@phdthesis{Lueffe2023, author = {L{\"u}ffe, Teresa Magdalena}, title = {Behavioral and pharmacological validation of genetic zebrafish models for ADHD}, doi = {10.25972/OPUS-25716}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-257168}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2023}, abstract = {Attention-deficit/hyperactivity disorder (ADHD) is the most prevalent neurodevelopmental disorder described in psychiatry today. ADHD arises during early childhood and is characterized by an age-inappropriate level of inattention, hyperactivity, impulsivity, and partially emotional dysregulation. Besides, substantial psychiatric comorbidity further broadens the symptomatic spectrum. Despite advances in ADHD research by genetic- and imaging studies, the etiopathogenesis of ADHD remains largely unclear. Twin studies suggest a heritability of 70-80 \% that, based on genome-wide investigations, is assumed to be polygenic and a mixed composite of small and large, common and rare genetic variants. In recent years the number of genetic risk candidates is continuously increased. However, for most, a biological link to neuropathology and symptomatology of the patient is still missing. Uncovering this link is vital for a better understanding of the disorder, the identification of new treatment targets, and therefore the development of a more targeted and possibly personalized therapy. The present thesis addresses the issue for the ADHD risk candidates GRM8, FOXP2, and GAD1. By establishing loss of function zebrafish models, using CRISPR/Cas9 derived mutagenesis and antisense oligonucleotides, and studying them for morphological, functional, and behavioral alterations, it provides novel insights into the candidate's contribution to neuropathology and ADHD associated phenotypes. Using locomotor activity as behavioral read-out, the present work identified a genetic and functional implication of Grm8a, Grm8b, Foxp2, and Gad1b in ADHD associated hyperactivity. Further, it provides substantial evidence that the function of Grm8a, Grm8b, Foxp2, and Gad1b in activity regulation involves GABAergic signaling. Preliminary indications suggest that the three candidates interfere with GABAergic signaling in the ventral forebrain/striatum. However, according to present and previous data, via different biological mechanisms such as GABA synthesis, transmitter release regulation, synapse formation and/or transcriptional regulation of synaptic components. Intriguingly, this work further demonstrates that the activity regulating circuit, affected upon Foxp2 and Gad1b loss of function, is involved in the therapeutic effect mechanism of methylphenidate. Altogether, the present thesis identified altered GABAergic signaling in activity regulating circuits in, presumably, the ventral forebrain as neuropathological underpinning of ADHD associated hyperactivity. Further, it demonstrates altered GABAergic signaling as mechanistic link between the genetic disruption of Grm8a, Grm8b, Foxp2, and Gad1b and ADHD symptomatology like hyperactivity. Thus, this thesis highlights GABAergic signaling in activity regulating circuits and, in this context, Grm8a, Grm8b, Foxp2, and Gad1b as exciting targets for future investigations on ADHD etiopathogenesis and the development of novel therapeutic interventions for ADHD related hyperactivity. Additionally, thigmotaxis measurements suggest Grm8a, Grm8b, and Gad1b as interesting candidates for prospective studies on comorbid anxiety in ADHD. Furthermore, expression analysis in foxp2 mutants demonstrates Foxp2 as regulator of ADHD associated gene sets and neurodevelopmental disorder (NDD) overarching genetic and functional networks with possible implications for ADHD polygenicity and comorbidity. Finally, with the characterization of gene expression patterns and the generation and validation of genetic zebrafish models for Grm8a, Grm8b, Foxp2, and Gad1b, the present thesis laid the groundwork for future research efforts, for instance, the identification of the functional circuit(s) and biological mechanism(s) by which Grm8a, Grm8b, Foxp2, and Gad1b loss of function interfere with GABAergic signaling and ultimately induce hyperactivity.}, language = {en} } @phdthesis{JimenezMartin2022, author = {Jim{\´e}nez Mart{\´i}n, Ovidio Manuel}, title = {Analysis of MYCN and MAX alterations in Wilms Tumor}, doi = {10.25972/OPUS-24291}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-242919}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2022}, abstract = {Wilms tumor (WT) is the most common renal tumor in childhood. Among others, MYCN copy number gain and MYCN P44L and MAX R60Q mutations have been identified in WT. The proto-oncogene MYCN encodes a transcription factor that requires dimerization with MAX to activate transcription of numerous target genes. MYCN gain has been associated with adverse prognosis. The MYCN P44L and MAX R60Q mutations, located in either the transactivating or basic helix-loop-helix domain, respectively, are predicted to be damaging by different pathogenicity prediction tools. These mutations have been reported in several other cancers and remain to be functionally characterized. In order to further describe these events in WT, we screened both mutations in a large cohort of unselected WT patients, to check for an association of the mutation status with certain histological or clinical features. MYCN P44L and MAX R60Q revealed frequencies of 3 \% and 0.9 \% and also were significantly associated to higher risk of relapse and metastasis, respectively. Furthermore, to get a better understanding of the MAX mutational landscape in WT, over 100 WT cases were analyzed by Sanger sequencing to identify other eventual MAX alterations in its coding sequence. R60Q remained the only MAX CDS alteration described in WT to date. To analyze the potential functional consequences of these mutations, we used a doxycycline-inducible system to overexpress each mutant in HEK293 cells. This biochemical characterization identified a reduced transcriptional activation potential for MAX R60Q, while the MYCN P44L mutation did not change activation potential or protein stability. The protein interactome of N-MYC-P44L was likewise not altered as shown by mass spectrometric analyses of purified N-MYC complexes. However, we could identify a number of novel N-MYC partner proteins, several of these known for their oncogenic potential. Their correlated expression in WT samples suggested a role in WT oncogenesis and they expand the range of potential biomarkers for WT stratification and targeting, especially for high-risk WT.}, subject = {Nephroblastom}, language = {en} } @phdthesis{Vardapour2022, author = {Vardapour, Romina}, title = {Mutations in the DROSHA/DGCR8 microprocessor complex in high-risk blastemal Wilms tumor}, doi = {10.25972/OPUS-23140}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-231404}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2022}, abstract = {Wilms tumor (WT) or nephroblastoma is the most common kidney tumor in childhood. Several genetic alterations have been identified in WT over the past years. However, a clear-cut underlying genetic defect has remained elusive. Growing evidence suggests that miRNA processing genes play a major role in the formation of pediatric tumors, including WT. We and others have identified the microprocessor genes DROSHA and DGCR8 as key players in Wilms tumorigenesis. Exome sequence analysis of a cohort of blastemal-type WTs revealed the recurrent hotspot mutations DROSHA E1147K and DGCR8 E518K mapping to regions important for catalyic activity and RNA-binding. These alterations were expected to affect processing of miRNA precursors, ultimately leading to altered miRNA expression. Indeed, mutated tumor samples were characterized by distinct miRNA patterns. Notably, these mutations have been observed almost exclusively in WT, suggesting that they play a specific role in WT formation. The aim of the present work was to first examine the mutation frequency of DROSHA E1147K and DGCR8 E518K in a larger cohort of WTs, and to further characterize these microprocessor gene mutations as potential oncogenic drivers for WT formation. Screening of additional 700 WT samples by allele-specific PCR revealed a high frequency of DROSHA E1147K and DGCR8 E518K mutations, with the highest incidence found in tumors of high-risk histology. DROSHA E1147K was heterozygously expressed in all cases, which strongly implies a dominant negative effect. In contrast, DGCR8 E518K exclusively exhibited homozygous expression, suggestive for the mutation to act recessive. To functionally assess the mutations of the microprocessor complex in vitro, I generated stable HEK293T cell lines with inducible overexpression of DROSHA E1147K, and stable mouse embryonic stem cell (mESC) lines with inducible overexpression of DGCR8 E518K. To mimic the homozygous expression observed in WT, DGCR8 mESC lines were generated on a DGCR8 knockout background. Inducible overexpression of wild-type or mutant DROSHA in HEK293T cells showed that DROSHA E1147K leads to a global downregulation of miRNA expression. It has previously been shown that the knockout of DGCR8 in mESCs also results in a significant downregulation of canonical miRNAs. Inducible overexpression of wild type DGCR8 rescued this processing defect. DGCR8 E518K on the other hand, only led to a partial rescue. Differentially expressed miRNAs comprised members of the ESC cell cycle (ESCC) and let-7 miRNA families whose antagonism is known to play a pivotal role in the regulation of stem cell properties. Along with altered miRNA expression, DGCR8-E518K mESCs exhibited alterations in target gene expression potentially affecting various biological processes. We could observe decreased proliferation rates, most likely due to reduced cell viability. DGCR8-E518K seemed to be able to overcome the block of G1-S transition and to rescue the cell cycle defect in DGCR8-KO mESCs, albeit not to the full extent like DGCR8-wild-type. Moreover, DGCR8-E518K appeared to be unable to completely block epithelial-to-mesenchymal transition (EMT). Embryoid bodies (EBs) with the E518K mutation, however, were still able to silence the self-renewal program rescuing the differentiation defect in DGCR8-KO mESCs. Taken together, I could show that DROSHA E1147K and DGCR8 E518K are frequent events in WT with the highest incidence in high-risk tumor entities. Either mutation led to altered miRNA expression in vitro confirming our previous findings in tumor samples. While the DROSHA E1147K mutation resulted in a global downregulation of canonical miRNAs, DGCR8 E518K was able to retain significant activity of the microprocessor complex, suggesting that partial reduction of activity or altered specificity may be critical in Wilms tumorigenesis. Despite the significant differences found in the miRNA and mRNA profiles of DGCR8 E518K and DGCR8-wild-type mESCs, functional analysis showed that DGCR8 E518K could mostly restore important cellular functions in the knockout and only slightly differed from the wild-type situation. Further studies in a rather physiological environment, such as in a WT blastemal model system, may additionally help to better assess the subtle differences between DGCR8 E518K and DGCR8 wild-type observed in our mESC lines. Together with our findings, these model systems may thus contribute to better understand the role of these microprocessor mutations in the formation of WT.}, subject = {Nephroblastom}, language = {en} } @phdthesis{Gruendl2021, author = {Gr{\"u}ndl, Marco}, title = {Biochemical characterization of the MMB-Hippo crosstalk and its physiological relevance for heart development}, doi = {10.25972/OPUS-21332}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-213328}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2021}, abstract = {The Myb-MuvB (MMB) complex plays an essential role in the time-dependent transcriptional activation of mitotic genes. Recently, our laboratory identified a novel crosstalk between the MMB-complex and YAP, the transcriptional coactivator of the Hippo pathway, to coregulate a subset of mitotic genes (Pattschull et al., 2019). Several genetic studies have shown that the Hippo-YAP pathway is essential to drive cardiomyocyte proliferation during cardiac development (von Gise et al., 2012; Heallen et al., 2011; Xin et al., 2011). However, the exact mechanisms of how YAP activates proliferation of cardiomyocytes is not known. This doctoral thesis addresses the physiological role of the MMB-Hippo crosstalk within the heart and characterizes the YAP-B-MYB interaction with the overall aim to identify a potent inhibitor of YAP. The results reported in this thesis indicate that complete loss of the MMB scaffold protein LIN9 in heart progenitor cells results in thinning of ventricular walls, reduced cardiomyocyte proliferation and early embryonic lethality. Moreover, genetic experiments using mice deficient in SAV1, a core component of the Hippo pathway, and LIN9-deficient mice revealed that the correct function of the MMB complex is critical for proliferation of cardiomyocytes due to Hippo-deficiency. Whole genome transcriptome profiling as well as genome wide binding studies identified a subset of Hippo-regulated cell cycle genes as direct targets of MMB. By proximity ligation assay (PLA), YAP and B-MYB were discovered to interact in embryonal cardiomyocytes. Biochemical approaches, such as co-immunoprecipitation assays, GST-pulldown assays, and µSPOT-based peptide arrays were employed to characterize the YAP-B-MYB interaction. Here, a PY motif within the N-terminus of B-MYB was found to directly interact with the YAP WW-domains. Consequently, the YAP WW-domains were important for the ability of YAP to drive proliferation in cardiomyocytes and to activate MMB target genes in differentiated C2C12 cells. The biochemical information obtained from the interaction studies was utilized to develop a novel competitive inhibitor of YAP called MY-COMP (Myb-YAP competition). In MY-COMP, the protein fragment of B-MYB containing the YAP binding domain is fused to a nuclear localization signal. Co-immunoprecipitation studies as well as PLA revealed that the YAP-B-MYB interaction is robustly blocked by expression of MY-COMP. Adenoviral overexpression of MY-COMP in embryonal cardiomyocytes suppressed entry into mitosis and blocked the pro-proliferative function of YAP. Strikingly, characterization of the cellular phenotype showed that ectopic expression of MY-COMP led to growth defects, nuclear abnormalities and polyploidization in HeLa cells. Taken together, the results of this thesis reveal the mechanism of the crosstalk between the Hippo signaling pathway and the MMB complex in the heart and form the basis for interference with the oncogenic activity of the Hippo coactivator YAP.}, subject = {Zellzyklus}, language = {en} } @phdthesis{Roeschert2021, author = {R{\"o}schert, Isabelle}, title = {Aurora-A prevents transcription-replication conflicts in MYCN-amplified neuroblastoma}, doi = {10.25972/OPUS-24303}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-243037}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2021}, abstract = {Neuroblastoma is the most abundant, solid, extracranial tumor in early childhood and the leading cause of cancer-related childhood deaths worldwide. Patients with high-risk neuroblastoma often show MYCN-amplification and elevated levels of Aurora-A. They have a low overall survival and despite multimodal therapy options a poor therapeutic prognosis. MYCN-amplified neuroblastoma cells depend on Aurora-A functionality. Aurora-A stabilizes MYCN and prevents it from proteasomal degradation by competing with the E3 ligase SCFFBXW7. Interaction between Aurora-A and MYCN can be observed only in S phase of the cell cycle and activation of Aurora-A can be induced by MYCN in vitro. These findings suggest the existence of a profound interconnection between Aurora-A and MYCN in S phase. Nevertheless, the details remain elusive and were investigated in this study. Fractionation experiments show that Aurora-A is recruited to chromatin in S phase in a MYCN-dependent manner. Albeit being unphosphorylated on the activating T288 residue, Aurora-A kinase activity was still present in S phase and several putative, novel targets were identified by phosphoproteomic analysis. Particularly, eight phosphosites dependent on MYCN-activated Aurora-A were identified. Additionally, phosphorylation of serine 10 on histone 3 was verified as a target of this complex in S phase. ChIP-sequencing experiments reveal that Aurora-A regulates transcription elongation as well as histone H3.3 variant incorporation in S phase. 4sU-sequencing as well as immunoblotting demonstrated that Aurora-A activity impacts splicing. PLA measurements between the transcription and replication machinery revealed that Aurora-A prevents the formation of transcription-replication conflicts, which activate of kinase ATR. Aurora-A inhibitors are already used to treat neuroblastoma but display dose-limiting toxicity. To further improve Aurora-A based therapies, we investigated whether low doses of Aurora-A inhibitor combined with ATR inhibitor could increase the efficacy of the treatment albeit reducing toxicity. The study shows that the combination of both drugs leads to a reduction in cell growth as well as an increase in apoptosis in MYCN-amplified neuroblastoma cells, which is not observable in MYCN non-amplified neuroblastoma cells. This new approach was also tested by a collaboration partner in vivo resulting in a decrease in tumor burden, an increase in overall survival and a cure of 25\% of TH-MYCN mice. These findings indicate indeed a therapeutic window for targeting MYCN-amplified neuroblastoma.}, subject = {Neuroblastom}, language = {en} } @phdthesis{Tomasovic2020, author = {Tomasovic, Angela}, title = {Die ERK-ERK Interaktionsfl{\"a}che als therapeutische Zielstruktur zur selektiven Inhibition nukle{\"a}rer ERK1/2-Funktionen zum Schutz vor pathologischer kardialer Hypertrophie}, doi = {10.25972/OPUS-15430}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-154304}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2020}, abstract = {Die Mitogen-aktivierten Proteinkinasen ERK1/2 (extrazellul{\"a}r Signal-regulierte Kinase 1 und 2) sind die Effektorkinasen der Raf/MEK/ERK-Kaskade und verkn{\"u}pfen externe Stimuli mit der intrazellul{\"a}-ren Antwort, wodurch sie wichtige Schl{\"u}sselmolek{\"u}le der zellul{\"a}ren Signaltransduktion darstellen. Zahlreiche Studien belegen die Beteiligung von ERK1/2 an der Entstehung pathologischer kardialer Hypertrophie. Genauso ist bekannt, dass ERK1/2 anti-apoptotische, kardioprotektive Eigenschaften besitzen. So f{\"u}hrte, wie in dieser Arbeit gezeigt, eine Hemmung der katalytischen ERK1/2-Aktivit{\"a}t durch den MEK-Inhibitor PD98059 zu einer signifikanten Reduktion der hypertrophen Antwort von Kardiomy-ozyten auf den Stimulus Phenylephrin. Dies war allerdings mit einem Anstieg der Apoptoserate in diesen Zellen verbunden, wodurch sich eine Hemmung der totalen ERK-Aktivit{\"a}t als nicht praktika-bel f{\"u}r die Behandlung pathologischer kardialer Hypertrophie herauskristallisierte. In fr{\"u}heren Un-tersuchungen wurde eine Autophosphorylierung von ERK an Threonin 188 (murines ERK2) entdeckt und als Trigger f{\"u}r ERK1/2-vermitteltes hypertrophes Wachstum identifiziert. Diese Autophospho-rylierung steuert die nukle{\"a}re Lokalisation von ERK1/2 und erm{\"o}glicht so die Aktivierung nukle{\"a}rer ERK-Zielproteine sowie hypertrophes Wachstum. Eine Interferenz mit der ERKThr188-Phosphorylierung konnte schon in vitro und in vivo erfolgreich einer pathologischen Hypertrophie entgegenwirken, ohne Einfluss auf physiologisches Herzwachstum oder die zytosolischen, anti-apoptotischen Effekte von ERK1/2 zu nehmen. Einen initialen Schritt f{\"u}r das Zustandekommen dieser Autophosphorylierung an Threonin 188 stellt dabei die Dimerisierung von ERK dar. In der vorliegenden Arbeit wurde daher die Inhibition der ERK-Dimerisierung im Hinblick auf die Behand-lung ERKThr188-vermittelter pathologischer Hypertrophie untersucht. Dabei sollte die endogene ERK-Dimerisierung mithilfe eines selbst generierten Peptids unterbunden werden. In {\"U}bereinstimmung mit den Ergebnissen zu einer dimerisierungsdefizienten ERK2-Mutante (ERK2-Δ4) konnte das Peptid in vitro und in vivo erfolgreich pathologisch hypertrophes Herzwachstum mindern. Dabei f{\"u}hrte es sogar zu einem R{\"u}ckgang des apoptotischen Zelltodes, ausgel{\"o}st durch eine Aortenligation, f{\"u}h-ren. Es zeigte sich, dass das Peptid die nukle{\"a}re Translokation von ERK2 verhindert und dadurch nukle{\"a}re ERK-Substrate geringer aktiviert werden. Da eine Dysregulation in der Raf/MEK/ERK-Kaskade auch die Entstehung von Tumoren beg{\"u}nstigen kann, sollte schließlich untersucht wer-den, ob das Prinzip der Hemmung nukle{\"a}rer ERK-Effekte auch die Proliferation von Krebszellen beeinflussen kann. Es stellte sich heraus, dass die Peptid-vermittelte Hemmung der ERK-Dimerisierung auch die Proliferation von Kolonkarzinomzelllinien mit unterschiedlichen Mutations-stadien der Raf/MEK/ERK-Kaskade reduziert. In der vorliegenden Arbeit konnte somit die Intervention mit der ERK-Dimerisierung als Target der ERKThr188-Autophosphorylierung als translationale Strategie zur Reduktion nukle{\"a}rer ERK-Effekte herausgearbeitet werden. Dies bietet die M{\"o}glichkeit ERK-vermittelte pathologische kardialer Hy-pertrophie und ERK-vermittelte Tumor-Proliferation zu behandeln, ohne kardiotoxische Nebenwir-kungen zu verursachen.}, subject = {Herzinsuffizienz}, language = {de} } @phdthesis{Kruber2019, author = {Kruber, Philip}, title = {Functional analysis of DROSHA and SIX1 mutations in kidney development and Wilms tumor}, doi = {10.25972/OPUS-16141}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-161418}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2019}, abstract = {Wilms tumor (WT) is the most common kidney cancer in childhood. It is a genetically heterogeneous tumor and several genetic alterations have been identified in WT patients. Recurrent mutations were found in the homeo-domain of SIX1 and SIX2 in high proliferative tumors (18.1\% of the blastemal-type tumors) as well as in the microprocessor genes DROSHA and DGCR8 (18.2\% of the blastemal-type tumors), indicating a critical role of the SIX-SALL pathway and aberrant miRNA processing in WT formation. Underlined by the fact that a significant overlap between mutations in DROSHA and SIX1 was found, indicating a synergistic effect. To characterize the in vivo role of DROSHA and SIX mutations during kidney development and their oncogenic potential, I analyzed mouse lines with either a targeted deletion of Drosha or an inducible expression of human DROSHA or SIX1 carrying a tumor-specific E1147K or Q177R mutation, respectively. The DROSHA mutation E1147K was predicted to act in a dominant negative manner. Six2-cre mediated deletion of Drosha in nephron progenitors led to a lethal phenotype with apoptotic loss of progenitor cells and early termination of nephrogenesis. Mosaic deletions via Wt1-creERT2 resulted in a milder phenotype with viable offspring that developed proteinuria after 2-4 weeks, but no evidence of tumor formation. Activation of the DROSHA-E1147K transgene via Six2-cre, on the other hand, induced a more severe phenotype with apoptosis of progenitor cells, proteinuria and glomerular sclerosis. The severely growth-retarded mice died within the first two months. This strong phenotype was consistent with the predicted dominant-negative effect of DROSHA-E1147K. Analysis of the SIX1-Q177R mutation suggested that the mutation leads to a shift in DNA binding specificity instead of a complete loss of DNA binding. This may end up in subtle changes of the gene regulatory capacity of SIX1. Six2-cre mediated activation of SIX1-Q177R lead to a viable phenotype with no alterations or shortened life span. Yet a global activation of SIX1-Q177R mediated by Zp3-cre resulted in bilateral hydronephrosis and juvenile death of the mice. To mimic the synergistic effect of DROSHA and SIX1 mutations, I generated compound mutants in two combinations: A homozygous deletion of Drosha combined with an activation of SIX1-Q177R and a compound mutant with activation of DROSHA-E1147K and SIX1-Q177R. Each mouse model variant displayed new phenotypical alterations. Mice with Six2-cre mediated homozygous deletion of Drosha and activation of SIX1-Q177R were not viable, yet heterozygous deletion of Drosha and activation of SIX1-Q177R led to hydronephrosis, proteinuria and an early death around stage P28. Combined activation of DROSHA-E1147K and SIX1-Q177R under Six2-cre resulted in proteinuria, glomerulosclerosis and lesions inside the kidney. These mice also suffered from juvenile death. Both mouse models could confirm the predicted synergistic effect. While these results underscore the importance of a viable self-renewing progenitor pool for kidney development, there was no evidence of tumor formation. This suggests that either additional alterations in mitogenic or antiapoptotic pathways are needed for malignant transformation, or premature loss of a susceptible target cell population and early lethality prevent WT formation.}, subject = {Nephroblastom}, language = {en} } @phdthesis{Hoffmann2017, author = {Hoffmann, Helene}, title = {Identifying regulators of tumor vascular morphology}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-142348}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2017}, abstract = {In contrast to normal vessels, tumor vasculature is structurally and functionally abnormal. Tumor vessels are highly disorganized, tortuous and dilated, with uneven diameter and excessive branching. Consequently, tumor blood flow is chaotic, which leads to hypoxic and acidic regions in tumors. These conditions lower the therapeutic effectiveness and select for cancer cells that are more malignant and metastatic. The therapeutic outcome could be improved by increasing the functionality and density of the tumor vasculature. Tumor angiogenesis also shows parallels to epithelial to mesenchymal transition (EMT), a process enabling metastasis. Metastasis is a multi-step process, during which tumor cells have to invade the surrounding host tissue to reach the circulation and to be transported to distant sites. We hypothesize that the variability in the phenotype of the tumor vasculature is controlled by the differential expression of key transcription factors. Inhibiting these transcription factors might be a promising way for angiogenic intervention and vascular re-engineering. Therefore, we investigated the interdependence of tumor-, stroma- and immune cell-derived angiogenic factors, transcription factors and resulting vessel phenotypes. Additionally, we evaluated whether transcription factors that regulate EMT are promising targets for vascular remodeling. We used formalin fixed paraffin embedded samples from breast cancer patients, classified according to estrogen-, progesterone- and human epidermal growth factor receptor (HER) 2 status. Establishing various techniques (CD34 staining, laser microdissection, RNA isolation and expression profiling) we systematically analyzed tumor and stroma-derived growths factors. In addition, vascular parameters such as microvessel size, area, circularity and density were assessed. Finally the established expression profiles were correlated with the observed vessel phenotype. As the SNAI1 transcriptional repressor is a key regulator of EMT, we examined the effect of vascular knockdown of Snai1 in murine cancer models (E0771, B16-F10 and lewis lung carcinoma). Among individual mammary carcinomas, but not among subtypes, strong differences of vascular parameters were observed. Also, little difference between lobular carcinomas and ductal carcinomas was found. Vessel phenotype of Her2 enriched carcinomas was similar to that of lobular carcinomas. Vessel morphology of luminal A and B and basal-like tumors resembled each other. Expression of angiogenic factors was variable across subtypes. We discovered an inverse correlation of PDGF-B and VEGF-A with vessel area in luminal A tumors. In these tumors expression of IL12A, an inhibitor of angiogenesis, was also correlated with vessel size. Treatment of endothelial cells with growth factors revealed an increased expression of transcription factors involved in the regulation of EMT. Knockdown of Snai1 in endothelial cells of mice increased tumor growth and decreased hypoxia in the E0771 and the B16-F10 models. In the lewis lung carcinomas, tumor vascularity and biodistribution of doxorubicin were improved. Here, doxorubicin treatment in combination with the endothelial cell-specific knockdown did slow tumor growth. This shows that SNAI1 is important for a tumor's vascularization, with the significance of its role depending on the tumor model. The methods established in this work open the way for the analysis of the expression of key transcription factors in vessels of formalin fixed paraffin embedded tumors. This research enables us to find novel targets for vascular intervention and to eventually design novel targeted drugs to inhibit these targets.}, subject = {Antiangiogenese}, language = {en} } @phdthesis{Stritt2017, author = {Stritt, Simon}, title = {The role of the cytoskeleton in platelet production and the pathogenesis of platelet disorders in humans and mice}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-122662}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2017}, abstract = {Platelets are continuously produced from megakaryocytes (MK) in the bone marrow by a cytoskeleton-driven process of which the molecular regulation is not fully understood. As revealed in this thesis, MK/ platelet-specific Profilin1 (Pfn1) deficiency results in micro- thrombocytopenia, a hallmark of the Wiskott-Aldrich syndrome (WAS) in humans, due to accelerated platelet turnover and premature platelet release into the bone marrow. Both Pfn1-deficient mouse platelets and platelets isolated from WAS patients contained abnormally organized and hyper-stable microtubules. These results reveal an unexpected function of Pfn1 as a regulator of microtubule organization and point to a previously unrecognized mechanism underlying the platelet formation defect in WAS patients. In contrast, Twinfilin2a (Twf2a) was established as a central regulator of platelet reactivity and turnover. Twf2a-deficient mice revealed an age-dependent macrothrombocytopenia that could be explained by a markedly decreased platelet half-life, likely due to the pronounced hyper-reactivity of \(Twf2a^{-/-}\) platelets. The latter was characterized by sustained integrin acti- vation and thrombin generation in vitro that translated into accelerated thrombus formation in vivo. To further elucidate mechanisms of integrin activation, Rap1-GTP-interacting adaptor molecule (RIAM)-null mice were generated. Despite the proposed critical role of RIAM for platelet integrin activation, no alterations in this process could be found and it was concluded that RIAM is dispensable for the activation of β1 and β3 integrins, at least in platelets. These findings change the current mechanistic understanding of platelet integrin activation. Outside-in signaling by integrins and other surface receptors was supposed to regulate MK migration, but also the temporal and spatial formation of proplatelet protrusions. In this the- sis, phospholipase D (PLD) was revealed as critical regulator of actin dynamics and podo- some formation in MKs. Hence, the unaltered platelet counts and production in \(Pld1/2^{-/-}\) mice and the absence of a premature platelet release in the bone marrow of \(Itga2^{-/-}\) mice question the role of podosomes in platelet production and raise the need to reconsider the proposed inhibitory signaling by α2β1 integrins on proplatelet formation. Non-muscle myosin IIA (NMMIIA) has been implicated as a downstream effector of the in- hibitory signals transmitted via α2β1 integrins. Besides Rho-GTPase signaling, also \(Mg^{2+}\) and transient receptor potential melastatin-like 7 (TRPM7) channel α-kinase are known regulators of NMMIIA activity. In this thesis, TRPM7 was identified as major regulator of \(Mg^{2+}\) homeostasis in MKs and platelets. Furthermore, decreased \([Mg^{2+}]_i\) led to deregulated NMMIIA activity and altered cytoskeletal dynamics that impaired thrombopoiesis and resulted in macrothrombocytopenia in humans and mice.}, subject = {Thrombozytopoese}, language = {en} } @phdthesis{Brockmann2015, author = {Brockmann, Markus}, title = {Inhibition von Aurora-A als neue Therapiestrategie gegen MYCN-amplifizierte Neuroblastome}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-135951}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2015}, abstract = {Im Neuroblastom ist die Amplifikation des MYCN-Gens, das f{\"u}r den Transkriptionsfaktor N-Myc kodiert, der klinisch bedeutendste Faktor f{\"u}r eine schlechte Prognose. Als Mitglied der onkogenen Myc-Familie induziert N-Myc die Expression von Genen, die in vielen biologischen Prozessen wie Metabolismus, Zellzyklusprogression, Zellwachstum und Apoptose eine wichtige Rolle spielen. Die Deregulation der MYCN-Expression f{\"u}hrt zu einem charakteristischen Genexpressionsprofil und einem aggressiven Phenotyp in den Tumorzellen. In normalen neuronalen Vorl{\"a}uferzellen wird N-Myc gew{\"o}hnlich sehr schnell proteasomal abgebaut. W{\"a}hrend der Mitose wird N-Myc an Serin 62 phosphoryliert. Diese Phosphorylierung dient als Erkennungssignal f{\"u}r die Kinase GSK3β, die die Phosphorylierung an Threonin 58 katalysiert. Das Phosphodegron wird von Fbxw7, einer Komponente des E3-Ubiquitinligase-Komplex SCFFbxw7, erkannt. Die anschließende Ubiquitinierung induziert den proteasomalen Abbau des Proteins. Die Reduktion der N-Myc-Proteinlevel erm{\"o}glicht den neuronalen Vorl{\"a}uferzellen den Austritt aus dem Zellzyklus und f{\"u}hrt zu einer terminalen Differenzierung. In einem shRNA Screen konnte AURKA als essentielles Gen f{\"u}r die Proliferation MYCN-amplifizierter Neuroblastomzellen identifiziert werden. Eine Aurora-A-Depletion hatte jedoch keinen Einfluss auf das Wachstum nicht-amplifizierter Zellen. W{\"a}hrend dieser Doktorarbeit konnte gezeigt werden, dass Aurora-A speziell den Fbxw7-vermittelten Abbau verhindert und dadurch N-Myc stabilisiert. F{\"u}r die Stabilisierung ist zwar die Interaktion der beiden Proteine von entscheidender Bedeutung, {\"u}berraschenderweise spielt die Kinaseaktivit{\"a}t von Aurora-A jedoch keine Rolle. Zwei spezifische Aurora-A-Inhibitoren, MLN8054 und MLN8237, sind allerdings in der Lage, nicht nur die Kinaseaktivit{\"a}t zu hemmen, sondern auch die N-Myc-Proteinlevel zu reduzieren. Beide Molek{\"u}le induzieren eine Konformations{\"a}nderung in der Kinasedom{\"a}ne von Aurora-A. Diese ungew{\"o}hnliche strukturelle Ver{\"a}nderung hat zur Folge, dass der N-Myc/Aurora-A-Komplex dissoziiert und N-Myc mit Hilfe von Fbxw7 proteasomal abgebaut werden kann. In MYCN-amplifizierten Zellen f{\"u}hrt diese Reduktion an N-Myc zu einem Zellzyklusarrest in der G1-Phase. Die in vitro Daten konnten in einem transgenen Maus-Modell f{\"u}r das MYCN-amplifizierte Neuroblastom best{\"a}tigt werden. Die Behandlung mit MLN8054 und MLN8237 f{\"u}hrte in den Tumoren ebenfalls zu einer N-Myc-Reduktion. Dar{\"u}ber hinaus konnte ein prozentualer Anstieg an differenzierten Zellen, die vollst{\"a}ndige Tumorregression in der Mehrzahl der Neuroblastome und eine gesteigerte Lebenserwartung beobachtet werden. Insgesamt zeigen die in vitro und in vivo Daten, dass die spezifischen Aurora-A-Inhibitoren ein hohes therapeutisches Potential gegen das MYCN-amplifizierte Neuroblastom besitzen.}, subject = {N-Myc}, language = {de} }