137 results on '"Thomas Hoenen"'
Search Results
2. Ebola virus inclusion bodies are liquid organelles whose formation is facilitated by nucleoprotein oligomerization
- Author
-
Bianca S. Bodmer, Melina Vallbracht, Dmitry S. Ushakov, Lisa Wendt, Petr Chlanda, and Thomas Hoenen
- Subjects
Ebolavirus, inclusion bodies, intrinsically disordered region, liquid organelle, liquid–liquid phase separation ,Infectious and parasitic diseases ,RC109-216 ,Microbiology ,QR1-502 - Abstract
Viral RNA synthesis of several non-segmented, negative-sense RNA viruses (NNSVs) takes place in inclusion bodies (IBs) that show properties of liquid organelles, which are formed by liquid–liquid phase separation of scaffold proteins. It is believed that this is driven by intrinsically disordered regions (IDRs) and/or multiple copies of interaction domains, which for NNSVs are usually located in their nucleo – and phosphoproteins. In contrast to other NNSVs, the Ebola virus (EBOV) nucleoprotein NP alone is sufficient to form IBs without the need for a phosphoprotein, and to facilitate the recruitment of other viral proteins into these structures. While it has been proposed that also EBOV IBs are liquid organelles, this has so far not been formally demonstrated. Here we used a combination of live cell microscopy, fluorescence recovery after photobleaching assays, and mutagenesis approaches together with reverse genetics-based generation of recombinant viruses to study the formation of EBOV IBs. Our results demonstrate that EBOV IBs are indeed liquid organelles, and that oligomerization but not IDRs of the EBOV nucleoprotein plays a key role in their formation. Additionally, VP35 (often considered the phosphoprotein-equivalent of EBOV) is not essential for IB formation, but alters their liquid behaviour. These findings define the molecular mechanism for the formation of EBOV IBs, which play a central role in the life cycle of this deadly virus.
- Published
- 2023
- Full Text
- View/download PDF
3. N6-methyladenosine is required for efficient RNA synthesis of Ebola virus and other haemorrhagic fever viruses
- Author
-
Lisa Wendt, Matthew J. Pickin, Bianca S. Bodmer, Sven Reiche, Lucie Fénéant, Julia E. Hölper, Walter Fuchs, Allison Groseth, and Thomas Hoenen
- Subjects
Ebola virus ,Junín virus ,Crimean-Congo haemorrhagic fever virus ,filovirus ,arenavirus ,orthonairovirus ,Infectious and parasitic diseases ,RC109-216 ,Microbiology ,QR1-502 - Abstract
N6-methyladenosine (m6A) is one of the most abundant modifications of cellular RNA, where it serves various functions. m6A methylation of many viral RNA species has also been described; however, little is known about the m6A epitranscriptome of haemorrhagic fever-causing viruses like Ebola virus (EBOV). Here, we analysed the importance of the methyltransferase METTL3 for the life cycle of this virus. We found that METTL3 interacts with the EBOV nucleoprotein and the transcriptional activator VP30 to support viral RNA synthesis, and that METTL3 is recruited into EBOV inclusions bodies, where viral RNA synthesis occurs. Analysis of the m6A methylation pattern of EBOV mRNAs showed that they are methylated by METTL3. Further studies revealed that METTL3 interaction with the viral nucleoprotein, as well as its importance for RNA synthesis and protein expression, is also observed for other haemorrhagic fever viruses such as Junín virus (JUNV) and Crimean-Congo haemorrhagic fever virus (CCHFV). The negative effects on viral RNA synthesis due to loss of m6A methylation are independent of innate immune sensing, as METTL3 knockout did not affect type I interferon induction in response to viral RNA synthesis or infection. Our results suggest a novel function for m6A that is conserved among diverse haemorrhagic fever-causing viruses (i.e. EBOV, JUNV and CCHFV), making METTL3 a promising target for broadly-acting antivirals.
- Published
- 2023
- Full Text
- View/download PDF
4. A dsRNA-binding mutant reveals only a minor role of exonuclease activity in interferon antagonism by the arenavirus nucleoprotein
- Author
-
Patrick Bohn, Irke Waßmann, Lisa Wendt, Anne Leske, Thomas Hoenen, Birke A. Tews, and Allison Groseth
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
The arenavirus nucleoprotein (NP) plays an important role in the virus’ ability to block interferon (IFN) production, and its exonuclease function appears to contribute to this activity. However, efforts to analyze this contribution are complicated by the functional overlap between the exonuclease active site and a neighboring region involved in IKKε-binding and subsequent inhibition of IRF3 activation, which also plays an important role in IFN production. To circumvent this issue, we mutated a residue located away from the active site that is involved in binding of the dsRNA substrate being targeted for exonuclease digestion, i.e. H426A. We found that expression of Tacaribe virus (TCRV) NP containing this RNA-binding H426A mutation was still able to efficiently block IFN-β promoter activity in response to Sendai virus infection, despite being strongly impaired in its exonuclease activity. This was in contrast to a conventional exonuclease active site mutant (E388A), which was impaired with respect to both exonuclease activity and IFN antagonism. Importantly, growth of a recombinant virus encoding the RNA-binding mutation (rTCRV-H426A) was similar to wild-type in IFN-deficient cells, unlike the active site mutant (rTCRV-E388A), which was already markedly impaired in these cells. Further, in IFN-competent cells, the TCRV-H426A RNA-binding mutant showed more robust growth and delayed IFN-β mRNA upregulation compared to the TCRV-E388A active site mutant. Taken together, this novel mutational approach, which allows us to now dissect the different contributions of the NP exonuclease activity and IKKε-binding/IRF3 inhibition to IFN antagonism, clearly suggests that conventional exonuclease mutants targeting the active site overestimate the contribution of the exonuclease function, and that rather other IFN antagonistic functions of NP play the dominant role in IFN-antagonism. Author summary The ability to suppress interferon production plays an important role in the successful establishment of viral infection, and for arenaviruses, the exonuclease domain of the nucleoprotein is suggested to play an important role in this process. However, analyzing its contribution is challenging because mutations that disrupt the exonuclease active site also block antagonism of IRF3 activation, which is also important for interferon production. Here we report a mutation that instead targets binding of the exonuclease’s dsRNA substrate. As expected, this mutation strongly impaired exonuclease activity; however, unlike classical mutations targeting the exonuclease active site, it was still able to efficiently block interferon production. This suggests that data based on active site mutants overestimate the contribution of the NP exonuclease function to interferon antagonism. This advance in our understanding regarding the specific contributions of different NP functions to interferon antagonism has important implications for understanding arenavirus pathogenesis and the development of future treatment strategies.
- Published
- 2023
5. iPSC screening for drug repurposing identifies anti‐RNA virus agents modulating host cell susceptibility
- Author
-
Keiko Imamura, Yasuteru Sakurai, Takako Enami, Ran Shibukawa, Yohei Nishi, Akira Ohta, Tsugumine Shu, Jitsutaro Kawaguchi, Sayaka Okada, Thomas Hoenen, Jiro Yasuda, and Haruhisa Inoue
- Subjects
Ebola virus ,human iPSC ,PPARγ ,SARS‐CoV‐2 ,Sendai virus ,SERMs ,Biology (General) ,QH301-705.5 - Abstract
Human pathogenic RNA viruses are threats to public health because they are prone to escaping the human immune system through mutations of genomic RNA, thereby causing local outbreaks and global pandemics of emerging or re‐emerging viral diseases. While specific therapeutics and vaccines are being developed, a broad‐spectrum therapeutic agent for RNA viruses would be beneficial for targeting newly emerging and mutated RNA viruses. In this study, we conducted a screen of repurposed drugs using Sendai virus (an RNA virus of the family Paramyxoviridae), with human‐induced pluripotent stem cells (iPSCs) to explore existing drugs that may present anti‐RNA viral activity. Selected hit compounds were evaluated for their efficacy against two important human pathogens: Ebola virus (EBOV) using Huh7 cells and severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) using Vero E6 cells. Selective estrogen receptor modulators (SERMs), including raloxifene, exhibited antiviral activities against EBOV and SARS‐CoV‐2. Pioglitazone, a PPARγ agonist, also exhibited antiviral activities against SARS‐CoV‐2, and both raloxifene and pioglitazone presented a synergistic antiviral effect. Finally, we demonstrated that SERMs blocked entry steps of SARS‐CoV‐2 into host cells. These findings suggest that the identified FDA‐approved drugs can modulate host cell susceptibility against RNA viruses.
- Published
- 2021
- Full Text
- View/download PDF
6. TRIM25 and ZAP target the Ebola virus ribonucleoprotein complex to mediate interferon-induced restriction.
- Author
-
Rui Pedro Galão, Harry Wilson, Kristina L Schierhorn, Franka Debeljak, Bianca S Bodmer, Daniel Goldhill, Thomas Hoenen, Sam J Wilson, Chad M Swanson, and Stuart J D Neil
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Ebola virus (EBOV) causes highly pathogenic disease in primates. Through screening a library of human interferon-stimulated genes (ISGs), we identified TRIM25 as a potent inhibitor of EBOV transcription-and-replication-competent virus-like particle (trVLP) propagation. TRIM25 overexpression inhibited the accumulation of viral genomic and messenger RNAs independently of the RNA sensor RIG-I or secondary proinflammatory gene expression. Deletion of TRIM25 strongly attenuated the sensitivity of trVLPs to inhibition by type-I interferon. The antiviral activity of TRIM25 required ZAP and the effect of type-I interferon was modulated by the CpG dinucleotide content of the viral genome. We find that TRIM25 interacts with the EBOV vRNP, resulting in its autoubiquitination and ubiquitination of the viral nucleoprotein (NP). TRIM25 is recruited to incoming vRNPs shortly after cell entry and leads to dissociation of NP from the vRNA. We propose that TRIM25 targets the EBOV vRNP, exposing CpG-rich viral RNA species to restriction by ZAP.
- Published
- 2022
- Full Text
- View/download PDF
7. Assessment of Life Cycle Modeling Systems as Prediction Tools for a Possible Attenuation of Recombinant Ebola Viruses
- Author
-
Bianca S. Bodmer and Thomas Hoenen
- Subjects
Ebola virus ,filovirus ,reverse genetics ,life cycle modeling system ,minigenome system ,trVLP system ,Microbiology ,QR1-502 - Abstract
Ebola virus (EBOV) causes hemorrhagic fever in humans with high case fatality rates. In the past, a number of recombinant EBOVs expressing different reporters from additional transcription units or as fusion proteins have been rescued. These viruses are important tools for the study of EBOV, and their uses include high throughput screening approaches, the analysis of intercellular localization of viral proteins and of tissue distribution of viruses, and the study of pathogenesis in vivo. However, they all show, at least in vivo, attenuation compared to wild type virus, and the basis of this attenuation is only poorly understood. Unfortunately, rescue of these viruses is a lengthy and not always successful process, and working with them is restricted to biosafety level (BSL)-4 laboratories, so that the search for non-attenuated reporter-expressing EBOVs remains challenging. However, several life cycle modeling systems have been developed to mimic different aspects of the filovirus life cycle under BSL-1 or -2 conditions, but it remains unclear whether these systems can be used to predict the viability and possible attenuation of recombinant EBOVs. To address this question, we systematically fused N- or C-terminally either a flag-HA tag or a green fluorescent protein (GFP) to different EBOV proteins, and analyzed the impact of these additions with respect to protein function in life cycle modeling systems. Based on these results, selected recombinant EBOVs encoding these tags/proteins were then rescued and characterized for a possible attenuation in vitro, and results compared with data from the life cycle modeling systems. While the results for the small molecular tags showed mostly good concordance, GFP-expressing viruses were more attenuated than expected based on the results from the life cycle modeling system, demonstrating a limitation of these systems and emphasizing the importance of work with infectious virus. Nevertheless, life cycle modeling system remain useful tools to exclude non-viable tagging strategies.
- Published
- 2022
- Full Text
- View/download PDF
8. Virus–Host Cell Interactions
- Author
-
Thomas Hoenen and Allison Groseth
- Subjects
n/a ,Cytology ,QH573-671 - Abstract
As obligate intracellular parasites, viruses are intimately interconnected with their host cells [...]
- Published
- 2022
- Full Text
- View/download PDF
9. A genome-wide siRNA screen identifies a druggable host pathway essential for the Ebola virus life cycle
- Author
-
Scott Martin, Abhilash I. Chiramel, Marie Luisa Schmidt, Yu-Chi Chen, Nadia Whitt, Ari Watt, Eric C. Dunham, Kyle Shifflett, Shelby Traeger, Anne Leske, Eugen Buehler, Cynthia Martellaro, Janine Brandt, Lisa Wendt, Andreas Müller, Stephanie Peitsch, Sonja M. Best, Jürgen Stech, Stefan Finke, Angela Römer-Oberdörfer, Allison Groseth, Heinz Feldmann, and Thomas Hoenen
- Subjects
Ebola virus ,Carbamoyl phosphate synthetase 2 aspartate transcarbamylase and dihydroorotase ,De novo pyrimidine synthesis ,Dihydroorotate dehydrogenase ,Host factor ,Teriflunomide ,Medicine ,Genetics ,QH426-470 - Abstract
Abstract Background The 2014–2016 Ebola virus (EBOV) outbreak in West Africa highlighted the need for improved therapeutic options against this virus. Approaches targeting host factors/pathways essential for the virus are advantageous because they can potentially target a wide range of viruses, including newly emerging ones and because the development of resistance is less likely than when targeting the virus directly. However, systematic approaches for screening host factors important for EBOV have been hampered by the necessity to work with this virus at biosafety level 4 (BSL4). Methods In order to identify host factors involved in the EBOV life cycle, we performed a genome-wide siRNA screen comprising 64,755 individual siRNAs against 21,566 human genes to assess their activity in EBOV genome replication and transcription. As a screening platform, we used reverse genetics-based life cycle modelling systems that recapitulate these processes without the need for a BSL4 laboratory. Results Among others, we identified the de novo pyrimidine synthesis pathway as an essential host pathway for EBOV genome replication and transcription, and confirmed this using infectious EBOV under BSL4 conditions. An FDA-approved drug targeting this pathway showed antiviral activity against infectious EBOV, as well as other non-segmented negative-sense RNA viruses. Conclusions This study provides a minable data set for every human gene regarding its role in EBOV genome replication and transcription, shows that an FDA-approved drug targeting one of the identified pathways is highly efficacious in vitro, and demonstrates the power of life cycle modelling systems for conducting genome-wide host factor screens for BSL4 viruses.
- Published
- 2018
- Full Text
- View/download PDF
10. Immunization with GP1 but Not Core-like Particles Displaying Isolated Receptor-Binding Epitopes Elicits Virus-Neutralizing Antibodies against Junín Virus
- Author
-
Gleyder Roman-Sosa, Anne Leske, Xenia Ficht, Tung Huy Dau, Julia Holzerland, Thomas Hoenen, Martin Beer, Robert Kammerer, Reinhold Schirmbeck, Felix A. Rey, Sandra M. Cordo, and Allison Groseth
- Subjects
arenavirus ,Junín virus ,immune response ,neutralizing antibodies ,glycoprotein ,GP1 ,Medicine - Abstract
New World arenaviruses are rodent-transmitted viruses and include a number of pathogens that are responsible for causing severe human disease. This includes Junín virus (JUNV), which is the causative agent of Argentine hemorrhagic fever. The wild nature and mobility of the rodent reservoir host makes it difficult to control the disease, and currently passive immunization with high-titer neutralizing antibody-containing plasma from convalescent patients is the only specific therapy. However, dwindling supplies of naturally available convalescent plasma, and challenges in developing similar resources for other closely related viruses, have made the development of alternative antibody-based therapeutic approaches of critical importance. In this study, we sought to induce a neutralizing antibody response in rabbits against the receptor-binding subunit of the viral glycoprotein, GP1, and the specific peptide sequences in GP1 involved in cellular receptor contacts. While these specific receptor-interacting peptides did not efficiently induce the production of neutralizing antibodies when delivered as a particulate antigen (as part of hepatitis B virus core-like particles), we showed that recombinant JUNV GP1 purified from transfected mammalian cells induced virus-neutralizing antibodies at high titers in rabbits. Further, neutralization was observed across a range of unrelated JUNV strains, a feature that is critical for effectiveness in the field. These results underscore the potential of GP1 alone to induce a potent neutralizing antibody response and highlight the importance of epitope presentation. In addition, effective virus neutralization by rabbit antibodies supports the potential applicability of this species for the future development of immunotherapeutics (e.g., based on humanized monoclonal antibodies). Such information can be applied in the design of vaccines and immunogens for both prevention and specific therapies against this and likely also other closely related pathogenic New World arenaviruses.
- Published
- 2022
- Full Text
- View/download PDF
11. Interferon-Induced HERC5 Inhibits Ebola Virus Particle Production and Is Antagonized by Ebola Glycoprotein
- Author
-
Ermela Paparisto, Nina R. Hunt, Daniel S. Labach, Macon D. Coleman, Eric J. Di Gravio, Mackenzie J. Dodge, Nicole J. Friesen, Marceline Côté, Andreas Müller, Thomas Hoenen, and Stephen D. Barr
- Subjects
Ebola virus ,Marburg virus ,HERC5 ,antiviral ,interferon ,Cytology ,QH573-671 - Abstract
Survival following Ebola virus (EBOV) infection correlates with the ability to mount an early and robust interferon (IFN) response. The host IFN-induced proteins that contribute to controlling EBOV replication are not fully known. Among the top genes with the strongest early increases in expression after infection in vivo is IFN-induced HERC5. Using a transcription- and replication-competent VLP system, we showed that HERC5 inhibits EBOV virus-like particle (VLP) replication by depleting EBOV mRNAs. The HERC5 RCC1-like domain was necessary and sufficient for this inhibition and did not require zinc finger antiviral protein (ZAP). Moreover, we showed that EBOV (Zaire) glycoprotein (GP) but not Marburg virus GP antagonized HERC5 early during infection. Our data identify a novel ‘protagonist–antagonistic’ relationship between HERC5 and GP in the early stages of EBOV infection that could be exploited for the development of novel antiviral therapeutics.
- Published
- 2021
- Full Text
- View/download PDF
12. Ebola virus VP24 interacts with NP to facilitate nucleocapsid assembly and genome packaging
- Author
-
Logan Banadyga, Thomas Hoenen, Xavier Ambroggio, Eric Dunham, Allison Groseth, and Hideki Ebihara
- Subjects
Medicine ,Science - Abstract
Abstract Ebola virus causes devastating hemorrhagic fever outbreaks for which no approved therapeutic exists. The viral nucleocapsid, which is minimally composed of the proteins NP, VP35, and VP24, represents an attractive target for drug development; however, the molecular determinants that govern the interactions and functions of these three proteins are still unknown. Through a series of mutational analyses, in combination with biochemical and bioinformatics approaches, we identified a region on VP24 that was critical for its interaction with NP. Importantly, we demonstrated that the interaction between VP24 and NP was required for both nucleocapsid assembly and genome packaging. Not only does this study underscore the critical role that these proteins play in the viral replication cycle, but it also identifies a key interaction interface on VP24 that may serve as a novel target for antiviral therapeutic intervention.
- Published
- 2017
- Full Text
- View/download PDF
13. Ebola Virus Glycoprotein Domains Associated with Protective Efficacy
- Author
-
Bharti Bhatia, Wakako Furuyama, Thomas Hoenen, Heinz Feldmann, and Andrea Marzi
- Subjects
vesicular stomatitis virus ,VSV ,EBOV ,filovirus ,mucin-like domain ,glycan cap ,Medicine - Abstract
Ebola virus (EBOV) is the cause of sporadic outbreaks of human hemorrhagic disease in Africa, and the best-characterized virus in the filovirus family. The West African epidemic accelerated the clinical development of vaccines and therapeutics, leading to licensure of vaccines and antibody-based therapeutics for human use in recent years. The most widely used vaccine is based on vesicular stomatitis virus (VSV) expressing the EBOV glycoprotein (GP) (VSV-EBOV). Due to its favorable immune cell targeting, this vaccine has also been used as a base vector for the development of second generation VSV-based vaccines against Influenza, Nipah, and Zika viruses. However, in these situations, it may be beneficial if the immunogenicity against EBOV GP is minimized to induce a better protective immune response against the other foreign immunogen. Here, we analyzed if EBOV GP can be truncated to be less immunogenic, yet still able to drive replication of the vaccine vector. We found that the EBOV GP glycan cap and the mucin-like domain are both dispensable for VSV-EBOV replication. The glycan cap, however, appears critical for mediating a protective immune response against lethal EBOV challenge in mice.
- Published
- 2021
- Full Text
- View/download PDF
14. The Merits of Malaria Diagnostics during an Ebola Virus Disease Outbreak
- Author
-
Emmie de Wit, Darryl Falzarano, Clayton Onyango, Kyle Rosenke, Andrea Marzi, Melvin Ochieng, Bonventure Juma, Robert J. Fischer, Joseph B. Prescott, David Safronetz, Victor Omballa, Collins Owuor, Thomas Hoenen, Allison Groseth, Neeltje van Doremalen, Galina Zemtsova, Joshua Self, Trenton Bushmaker, Kristin McNally, Thomas Rowe, Shannon L. Emery, Friederike Feldmann, Brandi Williamson, Tolbert G. Nyenswah, Allen Grolla, James E. Strong, Gary Kobinger, Ute Stroeher, Mark Rayfield, Fatorma K. Bolay, Kathryn C. Zoon, Jorgen Stassijns, Livia Tampellini, Martin de Smet, Stuart T. Nichol, Barry Fields, Armand Sprecher, Heinz Feldmann, Moses Massaquoi, and Vincent J. Munster
- Subjects
Ebola ,disease outbreak ,malaria ,Plasmodium ,diagnostics ,PCR ,Medicine ,Infectious and parasitic diseases ,RC109-216 - Abstract
Malaria is a major public health concern in the countries affected by the Ebola virus disease epidemic in West Africa. We determined the feasibility of using molecular malaria diagnostics during an Ebola virus disease outbreak and report the incidence of Plasmodium spp. parasitemia in persons with suspected Ebola virus infection.
- Published
- 2016
- Full Text
- View/download PDF
15. Nanopore Sequencing as a Rapidly Deployable Ebola Outbreak Tool
- Author
-
Thomas Hoenen, Allison Groseth, Kyle Rosenke, Robert J. Fischer, Andreas Hoenen, Seth D. Judson, Cynthia Martellaro, Darryl Falzarano, Andrea Marzi, R. Burke Squires, Kurt R. Wollenberg, Emmie de Wit, Joseph B. Prescott, David Safronetz, Neeltje van Doremalen, Trenton Bushmaker, Friederike Feldmann, Kristin McNally, Fatorma K. Bolay, Barry Fields, Tara Sealy, Mark Rayfield, Stuart T. Nichol, Kathryn C. Zoon, Moses Massaquoi, Vincent J. Munster, and Heinz Feldmann
- Subjects
Ebola hemorrhagic fever ,Ebola virus ,Ebolavirus ,viruses ,high-throughput nucleotide sequencing ,nanopore sequencing ,Medicine ,Infectious and parasitic diseases ,RC109-216 - Abstract
Rapid sequencing of RNA/DNA from pathogen samples obtained during disease outbreaks provides critical scientific and public health information. However, challenges exist for exporting samples to laboratories or establishing conventional sequencers in remote outbreak regions. We successfully used a novel, pocket-sized nanopore sequencer at a field diagnostic laboratory in Liberia during the current Ebola virus outbreak.
- Published
- 2016
- Full Text
- View/download PDF
16. Differences in Viral RNA Synthesis but Not Budding or Entry Contribute to the In Vitro Attenuation of Reston Virus Compared to Ebola Virus
- Author
-
Bianca S. Bodmer, Josephin Greßler, Marie L. Schmidt, Julia Holzerland, Janine Brandt, Stefanie Braun, Allison Groseth, and Thomas Hoenen
- Subjects
Ebola virus ,Reston virus ,filovirus ,pathogenicity ,RNA synthesis ,budding ,Biology (General) ,QH301-705.5 - Abstract
Most filoviruses cause severe disease in humans. For example, Ebola virus (EBOV) is responsible for the two most extensive outbreaks of filovirus disease to date, with case fatality rates of 66% and 40%, respectively. In contrast, Reston virus (RESTV) is apparently apathogenic in humans, and while transmission of RESTV from domestic pigs to people results in seroconversion, no signs of disease have been reported in such cases. The determinants leading to these differences in pathogenicity are not well understood, but such information is needed in order to better evaluate the risks posed by the repeated spillover of RESTV into the human population and to perform risk assessments for newly emerging filoviruses with unknown pathogenic potential. Interestingly, RESTV and EBOV already show marked differences in their growth in vitro, with RESTV growing slower and reaching lower end titers. In order to understand the basis for this in vitro attenuation of RESTV, we used various life cycle modeling systems mimicking different aspects of the virus life cycle. Our results showed that viral RNA synthesis was markedly slower when using the ribonucleoprotein (RNP) components from RESTV, rather than those for EBOV. In contrast, the kinetics of budding and entry were indistinguishable between these two viruses. These data contribute to our understanding of the molecular basis for filovirus pathogenicity by showing that it is primarily differences in the robustness of RNA synthesis by the viral RNP complex that are responsible for the impaired growth of RESTV in tissue culture.
- Published
- 2020
- Full Text
- View/download PDF
17. The Cellular Protein CAD is Recruited into Ebola Virus Inclusion Bodies by the Nucleoprotein NP to Facilitate Genome Replication and Transcription
- Author
-
Janine Brandt, Lisa Wendt, Bianca S. Bodmer, Thomas C. Mettenleiter, and Thomas Hoenen
- Subjects
Ebola virus ,filovirus ,inclusion bodies ,CAD ,pyrimidine synthesis ,Cytology ,QH573-671 - Abstract
Ebola virus (EBOV) is a zoonotic pathogen causing severe hemorrhagic fevers in humans and non-human primates with high case fatality rates. In recent years, the number and extent of outbreaks has increased, highlighting the importance of better understanding the molecular aspects of EBOV infection and host cell interactions to control this virus more efficiently. Many viruses, including EBOV, have been shown to recruit host proteins for different viral processes. Based on a genome-wide siRNA screen, we recently identified the cellular host factor carbamoyl-phosphate synthetase 2, aspartate transcarbamylase, and dihydroorotase (CAD) as being involved in EBOV RNA synthesis. However, mechanistic details of how this host factor plays a role in the EBOV life cycle remain elusive. In this study, we analyzed the functional and molecular interactions between EBOV and CAD. To this end, we used siRNA knockdowns in combination with various reverse genetics-based life cycle modelling systems and additionally performed co-immunoprecipitation and co-immunofluorescence assays to investigate the influence of CAD on individual aspects of the EBOV life cycle and to characterize the interactions of CAD with viral proteins. Following this approach, we could demonstrate that CAD directly interacts with the EBOV nucleoprotein NP, and that NP is sufficient to recruit CAD into inclusion bodies dependent on the glutaminase (GLN) domain of CAD. Further, siRNA knockdown experiments indicated that CAD is important for both viral genome replication and transcription, while substrate rescue experiments showed that the function of CAD in pyrimidine synthesis is indeed required for those processes. Together, this suggests that NP recruits CAD into inclusion bodies via its GLN domain in order to provide pyrimidines for EBOV genome replication and transcription. These results define a novel mechanism by which EBOV hijacks host cell pathways in order to facilitate genome replication and transcription and provide a further basis for the development of host-directed broad-spectrum antivirals.
- Published
- 2020
- Full Text
- View/download PDF
18. Characterization of the catalytic center of the Ebola virus L polymerase.
- Author
-
Marie Luisa Schmidt and Thomas Hoenen
- Subjects
Arctic medicine. Tropical medicine ,RC955-962 ,Public aspects of medicine ,RA1-1270 - Abstract
Ebola virus (EBOV) causes a severe hemorrhagic fever in humans and non-human primates. While no licensed therapeutics are available, recently there has been tremendous progress in developing antivirals. Targeting the ribonucleoprotein complex (RNP) proteins, which facilitate genome replication and transcription, and particularly the polymerase L, is a promising antiviral approach since these processes are essential for the virus life cycle. However, until now little is known about L in terms of its structure and function, and in particular the catalytic center of the RNA-dependent RNA polymerase (RdRp) of L, which is one of the most promising molecular targets, has never been experimentally characterized.Using multiple sequence alignments with other negative sense single-stranded RNA viruses we identified the putative catalytic center of the EBOV RdRp. An L protein with mutations in this center was then generated and characterized using various life cycle modelling systems. These systems are based on minigenomes, i.e. miniature versions of the viral genome, in which the viral genes are exchanged against a reporter gene. When such minigenomes are coexpressed with RNP proteins in mammalian cells, the RNP proteins recognize them as authentic templates for replication and transcription, resulting in reporter activity reflecting these processes. Replication-competent minigenome systems indicated that our L catalytic domain mutant was impaired in genome replication and/or transcription, and by using replication-deficient minigenome systems, as well as a novel RT-qPCR-based genome replication assay, we showed that it indeed no longer supported either of these processes. However, it still showed similar expression to wild-type L, and retained its ability to be incorporated into inclusion bodies, which are the sites of EBOV genome replication.We have experimentally defined the catalytic center of the EBOV RdRp, and thus a promising antiviral target regulating an essential aspect of the EBOV life cycle.
- Published
- 2017
- Full Text
- View/download PDF
19. The phosphatidylinositol-3-phosphate 5-kinase inhibitor apilimod blocks filoviral entry and infection.
- Author
-
Elizabeth A Nelson, Julie Dyall, Thomas Hoenen, Alyson B Barnes, Huanying Zhou, Janie Y Liang, Julia Michelotti, William H Dewey, Lisa Evans DeWald, Richard S Bennett, Patrick J Morris, Rajarshi Guha, Carleen Klumpp-Thomas, Crystal McKnight, Yu-Chi Chen, Xin Xu, Amy Wang, Emma Hughes, Scott Martin, Craig Thomas, Peter B Jahrling, Lisa E Hensley, Gene G Olinger, and Judith M White
- Subjects
Arctic medicine. Tropical medicine ,RC955-962 ,Public aspects of medicine ,RA1-1270 - Abstract
Phosphatidylinositol-3-phosphate 5-kinase (PIKfyve) is a lipid kinase involved in endosome maturation that emerged from a haploid genetic screen as being required for Ebola virus (EBOV) infection. Here we analyzed the effects of apilimod, a PIKfyve inhibitor that was reported to be well tolerated in humans in phase 2 clinical trials, for its effects on entry and infection of EBOV and Marburg virus (MARV). We first found that apilimod blocks infections by EBOV and MARV in Huh 7, Vero E6 and primary human macrophage cells, with notable potency in the macrophages (IC50, 10 nM). We next observed that similar doses of apilimod block EBOV-glycoprotein-virus like particle (VLP) entry and transcription-replication competent VLP infection, suggesting that the primary mode of action of apilimod is as an entry inhibitor, preventing release of the viral genome into the cytoplasm to initiate replication. After providing evidence that the anti-EBOV action of apilimod is via PIKfyve, we showed that it blocks trafficking of EBOV VLPs to endolysosomes containing Niemann-Pick C1 (NPC1), the intracellular receptor for EBOV. Concurrently apilimod caused VLPs to accumulate in early endosome antigen 1-positive endosomes. We did not detect any effects of apilimod on bulk endosome acidification, on the activity of cathepsins B and L, or on cholesterol export from endolysosomes. Hence by antagonizing PIKfyve, apilimod appears to block EBOV trafficking to its site of fusion and entry into the cytoplasm. Given the drug's observed anti-filoviral activity, relatively unexplored mechanism of entry inhibition, and reported tolerability in humans, we propose that apilimod be further explored as part of a therapeutic regimen to treat filoviral infections.
- Published
- 2017
- Full Text
- View/download PDF
20. The Ebola Virus Nucleoprotein Recruits the Nuclear RNA Export Factor NXF1 into Inclusion Bodies to Facilitate Viral Protein Expression
- Author
-
Lisa Wendt, Janine Brandt, Bianca S. Bodmer, Sven Reiche, Marie Luisa Schmidt, Shelby Traeger, and Thomas Hoenen
- Subjects
ebola virus ,filovirus ,inclusion bodies ,nxf1 ,liquid organelles ,mrna export ,Cytology ,QH573-671 - Abstract
Ebola virus (EBOV) causes severe outbreaks of viral hemorrhagic fever in humans. While virus-host interactions are promising targets for antivirals, there is only limited knowledge regarding the interactions of EBOV with cellular host factors. Recently, we performed a genome-wide siRNA screen that identified the nuclear RNA export factor 1 (NXF1) as an important host factor for the EBOV life cycle. NXF1 is a major component of the nuclear mRNA export pathway that is usurped by many viruses whose life cycles include nuclear stages. However, the role of NXF1 in the life cycle of EBOV, a virus replicating in cytoplasmic inclusion bodies, remains unknown. In order to better understand the role of NXF1 in the EBOV life cycle, we performed a combination of co-immunoprecipitation and double immunofluorescence assays to characterize the interactions of NXF1 with viral proteins and RNAs. Additionally, using siRNA-mediated knockdown of NXF1 together with functional assays, we analyzed the role of NXF1 in individual aspects of the virus life cycle. With this approach we identified the EBOV nucleoprotein (NP) as a viral interaction partner of NXF1. Further studies revealed that NP interacts with the RNA-binding domain of NXF1 and competes with RNA for this interaction. Co-localization studies showed that RNA binding-deficient, but not wildtype NXF1, accumulates in NP-derived inclusion bodies, and knockdown experiments demonstrated that NXF1 is necessary for viral protein expression, but not for viral RNA synthesis. Finally, our results showed that NXF1 interacts with viral mRNAs, but not with viral genomic RNAs. Based on these results we suggest a model whereby NXF1 is recruited into inclusion bodies to promote the export of viral mRNA:NXF1 complexes from these sites. This would represent a novel function for NXF1 in the life cycle of cytoplasmically replicating viruses, and may provide a basis for new therapeutic approaches against EBOV, and possibly other emerging viruses.
- Published
- 2020
- Full Text
- View/download PDF
21. Filovirus RefSeq Entries: Evaluation and Selection of Filovirus Type Variants, Type Sequences, and Names
- Author
-
Jens H. Kuhn, Kristian G. Andersen, Yīmíng Bào, Sina Bavari, Stephan Becker, Richard S. Bennett, Nicholas H. Bergman, Olga Blinkova, Steven Bradfute, J. Rodney Brister, Alexander Bukreyev, Kartik Chandran, Alexander A. Chepurnov, Robert A. Davey, Ralf G. Dietzgen, Norman A. Doggett, Olga Dolnik, John M. Dye, Sven Enterlein, Paul W. Fenimore, Pierre Formenty, Alexander N. Freiberg, Robert F. Garry, Nicole L. Garza, Stephen K. Gire, Jean-Paul Gonzalez, Anthony Griffiths, Christian T. Happi, Lisa E. Hensley, Andrew S. Herbert, Michael C. Hevey, Thomas Hoenen, Anna N. Honko, Georgy M. Ignatyev, Peter B. Jahrling, Joshua C. Johnson, Karl M. Johnson, Jason Kindrachuk, Hans-Dieter Klenk, Gary Kobinger, Tadeusz J. Kochel, Matthew G. Lackemeyer, Daniel F. Lackner, Eric M. Leroy, Mark S. Lever, Elke Mühlberger, Sergey V. Netesov, Gene G. Olinger, Sunday A. Omilabu, Gustavo Palacios, Rekha G. Panchal, Daniel J. Park, Jean L. Patterson, Janusz T. Paweska, Clarence J. Peters, James Pettitt, Louise Pitt, Sheli R. Radoshitzky, Elena I. Ryabchikova, Erica Ollmann Saphire, Pardis C. Sabeti, Rachel Sealfon, Aleksandr M. Shestopalov, Sophie J. Smither, Nancy J. Sullivan, Robert Swanepoel, Ayato Takada, Jonathan S. Towner, Guido van der Groen, Viktor E. Volchkov, Valentina A. Volchkova, Victoria Wahl-Jensen, Travis K. Warren, Kelly L. Warfield, Manfred Weidmann, and Stuart T. Nichol
- Subjects
Bundibugyo virus ,cDNA clone ,cuevavirus ,Ebola ,Ebola virus ,ebolavirus ,filovirid ,Filoviridae ,filovirus ,genome annotation ,ICTV ,International Committee on Taxonomy of Viruses ,Lloviu virus ,Marburg virus ,marburgvirus ,mononegavirad ,Mononegavirales ,mononegavirus ,Ravn virus ,RefSeq ,Reston virus ,reverse genetics ,Sudan virus ,Taï Forest virus ,virus classification ,virus isolate ,virus nomenclature ,virus strain ,virus taxonomy ,virus variant ,Microbiology ,QR1-502 - Abstract
Sequence determination of complete or coding-complete genomes of viruses is becoming common practice for supporting the work of epidemiologists, ecologists, virologists, and taxonomists. Sequencing duration and costs are rapidly decreasing, sequencing hardware is under modification for use by non-experts, and software is constantly being improved to simplify sequence data management and analysis. Thus, analysis of virus disease outbreaks on the molecular level is now feasible, including characterization of the evolution of individual virus populations in single patients over time. The increasing accumulation of sequencing data creates a management problem for the curators of commonly used sequence databases and an entry retrieval problem for end users. Therefore, utilizing the data to their fullest potential will require setting nomenclature and annotation standards for virus isolates and associated genomic sequences. The National Center for Biotechnology Information’s (NCBI’s) RefSeq is a non-redundant, curated database for reference (or type) nucleotide sequence records that supplies source data to numerous other databases. Building on recently proposed templates for filovirus variant naming [ ()////-], we report consensus decisions from a majority of past and currently active filovirus experts on the eight filovirus type variants and isolates to be represented in RefSeq, their final designations, and their associated sequences.
- Published
- 2014
- Full Text
- View/download PDF
22. Detection of Viral RNA in Tissues following Plasma Clearance from an Ebola Virus Infected Patient.
- Author
-
Mirella Biava, Claudia Caglioti, Licia Bordi, Concetta Castilletti, Francesca Colavita, Serena Quartu, Emanuele Nicastri, Francesco Nicola Lauria, Nicola Petrosillo, Simone Lanini, Thomas Hoenen, Gary Kobinger, Alimuddin Zumla, Antonino Di Caro, Giuseppe Ippolito, Maria Rosaria Capobianchi, and Eleonora Lalle
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
An unprecedented Ebola virus (EBOV) epidemic occurred in 2013-2016 in West Africa. Over this time the epidemic exponentially grew and moved to Europe and North America, with several imported cases and many Health Care Workers (HCW) infected. Better understanding of EBOV infection patterns in different body compartments is mandatory to develop new countermeasures, as well as to fully comprehend the pathways of human-to-human transmission. We have longitudinally explored the persistence of EBOV-specific negative sense genomic RNA (neg-RNA) and the presence of positive sense RNA (pos-RNA), including both replication intermediate (antigenomic-RNA) and messenger RNA (mRNA) molecules, in the upper and lower respiratory tract, as compared to plasma, in a HCW infected with EBOV in Sierra Leone, who was hospitalized in the high isolation facility of the National Institute for Infectious Diseases "Lazzaro Spallanzani" (INMI), Rome, Italy. We observed persistence of pos-RNA and neg-RNAs in longitudinally collected specimens of the lower respiratory tract, even after viral clearance from plasma, suggesting possible local replication. The purpose of the present study is to enhance the knowledge on the biological features of EBOV that can contribute to the human-to-human transmissibility and to develop effective intervention strategies. However, further investigation is needed in order to better understand the clinical meaning of viral replication and shedding in the respiratory tract.
- Published
- 2017
- Full Text
- View/download PDF
23. Enhanced light microscopy visualization of virus particles from Zika virus to filamentous ebolaviruses.
- Author
-
George G Daaboul, David S Freedman, Steven M Scherr, Erik Carter, Alexandru Rosca, David Bernstein, Chad E Mire, Krystle N Agans, Thomas Hoenen, Thomas W Geisbert, M Selim Ünlü, and John H Connor
- Subjects
Medicine ,Science - Abstract
Light microscopy is a powerful tool in the detection and analysis of parasites, fungi, and prokaryotes, but has been challenging to use for the detection of individual virus particles. Unlabeled virus particles are too small to be visualized using standard visible light microscopy. Characterization of virus particles is typically performed using higher resolution approaches such as electron microscopy or atomic force microscopy. These approaches require purification of virions away from their normal millieu, requiring significant levels of expertise, and can only enumerate small numbers of particles per field of view. Here, we utilize a visible light imaging approach called Single Particle Interferometric Reflectance Imaging Sensor (SP-IRIS) that allows automated counting and sizing of thousands of individual virions. Virions are captured directly from complex solutions onto a silicon chip and then detected using a reflectance interference imaging modality. We show that the use of different imaging wavelengths allows the visualization of a multitude of virus particles. Using Violet/UV illumination, the SP-IRIS technique is able to detect individual flavivirus particles (~40 nm), while green light illumination is capable of identifying and discriminating between vesicular stomatitis virus and vaccinia virus (~360 nm). Strikingly, the technology allows the clear identification of filamentous infectious ebolavirus particles and virus-like particles. The ability to differentiate and quantify unlabeled virus particles extends the usefulness of traditional light microscopy and can be embodied in a straightforward benchtop approach allowing widespread applications ranging from rapid detection in biological fluids to analysis of virus-like particles for vaccine development and production.
- Published
- 2017
- Full Text
- View/download PDF
24. To be or not to be phosphorylated: understanding the role of Ebola virus nucleoprotein in the dynamic interplay with the transcriptional activator VP30 and the host phosphatase PP2A-B56
- Author
-
Lennart Kämper, Ida Kuhl, Melina Vallbracht, Thomas Hoenen, Uwe Linne, Axel Weber, Petr Chlanda, Michael Kracht, and Nadine Biedenkopf
- Subjects
Ebola virus ,nucleoprotein ,VP30 ,host phosphatase PP2A ,viral transcription ,phosphorylation ,Infectious and parasitic diseases ,RC109-216 ,Microbiology ,QR1-502 - Abstract
Ebola virus (EBOV) transcription is essentially regulated via dynamic dephosphorylation of its viral transcription activator VP30 by the host phosphatase PP2A. The nucleoprotein NP has emerged as a third key player in the regulation of this process by recruiting both the regulatory subunit B56 of PP2A and its substrate VP30 to initiate VP30 dephosphorylation and hence viral transcription. Both binding sites are located in close proximity to each other in NP’s C-terminal-disordered region. This study investigates NP’s role in VP30 dephosphorylation and transcription activation, focussing on the spatial requirements of NP’s binding sites. Increasing the distance between PP2A-B56 and VP30 at the NP interface revealed that close spatial and orientational contact is necessary for efficient VP30 dephosphorylation and viral transcription. Longer distances were lethal for recombinant EBOV except when a compensatory mutation, NP-T603I, occurred. This mutation, located between the NP binding sites for PP2A-B56 and VP30, fully restored functionality. Mass spectrometry showed that T603 is phosphorylated in recEBOV-NPwt virions. Mutational analysis indicated that T603I facilitates VP30 dephosphorylation in otherwise lethal recEBOV and that dynamic phosphorylation of NP-T603 is important for efficient primary viral transcription in the WT context. These findings emphasize the critical and evolutionarily pressured interplay between VP30 and PP2A-B56 within the NP C-terminal-disordered region and highlight the important role of NP on the regulation of viral transcription during the EBOV life cycle.
- Published
- 2025
- Full Text
- View/download PDF
25. Sequencing of Ebola Virus Genomes Using Nanopore Technology
- Author
-
Thomas Hoenen
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Sequencing of virus genomes during disease outbreaks can provide valuable information for diagnostics, epidemiology, and evaluation of potential countermeasures. However, particularly in remote areas logistical and technical challenges can be significant. Nanopore sequencing provides an alternative to classical Sanger and next-generation sequencing methods, and was successfully used under outbreak conditions (Hoenen et al., 2016; Quick et al., 2016). Here we describe a protocol used for sequencing of Ebola virus under outbreak conditions using Nanopore technology, which we successfully implemented at the CDC/NIH diagnostic laboratory (de Wit et al., 2016) located at the ELWA-3 Ebola virus Treatment Unit in Monrovia, Liberia, during the recent Ebola virus outbreak in West Africa.
- Published
- 2016
- Full Text
- View/download PDF
26. A Rapid Screening Assay Identifies Monotherapy with Interferon-ß and Combination Therapies with Nucleoside Analogs as Effective Inhibitors of Ebola Virus.
- Author
-
Stephen D S McCarthy, Beata Majchrzak-Kita, Trina Racine, Hannah N Kozlowski, Darren P Baker, Thomas Hoenen, Gary P Kobinger, Eleanor N Fish, and Donald R Branch
- Subjects
Arctic medicine. Tropical medicine ,RC955-962 ,Public aspects of medicine ,RA1-1270 - Abstract
To date there are no approved antiviral drugs for the treatment of Ebola virus disease (EVD). While a number of candidate drugs have shown limited efficacy in vitro and/or in non-human primate studies, differences in experimental methodologies make it difficult to compare their therapeutic effectiveness. Using an in vitro model of Ebola Zaire replication with transcription-competent virus like particles (trVLPs), requiring only level 2 biosafety containment, we compared the activities of the type I interferons (IFNs) IFN-α and IFN-ß, a panel of viral polymerase inhibitors (lamivudine (3TC), zidovudine (AZT) tenofovir (TFV), favipiravir (FPV), the active metabolite of brincidofovir, cidofovir (CDF)), and the estrogen receptor modulator, toremifene (TOR), in inhibiting viral replication in dose-response and time course studies. We also tested 28 two- and 56 three-drug combinations against Ebola replication. IFN-α and IFN-ß inhibited viral replication 24 hours post-infection (IC50 0.038μM and 0.016μM, respectively). 3TC, AZT and TFV inhibited Ebola replication when used alone (50-62%) or in combination (87%). They exhibited lower IC50 (0.98-6.2μM) compared with FPV (36.8μM), when administered 24 hours post-infection. Unexpectedly, CDF had a narrow therapeutic window (6.25-25μM). When dosed >50μM, CDF treatment enhanced viral infection. IFN-ß exhibited strong synergy with 3TC (97.3% inhibition) or in triple combination with 3TC and AZT (95.8% inhibition). This study demonstrates that IFNs and viral polymerase inhibitors may have utility in EVD. We identified several 2 and 3 drug combinations with strong anti-Ebola activity, confirmed in studies using fully infectious ZEBOV, providing a rationale for testing combination therapies in animal models of lethal Ebola challenge. These studies open up new possibilities for novel therapeutic options, in particular combination therapies, which could prevent and treat Ebola infection and potentially reduce drug resistance.
- Published
- 2016
- Full Text
- View/download PDF
27. Ebola virus RNA editing depends on the primary editing site sequence and an upstream secondary structure.
- Author
-
Masfique Mehedi, Thomas Hoenen, Shelly Robertson, Stacy Ricklefs, Michael A Dolan, Travis Taylor, Darryl Falzarano, Hideki Ebihara, Stephen F Porcella, and Heinz Feldmann
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Ebolavirus (EBOV), the causative agent of a severe hemorrhagic fever and a biosafety level 4 pathogen, increases its genome coding capacity by producing multiple transcripts encoding for structural and nonstructural glycoproteins from a single gene. This is achieved through RNA editing, during which non-template adenosine residues are incorporated into the EBOV mRNAs at an editing site encoding for 7 adenosine residues. However, the mechanism of EBOV RNA editing is currently not understood. In this study, we report for the first time that minigenomes containing the glycoprotein gene editing site can undergo RNA editing, thereby eliminating the requirement for a biosafety level 4 laboratory to study EBOV RNA editing. Using a newly developed dual-reporter minigenome, we have characterized the mechanism of EBOV RNA editing, and have identified cis-acting sequences that are required for editing, located between 9 nt upstream and 9 nt downstream of the editing site. Moreover, we show that a secondary structure in the upstream cis-acting sequence plays an important role in RNA editing. EBOV RNA editing is glycoprotein gene-specific, as a stretch encoding for 7 adenosine residues located in the viral polymerase gene did not serve as an editing site, most likely due to an absence of the necessary cis-acting sequences. Finally, the EBOV protein VP30 was identified as a trans-acting factor for RNA editing, constituting a novel function for this protein. Overall, our results provide novel insights into the RNA editing mechanism of EBOV, further understanding of which might result in novel intervention strategies against this viral pathogen.
- Published
- 2013
- Full Text
- View/download PDF
28. An upstream open reading frame modulates ebola virus polymerase translation and virus replication.
- Author
-
Reed S Shabman, Thomas Hoenen, Allison Groseth, Omar Jabado, Jennifer M Binning, Gaya K Amarasinghe, Heinz Feldmann, and Christopher F Basler
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Ebolaviruses, highly lethal zoonotic pathogens, possess longer genomes than most other non-segmented negative-strand RNA viruses due in part to long 5' and 3' untranslated regions (UTRs) present in the seven viral transcriptional units. To date, specific functions have not been assigned to these UTRs. With reporter assays, we demonstrated that the Zaire ebolavirus (EBOV) 5'-UTRs lack internal ribosomal entry site function. However, the 5'-UTRs do differentially regulate cap-dependent translation when placed upstream of a GFP reporter gene. Most dramatically, the 5'-UTR derived from the viral polymerase (L) mRNA strongly suppressed translation of GFP compared to a β-actin 5'-UTR. The L 5'-UTR is one of four viral genes to possess upstream AUGs (uAUGs), and ablation of each uAUG enhanced translation of the primary ORF (pORF), most dramatically in the case of the L 5'-UTR. The L uAUG was sufficient to initiate translation, is surrounded by a "weak" Kozak sequence and suppressed pORF translation in a position-dependent manner. Under conditions where eIF2α was phosphorylated, the presence of the uORF maintained translation of the L pORF, indicating that the uORF modulates L translation in response to cellular stress. To directly address the role of the L uAUG in virus replication, a recombinant EBOV was generated in which the L uAUG was mutated to UCG. Strikingly, mutating two nucleotides outside of previously-defined protein coding and cis-acting regulatory sequences attenuated virus growth to titers 10-100-fold lower than a wild-type virus in Vero and A549 cells. The mutant virus also exhibited decreased viral RNA synthesis as early as 6 hours post-infection and enhanced sensitivity to the stress inducer thapsigargin. Cumulatively, these data identify novel mechanisms by which EBOV regulates its polymerase expression, demonstrate their relevance to virus replication and identify a potential therapeutic target.
- Published
- 2013
- Full Text
- View/download PDF
29. Clomiphene and Its Isomers Block Ebola Virus Particle Entry and Infection with Similar Potency: Potential Therapeutic Implications
- Author
-
Elizabeth A. Nelson, Alyson B. Barnes, Ronald D. Wiehle, Gregory K. Fontenot, Thomas Hoenen, and Judith M. White
- Subjects
Ebola ,filovirus ,enclomiphene ,zuclomiphene ,anti-viral ,Ebola Virus disease ,Ebola virus survivors ,Microbiology ,QR1-502 - Abstract
The 2014 outbreak of Ebola virus (EBOV) in Western Africa highlighted the need for anti-EBOV therapeutics. Clomiphene is a U.S. Food and Drug Administration (FDA)-approved drug that blocks EBOV entry and infection in cells and significantly protects EBOV-challenged mice. As provided, clomiphene is, approximately, a 60:40 mixture of two stereoisomers, enclomiphene and zuclomiphene. The pharmacokinetic properties of the two isomers vary, but both accumulate in the eye and male reproductive tract, tissues in which EBOV can persist. Here we compared the ability of clomiphene and its isomers to inhibit EBOV using viral-like particle (VLP) entry and transcription/replication-competent VLP (trVLP) assays. Clomiphene and its isomers inhibited the entry and infection of VLPs and trVLPs with similar potencies. This was demonstrated with VLPs bearing the glycoproteins from three filoviruses (EBOV Mayinga, EBOV Makona, and Marburg virus) and in two cell lines (293T/17 and Vero E6). Visual problems have been noted in EBOV survivors, and viral RNA has been isolated from semen up to nine months post-infection. Since the clomiphene isomers accumulate in these affected tissues, clomiphene or one of its isomers warrants consideration as an anti-EBOV agent, for example, to potentially help ameliorate symptoms in EBOV survivors.
- Published
- 2016
- Full Text
- View/download PDF
30. The Ebola virus glycoprotein contributes to but is not sufficient for virulence in vivo.
- Author
-
Allison Groseth, Andrea Marzi, Thomas Hoenen, Astrid Herwig, Don Gardner, Stephan Becker, Hideki Ebihara, and Heinz Feldmann
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Among the Ebola viruses most species cause severe hemorrhagic fever in humans; however, Reston ebolavirus (REBOV) has not been associated with human disease despite numerous documented infections. While the molecular basis for this difference remains unclear, in vitro evidence has suggested a role for the glycoprotein (GP) as a major filovirus pathogenicity factor, but direct evidence for such a role in the context of virus infection has been notably lacking. In order to assess the role of GP in EBOV virulence, we have developed a novel reverse genetics system for REBOV, which we report here. Together with a previously published full-length clone for Zaire ebolavirus (ZEBOV), this provides a unique possibility to directly investigate the role of an entire filovirus protein in pathogenesis. To this end we have generated recombinant ZEBOV (rZEBOV) and REBOV (rREBOV), as well as chimeric viruses in which the glycoproteins from these two virus species have been exchanged (rZEBOV-RGP and rREBOV-ZGP). All of these viruses could be rescued and the chimeras replicated with kinetics similar to their parent virus in tissue culture, indicating that the exchange of GP in these chimeric viruses is well tolerated. However, in a mouse model of infection rZEBOV-RGP demonstrated markedly decreased lethality and prolonged time to death when compared to rZEBOV, confirming that GP does indeed contribute to the full expression of virulence by ZEBOV. In contrast, rREBOV-ZGP did not show any signs of virulence, and was in fact slightly attenuated compared to rREBOV, demonstrating that GP alone is not sufficient to confer a lethal phenotype or exacerbate disease in this model. Thus, while these findings provide direct evidence that GP contributes to filovirus virulence in vivo, they also clearly indicate that other factors are needed for the acquisition of full virulence.
- Published
- 2012
- Full Text
- View/download PDF
31. Tacaribe virus but not junin virus infection induces cytokine release from primary human monocytes and macrophages.
- Author
-
Allison Groseth, Thomas Hoenen, Michaela Weber, Svenja Wolff, Astrid Herwig, Andreas Kaufmann, and Stephan Becker
- Subjects
Arctic medicine. Tropical medicine ,RC955-962 ,Public aspects of medicine ,RA1-1270 - Abstract
The mechanisms underlying the development of disease during arenavirus infection are poorly understood. However, common to all hemorrhagic fever diseases is the involvement of macrophages as primary target cells, suggesting that the immune response in these cells may be of paramount importance during infection. Thus, in order to identify features of the immune response that contribute to arenavirus pathogenesis, we have examined the growth kinetics and cytokine profiles of two closely related New World arenaviruses, the apathogenic Tacaribe virus (TCRV) and the hemorrhagic fever-causing Junin virus (JUNV), in primary human monocytes and macrophages. Both viruses grew robustly in VeroE6 cells; however, TCRV titres were decreased by approximately 10 fold compared to JUNV in both monocytes and macrophages. Infection of both monocytes and macrophages with TCRV also resulted in the release of high levels of IL-6, IL-10 and TNF-α, while levels of IFN-α, IFN-β and IL-12 were not affected. However, we could show that the presence of these cytokines had no direct effect on growth of either TCRV of JUNV in macrophages. Further analysis also showed that while the production of IL-6 and IL-10 are dependent on viral replication, production of TNF-α also occurs after exposure to UV-inactivated TCRV particles and is thus independent of productive virus infection. Surprisingly, JUNV infection did not have an effect on any of the cytokines examined indicating that, in contrast to other viral hemorrhagic fever viruses, macrophage-derived cytokine production is unlikely to play an active role in contributing to the cytokine dysregulation observed in JUNV infected patients. Rather, these results suggest that an early, controlled immune response by infected macrophages may be critical for the successful control of infection of apathogenic viruses and prevention of subsequent disease, including systemic cytokine dysregulation.
- Published
- 2011
- Full Text
- View/download PDF
32. Reverse Genetiksysteme zur Erforschung von Ebola-Viren
- Author
-
Thomas Hoenen and Allison Groseth
- Subjects
Molecular Biology ,Biotechnology - Abstract
Ebolaviruses are among the deadliest viruses known, and work with infectious virus is restricted to laboratories of the highest biosafety level (i. e. BSL4). To facilitate research on ebolaviruses, also for researchers without access to BSL4 laboratories, reverse genetics-based life cycle modelling systems have been developed. Here, we describe how these system work, provide examples of their applications, and discuss their advantages and limitations.
- Published
- 2023
- Full Text
- View/download PDF
33. The Ebola virus VP40 matrix layer undergoes endosomal disassembly essential for membrane fusion
- Author
-
Sophie L Winter, Gonen Golani, Fabio Lolicato, Melina Vallbracht, Keerthihan Thiyagarajah, Samy Sid Ahmed, Christian Lüchtenborg, Oliver T Fackler, Britta Brügger, Thomas Hoenen, Walter Nickel, Ulrich S Schwarz, Petr Chlanda, and Department of Physics
- Subjects
Ebola virus ,General Immunology and Microbiology ,Membrane modeling and molecular dynamics simulations ,General Neuroscience ,in situ cryo-ET ,Membrane fusion ,Virus entry and uncoating ,1182 Biochemistry, cell and molecular biology ,Molecular Biology ,General Biochemistry, Genetics and Molecular Biology - Abstract
Ebola viruses (EBOVs) assemble into filamentous virions, whose shape and stability are determined by the matrix viral protein 40 (VP40). Virus entry into host cells occurs via membrane fusion in late endosomes; however, the mechanism of how the remarkably long virions undergo uncoating, including virion disassembly and nucleocapsid release into the cytosol, remains unknown. Here, we investigate the structural architecture of EBOVs entering host cells and discover that the VP40 matrix disassembles prior to membrane fusion. We reveal that VP40 disassembly is caused by the weakening of VP40-lipid interactions driven by low endosomal pH that equilibrates passively across the viral envelope without a dedicated ion channel. We further show that viral membrane fusion depends on VP40 matrix integrity, and its disassembly reduces the energy barrier for fusion stalk formation. Thus, pH-driven structural remodeling of the VP40 matrix acts as a molecular switch coupling viral matrix uncoating to membrane fusion during EBOV entry.
- Published
- 2023
34. Single-dose treatment with VSV-EBOV expressing Ebola virus-specific artificial miRNAs does not protect mice from lethal disease
- Author
-
Kyle L O’Donnell, Julie Callison, Heinz Feldmann, Thomas Hoenen, and Andrea Marzi
- Subjects
Infectious Diseases ,Immunology and Allergy - Abstract
Although significant progress has been made in the development of therapeutics against Ebola virus (EBOV), we sought to expand upon existing strategies and combine an RNAi-based intervention with the approved VSV-EBOV vaccine to conjointly treat and vaccinate patients during an outbreak. We constructed VSV-EBOV vectors expressing artificial miRNAs (amiRNAs) targeting sequences of EBOV proteins. In vitro experiments demonstrated a robust decrease in EBOV replication using a minigenome system and infectious virus. For in vivo evaluation, MA-EBOV-infected CD-1 mice were treated 24 hours after infection with a single dose of the VSV-EBOV-amiRNA constructs. We observed no difference in disease progression or survival compared to the control-treated mice. In summary, while amiRNAs decrease viral replication in vitro, the effect is not sufficient to protect mice from lethal disease, and this therapeutic approach requires further optimization.
- Published
- 2023
- Full Text
- View/download PDF
35. Cryoelectron microscopic structure of the nucleoprotein–RNA complex of the European filovirus, Lloviu virus
- Author
-
Shangfan Hu, Yoko Fujita-Fujiharu, Yukihiko Sugita, Lisa Wendt, Yukiko Muramoto, Masahiro Nakano, Thomas Hoenen, and Takeshi Noda
- Subjects
Lloviu virus ,nucleocapsid ,cryo-EM ,Filovirus - Abstract
Lloviu virus (LLOV) is a novel filovirus detected in Schreiber’s bats in Europe. The isolation of the infectious LLOV from bats has raised public health concerns. However, the virological and molecular characteristics of LLOV remain largely unknown. The nucleoprotein (NP) of LLOV encapsidates the viral genomic RNA to form a helical NP-RNA complex, which acts as a scaffold for nucleocapsid formation and de novo viral RNA synthesis. In this study, using single-particle cryo-electron microscopy, we determined two structures of the LLOV NP-RNA helical complex, comprising a full-length and a C-terminally truncated NP. The two helical structures were identical, demonstrating that the N-terminal region determines the helical arrangement of the NP. The LLOV NP-RNA protomers displayed a structure similar to that in the Ebola and Marburg virus, but the spatial arrangements in the helix differed. Structure-based mutational analysis identified amino acids involved in the helical assembly and viral RNA synthesis. These structures advance our understanding of the filovirus nucleocapsid formation, and provide a structural basis for the development of anti-filoviral therapeutics., ヨーロッパに分布するエボラウイルス近縁ウイルスの増殖機構を解明 --広範囲の抗フィロウイルス療法の開発に期待--. 京都大学プレスリリース. 2023-04-10.
- Published
- 2023
- Full Text
- View/download PDF
36. Evidence for Viral mRNA Export from Ebola Virus Inclusion Bodies by the Nuclear RNA Export Factor NXF1
- Author
-
Lisa Wendt, Janine Brandt, Dmitry S. Ushakov, Bianca S. Bodmer, Matthew J. Pickin, Allison Groseth, and Thomas Hoenen
- Subjects
Nucleocytoplasmic Transport Proteins ,Immunology ,RNA-Binding Proteins ,Hemorrhagic Fever, Ebola ,Ebolavirus ,Microbiology ,Antiviral Agents ,Inclusion Bodies, Viral ,Virus-Cell Interactions ,Viral Proteins ,Virology ,Insect Science ,Humans ,RNA, Viral ,RNA, Messenger - Abstract
Many negative-sense RNA viruses, including the highly pathogenic Ebola virus (EBOV), use cytoplasmic inclusion bodies (IBs) for viral RNA synthesis. However, it remains unclear how viral mRNAs are exported from these IBs for subsequent translation. We recently demonstrated that the nuclear RNA export factor 1 (NXF1) is involved in a late step in viral protein expression, i.e., downstream of viral mRNA transcription, and proposed it to be involved in this mRNA export process. We now provide further evidence for this function by showing that NXF1 is not required for translation of viral mRNAs, thus pinpointing its function to a step between mRNA transcription and translation. We further show that RNA binding of both NXF1 and EBOV NP is necessary for export of NXF1 from IBs, supporting a model in which NP hands viral mRNA over to NXF1 for export. Mapping of NP-NXF1 interactions allowed refinement of this model, revealing two separate interaction sites, one of them directly involving the RNA binding cleft of NP, even though these interactions are RNA-independent. Immunofluorescence analyses demonstrated that individual NXF1 domains are sufficient for its recruitment into IBs, and complementation assays helped to define NXF1 domains important for its function in the EBOV life cycle. Finally, we show that NXF1 is also required for protein expression of other viruses that replicate in cytoplasmic IBs, including Lloviu and Junín virus. These data suggest a role for NXF1 in viral mRNA export from IBs for various viruses, making it a potential target for broadly active antivirals. IMPORTANCE Filoviruses such as the Ebola virus (EBOV) cause severe hemorrhagic fevers with high case fatality rates and limited treatment options. The identification of virus-host cell interactions shared among several viruses would represent promising targets for the development of broadly active antivirals. In this study, we reveal the mechanistic details of how EBOV usurps the nuclear RNA export factor 1 (NXF1) to export viral mRNAs from viral inclusion bodies (IBs). We further show that NXF1 is not only required for the EBOV life cycle but also necessary for other viruses known to replicate in cytoplasmic IBs, including the filovirus Lloviu virus and the highly pathogenic arenavirus Junín virus. This suggests NXF1 as a promising target for the development of broadly active antivirals.
- Published
- 2023
37. Current Therapies for Biosafety Level 4 Pathogens
- Author
-
Lucie Fénéant, Bianca Bodmer, Thomas C. Mettenleiter, Allison Groseth, and Thomas Hoenen
- Published
- 2021
- Full Text
- View/download PDF
38. Author Reply to Peer Reviews of The Ebola virus VP40 matrix layer undergoes endosomal disassembly essential for membrane fusion
- Author
-
Sophie L. Winter, Gonen Golani, Fabio Lolicato, Melina Vallbracht, Keerthihan Thiyagarajah, Samy Sid Ahmed, Christian Luechtenborg, Oliver T Fackler, Britta Bruegger, Thomas Hoenen, Walter Nickel, Ulrich S Schwarz, and Petr Chlanda
- Published
- 2023
- Full Text
- View/download PDF
39. The Ebola virus VP40 matrix undergoes endosomal disassembly essential for membrane fusion
- Author
-
Sophie L. Winter, Gonen Golani, Fabio Lolicato, Melina Vallbracht, Keerthihan Thiyagarajah, Samy Sid Ahmed, Christian Lüchtenborg, Oliver T. Fackler, Britta Brügger, Thomas Hoenen, Walter Nickel, Ulrich S. Schwarz, and Petr Chlanda
- Abstract
Ebola viruses (EBOVs) are filamentous particles, whose shape and stability are determined by the VP40 matrix. Virus entry into host cells occurs via membrane fusion in late endosomes; however, the mechanism of how the remarkably long virions undergo uncoating including virion disassembly and nucleocapsid release into the cytosol, remains unknown. Here, we investigate the structural architecture of EBOVs entering host cells and discover that the VP40 matrix disassembles prior to membrane fusion. We reveal that VP40 disassembly is caused by the weakening of VP40-lipid interactions driven by low endosomal pH that equilibrates passively across the viral envelope without a dedicated ion channel. We further show that viral membrane fusion depends on VP40 matrix integrity, and its disassembly reduces the energy barrier for fusion stalk formation. Thus, pH-driven structural remodeling of the VP40 matrix acts as a molecular switch coupling viral matrix uncoating to membrane fusion during EBOV entry.
- Published
- 2022
- Full Text
- View/download PDF
40. A dsRNA-binding mutant reveals only a minor role of exonuclease activity in interferon antagonism by the arenavirus nucleoprotein
- Author
-
Patrick Bohn, Irke Waßmann, Lisa Wendt, Anne Leske, Thomas Hoenen, Birke A. Tews, and Allison Groseth
- Subjects
Virology ,Immunology ,Genetics ,Parasitology ,Molecular Biology ,Microbiology - Abstract
The arenavirus nucleoprotein (NP) plays an important role in the virus’ ability to block interferon (IFN) production, and its exonuclease function appears to contribute to this activity. However, efforts to analyze this contribution are complicated by the functional overlap between the exonuclease active site and a neighboring region involved in IKKε-binding and subsequent inhibition of IRF3 activation, which also plays an important role in IFN production. To circumvent this issue, we mutated a residue located away from the active site that is involved in binding of the dsRNA substrate being targeted for exonuclease digestion, i.e. H426A. We found that expression of Tacaribe virus (TCRV) NP containing this RNA-binding H426A mutation was still able to efficiently block IFN-β promoter activity in response to Sendai virus infection, despite being strongly impaired in its exonuclease activity. This was in contrast to a conventional exonuclease active site mutant (E388A), which was impaired with respect to both exonuclease activity and IFN antagonism. Importantly, growth of a recombinant virus encoding the RNA-binding mutation (rTCRV-H426A) was similar to wild-type in IFN-deficient cells, unlike the active site mutant (rTCRV-E388A), which was already markedly impaired in these cells. Further, in IFN-competent cells, the TCRV-H426A RNA-binding mutant showed more robust growth and delayed IFN-β mRNA upregulation compared to the TCRV-E388A active site mutant. Taken together, this novel mutational approach, which allows us to now dissect the different contributions of the NP exonuclease activity and IKKε-binding/IRF3 inhibition to IFN antagonism, clearly suggests that conventional exonuclease mutants targeting the active site overestimate the contribution of the exonuclease function, and that rather other IFN antagonistic functions of NP play the dominant role in IFN-antagonism.
- Published
- 2022
41. Immunization with GP1 but Not Core-like Particles Displaying Isolated Receptor-Binding Epitopes Elicits Virus-Neutralizing Antibodies against Junín Virus
- Author
-
Gleyder Roman-Sosa, Anne Leske, Xenia Ficht, Tung Huy Dau, Julia Holzerland, Thomas Hoenen, Martin Beer, Robert Kammerer, Reinhold Schirmbeck, Felix A. Rey, Sandra M. Cordo, and Allison Groseth
- Subjects
Pharmacology ,Infectious Diseases ,Drug Discovery ,Immunology ,Pharmacology (medical) ,arenavirus ,Junín virus ,immune response ,neutralizing antibodies ,glycoprotein ,GP1 - Abstract
New World arenaviruses are rodent-transmitted viruses and include a number of pathogens that are responsible for causing severe human disease. This includes Junín virus (JUNV), which is the causative agent of Argentine hemorrhagic fever. The wild nature and mobility of the rodent reservoir host makes it difficult to control the disease, and currently passive immunization with high-titer neutralizing antibody-containing plasma from convalescent patients is the only specific therapy. However, dwindling supplies of naturally available convalescent plasma, and challenges in developing similar resources for other closely related viruses, have made the development of alternative antibody-based therapeutic approaches of critical importance. In this study, we sought to induce a neutralizing antibody response in rabbits against the receptor-binding subunit of the viral glycoprotein, GP1, and the specific peptide sequences in GP1 involved in cellular receptor contacts. While these specific receptor-interacting peptides did not efficiently induce the production of neutralizing antibodies when delivered as a particulate antigen (as part of hepatitis B virus core-like particles), we showed that recombinant JUNV GP1 purified from transfected mammalian cells induced virus-neutralizing antibodies at high titers in rabbits. Further, neutralization was observed across a range of unrelated JUNV strains, a feature that is critical for effectiveness in the field. These results underscore the potential of GP1 alone to induce a potent neutralizing antibody response and highlight the importance of epitope presentation. In addition, effective virus neutralization by rabbit antibodies supports the potential applicability of this species for the future development of immunotherapeutics (e.g., based on humanized monoclonal antibodies). Such information can be applied in the design of vaccines and immunogens for both prevention and specific therapies against this and likely also other closely related pathogenic New World arenaviruses.
- Published
- 2021
42. Therapeutic strategies to target the Ebola virus life cycle
- Author
-
Thomas Hoenen, Heinz Feldmann, and Allison Groseth
- Subjects
0303 health sciences ,Ebola virus ,General Immunology and Microbiology ,030306 microbiology ,medicine.drug_class ,viruses ,Viral pathogenesis ,Drug Evaluation, Preclinical ,Disease ,ZMapp ,Biology ,Ebolavirus ,medicine.disease_cause ,Monoclonal antibody ,Antiviral Agents ,Microbiology ,Virology ,03 medical and health sciences ,Infectious Diseases ,Viral life cycle ,Host-Pathogen Interactions ,Gene expression ,medicine ,Viral rna ,medicine.drug - Abstract
Following the Ebola virus disease epidemic in west Africa, there has been increased awareness of the need for improved therapies for emerging diseases, including viral haemorrhagic fevers such as those caused by Ebola virus and other filoviruses. Our continually improving understanding of the virus life cycle coupled with the increased availability of ‘omics’ analyses and high-throughput screening technologies has enhanced our ability to identify potential viral and host factors and aspects involved in the infection process that might be intervention targets. In this Review we address compounds that have shown promise to various degrees in interfering with the filovirus life cycle, including monoclonal antibodies such as ZMapp, mAb114 and REGN-EB3 and inhibitors of viral RNA synthesis such as remdesivir and TKM-Ebola. Furthermore, we discuss the general potential of targeting aspects of the virus life cycle such as the entry process, viral RNA synthesis and gene expression, as well as morphogenesis and budding. In this Review, Hoenen, Groseth and Feldmann address strategies that have shown promise in interfering with the filovirus life cycle, including the entry process, viral RNA synthesis and gene expression, as well as morphogenesis and budding.
- Published
- 2019
- Full Text
- View/download PDF
43. Assessment of the function and intergenus-compatibility of Ebola and Lloviu virus proteins
- Author
-
Thomas Hoenen, Julia Holzerland, Janine Brandt, Stefanie Braun, Eric Hartmann, Lukas Zierke, Lisa Wendt, Marie Luisa Schmidt, Andreas Müller, Allison Groseth, and Lennart Kämper
- Subjects
0301 basic medicine ,viruses ,030106 microbiology ,Virus Replication ,medicine.disease_cause ,Genome ,Virus ,Viral Proteins ,03 medical and health sciences ,Viral life cycle ,Virology ,medicine ,Humans ,Ebola virus ,Lloviu virus ,biology ,Genetic Complementation Test ,Filoviridae ,Marburgvirus ,biology.organism_classification ,Phenotype ,Recombinant Proteins ,Reverse Genetics ,Reverse genetics ,HEK293 Cells ,030104 developmental biology - Abstract
Sequences for Lloviu virus (LLOV), a putative novel filovirus, were first identified in Miniopterus schreibersii bats in Spain following a massive bat die-off in 2002, and also recently found in bats in Hungary. However, until now it is unclear if these sequences correspond to a fully functional, infectious virus, and whether it will show a pathogenic phenotype like African filoviruses, such as ebola- and marburgviruses, or be apathogenic for humans, like the Asian filovirus Reston virus. Since no infectious virus has been recovered, the only opportunity to study infectious LLOV is to use a reverse genetics-based full-length clone system to de novo generate LLOV. As a first step in this process, and to investigate whether the identified sequences indeed correspond to functional viral proteins, we have developed life cycle modelling systems for LLOV, which allow us to study genome replication and transcription as well as entry of this virus. We show that all LLOV proteins fulfill their canonical role in the virus life cycle as expected based on the well-studied related filovirus Ebola virus. Further, we have analysed the intergenus-compatibility of proteins that have to act in concert to facilitate the virus life cycle. We show that some but not all proteins from LLOV and Ebola virus are compatible with each other, emphasizing the close relationship of these viruses, and informing future studies of filovirus biology with respect to the generation of genus-chimeric proteins in order to probe virus protein-protein interactions on a functional level.
- Published
- 2019
- Full Text
- View/download PDF
44. Ebola Virus Glycoprotein Domains Associated with Protective Efficacy
- Author
-
Heinz Feldmann, Thomas Hoenen, Bharti Bhatia, Wakako Furuyama, and Andrea Marzi
- Subjects
0301 basic medicine ,filovirus ,Immunogen ,viruses ,030231 tropical medicine ,Immunology ,medicine.disease_cause ,Virus ,Article ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,glycan cap ,Drug Discovery ,medicine ,Pharmacology (medical) ,Vector (molecular biology) ,mucin-like domain ,Pharmacology ,chemistry.chemical_classification ,Ebola virus ,biology ,Immunogenicity ,biology.organism_classification ,Virology ,030104 developmental biology ,Infectious Diseases ,chemistry ,Vesicular stomatitis virus ,VSV ,Medicine ,vesicular stomatitis virus ,Glycoprotein ,EBOV - Abstract
Ebola virus (EBOV) is the cause of sporadic outbreaks of human hemorrhagic disease in Africa, and the best-characterized virus in the filovirus family. The West Africa epidemic accelerated the clinical development of vaccines and therapeutics leading to licensure of vaccines and antibody-based therapeutics for human use in recent years. The most widely used vaccine is based on vesicular stomatitis virus (VSV) expressing the EBOV glycoprotein (GP)(VSV-EBOV). Due to its favorable immune cell targeting, this vaccine has also been used as base-vector for the development of second generation VSV-based vaccines against Influenza, Nipah, and Zika viruses. However, in these situations it may be beneficial if the immunogenicity against EBOV GP is minimized to induce a better protective immune response against the other foreign immunogen. Here, we analyzed if EBOV GP can be truncated to be less immunogenic yet still able to drive replication of the vaccine vector. We found that the EBOV GP glycan cap and the mucin-like domain are both dispensable for VSV-EBOV replication. The glycan cap domain, however, appears critical for mediating a protective immune response against lethal EBOV challenge in mice.
- Published
- 2021
45. Interferon-induced HERC5 Inhibits Ebola Virus Particle Production and Is Antagonized by Ebola Glycoprotein
- Author
-
Thomas Hoenen, Marceline Côté, Macon D. Coleman, Nina R Hunt, Stephen D. Barr, Ermela Paparisto, Daniel S Labach, Eric Di Gravio, Andreas Müller, and Mackenzie J. Dodge
- Subjects
chemistry.chemical_classification ,Ebola virus ,viruses ,Biology ,ZINC FINGER ANTIVIRAL PROTEIN ,medicine.disease_cause ,Virology ,Marburg virus ,chemistry ,In vivo ,Transcription (biology) ,Interferon ,medicine ,biochemistry ,Glycoprotein ,Gene ,medicine.drug - Abstract
Survival following Ebola virus (EBOV) infection correlates with the ability to mount an early and robust interferon (IFN) response. The host IFN-induced proteins that contribute to controlling EBOV replication are not fully known. Among the top genes with the strongest early increases in expression after infection in vivo is IFN-induced HERC5. Using a transcription- and replication-competent VLP system, we showed that HERC5 inhibits EBOV virus-like particle (VLP) replication by depleting EBOV mRNAs. The HERC5 RCC1-like domain was necessary and sufficient for this inhibition and did not require zinc finger antiviral protein (ZAP). Moreover, we showed that EBOV (Zaire) glycoprotein (GP) but not Marburg virus GP antagonized HERC5 early during infection. Our data identifies a novel ‘protagonist-antagonistic’ relationship between HERC5 and GP in the early stages of EBOV infection that could be exploited for the development of novel antiviral therapeutics.
- Published
- 2021
- Full Text
- View/download PDF
46. Remdesivir inhibits the polymerases of the novel filoviruses Lloviu and Bombali virus
- Author
-
Lisa Wendt, Bianca S Bodmer, Allison Groseth, Josephin Greßler, Thomas Hoenen, and Lukas Zierke
- Subjects
0301 basic medicine ,viruses ,030106 microbiology ,Biology ,medicine.disease_cause ,Virus Replication ,Antiviral Agents ,Virus ,Cell Line ,03 medical and health sciences ,Inhibitory Concentration 50 ,Virology ,Ic50 values ,medicine ,Humans ,Polymerase ,Phylogeny ,Pharmacology ,Ebola virus ,Lloviu virus ,Alanine ,Treatment options ,Ebolavirus ,Filoviridae ,RNA-Dependent RNA Polymerase ,Reverse genetics ,Adenosine Monophosphate ,030104 developmental biology ,biology.protein ,Antibody - Abstract
In recent years, a number of novel filoviruses (e.g. Lloviu virus (LLOV) and Bombali virus (BOMV)) have been discovered. While antibody-based therapeutics have recently been approved for treatment of infections with the filovirus Ebola virus (EBOV), no treatment options for novel filoviruses currently exist. Further, the development of antivirals against them is complicated by the fact that only sequence information, but no actual virus isolates, are available. To address this issue, we developed a reverse genetics-based minigenome system for BOMV, which allows us to assess the activity of the BOMV polymerase. Together with similar systems that we have developed for other filoviruses in the past (i.e. LLOV and Reston virus (RESTV)), we then assessed the efficiency of remdesivir, a known inhibitor of the EBOV polymerase that has recently been tested in a clinical trial for efficacy against Ebola disease. We show that remdesivir is indeed also active against the polymerases of BOMV, LLOV, and RESTV, with comparable IC50 values to its activity against EBOV. This suggests that treatment with remdesivir might represent a viable option in case of infections with novel filoviruses.
- Published
- 2021
47. Differences in Viral RNA Synthesis but Not Budding or Entry Contribute to the In Vitro Attenuation of Reston Virus Compared to Ebola Virus
- Author
-
Thomas Hoenen, Stefanie Braun, Josephin Greßler, Marie Luisa Schmidt, Allison Groseth, Bianca S Bodmer, Julia Holzerland, and Janine Brandt
- Subjects
Microbiology (medical) ,filovirus ,Population ,budding ,Biology ,medicine.disease_cause ,Microbiology ,Article ,Virus ,03 medical and health sciences ,Ebola virus ,Viral life cycle ,Virology ,medicine ,pathogenicity ,Seroconversion ,RNA synthesis ,trVLP system ,education ,lcsh:QH301-705.5 ,030304 developmental biology ,Ribonucleoprotein ,0303 health sciences ,Budding ,education.field_of_study ,030306 microbiology ,Outbreak ,lcsh:Biology (General) ,Reston virus ,entry - Abstract
Most filoviruses cause severe disease in humans. For example, Ebola virus (EBOV) is responsible for the two most extensive outbreaks of filovirus disease to date, with case fatality rates of 66% and 40%, respectively. In contrast, Reston virus (RESTV) is apparently apathogenic in humans, and while transmission of RESTV from domestic pigs to people results in seroconversion, no signs of disease have been reported in such cases. The determinants leading to these differences in pathogenicity are not well understood, but such information is needed in order to better evaluate the risks posed by the repeated spillover of RESTV into the human population and to perform risk assessments for newly emerging filoviruses with unknown pathogenic potential. Interestingly, RESTV and EBOV already show marked differences in their growth in vitro, with RESTV growing slower and reaching lower end titers. In order to understand the basis for this in vitro attenuation of RESTV, we used various life cycle modeling systems mimicking different aspects of the virus life cycle. Our results showed that viral RNA synthesis was markedly slower when using the ribonucleoprotein (RNP) components from RESTV, rather than those for EBOV. In contrast, the kinetics of budding and entry were indistinguishable between these two viruses. These data contribute to our understanding of the molecular basis for filovirus pathogenicity by showing that it is primarily differences in the robustness of RNA synthesis by the viral RNP complex that are responsible for the impaired growth of RESTV in tissue culture.
- Published
- 2020
- Full Text
- View/download PDF
48. The Cellular Protein CAD is Recruited into Ebola Virus Inclusion Bodies by the Nucleoprotein NP to Facilitate Genome Replication and Transcription
- Author
-
Thomas Hoenen, Janine Brandt, Lisa Wendt, Bianca S Bodmer, and Thomas C. Mettenleiter
- Subjects
0301 basic medicine ,filovirus ,Transcription, Genetic ,030106 microbiology ,inclusion bodies ,Genome, Viral ,Computational biology ,Biology ,Virus Replication ,medicine.disease_cause ,Genome ,Article ,Virus ,Cell Line ,Inclusion Bodies, Viral ,Viral Proteins ,03 medical and health sciences ,Ebola virus ,Protein Domains ,Transcription (biology) ,pyrimidine synthesis ,Aspartate Carbamoyltransferase ,medicine ,Animals ,Humans ,CAD ,lcsh:QH301-705.5 ,Dihydroorotase ,Host factor ,General Medicine ,Ebolavirus ,Reverse genetics ,Nucleoprotein ,virology ,Nucleoproteins ,Pyrimidines ,030104 developmental biology ,lcsh:Biology (General) ,Gene Knockdown Techniques ,RNA ,Carbamoyl-Phosphate Synthase (Glutamine-Hydrolyzing) ,Viral genome replication ,Protein Binding - Abstract
Ebola virus (EBOV) is a zoonotic pathogen causing severe hemorrhagic fevers in humans and non-human primates with high case fatality rates. In recent years, the number and extent of outbreaks has increased, highlighting the importance of better understanding the molecular aspects of EBOV infection and host cell interactions to control this virus more efficiently. Many viruses, including EBOV, have been shown to recruit host proteins for different viral processes. Based on a genome-wide siRNA screen, we recently identified the cellular host factor carbamoyl-phosphate synthetase 2, aspartate transcarbamylase, and dihydroorotase (CAD) as being involved in EBOV RNA synthesis. However, mechanistic details of how this host factor plays a role in the EBOV life cycle remain elusive. In this study, we analyzed the functional and molecular interactions between EBOV and CAD. To this end, we used siRNA knockdowns in combination with various reverse genetics-based life cycle modelling systems and additionally performed co-immunoprecipitation and co-immunofluorescence assays to investigate the influence of CAD on individual aspects of the EBOV life cycle and to characterize the interactions of CAD with viral proteins. Following this approach, we could demonstrate that CAD directly interacts with the EBOV nucleoprotein NP, and that NP is sufficient to recruit CAD into inclusion bodies dependent on the glutaminase (GLN) domain of CAD. Further, siRNA knockdown experiments indicated that CAD is important for both viral genome replication and transcription, while substrate rescue experiments showed that the function of CAD in pyrimidine synthesis is indeed required for those processes. Together, this suggests that NP recruits CAD into inclusion bodies via its GLN domain in order to provide pyrimidines for EBOV genome replication and transcription. These results define a novel mechanism by which EBOV hijacks host cell pathways in order to facilitate genome replication and transcription and provide a further basis for the development of host-directed broad-spectrum antivirals.
- Published
- 2020
- Full Text
- View/download PDF
49. Structure and functions of the Ebola virus matrix protein VP40
- Author
-
Lisa Wendt, Thomas Hoenen, and Janine Brandt
- Subjects
VP40 ,Ebola virus ,Viral matrix protein ,viruses ,Virology ,medicine ,Biology ,medicine.disease_cause - Abstract
The matrix protein VP40 of the highly pathogenic Ebola virus (EBOV), a member of the filovirus family, is the most abundant protein in EBOV virions. During the viral life cycle it mediates assembly and budding from the host cell, and is responsible for the characteristic filamentous shape of EBOV particles. In addition to this classical function as a matrix protein, VP40 was also shown to have a regulatory function in viral transcription. To enable these distinct functions, VP40 can adopt different oligomeric states, in particular, dimers, hexamers and ring-like octameric RNA-binding structures. This review describes the properties and functions of the EBOV matrix protein VP40 and how these different conformations of VP40 contribute to its diverse functions.
- Published
- 2019
- Full Text
- View/download PDF
50. Ebola virus inclusion body formation and RNA synthesis are controlled by a novel domain of NP interacting with VP35
- Author
-
Tsuyoshi Miyake, Thomas Hoenen, Daniel A. Engel, Benjamin E. Neubauer, Charlotte M. Farley, and Thomas P. Beddow
- Subjects
Ebola virus ,Chemistry ,viruses ,medicine.disease_cause ,Inclusion bodies ,Nucleoprotein ,Cell biology ,chemistry.chemical_compound ,Viral replication ,Viral life cycle ,Cytoplasm ,RNA polymerase ,medicine ,Function (biology) - Abstract
Ebola virus (EBOV) inclusion bodies (IBs) are cytoplasmic sites of nucleocapsid formation and RNA replication, housing key steps in the virus life cycle that warrant further investigation. During infection IBs display dynamic properties regarding their size and location. Also, the contents of IBs must transition prior to further viral maturation, assembly and release, implying additional steps in IB function. Interestingly, expression of the viral nucleoprotein (NP) alone is sufficient for generation of IBs, indicating that it plays an important role in IB formation during infection. In addition to NP, other components of the nucleocapsid localize to IBs, including VP35, VP24, VP30 and the RNA polymerase L. Previously we defined and solved the crystal structure of the C-terminal domain of NP (NP-Ct), but its role in virus replication remained unclear. Here we show that NP-Ct is absolutely required for IB formation when NP is expressed alone. Interestingly, we find that NP-Ct is also required for production of infectious virus-like particles and retention of viral RNA within these particles. Furthermore, co-expression of the nucleocapsid component VP35 overcomes deletion of NP-Ct in triggering IB formation, demonstrating a functional interaction between the two proteins. Of all the EBOV proteins only VP35 is able to overcome the defect in IB formation caused by deletion of NP-Ct. This effect is mediated by a novel protein-protein interaction between VP35 and NP that controls both regulation of IB formation and RNA replication itself, and which is mediated by a newly identified domain of NP, the “central domain” (CD).ImportanceInclusion bodies (IBs) are cytoplasmic sites of RNA synthesis for a variety of negative sense RNA viruses including Ebola virus. In addition to housing important steps in the viral life cycle, IBs protect new viral RNA from innate immune attack and contain specific host proteins whose function is under study. A key viral factor in Ebola virus IB formation is the nucleoprotein, NP, which also is important in RNA encapsidation and synthesis. In this study, we have identified two domains of NP that control inclusion body formation. One of these, the central domain (CD), interacts with viral protein VP35 to control both inclusion body formation and RNA synthesis. The other is the NP C-terminal domain (NP-Ct), whose function has not previously been reported. These findings contribute to a model in which NP and its interactions with VP35 link the establishment of IBs to the synthesis of viral RNA.
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.