33 results on '"Dinnon III, Kenneth H."'
Search Results
2. A genome-wide arrayed CRISPR screen identifies PLSCR1 as an intrinsic barrier to SARS-CoV-2 entry that recent virus variants have evolved to resist.
- Author
-
Le Pen, Jérémie, Paniccia, Gabrielle, Kinast, Volker, Moncada-Velez, Marcela, Ashbrook, Alison W., Bauer, Michael, Hoffmann, H.-Heinrich, Pinharanda, Ana, Ricardo-Lax, Inna, Stenzel, Ansgar F., Rosado-Olivieri, Edwin A., Dinnon III, Kenneth H., Doyle, William C., Freije, Catherine A., Hong, Seon-Hui, Lee, Danyel, Lewy, Tyler, Luna, Joseph M., Peace, Avery, and Schmidt, Carltin
- Subjects
SARS-CoV-2 ,COVID-19 ,GENETIC testing ,HUMAN genes ,INTERFERONS - Abstract
Interferons (IFNs) play a crucial role in the regulation and evolution of host–virus interactions. Here, we conducted a genome-wide arrayed CRISPR knockout screen in the presence and absence of IFN to identify human genes that influence Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection. We then performed an integrated analysis of genes interacting with SARS-CoV-2, drawing from a selection of 67 large-scale studies, including our own. We identified 28 genes of high relevance in both human genetic studies of Coronavirus Disease 2019 (COVID-19) patients and functional genetic screens in cell culture, with many related to the IFN pathway. Among these was the IFN-stimulated gene PLSCR1. PLSCR1 did not require IFN induction to restrict SARS-CoV-2 and did not contribute to IFN signaling. Instead, PLSCR1 specifically restricted spike-mediated SARS-CoV-2 entry. The PLSCR1-mediated restriction was alleviated by TMPRSS2 overexpression, suggesting that PLSCR1 primarily restricts the endocytic entry route. In addition, recent SARS-CoV-2 variants have adapted to circumvent the PLSCR1 barrier via currently undetermined mechanisms. Finally, we investigate the functional effects of PLSCR1 variants present in humans and discuss an association between PLSCR1 and severe COVID-19 reported recently. Interferons (IFNs) have a role in the regulation of virus-host interactions. This study uses genome-wide CRISPR knockout screen data to identify 28 genes that impact SARS-CoV-2 infection, including PLSCR1, which restricts spike-mediated SARS-CoV-2 entry independently of its IFN-related role. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. SARS-CoV-2 infection is effectively treated and prevented by EIDD-2801
- Author
-
Wahl, Angela, Gralinski, Lisa E., Johnson, Claire E., Yao, Wenbo, Kovarova, Martina, Dinnon, III, Kenneth H., Liu, Hongwei, Madden, Victoria J., Krzystek, Halina M., De, Chandrav, White, Kristen K., Gully, Kendra, Schäfer, Alexandra, Zaman, Tanzila, Leist, Sarah R., Grant, Paul O., Bluemling, Gregory R., Kolykhalov, Alexander A., Natchus, Michael G., Askin, Frederic B., Painter, George, Browne, Edward P., Jones, Corbin D., Pickles, Raymond J., Baric, Ralph S., and Garcia, J. Victor
- Published
- 2021
- Full Text
- View/download PDF
4. SARS-CoV-2 RBD trimer protein adjuvanted with Alum-3M-052 protects from SARS-CoV-2 infection and immune pathology in the lung
- Author
-
Routhu, Nanda Kishore, Cheedarla, Narayanaiah, Bollimpelli, Venkata Satish, Gangadhara, Sailaja, Edara, Venkata Viswanadh, Lai, Lilin, Sahoo, Anusmita, Shiferaw, Ayalnesh, Styles, Tiffany M., Floyd, Katharine, Fischinger, Stephanie, Atyeo, Caroline, Shin, Sally A., Gumber, Sanjeev, Kirejczyk, Shannon, Dinnon, III, Kenneth H., Shi, Pei-Yong, Menachery, Vineet D., Tomai, Mark, Fox, Christopher B., Alter, Galit, Vanderford, Thomas H., Gralinski, Lisa, Suthar, Mehul S., and Amara, Rama Rao
- Published
- 2021
- Full Text
- View/download PDF
5. A mouse-adapted model of SARS-CoV-2 to test COVID-19 countermeasures
- Author
-
Dinnon, III, Kenneth H., Leist, Sarah R., Schäfer, Alexandra, Edwards, Caitlin E., Martinez, David R., Montgomery, Stephanie A., West, Ande, Yount, Jr, Boyd L., Hou, Yixuan J., Adams, Lily E., Gully, Kendra L., Brown, Ariane J., Huang, Emily, Bryant, Matthew D., Choong, Ingrid C., Glenn, Jeffrey S., Gralinski, Lisa E., Sheahan, Timothy P., and Baric, Ralph S.
- Published
- 2020
- Full Text
- View/download PDF
6. Host Genetic Variation Impacts SARS-CoV-2 Vaccination Response in the Diversity Outbred Mouse Population.
- Author
-
Cruz Cisneros, Marta C., Anderson, Elizabeth J., Hampton, Brea K., Parotti, Breantié, Sarkar, Sanjay, Taft-Benz, Sharon, Bell, Timothy A., Blanchard, Matthew, Dillard, Jacob A., Dinnon III, Kenneth H., Hock, Pablo, Leist, Sarah R., Madden, Emily A., Shaw, Ginger D., West, Ande, Baric, Ralph S., Baxter, Victoria K., Pardo-Manuel de Villena, Fernando, Heise, Mark T., and Ferris, Martin T.
- Subjects
GENETIC variation ,VACCINE effectiveness ,COVID-19 vaccines ,SARS-CoV-2 ,DNA vaccines ,VIRAL shedding ,HUMORAL immunity - Abstract
The COVID-19 pandemic led to the rapid and worldwide development of highly effective vaccines against SARS-CoV-2. However, there is significant individual-to-individual variation in vaccine efficacy due to factors including viral variants, host age, immune status, environmental and host genetic factors. Understanding those determinants driving this variation may inform the development of more broadly protective vaccine strategies. While host genetic factors are known to impact vaccine efficacy for respiratory pathogens such as influenza and tuberculosis, the impact of host genetic variation on vaccine efficacy against COVID-19 is not well understood. To model the impact of host genetic variation on SARS-CoV-2 vaccine efficacy, while controlling for the impact of non-genetic factors, we used the Diversity Outbred (DO) mouse model. We found that DO mice immunized against SARS-CoV-2 exhibited high levels of variation in vaccine-induced neutralizing antibody responses. While the majority of the vaccinated mice were protected from virus-induced disease, similar to human populations, we observed vaccine breakthrough in a subset of mice. Importantly, we found that this variation in neutralizing antibody, virus-induced disease, and viral titer is heritable, indicating that the DO serves as a useful model system for studying the contribution of genetic variation of both vaccines and disease outcomes. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
7. Investigation of the Host Kinome Response to Coronavirus Infection Reveals PI3K/mTOR Inhibitors as Betacoronavirus Antivirals.
- Author
-
Fritch, Ethan J., Mordant, Angie L., Gilbert, Thomas S.K., Wells, Carrow I., Yang, Xuan, Barker, Natalie K., Madden, Emily A., Dinnon III, Kenneth H., Hou, Yixuan J., Tse, Longping V., Castillo, Izabella N., Sims, Amy C., Moorman, Nathaniel J., Lakshmanane, Premkumar, Willson, Timothy M., Herring, Laura E., Graves, Lee M., and Baric, Ralph S.
- Published
- 2023
- Full Text
- View/download PDF
8. Publisher Correction: A mouse-adapted model of SARS-CoV-2 to test COVID-19 countermeasures
- Author
-
Dinnon, III, Kenneth H., Leist, Sarah R., Schäfer, Alexandra, Edwards, Caitlin E., Martinez, David R., Montgomery, Stephanie A., West, Ande, Yount, Jr, Boyd L., Hou, Yixuan J., Adams, Lily E., Gully, Kendra L., Brown, Ariane J., Huang, Emily, Bryant, Matthew D., Choong, Ingrid C., Glenn, Jeffrey S., Gralinski, Lisa E., Sheahan, Timothy P., and Baric, Ralph S.
- Published
- 2021
- Full Text
- View/download PDF
9. A C57BL/6 Mouse Model of SARS-CoV-2 Infection Recapitulates Age- and Sex-Based Differences in Human COVID-19 Disease and Recovery
- Author
-
Davis, Michael A., primary, Voss, Kathleen, additional, Turnbull, J. Bryan, additional, Gustin, Andrew T., additional, Knoll, Megan, additional, Muruato, Antonio, additional, Hsiang, Tien-Ying, additional, Dinnon III, Kenneth H., additional, Leist, Sarah R., additional, Nickel, Katie, additional, Baric, Ralph S., additional, Ladiges, Warren, additional, Akilesh, Shreeram, additional, Smith, Kelly D., additional, and Gale, Michael, additional
- Published
- 2022
- Full Text
- View/download PDF
10. A C57BL/6 Mouse model of SARS-CoV-2 infection recapitulates age- and sex-based differences in human COVID-19 disease and recovery
- Author
-
Davis, Michael, primary, Voss, Kathleen, additional, Turnbull, J. Bryan, additional, Gustin, Andrew T., additional, Knoll, Megan, additional, Muruato, Antonio, additional, Hsiang, Tien-Ying, additional, Dinnon, III Kenneth H., additional, Leist, Sarah R., additional, Nickel, Katie, additional, Baric, Ralph S., additional, Ladiges, Warren, additional, Akilesh, Shreeram, additional, Smith, Kelly D., additional, and Gale, Michael, additional
- Published
- 2022
- Full Text
- View/download PDF
11. A C57BL/6 Mouse Model of SARS-CoV-2 Infection Recapitulates Age- and Sex-Based Differences in Human COVID-19 Disease and Recovery.
- Author
-
Davis, Michael A., Voss, Kathleen, Turnbull, J. Bryan, Gustin, Andrew T., Knoll, Megan, Muruato, Antonio, Hsiang, Tien-Ying, Dinnon III, Kenneth H., Leist, Sarah R., Nickel, Katie, Baric, Ralph S., Ladiges, Warren, Akilesh, Shreeram, Smith, Kelly D., and Gale Jr., Michael
- Subjects
COVID-19 ,LABORATORY mice ,SARS-CoV-2 ,SEVERE combined immunodeficiency ,ANIMAL disease models ,CORONAVIRUS diseases ,SYMPTOMS - Abstract
We present a comprehensive analysis of SARS-CoV-2 infection and recovery using wild type C57BL/6 mice and a mouse-adapted virus, and we demonstrate that this is an ideal model of infection and recovery that phenocopies acute human disease arising from the ancestral SARS-CoV-2. Disease severity and infection kinetics are age- and sex-dependent, as has been reported for humans, with older mice and males in particular exhibiting decreased viral clearance and increased mortality. We identified key parallels with human pathology, including intense virus positivity in bronchial epithelial cells, wide-spread alveolar involvement, recruitment of immune cells to the infected lungs, and acute bronchial epithelial cell death. Moreover, older animals experienced increased virus persistence, delayed dispersal of immune cells into lung parenchyma, and morphologic evidence of tissue damage and inflammation. Parallel analysis of SCID mice revealed that the adaptive immune response was not required for recovery from COVID disease symptoms nor early phase clearance of virus but was required for efficient clearance of virus at later stages of infection. Finally, transcriptional analyses indicated that induction and duration of key innate immune gene programs may explain differences in age-dependent disease severity. Importantly, these data demonstrate that SARS-CoV-2-mediated disease in C57BL/6 mice phenocopies human disease across ages and establishes a platform for future therapeutic and genetic screens for not just SARS-CoV-2 but also novel coronaviruses that have yet to emerge. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
12. Mouse Adapted SARS-CoV-2 (MA10) Viral Infection Induces Neuroinflammation in Standard Laboratory Mice.
- Author
-
Amruta, Narayanappa, Ismael, Saifudeen, Leist, Sarah R., Gressett, Timothy E., Srivastava, Akhilesh, Dinnon III, Kenneth H., Engler-Chiurazzi, Elizabeth B., Maness, Nicholas J., Qin, Xuebin, Kolls, Jay K., Baric, Ralph S., and Bix, Gregory
- Subjects
COVID-19 ,SARS-CoV-2 ,LABORATORY mice ,VIRUS diseases ,GLIAL fibrillary acidic protein - Abstract
Increasing evidence suggests that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection impacts neurological function both acutely and chronically, even in the absence of pronounced respiratory distress. Developing clinically relevant laboratory mouse models of the neuropathogenesis of SARS-CoV-2 infection is an important step toward elucidating the underlying mechanisms of SARS-CoV-2-induced neurological dysfunction. Although various transgenic models and viral delivery methods have been used to study the infection potential of SARS-CoV-2 in mice, the use of commonly available laboratory mice would facilitate the study of SARS-CoV-2 neuropathology. Herein we show neuroinflammatory profiles of immunologically intact mice, C57BL/6J and BALB/c, as well as immunodeficient (Rag2
−/− ) mice, to a mouse-adapted strain of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2 (MA10)). Our findings indicate that brain IL-6 levels are significantly higher in BALB/c male mice infected with SARS-CoV-2 MA10. Additionally, blood-brain barrier integrity, as measured by the vascular tight junction protein claudin-5, was reduced by SARS-CoV-2 MA10 infection in all three strains. Brain glial fibrillary acidic protein (GFAP) mRNA was also elevated in male C57BL/6J infected mice compared with the mock group. Lastly, immune-vascular effects of SARS-CoV-2 (MA10), as measured by H&E scores, demonstrate an increase in perivascular lymphocyte cuffing (PLC) at 30 days post-infection among infected female BALB/c mice with a significant increase in PLC over time only in SARS-CoV-2 MA10) infected mice. Our study is the first to demonstrate that SARS-CoV-2 (MA10) infection induces neuroinflammation in laboratory mice and could be used as a novel model to study SARS-CoV-2-mediated cerebrovascular pathology. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
13. SARS-CoV-2 infection produces chronic pulmonary epithelial and immune cell dysfunction with fibrosis in mice.
- Author
-
Dinnon III, Kenneth H., Leist, Sarah R., Okuda, Kenichi, Dang, Hong, Fritch, Ethan J., Gully, Kendra L., De la Cruz, Gabriela, Evangelista, Mia D., Asakura, Takanori, Gilmore, Rodney C., Hawkins, Padraig, Nakano, Satoko, West, Ande, Schäfer, Alexandra, Gralinski, Lisa E., Everman, Jamie L., Sajuthi, Satria P., Zweigart, Mark R., Dong, Stephanie, and McBride, Jennifer
- Subjects
COVID-19 ,SARS-CoV-2 ,POST-acute COVID-19 syndrome ,ADULT respiratory distress syndrome ,MYOFIBROBLASTS - Abstract
A subset of individuals who recover from coronavirus disease 2019 (COVID-19) develop post-acute sequelae of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) (PASC), but the mechanistic basis of PASC-associated lung abnormalities suffers from a lack of longitudinal tissue samples. The mouse-adapted SARS-CoV-2 strain MA10 produces an acute respiratory distress syndrome in mice similar to humans. To investigate PASC pathogenesis, studies of MA10-infected mice were extended from acute to clinical recovery phases. At 15 to 120 days after virus clearance, pulmonary histologic findings included subpleural lesions composed of collagen, proliferative fibroblasts, and chronic inflammation, including tertiary lymphoid structures. Longitudinal spatial transcriptional profiling identified global reparative and fibrotic pathways dysregulated in diseased regions, similar to human COVID-19. Populations of alveolar intermediate cells, coupled with focal up-regulation of profibrotic markers, were identified in persistently diseased regions. Early intervention with antiviral EIDD-2801 reduced chronic disease, and early antifibrotic agent (nintedanib) intervention modified early disease severity. This murine model provides opportunities to identify pathways associated with persistent SARS-CoV-2 pulmonary disease and test countermeasures to ameliorate PASC. Pursuing pulmonary PASC: A potential consequence of SARS-CoV-2 infection is long COVID, also called post-acute sequelae of SARS-CoV-2 (PASC). PASC can include chronic lung dysfunction among other symptoms, but the mechanisms driving PASC-associated pulmonary disease are unknown. Here, Dinnon et al. infected mice with a mouse-adapted strain of SARS-CoV-2 to characterize long-term damage in the lungs over time. Using histology and spatial transcriptomics, the authors observed the development of a profibrotic phenotype at later time points after infection; severity of lung damage could be ameliorated by early antifibrotic or antiviral drug treatment. These data support the use of mouse-adapted SARS-CoV-2 as a PASC model and suggest that antiviral and antifibrotic treatments could be an option to prevent or reduce PASC in humans. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
14. Remdesivir Inhibits SARS-CoV-2 in Human Lung Cells and Chimeric SARS-CoV Expressing the SARS-CoV-2 RNA Polymerase in Mice
- Author
-
Yount, Boyd L., Baric, Ralph S., Ma, Bin, Agostini, Maria L., Graham, Rachel L., Feng, Joy Y., Stevens, Laura J., Du Pont, Venice, Dinnon III, Kenneth H., Lu, Xiaotao, Gralinski, Lisa E., George, Amelia S., Hughes, Tia M., Gully, Kendra, Pitts, Jared, Brown, Ariane J., Cihlar, Tomas, Martinez, David R., Bilello, John P., Sheahan, Timothy P., Murakami, Eisuke, Pruijssers, Andrea J., Schaefer, Alexandra, Leist, Sarah R., Babusis, Darius, Porter, Danielle P., Chappell, James D., and Perry, Jason K.
- Subjects
viruses ,virus diseases ,respiratory system ,respiratory tract diseases - Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the novel viral disease COVID-19. With no approved therapies, this pandemic illustrates the urgent need for broad-spectrum antiviral countermeasures against SARS-CoV-2 and future emerging CoVs. We report that remdesivir (RDV) potently inhibits SARS-CoV-2 replication in human lung cells and primary human airway epithelial cultures (EC50 = 0.01 μM). Weaker activity is observed in Vero E6 cells (EC50 = 1.65 μM) because of their low capacity to metabolize RDV. To rapidly evaluate in vivo efficacy, we engineered a chimeric SARS-CoV encoding the viral target of RDV, the RNA-dependent RNA polymerase of SARS-CoV-2. In mice infected with the chimeric virus, therapeutic RDV administration diminishes lung viral load and improves pulmonary function compared with vehicle-treated animals. These data demonstrate that RDV is potently active against SARS-CoV-2 in vitro and in vivo, supporting its further clinical testing for treatment of COVID-19.
- Published
- 2020
- Full Text
- View/download PDF
15. Trypsin Treatment Unlocks Barrier for Zoonotic Bat Coronavirus Infection
- Author
-
McAnarney, Eileen T., Baric, Ralph S., Lipkin, W. Ian, Graham, Barney, Dinnon III, Kenneth H., Wang, Lingshu, Scobey, Trevor, Randell, Scott H., Menachery, Vineet D., Gralinski, Lisa E., Yount, Jr., Boyd L., Hale, Andrew, Anthony, Simon J., and Graham, Rachel L.
- Subjects
viruses ,virus diseases - Abstract
Traditionally, the emergence of coronaviruses (CoVs) has been attributed to a gain in receptor binding in a new host. Our previous work with severe acute respiratory syndrome (SARS)-like viruses argued that bats already harbor CoVs with the ability to infect humans without adaptation. These results suggested that additional barriers limit the emergence of zoonotic CoV. In this work, we describe overcoming host restriction of two Middle East respiratory syndrome (MERS)-like bat CoVs using exogenous protease treatment. We found that the spike protein of PDF2180-CoV, a MERS-like virus found in a Ugandan bat, could mediate infection of Vero and human cells in the presence of exogenous trypsin. We subsequently show that the bat virus spike can mediate the infection of human gut cells but is unable to infect human lung cells. Using receptor-blocking antibodies, we show that infection with the PDF2180 spike does not require MERS-CoV receptor DPP4 and antibodies developed against the MERS spike receptor-binding domain and S2 portion are ineffective in neutralizing the PDF2180 chimera. Finally, we found that the addition of exogenous trypsin also rescues HKU5-CoV, a second bat group 2c CoV. Together, these results indicate that proteolytic cleavage of the spike, not receptor binding, is the primary infection barrier for these two group 2c CoVs. Coupled with receptor binding, proteolytic activation offers a new parameter to evaluate the emergence potential of bat CoVs and offers a means to recover previously unrecoverable zoonotic CoV strains. IMPORTANCE Overall, our studies demonstrate that proteolytic cleavage is the primary barrier to infection for a subset of zoonotic coronaviruses. Moving forward, the results argue that both receptor binding and proteolytic cleavage of the spike are critical factors that must be considered for evaluating the emergence potential and risk posed by zoonotic coronaviruses. In addition, the findings also offer a novel means to recover previously uncultivable zoonotic coronavirus strains and argue that other tissues, including the digestive tract, could be a site for future coronavirus emergence events in humans.
- Published
- 2020
- Full Text
- View/download PDF
16. Novel virus-like nanoparticle vaccine effectively protects animal model from SARS-CoV-2 infection.
- Author
-
Geng, Qibin, Tai, Wanbo, Baxter, Victoria K., Shi, Juan, Wan, Yushun, Zhang, Xiujuan, Montgomery, Stephanie A., Taft-Benz, Sharon A., Anderson, Elizabeth J., Knight, Audrey C., Dinnon III, Kenneth H., Leist, Sarah R., Baric, Ralph S., Shang, Jian, Hong, Sung-Wook, Drelich, Aleksandra, Tseng, Chien-Te K., Jenkins, Marc, Heise, Mark, and Du, Lanying
- Subjects
SARS-CoV-2 ,COVID-19 ,COVID-19 pandemic ,VIRAL vaccines ,PATHOLOGICAL physiology ,VACCINES - Abstract
The key to battling the COVID-19 pandemic and its potential aftermath is to develop a variety of vaccines that are efficacious and safe, elicit lasting immunity, and cover a range of SARS-CoV-2 variants. Recombinant viral receptor-binding domains (RBDs) are safe vaccine candidates but often have limited efficacy due to the lack of virus-like immunogen display pattern. Here we have developed a novel virus-like nanoparticle (VLP) vaccine that displays 120 copies of SARS-CoV-2 RBD on its surface. This VLP-RBD vaccine mimics virus-based vaccines in immunogen display, which boosts its efficacy, while maintaining the safety of protein-based subunit vaccines. Compared to the RBD vaccine, the VLP-RBD vaccine induced five times more neutralizing antibodies in mice that efficiently blocked SARS-CoV-2 from attaching to its host receptor and potently neutralized the cell entry of variant SARS-CoV-2 strains, SARS-CoV-1, and SARS-CoV-1-related bat coronavirus. These neutralizing immune responses induced by the VLP-RBD vaccine did not wane during the two-month study period. Furthermore, the VLP-RBD vaccine effectively protected mice from SARS-CoV-2 challenge, dramatically reducing the development of clinical signs and pathological changes in immunized mice. The VLP-RBD vaccine provides one potentially effective solution to controlling the spread of SARS-CoV-2. Author summary: Both mRNA-based and viral vector-based vaccines are currently being distributed to curtail the COVID-19 pandemic. Continued development of more varieties of SARS-CoV-2 vaccines will help battle the many variants of SARS-CoV-2. Here we have developed a virus-like particle (VLP) vaccine that combines the effectiveness of virus-based vaccines and safety of protein-based vaccines. Using the lumazine synthase nanoparticle protein as the structural scaffold and 120 copies of SARS-CoV-2 receptor-binding domain as the surface immunogen, this VLP vaccine induced high-titer neutralizing antibody responses in mice that lasted >2 months and potently inhibited SARS-CoV-2, SARS-CoV-1, and their variants. The VLP vaccine also protected mice from high-titer SARS-CoV-2 challenge. The novel VLP vaccine may contribute to the protection of the human population from SARS-CoV-2 and its variants. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
17. Shortening of Zika virus CD-loop reduces neurovirulence while preserving antigenicity
- Author
-
Dinnon III, Kenneth H., primary, Gallichotte, Emily N., additional, Fritch, Ethan J., additional, Menachery, Vineet D., additional, and Baric, Ralph S., additional
- Published
- 2019
- Full Text
- View/download PDF
18. A highly immunogenic and effective measles virus-based Th1-biased COVID-19 vaccine.
- Author
-
Hörner, Cindy, Schürmann, Christoph, Auste, Arne, Ebenig, Aileen, Muraleedharan, Samada, Dinnon III, Kenneth H., Scholz, Tatjana, Herrmann, Maike, Schnierle, Barbara S., Baric, Ralph S., and Mühlebach, Michael D.
- Subjects
COVID-19 vaccines ,MEASLES ,COVID-19 pandemic ,SARS-CoV-2 - Abstract
The COVID-19 pandemic is caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and has spread worldwide, with millions of cases and more than 1 million deaths to date. The gravity of the situation mandates accelerated efforts to identify safe and effective vaccines. Here, we generated measles virus (MeV)-based vaccine candidates expressing the SARS-CoV-2 spike glycoprotein (S). Insertion of the full-length S protein gene in two different MeV genomic positions resulted in modulated S protein expression. The variant with lower S protein expression levels was genetically stable and induced high levels of effective Th1-biased antibody and T cell responses in mice after two immunizations. In addition to neutralizing IgG antibody responses in a protective range, multifunctional CD8
+ and CD4+ T cell responses with S protein-specific killing activity were detected. Upon challenge using a mouse-adapted SARS-CoV-2, virus loads in vaccinated mice were significantly lower, while vaccinated Syrian hamsters revealed protection in a harsh challenge setup using an early-passage human patient isolate. These results are highly encouraging and support further development of MeV-based COVID-19 vaccines. [ABSTRACT FROM AUTHOR]- Published
- 2020
- Full Text
- View/download PDF
19. Swine acute diarrhea syndrome coronavirus replication in primary human cells reveals potential susceptibility to infection.
- Author
-
Edwards, Caitlin E., Yount, Boyd L., Graham, Rachel L., Leist, Sarah R., Yixuan J. Hou, Dinnon III, Kenneth H., Sims, Amy C., Swanstrom, Jesica, Gully, Kendra, Scobey, Trevor D., Cooley, Michelle R., Currie, Caroline G., Randell, Scott H., and Baric, Ralph S.
- Subjects
HEPATOCELLULAR carcinoma ,COVID-19 ,VIRUS identification ,DISEASE outbreaks ,PRIMARY cell culture - Abstract
Zoonotic coronaviruses represent an ongoing threat, yet the myriads of circulating animal viruses complicate the identification of higher-risk isolates that threaten human health. Swine acute diarrhea syndrome coronavirus (SADS-CoV) is a newly discovered, highly pathogenic virus that likely evolved from closely related HKU2 bat coronaviruses, circulating in Rhinolophus spp. bats in China and elsewhere. As coronaviruses cause severe economic losses in the pork industry and swine are key intermediate hosts of human disease outbreaks, we synthetically resurrected a recombinant virus (rSADS-CoV) as well as a derivative encoding tomato red fluorescent protein (tRFP) in place of ORF3. rSADS-CoV replicated efficiently in a variety of continuous animal and primate cell lines, including human liver and rectal carcinoma cell lines. Of concern, rSADS-CoV also replicated efficiently in several different primary human lung cell types, as well as primary human intestinal cells. rSADS-CoV did not use human coronavirus ACE-2, DPP4, or CD13 receptors for docking and entry. Contemporary human donor sera neutralized the group I human coronavirus NL63, but not rSADS-CoV, suggesting limited human group I coronavirus cross protective herd immunity. Importantly, remdesivir, a broad-spectrum nucleoside analog that is effective against other group 1 and 2 coronaviruses, efficiently blocked rSADS-CoV replication in vitro. rSADS-CoV demonstrated little, if any, replicative capacity in either immune-competent or immunodeficient mice, indicating a critical need for improved animal models. Efficient growth in primary human lung and intestinal cells implicate SADS-CoV as a potential higher-risk emerging coronavirus pathogen that could negatively impact the global economy and human health. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
20. An orally bioavailable broad-spectrum antiviral inhibits SARS-CoV-2 in human airway epithelial cell cultures and multiple coronaviruses in mice.
- Author
-
Sheahan, Timothy P., Sims, Amy C., Zhou, Shuntai, Graham, Rachel L., Pruijssers, Andrea J., Agostini, Maria L., Leist, Sarah R., Schäfer, Alexandra, Dinnon III, Kenneth H., Stevens, Laura J., Chappell, James D., Lu, Xiaotao, Hughes, Tia M., George, Amelia S., Hill, Collin S., Montgomery, Stephanie A., Brown, Ariane J., Bluemling, Gregory R., Natchus, Michael G., and Saindane, Manohar
- Subjects
COVID-19 ,EPITHELIAL cell culture ,CORONAVIRUSES ,EBOLA virus ,RNA viruses - Abstract
Catastrophic consequences: Broad-spectrum antivirals are desirable, particularly in the context of emerging zoonotic infections for which specific interventions do not yet exist. Sheahan et al. tested the potential of a ribonucleoside analog previously shown to be active against other RNA viruses such as influenza and Ebola virus to combat coronaviruses. This drug was effective in cell lines and primary human airway epithelial cultures against multiple coronaviruses including SARS-CoV-2. Mouse models of SARS and MERS demonstrated that early treatment reduced viral replication and damage to the lungs. Mechanistically, this drug is incorporated into the viral RNA, inducing mutations and eventually leading to error catastrophe in the virus. In this manner, inducing catastrophe could help avoid catastrophe by stemming the next pandemic. Coronaviruses (CoVs) traffic frequently between species resulting in novel disease outbreaks, most recently exemplified by the newly emerged SARS-CoV-2, the causative agent of COVID-19. Here, we show that the ribonucleoside analog β-d-N
4 -hydroxycytidine (NHC; EIDD-1931) has broad-spectrum antiviral activity against SARS-CoV-2, MERS-CoV, SARS-CoV, and related zoonotic group 2b or 2c bat-CoVs, as well as increased potency against a CoV bearing resistance mutations to the nucleoside analog inhibitor remdesivir. In mice infected with SARS-CoV or MERS-CoV, both prophylactic and therapeutic administration of EIDD-2801, an orally bioavailable NHC prodrug (β-d-N4 -hydroxycytidine-5′-isopropyl ester), improved pulmonary function and reduced virus titer and body weight loss. Decreased MERS-CoV yields in vitro and in vivo were associated with increased transition mutation frequency in viral, but not host cell RNA, supporting a mechanism of lethal mutagenesis in CoV. The potency of NHC/EIDD-2801 against multiple CoVs and oral bioavailability highlights its potential utility as an effective antiviral against SARS-CoV-2 and other future zoonotic CoVs. [ABSTRACT FROM AUTHOR]- Published
- 2020
- Full Text
- View/download PDF
21. Trypsin treatment unlocks barrier for zoonotic bat coronaviruses infection.
- Author
-
Menachery, Vineet D., Dinnon III, Kenneth H., Yount Jr., Boyd L., McAnarney, Eileen T., Gralinski, Lisa E., Hale, Andrew, Graham, Rachel L., Scobey, Trevor, Anthony, Simon J., Lingshu Wang, Graham, Barney, Randell, Scott H., Lipkin, W. Ian, and Baric, Ralph S.
- Subjects
- *
TRYPSIN , *BATS , *INFECTION , *RECEPTOR antibodies , *MERS coronavirus , *CORONAVIRUSES - Abstract
Traditionally, the emergence of coronaviruses (CoVs) has been attributed to a gain in receptor binding in a new host. Our previous work with SARS-like viruses argued that bats already harbor CoVs with the ability to infect humans without adaptation. These results suggested that additional barriers limit the emergence of zoonotic CoV. In this work, we describe overcoming host restriction of two MERS-like bat CoVs using exogenous protease treatment. We found that the spike protein of PDF2180-CoV, a MERS-like virus found in a Ugandan bat, could mediate infection of Vero and human cells in the presence of exogenous trypsin. We subsequently show that the bat virus spike can mediate infection of human gut cells, but is unable to infect human lung cells. Using receptor-blocking antibodies, we show that infection with the PDF2180 spike does not require MERS-CoV receptor DPP4 and antibodies developed against the MERS spike receptor-binding domain and S2 portion are ineffective in neutralizing the PDF2180 chimera. Finally, we found that addition of exogenous trypsin also rescues HKU5-CoV, a second bat group 2c CoV. Together, these results indicate that proteolytic cleavage of the spike, not receptor binding, is the primary infection barrier for these two group 2c CoVs. Coupled with receptor binding, proteolytic activation offers a new parameter to evaluate emergence potential of bat CoVs and offer a means to recover previously unrecoverable zoonotic CoV strains. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
22. CD-loop Extension in Zika Virus Envelope Protein Key for Stability and Pathogenesis.
- Author
-
Gallichotte, Emily N., Dinnon III, Kenneth H., Xin-Ni Lim, Thiam-Seng Ng, Lim, Elisa X. Y., Menachery, Vineet D., Shee-Mei Lok, Baric, Ralph S., Dinnon, Kenneth H 3rd, Lim, Xin-Ni, Ng, Thiam-Seng, and Lok, Shee-Mei
- Subjects
- *
HUMAN abnormalities , *GUILLAIN-Barre syndrome , *ZIKA virus infections , *MICROCEPHALY , *PATHOGENIC microorganisms , *PUBLIC health , *PROTEIN metabolism , *AMINO acids , *ANIMAL experimentation , *ANTIGENS , *BIOCHEMISTRY , *BIOLOGICAL models , *CELL lines , *PHYSICAL & theoretical chemistry , *IMMUNOGLOBULINS , *MATHEMATICAL models , *PHENOMENOLOGY , *MICE , *MOLECULAR structure , *GENETIC mutation , *PRIMATES , *PROTEINS , *RESEARCH funding , *TEMPERATURE , *VIRAL antibodies , *THEORY - Abstract
With severe disease manifestations including microcephaly, congenital malformation, and Guillain-Barré syndrome, Zika virus (ZIKV) remains a persistent global public health threat. Despite antigenic similarities with dengue viruses, structural studies have suggested the extended CD-loop and hydrogen-bonding interaction network within the ZIKV envelope protein contribute to stability differences between the viral families. This enhanced stability may lead to the augmented infection, disease manifestation, and persistence in body fluids seen following ZIKV infection. To examine the role of these motifs in infection, we generated a series of ZIKV recombinant viruses that disrupted the hydrogen-bonding network (350A, 351A, and 350A/351A) or the CD-loop extension (Δ346). Our results demonstrate a key role for the ZIKV extended CD-loop in cell-type-dependent replication, virion stability, and in vivo pathogenesis. Importantly, the Δ346 mutant maintains similar antigenicity to wild-type virus, opening the possibility for its use as a live-attenuated vaccine platform for ZIKV and other clinically relevant flaviviruses. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
23. Fc-mediated pan-sarbecovirus protection after alphavirus vector vaccination.
- Author
-
Adams, Lily E., Leist, Sarah R., Dinnon III, Kenneth H., West, Ande, Gully, Kendra L., Anderson, Elizabeth J., Loome, Jennifer F., Madden, Emily A., Powers, John M., Schäfer, Alexandra, Sarkar, Sanjay, Castillo, Izabella N., Maron, Jenny S., McNamara, Ryan P., Bertera, Harry L., Zweigert, Mark R., Higgins, Jaclyn S., Hampton, Brea K., Premkumar, Lakshmanane, and Alter, Galit
- Abstract
Group 2B β-coronaviruses (sarbecoviruses) have caused regional and global epidemics in modern history. Here, we evaluate the mechanisms of cross-sarbecovirus protective immunity, currently less clear yet important for pan-sarbecovirus vaccine development, using a panel of alphavirus-vectored vaccines covering bat to human strains. While vaccination does not prevent virus replication, it protects against lethal heterologous disease outcomes in both severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and clade 2 bat sarbecovirus challenge models. The spike vaccines tested primarily elicit a highly S1-specific homologous neutralizing antibody response with no detectable cross-virus neutralization. Rather, non-neutralizing antibody functions, mechanistically linked to FcgR4 and spike S2, mediate cross-protection in wild-type mice. Protection is lost in FcR knockout mice, further supporting a model for non-neutralizing, protective antibodies. These data highlight the importance of FcR-mediated cross-protective immune responses in universal pan-sarbecovirus vaccine designs. [Display omitted] • Non-neutralizing antibodies mediate pan-sarbecovirus protection • Antibody-mediated cross-protection is lost in absence of FcR function • S2-specific antibodies are a strong correlate of protective FcR effector function • Full-length spike elicits the broadest pan-sarbecovirus protection Using a lethal model for β-coronavirus infection, Adams et al. described heterologous protection from disease that was driven by non-neutralizing antibodies through Fc-receptor-dependent mechanisms. These results reveal important protective correlates for inclusion into the design and testing of future pan-coronavirus vaccines. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
24. Stabilized coronavirus spike stem elicits a broadly protective antibody.
- Author
-
Hsieh, Ching-Lin, Werner, Anne P., Leist, Sarah R., Stevens, Laura J., Falconer, Ester, Goldsmith, Jory A., Chou, Chia-Wei, Abiona, Olubukola M., West, Ande, Westendorf, Kathryn, Muthuraman, Krithika, Fritch, Ethan J., Dinnon III, Kenneth H., Schäfer, Alexandra, Denison, Mark R., Chappell, James D., Baric, Ralph S., Graham, Barney S., Corbett, Kizzmekia S., and McLellan, Jason S.
- Abstract
Current coronavirus (CoV) vaccines primarily target immunodominant epitopes in the S1 subunit, which are poorly conserved and susceptible to escape mutations, thus threatening vaccine efficacy. Here, we use structure-guided protein engineering to remove the S1 subunit from the Middle East respiratory syndrome (MERS)-CoV spike (S) glycoprotein and develop stabilized stem (SS) antigens. Vaccination with MERS SS elicits cross-reactive β-CoV antibody responses and protects mice against lethal MERS-CoV challenge. High-throughput screening of antibody-secreting cells from MERS SS-immunized mice led to the discovery of a panel of cross-reactive monoclonal antibodies. Among them, antibody IgG22 binds with high affinity to both MERS-CoV and severe acute respiratory syndrome (SARS)-CoV-2 S proteins, and a combination of electron microscopy and crystal structures localizes the epitope to a conserved coiled-coil region in the S2 subunit. Passive transfer of IgG22 protects mice against both MERS-CoV and SARS-CoV-2 challenge. Collectively, these results provide a proof of principle for cross-reactive CoV antibodies and inform the development of pan-CoV vaccines and therapeutic antibodies. [Display omitted] • Structure-guided design generates MERS-CoV spike stabilized stem (SS) antigens • Cross-reactive CoV-spike-stem-specific monoclonal antibodies • Structures of the Fab22-spike complexes reveal a conserved, protective epitope • Passive transfer of IgG22 protects mice against MERS-CoV and SARS-CoV-2 challenge Hsieh et al. generate MERS-CoV spike stabilized stem (SS) antigens using a structure-guided approach. Mice immunized with MERS SS are protected against MERS-CoV challenge and used for isolation of cross-reactive monoclonal antibodies, including IgG22, which protects mice against MERS-CoV and SARS-CoV-2 challenges. Structures of Fab22-spike complexes reveal a conserved epitope. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
25. A Newcastle Disease Virus (NDV) Expressing a Membrane-Anchored Spike as a Cost-Effective Inactivated SARS-CoV-2 Vaccine.
- Author
-
Sun, Weina, McCroskery, Stephen, Liu, Wen-Chun, Leist, Sarah R., Liu, Yonghong, Albrecht, Randy A., Slamanig, Stefan, Oliva, Justine, Amanat, Fatima, Schäfer, Alexandra, Dinnon III, Kenneth H., Innis, Bruce L., García-Sastre, Adolfo, Krammer, Florian, Baric, Ralph S., and Palese, Peter
- Subjects
NEWCASTLE disease virus ,COVID-19 ,SARS-CoV-2 ,INFLUENZA vaccines ,VACCINES - Abstract
A successful severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine must not only be safe and protective, but must also meet the demand on a global scale at a low cost. Using the current influenza virus vaccine production capacity to manufacture an egg-based inactivated Newcastle disease virus (NDV)/SARS-CoV-2 vaccine would meet that challenge. Here, we report pre-clinical evaluations of an inactivated NDV chimera stably expressing the membrane-anchored form of the spike (NDV-S) as a potent coronavirus disease 2019 (COVID-19) vaccine in mice and hamsters. The inactivated NDV-S vaccine was immunogenic, inducing strong binding and/or neutralizing antibodies in both animal models. More importantly, the inactivated NDV-S vaccine protected animals from SARS-CoV-2 infections. In the presence of an adjuvant, antigen-sparing could be achieved, which would further reduce the cost while maintaining the protective efficacy of the vaccine. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
26. A Mouse-Adapted SARS-CoV-2 Induces Acute Lung Injury and Mortality in Standard Laboratory Mice.
- Author
-
Leist, Sarah R., Dinnon III, Kenneth H., Schäfer, Alexandra, Tse, Longping V., Okuda, Kenichi, Hou, Yixuan J., West, Ande, Edwards, Caitlin E., Sanders, Wes, Fritch, Ethan J., Gully, Kendra L., Scobey, Trevor, Brown, Ariane J., Sheahan, Timothy P., Moorman, Nathaniel J., Boucher, Richard C., Gralinski, Lisa E., Montgomery, Stephanie A., and Baric, Ralph S.
- Subjects
- *
SARS-CoV-2 , *LABORATORY mice , *ADULT respiratory distress syndrome , *LUNG injuries , *COVID-19 , *AGE factors in memory - Abstract
The SARS-CoV-2 pandemic has caused extreme human suffering and economic harm. We generated and characterized a new mouse-adapted SARS-CoV-2 virus that captures multiple aspects of severe COVID-19 disease in standard laboratory mice. This SARS-CoV-2 model exhibits the spectrum of morbidity and mortality of COVID-19 disease as well as aspects of host genetics, age, cellular tropisms, elevated Th1 cytokines, and loss of surfactant expression and pulmonary function linked to pathological features of acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). This model can rapidly access existing mouse resources to elucidate the role of host genetics, underlying molecular mechanisms governing SARS-CoV-2 pathogenesis, and the protective or pathogenic immune responses related to disease severity. The model promises to provide a robust platform for studies of ALI and ARDS to evaluate vaccine and antiviral drug performance, including in the most vulnerable populations (i.e., the aged) using standard laboratory mice. • Serial in vivo evolution selected for a lethal mouse-adapted SARS-CoV-2 MA10 variant • SARS-CoV-2 MA10 shows a dose- and age-related increase in pathogenesis in BALB/c mice • Mice exhibit ALI, ARDS, and surfactant loss, key metrics in countermeasure performance • SARS-CoV-2 MA10 model enables access to immune reagents and genetically defined mice Leist et al. present a mouse model for COVID-19 by serially passaging human SARS-CoV-2 in vivo to create an evolution-selected lethal mouse-adapted virus variant, called MA10. MA10 shows a dose- and age-related increase in pathogenesis in standard laboratory mice and recapitulates key features of COVID-19 in humans. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
27. Remdesivir Inhibits SARS-CoV-2 in Human Lung Cells and Chimeric SARS-CoV Expressing the SARS-CoV-2 RNA Polymerase in Mice.
- Author
-
Pruijssers, Andrea J., George, Amelia S., Schäfer, Alexandra, Leist, Sarah R., Gralinksi, Lisa E., Dinnon III, Kenneth H., Yount, Boyd L., Agostini, Maria L., Stevens, Laura J., Chappell, James D., Lu, Xiaotao, Hughes, Tia M., Gully, Kendra, Martinez, David R., Brown, Ariane J., Graham, Rachel L., Perry, Jason K., Du Pont, Venice, Pitts, Jared, and Ma, Bin
- Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the novel viral disease COVID-19. With no approved therapies, this pandemic illustrates the urgent need for broad-spectrum antiviral countermeasures against SARS-CoV-2 and future emerging CoVs. We report that remdesivir (RDV) potently inhibits SARS-CoV-2 replication in human lung cells and primary human airway epithelial cultures (EC 50 = 0.01 μM). Weaker activity is observed in Vero E6 cells (EC 50 = 1.65 μM) because of their low capacity to metabolize RDV. To rapidly evaluate in vivo efficacy, we engineered a chimeric SARS-CoV encoding the viral target of RDV, the RNA-dependent RNA polymerase of SARS-CoV-2. In mice infected with the chimeric virus, therapeutic RDV administration diminishes lung viral load and improves pulmonary function compared with vehicle-treated animals. These data demonstrate that RDV is potently active against SARS-CoV-2 in vitro and in vivo , supporting its further clinical testing for treatment of COVID-19. • Remdesivir binding of active site of polymerase is conserved across all human CoVs • Remdesivir inhibits SARS-CoV-2 in primary and continuous human lung cell cultures • Remdesivir potency depends on cell-type-specific metabolism to its active form • Therapeutic remdesivir reduces viral loads and improves outcomes in mice SARS-CoV-2 causes severe lung disease (COVID-19) in humans. Pruijssers et al. demonstrate that the antiviral drug remdesivir potently inhibits SARS-CoV-2 in human lung cell cultures. Therapeutic treatment of infected mice with remdesivir reduces viral loads and improves clinical outcomes, further supporting use of remdesivir for the treatment of COVID-19. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
28. Shortening of Zika virus CD-loop reduces neurovirulence while preserving antigenicity
- Author
-
Menachery, Vineet D., Baric, Ralph S., Dinnon III, Kenneth H., Gallichotte, Emily N., and Fritch, Ethan
- Subjects
viruses ,3. Good health - Abstract
Zika virus (ZIKV) is a mosquito-borne positive sense RNA virus. Recently, ZIKV emerged into the Western hemisphere as a human health threat, with severe disease associated with developmental and neurological complications. The structural envelope protein of ZIKV and other neurotropic flaviviruses contains an extended CD-loop relative to non-neurotropic flaviviruses, and has been shown to augment ZIKV stability and pathogenesis. Here we show that shortening the CD-loop in ZIKV attenuates the virus in mice, by reducing the ability to invade and replicate in the central nervous system. The CD-loop mutation was genetically stable following infection in mice, though secondary site mutations arise adjacent to the CD-loop. Importantly, while shortening of the CD-loop attenuates the virus, the CD-loop mutant maintains antigenicity in immunocompetent mice, eliciting an antibody response that similarly neutralizes both the mutant and wildtype ZIKV. These findings suggest that the extended CD-loop in ZIKV is a determinant of neurotropism and may be a target in live-attenuated vaccine design, for not only ZIKV, but for other neurotropic flaviviruses.
29. Broad spectrum antiviral remdesivir inhibits human endemic and zoonotic deltacoronaviruses with a highly divergent RNA dependent RNA polymerase
- Author
-
Dinnon III, Kenneth H., Baric, Ralph S., Graham, Rachel L., Sheahan, Timothy P., Won, John J., Denison, Mark R., Feng, Joy Y., Sims, Amy C., Cihlar, Tomas, and Brown, Ariane J.
- Subjects
viruses ,virus diseases ,respiratory tract diseases ,3. Good health - Abstract
The genetically diverse Orthocoronavirinae (CoV) family is prone to cross species transmission and disease emergence in both humans and livestock. Viruses similar to known epidemic strains circulating in wild and domestic animals further increase the probability of emergence in the future. Currently, there are no approved therapeutics for any human CoV presenting a clear unmet medical need. Remdesivir (RDV, GS-5734) is a monophosphoramidate prodrug of an adenosine analog with potent activity against an array of RNA virus families including Filoviridae, Paramyxoviridae, Pneumoviridae, and Orthocoronavirinae, through the targeting of the viral RNA dependent RNA polymerase (RdRp). We developed multiple assays to further define the breadth of RDV antiviral activity against the CoV family. Here, we show potent antiviral activity of RDV against endemic human CoVs OC43 (HCoV-OC43) and 229E (HCoV-229E) with submicromolar EC50 values. Of known CoVs, the members of the deltacoronavirus genus have the most divergent RdRp as compared to SARS- and MERS-CoV and both avian and porcine members harbor a native residue in the RdRp that confers resistance in beta-CoVs. Nevertheless, RDV is highly efficacious against porcine deltacoronavirus (PDCoV). These data further extend the known breadth and antiviral activity of RDV to include both contemporary human and highly divergent zoonotic CoV and potentially enhance our ability to fight future emerging CoV.
30. Combination Attenuation Offers Strategy for Live Attenuated Coronavirus Vaccines.
- Author
-
Menachery, Vineet D., Gralinski, Lisa E., Mitchell, Hugh D., Dinnon III, Kenneth H., Leist, Sarah R., Yount Jr., Boyd L., McAnarney, Eileen T., Graham, Rachel L., Waters, Katrina M., and Baric, Ralph S.
- Subjects
- *
CORONAVIRUS diseases , *VIRAL vaccines , *CORONAVIRUSES , *VIRUS diseases , *METHYLTRANSFERASES , *VACCINATION - Abstract
With an ongoing threat posed by circulating zoonotic strains, new strategies are required to prepare for the next emergent coronavirus (CoV). Previously, groups had targeted conserved coronavirus proteins as a strategy to generate live attenuated vaccine strains against current and future CoVs. With this in mind, we explored whether manipulation of CoV NSP16, a conserved 2'O methyltransferase (MTase), could provide a broad attenuation platform against future emergent strains. Using the severe acute respiratory syndrome-CoV mouse model, an NSP16 mutant vaccine was evaluated for protection from heterologous challenge, efficacy in the aging host, and potential for reversion to pathogenesis. Despite some success, concerns for virulence in the aged and potential for reversion makes targeting NSP16 alone an untenable approach. However, combining a 2'O MTase mutation with a previously described CoV fidelity mutant produced a vaccine strain capable of protection from heterologous virus challenge, efficacy in aged mice, and no evidence for reversion. Together, the results indicate that targeting the CoV 2'O MTase in parallel with other conserved attenuating mutations may provide a platform strategy for rapidly generating live attenuated coronavirus vaccines. IMPORTANCE Emergent coronaviruses remain a significant threat to global public health and rapid response vaccine platforms are needed to stem future outbreaks. However, failure of many previous CoV vaccine formulations has clearly highlighted the need to test efficacy under different conditions and especially in vulnerable populations such as the aged and immunocompromised. This study illustrates that despite success in young models, the 2'O methyltransferase mutant carries too much risk for pathogenesis and reversion in vulnerable models to be used as a standalone vaccine strategy. Importantly, the 2'O methyltransferase mutation can be paired with other attenuating approaches to provide robust protection from heterologous challenge and in vulnerable populations. Coupled with increased safety and reduced pathogenesis, the study highlights the potential for 2'O methyltransferase attenuation as a major component of future live attenuated coronavirus vaccines. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
31. COVID-19 vaccine mRNA-1273 elicits a protective immune profile in mice that is not associated with vaccine-enhanced disease upon SARS-CoV-2 challenge.
- Author
-
DiPiazza, Anthony T., Leist, Sarah R., Abiona, Olubukola M., Moliva, Juan I., Werner, Anne, Minai, Mahnaz, Nagata, Bianca M., Bock, Kevin W., Phung, Emily, Schäfer, Alexandra, Dinnon III, Kenneth H., Chang, Lauren A., Loomis, Rebecca J., Boyoglu-Barnum, Seyhan, Alvarado, Gabriela S., Sullivan, Nancy J., Edwards, Darin K., Morabito, Kaitlyn M., Mascola, John R., and Carfi, Andrea
- Subjects
- *
HUMORAL immunity , *COVID-19 vaccines , *SARS-CoV-2 , *PATHOLOGICAL physiology , *LABORATORY mice - Abstract
Vaccine-associated enhanced respiratory disease (VAERD) was previously observed in some preclinical models of severe acute respiratory syndrome (SARS) and MERS coronavirus vaccines. We used the SARS coronavirus 2 (SARS-CoV-2) mouse-adapted, passage 10, lethal challenge virus (MA10) mouse model of acute lung injury to evaluate the immune response and potential for immunopathology in animals vaccinated with research-grade mRNA-1273. Whole-inactivated virus or heat-denatured spike protein subunit vaccines with alum designed to elicit low-potency antibodies and Th2-skewed CD4+ T cells resulted in reduced viral titers and weight loss post challenge but more severe pathological changes in the lung compared to saline-immunized animals. In contrast, a protective dose of mRNA-1273 induced favorable humoral and cellular immune responses that protected from viral replication in the upper and lower respiratory tract upon challenge. A subprotective dose of mRNA-1273 reduced viral replication and limited histopathological manifestations compared to animals given saline. Overall, our findings demonstrate an immunological signature associated with antiviral protection without disease enhancement following vaccination with mRNA-1273. [Display omitted] • mRNA-1273 elicits neutralizing antibodies and spike-binding germinal center B cells • Humoral immunity is accompanied by spike-specific CD4+ Th1, Tfh, and CD8+ T cells • mRNA-1273 demonstrates protection in the mouse MA10 acute lung injury model • mRNA-1273 vaccination does not result in enhanced disease following infection As vaccine-enhanced disease to respiratory viruses has been previously observed, a thorough safety evaluation of COVID-19 vaccines in preclinical animal models is essential. Here, DiPiazza and Leist et al. provide evidence for antiviral protection in the absence of lung disease following SARS-CoV-2 challenge in mice immunized with research-grade mRNA-1273. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
32. SARS-CoV-2 Reverse Genetics Reveals a Variable Infection Gradient in the Respiratory Tract.
- Author
-
Hou, Yixuan J., Okuda, Kenichi, Edwards, Caitlin E., Martinez, David R., Asakura, Takanori, Dinnon III, Kenneth H., Kato, Takafumi, Lee, Rhianna E., Yount, Boyd L., Mascenik, Teresa M., Chen, Gang, Olivier, Kenneth N., Ghio, Andrew, Tse, Longping V., Leist, Sarah R., Gralinski, Lisa E., Schäfer, Alexandra, Dang, Hong, Gilmore, Rodney, and Nakano, Satoko
- Subjects
- *
SARS disease , *VIRAL antibodies , *SARS virus , *SARS-CoV-2 , *COVID-19 , *RESPIRATORY infections , *REVERSE genetics - Abstract
The mode of acquisition and causes for the variable clinical spectrum of coronavirus disease 2019 (COVID-19) remain unknown. We utilized a reverse genetics system to generate a GFP reporter virus to explore severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pathogenesis and a luciferase reporter virus to demonstrate sera collected from SARS and COVID-19 patients exhibited limited cross-CoV neutralization. High-sensitivity RNA in situ mapping revealed the highest angiotensin-converting enzyme 2 (ACE2) expression in the nose with decreasing expression throughout the lower respiratory tract, paralleled by a striking gradient of SARS-CoV-2 infection in proximal (high) versus distal (low) pulmonary epithelial cultures. COVID-19 autopsied lung studies identified focal disease and, congruent with culture data, SARS-CoV-2-infected ciliated and type 2 pneumocyte cells in airway and alveolar regions, respectively. These findings highlight the nasal susceptibility to SARS-CoV-2 with likely subsequent aspiration-mediated virus seeding to the lung in SARS-CoV-2 pathogenesis. These reagents provide a foundation for investigations into virus-host interactions in protective immunity, host susceptibility, and virus pathogenesis. • A SARS-CoV-2 infectious cDNA clone and reporter viruses are generated • SARS-CoV-2 and SARS-CoV neutralization assays show limited cross neutralization • SARS-CoV-2 shows a gradient infectivity from the proximal to distal respiratory tract • Ciliated airway cells and AT-2 cells are primary targets for SARS-CoV-2 infection Hou et al. present a reverse genetics system for SARS-CoV-2, which is then used to make reporter viruses to quantify the ability of patient sera and antibodies to neutralize infectious virus and to examine viral tropism along the human respiratory tract. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
33. Broad spectrum antiviral remdesivir inhibits human endemic and zoonotic deltacoronaviruses with a highly divergent RNA dependent RNA polymerase.
- Author
-
Brown, Ariane J., Won, John J., Graham, Rachel L., Dinnon III, Kenneth H., Sims, Amy C., Feng, Joy Y., Cihlar, Tomas, Denison, Mark R., Baric, Ralph S., and Sheahan, Timothy P.
- Subjects
- *
RNA polymerases , *RNA , *INFECTIOUS disease transmission , *ANIMALS , *PARAMYXOVIRUSES , *ZOONOSES , *REMDESIVIR , *CORONAVIRUSES - Abstract
The genetically diverse Orthocoronavirinae (CoV) family is prone to cross species transmission and disease emergence in both humans and livestock. Viruses similar to known epidemic strains circulating in wild and domestic animals further increase the probability of emergence in the future. Currently, there are no approved therapeutics for any human CoV presenting a clear unmet medical need. Remdesivir (RDV, GS-5734) is a monophosphoramidate prodrug of an adenosine analog with potent activity against an array of RNA virus families including Filoviridae , Paramyxoviridae , Pneumoviridae , and Orthocoronavirinae , through the targeting of the viral RNA dependent RNA polymerase (RdRp). We developed multiple assays to further define the breadth of RDV antiviral activity against the CoV family. Here, we show potent antiviral activity of RDV against endemic human CoVs OC43 (HCoV-OC43) and 229E (HCoV-229E) with submicromolar EC 50 values. Of known CoVs, the members of the deltacoronavirus genus have the most divergent RdRp as compared to SARS- and MERS-CoV and both avian and porcine members harbor a native residue in the RdRp that confers resistance in beta-CoVs. Nevertheless, RDV is highly efficacious against porcine deltacoronavirus (PDCoV). These data further extend the known breadth and antiviral activity of RDV to include both contemporary human and highly divergent zoonotic CoV and potentially enhance our ability to fight future emerging CoV. • In vitro antiviral assays were developed for human CoV OC43 and 229E and the zoonotic PDCoV. • The nucleoside analog RDV inhibited HCoV-OC43 and 229E as well as deltacoronavirus member PDCoV. • RDV has broad-spectrum antiviral activity against CoV and should be evaluated for future emerging CoV. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.