@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} }