly machinery consisting of protein arginine methyltransferase (PRMT5)- and survival motor neuron (SMN)-complexes. The elucidation of the reaction mechanism of cellular U snRNP assembly would serve as a paradigm for our understanding of how RNA-protein complexes are formed in the cellular environment. The following key findings were obtained as part of this study: 1) Efforts were made to establish a full inventory of the subunits of the SMN-complex. This was achieved by the biochemical definition and characterization of an atypical component of this complex, the unrip protein. This protein is associated with the SMN-complex exclusively in the cytoplasm and influences its subcellular localization. 2) With a full inventory of the components in hand, the architecture of the SMN-complex was defined on the basis of an interaction map of all subunits. This study elucidated that the proteins SMN, Gemin7 and Gemin8 form a backbone, onto which the remaining subunits adhere in a modular manner. 3) The two studies mentioned above formed the basis to elucidate the reaction mechanism of cellular U snRNP assembly. Initially, an early phase in the SMN-assisted formation of U snRNPs was analyzed. Two subunits of the U7 snRNP (LSm10 and 11) were found to interact with the PRMT5-complex, without being methylated. This report suggests that the stimulatory role of the PRMT5-complex is independent of its methylation activity. 4) Key reaction intermediates in U snRNP assembly were found and characterized by a combination of biochemistry and structural studies. Initially, a precursor to U snRNPs with a sedimentation coefficient of 6S is formed by the pICln subunit of the PRMT5-complex and Sm proteins. This intermediate was shown to constitute a kinetic trap in the U snRNP assembly reaction. Progression towards the assembled U snRNP depends on the activity of the SMN-complex, which acts as a catalyst. The formation of U snRNPs is shown to be structurally similar to the way clamps are deposited onto DNA to tether poorly processive polymerases. 5) The human SMN-complex is composed of several subunits. However, it is unknown whether all subunits of this entity are essential for U snRNP assembly. A combination of bioinformatics and biochemistry was applied to tackle this question. By mining databases containing whole-genome assemblies, the SMN-Gemin2 heterodimer is recognized as the most ancestral form of the SMN-complex. Biochemical purification of the Drosophila melanogaster SMN-complex reveals that this complex is composed of the same two subunits. Furthermore, evidence is provided that the SMN-Gemin2 heterodimer is necessary and sufficient to promote faithful U snRNP assembly. Future studies will adress further details in the reaction mechanism of cellular U snRNP assembly. The results obtained in this thesis suggest that the SMN and Gemin2 subunits are sufficient to promote U snRNP formation. What then is the function of the remaining subunits of the SMN-complex? The reconstitution schemes established in this thesis will be instrumental to address this question. Furthermore, additional mechanistic insights into the U snRNP assembly reaction will require the elucidation of structures of the assembly machinery trapped at various states. The prerequisite for these structural studies, the capability to generate homogenous complexes in sufficient amounts, has been accomplished in this thesis.}, subject = {Small nuclear RNP}, language = {en} } @phdthesis{Christenn2005, author = {Christenn, Marcus}, title = {Charakterisierung von Somatostatinrezeptor-Subtyp 4 interagierenden Proteinen in der Ratte (Rattus norvegicus)}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-14253}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2005}, abstract = {Somatostatin ist ein regulatorisches Peptid, das eine Vielzahl von biologischen Prozessen innerhalb des K{\"o}rpers beeinflußt. Die Wirkung von Somatostatin wird auf zellul{\"a}rer Ebene {\"u}ber eine Familie von f{\"u}nf G-Protein-gekoppelten Rezeptoren vermittelt, die entweder in G Protein-abh{\"a}ngiger Weise oder vermutlich auch {\"u}ber andere interagierende intrazellul{\"a}re Proteine auf nachgeschaltete Signaltransduktionswege wirken. Der Somatostatinrezeptor Subtyp 4 (SSTR4) wird haupts{\"a}chlich im Gehirn exprimiert und wirkt dort inhibierend auf die exzitatorische Signalweiterleitung. Es sind aber auch stimulierende Effekte des SSTR4 bekannt. Um das subtypspezifische Signalverhalten des SSTR4 weiter zu untersuchen, wurden im Rahmen dieser Arbeit Proteine gesucht, die intrazellul{\"a}r mit dem SSTR4 interagieren und so seine physiologischen Effekte beeinflussen. In einem ersten Ansatz konnten drei m{\"o}gli-che Interaktionspartner mit Hilfe des Hefe-Zwei-Hybrid-Systems identifiziert werden, die aber in nachfolgenden Untersuchungen als unpezifisch eingestuft wurden. Mit Hilfe einer Affinit{\"a}tschromatografie wurden dann zwei Proteine identifiziert, die spezifisch mit dem SSTR4 interagieren. Sowohl PSD-95 als auch PSD-93 (Postsynaptic density protein of 95 kDa bzw. 93kDa) wurden mit einem immobilisierten Peptid pr{\"a}zipitiert, das die neun C-terminalen Aminos{\"a}uren des SSTR4 enth{\"a}lt. Die Interaktion des SSTR4 mit PSD 95 wurde im Weiteren n{\"a}her charakterisiert. In einem Bindungsexperiment mit rekombinaten Proteinen konnte gezeigt werden, dass die Interaktion durch die 1. und 2. PDZ-Dom{\"a}ne von PSD-95 vermittelt wird. In humanen embryonalen Nieren-Zellen (HEK293), die den SSTR4 stabil exprimieren, konnte PSD-95 mit dem Rezeptor koimmunpr{\"a}zipitiert werden. Nach Koexpression von PSD-95 und SSTR4 findet man eine partielle Kolokalisierung beider Proteine an der Zellmembran, wobei aber der Großteil des PSD-95 weiterhin eine diffuse zytoplasmatische Verteilung zeigt. Die Interaktion wurde in vivo sowohl immunhistochemisch in kultivierten Hippocampus-Neuronen als auch durch Koimmunpr{\"a}zipitation beider Proteine aus Rattengehirn-Lysaten nachgewiesen. Die Interaktion von PSD-95 mit dem SSTR4 beeinflußt weder die Agonisten-induzierte Internalisierung des Rezeptors in HEK293-Zellen, noch die Kopplung des Rezeptors an einen G-Protein-gekoppelten einw{\"a}rtsgleichrichtenden Kaliumkanal in Oozyten des afrikanischen Krallenfrosches Xenopus laevis. Durch die Interaktion mit PSD-95 wird der SSTR4 in physikalische N{\"a}he zu bestimmten Zielproteinen gebracht, {\"u}ber die nachfolgend die Somatostatineffekte weitervermittelt werden. So erm{\"o}glicht die Interaktion vermutlich eine Integration des SSTR4 in den postsynaptischen Komplex aus PSD-95 und Glutamatrezeptoren, wo der SSTR4 die bereits beschrieben regulatorischen Effekte auf die Glutamat-vermittelte exzitatorische Signaltransduktion aus{\"u}ben kann.}, subject = {Ratte}, language = {de} } @phdthesis{Colnot2001, author = {Colnot, Thomas}, title = {Beurteilung von Phyto- und Xeno{\"o}strogenen am Beispiel ausgew{\"a}hlter Substanzen}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-1180438}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2001}, abstract = {Bei Daidzein und Bisphenol A handelt es sich um zwei Vertreter einer Klasse von Stoffen, die als „Umwelthormone" (engl. endocrine disrupter) bezeichnet werden. Aus der Gruppe der Phyto{\"o}strogene wurde Daidzein als wichtiger Vertreter, der in hohen Konzentrationen in vielen Nutzpflanzen und Nahrungsmitteln vorkommt, ausgew{\"a}hlt. Sojaprodukte, die den gr{\"o}ßten Beitrag einer menschlichen Exposition gegen Daidzein liefern, werden in zunehmendem Maße auch in westlichen L{\"a}ndern konsumiert. Bisphenol A wurde als Vertreter der Xeno{\"o}strogene gew{\"a}hlt, da es - was Weltjahresproduktion und Verwendung angeht - die wohl wichtigste Substanz dieser Gruppe darstellt. Im ersten Teil der Arbeit wurde die Biotransformation und Toxikokinetik der beiden Verbindungen nach oraler Gabe in der Ratte aufgekl{\"a}rt. Dabei konnte gezeigt werden, daß die orale Bioverf{\"u}gbarkeit beider Substanzen in der Ratte sehr gering war. Maximal zehn Prozent der jeweils applizierten Dosis konnten im Urin der Tiere wiedergefunden werden. Als Hauptmetabolit wurden sowohl von Daidzein als auch von Bisphenol A das jeweilige Glucuronid-Konjugat gebildet. Bei Daidzein {\"u}berwog in der m{\"a}nnlichen Ratte zus{\"a}tzlich das Sulfat-Konjugat. Der Anteil an freier, d.h. unkonjugierter Verbindung betrug im Urin der Tiere zwischen 1 und 3 Prozent der Dosis. Außer den Phase II-Konjugaten, die aufgrund ihrer mangelnden {\"o}strogenen Wirksamkeit zu einer Detoxifizierung der beiden Verbindungen f{\"u}hrte, konnten nach Gabe von Bisphenol A in der Ratte keine weiteren Metabolite identifiziert werden. Nach Exposition mit Daidzein konnten in den Faeces der Tiere in geringem Umfang die beiden reduktiven Metabolite Equol und O-DMA gefunden werden. Diese wurden wahrscheinlich im Magen-Darm-Trakt durch die Bakterien der Darmflora gebildet. Sowohl Daidzein als auch Bisphenol A wurden bei der Ratte nur unvollst{\"a}ndig aus dem Magen-Darm-Trakt resorbiert; der Großteil der gegebenen Dosis wurde als unver{\"a}nderte Substanz in den Faeces wiedergefunden. Bei Bisphenol A wurde die Ausscheidung zudem durch einen ausgepr{\"a}gten enterohepatischen Kreislauf verz{\"o}gert. Im zweiten Teil der Arbeit wurden zun{\"a}chst empfindliche GC/MS- und HPLC-Methoden zur Quantifizierung der Verbindungen in humanen Plasma- und Urinproben entwickelt. Danach wurden freiwillige Probanden oral mit jeweils 5 mg Daidzein bzw. d16-Bisphenol A exponiert, um Daten zur Biotransformation und Toxikokinetik der beiden Substanzen im Mensch zu erhalten. Wegen des deutlich meßbaren Hintergrundes an Bisphenol A, das in allen Kontrollproben nachweisbar war, wurde f{\"u}r die Humanstudie die deuterierte Verbindung gegeben, f{\"u}r die kein st{\"o}render Hintergrund meßbar war. Die Bioverf{\"u}gbarkeit der Gesamt-Substanz (freie Verbindung + Konjugate) im Menschen war in beiden F{\"a}llen deutlich h{\"o}her als in der Ratte. Von Daidzein wurden 40 Prozent (Ratte 10 Prozent), von Bisphenol A > 95 Prozent (Ratte 13 Prozent) der applizierten Dosis im Urin der Probanden wiedergefunden. Dabei zeigte sich ein sehr effizienter Phase II-Metabolismus; weniger als 1 Prozent der Glucuronid-Konjugatkonzentrationen wurden als unver{\"a}nderte Substanz gefunden. Das Glucuronid stellte in beiden F{\"a}llen den einzigen nachweisbaren Metaboliten dar. Die Elimination von Daidzein und Bisphenol A verlief in den beiden Studien sehr schnell nach einer Kinetik erster Ordnung. Im Gegensatz zu der Ratte konnten auch bei Bisphenol A keine Auff{\"a}lligkeiten in den Ausscheidungskurven beobachtet werden, Hinweise auf einen enterohepatischen Kreislauf im Menschen wurden nicht gefunden. Im Falle von Bisphenol A wurde fast die komplette applizierte Dosis (> 95 Prozent) in Form des Glucuronides im Urin wiedergefunden. Anhand der erhobenen Daten wurde anschließend eine Beurteilung des Risikos f{\"u}r den Menschen abgegeben.}, subject = {Phyto{\"o}strogen}, language = {de} } @incollection{DasZografakisOeljeklausetal.2023, author = {Das, Hirakjyoti and Zografakis, Alexandros and Oeljeklaus, Silke and Warscheid, Bettina}, title = {Analysis of Yeast Peroxisomes via Spatial Proteomics}, series = {Peroxisomes}, booktitle = {Peroxisomes}, edition = {accepted version}, publisher = {Springer}, doi = {10.1007/978-1-0716-3048-8_2}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-327532}, publisher = {Universit{\"a}t W{\"u}rzburg}, pages = {13-31}, year = {2023}, abstract = {Peroxisomes are ubiquitous organelles with essential functions in numerous cellular processes such as lipid metabolism, detoxification of reactive oxygen species and signaling. Knowledge of the peroxisomal proteome including multi-localized proteins and, most importantly, changes of its composition induced by altering cellular conditions or impaired peroxisome biogenesis and function is of paramount importance for a holistic view on peroxisomes and their diverse functions in a cellular context. In this chapter, we provide a spatial proteomics protocol specifically tailored to the analysis of the peroxisomal proteome of baker's yeast that enables the definition of the peroxisomal proteome under distinct conditions and to monitor dynamic changes of the proteome including the relocation of individual proteins to a different cellular compartment. The protocol comprises subcellular fractionation by differential centrifugation followed by Nycodenz density gradient centrifugation of a crude peroxisomal fraction, quantitative mass spectrometric measurements of subcellular and density gradient fractions and advanced computational data analysis, resulting in the establishment of organellar maps on a global scale.}, language = {en} } @article{DeGiorgiBuonaguroWorschechetal.2013, author = {De Giorgi, Valeria and Buonaguro, Luigi and Worschech, Andrea and Tornesello, Maria Lina and Izzo, Francesco and Marincola, Francesco M. and Wang, Ena and Buonaguro, Franco M.}, title = {Molecular Signatures Associated with HCV-Induced Hepatocellular Carcinoma and Liver Metastasis}, series = {PLoS ONE}, volume = {8}, journal = {PLoS ONE}, number = {2}, doi = {10.1371/journal.pone.0056153}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-131155}, pages = {e56153}, year = {2013}, abstract = {Hepatocellular carcinomas (HCCs) are a heterogeneous group of tumors that differ in risk factors and genetic alterations. In Italy, particularly Southern Italy, chronic hepatitis C virus (HCV) infection represents the main cause of HCC. Using high-density oligoarrays, we identified consistent differences in gene-expression between HCC and normal liver tissue. Expression patterns in HCC were also readily distinguishable from those associated with liver metastases. To characterize molecular events relevant to hepatocarcinogenesis and identify biomarkers for early HCC detection, gene expression profiling of 71 liver biopsies from HCV-related primary HCC and corresponding HCV-positive non-HCC hepatic tissue, as well as gastrointestinal liver metastases paired with the apparently normal peri-tumoral liver tissue, were compared to 6 liver biopsies from healthy individuals. Characteristic gene signatures were identified when normal tissue was compared with HCV-related primary HCC, corresponding HCV-positive non-HCC as well as gastrointestinal liver metastases. Pathway analysis classified the cellular and biological functions of the genes differentially expressed as related to regulation of gene expression and post-translational modification in HCV-related primary HCC; cellular Growth and Proliferation, and Cell-To-Cell Signaling and Interaction in HCV-related non HCC samples; Cellular Growth and Proliferation and Cell Cycle in metastasis. Also characteristic gene signatures were identified of HCV-HCC progression for early HCC diagnosis. Conclusions: A diagnostic molecular signature complementing conventional pathologic assessment was identified.}, language = {en} } @article{DenkSchmidtSchurretal.2021, author = {Denk, S. and Schmidt, S. and Schurr, Y. and Schwarz, G. and Schote, F. and Diefenbacher, M. and Armendariz, C. and Dejure, F. and Eilers, M. and Wiegering, Armin}, title = {CIP2A regulates MYC translation (via its 5′UTR) in colorectal cancer}, series = {International Journal of Colorectal Disease}, volume = {36}, journal = {International Journal of Colorectal Disease}, number = {5}, doi = {10.1007/s00384-020-03772-y}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-280092}, pages = {911-918}, year = {2021}, abstract = {Background Deregulated expression of MYC is a driver of colorectal carcinogenesis, suggesting that decreasing MYC expression may have significant therapeutic value. CIP2A is an oncogenic factor that regulates MYC expression. CIP2A is overexpressed in colorectal cancer (CRC), and its expression levels are an independent marker for long-term outcome of CRC. Previous studies suggested that CIP2A controls MYC protein expression on a post-transcriptional level. Methods To determine the mechanism by which CIP2A regulates MYC in CRC, we dissected MYC translation and stability dependent on CIP2A in CRC cell lines. Results Knockdown of CIP2A reduced MYC protein levels without influencing MYC stability in CRC cell lines. Interfering with proteasomal degradation of MYC by usage of FBXW7-deficient cells or treatment with the proteasome inhibitor MG132 did not rescue the effect of CIP2A depletion on MYC protein levels. Whereas CIP2A knockdown had marginal influence on global protein synthesis, we could demonstrate that, by using different reporter constructs and cells expressing MYC mRNA with or without flanking UTR, CIP2A regulates MYC translation. This interaction is mainly conducted by the MYC 5′UTR. Conclusions Thus, instead of targeting MYC protein stability as reported for other tissue types before, CIP2A specifically regulates MYC mRNA translation in CRC but has only slight effects on global mRNA translation. In conclusion, we propose as novel mechanism that CIP2A regulates MYC on a translational level rather than affecting MYC protein stability in CRC.}, language = {en} } @phdthesis{Dill2012, author = {Dill, Holger}, title = {Functional characterization of the microRNA-26 family in zebrafish neurogenesis}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-70757}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2012}, abstract = {Formation oft the central nervous system (CNS) from multipotent neuronal stem cells (NSCs) requires a tightly controlled, step-wise activation of the neuronal gene expression program. Expression of neuronal genes at the transition from neural stem cell to mature neuron (i. e. neuronal cell differentiation) is controlled by the Repressor element 1 (RE1) silencing transcription factor (REST) complex. As a master transcriptional regulator, the REST-complex specifically inhibits expression of neuronal genes in non-neuronal tissues and neuronal progenitor cells. Differentiation of NSCs to mature neurons requires the activation of genes controlled by the REST-complex, but how abrogation of REST-complex mediated repression is achieved during neurogenesis is only poorly understood. MicroRNAs (miRNAs) are a class of small regulatory RNAs that posttranscriptionally control target gene expression. Binding of miRNAs to target sequences in the 3'UTR of mRNAs, leads either to degradation or translational inhibition of the mRNA. Distinct neuronal miRNAs (e.g. miR-124) were shown to modulate REST-complex activity by silencing expression of REST-complex components. Interestingly, these miRNAs are also under transcriptional control of the REST-complex and inactivation of the REST-complex precedes their expression. Hence, additional factors are required for derepression of neuronal genes at the onset of neurogenesis. In this study function of the miR-26 family during neurogenesis of the zebrafish (Danio rerio) was analyzed. Computational target prediction revealed a number of REST-complex components as putative miR-26 targets. One of these predicted target genes, the C-terminal domain small phosphatase 2 (Ctdsp2) was validated as an in vivo target for miR-26b. Ctdsps are important cofactors of REST and suppress neuronal gene expression by dephosphorylating the C-terminal domain (CTD) of RNA polymerase II (Pol II). Interestingly, miR-26b is encoded in an intron of the ctdsp2 primary transcript and is cotranscribed together with its host gene. Hence, miR-26b modulates expression of its host gene ctdsp2 in an intrinsic negative autoregulatory loop. This negative autoregulatory loop is inactive in NSCs because miR-26b biogenesis is inhibited at the precursor level. Generation of mature miR-26b is activated during neurogenesis, where it suppresses Ctdsp2 protein expression and is required for neuronal cell differentiation in vivo. Strikingly, miR-26b is expressed prior to miR-124 during neuronal cell differentiation. Thus, it is reasonable to speculate about a function of miR-26b in early events of neurogenesis. In line with this assumption, knockdown of miR-26b in zebrafish embryos results in downregulation of REST-complex controlled neuronal genes and a block in neuronal cell differentiation, most likely due to aberrant regulation of Ctdsp2 expression. This is evident by reduced numbers of secondary motor neurons compared to control siblings. In contrast, motor neuron progenitor cells and glia cells were not affected by depletion of miR-26b.This study identifies the ctdsp2/miR-26b autoregulatory loop as the first experimentally validated interaction between an intronic miRNA and its host gene transcript. Silencing of ctdsp2 by miR-26b in neurons is possible because biogenesis of the ctdsp2 mRNA and mature mir-26b is uncoupled at the posttranscriptional level. Furthermore the obtained data indicate a cell type specific role for miR-26b in vertebrate neurogenesis and CNS development.}, subject = {Zebrab{\"a}rbling}, language = {en} } @article{DonatRotherSchaeferetal.2014, author = {Donat, Ulrike and Rother, Juliane and Sch{\"a}fer, Simon and Hess, Michael and H{\"a}rtl, Barbara and Kober, Christina and Langbein-Laugwitz, Johanna and Stritzker, Jochen and Chen, Nanhai G. and Aguilar, Richard J. and Weibel, Stephanie and Szalay, Alandar A.}, title = {Characterization of Metastasis Formation and Virotherapy in the Human C33A Cervical Cancer Model}, series = {PLoS ONE}, volume = {9}, journal = {PLoS ONE}, number = {6}, issn = {1932-6203}, doi = {10.1371/journal.pone.0098533}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-119674}, pages = {e98533}, year = {2014}, abstract = {More than 90\% of cancer mortalities are due to cancer that has metastasized. Therefore, it is crucial to intensify research on metastasis formation and therapy. Here, we describe for the first time the metastasizing ability of the human cervical cancer cell line C33A in athymic nude mice after subcutaneous implantation of tumor cells. In this model, we demonstrated a steady progression of lumbar and renal lymph node metastases during tumor development. Besides predominantly occurring lymphatic metastases, we visualized the formation of hematogenous metastases utilizing red fluorescent protein (RFP) expressing C33A-RFP cells. RFP positive cancer cells were found migrating in blood vessels and forming micrometastases in lungs of tumor-bearing mice. Next, we set out to analyze the influence of oncolytic virotherapy in the C33A-RFP model and demonstrated an efficient virus-mediated reduction of tumor size and metastatic burden. These results suggest the C33A-RFP cervical cancer model as a new platform to analyze cancer metastases as well as to test novel treatment options to combat metastases.}, language = {en} } @article{DraganovSantidrianMinevetal.2019, author = {Draganov, Dobrin D. and Santidrian, Antonio F. and Minev, Ivelina and Duong, Nguyen and Kilinc, Mehmet Okyay and Petrov, Ivan and Vyalkova, Anna and Lander, Elliot and Berman, Mark and Minev, Boris and Szalay, Aladar A.}, title = {Delivery of oncolytic vaccinia virus by matched allogeneic stem cells overcomes critical innate and adaptive immune barriers}, series = {Journal of Translational Medicine}, volume = {17}, journal = {Journal of Translational Medicine}, issn = {100}, doi = {10.1186/s12967-019-1829-z}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-226312}, year = {2019}, abstract = {Background Previous studies have identified IFNγ as an important early barrier to oncolytic viruses including vaccinia. The existing innate and adaptive immune barriers restricting oncolytic virotherapy, however, can be overcome using autologous or allogeneic mesenchymal stem cells as carrier cells with unique immunosuppressive properties. Methods To test the ability of mesenchymal stem cells to overcome innate and adaptive immune barriers and to successfully deliver oncolytic vaccinia virus to tumor cells, we performed flow cytometry and virus plaque assay analysis of ex vivo co-cultures of stem cells infected with vaccinia virus in the presence of peripheral blood mononuclear cells from healthy donors. Comparative analysis was performed to establish statistically significant correlations and to evaluate the effect of stem cells on the activity of key immune cell populations. Results Here, we demonstrate that adipose-derived stem cells (ADSCs) have the potential to eradicate resistant tumor cells through a combination of potent virus amplification and sensitization of the tumor cells to virus infection. Moreover, the ADSCs demonstrate ability to function as a virus-amplifying Trojan horse in the presence of both autologous and allogeneic human PBMCs, which can be linked to the intrinsic immunosuppressive properties of stem cells and their unique potential to overcome innate and adaptive immune barriers. The clinical application of ready-to-use ex vivo expanded allogeneic stem cell lines, however, appears significantly restricted by patient-specific allogeneic differences associated with the induction of potent anti-stem cell cytotoxic and IFNγ responses. These allogeneic responses originate from both innate (NK)- and adaptive (T)- immune cells and might compromise therapeutic efficacy through direct elimination of the stem cells or the induction of an anti-viral state, which can block the potential of the Trojan horse to amplify and deliver vaccinia virus to the tumor. Conclusions Overall, our findings and data indicate the feasibility to establish simple and informative assays that capture critically important patient-specific differences in the immune responses to the virus and stem cells, which allows for proper patient-stem cell matching and enables the effective use of off-the-shelf allogeneic cell-based delivery platforms, thus providing a more practical and commercially viable alternative to the autologous stem cell approach.}, language = {en} } @article{DuggalGeissingerZhangetal.2013, author = {Duggal, Rohit and Geissinger, Ulrike and Zhang, Qian and Aguilar, Jason and Chen, Nanhai G. and Binda, Elena and Vescovi, Angelo L. and Szalay, Aladar A.}, title = {Vaccinia virus expressing bone morphogenetic protein-4 in novel glioblastoma orthotopic models facilitates enhanced tumor regression and long-term survival}, series = {Journal of Translational Medicine}, volume = {11}, journal = {Journal of Translational Medicine}, number = {155}, doi = {10.1186/1479-5876-11-155}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-129626}, year = {2013}, abstract = {No abstract availableBackground: Glioblastoma multiforme (GBM) is one of the most aggressive forms of cancer with a high rate of recurrence. We propose a novel oncolytic vaccinia virus (VACV)-based therapy using expression of the bone morphogenetic protein (BMP)-4 for treating GBM and preventing recurrence. Methods: We have utilized clinically relevant, orthotopic xenograft models of GBM based on tumor-biopsy derived, primary cancer stem cell (CSC) lines. One of the cell lines, after being transduced with a cDNA encoding firefly luciferase, could be used for real time tumor imaging. A VACV that expresses BMP-4 was constructed and utilized for infecting several primary glioma cultures besides conventional serum-grown glioma cell lines. This virus was also delivered intracranially upon implantation of the GBM CSCs in mice to determine effects on tumor growth. Results: We found that the VACV that overexpresses BMP-4 demonstrated heightened replication and cytotoxic activity in GBM CSC cultures with a broad spectrum of activity across several different patient-biopsy cultures. Intracranial inoculation of mice with this virus resulted in a tumor size equal to or below that at the time of injection. This resulted in survival of 100\% of the treated mice up to 84 days post inoculation, significantly superior to that of a VACV lacking BMP-4 expression. When mice with a higher tumor burden were injected with the VACV lacking BMP-4, 80\% of the mice showed tumor recurrence. In contrast, no recurrence was seen when mice were injected with the VACV expressing BMP-4, possibly due to induction of differentiation in the CSC population and subsequently serving as a better host for VACV infection and oncolysis. This lack of recurrence resulted in superior survival in the BMP-4 VACV treated group. Conclusions: Based on these findings we propose a novel VACV therapy for treating GBM, which would allow tumor specific production of drugs in the future in combination with BMPs which would simultaneously control tumor maintenance and facilitate CSC differentiation, respectively, thereby causing sustained tumor regression without recurrence.}, language = {en} } @phdthesis{Eggert2005, author = {Eggert, Christian}, title = {Untersuchungen zur Biogenese spleißosomaler UsnRNPs und ihrer Bedeutung f{\"u}r die Pathogenese der SMA}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-15334}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2005}, abstract = {Die neurodegenerative Krankheit Spinale Muskelatrophie (SMA) wird durch den Mangel an funktionellem Survival Motor Neuron Protein (SMN) verursacht. Eine Funktion von SMN liegt in der Biogenese spleißosomaler UsnRNPs (U-rich small nuclear ribonucleoprotein particles). Diese Arbeit zeigt in einem SMA-Modell in Hela-Zellkultur, dass der SMN-Mangel zu einer reduzierten de novo-Produktion der spleißosomalen UsnRNPs f{\"u}hrt. In einem Zebrafisch-Modell f{\"u}r SMA wurde nachgewiesen, dass die reduzierte UsnRNP-Produktion die Degenerationen von Axonen der Motoneuronen verursacht, einen Ph{\"a}notyp wie er bei SMA auftritt. Damit konnte erstmals eine direkte Verbindung zwischen einer zellul{\"a}ren Funktion von SMN und der Entstehung von SMA hergestellt werden.}, subject = {Spinale Muskelatrophie}, language = {de} } @phdthesis{Ehrig2012, author = {Ehrig, Klaas}, title = {Effects of stem cell transcription factor-expressing vaccinia viruses in oncolytic virotherapy}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-85139}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2012}, abstract = {Krebserkrankungen bleiben auch im Jahr 2012 die zweith{\"a}uftigste Todesursache in der industralisierten Welt. Zus{\"a}tzlich hat die Etablierung der Krebsstammzell-Hypothese grunds{\"a}tzliche Auswirkungen auf die Erfolgsaussichten konventioneller Krebstherapie, wie Chemotherapie oder Strahlentherapie. Deswegen ist es von gr{\"o}βter Notwendigkeit, dass neue Ans{\"a}tze zur Krebstherapie entwickelt werden, die den Ausgang der Behandlung verbessern und zu weniger Nebenwirkungen f{\"u}hren. Diverse vorklinische Studien haben gezeigt, dass die onkolytische Virotherapie mit Vaccinia-Viren ein potentes und gut tolerierbares neues Werkzeug in der Krebstherapie darstellt. Die Effizienz des Vaccinia-Virus als Therapeutikum allein oder in Kombination mit Strahlen- oder Chemotherapie wird aktuell in mehreren klinischen Studien der Phasen I \& II getestet. Krebsstammzellen und Stammzellen teilen eine Vielzahl von Eigenschaften, wie die F{\"a}higkeit zur Selbst-Erneuerung und Pluripotenz, Stilllegung der Zellproliferation, Resistenz gegen Medikamente oder Bestrahlung, die Expression von diversen Zelloberfl{\"a}chen-molek{\"u}len, die Aktivierung und Hemmung spezifischer Signaltransduktionswege oder die Expression von Stammzell-spezifischen Genen. In dieser Arbeit wurden zwei neue rekombinante Vaccinia-Viren entwickelt, welche die Stammzell-Transkriptionsfaktoren Nanog (GLV-1h205) und Oct4 (GLV-1h208) exprimieren, um tiefere Einblicke in die Rolle dieser Masterregulatoren in der Entstehung von Krebs und ihrem Einfluss auf die onkolytische Virotherapie zu gewinnen. Das Replikationspotential beider Virusst{\"a}mme in menschlichen A549-Zellen und PC-3-Zellen wurde anhand von Replikations-Assays bestimmt. Die Expression der Virus-spezifischen Markergene Ruc-GFP und beta-Galaktosidase, wie auch die Expression der Transkriptionsfaktoren Nanog und Oct4 wurde mit Hilfe von RT-PCR, SDS-PAGE und Western blotting, sowie immunozytochemischen Experimenten nachgewiesen. Des Weiteren wurde der Einfluss einer GLV-1h205-Infektion von A549-Zellen auf den Zellzyklus untersucht. Zudem wurde die Bedeutung der Virus-vermittelten Transkriptionsfaktor-Expression auf die Behandlung von subkutanen A549-Tumoren in einem Xenograft-Modell untersucht. Zur Untersuchung, ob die beobachteten Vorteile in der Behandlung von Lungenadenokarzinomen in M{\"a}usen mit GLV-1h205 Promoter- oder Transkriptionsfaktor-abh{\"a}ngig sind, wurde ein Kontroll-Virus (GLV-1h321) hergestellt, dass f{\"u}r eine unfunktionale Nanog-Mutante codiert. Mittels SDS-PAGE und Western blotting sowie Immunozytochemie wurde die Transgen-Expression analysiert. Ein weitere Aspekt dieser Arbeit war die Fragestellung, ob sich das onkolyische Vaccinia-Virus GLV-1h68 eignet, als neues und weniger invasives Therapeutikum effizient Darmkrebszellen zu infizieren um sich in ihnen zu replizieren und diese anschlieβend zu lysieren. Ein derartiger Therapieansatz w{\"u}rde besonders im Hinblick auf sp{\"a}t diagnostizierten, metastasierenden Darmkrebs eine interessante Behandlungsalternative darstellen. Virale Markergen-expression wurde anhand von Fluoreszenzmikroskopie und FACS-Analyse untersucht. Desweiteren wurde gezeigt, dass die einmalige Administration von GLV-1h68 in mindestens zwei verschiedenen Darmkrebszelllinien zu einer signifikanten Inhibierung des Tumorwachstums in vivo und zu signifikant verbessertem {\"U}berleben f{\"u}hrt. Der Transkriptionsfaktor Klf4 wird zwar stark in ruhenden, ausdifferenzierten Zellen des Darmepithels exprimiert, ist hingegen bei Darmkrebs generell dramatisch herabreguliert. Die Expression von Klf4 f{\"u}hrt zu einem Stop der Zellproliferation und inhibiert die Aktivit{\"a}t des Wnt-Signalweges, indem es im Zellkern an die Transaktivierungsdom{\"a}ne von beta-Catenin bindet. Um die Behandlung von Darmkrebs mit Hilfe onkolytischer Virotherapie weiter zu verbessern, wurden verschiedene Vaccinia-Viren (GLV-1h290-292) erzeugt, die durch verschiedene Promoterst{\"a}rken die Expression unterschiedlicher Mengen an Tumorsuppressor Klf4 vermitteln. Die anf{\"a}ngliche Charakterisierung der drei Virusst{\"a}mme mittels Replikations-Assay, Zytotoxizit{\"a}tstudien, SDS-PAGE und Western blotting, Immunozytochemie sowie die Analyse der Proteinfunktion mit Hilfe von qPCR- und ELISA-Analysen zur Bestimmung von zellul{\"a}rem beta-Catenin, zeigten eine Promoter-abh{\"a}ngige Expression und Wirkung von Klf4. F{\"u}r weitere Analysen wurde das Virus GLV-1h291 gew{\"a}hlt, welches nach Infektion die gr{\"o}βte Menge an Klf4 produziert und zus{\"a}tzlich durch die C-terminale Fusion einer TAT Transduktionsdom{\"a}ne Membran-g{\"a}ngig gemacht (GLV-1h391). Die erhaltenen Befunde machen das Klf4-TAT-kodierende Vaccinia-Virus GLV-1h391 zu einem vielversprechenden Kandidaten f{\"u}r eine Behandlung von Darmkrebs beim Menschen.}, subject = {Lungenkrebs}, language = {en} } @article{EhrigKilincChenetal.2013, author = {Ehrig, Klaas and Kilinc, Mehmet O. and Chen, Nanhai G. and Stritzker, Jochen and Buckel, Lisa and Zhang, Qian and Szalay, Aladar A.}, title = {Growth inhibition of different human colorectal cancer xenografts after a single intravenous injection of oncolytic vaccinia virus GLV-1h68}, series = {Journal of Translational Medicine}, volume = {11}, journal = {Journal of Translational Medicine}, number = {79}, doi = {10.1186/1479-5876-11-79}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-129619}, year = {2013}, abstract = {Background: Despite availability of efficient treatment regimens for early stage colorectal cancer, treatment regimens for late stage colorectal cancer are generally not effective and thus need improvement. Oncolytic virotherapy using replication-competent vaccinia virus (VACV) strains is a promising new strategy for therapy of a variety of human cancers. Methods: Oncolytic efficacy of replication-competent vaccinia virus GLV-1h68 was analyzed in both, cell cultures and subcutaneous xenograft tumor models. Results: In this study we demonstrated for the first time that the replication-competent recombinant VACV GLV-1h68 efficiently infected, replicated in, and subsequently lysed various human colorectal cancer lines (Colo 205, HCT-15, HCT-116, HT-29, and SW-620) derived from patients at all four stages of disease. Additionally, in tumor xenograft models in athymic nude mice, a single injection of intravenously administered GLV-1h68 significantly inhibited tumor growth of two different human colorectal cell line tumors (Duke's type A-stage HCT-116 and Duke's type C-stage SW-620), significantly improving survival compared to untreated mice. Expression of the viral marker gene ruc-gfp allowed for real-time analysis of the virus infection in cell cultures and in mice. GLV-1h68 treatment was well-tolerated in all animals and viral replication was confined to the tumor. GLV-1h68 treatment elicited a significant up-regulation of murine immune-related antigens like IFN-γ, IP-10, MCP-1, MCP-3, MCP-5, RANTES and TNF-γ and a greater infiltration of macrophages and NK cells in tumors as compared to untreated controls. Conclusion: The anti-tumor activity observed against colorectal cancer cells in these studies was a result of direct viral oncolysis by GLV-1h68 and inflammation-mediated innate immune responses. The therapeutic effects occurred in tumors regardless of the stage of disease from which the cells were derived. Thus, the recombinant vaccinia virus GLV-1h68 has the potential to treat colorectal cancers independently of the stage of progression.}, language = {en} } @phdthesis{Englert2005, author = {Englert, Markus}, title = {Mechanismus des pre-tRNA-Spleißens : Struktur und Funktion pflanzlicher und animaler RNA-Ligasen}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-14420}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2005}, abstract = {Transfer Ribonukleins{\"a}uren werden von der RNA Polymerase III als Vorl{\"a}ufer tRNA transkribiert und durchlaufen eine Vielzahl von Reifungsschritten hin zur maturen tRNA. Neben der Hydrolyse der 5´- und 3´-Flanke durch die RNase P und die tRNase Z, sowie einer Vielzahl von Basenmodifizierungen, wird bei einigen pre-tRNAs das Intron herausgespleißt. Die ersten intronhaltigen tRNA Gene wurden in der Hefe Saccharomyces cerevisiae nachgewiesen und folglich wurde der Spleißmechanismus in diesem Organismus als erstes untersucht. Eine tetramere tRNA Spleißendonuklease spaltet das Intron an den Exongrenzen heraus und eine tRNA Ligase ligiert die entstandenen tRNA H{\"a}lften zur gespleißten tRNA. Einzig in der Hefe und anderen Pilzen konnten bisher die Gene f{\"u}r die tRNA Ligase identifiziert werden. Weder molekularbiologische Ans{\"a}tze - wie z.B. DNA Hybridisierung, Expressions-"Screening" und funktionelle Komplementationsstudien mit einem tRNA Ligase-defizienten Hefestamm - noch Datenbanksuchen mit der bekannten Hefe tRNA Ligasesequenz haben in den vergangenen Jahren zur Identifizierung eines pflanzlichen oder animalen tRNA Ligase Gens gef{\"u}hrt. In dieser Arbeit ist es erstmals gelungen, das tRNA Ligase Protein aus Weizenkeimen bis zur Homogenit{\"a}t zu isolieren und mit Hilfe erhaltener Peptidsequenzen die entsprechenden Kern-codierten Gene in h{\"o}heren und niederer Pflanzen zu identifizieren. Die Ligaseaktivit{\"a}t wurde f{\"u}r das klonierte, rekombinant {\"u}berexprimierte tRNA Ligaseprotein best{\"a}tigt. Weiterhin wurde zum ersten Mal das Ligaseprotein aus Schweineleber aufgereinigt und das zugeh{\"o}rige Gen im humanen Genom identifiziert.}, subject = {Pflanzen}, language = {de} } @phdthesis{Faul2004, author = {Faul, Thomas}, title = {Lokalisation und Dynamik der Replikationsproteine des murinen pr{\"a}-replikativen Komplexes}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-10473}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2004}, abstract = {Ein Ziel der vorliegenden Arbeit war die Untersuchung der Lokalisation und der Dynamik der Replikationsproteine des murinen pr{\"a}-replikativen Komplexes in vivo. Dazu wurden die zu untersuchenden Replikationsproteine als EGFP-Fusionsproteine in LTK--Zellen exprimiert und am konfokalen Laserscanning-Mikroskop untersucht. CDC6-EGFP war in der G1-Phase diffus in Zellkern und Cytoplasma verteilt, am G1/S-{\"U}bergang ausschließlich im Zellkern lokalisiert und w{\"a}hrend der S-Phase in zahlreichen Foci im Kern akkumuliert. CDC6-EGFP war mit Replikationsfoci colokalisiert. Endogenes Cdc6p wies dieselbe subzellul{\"a}re Verteilung wie CDC6-EGFP auf. Auch Fusionsproteine des humanen Proteins Cdc6p waren in HEK-293T-Zellen in Replikationsfoci lokalisiert. FRAP-Studien ergaben, dass 80-90 \% von CDC6-EGFP w{\"a}hrend der gesamten S-Phase stabil mit der Replikationsmaschinerie assoziiert sind. Durch Mutation der Phosphoryliersstellen f{\"u}r Cyclin-abh{\"a}ngige Proteinkinasen wurde der Einfluss des Phosphorylierungsstatus der konservierten Serinreste der Cdk-Phosphorylierungsstellen auf die Lokalisation von CDC6-EGFP in vivo untersucht. Alle Mutanten bei denen die Cdk-Serinreste zu nicht-phosphorylierbaren Alaninresten mutiert wurden waren in Replikationsfoci lokalisiert. Dies zeigt, dass die Phosphorylierung dieser Serinreste f{\"u}r die Lokalisation von CDC6-EGFP an Stellen aktiver DNA-Replikation nicht essentiell ist. Durch Mutation der Serinreste zu Phosphatreste-simulierenden Aspartatresten konnte gezeigt werden, dass die Phosphorylierung des Serinrests S102 zum Export von CDC6-EGFP aus dem Zellkern f{\"u}hrt. FRAP-Studien ergaben, dass CDC6-EGFP in Replikationsfoci an Serinrest 82 phosphoryliert und an Serinrest 102 dephosphoryliert vorliegt. Mit Immunfluoreszenz-Analysen konnte gezeigt werden, dass Chromatin in Replikationsfoci nicht acetyliert ist. Dies deutet darauf hin, dass die Elongation der DNA-Replikation an nicht-acetyliertem Chromatin erfolgt. Trichostatin A-induzierte Hyperacetylierung des Chromatins hatte keinen Einfluss auf Lokalisation und Mobilit{\"a}t von CDC6-EGFP in Replikationsfoci. Die Mobilit{\"a}t des nucleoplasmatischen CDC6-EGFP-Pools wurde dadurch erh{\"o}ht. In der G1-Phase wurde die Mobilit{\"a}t von CDC6-EGFP durch TSA verringert, woraus gefolgert werden kann, dass der Acetylierungsstatus des Chromatins in der G1-Phase die Mobilit{\"a}t von CDC6-EGFP beeinflusst. ORC1-EGFP war im Zellkern in großen kugelf{\"o}rmigen Strukturen lokalisiert, ORC2-EGFP war diffus in Cytoplasma und Zellkern verteilt. ORC3-EGFP akkumulierte in PML nuclear bodies. W{\"a}hrend ORC4-EGFP und ORC5-EGFP am Centrosom lokalisiert waren konnte ORC6-EGFP in Nucleoli nachgewiesen werden. Die EGFP-Fusionsproteine von Cdc45p, PCNA und DNA-Ligase-I waren im Zellkern lokalisiert, die Nucleoli waren ausgespart. Ein weiterer Aspekt dieser Arbeit war die Untersuchung der Substratspezifit{\"a}t der murinen Cdc7p/Dbf4p-Proteinkinase. Die in Sf9-Zellen exprimierte und aufgereinigte Kinase phosphorylierte Orc2p, Orc6p, Cdc45p und Mcm6p. Mit Phosphopeptidkartierungen konnte gezeigt werden, dass Cdc7p von CylinE/Cdk2 an zwei Stellen und von CyclinA/Cdk2 an einer Stelle in vitro phosphoryliert wird. CDC7-EGFP war in der G1-Phase, am G1/S-{\"U}bergang und in der S-Phase im Kern lokalisiert. Durch FISH-Experimente konnte der genomische Locus des murinen Cdc7-Gens der Bande E von Chromosom 5 zugeordnet werden. Mit Kinase-Assays wurde untersucht, ob die murine Plk1p-Kinase Initiationsfaktoren der DNA-Replikation in vitro phosphoryliert. Die in Sf9-Zellen exprimierte Plk1p phosphorylierte Cdc7p, Orc2p und Orc6p. Cdc7p und Orc6p sind mit Plk1p am Midbody w{\"a}hrend der Telophase in vivo colokalisiert. Ein weiteres Ziel dieser Arbeit war die Messung der Mobilit{\"a}t des murinen Transkriptions-Terminationsfaktors TTF-I mittels FRAP. EGFP-TTF-I und EGFP-NRD waren diffus in den Nucleoli verteilt, einzelne Areale waren ausgespart. EGFP-TTFdeltaN185 war hingegen in distinkten nucleol{\"a}ren Stellen akkumuliert. Mit FRAP-Studien konnte gezeigt werden, dass EGFP-TTFdeltaN185 in einer 10 \%igen immobilen Fraktion vorlag w{\"a}hrend das Gesamtprotein EGFP-TTF-I zu 100\% mobil war. Das Protein TIP5 interagiert mit TTF-I. EGFP-TIP5 war diffus im Nucleoplasma verteilt, die Ncleoli waren ausgespart. Durch Cotransfektionen verschiedener EYFP-TTF-I-Konstrukte mit EGFP-TIP5 konnte gezeigt werden, dass EGFP-TIP5 von EYFP-TTFdeltaN185 nicht in Nucleoli cotransportiert wird. Mit BRET-Studien ergaben, dass Orc6p mit TTF-I in vivo interagiert. Eine Interaktion mit TTFdeltaN185 war nicht nachweisbar.}, subject = {Maus}, language = {de} } @article{FetivaLissGertzmannetal.2023, author = {Fetiva, Maria Camila and Liss, Franziska and Gertzmann, D{\"o}rthe and Thomas, Julius and Gantert, Benedikt and Vogl, Magdalena and Sira, Nataliia and Weinstock, Grit and Kneitz, Susanne and Ade, Carsten P. and Gaubatz, Stefan}, title = {Oncogenic YAP mediates changes in chromatin accessibility and activity that drive cell cycle gene expression and cell migration}, series = {Nucleic Acids Research}, volume = {51}, journal = {Nucleic Acids Research}, number = {9}, doi = {10.1093/nar/gkad107}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-350218}, pages = {4266-4283}, year = {2023}, abstract = {YAP, the key protein effector of the Hippo pathway, is a transcriptional co-activator that controls the expression of cell cycle genes, promotes cell growth and proliferation and regulates organ size. YAP modulates gene transcription by binding to distal enhancers, but the mechanisms of gene regulation by YAP-bound enhancers remain poorly understood. Here we show that constitutive active YAP5SA leads to widespread changes in chromatin accessibility in untransformed MCF10A cells. Newly accessible regions include YAP-bound enhancers that mediate activation of cycle genes regulated by the Myb-MuvB (MMB) complex. By CRISPR-interference we identify a role for YAP-bound enhancers in phosphorylation of Pol II at Ser5 at MMB-regulated promoters, extending previously published studies that suggested YAP primarily regulates the pause-release step and transcriptional elongation. YAP5SA also leads to less accessible 'closed' chromatin regions, which are not directly YAP-bound but which contain binding motifs for the p53 family of transcription factors. Diminished accessibility at these regions is, at least in part, a consequence of reduced expression and chromatin-binding of the p53 family member ΔNp63 resulting in downregulation of ΔNp63-target genes and promoting YAP-mediated cell migration. In summary, our studies uncover changes in chromatin accessibility and activity that contribute to the oncogenic activities of YAP.}, language = {en} } @phdthesis{Flegler2022, author = {Flegler, Vanessa Judith}, title = {Application of electron cryomicroscopy for structural and functional studies on the mechanosensitive channels of small conductance}, doi = {10.25972/OPUS-26897}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-268979}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2022}, abstract = {Bacteria thrive and survive in many different environments, and as a result, they have developed robust mechanisms to adapt rapidly to alterations in their surroundings. The protection against osmotic forces is provided by mechanosensitive channels: their primary function is to maintain the integrity of the cell upon a hypoosmotic shock. The mechanosensitive channel of small conductance (MscS) is not only the smallest common structural unit of a diverse family that allows for a tailored response in osmoregulation; it is also the most intensively studied homologue. Mechanosensitive channels directly sense elevated membrane tension levels generated by increased pressure within the cell and open transiently. Escherichia coli has six paralogues that differ in their gating properties and the number of additional transmembrane (TM) helices. These TM helices, termed sensor paddles, are essential for sensing, as they directly contact the surrounding membrane; however, the role of the additional TM helices is still unclear. Furthermore, lipids occupy hydrophobic pockets far away from the membrane plane. A recent gating model for MscS states that increased membrane tension triggers the expulsion of lipids out of those pockets, modulating different conformational states of MscS. This model focuses on bound lipids, but it is still unclear to what extent the direct interaction with the membrane influences sensing and how relevant it is for the larger paralogues. In the herein described work, structural studies on two larger paralogues, the medium-sized channel YnaI and the large channel YbiO were realised using electron cryomicroscopy (cryo-EM). Lipids were identified in YnaI in the pockets in a similar position and orientation as in MscS, suggesting a conserved sensing mechanism. Moreover, the copolymer diisobutylene/maleic acid (DIBMA) allowed the extraction of artificially activated YnaI from plasma membranes, leading to an open-like form of this channel. This novel conformation indicated that the pore helices bend at a GGxGG motif during gating, which is unique among the Escherichia coli paralogues, concomitant with a structural reorganisation of the sensor paddles. Thus, despite a high similarity of their closed states, the gating mechanisms of MscS and YnaI are surprisingly different. Furthermore, the comparison of MscS, YnaI, and YbiO accentuates variations and similarities between the differently sized family members, implying fine-tuning of channel properties in the pore regions and the cytosolic lateral entry sides into the channel. Structural analyses of MscS reconstituted into different systems showed the advantages and disadvantages of certain polymers and detergents. The novel DIBMA copolymer and the more conventional amphiphilic polymers, so-called Amphipols, perturb contacting transmembrane helices or lead to their denaturation. Due to this observation, the obtained structures of YnaI must also be cautiously considered. The structures obtained in detergents resulted in unaffected channels; however, the applicability of detergents for MscS-like channels is limited by the increased required sample concentration. The role of lipids for gating MscS in the absence of a membrane was examined by deliberately removing coordinated lipid molecules from MscS using different amounts and kinds of detergent. The effects on the channel were inspected by cryo-EM. These experiments showed that closed MscS adopts the open conformation when it is enough delipidated by incubation with the detergent n-dodecyl-β-D-maltoside, and adding lipids to the open channel reverses this process. The results agree with the state-of-the-art model that the amount of lipid molecules in the pockets and grooves is responsible for the conformational state of MscS. Furthermore, incubation with the detergent lauryl maltose neopentyl glycol, which has stabilising and delipidating characteristics, resulted in a high-resolution structure of open MscS exhibiting an intricate network of ligands. Based on this structure, an updated gating model is proposed, which states that upon opening, lipids from the pockets migrate into the cytosolic membrane leaflet, while lipids from the periplasmic leaflet enter the grooves that arise between the sensor paddles.}, language = {en} } @phdthesis{Geissinger2010, author = {Geissinger, Ulrike}, title = {Vaccinia Virus-mediated MR Imaging of Tumors in Mice: Overexpression of Iron-binding Proteins in Colonized Xenografts}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-48099}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2010}, abstract = {Vaccinia virus plays an important role in human medicine and molecular biology ever since the 18th century after E. Jenner discovered its value as a vaccination virus against smallpox. After the successful eradication of smallpox, vaccinia virus, apart from its use as a vaccine carrier, is today mainly used as a viral vector in molecular biology and increasingly in cancer therapy. The capability to specifically target and destroy cancer cells makes it a perfect agent for oncolytic virotherapy. Furthermore, the virus can easily be modified by inserting genes encoding therapeutic or diagnostic proteins to be expressed within the tumor. The emphasis in this study was the diagnosis of tumors using different vaccinia virus strains. Viruses with metal-accumulating capabilities for tumor detection via MRI technology were generated and tested for their usefulness in cell culture and in vivo. The virus strains GLV-1h131, GLV-1h132, and GLV-1h133 carry the gene encoding the two subunits of the iron storage protein ferritin under the control of three different promoters. GLV-1h110, GLV-1h111, and GLV-1h112 encode the bacterial iron storage protein bacterioferritin, whereas GLV-1h113 encodes the codon-optimized version of bacterioferritin for more efficient expression in human cells. GLV-1h22 contains the transferrin receptor gene, which plays an important role in iron uptake, and GLV-1h114 and GLV-1h115 contain the murine transferrin receptor gene. For possibly better iron uptake the virus strains GLV-1h154, GLV-1h155, GLV-1h156, and GLV-1h157 were generated, each with a version of a ferritin gene and a transferrin receptor gene. GLV-1h154 carries the genes that encode bacterioferritin and human transferrin receptor, GLV-1h155 the human ferritin H-chain gene and the human transferrin receptor gene. GLV-1h156 and GLV-1h157 infected cells both express the mouse transferrin receptor and bacterioferritin or human ferritin H-chain, respectively. The virus strains GLV-1h186 and GLV-1h187 were generated to contain a mutated form of the ferritin light chain, which was shown to result in iron overload and the wildtype light chain gene, respectively. The gene encoding the Divalent Metal Transporter 1, which is a major protein in the uptake of iron, was inserted in the virus strain GLV-1h102. The virus strain GLV-1h184 contains the magA gene of the magnetotactic bacterium Magnetospirillum magnetotacticum, which produces magnetic nanoparticles for orientation in the earth's magnetic field. Initially the infection and replication capability of all the virus strains were analyzed and compared to that of the parental virus strain GLV-1h68, revealing that all the viruses were able to infect cells of the human cancer cell lines A549 and GI-101A. All constructs exhibited a course of infection comparable to that of GLV-1h68. Next, to investigate the expression of the foreign proteins in GI-101A and A549 cells with protein analytical methods, SDS-gelelectrophoresis, Western blots and ELISAs were performed. The proteins, which were expressed under the control of the strong promoters, could be detected using these methods. To be able to successfully detect the protein expression of MagA and DMT1, which were expressed under the control of the weak promoter, the more sensitive method RT-PCR was used to at least confirm the transcription of the inserted genes. The determination of the iron content in infected GI-101A and A549 cells showed that infection with all used virus strains led to iron accumulation in comparison to uninfected cells, even infection with the parental virus strain GLV-1h68. The synthetic phytochelatin EC20 was also shown to enhance the accumulation of different heavy metals in bacterial cultures. In vivo experiments with A549 tumor-bearing athymic nude mice revealed that 24 days post infection virus particles were found mainly in the tumor. The virus-mediated expression of recombinant proteins in the tumors was detected successfully by Western blot. Iron accumulation in tumor lysates was investigated by using the ferrozine assay and led to the result that GLV-1h68-infected tumors had the highest iron content. Histological stainings confirmed the finding that iron accumulation was not a direct result of the insertion of genes encoding iron-accumulating proteins in the virus genome. Furthermore virus-injected tumorous mice were analyzed using MRI technology. Two different measurements were performed, the first scan being done with a seven Tesla small animal scanner seven days post infection whereas the second scan was performed using a three Tesla human scanner 21 days after virus injection. Tumors of mice injected with the virus strains GLV-1h113 and GLV-1h184 were shown to exhibit shortened T2 and T2* relaxation times, which indicates enhanced iron accumulation. In conclusion, the experiments in this study suggest that the bacterioferritin-encoding virus strain GLV-1h113 and the magA-encoding virus strain GLV-1h184 are promising candidates to be used for cancer imaging after further analyzation and optimization.}, subject = {Vaccinia-Virus}, language = {en} } @article{GentschevAdelfingerJosupeitetal.2012, author = {Gentschev, Ivaylo and Adelfinger, Marion and Josupeit, Rafael and Rudolph, Stephan and Ehrig, Klaas and Donat, Ulrike and Weibel, Stephanie and Chen, Nanhai G. and Yu, Yong A. and Zhang, Qian and Heisig, Martin and Thamm, Douglas and Stritzker, Jochen and MacNeill, Amy and Szalay, Aladar A.}, title = {Preclinical Evaluation of Oncolytic Vaccinia Virus for Therapy of Canine Soft Tissue Sarcoma}, series = {PLoS One}, volume = {7}, journal = {PLoS One}, number = {5}, doi = {10.1371/journal.pone.0037239}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-129998}, year = {2012}, abstract = {Virotherapy using oncolytic vaccinia virus (VACV) strains is one promising new strategy for canine cancer therapy. In this study we describe the establishment of an in vivo model of canine soft tissue sarcoma (CSTS) using the new isolated cell line STSA-1 and the analysis of the virus-mediated oncolytic and immunological effects of two different Lister VACV LIVP1.1.1 and GLV-1h68 strains against CSTS. Cell culture data demonstrated that both tested VACV strains efficiently infected and destroyed cells of the canine soft tissue sarcoma line STSA-1. In addition, in our new canine sarcoma tumor xenograft mouse model, systemic administration of LIVP1.1.1 or GLV-1h68 viruses led to significant inhibition of tumor growth compared to control mice. Furthermore, LIVP1.1.1 mediated therapy resulted in almost complete tumor regression and resulted in long-term survival of sarcoma-bearing mice. The replication of the tested VACV strains in tumor tissues led to strong oncolytic effects accompanied by an intense intratumoral infiltration of host immune cells, mainly neutrophils. These findings suggest that the direct viral oncolysis of tumor cells and the virus-dependent activation of tumor-associated host immune cells could be crucial parts of anti-tumor mechanism in STSA-1 xenografts. In summary, the data showed that both tested vaccinia virus strains and especially LIVP1.1.1 have great potential for effective treatment of CSTS.}, language = {en} } @article{GentschevMuellerAdelfingeretal.2011, author = {Gentschev, Ivaylo and M{\"u}ller, Meike and Adelfinger, Marion and Weibel, Stephanie and Grummt, Friedrich and Zimmermann, Martina and Bitzer, Michael and Heisig, Martin and Zhang, Qian and Yu, Yong A. and Chen, Nanhai G. and Stritzker, Jochen and Lauer, Ulrich M. and Szalay, Aladar A.}, title = {Efficient Colonization and Therapy of Human Hepatocellular Carcinoma (HCC) Using the Oncolytic Vaccinia Virus Strain GLV-1h68}, series = {PLOS ONE}, volume = {6}, journal = {PLOS ONE}, number = {7}, doi = {10.1371/journal.pone.0022069}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-135319}, pages = {e22069}, year = {2011}, abstract = {Virotherapy using oncolytic vaccinia virus strains is one of the most promising new strategies for cancer therapy. In this study, we analyzed for the first time the therapeutic efficacy of the oncolytic vaccinia virus GLV-1h68 in two human hepatocellular carcinoma cell lines HuH7 and PLC/PRF/5 (PLC) in cell culture and in tumor xenograft models. By viral proliferation assays and cell survival tests, we demonstrated that GLV-1h68 efficiently colonized, replicated in, and did lyse these cancer cells in culture. Experiments with HuH7 and PLC xenografts have revealed that a single intravenous injection (i.v.) of mice with GLV-1h68 resulted in a significant reduction of primary tumor sizes compared to uninjected controls. In addition, replication of GLV-1h68 in tumor cells led to strong inflammatory and oncolytic effects resulting in intense infiltration of MHC class II-positive cells like neutrophils, macrophages, B cells and dendritic cells and in up-regulation of 13 pro-inflammatory cytokines. Furthermore, GLV-1h68 infection of PLC tumors inhibited the formation of hemorrhagic structures which occur naturally in PLC tumors. Interestingly, we found a strongly reduced vascular density in infected PLC tumors only, but not in the non-hemorrhagic HuH7 tumor model. These data demonstrate that the GLV-1h68 vaccinia virus may have an enormous potential for treatment of human hepatocellular carcinoma in man.}, language = {en} } @article{GentschevPatilPetrovetal.2014, author = {Gentschev, Ivaylo and Patil, Sadeep S. and Petrov, Ivan and Cappello, Joseph and Adelfinger, Marion and Szalay, Aladar A.}, title = {Oncolytic Virotherapy of Canine and Feline Cancer}, series = {Viruses}, volume = {6}, journal = {Viruses}, number = {5}, doi = {10.3390/v6052122}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-119753}, pages = {2122-2137}, year = {2014}, abstract = {Cancer is the leading cause of disease-related death in companion animals such as dogs and cats. Despite recent progress in the diagnosis and treatment of advanced canine and feline cancer, overall patient treatment outcome has not been substantially improved. Virotherapy using oncolytic viruses is one promising new strategy for cancer therapy. Oncolytic viruses (OVs) preferentially infect and lyse cancer cells, without causing excessive damage to surrounding healthy tissue, and initiate tumor-specific immunity. The current review describes the use of different oncolytic viruses for cancer therapy and their application to canine and feline cancer.}, language = {en} } @article{GerovaWickeChiharaetal.2021, author = {Gerova, Milan and Wicke, Laura and Chihara, Kotaro and Schneider, Cornelius and Lavigne, Rob and Vogel, J{\"o}rg}, title = {A grad-seq view of RNA and protein complexes in Pseudomonas aeruginosa under standard and bacteriophage predation conditions}, series = {mbio}, volume = {12}, journal = {mbio}, number = {1}, doi = {10.1128/mBio.03454-20}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-259054}, pages = {e03454-20}, year = {2021}, abstract = {The Gram-negative rod-shaped bacterium Pseudomonas aeruginosa is not only a major cause of nosocomial infections but also serves as a model species of bacterial RNA biology. While its transcriptome architecture and posttranscriptional regulation through the RNA-binding proteins Hfq, RsmA, and RsmN have been studied in detail, global information about stable RNA-protein complexes in this human pathogen is currently lacking. Here, we implement gradient profiling by sequencing (Grad-seq) in exponentially growing P. aeruginosa cells to comprehensively predict RNA and protein complexes, based on glycerol gradient sedimentation profiles of >73\% of all transcripts and ∼40\% of all proteins. As to benchmarking, our global profiles readily reported complexes of stable RNAs of P. aeruginosa, including 6S RNA with RNA polymerase and associated product RNAs (pRNAs). We observe specific clusters of noncoding RNAs, which correlate with Hfq and RsmA/N, and provide a first hint that P. aeruginosa expresses a ProQ-like FinO domain-containing RNA-binding protein. To understand how biological stress may perturb cellular RNA/protein complexes, we performed Grad-seq after infection by the bacteriophage ΦKZ. This model phage, which has a well-defined transcription profile during host takeover, displayed efficient translational utilization of phage mRNAs and tRNAs, as evident from their increased cosedimentation with ribosomal subunits. Additionally, Grad-seq experimentally determines previously overlooked phage-encoded noncoding RNAs. Taken together, the Pseudomonas protein and RNA complex data provided here will pave the way to a better understanding of RNA-protein interactions during viral predation of the bacterial cell. IMPORTANCE Stable complexes by cellular proteins and RNA molecules lie at the heart of gene regulation and physiology in any bacterium of interest. It is therefore crucial to globally determine these complexes in order to identify and characterize new molecular players and regulation mechanisms. Pseudomonads harbor some of the largest genomes known in bacteria, encoding ∼5,500 different proteins. Here, we provide a first glimpse on which proteins and cellular transcripts form stable complexes in the human pathogen Pseudomonas aeruginosa. We additionally performed this analysis with bacteria subjected to the important and frequently encountered biological stress of a bacteriophage infection. We identified several molecules with established roles in a variety of cellular pathways, which were affected by the phage and can now be explored for their role during phage infection. Most importantly, we observed strong colocalization of phage transcripts and host ribosomes, indicating the existence of specialized translation mechanisms during phage infection. All data are publicly available in an interactive and easy to use browser.}, language = {en} } @phdthesis{Gimple2004, author = {Gimple, Olaf}, title = {Substratbindung und Katalyse in RNase P RNA vom cyanobakteriellen Typ}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-11283}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2004}, abstract = {Ribonuklease P (RNase P) ist eine essentielle Endonuklease, welche die 5'-Flanke von pre-tRNAs entfernt. Die RNase P RNA des Cyanobakteriums Prochlorococcus marinus ist in vitro katalytisch aktiv und bevorzugt in heterologen Prozessierungssystemen Substrate mit vollst{\"a}ndigem 3'-CCA-Ende. Diese Substratspezifit{\"a}t widerspricht den Erwartungen, da tRNAs in P. marinus nicht mit dem CCA-Ende codiert sind und die RNase P RNA auch nicht das GGU-Bindungsmotiv f{\"u}r diese CCA-Enden aufweist. Um die Substratspezifit{\"a}t und Aufbau des Ribozym-Substrat-Komplex von P. marinus RNase P RNA im homologen System untersuchen zu k{\"o}nnen, wurden Transkriptionsklone f{\"u}r P. marinus pre- und mat-tRNAArgCCU konstruiert, mit denen nach entsprechender Restriktionshydrolyse Transkripte mit stufenweise verk{\"u}rzten 3'-CCA-Ende synthetisiert werden k{\"o}nnen. Durch enzymkinetische Untersuchungen der Prozessierung durch P. marinus RNase P RNA wurde unter steady-state-Bedingungen f{\"u}r pre-tRNACCA eine Michaelis-Menten Konstante von 6,92 µM ermittelt. Die Entfernung von A76 und C75 des 3'-CCA-Endes f{\"u}hrt zu einer Erh{\"o}hung der KM (7,13 µM bzw. 19,68µM). Diese Substrate werden folglich weniger stark gebunden, was sich auch in der freien Bindungsenthalpie von 0,02 und 0,65 kcal/mol ausdr{\"u}ckt. Die Entfernung des vollst{\"a}ndigen 3'-CCA-Endes f{\"u}hrt zu einer erheblichen Erniedrigung der KM (0,83µM) und zu einer energetisch beg{\"u}nstigten, st{\"a}rkeren Substratbindung (-1,31 kcal/mol). P. marinus RNase RNase P RNA zeigt folglich bei der in vitro Prozessierung im homologen System unter steady-state-Bedingungen eine Substratspezifit{\"a}t f{\"u}r das Substrat mit deletiertem 3'-CCA-Ende. Durch die Methode des Crosslinking, die in dieser Arbeit etabliert und optimiert wurde, k{\"o}nnen RNA-Protein und RNA-RNA Interaktionen nachgewiesen werden. Mit ihr wurde die Bindung von Substrat und Produkt im Komplex mit der RNase P RNA untersucht. Durch interne Modifizierung der P. marinus RNase P RNA-Komponente mit dem photosensiblen Nukleotidanalogon s4U wurden Kontaktstellen in 5'-Flanke, Acceptor-Stamm, D-Stamm, D-Schleife, Anticodon-Schleife und in der variablen Schleife der P. marinus pre-tRNAArg identifiziert. Diese lokalisierten Kontaktstellen stehen denen in der 5'-Flanke, dem Acceptor-Stamm und der 3'-Flanke, wie sie f{\"u}r den Ribozym-Substrat-Komplex mit E. coli RNase P RNA identifiziert wurden, gegen{\"u}ber. In P. marinus RNase P RNA werden folglich alternative Kontaktstellen zur Substratbindung benutzt. Mit Hilfe der hier {\"u}berexprimierten E. coli Nukleotidyltransferase, konnte pre- und mat-tRNAArg durch eine neue Synthesestrategie am 3'-CCA-Ende mit dem Crosslink-Reagenz Azidophenacyl (APA) modifiziert werden. Durch die Positionierung von APA am 5'-Terminus von pre- und mat-tRNAArg wurden weitere modifizierte tRNAs synthetisiert. Durch Crosslink-Experimente im homologen P. marinus System mit diesen modifizierten pre- und mat-tRNAArg-Varianten wurden die selben Regionen des katalytischen Zentrums (J18/2, Region P15/P16, J5/15) der RNase P RNA identifiziert, wie sie von E. coli und B. subtilis RNase P RNA bekannt sind. Dies bedeutet, dass die 5'-Flanke, die Prozessierungsstelle und das 3'-CCA-Ende der tRNAs auf einer vergleichbaren Oberfl{\"a}che positioniert werden wie in anderen Ribozymen. Durch die fehlende Fixierung des 3'-CCA-Endes {\"u}ber Basenpaarungen mit dem GGU-Bindungsmotiv werden die tRNAs in P. marinus RNase P RNA weniger starr an das Ribozym gebunden und das 3'-CCA-Ende besitzt eine flexiblere Positionierung im Komplex mit dem Ribozym. Die Existenz unterschiedlicher Crosslink-Muster in P6, P18, J5/15 und J3/4 zeigt, dass pre-tRNAs und reife tRNAs durch verschiedene Modi an das P. marinus Ribozym gebunden werden. Die Identifizierung von vernetzten Nukleotiden in P15, J15/16, P16 und J16/15, die mit vergleichbaren modifizierten tRNAs in E. coli RNase P RNA nicht gefunden wurden, belegen, dass in P. marinus RNase P RNA ein anderer Produkt-Bindungs-Modus existiert als in E. coli. Erstmals konnten in dieser Arbeit auch zu erwartende Interaktionen mit dem katalytischen Zentrum identifiziert werden, die in bisherigen Crosslink-Experimenten in E. coli und B. subtilis RNase P RNA nicht oder nur geringf{\"u}gig auftraten. Um die erhaltenen Ergebnisse besser veranschaulichen zu k{\"o}nnen, wurde mit dem Programm ERNA 3D ein Raumstrukturmodell f{\"u}r P. marinus RNase P RNA und tRNAArg erstellt. Die RNase P RNA der Cyanellen von Cyanophora paradoxa, ist in vitro katalytisch inaktiv. Um zu kl{\"a}ren, ob die fehlende Ribozym-Aktivit{\"a}t dieser RNase P RNA auf eine fehlerhafte Substratbindung zur{\"u}ckzuf{\"u}hren ist, sollten Crosslink-Experimente mit den modifizierten P. marinus tRNAArg durchgef{\"u}hrt werden. Es konnte gezeigt werden, dass 5'- und 3'-modifizierte pre-tRNAs in C. paradoxa in einem anderen Modus gebunden werden, als durch die katalytisch aktive P. marinus RNase P RNA.}, subject = {Prochlorococcus marinus}, language = {de} } @article{GoncharovaRuzhenkovaPetrovetal.2016, author = {Goncharova, Elena P. and Ruzhenkova, Julia S. and Petrov, Ivan S. and Shchelkunov, Sergey N. and Zenkova, Marina A.}, title = {Oncolytic virus efficiency inhibited growth of tumour cells with multiple drug resistant phenotype in vivo and in vitro}, series = {Journal of Translational Medicine}, volume = {14}, journal = {Journal of Translational Medicine}, number = {241}, doi = {10.1186/s12967-016-1002-x}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-165714}, pages = {1-14}, year = {2016}, abstract = {Background Tumour resistance to a wide range of drugs (multiple drug resistant, MDR) acquired after intensive chemotherapy is considered to be the main obstacle of the curative treatment of cancer patients. Recent work has shown that oncolytic viruses demonstrated prominent potential for effective treatment of diverse cancers. Here, we evaluated whether genetically modified vaccinia virus (LIVP-GFP) may be effective in treatment of cancers displaying MDR phenotype. Methods LIVP-GFP replication, transgene expression and cytopathic effects were analysed in human cervical carcinomas KB-3-1 (MDR-), KB-8-5 (MDR+) and in murine melanoma B-16 (MDR-), murine lymphosarcomas RLS and RLS-40 (MDR+). To investigate the efficacy of this therapy in vivo, we treated immunocompetent mice bearing murine lymphosarcoma RLS-40 (MDR+) (6- to 8-week-old female CBA mice; n = 10/group) or melanoma B-16 (MDR-) (6- to 8-week-old female C57Bl mice; n = 6/group) with LIVP-GFP (5 × 107 PFU of virus in 0.1 mL of IMDM immediately and 4 days after tumour implantation). Results We demonstrated that LIVP-GFP replication was effective in human cervical carcinomas KB-3-1 (MDR-) and KB-8-5 (MDR+) and in murine melanoma B-16 (MDR-), whereas active viral production was not detected in murine lymphosarcomas RLS and RLS-40 (MDR+). Additionally, it was found that in tumour models in immunocompetent mice under the optimized regimen intratumoural injections of LIVP-GFP significantly inhibited melanoma B16 (33 \% of mice were with complete response after 90 days) and RLS-40 tumour growth (fourfold increase in tumour doubling time) as well as metastasis. Conclusion The anti-tumour activity of LIVP-GFP is a result of direct oncolysis of tumour cells in case of melanoma B-16 because the virus effectively replicates and destroys these cells, and virus-mediated activation of the host immune system followed by immunologically mediated destruction of of tumour cells in case of lymphosarcoma RLS-40. Thus, the recombinant vaccinia virus LIVP-GFP is able to inhibit the growth of malignant cells with the MDR phenotype and tumour metastasis when administered in the early stages of tumour development.}, language = {en} } @article{GowdaGodderKmieciaketal.2011, author = {Gowda, Madhu and Godder, Kamar and Kmieciak, Maciej and Worschech, Andrea and Ascierto, Maria-Libera and Wang, Ena and Francesco M., Marincola and Manjili, Masoud H.}, title = {Distinct signatures of the immune responses in low risk versus high risk neuroblastoma}, series = {Journal of Translational Medicine}, volume = {9}, journal = {Journal of Translational Medicine}, number = {170}, doi = {10.1186/1479-5876-9-170}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-135147}, pages = {1-8}, year = {2011}, abstract = {Background: Over 90\% of low risk (LR) neuroblastoma patients survive whereas less than 30\% of high risk (HR) patients are long term survivors. Age (children younger than 18 months old) is associated with LR disease. Considering that adaptive immune system is well developed in older children, and that T cells were shown to be involved in tumor escape and progression of cancers, we sought to determine whether HR patients may tend to show a signature of adaptive immune responses compared to LR patients who tend to have diminished T-cell responses but an intact innate immune response. Methods: We performed microarray analysis of RNA extracted from the tumor specimens of HR and LR patients. Flow cytometry was performed to determine the cellular constituents in the blood while multiplex cytokine array was used to detect the cytokine profile in patients' sera. A HR tumor cell line, SK-N-SH, was also used for detecting the response to IL-1 beta, a cytokines which is involved in the innate immune responses. Results: Distinct patterns of gene expression were detected in HR and LR patients indicating an active T-cell response and a diminished adaptive immune response, respectively. A diminished adaptive immune response in LR patients was evident by higher levels of IL-10 in the sera. In addition, HR patients had lower levels of circulating myeloid derived suppressor cells (MDSC) compared with a control LR patient. LR patients showed slightly higher levels of cytokines of the innate immune responses. Treatment of the HR tumor line with IL-1b induced expression of cytokines of the innate immune responses. Conclusions: This data suggests that adaptive immune responses may play an important role in the progression of HR disease whereas innate immune responses may be active in LR patients.}, language = {en} } @article{GrimmPelzSchneideretal.2020, author = {Grimm, Clemens and Pelz, Jann-Patrick and Schneider, Cornelius and Sch{\"a}ffler, Katrin and Fischer, Utz}, title = {Crystal Structure of a Variant PAM2 Motif of LARP4B Bound to the MLLE Domain of PABPC1}, series = {Biomolecules}, volume = {10}, journal = {Biomolecules}, number = {6}, issn = {2218-273X}, doi = {10.3390/biom10060872}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-207800}, year = {2020}, abstract = {Eukaryotic cells determine the protein output of their genetic program by regulating mRNA transcription, localization, translation and turnover rates. This regulation is accomplished by an ensemble of RNA-binding proteins (RBPs) that bind to any given mRNA, thus forming mRNPs. Poly(A) binding proteins (PABPs) are prominent members of virtually all mRNPs that possess poly(A) tails. They serve as multifunctional scaffolds, allowing the recruitment of diverse factors containing a poly(A)-interacting motif (PAM) into mRNPs. We present the crystal structure of the variant PAM motif (termed PAM2w) in the N-terminal part of the positive translation factor LARP4B, which binds to the MLLE domain of the poly(A) binding protein C1 cytoplasmic 1 (PABPC1). The structural analysis, along with mutational studies in vitro and in vivo, uncovered a new mode of interaction between PAM2 motifs and MLLE domains.}, language = {en} } @article{GrossHennardMasourisetal.2012, author = {Gross, Henrik and Hennard, Christine and Masouris, Ilias and Cassel, Christian and Barth, Stephanie and Stober-Gr{\"a}sser, Ute and Mamiani, Alfredo and Moritz, Bodo and Ostareck, Dirk and Ostareck-Lederer, Antje and Neuenkirchen, Nils and Fischer, Utz and Deng, Wen and Leonhardt, Heinrich and Noessner, Elfriede and Kremmer, Elisabeth and Gr{\"a}sser, Friedrich A.}, title = {Binding of the Heterogeneous Ribonucleoprotein K (hnRNP K) to the Epstein-Barr Virus Nuclear Antigen 2 (EBNA2) Enhances Viral LMP2A Expression}, series = {PLoS One}, volume = {7}, journal = {PLoS One}, number = {8}, doi = {10.1371/journal.pone.0042106}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-133707}, year = {2012}, abstract = {The Epstein-Barr Virus (EBV) -encoded EBNA2 protein, which is essential for the in vitro transformation of B-lymphocytes, interferes with cellular processes by binding to proteins via conserved sequence motifs. Its Arginine-Glycine (RG) repeat element contains either symmetrically or asymmetrically di-methylated arginine residues (SDMA and ADMA, respectively). EBNA2 binds via its SDMA-modified RG-repeat to the survival motor neurons protein (SMN) and via the ADMA-RG-repeat to the NP9 protein of the human endogenous retrovirus K (HERV-K (HML-2) Type 1). The hypothesis of this work was that the methylated RG-repeat mimics an epitope shared with cellular proteins that is used for interaction with target structures. With monoclonal antibodies against the modified RG-repeat, we indeed identified cellular homologues that apparently have the same surface structure as methylated EBNA2. With the SDMA-specific antibodies, we precipitated the Sm protein D3 (SmD3) which, like EBNA2, binds via its SDMA-modified RG-repeat to SMN. With the ADMA-specific antibodies, we precipitated the heterogeneous ribonucleoprotein K (hnRNP K). Specific binding of the ADMA-antibody to hnRNP K was demonstrated using E. coli expressed/ADMA-methylated hnRNP K. In addition, we show that EBNA2 and hnRNP K form a complex in EBV-infected B-cells. Finally, hnRNP K, when co-expressed with EBNA2, strongly enhances viral latent membrane protein 2A (LMP2A) expression by an unknown mechanism as we did not detect a direct association of hnRNP K with DNA-bound EBNA2 in gel shift experiments. Our data support the notion that the methylated surface of EBNA2 mimics the surface structure of cellular proteins to interfere with or co-opt their functional properties.}, language = {en} } @phdthesis{Goetz2018, author = {G{\"o}tz, Silvia}, title = {Zuo1 - ein neues G-Quadruplex-bindendes Protein in \(Saccharomyces\) \(cerevisiae\)}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-152158}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2018}, abstract = {G-Quadruplex (G4)-Strukturen sind sehr stabile und polymorphe DNA und RNA Sekund{\"a}rstrukturen mit einem konservierten Guanin-reichen Sequenzmotiv (G4-Motiv). Sie bestehen aus {\"u}bereinander gestapelten planaren G-Quartetts, in denen je vier Guanine durch Wasserstoffbr{\"u}ckenbindungen zusammengehalten werden. Da G4-Motive in Eukaryoten an bestimmten Stellen im Genom angereichert vorkommen, wird angenommen, dass die Funktion von G4-Strukturen darin besteht, biologische Prozesse positiv oder negativ zu regulieren. Aufgrund der hohen thermodynamischen Stabilit{\"a}t von G4 Strukturen ist davon auszugehen, dass Proteine in die Faltung, Stabilisierung und Entfaltung dieser Nukleins{\"a}ure-Strukturen regulatorisch involviert sind. Bis heute wurden viele Proteine in der Literatur beschrieben, die G4-Strukturen entwinden k{\"o}nnen. Jedoch konnten bisher nur wenige Proteine identifiziert werden, die in vivo die Faltung f{\"o}rdern oder G4-Strukturen stabilisieren. Durch Yeast One-Hybrid (Y1H)-Screenings habe ich Zuo1 als neues G4 bindendes Protein identifiziert. In vitro Analysen best{\"a}tigten diese Interaktion und es stellte sich heraus, dass Zuo1 G4-Strukturen stabilisiert. {\"U}bereinstimmend mit den in vitro Daten konnte gezeigt werden, dass Zuo1 signifikant an G4-Motive im Genom von Saccharomyces ceresivisiae bindet. Genomweit {\"u}berlappen G4-Motive, an die Zuo1 bindet, mit Stellen, an denen die DNA Replikation zum Stillstand kommt und vermehrt DNA Sch{\"a}den vorkommen. Diese Ergebnisse legen nahe, dass Zuo1 eine Funktion w{\"a}hrend der DNA Reparatur oder in Zusammenhang mit dem Vorankommen der DNA Replikationsgabel hat, indem G4-Strukturen stabilisiert werden. Diese Hypothese wird außerdem durch genetische Experimente gest{\"u}tzt, wonach in Abwesenheit von Zuo1 die Genominstabilit{\"a}t zunimmt. Aufgrund dieser Daten war es m{\"o}glich ein Model zu entwickeln, bei dem Zuo1 w{\"a}hrend der S-Phase G4-Strukturen bindet und stabilisiert wodurch die DNA Replikation blockiert wird. Diese Interaktion findet neben Stellen schadhafter DNA statt und unterst{\"u}tzt somit DNA Reparatur-Prozesse wie beispielsweise die Nukleotidexzisionsreparatur. Als weiteres potentielles G4-bindendes Protein wurde Slx9 in Y1H-Screenings identifiziert. In vitro Experimente zeigten zwar, dass Slx9 mit h{\"o}herer Affinit{\"a}t an G4-Strukturen bindet im Vergleich zu anderen getesteten DNA Konformationen, jedoch wurde in S. cerevisiae genomweit keine signifikante Bindung an G4-Motive festgestellt.}, subject = {Saccharomyces cerevisiae}, language = {de} } @phdthesis{Haake2001, author = {Haake, Markus}, title = {Stat6-vermittelte Genregulation in eukaryontischen Zellen}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-1181580}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2001}, abstract = {Der Transkriptionsfaktor Stat6 vermittelt zentrale Wirkungen von IL-4 und IL-13, die in der Pathologie atopischer Erkrankungen eine Rolle spielen. Seine Spezifit{\"a}t f{\"u}r diese beiden allergieassoziierten Cytokine ist eine wesentliche Motivation ihn n{\"a}her zu untersuchen. In dieser Arbeit sollte mehr {\"u}ber die Funktion von Stat6 herausgefunden werden. Außerdem wurden M{\"o}glichkeiten untersucht dieses Verhalten zu beinflussen. Einen Schwerpunkt der Arbeit bildete die Regulation des Eotaxin-1-Promotors. Eotaxin-1 ist einer der st{\"a}rksten Rekrutierungsfaktoren f{\"u}r Eosinophile, die eine zentrale Rolle bei der Immunpathologie allergischer Erkrankungen spielen. Mit Hilfe der Daten konnte eine neue Hypothese zur Regulation des Eotaxin-1-Promotors entwickelt werden. Zum Vergleich wurde mit der Untersuchung des Promotors eines weiteren Chemokins, des MCP-4, begonnen. In Zusammenarbeit mit Dr. Sascha Stolzenberger wurde ein Weg untersucht den Stat6-Signalweg zu hemmen. Dabei wurden mit Hilfe des Antennapedia-Peptides Stat6-Bindepeptide in die Zelle transportiert, um dort {\"u}ber eine kompetitive Hemmung die Signaltransduktion zu unterbinden. Ergebnis dieser Arbeiten ist ein hochspezifischer, aber nur transient wirkender Stat6 Inhibitor. Die Stat6/DNA-Wechselwirkung wurde mit der Magnetobead-Technik untersucht. Dabei werden Promotorfragmente an Magnetk{\"u}gelchen gekoppelt und unter Ausnutzung der Magnetisierung an die DNA bindende Proteine isoliert und {\"u}ber SDS-PAGE/Immunoblotanalyse untersucht. Mit dem Verfahren konnte die Stat6-Bindung an acht verschiedene Promotoren nachgewiesen werden. In Zusammenarbeit mit der Arbeitsgruppe Pallardy aus Paris wurde die Wechselwirkung von Stat6 mit dem Glucocorticoid-Rezeptor untersucht. Glucocorticoide kontrollieren Entz{\"u}ndungen und Interaktionen des aktivierten Rezeptors mit anderen Proteinen aus der Stat-Familie sind seit l{\"a}ngerem bekannt. Wie in dieser Arbeit gezeigt wurde, interagiert Stat6 mit dem Glucocorticoidrezeptor unabh{\"a}ngig von einer Bindung an DNA. Zus{\"a}tzlich wurde der Mucin-2-Promotor auf Stat6-Regulierung untersucht. Mucine sind wichtige Bestandteile des Schleimes. Verst{\"a}rkte Schleim-Sekretion ist ein klinisches Symptom asthmatischer Erkrankungen und tr{\"a}gt zur Zerst{\"o}rung der Lunge bei. Ein potentiell Stat6 reguliertes Fragment aus dem Mucinpromoter wurde mit Hilfe von PCR-Techniken isoliert und in Reportergenvektoren kloniert.}, subject = {Interleukin 4}, language = {de} } @article{HaddadChenCarlinetal.2012, author = {Haddad, Dana and Chen, Chun-Hao and Carlin, Sean and Silberhumer, Gerd and Chen, Nanhai G. and Zhang, Qian and Longo, Valerie and Carpenter, Susanne G. and Mittra, Arjun and Carson, Joshua and Au, Joyce and Gonen, Mithat and Zanzonico, Pat B. and Szalay, Aladar A. and Fong, Yuman}, title = {Imaging Characteristics, Tissue Distribution, and Spread of a Novel Oncolytic Vaccinia Virus Carrying the Human Sodium Iodide Symporter}, series = {PLoS One}, volume = {7}, journal = {PLoS One}, number = {8}, doi = {10.1371/journal.pone.0041647}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-130041}, pages = {e41647}, year = {2012}, abstract = {Introduction: Oncolytic viruses show promise for treating cancer. However, to assess therapy and potential toxicity, a noninvasive imaging modality is needed. This study aims to determine the in vivo biodistribution, and imaging and timing characteristics of a vaccinia virus, GLV-1h153, encoding the human sodium iodide symporter (hNIS. Methods: GLV-1h153 was modified from GLV-1h68 to encode the hNIS gene. Timing of cellular uptake of radioiodide \(^{131}\)I in human pancreatic carcinoma cells PANC-1 was assessed using radiouptake assays. Viral biodistribution was determined in nude mice bearing PANC-1 xenografts, and infection in tumors confirmed histologically and optically via Green Fluorescent Protein (GFP) and bioluminescence. Timing characteristics of enhanced radiouptake in xenografts were assessed via \(^{124}\)I-positron emission tomography (PET). Detection of systemic administration of virus was investigated with both \(^{124}\)I-PET and 99m-technecium gamma-scintigraphy. Results: GLV-1h153 successfully facilitated time-dependent intracellular uptake of \(^{131}\)I in PANC-1 cells with a maximum uptake at 24 hours postinfection (P < 0.05). In vivo, biodistribution profiles revealed persistence of virus in tumors 5 weeks postinjection at 10\(^9\) plaque-forming unit (PFU)/gm tissue, with the virus mainly cleared from all other major organs. Tumor infection by GLV-1h153 was confirmed via optical imaging and histology. GLV-1h153 facilitated imaging virus replication in tumors via PET even at 8 hours post radiotracer injection, with a mean \% ID/gm of 3.82 \(\pm\) 60.46 (P < 0.05) 2 days after intratumoral administration of virus, confirmed via tissue radiouptake assays. One week post systemic administration, GLV1h153-infected tumors were detected via \(^{124}\)I-PET and 99m-technecium-scintigraphy. Conclusion: GLV-1h153 is a promising oncolytic agent against pancreatic cancer with a promising biosafety profile. GLV-1h153 facilitated time-dependent hNIS-specific radiouptake in pancreatic cancer cells, facilitating detection by PET with both intratumoral and systemic administration. Therefore, GLV-1h153 is a promising candidate for the noninvasive imaging of virotherapy and warrants further study into longterm monitoring of virotherapy and potential radiocombination therapies with this treatment and imaging modality.}, language = {en} } @article{HaddadChenZhangetal.2011, author = {Haddad, Dana and Chen, Nanhai G. and Zhang, Qian and Chen, Chun-Hao and Yu, Yong A. and Gonzalez, Lorena and Carpenter, Susanne G. and Carson, Joshua and Au, Joyce and Mittra, Arjun and Gonen, Mithat and Zanzonico, Pat B. and Fong, Yuman and Szalay, Aladar A.}, title = {Insertion of the human sodium iodide symporter to facilitate deep tissue imaging does not alter oncolytic or replication capability of a novel vaccinia virus}, series = {Journal of Translational Medicine}, volume = {9}, journal = {Journal of Translational Medicine}, number = {36}, doi = {10.1186/1479-5876-9-36}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-140847}, pages = {1-14}, year = {2011}, abstract = {Introduction: Oncolytic viruses show promise for treating cancer. However, to assess therapeutic efficacy and potential toxicity, a noninvasive imaging modality is needed. This study aimed to determine if insertion of the human sodium iodide symporter (hNIS) cDNA as a marker for non-invasive imaging of virotherapy alters the replication and oncolytic capability of a novel vaccinia virus, GLV-1h153. Methods: GLV-1h153 was modified from parental vaccinia virus GLV-1h68 to carry hNIS via homologous recombination. GLV-1h153 was tested against human pancreatic cancer cell line PANC-1 for replication via viral plaque assays and flow cytometry. Expression and transportation of hNIS in infected cells was evaluated using Westernblot and immunofluorescence. Intracellular uptake of radioiodide was assessed using radiouptake assays. Viral cytotoxicity and tumor regression of treated PANC-1tumor xenografts in nude mice was also determined. Finally, tumor radiouptake in xenografts was assessed via positron emission tomography (PET) utilizing carrier-free (124)I radiotracer. Results: GLV-1h153 infected, replicated within, and killed PANC-1 cells as efficiently as GLV-1h68. GLV-1h153 provided dose-dependent levels of hNIS expression in infected cells. Immunofluorescence detected transport of the protein to the cell membrane prior to cell lysis, enhancing hNIS-specific radiouptake (P < 0.001). In vivo, GLV-1h153 was as safe and effective as GLV-1h68 in regressing pancreatic cancer xenografts (P < 0.001). Finally, intratumoral injection of GLV-1h153 facilitated imaging of virus replication in tumors via (124)I-PET. Conclusion: Insertion of the hNIS gene does not hinder replication or oncolytic capability of GLV-1h153, rendering this novel virus a promising new candidate for the noninvasive imaging and tracking of oncolytic viral therapy.}, language = {en} } @article{HaddadSocciChenetal.2016, author = {Haddad, Dana and Socci, Nicholas and Chen, Chun-Hao and Chen, Nanhai G and Zhang, Qian and Carpenter, Susanne G and Mittra, Arjun and Szalay, Aladar A and Fong, Yuman}, title = {Molecular network, pathway, and functional analysis of-time dependent gene changes associated with pancreatic cancer susceptibility to oncolytic vaccinia virotherapy}, series = {Molecular Therapy — Oncolytics}, volume = {3}, journal = {Molecular Therapy — Oncolytics}, doi = {10.1038/mto.2016.8}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-165855}, pages = {16008}, year = {2016}, abstract = {Background: Pancreatic cancer is a fatal disease associated with resistance to conventional therapies. This study aimed to determine changes in gene expression patterns associated with infection and susceptibility of pancreatic cancer cells to an oncolyticvaccinia virus, GLV-1h153, carrying the human sodium iodide symporter for deep tissue imaging of virotherapy. Methods: Replication and susceptibility of pancreatic adenocarcinoma PANC-1 cells to GLV-1h153 was confirmed with replication and cytotoxicity assays. PANC-1 cells were then infected with GLV-1h153 and near-synchronous infection confirmed via flow cytometry of viral-induced green fluorescent protein (GFP) expression. Six and 24 hours after infection, three samples of each time point were harvested, and gene expression patterns assessed using HG-U133A cDNA microarray chips as compared to uninfected control. Differentially expressed genes were identified using Bioconductor LIMMA statistical analysis package. A fold change of 2.0 or above was used as a cutoff, with a P value of 0.01. The gene list was then analyzed using Ingenuity Pathways Analysis software. Results: Differential gene analysis revealed a total of 12,412 up- and 11,065 downregulated genes at 6 and 24 hours postinfection with GLV-1h153 as compared to control. At 6 hours postinfection. A total of 139 genes were either up or downregulated >twofold (false discovery rate < 0.05), of which 124 were mapped by Ingenuity Pathway Analysis (IPA). By 24 hours postinfection, a total of 5,698 genes were identified and 5,563 mapped by IPA. Microarray revealed gene expression changes, with gene networks demonstrating downregulation of processes such as cell death, cell cycle, and DNA repair, and upregulation of infection mechanisms (P < 0.01). Six hours after infection, gene changes involved pathways such as HMGB-1, interleukin (IL)-2, IL-6, IL-8, janus kinase/signal tranducer and activator of transcription (JAK/STAT), interferon, and ERK 5 signaling (P < 0.01). By 24 hours, prominent pathways included P53- and Myc-induced apoptotic processes, pancreatic adenocarcinoma signaling, and phosphoinositide 3-kinase/v-akt murine thymoma vial oncogene homolog 1 (PI3/AKT) pathways. Conclusions: Our study reveals the ability to assess time-dependent changes in gene expression patterns in pancreatic cancer cells associated with infection and susceptibility to vaccinia viruses. This suggests that molecular assays may be useful to develop safer and more efficacious oncolyticvirotherapies and support the idea that these treatments may target pathways implicated in pancreatic cancer resistance to conventional therapies.}, language = {en} } @phdthesis{Handoko2007, author = {Handoko, Lusy Lusiana}, title = {Functional Characterization of IGHMBP2, the Disease Gene Product of Spinal Muscular Atrophy with Respiratory Distress Type 1 (SMARD1)}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-24984}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2007}, abstract = {Spinale Muskelatrophie mit Atemnot Type 1 (SMARD1) ist eine autosomal rezessive, neurodegenerative Erkrankung, die sich h{\"a}ufig schon im S{\"a}uglings- und Kleinkindalter manifestiert. Pathologisches Merkmal von SMARD1 ist eine fr{\"u}he und akut einsetzende Atemnot und eine progrediente, zun{\"a}chst distal betonte Muskelschw{\"a}che, die durch eine L{\"a}hmung des Zwerchfells und der Skelettmuskulatur aufgrund des Absterbens der motorischen Vordernhornzellen des R{\"u}ckenmarks eintritt. SMARD1 ist eine monogene Krankheit, die durch Mutationen im Gen f{\"u}r das Immunoglobulin µ-bindende Protein 2" (IGHMBP2) hervorgerufen wird. Obwohl Mutationen in IGHMBP2 ausschließlich die Degeneration von Motoneuronen ausl{\"o}sen, ist das Gen bei Menschen und M{\"a}usen ubiquit{\"a}r exprimiert. Deshalb scheint SMARD1 durch den Defekt eines „Haushaltsproteins" statt eines Neuron-spezifischen Faktors verursacht zu werden. IGHMBP2 verf{\"u}gt {\"u}ber eine N-terminale DEXDc-Helicase/ATPase-Dom{\"a}ne und geh{\"o}rt zur Superfamily 1 Helicase. Bislang war lediglich bekannt, dass das Protein in verschiedenen zellul{\"a}ren Aktivit{\"a}ten wie DNA Replikation, Transkription und pr{\"a}-mRNA Splicing zugewiesen wurde. Die pr{\"a}zise Funktion von IGHMBP2 in den obengenannten Prozessen, und damit auch die molekulare Ursache von SMARD1 sind jedoch noch v{\"o}llig unklar. Das Ziel der vorliegenden Arbeit war es daher, das IGHMBP2 Protein sowohl enzymatisch zu charakterisieren als auch den Prozess zu identifizieren, in dem dieses Protein in vivo agiert. Mit diesem Wissen sollten dann pathogene Mutanten von IGHMBP2 auf Defekte hin untersucht werden. Ein Schl{\"u}ssel f{\"u}r diese Arbeit war die Gewinnung von rekombinantem, biologisch aktivem IGHMBP2 durch eine zweistufige Aufreinigungsstrategie. Dieses hochreine Enzym zeigte eine ATP-abh{\"a}ngige Helikaseaktivit{\"a}t, die sowohl doppelstr{\"a}ngige DNA als auch RNA mit einer 5'\&\#8594;3' Direktionalit{\"a}t entwindet. Interessanterweise zeigte sich, dass dieses Enzym -im Gegensatz zu fr{\"u}heren Befunden- nahezu ausschließlich im Zytoplasma von Zellen lokalisiert ist. Dar{\"u}ber hinaus wiesen die Affinit{\"a}tsaufreinigungsexperimente und Grossenfraktionierungsuntersuchungen daraufhin, dass IGHMBP2 ein Bestandteil des RNase-empfindlichen Komplexes ist, der als Ribosomen identifiziert wurde. IGHMBP2 interagiert prim{\"a}r mit 80S Monosomen, wobei das Protein mit beiden Untereinheiten in Kontakt steht. Hingegen ist IGHMBP2 an Polysomen nur in geringen Mengen zu finden. Diese Befunde deuten stark auf eine Rolle von IGHMBP2 bei der mRNA Verarbeitung am Ribosom hin, wobei noch unklar ist, ob es sich um translationsrelevante Prozesse handelt oder die mRNA-Stabilit{\"a}t beeinflusst. Die biochemische und enzymatische Charakterisierung von IGHMBP2 erlaubte erstmals Einblicke in den Pathomechanismus von SMARD1. In den folgenden Untersuchungen wurden die enzymatischen Aktivit{\"a}ten der SMARD1-erregenden Ighmbp2 Mutante und ihre Assoziation mit ribosomalen Untereinheiten nachgeforscht. Interessanterweise konnten pathogene Missense-Mutanten von IGHMBP2 noch genauso gut wie das Wildtyp-Protein mit ribosomalen Untereinheiten wechselwirken. Jedoch inhibierten alle bisher getesteten Mutanten die RNA Helikaseaktivit{\"a}t, allerdings {\"u}ber unterschiedliche Mechanismen. Diese Daten weisen darauf hin, dass ein Defekt in den enzymatischen Aktivit{\"a}ten des IGHMBP2 direkt mit der Pathogenese der SMARD1 korreliert. Des Weiteren lassen die im Rahmen dieser Arbeit erhaltenen Ergebnisse vermuten, dass SMARD1 durch Defekte in der zellularen Translationsmaschinerie entsteht.}, subject = {IGHMBP2}, language = {en} } @unpublished{HennigPrustyKauferetal.2021, author = {Hennig, Thomas and Prusty, Archana B. and Kaufer, Benedikt and Whisnant, Adam W. and Lodha, Manivel and Enders, Antje and Thomas, Julius and Kasimir, Francesca and Grothey, Arnhild and Herb, Stefanie and J{\"u}rges, Christopher and Meister, Gunter and Erhard, Florian and D{\"o}lken, Lars and Prusty, Bhupesh K.}, title = {Selective inhibition of microRNA processing by a herpesvirus-encoded microRNA triggers virus reactivation from latency}, edition = {submitted version}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-267858}, year = {2021}, abstract = {Herpesviruses have mastered host cell modulation and immune evasion to augment productive infection, life-long latency and reactivation thereof 1,2. A long appreciated, yet elusively defined relationship exists between the lytic-latent switch and viral non-coding RNAs 3,4. Here, we identify miRNA-mediated inhibition of miRNA processing as a novel cellular mechanism that human herpesvirus 6A (HHV-6A) exploits to disrupt mitochondrial architecture, evade intrinsic host defense and drive the latent-lytic switch. We demonstrate that virus-encoded miR-aU14 selectively inhibits the processing of multiple miR-30 family members by direct interaction with the respective pri-miRNA hairpin loops. Subsequent loss of miR-30 and activation of miR-30/p53/Drp1 axis triggers a profound disruption of mitochondrial architecture, which impairs induction of type I interferons and is necessary for both productive infection and virus reactivation. Ectopic expression of miR-aU14 was sufficient to trigger virus reactivation from latency thereby identifying it as a readily drugable master regulator of the herpesvirus latent-lytic switch. Our results show that miRNA-mediated inhibition of miRNA processing represents a generalized cellular mechanism that can be exploited to selectively target individual members of miRNA families. We anticipate that targeting miR-aU14 provides exciting therapeutic options for preventing herpesvirus reactivations in HHV-6-associated disorders like myalgic encephalitis/chronic fatigue syndrome (ME/CFS) and Long-COVID.}, language = {en} } @unpublished{HennigPrustyKauferetal.2022, author = {Hennig, Thomas and Prusty, Archana B. and Kaufer, Benedikt and Whisnant, Adam W. and Lodha, Manivel and Enders, Antje and Thomas, Julius and Kasimir, Francesca and Grothey, Arnhild and Herb, Stefanie and J{\"u}rges, Christopher and Meister, Gunter and Erhard, Florian and D{\"o}lken, Lars and Prusty, Bhupesh K.}, title = {Selective inhibition of miRNA 1 processing by a herpesvirus encoded miRNA}, edition = {accepted version}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-267862}, year = {2022}, abstract = {Herpesviruses have mastered host cell modulation and immune evasion to augment productive infection, life-long latency and reactivation thereof 1,2. A long appreciated, yet elusively defined relationship exists between the lytic-latent switch and viral non-coding RNAs 3,4. Here, we identify miRNA-mediated inhibition of miRNA processing as a thus far unknown cellular mechanism that human herpesvirus 6A (HHV-6A) exploits to disrupt mitochondrial architecture, evade intrinsic host defense and drive the lytic-latent switch. We demonstrate that virus-encoded miR-aU14 selectively inhibits the processing of multiple miR-30 family members by direct interaction with the respective pri-miRNA hairpin loops. Subsequent loss of miR-30 and activation of the miR-30/p53/Drp1 axis triggers a profound disruption of mitochondrial architecture. This impairs induction of type I interferons and is necessary for both productive infection and virus reactivation. Ectopic expression of miR-aU14 triggered virus reactivation from latency, identifying viral miR-aU14 as a readily drugable master regulator of the herpesvirus lytic-latent switch. Our results show that miRNA-mediated inhibition of miRNA processing represents a generalized cellular mechanism that can be exploited to selectively target individual members of miRNA families. We anticipate that targeting miR-aU14 provides exciting therapeutic options for preventing herpesvirus reactivations in HHV-6-associated disorders.}, language = {en} } @phdthesis{Herrmann2004, author = {Herrmann, Thomas}, title = {Sequenz-spezifische Interaktion des murinen pr{\"a}replikativen Komplexes mit Origin-DNA}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-11241}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2004}, abstract = {Mit Hilfe von in vivo ChIP-Experimenten identifizierten wir eine pr{\"a}RC Bindungsstelle von -2519 bis -2152 (Fragment B) innerhalb eines „origin of bidirectional replication (OBR)" der 44 kb langen Maus-rDNA-Einheit. Diese Bindungsstelle befindet sich ungef{\"a}hr 2,3 kb ubstream des Transkriptionsstartpunktes der RNA-Poymerase I. An dieser Bindungsstelle konnte in der G1-Phase der komplette ORC-Komplex sowie Geminin, MCM3 und -6 nachgewiesen werden. F{\"u}r den G1/S-Phasen{\"u}bergang deuten die Ergebnisse darauf hin, dass sich ORC6 und Geminin von Fragment B abl{\"o}sen, w{\"a}hrend sich CDC6 und -45 an den ORC-Komplex anlagern. Mit Erreichen der S-Phase konnte gezeigt werden, dass sich CDC6 und -45 sowie ORC1 wieder abl{\"o}sen und ein Core-Komplex von ORC2-5 sowie MCM3 und -6 gebunden bleibt. Außerdem konnte an Fragment B eine spezifische Bindung eines aus FM3A-Zellkernprotein angereicherten pr{\"a}RC-Komplexes (Komplex A) auch in vitro mit Hilfe von EMSA-Experimenten beobachtet werden. Die Bindungsaktivit{\"a}t von Komplex A an Fragment B konnte durch ATP verst{\"a}rkt werden.}, subject = {Maus}, language = {de} }