63 results on '"Shaia C"'
Search Results
2. Temporal Progression of Lesions in Guinea Pigs Infected With Lassa Virus
- Author
-
Bell, T. M., primary, Shaia, C. I., additional, Bearss, J. J., additional, Mattix, M. E., additional, Koistinen, K. A., additional, Honnold, S. P., additional, Zeng, X., additional, Blancett, C. D., additional, Donnelly, G. C., additional, Shamblin, J. D., additional, Wilkinson, E. R., additional, and Cashman, K. A., additional
- Published
- 2016
- Full Text
- View/download PDF
3. Pathogenesis of Bolivian Hemorrhagic Fever in Guinea Pigs
- Author
-
Bell, T. M., primary, Bunton, T. E., additional, Shaia, C. I., additional, Raymond, J. W., additional, Honnold, S. P., additional, Donnelly, G. C., additional, Shamblin, J. D., additional, Wilkinson, E. R., additional, and Cashman, K. A., additional
- Published
- 2015
- Full Text
- View/download PDF
4. Temporal Progression of Lesions in Guinea Pigs Infected With Lassa Virus.
- Author
-
Bell, T. M., Shaia, C. I., Bearss, J. J., Mattix, M. E., Koistinen, K. A., Honnold, S. P., Zeng, X., Blancett, C. D., Donnelly, G. C., Shamblin, J. D., Wilkinson, E. R., and Cashman, K. A.
- Subjects
LASSA fever ,HEMORRHAGIC fever ,GUINEA pigs ,DIAGNOSIS ,INFECTIOUS disease transmission ,DISEASES - Abstract
Lassa virus (LASV) infection causes an acute, multisystemic viral hemorrhagic fever that annually infects an estimated 100 000 to 300 000 persons in West Africa. This pathogenesis study evaluated the temporal progression of disease in guinea pigs following aerosol and subcutaneous inoculation of the Josiah strain of LASV as well as the usefulness of Strain 13 guinea pigs as an animal model for Lassa fever. After experimental infection, guinea pigs (Cavia porcellus; n = 67) were serially sampled to evaluate the temporal progression of infection, gross and histologic lesions, and serum chemistry and hematologic changes. Guinea pigs developed viremia on day 5 to 6 postexposure (PE), with clinical signs appearing by day 7 to 8 PE. Complete blood counts revealed lymphopenia and thrombocytopenia. Gross pathologic findings included skin lesions and congested lungs. Histologic lesions consisted of cortical lymphoid depletion by day 6 to 7 PE with lymphohistiocytic interstitial pneumonia at 7 to 8 days PE. Scattered hepatocellular degeneration and cell death were also noted in the liver and, to a lesser extent, in other tissues including the haired skin, lung, heart, adrenal gland, lymph nodes, thymus, and spleen. The first cell types to demonstrate staining for viral antigen were fibroblastic reticular cells and macrophages/dendritic cells in the lymph nodes on day 5 to 6 PE. This study demonstrates similarities between Lassa viral disease in human infections and experimental guinea pig infection. These shared pathologic characteristics support the utility of guinea pigs as an additional animal model for vaccine and therapeutic development under the Food and Drug Administration’s Animal Rule. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
5. Pathology of Experimental Machupo Virus Infection, Chicava Strain, in Cynomolgus Macaques (Macaca fascicularis) by Intramuscular and Aerosol Exposure
- Author
-
Bell, T. M., primary, Shaia, C. I., additional, Bunton, T. E., additional, Robinson, C. G., additional, Wilkinson, E. R., additional, Hensley, L. E., additional, and Cashman, K. A., additional
- Published
- 2014
- Full Text
- View/download PDF
6. Experimental Aerosolized Guinea Pig–Adapted Zaire Ebolavirus (Variant
- Author
-
Twenhafel, N. A., primary, Shaia, C. I., additional, Bunton, T. E., additional, Shamblin, J. D., additional, Wollen, S. E., additional, Pitt, L. M., additional, Sizemore, D. R., additional, Ogg, M. M., additional, and Johnston, S. C., additional
- Published
- 2014
- Full Text
- View/download PDF
7. Pathogenesis of Bolivian Hemorrhagic Fever in Guinea Pigs.
- Author
-
Bell, T. M., Bunton, T. E., Shaia, C. I., Raymond, J. W., Honnold, S. P., Donnelly, G. C., Shamblin, J. D., Wilkinson, E. R., and Cashman, K. A.
- Subjects
HEMORRHAGIC fever ,GUINEA pigs ,VACCINES ,DISEASE progression ,HEMATOLOGY ,DISEASES - Abstract
Machupo virus, the cause of Bolivian hemorrhagic fever, is a highly lethal viral hemorrhagic fever with no Food and Drug Administration–approved vaccines or therapeutics. This study evaluated the guinea pig as a model using the Machupo virus–Chicava strain administered via aerosol challenge. Guinea pigs (Cavia porcellus) were serially sampled to evaluate the temporal progression of infection, gross and histologic lesions, and sequential changes in serum chemistry and hematology. The incubation period was 5 to 12 days, and complete blood counts revealed leukopenia with lymphopenia and thrombocytopenia. Gross pathologic findings included congestion and hemorrhage of the gastrointestinal mucosa and serosa, noncollapsing lungs with fluid exudation, enlarged lymph nodes, and progressive pallor and friability of the liver. Histologic lesions consisted of foci of degeneration and cell death in the haired skin, liver, pancreas, adrenal glands, lymph nodes, tongue, esophagus, salivary glands, renal pelvis, small intestine, and large intestine. Lymphohistiocytic interstitial pneumonia was also present. Inflammation within the central nervous system, interpreted as nonsuppurative encephalitis, was histologically apparent approximately 16 days postexposure and was generally progressive. Macrophages in the tracheobronchial lymph node, on day 5 postexposure, were the first cells to demonstrate visible viral antigen. Viral antigen was detected throughout the lymphoid system by day 9 postexposure, followed by prominent spread within epithelial tissues and then brain. This study provides insight into the course of Machupo virus infection and supports the utility of guinea pigs as an additional animal model for vaccine and therapeutic development. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
8. Biodetection grinder
- Author
-
Shaia, C. D and Jones, G. H
- Subjects
Biotechnology - Abstract
Work on a biodetection grinder is summarized. It includes development of the prototype grinder, second generation grinder, and the production version of the grinder. Tests showed the particle size distribution was satisfactory and biological evaluation confirmed the tests.
- Published
- 1971
9. Experimental Aerosolized Guinea Pig–Adapted Zaire Ebolavirus (Variant: Mayinga) Causes Lethal Pneumonia in Guinea Pigs.
- Author
-
Twenhafel, N. A., Shaia, C. I., Bunton, T. E., Shamblin, J. D., Wollen, S. E., Pitt, L. M., Sizemore, D. R., Ogg, M. M., and Johnston, S. C.
- Subjects
PULMONARY fibrosis ,GUINEA pigs as laboratory animals ,AEROSOLS ,EBOLA virus disease ,VETERINARY pathology ,ALVEOLAR macrophages ,ANIMAL models in research - Abstract
Eight guinea pigs were aerosolized with guinea pig–adapted Zaire ebolavirus (variant: Mayinga) and developed lethal interstitial pneumonia that was distinct from lesions described in guinea pigs challenged subcutaneously, nonhuman primates challenged by the aerosol route, and natural infection in humans. Guinea pigs succumbed with significant pathologic changes primarily restricted to the lungs. Intracytoplasmic inclusion bodies were observed in many alveolar macrophages. Perivasculitis was noted within the lungs. These changes are unlike those of documented subcutaneously challenged guinea pigs and aerosolized filoviral infections in nonhuman primates and human cases. Similar to findings in subcutaneously challenged guinea pigs, there were only mild lesions in the liver and spleen. To our knowledge, this is the first report of aerosol challenge of guinea pigs with guinea pig–adapted Zaire ebolavirus (variant: Mayinga). Before choosing this model for use in aerosolized ebolavirus studies, scientists and pathologists should be aware that aerosolized guinea pig–adapted Zaire ebolavirus (variant: Mayinga) causes lethal pneumonia in guinea pigs. [ABSTRACT FROM PUBLISHER]
- Published
- 2015
- Full Text
- View/download PDF
10. Pathology of Experimental Machupo Virus Infection, Chicava Strain, in Cynomolgus Macaques (Macaca fascicularis) by Intramuscular and Aerosol Exposure.
- Author
-
Bell, T. M., Shaia, C. I., Bunton, T. E., Robinson, C. G., Wilkinson, E. R., Hensley, L. E., and Cashman, K. A.
- Subjects
HEMORRHAGIC fever ,ANIMAL models in research ,KRA ,INCUBATION period (Communicable diseases) ,VETERINARY pathology ,ALANINE aminotransferase ,AEROSOLS ,VETERINARY histology ,DISEASES - Abstract
Machupo virus, the causative agent of Bolivian hemorrhagic fever (BHF), is a highly lethal viral hemorrhagic fever of which little is known and for which no Food and Drug Administration–approved vaccines or therapeutics are available. This study evaluated the cynomolgus macaque as an animal model using the Machupo virus, Chicava strain, via intramuscular and aerosol challenge. The incubation period was 6 to 10 days with initial signs of depression, anorexia, diarrhea, mild fever, and a petechial skin rash. These were often followed by neurologic signs and death within an average of 18 days. Complete blood counts revealed leukopenia as well as marked thrombocytopenia. Serum chemistry values identified a decrease in total protein, marked increases in alanine aminotransferase and aspartate aminotransferase, and moderate increases in alkaline phosphatase. Gross pathology findings included a macular rash extending across the axillary and inguinal regions beginning at approximately 10 days postexposure as well as enlarged lymph nodes and spleen, enlarged and friable liver, and sporadic hemorrhages along the gastrointestinal mucosa and serosa. Histologic lesions consisted of foci of degeneration and necrosis/apoptosis in the haired skin, liver, pancreas, adrenal glands, lymph nodes, tongue, esophagus, salivary glands, stomach, small intestine, and large intestine. Lymphohistiocytic interstitial pneumonia was also present. Inflammation within the central nervous system (nonsuppurative encephalitis) was histologically apparent approximately 16 days postexposure and was generally progressive. This study provides insight into the course of Machupo virus infection in cynomolgus macaques and supports the usefulness of cynomolgus macaques as a viable model of human Machupo virus infection. [ABSTRACT FROM PUBLISHER]
- Published
- 2015
- Full Text
- View/download PDF
11. Purification and expression of the Tf190 adhesin in Tritrichomonas foetus.
- Author
-
Shaia, C I, Voyich, J, Gillis, S J, Singh, B N, and Burgess, D E
- Abstract
Bovine trichomoniasis is a sexually transmitted disease caused by Tritrichomonas foetus and characterized by early embryo loss. The mechanism of this loss is not known, although the parasite is known to cause inflammation and to have the ability to kill host cells by a contact-dependent cytotoxic mechanism. Antibody specific for a 190,000-Da surface complex (Tf190) was previously shown to inhibit this adhesion. In this study we used immunoaffinity chromatography to purify Tf190 from T. foetus in order to analyze its composition and examine its expression. Analysis by sodium dodecyl sulfate-polyacrylamide gel electrophoresis of purified Tf190 followed by silver staining revealed three components of Tf190. Western blotting and antibody-binding experiments showed that the 140- and 60-kDa bands were immunogenic. By using a battery of monoclonal antibodies (MAbs) periodate-sensitive epitopes were identified on Tf190, suggesting that these epitopes contained carbohydrate structures. Analyses of affinity-purified Tf190 by high-performance liquid chromatography and gas-liquid chromatography demonstrated the presence of the monosaccharides and lipids known to be prominent constituents of the lipophosphoglycan (LPG) of T. foetus. Flow cytometry experiments on several isolates of T. foetus with Tf190-specific antibodies revealed that Tf190 was present on subpopulations of all isolates but that not all epitopes were present on every isolate. This pattern of reactivities on the different parasite isolates was confirmed by Western blots of whole-parasite extracts probed with MAbs and antiserum. These results suggest that although variation in the expression of epitopes of Tf190 occurs in different strains of T. foetus, the Tf190 adhesion complex is widespread in different populations of the parasite. The data further suggest that immunogenic structures, important in the adhesion of T. foetus to mammalian cells, are located in the LPG-like component of Tf190.
- Published
- 1998
12. A Bivalent Adenovirus-Vectored Vaccine Induces a Robust Humoral Response, but Does Not Protect Cynomolgus Macaques Against a Lethal Challenge With Sudan Virus.
- Author
-
van Tol S, Fletcher P, Feldmann F, Mukesh RK, Port JR, Gallogly S, Schulz JE, Rhoderick JF, Makinson R, Carmody A, Myers L, Lovaglio J, Smith BJ, Okumura A, Shaia C, Saturday G, Marzi A, Lambe T, Munster VJ, and van Doremalen N
- Subjects
- Animals, Immunity, Humoral, Ebola Vaccines immunology, Ebola Vaccines administration & dosage, Immunoglobulin G blood, Disease Models, Animal, Viral Load, Adenoviruses, Simian immunology, Adenoviruses, Simian genetics, Vaccination, Adenoviridae genetics, Adenoviridae immunology, Macaca fascicularis, Hemorrhagic Fever, Ebola prevention & control, Hemorrhagic Fever, Ebola immunology, Hemorrhagic Fever, Ebola virology, Ebolavirus immunology, Ebolavirus genetics, Antibodies, Viral blood, Genetic Vectors, Antibodies, Neutralizing blood, Antibodies, Neutralizing immunology
- Abstract
The most recent Sudan virus (SUDV) outbreak in Uganda was first detected in September 2022 and resulted in 164 laboratory-confirmed cases and 77 deaths. There are no approved vaccines against SUDV. Here, we investigated the protective efficacy of ChAdOx1-biEBOV in cynomolgus macaques using a prime or a prime-boost regimen. ChAdOx1-biEBOV is a replication-deficient simian adenovirus vector encoding SUDV and Ebola virus (EBOV) glycoproteins (GPs). Intramuscular vaccination induced SUDV and EBOV GP-specific immunoglobulin G responses and neutralizing antibodies. Upon challenge with SUDV, vaccinated animals showed signs of disease like those observed in control animals, and no differences in survival outcomes were measured among all 3 groups. Viral load in blood samples and in tissue samples obtained after necropsy were not significantly different between groups. Overall, this study highlights the importance of evaluating vaccines in multiple animal models and demonstrates the importance of understanding protective efficacy in both animal models and human hosts., Competing Interests: Potential conflicts of interest. T. L. reports consulting fees from Vaccitech on an unrelated project, has received honorarium from Seqirus on an unrelated project, and is named as an inventor on a patent application for a vaccine against SARS-CoV-2. All other authors report no potential conflicts. All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed., (Published by Oxford University Press on behalf of Infectious Diseases Society of America 2024.)
- Published
- 2024
- Full Text
- View/download PDF
13. Antibodies targeting the Crimean-Congo Hemorrhagic Fever Virus nucleoprotein protect via TRIM21.
- Author
-
Leventhal SS, Bisom T, Clift D, Rao D, Meade-White K, Shaia C, Murray J, Mihalakakos EA, Hinkley T, Reynolds SJ, Best SM, Erasmus JH, James LC, Feldmann H, and Hawman DW
- Subjects
- Animals, Mice, Ribonucleoproteins immunology, Ribonucleoproteins metabolism, Mice, Knockout, Humans, Female, Mice, Inbred C57BL, Viral Vaccines immunology, Antibodies, Neutralizing immunology, Immunization, Passive, Hemorrhagic Fever Virus, Crimean-Congo immunology, Antibodies, Viral immunology, Hemorrhagic Fever, Crimean immunology, Hemorrhagic Fever, Crimean prevention & control, Nucleoproteins immunology, Nucleoproteins metabolism
- Abstract
Crimean-Congo Hemorrhagic Fever Virus (CCHFV) is a negative-sense RNA virus spread by Hyalomma genus ticks across Europe, Asia, and Africa. CCHF disease begins as a non-specific febrile illness which may progress into a severe hemorrhagic disease with no widely approved or highly efficacious interventions currently available. Recently, we reported a self-replicating, alphavirus-based RNA vaccine that expresses the CCHFV nucleoprotein and is protective against lethal CCHFV disease in mice. This vaccine induces high titers of non-neutralizing anti-NP antibodies and we show here that protection does not require Fc-gamma receptors or complement. Instead, vaccinated mice deficient in the intracellular Fc-receptor TRIM21 were unable to control the infection despite mounting robust CCHFV-specific immunity. We also show that passive transfer of NP-immune sera confers significant TRIM21-dependent protection against lethal CCHFV challenge. Together our data identifies TRIM21-mediated mechanisms as the Fc effector function of protective antibodies against the CCHFV NP and provides mechanistic insight into how vaccines against the CCHFV NP confer protection., (© 2024. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)
- Published
- 2024
- Full Text
- View/download PDF
14. In vivo delivery of engineered synthetic DNA-encoded SARS-CoV-2 monoclonal antibodies for pre-exposure prophylaxis in non-human primates.
- Author
-
Patel A, Rosenke K, Parzych EM, Feldmann F, Bharti S, Griffin AJ, Schouest B, Lewis M, Choi J, Chokkalingam N, Machado V, Smith BJ, Frase D, Ali AR, Lovaglio J, Nguyen B, Hanley PW, Walker SN, Gary EN, Kulkarni A, Generotti A, Francica JR, Rosenthal K, Kulp DW, Esser MT, Smith TRF, Shaia C, Weiner DB, and Feldmann H
- Subjects
- Animals, Macaca mulatta, SARS-CoV-2 genetics, Antibodies, Viral, Antibodies, Monoclonal, Macaca fascicularis, DNA, Antibodies, Neutralizing, Spike Glycoprotein, Coronavirus genetics, Pre-Exposure Prophylaxis, COVID-19 prevention & control
- Abstract
COVID-19 remains a major public health concern. Monoclonal antibodies have received emergency use authorization (EUA) for pre-exposure prophylaxis against COVID-19 among high-risk groups for treatment of mild to moderate COVID-19. In addition to recombinant biologics, engineered synthetic DNA-encoded antibodies (DMAb) are an important strategy for direct in vivo delivery of protective mAb. A DMAb cocktail was synthetically engineered to encode the immunoglobulin heavy and light chains of two different two different Fc-engineered anti-SARS-CoV-2 antibodies. The DMAbs were designed to enhance in vivo expression and delivered intramuscularly to cynomolgus and rhesus macaques with a modified in vivo delivery regimen. Serum levels were detected in macaques, along with specific binding to SARS-CoV-2 spike receptor binding domain protein and neutralization of multiple SARS-CoV-2 variants of concern in pseudovirus and authentic live virus assays. Prophylactic administration was protective in rhesus macaques against signs of SARS-CoV-2 (USA-WA1/2020) associated disease in the lungs. Overall, the data support further study of DNA-encoded antibodies as an additional delivery mode for prevention of COVID-19 severe disease. These data have implications for human translation of gene-encoded mAbs for emerging infectious diseases and low dose mAb delivery against COVID-19.
- Published
- 2024
- Full Text
- View/download PDF
15. A replicating RNA vaccine confers protection in a rhesus macaque model of Crimean-Congo hemorrhagic fever.
- Author
-
Hawman DW, Leventhal SS, Meade-White K, Khandhar A, Murray J, Lovaglio J, Shaia C, Saturday G, Hinkley T, Erasmus J, and Feldmann H
- Abstract
Crimean-Congo hemorrhagic fever (CCHF) is a tick-borne febrile illness with a wide geographic distribution. In recent years the geographic range of Crimean-Congo hemorrhagic fever virus (CCHFV) and its tick vector have increased, placing an increasing number of people at risk of CCHFV infection. Currently, there are no widely available vaccines, and although the World Health Organization recommends ribavirin for treatment, its efficacy is unclear. Here we evaluate a promising replicating RNA vaccine in a rhesus macaque (Macaca mulatta) model of CCHF. This model provides an alternative to the established cynomolgus macaque model and recapitulates mild-to-moderate human disease. Rhesus macaques infected with CCHFV consistently exhibit viremia, detectable viral RNA in a multitude of tissues, and moderate pathology in the liver and spleen. We used this model to evaluate the immunogenicity and protective efficacy of a replicating RNA vaccine. Rhesus macaques vaccinated with RNAs expressing the CCHFV nucleoprotein and glycoprotein precursor developed robust non-neutralizing humoral immunity against the CCHFV nucleoprotein and had significant protection against the CCHFV challenge. Together, our data report a model of CCHF using rhesus macaques and demonstrate that our replicating RNA vaccine is immunogenic and protective in non-human primates after a prime-boost immunization., (© 2024. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)
- Published
- 2024
- Full Text
- View/download PDF
16. Single dose, dual antigen RNA vaccines protect against lethal Crimean-Congo haemorrhagic fever virus infection in mice.
- Author
-
Leventhal SS, Meade-White K, Shaia C, Tipih T, Lewis M, Mihalakakos EA, Hinkley T, Khandhar AP, Erasmus JH, Feldmann H, and Hawman DW
- Subjects
- Animals, Mice, mRNA Vaccines, Vaccination, Hemorrhagic Fever Virus, Crimean-Congo genetics, Hemorrhagic Fever, Crimean prevention & control, Vaccines
- Abstract
Background: Crimean-Congo Haemorrhagic Fever Virus is a tick-borne bunyavirus prevalent across Asia, Africa, the Middle East, and Europe. The virus causes a non-specific febrile illness which may develop into severe haemorrhagic disease. To date, there are no widely approved therapeutics. Recently, we reported an alphavirus-based replicon RNA vaccine which expresses the CCHFV nucleoprotein (repNP) or glycoprotein precursor (repGPC) and is protective against lethal disease in mice., Methods: Here, we evaluated engineered GPC constructs to find the minimal enhancing epitope of repGPC and test two RNA vaccine approaches to express multiple antigens in vivo to optimize protective efficacy of our repRNA., Findings: Vaccination with repNP and a construct expressing just the Gc antigen (repGc-FL) resulted in equivalent immunogenicity and protective efficacy compared to original repNP + repGPC vaccination. This vaccine was protective when prepared in either of two vaccine approaches, a mixed synthesis reaction producing two RNAs in a single tube and a single RNA expressing two antigens., Interpretation: Overall, our data illustrate two vaccine approaches to deliver two antigens in a single immunization. Both approaches induced protective immune responses against CCHFV in this model. These approaches support their continued development for this and future vaccine candidates for CCHFV and other vaccines where inclusion of multiple antigens would be optimal., Funding: This work was supported by the Intramural Research Program, NIAID/NIH, HDT Bio and MCDC Grant #MCDC2204-011., Competing Interests: Declaration of interests J.E. has equity interest in HDT Bio. J.E. and A.K. are a co-inventor on U.S. patent application no. 62/993,307 “Compositions and methods for delivery of RNA” pertaining to formulations for RNA delivery. DWH, JE and HF are inventors on U.S. patent application number 63/365,015 “Replicating RNA vaccine for Crimean-Congo haemorrhagic fever virus” regarding the repRNA for use against CCHFV., (Published by Elsevier B.V.)
- Published
- 2024
- Full Text
- View/download PDF
17. Host and viral determinants of airborne transmission of SARS-CoV-2 in the Syrian hamster.
- Author
-
Port JR, Morris DH, Riopelle JC, Yinda CK, Avanzato VA, Holbrook MG, Bushmaker T, Schulz JE, Saturday TA, Barbian K, Russell CA, Perry-Gottschalk R, Shaia C, Martens C, Lloyd-Smith JO, Fischer RJ, and Munster VJ
- Subjects
- Cricetinae, Animals, Male, Mesocricetus, Respiratory Aerosols and Droplets, SARS-CoV-2, COVID-19
- Abstract
It remains poorly understood how SARS-CoV-2 infection influences the physiological host factors important for aerosol transmission. We assessed breathing pattern, exhaled droplets, and infectious virus after infection with Alpha and Delta variants of concern (VOC) in the Syrian hamster. Both VOCs displayed a confined window of detectable airborne virus (24-48 hr), shorter than compared to oropharyngeal swabs. The loss of airborne shedding was linked to airway constriction resulting in a decrease of fine aerosols (1-10 µm) produced, which are suspected to be the major driver of airborne transmission. Male sex was associated with increased viral replication and virus shedding in the air. Next, we compared the transmission efficiency of both variants and found no significant differences. Transmission efficiency varied mostly among donors, 0-100% (including a superspreading event), and aerosol transmission over multiple chain links was representative of natural heterogeneity of exposure dose and downstream viral kinetics. Co-infection with VOCs only occurred when both viruses were shed by the same donor during an increased exposure timeframe (24-48 hr). This highlights that assessment of host and virus factors resulting in a differential exhaled particle profile is critical for understanding airborne transmission., Competing Interests: JP, DM, JR, CY, VA, MH, TB, JS, TS, KB, CR, RP, CS, CM, JL, RF, VM No competing interests declared
- Published
- 2024
- Full Text
- View/download PDF
18. An atlas of gross and histologic lesions and immunohistochemical immunoreactivity during the temporal progression of aerosolized Lassa virus induced hemorrhagic fever in cynomolgus macaques.
- Author
-
Bohler F, Cashman K, Wilkinson E, Johnson JC, Rosenke K, Shamblin J, Hensley L, Honko A, and Shaia C
- Subjects
- Humans, Animals, Macaca fascicularis, Antigens, Viral, Viremia, Lassa virus, Lassa Fever pathology
- Abstract
Lassa virus (LASV) causes an acute multisystemic hemorrhagic fever in humans known as Lassa fever, which is endemic in several African countries. This manuscript focuses on the progression of disease in cynomolgus macaques challenged with aerosolized LASV and serially sampled for the development and progression of gross and histopathologic lesions. Gross lesions were first noted in tissues on day 6 and persisted throughout day 12. Viremia and histologic lesions were first noted on day 6 commencing with the pulmonary system and hemolymphatic system and progressing at later time points to include all systems. Immunoreactivity to LASV antigen was first observed in the lungs of one macaque on day 3 and appeared localized to macrophages with an increase at later time points to include immunoreactivity in all organ systems. Additionally, this manuscript will serve as a detailed atlas of histopathologic lesions and disease progression for comparison to other animal models of aerosolized Arenaviral disease., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2024 Bohler, Cashman, Wilkinson, Johnson, Rosenke, Shamblin, Hensley, Honko and Shaia.)
- Published
- 2024
- Full Text
- View/download PDF
19. Favipiravir and Ribavirin protect immunocompetent mice from lethal CCHFV infection.
- Author
-
Tipih T, Meade-White K, Rao D, Bushmaker T, Lewis M, Shaia C, Feldmann H, and Hawman DW
- Subjects
- Humans, Male, Animals, Mice, Ribavirin pharmacology, Ribavirin therapeutic use, Tigecycline therapeutic use, Adrenal Cortex Hormones therapeutic use, Hemorrhagic Fever Virus, Crimean-Congo genetics, Hemorrhagic Fever, Crimean
- Abstract
Crimean-Congo hemorrhagic fever virus (CCHFV) causes Crimean-Congo hemorrhagic fever (CCHF) in humans with high morbidity and mortality. Currently, there is neither an approved antiviral drug nor a vaccine against CCHFV. In this study, we describe a lethal model of CCHFV infection using a mouse-adapted strain of CCHFV (MA-CCHFV) in adult wild-type male mice. Infected mice developed high viral loads, tissue pathology, and inflammatory immune responses before ultimately succumbing to the infection. We used the model to evaluate the protective efficacy of nucleoside analogs monulpiravir, favipiravir, ribavirin, the antibiotic tigecycline and the corticosteroids dexamethasone and methylprednisolone against lethal CCHFV infection. Tigecycline, monulpiravir and the corticosteroids failed to protect mice from lethal MA-CCHFV infection. In contrast, favipiravir and ribavirin protected animals from clinical disease and death even when treatment was delayed. Despite demonstrating uniform protection, CCHFV RNA persisted in survivors treated with favipiravir and ribavirin. Nevertheless, the study demonstrated the anti-CCHFV efficacy of favipiravir and ribavirin in a model with intact innate immunity and establishes this model for continued development of CCHFV countermeasures., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Published by Elsevier B.V.)
- Published
- 2023
- Full Text
- View/download PDF
20. Temporal changes in pathology and viral RNA distribution in guinea pigs following separate infection with two New World Arenaviruses.
- Author
-
Cline C, Zeng X, Bell TM, Shaia C, Facemire P, Williams J, Davis N, Babka A, Picado E, Fitzpatrick C, and Golden JW
- Subjects
- Humans, Guinea Pigs, Animals, RNA, Viral genetics, Liver, Brain, Arenaviruses, New World, Junin virus
- Abstract
Numerous arenaviruses have been identified throughout the Americas and a subset of these viruses cause viral hemorrhagic fever in humans. This study compared the pathology and viral RNA distribution in Hartley guinea pigs challenged with two human-disease causing New World arenaviruses, Junin virus (JUNV) or Guanarito virus (GTOV). Histopathologic analysis and RNA in situ hybridization revealed similar pathology and viral RNA distribution for both groups of animals challenged with either JUNV or GTOV on days 3, 7, 10 and 12 post exposure (PE). Gross lesions were first observed on day 7 and primarily involved the lungs and liver. The most severe histologic lesions occurred in the lymph nodes, spleen, and thymus and included lymphoid depletion and necrosis which increased in severity over time. Extensive necrosis was also observed in the bone marrow on day 12. Minimal to mild inflammation with and without necrosis was observed in the choroid plexus of the brain, choroid of the eye, intestinal tract, lung and adrenal gland. Significant liver lesions were rare, consisting predominantly of hepatocyte vacuolation. Viral RNA labeling was identified in nearly all organs examined, was often extensive in certain organs and generally increased over time starting on day 7. Our data demonstrate the guinea pig may serve as a useful model to study New World arenavirus infection in humans and for the evaluation and development of medical countermeasures., Competing Interests: The authors have declared that no competing interests exist., (Copyright: This is an open access article, free of all copyright, and may be freely reproduced, distributed, transmitted, modified, built upon, or otherwise used by anyone for any lawful purpose. The work is made available under the Creative Commons CC0 public domain dedication.)
- Published
- 2023
- Full Text
- View/download PDF
21. Single-dose VSV-based vaccine protects against Kyasanur Forest disease in nonhuman primates.
- Author
-
Bhatia B, Tang-Huau TL, Feldmann F, Hanley PW, Rosenke R, Shaia C, Marzi A, and Feldmann H
- Subjects
- Animals, Vaccination, Cross Reactions, Macaca, Kyasanur Forest Disease prevention & control, Vaccines
- Abstract
Kyasanur Forest disease virus (KFDV) is an endemic arbovirus in western India mainly transmitted by hard ticks of the genus Haemaphysalis . KFDV causes Kyasanur Forest disease (KFD), a syndrome including fever, gastrointestinal symptoms, and hemorrhages. There are no approved treatments, and the efficacy of the only vaccine licensed in India has recently been questioned. Here, we studied the protective efficacy of a vesicular stomatitis virus (VSV)-based vaccine expressing the KFDV precursor membrane and envelope proteins (VSV-KFDV) in pigtailed macaques. VSV-KFDV vaccination was found to be safe and elicited strong humoral and cellular immune responses. A single-dose vaccination reduced KFDV loads and pathology and protected macaques from KFD-like disease. Furthermore, VSV-KFDV elicited cross-reactive neutralizing immune responses to Alkhurma hemorrhagic fever virus, a KFDV variant found in Saudi Arabia.
- Published
- 2023
- Full Text
- View/download PDF
22. Immunological correlates of protection afforded by PHV02 live, attenuated recombinant vesicular stomatitis virus vector vaccine against Nipah virus disease.
- Author
-
Monath TP, Nichols R, Feldmann F, Griffin A, Haddock E, Callison J, Meade-White K, Okumura A, Lovaglio J, Hanley PW, Clancy CS, Shaia C, Rida W, and Fusco J
- Subjects
- Animals, Chlorocebus aethiops, Antibodies, Neutralizing, Nipah Virus, Ebola Vaccines, Henipavirus Infections prevention & control, Vesicular Stomatitis, Hemorrhagic Fever, Ebola
- Abstract
Introduction: Immune correlates of protection afforded by PHV02, a recombinant vesicular stomatitis (rVSV) vector vaccine against Nipah virus (NiV) disease, were investigated in the African green monkey (AGM) model. Neutralizing antibody to NiV has been proposed as the principal mediator of protection against future NiV infection., Methods: Two approaches were used to determine the correlation between neutralizing antibody levels and outcomes following a severe (1,000 median lethal doses) intranasal/intratracheal (IN/IT) challenge with NiV (Bangladesh): (1) reduction in vaccine dose given 28 days before challenge and (2) challenge during the early phase of the antibody response to the vaccine., Results: Reduction in vaccine dose to very low levels led to primary vaccine failure rather than a sub-protective level of antibody. All AGMs vaccinated with the nominal clinical dose (2 × 10
7 pfu) at 21, 14, or 7 days before challenge survived. AGMs vaccinated at 21 days before challenge had neutralizing antibodies (geometric mean titer, 71.3). AGMs vaccinated at 7 or 14 days before challenge had either undetectable or low neutralizing antibody titers pre-challenge but had a rapid rise in titers after challenge that abrogated the NiV infection. A simple logistic regression model of the combined studies was used, in which the sole explanatory variable was pre-challenge neutralizing antibody titers. For a pre-challenge titer of 1:5, the predicted survival probability is 100%. The majority of animals with pre-challenge neutralizing titer of ≥1:20 were protected against pulmonary infiltrates on thoracic radiograms, and a majority of those with titers ≥1:40 were protected against clinical signs of illness and against a ≥fourfold antibody increase following challenge (indicating sterile immunity). Controls receiving rVSV-Ebola vaccine rapidly succumbed to NiV challenge, eliminating the innate immunity stimulated by the rVSV vector as a contributor to survival in monkeys challenged as early as 7 days after vaccination., Discussion and Conclusion: It was concluded that PHV02 vaccine elicited a rapid onset of protection and that any detectable level of neutralizing antibody was a functional immune correlate of survival., Competing Interests: TM is employed by Crozet Biopharma LLC. RN and JF are employed by Public Health Vaccines Inc. WR is an independent consultant. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Monath, Nichols, Feldmann, Griffin, Haddock, Callison, Meade-White, Okumura, Lovaglio, Hanley, Clancy, Shaia, Rida and Fusco.)- Published
- 2023
- Full Text
- View/download PDF
23. Histopathologic Characterization of Experimental Peracute SARS-CoV-2 Infection in the Syrian Hamster.
- Author
-
Clancy CS, Meade-White K, Shaia C, Saturday G, Feldmann H, and Rosenke K
- Abstract
Coronavirus Infectious Disease 2019 (COVID-19) initiated a global pandemic that thus far has resulted in the death of over 6.5 million people internationally. Understanding the viral tropism during the initial, subclinical phase of infection is critical to develop targeted vaccines and therapeutics. With the continued emergence of variants of concern, particularly those that appear to have a tropism for the upper respiratory tract, understanding the complete pathogenesis is critical to develop more effective interventions. Thus far, the Syrian hamster has served as the most consistent small animal model of SARS-CoV-2 infection for mild to moderate respiratory disease. Herein, we utilize histopathology and immunohistochemistry to characterize the peracute phase of disease initiating at 6-h-post-inoculation in the intranasal inoculation route Syrian hamster model. Inflammation and viral replication initiates in the respiratory epithelium of nasal turbinates as early as 12-h-post-inoculation and moves caudally through the nasal cavity by 36-h-post inoculation. Lower respiratory involvement can be detected as early as 12-h-post inoculation in the intranasal inoculated hamster model. These data highlight the importance of rostral nasal cavity sampling at early timepoints for detection of SARS-CoV-2 in the Syrian hamster model.
- Published
- 2023
- Full Text
- View/download PDF
24. Genetically diverse mouse models of SARS-CoV-2 infection reproduce clinical variation in type I interferon and cytokine responses in COVID-19.
- Author
-
Robertson SJ, Bedard O, McNally KL, Shaia C, Clancy CS, Lewis M, Broeckel RM, Chiramel AI, Shannon JG, Sturdevant GL, Rosenke R, Anzick SL, Forte E, Preuss C, Baker CN, Harder JM, Brunton C, Munger S, Bruno DP, Lack JB, Leung JM, Shamsaddini A, Gardina P, Sturdevant DE, Sun J, Martens C, Holland SM, Rosenthal NA, and Best SM
- Subjects
- Humans, Mice, Animals, Cytokines, SARS-CoV-2, Mice, Transgenic, Inflammation genetics, Disease Models, Animal, Lung, COVID-19, Interferon Type I
- Abstract
Inflammation in response to severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection drives severity of coronavirus disease 2019 (COVID-19) and is influenced by host genetics. To understand mechanisms of inflammation, animal models that reflect genetic diversity and clinical outcomes observed in humans are needed. We report a mouse panel comprising the genetically diverse Collaborative Cross (CC) founder strains crossed to human ACE2 transgenic mice (K18-hACE2) that confers susceptibility to SARS-CoV-2. Infection of CC x K18-hACE2 resulted in a spectrum of survival, viral replication kinetics, and immune profiles. Importantly, in contrast to the K18-hACE2 model, early type I interferon (IFN-I) and regulated proinflammatory responses were required for control of SARS-CoV-2 replication in PWK x K18-hACE2 mice that were highly resistant to disease. Thus, virus dynamics and inflammation observed in COVID-19 can be modeled in diverse mouse strains that provide a genetically tractable platform for understanding anti-coronavirus immunity., (© 2023. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)
- Published
- 2023
- Full Text
- View/download PDF
25. Targeting 2-Oxoglutarate-Dependent Dioxygenases Promotes Metabolic Reprogramming That Protects against Lethal SARS-CoV-2 Infection in the K18-hACE2 Transgenic Mouse Model.
- Author
-
Jessop F, Schwarz B, Bohrnsen E, Miltko M, Shaia C, and Bosio CM
- Subjects
- Mice, Animals, Mice, Transgenic, Ketoglutaric Acids, SARS-CoV-2, Angiotensin-Converting Enzyme 2, COVID-19, Dioxygenases
- Abstract
Dysregulation of host metabolism is a feature of lethal SARS-CoV-2 infection. Perturbations in α-ketoglutarate levels can elicit metabolic reprogramming through 2-oxoglutarate-dependent dioxygenases (2-ODDGs), leading to stabilization of the transcription factor HIF-1α. HIF1-α activation has been reported to promote antiviral mechanisms against SARS-CoV-2 through direct regulation of ACE2 expression (a receptor required for viral entry). However, given the numerous pathways HIF-1α serves to regulate it is possible that there are other undefined metabolic mechanisms contributing to the pathogenesis of SARS-CoV-2 independent of ACE2 downregulation. In this study, we used in vitro and in vivo models in which HIF-1α modulation of ACE2 expression was negated, allowing for isolated characterization of the host metabolic response within SARS-CoV-2 disease pathogenesis. We demonstrated that SARS-CoV-2 infection limited stabilization of HIF-1α and associated mitochondrial metabolic reprogramming by maintaining activity of the 2-ODDG prolyl hydroxylases. Inhibition of 2-ODDGs with dimethyloxalylglycine promoted HIF-1α stabilization following SARS-CoV-2 infection, and significantly increased survival among SARS-CoV-2-infected mice compared with vehicle controls. However, unlike previous reports, the mechanism by which activation of HIF-1α responses contributed to survival was not through impairment of viral replication. Rather, dimethyloxalylglycine treatment facilitated direct effects on host metabolism including increased glycolysis and resolution of dysregulated pools of metabolites, which correlated with reduced morbidity. Taken together, these data identify (to our knowledge) a novel function of α-ketoglutarate-sensing platforms, including those responsible for HIF-1α stabilization, in the resolution of SARS-CoV-2 infection and support targeting these metabolic nodes as a viable therapeutic strategy to limit disease severity during infection., (Copyright © 2023 The Authors.)
- Published
- 2023
- Full Text
- View/download PDF
26. Of Murines and Humans: Modeling Persistent Powassan Disease in C57BL/6 Mice.
- Author
-
Scroggs SLP, Offerdahl DK, Stewart PE, Shaia C, Griffin AJ, and Bloom ME
- Subjects
- Humans, Female, Animals, Mice, Male, Mice, Inbred C57BL, Brain pathology, Inflammation, RNA, Viral, Encephalitis, Tick-Borne
- Abstract
Powassan infection is caused by two closely related, tick-transmitted viruses of the genus Flavivirus (family Flaviviridae ): Powassan virus lineage I (POWV) and lineage II (known as deer tick virus [DTV]). Infection is typically asymptomatic or mild but can progress to neuroinvasive disease. Approximately 10% of neuroinvasive cases are fatal, and half of the survivors experience long-term neurological sequelae. Understanding how these viruses cause long-term symptoms as well as the possible role of viral persistence is important for developing therapies. We intraperitoneally inoculated 6-week-old C57BL/6 mice (50% female) with 10
3 focus-forming units (FFU) DTV and assayed for infectious virus, viral RNA, and inflammation during acute infection and 21, 56, and 84 days postinfection (dpi). Although most mice (86%) were viremic 3 dpi, only 21% of the mice were symptomatic and 83% recovered. Infectious virus was detected only in the brains of mice sampled during the acute infection. Viral RNA was detected in the brain until 84 dpi, but the magnitude decreased over time. Meningitis and encephalitis were visible in acute mice and from mice sampled at 21 dpi. Inflammation was observed until 56 dpi in the brain and 84 dpi in the spinal cord, albeit at low levels. These results suggest that the long-term neurological symptoms associated with Powassan disease are likely caused by lingering viral RNA and chronic inflammation in the central nervous system rather than by a persistent, active viral infection. The C57BL/6 model of persistent Powassan mimics illness in humans and can be used to study the mechanisms of chronic disease. IMPORTANCE Half of Powassan infection survivors experience long-term, mild to severe neurological symptoms. The progression from acute to chronic Powassan disease is not well understood, severely limiting treatment and prevention options. Infection of C57BL/6 mice with DTV mimics clinical disease in humans, and the mice exhibit CNS inflammation and viral RNA persistence until at least 86 dpi, while infectious virus is undetectable after 12 dpi. These findings suggest that the long-term neurological symptoms of chronic Powassan disease are in part due the persistence of viral RNA and the corresponding long-term inflammation of the brain and spinal cord. Our work demonstrates that C57BL/6 mice can be used to study the pathogenesis of chronic Powassan disease.- Published
- 2023
- Full Text
- View/download PDF
27. Genetically diverse mouse models of SARS-CoV-2 infection reproduce clinical variation in type I interferon and cytokine responses in COVID-19.
- Author
-
Robertson S, Bedard O, McNally K, Shaia C, Clancy C, Lewis M, Broeckel R, Chiramel A, Shannon JG, Sturdevant G, Rosenke R, Anzick SL, Forte E, Preuss C, Baker C, Harder J, Brunton C, Munger SC, Bruno DP, Lack JB, Leung JM, Shamsaddini A, Gardina P, Sturdevant D, Sun J, Martens C, Holland S, Rosenthal N, and Best S
- Abstract
Inflammation in response to severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection drives severity of coronavirus disease 2019 (COVID-19) and is influenced by host genetics. To understand mechanisms of inflammation, animal models that reflect genetic diversity and clinical outcomes observed in humans are needed. We report a mouse panel comprising the genetically diverse Collaborative Cross (CC) founder strains crossed to human ACE2 transgenic mice (K18-hACE2) that confers susceptibility to SARS-CoV-2. Infection of CC x K18- hACE2 resulted in a spectrum of survival, viral replication kinetics, and immune profiles. Importantly, in contrast to the K18-hACE2 model, early type I interferon (IFN-I) and regulated proinflammatory responses were required for control of SARS-CoV-2 replication in PWK x K18-hACE2 mice that were highly resistant to disease. Thus, virus dynamics and inflammation observed in COVID-19 can be modeled in diverse mouse strains that provide a genetically tractable platform for understanding anti-coronavirus immunity.
- Published
- 2023
- Full Text
- View/download PDF
28. Combined molnupiravir-nirmatrelvir treatment improves the inhibitory effect on SARS-CoV-2 in macaques.
- Author
-
Rosenke K, Lewis MC, Feldmann F, Bohrnsen E, Schwarz B, Okumura A, Bohler WF, Callison J, Shaia C, Bosio CM, Lovaglio J, Saturday G, Jarvis MA, and Feldmann H
- Subjects
- Animals, Humans, Macaca mulatta, Ferrets, Lactams, Leucine, Nitriles, Antiviral Agents, SARS-CoV-2, COVID-19
- Abstract
The periodic emergence of SARS-CoV-2 variants of concern (VOCs) with unpredictable clinical severity and ability to escape preexisting immunity emphasizes the continued need for antiviral interventions. Two small molecule inhibitors, molnupiravir (MK-4482), a nucleoside analog, and nirmatrelvir (PF-07321332), a 3C-like protease inhibitor, have recently been approved as monotherapy for use in high-risk patients with COVID-19. As preclinical data are only available for rodent and ferret models, here we assessed the efficacy of MK-4482 and PF-07321332 alone and in combination against infection with the SARS-CoV-2 Delta VOC in the rhesus macaque COVID-19 model. Macaques were infected with the SARS-CoV-2 Delta variant and treated with vehicle, MK-4482, PF-07321332, or a combination of MK-4482 and PF-07321332. Clinical exams were performed at 1, 2, and 4 days postinfection to assess disease and virological parameters. Notably, use of MK-4482 and PF-07321332 in combination improved the individual inhibitory effect of both drugs, resulting in milder disease progression, stronger reduction of virus shedding from mucosal tissues of the upper respiratory tract, stronger reduction of viral replication in the lower respiratory tract, and reduced lung pathology. Our data strongly indicate superiority of combined MK-4482 and PF-07321332 treatment of SARS-CoV-2 infections as demonstrated in the closest COVID-19 surrogate model of human infection.
- Published
- 2023
- Full Text
- View/download PDF
29. Severe acute respiratory disease in American mink experimentally infected with SARS-CoV-2.
- Author
-
Adney DR, Lovaglio J, Schulz JE, Yinda CK, Avanzato VA, Haddock E, Port JR, Holbrook MG, Hanley PW, Saturday G, Scott D, Shaia C, Nelson AM, Spengler JR, Tansey C, Cossaboom CM, Wendling NM, Martens C, Easley J, Yap SW, Seifert SN, and Munster VJ
- Subjects
- Humans, Animals, Mink, Lung diagnostic imaging, SARS-CoV-2, COVID-19
- Abstract
An animal model that fully recapitulates severe COVID-19 presentation in humans has been a top priority since the discovery of SARS-CoV-2 in 2019. Although multiple animal models are available for mild to moderate clinical disease, models that develop severe disease are still needed. Mink experimentally infected with SARS-CoV-2 developed severe acute respiratory disease, as evident by clinical respiratory disease, radiological, and histological changes. Virus was detected in nasal, oral, rectal, and fur swabs. Deep sequencing of SARS-CoV-2 from oral swabs and lung tissue samples showed repeated enrichment for a mutation in the gene encoding nonstructural protein 6 in open reading frame 1ab. Together, these data indicate that American mink develop clinical features characteristic of severe COVID-19 and, as such, are uniquely suited to test viral countermeasures.
- Published
- 2022
- Full Text
- View/download PDF
30. SARS-CoV-2 Omicron BA.1 and BA.2 are attenuated in rhesus macaques as compared to Delta.
- Author
-
van Doremalen N, Singh M, Saturday TA, Yinda CK, Perez-Perez L, Bohler WF, Weishampel ZA, Lewis M, Schulz JE, Williamson BN, Meade-White K, Gallogly S, Okumura A, Feldmann F, Lovaglio J, Hanley PW, Shaia C, Feldmann H, de Wit E, Munster VJ, and Rosenke K
- Abstract
Since the emergence of SARS-CoV-2, five different variants of concern (VOCs) have been identified: Alpha, Beta, Gamma, Delta, and Omicron. Because of confounding factors in the human population, such as preexisting immunity, comparing severity of disease caused by different VOCs is challenging. Here, we investigate disease progression in the rhesus macaque model upon inoculation with the Delta, Omicron BA.1, and Omicron BA.2 VOCs. Disease severity in rhesus macaques inoculated with Omicron BA.1 or BA.2 was lower than those inoculated with Delta and resulted in significantly lower viral loads in nasal swabs, bronchial cytology brush samples, and lung tissue in rhesus macaques. Cytokines and chemokines were up-regulated in nasosorption samples of Delta animals compared to Omicron BA.1 and BA.2 animals. Overall, these data suggest that, in rhesus macaques, Omicron replicates to lower levels than the Delta VOC, resulting in reduced clinical disease.
- Published
- 2022
- Full Text
- View/download PDF
31. Characterization of Coxiella burnetii Dugway Strain Host-Pathogen Interactions In Vivo.
- Author
-
Tesfamariam M, Binette P, Cockrell D, Beare PA, Heinzen RA, Shaia C, and Long CM
- Abstract
Coxiella burnetii is a Gram-negative, intracellular bacterium that causes the zoonosis Q fever. Among the many natural isolates of C. burnetii recovered from various sources, the Dugway group exhibits unique genetic characteristics, including the largest C. burnetii genomes. These strains were isolated during 1954-1958 from wild rodents from the Utah, USA desert. Despite retaining phase I lipopolysaccharide and the type 4B secretion system, two critical virulence factors, avirulence has been reported in a guinea pig infection model. Using guinea pig models, we evaluated the virulence, whole-cell vaccine (WCV) efficacy, and post-vaccination hypersensitivity (PVH) potential of a representative Dugway strain. Consistent with prior reports, Dugway appeared to be highly attenuated compared to a virulent strain. Indeed, Dugway-infected animals showed similarly low levels of fever, body weight loss, and splenomegaly like Nine Mile II-infected animals. When compared to a human Q fever vaccine, QVax
® , Dugway WCV exhibited analogous protection against a heterologous Nine Mile I challenge. PVH was investigated in a skin-testing model which revealed significantly decreased maximum erythema in Dugway Δ dot/icm WCV-skin-tested animals compared to that of QVax® . These data provide insight into this unique bacterial strain and implicate its potential use as a mutated WCV candidate.- Published
- 2022
- Full Text
- View/download PDF
32. Contribution of Lipid Mediators in Divergent Outcomes following Acute Bacterial and Viral Lung Infections in the Obese Host.
- Author
-
Schwarz B, Roberts LM, Bohrnsen E, Jessop F, Wehrly TD, Shaia C, and Bosio CM
- Subjects
- Animals, Chemokines metabolism, Cytokines metabolism, Lipids, Lung microbiology, Mice, Mice, Inbred C57BL, Obesity metabolism, SARS-CoV-2, COVID-19, Francisella tularensis, Tularemia, Virus Diseases metabolism
- Abstract
Obesity is considered an important comorbidity for a range of noninfectious and infectious disease states including those that originate in the lung, yet the mechanisms that contribute to this susceptibility are not well defined. In this study, we used the diet-induced obesity (DIO) mouse model and two models of acute pulmonary infection, Francisella tularensis subspecies tularensis strain SchuS4 and SARS-CoV-2, to uncover the contribution of obesity in bacterial and viral disease. Whereas DIO mice were more resistant to infection with SchuS4, DIO animals were more susceptible to SARS-CoV-2 infection compared with regular weight mice. In both models, neither survival nor morbidity correlated with differences in pathogen load, overall cellularity, or influx of inflammatory cells in target organs of DIO and regular weight animals. Increased susceptibility was also not associated with exacerbated production of cytokines and chemokines in either model. Rather, we observed pathogen-specific dysregulation of the host lipidome that was associated with vulnerability to infection. Inhibition of specific pathways required for generation of lipid mediators reversed resistance to both bacterial and viral infection. Taken together, our data demonstrate disparity among obese individuals for control of lethal bacterial and viral infection and suggest that dysregulation of the host lipidome contributes to increased susceptibility to viral infection in the obese host.
- Published
- 2022
- Full Text
- View/download PDF
33. Combined Molnupiravir and Nirmatrelvir Treatment Improves the Inhibitory Effect on SARS-CoV-2 in Rhesus Macaques.
- Author
-
Rosenke K, Lewis MC, Feldmann F, Bohrnsen E, Schwarz B, Okumura A, Bohler WF, Callison J, Shaia C, Bosio CM, Lovaglio J, Saturday G, Jarvis MA, and Feldmann H
- Abstract
The periodic emergence of SARS-CoV-2 variants of concern (VOCs) with unpredictable clinical severity and ability to escape preexisting immunity emphasizes the continued need for antiviral interventions. Two small molecule inhibitors, molnupiravir (MK-4482), a nucleoside analog, and nirmatrelvir (PF-07321332), a 3C-like protease inhibitor, have each recently been approved as monotherapy for use in high risk COVID-19 patients. As preclinical data are only available for rodent and ferret models, we originally assessed the efficacy of MK-4482 and PF-07321332 alone and then in combination Against infection with the SARS-CoV-2 Delta VOC in the rhesus macaque COVID-19 model. Notably, use of MK-4482 and PF-07321332 in combination improved the individual inhibitory effect of both drugs. Combined treatment resulted in milder disease progression, stronger reduction of virus shedding from mucosal tissues of the upper respiratory tract, stronger reduction of viral replication in the lower respiratory tract, and reduced lung pathology. Our data strongly indicate superiority of combined MK-4482 and PF-07321332 treatment of SARS-CoV-2 infections as demonstrated here in the closest COVID-19 surrogate model., One Sentence Summary: The combination of molnupiravir and nirmatrelvir inhibits SARS-CoV-2 replication and shedding more effectively than individual treatments in the rhesus macaque model.
- Published
- 2022
- Full Text
- View/download PDF
34. Molnupiravir inhibits SARS-CoV-2 variants including Omicron in the hamster model.
- Author
-
Rosenke K, Okumura A, Lewis MC, Feldmann F, Meade-White K, Bohler WF, Griffin A, Rosenke R, Shaia C, Jarvis MA, and Feldmann H
- Subjects
- Animals, Cricetinae, Cytidine analogs & derivatives, Humans, Hydroxylamines, SARS-CoV-2, COVID-19 Drug Treatment
- Abstract
The recent emergence of the SARS-CoV-2 Omicron variant of concern (VOC), which contains a heavily mutated spike protein capable of escaping preexisting immunity, identifies a continued need for interventional measures. Molnupiravir (MK-4482), an orally administered nucleoside analog, has demonstrated efficacy against earlier SARS-CoV-2 lineages and was recently approved for SARS-CoV-2 infections in high-risk adults. Here, we assessed the efficacy of MK-4482 against the earlier Alpha, Beta, and Delta VOCs and Omicron in the hamster COVID-19 model. Omicron replication and associated lung disease in vehicle-treated hamsters was reduced compared with replication and lung disease associated with earlier VOCs. MK-4482 treatment inhibited virus replication in the lungs of hamsters infected with Alpha, Beta, or Delta VOCs. Importantly, MK-4482 profoundly inhibited virus replication in the upper and lower respiratory tract of hamsters infected with the Omicron VOC. Consistent with its mutagenic mechanism, MK-4482 treatment had a more pronounced inhibitory effect on infectious titers compared with viral RNA genome load. Histopathologic analysis showed that MK-4482 treatment caused a concomitant reduction in the level of lung disease and viral antigen load in infected hamsters across all VOCs examined. Together, our data indicate the potential of MK-4482 as an effective antiviral against known SARS-CoV-2 VOCs, especially Omicron, and likely future SARS-CoV-2 variants.
- Published
- 2022
- Full Text
- View/download PDF
35. Histologic pulmonary lesions of SARS-CoV-2 in 4 nonhuman primate species: An institutional comparative review.
- Author
-
Clancy CS, Shaia C, Munster V, de Wit E, Hawman D, Okumura A, Feldmann H, Saturday G, and Scott D
- Subjects
- Animals, Chlorocebus aethiops, Lung pathology, Macaca mulatta, Pandemics veterinary, COVID-19 veterinary, SARS-CoV-2
- Abstract
Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is an emergent, amphixenotic infection that resulted in a pandemic declaration in March 2020. A rapid search for appropriate animal models of this newly emergent viral respiratory disease focused initially on traditional nonhuman primate research species. Nonhuman primate models have previously been shown to be valuable in evaluation of emerging respiratory coronaviruses with pandemic potential (ie, SARS-CoV and Middle East respiratory syndrome coronavirus). In this article, we review the pulmonary histopathologic characteristics and immunohistochemical evaluation of experimental SARS-CoV-2 infection in the rhesus macaque, pigtail macaque, African green monkey, and squirrel monkey. Our results indicate that all evaluated nonhuman primate species developed variably severe histopathologic changes typical of coronavirus respiratory disease characterized by interstitial pneumonia with or without syncytial cell formation, alveolar fibrin, and pulmonary edema that progressed to type II pneumocyte hyperplasia. Lesion distribution was multifocal, frequently subpleural, and often more severe in lower lung lobes. However, squirrel monkeys showed the least severe and least consistent lesions of the evaluated nonhuman primates. Additionally, our results highlight the disparate physical relationship between viral antigen and foci of pulmonary lesions. While classic respiratory coronaviral lesions were observed in the lungs of all nonhuman primates evaluated, none of the primates exhibited severe lesions or evidence of diffuse alveolar damage and therefore are unlikely to represent the severe form of SARS-CoV-2 infection observed in fatal human cases.
- Published
- 2022
- Full Text
- View/download PDF
36. Murine Q Fever Vaccination Model Reveals Sex Dimorphism in Early Phase Delayed-Type Hypersensitivity Responses.
- Author
-
Binette P, Tesfamariam M, Cockrell D, Heinzen RA, Richards C, Shaia C, and Long CM
- Subjects
- Animals, Bacterial Vaccines, Female, Male, Mice, Sex Characteristics, Vaccination, Coxiella burnetii, Q Fever
- Abstract
Delayed-type hypersensitivity (DTH) responses to microbial vaccines and related components are a major roadblock for widespread licensing of whole cell vaccines such as that of Q fever. Q fever is a zoonotic disease caused by the intracellular bacterium Coxiella burnetii . The only currently licensed vaccine, Q-Vax
® , is a whole cell inactivated formulation that is associated with a potentially severe dermal post vaccination DTH response in previously sensitized individuals. To investigate the underlying immunologic mechanisms of this response and better represent the early-phase DTH response observed in humans, a murine sensitization and skin testing model was developed and employed. Female C57Bl/6J mice displayed the most robust early-phase DTH responses following sensitization and elicitation compared to their male counterparts and other mouse strains. Immunologic responses were measured within the skin, draining lymph nodes, and serum following both sensitization and elicitation with Q fever whole cell vaccines. Local immunologic responses in the dermis were characterized by inflammation primarily involving neutrophils, macrophages, and T cells. Secondary lymphoid organ profiling revealed distinct immunological signatures following both sensitization and elicitation with a sex-based dichotomy in T cell phenotypes and antigen presenting cell numbers. Beyond providing a post-Q fever vaccination DTH model that recapitulates early-phase DTH events, these data suggest that sex is a primary factor influencing the magnitude and composition of the ensuing response., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Binette, Tesfamariam, Cockrell, Heinzen, Richards, Shaia and Long.)- Published
- 2022
- Full Text
- View/download PDF
37. Toward Optimization of a Rabbit Model of Staphylococcus aureus (USA300) Skin and Soft Tissue Infection.
- Author
-
Malachowa N, McGuinness W, Kobayashi SD, Porter AR, Shaia C, Lovaglio J, Smith B, Rungelrath V, Saturday G, Scott DP, Falugi F, Missiakas D, Schneewind O, and DeLeo FR
- Subjects
- Animals, Rabbits, Reinfection, Staphylococcus aureus, United States, Methicillin-Resistant Staphylococcus aureus genetics, Soft Tissue Infections, Staphylococcal Infections microbiology, Staphylococcal Skin Infections microbiology, Vaccines
- Abstract
Staphylococcus aureus remains a leading cause of skin and soft tissue infections (SSTIs) globally. In the United States, many of these infections are caused by isolates classified as USA300. Our understanding of the success of USA300 as a human pathogen is due in part to data obtained from animal infection models, including rabbit SSTI models. These animal models have been used to study S. aureus virulence and pathogenesis and to gain an enhanced understanding of the host response to infection. Although significant knowledge has been gained, the need to use a relatively high inoculum of USA300 (1 × 10
8 to 5 × 108 CFU) is a caveat of these infection models. As a step toward addressing this issue, we created mutations in USA300 that mimic those found in S. aureus strains with naturally occurring rabbit tropism-namely, single nucleotide polymorphisms in dltB and/or deletion of rot . We then developed a rabbit SSTI model that utilizes an inoculum of 106 USA300 CFU to cause reproducible disease and tested whether primary SSTI protects rabbits against severe reinfection caused by the same strain. Although there was modest protection against severe reinfection, primary infection and reinfection with rabbit-tropic USA300 strains failed to increase the overall level of circulating anti-S. aureus antibodies significantly. These findings provide additional insight into the host response to S. aureus. More work is needed to further develop a low-inoculum infection model that can be used to better test the potential of new therapeutics or vaccine target antigens. IMPORTANCE Animal models of S. aureus infection are important for evaluating bacterial pathogenesis and host immune responses. These animal infection models are often used as an initial step in the testing of vaccine antigens and new therapeutics. The extent to which animal models of S. aureus infection approximate human infections remains a significant consideration for translation of results to human clinical trials. Although significant progress has been made with rabbit models of S. aureus infection, one concern is the high inoculum needed to cause reproducible disease. Here, we generated USA300 strains that have tropism for rabbits and developed a rabbit SSTI model that uses fewer CFU than previous models.- Published
- 2022
- Full Text
- View/download PDF
38. Molnupiravir (MK-4482) is efficacious against Omicron and other SARS-CoV-2 variants in the Syrian hamster COVID-19 model.
- Author
-
Rosenke K, Okumura A, Lewis MC, Feldmann F, Meade-White K, Bohler WF, Griffin A, Rosenke R, Shaia C, Jarvis MA, and Feldmann H
- Abstract
The recent emergence of the SARS-CoV-2 Omicron variant of concern (VOC) containing a heavily mutated spike protein capable of escaping preexisting immunity, identifies a continued need for interventional measures. Molnupiravir (MK-4482), an orally administered nucleoside analog, has demonstrated efficacy against earlier SARS-CoV-2 lineages and was recently approved for SARS-CoV-2 infections in high-risk adults. Here we assessed the efficacy of MK-4482 against the earlier Alpha, Beta and Delta VOCs and Omicron in the Syrian hamster COVID-19 model. Omicron replication and associated lung disease in vehicle treated hamsters was reduced compared to the earlier VOCs. MK-4482 treatment inhibited virus replication in the lungs of Alpha, Beta and Delta VOC infected hamsters. Importantly, MK-4482 profoundly inhibited virus replication in the upper and lower respiratory tract of hamsters infected with the Omicron VOC. Consistent with its mutagenic mechanism, MK-4482 treatment had a more pronounced inhibitory effect on infectious virus titers compared to viral RNA genome load. Histopathologic analysis showed that MK-4482 treatment caused a concomitant reduction in the level of lung disease and viral antigen load in infected hamsters across all VOCs examined. Together, our data indicate the potential of MK-4482 as an effective antiviral against known SARS-CoV-2 VOCs, especially Omicron, and likely future SARS-CoV-2 variants., Competing Interests: Competing interests: MAJ is employed, in part, by the Vaccine Group Ltd. All authors declare that they have no competing interests.
- Published
- 2022
- Full Text
- View/download PDF
39. SARS-CoV2 variant-specific replicating RNA vaccines protect from disease following challenge with heterologous variants of concern.
- Author
-
Hawman DW, Meade-White K, Archer J, Leventhal SS, Wilson D, Shaia C, Randall S, Khandhar AP, Krieger K, Hsiang TY, Gale M, Berglund P, Fuller DH, Feldmann H, and Erasmus JH
- Subjects
- Animals, Antibodies, Neutralizing, COVID-19 Vaccines, Cricetinae, Humans, Mice, RNA, Viral, Spike Glycoprotein, Coronavirus metabolism, Vaccines, Synthetic, mRNA Vaccines, COVID-19 prevention & control, SARS-CoV-2 genetics
- Abstract
Despite mass public health efforts, the SARS-CoV2 pandemic continues as of late 2021 with resurgent case numbers in many parts of the world. The emergence of SARS-CoV2 variants of concern (VoCs) and evidence that existing vaccines that were designed to protect from the original strains of SARS-CoV-2 may have reduced potency for protection from infection against these VoC is driving continued development of second-generation vaccines that can protect against multiple VoC. In this report, we evaluated an alphavirus-based replicating RNA vaccine expressing Spike proteins from the original SARS-CoV-2 Alpha strain and recent VoCs delivered in vivo via a lipid inorganic nanoparticle. Vaccination of both mice and Syrian Golden hamsters showed that vaccination induced potent neutralizing titers against each homologous VoC but reduced neutralization against heterologous challenges. Vaccinated hamsters challenged with homologous SARS-CoV2 variants exhibited complete protection from infection. In addition, vaccinated hamsters challenged with heterologous SARS-CoV-2 variants exhibited significantly reduced shedding of infectious virus. Our data demonstrate that this vaccine platform can be updated to target emergent VoCs, elicits significant protective immunity against SARS-CoV2 variants and supports continued development of this platform., Competing Interests: DH, KM, SL, DW, CS, SR, KK, TH, HF No competing interests declared, JA, MG has equity interest in HDT Bio, AK has equity interest in HDT Bio. Is a co-inventors on U.S. patent application no. 62/993,307 "Compositions and methods for delivery of RNA" pertaining to the LION formulation, PB has equity interest in HDT Bio. Is a consultant for Arcturus, Sensei, and Next Phase, DF has equity interest in HDT Bio. Is a consultant for Gerson Lehrman Group, Orlance, Abacus Bioscience, Neoleukin Therapeutics, JE has equity interest in HDT Bio. Is a consultant for InBios. Is a co-inventors on U.S. patent application no. 62/993,307 "Compositions and methods for delivery of RNA" pertaining to the LION formulation
- Published
- 2022
- Full Text
- View/download PDF
40. Three-Week Old Pigs Are Not Susceptible to Productive Infection with SARS-COV-2.
- Author
-
Haddock E, Callison J, Seifert SN, Okumura A, Tang-Huau TL, Leventhal SS, Lewis MC, Lovaglio J, Hanley PW, Shaia C, Hawman DW, Munster VJ, Jarvis MA, Richt JA, and Feldmann H
- Abstract
As the COVID-19 pandemic moves into its third year, there remains a need for additional animal models better recapitulating severe COVID to study SARS-CoV-2 pathogenesis and develop countermeasures, especially treatment options. Pigs are known intermediate hosts for many viruses with zoonotic potential and are susceptible to infection with alpha, beta and delta genera of coronaviruses. Herein, we infected young (3 weeks of age) pigs with SARS-CoV-2 using a combination of respiratory and parenteral inoculation routes. Pigs did not develop clinical disease, nor macroscopic or microscopic pathologic lesions upon SARS-CoV-2 infection. Despite occasional low levels of SARS-CoV-2 genomic RNA in the respiratory tract, subgenomic RNA and infectious virus were never found, and SARS-CoV-2-specific adaptive immune responses were not detectable over the 13-day study period. We concluded that pigs are not susceptible to productive SARS-CoV-2 infection and do not serve as a SARS-CoV-2 reservoir for zoonotic transmission.
- Published
- 2022
- Full Text
- View/download PDF
41. Increased small particle aerosol transmission of B.1.1.7 compared with SARS-CoV-2 lineage A in vivo.
- Author
-
Port JR, Yinda CK, Avanzato VA, Schulz JE, Holbrook MG, van Doremalen N, Shaia C, Fischer RJ, and Munster VJ
- Subjects
- Aerosols, Animals, COVID-19 virology, Female, Male, Mesocricetus, SARS-CoV-2 pathogenicity, Viral Load, Virus Shedding, COVID-19 transmission, SARS-CoV-2 genetics
- Abstract
The major transmission route for SARS-CoV-2 is airborne. However, previous studies could not elucidate the contribution between large droplets and aerosol transmission of SARS-CoV-2 and its variants. Here, we designed and validated an optimized transmission caging setup, which allows for the assessment of aerosol transmission efficiency at various distances. At a distance of 2 m, only particles of <5 μm traversed between cages. Using this setup, we investigated the relative efficiency of aerosol transmission between the SARS-CoV-2 Alpha variant (B.1.1.7) and lineage A in Syrian hamsters. Aerosol transmission of both variants was confirmed in all sentinels after 24 h of exposure as demonstrated by respiratory virus shedding and seroconversion. Productive transmission also occurred after 1 h of exposure, highlighting the efficiency of this transmission route. Interestingly, after donors were infected with a mix of both variants, the Alpha variant outcompeted the lineage A variant in an airborne transmission chain. Overall, these data indicate that a lower infectious dose of the Alpha variant, compared to lineage A, could be sufficient for successful transmission. This highlights the continuous need to assess emerging variants and the development for pre-emptive transmission mitigation strategies., (© 2022. This is a U.S. government work and not under copyright protection in the U.S.; foreign copyright protection may apply.)
- Published
- 2022
- Full Text
- View/download PDF
42. Age-related differences in immune dynamics during SARS-CoV-2 infection in rhesus macaques.
- Author
-
Speranza E, Purushotham JN, Port JR, Schwarz B, Flagg M, Williamson BN, Feldmann F, Singh M, Pérez-Pérez L, Sturdevant GL, Roberts LM, Carmody A, Schulz JE, van Doremalen N, Okumura A, Lovaglio J, Hanley PW, Shaia C, Germain RN, Best SM, Munster VJ, Bosio CM, and de Wit E
- Subjects
- Acute Disease, Animals, Antibody Formation immunology, Bronchoalveolar Lavage Fluid, COVID-19 complications, COVID-19 genetics, Cytokines blood, Gene Expression Regulation, Gene Regulatory Networks, Genomics, Immunity, Cellular genetics, Immunomodulation, Inflammation complications, Inflammation pathology, Lung immunology, Lung pathology, Lung virology, Lymphoid Tissue pathology, Macaca mulatta immunology, Macaca mulatta virology, Models, Biological, Single-Cell Analysis, T-Lymphocytes immunology, Transcription, Genetic, Aging immunology, COVID-19 immunology, COVID-19 veterinary, SARS-CoV-2 immunology
- Abstract
Advanced age is a key predictor of severe COVID-19. To gain insight into this relationship, we used the rhesus macaque model of SARS-CoV-2 infection. Eight older and eight younger macaques were inoculated with SARS-CoV-2. Animals were evaluated using viral RNA quantification, clinical observations, thoracic radiographs, single-cell transcriptomics, multiparameter flow cytometry, multiplex immunohistochemistry, cytokine detection, and lipidomics analysis at predefined time points in various tissues. Differences in clinical signs, pulmonary infiltrates, and virus replication were limited. Transcriptional signatures of inflammation-associated genes in bronchoalveolar lavage fluid at 3 dpi revealed efficient mounting of innate immune defenses in both cohorts. However, age-specific divergence of immune responses emerged during the post-acute phase. Older animals exhibited sustained local inflammatory innate responses, whereas local effector T-cell responses were induced earlier in the younger animals. Circulating lipid mediator and cytokine levels highlighted increased repair-associated signals in the younger animals, and persistent pro-inflammatory responses in the older animals. In summary, despite similar disease outcomes, multi-omics profiling suggests that age may delay or impair antiviral cellular immune responses and delay efficient return to immune homeostasis., (© 2022 Speranza et al.)
- Published
- 2022
- Full Text
- View/download PDF
43. High-Fat High-Sugar Diet-Induced Changes in the Lipid Metabolism Are Associated with Mildly Increased COVID-19 Severity and Delayed Recovery in the Syrian Hamster.
- Author
-
Port JR, Adney DR, Schwarz B, Schulz JE, Sturdevant DE, Smith BJ, Avanzato VA, Holbrook MG, Purushotham JN, Stromberg KA, Leighton I, Bosio CM, Shaia C, and Munster VJ
- Subjects
- Animals, Cricetinae, Cytokines blood, Disease Models, Animal, Edema, Fibrin, Hemorrhage, Humans, Interleukin-10, Interleukin-6, Lipidomics, Lipids blood, Liver pathology, Lung pathology, Male, Mesocricetus, Obesity, SARS-CoV-2, Sugars, Vasculitis pathology, Virus Shedding, COVID-19 pathology, Diet, High-Fat adverse effects, Dietary Carbohydrates adverse effects, Lipid Metabolism, Severity of Illness Index
- Abstract
Pre-existing comorbidities such as obesity or metabolic diseases can adversely affect the clinical outcome of COVID-19. Chronic metabolic disorders are globally on the rise and often a consequence of an unhealthy diet, referred to as a Western Diet. For the first time in the Syrian hamster model, we demonstrate the detrimental impact of a continuous high-fat high-sugar diet on COVID-19 outcome. We observed increased weight loss and lung pathology, such as exudate, vasculitis, hemorrhage, fibrin, and edema, delayed viral clearance and functional lung recovery, and prolonged viral shedding. This was accompanied by an altered, but not significantly different, systemic IL-10 and IL-6 profile, as well as a dysregulated serum lipid response dominated by polyunsaturated fatty acid-containing phosphatidylethanolamine, partially recapitulating cytokine and lipid responses associated with severe human COVID-19. Our data support the hamster model for testing restrictive or targeted diets and immunomodulatory therapies to mediate the adverse effects of metabolic disease on COVID-19.
- Published
- 2021
- Full Text
- View/download PDF
44. SARS-CoV2 variant-specific replicating RNA vaccines protect from disease and pathology and reduce viral shedding following challenge with heterologous SARS-CoV2 variants of concern.
- Author
-
Hawman DW, Meade-White K, Archer J, Leventhal S, Wilson D, Shaia C, Randall S, Khandhar AP, Hsiang TY, Gale M Jr, Berglund P, Fuller DH, Feldmann H, and Erasmus JH
- Abstract
Despite mass public health efforts, the SARS-CoV2 pandemic continues as of late-2021 with resurgent case numbers in many parts of the world. The emergence of SARS-CoV2 variants of concern (VoC) and evidence that existing vaccines that were designed to protect from the original strains of SARS-CoV-2 may have reduced potency for protection from infection against these VoC is driving continued development of second generation vaccines that can protect against multiple VoC. In this report, we evaluated an alphavirus-based replicating RNA vaccine expressing Spike proteins from the original SARS-CoV-2 Alpha strain and recent VoCs delivered in vivo via a lipid inorganic nanoparticle. Vaccination of both mice and Syrian Golden hamsters showed that vaccination induced potent neutralizing titers against each homologous VoC but reduced neutralization against heterologous challenges. Vaccinated hamsters challenged with homologous SARS-CoV2 variants exhibited complete protection from infection. In addition, vaccinated hamsters challenged with heterologous SARS-CoV-2 variants exhibited significantly reduced shedding of infectious virus. Our data demonstrate that this vaccine platform elicits significant protective immunity against SARS-CoV2 variants and supports continued development of this platform.
- Published
- 2021
- Full Text
- View/download PDF
45. UK B.1.1.7 (Alpha) variant exhibits increased respiratory replication and shedding in nonhuman primates.
- Author
-
Rosenke K, Feldmann F, Okumura A, Hansen F, Tang-Huau TL, Meade-White K, Kaza B, Callison J, Lewis MC, Smith BJ, Hanley PW, Lovaglio J, Jarvis MA, Shaia C, and Feldmann H
- Subjects
- Administration, Intranasal, Animals, COVID-19 epidemiology, Gastrointestinal Tract virology, Host Specificity, Polymorphism, Single Nucleotide, RNA, Viral isolation & purification, Random Allocation, Rectum virology, United Kingdom epidemiology, Vero Cells, Viral Load, COVID-19 virology, Chlorocebus aethiops virology, Respiratory System virology, Virus Replication, Virus Shedding
- Abstract
The continuing emergence of SARS-CoV-2 variants calls for regular assessment to identify differences in viral replication, shedding and associated disease. In this study, we compared African green monkeys infected intranasally with either the UK B.1.1.7 (Alpha) variant or its contemporary D614G progenitor. Both variants caused mild respiratory disease with no significant differences in clinical presentation. Significantly higher levels of viral RNA and infectious virus were found in upper and lower respiratory tract samples and tissues from B.1.1.7 infected animals. Interestingly, D614G infected animals showed significantly higher levels of viral RNA and infectious virus in rectal swabs and gastrointestinal tissues. Our results indicate that B.1.1.7 infection in African green monkeys is associated with increased respiratory replication and shedding but no disease enhancement similar to human B.1.1.7 cases.
- Published
- 2021
- Full Text
- View/download PDF
46. Alkhurma haemorrhagic fever virus causes lethal disease in IFNAR -/- mice.
- Author
-
Bhatia B, Haddock E, Shaia C, Rosenke R, Meade-White K, Griffin AJ, Marzi A, and Feldmann H
- Subjects
- Animals, Chlorocebus aethiops, Disease Models, Animal, Encephalitis, Tick-Borne genetics, Encephalitis, Tick-Borne pathology, Encephalitis, Tick-Borne virology, Female, Gene Knockout Techniques, Humans, Male, Mice, Vero Cells, Viral Load, Encephalitis Viruses, Tick-Borne pathogenicity, Encephalitis, Tick-Borne mortality, Receptor, Interferon alpha-beta genetics
- Abstract
ABSTRACT Alkhurma haemorrhagic fever virus (AHFV), a tick-borne flavivirus closely related to Kyasanur Forest disease virus, is the causative agent of a severe, sometimes fatal haemorrhagic/encephalitic disease in humans. To date, there are no specific treatments or vaccines available to combat AHFV infections. A challenge for the development of countermeasures is the absence of a reliable AHFV animal disease model for efficacy testing. Here, we used mice lacking the type I interferon (IFN) receptor (IFNAR
-/- ). AHFV strains Zaki-2 and 2003 both caused uniform lethality in these mice after intraperitoneal injection, but strain 2003 seemed more virulent with a median lethal dose of 0.4 median tissue culture infectious doses (TCID50 ). Disease manifestation in this animal model was similar to case reports of severe human AHFV infections with early generalized signs leading to haemorrhagic and neurologic complications. AHFV infection resulted in early high viremia followed by high viral loads (<108 TCID50 /g tissue) in all analyzed organs. Despite systemic viral replication, virus-induced pathology was mainly found in the spleen, lymph nodes, liver and heart. This uniformly lethal AHFV disease model will be instrumental for pathogenesis studies and countermeasure development against this neglected zoonotic pathogen.- Published
- 2021
- Full Text
- View/download PDF
47. Prior aerosol infection with lineage A SARS-CoV-2 variant protects hamsters from disease, but not reinfection with B.1.351 SARS-CoV-2 variant.
- Author
-
Yinda CK, Port JR, Bushmaker T, Fischer RJ, Schulz JE, Holbrook MG, Shaia C, de Wit E, van Doremalen N, and Munster VJ
- Subjects
- Animals, Antibodies, Viral blood, COVID-19 transmission, COVID-19 virology, Chlorocebus aethiops, Cricetinae, Disease Models, Animal, Female, High-Throughput Nucleotide Sequencing, Humans, SARS-CoV-2 genetics, SARS-CoV-2 immunology, Seroconversion, Severity of Illness Index, Vero Cells, Viral Load, Virus Replication, Broadly Neutralizing Antibodies blood, COVID-19 immunology, Fomites virology, SARS-CoV-2 pathogenicity, Sequence Analysis, RNA methods
- Abstract
The circulation of SARS-CoV-2 has resulted in the emergence of variants of concern (VOCs). It is currently unclear whether the previous infection with SARS-CoV-2 provides protection against reinfection with VOCs. Here, we show that low dose aerosol exposure to hCoV-19/human/USA/WA-CDC-WA1/2020 (WA1, lineage A), resulted in a productive mild infection. In contrast, a low dose of SARS-CoV-2 via fomites did not result in productive infection in the majority of exposed hamsters and these animals remained non-seroconverted. After recovery, hamsters were re-exposed to hCoV-19/South African/KRISP-K005325/2020 (VOC B.1.351) via an intranasal challenge. Seroconverted rechallenged animals did not lose weight and shed virus for three days. They had a little infectious virus and no pathology in the lungs. In contrast, shedding, weight loss and extensive pulmonary pathology caused by B.1.351 replication were observed in the non-seroconverted animals. The rechallenged seroconverted animals did not transmit the virus to naïve sentinels via direct contact transmission, in contrast to the non-seroconverted animals. Reinfection with B.1.351 triggered an anamnestic response that boosted not only neutralizing titres against lineage A, but also titres against B.1.351. Our results confirm that aerosol exposure is a more efficient infection route than fomite exposure. Furthermore, initial infection with SARS-CoV-2 lineage A does not prevent heterologous reinfection with B.1.351 but prevents disease and onward transmission. These data suggest that previous SARS-CoV-2 exposure induces partial protective immunity. The reinfection generated a broadly neutralizing humoral response capable of effectively neutralizing B.1.351 while maintaining its ability to neutralize the virus to which the initial response was directed against.
- Published
- 2021
- Full Text
- View/download PDF
48. Recovery from Acute SARS-CoV-2 Infection and Development of Anamnestic Immune Responses in T Cell-Depleted Rhesus Macaques.
- Author
-
Hasenkrug KJ, Feldmann F, Myers L, Santiago ML, Guo K, Barrett BS, Mickens KL, Carmody A, Okumura A, Rao D, Collins MM, Messer RJ, Lovaglio J, Shaia C, Rosenke R, van Doremalen N, Clancy C, Saturday G, Hanley P, Smith BJ, Meade-White K, Shupert WL, Hawman DW, and Feldmann H
- Subjects
- Animals, CD4-Positive T-Lymphocytes cytology, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes cytology, CD8-Positive T-Lymphocytes immunology, COVID-19 immunology, Female, Lymphocyte Depletion methods, Macaca mulatta immunology, Male, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, COVID-19 pathology, Immunologic Memory immunology, Lymphopenia immunology, SARS-CoV-2 immunology
- Abstract
Severe coronavirus disease 2019 (COVID-19) has been associated with T cell lymphopenia, but no causal effect of T cell deficiency on disease severity has been established. To investigate the specific role of T cells in recovery from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections, we studied rhesus macaques that were depleted of either CD4
+ , CD8+ , or both T cell subsets prior to infection. Peak virus loads were similar in all groups, but the resolution of virus in the T cell-depleted animals was slightly delayed compared to that in controls. The T cell-depleted groups developed virus-neutralizing antibody responses and class switched to IgG. When reinfected 6 weeks later, the T cell-depleted animals showed anamnestic immune responses characterized by rapid induction of high-titer virus-neutralizing antibodies, faster control of virus loads, and reduced clinical signs. These results indicate that while T cells play a role in the recovery of rhesus macaques from acute SARS-CoV-2 infections, their depletion does not induce severe disease, and T cells do not account for the natural resistance of rhesus macaques to severe COVID-19. Neither primed CD4+ nor CD8+ T cells appeared critical for immunoglobulin class switching, the development of immunological memory, or protection from a second infection. IMPORTANCE Patients with severe COVID-19 often have decreased numbers of T cells, a cell type important in fighting most viral infections. However, it is not known whether the loss of T cells contributes to severe COVID-19 or is a consequence of it. We studied rhesus macaques, which develop only mild COVID-19, similar to most humans. Experimental depletion of T cells slightly prolonged their clearance of virus, but there was no increase in disease severity. Furthermore, they were able to develop protection from a second infection and produced antibodies capable of neutralizing the virus. They also developed immunological memory, which allows a much stronger and more rapid response upon a second infection. These results suggest that T cells are not critical for recovery from acute SARS-CoV-2 infections in this model and point toward B cell responses and antibodies as the essential mediators of protection from re-exposure.- Published
- 2021
- Full Text
- View/download PDF
49. Increased aerosol transmission for B.1.1.7 (alpha variant) over lineage A variant of SARS-CoV-2.
- Author
-
Port J, Yinda CK, Avanzato V, Schulz J, Holbrook M, van Doremalen N, Shaia C, Fischer R, and Munster V
- Abstract
Airborne transmission, a term combining both large droplet and aerosol transmission, is thought to be the main transmission route of SARS-CoV-2. Here we investigated the relative efficiency of aerosol transmission of two variants of SARS-CoV-2, B.1.1.7 (alpha) and lineage A, in the Syrian hamster. A novel transmission caging setup was designed and validated, which allowed the assessment of transmission efficiency at various distances. At 2 meters distance, only particles <5 µm traversed between cages. In this setup, aerosol transmission was confirmed in 8 out of 8 (N = 4 for each variant) sentinels after 24 hours of exposure as demonstrated by respiratory shedding and seroconversion. Successful transmission occurred even when exposure time was limited to one hour, highlighting the efficiency of this transmission route. Interestingly, the B.1.1.7 variant outcompeted the lineage A variant in an airborne transmission chain after mixed infection of donors. Combined, this data indicates that the infectious dose of B.1.1.7 required for successful transmission may be lower than that of lineage A virus. The experimental proof for true aerosol transmission and the increase in the aerosol transmission potential of B.1.1.7 underscore the continuous need for assessment of novel variants and the development or preemptive transmission mitigation strategies.
- Published
- 2021
- Full Text
- View/download PDF
50. Increased aerosol transmission for B.1.1.7 (alpha variant) over lineage A variant of SARS-CoV-2.
- Author
-
Port JR, Yinda CK, Avanzato VA, Schulz JE, Holbrook MG, van Doremalen N, Shaia C, Fischer RJ, and Munster VJ
- Abstract
Airborne transmission, a term combining both large droplet and aerosol transmission, is thought to be the main transmission route of SARS-CoV-2. Here we investigated the relative efficiency of aerosol transmission of two variants of SARS-CoV-2, B.1.1.7 (alpha) and lineage A, in the Syrian hamster. A novel transmission caging setup was designed and validated, which allowed the assessment of transmission efficiency at various distances. At 2 meters distance, only particles <5 µm traversed between cages. In this setup, aerosol transmission was confirmed in 8 out of 8 (N = 4 for each variant) sentinels after 24 hours of exposure as demonstrated by respiratory shedding and seroconversion. Successful transmission occurred even when exposure time was limited to one hour, highlighting the efficiency of this transmission route. Interestingly, the B.1.1.7 variant outcompeted the lineage A variant in an airborne transmission chain after mixed infection of donors. Combined, this data indicates that the infectious dose of B.1.1.7 required for successful transmission may be lower than that of lineage A virus. The experimental proof for true aerosol transmission and the increase in the aerosol transmission potential of B.1.1.7 underscore the continuous need for assessment of novel variants and the development or preemptive transmission mitigation strategies.
- 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.