Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
J Virol ; 96(17): e0006522, 2022 09 14.
Article in English | MEDLINE | ID: covidwho-2001768

ABSTRACT

Swine acute diarrhea syndrome coronavirus (SADS-CoV) is a recently emerging bat-borne coronavirus responsible for high mortality rates in piglets. In vitro studies have indicated that SADS-CoV has a wide tissue tropism in different hosts, including humans. However, whether this virus potentially threatens other animals remains unclear. Here, we report the experimental infection of wild-type BALB/c and C57BL/6J suckling mice with SADS-CoV. We found that mice less than 7 days old are susceptible to the virus, which caused notable multitissue infections and damage. The mortality rate was the highest in 2-day-old mice and decreased in older mice. Moreover, a preliminary neuroinflammatory response was observed in 7-day-old SADS-CoV-infected mice. Thus, our results indicate that SADS-CoV has potential pathogenicity in young hosts. IMPORTANCE SADS-CoV, which likely has originated from bat coronaviruses, is highly pathogenic to piglets and poses a threat to the swine industry. Little is known about its potential to disseminate to other animals. No efficient treatment is available, and the quarantine strategy is the only preventive measure. In this study, we demonstrated that SADS-CoV can efficiently replicate in suckling mice younger than 7 days. In contrast to infected piglets, in which intestinal tropism is shown, SADS-CoV caused infection and damage in all murine tissues evaluated in this study. In addition, neuroinflammatory responses were detected in some of the infected mice. Our work provides a preliminary cost-effective model for the screening of antiviral drugs against SADS-CoV infection.


Subject(s)
Alphacoronavirus , Coronavirus Infections , Diarrhea , Mice , Swine Diseases , Alphacoronavirus/pathogenicity , Animals , Chiroptera/virology , Coronavirus Infections/complications , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Diarrhea/complications , Diarrhea/veterinary , Diarrhea/virology , Humans , Mice/virology , Mice, Inbred BALB C , Mice, Inbred C57BL , Neuroinflammatory Diseases/complications , Neuroinflammatory Diseases/veterinary , Neuroinflammatory Diseases/virology , Swine/virology , Swine Diseases/virology
2.
PLoS One ; 17(2): e0263582, 2022.
Article in English | MEDLINE | ID: covidwho-1910522

ABSTRACT

The membrane protein M of the Porcine Epidemic Diarrhea Virus (PEDV) is the most abundant component of the viral envelope. The M protein plays a central role in the morphogenesis and assembly of the virus through protein interactions of the M-M, M-Spike (S) and M-nucleocapsid (N) type. The M protein is known to induce protective antibodies in pigs and to participate in the antagonistic response of the cellular antiviral system coordinated by the type I and type III interferon pathways. The 3D structure of the PEDV M protein is still unknown. The present work exposes a predicted 3D model of the M protein generated using the Robetta protocol. The M protein model is organized into a transmembrane and a globular region. The obtained 3D model of the PEDV M protein was compared with 3D models of the SARS-CoV-2 M protein created using neural networks and with initial machine learning-based models created using trRosetta. The 3D model of the present study predicted four linear B-cell epitopes (RSVNASSGTG and KHGDYSAVSNPSALT peptides are noteworthy), six discontinuous B-cell epitopes, forty weak binding and fourteen strong binding T-cell epitopes in the CV777 M protein. A high degree of conservation of the epitopes predicted in the PEDV M protein was observed among different PEDV strains isolated in different countries. The data suggest that the M protein could be a potential candidate for the development of new treatments or strategies that activate protective cellular mechanisms against viral diseases.


Subject(s)
Coronavirus Infections/virology , Coronavirus M Proteins/chemistry , Porcine epidemic diarrhea virus/chemistry , Swine Diseases/virology , Swine/virology , Amino Acid Sequence , Animals , Coronavirus Infections/immunology , Coronavirus Infections/veterinary , Coronavirus M Proteins/immunology , Epitopes, B-Lymphocyte/chemistry , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/immunology , Models, Molecular , Porcine epidemic diarrhea virus/immunology , Protein Conformation , Swine Diseases/immunology
3.
Nature ; 600(7887): 133-137, 2021 12.
Article in English | MEDLINE | ID: covidwho-1521757

ABSTRACT

Coronaviruses have caused three major epidemics since 2003, including the ongoing SARS-CoV-2 pandemic. In each case, the emergence of coronavirus in our species has been associated with zoonotic transmissions from animal reservoirs1,2, underscoring how prone such pathogens are to spill over and adapt to new species. Among the four recognized genera of the family Coronaviridae, human infections reported so far have been limited to alphacoronaviruses and betacoronaviruses3-5. Here we identify porcine deltacoronavirus strains in plasma samples of three Haitian children with acute undifferentiated febrile illness. Genomic and evolutionary analyses reveal that human infections were the result of at least two independent zoonoses of distinct viral lineages that acquired the same mutational signature in the genes encoding Nsp15 and the spike glycoprotein. In particular, structural analysis predicts that one of the changes in the spike S1 subunit, which contains the receptor-binding domain, may affect the flexibility of the protein and its binding to the host cell receptor. Our findings highlight the potential for evolutionary change and adaptation leading to human infections by coronaviruses outside of the previously recognized human-associated coronavirus groups, particularly in settings where there may be close human-animal contact.


Subject(s)
Coronavirus Infections/epidemiology , Coronavirus Infections/virology , Deltacoronavirus/isolation & purification , Swine/virology , Viral Zoonoses/epidemiology , Viral Zoonoses/virology , Amino Acid Sequence , Animals , Bayes Theorem , Child , Chlorocebus aethiops , Conserved Sequence , Coronavirus Infections/blood , Deltacoronavirus/classification , Deltacoronavirus/genetics , Deltacoronavirus/pathogenicity , Female , Haiti/epidemiology , Humans , Male , Models, Molecular , Mutation , Phylogeny , Vero Cells , Viral Zoonoses/blood
4.
Viruses ; 13(10)2021 10 01.
Article in English | MEDLINE | ID: covidwho-1444334

ABSTRACT

Coronaviruses (CoVs) are a group of enveloped positive-sense RNA viruses and can cause deadly diseases in animals and humans. Cell entry is the first and essential step of successful virus infection and can be divided into two ongoing steps: cell binding and membrane fusion. Over the past two decades, stimulated by the global outbreak of SARS-CoV and pandemic of SARS-CoV-2, numerous efforts have been made in the CoV research. As a result, significant progress has been achieved in our understanding of the cell entry process. Here, we review the current knowledge of this essential process, including the viral and host components involved in cell binding and membrane fusion, molecular mechanisms of their interactions, and the sites of virus entry. We highlight the recent findings of host restriction factors that inhibit CoVs entry. This knowledge not only enhances our understanding of the cell entry process, pathogenesis, tissue tropism, host range, and interspecies-transmission of CoVs but also provides a theoretical basis to design effective preventive and therapeutic strategies to control CoVs infection.


Subject(s)
Coronavirus Infections/pathology , Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/metabolism , Virus Attachment , Virus Internalization , Animals , Cats/virology , Cattle/virology , Chickens/virology , Coronavirus/genetics , Dogs/virology , Livestock/virology , Membrane Fusion/physiology , Receptors, Virus/metabolism , Spike Glycoprotein, Coronavirus/genetics , Swine/virology , Viral Tropism/physiology
5.
Mol Cells ; 44(6): 377-383, 2021 Jun 30.
Article in English | MEDLINE | ID: covidwho-1289259

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.


Subject(s)
COVID-19/virology , Models, Theoretical , Organoids/virology , SARS-CoV-2/pathogenicity , Animals , COVID-19/immunology , COVID-19/pathology , Cats , Cell Line, Tumor , Chickens/virology , Chlorocebus aethiops/virology , Cricetinae , Dogs , Ferrets/virology , Humans , Mice , Organoids/immunology , Organoids/pathology , Rabbits , SARS-CoV-2/growth & development , Swine/virology , Vero Cells
7.
Mol Immunol ; 135: 254-267, 2021 07.
Article in English | MEDLINE | ID: covidwho-1209747

ABSTRACT

By definition no model is perfect, and this also holds for biology and health sciences. In medicine, murine models are, and will be indispensable for long, thanks to their reasonable cost and huge choice of transgenic strains and molecular tools. On the other side, non-human primates remain the best animal models although their use is limited because of financial and obvious ethical reasons. In the field of respiratory diseases, specific clinical models such as sheep and cotton rat for bronchiolitis, or ferret and Syrian hamster for influenza and Covid-19, have been successfully developed, however, in these species, the toolbox for biological analysis remains scarce. In this view the porcine medical model is appearing as the third, intermediate, choice, between murine and primate. Herein we would like to present the pros and cons of pig as a model for acquired respiratory conditions, through an immunological point of view. Indeed, important progresses have been made in pig immunology during the last decade that allowed the precise description of immune molecules and cell phenotypes and functions. These progresses might allow the use of pig as clinical model of human respiratory diseases but also as a species of interest to perform basic research explorations.


Subject(s)
COVID-19/immunology , Disease Models, Animal , SARS-CoV-2/immunology , Swine/immunology , Animals , COVID-19/pathology , COVID-19/therapy , Humans , Swine/virology
8.
Viruses ; 13(1)2020 12 22.
Article in English | MEDLINE | ID: covidwho-1025055

ABSTRACT

Bats are often claimed to be a major source for future viral epidemics, as they are associated with several viruses with zoonotic potential. Here we describe the presence and biodiversity of bats associated with intensive pig farms devoted to the production of heavy pigs in northern Italy. Since chiropters or signs of their presence were not found within animal shelters in our study area, we suggest that fecal viruses with high environmental resistance have the highest likelihood for spillover through indirect transmission. In turn, we investigated the circulation of mammalian orthoreoviruses (MRVs), coronaviruses (CoVs) and astroviruses (AstVs) in pigs and bats sharing the same environment. Results of our preliminary study did not show any bat virus in pigs suggesting that spillover from these animals is rare. However, several AstVs, CoVs and MRVs circulated undetected in pigs. Among those, one MRV was a reassortant strain carrying viral genes likely acquired from bats. On the other hand, we found a swine AstV and a MRV strain carrying swine genes in bat guano, indicating that viral exchange at the bat-pig interface might occur more frequently from pigs to bats rather than the other way around. Considering the indoor farming system as the most common system in the European Union (EU), preventive measures should focus on biosecurity rather than displacement of bats, which are protected throughout the EU and provide critical ecosystem services for rural settings.


Subject(s)
Chiroptera , Swine , Animals , Biodiversity , Chiroptera/virology , DNA Viruses/classification , DNA Viruses/genetics , Ecosystem , Phylogeny , RNA Viruses/classification , RNA Viruses/genetics , Reassortant Viruses/genetics , Swine/virology , Swine Diseases/epidemiology , Swine Diseases/transmission , Swine Diseases/virology , Virus Diseases/veterinary
9.
J Biol Chem ; 296: 100435, 2021.
Article in English | MEDLINE | ID: covidwho-1087033

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic represents a global threat, and the interaction between the virus and angiotensin-converting enzyme 2 (ACE2), the primary entry receptor for SARS-CoV-2, is a key determinant of the range of hosts that can be infected by the virus. However, the mechanisms underpinning ACE2-mediated viral entry across species remains unclear. Using infection assay, we evaluated SARS-CoV-2 entry mediated by ACE2 of 11 different animal species. We discovered that ACE2 of Rhinolophus sinicus (Chinese rufous horseshoe bat), Felis catus (domestic cat), Canis lupus familiaris (dog), Sus scrofa (wild pig), Capra hircus (goat), and Manis javanica (Malayan pangolin) facilitated SARS-CoV-2 entry into nonsusceptible cells. Moreover, ACE2 of the pangolin also mediated SARS-CoV-2 entry, adding credence to the hypothesis that SARS-CoV-2 may have originated from pangolins. However, the ACE2 proteins of Rhinolophus ferrumequinum (greater horseshoe bat), Gallus gallus (red junglefowl), Notechis scutatus (mainland tiger snake), or Mus musculus (house mouse) did not facilitate SARS-CoV-2 entry. In addition, a natural isoform of the ACE2 protein of Macaca mulatta (rhesus monkey) with the Y217N mutation was resistant to SARS-CoV-2 infection, highlighting the possible impact of this ACE2 mutation on SARS-CoV-2 studies in rhesus monkeys. We further demonstrated that the Y217 residue of ACE2 is a critical determinant for the ability of ACE2 to mediate SARS-CoV-2 entry. Overall, these results clarify that SARS-CoV-2 can use the ACE2 receptors of multiple animal species and show that tracking the natural reservoirs and intermediate hosts of SARS-CoV-2 is complex.


Subject(s)
Angiotensin-Converting Enzyme 2/genetics , COVID-19/epidemiology , COVID-19/transmission , Pandemics , SARS-CoV-2/pathogenicity , Spike Glycoprotein, Coronavirus/genetics , Angiotensin-Converting Enzyme 2/chemistry , Angiotensin-Converting Enzyme 2/immunology , Animals , COVID-19/diagnosis , COVID-19/immunology , Cats , Chickens/virology , Chiroptera/virology , Dogs , Elapidae/virology , Eutheria/virology , Gene Expression , Goats/virology , HEK293 Cells , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate , Macaca mulatta/virology , Mice , Models, Molecular , Mutation , Protein Binding , Protein Structure, Secondary , Recombinant Proteins/genetics , Recombinant Proteins/immunology , SARS-CoV-2/genetics , SARS-CoV-2/immunology , Species Specificity , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/immunology , Swine/virology , Virus Internalization
10.
Prev Vet Med ; 188: 105281, 2021 Mar.
Article in English | MEDLINE | ID: covidwho-1051106

ABSTRACT

Pigs (Sus scrofa) may be important surveillance targets for risk assessment and risk-based control planning against emerging zoonoses. Pigs have high contact rates with humans and other animals, transmit similar pathogens as humans including CoVs, and serve as reservoirs and intermediate hosts for notable human pandemics. Wild and domestic pigs both interface with humans and each other but have unique ecologies that demand different surveillance strategies. Three fundamental questions shape any surveillance program: where, when, and how can surveillance be conducted to optimize the surveillance objective? Using theory of mechanisms of zoonotic spillover and data on risk factors, we propose a framework for determining where surveillance might begin initially to maximize a detection in each host species at their interface. We illustrate the utility of the framework using data from the United States. We then discuss variables to consider in refining when and how to conduct surveillance. Recent advances in accounting for opportunistic sampling designs and in translating serology samples into infection times provide promising directions for extracting spatio-temporal estimates of disease risk from typical surveillance data. Such robust estimates of population-level disease risk allow surveillance plans to be updated in space and time based on new information (adaptive surveillance) thus optimizing allocation of surveillance resources to maximize the quality of risk assessment insight.


Subject(s)
Communicable Diseases, Emerging/epidemiology , Coronavirus Infections/epidemiology , Coronavirus Infections/veterinary , Public Health Surveillance/methods , Swine Diseases/epidemiology , Swine Diseases/virology , Zoonoses/epidemiology , Animals , Animals, Wild/virology , Coronavirus/isolation & purification , Disease Reservoirs/virology , Humans , Sus scrofa/virology , Swine/virology , Zoonoses/transmission
11.
PLoS One ; 15(12): e0244025, 2020.
Article in English | MEDLINE | ID: covidwho-992706

ABSTRACT

Coronaviruses such as SARS-CoV-2 regularly infect host tissues that express antiviral proteins (AVPs) in abundance. Understanding how they evolve to adapt or evade host immune responses is important in the effort to control the spread of infection. Two AVPs that may shape viral genomes are the zinc finger antiviral protein (ZAP) and the apolipoprotein B mRNA editing enzyme-catalytic polypeptide-like 3 (APOBEC3). The former binds to CpG dinucleotides to facilitate the degradation of viral transcripts while the latter frequently deaminates C into U residues which could generate notable viral sequence variations. We tested the hypothesis that both APOBEC3 and ZAP impose selective pressures that shape the genome of an infecting coronavirus. Our investigation considered a comprehensive number of publicly available genomes for seven coronaviruses (SARS-CoV-2, SARS-CoV, and MERS infecting Homo sapiens, Bovine CoV infecting Bos taurus, MHV infecting Mus musculus, HEV infecting Sus scrofa, and CRCoV infecting Canis lupus familiaris). We show that coronaviruses that regularly infect tissues with abundant AVPs have CpG-deficient and U-rich genomes; whereas those that do not infect tissues with abundant AVPs do not share these sequence hallmarks. Among the coronaviruses surveyed herein, CpG is most deficient in SARS-CoV-2 and a temporal analysis showed a marked increase in C to U mutations over four months of SARS-CoV-2 genome evolution. Furthermore, the preferred motifs in which these C to U mutations occur are the same as those subjected to APOBEC3 editing in HIV-1. These results suggest that both ZAP and APOBEC3 shape the SARS-CoV-2 genome: ZAP imposes a strong CpG avoidance, and APOBEC3 constantly edits C to U. Evolutionary pressures exerted by host immune systems onto viral genomes may motivate novel strategies for SARS-CoV-2 vaccine development.


Subject(s)
COVID-19/genetics , Coronavirus/genetics , Cytidine Deaminase/genetics , RNA-Binding Proteins/genetics , Repressor Proteins/genetics , APOBEC Deaminases , Animals , COVID-19/pathology , COVID-19/virology , Cattle , Coronavirus/classification , Coronavirus/pathogenicity , Dogs , Evolution, Molecular , Genome, Viral/genetics , Humans , Mice , Middle East Respiratory Syndrome Coronavirus/genetics , Middle East Respiratory Syndrome Coronavirus/pathogenicity , Severe acute respiratory syndrome-related coronavirus/genetics , Severe acute respiratory syndrome-related coronavirus/pathogenicity , SARS-CoV-2/genetics , SARS-CoV-2/pathogenicity , Swine/virology
12.
Vet Microbiol ; 252: 108933, 2021 Jan.
Article in English | MEDLINE | ID: covidwho-966338

ABSTRACT

There is strong evidence that severe acute respiratory syndrome 2 virus (SARS-CoV-2), the causative agent of the coronavirus disease 2019 (COVID-19) pandemic, originated from an animal reservoir. However, the exact mechanisms of emergence, the host species involved, and the risk to domestic and agricultural animals are largely unknown. Some domestic animal species, including cats, ferrets, and minks, have been demonstrated to be susceptible to SARS-CoV-2 infection, while others, such as pigs and chickens, are not. Importantly, the susceptibility of ruminants to SARS-CoV-2 is unknown, even though they often live in close proximity to humans. We investigated the replication and tissue tropism of two different SARS-CoV-2 isolates in the respiratory tract of three farm animal species - cattle, sheep, and pigs - using respiratory ex vivo organ cultures (EVOCs). We demonstrate that the respiratory tissues of cattle and sheep, but not of pigs, sustain viral replication in vitro of both isolates and that SARS-CoV-2 is associated to ACE2-expressing cells of the respiratory tract of both ruminant species. Intriguingly, a SARS-CoV-2 isolate containing an amino acid substitution at site 614 of the spike protein (mutation D614G) replicated at higher magnitude in ex vivo tissues of both ruminant species, supporting previous results obtained using human cells. These results suggest that additional in vivo experiments involving several ruminant species are warranted to determine their potential role in the epidemiology of this virus.


Subject(s)
Organ Culture Techniques , Respiratory System/virology , Ruminants/virology , SARS-CoV-2/physiology , Viral Tropism , Virus Replication , Angiotensin-Converting Enzyme 2/genetics , Animals , Cattle/virology , Host Specificity , SARS-CoV-2/genetics , Sheep/virology , Swine/virology
13.
Virology ; 553: 35-45, 2021 01 15.
Article in English | MEDLINE | ID: covidwho-922156

ABSTRACT

We report the generation of a full-length infectious cDNA clone for porcine deltacoronavirus strain USA/IL/2014/026. Similar to the parental strain, the infectious clone virus (icPDCoV) replicated efficiently in cell culture and caused mild clinical symptoms in piglets. To investigate putative viral interferon (IFN) antagonists, we generated two mutant viruses: a nonstructural protein 15 mutant virus that encodes a catalytically-inactive endoribonuclease (icEnUmut), and an accessory gene NS6-deletion virus in which the NS6 gene was replaced with the mNeonGreen sequence (icDelNS6/nG). By infecting PK1 cells with these recombinant PDCoVs, we found that icDelNS6/nG elicited similar levels of type I IFN responses as icPDCoV, however icEnUmut stimulated robust type I IFN responses, demonstrating that the deltacoronavirus endoribonuclease, but not NS6, functions as an IFN antagonist in PK1 cells. Collectively, the construction of a full-length infectious clone and the identification of an IFN-antagonistic endoribonuclease will aid in the development of live-attenuated deltacoronavirus vaccines.


Subject(s)
DNA, Complementary/isolation & purification , Deltacoronavirus/genetics , Swine/virology , Animals , Clone Cells , Coronavirus Infections/pathology , Deltacoronavirus/pathogenicity , Deltacoronavirus/physiology , Endoribonucleases/physiology , Interferons/antagonists & inhibitors , Virus Replication
14.
Med Sci (Paris) ; 36(6-7): 642-646, 2020.
Article in French | MEDLINE | ID: covidwho-851322

ABSTRACT

TITLE: Épidémies: Leçons d'Histoire. ABSTRACT: Jusqu'au milieu du XVIIIe siècle, l'espérance de vie était de 25 ans dans les pays d'Europe, proche alors de celle de la préhistoire. À cette époque, nos ancêtres succombaient, pour la plupart, à une infection bactérienne ou virale, quand la mort n'était pas le résultat d'un épisode critique, comme la guerre ou la famine. Un seul microbe suffisait à terrasser de nombreuses victimes. L'épidémie de SARS-CoV-2 est là pour nous rappeler que ce risque est désormais à nouveau d'actualité. Si son origine zoonotique par la chauve-souris est probable, la contamination interhumaine montre son adaptation rapide à l'homme et permet d'évoquer ainsi la transmission des épidémies, qu'elle soit ou non liée à des vecteurs, ces derniers pouvant représenter dans d'autres occasions un des maillons de la chaîne.


Subject(s)
Bacterial Infections/epidemiology , Epidemics/history , Virus Diseases/epidemiology , Adult , Animals , Bacterial Infections/history , Betacoronavirus/physiology , COVID-19 , Cattle , Chiroptera/virology , Communicable Diseases, Emerging/epidemiology , Communicable Diseases, Emerging/history , Communicable Diseases, Emerging/microbiology , Communicable Diseases, Emerging/virology , Coronavirus Infections/epidemiology , Disease Reservoirs/microbiology , Disease Reservoirs/veterinary , Disease Reservoirs/virology , Dogs , History, 18th Century , History, 19th Century , History, 20th Century , History, 21st Century , History, Ancient , Humans , Life Expectancy/history , Life Expectancy/trends , Longevity/physiology , Pandemics , Pneumonia, Viral/epidemiology , SARS-CoV-2 , Sheep/microbiology , Sheep/virology , Swine/microbiology , Swine/virology , Virus Diseases/history , Zoonoses/epidemiology , Zoonoses/virology
15.
Vet Microbiol ; 247: 108785, 2020 Aug.
Article in English | MEDLINE | ID: covidwho-827867

ABSTRACT

Porcine deltacoronavirus (PDCoV) is a novel swine enteropathogenic coronavirus that causes watery diarrhea, vomiting and mortality in nursing piglets. Type III interferons (IFN-λs) are the major antiviral cytokines in intestinal epithelial cells, the target cells in vivo for PDCoV. In this study, we found that PDCoV infection remarkably inhibited Sendai virus-induced IFN-λ1 production by suppressing transcription factors IRF and NF-κB in IPI-2I cells, a line of porcine intestinal mucosal epithelial cells. We also confirmed that PDCoV infection impeded the activation of IFN-λ1 promoter stimulated by RIG-I, MDA5 and MAVS, but not by TBK1 and IRF1. Although the expression levels of IRF1 and MAVS were not changed, PDCoV infection resulted in reduction of the number of peroxisomes, the platform for MAVS to activate IRF1, and subsequent type III IFN production. Taken together, our study demonstrates that PDCoV suppresses type III IFN responses to circumvent the host's antiviral immunity.


Subject(s)
Coronavirus Infections/veterinary , Epithelial Cells/immunology , Epithelial Cells/virology , Host-Pathogen Interactions/immunology , Interferons/antagonists & inhibitors , Animals , Cell Line , Coronavirus , Coronavirus Infections/immunology , Coronavirus Infections/virology , Interferon Regulatory Factor-1/antagonists & inhibitors , Interferon Regulatory Factor-1/immunology , Interferons/immunology , Intestines/cytology , Intestines/virology , Kidney/cytology , Kidney/virology , NF-kappa B/antagonists & inhibitors , NF-kappa B/immunology , Sendai virus/immunology , Signal Transduction/immunology , Swine/virology , Swine Diseases/immunology , Swine Diseases/virology , Interferon Lambda
17.
Arch Virol ; 165(2): 345-354, 2020 Feb.
Article in English | MEDLINE | ID: covidwho-824852

ABSTRACT

Porcine hemagglutinating encephalomyelitis virus (PHEV) is a typical neurotropic coronavirus that mainly invades the central nervous system (CNS) in piglets and causes vomiting and wasting disease. Emerging evidence suggests that PHEV alters microRNA (miRNA) expression profiles, and miRNA has also been postulated to be involved in its pathogenesis, but the mechanisms underlying this process have not been fully explored. In this study, we found that PHEV infection upregulates miR-142a-3p RNA expression in N2a cells and in the CNS of mice. Downregulation of miR-142a-3p by an miRNA inhibitor led to a significant repression of viral proliferation, implying that it acts as a positive regulator of PHEV proliferation. Using a dual-luciferase reporter assay, miR-142a-3p was found to bind directly bound to the 3' untranslated region (3'UTR) of Rab3a mRNA and downregulate its expression. Knockdown of Rab3a expression by transfection with an miR-142a-3p mimic or Rab3a siRNA significantly increased PHEV replication in N2a cells. Conversely, the use of an miR-142a-3p inhibitor or overexpression of Rab3a resulted in a marked restriction of viral production at both the mRNA and protein level. Our data demonstrate that miR-142a-3p promotes PHEV proliferation by directly targeting Rab3a mRNA, and this provides new insights into the mechanisms of PHEV-related pathogenesis and virus-host interactions.


Subject(s)
Betacoronavirus 1/genetics , Cell Proliferation/genetics , Coronavirus Infections/genetics , MicroRNAs/genetics , Swine/virology , rab3A GTP-Binding Protein/genetics , 3' Untranslated Regions/genetics , Animals , Cell Line , Cell Line, Tumor , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Down-Regulation/genetics , HEK293 Cells , Humans , Mice , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Up-Regulation/genetics
18.
Vet Res Commun ; 44(3-4): 119-130, 2020 Nov.
Article in English | MEDLINE | ID: covidwho-756542

ABSTRACT

Coronaviruses are a large family of viruses that are known to infect both humans and animals. However, the evidence of inter-transmission of coronavirus between humans and companion animals is still a debatable issue. There is substantial evidence that the virus outbreak is fueled by zoonotic transmission because this new virus belongs to the same family of viruses as SARS-CoV associated with civet cats, and MERS-CoV associated with dromedary camels. While the whole world is investigating the possibility about the transmission of this virus, the transmission among humans is established, but the interface between humans and animals is not much evident. Not only are the lives of human beings at risk, but there is an equal potential threat to the animal world. With multiple reports claiming about much possibility of transmission of COVID-19 from humans to animals, there has been a significant increase in the number of pets being abandoned by their owners. Additionally, the risk of reverse transmission of COVID-19 virus from companion pets like cats and dogs at home is yet another area of concern. The present article highlights different evidence of human-animal interface and necessitates the precautionary measures required to combat with the consequences of this interface. The Centers for Disease Control and Prevention (CDC) and the World Health Organization (WHO) have suggested various ways to promote awareness and corroborate practices for helping people as well as animals to stay secure and healthy.


Subject(s)
Betacoronavirus , Coronavirus Infections/transmission , Pneumonia, Viral/transmission , Zoonoses/transmission , Animals , Betacoronavirus/pathogenicity , COVID-19 , Cats/virology , Coronavirus Infections/veterinary , Dogs/virology , Ferrets/virology , Humans , Pandemics/veterinary , Pneumonia, Viral/veterinary , Poultry/virology , SARS-CoV-2 , Swine/virology , Zoonoses/virology
20.
Vet Res Commun ; 44(3-4): 101-110, 2020 Nov.
Article in English | MEDLINE | ID: covidwho-639440

ABSTRACT

The outbreak of the SARS-CoV-2 in mainland China with subsequent human to human transmission worldwide had taken up the shape of a devastating pandemic. The ability of the virus to infect multiple species other than humans has currently been reported in experimental conditions. Non-human primates, felines, ferrets, rodents and host of other animals could previously be infected in experimental conditions with SARS-CoV and recently with SARS-CoV-2, both virus using Angiotensin-converting-enzyme 2 receptor for cellular entry. The variations in sequence homology of ACE2 receptor across species is identified as one of the factors determining virulence and pathogenicity in animals. The infection in experimental animals with SARS-CoV or SARS-CoV-2 on most occasions are asymptomatic, however, the virus could multiply within the respiratory tract and extra-pulmonary organs in most of the species. Here, we discuss about the pathogenicity, transmission, variations in angiotensin-converting-enzyme 2 receptor-binding across species and host pathogen interactions of SARS and SARS-CoV-2 in laboratory animals used in research.


Subject(s)
Betacoronavirus/pathogenicity , Coronavirus Infections/veterinary , Host-Pathogen Interactions , Pandemics/veterinary , Pneumonia, Viral/veterinary , Severe Acute Respiratory Syndrome/veterinary , Severe acute respiratory syndrome-related coronavirus/pathogenicity , Animals , COVID-19 , Callithrix/virology , Cats/virology , Chickens/virology , Chiroptera/virology , Chlorocebus aethiops/virology , Coronavirus Infections/transmission , Coronavirus Infections/virology , Cricetinae/virology , Ferrets/virology , Macaca fascicularis/virology , Macaca mulatta/virology , Mice , Mice, Inbred Strains/virology , Pneumonia, Viral/transmission , Pneumonia, Viral/virology , Rodentia/virology , SARS-CoV-2 , Severe Acute Respiratory Syndrome/transmission , Severe Acute Respiratory Syndrome/virology , Swine/virology
SELECTION OF CITATIONS
SEARCH DETAIL