1
|
Schäfer A, Leist SR, Powers JM, Baric RS. Animal models of Long Covid: A hit-and-run disease. Sci Transl Med 2024; 16:eado2104. [PMID: 39536118 DOI: 10.1126/scitranslmed.ado2104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV 2) pandemic has caused more than 7 million deaths globally. Despite the presence of infection- and vaccine-induced immunity, SARS-CoV-2 infections remain a major global health concern because of the emergence of SARS-CoV-2 variants that can cause severe acute coronavirus disease 2019 (COVID-19) or enhance Long Covid disease phenotypes. About 5 to 10% of SARS-CoV-2-infected individuals develop Long Covid, which, similar to acute COVID 19, often affects the lung. However, Long Covid can also affect other peripheral organs, especially the brain. The causal relationships between acute disease phenotypes, long-term symptoms, and involvement of multiple organ systems remain elusive, and animal model systems mimicking both acute and post-acute phases are imperative. Here, we review the current state of Long Covid animal models, including current and possible future applications.
Collapse
Affiliation(s)
- Alexandra Schäfer
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sarah R Leist
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - John M Powers
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
2
|
Rowe T, Fletcher A, Svoboda P, Pohl J, Hatta Y, Jasso G, Wentworth DE, Ross TM. Interferon as an immunoadjuvant to enhance antibodies following influenza B infection and vaccination in ferrets. NPJ Vaccines 2024; 9:199. [PMID: 39448628 PMCID: PMC11502657 DOI: 10.1038/s41541-024-00973-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 09/19/2024] [Indexed: 10/26/2024] Open
Abstract
Despite annual vaccination, influenza B viruses (IBV) continue to cause significant morbidity and mortality in humans. We have found that IBV infection resulted in a weaker innate and adaptive immune response than influenza A viruses (IAV) in ferrets. To understand and overcome the weak immune responses to IBV in ferrets, we administered type-I or type-III interferon (IFN) to ferrets following infection or vaccination and evaluated their effects on the immune response. IFN signaling following viral infection plays an important role in the initial innate immune response and affects subsequent adaptive immune responses. In the respiratory tract, IFN lambda (IFNL) has regulatory effects on adaptive immunity indirectly through thymic stromal lymphopoietin (TSLP), which then acts on immune cells to stimulate the adaptive response. Following IBV infection or vaccination, IFN treatment (IFN-Tx) upregulated gene expression of early inflammatory responses in the upper respiratory tract and robust IFN, TSLP, and inflammatory responses in peripheral blood cells. These responses were sustained following challenge or vaccination in IFN-Tx animals. Serum IFNL and TSLP levels were enhanced in IFN-Tx animals following challenge/rechallenge over mock-Tx; however, this difference was not observed following vaccination. Antibody responses in serum of IFN-Tx animals following IBV infection or vaccination increased more quickly and to higher titers and were sustained longer than mock-Tx animals over 3 months. Following rechallenge of infected animals 3 months post treatment, antibody levels remained higher than mock-Tx. However, IFN-Tx did not have an effect on antibody responses following challenge of vaccinated animals. A strong direct correlation was found between TSLP levels and antibody responses following challenge-rechallenge and vaccination-challenge indicating it as a useful tool for predicting adaptive immune responses following IBV infection or vaccination. The effects of IFN on strengthening both innate and adaptive responses to IBV may aid in development of more effective treatments following infection and improved influenza vaccines.
Collapse
Affiliation(s)
- Thomas Rowe
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA, USA.
- Department of Infectious Diseases, University of Georgia, Athens, GA, USA.
| | | | - Pavel Svoboda
- Division of Core Laboratory Services and Response, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Jan Pohl
- Division of Core Laboratory Services and Response, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Yasuko Hatta
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Gabriela Jasso
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - David E Wentworth
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Ted M Ross
- Department of Infectious Diseases, University of Georgia, Athens, GA, USA
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL, USA
- Department of Infection Biology, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
3
|
Gildemann K, Tsernant ML, Liivand L, Ennomäe R, Poikalainen V, Lepasalu L, Rom S, Kavak A, Cox RM, Wolf JD, Lieber CM, Plemper RK, Männik A, Ustav M, Ustav M, Gerhold JM. Anti-SARS-CoV-2 antibodies in a nasal spray efficiently block viral transmission between ferrets. iScience 2024; 27:110326. [PMID: 39045097 PMCID: PMC11263742 DOI: 10.1016/j.isci.2024.110326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/17/2024] [Accepted: 06/18/2024] [Indexed: 07/25/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to spread in the population. We recently reported the production of bovine colostrum-derived antibodies that can neutralize the virus. These have been formulated into a nasal spray. The immunoglobulin preparation is capable of blocking interaction of the trimeric spike protein (Tri S) of SARS-CoV-2 with the cellular receptor angiotensin-converting enzyme 2 (ACE2), entry of a pseudovirus carrying the Tri S into ACE2 over-expressing human embryonic kidney (HEK) cells, and entry of the virus into live Vero E6 cells. Using an ELISA assay, we demonstrate here that this holds true for different SARS-CoV-2 variants of concern. Using the ferret transmission model, we show that the nasal spray formulation of anti-SARS-CoV-2 immunoglobulins efficiently blocks transmission of SARS-CoV-2 from infected to uninfected ferrets. The results indicate that the use of the nasal spray in humans can add an effective additional layer of protection against the virus, and might be applicable for other viruses of the upper respiratory tract.
Collapse
Affiliation(s)
- Kiira Gildemann
- Icosagen Cell Factory OÜ, Õssu, Kambja vald, 61713 Tartumaa, Estonia
| | | | - Laura Liivand
- Icosagen Cell Factory OÜ, Õssu, Kambja vald, 61713 Tartumaa, Estonia
| | - Retti Ennomäe
- Icosagen Cell Factory OÜ, Õssu, Kambja vald, 61713 Tartumaa, Estonia
| | | | | | - Siimu Rom
- Chemi-Pharm AS, Tänassilma, 76404 Harjumaa, Estonia
| | - Ants Kavak
- Department of Clinical Veterinary Medicine, Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, 51014 Tartu, Estonia
| | - Robert Marsden Cox
- Center for Translational Antiviral Research, Georgia State University Institute for Biomedical Sciences, Atlanta, GA 30303, USA
| | - Josef Dieter Wolf
- Center for Translational Antiviral Research, Georgia State University Institute for Biomedical Sciences, Atlanta, GA 30303, USA
| | - Carolin Maria Lieber
- Center for Translational Antiviral Research, Georgia State University Institute for Biomedical Sciences, Atlanta, GA 30303, USA
| | - Richard Karl Plemper
- Center for Translational Antiviral Research, Georgia State University Institute for Biomedical Sciences, Atlanta, GA 30303, USA
| | - Andres Männik
- Icosagen Cell Factory OÜ, Õssu, Kambja vald, 61713 Tartumaa, Estonia
| | - Mart Ustav
- Icosagen Cell Factory OÜ, Õssu, Kambja vald, 61713 Tartumaa, Estonia
| | - Mart Ustav
- Icosagen Cell Factory OÜ, Õssu, Kambja vald, 61713 Tartumaa, Estonia
| | | |
Collapse
|
4
|
Hickey AJ, Maloney SE, Kuehl PJ, Phillips JE, Wolff RK. Practical Considerations in Dose Extrapolation from Animals to Humans. J Aerosol Med Pulm Drug Deliv 2024; 37:77-89. [PMID: 38237032 DOI: 10.1089/jamp.2023.0041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2024] Open
Abstract
Animal studies are an important component of drug product development and the regulatory review process since modern practices have been in place, for almost a century. A variety of experimental systems are available to generate aerosols for delivery to animals in both liquid and solid forms. The extrapolation of deposited dose in the lungs from laboratory animals to humans is challenging because of genetic, anatomical, physiological, pharmacological, and other biological differences between species. Inhaled drug delivery extrapolation requires scrutiny as the aerodynamic behavior, and its role in lung deposition is influenced not only by the properties of the drug aerosol but also by the anatomy and pulmonary function of the species in which it is being evaluated. Sources of variability between species include the formulation, delivery system, and species-specific biological factors. It is important to acknowledge the underlying variables that contribute to estimates of dose scaling between species.
Collapse
Affiliation(s)
- Anthony J Hickey
- Department of Technology Advancement and Commercialization, RTI International, Research Triangle Park, North Carolina, USA
| | - Sara E Maloney
- Department of Technology Advancement and Commercialization, RTI International, Research Triangle Park, North Carolina, USA
| | - Phillip J Kuehl
- Division: Scientific Core Laboratories; Lovelace Respiratory Research Institute, Albuquerque, New Mexico, USA
| | - Jonathan E Phillips
- Amgen, Inc., Inflammation Discovery Research, Thousand Oaks, California, USA
| | | |
Collapse
|
5
|
Pramod RK, Atul PK, Pandey M, Anbazhagan S, Mhaske ST, Barathidasan R. Care, management, and use of ferrets in biomedical research. Lab Anim Res 2024; 40:10. [PMID: 38532510 DOI: 10.1186/s42826-024-00197-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 03/02/2024] [Accepted: 03/14/2024] [Indexed: 03/28/2024] Open
Abstract
The ferret (Mustela putorius furo) is a small domesticated species of the family Mustelidae within the order Carnivora. The present article reviews and discusses the current state of knowledge about housing, care, breeding, and biomedical uses of ferrets. The management and breeding procedures of ferrets resemble those used for other carnivores. Understanding its behavior helps in the use of environmental enrichment and social housing, which promote behaviors typical of the species. Ferrets have been used in research since the beginning of the twentieth century. It is a suitable non-rodent model in biomedical research because of its hardy nature, social behavior, diet and other habits, small size, and thus the requirement of a relatively low amount of test compounds and early sexual maturity compared with dogs and non-human primates. Ferrets and humans have numerous similar anatomical, metabolic, and physiological characteristics, including the endocrine, respiratory, auditory, gastrointestinal, and immunological systems. It is one of the emerging animal models used in studies such as influenza and other infectious respiratory diseases, cystic fibrosis, lung cancer, cardiac research, gastrointestinal disorders, neuroscience, and toxicological studies. Ferrets are vulnerable to many human pathogenic organisms, like severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), because air transmission of this virus between them has been observed in the laboratory. Ferrets draw the attention of the medical community compared to rodents because they occupy a distinct niche in biomedical studies, although they possess a small representation in laboratory research.
Collapse
Affiliation(s)
- Ravindran Kumar Pramod
- ICMR-National Animal Resource Facility for Biomedical Research, Genome Valley, Hyderabad, Telangana, 500101, India.
| | - Pravin Kumar Atul
- ICMR-National Animal Resource Facility for Biomedical Research, Genome Valley, Hyderabad, Telangana, 500101, India
| | - Mamta Pandey
- ICMR-National Animal Resource Facility for Biomedical Research, Genome Valley, Hyderabad, Telangana, 500101, India
| | - S Anbazhagan
- ICMR-National Animal Resource Facility for Biomedical Research, Genome Valley, Hyderabad, Telangana, 500101, India
| | - Suhas T Mhaske
- ICMR-National Animal Resource Facility for Biomedical Research, Genome Valley, Hyderabad, Telangana, 500101, India
| | - R Barathidasan
- ICMR-National Animal Resource Facility for Biomedical Research, Genome Valley, Hyderabad, Telangana, 500101, India
| |
Collapse
|
6
|
Belser JA, Kieran TJ, Mitchell ZA, Sun X, Mayfield K, Tumpey TM, Spengler JR, Maines TR. Key considerations to improve the normalization, interpretation and reproducibility of morbidity data in mammalian models of viral disease. Dis Model Mech 2024; 17:dmm050511. [PMID: 38440823 PMCID: PMC10941659 DOI: 10.1242/dmm.050511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 02/05/2024] [Indexed: 03/06/2024] Open
Abstract
Viral pathogenesis and therapeutic screening studies that utilize small mammalian models rely on the accurate quantification and interpretation of morbidity measurements, such as weight and body temperature, which can vary depending on the model, agent and/or experimental design used. As a result, morbidity-related data are frequently normalized within and across screening studies to aid with their interpretation. However, such data normalization can be performed in a variety of ways, leading to differences in conclusions drawn and making comparisons between studies challenging. Here, we discuss variability in the normalization, interpretation, and presentation of morbidity measurements for four model species frequently used to study a diverse range of human viral pathogens - mice, hamsters, guinea pigs and ferrets. We also analyze findings aggregated from influenza A virus-infected ferrets to contextualize this discussion. We focus on serially collected weight and temperature data to illustrate how the conclusions drawn from this information can vary depending on how raw data are collected, normalized and measured. Taken together, this work supports continued efforts in understanding how normalization affects the interpretation of morbidity data and highlights best practices to improve the interpretation and utility of these findings for extrapolation to public health contexts.
Collapse
Affiliation(s)
- Jessica A. Belser
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Troy J. Kieran
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Zoë A. Mitchell
- Franklin College of Arts and Sciences, University of Georgia, Athens, GA 30602, USA
- Division of Scientific Resources, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Xiangjie Sun
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Kristin Mayfield
- Division of Scientific Resources, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Terrence M. Tumpey
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Jessica R. Spengler
- Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Taronna R. Maines
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| |
Collapse
|
7
|
Kieran TJ, Sun X, Maines TR, Beauchemin CAA, Belser JA. Exploring associations between viral titer measurements and disease outcomes in ferrets inoculated with 125 contemporary influenza A viruses. J Virol 2024; 98:e0166123. [PMID: 38240592 PMCID: PMC10878272 DOI: 10.1128/jvi.01661-23] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 12/16/2023] [Indexed: 02/21/2024] Open
Abstract
As use of the ferret model to study influenza A virus (IAV) pathogenicity increases, periodic assessment of data generated in this model is warranted, to identify features associated with virus replication throughout the respiratory tract and to refine future analyses. However, protocol-specific differences present between independent laboratories limit easy aggregation of virological data. We compiled viral titer and clinical data from >1,000 ferrets inoculated with 125 contemporary IAV under a consistent experimental protocol (including high- and low-pathogenicity avian, swine-origin, and human viruses, spanning H1, H2, H3, H5, H7, and H9 subtypes) and examined which meaningful and statistically supported associations were present among numerous quantitative measurements. Viral titers correlated positively between ferret nasal turbinate tissue, lung tissue, and nasal wash specimens, though the strength of the associations varied, notably regarding the particular nasal wash summary measure employed and properties of the virus itself. Use of correlation coefficients and mediation analyses further supported the interconnectedness of viral titer measurements taken at different sites throughout the respiratory tract. IAV possessing mammalian host adaptation markers in the HA and PB2 exhibited more rapid growth in the ferret upper respiratory tract early after infection, supported by quantities derived from infectious titer data to capture infection progression, compared with viruses bearing hallmarks of avian IAV. Collectively, this work identifies summary metrics most closely linked with virological and phenotypic outcomes in ferrets, supporting continued refinement of data analyzed from in vivo experimentation, notably from studies conducted to evaluate the public health risk posed by novel and emerging IAV.IMPORTANCEFerrets are frequently employed to study the pandemic potential of novel and emerging influenza A viruses. However, systematic retrospective analyses of data generated from these experiments are rarely performed, limiting our ability to identify trends in this data and explore how analyses can be refined. Using logarithmic viral titer and clinical data aggregated from one research group over 20 years, we assessed which meaningful and statistically supported associations were present among numerous quantitative measurements obtained from influenza A virus (IAV)-infected ferrets, including those capturing viral titers, infection progression, and disease severity. We identified numerous linear correlations between parameters assessing virus replication at discrete sites in vivo, including parameters capturing infection progression not frequently employed in the field, and sought to investigate the interconnected nature of these associations. This work supports continued refinement of data analyzed from in vivo experimentation, notably from studies which evaluate the public health risk posed by IAV.
Collapse
Affiliation(s)
- Troy J. Kieran
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Xiangjie Sun
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Taronna R. Maines
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Catherine A. A. Beauchemin
- Department of Physics, Toronto Metropolitan University, Toronto, Canada
- Interdisciplinary Theoretical and Mathematical Sciences (iTHEMS) at RIKEN, Wako, Japan
| | - Jessica A. Belser
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|
8
|
Dong C, Shang L. Can animal models help to explore functions of nanoparticles in health and diseases? Nanomedicine (Lond) 2024; 19:181-183. [PMID: 38059455 DOI: 10.2217/nnm-2023-0323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2023] Open
Affiliation(s)
- Caijuan Dong
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Lijun Shang
- School of Human Sciences, London Metropolitan University, London, N7 8DB, UK
| |
Collapse
|
9
|
Freeman TL, McElroy AK. Laboratory Animal Models for Rift Valley Fever Virus Disease. Methods Mol Biol 2024; 2824:425-445. [PMID: 39039428 DOI: 10.1007/978-1-0716-3926-9_27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Rift Valley fever virus (RVFV) is an arboviral pathogen of clinical and agricultural relevance. The ongoing development of targeted RVFV prophylactics and therapeutics is overwhelmingly dependent on animal models due to both natural, that is, sporadic outbreaks, and structural, for example, underresourcing of endemic regions, limitations in accessing human patient samples and cohorts. Elucidating mechanisms of viral pathogenesis and testing therapeutics is further complicated by the diverse manifestations of RVFV disease and the heterogeneity of the host response to infection. In this chapter, we describe major clinical manifestations of RVFV infection and discuss the laboratory animal models used to study each.
Collapse
Affiliation(s)
- Tracey L Freeman
- University of Pittsburgh School of Medicine, Department of Pediatrics, Division of Pediatric Infectious Disease, Pittsburgh, PA, USA
- University of Pittsburgh Center for Vaccine Research, Pittsburgh, PA, USA
| | - Anita K McElroy
- University of Pittsburgh School of Medicine, Department of Pediatrics, Division of Pediatric Infectious Disease, Pittsburgh, PA, USA.
- University of Pittsburgh Center for Vaccine Research, Pittsburgh, PA, USA.
| |
Collapse
|
10
|
Zumbrun EE, Zak SE, Lee ED, Bowling PA, Ruiz SI, Zeng X, Koehler JW, Delp KL, Bakken RR, Hentschel SS, Bloomfield HA, Ricks KM, Clements TL, Babka AM, Dye JM, Herbert AS. SARS-CoV-2 Aerosol and Intranasal Exposure Models in Ferrets. Viruses 2023; 15:2341. [PMID: 38140582 PMCID: PMC10747480 DOI: 10.3390/v15122341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the worldwide COVID-19 pandemic. Animal models are extremely helpful for testing vaccines and therapeutics and for dissecting the viral and host factors that contribute to disease severity and transmissibility. Here, we report the assessment and comparison of intranasal and small particle (~3 µm) aerosol SARS-CoV-2 exposure in ferrets. The primary endpoints for analysis were clinical signs of disease, recovery of the virus in the upper respiratory tract, and the severity of damage within the respiratory tract. This work demonstrated that ferrets were productively infected with SARS-CoV-2 following either intranasal or small particle aerosol exposure. SARS-CoV-2 infection of ferrets resulted in an asymptomatic disease course following either intranasal or small particle aerosol exposure, with no clinical signs, significant weight loss, or fever. In both aerosol and intranasal ferret models, SARS-CoV-2 replication, viral genomes, and viral antigens were detected within the upper respiratory tract, with little to no viral material detected in the lungs. The ferrets exhibited a specific IgG immune response to the SARS-CoV-2 full spike protein. Mild pathological findings included inflammation, necrosis, and edema within nasal turbinates, which correlated to positive immunohistochemical staining for the SARS-CoV-2 virus. Environmental sampling was performed following intranasal exposure of ferrets, and SARS-CoV-2 genomic material was detected on the feeders and nesting areas from days 2-10 post-exposure. We conclude that both intranasal and small particle aerosol ferret models displayed measurable parameters that could be utilized for future studies, including transmission studies and testing SARS-CoV-2 vaccines and therapeutics.
Collapse
Affiliation(s)
- Elizabeth E. Zumbrun
- Division of Virology, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (S.E.Z.); (R.R.B.); (S.S.H.); (J.M.D.); (A.S.H.)
| | - Samantha E. Zak
- Division of Virology, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (S.E.Z.); (R.R.B.); (S.S.H.); (J.M.D.); (A.S.H.)
| | - Eric D. Lee
- Division of Pathology, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (E.D.L.); (X.Z.); (H.A.B.); (A.M.B.)
| | - Philip A. Bowling
- Division of Veterinary Medicine, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA;
| | - Sara I. Ruiz
- Division of Bacteriology, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA;
| | - Xiankun Zeng
- Division of Pathology, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (E.D.L.); (X.Z.); (H.A.B.); (A.M.B.)
| | - Jeffrey W. Koehler
- Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (J.W.K.); (K.L.D.); (K.M.R.); (T.L.C.)
| | - Korey L. Delp
- Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (J.W.K.); (K.L.D.); (K.M.R.); (T.L.C.)
| | - Russel R. Bakken
- Division of Virology, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (S.E.Z.); (R.R.B.); (S.S.H.); (J.M.D.); (A.S.H.)
| | - Shannon S. Hentschel
- Division of Virology, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (S.E.Z.); (R.R.B.); (S.S.H.); (J.M.D.); (A.S.H.)
| | - Holly A. Bloomfield
- Division of Pathology, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (E.D.L.); (X.Z.); (H.A.B.); (A.M.B.)
| | - Keersten M. Ricks
- Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (J.W.K.); (K.L.D.); (K.M.R.); (T.L.C.)
| | - Tamara L. Clements
- Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (J.W.K.); (K.L.D.); (K.M.R.); (T.L.C.)
| | - April M. Babka
- Division of Pathology, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (E.D.L.); (X.Z.); (H.A.B.); (A.M.B.)
| | - John M. Dye
- Division of Virology, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (S.E.Z.); (R.R.B.); (S.S.H.); (J.M.D.); (A.S.H.)
| | - Andrew S. Herbert
- Division of Virology, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (S.E.Z.); (R.R.B.); (S.S.H.); (J.M.D.); (A.S.H.)
| |
Collapse
|
11
|
Anderson-Mondella CJJ, Maines TR, Tansey CM, Belser JA. Meeting Ferret Enrichment Needs in Infectious Disease Laboratory Settings. JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE : JAALAS 2023; 62:518-524. [PMID: 37857467 PMCID: PMC10772907 DOI: 10.30802/aalas-jaalas-23-000057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/07/2023] [Accepted: 09/12/2023] [Indexed: 10/21/2023]
Abstract
Environmental enrichment is a necessary component of all research vivarium settings. However, appropriate enrichment decisions vary greatly depending on the species involved and the research use of the animals. The increasing use of ferrets in research settings-notably for modeling the pathogenicity and transmissibility of viral pathogens that require containment in ABSL-2 to -4 environments-presents a particular challenge for veterinary and research staff to ensure that enrichment needs for these animals are met consistently. Here, we discuss the species-specific enrichment needs of ferrets, enrichment considerations for ferrets housed in research settings, and the challenges and importance of providing appropriate enrichment during experimentation, including when ferrets are housed in high-containment facilities. This article is organized to support the easy availability of information that will facilitate the design and implementation of optimal environmental enrichment for ferrets used in diverse research efforts in vivarium settings.
Collapse
Affiliation(s)
- Challie JJ Anderson-Mondella
- Comparative Medicine Branch, Division of Scientific Resources, Centers for Disease Control and Prevention, Atlanta, Georgia
- Georgia Gwinnett College, Lawrenceville, Georgia; and
| | - Taronna R Maines
- Immunology and Pathogenesis Branch, Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Cassandra M Tansey
- Comparative Medicine Branch, Division of Scientific Resources, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Jessica A Belser
- Immunology and Pathogenesis Branch, Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia
| |
Collapse
|
12
|
Rouhana S, Jacyniak K, Francis ME, Falzarano D, Kelvin AA, Pyle WG. Sex differences in the cardiac stress response following SARS-CoV-2 infection of ferrets. Am J Physiol Heart Circ Physiol 2023; 325:H1153-H1167. [PMID: 37737732 PMCID: PMC10894670 DOI: 10.1152/ajpheart.00101.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 09/15/2023] [Accepted: 09/15/2023] [Indexed: 09/23/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection damages the heart, increasing the risk of adverse cardiovascular events. Female sex protects against complications of infection; females are less likely to experience severe illness or death, although their risk for postacute sequelae of COVID-19 ("long COVID") is higher than in males. Despite the important role of the heart in COVID-19 outcomes, molecular elements in the heart impacted by SARS-CoV-2 are poorly understood. Similarly, the role sex has on the myocardial effects of SARS-CoV-2 infection has not been investigated at a molecular level. We intranasally inoculated female and male ferrets with SARS-CoV-2 and assessed myocardial stress signals, inflammation, and the innate immune response for 14 days. Myocardial phosphorylated GSK3α/β decreased at day 2 postinfection (pi) in male ferrets, whereas females showed no changes. Myocardial levels of p62/SQSTM1 decreased in male ferrets at days 2, 7, and 14 pi while lower baseline levels in females increased on day 2. Phosphorylated ERK1/2 increased in cardiomyocyte nuclei in females on days 2 and 14 pi, whereas male ferrets had no changes. Only hearts from females increased fibrosis on day 14 pi. Immune and inflammation markers increased in hearts, with some sex differences. These results are the first to identify myocardial stress responses following SARS-CoV-2 infection and reveal sex differences that may contribute to differential outcomes. Future research is required to define the pathways involving these stress signals to fully understand the myocardial effects of COVID-19 and identify targets that mitigate cardiac injury following SARS-CoV-2 infection.NEW & NOTEWORTHY Cardiovascular disease is a leading risk factor for severe COVID-19, and cardiovascular pathologies are among the most common adverse outcomes following SARS-CoV-2 infection. Females and males have different outcomes and adverse cardiovascular events following SARS-CoV-2 infection. This study shows sex differences in stress proteins p62/SQSTM1, ERK1/2, and GSK3α/β, along with innate immunity and inflammation in hearts of ferrets infected with SARS-CoV-2, identifying mechanisms of COVID-19 cardiac injury and cardiac complications of long COVID.
Collapse
Affiliation(s)
- Sarah Rouhana
- IMPART Investigator Team, Dalhousie Medicine, Saint John, New Brunswick, Canada
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Kathy Jacyniak
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Magen E Francis
- Vaccine and Infectious Disease Organization-International Vaccine Centre, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Darryl Falzarano
- Vaccine and Infectious Disease Organization-International Vaccine Centre, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Alyson A Kelvin
- Vaccine and Infectious Disease Organization-International Vaccine Centre, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - W Glen Pyle
- IMPART Investigator Team, Dalhousie Medicine, Saint John, New Brunswick, Canada
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
13
|
Kim EH, Park SJ. Emerging Tick-Borne Dabie bandavirus: Virology, Epidemiology, and Prevention. Microorganisms 2023; 11:2309. [PMID: 37764153 PMCID: PMC10536723 DOI: 10.3390/microorganisms11092309] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/09/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Severe Fever with Thrombocytopenia Syndrome (SFTS), caused by Dabie bandavirus (SFTSV), is an emerging infectious disease first identified in China. Since its discovery, infections have spread throughout East Asian countries primarily through tick bites but also via transmission between animals and humans. The expanding range of ticks, the primary vectors for SFTSV, combined with migration patterns of tick-carrying birds, sets the stage for the global spread of this virus. SFTSV rapidly evolves due to continuous mutation and reassortment; currently, no approved vaccines or antiviral drugs are available. Thus, the threat this virus poses to global health is unmistakable. This review consolidates the most recent research on SFTSV, including its molecular characteristics, transmission pathways through ticks and other animals, as well as the progress in antiviral drug and vaccine development, encompassing animal models and clinical trials.
Collapse
Affiliation(s)
- Eun-Ha Kim
- Center for Study of Emerging and Re-Emerging Viruses, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea;
| | - Su-Jin Park
- Division of Life Science, Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| |
Collapse
|
14
|
Das T, Sikdar S, Chowdhury MHU, Nyma KJ, Adnan M. SARS-CoV-2 prevalence in domestic and wildlife animals: A genomic and docking based structural comprehensive review. Heliyon 2023; 9:e19345. [PMID: 37662720 PMCID: PMC10474441 DOI: 10.1016/j.heliyon.2023.e19345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 08/08/2023] [Accepted: 08/19/2023] [Indexed: 09/05/2023] Open
Abstract
The SARS-CoV-2 virus has been identified as the infectious agent that led to the COVID-19 pandemic, which the world has seen very recently. Researchers have linked the SARS-CoV-2 outbreak to bats for the zoonotic spread of the virus to humans. Coronaviruses have a crown-like shape and positive-sense RNA nucleic acid. It attaches its spike glycoprotein to the host angiotensin-converting enzyme 2 (ACE2) receptor. Coronavirus genome comprises 14 ORFs and 27 proteins, spike glycoprotein being one of the most critical proteins for viral pathogenesis. Many mammals and reptiles, including bats, pangolins, ferrets, snakes, and turtles, serve as the principal reservoirs for this virus. But many experimental investigations have shown that certain domestic animals, including pigs, chickens, dogs, cats, and others, may also be able to harbor this virus, whether they exhibit any symptoms. These animals act as reservoirs for SARS-CoV, facilitating its zoonotic cross-species transmission to other species, including humans. In this review, we performed a phylogenetic analysis with multiple sequence alignment and pairwise evolutionary distance analysis, which revealed the similarity of ACE2 receptors in humans, chimpanzees, domestic rabbits, house mice, and golden hamsters. Pairwise RMSD analysis of the spike protein from some commonly reported SARS-CoV revealed that bat and pangolin coronavirus shared the highest structural similarity with human coronavirus. In a further experiment, molecular docking confirmed a higher affinity of pig, bat, and pangolin coronavirus spike proteins' affinity to the human ACE2 receptor. Such comprehensive structural and genomic analysis can help us to forecast the next likely animal source of these coronaviruses that may infect humans. To combat these zoonotic illnesses, we need a one health strategy that considers the well-being of people and animals and the local ecosystem.
Collapse
Affiliation(s)
- Tuhin Das
- Department of Microbiology, University of Chittagong, Chattogram, 4331, Bangladesh
| | - Suranjana Sikdar
- Department of Microbiology, University of Chittagong, Chattogram, 4331, Bangladesh
| | - Md. Helal Uddin Chowdhury
- Ethnobotany and Pharmacognosy Lab, Department of Botany, University of Chittagong, Chattogram, 4331, Bangladesh
| | | | - Md. Adnan
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, 84112, United States
- Department of Pharmacy, International Islamic University Chittagong, Chattogram, 4318, Bangladesh
| |
Collapse
|
15
|
Jimenez IA, Craney MC, Painter MC, Burch-Strong KE, Plunkard JCM, Villano JS, Hopper LM. Behavioral Evaluation of Laboratory-housed Ferrets ( Mustela Putorius Furo) in Different Enclosure Sizes. JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE : JAALAS 2023; 62:382-394. [PMID: 37673662 PMCID: PMC10597328 DOI: 10.30802/aalas-jaalas-23-000046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 07/19/2023] [Accepted: 07/19/2023] [Indexed: 09/08/2023]
Abstract
The domestic ferret (Mustela putorius furo) is a common research model for infectious disease and behavioral studies. Ferrets are social animals that are commonly pair-housed. The United States has no species-specific regulatory standards for housing ferrets. Optimal enclosure dimensions have also not been investigated in this species, and cage sizes reported in the literature vary. Adequate space is an important animal welfare consideration, as smaller cages have been linked to increased incidence of stress- or boredom-related behaviors in some species. Here, we evaluated activity budget and space utilization in 2 different enclosure sizes for pair-housed female ferrets (n = 12). Single cages measured 78.7×78.7×45.7cm; double cages were comprised of 2 single cages connected by a short tunnel measuring 17.8 cm. Three pairs of ferrets were housed in each cage size and continuous video recordings were captured for 2 wk prior to crossover to the other cage size. The overall activity budget was similar between groups, with the predominant behavior being inactivity (89%). Stereotypic behaviors, such as cage biting or escape attempts, were infrequent (<0.1%) in both groups. Ferrets in double cages remained in the same cage as their partner 96% of the time, suggesting that social support is very valuable. Our results suggest that ferrets in both cage sizes experienced satisfactory welfare conditions. Our findings also suggest that while cage size is not the only determinant of conspecific aggression, larger cages may be an effective intervention to ameliorate aggression in certain ferrets based on signalment or behavioral history, with particular utility as a potential alternative to re-pairing or single-housing. This study provides valuable information to guide animal care and use programs regarding appropriate ferret housing.
Collapse
Affiliation(s)
- Isabel A Jimenez
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Morgan C Craney
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Melissa C Painter
- Research Animal Resources, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kayla E Burch-Strong
- Research Animal Resources, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jessica C M Plunkard
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jason S Villano
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
- Research Animal Resources, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Lydia M Hopper
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
- Research Animal Resources, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
16
|
Smith RE, Choudhary S, Ramirez JA. Ferrets as Models for Viral Respiratory Disease. Comp Med 2023; 73:187-193. [PMID: 37258084 PMCID: PMC10290486 DOI: 10.30802/aalas-cm-22-000064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/26/2022] [Accepted: 01/03/2023] [Indexed: 06/02/2023]
Abstract
Domestic ferrets (Mustela putorius furo) have been used in biomedical research to study influenza viruses since the early 20th century. Ferrets have continued to gain importance for the study of viral respiratory disease due to their disease susceptibility and anatomic similarities to humans. Here we review features of ferret biology and management that should be considered when planning to work with this species, particularly in models of respiratory disease. We specifically discuss biosafety and husbandry, clinical and pathologic assessments, and anesthetic considerations for ferrets with respiratory disease and systemic illness. These considerations are important for animal welfare, fidelity of the model to human disease, and ensuring accuracy and reproducibility of acquired data. Finally, we briefly review the use of ferrets to study respiratory diseases by discussing their respiratory anatomy and 2 frequently studied viral respiratory diseases, influenza and coronavirus disease 2019 (COVID-19).
Collapse
Affiliation(s)
| | | | - Julita A Ramirez
- Pfizer Worldwide Research, Development & Medical, Pearl River, New York 10965, USA;,
| |
Collapse
|
17
|
Clever S, Volz A. Mouse models in COVID-19 research: analyzing the adaptive immune response. Med Microbiol Immunol 2023; 212:165-183. [PMID: 35661253 PMCID: PMC9166226 DOI: 10.1007/s00430-022-00735-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 03/15/2022] [Indexed: 11/29/2022]
Abstract
The emergence of SARS-CoV-2, the severe acute respiratory syndrome coronavirus type 2 causing the COVID-19 pandemic, resulted in a major necessity for scientific countermeasures. Investigations revealing the exact mechanisms of the SARS-CoV-2 pathogenesis provide the basis for the development of therapeutic measures and protective vaccines against COVID-19. Animal models are inevitable for infection and pre-clinical vaccination studies as well as therapeutic testing. A well-suited animal model, mimicking the pathology seen in human COVID-19 patients, is an important basis for these investigations. Several animal models were already used during SARS-CoV-2 studies with different clinical outcomes after SARS-CoV-2 infection. Here, we give an overview of different animal models used in SARS-CoV-2 infection studies with a focus on the mouse model. Mice provide a well-established animal model for laboratory use and several different mouse models have been generated and are being used in SARS-CoV-2 studies. Furthermore, the analysis of SARS-CoV-2-specific T cells during infection and in vaccination studies in mice is highlighted.
Collapse
Affiliation(s)
- Sabrina Clever
- Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Asisa Volz
- Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Hannover, Germany
| |
Collapse
|
18
|
Martins M, do Nascimento GM, Conforti A, Noll JCG, Impellizeri JA, Sanchez E, Wagner B, Lione L, Salvatori E, Pinto E, Compagnone M, Viscount B, Hayward J, Shorrock C, Aurisicchio L, Diel DG. A linear SARS-CoV-2 DNA vaccine candidate reduces virus shedding in ferrets. Arch Virol 2023; 168:124. [PMID: 36988739 PMCID: PMC10052258 DOI: 10.1007/s00705-023-05746-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 03/03/2023] [Indexed: 03/30/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), has caused more than 760 million cases and over 6.8 million deaths as of March 2023. Vaccination has been the main strategy used to contain the spread of the virus and to prevent hospitalizations and deaths. Currently, two mRNA-based vaccines and one adenovirus-vectored vaccine have been approved and are available for use in the U.S. population. The versatility, low cost, and rapid production of DNA vaccines provide important advantages over other platforms. Additionally, DNA vaccines efficiently induce both B- and T-cell responses by expressing the antigen within transfected host cells, and the antigen, after being processed into peptides, can associate with MHC class I or II of antigen-presenting cells (APCs) to stimulate different T cell responses. However, the efficiency of DNA vaccination needs to be improved for use in humans. Importantly, in vivo DNA delivery combined with electroporation (EP) has been used successfully in the field of veterinary oncology, resulting in high rates of response after electrochemotherapy. Here, we evaluate the safety, immunogenicity, and protective efficacy of a novel linear SARS-CoV-2 DNA vaccine candidate delivered by intramuscular injection followed by electroporation (Vet-ePorator™) in ferrets. The linear SARS-CoV-2 DNA vaccine candidate did not cause unexpected side effects. Additionally, the vaccine elicited neutralizing antibodies and T cell responses on day 42 post-immunization using a low dose of the linear DNA construct in a prime-boost regimen. Most importantly, vaccination significantly reduced shedding of infectious SARS-CoV-2 through oral and nasal secretions in a ferret model.
Collapse
Affiliation(s)
- Mathias Martins
- Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Gabriela M do Nascimento
- Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | | | - Jessica C G Noll
- Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | | | | | - Bettina Wagner
- Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | | | | | | | | | - Brian Viscount
- Applied DNA Sciences, Inc., New York, NY, USA
- LineaRx, Inc. , New York, NY, USA
| | - James Hayward
- Applied DNA Sciences, Inc., New York, NY, USA
- LineaRx, Inc. , New York, NY, USA
| | - Clay Shorrock
- Applied DNA Sciences, Inc., New York, NY, USA
- LineaRx, Inc. , New York, NY, USA
| | - Luigi Aurisicchio
- Takis Biotech, Rome, Italy
- Evvivax Biotech, Rome, Italy
- Neomatrix Biotech, Rome, Italy
| | - Diego G Diel
- Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
19
|
Vilas Boas de Melo C, Peters F, van Dijken H, Lenz S, van de Ven K, Wijsman L, Gomersbach A, Schouten T, van Kasteren PB, van den Brand J, de Jonge J. Influenza Infection in Ferrets with SARS-CoV-2 Infection History. Microbiol Spectr 2022; 10:e0138622. [PMID: 36301107 PMCID: PMC9784775 DOI: 10.1128/spectrum.01386-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 09/23/2022] [Indexed: 01/05/2023] Open
Abstract
Nonpharmaceutical interventions (NPIs) to contain the SARS-CoV-2 pandemic drastically reduced human-to-human interactions, decreasing the circulation of other respiratory viruses, as well. Consequently, influenza virus circulation, which is normally responsible for 3 to 5 million hospitalizations per year globally, was significantly reduced. With the downscaling of the NPI countermeasures, there is a concern for increased influenza disease, particularly in individuals suffering from postacute effects of SARS-CoV-2 infection. To investigate this, we performed a sequential influenza H1N1 infection 4 weeks after an initial SARS-CoV-2 infection in ferrets. Upon H1N1 infection, ferrets that were previously infected with SARS-CoV-2 showed an increased tendency to develop clinical signs, compared to the control H1N1-infected animals. A histopathological analysis indicated only a slight increase for type II pneumocyte hyperplasia and bronchitis. Thus, the effects of the sequential infection appeared minor. However, ferrets were infected with B.1.351-SARS-CoV-2, the beta variant of concern, which replicated poorly in our model. The histopathology of the respiratory organs was mostly resolved 4 weeks after the SARS-CoV-2 infection, with only reminiscent histopathological features in the upper respiratory tract. Nevertheless, SARS-CoV-2 specific cellular and humoral responses were observed, confirming an established infection. On account of a modest trend toward the enhancement of the influenza disease, even upon a mild SARS-CoV-2 infection, our findings suggest that a stronger SARS-CoV-2 infection and its consequent, long-term effects could have a greater impact on the outcome of disease after a sequential influenza infection. Hence, the influenza vaccination of individuals suffering from postacute SARS-CoV-2 infection effects may be considered an avertible measure for such a scenario. IMPORTANCE During the COVID-19 pandemic, the use of face masks, social distancing, and isolation were effective not only in decreasing the circulation of SARS-CoV-2 but also in reducing other respiratory viruses, such as influenza. With fewer restrictions currently in place, influenza is slowly returning. In the meantime, people who are still suffering from long-COVID could be more vulnerable to an influenza virus infection and could develop a more severe influenza disease. This study provides directions to the effect of a previous SARS-CoV-2 exposure on influenza disease severity in a ferret model. This model is highly valuable to test sequential infections under controlled settings for translation to humans. We could not induce clear long-term COVID-19 effects, as the SARS-CoV-2 infections in the ferrets were mild. However, we still observed a slight increase in influenza disease severity compared to ferrets that had not encountered SARS-CoV-2 before. Therefore, it may be advisable to include long-COVID patients as a risk group for influenza vaccination.
Collapse
Affiliation(s)
| | - Florence Peters
- National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Harry van Dijken
- National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Stefanie Lenz
- National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Koen van de Ven
- National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Lisa Wijsman
- National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Angéla Gomersbach
- Animal Research Centre, Poonawalla Science Park, Bilthoven, The Netherlands
| | - Tanja Schouten
- Animal Research Centre, Poonawalla Science Park, Bilthoven, The Netherlands
| | - Puck B. van Kasteren
- National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | | | - Jørgen de Jonge
- National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| |
Collapse
|
20
|
Belser JA, Sun X, Kieran TJ, Brock N, Pulit-Penaloza JA, Pappas C, Basu Thakur P, Jones J, Wentworth DE, Zhou B, Tumpey TM, Maines TR. Detection of Airborne Influenza A and SARS-CoV-2 Virus Shedding following Ocular Inoculation of Ferrets. J Virol 2022; 96:e0140322. [PMID: 36448801 PMCID: PMC9769371 DOI: 10.1128/jvi.01403-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 11/04/2022] [Indexed: 12/03/2022] Open
Abstract
Despite reports of confirmed human infection following ocular exposure with both influenza A virus (IAV) and SARS-CoV-2, the dynamics of virus spread throughout oculonasal tissues and the relative capacity of virus transmission following ocular inoculation remain poorly understood. Furthermore, the impact of exposure route on subsequent release of airborne viral particles into the air has not been examined previously. To assess this, ferrets were inoculated by the ocular route with A(H1N1)pdm09 and A(H7N9) IAVs and two SARS-CoV-2 (early pandemic Washington/1 and Delta variant) viruses. Virus replication was assessed in both respiratory and ocular specimens, and transmission was evaluated in direct contact or respiratory droplet settings. Viral RNA in aerosols shed by inoculated ferrets was quantified with a two-stage cyclone aerosol sampler (National Institute for Occupational Safety and Health [NIOSH]). All IAV and SARS-CoV-2 viruses mounted a productive and transmissible infection in ferrets following ocular inoculation, with peak viral titers and release of virus-laden aerosols from ferrets indistinguishable from those from ferrets inoculated by previously characterized intranasal inoculation methods. Viral RNA was detected in ferret conjunctival washes from all viruses examined, though infectious virus in this specimen was recovered only following IAV inoculation. Low-dose ocular-only aerosol exposure or inhalation aerosol exposure of ferrets to IAV similarly led to productive infection of ferrets and shedding of aerosolized virus. Viral evolution during infection was comparable between all inoculation routes examined. These data support that both IAV and SARS-CoV-2 can establish a high-titer mammalian infection following ocular exposure that is associated with rapid detection of virus-laden aerosols shed by inoculated animals. IMPORTANCE Documented human infection with influenza viruses and SARS-CoV-2 has been reported among individuals wearing respiratory protection in the absence of eye protection, highlighting the capacity of these respiratory tract-tropic viruses to exploit nonrespiratory routes of exposure to initiate productive infection. However, comprehensive evaluations of how ocular exposure may modulate virus pathogenicity and transmissibility in mammals relative to respiratory exposure are limited and have not investigated multiple virus families side by side. Using the ferret model, we show that ocular exposure with multiple strains of either coronaviruses or influenza A viruses leads to an infection that results in shedding of detectable aerosolized virus from inoculated animals, contributing toward onward transmission of both viruses to susceptible contacts. Collectively, these studies support that the ocular surface represents a susceptible mucosal surface that, if exposed to a sufficient quantity of either virus, permits establishment of an infection which is similarly transmissible as that following respiratory exposure.
Collapse
Affiliation(s)
- Jessica A. Belser
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Xiangjie Sun
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Troy J. Kieran
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Nicole Brock
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | | | - Claudia Pappas
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Poulami Basu Thakur
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Joyce Jones
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - David E. Wentworth
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Bin Zhou
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Terrence M. Tumpey
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Taronna R. Maines
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|
21
|
Khoury O, Clouse C, McSwain MK, Applegate J, Kock ND, Atala A, Murphy SV. Ferret acute lung injury model induced by repeated nebulized lipopolysaccharide administration. Physiol Rep 2022; 10:e15400. [PMID: 36268626 PMCID: PMC9585421 DOI: 10.14814/phy2.15400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 06/29/2022] [Accepted: 07/05/2022] [Indexed: 06/16/2023] Open
Abstract
Inflammatory lung diseases affect millions of people worldwide. These diseases are caused by a number of factors such as pneumonia, sepsis, trauma, and inhalation of toxins. Pulmonary function testing (PFT) is a valuable functional methodology for better understanding mechanisms of lung disease, measuring disease progression, clinical diagnosis, and evaluating therapeutic interventions. Animal models of inflammatory lung diseases are needed that accurately recapitulate disease manifestations observed in human patients and provide an accurate prediction of clinical outcomes using clinically relevant pulmonary disease parameters. In this study, we evaluated a ferret lung inflammation model that closely represents multiple clinical manifestations of acute lung inflammation and injury observed in human patients. Lipopolysaccharide (LPS) from Pseudomonas aeruginosa was nebulized into ferrets for 7 repeated daily doses. Repeated exposure to nebulized LPS resulted in a restrictive pulmonary injury characterized using Buxco forced maneuver PFT system custom developed for ferrets. This is the first study to report repeated forced maneuver PFT in ferrets, establishing lung function measurements pre- and post-injury in live animals. Bronchoalveolar lavage and histological analysis confirmed that LPS exposure elicited pulmonary neutrophilic inflammation and structural damage to the alveoli. We believe this ferret model of lung inflammation, with clinically relevant disease manifestations and parameters for functional evaluation, is a useful pre-clinical model for understanding human inflammatory lung disease and for the evaluation of potential therapies.
Collapse
Affiliation(s)
- Oula Khoury
- Wake Forest Institute for Regenerative MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Cara Clouse
- Wake Forest Institute for Regenerative MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Malcolm K. McSwain
- Wake Forest Institute for Regenerative MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Jeffrey Applegate
- Department of Clinical Sciences, College of Veterinary MedicineNorth Carolina State UniversityRaleighNorth CarolinaUSA
| | - Nancy D. Kock
- Department of Pathology/Comparative MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Anthony Atala
- Wake Forest Institute for Regenerative MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Sean V. Murphy
- Wake Forest Institute for Regenerative MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| |
Collapse
|
22
|
Abstract
The continued spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in humans necessitates evaluation of variants for enhanced virulence and transmission. We used the ferret model to perform a comparative analysis of four SARS-CoV-2 strains, including an early pandemic isolate from the United States (WA1), and representatives of the Alpha, Beta, and Delta lineages. While Beta virus was not capable of pronounced replication in ferrets, WA1, Alpha, and Delta viruses productively replicated in the ferret upper respiratory tract, despite causing only mild disease with no overt histopathological changes. Strain-specific transmissibility was observed; WA1 and Delta viruses transmitted in a direct contact setting, whereas Delta virus was also capable of limited airborne transmission. Viral RNA was shed in exhaled air particles from all inoculated animals but was highest for Delta virus. Prior infection with SARS-CoV-2 offered varied protection against reinfection with either homologous or heterologous variants. Notable genomic variants in the spike protein were most frequently detected following WA1 and Delta virus infection.
Collapse
|
23
|
Jansen EB, Orvold SN, Swan CL, Yourkowski A, Thivierge BM, Francis ME, Ge A, Rioux M, Darbellay J, Howland JG, Kelvin AA. After the virus has cleared-Can preclinical models be employed for Long COVID research? PLoS Pathog 2022; 18:e1010741. [PMID: 36070309 PMCID: PMC9451097 DOI: 10.1371/journal.ppat.1010741] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV-2) can cause the life-threatening acute respiratory disease called COVID-19 (Coronavirus Disease 2019) as well as debilitating multiorgan dysfunction that persists after the initial viral phase has resolved. Long COVID or Post-Acute Sequelae of COVID-19 (PASC) is manifested by a variety of symptoms, including fatigue, dyspnea, arthralgia, myalgia, heart palpitations, and memory issues sometimes affecting between 30% and 75% of recovering COVID-19 patients. However, little is known about the mechanisms causing Long COVID and there are no widely accepted treatments or therapeutics. After introducing the clinical aspects of acute COVID-19 and Long COVID in humans, we summarize the work in animals (mice, Syrian hamsters, ferrets, and nonhuman primates (NHPs)) to model human COVID-19. The virology, pathology, immune responses, and multiorgan involvement are explored. Additionally, any studies investigating time points longer than 14 days post infection (pi) are highlighted for insight into possible long-term disease characteristics. Finally, we discuss how the models can be leveraged for treatment evaluation, including pharmacological agents that are currently in human clinical trials for treating Long COVID. The establishment of a recognized Long COVID preclinical model representing the human condition would allow the identification of mechanisms causing disease as well as serve as a vehicle for evaluating potential therapeutics.
Collapse
Affiliation(s)
- Ethan B. Jansen
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Spencer N. Orvold
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Cynthia L. Swan
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Anthony Yourkowski
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Brittany M. Thivierge
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Magen E. Francis
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Anni Ge
- Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Melissa Rioux
- Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Joseph Darbellay
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - John G. Howland
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Alyson A. Kelvin
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
24
|
Schindell BG, Allardice M, McBride JA, Dennehy B, Kindrachuk J. SARS-CoV-2 and the Missing Link of Intermediate Hosts in Viral Emergence - What We Can Learn From Other Betacoronaviruses. FRONTIERS IN VIROLOGY 2022; 2. [DOI: 10.3389/fviro.2022.875213] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
The emergence of SARS-CoV-2 in 2019 has resulted in a global pandemic with devastating human health and economic consequences. The development of multiple vaccines, antivirals and supportive care modalities have aided in our efforts to gain control of the pandemic. However, the emergence of multiple variants of concern and spillover into numerous nonhuman animal species could protract the pandemic. Further, these events also increase the difficulty in simultaneously monitoring viral evolution across multiple species and predicting future spillback potential into the human population. Here, we provide historic context regarding the roles of reservoir and intermediate hosts in coronavirus circulation and discuss current knowledge of these for SARS-CoV-2. Increased understanding of SARS-CoV-2 zoonoses are fundamental for efforts to control the global health and economic impacts of COVID-19.
Collapse
|
25
|
Lin Q, Lu C, Hong Y, Li R, Chen J, Chen W, Chen J. Animal models for studying coronavirus infections and developing antiviral agents and vaccines. Antiviral Res 2022; 203:105345. [PMID: 35605699 PMCID: PMC9122840 DOI: 10.1016/j.antiviral.2022.105345] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 04/30/2022] [Accepted: 05/17/2022] [Indexed: 01/17/2023]
Abstract
In addition to severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV), SARS-CoV-2 has become the third deadly coronavirus that infects humans and causes the new coronavirus disease (COVID-19). COVID-19 has already caused more than six million deaths worldwide and it is likely the biggest pandemic of this century faced by mankind. Although many studies on SARS-CoV-2 have been conducted, a detailed understanding of SARS-CoV-2 and COVID-19 is still lacking. Animal models are indispensable for studying its pathogenesis and developing vaccines and antivirals. In this review, we analyze animal models of coronavirus infections and explore their applications on antivirals and vaccines.
Collapse
Affiliation(s)
- Qisheng Lin
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Chunni Lu
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Monash University, Clayton, Victoria 3168, Australia
| | - Yuqi Hong
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Runfeng Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Jinding Chen
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Weisan Chen
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia.
| | - Jianxin Chen
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
26
|
Lossi L. Anatomical features for an adequate choice of the experimental animal model in biomedicine: III. Ferret, goat, sheep, and horse. Ann Anat 2022; 244:151978. [PMID: 35787443 DOI: 10.1016/j.aanat.2022.151978] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/08/2022] [Accepted: 06/09/2022] [Indexed: 11/17/2022]
Abstract
The anatomical characteristics of each of the many species today employed in biomedical research are very important when selecting the correct animal model(s), especially for conducting translational research. In previous papers, these features have been considered for fish (D'Angelo et al. Ann. Anat, 2016, 205:75), the most common laboratory rodents, rabbits, and pigs (Lossi et al. 2016). I here follow this line of discussion by dealing with the importance of proper knowledge of ferrets, goats, sheep, and horses' main anatomical features in translational research.
Collapse
Affiliation(s)
- Laura Lossi
- University of Turin, Department of Veterinary Sciences, Turin, Italy; INN, Istituto Nazionale di Neuroscienze, Turin, Italy.
| |
Collapse
|
27
|
Ciurkiewicz M, Armando F, Schreiner T, de Buhr N, Pilchová V, Krupp-Buzimikic V, Gabriel G, von Köckritz-Blickwede M, Baumgärtner W, Schulz C, Gerhauser I. Ferrets are valuable models for SARS-CoV-2 research. Vet Pathol 2022; 59:661-672. [PMID: 35001763 PMCID: PMC9207987 DOI: 10.1177/03009858211071012] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), resulted in an ongoing pandemic with millions of deaths worldwide. Infection of humans can be asymptomatic or result in fever, fatigue, dry cough, dyspnea, and acute respiratory distress syndrome with multiorgan failure in severe cases. The pathogenesis of COVID-19 is not fully understood, and various models employing different species are currently applied. Ferrets can be infected with SARS-CoV-2 and efficiently transmit the virus to contact animals. In contrast to hamsters, ferrets usually show mild disease and viral replication restricted to the upper airways. Most reports have used the intranasal inoculation route, while the intratracheal infection model is not well characterized. Herein, we present clinical, virological, and pathological data from young ferrets intratracheally inoculated with SARS-CoV-2. Infected animals showed no significant clinical signs, and had transient infection with peak viral RNA loads at 4 days postinfection, mild to moderate rhinitis, and pulmonary endothelialitis/vasculitis. Viral antigen was exclusively found in the respiratory epithelium of the nasal cavity, indicating a particular tropism for cells in this location. Viral antigen was associated with epithelial damage and influx of inflammatory cells, including activated neutrophils releasing neutrophil extracellular traps. Scanning electron microscopy of the nasal respiratory mucosa revealed loss of cilia, shedding, and rupture of epithelial cells. The currently established ferret SARS-CoV-2 infection models are comparatively discussed with SARS-CoV-2 pathogenesis in mink, and the advantages and disadvantages of both species as research models for zoonotic betacoronaviruses are highlighted.
Collapse
Affiliation(s)
| | - Federico Armando
- University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Tom Schreiner
- University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Nicole de Buhr
- University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Veronika Pilchová
- University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Vanessa Krupp-Buzimikic
- University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Gülşah Gabriel
- University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | | | | | - Claudia Schulz
- University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Ingo Gerhauser
- University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| |
Collapse
|
28
|
Coinfection of Chickens with H9N2 and H7N9 Avian Influenza Viruses Leads to Emergence of Reassortant H9N9 Virus with Increased Fitness for Poultry and a Zoonotic Potential. J Virol 2022; 96:e0185621. [PMID: 35019727 PMCID: PMC8906417 DOI: 10.1128/jvi.01856-21] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
An H7N9 low-pathogenicity avian influenza virus (LPAIV) emerged in 2013 through genetic reassortment between H9N2 and other LPAIVs circulating in birds in China. This virus causes inapparent clinical disease in chickens, but zoonotic transmission results in severe and fatal disease in humans. To examine a natural reassortment scenario between H7N9 and G1 lineage H9N2 viruses predominant in the Indian subcontinent, we performed an experimental coinfection of chickens with A/Anhui/1/2013/H7N9 (Anhui/13) virus and A/Chicken/Pakistan/UDL-01/2008/H9N2 (UDL/08) virus. Plaque purification and genotyping of the reassortant viruses shed via the oropharynx of contact chickens showed H9N2 and H9N9 as predominant subtypes. The reassortant viruses shed by contact chickens also showed selective enrichment of polymerase genes from H9N2 virus. The viable "6+2" reassortant H9N9 (having nucleoprotein [NP] and neuraminidase [NA] from H7N9 and the remaining genes from H9N2) was successfully shed from the oropharynx of contact chickens, plus it showed an increased replication rate in human A549 cells and a significantly higher receptor binding to α2,6 and α2,3 sialoglycans compared to H9N2. The reassortant H9N9 virus also had a lower fusion pH, replicated in directly infected ferrets at similar levels compared to H7N9 and transmitted via direct contact. Ferrets exposed to H9N9 via aerosol contact were also found to be seropositive, compared to H7N9 aerosol contact ferrets. To the best of our knowledge, this is the first study demonstrating that cocirculation of H7N9 and G1 lineage H9N2 viruses could represent a threat for the generation of novel reassortant H9N9 viruses with greater virulence in poultry and a zoonotic potential. IMPORTANCE We evaluated the consequences of reassortment between the H7N9 and the contemporary H9N2 viruses of the G1 lineage that are enzootic in poultry across the Indian subcontinent and the Middle East. Coinfection of chickens with these viruses resulted in the emergence of novel reassortant H9N9 viruses with genes derived from both H9N2 and H7N9 viruses. The "6+2" reassortant H9N9 (having NP and NA from H7N9) virus was shed from contact chickens in a significantly higher proportion compared to most of the reassortant viruses, showed significantly increased replication fitness in human A549 cells, receptor binding toward human (α2,6) and avian (α2,3) sialic acid receptor analogues, and the potential to transmit via contact among ferrets. This study demonstrated the ability of viruses that already exist in nature to exchange genetic material, highlighting the potential emergence of viruses from these subtypes with zoonotic potential.
Collapse
|
29
|
Kim YI, Casel MAB, Choi YK. Transmissibility and pathogenicity of SARS-CoV-2 variants in animal models. J Microbiol 2022; 60:255-267. [PMID: 35235177 PMCID: PMC8890026 DOI: 10.1007/s12275-022-2033-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 12/13/2022]
Abstract
As of February 2022, SARS-CoV-2 is still one of the most serious public health threats due to its high mortality rate and rapid spread of novel variants. Since the first outbreak in 2019, general understanding of SARS-CoV-2 has been improved through basic and clinical studies; however, knowledge gaps still exist in our understanding of the emerging novel SARSCoV-2 variants, which impacts the corresponding development of vaccines and therapeutics. Especially, accumulation of mutations in SARS-CoV-2 and rapid spread in populations with previous immunity has resulted in selection of variants that evade the host immune response. This phenomenon threatens to render current SARS-CoV-2 vaccines ineffective for controlling the pandemic. Proper animal models are essential for detailed investigations into the viral etiology, transmission and pathogenesis mechanisms, as well as evaluation of the efficacy of vaccine candidates against recent SARS-CoV-2 variants. Further, the choice of animal model for each research topic is important for researchers to gain better knowledge of recent SARS-CoV-2 variants. Here, we review the advantages and limitations of each animal model, including mice, hamsters, ferrets, and non-human primates, to elucidate variant SARS-CoV-2 etiology and transmission and to evaluate therapeutic and vaccine efficacy.
Collapse
Affiliation(s)
- Young-Il Kim
- Center for Study of Emerging and Re-emerging Viruses, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon, 34126, Republic of Korea
| | - Mark Anthony B Casel
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Young Ki Choi
- Center for Study of Emerging and Re-emerging Viruses, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon, 34126, Republic of Korea.
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, 28644, Republic of Korea.
| |
Collapse
|
30
|
Zhang Y, Almazi JG, Ong HX, Johansen MD, Ledger S, Traini D, Hansbro PM, Kelleher AD, Ahlenstiel CL. Nanoparticle Delivery Platforms for RNAi Therapeutics Targeting COVID-19 Disease in the Respiratory Tract. Int J Mol Sci 2022; 23:2408. [PMID: 35269550 PMCID: PMC8909959 DOI: 10.3390/ijms23052408] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/14/2022] [Accepted: 02/18/2022] [Indexed: 02/06/2023] Open
Abstract
Since December 2019, a pandemic of COVID-19 disease, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has rapidly spread across the globe. At present, the Food and Drug Administration (FDA) has issued emergency approval for the use of some antiviral drugs. However, these drugs still have limitations in the specific treatment of COVID-19, and as such, new treatment strategies urgently need to be developed. RNA-interference-based gene therapy provides a tractable target for antiviral treatment. Ensuring cell-specific targeted delivery is important to the success of gene therapy. The use of nanoparticles (NPs) as carriers for the delivery of small interfering RNA (siRNAs) to specific tissues or organs of the human body could play a crucial role in the specific therapy of severe respiratory infections, such as COVID-19. In this review, we describe a variety of novel nanocarriers, such as lipid NPs, star polymer NPs, and glycogen NPs, and summarize the pre-clinical/clinical progress of these nanoparticle platforms in siRNA delivery. We also discuss the application of various NP-capsulated siRNA as therapeutics for SARS-CoV-2 infection, the challenges with targeting these therapeutics to local delivery in the lung, and various inhalation devices used for therapeutic administration. We also discuss currently available animal models that are used for preclinical assessment of RNA-interference-based gene therapy. Advances in this field have the potential for antiviral treatments of COVID-19 disease and could be adapted to treat a range of respiratory diseases.
Collapse
Affiliation(s)
- Yuan Zhang
- Kirby Institute, UNSW, Sydney, NSW 2052, Australia; (Y.Z.); (S.L.); (A.D.K.)
| | - Juhura G. Almazi
- Respiratory Technology, Woolcock Institute of Medical Research, Sydney, NSW 2037, Australia; (J.G.A.); (H.X.O.); (D.T.)
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Ryde, NSW 2109, Australia
| | - Hui Xin Ong
- Respiratory Technology, Woolcock Institute of Medical Research, Sydney, NSW 2037, Australia; (J.G.A.); (H.X.O.); (D.T.)
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Ryde, NSW 2109, Australia
| | - Matt D. Johansen
- Centre for Inflammation, Faculty of Science, Centenary Institute and University of Technology Sydney, Sydney, NSW 2050, Australia; (M.D.J.); (P.M.H.)
| | - Scott Ledger
- Kirby Institute, UNSW, Sydney, NSW 2052, Australia; (Y.Z.); (S.L.); (A.D.K.)
| | - Daniela Traini
- Respiratory Technology, Woolcock Institute of Medical Research, Sydney, NSW 2037, Australia; (J.G.A.); (H.X.O.); (D.T.)
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Ryde, NSW 2109, Australia
| | - Philip M. Hansbro
- Centre for Inflammation, Faculty of Science, Centenary Institute and University of Technology Sydney, Sydney, NSW 2050, Australia; (M.D.J.); (P.M.H.)
| | - Anthony D. Kelleher
- Kirby Institute, UNSW, Sydney, NSW 2052, Australia; (Y.Z.); (S.L.); (A.D.K.)
| | | |
Collapse
|
31
|
Abstract
For four decades, genetically altered laboratory animals have provided invaluable information. Originally, genetic modifications were performed on only a few animal species, often chosen because of the ready accessibility of embryonic materials and short generation times. The methods were often slow, inefficient and expensive. In 2013, a new, extremely efficient technology, namely CRISPR/Cas9, not only made the production of genetically altered organisms faster and cheaper, but also opened it up to non-conventional laboratory animal species. CRISPR/Cas9 relies on a guide RNA as a 'location finder' to target DNA double strand breaks induced by the Cas9 enzyme. This is a prerequisite for non-homologous end joining repair to occur, an error prone mechanism often generating insertion or deletion of genetic material. If a DNA template is also provided, this can lead to homology directed repair, allowing precise insertions, deletions or substitutions. Due to its high efficiency in targeting DNA, CRISPR/Cas9-mediated genetic modification is now possible in virtually all animal species for which we have genome sequence data. Furthermore, modifications of Cas9 have led to more refined genetic alterations from targeted single base-pair mutations to epigenetic modifications. The latter offer altered gene expression without genome alteration. With this ever growing genetic toolbox, the number and range of genetically altered conventional and non-conventional laboratory animals with simple or complex genetic modifications is growing exponentially.
Collapse
|
32
|
Kim YI, Yu KM, Koh JY, Kim EH, Kim SM, Kim EJ, Casel MAB, Rollon R, Jang SG, Song MS, Park SJ, Jeong HW, Kim EG, Lee OJ, Kim YD, Choi Y, Lee SA, Choi YJ, Park SH, Jung JU, Choi YK. Age-dependent pathogenic characteristics of SARS-CoV-2 infection in ferrets. Nat Commun 2022; 13:21. [PMID: 35013229 PMCID: PMC8748994 DOI: 10.1038/s41467-021-27717-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 12/03/2021] [Indexed: 12/15/2022] Open
Abstract
While the seroprevalence of SARS-CoV-2 in healthy people does not differ significantly among age groups, those aged 65 years or older exhibit strikingly higher COVID-19 mortality compared to younger individuals. To further understand differing COVID-19 manifestations in patients of different ages, three age groups of ferrets are infected with SARS-CoV-2. Although SARS-CoV-2 is isolated from all ferrets regardless of age, aged ferrets (≥3 years old) show higher viral loads, longer nasal virus shedding, and more severe lung inflammatory cell infiltration, and clinical symptoms compared to juvenile (≤6 months) and young adult (1–2 years) groups. Furthermore, direct contact ferrets co-housed with the virus-infected aged group shed more virus than direct-contact ferrets co-housed with virus-infected juvenile or young adult ferrets. Transcriptome analysis of aged ferret lungs reveals strong enrichment of gene sets related to type I interferon, activated T cells, and M1 macrophage responses, mimicking the gene expression profile of severe COVID-19 patients. Thus, SARS-CoV-2-infected aged ferrets highly recapitulate COVID-19 patients with severe symptoms and are useful for understanding age-associated infection, transmission, and pathogenesis of SARS-CoV-2. Here, Kim et al. characterize SARS-CoV-2 infection in juvenile, young, and old aged ferrets to provide a further understanding of differences in COVID-19 severity in humans at different ages. Aged ferrets have higher viral loads, shed virus longer, and mimic the transcriptomic profile of severely infected patients.
Collapse
Affiliation(s)
- Young-Il Kim
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea.,Zoonotic Infectious Diseases Research Center, Chungbuk National University, Cheongju, Korea.,Center for Study of Emerging and Re-emerging Viruses, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon, 34126, Republic of Korea
| | - Kwang-Min Yu
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea.,Zoonotic Infectious Diseases Research Center, Chungbuk National University, Cheongju, Korea
| | - June-Young Koh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Eun-Ha Kim
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea.,Zoonotic Infectious Diseases Research Center, Chungbuk National University, Cheongju, Korea
| | - Se-Mi Kim
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea.,Zoonotic Infectious Diseases Research Center, Chungbuk National University, Cheongju, Korea.,Center for Study of Emerging and Re-emerging Viruses, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon, 34126, Republic of Korea
| | - Eun Ji Kim
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea.,Zoonotic Infectious Diseases Research Center, Chungbuk National University, Cheongju, Korea
| | - Mark Anthony B Casel
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea.,Zoonotic Infectious Diseases Research Center, Chungbuk National University, Cheongju, Korea
| | - Rare Rollon
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea
| | - Seung-Gyu Jang
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea
| | - Min-Suk Song
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea.,Zoonotic Infectious Diseases Research Center, Chungbuk National University, Cheongju, Korea
| | - Su-Jin Park
- Division of Life Science, Research Institute of Life Science, Gyeongsang National University, Jinju, 52828, Korea
| | - Hye Won Jeong
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea
| | - Eung-Gook Kim
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea
| | - Ok-Jun Lee
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea
| | - Yong-Dae Kim
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea
| | - Younho Choi
- Cancer Biology Department and Global Center for Pathogens Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Shin-Ae Lee
- Cancer Biology Department and Global Center for Pathogens Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Youn Jung Choi
- Cancer Biology Department and Global Center for Pathogens Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Su-Hyung Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Jae U Jung
- Cancer Biology Department and Global Center for Pathogens Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| | - Young Ki Choi
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea. .,Zoonotic Infectious Diseases Research Center, Chungbuk National University, Cheongju, Korea. .,Center for Study of Emerging and Re-emerging Viruses, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon, 34126, Republic of Korea.
| |
Collapse
|
33
|
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and seasonal influenza viruses are co-circulating in the human population. However, only a few cases of viral co-infection with these two viruses have been documented in humans with some people having severe disease and others mild disease. In order to examine this phenomenon, ferrets were co-infected with SARS-CoV-2 and human seasonal influenza A viruses (IAVs) (H1N1 or H3N2) and were compared to animals that received each virus alone. Ferrets were either immunologically naïve to both viruses or vaccinated with the 2019-2020 split-inactivated influenza virus vaccine. Co-infected naive ferrets lost significantly more body weight than ferrets infected with each virus alone and induced more severe inflammation in both the nose and lungs than ferrets single-infected with each virus. Co-infected naïve animals had predominantly higher IAV titers than SARS-CoV-2 titers, and IAVs efficiently transmitted to the co-housed ferrets by direct contact. Comparatively, SARS-CoV-2 failed to transmit to the ferrets that co-housed with co-infected ferrets by direct contact. Moreover, vaccination significantly reduced IAVs virus titers and shortened the viral shedding, but did not completely block influenza virus direct contact transmission. Notably, vaccination significantly ameliorated the influenza associated disease by protecting vaccinated animals from severe morbidity after IAV single infection or IAV and SARS-CoV-2 co-infection, suggesting that seasonal influenza virus vaccination is pivotal to prevent severe disease induced by IAVs and SARS-CoV-2 co-infection during the COVID-19 pandemic.
Importance
Influenza A viruses cause severe morbidity and mortality during each influenza virus season. The emergence of SARS-CoV-2 infection in the human population offers the opportunity to potential co-infections of both viruses. The development of useful animal models to asses pathogenesis, transmission, and viral evolution of these viruses as the co-infect a host is of critical importance for the development of vaccines and therapeutics. The ability to prevent the most severe effects of viral co-infections can be studied using effect co-infection ferret models described in this report.
Collapse
|
34
|
Bi Z, Hong W, Yang J, Lu S, Peng X. Animal models for SARS-CoV-2 infection and pathology. MedComm (Beijing) 2021; 2:548-568. [PMID: 34909757 PMCID: PMC8662225 DOI: 10.1002/mco2.98] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 10/10/2021] [Accepted: 10/13/2021] [Indexed: 02/05/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the etiology of coronavirus disease 2019 (COVID-19) pandemic. Current variants including Alpha, Beta, Gamma, Delta, and Lambda increase the capacity of infection and transmission of SARS-CoV-2, which might disable the in-used therapies and vaccines. The COVID-19 has now put an enormous strain on health care system all over the world. Therefore, the development of animal models that can capture characteristics and immune responses observed in COVID-19 patients is urgently needed. Appropriate models could accelerate the testing of therapeutic drugs and vaccines against SARS-CoV-2. In this review, we aim to summarize the current animal models for SARS-CoV-2 infection, including mice, hamsters, nonhuman primates, and ferrets, and discuss the details of transmission, pathology, and immunology induced by SARS-CoV-2 in these animal models. We hope this could throw light to the increased usefulness in fundamental studies of COVID-19 and the preclinical analysis of vaccines and therapeutic agents.
Collapse
Affiliation(s)
- Zhenfei Bi
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanChina
| | - Weiqi Hong
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanChina
| | - Jingyun Yang
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanChina
| | - Shuaiyao Lu
- National Kunming High‐level Biosafety Primate Research CenterInstitute of Medical BiologyChinese Academy of Medical Sciences and Peking Union Medical CollegeYunnanChina
| | - Xiaozhong Peng
- National Kunming High‐level Biosafety Primate Research CenterInstitute of Medical BiologyChinese Academy of Medical Sciences and Peking Union Medical CollegeYunnanChina
| |
Collapse
|
35
|
Renn M, Bartok E, Zillinger T, Hartmann G, Behrendt R. Animal models of SARS-CoV-2 and COVID-19 for the development of prophylactic and therapeutic interventions. Pharmacol Ther 2021; 228:107931. [PMID: 34171328 PMCID: PMC8219947 DOI: 10.1016/j.pharmthera.2021.107931] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 05/20/2021] [Accepted: 05/24/2021] [Indexed: 01/01/2023]
Abstract
Infections of the Coronavirus SARS-CoV-2 continue to spread around the globe, causing Coronavirus Disease (COVID)-19. Infected people are at risk of developing acute interstitial pneumonia, which can result in lethal complications, particularly in patients with pre-existing co-morbidities. Novel prophylactic and therapeutic interventions are urgently needed to limit the infection-associated health risk for the population and to contain the pandemic. Animal models are indispensable to assessing the efficacy and safety of potential new antivirals, vaccines, and other innovative therapies, such as nucleic acid agonists of innate immune sensing receptors. In this review, we provide an overview of the commonly used animal models to study SARS-CoV-2 and COVID-19, including a summary of their susceptibility to infection, the spectrum of symptoms elicited, and the potential for drug development in each model. We hope that this review will help researchers to decide on the right model organism to quickly address their specific scientific questions.
Collapse
Affiliation(s)
- Marcel Renn
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127 Bonn, Germany; Mildred Scheel School of Oncology, University Hospital Bonn, Medical Faculty, 53127 Bonn, Germany
| | - Eva Bartok
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127 Bonn, Germany; Unit of Experimental Immunology, Department of Biomedical Sciences, Institute of Tropical Medicine, 2000 Antwerp, Belgium
| | - Thomas Zillinger
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127 Bonn, Germany; Institute of Immunology, School of Medicine, Philipps University Marburg, 35043 Marburg, Germany
| | - Gunther Hartmann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127 Bonn, Germany
| | - Rayk Behrendt
- Institute for Immunology, Technische Universität Dresden, Medical Faculty Carl Gustav Carus, 01307 Dresden, Germany.
| |
Collapse
|
36
|
Escobedo RA, Singh DK, Kaushal D. Understanding COVID-19: From Dysregulated Immunity to Vaccination Status Quo. Front Immunol 2021; 12:765349. [PMID: 34858417 PMCID: PMC8632224 DOI: 10.3389/fimmu.2021.765349] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 10/25/2021] [Indexed: 12/27/2022] Open
Abstract
The development of vaccines against infectious diseases has helped us battle the greatest threat to public health. With the emergence of novel viruses, targeted immunotherapeutics ranging from informed vaccine development to personalized medicine may be the very thing that separates us between life and death. Late in 2019, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the etiological agent of coronavirus disease 2019 (COVID-19), made a remarkable entrance to human civilization, being one of many to cross the species barrier. This review discusses the important aspects of COVID-19, providing a brief overview of our current understanding of dysregulated immune responses developed using various experimental models, a brief outline of experimental models of COVID-19 and more importantly, the rapid development of vaccines against COVID-19.
Collapse
Affiliation(s)
- Ruby A. Escobedo
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, United States
- The Integrated Biomedical Sciences (IBMS) Graduate Program, University of Texas Health Sciences Center at San Antonio, San Antonio, TX, United States
| | - Dhiraj K. Singh
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, United States
| |
Collapse
|
37
|
Development of an Inactivated Vaccine against SARS CoV-2. Vaccines (Basel) 2021; 9:vaccines9111266. [PMID: 34835197 PMCID: PMC8624180 DOI: 10.3390/vaccines9111266] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/26/2021] [Accepted: 10/30/2021] [Indexed: 12/15/2022] Open
Abstract
The rapid spread of SARS-CoV-2 with its mutating strains has posed a global threat to safety during this COVID-19 pandemic. Thus far, there are 123 candidate vaccines in human clinical trials and more than 190 candidates in preclinical development worldwide as per the WHO on 1 October 2021. The various types of vaccines that are currently approved for emergency use include viral vectors (e.g., adenovirus, University of Oxford/AstraZeneca, Gamaleya Sputnik V, and Johnson & Johnson), mRNA (Moderna and Pfizer-BioNTech), and whole inactivated (Sinovac Biotech and Sinopharm) vaccines. Amidst the emerging cases and shortages of vaccines for global distribution, it is vital to develop a vaccine candidate that recapitulates the severe and fatal progression of COVID-19 and further helps to cope with the current outbreak. Hence, we present the preclinical immunogenicity, protective efficacy, and safety evaluation of a whole-virion inactivated SARS-CoV-2 vaccine candidate (ERUCoV-VAC) formulated in aluminium hydroxide, in three animal models, BALB/c mice, transgenic mice (K18-hACE2), and ferrets. The hCoV-19/Turkey/ERAGEM-001/2020 strain was used for the safety evaluation of ERUCoV-VAC. It was found that ERUCoV-VAC was highly immunogenic and elicited a strong immune response in BALB/c mice. The protective efficacy of the vaccine in K18-hACE2 showed that ERUCoV-VAC induced complete protection of the mice from a lethal SARS-CoV-2 challenge. Similar viral clearance rates with the safety evaluation of the vaccine in upper respiratory tracts were also positively appreciable in the ferret models. ERUCoV-VAC has been authorized by the Turkish Medicines and Medical Devices Agency and has now entered phase 3 clinical development (NCT04942405). The name of ERUCoV-VAC has been changed to TURKOVAC in the phase 3 clinical trial.
Collapse
|
38
|
78495111110.1152/physrev.00046.2020" />
Abstract
This medical review addresses the hypothesis that CD38/NADase is at the center of a functional axis (i.e., intracellular Ca2+ mobilization/IFNγ response/reactive oxygen species burst) driven by severe acute respiratory syndrome coronavirus 2 infection, as already verified in respiratory syncytial virus pathology and CD38 activity in other cellular settings. Key features of the hypothesis are that 1) the substrates of CD38 (e.g., NAD+ and NADP+) are depleted by viral-induced metabolic changes; 2) the products of the enzymatic activity of CD38 [e.g., cyclic adenosine diphosphate-ribose (ADPR)/ADPR/nicotinic acid adenine dinucleotide phosphate] and related enzymes [e.g., poly(ADP-ribose)polymerase, Sirtuins, and ADP-ribosyl hydrolase] are involved in the anti‐viral and proinflammatory response that favors the onset of lung immunopathology (e.g., cytokine storm and organ fibrosis); and 3) the pathological changes induced by this kinetic mechanism may be reduced by distinct modulators of the CD38/NAD+ axis (e.g., CD38 blockers, NAD+ suppliers, among others). This view is supported by arrays of associative basic and applied research data that are herein discussed and integrated with conclusions reported by others in the field of inflammatory, immune, tumor, and viral diseases.
Collapse
Affiliation(s)
- Alberto L. Horenstein
- Department of Medical Science, University of Turin, Turin, Italy; and Centro Ricerca Medicina, Sperimentale (CeRMS) and Fondazione Ricerca Molinette Onlus, Turin, Italy
| | - Angelo C. Faini
- Department of Medical Science, University of Turin, Turin, Italy; and Centro Ricerca Medicina, Sperimentale (CeRMS) and Fondazione Ricerca Molinette Onlus, Turin, Italy
| | - Fabio Malavasi
- Department of Medical Science, University of Turin, Turin, Italy; and Centro Ricerca Medicina, Sperimentale (CeRMS) and Fondazione Ricerca Molinette Onlus, Turin, Italy
| |
Collapse
|
39
|
de Vries RD, Rockx B, Haagmans BL, Herfst S, Koopmans MP, de Swart RL. Animal models of SARS-CoV-2 transmission. Curr Opin Virol 2021; 50:8-16. [PMID: 34256352 PMCID: PMC8238653 DOI: 10.1016/j.coviro.2021.06.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/17/2021] [Accepted: 06/21/2021] [Indexed: 02/06/2023]
Abstract
SARS-CoV-2 emerged in China as a zoonotic virus in December 2019. The virus proved to be human-to-human transmissible and its global spread resulted in the ongoing COVID-19 pandemic, associated with high morbidity and mortality. Vaccines were developed at an unprecedented speed and proved to be efficacious in preventing disease, but it remains to be determined if vaccines are able to interrupt transmission. Moreover, virus variants of concern continue to emerge that appear more transmissible and/or less sensitive to virus-specific immune responses. Here, we briefly review the role of animal models in assessing prophylactic and therapeutic options to interrupt SARS-CoV-2 transmission.
Collapse
Affiliation(s)
- Rory D de Vries
- Department Viroscience, Erasmus MC, University Medical Center Rotterdam, The Netherlands
| | - Barry Rockx
- Department Viroscience, Erasmus MC, University Medical Center Rotterdam, The Netherlands
| | - Bart L Haagmans
- Department Viroscience, Erasmus MC, University Medical Center Rotterdam, The Netherlands
| | - Sander Herfst
- Department Viroscience, Erasmus MC, University Medical Center Rotterdam, The Netherlands
| | - Marion Pg Koopmans
- Department Viroscience, Erasmus MC, University Medical Center Rotterdam, The Netherlands
| | - Rik L de Swart
- Department Viroscience, Erasmus MC, University Medical Center Rotterdam, The Netherlands.
| |
Collapse
|
40
|
Wang S, Li L, Yan F, Gao Y, Yang S, Xia X. COVID-19 Animal Models and Vaccines: Current Landscape and Future Prospects. Vaccines (Basel) 2021; 9:1082. [PMID: 34696190 PMCID: PMC8537799 DOI: 10.3390/vaccines9101082] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/17/2021] [Accepted: 09/23/2021] [Indexed: 12/23/2022] Open
Abstract
The worldwide pandemic of coronavirus disease 2019 (COVID-19) has become an unprecedented challenge to global public health. With the intensification of the COVID-19 epidemic, the development of vaccines and therapeutic drugs against the etiological agent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is also widespread. To prove the effectiveness and safety of these preventive vaccines and therapeutic drugs, available animal models that faithfully recapitulate clinical hallmarks of COVID-19 are urgently needed. Currently, animal models including mice, golden hamsters, ferrets, nonhuman primates, and other susceptible animals have been involved in the study of COVID-19. Moreover, 117 vaccine candidates have entered clinical trials after the primary evaluation in animal models, of which inactivated vaccines, subunit vaccines, virus-vectored vaccines, and messenger ribonucleic acid (mRNA) vaccines are promising vaccine candidates. In this review, we summarize the landscape of animal models for COVID-19 vaccine evaluation and advanced vaccines with an efficacy range from about 50% to more than 95%. In addition, we point out future directions for COVID-19 animal models and vaccine development, aiming at providing valuable information and accelerating the breakthroughs confronting SARS-CoV-2.
Collapse
Affiliation(s)
- Shen Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (S.W.); (X.X.)
| | - Ling Li
- National Research Center for Exotic Animal Diseases, China Animal Health and Epidemiology Center, Qingdao 266000, China;
| | - Feihu Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (S.W.); (X.X.)
| | - Yuwei Gao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (S.W.); (X.X.)
| | - Songtao Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (S.W.); (X.X.)
| | - Xianzhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (S.W.); (X.X.)
| |
Collapse
|
41
|
Viral and Bacterial Co-Infections in the Lungs: Dangerous Liaisons. Viruses 2021; 13:v13091725. [PMID: 34578306 PMCID: PMC8472850 DOI: 10.3390/v13091725] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 12/23/2022] Open
Abstract
Respiratory tract infections constitute a significant public health problem, with a therapeutic arsenal that remains relatively limited and that is threatened by the emergence of antiviral and/or antibiotic resistance. Viral–bacterial co-infections are very often associated with the severity of these respiratory infections and have been explored mainly in the context of bacterial superinfections following primary influenza infection. This review summarizes our current knowledge of the mechanisms underlying these co-infections between respiratory viruses (influenza viruses, RSV, and SARS-CoV-2) and bacteria, at both the physiological and immunological levels. This review also explores the importance of the microbiome and the pathological context in the evolution of these respiratory tract co-infections and presents the different in vitro and in vivo experimental models available. A better understanding of the complex functional interactions between viruses/bacteria and host cells will allow the development of new, specific, and more effective diagnostic and therapeutic approaches.
Collapse
|
42
|
COVID-19 Subunit Vaccine with a Combination of TLR1/2 and TLR3 Agonists Induces Robust and Protective Immunity. Vaccines (Basel) 2021; 9:vaccines9090957. [PMID: 34579194 PMCID: PMC8473206 DOI: 10.3390/vaccines9090957] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/20/2021] [Accepted: 08/25/2021] [Indexed: 12/23/2022] Open
Abstract
The development of COVID-19 vaccines is critical in controlling global health issues under the COVID-19 pandemic. The subunit vaccines are the safest and most widely used vaccine platform and highly effective against a multitude of infectious diseases. An adjuvant is essential for subunit vaccines to enhance the magnitude and durability of immune responses. In this study, we determined whether a combination of toll-like receptor (TLR)1/2 and TLR3 agonists (L-pampo) can be a potent adjuvant for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) subunit vaccine. We measured a neutralizing antibody (nAb) and an angiotensin-converting enzyme 2 (ACE2) receptor-blocking antibody against SARS-CoV-2 receptor-binding domain (RBD). We also detected interferon-gamma (IFN-γ) production by using ELISPOT and ELISA assays. By employing a ferret model, we detected nAbs and IFN-γ producing cells and measured viral load in nasal wash after the challenge of SARS-CoV-2. We found that SARS-CoV-2 antigens with L-pampo stimulated robust humoral and cellular immune responses. The efficacy of L-pampo was higher than the other adjuvants. Furthermore, in the ferret model, SARS-CoV-2 antigens with L-pampo elicited nAb response and antigen-specific cellular immune response against SARS-CoV-2, resulting in substantially decreased viral load in their nasal wash. Our study suggests that SARS-CoV-2 antigens formulated with TLR agonists, L-pampo, can be a potent subunit vaccine to promote sufficient protective immunity against SARS-CoV-2.
Collapse
|
43
|
Cummings TH, Magagnoli J, Hardin JW, Sutton SS. Drug repurposing of dextromethorphan as a cellular target for the management of influenza. Pharmacotherapy 2021; 41:796-803. [PMID: 34428315 DOI: 10.1002/phar.2618] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 08/10/2021] [Accepted: 08/10/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND Influenza viruses are responsible for seasonal epidemics and sporadic pandemics of varying severity in humans, and additional treatment options are needed. High-throughput siRNA screens and a pre-clinical research model demonstrated that dextromethorphan (DM) has anti-viral activity as a cellular target for treatment of influenza. This study examined DM usage and hospitalization rates among patients with laboratory-confirmed influenza in a national cohort of United States veterans. We aimed to evaluate the potential drug repurposing of DM as a cellular target for the management of influenza utilizing a large, national claims and electronic health record database. METHODS This retrospective drug-disease association cohort study was conducted using data from the Veterans Affairs Informatics and Computing Infrastructure (VINCI). We used a cohort with laboratory-confirmed diagnosis of influenza and international classification of disease (ICD)-9/10 diagnosis codes of fever, cough, influenza, or acute upper respiratory infection in an outpatient setting. The study outcome is inpatient hospitalization (all-cause and respiratory) within 30 days of influenza diagnosis. We estimated the relative risk for all-cause and respiratory hospitalizations using Poisson generalized linear model (GLM) and a greedy nearest neighbor propensity score 1:1 matched sub-analysis for both hospitalization models. FINDINGS A total of 18,677 patients met the inclusion and exclusion criteria and were evaluated in our study. The cohorts consisted of 2801 patients dispensed DM and 15,876 untreated patients (no DM). The Poisson GLM adjusted for covariates demonstrated a relative risk reduction of 34% for all-cause hospitalizations (Relative Risk (RR) 0.66, 95% Confidence Interval (CI) 0.525-0.832) and 40% for respiratory hospitalizations (RR 0.597, 95% CI 0.423-0.843) in patients with influenza treated with DM. CONCLUSION Influenza viruses continue to emerge and cause infection (including pandemics) in humans, so there remains a critical need to advance the understanding of influenza treatment. Our results demonstrated reduced hospitalization rates for influenza patients treated with DM. Further research on cellular targets and/or DM is warranted for the treatment of influenza.
Collapse
Affiliation(s)
- Tammy H Cummings
- Department of Clinical Pharmacy and Outcomes Sciences, College of Pharmacy, University of South Carolina, Columbia, South Carolina, USA.,Columbia VA Health Care System, Dorn Research Institute, Columbia, South Carolina, USA
| | - Joseph Magagnoli
- Department of Clinical Pharmacy and Outcomes Sciences, College of Pharmacy, University of South Carolina, Columbia, South Carolina, USA.,Columbia VA Health Care System, Dorn Research Institute, Columbia, South Carolina, USA
| | - James W Hardin
- Columbia VA Health Care System, Dorn Research Institute, Columbia, South Carolina, USA.,Department of Epidemiology & Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina, USA
| | - S Scott Sutton
- Department of Clinical Pharmacy and Outcomes Sciences, College of Pharmacy, University of South Carolina, Columbia, South Carolina, USA.,Columbia VA Health Care System, Dorn Research Institute, Columbia, South Carolina, USA
| |
Collapse
|
44
|
Tardiolo G, Brianti P, Sapienza D, dell’Utri P, Dio VD, Rao G, Calabrò RS. Are We Paving the Way to Dig Out of the "Pandemic Hole"? A Narrative Review on SARS-CoV-2 Vaccination: From Animal Models to Human Immunization. Med Sci (Basel) 2021; 9:53. [PMID: 34449681 PMCID: PMC8395838 DOI: 10.3390/medsci9030053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 01/08/2023] Open
Abstract
The Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is a new pathogen agent causing the coronavirus infectious disease (COVID-19). This novel virus originated the most challenging pandemic in this century, causing economic and social upheaval internationally. The extreme infectiousness and high mortality rates incentivized the development of vaccines to control this pandemic to prevent further morbidity and mortality. This international scenario led academic scientists, industries, and governments to work and collaborate strongly to make a portfolio of vaccines available at an unprecedented pace. Indeed, the robust collaboration between public systems and private companies led to resolutive actions for accelerating therapeutic interventions and vaccines mechanism. These strategies contributed to rapidly identifying safe and effective vaccines as quickly and efficiently as possible. Preclinical research employed animal models to develop vaccines that induce protective and long-lived immune responses. A spectrum of vaccines is worldwide under investigation in various preclinical and clinical studies to develop both individual protection and safe development of population-level herd immunity. Companies employed and developed different technological approaches for vaccines production, including inactivated vaccines, live-attenuated, non-replicating viral vector vaccines, as well as acid nucleic-based vaccines. In this view, the present narrative review provides an overview of current vaccination strategies, taking into account both preclinical studies and clinical trials in humans. Furthermore, to better understand immunization, animal models on SARS-CoV-2 pathogenesis are also briefly discussed.
Collapse
Affiliation(s)
- Giuseppe Tardiolo
- Department of Veterinary Sciences, University of Messina, Via Palatucci snc, 98168 Messina, Italy;
| | - Pina Brianti
- Unit of Dermatology, IRCCS San Raffaele Hospital, Via Olgettina 60, 20132 Milan, Italy;
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Piazza Pugliatti 1, 98122 Messina, Italy;
| | - Daniela Sapienza
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Piazza Pugliatti 1, 98122 Messina, Italy;
| | - Pia dell’Utri
- IRCCS Centro Neurolesi “Bonino Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (P.d.); (V.D.D.); (G.R.)
| | - Viviane Di Dio
- IRCCS Centro Neurolesi “Bonino Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (P.d.); (V.D.D.); (G.R.)
| | - Giuseppe Rao
- IRCCS Centro Neurolesi “Bonino Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (P.d.); (V.D.D.); (G.R.)
| | - Rocco Salvatore Calabrò
- IRCCS Centro Neurolesi “Bonino Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (P.d.); (V.D.D.); (G.R.)
| |
Collapse
|
45
|
Lee JS, Koh JY, Yi K, Kim YI, Park SJ, Kim EH, Kim SM, Park SH, Ju YS, Choi YK, Park SH. Single-cell transcriptome of bronchoalveolar lavage fluid reveals sequential change of macrophages during SARS-CoV-2 infection in ferrets. Nat Commun 2021; 12:4567. [PMID: 34315893 PMCID: PMC8316405 DOI: 10.1038/s41467-021-24807-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 07/07/2021] [Indexed: 12/15/2022] Open
Abstract
Few studies have used a longitudinal approach to describe the immune response to SARS-CoV-2 infection. Here, we perform single-cell RNA sequencing of bronchoalveolar lavage fluid cells longitudinally obtained from SARS-CoV-2-infected ferrets. Landscape analysis of the lung immune microenvironment shows distinct changes in cell proportions and characteristics compared to uninfected control, at 2 and 5 days post-infection (dpi). Macrophages are classified into 10 distinct subpopulations with transcriptome changes among monocyte-derived infiltrating macrophages and differentiated M1/M2 macrophages, notably at 2 dpi. Moreover, trajectory analysis reveals gene expression changes from monocyte-derived infiltrating macrophages toward M1 or M2 macrophages and identifies a macrophage subpopulation that has rapidly undergone SARS-CoV-2-mediated activation of inflammatory responses. Finally, we find that M1 or M2 macrophages show distinct patterns of gene modules downregulated by immune-modulatory drugs. Overall, these results elucidate fundamental aspects of the immune response dynamics provoked by SARS-CoV-2 infection.
Collapse
Affiliation(s)
| | - June-Young Koh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Kijong Yi
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Young-Il Kim
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea
| | - Su-Jin Park
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea
- Division of Life Science, Research Institute of Life Science, Gyeongsang National University, Jinju, Korea
| | - Eun-Ha Kim
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea
| | - Se-Mi Kim
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea
| | - Sung Ho Park
- School of Life Sciences, Ulsan National Institute of Science & Technology (UNIST), Ulsan, Republic of Korea
| | - Young Seok Ju
- GENOME INSIGHT Inc., Daejeon, Republic of Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- The Center for Epidemic Preparedness, KAIST Institute, KAIST, Daejeon, Republic of Korea
| | - Young Ki Choi
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea.
- Center for Study of Emerging and Re-emerging Viruses, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon, Republic of Korea.
| | - Su-Hyung Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.
- The Center for Epidemic Preparedness, KAIST Institute, KAIST, Daejeon, Republic of Korea.
| |
Collapse
|
46
|
Ingrole RSJ, Tao W, Joshi G, Gill HS. M2e conjugated gold nanoparticle influenza vaccine displays thermal stability at elevated temperatures and confers protection to ferrets. Vaccine 2021; 39:4800-4809. [PMID: 34301431 DOI: 10.1016/j.vaccine.2021.07.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 06/03/2021] [Accepted: 07/12/2021] [Indexed: 11/16/2022]
Abstract
Currently approved influenza vaccines are not only limited in breadth of protection but also have a limited shelf-life of 12-18 months when stored under appropriate conditions (2-8 °C). Inadvertent alteration in storage temperatures during manufacturing, transportation, distribution until delivery to patient, can damage the vaccine thus reducing its efficacy. A thermally stable vaccine can decrease the economic burden by reducing reliance on refrigeration system and can also enhance outreach of the vaccination program by allowing transportation to remote areas of the world where refrigerated conditions are scarce. We have previously developed a broadly protective influenza A vaccine by coupling the highly conserved extracellular region of the matrix 2 protein (M2e) of influenza A virus to gold nanoparticles (AuNPs) and upon subsequent addition of toll-like receptor 9 agonist - CpG, as an adjuvant, have shown its breadth of protection in a mouse model. In this study, we show that the vaccine is thermally stable when stored at 4 °C for 3 months, 37 °C for 3 months and 50 °C for 2 weeks in its lyophilized form, and later it was possible to readily reconstitute it in water without aggregation. Intranasal vaccination of mice using reconstituted vaccine induced M2e-specific IgG and IgG subtypes in serum similar to the freshly formulated vaccine, and fully protected mice against lethal influenza A challenge. Immunization of ferrets intranasally or intramuscularly with the vaccine induced M2e-specific IgG and there was reduced virus level in nasal wash of ferrets immunized through intranasal route.
Collapse
Affiliation(s)
- Rohan S J Ingrole
- Department of Chemical Engineering, Texas Tech University, 8th street and Canton Ave, Mail Stop 3121, Lubbock TX 79409-3121, USA
| | - Wenqian Tao
- Department of Chemical Engineering, Texas Tech University, 8th street and Canton Ave, Mail Stop 3121, Lubbock TX 79409-3121, USA
| | - Gaurav Joshi
- Department of Chemical Engineering, Texas Tech University, 8th street and Canton Ave, Mail Stop 3121, Lubbock TX 79409-3121, USA
| | - Harvinder Singh Gill
- Department of Chemical Engineering, Texas Tech University, 8th street and Canton Ave, Mail Stop 3121, Lubbock TX 79409-3121, USA.
| |
Collapse
|
47
|
Kim T, Lee JS, Ju YS. Experimental Models for SARS-CoV-2 Infection. Mol Cells 2021; 44:377-383. [PMID: 34187969 PMCID: PMC8245318 DOI: 10.14348/molcells.2021.0094] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/16/2021] [Accepted: 06/16/2021] [Indexed: 12/13/2022] Open
Abstract
Severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) is a novel virus that causes coronavirus disease 2019 (COVID-19). To understand the identity, functional characteristics and therapeutic targets of the virus and the diseases, appropriate infection models that recapitulate the in vivo pathophysiology of the viral infection are necessary. This article reviews the various infection models, including Vero cells, human cell lines, organoids, and animal models, and discusses their advantages and disadvantages. This knowledge will be helpful for establishing an efficient system for defense against emerging infectious diseases.
Collapse
Affiliation(s)
- Taewoo Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | | | - Young Seok Ju
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
- GENOME INSIGHT Inc., Daejeon 34051, Korea
| |
Collapse
|
48
|
Nguyen TQ, Rollon R, Choi YK. Animal Models for Influenza Research: Strengths and Weaknesses. Viruses 2021; 13:1011. [PMID: 34071367 PMCID: PMC8228315 DOI: 10.3390/v13061011] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/26/2021] [Accepted: 05/26/2021] [Indexed: 12/16/2022] Open
Abstract
Influenza remains one of the most significant public health threats due to its ability to cause high morbidity and mortality worldwide. Although understanding of influenza viruses has greatly increased in recent years, shortcomings remain. Additionally, the continuous mutation of influenza viruses through genetic reassortment and selection of variants that escape host immune responses can render current influenza vaccines ineffective at controlling seasonal epidemics and potential pandemics. Thus, there is a knowledge gap in the understanding of influenza viruses and a corresponding need to develop novel universal vaccines and therapeutic treatments. Investigation of viral pathogenesis, transmission mechanisms, and efficacy of influenza vaccine candidates requires animal models that can recapitulate the disease. Furthermore, the choice of animal model for each research question is crucial in order for researchers to acquire a better knowledge of influenza viruses. Herein, we reviewed the advantages and limitations of each animal model-including mice, ferrets, guinea pigs, swine, felines, canines, and non-human primates-for elucidating influenza viral pathogenesis and transmission and for evaluating therapeutic agents and vaccine efficacy.
Collapse
Affiliation(s)
- Thi-Quyen Nguyen
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju 28644, Korea; (T.-Q.N.); (R.R.)
| | - Rare Rollon
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju 28644, Korea; (T.-Q.N.); (R.R.)
| | - Young-Ki Choi
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju 28644, Korea; (T.-Q.N.); (R.R.)
- Zoonotic Infectious Diseases Research Center, Chungbuk National University, Cheongju 28644, Korea
| |
Collapse
|
49
|
Schiffman Z, Liu G, Cao W, Zhu W, Emeterio K, Qiu X, Banadyga L. The Ferret as a Model for Filovirus Pathogenesis and Countermeasure Evaluation. ILAR J 2021; 61:62-71. [PMID: 33951727 DOI: 10.1093/ilar/ilab011] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 12/04/2020] [Accepted: 01/14/2021] [Indexed: 11/13/2022] Open
Abstract
The domestic ferret (Mustela putorius furo) has long been a popular animal model for evaluating viral pathogenesis and transmission as well as the efficacy of candidate countermeasures. Without question, the ferret has been most widely implemented for modeling respiratory viruses, particularly influenza viruses; however, in recent years, it has gained attention as a novel animal model for characterizing filovirus infections. Although ferrets appear resistant to infection and disease caused by Marburg and Ravn viruses, they are highly susceptible to lethal disease caused by Ebola, Sudan, Bundibugyo, and Reston viruses. Notably, unlike the immunocompetent rodent models of filovirus infection, ferrets are susceptible to lethal disease caused by wild-type viruses, and they recapitulate many aspects of human filovirus disease, including systemic virus replication, coagulation abnormalities, and a dysregulated immune response. Along with the stringency with which they reproduce Ebola disease, their relatively small size and availability make ferrets an attractive choice for countermeasure evaluation and pathogenesis modeling. Indeed, they are so far the only small animal model available for Bundibugyo virus. Nevertheless, ferrets do have their limitations, including the lack of commercially available reagents to dissect host responses and their unproven predictive value in therapeutic evaluation. Although the use of the ferret model in ebolavirus research has been consistent over the last few years, its widespread use and utility remains to be fully proven. This review provides a comprehensive overview of the ferret models of filovirus infection and perspective on their ongoing use in pathogenesis modeling and countermeasure evaluation.
Collapse
Affiliation(s)
- Zachary Schiffman
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada.,Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Guodong Liu
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Wenguang Cao
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Wenjun Zhu
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Karla Emeterio
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada.,Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Xiangguo Qiu
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Logan Banadyga
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| |
Collapse
|
50
|
Case JB, Winkler ES, Errico JM, Diamond MS. On the road to ending the COVID-19 pandemic: Are we there yet? Virology 2021; 557:70-85. [PMID: 33676349 PMCID: PMC7908885 DOI: 10.1016/j.virol.2021.02.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/12/2021] [Accepted: 02/12/2021] [Indexed: 02/08/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged into the human population in late 2019 and caused the global COVID-19 pandemic. SARS-CoV-2 has spread to more than 215 countries and infected many millions of people. Despite the introduction of numerous governmental and public health measures to control disease spread, infections continue at an unabated pace, suggesting that effective vaccines and antiviral drugs will be required to curtail disease, end the pandemic, and restore societal norms. Here, we review the current developments in antibody and vaccine countermeasures to limit or prevent disease.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/biosynthesis
- COVID-19/epidemiology
- COVID-19/immunology
- COVID-19/prevention & control
- COVID-19/therapy
- COVID-19/virology
- COVID-19 Vaccines/administration & dosage
- COVID-19 Vaccines/biosynthesis
- COVID-19 Vaccines/immunology
- Clinical Trials as Topic
- Disease Models, Animal
- Genetic Vectors/chemistry
- Genetic Vectors/immunology
- Humans
- Immunity, Innate/drug effects
- Immunization, Passive/methods
- Immunogenicity, Vaccine
- Pandemics
- Patient Safety
- SARS-CoV-2/drug effects
- SARS-CoV-2/immunology
- SARS-CoV-2/pathogenicity
- Vaccines, Attenuated
- Vaccines, DNA
- Vaccines, Subunit
- Vaccines, Virus-Like Particle/administration & dosage
- Vaccines, Virus-Like Particle/biosynthesis
- Vaccines, Virus-Like Particle/immunology
- COVID-19 Serotherapy
Collapse
Affiliation(s)
- James Brett Case
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Emma S Winkler
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA; Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - John M Errico
- Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA; Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA; Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, 63110, USA; The Andrew M. and Jane M. Bursky Center for Human Immunology & Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|