AdobeStock_©-razihusin_179789354_2000px_200131

Publications

Please check http://www.researcherid.com/rid/B-6121-2009 for a full publication list of Roland Eils

 

Chua, R.L.*, Lukassen, S.*,Trump, S.*, Hennig, B.P.*, Wendisch, D.*, Pott, F., Debnath, O., Thürmann, L., Kurth, F., Völker, M.T., Kazmierski, J., Timmermann, B., Twardziok, S., Schneider, S., Machleidt, F., Müller-Redetzky, H., Maier, M., Krannich, A., Schmidt, S., Balzer, F., Liebig, J., Loske, J., Suttorp, N., Eils, J., Ishaque, N., Liebert, U.G., von Kalle, C., Hocke, A., Witzenrath, M., Goffinet, C., Drosten, C., Laudi, S.§,Lehmann, I., Conrad, C.§, Sander, L.-E.§ & Eils, R.§ (2020). COVID-19 severity correlates with airway epithelium-immune cell interactions identified by single-cell analysis. Nature Biotechnology doi: 10.1038/s41587-020-0602-4

 

Lukassen, S.*, Chua, R.L.*, Trefzer, T.*, Kahn, N.C.*, Schneider, M.A.*, Muley, T., Winter, H., Meister, M., Veith, C., Boots, A.W., Hennig, B.P., Kreuter, M.§, Conrad, C.§, & Eils, R.§ (2020). SARS-CoV-2 receptor ACE2 and TMPRSS2 are primarily expressed in bronchial transient secretory cells. EMBO Journal, 39(10), doi: 10.15252/embj.20105114

 

Aschenbrenner, S.*, Kallenberger, S.*, Hoffmann, M. D., Huck, A., Eils, R.§ & Niopek, D.§ (2020). Coupling Cas9 to artificial inhibitory domains enhances CRISPR-Cas9 target specificity. Science Advances, 6(6), doi:10.1126/sciadv.aay0187

 

ICGC/TCGA Pan-Cancer Analysis of Whole Genomes Consortium (2020). Pan-cancer Analysis of Whole Genomes, Nature, 578(7793), 82-93, doi: 10.1038/s41586-020-1969-6

 

Yakneen, S.§, Waszak, S.M., PCAWG Technical Working Group, Gertz, M., Korbel, J. O.§, PCAWG Consortium (2020). Bulter enables rapid cloud-based analysis of thousands of human genomes. Nature Biotechnology, 38(3), doi: 10.1038/s41587-019-0360-3

 

Zapatka, M.*, Borozan, I.*, Brewer, D.S.*, Iskar, M.*, Grundhoff, A., Alawi, M., Desai, N., Cooper, C.S., Eils, R., Ferretti, V. & Lichter, P.§ (2020). The landscape of viral association in human cancers. Nature Genetics, doi: 10.1101/465757

 

Chan, C.W.Y., Gu, Z., Bieg, M., Eils, R. & Herrmann, C.§ (2019). Impact of cancer mutational signatures on transcription factor motifs in the human genome. BMC Medical Genomics, 12(1), 64. doi: 10.1186/s12920-019-0525-4

 

Hoffmann, M. D., Aschenbrenner, S., Grosse, S., Rapti, K., Domenger, C., Mastel, M., Eils, R.§, Grimm, D.§ & Niopek, D.§ (2019). Cell-specific CRISPR-Cas9 activation by microRNA-dependent expression of anti-CRISPR proteins. Nucleic Acids Research, 47(13), doi:10.1093/nar/gkz271

 

Liu, L.*, Liu, C.*, Quintero, A.*, Wu, L*., Yuan, Y.*, Wang, M., Cheng, M., Leng, L., Xu, L., Dong, G., Li, R., Liu, Y., Wei, X., Xu, J., Chen, X., Lu, H., Chen, D., Wang, Q., Zhou, Q., Lin, X., Li, G., Liu, S., Wang, Q., Wang, H., Fink, J.L., Gao, Z., Liu, X., Hou, Y., Zhu, S., Yang, H., Ye, Y., Lin, G., Chen, F., Herrmann, C., Eils, R.§, Shang, Z.§ & Xu, X.§ (2019). Deconvolution of single-cell multi-omics layers reveals regulatory heterogeneity. Nature Communications, 10(1), 470. doi: 10.1038/s41467-018-08205-7

 

Mallm, J.-P.*, Iskar, M.*, Ishaque, N.*, Klett, L.C., Kugler, S.J., Muino, J.M., Teif, V.B., Poos, A.M., Großmann, S., Erdel, F., Tavernari, D., Koser, S.D., Schumacher, S., Brors, B., König, R., Remondini, D., Vingron, M., Stilgenbauer, S., Lichter, P., Zapatka, M., Mertens, D.§ & Rippe, K.§ (2019). Linking aberrant chromatin features in chronic lymphocytic leukemia to transcription factor networks. Molecular Systems Biology, 22;15(5):e8339. doi: 10.15252/msb.20188339

 

Park, J.*, Choi, W.*, Tiesmeyer, S., Long, B., Borm, L.E., Garren, E., Nguyen, T.N., Codeluppi, S., Schlesner, M., Tasic, B., Eils, R.§ & Ishaque, N.§ (2019). Segmentation-free inference of cell types from in situ transcriptomics data. bioRxiv, doi: 10.1101/800748

 

Smits, M., Zoldan, K., Ishaque, N., Gu, Z., Jechow, K., Wieland, D., Conrad, C., Eils, R., Fauvelle, C., Baumert, T.F., Emmerich, F., Bengsch, B., Neumann-Haefelin, C., Hofmann, M., Thimme, R. & Boettler, T.§ (2019). Follicular T helper cells shape the HCV-specific CD4 T cell repertoire after viral elimination. The Journal of Clinocal Investigation, 3;130(2):998-1009. doi: 10.1172/JCI129642

 

Tirier, S. M., Park, J., Preusser, F., Amrhein, L., Gu, Z., Steiger, S., Mallm, J. P., Krieger, T., Waschow, M., Eismann, B., Gut, M., Gut, I. G., Rippe, K., Schlesner, M., Theis, F., Fuchs, C., Ball, C. R., Glimm, H., Eils, R. & Conrad, C.§ (2019). Pheno-seq - linking visual features and gene expression in 3D cell culture systems. Scientific Reports, 9(1), 12367. doi: 10.1038/s41598-019-48771-4

 

Upmeier zu Belzen, J., Bürgel, T., Holderbach, S., Bubeck, F., Adam, L., Gandor, C., Klein, M., Mathony, J., Pfuderer, P. L., Platz, L., Przybilla, M., Schwendemann, M., Heid, D., Hoffmann, M. D., Jendrusch, M., Schmelas, C., Waldhauer, M., Lehmann, I., Niopek, D.§ & Eils, R.§ (2019). Leveraging implicit knowledge in neural networks for functional dissection and engineering of proteins. Nature Machine Intelligence 1, 225-235. doi: 10.1038/s42256-019-0049-9

 

Bubeck, F., Hoffmann, M. D., Harteveld, Z., Aschenbrenner, S., Bietz, A., Waldhauer, M. C., Börner, K., Fakhiri, J., Schmelas, C., Dietz, L., Grimm, D., Correia, B. E., Eils, R.§ & Niopek, D.§ (2018). Engineered anti-CRISPR proteins for optogenetic control of CRISPR/Cas9. Nature Methods. 15(11), 924-927, doi:10.1038/s41592-018-0178-9

 

Gröbner, S.N.*, Worst, B.C.*, Weischenfeldt, J., Buchhalter, I., Kleinheinz, K., Rudneva, V.A., Johann, P.D., Balasubramanian, G.P., Segura-Wang, M., Brabetz, S., Bender, S., Hutter, B., Sturm, D., Pfaff, E., Hubschmann, D., Zipprich, G., Heinold, M., Eils, J., Lawerenz, C., Erkek, S., Lambo, S., Waszak, S., Blattmann, C., Borkhardt, A., Kuhlen, M., Eggert, A., Fulda, S., Gessler, M., Wegert, J., Kappler, R., Baumhoer, D., Burdach, S., Kirschner-Schwabe, R., Kontny, U., Kulozik, A.E., Lohmann, D., Hettmer, S., Eckert, C., Bielack, S., Nathrath, M., Niemeyer, C., Richter, G.H., Schulte, J., Siebert, R., Westermann, F., Molenaar, J.J., Vassal, G., Witt, H., Burkhardt, B., Kratz, C.P., Witt, O., van Tilburg, C.M., Kramm, C.M., Fleischhack, G., Dirksen, U., Rutkowski, S., Fruhwald, M., von Hoff, K., Wolf, S., Klingebiel, T., Koscielniak, E., Landgraf, P., Koster, J., Resnick, A.C., Zhang, J., Liu, Y., Zhou, X., Waanders, A.J., Zwijnenburg, D.A., Raman, P., Brors, B., Weber, U.D., Northcott, P.A., Pajtler, K.W., Kool, M., Piro, R.M., Korbel, J.O., Schlesner, M., Eils, R., Jones, D.T.W., Lichter, P., Chavez, L.§, Zapatka, M.§, Pfister, S.M.§ (2018). The landscape of genomic alterations across childhood cancers. Nature, 559(7714), 321-327, doi: 10.1038/s41586-018-0167-2

 

Ishaque, N.*, Abba, M.L.*, Hauser, C., Patil, N., Paramasivam, N., Huebschmann, D., Leupold, J.H., Balasubramanian, G.P., Kleinheinz, K., Toprak, U.H., Hutter, B., Benner, A., Shavinskaya, A., Zhou, C., Gu, Z., Kerssemakers, J., Marx, A., Moniuszko, M., Kozlowski, M., Reszec, J., Niklinski, J., Eils, J., Schlesner, M., Eils, R., Brors, B. & Allgayer, H.§ (2018). Whole genome sequencing puts forward hypotheses on metastasis evolution and therapy in colorectal cancer. Nature Communications, 9(1), 4782. doi: 10.1038/s41467-018-07041-z

 

Kumar S.*, Warrel J.*, Shantao L., McGillivray P., Meyerson W., Salichos L., Harmanci, A., Martinez-FGundichely, A., Chan, C. W. Y., Nielsen, M. M., Locjovsky, L., Zhang, Y., Li, X., Pedersen, J. S., Herrmann C., Getz G., Khurana E. & Gerstein M. B.§ (2018). Passenger mutations in 2500 cancer genomes: Overall molecular functional impact and consequences, Cell, in Press, bioRxiv, doi: 10.1101/280446

 

Jabs, J., Zickgraf, F. M., Park, J., Wagner, S., Jiang, X., Jechow, K., Kleinheinz, K., Toprak, U. H., Schneider, M. A., Meister, M., Spaich, S., Sütterlin, M., Schlesner, M., Trumpp, A., Sprick, M., Eils, R.§ & Conrad, C.§ (2017). Screening drug effects in patient-derived cancer cells links organoid responses to genome alterations. Molecular Systems Biology, 13(11):955. doi: 10.15252/msb.20177697

 

Northcott, P. A.*, Buchhalter, I.*, Morrissy, A. S.*, Hovestadt, V., Weischenfeldt, J., Ehrenberger, T., Gröbner, S., Segura-Wang, M., Zichner, T., Rudneva, V. A., Warnatz, H. J., Sidiropoulos, N., Phillips, A. H., Schumacher, S., Kleinheinz, K., Waszak, S. M., Erkek, S., Jones, D. T. W., Worst, B. C., Kool, M., Zapatka, M., Jäger, N., Chavez, L., Hutter, B., Bieg, M., Paramasivam, N., Heinold, M., Gu, Z., Ishaque, N., Jäger-Schmidt, C., Imbusch, C. D., Jugold, A., Hübschmann, D., Risch, T., Amstislavskiy, V., Gonzalez, F. G. R., Weber, U. D., Wolf, S., Robinson, G. W., Zhou, X., Wu, G., Finkelstein, D., Liu, Y., Cavalli, F. M. G., Luu, B., Ramaswamy, V., Wu, X., Koster, J., Ryzhova, M., Cho, Y. J., Pomeroy, S. L., Herold-Mende, C., Schuhmann, M., Ebinger, M.,  Liau, L. M., Mora, J., McLendon, R. E., Jabado, N., Kumabe, T., Chuah, E., Ma, Y., Moore, R. A., Mungall, A. J., Mungall, K. L., Thiessen, N., Tse, K., Wong, T., Jones, S. J. M., Witt, O., Milde, T., Von Deimling, A., Capper, D., Korshunov, A., Yaspo, M. L., Kriwacki, R., Gajjar, A., Zhang, J., Beroukhim, R., Fraenkel, E., Korbel, J. O., Brors, B., Schlesner, M., Eils, R.§, Marra, M. A.§, Pfister, S. M.§, Taylor, M. D.§ & Lichter, P.§ (2017). The whole-genome landscape of medulloblastoma subtypes. Nature. 2017 Jul 19;547(7663):311-317. doi: 10.1038/nature22973

 

Reisinger, E.§, Genthner, L., Kerssemakers, J., Kensche, P., Borufka, S., Jugold, A., Kling, A., Prinz, M., Scholz, I., Zipprich, G., Eils, R., Lawerenz, C., Eils, J. (2017). OTP: An automatized system for managing and processing NGS data. Journal of Biotechnology, 10(261), 53-62, doi: 10.1016/j.jbiotec.2017.08.006

 

Bauer, T.*, Trump, S.*, Ishaque, N.*, Thurmann, L.*, Gu, L.*, Bauer, M.*, Bieg, M., Gu, Z.G., Weichenhan, D., Mallm, J.P., Roder, S., Herberth, G., Takada, E., Mucke, O., Winter, M., Junge, K.M., Grutzmann, K., Rolle-Kampczyk, U., Wang, Q., Lawerenz, C., Borte, M., Polte, T., Schlesner, M., Schanne, M., Wiemann, S., Georg, C., Stunnenberg, H.G., Plass, C., Rippe, K., Mizuguchi, J., Herrmann, C.*, Eils, R.§ & Lehmann, I.§ (2016). Environment-induced epigenetic reprogramming in genomic regulatory elements in smoking mothers and their children. Molecular Systems Biology, 12(3). doi: 10.15252/msb.20156520

 

Niopek, D., Wehler, P., Roensch, J., Eils, R.§ & Di Ventura, B.§ (2016). Optogenetic Control of Nuclear Protein Export. Nature Communication. 7. doi: 10.1038/ncomms10624

 

Peifer, M.*§, Hertwig, F.*, Roels, F.*, Dreidax, D.*, Gartlgruber, M.*, Menon, R., Krämer, A., Roncaioli, J.L., Sand, F., Heukmann J. M., Ikram, F., Ackermann, S., Engesser, A., Kahlert, Y., Altmüller, J., Nürnberg, P., Thierry-Mieg, J., Thierry-Mieg, D., Mariappan, A., Heynck, S., Mariotti, E., Henrich, K.-O., Gloeckner, C., Bosco, G., Leuscher, I., Schweiger, M. R., Savelyeya, L., Watkins, S. C., Shao, C., Bell, E., Höfer, T., Achter, V., Lang, U., Theissen, J., Volland, R., Saadati, M., Eggert, A., de Wilde, B., Peng, Z., Zhao, C., Shi, L., Ortmann, M., Büttner, R., Perner, S., Hero, B., Schramm, A., Schulte, J. H., Herrmann C., O’Sullivan R., Westermann F.§, Thomas R. K.§ & Fischer M.§ (2015). Telomerase activation by genomic rearrangements in high-risk neuroblastoma. Nature, 526(7575), 700–704. doi: 10.1038/nature14980

 

Wachsmuth, M., Conrad, C., Bulkescher, J., Koch, B., Mahen, R., Isokane R., Pepperkok, R.§ & Ellenberg, J.§ (2015). High-throughput fluorescence correlation spectroscopy enables analysis of proteome dynamics in living cells. Nature Biotechnology, 33(4), 384-389, doi: 10.1038/nbt.3146

 

Wegert, J.*, Ishaque, N.*, Vardapour, R., Geörg, C., Gu, Z., Bieg, M., Ziegler, B., Bausenwein, S., Nourkami, N., Ludwig, N., Keller, A., Grimm, C., Kneitz, S., Williams, R.D., Chagtai, T., Pritchard-Jones, K., van Sluis, P., Volckmann, R., Koster, J., Versteeg, R., Acha, T., O'Sullivan, M.J., Bode, P.K., Niggli, F., Tytgat, G.A., van Tinteren, H., van den Heuvel-Eibrink, M.M., Meese, E., Vokuhl, C., Leuschner, I., Graf, N., Eils, R., Pfister, S.M., Kool, M.§ & Gessler, M.§ (2015). Mutations in the SIX1/2 pathway and the DROSHA/DGCR8 miRNA microprocessor complex underlie high-risk blastemal type Wilms tumors. Cancer Cell, 9;27(2):298-311. doi: 10.1016/j.ccell.2015.01.002 

 

Jäger, N., Schlesner, M., Jones, David T.W., Raffel, S., Mallm, J.-P., Junge, Kristin M., Weichenhan, D., Bauer, T., Ishaque, N., Kool, M., Northcott, Paul A., Korshunov, A., Drews, Ruben M., Koster, J., Versteeg, R., Richter, J., Hummel, M., Mack, Stephen C., Taylor, Michael D., Witt, H., Swartman, B., Schulte-Bockholt, D., Sultan, M., Yaspo, M.-L., Lehrach, H., Hutter, B., Brors, B., Wolf, S., Plass, C., Siebert, R., Trumpp, A., Rippe, K., Lehmann, I., Lichter, P., Pfister, Stefan M. & Eils, R.§ (2013). Hypermutation of the Inactive X Chromosome Is a Frequent Event in Cancer. Cell, 155(3), 567-581. doi: 10.1016/j.cell.2013.09.042

 

Conrad, C., Wünsche, A., Tan, T. H., Bulkescher, J., Sieckmann, F., Verissimo, F., Edelstein, A., Walter, T., Liebel, U., Pepperkok, R.§ & Ellenberg, J.§ (2011) Micropilot: automation of fluorescence microscopy-based imaging for systems biology. Nature Methods, 8(3), 246–249, doi: 10.1038/nmeth.1558

 

Conrad, C.*, Erfle, H.*, Warnat, P., Daigle, N., Lorch, T., Ellenberg, J., Pepperkok, R. & Eils, R.§ (2004). Automatic identification of subcellular phenotypes on human cell arrays. Genome Research, 14(6), 1130-1136. doi: 10.1101/gr.2383804

 

*these authors contributed equally

§corresponding author

Latest News

SBHD 2024

Our colleaugues at Vanderbilt University organise the 16th INTERNATIONAL CONFERENCE ON SYSTEMS BIOLOGY OF HUMAN DISEASE – SBHD 2024 this year from June 10-12. Don’t miss the opportunity to

Top 100 Köpfe der Wissenschaft 2023

Am 11.10.2023 wurde Prof. Roland Eils im Tagesspiegel als einer der 100 wichtigsten Köpfe der Hauptstadt-Wissenschaft gewürdigt. So schreibt der Tagesspiegel: "Um Big Data dreht sich alles in der

Young BIH scientists attend 72nd Lindau Nobel Laureate Meeting

The Lindau Nobel Laureate Meetings are annual conferences where some of the brightest minds in science converge to exchange knowledge, foster collaboration, and inspire the next generation of

Roland Eils wird als Highly Cited Researcher international gelistet.

Roland Eils gehört zu den weltweit am häufigsten zitierten Forschenden des Jahres 2022, wie aus der Liste der „Highly Cited Researchers“ hervorgeht, die das Unternehmen Clarivate heute veröffentlicht