@phdthesis{Juerges2022, author = {J{\"u}rges, Christopher Sebastian}, title = {Algorithmic methods for elucidating the transcriptomic landscape of herpesviruses}, doi = {10.25972/OPUS-27282}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-272825}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2022}, abstract = {Transcription describes the process of converting the information contained in DNA into RNA. Although, tremendous progress has been made in recent decades to uncover this complex mechanism, it is still not fully understood. Given the advances and reduction in cost of high-throughput sequencing experiments, more and more data have been generated to help elucidating this complex process. Importantly, these sequencing experiments produce massive amounts of data that are incomprehensible in their raw form for humans. Further, sequencing techniques are not always 100\% accurate and are subject to a certain degree of variability and, in special cases, they might introduce technical artifacts. Thus, computational and statistical methods are indispensable to uncover the information buried in these datasets. In this thesis, I worked with multiple high throughput datasets from herpes simplex virus 1 (HSV-1) and human cytomegalovirus (HCMV) infections. During the last decade, it has became clear that a gene might not have a single, but multiple sites at which transcription initiates. These multiple transcription start sites (TiSS) demonstrated to have regulatory effects on the gene itself depending on which TiSS is used. Specialized experimental approaches were developed to help identify TiSS (TiSS-profiling). In order to facilitate the identification of all potential TiSS that are used for cell type- and condition-specific transcription, I developed the tool iTiSS. By using a new general enrichment-based approach to predict TiSS, iTiSS proved to be applicable in integrated studies and made it less prone to false positives compared to other TiSS-calling tools. Another improvement in recent years was made in metabolic labeling experiments such as SLAM-seq. Here, they removed the time consuming and laborious step of physically separating new from old RNA in the samples. This was achieved by inducing specific nucleotide conversions in newly synthesized RNA that are later visible in the data. Consequently, the separation of new and old RNA is now done computationally and, hence, tools are needed that accurately quantify these fold-changes. My second tool that I developed, called GRAND-SLAM proved to be capable to accomplish this task and outperform competing programs. As both of my tools, iTiSS and GRAND-SLAM are not specifically tailored to my own goals, but could also facilitate the research of other groups in this field, I made them publicly available on GitHub. I applied my tools to datasets generated in our lab as well as to publicly available data sets from HSV-1 and HCMV, respectively. For HSV-1, I was able to predict and validate TiSS with nucleotide precision using iTiSS. This has lead to the most comprehensive annotation for HSV-1 to date, which now serves as the fundamental basis of any future transcriptomic research on HSV-1. By combining both my tools, I was further able to uncover parts of the highly complex gene kinetics in HCMV and to resolve the limitations caused by the densely packed genome of HCMV. With the ever-increasing advances in sequencing techniques and their decrease in cost, the amounts of data produced will continue to rise massively in the future. Additionally, more and more specialized omics approaches are appearing, calling for new tools to leverage their full information potential. Consequently, it has become apparent that specialized computational tools such as iTiSS and GRAND-SLAM are needed and will become an essential and indispensable part of the analysis.}, subject = {Herpesviren}, language = {en} } @unpublished{HennigPrustyKauferetal.2021, author = {Hennig, Thomas and Prusty, Archana B. and Kaufer, Benedikt and Whisnant, Adam W. and Lodha, Manivel and Enders, Antje and Thomas, Julius and Kasimir, Francesca and Grothey, Arnhild and Herb, Stefanie and J{\"u}rges, Christopher and Meister, Gunter and Erhard, Florian and D{\"o}lken, Lars and Prusty, Bhupesh K.}, title = {Selective inhibition of microRNA processing by a herpesvirus-encoded microRNA triggers virus reactivation from latency}, edition = {submitted version}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-267858}, year = {2021}, abstract = {Herpesviruses have mastered host cell modulation and immune evasion to augment productive infection, life-long latency and reactivation thereof 1,2. A long appreciated, yet elusively defined relationship exists between the lytic-latent switch and viral non-coding RNAs 3,4. Here, we identify miRNA-mediated inhibition of miRNA processing as a novel cellular mechanism that human herpesvirus 6A (HHV-6A) exploits to disrupt mitochondrial architecture, evade intrinsic host defense and drive the latent-lytic switch. We demonstrate that virus-encoded miR-aU14 selectively inhibits the processing of multiple miR-30 family members by direct interaction with the respective pri-miRNA hairpin loops. Subsequent loss of miR-30 and activation of miR-30/p53/Drp1 axis triggers a profound disruption of mitochondrial architecture, which impairs induction of type I interferons and is necessary for both productive infection and virus reactivation. Ectopic expression of miR-aU14 was sufficient to trigger virus reactivation from latency thereby identifying it as a readily drugable master regulator of the herpesvirus latent-lytic switch. Our results show that miRNA-mediated inhibition of miRNA processing represents a generalized cellular mechanism that can be exploited to selectively target individual members of miRNA families. We anticipate that targeting miR-aU14 provides exciting therapeutic options for preventing herpesvirus reactivations in HHV-6-associated disorders like myalgic encephalitis/chronic fatigue syndrome (ME/CFS) and Long-COVID.}, language = {en} } @article{WhisnantJuergesHennigetal.2020, author = {Whisnant, Adam W. and J{\"u}rges, Christopher S. and Hennig, Thomas and Wyler, Emanuel and Prusty, Bhupesh and Rutkowski, Andrzej J. and L'hernault, Anne and Djakovic, Lara and G{\"o}bel, Margarete and D{\"o}ring, Kristina and Menegatti, Jennifer and Antrobus, Robin and Matheson, Nicholas J. and K{\"u}nzig, Florian W. H. and Mastrobuoni, Guido and Bielow, Chris and Kempa, Stefan and Liang, Chunguang and Dandekar, Thomas and Zimmer, Ralf and Landthaler, Markus and Gr{\"a}sser, Friedrich and Lehner, Paul J. and Friedel, Caroline C. and Erhard, Florian and D{\"o}lken, Lars}, title = {Integrative functional genomics decodes herpes simplex virus 1}, series = {Nature Communications}, volume = {11}, journal = {Nature Communications}, doi = {10.1038/s41467-020-15992-5}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-229884}, year = {2020}, abstract = {The predicted 80 open reading frames (ORFs) of herpes simplex virus 1 (HSV-1) have been intensively studied for decades. Here, we unravel the complete viral transcriptome and translatome during lytic infection with base-pair resolution by computational integration of multi-omics data. We identify a total of 201 transcripts and 284 ORFs including all known and 46 novel large ORFs. This includes a so far unknown ORF in the locus deleted in the FDA-approved oncolytic virus Imlygic. Multiple transcript isoforms expressed from individual gene loci explain translation of the vast majority of ORFs as well as N-terminal extensions (NTEs) and truncations. We show that NTEs with non-canonical start codons govern the subcellular protein localization and packaging of key viral regulators and structural proteins. We extend the current nomenclature to include all viral gene products and provide a genome browser that visualizes all the obtained data from whole genome to single-nucleotide resolution. Here, using computational integration of multi-omics data, the authors provide a detailed transcriptome and translatome of herpes simplex virus 1 (HSV-1), including previously unidentified ORFs and N-terminal extensions. The study also provides a HSV-1 genome browser and should be a valuable resource for further research.}, language = {en} } @article{DjakovicHennigReinischetal.2023, author = {Djakovic, Lara and Hennig, Thomas and Reinisch, Katharina and Milić, Andrea and Whisnant, Adam W. and Wolf, Katharina and Weiß, Elena and Haas, Tobias and Grothey, Arnhild and J{\"u}rges, Christopher S. and Kluge, Michael and Wolf, Elmar and Erhard, Florian and Friedel, Caroline C. and D{\"o}lken, Lars}, title = {The HSV-1 ICP22 protein selectively impairs histone repositioning upon Pol II transcription downstream of genes}, series = {Nature Communications}, volume = {14}, journal = {Nature Communications}, doi = {10.1038/s41467-023-40217-w}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-358161}, year = {2023}, abstract = {Herpes simplex virus 1 (HSV-1) infection and stress responses disrupt transcription termination by RNA Polymerase II (Pol II). In HSV-1 infection, but not upon salt or heat stress, this is accompanied by a dramatic increase in chromatin accessibility downstream of genes. Here, we show that the HSV-1 immediate-early protein ICP22 is both necessary and sufficient to induce downstream open chromatin regions (dOCRs) when transcription termination is disrupted by the viral ICP27 protein. This is accompanied by a marked ICP22-dependent loss of histones downstream of affected genes consistent with impaired histone repositioning in the wake of Pol II. Efficient knock-down of the ICP22-interacting histone chaperone FACT is not sufficient to induce dOCRs in ΔICP22 infection but increases dOCR induction in wild-type HSV-1 infection. Interestingly, this is accompanied by a marked increase in chromatin accessibility within gene bodies. We propose a model in which allosteric changes in Pol II composition downstream of genes and ICP22-mediated interference with FACT activity explain the differential impairment of histone repositioning downstream of genes in the wake of Pol II in HSV-1 infection.}, language = {en} } @article{LodhaMuchsinJuergesetal.2023, author = {Lodha, Manivel and Muchsin, Ihsan and J{\"u}rges, Christopher and Juranic Lisnic, Vanda and L'Hernault, Anne and Rutkowski, Andrzej J. and Prusty, Bhupesh K. and Grothey, Arnhild and Milic, Andrea and Hennig, Thomas and Jonjic, Stipan and Friedel, Caroline C. and Erhard, Florian and D{\"o}lken, Lars}, title = {Decoding murine cytomegalovirus}, series = {PLOS Pathogens}, volume = {19}, journal = {PLOS Pathogens}, number = {5}, issn = {1553-7374}, doi = {10.1371/journal.ppat.1010992}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-350480}, year = {2023}, abstract = {The genomes of both human cytomegalovirus (HCMV) and murine cytomegalovirus (MCMV) were first sequenced over 20 years ago. Similar to HCMV, the MCMV genome had initially been proposed to harbor ≈170 open reading frames (ORFs). More recently, omics approaches revealed HCMV gene expression to be substantially more complex comprising several hundred viral ORFs. Here, we provide a state-of-the art reannotation of lytic MCMV gene expression based on integrative analysis of a large set of omics data. Our data reveal 365 viral transcription start sites (TiSS) that give rise to 380 and 454 viral transcripts and ORFs, respectively. The latter include 200 small ORFs, some of which represented the most highly expressed viral gene products. By combining TiSS profiling with metabolic RNA labelling and chemical nucleotide conversion sequencing (dSLAM-seq), we provide a detailed picture of the expression kinetics of viral transcription. This not only resulted in the identification of a novel MCMV immediate early transcript encoding the m166.5 ORF, which we termed ie4, but also revealed a group of well-expressed viral transcripts that are induced later than canonical true late genes and contain an initiator element (Inr) but no TATA- or TATT-box in their core promoters. We show that viral upstream ORFs (uORFs) tune gene expression of longer viral ORFs expressed in cis at translational level. Finally, we identify a truncated isoform of the viral NK-cell immune evasin m145 arising from a viral TiSS downstream of the canonical m145 mRNA. Despite being ≈5-fold more abundantly expressed than the canonical m145 protein it was not required for downregulating the NK cell ligand, MULT-I. In summary, our work will pave the way for future mechanistic studies on previously unknown cytomegalovirus gene products in an important virus animal model.}, language = {en} }