123 results on '"Hume, Adam J."'
Search Results
2. 2022 taxonomic update of phylum Negarnaviricota (Riboviria: Orthornavirae), including the large orders Bunyavirales and Mononegavirales
- Author
-
Kuhn, Jens H., Adkins, Scott, Alkhovsky, Sergey V., Avšič-Županc, Tatjana, Ayllón, María A., Bahl, Justin, Balkema-Buschmann, Anne, Ballinger, Matthew J., Bandte, Martina, Beer, Martin, Bejerman, Nicolas, Bergeron, Éric, Biedenkopf, Nadine, Bigarré, Laurent, Blair, Carol D., Blasdell, Kim R., Bradfute, Steven B., Briese, Thomas, Brown, Paul A., Bruggmann, Rémy, Buchholz, Ursula J., Buchmeier, Michael J., Bukreyev, Alexander, Burt, Felicity, Büttner, Carmen, Calisher, Charles H., Candresse, Thierry, Carson, Jeremy, Casas, Inmaculada, Chandran, Kartik, Charrel, Rémi N., Chiaki, Yuya, Crane, Anya, Crane, Mark, Dacheux, Laurent, Bó, Elena Dal, de la Torre, Juan Carlos, de Lamballerie, Xavier, de Souza, William M., de Swart, Rik L., Dheilly, Nolwenn M., Di Paola, Nicholas, Di Serio, Francesco, Dietzgen, Ralf G., Digiaro, Michele, Drexler, J. Felix, Duprex, W. Paul, Dürrwald, Ralf, Easton, Andrew J., Elbeaino, Toufic, Ergünay, Koray, Feng, Guozhong, Feuvrier, Claudette, Firth, Andrew E., Fooks, Anthony R., Formenty, Pierre B. H., Freitas-Astúa, Juliana, Gago-Zachert, Selma, García, María Laura, García-Sastre, Adolfo, Garrison, Aura R., Godwin, Scott E., Gonzalez, Jean-Paul J., de Bellocq, Joëlle Goüy, Griffiths, Anthony, Groschup, Martin H., Günther, Stephan, Hammond, John, Hepojoki, Jussi, Hierweger, Melanie M., Hongō, Seiji, Horie, Masayuki, Horikawa, Hidenori, Hughes, Holly R., Hume, Adam J., Hyndman, Timothy H., Jiāng, Dàohóng, Jonson, Gilda B., Junglen, Sandra, Kadono, Fujio, Karlin, David G., Klempa, Boris, Klingström, Jonas, Koch, Michel C., Kondō, Hideki, Koonin, Eugene V., Krásová, Jarmila, Krupovic, Mart, Kubota, Kenji, Kuzmin, Ivan V., Laenen, Lies, Lambert, Amy J., Lǐ, Jiànróng, Li, Jun-Min, Lieffrig, François, Lukashevich, Igor S., Luo, Dongsheng, Maes, Piet, Marklewitz, Marco, Marshall, Sergio H., Marzano, Shin-Yi L., McCauley, John W., Mirazimi, Ali, Mohr, Peter G., Moody, Nick J. G., Morita, Yasuaki, Morrison, Richard N., Mühlberger, Elke, Naidu, Rayapati, Natsuaki, Tomohide, Navarro, José A., Neriya, Yutaro, Netesov, Sergey V., Neumann, Gabriele, Nowotny, Norbert, Ochoa-Corona, Francisco M., Palacios, Gustavo, Pallandre, Laurane, Pallás, Vicente, Papa, Anna, Paraskevopoulou, Sofia, Parrish, Colin R., Pauvolid-Corrêa, Alex, Pawęska, Janusz T., Pérez, Daniel R., Pfaff, Florian, Plemper, Richard K., Postler, Thomas S., Pozet, Françoise, Radoshitzky, Sheli R., Ramos-González, Pedro L., Rehanek, Marius, Resende, Renato O., Reyes, Carina A., Romanowski, Víctor, Rubbenstroth, Dennis, Rubino, Luisa, Rumbou, Artemis, Runstadler, Jonathan A., Rupp, Melanie, Sabanadzovic, Sead, Sasaya, Takahide, Schmidt-Posthaus, Heike, Schwemmle, Martin, Seuberlich, Torsten, Sharpe, Stephen R., Shi, Mang, Sironi, Manuela, Smither, Sophie, Song, Jin-Won, Spann, Kirsten M., Spengler, Jessica R., Stenglein, Mark D., Takada, Ayato, Tesh, Robert B., Těšíková, Jana, Thornburg, Natalie J., Tischler, Nicole D., Tomitaka, Yasuhiro, Tomonaga, Keizō, Tordo, Noël, Tsunekawa, Kenta, Turina, Massimo, Tzanetakis, Ioannis E., Vaira, Anna Maria, van den Hoogen, Bernadette, Vanmechelen, Bert, Vasilakis, Nikos, Verbeek, Martin, von Bargen, Susanne, Wada, Jiro, Wahl, Victoria, Walker, Peter J., Whitfield, Anna E., Williams, John V., Wolf, Yuri I., Yamasaki, Junki, Yanagisawa, Hironobu, Ye, Gongyin, Zhang, Yong-Zhen, and Økland, Arnfinn Lodden
- Published
- 2022
- Full Text
- View/download PDF
3. Renaming of genera Ebolavirus and Marburgvirus to Orthoebolavirus and Orthomarburgvirus, respectively, and introduction of binomial species names within family Filoviridae
- Author
-
Biedenkopf, Nadine, Bukreyev, Alexander, Chandran, Kartik, Di Paola, Nicholas, Formenty, Pierre B. H., Griffiths, Anthony, Hume, Adam J., Mühlberger, Elke, Netesov, Sergey V., Palacios, Gustavo, Pawęska, Janusz T., Smither, Sophie, Takada, Ayato, Wahl, Victoria, and Kuhn, Jens H.
- Published
- 2023
- Full Text
- View/download PDF
4. Ebola virus infection induces a delayed type I IFN response in bystander cells and the shutdown of key liver genes in human iPSC-derived hepatocytes
- Author
-
Scoon, Whitney A., Mancio-Silva, Liliana, Suder, Ellen L., Villacorta-Martin, Carlos, Lindstrom-Vautrin, Jonathan, Bernbaum, John G., Mazur, Steve, Johnson, Reed F., Olejnik, Judith, Flores, Elizabeth Y., Mithal, Aditya, Wang, Feiya, Hume, Adam J., Kaserman, Joseph E., March-Riera, Sandra, Wilson, Andrew A., Bhatia, Sangeeta N., Mühlberger, Elke, and Mostoslavsky, Gustavo
- Published
- 2022
- Full Text
- View/download PDF
5. Interferon-alpha or -beta facilitates SARS-CoV-2 pulmonary vascular infection by inducing ACE2
- Author
-
Klouda, Timothy, Hao, Yuan, Kim, Hyunbum, Kim, Jiwon, Olejnik, Judith, Hume, Adam J., Ayyappan, Sowntharya, Hong, Xuechong, Melero-Martin, Juan, Fang, Yinshan, Wang, Qiong, Zhou, Xiaobo, Mühlberger, Elke, Jia, Hongpeng, Padera, Jr., Robert F., Raby, Benjamin A., and Yuan, Ke
- Published
- 2022
- Full Text
- View/download PDF
6. Author Correction: Isolation of infectious Lloviu virus from Schreiber’s bats in Hungary
- Author
-
Kemenesi, Gábor, Tóth, Gábor E., Mayora-Neto, Martin, Scott, Simon, Temperton, Nigel, Wright, Edward, Mühlberger, Elke, Hume, Adam J., Suder, Ellen L., Zana, Brigitta, Boldogh, Sándor A., Görföl, Tamás, Estók, Péter, Szentiványi, Tamara, Lanszki, Zsófia, Somogyi, Balázs A., Nagy, Ágnes, Pereszlényi, Csaba I., Dudás, Gábor, Földes, Fanni, Kurucz, Kornélia, Madai, Mónika, Zeghbib, Safia, Maes, Piet, Vanmechelen, Bert, and Jakab, Ferenc
- Published
- 2022
- Full Text
- View/download PDF
7. Isolation of infectious Lloviu virus from Schreiber’s bats in Hungary
- Author
-
Kemenesi, Gábor, Tóth, Gábor E., Mayora-Neto, Martin, Scott, Simon, Temperton, Nigel, Wright, Edward, Mühlberger, Elke, Hume, Adam J., Suder, Ellen L., Zana, Brigitta, Boldogh, Sándor A., Görföl, Tamás, Estók, Péter, Szentiványi, Tamara, Lanszki, Zsófia, Somogyi, Balázs A., Nagy, Ágnes, Pereszlényi, Csaba I., Dudás, Gábor, Földes, Fanni, Kurucz, Kornélia, Madai, Mónika, Zeghbib, Safia, Maes, Piet, Vanmechelen, Bert, and Jakab, Ferenc
- Published
- 2022
- Full Text
- View/download PDF
8. Low-Input, High-Resolution 5′ Terminal Filovirus RNA Sequencing with ViBE-Seq.
- Author
-
Ross, Stephen J., Hume, Adam J., Olejnik, Judith, Turcinovic, Jacquelyn, Honko, Anna N., McKay, Lindsay G. A., Connor, John H., Griffiths, Anthony, Mühlberger, Elke, and Cifuentes, Daniel
- Subjects
- *
MARBURG virus , *RNA sequencing , *EBOLA virus , *NUCLEOTIDE sequencing , *RNA viruses - Abstract
Although next-generation sequencing (NGS) has been instrumental in determining the genomic sequences of emerging RNA viruses, de novo sequence determination often lacks sufficient coverage of the 5′ and 3′ ends of the viral genomes. Since the genome ends of RNA viruses contain the transcription and genome replication promoters that are essential for viral propagation, a lack of terminal sequence information hinders the efforts to study the replication and transcription mechanisms of emerging and re-emerging viruses. To circumvent this, we have developed a novel method termed ViBE-Seq (Viral Bona Fide End Sequencing) for the high-resolution sequencing of filoviral genome ends using a simple yet robust protocol with high fidelity. This technique allows for sequence determination of the 5′ end of viral RNA genomes and mRNAs with as little as 50 ng of total RNA. Using the Ebola virus and Marburg virus as prototypes for highly pathogenic, re-emerging viruses, we show that ViBE-Seq is a reliable technique for rapid and accurate 5′ end sequencing of filovirus RNA sourced from virions, infected cells, and tissue obtained from infected animals. We also show that ViBE-Seq can be used to determine whether distinct reverse transcriptases have terminal deoxynucleotidyl transferase activity. Overall, ViBE-Seq will facilitate the access to complete sequences of emerging viruses. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
9. A Comparative Assessment of the Pathogenic Potential of Newly Discovered Henipaviruses.
- Author
-
Meier, Kristina, Olejnik, Judith, Hume, Adam J., and Mühlberger, Elke
- Subjects
HENDRA virus ,HENIPAVIRUSES ,NIPAH virus ,VIRAL transmission ,NUCLEOTIDE sequencing - Abstract
Recent advances in high-throughput sequencing technologies have led to the discovery of a plethora of previously unknown viruses in animal samples. Some of these newly detected viruses are closely related to human pathogens. A prime example are the henipaviruses. Both Nipah (NiV) and Hendra virus (HeV) cause severe disease in humans. Henipaviruses are of zoonotic origin, and animal hosts, including intermediate hosts, play a critical role in viral transmission to humans. The natural reservoir hosts of NiV and HeV seem to be restricted to a few fruit bat species of the Pteropus genus in distinct geographic areas. However, the recent discovery of novel henipa- and henipa-like viruses suggests that these viruses are far more widespread than was originally thought. To date, these new viruses have been found in a wide range of animal hosts, including bats, shrews, and rodents in Asia, Africa, Europe, and South America. Since these viruses are closely related to human pathogens, it is important to learn whether they pose a threat to human health. In this article, we summarize what is known about the newly discovered henipaviruses, highlight differences to NiV and HeV, and discuss their pathogenic potential. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
10. Ebolavirus polymerase uses an unconventional genome replication mechanism
- Author
-
Deflubé, Laure R., Cressey, Tessa N., Hume, Adam J., Olejnik, Judith, Haddock, Elaine, Feldmann, Friederike, Ebihara, Hideki, Fearns, Rachel, and Mühlberger, Elke
- Published
- 2019
11. Heat Inactivation of Nipah Virus for Downstream Single-Cell RNA Sequencing Does Not Interfere with Sample Quality.
- Author
-
Hume, Adam J., Olejnik, Judith, White, Mitchell R., Huang, Jessie, Turcinovic, Jacquelyn, Heiden, Baylee, Bawa, Pushpinder S., Williams, Christopher J., Gorham, Nickolas G., Alekseyev, Yuriy O., Connor, John H., Kotton, Darrell N., and Mühlberger, Elke
- Subjects
NIPAH virus ,RNA sequencing ,BIOLOGICAL systems ,VIRUS inactivation ,CHEMICAL testing ,RNA viruses - Abstract
Single-cell RNA sequencing (scRNA-seq) technologies are instrumental to improving our understanding of virus–host interactions in cell culture infection studies and complex biological systems because they allow separating the transcriptional signatures of infected versus non-infected bystander cells. A drawback of using biosafety level (BSL) 4 pathogens is that protocols are typically developed without consideration of virus inactivation during the procedure. To ensure complete inactivation of virus-containing samples for downstream analyses, an adaptation of the workflow is needed. Focusing on a commercially available microfluidic partitioning scRNA-seq platform to prepare samples for scRNA-seq, we tested various chemical and physical components of the platform for their ability to inactivate Nipah virus (NiV), a BSL-4 pathogen that belongs to the group of nonsegmented negative-sense RNA viruses. The only step of the standard protocol that led to NiV inactivation was a 5 min incubation at 85 °C. To comply with the more stringent biosafety requirements for BSL-4-derived samples, we included an additional heat step after cDNA synthesis. This step alone was sufficient to inactivate NiV-containing samples, adding to the necessary inactivation redundancy. Importantly, the additional heat step did not affect sample quality or downstream scRNA-seq results. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
12. The 3′ Untranslated Regions of Ebola Virus mRNAs Contain AU-Rich Elements Involved in Posttranscriptional Stabilization and Decay.
- Author
-
Nelson, Emily V, Ross, Stephen J, Olejnik, Judith, Hume, Adam J, Deeney, Dylan J, King, Emily, Grimins, Autumn O, Lyons, Shawn M, Cifuentes, Daniel, and Mühlberger, Elke
- Subjects
EBOLA virus ,RNA-binding proteins ,MAMMALIAN embryos ,REPORTER genes ,PROTEIN expression - Abstract
The 3′ untranslated regions (UTRs) of Ebola virus (EBOV) mRNAs are enriched in their AU content and therefore represent potential targets for RNA binding proteins targeting AU-rich elements (ARE-BPs). ARE-BPs are known to fine-tune RNA turnover and translational activity. We identified putative AREs within EBOV mRNA 3′ UTRs and assessed whether they might modulate mRNA stability. Using mammalian and zebrafish embryo reporter assays, we show a conserved, ARE-BP-mediated stabilizing effect and increased reporter activity with the tested EBOV 3′ UTRs. When coexpressed with the prototypic ARE-BP tristetraprolin (TTP, ZFP36) that mainly destabilizes its target mRNAs, the EBOV nucleoprotein (NP) 3′ UTR resulted in decreased reporter gene activity. Coexpression of NP with TTP led to reduced NP protein expression and diminished EBOV minigenome activity. In conclusion, the enrichment of AU residues in EBOV 3′ UTRs makes them possible targets for cellular ARE-BPs, leading to modulation of RNA stability and translational activity. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
13. Pathogenicity of Lloviu and Bombali Viruses in Type I Interferon Receptor Knockout Mice.
- Author
-
Fletcher, Paige, Feldmann, Friederike, Takada, Ayato, Crossland, Nicholas A, Hume, Adam J, Albariño, César, Kemenesi, Gábor, Feldmann, Heinz, Mühlberger, Elke, and Marzi, Andrea
- Subjects
TYPE I interferons ,INTERFERON receptors ,KNOCKOUT mice ,VIRAL load - Abstract
Type I interferon receptor knockout (IFNAR
−/− ) mice are not able to generate a complete innate immune response; therefore, these mice are often considered to assess the pathogenicity of emerging viruses. We infected IFNAR−/− mice with a low or high dose of Lloviu virus (LLOV) or Bombali virus (BOMV) by the intranasal (IN) or intraperitoneal (IP) route and compared virus loads at early and late time points after infection. No signs of disease and no viral RNA were detected after IN infection regardless of LLOV dose. In contrast, IP infections resulted in increased viral loads in the high-dose LLOV and BOMV groups at the early time point. The low-dose LLOV and BOMV groups achieved higher viral loads at the late time point. However, there was 100% survival in all groups and no signs of disease. In conclusion, our results indicate a limited value of the IFNAR−/− mouse model for investigation of the pathogenicity of LLOV and BOMV. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
14. Art of the Kill: Designing and Testing Viral Inactivation Procedures for Highly Pathogenic Negative Sense RNA Viruses.
- Author
-
Olejnik, Judith, Hume, Adam J., Ross, Stephen J., Scoon, Whitney A., Seitz, Scott, White, Mitchell R., Slutzky, Ben, Yun, Nadezhda E., and Mühlberger, Elke
- Subjects
RNA viruses ,PATHOGENIC viruses ,CHEMICAL reagents ,TEST design ,EBOLA virus ,SAFETY regulations - Abstract
The study of highly pathogenic viruses handled under BSL-4 conditions and classified as Select Agents frequently involves the transfer of inactivated materials to lower containment levels for downstream analyses. Adhering to Select Agent and BSL-4 safety regulations requires validation or verification of the inactivation procedures, which comes with an array of challenges for each method. This includes the use of cytotoxic reagents for chemical inactivation and defining the precise inactivation parameters for physical inactivation. Here, we provide a workflow for various inactivation methods using Ebola, Nipah, and Lassa viruses as our examples. We choose three distinct inactivation methods (TRIzol/TRIzol LS, aldehyde fixation using different fixatives, and heat) to highlight the challenges of each method and provide possible solutions. We show that, whereas published chemical inactivation methods are highly reliable, the parameters for heat inactivation must be clearly defined to ensure complete inactivation. In addition to the inactivation data, we also provide examples and templates for the documentation required for approval and use of inactivation SOPs, including an inactivation report, the procedure sections of developed SOPs, and an electronic inactivation certificate that accompanies inactivated samples. The provided information can be used as a roadmap for similar studies at high and maximum containment laboratories. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
15. An RNA polymerase II-driven Ebola virus minigenome system as an advanced tool for antiviral drug screening
- Author
-
Nelson, Emily V., Pacheco, Jennifer R., Hume, Adam J., Cressey, Tessa N., Deflubé, Laure R., Ruedas, John B., Connor, John H., Ebihara, Hideki, and Mühlberger, Elke
- Published
- 2017
- Full Text
- View/download PDF
16. Human Papillomavirus 16 E7 Inactivator of Retinoblastoma Family Proteins Complements Human Cytomegalovirus Lacking UL97 Protein Kinase
- Author
-
Kamil, Jeremy P., Hume, Adam J., Jurak, Igor, Münger, Karl, Kalejta, Robert F., Coen, Donald M., and Harlow, Ed
- Published
- 2009
- Full Text
- View/download PDF
17. Phosphorylation of Retinoblastoma Protein by Viral Protein with Cyclin-Dependent Kinase Function
- Author
-
Hume, Adam J., Finkel, Jonathan S., Kamil, Jeremy P., Coen, Donald M., Culbertson, Michael R., and Kalejta, Robert F.
- Published
- 2008
- Full Text
- View/download PDF
18. Establishment of an Inactivation Method for Ebola Virus and SARS-CoV-2 Suitable for Downstream Sequencing of Low Cell Numbers.
- Author
-
Olejnik, Judith, Leon, Juliette, Michelson, Daniel, Chowdhary, Kaitavjeet, Galvan-Pena, Silvia, Benoist, Christophe, Mühlberger, Elke, and Hume, Adam J.
- Subjects
SARS-CoV-2 ,EBOLA virus ,RNA sequencing ,HEAT treatment ,BIOSAFETY ,VIRUS inactivation - Abstract
Technologies that facilitate the bulk sequencing of small numbers of cells as well as single-cell RNA sequencing (scRNA-seq) have aided greatly in the study of viruses as these analyses can be used to differentiate responses from infected versus bystander cells in complex systems, including in organoid or animal studies. While protocols for these analyses are typically developed with biosafety level 2 (BSL-2) considerations in mind, such analyses are equally useful for the study of viruses that require higher biosafety containment levels. Many of these workstreams, however, are not directly compatible with the more stringent biosafety regulations of BSL-3 and BSL-4 laboratories ensuring virus inactivation and must therefore be modified. Here we show that TCL buffer (Qiagen), which was developed for bulk sequencing of small numbers of cells and also facilitates scRNA-seq, inactivates both Ebola virus (EBOV) and SARS-CoV-2, BSL-4 and BSL-3 viruses, respectively. We show that additional heat treatment, necessary for the more stringent biosafety concerns for BSL-4-derived samples, was additionally sufficient to inactivate EBOV-containing samples. Critically, this heat treatment had minimal effects on extracted RNA quality and downstream sequencing results. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
19. Labeling Ebola Virus with a Self-Splicing Fluorescent Reporter.
- Author
-
Heiden, Baylee, Mühlberger, Elke, Lennon, Christopher W., and Hume, Adam J.
- Subjects
EBOLA virus ,FLUORESCENT proteins ,VIRAL proteins ,CHIMERIC proteins ,PEPTIDE bonds ,REPORTER genes ,RNA splicing - Abstract
Inteins (intervening proteins) are polypeptides that interrupt the sequence of other proteins and remove themselves through protein splicing. In this intein-catalyzed reaction, the two peptide bonds surrounding the intein are rearranged to release the intein from the flanking protein sequences, termed N- and C-exteins, which are concurrently joined by a peptide bond. Because of this unique functionality, inteins have proven exceptionally useful in protein engineering. Previous work has demonstrated that heterologous proteins can be inserted within an intein, with both the intein and inserted protein retaining function, allowing for intein-containing genes to coexpress additional coding sequences. Here, we show that a fluorescent protein (ZsGreen) can be inserted within the Pyrococcus horikoshii RadA intein, with the hybrid protein (ZsG-Int) maintaining fluorescence and splicing capability. We used this system to create a recombinant Ebola virus expressing a fluorescent protein. We first tested multiple potential insertion sites for ZsG-Int within individual Ebola virus proteins, identifying a site within the VP30 gene that facilitated efficient intein splicing in mammalian cells while also preserving VP30 function. Next, we successfully rescued a virus containing the ZsG-Int-VP30 fusion protein, which displayed fluorescence in the infected cells. We thus report a new intein-based application for adding reporters to systems without the need to add additional genes. Further, this work highlights a novel reporter design, whereby the reporter is only made if the protein of interest is translated and does not remain fused to the protein of interest. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
20. Human airway lineages derived from pluripotent stem cells reveal the epithelial responses to SARS-CoV-2 infection.
- Author
-
Ruobing Wang, Hume, Adam J., Beermann, Mary Lou, Simone-Roach, Chantelle, Lindstrom-Vautrin, Jonathan, Le Suer, Jake, Huang, Jessie, Olejnik, Judith, Villacorta-Martin, Carlos, Bullitt, Esther, Hinds, Anne, Ghaedi, Mahboobe, Rollins, Stuart, Werder, Rhiannon B., Abo, Kristine M., Wilson, Andrew A., Mühlberger, Elke, Kotton, Darrell N., and Hawkins, Finn J.
- Subjects
- *
PLURIPOTENT stem cells , *INDUCED pluripotent stem cells , *EPITHELIAL cells , *SARS-CoV-2 , *ANGIOTENSIN converting enzyme - Abstract
There is an urgent need to understand how SARS-CoV-2 infects the airway epithelium and in a subset of individuals leads to severe illness or death. Induced pluripotent stem cells (iPSCs) provide a near limitless supply of human cells that can be differentiated into cell types of interest, including airway epithelium, for disease modeling. We present a human iPSC-derived airway epithelial platform, composed of the major airway epithelial cell types, that is permissive to SARS-CoV-2 infection. Subsets of iPSC-airway cells express the SARS-CoV-2 entry factors angiotensin-converting enzyme 2 (ACE2), and transmembrane protease serine 2 (TMPRSS2). Multiciliated cells are the primary initial target of SARS-CoV-2 infection. On infection with SARS-CoV-2, iPSC-airway cells generate robust interferon and inflammatory responses, and treatment with remdesivir or camostat mesylate causes a decrease in viral propagation and entry, respectively. In conclusion, iPSC-derived airway cells provide a physiologically relevant in vitro model system to interrogate the pathogenesis of, and develop treatment strategies for, COVID-19 pneumonia. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
21. Recombinant Lloviu virus as a tool to study viral replication and host responses.
- Author
-
Hume, Adam J., Heiden, Baylee, Olejnik, Judith, Suder, Ellen L., Ross, Stephen, Scoon, Whitney A., Bullitt, Esther, Ericsson, Maria, White, Mitchell R., Turcinovic, Jacquelyn, Thao, Tran T. N., Hekman, Ryan M., Kaserman, Joseph E., Huang, Jessie, Alysandratos, Konstantinos-Dionysios, Toth, Gabor E., Jakab, Ferenc, Kotton, Darrell N., Wilson, Andrew A., and Emili, Andrew
- Subjects
- *
RECOMBINANT viruses , *EBOLA virus disease , *VIRAL replication , *PATHOGENIC viruses , *EBOLA virus , *VIRAL tropism , *RNA viruses , *AVIAN influenza A virus - Abstract
Next generation sequencing has revealed the presence of numerous RNA viruses in animal reservoir hosts, including many closely related to known human pathogens. Despite their zoonotic potential, most of these viruses remain understudied due to not yet being cultured. While reverse genetic systems can facilitate virus rescue, this is often hindered by missing viral genome ends. A prime example is Lloviu virus (LLOV), an uncultured filovirus that is closely related to the highly pathogenic Ebola virus. Using minigenome systems, we complemented the missing LLOV genomic ends and identified cis-acting elements required for LLOV replication that were lacking in the published sequence. We leveraged these data to generate recombinant full-length LLOV clones and rescue infectious virus. Similar to other filoviruses, recombinant LLOV (rLLOV) forms filamentous virions and induces the formation of characteristic inclusions in the cytoplasm of the infected cells, as shown by electron microscopy. Known target cells of Ebola virus, including macrophages and hepatocytes, are permissive to rLLOV infection, suggesting that humans could be potential hosts. However, inflammatory responses in human macrophages, a hallmark of Ebola virus disease, are not induced by rLLOV. Additional tropism testing identified pneumocytes as capable of robust rLLOV and Ebola virus infection. We also used rLLOV to test antivirals targeting multiple facets of the replication cycle. Rescue of uncultured viruses of pathogenic concern represents a valuable tool in our arsenal for pandemic preparedness. Author summary: Due to increasing utilization of high-throughput sequencing technologies, RNA sequences of many unknown viruses have been discovered in bats and other animal species. Research on the pathogenic potential of these viruses is hampered by incomplete viral genome sequences and difficulties in isolating infectious virus from the animal hosts. One example of these potentially zoonotic pathogens is Lloviu virus (LLOV), a filovirus which is closely related to Ebola virus. Here we applied molecular virological approaches, including minigenome assays, to complement the incomplete LLOV genome ends with sequences from related viruses and identify cis-acting elements required for LLOV replication and transcription that were missing in the published LLOV sequence. The resulting full-length clones were used to generate infectious recombinant LLOV. We used this virus for electron microscopic analyses, infection studies in human cells, host response analysis, and antiviral drug testing. Our results provide new insights into the pathogenic potential of LLOV and delineate a roadmap for studying uncultured viruses. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
22. A virus-specific monocyte inflammatory phenotype is induced by SARS-CoV-2 at the immune-epithelial interface.
- Author
-
Leon, Juliette, Michelson, Daniel A., Olejnik, Judith, Chowdhary, Kaitavjeet, Hyung Suk Oh, Hume, Adam J., Galván-Peña, Silvia, Yangyang Zhu, Chen, Felicia, Vijaykumar, Brinda, Liang Yang, Crestani, Elena, Yonker, Lael M., Knipe, David M., Mühlberger, Elke, and Benoist, Christophe
- Subjects
SARS-CoV-2 ,COMMERCIAL products ,COVID-19 ,MYELOID cells - Abstract
Infection by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) provokes a potentially fatal pneumonia with multiorgan failure, and high systemic inflammation. To gain mechanistic insight and ferret out the root of this immune dysregulation, we modeled, by in vitro coculture, the interactions between infected epithelial cells and immunocytes. A strong response was induced in monocytes and B cells, with a SARS-CoV-2-specific inflammatory gene cluster distinct from that seen in influenza A or Ebola virus-infected cocultures, and which reproduced deviations reported in blood or lung myeloid cells from COVID-19 patients. A substantial fraction of the effect could be reproduced after individual transfection of several SARS-CoV-2 proteins (Spike and some nonstructural proteins), mediated by soluble factors, but not via transcriptional induction. This response was greatly muted in monocytes from healthy children, perhaps a clue to the age dependency of COVID-19. These results suggest that the inflammatory malfunction in COVID-19 is rooted in the earliest perturbations that SARS-CoV-2 induces in epithelia. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
23. Early Developmental 2,3,7,8-Tetrachlorodibenzo-p-Dioxin Exposure Decreases Chick Embryo Heart Chronotropic Response to Isoproterenol but Not to Agents Affecting Signals Downstream of the Beta-Adrenergic Receptor
- Author
-
Sommer, Rebecca J., Hume, Adam J., Ciak, Jessica M., VanNostrand, John J., Friggens, Megan, and Walker, Mary K.
- Published
- 2005
24. Regulation of the retinoblastoma proteins by the human herpesviruses
- Author
-
Kalejta Robert F and Hume Adam J
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 ,Cytology ,QH573-671 - Abstract
Abstract Viruses are obligate intracellular parasites that alter the environment of infected cells in order to replicate more efficiently. One way viruses achieve this is by modulating cell cycle progression. The main regulators of progression out of G0, through G1, and into S phase are the members of the retinoblastoma (Rb) family of tumor suppressors. Rb proteins repress the transcription of genes controlled by the E2F transcription factors. Because the expression of E2F-responsive genes is required for cell cycle progression into the S phase, Rb arrests the cell cycle in G0/G1. A number of viral proteins directly target Rb family members for inactivation, presumably to create an environment more hospitable for viral replication. Such viral proteins include the extensively studied oncoproteins E7 (from human papillomavirus), E1A (from adenovirus), and the large T (tumor) antigen (from simian virus 40). Elucidating how these three viral proteins target and inactivate Rb has proven to be an invaluable approach to augment our understanding of both normal cell cycle progression and carcinogenesis. In addition to these proteins, a number of other virally-encoded inactivators of the Rb family have subsequently been identified including a surprising number encoded by human herpesviruses. Here we review how the human herpesviruses modulate Rb function during infection, introduce the individual viral proteins that directly or indirectly target Rb, and speculate about what roles Rb modulation by these proteins may play in viral replication, pathogenesis, and oncogenesis.
- Published
- 2009
- Full Text
- View/download PDF
25. MHC class II transactivator CIITA induces cell resistance to Ebola virus and SARS-like coronaviruses.
- Author
-
Bruchez, Anna, Sha, Ky, Johnson, Joshua, Chen, Li, Stefani, Caroline, McConnell, Hannah, Gaucherand, Lea, Prins, Rachel, Matreyek, Kenneth A., Hume, Adam J., Mühlberger, Elke, Schmidt, Emmett V., Olinger, Gene G., Stuart, Lynda M., and Lacy-Hulbert, Adam
- Published
- 2020
- Full Text
- View/download PDF
26. Egyptian Rousette IFN-ω Subtypes Elicit Distinct Antiviral Effects and Transcriptional Responses in Conspecific Cells.
- Author
-
Pavlovich, Stephanie S., Darling, Tamarand, Hume, Adam J., Davey, Robert A., Feng, Feng, Mühlberger, Elke, and Kepler, Thomas B.
- Subjects
MARBURG virus ,HUMAN genome ,SYMPTOMS ,RNA viruses ,T cell receptors ,BATS - Abstract
Bats host a number of viruses that cause severe disease in humans without experiencing overt symptoms of disease themselves. While the mechanisms underlying this ability to avoid sickness are not known, deep sequencing studies of bat genomes have uncovered genetic adaptations that may have functional importance in the antiviral response of these animals. Egyptian rousette bats (Rousettus aegyptiacus) are the natural reservoir hosts of Marburg virus (MARV). In contrast to humans, these bats do not become sick when infected with MARV. A striking difference to the human genome is that Egyptian rousettes have an expanded repertoire of IFNW genes. To probe the biological implications of this expansion, we synthesized IFN-ω4 and IFN-ω9 proteins and tested their antiviral activity in Egyptian rousette cells. Both IFN-ω4 and IFN-ω9 showed antiviral activity against RNA viruses, including MARV, with IFN-ω9 being more efficient than IFN-ω4. Using RNA-Seq, we examined the transcriptional response induced by each protein. Although the sets of genes induced by the two IFNs were largely overlapping, IFN-ω9 induced a more rapid and intense response than did IFN-ω4. About 13% of genes induced by IFN-ω treatment are not found in the Interferome or other ISG databases, indicating that they may be uniquely IFN-responsive in this bat. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
27. Distinct Genome Replication and Transcription Strategies within the Growing Filovirus Family.
- Author
-
Hume, Adam J. and Mühlberger, Elke
- Subjects
- *
MARINE fishes , *EBOLA virus , *FILOVIRIDAE , *PROTEIN domains , *GENOMES - Abstract
Research on filoviruses has historically focused on the highly pathogenic ebola- and marburgviruses. Indeed, until recently, these were the only two genera in the filovirus family. Recent advances in sequencing technologies have facilitated the discovery of not only a new ebolavirus, but also three new filovirus genera and a sixth proposed genus. While two of these new genera are similar to the ebola- and marburgviruses, the other two, discovered in saltwater fishes, are considerably more diverse. Nonetheless, these viruses retain a number of key features of the other filoviruses. Here, we review the key characteristics of filovirus replication and transcription, highlighting similarities and differences between the viruses. In particular, we focus on key regulatory elements in the genomes, replication and transcription strategies, and the conservation of protein domains and functions among the viruses. In addition, using computational analyses, we were able to identify potential homology and functions for some of the genes of the novel filoviruses with previously unknown functions. Although none of the newly discovered filoviruses have yet been isolated, initial studies of some of these viruses using minigenome systems have yielded insights into their mechanisms of replication and transcription. In general, the Cuevavirus and proposed Dianlovirus genera appear to follow the transcription and replication strategies employed by the ebola- and marburgviruses, respectively. While our knowledge of the fish filoviruses is currently limited to sequence analysis, the lack of certain conserved motifs and even entire genes necessitates that they have evolved distinct mechanisms of replication and transcription. Unlabelled Image [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
28. Toll-like receptor 4 in acute viral infection: Too much of a good thing.
- Author
-
Olejnik, Judith, Hume, Adam J., and Mühlberger, Elke
- Subjects
- *
TOLL-like receptors , *INFLAMMATION , *VIRUS diseases , *LIGANDS (Biochemistry) , *IMMUNE response - Abstract
The article examines the role of toll-like receptor 4 (TLR4) in damaging inflammatory responses during acute viral infections. Topics discussed include the ligands of TLR4, activation of viruses that induce an inflammatory response during acute infection through TLR4, and the therapeutic potential of TLR4. The role of over-stimulation of TLR4 in excessive inflammatory response that is damaging to the host and its benefits to the host by establishing a protective immune response are explained.
- Published
- 2018
- Full Text
- View/download PDF
29. Correction: Recombinant Lloviu virus as a tool to study viral replication and host responses.
- Author
-
Hume, Adam J., Heiden, Baylee, Olejnik, Judith, Suder, Ellen L., Ross, Stephen, Scoon, Whitney A., Bullitt, Esther, Ericsson, Maria, White, Mitchell R., Turcinovic, Jacquelyn, Thao, Tran T. N., Hekman, Ryan M., Kaserman, Joseph E., Huang, Jessie, Alysandratos, Konstantinos-Dionysios, Toth, Gabor E., Jakab, Ferenc, Kotton, Darrell N., Wilson, Andrew A., and Emili, Andrew
- Subjects
- *
RECOMBINANT viruses , *VIRAL replication - Abstract
Reference 1 Hume AJ, Heiden B, Olejnik J, Suder EL, Ross S, Scoon WA, et al. (2022) Recombinant Lloviu virus as a tool to study viral replication and host responses. The x-axis label for panel D of Fig 5 is incorrect. [Extracted from the article]
- Published
- 2022
- Full Text
- View/download PDF
30. Inactivation of RNA Viruses by Gamma Irradiation: A Study on Mitigating Factors.
- Author
-
Hume, Adam J., Ames, Joshua, Rennick, Linda J., Duprex, W. Paul, Marzi, Andrea, Tonkiss, John, and Mühlberger, Elke
- Subjects
- *
RNA viruses , *BIOSAFETY , *PATHOGENIC microorganisms , *VIRION , *RECOMBINANT viruses , *FLUORESCENT proteins - Abstract
Effective inactivation of biosafety level 4 (BSL-4) pathogens is vital in order to study these agents safely. Gamma irradiation is a commonly used method for the inactivation of BSL-4 viruses, which among other advantages, facilitates the study of inactivated yet morphologically intact virions. The reported values for susceptibility of viruses to inactivation by gamma irradiation are sometimes inconsistent, likely due to differences in experimental protocols. We analyzed the effects of common sample attributes on the inactivation of a recombinant vesicular stomatitis virus expressing the Zaire ebolavirus glycoprotein and green fluorescent protein. Using this surrogate virus, we found that sample volume and protein content of the sample modulated viral inactivation by gamma irradiation but that air volume within the sample container and the addition of external disinfectant surrounding the sample did not. These data identify several factors which alter viral susceptibility to inactivation and highlight the usefulness of lower biosafety level surrogate viruses for such studies. Our results underscore the need to validate inactivation protocols of BSL-4 pathogens using “worst-case scenario” procedures to ensure complete sample inactivation. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
31. Sa1125: USING HUMAN IPSC DERIVED INTESTINAL ORGANOIDS TO MODEL INFECTIOUS DISEASES.
- Author
-
Mithal, Aditya, Hume, Adam J., Mühlberger, Elke, and Mostoslavsky, Gustavo
- Published
- 2022
- Full Text
- View/download PDF
32. Interferon‐alpha or ‐beta Facilitates SARS‐CoV‐2 Pulmonary Vascular Infection by Inducing ACE2.
- Author
-
Yuan, Ke, Klouda, Timothy, Yuan, Hao, Kim, Hyunbum, Kim, Jiwon, Olejnik, Judith, Hume, Adam J., Melero‐Martin, Juan, Mühlberger, Elke, Jia, Hongpeng, Padera, Robert F., and Raby, Benjamin A.
- Published
- 2022
- Full Text
- View/download PDF
33. Forty-Five Years of Marburg Virus Research.
- Author
-
Brauburger, Kristina, Hume, Adam J., Mühlberger, Elke, and Olejnik, Judith
- Subjects
- *
HEMORRHAGIC fever , *EPIDEMICS , *MARBURG virus , *MARBURG virus disease , *MOLECULAR biology - Abstract
In 1967, the first reported filovirus hemorrhagic fever outbreak took place in Germany and the former Yugoslavia. The causative agent that was identified during this outbreak, Marburg virus, is one of the most deadly human pathogens. This article provides a comprehensive overview of our current knowledge about Marburg virus disease ranging from ecology to pathogenesis and molecular biology. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
34. Activation of STAT3-mediated ciliated cell survival protects against severe infection by respiratory syncytial virus.
- Author
-
Caiqi Zhao, Yan Bai, Wei Wang, Amonkar, Gaurang M., Hongmei Mou, Olejnik, Judith, Hume, Adam J., Mühlberger, Elke, Lukacs, Nicholas W., Fearns, Rachel, Lerou, Paul H., and Xingbin Ai
- Subjects
- *
RESPIRATORY syncytial virus infections , *BRONCHIOLITIS , *CELL survival , *VIRAL transmission , *INFANT diseases , *PNEUMONIA - Abstract
Respiratory syncytial virus (RSV) selectively targets ciliated cells in human bronchial epithelium and can cause bronchiolitis and pneumonia, mostly in infants. To identify molecular targets of intervention during RSV infection in infants, we investigated how age regulates RSV interaction with the bronchial epithelium barrier. Employing precision-cut lung slices and air-liquid interface cultures generated from infant and adult human donors, we found robust RSV virus spread and extensive apoptotic cell death only in infant bronchial epithelium. In contrast, adult bronchial epithelium showed no barrier damage and limited RSV infection. Single nuclear RNA-Seq revealed age-related insufficiency of an antiapoptotic STAT3 activation response to RSV infection in infant ciliated cells, which was exploited to facilitate virus spread via the extruded apoptotic ciliated cells carrying RSV. Activation of STAT3 and blockade of apoptosis rendered protection against severe RSV infection in infant bronchial epithelium. Lastly, apoptotic inhibitor treatment of a neonatal mouse model of RSV infection mitigated infection and inflammation in the lung. Taken together, our findings identify a STAT3-mediated antiapoptosis pathway as a target to battle severe RSV disease in infants. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
35. Actionable Cytopathogenic Host Responses of Human Alveolar Type 2 Cells to SARS-CoV-2.
- Author
-
Hekman, Ryan M., Hume, Adam J., Goel, Raghuveera Kumar, Abo, Kristine M., Huang, Jessie, Blum, Benjamin C., Werder, Rhiannon Bree, Suder, Ellen L., Paul, Indranil, Phanse, Sadhna, Youssef, Ahmed, Alysandratos, Kostantinos D., Padhorny, Dzmitry, Ojha, Sandeep, Mora-Martin, Alexandra, Kretov, Dmitry, Ash, Peter, Varma, Mamta, Zhao, Jian, and Patten, J.J.
- Subjects
- *
PROTEOMICS , *SARS-CoV-2 , *PLURIPOTENT stem cells , *COVID-19 pandemic , *MIDDLE East respiratory syndrome , *CELL communication , *COVID-19 - Abstract
Human transmission of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), causative pathogen of the COVID-19 pandemic, exerts a massive health and socioeconomic crisis. The virus infects alveolar epithelial type 2 cells (AT2s), leading to lung injury and impaired gas exchange, but the mechanisms driving infection and pathology are unclear. We performed a quantitative phosphoproteomic survey of induced pluripotent stem cell-derived AT2s (iAT2s) infected with SARS-CoV-2 at air-liquid interface (ALI). Time course analysis revealed rapid remodeling of diverse host systems, including signaling, RNA processing, translation, metabolism, nuclear integrity, protein trafficking, and cytoskeletal-microtubule organization, leading to cell cycle arrest, genotoxic stress, and innate immunity. Comparison to analogous data from transformed cell lines revealed respiratory-specific processes hijacked by SARS-CoV-2, highlighting potential novel therapeutic avenues that were validated by a high hit rate in a targeted small molecule screen in our iAT2 ALI system. • SARS-CoV-2 infection in induced lung cells is characterized by phosphoproteomics • Analysis of response reveals host cell signaling and protein expression profile • Comparison to studies in undifferentiated cell lines shows unique pathology in iAT2s • Systems-level predictions find druggable pathways that can impede viral life cycle Hekman et al. describe how a layer of primary stem cells (iAT2s) recapitulating lung biology responds to infection with SARS-CoV-2. They compare their work to previous studies with immortalized cell lines. Their data predict what effect the virus has on a lung cell and which drugs may slow infection. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
36. A Chimeric Lloviu Virus Minigenome System Reveals that the Bat-Derived Filovirus Replicates More Similarly to Ebolaviruses than Marburgviruses.
- Author
-
Manhart, Whitney A., Pacheco, Jennifer R., Hume, Adam J., Cressey, Tessa N., Deflubé, Laure R., and Mühlberger, Elke
- Abstract
Summary Recently, traces of zoonotic viruses have been discovered in bats and other species around the world, but despite repeated attempts, full viral genomes have not been rescued. The absence of critical genetic sequences from these viruses and the difficulties to isolate infectious virus from specimens prevent research on their pathogenic potential for humans. One example of these zoonotic pathogens is Lloviu virus (LLOV), a filovirus that is closely related to Ebola virus. Here, we established LLOV minigenome systems based on sequence complementation from other filoviruses. Our results show that the LLOV replication and transcription mechanisms are, in general, more similar to ebolaviruses than to marburgviruses. We also show that a single nucleotide at the 3′ genome end determines species specificity of the LLOV polymerase. The data obtained here will be instrumental for the rescue of infectious LLOV clones for pathogenesis studies. Graphical Abstract Highlights • The LLOV replication complex supports replication of chimeric LLOV minigenomes • LLOV utilizes an ebolavirus-like replication strategy • LLOV does not recognize the marburgvirus leader sequence • The terminal genomic 3′ nucleotides determine the specificity of the LLOV polymerase Lloviu virus (LLOV) is a filovirus of unknown pathogenicity, and the viral genome ends have not been recovered. Manhart et al. established a minigenome system for studying LLOV by complementing missing sequence information with that of other filoviruses. This minigenome provides a blueprint for generating recombinant LLOV for pathogenesis studies. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
37. Ebolaviruses Associated with Differential Pathogenicity Induce Distinct Host Responses in Human Macrophages.
- Author
-
Olejnik, Judith, Forero, Forero, Deflubé, Laure R., Hume, Adam J., Manhart, Whitney A., Nishida, Andrew, Marzi, Andrea, Katze, Michael G., Ebihara, Hideki, Rasmussen, Angela L., and Mühlberger, Elke
- Subjects
- *
MACROPHAGES , *MICROBIAL virulence , *PHAGOCYTES , *RETICULO-endothelial system , *PHAGOCYTOSIS , *MULTINUCLEATED giant cells - Abstract
Ebola virus (EBOV) and Reston virus (RESTV) are members of the Ebolavirus genus which greatly differ in their pathogenicity. While EBOV causes a severe disease in humans characterized by a dysregulated inflammatory response and elevated cytokine and chemokine production, there are no reported disease-associated human cases of RESTV infection, suggesting that RESTV is nonpathogenic for humans. The underlying mechanisms determining the pathogenicity of different ebolavirus species are not yet known. In this study, we dissected the host response to EBOV and RESTV infection in primary human monocyte-derived macrophages (MDMs). As expected, EBOV infection led to a profound proinflammatory response, including strong induction of type I and type III interferons (IFNs). In contrast, RESTV-infected macrophages remained surprisingly silent. Early activation of IFN regulatory factor 3 (IRF3) and NF-kB was observed in EBOV-infected, but not in RESTV-infected, MDMs. In concordance with previous results, MDMs treated with inactivated EBOV and Ebola virus-like particles (VLPs) induced NF--B activation mediated by Toll-like receptor 4 (TLR4) in a glycoprotein (GP)-dependent manner. This was not the case in cells exposed to live RESTV, inactivated RESTV, or VLPs containing RESTV GP, indicating that RESTV GP does not trigger TLR4 signaling. Our results suggest that the lack of immune activation in RESTV-infected MDMs contributes to lower pathogenicity by preventing the cytokine storm observed in EBOV infection. We further demonstrate that inhibition of TLR4 signaling abolishes EBOV GP-mediated NF-kB activation. This finding indicates that limiting the excessive TLR4-mediated proinflammatory response in EBOV infection should be considered as a potential supportive treatment option for EBOV disease. IMPORTANCE Emerging infectious diseases are a major public health concern, as exemplified by the recent devastating Ebola virus (EBOV) outbreak. Different ebolavirus species are associated with widely varying pathogenicity in humans, ranging from asymptomatic infections for Reston virus (RESTV) to severe disease with fatal outcomes for EBOV. In this comparative study of EBOV- and RESTV-infected human macrophages, we identified key differences in host cell responses. Consistent with previous data, EBOV infection is associated with a proinflammatory signature triggered by the surface glycoprotein (GP), which can be inhibited by blocking TLR4 signaling. In contrast, infection with RESTV failed to stimulate a strong host response in infected macrophages due to the inability of RESTV GP to stimulate TLR4. We propose that disparate proinflammatory host signatures contribute to the differences in pathogenicity reported for ebolavirus species and suggest that proinflammatory pathways represent an intriguing target for the development of novel therapeutics. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
38. Ebola Virus Does Not Induce Stress Granule Formation during Infection and Sequesters Stress Granule Proteins within Viral Inclusions.
- Author
-
Nelson, Emily V., Schmidt, Kristina M., Deflubé, Laure R., Doğanay, Sultan, Banadyga, Logan, Olejnik, Judith, Hume, Adam J., Ryabchikova, Elena, Ebihara, Hideki, Kedersha, Nancy, Taekjip Ha, and Mühlberger, Elke
- Subjects
- *
EBOLA virus , *GRANULE cells , *NUCLEOCAPSIDS , *VIRAL replication , *INITIATION factors (Biochemistry) , *PHOSPHORYLATION , *SODIUM arsenite - Abstract
A hallmark of Ebola virus (EBOV) infection is the formation of viral inclusions in the cytoplasm of infected cells. These viral inclusions contain the EBOV nucleocapsids and are sites of viral replication and nucleocapsid maturation. Although there is growing evidence that viral inclusions create a protected environment that fosters EBOV replication, little is known about their role in the host response to infection. The cellular stress response is an effective antiviral strategy that leads to stress granule (SG) formation and translational arrest mediated by the phosphorylation of a translation initiation factor, the subunit of eukaryotic initiation factor 2 (eIF2α). Here, we show that selected SG proteins are sequestered within EBOV inclusions, where they form distinct granules that colocalize with viral RNA. These inclusion-bound (IB) granules are functionally and structurally different from canonical SGs. Formation of IB granules does not indicate translational arrest in the infected cells. We further show that EBOV does not induce formation of canonical SGs or eIF2α phosphorylation at any time postinfection but is unable to fully inhibit SG formation induced by different exogenous stressors, including sodium arsenite, heat, and hippuristanol. Despite the sequestration of SG marker proteins into IB granules, canonical SGs are unable to form within inclusions, which we propose might be mediated by a novel function of VP35, which disrupts SG formation. This function is independent of VP35's RNA binding activity. Further studies aim to reveal the mechanism for SG protein sequestration and precise function within inclusions. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
39. The Egyptian Rousette Genome Reveals Unexpected Features of Bat Antiviral Immunity.
- Author
-
Pavlovich, Stephanie S., Lovett, Sean P., Koroleva, Galina, Guito, Jonathan C., Arnold, Catherine E., Nagle, Elyse R., Kulcsar, Kirsten, Lee, Albert, Thibaud-Nissen, Françoise, Hume, Adam J., Mühlberger, Elke, Uebelhoer, Luke S., Towner, Jonathan S., Rabadan, Raul, Sanchez-Lockhart, Mariano, Kepler, Thomas B., and Palacios, Gustavo
- Subjects
- *
ROUSETTUS aegyptiacus , *MAMMAL genomes , *ANTIVIRAL agents , *VIRUS diseases , *MAMMAL evolution , *TYPE I interferons - Abstract
Summary Bats harbor many viruses asymptomatically, including several notorious for causing extreme virulence in humans. To identify differences between antiviral mechanisms in humans and bats, we sequenced, assembled, and analyzed the genome of Rousettus aegyptiacus , a natural reservoir of Marburg virus and the only known reservoir for any filovirus. We found an expanded and diversified KLRC/KLRD family of natural killer cell receptors, MHC class I genes, and type I interferons, which dramatically differ from their functional counterparts in other mammals. Such concerted evolution of key components of bat immunity is strongly suggestive of novel modes of antiviral defense. An evaluation of the theoretical function of these genes suggests that an inhibitory immune state may exist in bats. Based on our findings, we hypothesize that tolerance of viral infection, rather than enhanced potency of antiviral defenses, may be a key mechanism by which bats asymptomatically host viruses that are pathogenic in humans. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
40. Activation of STAT3-mediated ciliated cell survival protects against severe infection by respiratory syncytial virus.
- Author
-
Zhao C, Bai Y, Wang W, Amonkar GM, Mou H, Olejnik J, Hume AJ, Mühlberger E, Lukacs NW, Fearns R, Lerou PH, and Ai X
- Subjects
- Humans, Animals, Mice, Infant, Cell Survival, Respiratory Mucosa virology, Respiratory Mucosa metabolism, Respiratory Mucosa pathology, Female, Male, Adult, Cilia metabolism, Cilia pathology, Cilia virology, STAT3 Transcription Factor metabolism, STAT3 Transcription Factor genetics, Respiratory Syncytial Virus Infections virology, Respiratory Syncytial Virus Infections pathology, Respiratory Syncytial Virus Infections metabolism, Respiratory Syncytial Virus Infections genetics, Apoptosis, Respiratory Syncytial Virus, Human physiology
- Abstract
Respiratory syncytial virus (RSV) selectively targets ciliated cells in human bronchial epithelium and can cause bronchiolitis and pneumonia, mostly in infants. To identify molecular targets of intervention during RSV infection in infants, we investigated how age regulates RSV interaction with the bronchial epithelium barrier. Employing precision-cut lung slices and air-liquid interface cultures generated from infant and adult human donors, we found robust RSV virus spread and extensive apoptotic cell death only in infant bronchial epithelium. In contrast, adult bronchial epithelium showed no barrier damage and limited RSV infection. Single nuclear RNA-Seq revealed age-related insufficiency of an antiapoptotic STAT3 activation response to RSV infection in infant ciliated cells, which was exploited to facilitate virus spread via the extruded apoptotic ciliated cells carrying RSV. Activation of STAT3 and blockade of apoptosis rendered protection against severe RSV infection in infant bronchial epithelium. Lastly, apoptotic inhibitor treatment of a neonatal mouse model of RSV infection mitigated infection and inflammation in the lung. Taken together, our findings identify a STAT3-mediated antiapoptosis pathway as a target to battle severe RSV disease in infants.
- Published
- 2024
- Full Text
- View/download PDF
41. A high-throughput, polymerase-targeted RT-PCR for broad detection of mammalian filoviruses.
- Author
-
Cui N, Perez YL, Hume AJ, Nunley BE, Kong K, Mills MG, Xie H, and Greninger AL
- Subjects
- Humans, Animals, RNA, Viral genetics, Sensitivity and Specificity, High-Throughput Nucleotide Sequencing methods, Mammals virology, High-Throughput Screening Assays methods, Viral Proteins genetics, Filoviridae genetics, Filoviridae isolation & purification, Filoviridae classification, Filoviridae Infections virology, Reverse Transcriptase Polymerase Chain Reaction methods
- Abstract
Filoviruses are some of the most lethal viruses in the modern world, and increasing numbers of filovirus species and genera have been discovered in recent years. Despite the potential severity of filovirus outbreaks in the human population, comparably few sensitive pan-filovirus RT-PCR assays have been described that might facilitate early detection and prevention. Here, we present a new pan-filovirus RT-PCR assay targeting the L polymerase gene for detection of all known mammalian filoviruses. We demonstrate the detection of 10 synthetic filovirus RNA templates with analytical sensitivity ranging from 178 to 3,354 copies/mL, without cross-reactivity on 10 non-filoviral human viral species. We verified assay performance on 10 inactivated filovirus isolates, yielding initial sensitivities of 0.012-44.17 TCID
50 /mL. We coupled this broadly reactive RT-PCR with a deep sequencing workflow that is amenable to high-throughput pooling to maximize detection and discovery potential. In summary, this pan-filovirus RT-PCR assay targets the most conserved filovirus gene, offers the widest breadth of coverage to date, and may help in the detection and discovery of novel filoviruses.IMPORTANCEFiloviruses remain some of the most mysterious viruses known to the world, with extremely high lethality rates and significant pandemic potential. Yet comparably few filovirus species and genera have been discovered to date and questions surround the definitive host species for zoonotic infections. Here, we describe a novel broadly reactive RT-PCR assay targeting the conserved L polymerase gene for high-throughput screening for filoviruses in a variety of clinical and environmental specimens. We demonstrate the assay can detect all known mammalian filoviruses and determine the sensitivity and specificity of the assay on synthetic RNA sequences, inactivated filovirus isolates, and non-filoviral species., Competing Interests: The authors declare no conflict of interest.- Published
- 2024
- Full Text
- View/download PDF
42. Improved protein splicing through viral passaging.
- Author
-
Hume AJ, Deeney DJ, Smetana JS, Turcinovic J, Connor JH, Belfort M, Mühlberger E, and Lennon CW
- Subjects
- Humans, Virus Replication, Viral Proteins genetics, Viral Proteins metabolism, Genes, Reporter, Inteins genetics, Protein Splicing, Ebolavirus genetics, Ebolavirus physiology
- Abstract
In tervening pro teins (inteins) are translated as subdomains within host proteins and removed through an intein-driven splicing reaction where the flanking sequences (exteins) are joined with a peptide bond. Previously, we developed a self-removing translation reporter for labeling Ebola virus (EBOV). In this reporter, an intein (RadA) containing the fluorescent protein ZsGreen (ZsG) is inserted within the EBOV protein VP30. Upon VP30-RadA-ZsG expression from the viral genome, RadA-ZsG is removed from VP30 through the protein splicing activity of RadA, generating functional, non-tagged VP30 and functional ZsGreen. While incorporation of our VP30-RadA-ZsG fusion reporter into recombinant EBOV (rEBOV-RadA-ZsG) resulted in an infectious virus that expresses ZsG upon infection of cells, this virus displayed a replication defect compared to wild-type EBOV, which might be the result of insufficient RadA splicing. Here, we demonstrate that the serial passaging of rEBOV-RadA-ZsG in human cells led to an increase in replication efficiency compared to unpassaged rEBOV-RadA-ZsG. Sequencing of passaged viruses revealed intein-specific mutations. These mutations improve intein activity in both prokaryotic and eukaryotic systems, as well as in multiple extein contexts. Taken together, our findings offer a novel means to select for inteins with enhanced catalytic properties that appear independent of extein context and expression system.IMPORTANCE In tervening pro teins (inteins) are self-removing protein elements that have been utilized to develop a variety of innovative protein engineering technologies. Here, we report the isolation of inteins with improved catalytic activity through viral passaging. Specifically, we inserted a highly active intein within an essential protein of Ebola virus and serially passaged this recombinant virus, which led to intein-specific hyper-activity mutations. The identified mutations showed improved intein activity within both bacterial and eukaryotic expression systems and in multiple extein contexts. These results present a new strategy for developing inteins with improved splicing activity., Competing Interests: The authors declare no conflict of interest.
- Published
- 2024
- Full Text
- View/download PDF
43. PARP14 is pro- and anti-viral host factor that promotes IFN production and affects the replication of multiple viruses.
- Author
-
Parthasarathy S, Saenjamsai P, Hao H, Ferkul A, Pfannenstiel JJ, Suder EL, Bejan DS, Chen Y, Schwarting N, Aikawa M, Muhlberger E, Orozco RC, Sullivan CS, Cohen MS, Davido DJ, Hume AJ, and Fehr AR
- Abstract
PARP14 is a 203 kDa multi-domain protein that is primarily known as an ADP-ribosyltransferase, and is involved in a variety of cellular functions including DNA damage, microglial activation, inflammation, and cancer progression. In addition, PARP14 is upregulated by interferon (IFN), indicating a role in the antiviral response. Furthermore, PARP14 has evolved under positive selection, again indicating that it is involved in host-pathogen conflict. We found that PARP14 is required for increased IFN-I production in response to coronavirus infection lacking ADP-ribosylhydrolase (ARH) activity and poly(I:C), however, whether it has direct antiviral function remains unclear. Here we demonstrate that the catalytic activity of PARP14 enhances IFN-I and IFN-III responses and restricts ARH-deficient murine hepatitis virus (MHV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) replication. To determine if PARP14's antiviral functions extended beyond CoVs, we tested the ability of herpes simplex virus 1 (HSV-1) and several negative-sense RNA viruses, including vesicular stomatitis virus (VSV), Ebola virus (EBOV), and Nipah virus (NiV), to infect A549 PARP14 knockout (KO) cells. HSV-1 had increased replication in PARP14 KO cells, indicating that PARP14 restricts HSV-1 replication. In contrast, PARP14 was critical for the efficient infection of VSV, EBOV, and NiV, with EBOV infectivity at less than 1% of WT cells. A PARP14 active site inhibitor had no impact on HSV-1 or EBOV infection, indicating that its effect on these viruses was independent of its catalytic activity. These data demonstrate that PARP14 promotes IFN production and has both pro- and anti-viral functions targeting multiple viruses., Competing Interests: Competing interests: ARF is in the process of submitting a patent application for use of SARS-CoV-2 ΔMac1 virus as a live-attenuated vaccine.
- Published
- 2024
- Full Text
- View/download PDF
44. ICTV Virus Taxonomy Profile: Filoviridae 2024.
- Author
-
Biedenkopf N, Bukreyev A, Chandran K, Di Paola N, Formenty PBH, Griffiths A, Hume AJ, Mühlberger E, Netesov SV, Palacios G, Pawęska JT, Smither S, Takada A, Wahl V, and Kuhn JH
- Subjects
- Animals, Humans, Phylogeny, Genome, Viral, Virus Replication, Mammals genetics, Ebolavirus genetics, Rhabdoviridae genetics, Marburgvirus
- Abstract
Filoviridae is a family of negative-sense RNA viruses with genomes of about 13.1-20.9 kb that infect fish, mammals and reptiles. The filovirid genome is a linear, non-segmented RNA with five canonical open reading frames (ORFs) that encode a nucleoprotein (NP), a polymerase cofactor (VP35), a glycoprotein (GP
1,2 ), a transcriptional activator (VP30) and a large protein (L) containing an RNA-directed RNA polymerase (RdRP) domain. All filovirid genomes encode additional proteins that vary among genera. Several filovirids (e.g., Ebola virus, Marburg virus) are pathogenic for humans and highly virulent. This is a summary of the International Committee on Taxonomy of Viruses (ICTV) Report on the family Filoviridae , which is available at www.ictv.global/report/filoviridae.- Published
- 2024
- Full Text
- View/download PDF
45. Minor intron-containing genes as an ancient backbone for viral infection?
- Author
-
Wuchty S, White AK, Olthof AM, Drake K, Hume AJ, Olejnik J, Aguiar-Pulido V, Mühlberger E, and Kanadia RN
- Abstract
Minor intron-containing genes (MIGs) account for <2% of all human protein-coding genes and are uniquely dependent on the minor spliceosome for proper excision. Despite their low numbers, we surprisingly found a significant enrichment of MIG-encoded proteins (MIG-Ps) in protein-protein interactomes and host factors of positive-sense RNA viruses, including SARS-CoV-1, SARS-CoV-2, MERS coronavirus, and Zika virus. Similarly, we observed a significant enrichment of MIG-Ps in the interactomes and sets of host factors of negative-sense RNA viruses such as Ebola virus, influenza A virus, and the retrovirus HIV-1. We also found an enrichment of MIG-Ps in double-stranded DNA viruses such as Epstein-Barr virus, human papillomavirus, and herpes simplex viruses. In general, MIG-Ps were highly connected and placed in central positions in a network of human-host protein interactions. Moreover, MIG-Ps that interact with viral proteins were enriched with essential genes. We also provide evidence that viral proteins interact with ancestral MIGs that date back to unicellular organisms and are mainly involved in basic cellular functions such as cell cycle, cell division, and signal transduction. Our results suggest that MIG-Ps form a stable, evolutionarily conserved backbone that viruses putatively tap to invade and propagate in human host cells., (© The Author(s) 2024. Published by Oxford University Press on behalf of National Academy of Sciences.)
- Published
- 2024
- Full Text
- View/download PDF
46. Age-related STAT3 signaling regulates severity of respiratory syncytial viral infection in human bronchial epithelial cells.
- Author
-
Zhao C, Wang W, Bai Y, Amonkar G, Mou H, Olejnik J, Hume AJ, Mühlberger E, Fang Y, Que J, Fearns R, Ai X, and Lerou PH
- Abstract
Respiratory syncytial virus (RSV) can cause severe disease especially in infants; however, mechanisms of age-associated disease severity remain elusive. Here, employing human bronchial epithelium models generated from tracheal aspirate-derived basal stem cells of neonates and adults, we investigated whether age regulates RSV-epithelium interaction to determine disease severity. We show that following RSV infection, only neonatal epithelium model exhibited cytopathy and mucus hyperplasia, and neonatal epithelium had more robust viral spread and inflammatory responses than adult epithelium. Mechanistically, RSV-infected neonatal ciliated cells displayed age-related impairment of STAT3 activation, rendering susceptibility to apoptosis, which facilitated viral spread. In contrast, SARS-CoV-2 infection of ciliated cells had no effect on STAT3 activation and was not affected by age. Taken together, our findings identify an age-related and RSV-specific interaction with neonatal bronchial epithelium that critically contributes to severity of infection, and STAT3 activation offers a potential strategy to battle severe RSV disease in infants.
- Published
- 2023
- Full Text
- View/download PDF
47. Annual (2023) taxonomic update of RNA-directed RNA polymerase-encoding negative-sense RNA viruses (realm Riboviria : kingdom Orthornavirae : phylum Negarnaviricota ).
- Author
-
Kuhn JH, Abe J, Adkins S, Alkhovsky SV, Avšič-Županc T, Ayllón MA, Bahl J, Balkema-Buschmann A, Ballinger MJ, Kumar Baranwal V, Beer M, Bejerman N, Bergeron É, Biedenkopf N, Blair CD, Blasdell KR, Blouin AG, Bradfute SB, Briese T, Brown PA, Buchholz UJ, Buchmeier MJ, Bukreyev A, Burt F, Büttner C, Calisher CH, Cao M, Casas I, Chandran K, Charrel RN, Kumar Chaturvedi K, Chooi KM, Crane A, Dal Bó E, Carlos de la Torre J, de Souza WM, de Swart RL, Debat H, Dheilly NM, Di Paola N, Di Serio F, Dietzgen RG, Digiaro M, Drexler JF, Duprex WP, Dürrwald R, Easton AJ, Elbeaino T, Ergünay K, Feng G, Firth AE, Fooks AR, Formenty PBH, Freitas-Astúa J, Gago-Zachert S, Laura García M, García-Sastre A, Garrison AR, Gaskin TR, Gong W, Gonzalez JJ, de Bellocq J, Griffiths A, Groschup MH, Günther I, Günther S, Hammond J, Hasegawa Y, Hayashi K, Hepojoki J, Higgins CM, Hongō S, Horie M, Hughes HR, Hume AJ, Hyndman TH, Ikeda K, Jiāng D, Jonson GB, Junglen S, Klempa B, Klingström J, Kondō H, Koonin EV, Krupovic M, Kubota K, Kurath G, Laenen L, Lambert AJ, Lǐ J, Li JM, Liu R, Lukashevich IS, MacDiarmid RM, Maes P, Marklewitz M, Marshall SH, Marzano SL, McCauley JW, Mirazimi A, Mühlberger E, Nabeshima T, Naidu R, Natsuaki T, Navarro B, Navarro JA, Neriya Y, Netesov SV, Neumann G, Nowotny N, Nunes MRT, Ochoa-Corona FM, Okada T, Palacios G, Pallás V, Papa A, Paraskevopoulou S, Parrish CR, Pauvolid-Corrêa A, Pawęska JT, Pérez DR, Pfaff F, Plemper RK, Postler TS, Rabbidge LO, Radoshitzky SR, Ramos-González PL, Rehanek M, Resende RO, Reyes CA, Rodrigues TCS, Romanowski V, Rubbenstroth D, Rubino L, Runstadler JA, Sabanadzovic S, Sadiq S, Salvato MS, Sasaya T, Schwemmle M, Sharpe SR, Shi M, Shimomoto Y, Kavi Sidharthan V, Sironi M, Smither S, Song JW, Spann KM, Spengler JR, Stenglein MD, Takada A, Takeyama S, Tatara A, Tesh RB, Thornburg NJ, Tian X, Tischler ND, Tomitaka Y, Tomonaga K, Tordo N, Tu C, Turina M, Tzanetakis IE, Maria Vaira A, van den Hoogen B, Vanmechelen B, Vasilakis N, Verbeek M, von Bargen S, Wada J, Wahl V, Walker PJ, Waltzek TB, Whitfield AE, Wolf YI, Xia H, Xylogianni E, Yanagisawa H, Yano K, Ye G, Yuan Z, Zerbini FM, Zhang G, Zhang S, Zhang YZ, Zhao L, and Økland AL
- Subjects
- RNA-Dependent RNA Polymerase genetics, Negative-Sense RNA Viruses, RNA Viruses genetics
- Abstract
In April 2023, following the annual International Committee on Taxonomy of Viruses (ICTV) ratification vote on newly proposed taxa, the phylum Negarnaviricota was amended and emended. The phylum was expanded by one new family, 14 new genera, and 140 new species. Two genera and 538 species were renamed. One species was moved, and four were abolished. This article presents the updated taxonomy of Negarnaviricota as now accepted by the ICTV.
- Published
- 2023
- Full Text
- View/download PDF
48. Human airway lineages derived from pluripotent stem cells reveal the epithelial responses to SARS-CoV-2 infection.
- Author
-
Wang R, Hume AJ, Beermann ML, Simone-Roach C, Lindstrom-Vautrin J, Le Suer J, Huang J, Olejnik J, Villacorta-Martin C, Bullitt E, Hinds A, Ghaedi M, Rollins S, Werder RB, Abo KM, Wilson AA, Mühlberger E, Kotton DN, and Hawkins FJ
- Subjects
- Epithelial Cells, Humans, SARS-CoV-2, COVID-19, Induced Pluripotent Stem Cells, Pluripotent Stem Cells
- Abstract
There is an urgent need to understand how SARS-CoV-2 infects the airway epithelium and in a subset of individuals leads to severe illness or death. Induced pluripotent stem cells (iPSCs) provide a near limitless supply of human cells that can be differentiated into cell types of interest, including airway epithelium, for disease modeling. We present a human iPSC-derived airway epithelial platform, composed of the major airway epithelial cell types, that is permissive to SARS-CoV-2 infection. Subsets of iPSC-airway cells express the SARS-CoV-2 entry factors angiotensin-converting enzyme 2 ( ACE2 ), and transmembrane protease serine 2 ( TMPRSS2 ). Multiciliated cells are the primary initial target of SARS-CoV-2 infection. On infection with SARS-CoV-2, iPSC-airway cells generate robust interferon and inflammatory responses, and treatment with remdesivir or camostat mesylate causes a decrease in viral propagation and entry, respectively. In conclusion, iPSC-derived airway cells provide a physiologically relevant in vitro model system to interrogate the pathogenesis of, and develop treatment strategies for, COVID-19 pneumonia.
- Published
- 2022
- Full Text
- View/download PDF
49. The oral drug nitazoxanide restricts SARS-CoV-2 infection and attenuates disease pathogenesis in Syrian hamsters.
- Author
-
Miorin L, Mire CE, Ranjbar S, Hume AJ, Huang J, Crossland NA, White KM, Laporte M, Kehrer T, Haridas V, Moreno E, Nambu A, Jangra S, Cupic A, Dejosez M, Abo KA, Tseng AE, Werder RB, Rathnasinghe R, Mutetwa T, Ramos I, de Aja JS, de Alba Rivas CG, Schotsaert M, Corley RB, Falvo JV, Fernandez-Sesma A, Kim C, Rossignol JF, Wilson AA, Zwaka T, Kotton DN, Mühlberger E, García-Sastre A, and Goldfeld AE
- Abstract
A well-tolerated and cost-effective oral drug that blocks SARS-CoV-2 growth and dissemination would be a major advance in the global effort to reduce COVID-19 morbidity and mortality. Here, we show that the oral FDA-approved drug nitazoxanide (NTZ) significantly inhibits SARS-CoV-2 viral replication and infection in different primate and human cell models including stem cell-derived human alveolar epithelial type 2 cells. Furthermore, NTZ synergizes with remdesivir, and it broadly inhibits growth of SARS-CoV-2 variants B.1.351 (beta), P.1 (gamma), and B.1617.2 (delta) and viral syncytia formation driven by their spike proteins. Strikingly, oral NTZ treatment of Syrian hamsters significantly inhibits SARS-CoV-2-driven weight loss, inflammation, and viral dissemination and syncytia formation in the lungs. These studies show that NTZ is a novel host-directed therapeutic that broadly inhibits SARS-CoV-2 dissemination and pathogenesis in human and hamster physiological models, which supports further testing and optimization of NTZ-based therapy for SARS-CoV-2 infection alone and in combination with antiviral drugs.
- Published
- 2022
- Full Text
- View/download PDF
50. Reconstructed signaling and regulatory networks identify potential drugs for SARS-CoV-2 infection.
- Author
-
Ding J, Lugo-Martinez J, Yuan Y, Huang J, Hume AJ, Suder EL, Mühlberger E, Kotton DN, and Bar-Joseph Z
- Abstract
Several molecular datasets have been recently compiled to characterize the activity of SARS-CoV-2 within human cells. Here we extend computational methods to integrate several different types of sequence, functional and interaction data to reconstruct networks and pathways activated by the virus in host cells. We identify key proteins in these networks and further intersect them with genes differentially expressed at conditions that are known to impact viral activity. Several of the top ranked genes do not directly interact with virus proteins. We experimentally tested treatments for a number of the predicted targets. We show that blocking one of the predicted indirect targets significantly reduces viral loads in stem cell-derived alveolar epithelial type II cells (iAT2s)., Competing Interests: Conflict of interest None.
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.