Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Virology ; 582: 57-61, 2023 05.
Article in English | MEDLINE | ID: covidwho-2304372

ABSTRACT

Competition assays were conducted in vitro and in vivo to examine how the Delta (B.1.617.2) variant displaced the prototype Washington/1/2020 (WA/1) strain. While WA/1 virus exhibited a moderately increased proportion compared to that in the inoculum following co-infection in human respiratory cells, Delta variant possessed a substantial in vivo fitness advantage as this virus becoming predominant in both inoculated and contact animals. This work identifies critical traits of the Delta variant that likely played a role in it becoming a dominant variant and highlights the necessities of employing multiple model systems to assess the fitness of newly emerged SARS-CoV-2 variants.


Subject(s)
COVID-19 , Ferrets , Animals , Humans , SARS-CoV-2/genetics , Biological Assay
2.
Food and Agriculture Organization of the United Nations ; 2021.
Article in English, Arabic, Ru fr, Es zh | CAB Abstracts | ID: covidwho-2247079

ABSTRACT

SARS-CoV-2 was first identified in humans in December 2019 and has since affected almost 68 million people causing over 1.5 million deaths worldwide. Animal-to-human and animal-to-animal transmission has been documented within farmed minks in several countries. SARS-CoV-2 has been identified in a farmed mink population in a number of countries. Some of the affected farms reported also workers SARS-CoV-2 infection and it is hypothesized that the mink farms were infected through human-mink transmission proving SARS-CoV-2 capability of reverse zoonosis. This Tripartite Risk Assessment, as a joint effort under the GLEWS+ initiative, completed with the Food and Agriculture Organization (FAO), the World Health Organization (WHO), and the World Organisation for Animal Health (OIE), evaluates the risk of introduction and spread of SARS-CoV-2 within fur farming systems as well as whether farmed fur animals could play a significant role in the spread of SARS-CoV-2 to humans via spillover. Additionally, using a One Health approach, the Tripartite evaluated the risk of the escaped minks leading to the establishment of a viral reservoir in susceptible wildlife populations. This work provides guidance to Members on this newly emerging threat.

3.
Convegno SISVet ; 2022.
Article in English | CAB Abstracts | ID: covidwho-2279456

ABSTRACT

This paper describes the pathogenesis and immunology of Macaca mulatta, Macaca fascicularis, ferrets, Syrian golden hamsters (Mesocricetus auratus), mice, cats, mink, pigs and rabbits used as models for SARS-CoV-2 infection.

4.
J Nanobiotechnology ; 21(1): 60, 2023 Feb 22.
Article in English | MEDLINE | ID: covidwho-2252536

ABSTRACT

BACKGROUND: Unlike the injectable vaccines, intranasal lipid nanoparticle (NP)-based adjuvanted vaccine is promising to protect against local infection and viral transmission. Infection of ferrets with SARS-CoV-2 results in typical respiratory disease and pathology akin to in humans, suggesting that the ferret model may be ideal for intranasal vaccine studies. RESULTS: We developed SARS-CoV-2 subunit vaccine containing both Spike receptor binding domain (S-RBD) and Nucleocapsid (N) proteins (NP-COVID-Proteins) or their mRNA (NP-COVID-mRNA) and NP-monosodium urate adjuvant. Both the candidate vaccines in intranasal vaccinated aged ferrets substantially reduced the replicating virus in the entire respiratory tract. Specifically, the NP-COVID-Proteins vaccine did relatively better in clearing the virus from the nasal passage early post challenge infection. The immune gene expression in NP-COVID-Proteins vaccinates indicated increased levels of mRNA of IFNα, MCP1 and IL-4 in lungs and nasal turbinates, and IFNγ and IL-2 in lungs; while proinflammatory mediators IL-1ß and IL-8 mRNA levels in lungs were downregulated. In NP-COVID-Proteins vaccinated ferrets S-RBD and N protein specific IgG antibodies in the serum were substantially increased at both day post challenge (DPC) 7 and DPC 14, while the virus neutralizing antibody titers were relatively better induced by mRNA versus the proteins-based vaccine. In conclusion, intranasal NP-COVID-Proteins vaccine induced balanced Th1 and Th2 immune responses in the respiratory tract, while NP-COVID-mRNA vaccine primarily elicited antibody responses. CONCLUSIONS: Intranasal NP-COVID-Proteins vaccine may be an ideal candidate to elicit increased breadth of immunity against SARS-CoV-2 variants.


Subject(s)
COVID-19 , Influenza Vaccines , Humans , Animals , Aged , Ferrets , Immunity, Mucosal , SARS-CoV-2 , Viral Load , Antibodies, Viral , Lung/pathology , Antibodies, Neutralizing , Adjuvants, Immunologic , COVID-19 Vaccines , mRNA Vaccines
5.
Viruses ; 14(10)2022 10 04.
Article in English | MEDLINE | ID: covidwho-2066555

ABSTRACT

A preliminary vaccination trial against the emergent pathogen, SARS-CoV-2, was completed in captive black-footed ferrets (Mustela nigripes; BFF) to assess safety, immunogenicity, and anti-viral efficacy. Vaccination and boosting of 15 BFF with purified SARS-CoV-2 S1 subunit protein produced a nearly 150-fold increase in mean antibody titers compared to pre-vaccination titers. Serum antibody responses were highest in young animals, but in all vaccinees, antibody response declined rapidly. Anti-viral activity from vaccinated and unvaccinated BFF was determined in vitro, as well as in vivo with a passive serum transfer study in mice. Transgenic mice that received BFF serum transfers and were subsequently challenged with SARS-CoV-2 had lung viral loads that negatively correlated (p < 0.05) with the BFF serum titer received. Lastly, an experimental challenge study in a small group of BFF was completed to test susceptibility to SARS-CoV-2. Despite viral replication and shedding in the upper respiratory tract for up to 7 days post-challenge, no clinical disease was observed in either vaccinated or naive animals. The lack of morbidity or mortality observed indicates SARS-CoV-2 is unlikely to affect wild BFF populations, but infected captive animals pose a potential risk, albeit low, for humans and other animals.


Subject(s)
COVID-19 Vaccines , COVID-19 , Animals , Antibodies, Viral , Antiviral Agents , COVID-19/prevention & control , COVID-19 Vaccines/adverse effects , COVID-19 Vaccines/immunology , Ferrets , SARS-CoV-2
6.
Chinese Journal of Nosocomiology ; 32(9):1430-1434, 2022.
Article in English, Chinese | CAB Abstracts | ID: covidwho-2012147

ABSTRACT

Since the outbreak of COVID-19, the opinion that the aerosol plays a key role in the transmission of SARS-CoV-2 has been controversial. The COVID-19 pandemic has continued interpersonal transmission for more than two years, especially after the emergence of Delta and Omicron variants, making the situation of COVID-19 pandemic more severe. The transmission of SARS-COV-2 variants was significantly accelerated and the time of transmission between generations was significantly shortened. Therefore, it has been questioned to attribute the close-range infection to droplet transmission. The point that the aerosol can also has a close-range transmission and may play a dominant role is neglected under the influence of traditional transmission mode of respiratory infectious diseases. A large number of studies have shown that normal breath, talk and cough could release a large number of respiratory aerosol particles, and the virus particles were mainly tiny particles(=5.0 micro m). The biological activity and infectivity of the droplet nuclei have been questioned in the studies on their physicochemical properties. Animal models of ferrets and hamsters showed that SARS-COV-2 could transmit via aerosol. Therefore, the new evidence for the aerosol transmission of SARS-COV-2 was reviewed in the article so as to provide latest evidence-based evidence for prevention and control of COVID-19.

7.
Vaccines (Basel) ; 10(8)2022 Jul 25.
Article in English | MEDLINE | ID: covidwho-1957472

ABSTRACT

The COVID-19 pandemic is entering a new era with the approval of many SARS-CoV-2 vaccines. In spite of the restoration of an almost normal way of life thanks to the immune protection elicited by these innovative vaccines, we are still facing high viral circulation, with a significant number of deaths. To further explore alternative vaccination platforms, we developed COVID-eVax-a genetic vaccine based on plasmid DNA encoding the RBD domain of the SARS-CoV-2 spike protein. Here, we describe the correlation between immune responses and the evolution of viral infection in ferrets infected with the live virus. We demonstrate COVID-eVax immunogenicity as means of antibody response and, above all, a significant T-cell response, thus proving the critical role of T-cell immunity, in addition to the neutralizing antibody activity, in controlling viral spread.

8.
Infectious disease management in animal shelters ; 2(656), 2021.
Article in English | CAB Abstracts | ID: covidwho-1918436

ABSTRACT

This second edition contains 24 new and updated chapters on aetiology, epidemiology, prevalence, pathogenesis, clinical signs, treatment, prevention and control of infectious diseases in cats, dogs and exotic small companion mammals in animal shelters. These include an introduction to infectious disease management in animal shelters, wellness, data surveillance, diagnostic testing, necropsy techniques, outbreak management, pharmacology, sanitation, canine and feline vaccinations and immunology, canine infectious respiratory disease, canine distemper virus, canine influenza, feline infectious respiratory disease, canine parvovirus and other canine enteropathogens, feline panleukopenia, feline coronavirus and feline infectious peritonitis, internal parasites, heartworm disease, external parasites, dermatophytoses, zoonoses, rabies, feline leukaemia and feline immunodeficiency viruses and conditions in exotic companion mammals (ferrets, rabbits, guineapigs and rodents). It is intended for shelter veterinarians, managers and workers.

9.
Veterinar ; 59(1):14-23, 2021.
Article in Croatian | CAB Abstracts | ID: covidwho-1837993

ABSTRACT

The first cases of human disease from COVID-19 were recorded in December 2019 in China, from where it spread rapidly around the world. COVID-19, the third emerging coronavirus infection in humans, is caused by the new SARS-CoV-2 virus, which can cause a severe acute respiratory syndrome in some infected individuals. Previous research has revealed the possible animal origin of SARS-CoV-2, with bats considered as natural reservoirs and pangolins as intermediate hosts. To better understand COVID-19 and more successfully control the spread, domestic and wild animals have been infected in experimental conditions. On the other hand, in some species, infections have been recorded in field conditions. Natural infections have been reported in dogs, cats, tigers, lions, and minks, who have been in contact with SARS-CoV-2 positive humans. The reverse transmission of the pathogen, from infected animals to humans, has only been recorded on intensive mink farms. To better understand the pathogenesis of this disease's causative agent, drugs and vaccine trials, some experimental infections were performed on animal models, of which ferrets, rhesus macaques, and hamsters proved to be the most suitable. This article aimed to consolidate known data on the potential origin of SARS-CoV-2, its transmission to humans, infections in animals, and their significance in the epidemiology of COVID-19.

10.
Settimana Veterinaria ; : 35-36, 2021.
Article in Italian | CAB Abstracts | ID: covidwho-1837569
11.
Animals ; 12(9):1065, 2022.
Article in English | ProQuest Central | ID: covidwho-1837225

ABSTRACT

Simple SummaryHow ferrets across sectors are housed and the environmental enrichment provided (e.g., toys, beds, exploration of new sights and smells) can directly impact their health and wellbeing. Through an online questionnaire reaching ferret caretakers from pet owner, laboratory, zoo, rescue, and working (e.g., pest control) sectors, we describe how ferrets are housed, the enrichment they receive, enrichment types that ferrets most enjoy and those which may be harmful or problematic. Of 754 responses, 82.4% were from pet owners. Most ferrets were housed with at least one other ferret, and the type of housing varied across sectors from single-level cages to free-ranging housing. Environmental enrichments most commonly reported were hammocks, tunnels and human interaction, with ferrets reported to most enjoy digging, tunnels, human interaction and exploration. Scent trails were also reported to be among the most enjoyable enrichments but were rarely provided, suggesting that they could be used more. Problematic enrichment included rubber items, such as Kongs®, which could be chewed and swallowed, narrow tunnels trapping ferrets, and fabrics catching claws. These items should therefore be avoided. Our results suggest that all sectors have room to improve both housing and enrichment to better ferret welfare.Ferrets (Mustela putorius furo) are kept and used in multiple sectors of society, but little is known about how they are housed and what environmental enrichment (EE) they may benefit from. We aimed to help guide caretakers about what housing and EE can be provided for ferrets. Through an online questionnaire of ferret caretakers, including pet, laboratory, zoological collection, rescue and working animal sectors internationally, we described ferret housing, opportunities for exploration, EE provision and caretaker opinions on ferrets’ preferred EE types, and problematic EE. In total, 754 valid responses from 17 countries were analysed, with most (82.4%) coming from pet owners. Most ferrets were housed socially, with housing varying across sectors from single-level cages to free-range housing in a room or outdoor enclosure;pet owners mostly used multi-level cages. The most commonly reported EE included hammocks, tunnels and tactile interaction with caretakers. Respondents reported that ferrets particularly enjoyed digging substrates, tunnels, human interaction and exploration. The most frequently reported problems were that ingestion of unsuitable chew toys and rubber items could cause internal blockages, narrow tunnels could trap ferrets, and certain fabrics that could catch claws. This suggests a need for increased awareness of the risks of these EE types and for more commercially available safety-tested ferret EE. Scent trails were relatively rarely provided but were reported to be enjoyed and harmless, so we recommend that these should be provided more commonly. Our results suggest that there is scope to improve ferret housing and EE provision to benefit ferret welfare across all sectors.

12.
J Virol ; 96(5): e0179121, 2022 03 09.
Article in English | MEDLINE | ID: covidwho-1799229

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and seasonal influenza viruses are cocirculating in the human population. However, only a few cases of viral coinfection with these two viruses have been documented in humans with some people having severe disease and others mild disease. To examine this phenomenon, ferrets were coinfected 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 naive to both viruses or vaccinated with the 2019 to 2020 split-inactivated influenza virus vaccine. Coinfected naive ferrets lost significantly more body weight than ferrets infected with each virus alone and had more severe inflammation in both the nose and lungs compared to that of ferrets that were single infected with each virus. Coinfected, naive animals had predominantly higher IAV titers than SARS-CoV-2 titers, and IAVs were efficiently transmitted by direct contact to the cohoused ferrets. Comparatively, SARS-CoV-2 failed to transmit to the ferrets that cohoused with coinfected ferrets by direct contact. Moreover, vaccination significantly reduced IAV titers and shortened the viral shedding but did not completely block direct contact transmission of the influenza virus. Notably, vaccination significantly ameliorated influenza-associated disease by protecting vaccinated animals from severe morbidity after IAV single infection or IAV and SARS-CoV-2 coinfection, suggesting that seasonal influenza virus vaccination is pivotal to prevent severe disease induced by IAV and SARS-CoV-2 coinfection 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 coinfections of both viruses. The development of useful animal models to assess the pathogenesis, transmission, and viral evolution of these viruses as they coinfect a host is of critical importance for the development of vaccines and therapeutics. The ability to prevent the most severe effects of viral coinfections can be studied using effect coinfection ferret models described in this report.


Subject(s)
Antibodies, Viral/blood , COVID-19/prevention & control , Coinfection/prevention & control , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Animals , COVID-19/immunology , Female , Ferrets/immunology , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/immunology , Orthomyxoviridae Infections/immunology , Vaccination , Virus Shedding
13.
Journal of Virology ; 96(3):16, 2022.
Article in English | Web of Science | ID: covidwho-1755903

ABSTRACT

Susceptibility to severe acute respiratory syndrome coronavirus 2 (SARSCoV-2) and the outcome of coronavirus disease 2019 (COVID-19) have been linked to underlying health conditions and the age of affected individuals. Here, we assessed the effect of age on SARS-CoV-2 infection using a ferret model. For this, young (6-monthold) and aged (18- to 39-month-old) ferrets were inoculated intranasally with various doses of SARS-CoV-2. By using infectious virus shedding in respiratory secretions and seroconversion, we estimated that the infectious dose of SARS-CoV-2 in aged animals is similar to 32 PFU per animal, while in young animals it was estimated to be similar to 100 PFU. We showed that viral replication in the upper respiratory tract and shedding in respiratory secretions is enhanced in aged ferrets compared to young animals. Similar to observations in humans, this was associated with higher transcription levels of two key viral entry factors, ACE2 and TMPRSS2, in the upper respiratory tract of aged ferrets. IMPORTANCE In humans, ACE2 and TMPRSS2 are expressed in various cells and tissues, and differential expression has been described in young and old people, with a higher level of expressing cells being detected in the nasal brushing of older people than young individuals. We described the same pattern occurring in ferrets, and we demonstrated that age affects susceptibility of ferrets to SARS-CoV-2. Aged animals were more likely to get infected when exposed to lower infectious dose of the virus than young animals, and the viral replication in the upper respiratory tract and shedding are enhanced in aged ferrets. Together, these results suggest that the higher infectivity and enhanced ability of SARS-CoV-2 to replicate in aged individuals is associated, at least in part, with transcription levels of ACE2 and TMPRSS2 at the sites of virus entry. The young and aged ferret model developed here may represent a great platform to assess age-related differences in SARS-CoV-2 infection dynamics and replication.

14.
mBio ; 11(3)2020 05 22.
Article in English | MEDLINE | ID: covidwho-1723548

ABSTRACT

Due to the urgent need of a therapeutic treatment for coronavirus (CoV) disease 2019 (COVID-19) patients, a number of FDA-approved/repurposed drugs have been suggested as antiviral candidates at clinics, without sufficient information. Furthermore, there have been extensive debates over antiviral candidates for their effectiveness and safety against severe acute respiratory syndrome CoV 2 (SARS-CoV-2), suggesting that rapid preclinical animal studies are required to identify potential antiviral candidates for human trials. To this end, the antiviral efficacies of lopinavir-ritonavir, hydroxychloroquine sulfate, and emtricitabine-tenofovir for SARS-CoV-2 infection were assessed in the ferret infection model. While the lopinavir-ritonavir-, hydroxychloroquine sulfate-, or emtricitabine-tenofovir-treated group exhibited lower overall clinical scores than the phosphate-buffered saline (PBS)-treated control group, the virus titers in nasal washes, stool specimens, and respiratory tissues were similar between all three antiviral-candidate-treated groups and the PBS-treated control group. Only the emtricitabine-tenofovir-treated group showed lower virus titers in nasal washes at 8 days postinfection (dpi) than the PBS-treated control group. To further explore the effect of immune suppression on viral infection and clinical outcome, ferrets were treated with azathioprine, an immunosuppressive drug. Compared to the PBS-treated control group, azathioprine-immunosuppressed ferrets exhibited a longer period of clinical illness, higher virus titers in nasal turbinate, delayed virus clearance, and significantly lower serum neutralization (SN) antibody titers. Taken together, all antiviral drugs tested marginally reduced the overall clinical scores of infected ferrets but did not significantly affect in vivo virus titers. Despite the potential discrepancy of drug efficacies between animals and humans, these preclinical ferret data should be highly informative to future therapeutic treatment of COVID-19 patients.IMPORTANCE The SARS-CoV-2 pandemic continues to spread worldwide, with rapidly increasing numbers of mortalities, placing increasing strain on health care systems. Despite serious public health concerns, no effective vaccines or therapeutics have been approved by regulatory agencies. In this study, we tested the FDA-approved drugs lopinavir-ritonavir, hydroxychloroquine sulfate, and emtricitabine-tenofovir against SARS-CoV-2 infection in a highly susceptible ferret infection model. While most of the drug treatments marginally reduced clinical symptoms, they did not reduce virus titers, with the exception of emtricitabine-tenofovir treatment, which led to diminished virus titers in nasal washes at 8 dpi. Further, the azathioprine-treated immunosuppressed ferrets showed delayed virus clearance and low SN titers, resulting in a prolonged infection. As several FDA-approved or repurposed drugs are being tested as antiviral candidates at clinics without sufficient information, rapid preclinical animal studies should proceed to identify therapeutic drug candidates with strong antiviral potential and high safety prior to a human efficacy trial.


Subject(s)
Antiviral Agents/therapeutic use , Betacoronavirus/drug effects , Coronavirus Infections/drug therapy , Pneumonia, Viral/drug therapy , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Antiviral Agents/pharmacology , Betacoronavirus/immunology , COVID-19 , Coronavirus Infections/virology , Disease Models, Animal , Female , Ferrets , Humans , Hydroxychloroquine/therapeutic use , Pandemics , Pneumonia, Viral/virology , SARS-CoV-2 , United States , United States Food and Drug Administration , Viral Load
15.
Thromb Res ; 210: 6-11, 2022 02.
Article in English | MEDLINE | ID: covidwho-1634248

ABSTRACT

BACKGROUND: Many patients who are diagnosed with coronavirus disease 2019 (COVID-19) suffer from venous thromboembolic complications despite the use of stringent anticoagulant prophylaxis. Studies on the exact mechanism(s) underlying thrombosis in COVID-19 are limited as animal models commonly used to study venous thrombosis pathophysiology (i.e. rats and mice) are naturally not susceptible to Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). Ferrets are susceptible to SARS-CoV-2 infection, successfully used to study virus transmission, and have been previously used to study activation of coagulation and thrombosis during influenza virus infection. OBJECTIVES: This study aimed to explore the use of (heat-inactivated) plasma and lung material from SARS-CoV-2-inoculated ferrets studying COVID-19-associated changes in coagulation and thrombosis. MATERIAL AND METHODS: Histology and longitudinal plasma profiling using mass spectrometry-based proteomics approach was performed. RESULTS: Lungs of ferrets inoculated intranasally with SARS-CoV-2 demonstrated alveolar septa that were mildly expanded by macrophages, and diffuse interstitial histiocytic pneumonia. However, no macroscopical or microscopical evidence of vascular thrombosis in the lungs of SARS-CoV-2-inoculated ferrets was found. Longitudinal plasma profiling revealed minor differences in plasma protein profiles in SARS-CoV-2-inoculated ferrets up to 2 weeks post-infection. The majority of plasma coagulation factors were stable and demonstrated a low coefficient of variation. CONCLUSIONS: We conclude that while ferrets are an essential and well-suited animal model to study SARS-CoV-2 transmission, their use to study SARS-CoV-2-related changes relevant to thrombotic disease is limited.


Subject(s)
COVID-19 , Thrombosis , Venous Thrombosis , Animals , Blood Proteins , Ferrets , Humans , Lung , Mice , Rats , SARS-CoV-2
16.
J Virol ; 96(3): e0145521, 2022 02 09.
Article in English | MEDLINE | ID: covidwho-1532961

ABSTRACT

Susceptibility to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the outcome of coronavirus disease 2019 (COVID-19) have been linked to underlying health conditions and the age of affected individuals. Here, we assessed the effect of age on SARS-CoV-2 infection using a ferret model. For this, young (6-month-old) and aged (18- to 39-month-old) ferrets were inoculated intranasally with various doses of SARS-CoV-2. By using infectious virus shedding in respiratory secretions and seroconversion, we estimated that the infectious dose of SARS-CoV-2 in aged animals is ∼32 PFU per animal, while in young animals it was estimated to be ∼100 PFU. We showed that viral replication in the upper respiratory tract and shedding in respiratory secretions is enhanced in aged ferrets compared to young animals. Similar to observations in humans, this was associated with higher transcription levels of two key viral entry factors, ACE2 and TMPRSS2, in the upper respiratory tract of aged ferrets. IMPORTANCE In humans, ACE2 and TMPRSS2 are expressed in various cells and tissues, and differential expression has been described in young and old people, with a higher level of expressing cells being detected in the nasal brushing of older people than young individuals. We described the same pattern occurring in ferrets, and we demonstrated that age affects susceptibility of ferrets to SARS-CoV-2. Aged animals were more likely to get infected when exposed to lower infectious dose of the virus than young animals, and the viral replication in the upper respiratory tract and shedding are enhanced in aged ferrets. Together, these results suggest that the higher infectivity and enhanced ability of SARS-CoV-2 to replicate in aged individuals is associated, at least in part, with transcription levels of ACE2 and TMPRSS2 at the sites of virus entry. The young and aged ferret model developed here may represent a great platform to assess age-related differences in SARS-CoV-2 infection dynamics and replication.


Subject(s)
COVID-19/virology , Disease Susceptibility , Host-Pathogen Interactions , SARS-CoV-2/physiology , Age Factors , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/metabolism , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Biomarkers , COVID-19/genetics , COVID-19/immunology , Disease Models, Animal , Ferrets , Gene Expression , Host-Pathogen Interactions/immunology , Organ Specificity , RNA, Viral , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Viral Load
17.
Viruses ; 13(8)2021 08 18.
Article in English | MEDLINE | ID: covidwho-1360824

ABSTRACT

The pre-clinical development of antiviral agents involves experimental trials in animals and ferrets as an animal model for the study of SARS-CoV-2. Here, we used mathematical models and experimental data to characterize the within-host infection dynamics of SARS-CoV-2 in ferrets. We also performed a global sensitivity analysis of model parameters impacting the characteristics of the viral infection. We provide estimates of the viral dynamic parameters in ferrets, such as the infection rate, the virus production rate, the infectious virus proportion, the infected cell death rate, the virus clearance rate, as well as other related characteristics, including the basic reproduction number, pre-peak infectious viral growth rate, post-peak infectious viral decay rate, pre-peak infectious viral doubling time, post-peak infectious virus half-life, and the target cell loss in the respiratory tract. These parameters and indices are not significantly different between animals infected with viral strains isolated from the environment and isolated from human hosts, indicating a potential for transmission from fomites. While the infection period in ferrets is relatively short, the similarity observed between our results and previous results in humans supports that ferrets can be an appropriate animal model for SARS-CoV-2 dynamics-related studies, and our estimates provide helpful information for such studies.


Subject(s)
COVID-19/virology , Disease Models, Animal , Ferrets , SARS-CoV-2/physiology , Animals , Basic Reproduction Number , COVID-19/immunology , COVID-19/pathology , COVID-19/transmission , Cell Death , Humans , Immunity, Innate , Models, Biological , Respiratory System/pathology , Respiratory System/virology , SARS-CoV-2/immunology , Sensitivity and Specificity , Viral Load , Virus Shedding
18.
J Virol ; 95(14): e0011121, 2021 06 24.
Article in English | MEDLINE | ID: covidwho-1358015

ABSTRACT

The current fears of a future influenza pandemic have resulted in an increased emphasis on the development and testing of novel therapeutic strategies against the virus. Fundamental to this is the ferret model of influenza infection, which is critical in examining pathogenesis and treatment. Nevertheless, a precise evaluation of the efficacy of any treatment strategy in ferrets is reliant on understanding the immune response in this model. Interferon-inducible transmembrane proteins (IFITMs) are interferon-stimulated proteins shown to be critically important in the host immune response against viral infections. These proteins confer intrinsic innate immunity to pH-dependent viruses such as influenza viruses and can inhibit cytosolic entry of such viruses to limit the severity of infection following interferon upregulation. Mutations in IFITM genes in humans have been identified as key risk factors for worsened disease progression, particularly in the case of avian influenza viruses such as H7N9. While the IFITM genes of humans and mice have been well characterized, no studies have been conducted to classify the IFITM locus and interferon-driven upregulation of IFITMs in ferrets. Here, we show the architecture of the ferret IFITM locus and its synteny to the IFITM locus of other mammalian and avian species. Furthermore, we show that ferret IFITM1, -2, and -3 are functionally responsive to both interferon-α (IFN-α) and influenza virus stimulation. Thus, we show that ferret IFITMs exhibit interferon-stimulated properties similar to those shown in other species, furthering our knowledge of the innate immune response in the ferret model of human influenza virus infections. IMPORTANCE IFITM proteins can prevent the entry of several pH-dependent viruses, including high-consequence viruses such as HIV, influenza viruses, and SARS-coronaviruses. Mutations in these genes have been associated with worsened disease outcomes with mutations in their IFITM genes, highlighting these genes as potential disease risk factors. Ferrets provide a valuable tool to model infectious diseases; however, there is a critical shortage of information regarding their interferon-stimulated genes. We identified the putative ferret IFITM genes and mapped their complete gene locus. Thus, our study fills a critical gap in knowledge and supports the further use of the ferret model to explore the importance of IFITMs in these important diseases.


Subject(s)
Ferrets , Influenza A Virus, H1N1 Subtype , Interferon-alpha/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Orthomyxoviridae Infections/immunology , Animals , Cell Line , Conserved Sequence , Disease Models, Animal , Ferrets/immunology , Ferrets/metabolism , Ferrets/virology , Humans , Models, Molecular , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/metabolism , Polymerase Chain Reaction , Sequence Analysis, Protein , Up-Regulation
19.
ILAR J ; 62(1-2): 232-237, 2021 12 31.
Article in English | MEDLINE | ID: covidwho-1280105

ABSTRACT

This case report discusses Type I hypersensitivity in ferrets following exposure to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) inoculum, observed during a study investigating the efficacy of candidate COVID-19 vaccines. Following a comprehensive internal root-cause investigation, it was hypothesized that prior prime-boost immunization of ferrets with a commercial canine C3 vaccine to protect against the canine distemper virus had resulted in primary immune response to fetal bovine serum (FBS) in the C3 preparation. Upon intranasal exposure to SARS-CoV-2 virus cultured in medium containing FBS, an allergic airway response occurred in 6 out of 56 of the ferrets. The 6 impacted ferrets were randomly dispersed across study groups, including different COVID-19 vaccine candidates, routes of vaccine candidate administration, and controls (placebo). The root-cause investigation and subsequent analysis determined that the allergic reaction was unrelated to the COVID-19 vaccine candidates under evaluation. Histological assessment suggested that the allergic response was characterized by eosinophilic airway disease; increased serum immunoglobulin levels reactive to FBS further suggested this response was caused by immune priming to FBS present in the C3 vaccine. This was further supported by in vivo studies demonstrating ferrets administered diluted FBS also presented clinical signs consistent with a hyperallergic response, while clinical signs were absent in ferrets that received a serum-free SARS-CoV-2 inoculum. It is therefore recommended that vaccine studies in higher order animals should consider the impact of welfare vaccination and use serum-free inoculum whenever possible.


Subject(s)
COVID-19 , Hypersensitivity, Immediate , Viral Vaccines , Animals , COVID-19 Vaccines , Dogs , Ferrets , SARS-CoV-2
20.
Emerg Infect Dis ; 27(7): 1994-1996, 2021 07.
Article in English | MEDLINE | ID: covidwho-1278361

ABSTRACT

We found severe acute respiratory syndrome coronavirus 2 RNA in 6 (8.4%) of 71 ferrets in central Spain and isolated and sequenced virus from 1 oral and 1 rectal swab specimen. Natural infection occurs in kept ferrets when virus circulation among humans is high. However, small ferret collections probably cannot maintain virus circulation.


Subject(s)
COVID-19 , Ferrets , Animals , Humans , SARS-CoV-2 , Spain/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL