The impact of protein kinase signaling networks on influenza A virus (IAV) protein phosphorylation and virus replication

Research areas: Biochemistry, Signal Transduction, Virology

Influenza A viruses are characterized by a high degree of genomic plasticity which enables them to quickly adapt to new environmental conditions and to cross species barriers, causing epidemics and occasional pandemics. In the first funding period, CRISPR-Cas9-mediated gene editing was employed to generate mouse lung epithelial cells deficient in specific key proteins of the NF-B pathway. The functional analysis of these cells revealed that NF-κB was not relevant for replication of a mouse-adapted SC35M, while the absence of NF-κB activity increased replication of the non-adapted SC35 virus. The analysis of reassortant viruses showed that the anti-viral effect of NF-B is determined by the IAV genotype. In addition, the group has identified thousands of IAV-regulated phosphorylation sites of host cell and viral proteins were identified in a phospho-proteomic screen, which led to the discovery of new IAV-regulated kinase pathways, phosphorylation motifs, and cellular processes. This first comprehensive phospho-proteomic screen now allows to study the contribution of key phosphorylation sites and newly discovered pathways for IAV replication.

Project-related publications of the investigators:

  • Schloer S, Goretzko J, Pleschka S, Ludwig S, Rescher U. 2020. Combinatory Treatment with Oseltamivir and Itraconazole Targeting Both Virus and Host Factors in Influenza A Virus Infection. Viruses 12:703.
  • Mostafa A, Mahmoud SH, Shehata M, Muller C, Kandeil A, El-Shesheny R, Nooh HZ, Kayali G, Ali MA, Pleschka S. 2020. PA from a Recent H9N2 (G1-Like) Avian Influenza a Virus (AIV) Strain Carrying Lysine 367 Confers Altered Replication Efficiency and Pathogenicity to Contemporaneous H5N1 in Mammalian Systems. Viruses 12: E1046.
  • Harbig A, Mernberger M, Bittel L, Pleschka S, Schughart K, Steinmetzer T, Stiewe T, Nist A, Bottcher-Friebertshauser E. 2020. Transcriptome profiling and protease inhibition experiments identify proteases that activate H3N2 influenza A and influenza B viruses in murine airways. J Biol Chem 295:11388-11407.
  • Weber A, Dam S, Saul VV, Kuznetsova I, Muller C, Fritz-Wolf K, Becker K, Linne U, Gu H, Stokes MP, Pleschka S, Kracht M, Schmitz ML. 2019. Phosphoproteome Analysis of Cells Infected with Adapted and Nonadapted Influenza A Virus Reveals Novel Pro- and Antiviral Signaling Networks. J Virol 93: e00528-19.
  • Seibert M, Kruger M, Watson NA, Sen O, Daum JR, Slotman JA, Braun T, Houtsmuller AB, Gorbsky GJ, Jacob R, Kracht M, Higgins JMG, Schmitz ML. 2019. CDK1-mediated phosphorylation at H2B serine 6 is required for mitotic chromosome segregation. J Cell Biol 218:1164-1181.
  • Saul VV, Seibert M, Kruger M, Jeratsch S, Kracht M, Schmitz ML. 2019. ULK1/2 Restricts the Formation of Inducible SINT-Speckles, Membraneless Organelles Controlling the Threshold of TBK1 Activation. iScience 19:527-544.
  • Schmitz ML, Shaban MS, Albert BV, Gokcen A, Kracht M. 2018. The Crosstalk of Endoplasmic Reticulum (ER) Stress Pathways with NF-kappaB: Complex Mechanisms Relevant for Cancer, Inflammation and Infection. Biomedicines 6:59.
  • Poppe M, Wittig S, Jurida L, Bartkuhn M, Wilhelm J, Muller H, Beuerlein K, Karl N, Bhuju S, Ziebuhr J, Schmitz ML, Kracht M. 2017. The NF-kappaB-dependent and -independent transcriptome and chromatin landscapes of human coronavirus 229E-infected cells. PLoS Pathog 13:e1006286.
  • Haasbach E, Muller C, Ehrhardt C, Schreiber A, Pleschka S, Ludwig S, Planz O. 2017. The MEK-inhibitor CI-1040 displays a broad anti-influenza virus activity in vitro and provides a prolonged treatment window compared to standard of care in vivo. Antiviral Res 142:178-184.
  • Dam S, Kracht M, Pleschka S, Schmitz ML. 2016. The Influenza A Virus Genotype Determines the Antiviral Function of NF-kappaB. J Virol 90:7980-90.

Morbillivirus Intra- and Inter-Host Dynamics and Genetic Plasticity

Research area: Molecular Virology

The genomes of RNA viruses exist as quasispecies, due to the lack of proof-reading ability of their RNA-dependent RNA polymerase. The next-generation sequencing technology has provided new insights into the contribution of this genetic plasticity to adaptation and emergence of resistant variants in vitro, but the role of this plasticity in pathogenesis remains largely unknown. Canine distemper virus (CDV), a non-segmented negative-stranded RNA virus of the Morbillivirus genus in the Paramyxoviridae family, causes a lethal disease in ferrets. The von Messling group has shown that the virus initially amplifies in the immune system and that the subsequent spread to epithelial tissues coincides with the onset of clinical signs and transmission to a new host. Since the infection leads to extensive virus amplification in the respective target tissues, this system is ideally suited to directly characterize the viral genetic plasticity in vivo. First preliminary analyses of the sequence diversity in immune and epithelial tissues revealed distinct differences between virus inoculum and populations found at the end stages of infection. Based on these observations, this project will test the hypothesis that quasispecies contribute to virulence through modulation of dissemination and replication efficiency. It will provide new insights into the contribution of genetic plasticity to Morbillivirus biology, and pinpoint genome regions or motifs that are critical for virulence. The mechanistic concepts and experimental approaches developed in this system may also be applicable to other negative-stranded RNA viruses.

Project-related publications of the investigator:

  • Höper D, Freuling CM, Müller T, Hanke D, von Messling V, Duchow K, Beer M, Mettenleiter TC. High definition viral vaccine strain identity and stability testing using full-genome population data–The next generation of vaccine quality control. Vaccine 2015; 33: 5829-37.
  • Krumm SA, Yan D, Hovingh E, Evers TJ, Enkirch T, Reddy GP, Sun A, Saindane MT,Arrendale RF, Painter G, Liotta DC, Natchus MG, von Messling V, Plemper RK. An orally available, small-molecule polymerase inhibitor shows efficacy against a lethal morbillivirus infection in a large animal model. Sci. Transl. Med. 2014; 6: 232.
  • Svitek N, Gerhauser I, Goncalves C, Grabski E, Döring M, Kalinke U, Anderson DE, Cattaneo R, von Messling V. Morbillivirus control of the interferon response:relevance of STAT2 and mda5 but not STAT1 for canine distemper virus virulence in ferrets. J. Virol. 2014; 88: 2941-2950.
  • Frenzke M, Sawatsky B, Wong XX, Depeut S, Mateo M, Cattaneo R, von Messling V. Nectin-4-dependent measles virus spread to the cynomolgus monkey tracheal epithelium: role of infecting immune cells infiltrating the lamina propria. 2013; J. Virol. 87: 2526-2534.
  • Mühlebach MD, Mateo M, Sinn PL, Prüfer S, Uhlig KM, Leonard VHJ, Navaratnarajah CK, Frenzke M, Wong XX, Sawatsky B, Ramachandran S, McCray PB, Cichutek K, von Messling V, Lopez M, Cattaneo R. Adherens junction protein nectin-4 is the epithelial receptor for measles virus. Nature 2011; 480: 530-533.
  • Pillet S and von Messling V. Canine distemper virus selectively inhibits apoptosis progression in infected immune cells. J. Virol. 2009; 83: 6279-6287.
  • Anderson D and von Messling V. The region between the canine distemper virus M and F genes modulates virulence by controlling fusion protein expression. J. Virol. 2008; 82: 10510-10518.
  • Bonami F, Rudd PA, and von Messling V. Disease duration determines canine distemper virus neurovirulence. J. Virol. 2007; 81: 12066-12070.
  • Rudd PA, Cattaneo R, and von Messling V. Canine distemper virus uses both the anterograde and the hematogenous pathway for neuroinvasion. J. Virol. 2006; 80: 9361-9370.
  • von Messling V, Milosevic D, Cattaneo R. Tropism illuminated: lymphocyte-based pathways blazed by lethal morbillivirus through the host immune system. Proc. Natl. Acad. Sci. 2004; 101: 14216-14221.

Structural and functional factors determining entry and replication of hepatitis D and B viruses during co-infection

Research areas: Molecular Virology, Viral and cellular factors involved in RNA virus tropism and pathogenicity

Human hepatitis D virus (HDV) is a small defective RNA-virus that is dependent on the presence of hepatitis B virus (HBV) as a helper virus in coinfected patients. Both viruses infect hepatocytes within the liver via blood and sexual contacts and can cause acute and chronic liver disease resulting in liver cirrhosis and liver cancer. Worldwide, at least 12 million people are chronically infected with HDV with limited therapeutic options available. The completion of the full HDV replication cycle is dependent on a helper virus and cellular host factors within co-infected hepatocytes. Since HDV uses HBV surface proteins (HBsAg) for envelopment, budding and secretion of virions into the blood, both viruses are supposed to use comparable entry pathways for infection. The liver-specific bile acid transporter (NTCP) was discovered in 2012 as high-affinity HBV/HDV receptor and was first confirmed to be essential for HDV infection in 2013 by the principal investigators of this proposal. Our establishment of highly susceptible human hepatoma cells stably expressing NTCP at physiological levels enabled us to study viral binding, entry and replication of HDV and HBV at the cellular level. In addition to NTCP, other cellular cofactors appear to be involved in the quantitative regulation of virus entry via NTCP, but these have not yet been fully identified and characterized. Recently, interaction of HDV with envelope proteins of other coinfecting hepatotropic viruses, e.g. hepatitis C virus (HCV), has been demonstrated, suggesting that HDV might alter its infection pattern by switching viral surface proteins. Besides human HDV, HDV-like viruses have recently been found in other vertebrates (e.g. snakes, birds). In this context, we have isolated the first non-human mammalian HDV isolate from rodents, which is closely related to human HDV with robust replication in human liver cells, but its replication pattern has not yet been characterized. During the next funding period, we will (i) identify and analyze cellular NTCP cofactors defining viral binding and entry, (ii) characterize structural determinants of the overlapping virus/substrate binding domains of NTCP, (iii) compare replication of HDV with phylogenetically related animal HDV, and (iv) analyze determinants of HDV packaging upon coinfection with other hepatotropic viruses (HBV/HCV).

Project-related publications of the investigators:

  • Rasche A, Lehmann F, Goldmann N, Nagel M, Moreira-Soto A, Nobach D, de Oliveira Carneiro I, Osterrieder N, Greenwood AD, Steinmann E, Lukashev AN, Schuler G, Glebe D, Drexler JF, Equid HBVC. 2021. A hepatitis B virus causes chronic infections in equids worldwide. Proc Natl Acad Sci U S A 118.
  • Pfluger LS, Norz D, Volz T, Giersch K, Giese A, Goldmann N, Glebe D, Bockmann JH, Pfefferle S, Dandri M, Schulze Zur Wiesch J, Lutgehetmann M. 2021. Clinical establishment of a laboratory developed quantitative HDV PCR assay on the cobas6800 high-throughput system. JHEP Rep 3:100356.
  • Pfefferkorn M, Schott T, Bohm S, Deichsel D, Felkel C, Gerlich WH, Glebe D, Wat C, Pavlovic V, Heyne R, Berg T, van Bommel F. 2021. Composition of HBsAg is predictive of HBsAg loss during treatment in patients with HBeAg-positive chronic hepatitis B. J Hepatol 74:283-292.
  • Palatini M, Muller SF, Lowjaga K, Noppes S, Alber J, Lehmann F, Goldmann N, Glebe D, Geyer J. 2021. Mutational Analysis of the GXXXG/A Motifs in the Human Na(+)/Taurocholate Co-Transporting Polypeptide NTCP on Its Bile Acid Transport Function and Hepatitis B/D Virus Receptor Function. Front Mol Biosci 8:699443.
  • Lowjaga K, Kirstgen M, Muller SF, Goldmann N, Lehmann F, Glebe D, Geyer J. 2021. Long-term trans-inhibition of the hepatitis B and D virus receptor NTCP by taurolithocholic acid. Am J Physiol Gastrointest Liver Physiol 320:G66-G80.
  • Kirstgen M, Muller SF, Lowjaga K, Goldmann N, Lehmann F, Alakurtti S, Yli-Kauhaluoma J, Baringhaus KH, Krieg R, Glebe D, Geyer J. 2021. Identification of Novel HBV/HDV Entry Inhibitors by Pharmacophore- and QSAR-Guided Virtual Screening. Viruses 13.
  • Kirstgen M, Lowjaga K, Muller SF, Goldmann N, Lehmann F, Glebe D, Baringhaus KH, Geyer J. 2021. Hepatitis D Virus Entry Inhibitors Based on Repurposing Intestinal Bile Acid Reabsorption Inhibitors. Viruses 13.
  • Grosser G, Muller SF, Kirstgen M, Doring B, Geyer J. 2021. Substrate Specificities and Inhibition Pattern of the Solute Carrier Family 10 Members NTCP, ASBT and SOAT. Front Mol Biosci 8:689757.
  • Glebe D, Goldmann N, Lauber C, Seitz S. 2021. HBV evolution and genetic variability: Impact on prevention, treatment and development of antivirals. Antiviral Res 186:104973.
  • Paraskevopoulou S, Pirzer F, Goldmann N, Schmid J, Corman VM, Gottula LT, Schroeder S, Rasche A, Muth D, Drexler JF, Heni AC, Eibner GJ, Page RA, Jones TC, Muller MA, Sommer S, Glebe D, Drosten C. 2020. Mammalian deltavirus without hepadnavirus coinfection in the neotropical rodent Proechimys semispinosus. Proc Natl Acad Sci U S A 117:17977-17983.
  • Kirstgen M, Lowjaga K, Muller SF, Goldmann N, Lehmann F, Alakurtti S, Yli-Kauhaluoma J, Glebe D, Geyer J. 2020. Selective hepatitis B and D virus entry inhibitors from the group of pentacyclic lupane-type betulin-derived triterpenoids. Sci Rep 10:21772.
  • Jensen O, Ansari S, Gebauer L, Muller SF, Lowjaga K, Geyer J, Tzvetkov MV, Brockmoller J. 2020. A double-Flp-in method for stable overexpression of two genes. Sci Rep 10:14018.
  • Rasche A, Lehmann F, Konig A, Goldmann N, Corman VM, Moreira-Soto A, Geipel A, van Riel D, Vakulenko YA, Sander AL, Niekamp H, Kepper R, Schlegel M, Akoua-Koffi C, Souza B, Sahr F, Olayemi A, Schulze V, Petraityte-Burneikiene R, Kazaks A, Lowjaga K, Geyer J, Kuiken T, Drosten C, Lukashev AN, Fichet-Calvet E, Ulrich RG, Glebe D, Drexler JF. 2019. Highly diversified shrew hepatitis B viruses corroborate ancient origins and divergent infection patterns of mammalian hepadnaviruses. Proc Natl Acad Sci U S A 116:17007-17012.
  • Noppes S, Muller SF, Bennien J, Holtemeyer M, Palatini M, Leidolf R, Alber J, Geyer J. 2019. Homo- and heterodimerization is a common feature of the solute carrier family SLC10 members. Biol Chem 400:1371-1384.
  • Gerresheim GK, Bathke J, Michel AM, Andreev DE, Shalamova LA, Rossbach O, Hu P, Glebe D, Fricke M, Marz M, Goesmann A, Kiniry SJ, Baranov PV, Shatsky IN, Niepmann M. 2019. Cellular Gene Expression during Hepatitis C Virus Replication as Revealed by Ribosome Profiling. Int J Mol Sci 20.
  • Pfefferkorn M, Bohm S, Schott T, Deichsel D, Bremer CM, Schroder K, Gerlich WH, Glebe D, Berg T, van Bommel F. 2018. Quantification of large and middle proteins of hepatitis B virus surface antigen (HBsAg) as a novel tool for the identification of inactive HBV carriers. Gut 67:2045-2053.
  • Nielsen KO, Mirza AH, Kaur S, Jacobsen KS, Winther TN, Glebe D, Pociot F, Hogh B, Storling J. 2018. Hepatitis B virus suppresses the secretion of insulin-like growth factor binding protein 1 to facilitate anti-apoptotic IGF-1 effects in HepG2 cells. Exp Cell Res 370:399-408.
  • Muller SF, Konig A, Doring B, Glebe D, Geyer J. 2018. Characterisation of the hepatitis B virus cross-species transmission pattern via Na+/taurocholate co-transporting polypeptides from 11 New World and Old World primate species. PLoS One 13:e0199200.
  • de Carvalho Dominguez Souza BF, Konig A, Rasche A, de Oliveira Carneiro I, Stephan N, Corman VM, Roppert PL, Goldmann N, Kepper R, Muller SF, Volker C, de Souza AJS, Gomes-Gouvea MS, Moreira-Soto A, Stocker A, Nassal M, Franke CR, Rebello Pinho JR, Soares M, Geyer J, Lemey P, Drosten C, Netto EM, Glebe D, Drexler JF. 2018. A novel hepatitis B virus species discovered in capuchin monkeys sheds new light on the evolution of primate hepadnaviruses. J Hepatol 68:1114-1122.

The protease specificity of influenza virus hemagglutinin and coronavirus spike protein with monobasic cleavage site: underlying mechanisms and host proteases involved

Research area: Molecular Virology

Proteolytic activation of enveloped viruses including influenza A and B virus (IAV/IBV) and coronaviruses (CoV) by host cell proteases is crucial for virus infectivity. We previously demonstrated in vitro that the transmembrane serine protease 2 (TMPRSS2) cleaves influenza virus hemagglutinin possessing a monobasic cleavage site. Further studies by us and others revealed that TMPRSS2 is capable of activating many respiratory viruses. Recently, we identified TMPRSS2 as major activating protease of IAV of almost all 16 HA-subtypes in human and murine airway cells and of IBV in human lung. Having established the crucial role of TMPRSS2 in influenza virus activation in lower human airways we now want to validate its role in IAV/IBV activation in the human upper respiratory tract. Furthermore, we aim at investigating the tissue distribution and role of TMPRSS2 orthologues in IAV activation in chicken and ducks that are important IAV hosts.

Project-related publications of the investigator:

  • Bestle D, Limburg H, Kruhl D, Harbig A, Stein DA, Moulton H, Matrosovich M, Abdelwhab EM, Stech J, Bottcher-Friebertshauser E. 2021. Hemagglutinins of Avian Influenza Viruses Are Proteolytically Activated by TMPRSS2 in Human and Murine Airway Cells. J Virol 95:e0090621.
  • Harbig A, Mernberger M, Bittel L, Pleschka S, Schughart K, Steinmetzer T, Stiewe T, Nist A, Bottcher-Friebertshauser E. 2020. Transcriptome profiling and protease inhibition experiments identify proteases that activate H3N2 influenza A and influenza B viruses in murine airways. J Biol Chem 295:11388-11407.
  • Bestle D, Heindl MR, Limburg H, Van Lam van T, Pilgram O, Moulton H, Stein DA, Hardes K, Eickmann M, Dolnik O, Rohde C, Klenk HD, Garten W, Steinmetzer T, Bottcher-Friebertshauser E. 2020. TMPRSS2 and furin are both essential for proteolytic activation of SARS-CoV-2 in human airway cells. Life Sci Alliance 3.
  • Limburg H, Harbig A, Bestle D, Stein DA, Moulton HM, Jaeger J, Janga H, Hardes K, Koepke J, Schulte L, Koczulla AR, Schmeck B, Klenk HD, Bottcher-Friebertshauser E. 2019. TMPRSS2 Is the Major Activating Protease of Influenza A Virus in Primary Human Airway Cells and Influenza B Virus in Human Type II Pneumocytes. J Virol 93.
  • Bottcher-Friebertshauser E. 2018. Membrane-Anchored Serine Proteases: Host Cell Factors in Proteolytic Activation of Viral Glycoproteins. In: Böttcher-Friebertshäuser E., Garten W., Klenk H. (eds) Activation of Viruses by Host Proteases. Springer, Cham, p.153-203.
  • Tarnow C, Engels G, Arendt A, Schwalm F, Sediri H, Preuss A, Nelson PS, Garten W, Klenk HD, Gabriel G, Bottcher-Friebertshauser E. 2014. TMPRSS2 is a host factor that is essential for pneumotropism and pathogenicity of H7N9 influenza A virus in mice. J Virol 88:4744-51.
  • Baron J, Tarnow C, Mayoli-Nussle D, Schilling E, Meyer D, Hammami M, Schwalm F, Steinmetzer T, Guan Y, Garten W, Klenk HD, Bottcher-Friebertshauser E. 2013. Matriptase, HAT, and TMPRSS2 activate the hemagglutinin of H9N2 influenza A viruses. J Virol 87:1811-20.
  • Bottcher-Friebertshauser E, Lu Y, Meyer D, Sielaff F, Steinmetzer T, Klenk HD, Garten W. 2012. Hemagglutinin activating host cell proteases provide promising drug targets for the treatment of influenza A and B virus infections. Vaccine 30:7374-80.
  • Bottcher-Friebertshauser E, Stein DA, Klenk HD, Garten W. 2011. Inhibition of influenza virus infection in human airway cell cultures by an antisense peptide-conjugated morpholino oligomer targeting the hemagglutinin-activating protease TMPRSS2. J Virol 85:1554-62.
  • Bottcher-Friebertshauser E, Freuer C, Sielaff F, Schmidt S, Eickmann M, Uhlendorff J, Steinmetzer T, Klenk HD, Garten W. 2010. Cleavage of influenza virus hemagglutinin by airway proteases TMPRSS2 and HAT differs in subcellular localization and susceptibility to protease inhibitors. J Virol 84:5605-14.
  • Bottcher E, Matrosovich T, Beyerle M, Klenk HD, Garten W, Matrosovich M. 2006. Proteolytic activation of influenza viruses by serine proteases TMPRSS2 and HAT from human airway epithelium. J Virol 80:9896-8.

Functional comparison of the NSs virulence factors from the genus Phlebovirus

Research area: Molecular Virology

The genus Phlebovirus (family Phenuiviridae, order Bunyavirales) contains virus species covering a wide spectrum of virulence. Rift Valley fever virus (RVFV) is highly pathogenic representative, whereas the Sandfly fever Sicilian virus (SFSV) displays an intermediate level of virulence. Although the importance of phleboviruses is increasingly recognized, we are only beginning to understand the mechanisms of pathogenicity. A key virulence factor of phleboviruses is the non-structural protein NSs, an inhibitor of the antiviral type I interferon (IFN) system. In the previous two funding periods, the Weber group identified the mechanisms by which the NSs proteins of both RVFV and SFSV inhibit the transactivation of the IFN genes and abrogate the antiviral protein kinase R (PKR) pathway. For RVFV, the NSs was found to recruit several E3 ubiquitin ligases of the F-Box type in order to destroy the general host cell transcription factor TF-IIH and the antiviral mRNA translation inhibitor PKR. SFSV NSs, by contrast, is occluding the DNA-binding domain of the IFN transcription factor IRF-3 to inhibit IFN induction, and also binds to the translation initiation factor eIF2B to protect the ribosomal machinery against the inhibitory PKR signaling. Expectedly, phleboviruses with a deleted NSs gene are strong activators of the IFN system. Using an NSs-deleted RVFV, the viral panhandle was identified the main trigger of IFN induction, RIG-I as its cytoplasmic sensor, and the full set of upregulated innate immunity genes was characterized by transcriptome analysis.

Project-related publications of the investigator:

  • Wuerth JD, Weber F. 2022. Shielding the mRNA-translation factor eIF2B from inhibitory p-eIF2 as a viral strategy to evade protein kinase R-mediated innate immunity. Curr Opin Immunol 78:102251.
  • Shalamova L, Felgenhauer U, Wilhelm J, Schaubmar AR, Buettner K, Schoen A, Widera M, Ciesek S, Weber F. 2022. Omicron variant of SARS-CoV-2 exhibits an increased resilience to the antiviral type I interferon response. PNAS Nexus 1:67.
  • Hufsky F, Beslic D, Boeckaerts D, Duchene S, Gonzalez-Tortuero E, Gruber AJ, Guo J, Jansen D, Juma J, Kongkitimanon K, Luque A, Ritsch M, Lencioni Lovate G, Nishimura L, Pas C, Domingo E, Hodcroft E, Lemey P, Sullivan MB, Weber F, Gonzalez-Candelas F, Krautwurst S, Perez-Cataluna A, Randazzo W, Sanchez G, Marz M. 2022. The International Virus Bioinformatics Meeting 2022. Viruses 14.
  • Hollidge BS, Salzano MV, Ibrahim JM, Fraser JW, Wagner V, Leitner NE, Weiss SR, Weber F, Gonzalez-Scarano F, Soldan SS. 2022. Targeted Mutations in the Fusion Peptide Region of La Crosse Virus Attenuate Neuroinvasion and Confer Protection against Encephalitis. Viruses 14.
  • Wuerth JD, Weber F. 2021. NSs of the mildly virulent sandfly fever Sicilian virus is unable to inhibit interferon signaling and upregulation of interferon-stimulated genes. J Gen Virol 102.
  • Kashiwagi K, Shichino Y, Osaki T, Sakamoto A, Nishimoto M, Takahashi M, Mito M, Weber F, Ikeuchi Y, Iwasaki S, Ito T. 2021. eIF2B-capturing viral protein NSs suppresses the integrated stress response. Nat Commun 12:7102.
  • Borrego B, Moreno S, de la Losa N, Weber F, Brun A. 2021. The Change P82L in the Rift Valley Fever Virus NSs Protein Confers Attenuation in Mice. Viruses 13.
  • Wuerth JD, Habjan M, Kainulainen M, Berisha B, Bertheloot D, Superti-Furga G, Pichlmair A, Weber F. 2020. eIF2B as a Target for Viral Evasion of PKR-Mediated Translation Inhibition. mBio 11.
  • Schoen A, Lau S, Verbruggen P, Weber F. 2020. Elongin C Contributes to RNA Polymerase II Degradation by the Interferon Antagonist NSs of La Crosse Orthobunyavirus. J Virol 94.
  • Lau S, Weber F. 2020. Nuclear pore protein Nup98 is involved in replication of Rift Valley fever virus and nuclear import of virulence factor NSs. J Gen Virol 101:712-716.
  • Barr, JD, Weber F., Schmaljohn CS. 2020. Bunyaviruses. Chapter 17, Fields Virology, 7th edition. Lippincott Williams & Wilkins – Philadelphia, USA.
  • Wuerth JD, Weber F. 2019. Ferreting out viral pathogenesis. Nat Microbiol 4:384-385.
  • Jones R, Lessoued S, Meier K, Devignot S, Barata-Garcia S, Mate M, Bragagnolo G, Weber F, Rosenthal M, Reguera J. 2019. Structure and function of the Toscana virus cap-snatching endonuclease. Nucleic Acids Res 47:10914-10930.
  • Holzer M, Schoen A, Wulle J, Muller MA, Drosten C, Marz M, Weber F. 2019. Virus- and Interferon Alpha-Induced Transcriptomes of Cells from the Microbat Myotis daubentonii. iScience 19:647-661.
  • Frantz R, Teubner L, Schultze T, La Pietra L, Muller C, Gwozdzinski K, Pillich H, Hain T, Weber-Gerlach M, Panagiotidis GD, Mostafa A, Weber F, Rohde M, Pleschka S, Chakraborty T, Abu Mraheil M. 2019. The secRNome of Listeria monocytogenes Harbors Small Noncoding RNAs That Are Potent Inducers of Beta Interferon. mBio 10.
  • Wuerth JD, Habjan M, Wulle J, Superti-Furga G, Pichlmair A, Weber F. 2018. NSs Protein of Sandfly Fever Sicilian Phlebovirus Counteracts Interferon (IFN) Induction by Masking the DNA-Binding Domain of IFN Regulatory Factor 3. J Virol 92.
  • Kiening M, Weber F, Frishman D. 2017. Conserved RNA structures in the intergenic regions of ambisense viruses. Sci Rep 7:16625.
  • Ferron F, Weber F, de la Torre JC, Reguera J. 2017. Transcription and replication mechanisms of Bunyaviridae and Arenaviridae L proteins. Virus Res 234:118-134.

Lassa virus: host cell tropism and molecular pathogenesis

Research area: Molecular Virology

Lassa virus (LASV) is a zoonotic, hemorrhagic fever-causing virus that is endemic to West Africa.  Approved vaccines or specific antiviral drugs suitable to combat these infections are not available. The primary transmission route of LASV from its reservoir host to humans involves direct exposure to virus-containing rodent excretions. Although the initial infection involves the respiratory tract through inhalation of contaminated dust or fomites, information on LASV infection of human respiratory epithelial cells is limited. In the current funding period, the Strecker group observed that LASV infects polarized bronchial epithelial cells via the apical or basolateral side, while progeny virus particles are released predominantly from the apical surface. Goblet cells and basal cells were identified as initial LASV target cells in a primary human bronchial epithelial cell culture model. Together with collaborators, the Strecker group also solved the three-dimensional architecture of LASV virions and the spike envelope glycoprotein using high-resolution electron cryomicroscopy and tomography techniques.

Project-related publications of the investigator:

  • Thom R, Tipton T, Strecker T, Hall Y, Akoi Bore J, Maes P, Raymond Koundouno F, Fehling SK, Krahling V, Steeds K, Varghese A, Bailey G, Matheson M, Kouyate S, Cone M, Moussa Keita B, Kouyate S, Richard Ablam A, Laenen L, Vergote V, Guiver M, Timothy J, Atkinson B, Ottowell L, Richards KS, Bosworth A, Longet S, Mellors J, Pannetier D, Duraffour S, Munoz-Fontela C, Sow O, Koivogui L, Newman E, Becker S, Sprecher A, Raoul H, Hiscox J, Henao-Restrepo AM, Sakoba K, Magassouba N, Gunther S, Kader Konde M, Carroll MW. 2021. Longitudinal antibody and T cell responses in Ebola virus disease survivors and contacts: an observational cohort study. Lancet Infect Dis 21:507-516.
  • Olayemi A, Adesina AS, Strecker T, Magassouba N, Fichet-Calvet E. 2020. Determining Ancestry between Rodent- and Human-Derived Virus Sequences in Endemic Foci: Towards a More Integral Molecular Epidemiology of Lassa Fever within West Africa. Biology (Basel) 9.
  • Muller H, Fehling SK, Dorna J, Urbanowicz RA, Oestereich L, Krebs Y, Kolesnikova L, Schauflinger M, Krahling V, Magassouba N, Fichet-Calvet E, Ball JK, Kaufmann A, Bauer S, Becker S, von Messling V, Strecker T. 2020. Adjuvant formulated virus-like particles expressing native-like forms of the Lassa virus envelope surface glycoprotein are immunogenic and induce antibodies with broadly neutralizing activity. NPJ Vaccines 5:71.
  • Blok AJ, Gurnani P, Xenopoulos A, Burroughs L, Duncan J, Urbanowicz RA, Tsoleridis T, Muller-Krauter H, Strecker T, Ball JK, Alexander C, Alexander MR. 2020. Polymer microarrays rapidly identify competitive adsorbents of virus-like particles. Biointerphases 15:061005.
  • Timothy JWS, Hall Y, Akoi-Bore J, Diallo B, Tipton TRW, Bower H, Strecker T, Glynn JR, Carroll MW. 2019. Early transmission and case fatality of Ebola virus at the index site of the 2013-16 west African Ebola outbreak: a cross-sectional seroprevalence survey. Lancet Infect Dis 19:429-438.
  • Whitmer SLM, Strecker T, Cadar D, Dienes HP, Faber K, Patel K, Brown SM, Davis WG, Klena JD, Rollin PE, Schmidt-Chanasit J, Fichet-Calvet E, Noack B, Emmerich P, Rieger T, Wolff S, Fehling SK, Eickmann M, Mengel JP, Schultze T, Hain T, Ampofo W, Bonney K, Aryeequaye JND, Ribner B, Varkey JB, Mehta AK, Lyon GM, 3rd, Kann G, De Leuw P, Schuettfort G, Stephan C, Wieland U, Fries JWU, Kochanek M, Kraft CS, Wolf T, Nichol ST, Becker S, Stroher U, Gunther S. 2018. New Lineage of Lassa Virus, Togo, 2016. Emerg Infect Dis 24:599-602.
  • Watanabe Y, Raghwani J, Allen JD, Seabright GE, Li S, Moser F, Huiskonen JT, Strecker T, Bowden TA, Crispin M. 2018. Structure of the Lassa virus glycan shield provides a model for immunological resistance. Proc Natl Acad Sci U S A 115:7320-7325.
  • Sauerhering L, Muller H, Behner L, Elvert M, Fehling SK, Strecker T, Maisner A. 2017. Variability of interferon-lambda induction and antiviral activity in Nipah virus infected differentiated human bronchial epithelial cells of two human donors. J Gen Virol 98:2447-2453.
  • Huber M, Suprunenko T, Ashhurst T, Marbach F, Raifer H, Wolff S, Strecker T, Viengkhou B, Jung SR, Obermann HL, Bauer S, Xu HC, Lang PA, Tom A, Lang KS, King NJC, Campbell IL, Hofer MJ. 2017. IRF9 Prevents CD8(+) T Cell Exhaustion in an Extrinsic Manner during Acute Lymphocytic Choriomeningitis Virus Infection. J Virol 91.

Nipah virus replication in respiratory epithelial cells

Research area: Molecular Virology

The Malaysian Nipah virus (NiV) strain, a BSL-4 classified zoonotic paramyxovirus, emerged for the first time in 1998 and caused clinical diseases in pigs, which spread the infection to humans. While pigs developed an acute respiratory disease due to a highly efficient virus replication and associated inflammation processes in the airway epithelium, humans developed severe encephalitis with limited respiratory symptoms and little virus shedding via respiratory secretions. These differences in the clinical symptoms suggest species-specific differences in the infection of respiratory epithelial cells. Aiming to identify relevant host factors, we compared NiV infection in primary airway cultures of both species in terms of virus replication efficiencies and innate immune responses. A major finding was that NiV infections resulted in the expression of type III interferons (IFN-λ) in cells of both species with substantial quantitative differences. IFN induction was much lower in porcine airway cells, indicating an inherently limited IFN response. Surprisingly, the expression of proinflammatory cytokines (IL-6, IL-8) was not decreased in porcine epithelia. To evaluate the model that limited antiviral activity in porcine airway cells in the presence of a functional proinflammatory cytokine response is one of the species-specific factors contributing to the differences in NiV replication and virus-induced inflammatory processes, we will investigate the underlying signaling pathways in porcine and human airway epithelia. Here, we will focus on IFN-dependent and -independent expression of antiviral genes and proinflammatory cytokines in infected and uninfected bystander cells. The second part of the project is based on our recent findings that NiV not only forms cytosolic inclusion bodies (IBs), as is typical for all mononegaviruses, but additionally builds inclusions at the plasma membrane. Since the functional role of the two different types of inclusions in airway epithelia is still unknown, we will investigate their role in triggering or counteracting innate immune responses.

Project-related publications of the investigator:

  • Becker N, Heiner A, & Maisner A. 2022. Cytosolic Nipah Virus Inclusion Bodies Recruit Proteins Without Using Canonical Aggresome Pathways. Front. Virol. 1:821004.
  • Ringel M, Behner L, Heiner A, Sauerhering L, Maisner A. 2020. Replication of a Nipah Virus Encoding a Nuclear-Retained Matrix Protein. J Infect Dis 221:S389-S394.
  • Elvert M, Sauerhering L, Maisner A. 2020. Cytokine Induction in Nipah Virus-Infected Primary Human and Porcine Bronchial Epithelial Cells. J Infect Dis 221:S395-S400.
  • Ringel M, Heiner A, Behner L, Halwe S, Sauerhering L, Becker N, Dietzel E, Sawatsky B, Kolesnikova L, Maisner A. 2019. Nipah virus induces two inclusion body populations: Identification of novel inclusions at the plasma membrane. PLoS Pathog 15:e1007733.
  • Hoffmann M, Nehlmeier I, Brinkmann C, Krahling V, Behner L, Moldenhauer AS, Kruger N, Nehls J, Schindler M, Hoenen T, Maisner A, Becker S, Pohlmann S. 2019. Tetherin Inhibits Nipah Virus but Not Ebola Virus Replication in Fruit Bat Cells. J Virol 93.
  • Behner L, Zimmermann L, Ringel M, Weis M, Maisner A. 2018. Formation of high-order oligomers is required for functional bioactivity of an African bat henipavirus surface glycoprotein. Vet Microbiol 218:90-97.
  • Sauerhering L, Muller H, Behner L, Elvert M, Fehling SK, Strecker T, Maisner A. 2017. Variability of interferon-lambda induction and antiviral activity in Nipah virus infected differentiated human bronchial epithelial cells of two human donors. J Gen Virol 98:2447-2453.
  • Sauerhering L, Zickler M, Elvert M, Behner L, Matrosovich T, Erbar S, Matrosovich M, Maisner A. 2016. Species-specific and individual differences in Nipah virus replication in porcine and human airway epithelial cells. J Gen Virol 97:1511-1519.

Adaptation of Marburg virus to rodents as a model to study viral pathogenesis: Response of the mononuclear phagocyte system

Research area: Molecular Virology

Marburg virus (MARV) causes lethal fever in humans and non-lethal infections in rodents. Sequential passaging of MARV in guinea pigs resulted in a guinea pig-adapted strain that contains 4 non-synonymous mutations and causes lethal disease. Cells of the mononuclear phagocyte system (MPS) were found to be the primary MARV targets in both lethal and non-lethal infection. The Becker group discovered that a single mutation in the viral matrix protein VP40 (D184N) improved the growth of recombinant MARV (rMARVVP40(D184N)) specifically in guinea pig cells. This phenotype correlated with a decreased ability of VP40D184N to suppress viral RNA synthesis in these cells. In primary MPS cells derived from a newly established ex vivo model, rMARVVP40(D184N) had a clear growth advantage over wild-type MARV. This correlated with the observation that rMARVVP40(D184N), in contrast to the non-lethal wild type virus, no longer induced cell death in the infected macrophages or in non-infected bystander cells. Other mutations in the guinea pig-adapted MARV were found to be located in the polymerase (L) coding sequence. One of the mutations located in the polymerase active site was shown to dramatically increase the viral transcription/replication activity.

Project-related publications of the investigator:

  • Dietzel E, Schudt G, Krähling V, Matrosovich M, Becker S. 2016. Functional characterization of adaptive mutations during the West African Ebola virus outbreak. J. Virol. 91:e01913-16.
  • Koehler A, Kolesnikova L, Becker S. 2016. An active site mutation increases the polymerase activity of the guinea pig-lethal Marburg virus. J Gen Virol. 97(10):2494-2500.
  • Koehler A, Kolesnikova L, Welzel U, Schudt G, Herwig A, Becker S. 2016. A Single Amino Acid Change in the Marburg Virus Matrix Protein VP40 Provides a Replicative Advantage in a Species-Specific Manner. J Virol 90:1444-1454.
  • Wolff S, Groseth A, Meyer B, Jackson D, Strecker T, Kaufmann A, Becker S. 2016. The New World arenavirus Tacaribe virus induces caspase-dependent apoptosis in infected cells. J Gen Virol 97:855-866.
  • Dietzel E, Kolesnikova L, Sawatsky B, Heiner A, Weis M, Kobinger GP, Becker S, von Messling V, Maisner A. 2015. Nipah virus matrix protein influences fusogenicity and is essential for particle infectivity and stability. J Virol. 90(5):2514-22.
  • Schudt, G., O. Dolnik, L. Kolesnikova, N. Biedenkopf, A. Herwig and S. Becker. 2015. Transport of Ebolavirus Nucleocapsids Is Dependent on Actin Polymerization: Live-Cell Imaging Analysis of Ebolsvirus-Infected Cells. J. Infect Dis doi: 10.1093/infdis/jiv083
  • Kolesnikova, L., E. Mittler, G. Schudt, H. Shams-Eldin, and S. Becker. 2012. Phosphorylation of Marburg virus matrix protein VP40 triggers assembly of nucleocapsids with the viral envelope at the plasma membrane. Cell Microbiol 14:182-97
  • Mateo, M., C. Carbonnelle, O. Reynard, L. Kolesnikova, K. Nemirov, A. Page, V. A. Volchkova, and V. E. Volchkov. 2011. VP24 is a molecular determinant of Ebola virus virulence in guinea pigs. J Infect Dis 204 Suppl 3:S1011-20.
  • Welsch, S., L. Kolesnikova, V. Krahling, J. D. Riches, S. Becker, and J. A. Briggs. 2010. Electron tomography reveals the steps in filovirus budding. PLoS Pathog 6:e1000875.
  • Krahling, V., O. Dolnik, L. Kolesnikova, J. Schmidt-Chanasit, I. Jordan, V. Sandig, S. Gunther, and S. Becker. 2010. Establishment of fruit bat cells (Rousettus aegyptiacus) as a model system for the investigation of filoviral infection. PLoS Negl Trop Dis 4:e802.
  • Dolnik, O., L. Kolesnikova, and S. Becker. 2008. Filoviruses: Interactions with the host cell. Cell Mol Life Sci 65:756-76.
  • Kolesnikova, L., A. B. Bohil, R. E. Cheney, and S. Becker. 2007. Budding of Marburgvirus is associated with filopodia. Cell Microbiol 9:939-51.
  • Kolesnikova, L., B. Berghofer, S. Bamberg, and S. Becker. 2004. Multivesicular bodies as a platform for formation of the Marburg virus envelope. J Virol 78:12277-87.
  • Kolesnikova, L., S. Bamberg, B. Berghofer, and S. Becker. 2004. The matrix protein of Marburg virus is transported to the plasma membrane along cellular membranes: exploiting the retrograde late endosomal pathway. J Virol 78:2382-93.
  • Ryabchikova, E. I., L.  Kolesnikova, and S. V. Luchko. 1999. An analysis of features of pathogenesis in two animal models of Ebola virus infection. J Infect Dis 179 Suppl 1:S199-202.
  • Mühlberger, E., B. Lotfering, H.-D. Klenk, and S. Becker. 1998. Three of the four nucleocapsid proteins of Marburg virus, NP, VP35, and L, are sufficient to mediate replication and transcription of Marburg virus-specific monocistronic minigenomes. J Virol 72:8756-64.
  • Ryabchikova, E., L. Kolesnikova, M. Smolina, V. Tkachev, L. Pereboeva, S. Baranova, A. Grazhdantseva, and Y. Rassadkin. 1996. Ebola virus infection in guinea pigs: presumable role of granulomatous inflammation in pathogenesis. Arch Virol 141:909-21.
  • Ryabchikova, E., L. Strelets, L. Kolesnikova, O. Pyankov, and A. Sergeev. 1996. Respiratory Marburg virus infection in guinea pigs. Arch Virol 141:2177-90.

Role of receptor specificity and membrane fusion activity of influenza viruses in host range, cell tropism and pathogenicity

Research area: Molecular Virology/Molecular Immunology

Humans can be infected by seasonal, pandemic, and zoonotic influenza A viruses (IAVs). The hemagglutinin (HA) and neuraminidase (NA) surface glycoproteins of these viruses vary substantially depending on viral host species, evolutionary lineage, and strain. The long-term goal of the project is to characterize how this genetic variation affects viral replication, host range, and pathogenicity. During the first funding period, a large panel of fully characterized recombinant IAVs were generated and used to study effects of HA receptor specificity and membrane fusion activity on viral tropism to epithelial and endothelial cells and sensitivity to antiviral innate immune factors. The Matrosovich group found that the pH optimum of HA-mediated membrane fusion in specific IAVs affects their sensitivity to an interferon-induced antiviral state, and that the high pH fusion pH optimum seen in zoonotic H5N1 and H7N9 viruses is responsible for their enhanced tropism to human endothelial cells. Furthermore, the group discovered that adaptation of avian influenza viruses to pigs under natural conditions requires HA mutations that increase the fusion pH optimum. Finally, preliminary evidence was obtained to suggest a role of viral receptor specificity in IAV interactions with human alveolar macrophages and plasmacytoid dendritic cells.

Project-related publications of the principal investigators:

  • Gerlach T, Hensen L, Matrosovich T, Bergmann J, Winkler M, Peteranderl C, Klenk HD, Weber F, Herold S, Pöhlmann S, Matrosovich M. 2017. pH-optimum of hemagglutinin-mediated membrane fusion determines sensitivity of influenza A viruses to the interferon-induced antiviral state and IFITMs. J. Virol., accepted
  • Gambaryan AS, Matrosovich MN. 2015. What adaptive changes in hemagglutinin and neuraminidase are necessary for emergence of pandemic influenza virus from its avian precursor? Biochemistry (Moscow) 80:872-880.
  • Baumann J, Kouassi NM, Foni E, Klenk HD, Matrosovich M. 2015. H1N1 swine influenza viruses differ from avian precursors by a higher pH optimum of membrane fusion. J Virol 90:1569-1577.
  • Wendel I, Rubbenstroth D, Doedt J, Kochs G, Wilhelm J, Staeheli P, Klenk HD, Matrosovich M. 2015. The avian-origin PB1 gene segment facilitated replication and transmissibility of the H3N2/1968 pandemic influenza virus. J Virol 89:4170-4179.
  • Wendel I, Matrosovich M, Klenk HD. 2015. SnapShot: Evolution of Human Influenza A Viruses. Cell Host & Microbe 17:416-416.e411.
  • Heider A, Mochalova L, Harder T, Tuzikov A, Bovin N, Wolff T, Matrosovich M, Schweiger B. 2015. Alterations in hemagglutinin receptor-binding specificity accompany the emergence of highly pathogenic avian influenza viruses. J Virol 89:5395-5405.
  • Van Poucke S, Doedt J, Baumann J, Qiu Y, Matrosovich T, Klenk HD, Van Reeth K, Matrosovich M. 2015. Role of substitutions in the hemagglutinin in the emergence of the 1968 pandemic influenza virus. J Virol 89:12211-12216.
  • Bottcher-Friebertshauser E, Garten W, Matrosovich M, Klenk HD. 2014. The hemagglutinin: a determinant of pathogenicity. Curr Top Microbiol Immunol 385:3-34.
  • Linster M, Boheemen S, de Graaf M, Schrauwen EJA, Lexmond P, Manz B, Bestebroer TM, Baumann J, van Riel D, Rimmelzwaan GF, Osterhaus ADME, Matrosovich M, Fouchier RAM, Herfst S. 2014. Identification, characterization, and natural selection of mutations driving airborne transmission of A/H5N1 virus. Cell 157:329-339.
  • Hogner K, Wolff T, Pleschka S, Plog S, Gruber AD, Kalinke U, Walmrath HD, Bodner J, Gattenlohner S, Lewe-Schlosser P, Matrosovich M, Seeger W, Lohmeyer J, Herold S. 2013. Macrophage-expressed IFN-beta contributes to apoptotic alveolar epithelial cell injury in severe influenza virus pneumonia. Plos Pathog 9:e1003188.
  • Van Poucke S, Uhlendorff J, Wang ZF, Billiau V, Nicholls J, Matrosovich M, Van Reeth K. 2013. Effect of receptor specificity of A/Hong Kong/1/68 (H3N2) influenza virus variants on replication and transmission in pigs. Influenza and Other Respiratory Viruses 7:151-159.
  • Matrosovich M, Herrler G, Klenk HD. 2013. Sialic Acid Receptors of Viruses. Top Curr Chem 20:20.
  • Kolesnikova L, Heck S, Matrosovich T, Klenk HD, Becker S, Matrosovich M. 2013. Influenza virus budding from the tips of cellular microvilli in differentiated human airway epithelial cells. Journal of General Virology 94:971-976.
  • Klenk HD, Garten W, Matrosovich M. 2013. Pathogenesis, p 157-172. In Webster RG (ed), Textbook of Influenza, 2nd Edition. Wiley-Blackwell, Chichester, West Sussex, UK.
  • Crusat M, Liu JF, Palma AS, Childs RA, Liu Y, Wharton SA, Lin YP, Coombs PJ, Martin SR, Matrosovich M, Chen Z, Stevens DJ, Hien VM, Thanh TT, Nhu LNT, Nguyet LA, Ha DQ, van Doorn HR, Hien TT, Conradt HS, Kiso M, Gamblin SJ, Chai WG, Skehel JJ, Hay AJ, Farrar J, de Jong MD, Feizi T. 2013. Changes in the hemagglutinin of H5N1 viruses during human infection – Influence on receptor binding. Virology 447:326-337.
  • Corman VM, Eickmann M, Landt O, Bleicker T, Brunink S, Eschbach-Bludau M, Matrosovich M, Becker S, Drosten C. 2013. Specific detection by real-time reverse-transcription PCR assays of a novel avian influenza A(H7N9) strain associated with human spillover infections in China. Eurosurveillance 18:10-16.
  • Gambaryan, A. S., T. Y. Matrosovich, J. Philipp, V. J. Munster, R. A. Fouchier, G. Cattoli, I. Capua, S. L. Krauss, R. G. Webster, J. Banks, N. V. Bovin, H. D. Klenk, and M. N. Matrosovich. 2012. Receptor-binding profiles of H7 subtype influenza viruses in different host species. J.Virol. 86:4370-4379.
  • Liu, Y., R. A. Childs, T. Matrosovich, S. Wharton, A. S. Palma, W. Chai, R. Daniels, V. Gregory, J. Uhlendorff, M. Kiso, H. D. Klenk, A. Hay, T. Feizi, and M. Matrosovich. 2010. Altered receptor specificity and cell tropism of D222G hemagglutinin mutants isolated from fatal cases of pandemic A(H1N1) 2009 influenza virus. J.Virol. 84:12069-12074.
  • Childs, R. A., A. S. Palma, S. Wharton, T. Matrosovich, Y. Liu, W. Chai, M. A. Campanero-Rhodes, Y. Zhang, M. Eickmann, M. Kiso, A. Hay, M. Matrosovich, and T. Feizi. 2009. Receptor-binding specificity of pandemic influenza A (H1N1) 2009 virus determined by carbohydrate microarray. Nat.Biotechnol. 27:797-799.
  • Ocana-Macchi, M., M. Bel, L. Guzylack-Piriou, N. Ruggli, M. Liniger, K. C. McCullough, Y. Sakoda, N. Isoda, M. Matrosovich, and A. Summerfield. 2009. Hemagglutinin-dependent tropism of H5N1 avian influenza virus for human endothelial cells. J.Virol. 83:12947-12955.
  • Matrosovich, M., T. Matrosovich, J. Uhlendorff, W. Garten, and H. D. Klenk. 2007. Avian-virus-like receptor specificity of the hemagglutinin impedes influenza virus replication in cultures of human airway epithelium. Virology. 361:384-390.
  • Gambaryan, A., S. Yamnikova, D. Lvov, A. Tuzikov, A. Chinarev, G. Pazynina, R. Webster, M. Matrosovich, and N. Bovin. 2005. Receptor specificity of influenza viruses from birds and mammals: new data on involvement of the inner fragments of the carbohydrate chain. Virology 334:276-283.
  • Matrosovich, M. N., T. Y. Matrosovich, T. Gray, N. A. Roberts, and H. D. Klenk. 2004. Human and avian influenza viruses target different cell types in cultures of human airway epithelium. Proc.Natl.Acad.Sci U.S.A 101:4620-4624.
  • Matrosovich, M., A. Tuzikov, N. Bovin, A. Gambaryan, A. Klimov, M. R. Castrucci, I. Donatelli, and Y. Kawaoka. 2000. Early alterations of the receptor-binding properties of H1, H2, and H3 avian influenza virus hemagglutinins after their introduction into mammals. J Virol. 74:8502-8512.
  • Jung S, von Thülen T, Laukemper V, Pigisch S, Hangel D, Wagner H, Kaufmann A, Bauer S. A single naturally occurring 2′-O-methylation converts a TLR7- and TLR8-activating RNA into a TLR8-specific ligand. PLoS One. 2015 Mar 18;10(3):e0120498. doi:10.1371/journal.pone.0120498. eCollection 2015.
  • Oldenburg M, Krüger A, Ferstl R, Kaufmann A, Nees G, Sigmund A, Bathke B, Lauterbach H, Suter M, Dreher S, Koedel U, Akira S, Kawai T, Buer J, Wagner H, Bauer S, Hochrein H, Kirschning CJ. TLR13 recognizes bacterial 23S rRNA devoid of erythromycin resistance-forming modification. Science. 2012 Aug 31;337(6098):1111-5. doi: 10.1126/science.1220363. Epub 2012 Jul 19.
  • Bauer, S., C. J. Kirschning, H. Hacker, V. Redecke, S. Hausmann, S. Akira, H. Wagner, and G. B. Lipford. 2001. Human TLR9 confers responsiveness to bacterial DNA via species-specific CpG motif recognition. Proc Natl Acad Sci U S A 98:9237-42.
  • Hamm, S., A. Heit, M. Koffler, K. M. Huster, S. Akira, D. H. Busch, H. Wagner, and S. Bauer. 2007. Immunostimulatory RNA is a potent inducer of antigen-specific cytotoxic and humoral immune response in vivo. Int Immunol 19:297-304.
  • Hamm, S., E. Latz, D. Hangel, T. Muller, P. Yu, D. Golenbock, T. Sparwasser, H. Wagner, and S. Bauer. 2010. Alternating 2′-O-ribose methylation is a universal approach for generating non-stimulatory siRNA by acting as TLR7 antagonist. Immunobiology 215:559-69.
  • Heil, F., H. Hemmi, H. Hochrein, F. Ampenberger, C. Kirschning, S. Akira, G. Lipford, H. Wagner, and S. Bauer. 2004. Species-specific recognition of single-stranded RNA via toll-like receptor 7 and 8. Science 303:1526-9.
  • Heil, F., P. Ahmad-Nejad, H. Hemmi, H. Hochrein, F. Ampenberger, T. Gellert, H. Dietrich, G. Lipford, K. Takeda, S. Akira, H. Wagner, and S. Bauer. 2003. The Toll-like receptor 7 (TLR7)-specific stimulus loxoribine uncovers a strong relationship within the TLR7, 8 and 9 subfamily. Eur J Immunol 33:2987-97.
  • Jöckel, S., G. Nees, R. Sommer, Y. Zhao, D. Cherkasov, H. Hori, G. Ehm, M. Schnare, M. Nain, A. Kaufmann, and S. Bauer. 2012. The 2′-O-methylation status of a single guanosine controls transfer RNA-mediated Toll-like receptor 7 activation or inhibition. J Exp Med 209:235-41.
  • Jurk, M., F. Heil, J. Vollmer, C. Schetter, A. M. Krieg, H. Wagner, G. Lipford, and S. Bauer. 2002. Human TLR7 or TLR8 independently confer responsiveness to the antiviral compound R-848. Nat Immunol 3:499.
  • Rutz, M., J. Metzger, T. Gellert, P. Luppa, G. B. Lipford, H. Wagner, and S. Bauer. 2004. Toll-like receptor 9 binds single-stranded CpG-DNA in a sequence- and pH-dependent manner. Eur J Immunol 34:2541-50.

Pathogenesis of feline infectious peritonitis

Research area: Molecular Virology 

Feline coronaviruses (FCoVs) are highly prevalent in the cat population and classified into two biotypes: feline enteric coronavirus (FECV) leads to inapparent persistent infections of the gut, whereas feline infectious peritonitis virus (FIPV) causes a systemic disease with fatal outcome. According to the „internal mutation theory“, FIPV evolves from FECV by acquiring biotype-changing mutations, which is thought to occur in 5–10% of persistently infected cats. To date, the genetic changes responsible for the biotype switch have not been identified. The unambiguous identification of mutations critically involved in this process requires reverse genetics approaches suitable to produce, characterize, and manipulate genetically defined pairs of FECV-FIPV. The development of reverse genetic systems for FCoV field viruses represents a major technical challenge because these viruses do not grow in standard cell culture systems. In the first CRC 1021 funding period, the Tekes and Ziebuhr groups have resolved this problem and managed to establish reverse genetic systems suitable to produce wild-type and genetically manipulated serotype I field viruses, which will now be employed to identify mutations that contribute to the development of feline infectious peritonitis using both in vitro and in vivo experiments and to study functions of virus-encoded „accessory“ proteins in viral pathogenesis.

Project-related publications of the investigators:

  • Lemmermeyer T, Lamp B, Schneider R, Ziebuhr J, Tekes G, Thiel HJ. 2016. Characterization of monoclonal antibodies against feline coronavirus accessory protein 7b. Vet Microbiol 184: 11-19.
  • Thiel V, Thiel HJ, Tekes G. 2014. Tackling feline infectious peritonitis via reverse genetics. Bioengineered 5: 396-400.
  • Bank-Wolf B, Stallkamp I, Wiese S, Moritz A, Tekes G, Thiel HJ. 2014. Mutations of 3c and spike protein genes correlate with the occurrence of feline infectious peritonitis. Vet Microbiol 173: 177-188.
  • Madhugiri R, Fricke M, Marz M, Ziebuhr J. 2014. RNA structure analysis of alphacoronavirus terminal genome regions. Virus Res 194: 76-89.
  • de Groot, R. J., S. C. Baker, R. Baric, L. Enjuanes, A. E. Gorbalenya, K. V. Holmes, S. Perlman, L. Poon, P. J. M. Rottier, P. J. Talbot, P. C. Y. Woo, and J. Ziebuhr. 2012. FamilyCoronaviridae, p. 806-828. In A. M. Q. King, M. J. Adams, E. B. Carstens, and E. J. Lefkowitz (ed.), Virus Taxonomy. Elsevier, Amsterdam.
  • Tekes, G., D. Spies., B. Bank-Wolf, V. Thiel and H.-J. Thiel. 2012. A reverse genetic approach to study feline infectious peritonitis. J Virol., JVI.00023-12 [pii], 10.1128/JVI.00023-12.
  • Lamp, B., C. Riedel, G. Roman-Sosa, M. Heimann, S. Jacobi, P. Becher, H.-J. Thiel and T. Rümenapf. 2011. Biosynthesis of classical Swine Fever virus nonstructural proteins. J. Virol. 84: 3607-3620.
  • Züst, R., L. Cervantes-Barragan, M. Habjan, R. Maier, B.W. Neuman, J. Ziebuhr, K.J. Szretter, S.C. Baker, W.  Barchet, M.S. Diamond, S.G. Siddell, B. Ludewig, and V. Thiel. 2011. Ribose 2′-O-methylation provides a molecular signature for the distinction of self and non-self mRNA dependent on the RNA sensor Mda5. Nature Immunol. 12: 137-143.
  • Tekes, G., R. Hofmann-Lehmann, B. Bank-Wolf, R. Maier, H.-J. Thiel and V. Thiel. 2010. Chimeric feline coronaviruses that encode type II spike protein on type I genetic background display accelerated viral growth and altered receptor usage. J. Virol. 84: 1326-1333.
  • Tekes, G., R. Hofmann, I. Stallkamp, V. Thiel and H.-J. Thiel. 2008. Genome organization and reverse genetic analysis of a type I feline coronavirus. J. Virol. 82: 1851-1859.
  • Ziebuhr, J. 2008. Coronavirus replicative proteins, p. 65-81. In S. Perlman, T. Gallagher, and E. J. Snijder (ed.), Nidoviruses. ASM Press, Washington, DC.
  • Putics, A., A. E. Gorbalenya, and J. Ziebuhr. 2006. Identification of protease and ADP-ribose 1“-monophosphatase activities associated with transmissible gastroenteritis virus non-structural protein 3. J. Gen. Virol. 87: 651-656.
  • Minskaia, E., T. Hertzig, A.E. Gorbalenya, V. Campanacci, C. Cambillau, B. Canard, J. Ziebuhr. 2006. Discovery of an RNA virus 3′-5′ exoribonuclease that is critically involved in coronavirus RNA synthesis. Proc. Natl. Acad. Sci. USA 103: 5108-5113.
  • Putics, A., W. Filipowicz, J. Hall, A.E. Gorbalenya, and J. Ziebuhr. 2005. ADP-ribose-1″-monophosphatase: a conserved coronavirus enzyme that is dispensable for viral replication in tissue culture. J. Virol. 79: 12721-12731.
  • Ivanov, K. A., T. Hertzig, M. Rozanov, S. Bayer, V. Thiel, A. E. Gorbalenya, and J. Ziebuhr. 2004. Major genetic marker of nidoviruses encodes a replicative endoribonuclease. Proc. Natl. Acad. Sci. USA 101: 12694-12699.
  • Conzelmann, K.-K., N. Visser, P. v. Woensel, H.-J.Thiel. 1993. Molecular characterization of porcine reproductive and respiratory syndrome virus, a member of the arterivirus group. Virology 193: 329-339.