@article{StepniakKaestnerPoggietal.2015, author = {Stepniak, Beata and K{\"a}stner, Anne and Poggi, Giulia and Mitjans, Marina and Begemann, Martin and Hartmann, Annette and Van der Auwera, Sandra and Sananbenesi, Farahnaz and Kr{\"u}ger-Burg, Dilja and Matuszko, Gabriela and Brosi, Cornelia and Homuth, Georg and V{\"o}lzke, Henry and Benseler, Fritz and Bagni, Claudia and Fischer, Utz and Dityatev, Alexander and Grabe, Hans-J{\"o}rgen and Rujescu, Dan and Fischer, Andre and Ehrenreich, Hannelore}, title = {Accumulated common variants in the broader fragile X gene family modulate autistic phenotypes}, series = {EMBO Molecular Medicine}, volume = {7}, journal = {EMBO Molecular Medicine}, number = {12}, doi = {10.15252/emmm.201505696}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-136893}, pages = {1565-1579}, year = {2015}, abstract = {Fragile X syndrome (FXS) is mostly caused by a CGG triplet expansion in the fragile X mental retardation 1 gene (FMR1). Up to 60\% of affected males fulfill criteria for autism spectrum disorder (ASD), making FXS the most frequent monogenetic cause of syndromic ASD. It is unknown, however, whether normal variants (independent of mutations) in the fragile X gene family (FMR1, FXR1, FXR2) and in FMR2 modulate autistic features. Here, we report an accumulation model of 8 SNPs in these genes, associated with autistic traits in a discovery sample of male patients with schizophrenia (N = 692) and three independent replicate samples: patients with schizophrenia (N = 626), patients with other psychiatric diagnoses (N = 111) and a general population sample (N = 2005). For first mechanistic insight, we contrasted microRNA expression in peripheral blood mononuclear cells of selected extreme group subjects with high-versus low-risk constellation regarding the accumulation model. Thereby, the brain-expressed miR-181 species emerged as potential "umbrella regulator", with several seed matches across the fragile X gene family and FMR2. To conclude, normal variation in these genes contributes to the continuum of autistic phenotypes.}, language = {en} } @phdthesis{Wollny2019, author = {Wollny, Claudia}, title = {Der p97-Kofaktor UBXD1 ist ein neuer Regulator des NF-kB-Signalweges}, doi = {10.25972/OPUS-13243}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-132430}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2019}, abstract = {Die essenzielle, Ubiquitin-selektive ATPase p97 reguliert eine Vielzahl unterschiedlicher Prozesse in Eukaryoten. Dazu z{\"a}hlen Proteinqualit{\"a}tskontrolle, DNA-Reparatur, Signaltransduktion, Zellzykluskontrolle, Autophagie sowie das endolysosomale System. Diese unterschiedlichen Funktionen von p97 werden durch die Bindung von Kofaktoren engmaschig gesteuert und kontrolliert. Die gr{\"o}ßte und am besten untersuchte Gruppe von p97-Kofaktoren sind die Proteine der UBX Familie. Diese zeichnen sich durch den Besitz einer UBX-Dom{\"a}ne aus, welche die Bindung an p97 vermittelt. Das in h{\"o}heren Eukaryoten konservierte Familienmitglied UBXD1 besitzt dar{\"u}ber hinaus mit einer PUB-Dom{\"a}ne und einem VIM-Motiv noch mindestens zwei weitere p97-Bindemodule. UBXD1 kann an Vesikel des endolysosomalen Degradationssytems lokalisieren, seine genauen zellul{\"a}ren Funktionen sind jedoch noch weitgehend unbekannt. Ziel dieser Arbeit war die funktionelle Charakterisierung von humanem UBXD1. Daf{\"u}r wurden Kandidaten eines zuvor durchgef{\"u}hrten Yeast-Two-Hybrid-Screens auf ihre Two Hybrid-Interaktion mit unterschiedlichen UBXD1-Varianten getestet. Dar{\"u}ber hinaus wurde durch Immunpr{\"a}zipitationsexperimente untersucht, ob die Kandidatenproteine auch in S{\"a}ugerzellen mit UBXD1 interagieren. Als vielversprechende neue Bindungspartner von UBXD1 wurden so die Ubiquitin-Ligase TRIAD3A und das Ubiquitin-editierende Protein A20 identifiziert. Desweiteren konnte gezeigt werden, dass die Interaktion zwischen UBXD1 und A20 von einer funktionellen PUB Dom{\"a}ne und dem siebten Zinkfinger Motiv von A20 abh{\"a}ngig ist. Da sowohl TRIAD3A als auch A20 negative Regulatoren des NF B Signalweges sind, wurde daraufhin untersucht, ob auch UBXD1 eine Funktion in diesem Signalweg besitzt. Tats{\"a}chlich war in UBXD1-depletierten HeLa 57A-Zellen die NF B-abh{\"a}ngige Expression eines Reportgens nach Aktivierung des Signalweges durch TNF, IL-1, Doxorubicin und H2O2 stark reduziert. Dabei spricht die verringerte Aktivierung nach unterschiedlichen Stimuli f{\"u}r eine generelle Rolle von UBXD1 im NF B Signalweg. Durch quantitative Echtzeit-PCR konnte gezeigt werden, dass in HeLa- und HEK293T-Zellen nach UBXD1-Depletion auch die Expression endogener NF B Zielgene verringert ist. Da in UBXD1-depletierten Zellen nach Stimulation mit TNF oder IL-1 bereits die Kerntranslokation des NF B-Transkriptionsfaktor p65 reduziert ist, ist davon auszugehen, dass UBXD1 an einer fr{\"u}heren Phase der Aktivierung des Signalweges beteiligt ist. M{\"o}glicherweise ist dies darauf zur{\"u}ckzuf{\"u}hren, dass UBXD1 bekannte Funktionen von A20 reguliert und etwa die Bindung von A20 an Vesikel des endolysosomalen Systems oder an lineare Ubiquitinketten beeinflusst. Diese Arbeit beschreibt somit eine neue Funktion des p97-Kofaktors UBXD1 im NF B-Signalweg.}, subject = {Ubiquitin}, language = {de} } @article{SpiveyDeGiorgiZhaoetal.2012, author = {Spivey, Tara L. and De Giorgi, Valeria and Zhao, Yingdong and Bedognetti, Davide and Pos, Zoltan and Liu, Qiuzhen and Tomei, Sara and Ascierto, Maria Libera and Uccellini, Lorenzo and Reinboth, Jennifer and Chouchane, Lotfi and Stroncek, David F. and Wang, Ena and Marincola, Francesco M.}, title = {The stable traits of melanoma genetics: an alternate approach to target discovery}, series = {BMC Genomics}, volume = {13}, journal = {BMC Genomics}, number = {156}, doi = {10.1186/1471-2164-13-156}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-131992}, year = {2012}, abstract = {Background: The weight that gene copy number plays in transcription remains controversial; although in specific cases gene expression correlates with copy number, the relationship cannot be inferred at the global level. We hypothesized that genes steadily expressed by 15 melanoma cell lines (CMs) and their parental tissues (TMs) should be critical for oncogenesis and their expression most frequently influenced by their respective copy number. Results: Functional interpretation of 3,030 transcripts concordantly expressed (Pearson's correlation coefficient p-value < 0.05) by CMs and TMs confirmed an enrichment of functions crucial to oncogenesis. Among them, 968 were expressed according to the transcriptional efficiency predicted by copy number analysis (Pearson's correlation coefficient p-value < 0.05). We named these genes, "genomic delegates" as they represent at the transcriptional level the genetic footprint of individual cancers. We then tested whether the genes could categorize 112 melanoma metastases. Two divergent phenotypes were observed: one with prevalent expression of cancer testis antigens, enhanced cyclin activity, WNT signaling, and a Th17 immune phenotype (Class A). This phenotype expressed, therefore, transcripts previously associated to more aggressive cancer. The second class (B) prevalently expressed genes associated with melanoma signaling including MITF, melanoma differentiation antigens, and displayed a Th1 immune phenotype associated with better prognosis and likelihood to respond to immunotherapy. An intermediate third class (C) was further identified. The three phenotypes were confirmed by unsupervised principal component analysis. Conclusions: This study suggests that clinically relevant phenotypes of melanoma can be retraced to stable oncogenic properties of cancer cells linked to their genetic back bone, and offers a roadmap for uncovering novel targets for tailored anti-cancer therapy.}, 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{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} } @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{WeibelBasseLuesebrinkHessetal.2013, author = {Weibel, Stephanie and Basse-Luesebrink, Thomas Christian and Hess, Michael and Hofmann, Elisabeth and Seubert, Carolin and Langbein-Laugwitz, Johanna and Gentschev, Ivaylo and Sturm, Volker J{\"o}rg Friedrich and Ye, Yuxiang and Kampf, Thomas and Jakob, Peter Michael and Szalay, Aladar A.}, title = {Imaging of Intratumoral Inflammation during Oncolytic Virotherapy of Tumors by \(^{19}\)F-Magnetic Resonance Imaging (MRI)}, series = {PLoS ONE}, volume = {8}, journal = {PLoS ONE}, number = {3}, doi = {10.1371/journal.pone.0056317}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-130311}, pages = {e56317}, year = {2013}, abstract = {Background Oncolytic virotherapy of tumors is an up-coming, promising therapeutic modality of cancer therapy. Unfortunately, non-invasive techniques to evaluate the inflammatory host response to treatment are rare. Here, we evaluate \(^{19}\)F magnetic resonance imaging (MRI) which enables the non-invasive visualization of inflammatory processes in pathological conditions by the use of perfluorocarbon nanoemulsions (PFC) for monitoring of oncolytic virotherapy. Methodology/Principal Findings The Vaccinia virus strain GLV-1h68 was used as an oncolytic agent for the treatment of different tumor models. Systemic application of PFC emulsions followed by \(^1H\)/\(^{19}\)F MRI of mock-infected and GLV-1h68-infected tumor-bearing mice revealed a significant accumulation of the \(^{19}\)F signal in the tumor rim of virus-treated mice. Histological examination of tumors confirmed a similar spatial distribution of the \(^{19}\)F signal hot spots and \(CD68^+\)-macrophages. Thereby, the \(CD68^+\)-macrophages encapsulate the GFP-positive viral infection foci. In multiple tumor models, we specifically visualized early inflammatory cell recruitment in Vaccinia virus colonized tumors. Furthermore, we documented that the \(^{19}\)F signal correlated with the extent of viral spreading within tumors. Conclusions/Significance These results suggest \(^{19}\)F MRI as a non-invasive methodology to document the tumor-associated host immune response as well as the extent of intratumoral viral replication. Thus, \(^{19}\)F MRI represents a new platform to non-invasively investigate the role of the host immune response for therapeutic outcome of oncolytic virotherapy and individual patient response.}, language = {en} } @article{KrehanHeubeckMenzeletal.2012, author = {Krehan, Mario and Heubeck, Christian and Menzel, Nicolas and Seibel, Peter and Sch{\"o}n, Astrid}, title = {RNase MRP RNA and RNase P activity in plants are associated with a Pop1p containing complex}, series = {Nucleic Acids Research}, volume = {40}, journal = {Nucleic Acids Research}, number = {16}, doi = {10.1093/nar/gks476}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-130648}, pages = {7956- 7966}, year = {2012}, abstract = {RNase P processes the 5'-end of tRNAs. An essential catalytic RNA has been demonstrated in Bacteria, Archaea and the nuclei of most eukaryotes; an organism-specific number of proteins complement the holoenzyme. Nuclear RNase P from yeast and humans is well understood and contains an RNA, similar to the sister enzyme RNase MRP. In contrast, no protein subunits have yet been identified in the plant enzymes, and the presence of a nucleic acid in RNase P is still enigmatic. We have thus set out to identify and characterize the subunits of these enzymes in two plant model systems. Expression of the two known Arabidopsis MRP RNA genes in vivo was verified. The first wheat MRP RNA sequences are presented, leading to improved structure models for plant MRP RNAs. A novel mRNA encoding the central RNase P/MRP protein Pop1p was identified in Arabidopsis, suggesting the expression of distinct protein variants from this gene in vivo. Pop1p-specific antibodies precipitate RNase P activity and MRP RNAs from wheat extracts. Our results provide evidence that in plants, Pop1p is associated with MRP RNAs and with the catalytic subunit of RNase P, either separately or in a single large complex.}, 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{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} } @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} } @article{WangChenMinevetal.2012, author = {Wang, Huiqiang and Chen, Nanhai G. and Minev, Boris R. and Szalay, Aladar A.}, title = {Oncolytic vaccinia virus GLV-1h68 strain shows enhanced replication in human breast cancer stem-like cells in comparison to breast cancer cells}, series = {Journal of Translational Medicine}, volume = {10}, journal = {Journal of Translational Medicine}, number = {167}, doi = {10.1186/1479-5876-10-167}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-130019}, year = {2012}, abstract = {Background: Recent data suggest that cancer stem cells (CSCs) play an important role in cancer, as these cells possess enhanced tumor-forming capabilities and are responsible for relapses after apparently curative therapies have been undertaken. Hence, novel cancer therapies will be needed to test for both tumor regression and CSC targeting. The use of oncolytic vaccinia virus (VACV) represents an attractive anti-tumor approach and is currently under evaluation in clinical trials. The purpose of this study was to demonstrate whether VACV does kill CSCs that are resistant to irradiation and chemotherapy. Methods: Cancer stem-like cells were identified and separated from the human breast cancer cell line GI-101A by virtue of increased aldehyde dehydrogenase 1 (ALDH1) activity as assessed by the ALDEFLUOR assay and cancer stem cell-like features such as chemo-resistance, irradiation-resistance and tumor-initiating were confirmed in cell culture and in animal models. VACV treatments were applied to both ALDEFLUOR-positive cells in cell culture and in xenograft tumors derived from these cells. Moreover, we identified and isolated CD44\(^+\)CD24\(^+\)ESA\(^+\) cells from GI-101A upon an epithelial-mesenchymal transition (EMT). These cells were similarly characterized both in cell culture and in animal models. Results: We demonstrated for the first time that the oncolytic VACV GLV-1h68 strain replicated more efficiently in cells with higher ALDH1 activity that possessed stem cell-like features than in cells with lower ALDH1 activity. GLV-1h68 selectively colonized and eventually eradicated xenograft tumors originating from cells with higher ALDH1 activity. Furthermore, GLV-1h68 also showed preferential replication in CD44\(^+\)CD24\(^+\)ESA\(^+\) cells derived from GI-101A upon an EMT induction as well as in xenograft tumors originating from these cells that were more tumorigenic than CD44\(^+\)CD24\(^-\)ESA\(^+\) cells. Conclusions: Taken together, our findings indicate that GLV-1h68 efficiently replicates and kills cancer stem-like cells. Thus, GLV-1h68 may become a promising agent for eradicating both primary and metastatic tumors, especially tumors harboring cancer stem-like cells that are resistant to chemo and/or radiotherapy and may be responsible for recurrence of tumors.}, language = {en} } @article{PatilGentschevAdelfingeretal.2012, author = {Patil, Sandeep S. and Gentschev, Ivaylo and Adelfinger, Marion and Donat, Ulrike and Hess, Michael and Weibel, Stephanie and Nolte, Ingo and Frentzen, Alexa and Szalay, Aladar A.}, title = {Virotherapy of Canine Tumors with Oncolytic Vaccinia Virus GLV-1h109 Expressing an Anti-VEGF Single-Chain Antibody}, series = {PLoS One}, volume = {7}, journal = {PLoS One}, number = {10}, doi = {10.1371/journal.pone.0047472}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-130039}, pages = {e47472}, year = {2012}, abstract = {Virotherapy using oncolytic vaccinia virus (VACV) strains is one promising new strategy for cancer therapy. We have previously reported that oncolytic vaccinia virus strains expressing an anti-VEGF (Vascular Endothelial Growth Factor) single-chain antibody (scAb) GLAF-1 exhibited significant therapeutic efficacy for treatment of human tumor xenografts. Here, we describe the use of oncolytic vaccinia virus GLV-1h109 encoding GLAF-1 for canine cancer therapy. In this study we analyzed the virus-mediated delivery and production of scAb GLAF-1 and the oncolytic and immunological effects of the GLV-1h109 vaccinia virus strain against canine soft tissue sarcoma and canine prostate carcinoma in xenograft models. Cell culture data demonstrated that the GLV-1h109 virus efficiently infect, replicate in and destroy both tested canine cancer cell lines. In addition, successful expression of GLAF-1 was demonstrated in virus-infected canine cancer cells and the antibody specifically recognized canine VEGF. In two different xenograft models, the systemic administration of the GLV-1h109 virus was found to be safe and led to anti-tumor and immunological effects resulting in the significant reduction of tumor growth in comparison to untreated control mice. Furthermore, tumor-specific virus infection led to a continued production of functional scAb GLAF-1, resulting in inhibition of angiogenesis. Overall, the GLV-1h109-mediated cancer therapy and production of immunotherapeutic anti-VEGF scAb may open the way for combination therapy concept i.e. vaccinia virus mediated oncolysis and intratumoral production of therapeutic drugs in canine cancer patients.}, language = {en} } @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{WangChenMinevetal.2013, author = {Wang, Huiqiang and Chen, Nanhai G. and Minev, Boris R. and Zimmermann, Martina and Aguilar, Richard J. and Zhang, Qian and Sturm, Julia B. and Fend, Falko and Yu, Yong A. and Cappello, Joseph and Lauer, Ulrich M. and Szalay, Aladar A.}, title = {Optical Detection and Virotherapy of Live Metastatic Tumor Cells in Body Fluids with Vaccinia Strains}, series = {PLoS ONE}, volume = {8}, journal = {PLoS ONE}, number = {9}, doi = {10.1371/journal.pone.0071105}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-130059}, pages = {e71105}, year = {2013}, abstract = {Metastatic tumor cells in body fluids are important targets for treatment, and critical surrogate markers for evaluating cancer prognosis and therapeutic response. Here we report, for the first time, that live metastatic tumor cells in blood samples from mice bearing human tumor xenografts and in blood and cerebrospinal fluid samples from patients with cancer were successfully detected using a tumor cell-specific recombinant vaccinia virus (VACV). In contrast to the FDA-approved CellSearch system, VACV detects circulating tumor cells (CTCs) in a cancer biomarker-independent manner, thus, free of any bias related to the use of antibodies, and can be potentially a universal system for detection of live CTCs of any tumor type, not limited to CTCs of epithelial origin. Furthermore, we demonstrate for the first time that VACV was effective in preventing and reducing circulating tumor cells in mice bearing human tumor xenografts. Importantly, a single intra-peritoneal delivery of VACV resulted in a dramatic decline in the number of tumor cells in the ascitic fluid from a patient with gastric cancer. Taken together, these results suggest VACV to be a useful tool for quantitative detection of live tumor cells in liquid biopsies as well as a potentially effective treatment for reducing or eliminating live tumor cells in body fluids of patients with metastatic disease.}, 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} } @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} } @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{Pelz2015, author = {Pelz, Jann-Patrick}, title = {Strukturbiologische Untersuchungen zur Chaperone-vermittelten Zusammenlagerung spleißosomaler U-snRNPs}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-116973}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2015}, abstract = {Durch die Spleißreaktion werden nicht-kodierende Sequenzelemente (Introns) aus eukaryotischen Vorl{\"a}ufer-mRNAs entfernt und die kodierenden Sequenzelemente (Exons) miteinander zu einem offenen Leserahmen verbunden. Dieser zentrale Prozessierungsschritt w{\"a}hrend der eukaryotischen Genexpression wird durch das Spleißosom katalysiert, das aus den vier kleinen nukle{\"a}ren Ribonucleoproteinpartikeln (snRNPs) U1, U2, U4/U6 und U5, sowie einer Vielzahl weiterer Proteinfaktoren gebildet wird. Alle snRNPs besitzen eine gemeinsame ringf{\"o}rmige Kernstruktur, die aus sieben gemeinsamen Sm-Proteinen (SmB/B'-D1-D2-D3-E-F-G) besteht, die ein einzelstr{\"a}ngiges Sequenzmotiv auf der snRNAs binden. W{\"a}hrend sich diese, als Sm-Core-Dom{\"a}ne bezeichnete Struktur in vitro spontan ausbilden kann, erfolgt die Zusammenlagerung in vivo in einem assistierten und hochregulierten Prozess. Dieser ist abh{\"a}ngig von insgesamt mindestens 12 trans-agierenden Faktoren, die in den PRMT5- und SMN-Komplexen organisiert sind. Der PRMT5-Komplex agiert in der fr{\"u}hen Phase der Zusammenlagerung, indem er die Sm-Proteine durch die Untereinheit pICln rekrutiert und die symmetrische Methylierung von Argininresten in den C terminalen Schw{\"a}nzen von SmB/B', SmD1 und SmD3 katalysiert. Als Resultat dieser fr{\"u}hen Phase befinden sich die Sm-Proteine SmD1-D2-E-F-G und SmB/B'-D3 in zwei getrennten und durch pICln organisierten Komplexen. W{\"a}hrend SmB/B'-D3-pICln am PRMT5-Komplex gebunden bleibt, existiert der zweite Komplex als freies Intermediat mit einem Sedimentationskoeffizienten von 6S. Diese Intermediate k{\"o}nnen nicht mit RNA assoziieren, sodass f{\"u}r die Fortsetzung des Zusammenlagerungsprozesses die Interaktion der Sm-Proteine mit pICln aufgel{\"o}st werden muss. Dies geschieht in der sp{\"a}ten Phase der Sm-Core-Zusammenlagerung, in der die Sm-Proteine vom SMN-Komplex (bestehend aus SMN, Gemin2-8 und unrip) {\"u}bernommen werden und pICln dissoziiert wird. Dadurch werden die Sm-Proteine f{\"u}r ihre Interaktion mit der snRNA aktiviert und k{\"o}nnen auf die Sm-Bindestelle transferiert werden, wodurch die Formierung des Sm-Core abgeschlossen wird. Im Rahmen dieser Arbeit konnten mit Hilfe einer Kombination r{\"o}ntgenkristallographischer und elektronenmikroskopischer Methoden zwei wichtige Intermediate dieses Zusammenlagerungs-prozesses strukturbiologisch charakterisiert werden. Bei diesen Intermediaten handelt es sich um den 6S-Komplex, sowie um ein Sm-Protein-Transferintermediat mit einem Sedimentations-koeffizienten von 8S. In diesem ist der 6S-Komplex an zwei zentrale Untereinheiten des SMN-Komplexes (SMN und Gemin2) gebunden, w{\"a}hrend pICln den Komplex noch nicht verlassen hat. Der 8S-Komplex stellt daher ein „gefangenes" Intermediat zwischen der fr{\"u}hen und sp{\"a}ten Phase der Zusammenlagerung dar. Zun{\"a}chst gelang es eine erste Kristallform des rekombinant hergestellten 8S-Komplexes zu erhalten, die jedoch keine Strukturl{\"o}sung erlaubte. Durch eine kombinierte Optimierung der Kristallisationsbedingung und der verwendeten Proteine wurde eine weitere {\"a}hnliche Kristallform erhalten, mit der die Kristallstruktur des 8S-Komplexes gel{\"o}st werden konnte. Die Kristallisation des 6S-Komplexes gelang im Anschluss auf Basis der Hypothese, dass Kristalle beider Komplexe aufgrund der kompositionellen Verwandtschaft zwischen 6S und 8S auch {\"A}hnlichkeiten in der Architektur ihrer Kristallgitter aufweisen k{\"o}nnten. Daher wurden innerhalb von pICln gezielt Aminos{\"a}uren substituiert, die sich innerhalb von Kristallkontakten der 8S-Kristalle befanden und konformationell eingeschr{\"a}nkt waren. Mit entsprechend rekonstituierten 6S-Pr{\"a}parationen konnten dann zwei Kristallformen erzeugt werden, die eine Strukturl{\"o}sung des 6S-Komplexes erm{\"o}glichten. Durch die Kristallstruktur des 6S-Komplexes konnte f{\"u}r pICln eine strukturelle Mimikry der Sm-Proteine identifiziert werden. Diese erm{\"o}glicht eine Bindung der Sm-Proteine und eine fr{\"u}hzeitige topologische Organisation des Sm-Pentamers D1-D2-F-E-G in einer geschlossenen hexameren Ringstruktur. Die Kristallstruktur des 8S-Komplexes zeigt, wie der SMN-Komplex {\"u}ber Gemin2 an das Sm-Pentamer bindet. In Kombination mit einer EM-Struktur des 8S-Komplexes gelang es weiterhin, einen plausiblen Mechanismus f{\"u}r die Elimination von pICln und die Aktivierung der Sm-Proteine f{\"u}r die snRNA-Bindung zu formulieren. Somit konnten diese Arbeiten zu einem besseren Verst{\"a}ndnis der Funktionen von trans-agierenden Faktoren bei Zusammenlagerung von RNA-Protein-Komplexen in vivo beitragen.}, subject = {Spleißosom}, language = {de} } @article{HofmannWeibelSzalay2014, author = {Hofmann, Elisabeth and Weibel, Stephanie and Szalay, Aladar A.}, title = {Combination treatment with oncolytic Vaccinia virus and cyclophosphamide results in synergistic antitumor effects in human lung adenocarcinoma bearing mice}, doi = {10.1186/1479-5876-12-197}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-110168}, year = {2014}, abstract = {Background The capacity of the recombinant Vaccinia virus GLV-1h68 as a single agent to efficiently treat different human or canine cancers has been shown in several preclinical studies. Currently, its human safety and efficacy are investigated in phase I/II clinical trials. In this study we set out to evaluate the oncolytic activity of GLV-1h68 in the human lung adenocarcinoma cell line PC14PE6-RFP in cell cultures and analyzed the antitumor potency of a combined treatment strategy consisting of GLV-1h68 and cyclophosphamide (CPA) in a mouse model of PC14PE6-RFP lung adenocarcinoma. Methods PC14PE6-RFP cells were treated in cell culture with GLV-1h68. Viral replication and cell survival were determined by plaque assays and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays, respectively. Subcutaneously implanted PC14PE6-RFP xenografts were treated by systemic injection of GLV-1h68, CPA or a combination of both. Tumor growth and viral biodistribution were monitored and immune-related antigen profiling of tumor lysates was performed. Results GLV-1h68 efficiently infected, replicated in and lysed human PC14PE6-RFP cells in cell cultures. PC14PE6-RFP tumors were efficiently colonized by GLV-1h68 leading to much delayed tumor growth in PC14PE6-RFP tumor-bearing nude mice. Combination treatment with GLV-1h68 and CPA significantly improved the antitumor efficacy of GLV-1h68 and led to an increased viral distribution within the tumors. Pro-inflammatory cytokines and chemokines were distinctly elevated in tumors of GLV-1h68-treated mice. Factors expressed by endothelial cells or present in the blood were decreased after combination treatment. A complete loss in the hemorrhagic phenotype of the PC14PE6-RFP tumors and a decrease in the number of blood vessels after combination treatment could be observed. Conclusions CPA and GLV-1h68 have synergistic antitumor effects on PC14PE6-RFP xenografts. We strongly suppose that in the PC14PE6-RFP model the enhanced tumor growth inhibition achieved by combining GLV-1h68 with CPA is due to an effect on the vasculature rather than an immunosuppressive action of CPA. These results provide evidence to support further preclinical studies of combining GLV-1h68 and CPA in other highly angiogenic tumor models. Moreover, data presented here demonstrate that CPA can be combined successfully with GLV-1h68 based oncolytic virus therapy and therefore might be promising as combination therapy in human clinical trials.}, language = {en} }