Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Front Cell Neurosci ; 17: 1199518, 2023.
Article in English | MEDLINE | ID: covidwho-2301891
2.
Zool Res ; 44(2): 323-330, 2023 Mar 18.
Article in English | MEDLINE | ID: covidwho-2288820

ABSTRACT

The rapid spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in recent years not only caused a global pandemic but resulted in enormous social, economic, and health burdens worldwide. Despite considerable efforts to combat coronavirus disease 2019 (COVID-19), various SARS-CoV-2 variants have emerged, and their underlying mechanisms of pathogenicity remain largely unknown. Furthermore, effective therapeutic drugs are still under development. Thus, an ideal animal model is crucial for studying the pathogenesis of COVID-19 and for the preclinical evaluation of vaccines and antivirals against SARS-CoV-2 and variant infections. Currently, several animal models, including mice, hamsters, ferrets, and non-human primates (NHPs), have been established to study COVID-19. Among them, ferrets are naturally susceptible to SARS-CoV-2 infection and are considered suitable for COVID-19 study. Here, we summarize recent developments and application of SARS-CoV-2 ferret models in studies on pathogenesis, therapeutic agents, and vaccines, and provide a perspective on the role of these models in preventing COVID-19 spread.


Subject(s)
COVID-19 , Rodent Diseases , Cricetinae , Animals , Mice , SARS-CoV-2 , COVID-19/veterinary , Ferrets , Peptidyl-Dipeptidase A
3.
Acta Vet Scand ; 65(1): 9, 2023 Feb 28.
Article in English | MEDLINE | ID: covidwho-2265151

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was first identified in China by the end of 2019 and was responsible for a pandemic in the human population that resulted in millions of deaths worldwide. Since the beginning of the pandemic, the role of animals as spill-over or reservoir hosts was discussed. In addition to cats and dogs, ferrets are becoming increasingly popular as companion animals. Under experimental conditions, ferrets are susceptible to SARS-CoV-2 and it appears that they can also be infected through contact with a SARS-CoV-2 positive owner. However, there is still little information available regarding these natural infections. Here, we serologically tested samples collected from pet ferrets (n = 45) from Poland between June and September 2021. Of the ferrets that were included in the study, 29% (13/45) had contact with owners with confirmed SARS-CoV-2 infections. Nevertheless, SARS-CoV-2-specific antibodies could not be detected in any of the animals, independent of the infection status of the owner. The obtained results suggest that ferrets cannot be readily infected with SARS-CoV-2 under natural conditions, even after prolonged contact with infected humans. However, due to the rapid mutation rate of this virus, it is important to include ferrets in future monitoring studies.


Subject(s)
COVID-19 , Cat Diseases , Dog Diseases , Humans , Animals , Cats , Dogs , SARS-CoV-2 , Poland/epidemiology , COVID-19/veterinary , Cities , Ferrets , Antibodies, Viral , COVID-19 Testing/veterinary
4.
Oxf Open Immunol ; 3(1): iqac004, 2022.
Article in English | MEDLINE | ID: covidwho-2271603

ABSTRACT

COVID-19 has demonstrated the power of RNA vaccines as part of a pandemic response toolkit. Another virus with pandemic potential is influenza. Further development of RNA vaccines in advance of a future influenza pandemic will save time and lives. As RNA vaccines require formulation to enter cells and induce antigen expression, the aim of this study was to investigate the impact of a recently developed bioreducible cationic polymer, pABOL for the delivery of a self-amplifying RNA (saRNA) vaccine for seasonal influenza virus in mice and ferrets. Mice and ferrets were immunized with pABOL formulated saRNA vaccines expressing either haemagglutinin (HA) from H1N1 or H3N2 influenza virus in a prime boost regime. Antibody responses, both binding and functional were measured in serum after immunization. Animals were then challenged with a matched influenza virus either directly by intranasal inoculation or in a contact transmission model. While highly immunogenic in mice, pABOL-formulated saRNA led to variable responses in ferrets. Animals that responded to the vaccine with higher levels of influenza virus-specific neutralizing antibodies were more protected against influenza virus infection. pABOL-formulated saRNA is immunogenic in ferrets, but further optimization of RNA vaccine formulation and constructs is required to increase the quality and quantity of the antibody response to the vaccine.

5.
Microbiol Spectr ; : e0138622, 2022 Oct 27.
Article in English | MEDLINE | ID: covidwho-2256572

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.

6.
Emerging Animal Species ; : 100023.0, 2023.
Article in English | ScienceDirect | ID: covidwho-2234251

ABSTRACT

Ferrets are an emergent companion animal species, and a possible animal model for conditions such as COVID-19 infection. As such, they are more present in veterinary practice than in the past. This study aimed at assessing how ferrets' electrocardiographic parameters varied according to the age of the animal during isoflurane anesthesia. Eighteen ferrets, divided into three age categories (3-6 months: 7 ferrets;32-36 months: 4 ferrets;>55 months: 7 ferrets), were included in the study. Generalized Estimation Equations, with Bonferroni corrected pairwise comparisons for age categories, were run. Most of the measured electrocardiographic parameters varied depending on age (all p < 0.05), although their trend across the three age groups was not the same for all the parameters. S wave amplitude and P wave duration varied according to sex. Findings of the present study can be useful for veterinary practitioners dealing with ferrets.

7.
J Gen Virol ; 103(11)2022 11.
Article in English | MEDLINE | ID: covidwho-2231373

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19) in humans, has a wide host range, naturally infecting felids, canids, cervids, rodents and mustelids. Transmission of SARS-CoV-2 is universally accepted to occur via contact with contaminated secretions from the respiratory epithelium, either directly or indirectly. Transmission via droplet nuclei, generated from a cough or sneeze, has also been reported in several human and experimental animal scenarios. However, the role of droplet transmission at the human-animal interface remains to be fully elucidated. Here, the ferret infection model was used to investigate the routes of infection for the SARS-CoV-2 beta variant (B.1.351). Ferrets were exposed to droplets containing infectious SARS-CoV-2, ranging between 4 and 106 µm in diameter, simulating larger droplets produced by a cough from an infected person. Following exposure, viral RNA was detected on the fur of ferrets, and was deposited onto environmental surfaces, as well as the fur of ferrets placed in direct contact; SARS-CoV-2 remained infectious on the fur for at least 48 h. Low levels of viral RNA were detected in the nasal washes early post-exposure, yet none of the directly exposed, or direct-contact ferrets, became robustly infected or seroconverted to SARS-CoV-2. In comparison, ferrets intranasally inoculated with the SARS-CoV-2 beta variant became robustly infected, shedding viral RNA and infectious virus from the nasal cavity, with transmission to 75 % of naive ferrets placed in direct contact. These data suggest that larger infectious droplet nuclei and contaminated fur play minor roles in SARS-CoV-2 transmission among mustelids and potentially other companion animals.


Subject(s)
COVID-19 , Animals , Humans , SARS-CoV-2 , Ferrets , Cough , Aerosolized Particles and Droplets , RNA, Viral/genetics
8.
Springer Protocols Handbooks ; : 171-188, 2022.
Article in English | EMBASE | ID: covidwho-2173512

ABSTRACT

Ferret coronavirus is associated with two disease presentations in ferrets, namely, epizootic catarrhal enteritis and a feline infectious peritonitis (FIP)-like systemic disease. In this chapter, we describe conventional and real-time one-step reverse transcription polymerase chain reaction assays that are routinely used in our laboratory to detect either genotype 1 or genotype 2 ferret coronavirus in clinical specimens. These assays were designed based upon the conserved spike gene sequence difference found between three strains of ferret systemic coronavirus and three strains of ferret enteric coronavirus. Recent literature evidence indicates that pathotype is not associated with a specific genotype, and therefore, it is important to test for both genotypes either in enteric or systemic disease. Copyright © 2016 Springer Science+Business Media New York.

9.
Microorganisms ; 11(1)2023 Jan 11.
Article in English | MEDLINE | ID: covidwho-2200532

ABSTRACT

In this study, we investigated the features of the infectious process by simulating co-infection with SARS-CoV-2 and human adenovirus type 5 (HAdV-5) or influenza A virus (IAV) in vitro and in vivo. The determination of infectious activity of viruses and digital PCR demonstrated that during simultaneous and sequential HAdV-5 followed by SARS-CoV-2 infection in vitro and in vivo, the HAdV-5 infection does not interfere with replication of SARS-CoV-2. The hamsters co-infected and mono-infected with SARS-CoV-2 exhibited nearly identical viral titers and viral loads of SARS-CoV-2 in the lungs. The hamsters and ferrets co-infected by SARS-CoV-2- and IAV demonstrated more pronounced clinical manifestations than mono-infected animals. Additionally, the lung histological data illustrate that HAdV-5 or IAV and SARS-CoV-2 co-infection induces more severe pathological changes in the lungs than mono-infection. The expression of several genes specific to interferon and cytokine signaling pathways in the lungs of co-infected hamsters was more upregulated compared to single infected with SARS-CoV-2 animals. Thus, co-infection with HAdV-5 or IAV and SARS-CoV-2 leads to more severe pulmonary disease in animals.

10.
Metabolites ; 12(11)2022 Nov 21.
Article in English | MEDLINE | ID: covidwho-2116091

ABSTRACT

The global threat of COVID-19 has led to an increased use of metabolomics to study SARS-CoV-2 infections in animals and humans. In spite of these efforts, however, understanding the metabolome of SARS-CoV-2 during an infection remains difficult and incomplete. In this study, metabolic responses to a SAS-CoV-2 challenge experiment were studied in nasal washes collected from an asymptomatic ferret model (n = 20) at different time points before and after infection using an LC-MS-based metabolomics approach. A multivariate analysis of the nasal wash metabolome data revealed several statistically significant features. Despite no effects of sex or interaction between sex and time on the time course of SARS-CoV-2 infection, 16 metabolites were significantly different at all time points post-infection. Among these altered metabolites, the relative abundance of taurine was elevated post-infection, which could be an indication of hepatotoxicity, while the accumulation of sialic acids could indicate SARS-CoV-2 invasion. Enrichment analysis identified several pathways influenced by SARS-CoV-2 infection. Of these, sugar, glycan, and amino acid metabolisms were the key altered pathways in the upper respiratory channel during infection. These findings provide some new insights into the progression of SARS-CoV-2 infection in ferrets at the metabolic level, which could be useful for the development of early clinical diagnosis tools and new or repurposed drug therapies.

11.
Pathogenic Coronaviruses of Humans and Animals ; : 275-340, 2023.
Article in English | ScienceDirect | ID: covidwho-2082391

ABSTRACT

Coronaviruses are present in most animal species. Some animals may then serve as a reservoir or intermediate hosts of viruses causing mild or severe to fatal diseases in humans and other animals. Infected humans may also transmit coronaviruses, such as severe acute respiratory syndrome virus (SARS-CoV)-2, to animals, including captive endangered animal species. This chapter focuses on coronaviruses of wild and semidomesticated animals, including viruses from bats, rodents, nonhuman primates, ferrets, minks, and rabbits. The ability of coronaviruses to rapidly mutate and to exchange their genetic material with other coronaviruses leads to the production of variants able to infect and adapt to new host species. Special attention is given to coronaviruses of bats and rodents since they appear to have hosted ancestral coronaviruses that indirectly lead to zoonotic transmission of highly pathogenic human viruses, including SARS-CoV, the closely related SARS-CoV-2, and Middle East respiratory syndrome virus. The RNA genomes of several bat coronaviruses, such as WIV1 and WIV16, are very similar to SARS-CoV. Coronaviruses in animals primarily cause severe disease in the respiratory, central nervous, and digestive systems but may damage other organ systems as well. Further studies on wildlife coronaviruses are advisable to avoid human epidemics or pandemics as well as to protect endangered animal species.

12.
mBio ; 13(5): e0242122, 2022 10 26.
Article in English | MEDLINE | ID: covidwho-2038244

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. IMPORTANCE Continued surveillance and risk assessment of emerging SARS-CoV-2 variants are critical for pandemic response and preparedness. As such, in vivo evaluations are indispensable for early detection of variants with enhanced virulence and transmission. Here, we used the ferret model to compare the pathogenicity and transmissibility of an original SARS-CoV-2 isolate (USA-WA1/2020 [WA1]) to those of a panel of Alpha, Beta, and Delta variants, as well as to evaluate protection from homologous and heterologous reinfection. We observed strain-specific differences in replication kinetics in the ferret respiratory tract and virus load emitted into the air, revealing enhanced transmissibility of the Delta virus relative to previously detected strains. Prior infection with SARS-CoV-2 provided varied levels of protection from reinfection, with the Beta strain eliciting the lowest level of protection. Overall, we found that ferrets represent a useful model for comparative assessments of SARS-CoV-2 infection, transmission, and reinfection.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Humans , Ferrets , Reinfection , RNA, Viral/genetics , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus
13.
Front Cell Infect Microbiol ; 12: 873416, 2022.
Article in English | MEDLINE | ID: covidwho-2009846

ABSTRACT

Even with the COVID-19 pandemic, tuberculosis remains a leading cause of human death due to a single infectious agent. Until successfully treated, infected individuals may continue to transmit Mycobacterium tuberculosis bacilli to contacts. As with other respiratory pathogens, such as SARS-CoV-2, modeling the process of person-to-person transmission will inform efforts to develop vaccines and therapies that specifically impede disease transmission. The ferret (Mustela furo), a relatively inexpensive, small animal has been successfully employed to model transmissibility, pathogenicity, and tropism of influenza and other respiratory disease agents. Ferrets can become naturally infected with Mycobacterium bovis and are closely related to badgers, well known in Great Britain and elsewhere as a natural transmission vehicle for bovine tuberculosis. Herein, we report results of a study demonstrating that within 7 weeks of intratracheal infection with a high dose (>5 x 103 CFU) of M. tuberculosis bacilli, ferrets develop clinical signs and pathological features similar to acute disease reported in larger animals, and ferrets infected with very-high doses (>5 x 104 CFU) develop severe signs within two to four weeks, with loss of body weight as high as 30%. Natural transmission of this pathogen was also examined. Acutely-infected ferrets transmitted M. tuberculosis bacilli to co-housed naïve sentinels; most of the sentinels tested positive for M. tuberculosis in nasal washes, while several developed variable disease symptomologies similar to those reported for humans exposed to an active tuberculosis patient in a closed setting. Transmission was more efficient when the transmitting animal had a well-established acute infection. The findings support further assessment of this model system for tuberculosis transmission including the testing of prevention measures and vaccine efficacy.


Subject(s)
COVID-19 , Tuberculosis , Animals , Disease Models, Animal , Ferrets , Humans , Pandemics , SARS-CoV-2
14.
Settimana Veterinaria ; : 35-36, 2021.
Article in Italian | CAB Abstracts | ID: covidwho-1837569
15.
Monoclon Antib Immunodiagn Immunother ; 41(2): 101-109, 2022 Apr.
Article in English | MEDLINE | ID: covidwho-1806236

ABSTRACT

Ferrets (Mustela putorius furo) have been used as small animal models to investigate severe acute respiratory syndrome coronaviruses (SARS-CoV and SARS-CoV-2) infections. Pathological analyses of these tissue samples, including those of the lung, are, therefore, essential to understand the pathogenesis of SARS-CoVs and evaluate the action of therapeutic monoclonal antibodies (mAbs) against this disease. However, mAbs that recognize ferret-derived proteins and distinguish between specific cell types, such as lung epithelial cells, are limited. Podoplanin (PDPN) has been identified as an essential marker in lung type I alveolar epithelial cells, kidney podocytes, and lymphatic endothelial cells. In this study, an anti-ferret PDPN (ferPDPN) mAb PMab-292 (mouse IgG1, kappa) was established using the Cell-Based Immunization and Screening (CBIS) method. PMab-292 recognized ferPDPN-overexpressed Chinese hamster ovary-K1 (CHO/ferPDPN) cells by flow cytometry and Western blotting. The kinetic analysis using flow cytometry showed that the KD of PMab-292 for CHO/ferPDPN was 3.4 × 10-8 M. Furthermore, PMab-292 detected lung type I alveolar epithelial cells, lymphatic endothelial cells, and glomerular/Bowman's capsule in the kidney using immunohistochemistry. Hence, these results propose the usefulness of PMab-292 in analyzing ferret-derived tissues for SARS-CoV-2 research.


Subject(s)
Antineoplastic Agents, Immunological , COVID-19 , Severe acute respiratory syndrome-related coronavirus , Animals , Antibodies, Monoclonal , Antibody Specificity , CHO Cells , Cricetinae , Cricetulus , Endothelial Cells , Epitope Mapping/methods , Ferrets , Kinetics , Membrane Glycoproteins/genetics , Mice , SARS-CoV-2 , Transcription Factors
16.
J Virol ; 96(7): e0010022, 2022 04 13.
Article in English | MEDLINE | ID: covidwho-1728835

ABSTRACT

Understanding how animal influenza A viruses (IAVs) acquire airborne transmissibility in humans and ferrets is needed to prepare for and respond to pandemics. Here, we investigated in ferrets the replication and transmission of swine H1N1 isolates P4 and G15, whose majority population had decreased polymerase activity and poor hemagglutinin (HA) stability, respectively. For both isolates, a minor variant was selected and transmitted in ferrets. Polymerase-enhancing variant PA-S321 airborne-transmitted and propagated in one ferret. HA-stabilizing variant HA1-S210 was selected in all G15-inoculated ferrets and was transmitted by contact and airborne routes. With an efficient polymerase and a stable HA, the purified minor variant G15-HA1-S210 had earlier and higher peak titers in inoculated ferrets and was recovered at a higher frequency after airborne transmission than P4 and G15. Overall, HA stabilization played a more prominent role than polymerase enhancement in the replication and transmission of these viruses in ferrets. The results suggest pandemic risk-assessment studies may benefit from deep sequencing to identify minor variants with human-adapted traits. IMPORTANCE Diverse IAVs circulate in animals, yet few acquire the viral traits needed to start a human pandemic. A stabilized HA and mammalian-adapted polymerase have been shown to promote the adaptation of IAVs to humans and ferrets (the gold-standard model for IAV replication, pathogenicity, and transmissibility). Here, we used swine IAV isolates of the gamma lineage as a model to investigate the importance of HA stability and polymerase activity in promoting replication and transmission in ferrets. These are emerging viruses that bind to both α-2,6- and α-2,3-linked receptors. Using isolates containing mixed populations, a stabilized HA was selected within days in inoculated ferrets. An enhanced polymerase was also selected and propagated after airborne transmission to a ferret. Thus, HA stabilization was a stricter requirement, yet both traits promoted transmissibility. Knowing the viral traits needed for pandemic potential, and the relative importance of each, will help identify emerging viruses of greatest concern.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus , Influenza A Virus, H1N1 Subtype , Orthomyxoviridae Infections , Animals , Ferrets , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Humans , Orthomyxoviridae Infections/transmission , Orthomyxoviridae Infections/virology , Protein Stability , Swine
17.
J Infect Dis ; 225(3): 404-412, 2022 02 01.
Article in English | MEDLINE | ID: covidwho-1672208

ABSTRACT

Cocirculation of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and influenza viruses could pose unpredictable risks to health systems globally, with recent studies suggesting more severe disease outcomes in coinfected patients. The initial lack of a readily available coronavirus disease 2019 (COVID-19) vaccine has reinforced the importance of influenza vaccine programs during the COVID-19 pandemic. Live attenuated influenza vaccine (LAIV) is an important tool in protecting against influenza, particularly in children. However, it is unknown whether LAIV administration influences the outcomes of acute SARS-CoV-2 infection or disease. To investigate this, quadrivalent LAIV was administered to ferrets 3 days before or after SARS-CoV-2 infection. LAIV administration did not exacerbate the SARS-CoV-2 disease course or lung pathology with either regimen. In addition, LAIV administered before SARS-CoV-2 infection significantly reduced SARS-CoV-2 replication and shedding in the upper respiratory tract. This study demonstrated that LAIV administration in close proximity to SARS-CoV-2 infection does not exacerbate mild disease and can reduce SARS-CoV-2 shedding.


Subject(s)
COVID-19 , Influenza Vaccines , Virus Shedding , Animals , COVID-19/therapy , Disease Models, Animal , Ferrets , Influenza Vaccines/therapeutic use , Lung , Respiratory System/virology , SARS-CoV-2/physiology , Vaccines, Attenuated/therapeutic use , Virus Replication
18.
Cell Rep ; 38(6): 110344, 2022 02 08.
Article in English | MEDLINE | ID: covidwho-1639571

ABSTRACT

SARS-CoV-2 has a broad mammalian species tropism infecting humans, cats, dogs, and farmed mink. Since the start of the 2019 pandemic, several reverse zoonotic outbreaks of SARS-CoV-2 have occurred in mink, one of which reinfected humans and caused a cluster of infections in Denmark. Here we investigate the molecular basis of mink and ferret adaptation and demonstrate the spike mutations Y453F, F486L, and N501T all specifically adapt SARS-CoV-2 to use mustelid ACE2. Furthermore, we risk assess these mutations and conclude mink-adapted viruses are unlikely to pose an increased threat to humans, as Y453F attenuates the virus replication in human cells and all three mink adaptations have minimal antigenic impact. Finally, we show that certain SARS-CoV-2 variants emerging from circulation in humans may naturally have a greater propensity to infect mustelid hosts and therefore these species should continue to be surveyed for reverse zoonotic infections.


Subject(s)
Adaptation, Biological/immunology , SARS-CoV-2/genetics , Viral Zoonoses/genetics , Animals , COVID-19 , Ferrets/immunology , Genetic Fitness/genetics , Humans , Mink/immunology , Mutation , Pandemics , Respiratory System/virology , SARS-CoV-2/pathogenicity , Spike Glycoprotein, Coronavirus/immunology
19.
Vet Pathol ; 59(4): 661-672, 2022 07.
Article in English | MEDLINE | ID: covidwho-1613175

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.


Subject(s)
COVID-19 , Rodent Diseases , Animals , Antigens, Viral , COVID-19/veterinary , Cricetinae , Disease Models, Animal , Ferrets , Respiratory Mucosa , SARS-CoV-2
20.
J Microbiol ; 59(11): 1056-1062, 2021 Nov.
Article in English | MEDLINE | ID: covidwho-1453895

ABSTRACT

The COVID-19 pandemic has caused unprecedented health, social, and economic crises worldwide. However, to date, there is an only a limited effective treatment for this disease. Human placenta hydrolysate (hPH) has previously been shown to be safe and to improve the health condition in patients with hyperferritinemia and COVID-19. In this study, we aimed to determine the antiviral effects of hPH against SARS-CoV-2 in vitro and in vivo models and compared with Remdesivir, an FDA-approved drug for COVID-19 treatment. To assess whether hPH inhibited SARS-CoV-2 replication, we determined the CC50, EC50, and selective index (SI) in Vero cells by infection with a SARS-CoV-2 at an MOI of 0.01. Further, groups of ferrets infected with 105.8 TCID50/ml of SARS-CoV-2 and treated with hPH at 2, 4, 6 dpi, and compared their clinical manifestation and virus titers in respiratory tracts with PBS control-treated group. The mRNA expression of immune-related cytokines was determined by qRT-PCR. hPH treatment attenuated virus replication in a dose-dependent manner in vitro. In a ferret infection study, treatment with hPH resulted in minimal bodyweight loss and attenuated virus replication in the nasal wash, turbinates, and lungs of infected ferrets. In addition, qRT-PCR results revealed that the hPH treatment remarkably upregulated the gene expression of type I (IFN-α and IFN-ß) and II (IFN-γ) IFNs in SARS-CoV-2 infected ferrets. Our data collectively suggest that hPH has antiviral efficacy against SARS-CoV-2 and might be a promising therapeutic agent for the treatment of SARS-CoV-2 infection.


Subject(s)
Adenosine Monophosphate/analogs & derivatives , Alanine/analogs & derivatives , Antiviral Agents/therapeutic use , COVID-19 Drug Treatment , Placenta/chemistry , Protein Hydrolysates , SARS-CoV-2/drug effects , Adenosine Monophosphate/pharmacology , Adenosine Monophosphate/therapeutic use , Alanine/pharmacology , Alanine/therapeutic use , Animals , Chlorocebus aethiops , Female , Ferrets , Humans , Male , Pregnancy , Protein Hydrolysates/pharmacology , Protein Hydrolysates/therapeutic use , Vero Cells , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL