@phdthesis{Schmidt2012, author = {Schmidt, Traudel}, title = {Establishment of Hey-triple-KO-ES cells and characterisation of Bre, a Hey binding partner}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-85459}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2012}, abstract = {Hey1, Hey2 and HeyL are downstream effectors of the Notch signalling pathway. Hey genes play decisive roles during embryonic development for example in cardiovascular development. However, the precise transcriptional programmes and genes, which are affected by each single Hey gene, are still poorly understood. One drawback for the analysis of Hey1, Hey2 or HeyL single gene function is that these genes are co-expressed in many tissues and share a high degree of functional redundancy. Thus, it was necessary to establish a system, which is either devoid of Hey expression, or just comprises one single Hey gene family member. For this, Hey1(fl/fl)/Hey2(-/-)/HeyL(-/-)- as well as Hey-triple- knock out (KO)-ES cells (embryonic stem cells) were generated in this work, because ES cells and their differentiation as EBs (embryoid bodies) represent a valuable tool for the in vitro analysis of embryonic developmental processes. After the establishment of Hey1(fl/fl)/Hey2(-/-)/HeyL(-/-)- and Hey-triple- KO-ES cells, it could be seen by ALP staining and pluripotency marker expression that loss of Hey expression did not affect ES cell pluripotency features. Thus, these ES cells represent bona fide ES cells and could be further used for the differentiation as EBs. Here, differences in gene expression between Hey1(fl/fl)/Hey2(-/-)/HeyL(-/-)- and Hey-triple- KO-ES cells (after the loss of Hey1) could be observed in realtime-RT-PCR analysis for the endodermal marker AFP as well as for neural and myogenic markers in d10 EBs. However, the establishment of inducible Hey1, Hey2 or HeyL ES cell lines will be essential to confirm these findings and to search for novel Hey target genes. To get further insight into the mode of Hey action, the analysis of Hey interaction partners is necessary. One such binding partner, the Bre protein, has previously been found in a yeast-two-hybrid screen. Bre has been described to be a member of two distinct complexes (i.e. the nuclear BRCA1-A complex with a function in DNA damage response and the cytoplasmic BRISC complex), to directly interact with the TNF-receptor and Fas and to interfere with apoptotic signalling. The Hey-Bre interaction could be further corroborated in this work; yet, it was not possible to narrow down the interaction site of Bre with Hey1. It rather seems that non-overlapping parts of the Bre protein may bind to Hey. This interaction may be direct- pointing to more than one interaction site inside the Bre protein - or via a common binding partner such as the endogenous Bre protein itself. Besides the interaction studies, functional assays were performed for a more detailed characterisation of Hey1 and Bre interaction. Here, it could be shown that Hey1 over-expression did not have any influence on Bre sub-cellular localisation. Interestingly, it could be demonstrated that Bre positively interfered with Hey1 repressive function in luciferase assays at three of four promoters analysed. Moreover, interaction with Bre seems to lead to a stabilisation of Hey1. As Bre has been described to modulate the E3-ligase activity intrinsic to the BRCC complex it was analysed whether Bre over-expression results in an ubiquitination of Hey1. Yet, this could not be observed in the present work. Furthermore, an interaction of Bre with ubiquitinated proteins could not be demonstrated in an ubiquitin binding assay. To obtain a better insight into Bre function, Bre LacZ gene trap-ES cells and animals were generated. However, realtime-RT-analyses revealed that these cells and mice did not show a loss of Bre expression on mRNA level indicating that insertion mutagenesis did not occur as expected. However, embryos derived from these mice could nevertheless be used for the detection of tissues with Bre expression by β-galactosidase staining. Bre deficiency on mRNA levels was only achieved after the deletion of the floxed exon 3 resulting in the generation of Bre del-mice. Bre del-mice were fertile and without any obvious phenotype and they were used for the generation of Bre del- and wt-MEFs (murine embryonic fibroblasts). Characterisation of these cells showed that proliferation was not affected after loss of Bre (neither under normal nor under stress conditions). However, loss of Bre notably resulted in a reduction in the BRCA1 DNA damage response, in a slightly increased sensitivity towards apoptosis induction by FasL treatment and in an increase in the K63-poly-ubiquitin content in Bre del-cytoplasmic fractions, probably linked to a change in the BRISC de-ubiquitinase activity. Even though these results have the same tendencies as observed in former studies, the effects in the present work are less striking. Further studies as well as intercrossing of Bre del- to Hey KO-animals will be necessary to further understand the functional relevance of Hey and Bre interaction.}, subject = {Embryonale Stammzelle}, language = {en} } @phdthesis{Hofstetter2014, author = {Hofstetter, Christine}, title = {Inhibition of H3K27me-Specific Demethylase Activity During Murine ES cell Differentiation Induces DNA Damage Response}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-107023}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2014}, abstract = {Stem cells are defined by their capacity to self-renew and their potential to differentiate into multiple cell lineages. Pluripotent embryonic stem (ES) cells can renew indefinitely while keeping the potential to differentiate into any of the three germ layers (ectoderm, endoderm or mesoderm). For decades, ES cells are in the focus of research because of these unique features. When ES cells differentiate they form spheroid aggregates termed "embryoid bodies" (EBs). These EBs mimic post- implantation embryonic development and therefore facilitate the understanding of developmented mechanisms. During ES cell differentiation, de-repression or repression of genes accompanies the changes in chromatin structure. In ES cells, several mechanisms are involved in the regulation of the chromatin architecture, including post-translational modifications of histones. Post-translational histone methylation marks became one of the best- investigated epigenetic modifications, and they are essential for maintaining pluripotency. Until the first histone demethylase KDM1A was discovered in 2004 histone modifications were considered to be irreversible. Since then, a great number of histone demethylases have been identified. Their activity is linked to gene regulation as well as to stem cell self-renewal and differentiation. KDM6A and KDM6B are H3K27me3/2-specific histone demethylases, which are known to play a central role in the regulation of posterior development by regulating HOX gene expression. So far less is known about the molecular function of KDM6A or KDM6B in undifferentiated and differentiating ES cells. In order to completely abrogate KDM6A and KDM6B demethylase activity in undifferentiated and differentiating ES cells, a specific inhibitor (GSK-J4) was employed. Treatment with GSK-J4 had no effect on the viability or proliferation on ES cells. However, in the presence of GSK-J4 ES cell differentiation was completely abrogated with cells arrested in G1-phase and an increased rate of apoptosis. Global transcriptome analyses in early-differentiating ES cells revealed that only a limited set of genes were differentially regulated in response to GSK-J4 treatment with more genes up- regulated than down-regulated. Many of the up-regulated genes are linked to DNA damage response (DDR). In agreement with this, DNA damage was found in EBs incubated with GSK-J4. A co-localization of H3K27me3 or KDM6B with γH2AX foci, marking DNA breaks, could be excluded. However, differentiating Eed knockout (KO) ES cells, which are devoid of the H3K27me3 mark, showed an attenuated GSK-J4- induced DDR. Finally, hematopoietic differentiation in the presence of GSK-J4 resulted in a reduced colony-forming potential. This leads to the conclusion that differentiation in the presence of GSK-J4 is also restricted to hematopoietic differentiation. In conclusion, my results show that the enzymatic activity of KDM6A and KDM6B is not essential for maintaining the pluripotent state of ES cells. In contrast, the enzymatic activity of both proteins is indispensable for ES cell and hematopoietic differentiation. Additionally KDM6A and KDM6B enzymatic inhibition in differentiating ES cells leads to increased DNA damage with an activated DDR. Therefore, KDM6A and KDM6B are associated with DNA damage and in DDR in differentiating ES cells.}, subject = {Embryonale Stammzelle}, language = {en} }