Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
Front Microbiol ; 14: 1226877, 2023.
Article in English | MEDLINE | ID: mdl-37614595

ABSTRACT

Feline calicivirus (FCV) causes upper respiratory tract diseases and even death in cats, thereby acting as a great threat to feline animals. Currently, FCV prevention is mainly achieved through vaccination, but the effectiveness of vaccination is limited. In this study, 105 FCV strain VP1 sequences with clear backgrounds were downloaded from the NCBI and subjected to a maximum likelihood method for systematic evolutionary analysis. Based on the genetic analysis results, FCV-positive sera were prepared using SPF mice and Chinese field cats as target animals, followed by a cross-neutralization assay conducted on the different genotype strains and in vivo challenge tests were carried out to further verify with the strain with best cross-protection effect. The results revealed that FCV was mainly divided into two genotypes: GI and GII. The GI genotype strains are prevalent worldwide, but all GII genotype strains were isolated from Asia, indicating a clear geographical feature. This may form resistance to FCV prevention in Asia. The in vitro neutralization assay conducted using murine serum demonstrated that the cross-protection effect varied among strains. A strain with broad-spectrum neutralization properties, DL39, was screened. This strain could produce neutralizing titers (10 × 23.08-10 × 20.25) against all strains used in this study. The antibody titers against the GI strains were 10 × 23.08-10 × 20.5 and those against the GII strains were 10 × 20.75-10 × 20.25. Preliminary evidence suggested that the antibody titer of the DL39 strain against GI was higher than that against GII. Subsequent cross-neutralization assays with cat serum prepared with the DL39 strain and each strain simultaneously yielded results similar to those described above. In vivo challenge tests revealed that the DL39 strain-immunized cats outperformed the positive controls in all measures. The results of several trials demonstrated that strain DL39 can potentially be used as a vaccine strain. The study attempted to combine the genetic diversity and phylogenetic analysis of FCV with the discovery of potential vaccines, which is crucial for developing highly effective FCV vaccines.

2.
Infect Immun ; 91(1): e0019322, 2023 01 24.
Article in English | MEDLINE | ID: mdl-36541752

ABSTRACT

Pasteurella multocida primarily causes hemorrhagic septicemia and pneumonia in poultry and livestock. Identification of the relevant virulence factors is therefore essential for understanding its pathogenicity. Pmorf0222, encoding the PM0222 protein, is located on a specific prophage island of the pathogenic strain C48-1 of P. multocida. Its role in the pathogenesis of P. multocida infection is still unknown. The proinflammatory cytokine plays an important role in P. multocida infection; therefore, murine peritoneal exudate macrophages were treated with the purified recombinant PM0222, which induced the secretion of tumor necrosis factor alpha (TNF-α) and interleukin-1ß (IL-1ß) via the Toll-like receptor 1/2 (TLR1/2)-nuclear factor kappa B (NF-κB)/mitogen-activated protein kinase (MAPK) signaling and inflammasome activation. Additionally, the mutant strain and complemented strain were evaluated in the mouse model with P. multocida infection, and PM0222 was identified as a virulence factor, which was secreted by outer membrane vesicles of P. multocida. Further results revealed that Pmorf0222 affected the synthesis of the capsule, adhesion, serum sensitivity, and biofilm formation. Thus, we identified Pmorf0222 as a novel virulence factor in the C48-1 strain of P. multocida, explaining the high pathogenicity of this pathogenic strain.


Subject(s)
Pasteurella Infections , Pasteurella multocida , Mice , Animals , Pasteurella multocida/genetics , NF-kappa B/metabolism , Toll-Like Receptor 1 , Virulence Factors/genetics , Mitogen-Activated Protein Kinases/metabolism
3.
Transbound Emerg Dis ; 69(6): 3300-3316, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35964328

ABSTRACT

The number of parainfluenza virus 5 (PIV5) infection cases has increased worldwide over the past six decades; however, factors underlying this increase remain unclear. PIV5 has been emerging or re-emerging in humans and animal species. To date, no information is yet available regarding PIV5 infection in arthropod ticks. Here, we successfully isolated tick-derived PIV5 from the Ixodes persulcatus species designated as HLJ/Tick/2019 in Heilongjiang, China. Phylogenetic analysis revealed that the tick-derived PIV5 is closely related to subclade 2.2.6, which has become the dominant subtype prevalent in dogs, pigs and wildlife across China. Further experiments to understand the importance of this virus as an infectious vector revealed that a ferret animal model experimentally infected with Tick/HLJ/2019 via the oronasal and ocular inoculation routes developed moderate respiratory distress with pneumonia and neurologic tissue damage from inflammation for the first time. Further surveillance of PIV5 in vectors of viral transmission is necessary to enhance our knowledge of its ecology in reservoirs and facilitate the control of re-emerging diseases.


Subject(s)
Ixodes , Parainfluenza Virus 5 , Animals , Dogs , Humans , Ferrets , Ixodes/virology , Parainfluenza Virus 5/classification , Parainfluenza Virus 5/genetics , Parainfluenza Virus 5/isolation & purification , Phylogeny , Rubulavirus Infections/epidemiology , Rubulavirus Infections/pathology , Rubulavirus Infections/virology , Swine
4.
J Gen Virol ; 103(5)2022 05.
Article in English | MEDLINE | ID: mdl-35579608

ABSTRACT

The purification of virus particles is an essential process for the manufacture of vaccines. However, the application of different purification processes may affect the quality of the virus particles, such as structural integrity and homogeneity, which may further influence the infectivity and immunogenicity of the purified virus. In this study, we took Feline calicivirus (FCV), a common natural pathogen in cats belonging to Caliciviridae, as a research model. By using cryo-electron microscopy (cryo-EM), we incorporated the 3D classification process as a virus flexibility evaluation system. Cryo-EM images of virus particles resulting from different purification processes were compared at near-atomic resolution. The results indicated that molecular sieving purification will impact the stability of P-domains through increasing flexibility as determined by the evaluation system, which can be extended to assess the purification effect on the entire particle. This evaluation process can be further applied to all non-enveloped viruses.


Subject(s)
Caliciviridae Infections , Caliciviridae , Calicivirus, Feline , Cat Diseases , Viruses , Animals , Caliciviridae Infections/veterinary , Cats , Cryoelectron Microscopy/methods , Virion/chemistry
5.
J Neurosci Methods ; 370: 109476, 2022 Mar 15.
Article in English | MEDLINE | ID: mdl-35007653

ABSTRACT

BACKGROUND: Astrocytes are considered key players in neuroimmunopathological processes, and they play a certain role in neuroinflammation. Rodent primary astrocyte cultures are commonly used in the study of human neuroinflammation. However, gene sequence homologies are closer between humans and dogs than between humans and rodents. NEW METHOD: We established protocols to isolate astrocytes from the canine forebrain. Cerebral hemispheres of 3-4-week-old dogs were used. The isolation procedure included the use of the Neural Tissue Dissociation Kit P, demyelination by the magnetic bead method, and separation and preparation by differential adhesion. RESULTS: We found a 96% astrocyte purification rate after isolation by differential adhesion. Purified canine astrocytes increased the secretion of interleukin-1ß, interleukin-6, and tumor necrosis factor-alpha, and increased the expression of glial fibrillary acidic protein after lipopolysaccharide stimulation. We sequenced the transcriptome of the purified canine astrocytes and analyzed the differentially expressed genes among the rodent, human, and canine astrocytes. Transcriptome profiling and gene ontology analysis of the genes co-expressed in humans and canines indicate that human and canine astrocytes may be different from their rodent counterparts in terms of mediated interactions with metals. COMPARED WITH THE EXISTING METHODS: The cells prepared by our method allow for the rapid separation of astrocytes with a relatively small resource scheme. The method also retains the cell phenotype and has an in vitro culture lifetime of approximately 2-3 months. CONCLUSION: We established a method for preparing canine astrocytes with high purity, which can be used to study the biological function of astrocytes in vitro.


Subject(s)
Astrocytes , Cerebral Cortex , Animals , Astrocytes/metabolism , Cells, Cultured , Cerebral Cortex/metabolism , Dogs , Glial Fibrillary Acidic Protein/metabolism , Interleukin-6/genetics , Lipopolysaccharides/metabolism , Transcriptome
6.
Appl Microbiol Biotechnol ; 106(4): 1651-1661, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35089398

ABSTRACT

Feline calicivirus (FCV) has a single-stranded, positive-sense RNA genome, and it is responsible for many infectious respiratory diseases in cats. In addition, more worryingly, highly virulent strains of FCV can cause high mortality in felines. Therefore, a rapid and reliable diagnosis tool plays an important role in controlling the outbreak of FCV. In this study, enzymatic recombinase amplification (ERA) assay combined with lateral flow dipstick (LFD) was developed for the detection of FCV, targeting a relatively conversed position of FCV-ORF1. The results showed that the optimal reaction condition was at 40 °C for 30 min. ERA-LFD method was highly sensitive with the detection limit as low as 3.2 TCID50 of FCV RNA per reaction. The specificity analysis demonstrated no cross-reactivity with feline parvovirus (FPV), feline herpesvirus (FHV) and feline infectious peritonitis virus (FIPV). ERA-LFD was highly repeatable and reproducible, with the intra-assay and inter-assay coefficients of variation for this method both less than 7%. The general test showed that all the recombinant plasmids with known mutant sites and FCV strains with different mutant sites stored in our laboratory were all detected by this method. Of the 23 samples, 14 samples were tested positive for FCV by ERA-LFD and RT-qPCR, respectively. In summary, ERA-LFD assay was a fast, accurate and convenient diagnosis tool for the detection of FCV. KEY POINTS: • The detection principle of ERA-LFD was introduced. • Almost all the currently known FCV strains can be detected. • ERA-LFD is easy to operate and can be used for field detection.


Subject(s)
Caliciviridae Infections , Calicivirus, Feline , Communicable Diseases , Animals , Caliciviridae Infections/diagnosis , Caliciviridae Infections/veterinary , Calicivirus, Feline/genetics , Cats , Real-Time Polymerase Chain Reaction , Recombinases
7.
J Virol ; 95(13): e0033621, 2021 06 10.
Article in English | MEDLINE | ID: mdl-33853967

ABSTRACT

To replicate efficiently and evade the antiviral immune response of the host, some viruses degrade host mRNA to induce host gene shutoff via encoding shutoff factors. In this study, we found that feline calicivirus (FCV) infection promotes the degradation of endogenous and exogenous mRNAs and induces host gene shutoff, which results in global inhibition of host protein synthesis. Screening assays revealed that proteinase-polymerase (PP) is a most effective factor in reducing mRNA expression. Moreover, PP from differently virulent strains of FCV could induce mRNA degradation. Further, we found that the key sites of the PP protein required for its proteinase activity are also essential for its shutoff activity but also required for viral replication. The mechanism analysis showed that PP mainly targets Pol II-transcribed RNA in a ribosome-, 5' cap-, and 3' poly(A) tail-independent manner. Moreover, purified glutathione S-transferase (GST)-PP fusion protein exhibits RNase activity in vitro in assays using green fluorescent protein (GFP) RNA transcribed in vitro as a substrate in the absence of other viral or cellular proteins. Finally, PP-induced shutoff requires host Xrn1 to complete further RNA degradation. This study provides a newly discovered strategy in which FCV PP protein induces host gene shutoff by promoting the degradation of host mRNAs. IMPORTANCE Virus infection-induced shutoff is the result of targeted or global manipulation of cellular gene expression and leads to efficient viral replication and immune evasion. FCV is a highly contagious pathogen that persistently infects cats. It is unknown how FCV blocks the host immune response and persistently exists in cats. In this study, we found that FCV infection promotes the degradation of host mRNAs and induces host gene shutoff via a common strategy. Further, PP protein for different FCV strains is a key factor that enhances mRNA degradation. An in vitro assay showed that the GST-PP fusion protein possesses RNase activity in the absence of other viral or cellular proteins. This study demonstrates that FCV induces host gene shutoff by promoting the degradation of host mRNAs, thereby introducing a potential mechanism by which FCV infection inhibits the immune response.


Subject(s)
Calicivirus, Feline/growth & development , Immune Evasion/immunology , Peptide Hydrolases/metabolism , RNA Stability/physiology , RNA, Messenger/metabolism , Ribonucleases/metabolism , Animals , Caliciviridae Infections/pathology , Calicivirus, Feline/genetics , Calicivirus, Feline/metabolism , Cats , Cell Line , HEK293 Cells , Humans , Immune Evasion/genetics , Peptide Hydrolases/genetics , Protein Biosynthesis/physiology , RNA Interference , RNA, Small Interfering/genetics , Ribonucleases/genetics , Virus Replication
8.
Ticks Tick Borne Dis ; 12(1): 101554, 2021 01.
Article in English | MEDLINE | ID: mdl-33002807

ABSTRACT

Methionine aminopeptidases (MetAPs), which remove the initiator methionine from nascent peptides, are essential in all organisms and considered to be a valuable targets for the treatment of various diseases, including cancer, malaria, and bacterial infections. However, MetAPs have not been reported in hard ticks (family Ixodidae), and their bioinformatics characterisation in tick's genome sequences is limited. In this study, we cloned, identified, and characterised a novel MetAP from Ixodes persulcatus, a vector for pathogens causing Lyme borreliosis and tick-borne encephalitis. The sequence analysis showed that I. persulcatus MetAP was a type 1 enzyme carrying C-terminal motifs conserved in the M24A family of metallopeptidases. Protein-protein docking simulations using human MetAP revealed conservation of substrate and metal-binding residues in the catalytic site cleft of the novel enzyme, which was designated IpMetAP. Recombinant IpMetAP expressed in Escherichia coli revealed its significant enzymatic activity with the synthetic substrate H-Met-4-methyl-coumaryl-7-amide at pH 7.5 with Km of 0.014 mM, kcat of 0.25 s-1, and overall catalytic efficiency (kcat/Km) of 18.36 mM-1 s-1. The activity of IpMetAP was enhanced by the addition of divalent cations Mn2+ and Co2+ and significantly inhibited by EDTA and bestatin. Site-directed mutagenesis of conserved amino acids indicated that the substitution of metal-binding residues D226 and H288 completely abolished the IpMetAP enzymatic activity, whereas that of the other sites had only moderate effects on substrate hydrolysis. The catalytic properties of IpMetAP suggest that the enzyme behaves similar to other MetAPs and such characterization expands our knowledge of aminopeptidases and protein metabolism of tick.


Subject(s)
Aminopeptidases/genetics , Arthropod Proteins/genetics , Ixodes/genetics , Amino Acid Sequence , Aminopeptidases/chemistry , Aminopeptidases/metabolism , Animals , Arthropod Proteins/chemistry , Arthropod Proteins/metabolism , China , Ixodes/metabolism , Molecular Docking Simulation , Phylogeny , Protein Domains , Sequence Alignment
9.
PLoS Pathog ; 16(10): e1008944, 2020 10.
Article in English | MEDLINE | ID: mdl-33075108

ABSTRACT

Feline calicivirus (FCV) belongs to the Caliciviridae, which comprises small RNA viruses of both medical and veterinary importance. Once infection has occurred, FCV can persist in the cat population, but the molecular mechanism of how it escapes the innate immune response is still unknown. In this study, we found FCV strain 2280 to be relatively resistant to treatment with IFN-ß. FCV 2280 infection inhibited IFN-induced activation of the ISRE (Interferon-stimulated response element) promoter and transcription of ISGs (Interferon-stimulated genes). The mechanistic analysis showed that the expression of IFNAR1, but not IFNAR2, was markedly reduced in FCV 2280-infected cells by inducing the degradation of IFNAR1 mRNA, which inhibited the phosphorylation of downstream adaptors. Further, overexpression of the FCV 2280 nonstructural protein p30, but not p30 of the attenuated strain F9, downregulated the expression of IFNAR1 mRNA. His-p30 fusion proteins were produced in Escherichia coli and purified, and an in vitro digestion assay was performed. The results showed that 2280 His-p30 could directly degrade IFNAR1 RNA but not IFNAR2 RNA. Moreover, the 5'UTR of IFNAR1 mRNA renders it directly susceptible to cleavage by 2280 p30. Next, we constructed two chimeric viruses: rFCV 2280-F9 p30 and rFCV F9-2280 p30. Compared to infection with the parental virus, rFCV 2280-F9 p30 infection displayed attenuated activities in reducing the level of IFNAR1 and inhibiting the phosphorylation of STAT1 and STAT2, whereas rFCV F9-2280 p30 displayed enhanced activities. Animal experiments showed that the virulence of rFCV 2280-F9 p30 infection was attenuated but that the virulence of rFCV F9-2280 p30 was increased compared to that of the parental viruses. Collectively, these data show that FCV 2280 p30 could directly and selectively degrade IFNAR1 mRNA, thus blocking the type I interferon-induced activation of the JAK-STAT signalling pathway, which may contribute to the pathogenesis of FCV infection.


Subject(s)
Antiviral Agents/pharmacology , Caliciviridae Infections/drug therapy , Calicivirus, Feline/pathogenicity , Immunity, Innate/drug effects , Interferon Type I/metabolism , Animals , Caliciviridae Infections/virology , Calicivirus, Feline/drug effects , Calicivirus, Feline/immunology , Cat Diseases/virology , Cats , Interferon Type I/immunology , Interferon-beta/genetics , Viruses/drug effects , Viruses/genetics
10.
Vet Microbiol ; 245: 108707, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32456815

ABSTRACT

Feline viral rhinotracheitis is a prevalent disease among cats caused by feline herpesvirus 1 (FHV-1). microRNAs (miRNAs), which serve as important regulatory factors in the host, participate in the regulation of the host innate immune response to virus infection. However, the roles of miRNAs in the FHV-1 life cycle remain unclear. In this study, we found that a new miRNA, miR-101, could suppress FHV-1 replication. FHV-1 infection upregulated the expression level of miR-101 in a cGAS-dependent manner. Furthermore, miR-101 could significantly enhance type I interferon antiviral signaling by targeting suppressor of cytokine signaling 5 (SOCS5), a negative regulator of the JAK-STAT pathway. Likewise, knockdown of cellular SOCS5 also suppressed FHV-1 replication due to the enhancement of IFN-I-induced signaling cascades. Taken together, our data demonstrated a new strategy for miR-101-mediated defense against FHV-1 infection by enhancing IFN-I antiviral signaling and increased the knowledge of miRNAs regulating innate immune signaling pathways.


Subject(s)
Herpesviridae Infections/veterinary , Host-Pathogen Interactions , MicroRNAs/genetics , Suppressor of Cytokine Signaling Proteins/genetics , Varicellovirus/physiology , Virus Replication , Animals , Cats , Cell Line , Herpesviridae Infections/virology , Signal Transduction , Suppressor of Cytokine Signaling Proteins/immunology , Varicellovirus/pathogenicity
11.
Vet Microbiol ; 240: 108543, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31902487

ABSTRACT

Since 2011, to control the spread of pseudorabies (PR), US7/US8/UL23-deleted recombinant PRV (rPRV) vaccines based on current variants have been developed. The vaccines can provide effective immune protection to pigs, but fur-bearing animals, such as dogs, foxes, and minks, are increasingly infected by PRV due to consuming contaminated raw meat or offal from immunized pigs. It is suspected that the attenuated PRV vaccine strain is not safe for these fur-bearing animals. To confirm this, we construct a US7/US8/UL23-deleted and a US7/US8/UL23/US3-deleted rPRV based on PRV GL isolated from fox using the CRISPR/Cas9 method. Growth kinetics in vitro and pathogenicity in dogs were compared between the wild type and both rPRVs. The results showed that the growth kinetics of wild-type PRV and US7/US8/UL23-deleted rPRV were faster than those of US7/US8/UL23/US3-deleted recombinant PRV from 24 h to 48 h post infection. Moreover, PRV GL- and rPRVdelUS7/US8/UL23-infected cells formed cell-cell fusion, but the rPRVdelUS7/US8/UL23/US3-infected cells did not. Dogs challenged with wild-type PRV or US7/US8/UL23-deleted rPRV showed obvious nervous symptoms, and all the dogs died, but the group challenged with the US7/US8/UL23/US3-deleted rPRV did not show any nervous symptoms, and all the dogs survived for the duration of the experiment. Tissue viral load analyses also showed that the virulence of the US7/US8/UL23/US3-deleted rPRV was significantly reduced in dogs. This study provides evidence that the US7/US8/UL23-deleted rPRV variant still exhibits high virulence for dogs and also highlights the role of the US3 gene in the pathogenicity of PRV in dogs and provides a strategy for developing a safer vaccine.


Subject(s)
Gene Deletion , Herpesvirus 1, Suid/genetics , Herpesvirus 1, Suid/pathogenicity , Pseudorabies/virology , Rabies Vaccines/immunology , Viral Proteins/genetics , Animals , Antibodies, Viral/blood , CRISPR-Cas Systems , Dogs , Herpesvirus 1, Suid/growth & development , Pseudorabies/immunology , Rabies Vaccines/administration & dosage , Rabies Vaccines/genetics , Vaccination , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Viral Envelope Proteins/genetics , Virulence
12.
Viruses ; 12(1)2019 12 18.
Article in English | MEDLINE | ID: mdl-31861450

ABSTRACT

In response to viral infection, host cells activate various antiviral responses to inhibit virus replication. While feline herpesvirus 1 (FHV-1) manipulates the host early innate immune response in many different ways, the host could activate the antiviral response to counteract it through some unknown mechanisms. MicroRNAs (miRNAs) which serve as a class of regulatory factors in the host, participate in the regulation of the host innate immune response against virus infection. In this study, we found that the expression levels of miR-26a were significantly upregulated upon FHV-1 infection. Furthermore, FHV-1 infection induced the expression of miR-26a via a cGAS-dependent pathway, and knockdown of cellular cGAS significantly blocked the expression of miR-26a induced by poly (dA:dT) or FHV-1 infection. Next, we investigated the biological function of miR-26a during viral infection. miR-26a was able to increase the phosphorylation of STAT1 and promote type I IFN signaling, thus inhibiting viral replication. The mechanism study showed that miR-26a directly targeted host SOCS5. Knockdown of SOCS5 increased the phosphorylation of STAT1 and enhanced the type I IFN-mediated antiviral response, and overexpression of suppressor of the cytokine signalling 5 (SOCS5) decreased the phosphorylation of STAT1 and inhibited the type I IFN-mediated antiviral response. Meanwhile, with the knockdown of SOCS5, the upregulated expression of phosphorylated STAT1 and the anti-virus effect induced by miR-26a were significantly inhibited. Taken together, our data demonstrated a new strategy of host miRNAs against FHV-1 infection by enhancing IFN antiviral signaling.


Subject(s)
Gene Expression Regulation , Interferon Type I/metabolism , MicroRNAs/genetics , Signal Transduction , Suppressor of Cytokine Signaling Proteins/genetics , Varicellovirus/physiology , Virus Replication/genetics , 3' Untranslated Regions , Animals , Cat Diseases/genetics , Cat Diseases/metabolism , Cat Diseases/virology , Cats , Cell Line , Cells, Cultured , Herpesviridae Infections/veterinary , Host-Pathogen Interactions/genetics , Humans , Interferon Type I/biosynthesis , Janus Kinases/metabolism , Nucleotidyltransferases/metabolism , RNA Interference , STAT Transcription Factors/metabolism
13.
Arch Virol ; 164(8): 2159-2164, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31152250

ABSTRACT

Canine enteric coronaviruses (CCoVs) are important enteric pathogens of dogs. CCoVs with different variations are typically pantropic and pathogenic in dogs. In this study, we isolated a CCoV, designated HLJ-073, from a dead 6-week-old male Pekingese with gross lesions and diarrhea. Interestingly, sequence analysis suggested that HLJ-073 contained a 350-nt deletion in ORF3abc compared with reference CCoV isolates, resulting in the loss of portions of ORF3a and ORF3c and the complete loss of ORF3b. Phylogenetic analysis based on the S gene showed that HLJ-073 was more closely related to members of the FCoV II cluster than to members of the CCoV I or CCoV II cluster. Furthermore, recombination analysis suggested that HLJ-073 originated from the recombination of FCoV 79-1683 and CCoV A76, which were both isolated in the United States. Cell tropism experiments suggested that HLJ-073 could effectively replicate in canine macrophages/monocytes and human THP-1 cells. This is the first report of the isolation of strain HLJ-073 in China, and this virus has biological characteristics that are different from those of other reported CCoVs.


Subject(s)
Coronavirus, Canine/genetics , Sequence Deletion/genetics , Animals , Cells, Cultured , China , Coronavirus Infections/virology , Diarrhea/virology , Dog Diseases/virology , Dogs , Humans , Male , Phylogeny , Sequence Analysis, DNA/methods , Spike Glycoprotein, Coronavirus/genetics , THP-1 Cells
14.
Viruses ; 12(1)2019 12 30.
Article in English | MEDLINE | ID: mdl-31905881

ABSTRACT

Feline infectious peritonitis (FIP), caused by virulent feline coronavirus, is the leading infectious cause of death in cats. The type I interferon (type I IFN)-mediated immune responses provide host protection from infectious diseases. Several coronaviruses have been reported to evolve diverse strategies to evade host IFN response. However, whether feline infectious peritonitis virus (FIPV) antagonizes the type I IFN signaling remains unclear. In this study, we demonstrated that FIPV strain DF2 infection not only failed to induce interferon-ß (IFN-ß) and interferon-stimulated gene (ISG) production, but also inhibited Sendai virus (SEV) or polyinosinic-polycytidylic acid (poly(I:C))-induced IFN-ß production. Subsequently, we found that one of the non-structural proteins encoded by the FIPV genome, nsp5, interrupted type I IFN signaling in a protease-dependent manner by cleaving the nuclear factor κB (NF-κB) essential modulator (NEMO) at three sites-glutamine132 (Q132), Q205, and Q231. Further investigation revealed that the cleavage products of NEMO lost the ability to activate the IFN-ß promoter. Mechanistically, the nsp5-mediated NEMO cleavage disrupted the recruitment of the TRAF family member-associated NF-κB activator (TANK) to NEMO, which reduced the phosphorylation of interferon regulatory factor 3 (IRF3), leading to the inhibition of type I IFN production. Our research provides new insights into the mechanism for FIPV to counteract host innate immune response.


Subject(s)
Coronavirus Infections/immunology , Coronavirus, Feline/physiology , Cysteine Endopeptidases/metabolism , I-kappa B Kinase/metabolism , Interferon Type I/antagonists & inhibitors , Viral Nonstructural Proteins/metabolism , Animals , Cats , Cell Line , Coronavirus 3C Proteases , Coronavirus, Feline/metabolism , Cysteine Endopeptidases/genetics , I-kappa B Kinase/genetics , Immune Evasion , Immunity, Innate , Interferon Regulatory Factor-3 , Interferon Type I/metabolism , Interferon-beta/antagonists & inhibitors , Interferon-beta/genetics , Interferon-beta/metabolism , Mutation , NF-kappa B/metabolism , Promoter Regions, Genetic , Signal Transduction , Viral Nonstructural Proteins/genetics
15.
Biomed Res Int ; 2018: 2739830, 2018.
Article in English | MEDLINE | ID: mdl-30009167

ABSTRACT

Interferons (IFNs) can inhibit most, if not all, viral infections by eliciting the transcription of hundreds of interferon-stimulated genes (ISGs). Feline calicivirus (FCV) is a highly contagious pathogen of cats and a surrogate for Norwalk virus. Interferon efficiently inhibits the replication of FCV, but the mechanism of the antiviral activity is poorly understood. Here, we evaluated the anti-FCV activity of ten ISGs, whose antiviral activities were previously reported. The results showed that interferon regulatory factor 1 (IRF1) can significantly inhibit the replication of FCV, whereas the other ISGs tested in this study failed. Further, we found that IRF1 was localized in the nucleus and efficiently activated IFN-ß and the ISRE promoter. IRF1 can trigger the production of endogenous interferon and the expression of ISGs, suggesting that IRF1 can positively regulate IFN signalling. Importantly, the mRNA and protein levels of IRF1 were reduced upon FCV infection, which may be a new strategy for FCV to evade the innate immune system. Finally, the antiviral activity of IRF1 against feline panleukopenia virus, feline herpesvirus, and feline infectious peritonitis virus was demonstrated. These data indicate that feline IRF1 plays an important role in regulating the host type I IFN response and inhibiting feline viral infections.


Subject(s)
Antiviral Agents/pharmacology , Calicivirus, Feline/immunology , Interferon Regulatory Factor-1/immunology , Virus Replication , Animals , Caliciviridae Infections/immunology , Caliciviridae Infections/veterinary , Cat Diseases/immunology , Cats , Viruses
16.
J Virol Methods ; 262: 61-64, 2018 12.
Article in English | MEDLINE | ID: mdl-30016702

ABSTRACT

Mammalian reovirus (MRV) infects many species. Over the past decades, MRV infections in pigs have been reported, and several highly pathogenic MRV strains have recently been isolated in the United States. In this study, an indirect enzyme-linked immunosorbent assay (ELISA) against the σ1 protein from a serotype 3 reovirus strain (MPC/04) was established to detect antibodies in pigs. The assay did not react with antisera against other pig pathogens and was consistent with the indirect immunofluorescence assay (IFA) and virus neutralization test (VNT). In conclusion, the assay is specific and highly sensitive, providing a method for large-scale monitoring of the serotype 3 MRV infection epidemiology in pigs.


Subject(s)
Antibodies, Viral/blood , Enzyme-Linked Immunosorbent Assay/methods , Reoviridae Infections/veterinary , Reoviridae/immunology , Swine Diseases/diagnosis , Animals , Fluorescent Antibody Technique, Indirect , Neutralization Tests , Reoviridae/isolation & purification , Reoviridae Infections/diagnosis , Reoviridae Infections/immunology , Serogroup , Swine/virology , Swine Diseases/virology , Viral Fusion Proteins/immunology
17.
J Virol ; 92(12)2018 06 15.
Article in English | MEDLINE | ID: mdl-29618645

ABSTRACT

As a prevalent agent in cats, feline herpesvirus 1 (FHV-1) infection contributes to feline respiratory disease and acute and chronic conjunctivitis. FHV-1 can successfully evade the host innate immune response and persist for the lifetime of the cat. Several mechanisms of immune evasion by human herpesviruses have been elucidated, but the mechanism of immune evasion by FHV-1 remains unknown. In this study, we screened for FHV-1 open reading frames (ORFs) responsible for inhibiting the type I interferon (IFN) pathway with an IFN-ß promoter reporter and analysis of IFN-ß mRNA levels in HEK 293T cells and the Crandell-Reese feline kidney (CRFK) cell line, and we identified the Ser/Thr kinase US3 as the most powerful inhibitor. Furthermore, we found that the anti-IFN activity of US3 depended on its N terminus (amino acids 1 to 75) and was independent of its kinase activity. Mechanistically, the ectopic expression of US3 selectively inhibited IFN regulatory factor 3 (IRF3) promoter activation. Furthermore, US3 bound to the IRF association domain (IAD) of IRF3 and prevented IRF3 dimerization. Finally, US3-deleted recombinant FHV-1 and US3-repaired recombinant FHV-1 (rFHV-dUS3 and rFHV-rUS3, respectively) were constructed. Compared with wild-type FHV-1 and rFHV-rUS3, infection with rFHV-dUS3 induced large amounts of IFN-ß in vitro and in vivo More importantly, US3 deletion significantly attenuated virulence, reduced virus shedding, and blocked the invasion of trigeminal ganglia. These results indicate that FHV-1 US3 efficiently inhibits IFN induction by using a novel immune evasion mechanism and that FHV-1 US3 is a potential regulator of neurovirulence.IMPORTANCE Despite widespread vaccination, the prevalence of FHV-1 remains high, suggesting that it can successfully evade the host innate immune response and infect cats. In this study, we screened viral proteins for inhibiting the IFN pathway and identified the Ser/Thr kinase US3 as the most powerful inhibitor. In contrast to other members of the alphaherpesviruses, FHV-1 US3 blocked the host type I IFN pathway in a kinase-independent manner and via binding to the IRF3 IAD and preventing IRF3 dimerization. More importantly, the depletion of US3 attenuated the anti-IFN activity of FHV-1 and prevented efficient viral replication in vitro and in vivo Also, US3 deletion significantly attenuated virulence and blocked the invasion of trigeminal ganglia. We believe that these findings not only will help us to better understand the mechanism of how FHV-1 manipulates the host IFN response but also highlight the potential role of US3 in the establishment of latent infection in vivo.


Subject(s)
Alphaherpesvirinae/pathogenicity , Interferon Regulatory Factor-3/antagonists & inhibitors , Interferon-beta/antagonists & inhibitors , Nucleotidyltransferases/genetics , Protein Serine-Threonine Kinases/metabolism , Viral Proteins/metabolism , Alphaherpesvirinae/genetics , Animals , Cat Diseases/virology , Cats , Dimerization , HEK293 Cells , Humans , Interferon Regulatory Factor-3/metabolism , Interferon-beta/genetics , Interferon-beta/metabolism , Membrane Proteins/genetics , Protein Binding/physiology , Protein Serine-Threonine Kinases/genetics , RNA Interference , RNA, Small Interfering/genetics , Signal Transduction/physiology , Viral Proteins/genetics
18.
J Vet Med Sci ; 80(4): 731-735, 2018 Apr 27.
Article in English | MEDLINE | ID: mdl-29459503

ABSTRACT

Feline bocavirus (FBoV) is a newly identified bocavirus of cats in the family Parvoviridae. A novel FBoV HRB2015-LDF was first identified from the cat with severe enteritis in Northeast China, with an overall positive rate of 2.78% (1/36). Phylogenetic and homologous analysis of the complete genome showed that FBoV HRB2015-LDF was clustered into the FBoV branch and closely related to other FBoVs, with 68.7-97.5% identities. And the genes of VP1, NPA and NS1 shared 70.7-97.6, 72.4-98.6 and 67.2-98.0% nucleotide identities with other FBoVs, respectively. The results suggested that the FBoVs could be divided into two distinct lineages, and the difference of nucleotide identities was >20-30% between the lineages.


Subject(s)
Bocavirus , Cat Diseases/epidemiology , Enteritis/veterinary , Parvoviridae Infections/veterinary , Animals , Bocavirus/genetics , Cat Diseases/virology , Cats , China/epidemiology , Enteritis/epidemiology , Enteritis/virology , Male , Parvoviridae Infections/epidemiology , Parvoviridae Infections/virology , Phylogeny , Sequence Analysis, DNA/veterinary
19.
Vet Microbiol ; 213: 15-20, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29291998

ABSTRACT

Rabbit hemorrhagic disease virus (RHDV) is responsible for rabbit hemorrhagic disease (RHD), which is an acute, lethal and highly contagious disease in both wild and domestic rabbits. Although current vaccines are highly effective for controlling RHD, they are derived from infected rabbit livers and their use is thus associated with safety and animal-welfare concerns. In this study, we generated a recombinant lentogenic canine adenovirus type 2 (CAV2) vector expressing the RHDV vp60 gene, named rCAV2-VP60. rCAV2-VP60 expressed VP60 protein in Madin-Darby canine kidney cells as demonstrated by western blot and immunofluorescence assay. Polymerase chain reaction confirmed that the vp60 gene was successfully inserted into rCAV2-VP60 and was still detectable after 20 passages, indicating its stable genetic character. We evaluated the feasibility of rCAV2-VP60 as a live-virus-vectored RHD vaccine in rabbits. rCAV2-VP60 significantly induced specific antibodies to RHDV and provided effective protection against RHDV lethal challenge. These results suggest that rCAV2 expressing RHDV VP60 could be a safe and efficient candidate vaccine against RHDV in rabbits.


Subject(s)
Adenoviruses, Canine/genetics , Caliciviridae Infections/prevention & control , Hemorrhagic Disease Virus, Rabbit/immunology , Viral Structural Proteins/immunology , Viral Vaccines/immunology , Adenoviruses, Canine/metabolism , Animals , Blotting, Western , Caliciviridae Infections/virology , Dogs , Feasibility Studies , Gene Expression , Genetic Vectors , Hemorrhagic Disease Virus, Rabbit/genetics , Madin Darby Canine Kidney Cells , Rabbits , Recombinant Proteins , Viral Structural Proteins/genetics , Viral Structural Proteins/metabolism
20.
Biomed Res Int ; 2017: 7089091, 2017.
Article in English | MEDLINE | ID: mdl-29201911

ABSTRACT

Coronaviruses (CoVs), such as human coronavirus NL63 (HCoV-NL63), severe acute respiratory syndrome CoV (SARS-CoV), murine hepatitis virus (MHV), porcine epidemic diarrhea virus (PEDV), and Middle East Respiratory Syndrome Coronavirus (MERS-CoV), encode papain-like (PL) proteases that inhibit Sendai virus- (SeV-) induced interferon (IFN-ß) production. Recently, the crystal structure of transmissible gastroenteritis virus (TGEV) PL1 has been solved, which was similar to that of SARS-CoV PL2pro, which may antagonize host innate immunity. However, very little is known about whether TGEV PL1 can antagonize host innate immune response. Here, we presented evidence that TGEV PL1 encoded by the replicase gene could suppress the IFN-ß expression and inhibit the nuclear translocation of interferon regulatory factor 3 (IRF3). The ability to antagonize IFN-ß production was dependent on the intact catalytic activity of PL1. Furthermore, TGEV PL1 exerted deubiquitinase (DUB) activity which strongly inhibited the retinoic acid-induced gene I- (RIG-1-) and stimulator of interferon gene- (STING-) dependent IFN expression. Our data collectively suggest that TGEV PL1 can inhibit the IFN-ß expression and interfere with RIG-1- and STING-mediated signaling through a viral DUB activity. Our study has yielded strong evidence for the TGEV PL1 mechanisms that counteract the host innate immunity.


Subject(s)
Host-Pathogen Interactions/genetics , Immunity, Innate/genetics , Interferon-beta/genetics , Papain/genetics , Transmissible gastroenteritis virus/genetics , Animals , Coronavirus Papain-Like Proteases , DEAD Box Protein 58/genetics , Deubiquitinating Enzymes/genetics , HEK293 Cells , Humans , Interferon Regulatory Factor-3/genetics , Interferon-beta/biosynthesis , Membrane Proteins/genetics , Papain/chemistry , Papain/immunology , RNA-Dependent RNA Polymerase/genetics , Receptors, Immunologic , Swine , Transmissible gastroenteritis virus/chemistry , Transmissible gastroenteritis virus/pathogenicity , Ubiquitin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...