• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 500
  • 122
  • 70
  • 35
  • 30
  • 18
  • 9
  • 9
  • 6
  • 3
  • 3
  • 3
  • 3
  • 3
  • 3
  • Tagged with
  • 1022
  • 188
  • 187
  • 175
  • 174
  • 173
  • 138
  • 129
  • 125
  • 116
  • 108
  • 86
  • 85
  • 85
  • 79
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
211

Functional Analysis of the Caenorhabditis elegans HP1 Homolog HPL-2 in a Chromatin Context

Miller, Elizabeth Victoria 09 1900 (has links)
The heterochromatin 1 (HP1) family of non-histone chromosomal proteins is evolutionarily conserved and is involved in numerous biological processes, including the stabilization of heterochromatin, a state of compacted DNA along a protein scaffold. HP1 proteins and trimethylated histone H3 on lysine 9 (H3K9me3) are major constituents of heterochromatin and have been characterized extensively in vitro. The binding of HP1 proteins to H3K9 methylation marks plays an essential role in mammalian development and chromatin organization. However, due to their critical function, dissecting the molecular mechanism by which HP1 proteins exert their function in vivo is difficult. C. elegans is a unique model because not only are deletion mutants of the two HP1 homologs, HPL-1 and HPL-2, viable, but also H3K9 methylation is not essential to worm development. Interestingly, HPL-2 is alternatively spliced to generate two HP1 proteins, but in vivo experimentation has vastly ignored the potential contributions of the alternative transcripts to hpl-2 function, thus obfuscating which phenotypes associated with hpl-2 knockdown are due to the loss of one or more of the splicing variants. In this dissertation, I characterized the HPL-2 splicing variants (A and B) on a biochemical level in relation to the canonical human HP1b protein and on a physiological level in splicing variant-specific knockout worms. I show that both recombinant HPL-2A and HPL-2B bind H3K9me3 through their chromodomain (CD). But while HPL-2A acts as a canonical HP1 protein, namely it dimerizes and phase-separates like hHP1b, HPL-2B does not. In contrast to recombinant protein, in extracts both proteins rely on other factors, such as the MBT domain-containing protein LIN-61, for their recruitment to H3K9me3. Although HPL-2A and HPL-2B display distinct characteristics in vitro, both hpl-2a and hpl-2b worms are phenotypically wildtype. In agreement, knockout of either splicing variant leads to upregulated expression of the other one, suggesting a certain level of functional redundancy. Nevertheless, I show that the C-terminal extension of HPL-2B, which is absent in HPL-2A, resembles that of the CEC-4 heterochromatin anchor. I therefore hypothesize that the main functions of HPL-2 are distinct: HPL-2A mediates chromatin compaction and HPL-2B facilitates heterochromatin anchoring to the nuclear periphery.
212

Conservation and Regulation of the Essential Epigenetic Regulator UHRF1 Across Vertebrata Orthologs

Aljahani, Abrar 05 1900 (has links)
UHRF1 is a critical epigenetic regulator which serves as a molecular model for understanding the crosstalk between histone modification and DNA methylation. It is integrated in the process of DNA maintenance methylation through its histone ubiquitylation activity, ultimately functioning as a recruiter of DNA methyltransferase 1 (DNMT1). As the faithful propagation of DNA methylation patterns during cell division is a common molecular phenomenon among vertebrates, understanding the underlying conserved mechanism of UHRF1 for executing such a key process is important. Here, I present a broad-range evolutionary comparison of UHRF1 binding behavior and enzymatic activity of six species spanning across the vertebrata subphylum. According to their distinct binding modes to differentially methylated histone H3, a pattern is emerging which separates between mammalian and nonmammalian orthologs. H. sapiens, P. troglodytes and M. musculus UHRF1 orthologs utilize the functionality of both TTD and PHD domains to interact with histone H3 peptides, while G. gallus, X. laevis, and D. rerio employ either TTD or PHD. Further, UHRF1 allosteric regulation by 16:0 PI5P is a unique case to primate orthologs where H3K9me3 peptide binding is enhanced upon hUHRF1 and pUHRF1 interacting with 16:0 PI5P. This is due to their closed and autoinhibited conformation wherein TTD is blocked by the PBR region in linker 4. 16:0 PI5P outcompetes TTD for PBR binding resulting in a release of TTD blockage, hence, enhanced H3K9me3 binding. However, owing to the lack of phosphatidylinositol binding specificity and reduced sequence conservation of linker 4, the regulatory impact of 16:0 PI5P in avian and lower vertebrate orthologs could not be detected. Additionally, all UHRF1 orthologs exert their ubiquitylation enzymatic activity on histone H3 substrates, supporting the notion that the overall functionality of UHRF1 orthologs is conserved, despite their divergent molecular approaches. Taken together, my findings suggest that UHRF1 orthologs adopt distinct conformational states with a differential response to the allosteric regulators 16:0 PI5P and hemi-methylated DNA.
213

Regulation of heterochromatin formation by the JmjC-domain protein Epe1

Bao, Kehan January 2021 (has links)
In eukaryotic cells, DNA wraps around histones to form nucleosomes, which are the basic units of chromatin. Chromatin is classified as active euchromatin or repressive heterochromatin, depending on the modifications on histones and DNA. Heterochromatin, which is defined by the presence of histone modifications such as H3K9 methylation, serves important functions in cells such as silencing transposable elements, preventing aberrant recombination, and regulating gene expression.The fission yeast, which shares basic chromatin modification pathways with higher eukaryotes, is a premier model system for study heterochromatin formation. One important heterochromatin regulator is the JmjC-domain protein Epe1. It contains a conserved JmjC domain, which is commonly found in active demethylases. Despite that no in vitro demethylase activity has been demonstrated, Epe1 has been regarded as an H3K9 demethylase based on genetic evidence. However, the mechanism of its regulation is unclear at the beginning of my studies. In this thesis, I investigated the regulation of Epe1 through an unbiased genetic screen to identify factors important for Epe1 functions. From the screen, I identified multiple subunits within a transcriptional coactivator SAGA complex. I determined that Epe1 physically recruits SAGA to heterochromatin to promote histone acetylation and transcription, which provides a mechanism for a long-standing paradox regarding heterochromatin at repetitive DNA elements: heterochromatin normally represses transcription but the formation of heterochromatin requires transcription of the repeats. While past results suggest a role of Epe1 in promoting transcription of repeats, our results demonstrate how Epe1 promotes transcription. From this screen, I also identified multiple genes in the cAMP signaling pathway that are important for Epe1 function. I demonstrated that the cAMP signaling pathway regulates Epe1 protein levels post-transcriptionally, and this effect was also seen in cells experiencing glucose starvation, which dampens the cAMP signaling. This study uncovers another layer of control of Epe1 and provides a critical link between nutrient conditions and heterochromatin regulation. Altogether, my studies identified both a mechanism by which Epe1 promotes transcription within heterochromatin and a layer of Epe1 regulation by the glucose-sensing cAMP signaling pathway. These results will help future studies on Epe1 functions and how it is involved in epigenetic adaptation to changing nutrient conditions.
214

H2A.Z – a molecular guardian of RNA polymerase II transcription in African trypanosomes / H2A.Z – eine molekulare Wächterin der RNA Polymerase II Transkription in Afrikanischen Trypanosomen

Kraus, Amelie Johanna January 2021 (has links) (PDF)
In eukaryotes, the enormously long DNA molecules need to be packaged together with histone proteins into nucleosomes and further into compact chromatin structures to fit it into the nucleus. This nuclear organisation interferes with all phases of transcription that require the polymerase to bind to DNA. During transcription – the process in which the hereditary information stored in DNA is transferred to many transportable RNA molecules - nucleosomes form a physical obstacle for polymerase progression. Thus, transcription is usually accompanied by processes mediating nucleosome destabilisation, including post-translational histone modifications (PTMs) or exchange of canonical histones by their variant forms. To the best of our knowledge, acetylation of histones has the highest capability to induce chromatin opening. The lysine modification can destabilise histone-DNA interactions within a nucleosome and can serve as a binding site for various chromatin remodelers that can modify the nucleosome composition. For example, H4 acetylation can impede chromatin folding and can stimulate the exchange of canonical H2A histone by its variant form H2A.Z at transcription start sites (TSSs) in many eukaryotes, including humans. As histone H4, H2A.Z can be post-translationally acetylated and as acetylated H4, acetylated H2A.Z is enriched at TSSs suggested to be critical for transcription. However, thus far, it has been difficult to study the cause and consequence of H2A.Z acetylation. Even though, genome-wide chromatin profiling studies such as ChIP-seq have already revealed the genomic localisation of many histone PTMs and variant proteins, they can only be used to study individual chromatin marks and not to identify all factors important for establishing a distinct chromatin structure. This would require a comprehensive understanding of all marks associated to a specific genomic locus. However, thus far, such analyses of locus-specific chromatin have only been successful for repetitive regions, such as telomeres. In my doctoral thesis, I used the unicellular parasite Trypanosoma brucei as a model system for chromatin biology and took advantage of its chromatin landscape with TSSs comprising already 7% of the total T. brucei genome (humans: 0.00000156%). Atypical for a eukaryote, the protein-coding genes are arranged in long polycistronic transcription units (PTUs). Each PTU is controlled by its own ~10 kb-wide TSS, that lies upstream of the PTU. As observed in other eukaryotes, TSSs are enriched with nucleosomes containing acetylated histones and the histone variant H2A.Z. This is why I used T. brucei to particularly investigate the TSS-specific chromatin structures and to identify factors involved in H2A.Z deposition and transcription regulation in eukaryotes. To this end, I established an approach for locus-specific chromatin isolation that would allow me to identify the TSSs- and non-TSS-specific chromatin marks. Later, combining the approach with a method for quantifying lysine-specific histone acetylation levels, I found H2A.Z and H4 acetylation enriched in TSSs-nucleosomes and mediated by the histone acetyltransferases HAT1 and HAT2. Depletion of HAT2 reduced the levels of TSS-specific H4 acetylation, affected targeted H2A.Z deposition and shifted the sites of transcription initiation. Whereas HAT1 depletion had only a minor effect on H2A.Z deposition, it had a strong effect on H2A.Z acetylation and transcription levels. My findings demonstrate a clear link between histone acetylation, H2A.Z deposition and transcription initiation in the early diverged unicellular parasite T. brucei, which was thus far not possible to determine in other eukaryotes. Overall, my study highlights the usefulness of T. brucei as a model system for studying chromatin biology. My findings allow the conclusion that H2A.Z regardless of its modification state defines sites of transcription initiation, whereas H2A.Z acetylation is essential co-factor for transcription initiation. Altogether, my data suggest that TSS-specific chromatin establishment is one of the earliest developed mechanisms to control transcription initiation in eukaryotes. / In Eukaryoten muss die genomische DNA zusammen mit Histonproteinen zu Nukleosomen und weiter zu kompakten Chromatinstrukturen verpackt werden, damit sie in den Zellkern passt. Diese Organisation behindert die Transkription bei jedem Schritt, bei dem die Polymerase an der DNA bindet. Während der Transkription – dem Prozess bei dem die in der DNA gespeicherte Erbinformation in viele transportable RNA Molekülen umgewandelt wird – stellen Nukleosomen ein physikalisches Hindernis für das Vorankommen der Polymerase dar. Aus diesem Grund wird die Transkription üblicherweise von Prozessen begleitet, die für die Destabilisierung der Nukleosomen sorgen, wie zum Beispiel post-translationale Modifizierung (PTM) der Histone oder der Austausch von kanonischen Histonproteinen durch eine ihrer Varianten. Soweit bisher bekannt ist Histonacetylierung am besten dafür geeignet, eine offene Chromatinstruktur bereit zu stellen. Die Lysinmodifizierung kann Interaktionen zwischen der DNA und den Histonen innerhalb eines Nukleosomes destabilisieren und als Andockstelle für einige Proteinkomplexe sogenannte Chromatin-Modellierer fungieren, die die Zusammensetzung eines Nukleosomes verändern können. Zum Beispiel, kann Acetylierung am Histon H4 das „Zusammenfalten“ des Chromatins erschweren und den Austausch von kanonischem H2A mit ihrer Variante H2A.Z an den Transkriptiosinitiationsstellen (TSSen) in vielen eukaryotischen Organismen, Menschen eingeschlossen, stimulieren. Wie Histon H4, kann auch H2A.Z post-translationell acetyliert werden und wie acetyliertes H4, findet man auch acetyliertes H2A.Z vor allem an TSSen. Deswegen geht man davon aus, dass es sehr wichtig für die Transkriptioninitiierung ist. Allerdings war es bisher nicht möglich, die Ursache und Wirkung von H2A.Z Acetylierung genauer zu untersuchen. Genom-weite Chromatinprofilstudien wie z.B. ChIP-Seq ermöglichen es die genomische Lokalisierung von vielen Histon-Modifizierungen und -Varianten zu bestimmen. Dennoch reichen sie nicht dafür aus alle Faktoren, die für die Bildung einer bestimmten Chromatinstruktur notwendig sind, gleichzeitig herauszufinden. Das würde voraussetzen, dass man alle Merkmale der genomischen Stelle kennt. Bisher waren Analysen von spezifischen Chromatinstellen nur erfolgreich, wenn das Chromatin von einer repetitiven Region, wie z.B. Telomeren, stammt. In meiner Doktorarbeit verwendete ich den einzelligen Parasiten Trypanosoma brucei als Modelsystem für Chromatinbiologie. Dabei machte ich mir dessen Chromatinorganisation zunutze, die eher untypisch für einen eukaryotische Organismus ist. TSSen machen hier ungefähr 7% des gesamten Genoms aus (Mensch: 0.00000156%). Protein-kodierende Gene sind in langen polycistronischen Transkriptionseinheiten (PTE) angeordnet. Jede dieser Einheiten besitzt eine eigene TSS, die vor der PTE liegt, und bis zu 10 kb lang sein kann. Jedoch, wie in anderen Eukaryoten, sind an den TSSen Nukleosomen angereichert, die sich durch acetylierte Histone und den Einbau der Histonvariante H2A.Z auszeichnen. Aus diesen Gründen verwendete ich T. brucei, um während meiner Doktorarbeit die Chromatinstrukturen, die TSSen auszeichnen, genauer zu untersuchen und die Faktoren, die bei der H2A.Z Positionierung und dadurch bei der Transkriptionsregulation in Eukaryoten eine Rolle spielen, herauszufinden. Dafür etablierte ich zuerst eine Methode, mit der man Chromatin von einer bestimmten genomischen Stelle isolieren kann und die es mir ermöglichen würde, die Merkmale von TSS-spezifischen und -unspezifischen Chromatin zu identifizieren. Später konnte ich das entwickelte Protokoll mit einer Methode zur Quantifizierung von Lysin-spezifischen Histonacetylierung kombinieren. Dadurch konnte ich herausfinden, dass Nukleosomen an trypanosomischen TSSen stark acetyliertes H2A.Z und H4 enthalten und dass für diese Modifizierungen die Histonacetyltransferasen HAT1 und HAT2 verantwortlich sind. Eine Reduzierung der HAT2-Levels führte zu einer Reduzierung von H4 Acetylierung, verschlechterte die gezielte H2A.Z Positionierung und führte dazu, dass die Transkriptioninitiierung sich verlagerte. Wohingegen eine Reduzierung von HAT1, die zwar nur einen kleinen Einfluss auf die H2A.Z Positionierung hatte, eine sehr starke Verringerung von acetyliertem H2A.Z und der Transkriptionsrate zur Folge hatte. Durch meine Ergebnisse konnte ich zeigen, dass in T. brucei, einem evolutionär divergenten eukaryotischem Organismus, die Prozesse der Histonacetylierung, H2A.Z Positionierung und Transkriptionsinitiierung sehr stark miteinander verbunden sind. Meine Arbeit ist des weiteren ein Beweis dafür, dass T. brucei ein sehr wichtiger Modellorganismus für die Forschung an Chromatin ist. Insgesamt erlauben meine Ergebnisse die Schlussfolgerung, dass H2A.Z, egal ob modifiziert oder nicht, ein Herausstellungsmerkmal für TSSen ist, während acetyliertes H2A.Z essentiell für die Transkriptionsinitiierung darstellt. Zusammengefasst, weisen die Daten meiner Doktorarbeit darauf hin, dass die Etablierung von bestimmten Chromatinstrukturen an TSSen eines der frühesten entwickelten Mechanismen zur Kontrolle der Transkriptionsinitiierung in Eukaryoten ist.
215

Identification of New Roles for Histone Acetyltransferase 1

Agudelo Garcia, Paula A. 11 August 2017 (has links)
No description available.
216

The Chimeric Fusion Protein SETMAR Functions as a Chromatin Organizing Factor

Bates, Alison Melissa 08 1900 (has links)
Indiana University-Purdue University Indianapolis (IUPUI) / About 50 million years ago, an Hsmar1 transposon invaded an early primate genome and inserted itself downstream of a SET methyltransferase gene, leading to the birth of a new chimeric protein now called SETMAR. While all other Hsmar1 sequences in the human genome have suffered inactivating mutational damage, the transposase domain of SETMAR has remained remarkably intact, suggesting that it has gained a novel, evolutionarily advantageous function. While SETMAR can no longer transpose itself throughout the genome, it has retained its ancestral sequence-specific DNA binding activity, the importance of which is currently unknown. To investigate this, we performed ChIP-seq to examine SETMAR binding in the human genome. We also utilized RNA-sequencing to assess SETMAR overexpression as well as SETMAR deletion on the human transcriptome. Additionally, we explored SETMAR’s transposase-derived chromatin-looping ability using chromosome-conformation-capture-on-ChIP (4C) in the presence of SETMAR overexpression and performed genome-wide Hi-C to assess the impact of complete SETMAR silencing on global chromatin interactions. ChIP-seq revealed that SETMAR amassed 7,332 unique binding sites, 69% of which included a TIR motif. RNA-sequencing in cells overexpressing SETMAR indicated 177 differentially regulated transcripts, including repression of 17 histone transcripts, suggesting a possible role in chromatin dynamics. RNA-sequencing of parental and SETMAR knockout clones highlighted an average of 5,000 altered transcripts in each cell line, with 343 transcripts significantly differentially expressed in all three knockout clones, many of which participate in embryonic development pathways. 4C analysis in the presence of SETMAR overexpression discovered multiple intrachromosomal looping interactions, and Hi-C analysis of SETMAR knockout cell lines uncovered genome-wide loss of chromatin interactions and disruption of TAD boundaries. The prevalence of SETMAR binding in the human genome combined with its chromatin looping capability and its dramatic effects on the transcriptome suggest a previously undiscovered role for SETMAR as a novel chromatin organizing factor. / 2022-08-17
217

Nucleosome Regulation of Transcription Factor Binding Kinetics: Implications for Gene Expression

Donovan, Benjamin Thomas January 2019 (has links)
No description available.
218

3D Epigenome Dynamics in Normal and Stalled Development / 正常および遅延発生における3Dエピゲノムダイナミクス

Hu, Bo 26 September 2022 (has links)
京都大学 / マギル大学 / 新制・課程博士 / 博士(ゲノム医学) / 甲第24204号 / 医博JD第2号 / 新制||医||JD1(附属図書館) / 京都大学大学院医学研究科京都大学マギル大学ゲノム医学国際連携専攻 / (主査)准教授 Wilson Michael (トロント大学), 教授 竹内 理, 准教授 Bailey Swneke (マギル大学), 教授 伊藤 貴浩, 教授 Shoubridge Eric (マギル大学) / 学位規則第4条第1項該当 / Doctor of Philosophy in Human Genetics / Kyoto University / McGill University / DFAM
219

Modelling Sifrim-Hitz-Weiss Syndrome Using Mouse Genetics

Larrigan, Sarah 25 May 2023 (has links)
Neurodevelopmental disorders encompass a spectrum of different conditions with both genetic and environmental etiologies. Although rapid progress has been made in deciphering the genetic landscape of these disorders, in most cases, it remains unclear how mutations undermine neurodevelopmental mechanisms. However, increasing identification of risk genes suggests chromatin remodelling is frequently impacted. For instance, de novo variants encoding the chromatin remodeller CHD4 causes Sifrim-Hitz-Weiss syndrome, which manifests as an overgrowth-intellectual disability syndrome. To further understand Chd4’s role during cortical development, we excised the ATPase domain of Chd4 in the germline or specifically in the developing telencephalon, creating three mouse models. Germline heterozygotes presented a slight decrease in brain weight, cortex area and Ctip2+ cells, with females displaying more overt impairments in learning and memory. Telencephalon-specific conditional heterozygotes exhibited slight changes in white matter, increased repetitive movements and altered social behaviours. Telencephalon-specific conditional knockouts presented with decreased brain size, brain weight, and cortex thickness due to decreased upper layer neurons, and anxiety phenotypes. These data reveal an unexpected complexity in the impacts of Chd4 mutations on neurodevelopmental processes and behaviour.
220

Changes in chromatin accessibility by oncogenic YAP and its relevance for regulation of cell cycle gene expression and cell migration / Änderungen der Chromatinzugänglichkeit durch onkogene YAP und seine Relevanz für die Genexpression während des Zellzyklus und für die Zellmigration

Fetiva Mora, Maria Camila January 2023 (has links) (PDF)
Various types of cancer involve aberrant cell cycle regulation. Among the pathways responsible for tumor growth, the YAP oncogene, a key downstream effector of the Hippo pathway, is responsible for oncogenic processes including cell proliferation, and metastasis by controlling the expression of cell cycle genes. In turn, the MMB multiprotein complex (which is formed when B-MYB binds to the MuvB core) is a master regulator of mitotic gene expression, which has also been associated with cancer. Previously, our laboratory identified a novel crosstalk between the MMB-complex and YAP. By binding to enhancers of MMB target genes and promoting B-MYB binding to promoters, YAP and MMB co-regulate a set of mitotic and cytokinetic target genes which promote cell proliferation. This doctoral thesis addresses the mechanisms of YAP and MMB mediated transcription, and it characterizes the role of YAP regulated enhancers in transcription of cell cycle genes. The results reported in this thesis indicate that expression of constitutively active, oncogenic YAP5SA leads to widespread changes in chromatin accessibility in untransformed human MCF10A cells. ATAC-seq identified that newly accessible and active regions include YAP-bound enhancers, while the MMB-bound promoters were found to be already accessible and remain open during YAP induction. By means of CRISPR-interference (CRISPRi) and chromatin immuniprecipitation (ChIP), we identified a role of YAP-bound enhancers in recruitment of CDK7 to MMB-regulated promoters and in RNA Pol II driven transcriptional initiation and elongation of G2/M genes. Moreover, by interfering with the YAP-B-MYB protein interaction, we can show that binding of YAP to B-MYB is also critical for the initiation of transcription at MMB-regulated genes. Unexpectedly, overexpression of YAP5SA also leads to less accessible chromatin regions or chromatin closing. Motif analysis revealed that the newly closed regions contain binding motifs for the p53 family of transcription factors. Interestingly, chromatin closing by YAP is linked to the reduced expression and loss of chromatin-binding of the p53 family member Np63. Furthermore, I demonstrate that downregulation of Np63 following expression of YAP is a key step in driving cellular migration. Together, the findings of this thesis provide insights into the role of YAP in the chromatin changes that contribute to the oncogenic activities of YAP. The overexpression of YAP5SA not only leads to the opening of chromatin at YAP-bound enhancers which together with the MMB complex stimulate the expression of G2/M genes, but also promotes the closing of chromatin at ∆Np63 -bound regions in order to lead to cell migration. / Ein Kennzeichen vieler Tumoren ist die fehlerhafte Aktivierung von zellzyklusregulierenden Signalwegen. Ein für das Tumorwachstum wichtiger Signalwege ist der Hippo-Signalweg und das durch ihn regulierte Onkogen YAP, ein transkriptioneller Koaktivator. Durch die Regulierung von Zellzyklusgenen ist YAP verantwortlich für onkogene Prozesse wie Zellproliferation und Metastasierung. Der MMB-Multiproteinkomplex wiederum – er entsteht, wenn B-MYB an das MuvB-Kernmodul bindet – ist ein wichtiger Regulator der mitotischen Genexpression, welche ebenso mit der Tumorentstehung in Verbindung gebracht wurde. Unser Labor hat zuvor einen neuen Mechanismus der Regulation mitotischer Gene durch den MMB-Komplex und YAP identifiziert: Durch die Bindung an Enhancer der MMB-Zielgene und die Förderung der B-MYB-Bindung an Promotoren reguliert YAP eine Reihe von mitotischen und zytokinetischen Zielgenen, welche die Zellproliferation fördern. Diese Doktorarbeit befasst sich mit den Mechanismen der YAP- und MMB-vermittelten Transkription und charakterisiert die Rolle der YAP regulierten Enhancer während der Transkription von Zellzyklusgenen. Die in dieser Dissertation dargelegten Ergebnisse zeigen, dass die Expression von konstitutiv aktivem, onkogenem YAP5SA zu weitreichenden Veränderungen in der Chromatinzugänglichkeit nicht-transformierter humaner MCF10A Zellen führt. ATAC-seq zeigte, dass ein grosse Anzahl YAP-gebundene Enhancer zugänglich und aktiviert werden. Gleichzeitig konnte festgestellt werden, dass die MMB-gebundenen Promotoren bereits vor der Expression von YAP zugänglich sind und während der YAP-Induktion offen bleiben. Mittels CRISP-Interferenz (CRISPRi) und Chromatin-Immunpräzipitationen (ChIP) konnten wir zeigen, dass YAP-gebundene Enhancer die Rekrutierung von CDK7 an MMB-regulierten Promotoren sowie die Initiation und Elongation der Transkription von G2/M Genen fördert. Durch die experimentelle Blockade der YAP-B-MYB Proteininteraktion konnten wir darüber hinaus belegen, dass auch die Bindung von YAP an B-MYB für die Initiation der Transkription an MMB-regulierten Genen entscheidend ist. Unerwarteterweise führte die Überexpression von YAP5SA auch dazu, dass bestimmte Regionen im Genom weniger zugänglich werden. Motivanalysen ergaben, dass diese neu geschlossenen Regionen Bindungsmotive für die p53-Familie von Transkriptionsfaktoren enthalten. Die Chromatinschließung durch YAP ist an eine reduzierte Expression und an den Verlust der Chromatinbindung des p53-Familienmitglieds Np63 gekoppelt. Schließlich konnte gezeigt werden, dass die Inhibition von Np63 durch YAP ein wichtiger Schritt in der YAP-abhängigen Förderung der Zellmigration ist. Zusammenfassend liefern die Ergebnisse dieser Dissertation Einblicke in die Rolle von YAP bei Chromatinveränderungen welche zu den onkogenen Aktivitäten von YAP beitragen. Die Überexpression von YAP führt dabei einerseits zur Öffnung des Chromatins an YAP-gebundenen Enhancern, die zusammen mit dem MMB-Komplex die Expression von G2/M Genen stimulieren. Andererseits fördert YAP auch das Schließen von Chromatin an Np63-gebundenen Regionen, was wiederum Zellmigration nach sich zieht.

Page generated in 0.0385 seconds