@phdthesis{Eltschkner2020, author = {Eltschkner, Sandra}, title = {Targeting the Bacterial Fatty-Acid Synthesis Pathway: Towards the Development of Slow-Onset Inhibitors and the Characterisation of Protein-Protein Interactions}, doi = {10.25972/OPUS-15664}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-156643}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2020}, abstract = {A continuous arms race between the development of novel antibiotics and the evolution of corresponding resistance mechanisms in bacteria has been observed, since antibiotic agents like arsphenamines (e.g. Salvarsan, developed by Paul Ehrlich [1]), sulphonamides (e.g. Prontosil, Gerhard Domagk [2]) and penicillin (Alexander Fleming [3]) were first applied to effectively cure bacterial infections in the early 20th century. The rapid emergence of resistances in contrast to the currently lagging discovery of antibiotics displays a severe threat to human health. Some serious infectious diseases, such as tuberculosis or melioidosis, which were either thought to be an issue only in Third-World countries in case of tuberculosis, or regionally restricted with respect to melioidosis, are now on the rise to expand to other areas. In contrast, methicillin-resistant Staphylococcus aureus (MRSA) is already present in clinical setups all over the world and causes severe infections in immunocompromised patients. Thus, there is an urgent need for new and effective antimicrobial agents, which impair vital functions of the pathogen's metabolism. One central metabolic pathway is represented by the bacterial fatty-acid synthesis pathway (FAS II), which is essential for the synthesis of long and branched-chain fatty acids, as well as mycolic acids. These substances play a major role as modulating components of the properties of the most important protective barrier - the cell envelope. The integrity of the bacterial cell wall and the associated membrane(s) is crucial for cell growth and for protection against physical strain, intrusion of antibiotic agents and regulation of uptake of ions and other small molecules. Thus, this central pathway represents a promising target for antibiotic action against pathogens to combat infectious diseases. The last and rate-limiting step is catalysed by the trans-2-enoyl-ACP reductase (ENR) FabI or InhA (in mycobacteria), which has been demonstrated to be a valuable target for drug design and can be addressed, amongst others, by diphenyl ether (DPE) compounds, derived from triclosan (TCL) - the first one of this class which was discovered to bind to ENR enzymes [4, 5]. Based on this scaffold, inhibitors containing different combinations of substituents at crucial positions, as well as a novel type of substituent at position five were investigated regarding their binding behaviour towards the Burkholderia pseudomallei and Mycobacterium tuberculosis ENR enzymes bpFabI and InhA, respectively, by structural, kinetic and in-vivo experiments. Generally, substitution patterns modulate the association and dissociation velocities of the different ENR inhibitors in the context of the two-step slow-onset binding mechanism, which is observed for both enzymes. These alterations in the rapidity of complex formation and decomposition have a crucial impact on the residence time of a compound and hence, on the pharmacokinetic properties of potential drug candidates. For example, the substituents at the 2'-position of the DPE scaffold influence the ground- and transition state stability during the binding process to bpFabI, whereas 4'-substituents primarily alter the transition state [6]. The novel triazole group attached to the 5-position of the scaffold, targeting the hydrophobic part of the substrate-binding pocket in InhA, significantly enhances the energy barrier of the transition state of inhibitor binding [7] and decelerates the association- as well as the dissociation processes. Combinations with different substituents at the 2'-position can enhance or diminish this effect, e.g. by ground-state stabilisation, which will result in an increased residence time of the respective inhibitor on InhA. Further structural investigations carried out in this work, confirm the proposed binding mode of a customised saFabI inhibitor [8], carrying a pyridone moiety on the DPE scaffold to expand interactions with the protein environment. Structural and preliminary kinetic data confirm the binding of the same inhibitor to InhA in a related fashion. Comparisons with structures of the ENR inhibitor AFN-1252 [9] bound to ENR enzymes from other organisms, addressing a similar region as the pyridone-moiety of the DPE inhibitor, suggest that also the DPE inhibitor bears the potential to display binding to homologues of saFabI and InhA and may be optimised accordingly. Both of the newly investigated substituents, the pyridone moiety at the 4'-position as well as the 5-triazole substituent, provide a good starting point to modify the DPE scaffold also towards improved kinetic properties against ENR enzymes other than the herein studied and combining both groups on the DPE scaffold may have beneficial effects. The understanding of the underlying binding mechanism is a crucial factor to promote the dedicated design of inhibitors with superior pharmacokinetic characteristics. A second target for a structure-based drug-design approach is the interaction surface between ENR enzymes and the acyl-carrier protein (ACP), which delivers the growing acyl chain to each distinct enzyme of the dissociated FAS-II system and presumably recognises its respective interaction partner via electrostatic contacts. The interface between saACP and saFabI was investigated using different approaches including crosslinking experiments and the design of fusion constructs connecting the ACP and the FabI subunits via a flexible linker region of varying lengths and compositions. The crosslinking studies confirmed a set of residues to be part of the contact interface of a previously proposed complex model [10] and displayed high crosslinking efficiency of saACP to saFabI when mutated to cysteine residues. However, crystals of the complex obtained from either the single components, or of the fusion constructs usually displayed weak diffraction, which supports the assumption that complex formation is highly transient. To obtain ordered crystals for structural characterisation of the complex it is necessary to trap the complex in a fixed state, e.g. by a high-affinity substrate attached to ACP [11], which abolishes rapid complex dissociation. For this purpose, acyl-coupled long-residence time inhibitors might be a valuable tool to elucidate the detailed architecture of the ACP-FabI interface. This may provide a novel basis for the development of inhibitors that specifically target the FAS-II biosynthesis pathway.}, subject = {Fetts{\"a}urestoffwechsel}, language = {en} } @phdthesis{Jung2016, author = {Jung, Lisa Anna}, title = {Targeting MYC Function as a Strategy for Tumor Therapy}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-146993}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2016}, abstract = {A large fraction of human tumors exhibits aberrant expression of the oncoprotein MYC. As a transcription factor regulating various cellular processes, MYC is also crucially involved in normal development. Direct targeting of MYC has been a major challenge for molecular cancer drug discovery. The proof of principle that its inhibition is nevertheless feasible came from in vivo studies using a dominant-negative allele of MYC termed OmoMYC. Systemic expression of OmoMYC triggered long-term tumor regression with mild and fully reversible side effects on normal tissues. In this study, OmoMYC's mode of action was investigated combining methods of structural biology and functional genomics to elucidate how it is able to preferentially affect oncogenic functions of MYC. The crystal structure of the OmoMYC homodimer, both in the free and the E-box-bound state, was determined, which revealed that OmoMYC forms a stable homodimer, and as such, recognizes DNA via the same base-specific DNA contacts as the MYC/MAX heterodimer. OmoMYC binds DNA with an equally high affinity as MYC/MAX complexes. RNA-sequencing showed that OmoMYC blunts both MYC-dependent transcriptional activation and repression. Genome-wide DNA-binding studies using chromatin immunoprecipitation followed by high-throughput sequencing revealed that OmoMYC competes with MYC/MAX complexes on chromatin, thereby reducing their occupancy at consensus DNA binding sites. The most prominent decrease in MYC binding was seen at low-affinity promoters, which were invaded by MYC at oncogenic levels. Strikingly, gene set enrichment analyses using OmoMYC-regulated genes enabled the identification of tumor subgroups with high MYC levels in multiple tumor entities. Together with a targeted shRNA screen, this identified novel targets for the eradication of MYC-driven tumors, such as ATAD3A, BOP1, and ADRM1. In summary, the findings suggest that OmoMYC specifically inhibits tumor cell growth by attenuating the expression of rate-limiting proteins in cellular processes that respond to elevated levels of MYC protein using a DNA-competitive mechanism. This opens up novel strategies to target oncogenic MYC functions for tumor therapy.}, subject = {Myc}, language = {en} } @phdthesis{Wencker2022, author = {Wencker, Freya Dorothea Ruth}, title = {The methionine biosynthesis operon in \(Staphylococcus\) \(aureus\): Role of concerted RNA decay in transcript stability and T-box riboswitch turnover}, doi = {10.25972/OPUS-20712}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-207124}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2022}, abstract = {Methionine is the first amino acid of every newly synthesised protein. In combination with its role as precursor for the vital methyl-group donor S-adenosylmethionine, methionine is essential for every living cell. The opportunistic human pathogen Staphylococcus aureus is capable of synthesising methionine de novo, when it becomes scarce in the environment. All genes required for the de novo biosynthesis are encoded by the metICFE-mdh operon, except for metX. Expression is controlled by a hierarchical network with a methionyl-tRNA-specific T-box riboswitch (MET-TBRS) as centrepiece, that is also referred to as met leader (RNA). T-box riboswitches (TBRS) are regulatory RNA elements located in the 5'-untranslated region (5'-UTR) of genes. The effector molecule of T-box riboswitches is uncharged cognate tRNA. The prevailing mechanism of action is premature termination of transcription of the nascent RNA in the absence of the effector (i.e. uncharged cognate tRNA) due to formation of a hairpin structure, the Terminator stem. In presence of the effector, a transient stabilisation of the alternative structure, the Antiterminator, enables transcription of the downstream genes ('read-through'). Albeit, after the read-through the thermodynamically more stable Terminator eventually forms. The Terminator and the Antiterminator are two mutually exclusive structures. Previous work of the research group showed that in staphylococci the MET-TBRS ensures strictly methionine-dependent control of met operon expression. Uncharged methionyl-tRNA that activates the system is only present in sufficient amounts under methionine-deprived conditions. In contrast to other bacterial TBRS, the staphylococcal MET-TBRS has some characteristic features regarding its length and predicted secondary structure whose relevance for the function are yet unkown. Aim of the present thesis was to experimentally determine the structure of the met leader RNA and to investigate the stability of the met operon-specific transcripts in the context of methionine biosynthesis control. Furthermore, the yet unknown function of the mdh gene within the met operon was to be determined. In the context of this thesis, the secondary structure of the met leader was determined employing in-line probing. The structural analysis revealed the presence of almost all highly conserved T-box riboswitch structural characteristics. Furthermore, three additional stems, absent in all T-box riboswitches analysed to date, could be identified. Particularly remarkable is the above average length of the Terminator stem which renders it a potential target of the double-strand-specific endoribonuclease III (RNase III). The RNase III-dependent cleavage of the met leader could be experimentally verified by the use of suitable mutants. Moreover, the exact cleavage site within the Terminator was determined. The unusual immediate separation of the met leader from the met operon mRNA via the RNase III cleavage within the Terminator stem induces the rapid degradation of the met leader RNA and, most likely, that of the 5'-region of the met mRNA. The met mRNA is degraded from its 5'-end by the exoribonuclease RNase J. The stability of the met mRNA was found to vary over the length of the transcript with an instable 5'-end (metI and metC) and a longer half-life towards the 3'-end (metE and mdh). The varying transcript stability is reflected by differences in the available cellular protein levels. The obtained data suggest that programmed mRNA degradation is another level of regulation in the complex network of staphylococcal de novo methionine biosynthesis control. In addition, the MET-TBRS was studied with regard to a future use as a drug target for novel antimicrobial agents. To this end, effects of a dysregulated methionine biosynthesis on bacterial growth and survival were investigated in met leader mutants that either caused permanent transcription of the met operon ('ON') or prevented operon transcription ('OFF'), irrespective of the methionine status in the cell. Methionine deprivation turned out to be a strong selection pressure, as 'OFF' mutants acquired adaptive mutations within the met leader to restore met operon expression that subsequently re-enabled growth. The second part of the thesis was dedicated to the characterisation of the Mdh protein that is encoded by the last gene of the met operon and whose function is unknown yet. At first, co-transcription and -expression with the met operon could be demonstrated. Next, the Mdh protein was overexpressed and purified and the crystal structure of Mdh was solved to high resolution by the Kisker research group (Rudolf-Virchow-Zentrum W{\"u}rzburg). Analysis of the structure revealed the amino acid residues crucial for catalytic activity, and zinc was identified as a co-factor of Mdh. Also, Mdh was shown to exist as a dimer. However, identification of the Mdh substrate was, in the context of this thesis, (still) unsuccessful. Nevertheless, interactions of Mdh with enzymes of the met operon could be demonstrated by employing the bacterial two-hybrid system. This fact and the high conservation of mdh/Mdh on nucleotide and amino acid level among numerous staphylococcal species suggests an important role of Mdh within the methionine metabolism that should be a worthwhile subject of future research.}, subject = {Staphylococcus aureus}, language = {en} } @phdthesis{Klemm2020, author = {Klemm, Theresa Antonia}, title = {Minor differences cause major effects: How differential oligomerization regulates the activities of USP25 and USP28}, doi = {10.25972/OPUS-19108}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-191080}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2020}, abstract = {Deubiquitinases are regulators of the ubiquitin proteasome system that counteract the ubiquitination cascade by removing ubiquitin from substrates and cleaving ubiquitin chains. Due to their involvment in various important pathways, they are associated with several diseases and may thus present promising drug targets. The two related ubiquitin specific proteases USP25 and USP28 share a highly conserved amino acid sequence but perform distinct biological functions. USP28 plays roles in cell cycle regulation and was also linked to several types of cancer. It adopts oncogenic functions by rescuing the oncoproteins MYC and JUN from proteasomal degradation, which is induced by the E3-ligase SCF (FBW7). Opposingly, USP28 also regulates the stability of the tumor suppressor FBW7 itself. USP25 contributes to a balanced innate immune system by stabilizing TRAF3 and TRAF6 and lately was found to promote Wnt-signaling by deubiquitinating TNKS. Due to the high level of identity of both proteases, a recent attempt to inhibit USP28 led to cross reactivity against USP25. In our study, we characterized both USP25 and USP28 structurally and functionally using x-ray crystallography, biochemical as well as biophysical approaches to determine similarities and differences that can be exploited for the development of specific inhibitors. The crystal structure of the USP28 catalytic domain revealed a cherry-couple like dimer that mediates self-association by an inserted helical subdomain, the USP25/28 catalytic domain inserted domain (UCID). In USP25, the UCID leads to formation of a tetramer composed of two interlinked USP28-like dimers. Structural and functional analysis revealed that the dimeric USP28 is active, whereas the tetrameric USP25 is auto inhibited. Disruption of the tetramer by a cancer-associated mutation or a deletion-variant activates USP25 through dimer formation in in vitro assays and leads to an increased stability of TNKS in cell studies. Furthermore, in vitro data showed that neither ubiquitin nor substrate binding led to the activation of the USP25 tetramer construct. With the structure of the C-terminal domain of USP25, we determined the last unknown region in the enzyme as a separately folded domain that mediates substrate interactions. Combined the structures of the USP25 and USP28 catalytic domains and the functional characterization of both enzymes provide novel insights into the regulation of USPs by oligomerization. Furthermore, we identified individual features of each protease that might be explored for the development of specific small molecule inhibitors.}, subject = {Oligomerisation}, language = {en} } @phdthesis{Schoenwetter2021, author = {Sch{\"o}nwetter, Elisabeth Sofie}, title = {Towards an understanding of the intricate interaction network of TFIIH}, doi = {10.25972/OPUS-16892}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-168926}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2021}, abstract = {The integrity of its DNA is fundamental for every living cell. However, DNA is constantly threatened by exogenous and endogenous damaging agents that can cause a variety of different DNA lesions. The severe consequences of an accumulation of DNA lesions are reflected in cancerogenesis and aging. Several DNA repair mechanisms ensure the repair of DNA lesions and thus maintain DNA integrity. One of these DNA repair mechanisms is nucleotide excision repair (NER), which is famous for its ability to address a large variety of structurally unrelated DNA lesions. A key component of eukaryotic NER is the transcription factor II H (TFIIH) complex, which is not only essential for DNA repair but also for transcription. The TFIIH complex is composed of ten subunits. How these subunits work together during NER to unwind the DNA around the lesion is, however, not yet fully understood. High-resolution structural data and biochemical insights into the function of every subunit are thus indispensable to understand the functional networks within TFIIH. The importance of an intact TFIIH complex is reflected in the severe consequences of patient mutations in the TFIIH subunits XPB, XPD or p8 leading to the hallmark diseases xeroderma pigmentosum, Cockayne syndrome and trichothiodystrophy. Defects in the NER pathway are further associated with several types of cancer including skin cancer. The herein described work focused on five TFIIH subunits derived from the thermophilic fungus Chaetomium thermophilum, the p34/p44 pair and the ternary XPB/p52/p8 complex. The interaction between p34 and p44 was characterized based on a high-resolution structure of the p34_vWA/p44_RING minimal complex. Biochemical studies of the p34/p44 interaction led to the disclosure of an additional interaction between the p34 and p44 subunits, which had not been characterized so far. The p34/p44 interaction was shown to be central to TFIIH, which justifies the presence of several redundant interfaces to safeguard the interaction between the two proteins and might explain why so far, no patient mutations in these subunits have been identified. The p52 subunit of TFIIH was known to be crucial to stimulate the ATPase activity of XPB, which is required during NER. This work presents the first entire atomic resolution structural characterization of p52, which was derived of several crystal structures of p52 variants and a p52/p8 variant thereby demonstrating the interaction between p52 and p8. The precise structural model of p52 offered the possibility to investigate interactions with other TFIIH subunits in more detail. The middle domain 2 of p52 and the N-terminal domain of XPB were shown to mediate the main interaction between the two subunits. An analysis of the p52 crystal structures within recently published cryo-electron microscopy structures of TFIIH provides a model of how p52 and p8 stimulate the ATPase activity of XPB, which is essential for NER and transcription. The structural and biochemical findings of this work provide an additional building block towards the uncovering of the architecture and function of this essential transcription factor.}, subject = {DNS-Reparatur}, language = {en} } @phdthesis{Bathon2019, author = {Bathon, Kerstin}, title = {Mutations in protein kinase A catalytic subunit as a cause of adrenal Cushing's syndrome: mechanisms and functional consequences}, doi = {10.25972/OPUS-16893}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-168937}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2019}, abstract = {Protein kinase A (PKA) is the main effector of cyclic-adenosine monophosphate (cAMP) and plays an important role in steroidogenesis and proliferation of adrenal cells. In a previous study we found two mutations (L206R, 199_200insW) in the main catalytic subunit of protein kinase A (PKA C) to be responsible for cortisol-producing adrenocortical adenomas (CPAs). These mutations interfere with the formation of a stable holoenzyme, thus causing constitutive PKA activation. More recently, we identified additional mutations affecting PKA C in CPAs associated with overt Cushing syndrome: S213R+insIILR, 200_201insV, W197R, d244 248+E249Q, E32V. This study reports a functional characterization of those PKA Cmutations linked to CPAs of Cushing's patients. All analyzed mutations except for E32V showed a reduced interaction with at least one tested regulatory (R) subunit. Interestingly the results of the activity differed among the mutants and between the assays employed. For three mutants (L206R, 199_200insW, S213R+insIILR), the results showed enhanced translocation to the nucleus. This was also observed in CRISPR/Cas9 generated PRKACA L206R mutated HEK293T cells. The enhanced nuclear translocation of this mutants could be due to the lack of R subunit binding, but also other mechanisms could be at play. Additionally, I used an algorithm, which predicted an effect of the mutation on substrate specificity for four mutants (L206R, 199_200insW, 200_201insV, d244 248+E249Q). This was proven using phosphoproteomics for three mutants (L206R, 200_201insV, d244 248+E249Q). In PRKACA L206R mutated CPAs this change in substrate specificity also caused hyperphosphorylation of H1.4 on serine 36, which has been reported to be implicated in mitosis. Due to these observations, I hypothesized, that there are several mechanisms of action of PRKACA mutations leading to increased cortisol secretion and cell proliferation in adrenal cells: interference with the formation of a stable holoenzyme, altered subcellular localization and a change in substrate specificity. My data indicate that some PKA C mutants might act via just one, others by a combination of these mechanisms. Altogether, these findings indicate that several mechanisms contribute to the development of CPAs caused by PRKACA mutations. Moreover, these findings provide a highly illustrative example of how alterations in a protein kinase can cause a human disease.}, subject = {Proteinkinase A}, language = {en} } @phdthesis{Schmitt2017, author = {Schmitt, Dominik}, title = {Structural Characterization of the TFIIH Subunits p34 and p44 from C. thermophilum}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-104851}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2017}, abstract = {Several important cellular processes, including transcription, nucleotide excision repair and cell cycle control are mediated by the multifaceted interplay of subunits within the general transcription factor II H (TFIIH). A better understanding of the molecular structure of TFIIH is the key to unravel the mechanism of action of this versatile protein complex within these pathways. This becomes especially important in the context of severe diseases like xeroderma pigmentosum, Cockayne syndrome and trichothiodystrophy, that arise from single point mutations in some of the TFIIH subunits. In an attempt to structurally characterize the TFIIH complex, we harnessed the qualities of the eukaryotic thermophile Chaetomium thermophilum, a remarkable fungus, which has only recently been recognized as a novel model organism. Homologues of TFIIH from C. thermophilum were expressed in E. coli, purified to homogeneity and subsequently utilized for crystallization trials and biochemical studies. The results of the present work include the first crystal structure of the p34 subunit of TFIIH, comprising the N-terminal domain of the protein. The structure revealed a von Willebrand Factor A (vWA) like fold, which is generally known to be involved in a multitude of protein-protein interactions. Structural comparison allowed to delineate similarities as well as differences to already known vWA domains, providing insight into the role of p34 within TFIIH. These results indicate that p34 assumes the role of a structural scaffold for other TFIIH subunits via its vWA domain, while likely serving additional functions, which are mediated through its C-terminal zinc binding domain and are so far unknown. Within TFIIH p34 interacts strongly with the p44 subunit, a positive regulator of the XPD helicase, which is required for regulation of RNA Polymerase II mediated transcription and essential for eukaryotic nucleotide excision repair. Based on the p34 vWA structure putative protein-protein interfaces were analyzed and binding sites for the p34 p44 interaction suggested. Continuous crystallization efforts then led to the first structure of a p34 p44 minimal complex, comprising the N-terminal vWA domain of p34 and the C-terminal C4C4 RING domain of p44. The structure of the p34 p44 minimal complex verified the previous hypothesis regarding the involved binding sites. In addition, careful analysis of the complex interface allowed to identify critical residues, which were subsequently mutated and analyzed with respect to their significance in mediating the p34 p44 interaction, by analytical size exclusion chromatography, electrophoretic mobility shift assays and isothermal titration calorimetry. The structure of the p34 p44 complex also revealed a binding mode of the p44 C4C4 RING domain, which differed from that of other known RING domains in several aspects, supporting the hypothesis that p44 contains a novel variation of this domain.}, subject = {DNA-Reparatur}, language = {en} } @phdthesis{Buechner2014, author = {B{\"u}chner, Claudia Nadine}, title = {Single molecule studies of DNA lesion search and recognition strategies}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-111886}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2014}, abstract = {The integrity of our genome is continuously endangered by DNA damaging factors. Several cellular mechanisms have evolved to recognize and remove different types of DNA lesions. Despite the wealth of information on the three-dimensional structure and the catalytic mechanism of DNA repair enzymes, the essential process of target site search and identification remains more elusive. How can a small number of repair proteins find and detect the rare sites of damage rapidly and efficiently over an excess of millions of undamaged bases? To address this pivotal question in DNA repair, I focused on the central players from the two DNA damage excision repair pathways in my studies: nucleotide excision repair (NER) and base excision repair (BER). As examples for completely different approaches of damage search, recognition and verification, I compared the NER protein Xeroderma pigmentosum group D (XPD) with the BER proteins human thymine DNA glycosylase (hTDG) and human 8-oxoguanine glycosylase (hOgg1). In particular, the single molecule approach of atomic force microscopy (AFM) imaging and complementary biochemical and biophysical techniques were applied. I established a simple, optimized preparation approach, which yields homogeneous and pure samples of long (several hundreds to thousands of base pairs) DNA substrates suitable for the AFM studies with DNA repair proteins. Via this sample preparation, a single target site of interest can be introduced into DNA at a known position, which allows separate analysis of specific protein-DNA complexes bound to the lesion site and nonspecific complexes bound to non-damaged DNA. The first part of the thesis investigates the XPD protein involved in eukaryotic NER. In general, the NER mechanism removes helix-distorting lesions - carcinogenic UV light induced photoproducts, such as cyclobutane pyrimidine dimers (CPDs) as well as bulky DNA adducts. The 5'-3' helicase XPD has been proposed to be one of the key players in DNA damage verification in eukaryotic NER, which is still a matter of hot debate. In the studies, I focused on XPD from the archaeal species Thermoplasma acidophilum (taXPD), which shares a relatively high sequence homology with the sequence of the human protein and may serve as a good model for its eukaryotic counterpart. Based on AFM experiments and accompanying DNA binding affinity measurements with the biosensor technology Biolayer Interferometry (BLI), a clear role of XPD in damage verification was deciphered. Specifically, the data suggested that the ATP-dependent 5'-3' helicase activity of XPD was blocked by the presence of damage leading to stalled XPD-DNA damage verification complexes at the lesion sites. Successful damage verification led to ATP-dependent conformational changes visible by a significant transition in DNA bend angles from ~ 50° to ~ 65° at the site of the bound protein. Remarkably, this DNA bend angle shift was observed both in the presence of ATP and ATPγs (non-hydrolyzable ATP analog) indicating that ATP-binding instead of ATP hydrolysis was sufficient to induce repair competent conformational changes of XPD. Most importantly, detailed protein binding position and DNA bend angle analyses revealed for the first time that XPD preferably recognizes a bulky fluorescein lesion on the translocated strand, whereas a CPD lesion is preferentially detected on the opposite, non-translocated strand. Despite the different recognition strategies for both types of damages, they share a common verification complex conformation, which may serve as a signal for the recruitment of further NER factors. In the second part of the thesis, AFM imaging and a 2-Aminopurine fluorescence-based base-flipping assay were combined to investigate damage search and recognition by DNA glycosylases in BER. Exemplarily, I chose to study hTDG as a representative of the vast glycosylase family. hTDG excises thymine and uracil from mutagenic G:T and G:U mispairs contributing to cancer and genetic disease. The AFM data suggested that hTDG uses the intrinsic flexibility of G:T and G:U wobble pairs for initial damage sensing, while scanning DNA as a search complex (SC, slightly bent DNA). Remarkably, hTDG has been indicated to continuously switch between the search and interrogation conformation (IC, stronger bent DNA) during damage search. In the IC, target bases are interrogated by extrahelical base flipping, which is facilitated by protein-induced DNA bending and enhanced DNA flexibility at mismatches. AFM and fluorescence analyses revealed that the flipped base is stabilized via hTDG's arginine finger. Correct target bases are perfectly stabilized within the enzyme's catalytic pocket resulting in prolonged residence time and enhanced excision probability. To test for the generalizability of the proposed hTDG damage search model to BER glycosylases, identical studies were performed with a second glycosylase, hOgg1. The data on hOgg1, which removes structurally more stable 8-oxoguanine lesions, supported the hypothesis developed for lesion recognition by hTDG as a common strategy employed by BER glycosylases}, subject = {Rasterionenmikroskop}, language = {en} } @phdthesis{Selle2018, author = {Selle, Martina}, title = {Interaktionen zwischen sekretierten Proteinen von Staphylococcus aureus und der Immunantwort des Wirtes}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-128031}, school = {Universit{\"a}t W{\"u}rzburg}, pages = {XVII, 216}, year = {2018}, abstract = {Staphylococcus aureus ist ein grampositives Bakterium, welches h{\"a}ufig als kommensaler Besiedler auf der Nasen- und Rachenschleimhaut von S{\"a}ugetieren vorkommt. Dar{\"u}ber hinaus besitzt dieser fakultativ pathogene Mikroorganismus die F{\"a}higkeit schwer zu behandelnde Krankenhausinfektionen auszul{\"o}sen. Aufgrund der weiten Verbreitung von Antibiotikaresistenzen und dem Mangel an effektiven Therapien, verursachen S. aureus Infektionen j{\"a}hrlich enorme Kosten f{\"u}r das Gesundheitssystem. S. aureus wird meist von der Nase zum prim{\"a}ren Infektionsort {\"u}bertragen, wodurch zun{\"a}chst sehr h{\"a}ufig Wund- und Weichteilinfektionen hervor gerufen werden. Von diesem prim{\"a}ren Infektionsort ausgehend, kann der Erreger tiefer liegende Gewebsschichten infizieren oder sich {\"u}ber den Blutstrom im gesamten Organismus ausbreiten. Das Spektrum an Krankheitsbildern reicht von leichten Abszessen der Haut bis zu schweren, lebensbedrohlichen Erkrankungen wie Pneumonien und akuter Sepsis. F{\"u}r die erfolgreiche Kolonisierung und Infektion des Wirtes exprimiert S. aureus eine Vielzahl unterschiedlicher Virulenzfaktoren. Die wohl gr{\"o}ßte Gruppe an Virulenzfaktoren umfasst die Proteine, die an der Immunevasion und der Umgehung von verschiedenen Abwehrstrategien des Immunsystems beteiligt sind. Das bisherige Wissen {\"u}ber die Interaktion von S. aureus mit dem Immunsystem des Wirtes und die zugrunde liegenden Pathogenit{\"a}tsmechanismen ist bisher limitiert. Um neue Erkenntnisse {\"u}ber die Interaktion von Wirt und Pathogen zu erlangen, wurden im Rahmen dieser Arbeit bislang unbekannte sekretierte und Oberfl{\"a}chen-assoziierte Proteine von S. aureus funktionell charakterisiert. Die Funktion der ausgew{\"a}hlten Proteine wurde in vitro hinsichtlich Einfluss auf Komponenten des Immunsystems, Adh{\"a}sion an Wirtsfaktoren und Invasion in eukaryotische Zellen untersucht. Mit Hilfe der vorangegangenen in-vitro-Charakterisierung der putativen Virulenzfaktoren, konnte f{\"u}r die cytoplasmatische Adenylosuccinat-Synthase PurA eine neuartige Funktion identifiziert werden. PurA ist bekannt als essentielles Enzym der de novo Purin-Synthese. In dieser Arbeit wurde nun gezeigt, dass PurA zudem an der Immunevasion beteiligt ist. Durch die Bindung des humanen Faktor H des Komplementsystems sch{\"u}tzt PurA S. aureus vor der lytischen Aktivit{\"a}t des Komplementsystems und verhindert die Opsonisierung des Pathogens. Basierend auf diesen Ergebnissen wurde PurA detailliert charakterisiert. In Bindungsstudien mit rekombinantem Faktor H und PurA wurde eine direkte Interaktion beider Proteine nachgewiesen, wobei Faktor H mit dem N-terminalen Bereich von PurA interagiert. Weiterhin konnte PurA durch Immunfluoreszenz und FACS-Analysen auf der Zelloberfl{\"a}che nachgewiesen werden, wo es wahrscheinlich mit der Zellwand assoziiert vorliegt. Dort rekrutiert es Faktor H an die bakterielle Oberfl{\"a}che und verhindert das Fortschreiten der Komplement-Kaskade und damit die Lyse des Pathogens. Aufgrund der Multifunktionalit{\"a}t z{\"a}hlt PurA somit zur Gruppe der Moonlighting Proteine. Des Weiteren wurde die Rolle von PurA im Infektionsgeschehen in zwei unabh{\"a}ngigen Tiermodellen untersucht. In beiden Modellen wurde ein signifikant reduziertes Virulenzpotential der ΔpurA-Mutante beobachtet. Zuk{\"u}nftig soll gekl{\"a}rt werden, ob die verminderte Virulenz in der fehlenden Komplementevasion oder im Defekt in der Purin-Synthese begr{\"u}ndet ist. Aufgrund der sehr starken Attenuation in allen untersuchten Infektionsmodellen sollte PurA als potentielles Target f{\"u}r eine Therapie von S. aureus Infektionen weiter charakterisiert werden. Im Ergebnis dieser Arbeit wurde demnach mit PurA ein neues Moonlighting Protein identifiziert, das als Inhibitor des Komplementsystems wesentlich zur Immunevasion von S. aureus beitr{\"a}gt. F{\"u}r das bessere Verst{\"a}ndnis der humoralen S. aureus-spezifischen Immunantwort, Unterschieden in der Antik{\"o}rperantwort und der gebildeten Antik{\"o}rperspezifit{\"a}ten wurde weiterhin das w{\"a}hrend der Kolonisierung und Infektion gebildete S. aureus-spezifische Antik{\"o}rperprofil untersucht. Dazu wurden Plasmen von humanen nasalen Tr{\"a}gern und Nicht-Tr{\"a}gern sowie murine Seren von infizierten Tieren untersucht. Insbesondere wurde das Pathogen-spezifische Antik{\"o}rperprofil in unterschiedlichen Infektionsmodellen mit Hilfe eines Proteinarrays analysiert, der im Rahmen dieser Arbeit in einer Kooperation mit der Firma Alere Technologies (Jena, Deutschland) und universit{\"a}ren Forschergruppen der Universit{\"a}ten Greifswald, M{\"u}nster und Jena mitentwickelt wurde. Die Antik{\"o}rperprofile von intramuskul{\"a}r und intraven{\"o}s infizierten Tieren resultierten in jeweils spezifischen Antik{\"o}rperprofilen. Diese Ergebnisse deuten auf einen Zusammenhang zwischen der Art der Infektion und der gebildeten Antik{\"o}rperspezifit{\"a}ten hin. Wahrscheinlich beruht dies auf einer gewebespezifischen Genexpression als Anpassung an die individuellen Bed{\"u}rfnisse im Wirtsorganismus. Das ausgebildete Antik{\"o}rperprofil gibt somit einen Einblick in das Expressionsmuster von Virulenzfaktoren von S. aureus unter in vivo Bedingungen und tr{\"a}gt damit zum Verst{\"a}ndnis der komplexen Interaktion von Pathogen und Wirt bei. Diese Untersuchungen erg{\"a}nzen zudem die bisherigen Kenntnisse {\"u}ber die Anpassung der humoralen Immunantwort an eine asymptomatische Kolonisierung im Gegensatz zu einer akuten Infektion durch S. aureus. Dar{\"u}ber hinaus k{\"o}nnen die gewonnenen Ergebnisse f{\"u}r diagnostische Zwecke und zur Identifikation von neuen Zielstrukturen f{\"u}r eine Vakzin-Entwicklung genutzt werden.}, subject = {Staphylococcus aureus}, language = {de} } @phdthesis{Rohleder2014, author = {Rohleder, Florian}, title = {The Intricate Network of Replication-dependent Interstrand Crosslink DNA Repair}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-113121}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2014}, abstract = {The Fanconi anemia (FA) pathway is a replication-dependent DNA repair mechanism which is essential for the removal of interstrand crosslink (ICL) DNA damages in higher eukaryotes (Moldovan and D'Andrea, 2009). Malfunctions in this highly regulated repair network lead to genome instability (Deans and West, 2011). Pathological phenotypes of the disease FA which is caused by mutations in the eponymous pathway are very heterogeneous, involving congenital abnormalities, bone-marrow failure, cancer predisposition and infertility (Auerbach, 2009). The FA pathway comprises a complex interaction network and to date 16 FA complementation groups and associated factors have been identified (Kottemann and Smogorzewska, 2013). Additionally, components of nucleotide excision repair (NER), homologous recombination repair (HRR), and translesion synthesis (TLS) are involved and coordinated by the FA proteins (Niedzwiedz et al., 2004; Knipscheer et al., 2009). One of the FA proteins is the DEAH helicase FANCM. In complex with its binding partners FAAP24 and MHF1/2 it binds the stalled replication fork and activates the FA damage response (Wang et al., 2013). However, the exact steps towards removal of the ICL damage still remain elusive. To decipher the underlying process of FA initiation by FANCM, this thesis mainly focuses on the archaeal FANCM homolog helicase-associated endonuclease for fork-structured DNA (Hef). Hef from the archaeal organism Thermoplasma acidophilum (taHef) differs from other archaeal Hef proteins and exclusively comprises an N-terminal helicase entity with two RecA and a thumb-like domain while others additionally contain a nuclease portion at the C-terminus. I solved the crystal structure of full-length taHef at a resolution of 2.43 {\AA}. In contrast to the crystal structure of the helicase domain of Hef from Pyrococcus furiosus (pfHef), taHef exhibits an extremely open conformation (Nishino et al., 2005b) which implies that a domain movement of the RecA-like helicase motor domains of 61° is possible thus highlighting the flexibility of helicases which is required to translocate along the DNA. However, small-angle x-ray scattering (SAXS) measurements confirm an intermediate conformation of taHef in solution indicating that both crystal structures represent rather edge states. Most importantly, proliferating cell nuclear antigen (PCNA) was identified as an interaction partner of Hef. This interaction is mediated by a highly conserved canonical PCNA interacting peptide (PIP) motif. Intriguingly, the presence of PCNA does not alter the ATPase nor the helicase activity of taHef, thus suggesting that the interaction is entirely dedicated to recruit taHef to the replication fork to fulfill its function. Due to a high level of flexibility the taHef-taPCNA complex could not be crystallized and therefore SAXS was utilized to determine a low-resolution model of this quaternary structure. This newly discovered PCNA interaction could also be validated for the eukaryotic FANCM homolog Mph1 from the thermophilic fungus Chaetomium thermophilum (ctMph1). As the first step towards the characterization of this interaction I solved the crystal structure of PCNA from Chaetomium thermophilum (ctPCNA). Furthermore, it was possible to achieve preliminary results on the putative interaction between the human proteins FANCM and PCNA (hsFANCM, hsPCNA). In collaboration with Detlev Schindler (Human Genetics, W{\"u}rzburg) and Weidong Wang (National Institute on Aging, Baltimore, USA) co-immunoprecipitation (CoIP) experiments were performed using hsFANCM and hsPCNA expressed in HEK293 cells. Although an interaction was reproducibly observed in hydroxyurea stimulated cells further experiments and optimization procedures are required and ongoing.}, subject = {DNS-Reparatur}, language = {en} } @phdthesis{Schubert2015, author = {Schubert, Andreas}, title = {Protein kinases as targets for the development of novel drugs against alveolar echinococcosis}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-113694}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2015}, abstract = {The metacestode larval stage of the fox tapeworm Echinococcus multilocularis is the causative agent of alveolar echinococcosis (AE), one of the most lethal zoonosis of the northern hemisphere. The development of metacestode vesicles by asexual multiplication and the almost unrestricted infiltrative growth within the host organs is ensured from a population of undifferentiated, proliferative cells, so-called germinative cells. AE treatment options include surgery, if possible, as well as Benzimidazole-based chemotherapy (BZ). Given that the cellular targets of BZs, the -tubulins, are highly conserved between cestodes and humans, the chemotherapy is associated with considerable side-effects. Therefore, BZ can only be applied in parasitostatic doses and has to be given lifelong. Furthermore, the current anti-AE chemotherapy is ineffective in eliminating the germinative cell population of the parasite, which leads to remission of parasite growth as soon as therapy is discontinued. This work focuses on protein kinases involved in the proliferation and development of the parasite with the intention of developing novel anti-AE therapies. Polo-like kinases (Plks) are important regulators of the eukaryotic cell cycle and are involved in the regulation and formation of the mitotic spindles during the M-phase of the cell cycle. Plks have already been shown to be associated with deregulated cellular growth in human cancers and have been investigated as novel drug targets in the flatworm parasite Schistosoma mansoni. In the first part of this work, the characterisation of a novel and druggable parasite enzyme, EmPlk1, which is homologous to the polo-like kinase 1 (Plk1) of humans and S. mansoni (SmPlk1), is presented. Through in situ hybridisation, it could be demonstrated that emplk1 is specifically expressed in the Echinococcus germinative cells. Upon heterologous expression in the Xenopus oocyte system, EmPlk1 induced germinal vesicle breakdown, thus indicating that it is an active kinase. Furthermore, BI 2536, a compound originally designed to inhibit the human ortholog of EmPlk1, inhibited the EmPlk1 activity at a concentration of 25 nM. In vitro treatment of parasite vesicles with similar concentrations of BI 2536 led to the elimination of the germinative cells from Echinococcus larvae, thus preventing the growth and further development of the parasite. In in vitro cultivation systems for parasite primary cells, BI 2536 effectively inhibited the formation of new metacestode vesicles from germinative cells. Thus, BI 2536 has profound anti-parasitic activities in vitro at concentrations well within the range of plasma levels measured after the administration of safe dosages to patients (50 nM after 24 h). This implies that EmPlk1 is a promising new drug target for the development of novel anti-AE drugs that would specifically affect the parasite's stem cell population, namely the only parasite cells capable of proliferation. In addition to the chemotherapeutic aspects of this work, the inhibitor BI 2536 could be further used to study the function of stem cells in this model organism, utilising a method of injection of parasite stem cells into metacestode vesicles, for instance, as has been developed in this work. In the second part of this work, a novel receptor tyrosine kinase, the Venus flytrap kinase receptor (EmVKR) of E. multilocularis has been characterised. Members of this class of single-pass transmembrane receptors have recently been discovered in the related trematode S. mansoni and are associated with the growth and differentiation of sporocyst germinal cells and ovocytes. The ortholog receptor in EmVKR is characterised by an unusual domain composition of an extracellular Venus flytrap module (VFT), which shows significant similarity to GABA receptors, such as the GABAB receptor (γ-amino butyric acid type B) and is linked through a single transmembrane domain to an intracellular tyrosine kinase domain with similarities to the kinase domains of human insulin receptors. Based upon the size (5112bp) of emvkr and nucleotide sequence specificities, efforts have been made to isolate the gene from cell culture samples to study the ligand for the activation of this receptor type in Xenopus oocytes. To date, this type of receptor has only been described in invertebrates, thus making it an attractive target for drug screening. In a first trial, the ATP competitive inhibitor AG 1024 was tested in our in vitro cell culture. In conclusion, the EmVKR represents a novel receptor tyrosine kinase in E. multilocularis. Further efforts have to be made to identify the activating ligand of the receptor and its cellular function, which might strengthen the case for EmVKR as a potential drug target. The successful depletion of stem cells in the metacestode vesicle by the Plk1 inhibitor BI 2536 gives rise to optimising the chemical component for EmPlk1 as a new potential drug target. Furthermore, this inhibitor opens a new cell culture technique with high potential to study the cellular behaviour and influencing factors of stem cells in vitro.}, subject = {Chemotherapie}, language = {en} } @phdthesis{Schaefer2014, author = {Sch{\"a}fer, Christin Marliese}, title = {Approaching antimicrobial resistance - Structural and functional characterization of the fungal transcription factor Mrr1 from Candida albicans and the bacterial ß-ketoacyl-CoA thiolase FadA5 from Mycobacterium tuberculosis}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-108400}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2014}, abstract = {The number of fungal infections is rising in Germany and worldwide. These infections are mainly caused by the opportunistic fungal pathogen C. albicans, which especially harms immunocompromised people. With increasing numbers of fungal infections, more frequent and longer lasting treatments are necessary and lead to an increase of drug resistances, for example against the clinically applied therapeutic fluconazole. Drug resistance in C. albicans can be mediated by the Multidrug resistance pump 1 (Mdr1), a membrane transporter belonging to the major facilitator family. However, Mdr1-mediated fluconazole drug resistance is caused by the pump's regulator, the transcription factor Mrr1 (Multidrug resistance regulator 1). It was shown that Mrr1 is hyperactive without stimulation or further activation in resistant strains which is due to so called gain of function mutations in the MRR1 gene. To understand the mechanism that lays behind this constitutive activity of Mrr1, the transcription factor should be structurally and functionally (in vitro) characterized which could provide a basis for successful drug development to target Mdr1-mediated drug resistance caused by Mrr1. Therefore, the entire 1108 amino acid protein was successfully expressed in Escherichia coli. However, further purification was compromised as the protein tended to form aggregates, unsuitable for crystallization trials or further characterization experiments. Expression trials in the eukaryote Pichia pastoris neither yielded full length nor truncated Mrr1 protein. In order to overcome the aggregation problem, a shortened variant, missing the N-terminal 249 amino acids named Mrr1 '250', was successfully expressed in E. coli and could be purified without aggregation. Similar to the wild type Mrr1 '250', selected gain of function variants were successfully cloned, expressed and purified with varying yields and with varying purity. The Mrr1 `250' construct contains most of the described regulatory domains of Mrr1. It was used for crystallization and an initial comparative analysis between the wild type protein and the variants. The proposed dimeric form of the transcription factor, necessary for DNA binding, could be verified for both, the wild type and the mutant proteins. Secondary structure analysis by circular dichroism measurements revealed no significant differences in the overall fold of the wild type and variant proteins. In vitro, the gain of function variants seem to be less stable compared to the wild type protein, as they were more prone to degradation. Whether this observation holds true for the full length protein's stability in vitro and in vivo remains to be determined. The crystallization experiments, performed with the Mrr1 '250' constructs, led to few small needle shaped or cubic crystals, which did not diffract very well and were hardly reproducible. Therefore no structural information of the transcription factor could be gained so far. Infections with M. tuberculosis, the causative agent of tuberculosis, are the leading cause of mortality among bacterial diseases. Especially long treatment times, an increasing number of resistant strains and the prevalence of for decades persisting bacteria create the necessity for new drugs against this disease. The cholesterol import and metabolism pathways were discovered as promising new targets and interestingly they seem to play an important role for the chronic stage of the tuberculosis infection and for persisting bacteria. In this thesis, the 3-ketoacyl-CoA thiolase FadA5 from M. tuberculosis was characterized and the potential for specifically targeting this enzyme was investigated. FadA5 catalyzes the last step of the β-oxidation reaction in the side-chain degradation pathway of cholesterol. We solved the three dimensional structure of this enzyme by X-ray crystallography and obtained two different apo structures and three structures in complex with acetyl-CoA, CoA and a hydrolyzed steroid-CoA, which is the natural product of FadA5. Analysis of the FadA5 apo structures revealed a typical thiolase fold as it is common for biosynthetic and degradative enzymes of this class for one of the structures. The second apo structure showed deviations from the typical thiolase fold. All obtained structures show the enzyme as a dimer, which is consistent with the observed dimer formation in solution. Thus the dimer is likely to be the catalytically active form of the enzyme. Besides the characteristic structural fold, the catalytic triad, comprising two cysteines and one histidine, as well as the typical coenzyme A binding site of enzymes belonging to the thiolase class could be identified. The two obtained apo structures differed significantly from each other. One apo structure is in agreement with the characteristic thiolase fold and the well-known dimer interface could be identified in our structure. The same characteristics were observed in all complex structures. In contrast, the second apo structure followed the thiolase fold only partially. One subdomain, spanning 30 amino acids, was in a different orientation. This reorientation was caused by the formation of two disulfide bonds, including the active site cysteines, which rendered the enzyme inactive. The disulfide bonds together with the resulting domain swap still permitted dimer formation, yet with a significantly shifted dimer interface. The comparison of the apo structures together with the preliminary activity analysis performed by our collaborator suggest, that FadA5 can be inactivated by oxidation and reactivated by reduction. If this redox switch is of biological importance requires further evaluation, however, this would be the first reported example of a bacterial thiolase employing redox regulation. Our obtained complex structures represent different stages of the thiolase reaction cycle. In some complex structures, FadA5 was found to be acetylated at the catalytic cysteine and it was in complex with acetyl-CoA or CoA. These structures, together with the FadA5 structure in complex with a hydrolyzed steroid-CoA, revealed important insights into enzyme dynamics upon ligand binding and release. The steroid-bound structure is as yet a unique example of a thiolase enzyme interacting with a complex ligand. The characterized enzyme was used as platform for modeling studies and for comparison with human thiolases. These studies permitted initial conclusions regarding the specific targetability of FadA5 as a drug target against M. tuberculosis infection, taking the closely related human enzymes into account. Additional analyses led to the proposal of a specific lead compound based on the steroid and ligand interactions within the active site of FadA5.}, subject = {Multidrug-Resistenz}, language = {en} } @phdthesis{Mishra2013, author = {Mishra, Shambhavi}, title = {Structural and Functional Characterization of the Enzymes Involved in the Menaquinone Biosynthesis and Benzoate Degradation}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-90848}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2013}, abstract = {The present work illustrates the structural and biochemical characterization of two diverse proteins, BadI and MenD from Rhodopseudomonas palustris and Staphylococcus aureus, respectively. BadI or 2-ketocyclohexanecarboxyl-CoA is one of the key enzymes involved in the anaerobic degradation of aromatic compounds. The degradation of aromatic compounds is a vital process for the maintenance of the biogeochemical carbon cycle and bioremediation of xenobiotic compounds, which if present at higher concentrations can cause potential hazards to humans. Due to the relatively inert nature of aromatic compounds, enzymes catalyzing their degradation are of special interest for industrial applications. BadI is one of the key enzymes involved in the anaerobic degradation of aromatic compounds into an aliphatic moiety. The major focus of this study was to provide mechanistic insights into the reaction catalyzed by BadI. BadI belongs to the crotonase superfamily and shares high sequence homology with the family members of MenB or dihydroxynaphthoate synthase. BadI is known to catalyze the cleavage of the cyclic ring of 2-ketocyclohexane carboxyl-CoA by hydrolyzing the C-C bond leading to the formation of the aliphatic compound pimelyl CoA. On the other hand MenB catalyzes the condensation reaction of o-succinylbenzoyl-CoA to dihydroxylnaphthoyl-CoA. A comprehensive amino acid sequence analysis between BadI and MenB showed that the active site residues of MenB from Mycobacterium tuberculosis (mtMenB) are conserved in BadI from Rhodopseudomonas palustris. MenB is involved in the menaquinone biosynthesis pathway and is a potential drug target against Mycobacterium tuberculosis as it has no known human homologs. Due to the high homology between MenB and BadI and the inability to obtain MenB-inhibitor complex structures we extended our interest to BadI to explore a potential substitute model for mtMenB as a drug target. In addition, BadI possesses some unique mechanistic characteristics. As mentioned before, it hydrolyzes the substrate via a retro Dieckmann's reaction contrasting its closest homolog MenB that catalyzes a ring closing reaction through a Dieckmann's reaction. Nevertheless the active site residues in both enzymes seem to be highly conserved. We therefore decided to pursue the structural characterization of BadI to shed light on the similarities and differences between BadI and MenB and thereby provide some insights how they accomplish the contrasting reactions described above. We determined the first structures of BadI, in its apo and a substrate mimic bound form. The crystal structures revealed that the overall fold of BadI is similar to other crotonase superfamily members. However, there is no indication of domain swapping in BadI as observed for MenB. The absence of domain swapping is quite remarkable because the domain swapped C-terminal helical domain in MenB provides a tyrosine that is imperative for catalysis and is also conserved in the BadI sequence. Comparison of the active sites revealed that the C-terminus of BadI folds onto its core in such a way that the conserved tyrosine is located in the same position as in MenB and can form interactions with the ligand molecule. The structure of BadI also confirms the role of a serine and an aspartate in ligand interaction, thus validating that the conserved active site triad participates in the enzymatic reaction. The structures also reveal a noteworthy movement of the active site aspartate that adopts two major conformations. Structural studies further illuminated close proximity of the active site serine to a water and chlorine molecule and to the carbon atom at which the carbonyl group of the true substrate would reside. Biochemical characterization of BadI using enzyme kinetics validated that the suggested active site residues are involved in substrate interaction. However, the role of these residues is very distinct, with the serine assuming a major role. Thus, the present work ascertain the participation of putative active site residues and demonstrates that the active site residues of BadI adopt very distinctive roles compared to their closest homolog MenB. The MenD protein also referred to as SEPHCHC (2-succinyl-5-enolpyruvyl-6- hydroxy-3-cyclohexene-1-carboxylic acid) synthase is one of the enzymes involved in menaquinone biosynthesis in Staphylococcous aureus. Though S. aureus is usually considered as a commensal it can act as a remarkable pathogen when it crosses the epithelium, causing a wide spectrum of disorders ranging from skin infection to life threatening diseases. Small colony variants (SCVs), a slow growing, small sized subpopulation of the bacteria has been associated with persistent, recurrent and antibiotic resistant infections. These variants show autotrophy for thiamine, menaquinone or hemin. Menaquinone is an essential component in the electron transport pathway in gram-positive organisms. Therefore, enzymes partaking in this pathway are attractive drug targets against pathogens such as Mycobacterium tuberculosis and Bacillus subtilis. MenD, an enzyme catalyzing the first irreversible step in the menaquinone biosynthetic pathway has been implicated in the SCV phenotype of S. aureus. In the present work we explored biochemical and structural properties of this important enzyme. Our structural analysis revealed that despite its low sequence identity of 28\%, the overall fold of staphylococcal MenD (saMenD) is similar to Escherichia coli MenD (ecMenD) albeit with some significant disparities. Major structural differences can be observed near the active site region of the protein and are profound in the C-terminal helix and a loop near the active site. The loop contains critical residues for cofactor binding and is well ordered only in the ecMenD-ThDP structure, while in the apo and substrate bound structures of ecMenD the loop is primarily disordered. In our saMenD structure the loop is for the first time completely ordered in the apo form and displays a novel conformation of the cofactor-binding loop. The loop adopts an unusual open conformation and the conserved residues, which are responsible for cofactor binding are located too far away to form a productive complex with the cofactor in this conformation. Additionally, biochemical studies in conjugation with the structural data aided in the identification of the substrate-binding pocket and delineated residues contributing to its binding and catalysis. Thus the present work successfully divulged the unique biochemical and structural characteristics of saMenD.}, subject = {Benzoate}, language = {en} } @phdthesis{Narkhede2018, author = {Narkhede, Yogesh}, title = {In silico structure-based optimisation of pyrrolidine carboxamides as Mycobacterium tuberculosis enoyl-ACP reductase inhibitors}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-152468}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2018}, abstract = {The high infection rates and recent emergence of extremely drug resistant forms of Mycobacterium tuberculosis pose a significant challenge for global health. The NADH- dependent enoyl-ACP-reductase InhA of the type II mycobacterial fatty acid biosynthesis pathway is a well-validated target for inhibiting mycobacterial growth. InhA has been shown to be inhibited by a variety of compound series. Prominent classes of InhA inhibitors from literature include diaryl ethers, pyrrolidine carboxamides and arylamides which can be subjected to further development. Despite the progress in this area, very few compounds are in clinical development phase. The present work involves a detailed computational investigation of the binding modes and structure-based optimisation of pyrrolidine carboxamides as InhA inhibitors. With substituents of widely varying bulkiness, the pyrrolidine carboxamide dataset presented a challenge for prediction of binding mode as well as affinity. Using advanced docking protocols and in-house developed pose selection procedures, the binding modes of 44 compounds were predicted. The poses from docking were used in short molecular dynamics (MD) simulations to ascertain the dominant binding conformations for the bulkier members of the series. Subsequently, an activity-based classification strategy could be developed to circumvent the affinity prediction problems observed with this dataset. The prominent motions of the bound ligand and the active site residues were then ascertained using Essential Dynamics (ED). The information from ED and literature was subsequently used to design a total of 20 compounds that were subjected to extensive in-silico evaluations. Finally, the molecular determinants of rapid-reversible binding of pyrrolidine carboxamides were investigated using long MD simulations.}, subject = {Tuberkelbakterium}, language = {en} } @phdthesis{Endres2021, author = {Endres, Theresa}, title = {PAF1 complex and MYC couple transcription elongation with double-strand break repair}, doi = {10.25972/OPUS-24955}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-249557}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2021}, abstract = {The oncogene MYC is deregulated and overexpressed in a high variety of human cancers and is considered an important driver in tumorigenesis. The MYC protein binds to virtually all active promoters of genes which are also bound by the RNA Polymerase II (RNAPII). This results in the assumption that MYC is a transcription factor regulating gene expression. The effects of gene expression are weak and often differ depending on the tumor entities or MYC levels. These observations could argue that the oncogene MYC has additional functions independent of altering gene expression. In relation to this, the high diversity of interaction partners might be important. One of them is the RNAPII associated Factor I complex (PAF1c). In this study, direct interaction between PAF1c and MYC was confirmed in an in-vitro pulldown assay. ChIP sequencing analyses revealed that knockdown of PAF1c components resulted in reduced MYC occupancy at active promoters. Depletion or activation as well as overexpression of MYC led to reduced or enhanced global occupancy of PAF1c in the body of active genes, arguing that MYC and PAF1c bind cooperatively to chromatin. Upon PAF1c knockdown cell proliferation was reduced and additionally resulted in an attenuation of activation or repression of MYC-regulated genes. Interestingly, knockdown of PAF1c components caused an accumulation in S-phase of cells bearing oncogenic MYC levels. Remarkably, enhanced DNA damage, measured by elevated gH2AX and pKAP1 protein levels, was observed in those cells and this DNA damage occurs specifically during DNA synthesis. Strikingly, MYC is involved in double strand break repair in a PAF1c-dependent manner at oncogenic MYC levels. Collectively the data show that the transfer of PAF1c from MYC onto the RNAPII couples the transcriptional elongation with double strand break repair to maintain the genomic integrity in MYC-driven tumor cells.}, language = {en} } @phdthesis{Tiffe2019, author = {Tiffe, Theresa}, title = {Pr{\"a}valenz und Determinanten f{\"u}r die Einhaltung der leitliniengerechten Therapie kardiovaskul{\"a}rer Risikofaktoren in der Prim{\"a}r- und Sekund{\"a}rpr{\"a}vention von Herz-Kreislauf-Erkrankungen in Deutschland}, doi = {10.25972/OPUS-19272}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-192723}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2019}, abstract = {Die Einhaltung eines gesunden Lebensstils, einschließlich der Behandlung modifizierbarer kardiovaskul{\"a}rer Risikofaktoren, beeinflusst maßgeblich die Entstehung und Progression von Herz-Kreislauf-Erkrankungen (HKE). So reduziert eine ausgewogene Ern{\"a}hrungsweise, ausreichend k{\"o}rperliche Aktivit{\"a}t, Tabakverzicht, das Halten des Normalgewichtes sowie die Behandlung einer Hypertonie, Hyperlipid{\"a}mie und Diabetes mellitus, die kardiovaskul{\"a}re Morbidit{\"a}t und Mortalit{\"a}t. Die vorliegende Arbeit widmet sich (a) der Pr{\"a}valenz und leitliniengerechten Kontrolle kardiovaskul{\"a}rer Risikofaktoren von Teilnehmern aus der Allgemeinbev{\"o}lkerung der STAAB Kohortenstudie („H{\"a}ufigkeit und Einflussfaktoren auf fr{\"u}he Stadien A und B der Herzinsuffizienz in der Bev{\"o}lkerung") sowie der Sch{\"a}tzung des 10-Jahres Risikos f{\"u}r t{\"o}dliche HKE in diesem Kollektiv. Weiterhin wurde (b) der Einfluss von medikamentenbezogenen {\"U}berzeugungen auf die Blutdruckkontrolle von Teilnehmern der STAAB Kohortenstudie untersucht. Schließlich wurde (c) der Erhalt von {\"a}rztlichen Lebensstilempfehlungen sowie deren Determinanten bei Teilnehmern der STAAB Kohortenstudie sowie der EUROASPIRE IV Studie („European Action on Secondary and Primary Prevention by Intervention to Reduce Events") in Deutschland betrachtet. Die STAAB Kohortenstudie untersucht die fr{\"u}hen asymptomatischen Formen der Herzinsuffizienz-Stadien A und B in einer repr{\"a}sentativen Stichprobe von 5.000 Personen ohne symptomatische Herzinsuffizienz im Alter von 30 bis 79 Jahren aus der Allgemeinbev{\"o}lkerung mit Wohnsitz in der Stadt W{\"u}rzburg. Die EUROASPIRE IV Studie untersuchte bei 7.998 Koronarpatienten im Alter von 18 bis 79 Jahren aus insgesamt 24 Europ{\"a}ischen L{\"a}ndern (536 Patienten aus Deutschland) im Zeitraum 2012 bis 2013 die Risikofaktoren sowie die Umsetzung der leitliniengerechten Versorgung und Pr{\"a}vention von HKE im europ{\"a}ischen Vergleich. Die Datenerhebung beider Studien erfolgte durch ein geschultes Studienpersonal nach standardisierten Vorgaben. Die Pr{\"a}valenz und Kontrolle kardiovaskul{\"a}rer Risikofaktoren nach den aktuellen Vorgaben der „European Society of Cardiology" (ESC) wurde bei insgesamt 1.379 Teilnehmern, die zwischen Dezember 2013 und April 2015 an der STAAB Kohortenstudie teilgenommen haben, untersucht. Es zeigte sich eine hohe Pr{\"a}valenz der kardiovaskul{\"a}ren Risikofaktoren Hypertonie (31.8\%), Hyperlipid{\"a}mie (57.6\%) und Diabetes mellitus (3.5\%). Hierbei erreichten trotz Pharmakotherapie {\"u}ber die H{\"a}lfte der Teilnehmer mit einem Bluthochdruck (52.7\%) oder erh{\"o}hten LDL-Cholesterinwerten (56.7\%) sowie 44.0\% der Personen mit einem Diabetes mellitus die empfohlenen Grenzwerte nicht. Weiterhin wurde erstmalig zu Studienbesuch eine Hypertonie (36.0\%), Hyperlipid{\"a}mie (54.2\%) oder ein Langzeitzuckerwert (HbA1c) >6.5\% (23.3\%) detektiert. In der j{\"u}ngsten Altersgruppe (30-39 Jahre) fand sich der h{\"o}chste Anteil von unbekanntem Bluthochdruck (76.5\%) sowie hohem LDL-Cholesterin (78.0\%) und die Altersgruppe 60-69 Jahren wies mit 43.5\% die h{\"o}chste Pr{\"a}valenz f{\"u}r einen bislang nicht detektierten HbA1c >6.5\% auf. Die Akkumulation von drei oder mehr kardiovaskul{\"a}ren Risikofaktoren war mit dem m{\"a}nnlichen Geschlecht, einem h{\"o}heren Alter und einem niedrigeren Bildungsgrad assoziiert. Von 980 mittels SCORE („Systematic Coronary Risk Evaluation") Risiko-Chart untersuchten Teilnehmern befanden sich jeweils 56.6\%, 35.8\% und 7.5\% in der niedrigen, mittleren und hohen bis sehr hohen SCORE-Risikogruppe f{\"u}r t{\"o}dliche HKE. Das Hochrisiko-Kollektiv f{\"u}r t{\"o}dliche HKE war vorwiegend m{\"a}nnlich und wies h{\"a}ufiger eine Hypertonie oder ein hohes LDL-Cholesterin auf. Der Einfluss von {\"U}berzeugungen gegen{\"u}ber antihypertensiver Medikation auf die Blutdruckkontrolle wurde an 293 Teilnehmern, die von Oktober 2014 bis M{\"a}rz 2017 an der STAAB Kohortenstudie teilgenommen haben, untersucht. Auf ihre Medikamente gesundheitlich angewiesen zu sein gaben 87\% der Teilnehmer an, 78.1\% stimmten der Aussage zu, dass ihre Medikamente sie vor einer Verschlechterung ihrer Gesundheit sch{\"u}tzen. Es zeigte sich ein inverser Zusammenhang zwischen einem h{\"o}heren Maß an Bedenken gegen{\"u}ber der verordneten blutdrucksenkenden Medikation und einer besseren Blutdruckkontrolle bei Frauen. Ein signifikanter Zusammenhang zwischen Bedenken gegen{\"u}ber einer antihypertensiven Medikation und der Blutdruckkontrolle bei M{\"a}nnern ließ sich hingegen nicht feststellen. Es konnten keine statistisch signifikanten Assoziationen f{\"u}r die Notwendigkeit von Medikation in der vorliegen Untersuchung gezeigt werden. Die H{\"a}ufigkeit und Determinanten f{\"u}r die Empfehlung eines {\"a}rztlichen Lebensstils wurde bei 665 Teilnehmern der STAAB Kohortenstudie ohne vorbestehende HKE (Prim{\"a}rpr{\"a}vention) und bei 536 Koronarpatienten der EUROASPIRE IV Studie (Sekund{\"a}rpr{\"a}vention) untersucht. Mit Ausnahme der Empfehlung zum Rauchverzicht erhielten die Patienten der EUROASPIRE IV Studie h{\"a}ufiger {\"a}rztliche Lebensstilempfehlungen verglichen mit Teilnehmern der STAAB Kohortenstudie: (Rauchverzicht: STAAB 44.0\%, EUROASPIRE 36.7\%; Gewichtsreduktion: STAAB 43.9\%, EUROASPIRE 69.2\%; k{\"o}rperliche Aktivit{\"a}t steigern: STAAB 52.1\%, EUROASPIRE 71.4\%; gesundes Ern{\"a}hrungsverhalten: STAAB 43.9\%, EUROASPIRE 73.1\%). Die Chance f{\"u}r den Erhalt von mindestens 50\% aufgrund der individuellen Risikofaktoren ad{\"a}quaten {\"a}rztlichen Lebensstilempfehlungen war bei STAAB Teilnehmern mit offensichtlichen oder beobachtbaren kardiovaskul{\"a}ren Risikofaktoren signifikant erh{\"o}ht (BMI >25kg/m2, Hypertonie, Hyperlipid{\"a}mie und Diabetes mellitus). Hingegen erhielten Patienten mit einer vorbestehenden HKE signifikant h{\"a}ufiger eine {\"a}rztliche Lebensstilempfehlung bei einem Diabetes mellitus, wobei die Empfehlungsh{\"a}ufigkeit mit zunehmendem Alter abnahm. Die weitergehende nicht publizierte Analyse des Interaktions Modells zeigte, dass der Zusammenhang zwischen dem Alter und der Empfehlungsh{\"a}ufigkeit bei Patienten mit bereits bestehender HKE st{\"a}rker ausgepr{\"a}gt war, als bei Teilnehmern der STAAB Kohortenstudie ohne koronare HKE. Weiterhin war der Zusammenhang zwischen einer ad{\"a}quaten Lebensstilempfehlung und Hyperlipid{\"a}mie bei Teilnehmern ohne koronares Ereignis signifikant st{\"a}rker ausgepr{\"a}gt, im Vergleich zu Patienten mit einer bereits bestehender HKE. Die Ergebnisse zeigten ein erhebliches Potenzial f{\"u}r eine verbesserte Umsetzung leitliniengerechter Behandlung modifizierbarer kardiovaskul{\"a}rer Risikofaktoren in der Prim{\"a}r- und Sekund{\"a}rpr{\"a}vention. Vor dem Hintergrund einer hohen Anzahl kardiovaskul{\"a}rer Risikofaktoren bei jungen Erwachsenen sollte die Bedeutung der Langzeitfolgen im Arzt Patienten-Gespr{\"a}ch hervorgehoben und bei der Erarbeitung von Pr{\"a}ventionsstrategien, insbesondere f{\"u}r junge Altersgruppen, Beachtung finden. Geschlechtsspezifische Determinanten hinsichtlich der Kontrolle kardiovaskul{\"a}rer Risikofaktoren sowie Bef{\"u}rchtungen gegen{\"u}ber der Medikation sollten st{\"a}rker im Arzt-Patientengespr{\"a}ch ber{\"u}cksichtigt werden. Zur St{\"a}rkung der Compliance des Patienten bei der Umsetzung eines gesunden Lebensstils, sollte der Arzt hinsichtlich der Bedeutung von Lebensstilintervention, aber auch im Umgang mit schwierigen Situationen, wie die Empfehlung einer Gewichtsreduktion, sensibilisiert und bei der richtigen Handhabung der Leitlinienempfehlung st{\"a}rker unterst{\"u}tzt werden.}, subject = {Kardiovaskul{\"a}re Krankheit}, language = {de} } @phdthesis{Kaiser2020, author = {Kaiser, Sebastian}, title = {A RecQ helicase in disguise: Characterization of the unconventional Structure and Function of the human Genome Caretaker RecQ4}, doi = {10.25972/OPUS-16041}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-160414}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2020}, abstract = {From the simplest single-cellular organism to the most complex multicellular life forms, genetic information in form of DNA represents the universal basis for all biological processes and thus for life itself. Maintaining the structural and functional integrity of the genome is therefore of paramount importance for every single cell. DNA itself, as an active and complex macromolecular structure, is both substrate and product of many of these biochemical processes. A cornerstone of DNA maintenance is thus established by the tight regulation of the multitude of reactions in DNA metabolism, repressing adverse side reactions and ensuring the integrity of DNA in sequence and function. The family of RecQ helicases has emerged as a vital class of enzymes that facilitate genomic integrity by operating in a versatile spectrum of nucleic acid metabolism processes, such as DNA replication, repair, recombination, transcription and telomere stability. RecQ helicases are ubiquitously expressed and conserved in all kingdoms of life. Human cells express five different RecQ enzymes, RecQ1, BLM, WRN, RecQ4 and RecQ5, which all exhibit individual as well as overlapping functions in the maintenance of genomic integrity. Dysfunction of three human RecQ helicases, BLM, WRN and RecQ4, causes different heritable cancer susceptibility syndromes, supporting the theory that genomic instability is a molecular driving force for cancer development. However, based on their inherent DNA protective nature, RecQ helicases represent a double-edged sword in the maintenance of genomic integrity. While their activity in normal cells is essential to prevent cancerogenesis and cellular aging, cancer cells may exploit this DNA protective function by the overexpression of many RecQ helicases, aiding to overcome the disadvantageous results of unchecked DNA replication and simultaneously gaining resistance against chemotherapeutic drugs. Therefore, detailed knowledge how RecQ helicases warrant genomic integrity is required to understand their implication in cancerogenesis and aging, thus setting the stage to develop new strategies towards the treatment of cancer. The current study presents and discusses the first high-resolution X-ray structure of the human RecQ4 helicase. The structure encompasses the conserved RecQ4 helicase core, including a large fraction of its unique C- terminus. Our structural analysis of the RecQ4 model highlights distinctive differences and unexpected similarities to other, structurally conserved, RecQ helicases and permits to draw conclusions about the functional implications of the unique domains within the RecQ4 C-terminus. The biochemical characterization of various RecQ4 variants provides functional insights into the RecQ4 helicase mechanism, suggesting that RecQ4 might utilize an alternative DNA strand separation technique, compared to other human RecQ family members. Finally, the RecQ4 model permits for the first time the analysis of multiple documented RecQ4 patient mutations at the atomic level and thus provides the possibility for an advanced interpretation of particular structure-function relationships in RecQ4 pathogenesis.}, subject = {Helikasen}, language = {en} } @phdthesis{Koelmel2020, author = {K{\"o}lmel, Wolfgang}, title = {Structural and functional characterization of TFIIH from \(Chaetomium\) \(thermophilum\)}, doi = {10.25972/OPUS-16176}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-161769}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2020}, abstract = {Gene expression and transfer of the genetic information to the next generation forms the basis of cellular life. These processes crucially rely on DNA, thus the preservation, transcription and translation of DNA is of fundamental importance for any living being. The general transcription factor TFIIH is a ten subunit protein complex, which consists of two subcomplexes: XPB, p62, p52, p44, p34, and p8 constitute the TFIIH core, CDK7, CyclinH, and MAT1 constitute the CAK. These two subcomplexes are connected via XPD. TFIIH is a crucial factor involved in both, DNA repair and transcription. The central role of TFIIH is underlined by three severe disorders linked to failure of TFIIH in these processes: xeroderma pigmentosum, Cockayne syndrome, and trichothiodystrophy. Only limited structural and functional data of TFIIH are available so far. Here, the model organism Chaetomium thermophilum was utilized with the aim to structurally and functionally characterize TFIIH. By combining the expression and purification of single TFIIH subunits with the co-expression and co-purification of dual complexes, a unique and powerful modular system of the TFIIH core subunits could be established, encompassing all proteins in high quality and fully functional. This system permits the step-wise assembly of TFIIH core, thereby making it possible to assess the influence of the intricate interaction network within TFIIH core on the overall enzymatic activities of TFIIH, which has not been possible so far. Utilizing the single subunits and dual complexes, a detailed interaction network of TFIIH core was established, revealing the crucial role of the p34 subunit as a central scaffold of TFIIH by linking the two proteins p44 and p52. Our studies also suggest that p62 constitutes the central interface of TFIIH to the environment rather than acting as a scaffold. TFIIH core complexes were assembled and investigated via electron microscopy. Preliminary data indicate that TFIIH adopts different conformational states, which are important to fulfill its functions in transcription and DNA repair. Additionally, a shortened construct of p62 was used to develop an easy-to-use, low cost strategy to overcome the crystallographic phase problem via cesium derivatization.}, subject = {Transkriptionsfaktor}, language = {en} } @phdthesis{Dehmer2024, author = {Dehmer, Markus}, title = {A novel USP11-TCEAL1-mediated mechanism protects transcriptional elongation by RNA Polymerase II}, doi = {10.25972/OPUS-36054}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-360544}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2024}, abstract = {Deregulated expression of MYC oncoproteins is a driving event in many human cancers. Therefore, understanding and targeting MYC protein-driven mechanisms in tumor biology remain a major challenge. Oncogenic transcription in MYCN-amplified neuroblastoma leads to the formation of the MYCN-BRCA1-USP11 complex that terminates transcription by evicting stalling RNAPII from chromatin. This reduces cellular stress and allows reinitiation of new rounds of transcription. Basically, tumors with amplified MYC genes have a high demand on well orchestration of transcriptional processes-dependent and independent from MYC proteins functions in gene regulation. To date, the cooperation between promoter-proximal termination and transcriptional elongation in cancer cells remains still incomplete in its understanding. In this study the putative role of the dubiquitinase Ubiquitin Specific Protease 11 (USP11) in transcription regulation was further investigated. First, several USP11 interaction partners involved in transcriptional regulation in neuroblastoma cancer cells were identified. In particular, the transcription elongation factor A like 1 (TCEAL1) protein, which assists USP11 to engage protein-protein interactions in a MYCN-dependent manner, was characterized. The data clearly show that TCEAL1 acts as a pro-transcriptional factor for RNA polymerase II (RNAPII)-medi- ated transcription. In detail, TCEAL1 controls the transcription factor S-II (TFIIS), a factor that assists RNAPII to escape from paused sites. The findings claim that TCEAL1 outcompetes the transcription elongation factor TFIIS in a non-catalytic manner on chromatin of highly expressed genes. This is reasoned by the need regulating TFIIS function in transcription. TCEAL1 equili- brates excessive backtracking and premature termination of transcription caused by TFIIS. Collectively, the work shed light on the stoichiometric control of TFIIS demand in transcriptional regulation via the USP11-TCEAL1-USP7 complex. This complex protects RNAPII from TFIIS-mediated termination helping to regulate productive transcription of highly active genes in neuroblastoma.}, subject = {Transkription}, language = {en} } @phdthesis{Nair2024, author = {Nair, Radhika Karal}, title = {Structural and biochemical characterization of USP28 inhibition by small molecule inhibitors}, doi = {10.25972/OPUS-28174}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-281742}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2024}, abstract = {Ubiquitination is an important post-translational modification that maintains cellular homeostasis by regulating various biological processes. Deubiquitinases (DUBs) are enzymes that reverse the ubiquitination process by catalyzing the removal of ubiquitin from a substrate. Abnormal expression or function of DUBs is often associated with the onset and progression of various diseases, including cancer. Ubiquitin specific proteases (USPs), which constitute the largest family of DUBs in humans, have become the center of interest as potential targets in cancer therapy as many of them display increased activity or are overexpressed in a range of malignant tumors or the tumor microenvironment. Two related members of the USP family, USP28 and USP25, share high sequence identities but play diverse biological roles. USP28 regulates cell proliferation, oncogenesis, DNA damage repair and apoptosis, whereas USP25 is involved in the anti-viral response, innate immunity and ER-associated degradation in addition to carcinogenesis. USP28 and USP25 also exhibit different oligomeric states - while USP28 is a constitutively active dimer, USP25 assumes an auto-inhibited tetrameric structure. The catalytic domains of both USP28 and USP25 comprise the canonical, globular USP-domain but contain an additional, extended insertion site called USP25/28 catalytic domain inserted domain (UCID) that mediates oligomerization of the proteins. Disruption of the USP25 tetramer leads to the formation of an activated dimeric protein. However, it is still not clear what triggers its activation. Due to their role in maintaining and stabilizing numerous oncoproteins, USP28 and USP25 have emerged as interesting candidates for anti-cancer therapy. Recent advances in small-molecular inhibitor development have led to the discovery of relatively potent inhibitors of USP28 and USP25. This thesis focuses on the structural elucidation of USP28 and the biochemical characterization of USP28/USP25, both in complex with representatives of three out of the eight compound classes reported as USP28/USP25-specific inhibitors. The crystal structures of USP28 in complex with the AZ compounds, Vismodegib and FT206 reveal that all three inhibitor classes bind into the same allosteric pocket distant from the catalytic center, located between the palm and the thumb subdomains (the S1-site). Intriguingly, this binding pocket is identical to the UCID-tip binding interface in the USP25 tetramer, rendering the protein in a locked, inactive conformation. Formation of the binding pocket in USP28 requires a shift in the helix α5, which induces conformational changes and local distortion of the binding channel that typically accommodates the C-terminal tail of Ubiquitin, thus preventing catalysis and abrogating USP28 activity. The key residues of the USP28-inhibitor binding pocket are highly conserved in USP25. Mutagenesis studies of these residues accompanied by biochemical and biophysical assays confirm the proposed mechanism of inhibition and similar binding to USP25. This work provides valuable insights into the inhibition mechanism of the small molecule compounds specifically for the DUBs USP28 and USP25. The USP28-inhibitor complex structures offer a framework to develop more specific and potent inhibitors.}, subject = {Unique Selling Proposition}, language = {en} }