Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 610
Filter
1.
Vet Res ; 55(1): 128, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39350170

ABSTRACT

Picornaviridae are non-enveloped ssRNA viruses that cause diseases such as poliomyelitis, hand-foot-and-mouth disease (HFMD), hepatitis A, encephalitis, myocarditis, and foot-and-mouth disease (FMD). Virus-like particles (VLPs) vaccines mainly comprise particles formed through the self-assembly of viral capsid proteins (for enveloped viruses, envelope proteins are also an option). They do not contain the viral genome. On the other hand, the nanoparticles vaccine (NPs) is mainly composed of self-assembling biological proteins or nanomaterials, with viral antigens displayed on the surface. The presentation of viral antigens on these particles in a repetitive array can elicit a strong immune response in animals. VLPs and NPs can be powerful platforms for multivalent antigen presentation. This review summarises the development of virus-like particle vaccines (VLPs) and nanoparticle vaccines (NPs) against picornaviruses. By detailing the progress made in the fight against various picornaviruses such as poliovirus (PV), foot-and-mouth disease virus (FMDV), enterovirus (EV), Senecavirus A (SVA), and encephalomyocarditis virus (EMCV), we in turn highlight the significant strides made in vaccine technology. These advancements include diverse construction methods, expression systems, elicited immune responses, and the use of various adjuvants. We see promising prospects for the continued development and optimisation of VLPs and NPs vaccines. Future research should focus on enhancing these vaccines' immunogenicity, stability, and delivery methods. Moreover, expanding our understanding of the interplay between these vaccines and the immune system will be crucial. We hope these insights will inspire and guide fellow researchers in the ongoing quest to combat picornavirus infections more effectively.


Subject(s)
Nanoparticles , Picornaviridae Infections , Picornaviridae , Vaccines, Virus-Like Particle , Viral Vaccines , Animals , Viral Vaccines/immunology , Picornaviridae Infections/veterinary , Picornaviridae Infections/prevention & control , Picornaviridae Infections/immunology , Vaccines, Virus-Like Particle/immunology , Picornaviridae/immunology
2.
Infect Genet Evol ; 124: 105665, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39233257

ABSTRACT

BACKGROUND: Senecavirus A (SVA) is the only member of the genus Senecavirus in the family Picornaviridae, and is one of the pathogens of porcine blistering disease. SVA has been reported in the United States, Canada, China, Thailand, and Colombia. METHODS: In this study, positive SVA infection was detected by RT-PCR in sick materials collected from pig farms of different sizes in Anhui Province. RESULTS: In this study, a virulent strain of SVA was successfully obtained by viral isolation on BHK21 cells and named SVA-CH-AHAU-1. Meanwhile, a simple, rapid and accurate nano-PCR method for the detection of SVA infection was established in this study, using the recombinant plasmid pClone-SVA-3D as a template. CONCLUSIONS: The complete genome of SVA-CH-AHAU-1 is 7286 bp, including a 5' non-coding region (UTR), an open reading frame (ORF) of 6546 nucleotides, encoding 2182 amino acids (aa), and a 3' UTR with Poly(A) features, and phylogenetic analysis showed that this isolate had the highest nucleotide homology (97.9 %) with the US isolate US-15-41901SD. In this study, the virulent strain SVA-CH-AHAU-1 was found to recombine in the ORF region with isolates SVA-CH-SDGT-2017 and SVA/Canada/ON/FMA-2015-0024 T2/2015. The complete genome has been submitted to GeneBank with the accession number OM654411. In addition, our results suggest that the established nano-PCR assay can be used as an economical, reliable and sensitive method for the field diagnosis of SVA method, especially in resource-limited areas.


Subject(s)
Genome, Viral , Phylogeny , Picornaviridae Infections , Picornaviridae , Picornaviridae/genetics , Picornaviridae/classification , Picornaviridae/isolation & purification , Animals , Picornaviridae Infections/veterinary , Picornaviridae Infections/virology , Picornaviridae Infections/diagnosis , Swine , China , Evolution, Molecular , Swine Diseases/virology
3.
Arch Virol ; 169(10): 200, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39285064

ABSTRACT

Novel bovine parechoviruses (Bo ParVs) were isolated from the feces of Japanese black cattle. Phylogenetic analysis revealed that the novel Bo ParVs formed an independent cluster, exhibiting 72.2-75.6% nucleotide sequence identity to previous Bo ParVs, suggesting that they represent a new genotype. Bo ParVs, including the novel Bo ParVs, shared sequence similarity with each other in the 3' untranslated region (3'UTR) and exhibited low sequence similarity (<38.9% identity) to other parechoviruses. However, a secondary structure prediction of the 3'UTR revealed that the Bo ParVs shared conserved motifs in domain 2 with parechovirus B and E, suggesting some evolutionary constrains in this region.


Subject(s)
Cattle Diseases , Feces , Parechovirus , Phylogeny , Picornaviridae Infections , Animals , Cattle , Parechovirus/genetics , Parechovirus/isolation & purification , Parechovirus/classification , Picornaviridae Infections/veterinary , Picornaviridae Infections/virology , Feces/virology , Cattle Diseases/virology , 3' Untranslated Regions/genetics , Japan , Genotype , Nucleic Acid Conformation , RNA, Viral/genetics
4.
Sci Rep ; 14(1): 22575, 2024 09 29.
Article in English | MEDLINE | ID: mdl-39343789

ABSTRACT

DHAV-1 is a highly infectious pathogen that can cause acute hepatitis in ducklings. MicroRNA (miRNA) plays an essential regulatory role in virus response. We characterized and compared miRNA and mRNA expression profiles in duck embryonic fibroblasts (DEF) and the liver of ducklings infected with DHAV-1. DHAV-1 infected DEF was divided into infection group (D group) and blank group (M group), and DHAV-1 infected duckling group was divided into infection group (H group) and blank group (N group). D vs. M have 130 differentially expressed (DE) miRNA (DEM) and 2204 differentially expressed (DE) mRNA (DEG), H vs. N have 72 DEM and 1976 DEG. By the intersection of D vs. M and H vs. N comparisons, 15 upregulated DEM, 5 downregulated DEM, 340 upregulated DEG and 50 downregulated DEG were found with both in vivo and in vitro DHAV-1 infection. In particular, we identified the same DE miRNA target genes and functional annotations of DE mRNA. We enriched with multiple gene ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, which may have important roles in viral virulence, host immunity, and metabolism. We selected miR-155, which is co-upregulated, and found that miR-155 targets SOCS1 to inhibit DHVA-1 replication.


Subject(s)
Ducks , Fibroblasts , MicroRNAs , Poultry Diseases , RNA, Messenger , Animals , Ducks/virology , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Fibroblasts/metabolism , Fibroblasts/virology , Poultry Diseases/virology , Poultry Diseases/genetics , Poultry Diseases/metabolism , Gene Expression Profiling , Picornaviridae Infections/virology , Picornaviridae Infections/genetics , Picornaviridae Infections/veterinary , Picornaviridae Infections/metabolism , Mardivirus/genetics , Liver/metabolism , Liver/virology , Host-Pathogen Interactions/genetics , Gene Expression Regulation
5.
Vet Res ; 55(1): 110, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39300570

ABSTRACT

Duck hepatitis A virus type 1 (DHAV-1) is an important member of the Picornaviridae family that causes highly fatal hepatitis in ducklings. Since picornaviruses have small genomes with limited coding capacity, they must utilize host proteins for viral cap-independent translation and RNA replication. Here, we report the role of duck poly(rC)-binding protein 2 (PCBP2) in regulating the replication and translation of DHAV-1. During DHAV-1 infection, PCBP2 expression was upregulated. A biotinylated RNA pull-down assay revealed that PCBP2 positively regulates DHAV-1 translation through specific interactions with structural domains II and III of the DHAV-1 internal ribosome entry site (IRES). Further studies revealed that PCBP2 promotes DHAV-1 replication via an interaction of its KH1 domain (aa 1-92) with DHAV-1 3Dpol. Thus, our studies demonstrated the specific role of PCBP2 in regulating DHAV-1 translation and replication, revealing a novel mechanism by which host‒virus interactions regulate viral translation and replication. These findings contribute to further understanding of the pathogenesis of picornavirus infections.


Subject(s)
Ducks , Hepatitis Virus, Duck , Picornaviridae Infections , Poultry Diseases , RNA-Binding Proteins , Virus Replication , Animals , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Hepatitis Virus, Duck/physiology , Hepatitis Virus, Duck/genetics , Poultry Diseases/virology , Picornaviridae Infections/veterinary , Picornaviridae Infections/virology , Hepatitis, Viral, Animal/virology , Protein Biosynthesis
6.
J Vet Sci ; 25(5): e59, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39237364

ABSTRACT

IMPORTANCE: Despite advancements in herd management, feeding, and pharmaceutical interventions, neonatal calf diarrhea (NCD) remains a major global health concern. Bacteria, viruses, and parasites are the major contributors to NCD. Although several pathogens have been identified in the Republic of Korea (ROK), the etiological agents of numerous NCD cases have not been identified. OBJECTIVE: To identify, for the first time, the prevalence and impact of Boosepivirus (BooV) on calf diarrhea in the ROK. METHODS: Here, the unknown cause of calf diarrhea was determined using metagenomics We then explored the prevalence of certain pathogens, including BooV, that cause NCD. Seventy diarrheal fecal samples from Hanwoo (Bos taurus coreanae) calves were analyzed using reverse transcriptase and quantitative real-time polymerase chain reaction for pathogen detection and BooV isolate sequencing. RESULTS: The complete genome of BooV was detected from unknown causes of calf diarrhea. And also, BooV was the most frequently detected pathogen (35.7%) among 8 pathogens in 70 diarrheic feces from Hanwoo calves. Co-infection analyses indicated that most BooV-positive samples were solely infected with BooV, indicating its significance in NCD in the ROK. All isolates were classified as BooV B in phylogenetic analysis. CONCLUSIONS AND RELEVANCE: This is the first study to determine the prevalence and molecular characteristics of BooV in calf diarrhea in the ROK, highlighting the potential importance of BooV as a causative agent of calf diarrhea and highlighting the need for further research on its epidemiology and pathogenicity.


Subject(s)
Cattle Diseases , Diarrhea , Phylogeny , Picornaviridae Infections , Picornaviridae , Animals , Cattle , Republic of Korea/epidemiology , Cattle Diseases/virology , Cattle Diseases/epidemiology , Picornaviridae Infections/veterinary , Picornaviridae Infections/epidemiology , Picornaviridae Infections/virology , Picornaviridae/isolation & purification , Picornaviridae/genetics , Picornaviridae/classification , Diarrhea/veterinary , Diarrhea/virology , Diarrhea/epidemiology , Prevalence , Feces/virology , Genome, Viral , Animals, Newborn
7.
Vet Res ; 55(1): 115, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39334325

ABSTRACT

Senecavirus A (SVA), an emerging virus that causes blisters on the nose and hooves, reduces the production performance of pigs. RSAD2 is a radical S-adenosylmethionine (SAM) enzyme, and its expression can suppress various viruses due to its broad antiviral activity. However, the regulatory relationship between SVA and RSAD2 and the mechanism of action remain unclear. Here, we demonstrated that SVA infection increased RSAD2 mRNA levels, whereas RSAD2 expression negatively regulated viral replication, as evidenced by decreased viral VP1 protein expression, viral titres, and infected cell numbers. Viral proteins that interact with RSAD2 were screened, and the interaction between the 2 C protein and RSAD2 was found to be stronger than that between other proteins. Additionally, amino acids (aa) 43-70 of RSAD2 were crucial for interacting with the 2 C protein and played an important role in its anti-SVA activity. RSAD2 was induced by type I interferon (IFN-I) via Janus kinase signal transducer and activator of transcription (JAK-STAT), and had antiviral activity. Ruxolitinib, a JAK-STAT pathway inhibitor, and the knockdown of JAK1 expression substantially reduced RSAD2 expression levels and antiviral activity. Taken together, these results revealed that RSAD2 blocked SVA infection by interacting with the viral 2 C protein and provide a strategy for preventing and controlling SVA infection.


Subject(s)
Picornaviridae Infections , Picornaviridae , Virus Replication , Animals , Virus Replication/drug effects , Picornaviridae/physiology , Swine , Picornaviridae Infections/veterinary , Picornaviridae Infections/virology , Swine Diseases/virology , Viral Proteins/metabolism , Viral Proteins/genetics
8.
Poult Sci ; 103(10): 104087, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39094497

ABSTRACT

Duck hepatitis A virus type 3 (DHAV-3) is an infectious virus that is highly fatal to ducklings and causes significant economic losses in the duck industry worldwide. Biosecurity and vaccination are required to control the pathogen. In the present study, we attenuated a lowly pathogenic DHAV-3 clinical isolate, named as HB, by serial passaging in duck embryos, and followed by several adaptive proliferations in specific-pathogen-free (SPF) chicken embryos. The virulence of DHAV-3 at different passages was assessed by infecting 3-day-old ducklings. We found that the HB strain lost pathogenicity to ducklings from the 55th passage onwards. The 80th passage strain (HB80), which achieved good growth capacity in duck embryos with a viral titer of 108.17 50% egg lethal dose per milliliter (ELD50/mL), was selected as a live attenuated vaccine candidate. The HB80 strain did not induce clinical symptoms or pathological lesions in 3-day-old ducklings and showed no virulence reversion after 5 rounds of in vivo back-passage. The minimum effective dose of HB80 was determined to be 104.5 ELD50 by hypodermic inoculation of the neck. Importantly, a single dose of HB80 elicited good immune responses and provided complete protection against challenge with the lethal DHAV-3 strain. Compared with the genomic sequence of the parental HB strain, HB80 had 7 amino acid substitutions, two of them are in the hypervariable region of the VP1 and polymerase-encoding 3D regions, which may play a role in virulence attenuation. Our data suggest that the attenuated HB80 strain is a promising vaccine candidate for the prevention of DHAV-3 infections in China. HB80 has been registered as a New Veterinary Drug Registration Certificate by the Chinese Ministry of Agriculture and Rural Affairs (MARA), and is the first live attenuated DHAV-3 vaccine strain to be officially licensed in China.


Subject(s)
Ducks , Picornaviridae Infections , Poultry Diseases , Vaccines, Attenuated , Viral Vaccines , Animals , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Poultry Diseases/prevention & control , Poultry Diseases/virology , China , Picornaviridae Infections/veterinary , Picornaviridae Infections/prevention & control , Picornaviridae Infections/virology , Viral Vaccines/immunology , Virulence , Specific Pathogen-Free Organisms , Hepatitis Virus, Duck/pathogenicity
9.
Viruses ; 16(8)2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39205178

ABSTRACT

Equine rhinitis A (ERAV) and B (ERBV) viruses are respiratory pathogens with worldwide distribution. The current study aimed to determine the frequency of infection of ERAV and ERBV among horses and foals at Polish national studs, and to determine genetic variability within the viruses obtained. Virus-specific quantitative RT-PCR assays targeting a 5' untranslated region were used to screen nasal swabs collected from 621 horses at 16 national horse studs from throughout Poland, including 553 healthy horses and 68 horses with respiratory disease. A partial DNA polymerase gene was amplified and sequenced from the qRT-PCR-positive samples. The obtained sequences were analysed using phylogeny and genetic network analysis. None of the nasal swabs were positive for ERAV, whereas ERBV was found in 11/621 (1.78%) samples collected from 10 healthy horses and one foal affected by respiratory disease. Partial DNA polymerase gene sequence variability was correlated with individual horses and studs from which samples were collected when only Polish sequences were analysed, but there was no correlation between country of origin and ERBV sequence when Polish and international sequences were included in the network. The report presents the first detection of ERBV in Poland.


Subject(s)
Horse Diseases , Phylogeny , Picornaviridae Infections , Horses/virology , Animals , Poland/epidemiology , Horse Diseases/virology , Horse Diseases/epidemiology , Picornaviridae Infections/veterinary , Picornaviridae Infections/virology , Picornaviridae Infections/epidemiology , Prevalence , Genetic Variation , Erbovirus/genetics , Erbovirus/isolation & purification , Rhinovirus/genetics , Rhinovirus/isolation & purification , Rhinovirus/classification , Sequence Analysis, DNA
10.
Viruses ; 16(8)2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39205161

ABSTRACT

The SARS-CoV-2 pandemic resulted in a scale-up of viral genomic surveillance globally. However, the wet lab constraints (economic, infrastructural, and personnel) of translating novel virus variant sequence information to meaningful immunological and structural insights that are valuable for the development of broadly acting countermeasures (especially for emerging and re-emerging viruses) remain a challenge in many resource-limited settings. Here, we describe a workflow that couples wastewater surveillance, high-throughput sequencing, phylogenetics, immuno-informatics, and virus capsid structure modeling for the genotype-to-serotype characterization of uncultivated picornavirus sequences identified in wastewater. Specifically, we analyzed canine picornaviruses (CanPVs), which are uncultivated and yet-to-be-assigned members of the family Picornaviridae that cause systemic infections in canines. We analyzed 118 archived (stored at -20 °C) wastewater (WW) samples representing a population of ~700,000 persons in southwest USA between October 2019 to March 2020 and October 2020 to March 2021. Samples were pooled into 12 two-liter volumes by month, partitioned (into filter-trapped solids [FTSs] and filtrates) using 450 nm membrane filters, and subsequently concentrated to 2 mL (1000×) using 10,000 Da MW cutoff centrifugal filters. The 24 concentrates were subjected to RNA extraction, CanPV complete capsid single-contig RT-PCR, Illumina sequencing, phylogenetics, immuno-informatics, and structure prediction. We detected CanPVs in 58.3% (14/24) of the samples generated 13,824,046 trimmed Illumina reads and 27 CanPV contigs. Phylogenetic and pairwise identity analyses showed eight CanPV genotypes (intragenotype divergence <14%) belonging to four clusters, with intracluster divergence of <20%. Similarity analysis, immuno-informatics, and virus protomer and capsid structure prediction suggested that the four clusters were likely distinct serological types, with predicted cluster-distinguishing B-cell epitopes clustered in the northern and southern rims of the canyon surrounding the 5-fold axis of symmetry. Our approach allows forgenotype-to-serotype characterization of uncultivated picornavirus sequences by coupling phylogenetics, immuno-informatics, and virus capsid structure prediction. This consequently bypasses a major wet lab-associated bottleneck, thereby allowing resource-limited settings to leapfrog from wastewater-sourced genomic data to valuable immunological insights necessary for the development of prophylaxis and other mitigation measures.


Subject(s)
High-Throughput Nucleotide Sequencing , Phylogeny , Picornaviridae , Wastewater , Picornaviridae/genetics , Picornaviridae/classification , Picornaviridae/isolation & purification , Animals , Dogs , Wastewater/virology , Capsid Proteins/genetics , Capsid Proteins/chemistry , Genome, Viral , Capsid/immunology , Capsid/chemistry , United States/epidemiology , Picornaviridae Infections/veterinary , Picornaviridae Infections/virology , Picornaviridae Infections/epidemiology , Dog Diseases/virology , Dog Diseases/epidemiology , Genotype , Genetic Variation
11.
Curr Microbiol ; 81(10): 309, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39150576

ABSTRACT

Clarifying the etiology of diarrhea cases of unknown cause is important in the fight against enteric infections. In this study, we aimed to investigate the role of canine kobuvirus (CaKoV), in cases of diarrhea of unknown origin in dogs. A total 121 swab samples from dogs with diarrhea were collected. Molecular analyses of the samples were performed. For this purpose, after the sequence reaction, a phylogenetic tree was created, and bioinformatics analyses were performed. The prevalence rate of CaKoV in the sampled population was determined as 16.5% (20/121). The presence of parvovirus and coronavirus, which are common viral agents in CaKoV-positive dogs, was determined as 35% (7/20) and 10% (2/20), respectively. The rate of dogs with only CaKoV detected was 65% (13/20). Phylogenetic analysis of CaKoV strains clustered together closely related to reference strains. There are very limited studies on the role of CaKoV in the etiology of diarrhea cases of unknown cause in dogs around the world. So far, only one study has been done on CaKoV in Turkey. In this report which includes molecular characterization and epidemiological data on CaKoV determined the importance of CaKoV in cases of diarrhea of unknown origin. More comprehensive studies are needed to better understand the pathogenesis, epidemiology, and biology of CaKoV and to determine effective strategies to combat it.


Subject(s)
Diarrhea , Dog Diseases , Kobuvirus , Phylogeny , Picornaviridae Infections , Dogs , Animals , Kobuvirus/genetics , Kobuvirus/isolation & purification , Kobuvirus/classification , Diarrhea/virology , Diarrhea/veterinary , Diarrhea/epidemiology , Dog Diseases/virology , Picornaviridae Infections/veterinary , Picornaviridae Infections/virology , Picornaviridae Infections/epidemiology , Turkey/epidemiology , Feces/virology
12.
Virol J ; 21(1): 193, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39175061

ABSTRACT

BACKGROUND: Emerging zoonotic diseases arise from cross-species transmission events between wild or domesticated animals and humans, with bats being one of the major reservoirs of zoonotic viruses. Viral metagenomics has led to the discovery of many viruses, but efforts have mainly been focused on some areas of the world and on certain viral families. METHODS: We set out to describe full-length genomes of new picorna-like viruses by collecting feces from hundreds of bats captured in different regions of Spain. Viral sequences were obtained by high-throughput Illumina sequencing and analyzed phylogenetically to classify them in the context of known viruses. Linear discriminant analysis (LDA) was performed to infer likely hosts based on genome composition. RESULTS: We found five complete or nearly complete genomes belonging to the family Picornaviridae, including a new species of the subfamily Ensavirinae. LDA suggested that these were true vertebrate viruses, rather than viruses from the bat diet. Some of these viruses were related to picornaviruses previously found in other bat species from distant geographical regions. We also found a calhevirus genome that most likely belongs to a proposed new family within the order Picornavirales, and for which genome composition analysis suggested a plant host. CONCLUSIONS: Our findings describe new picorna-like viral species and variants circulating in the Iberian Peninsula, illustrate the wide geographical distribution and interspecies transmissibility of picornaviruses, and suggest new hosts for calheviruses.


Subject(s)
Chiroptera , Feces , Genetic Variation , Genome, Viral , Phylogeny , Picornaviridae Infections , Picornaviridae , Chiroptera/virology , Animals , Spain , Picornaviridae/genetics , Picornaviridae/classification , Picornaviridae/isolation & purification , Feces/virology , Picornaviridae Infections/transmission , Picornaviridae Infections/virology , Picornaviridae Infections/veterinary , High-Throughput Nucleotide Sequencing , Metagenomics , Humans , Zoonoses/virology , Zoonoses/transmission
13.
J Vet Med Sci ; 86(9): 986-991, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39069477

ABSTRACT

Fesaviruses, picorna-like RNA viruses, were discovered in 2014 in feces from cats in an animal shelter in the United States but have not since been reported elsewhere. In this study, we collected cat fecal samples from 20 adult cats from an animal shelter in Tokyo, Japan, and examined them for viral pathogens. Next generation sequencing (NGS) was performed to detect both RNA and DNA virus sequences. Sequences of a total of 7 RNA viruses including some common feline pathogenic viruses were detected across 8 samples, while no DNA virus sequences were identified in any sample. Of the RNA virus sequences detected in the samples, two sequences, 4,746 and 4,439 bp, demonstrated 90.3% and 85.0% similarity, respectively, to the fesavirus 4 sequence in the database. To confirm the NGS results, quantitative RT-PCR (qRT-PCR) assays were developed using specific primers and probes designed based on the contig sequences. Based on the qRT-PCR assays, we detected relatively high copy-numbers of fesavirus 4 RNA in the two fecal samples from which the fesavirus 4 sequences were originally obtained, and low copy numbers in other samples. These results demonstrate the presence of fesavirus 4 in cats in Japan for the first time.


Subject(s)
Cat Diseases , Feces , Animals , Cats , Japan/epidemiology , Feces/virology , Cat Diseases/virology , Cat Diseases/diagnosis , Picornaviridae/isolation & purification , Picornaviridae/genetics , Picornaviridae/classification , Picornaviridae Infections/veterinary , Picornaviridae Infections/virology , Picornaviridae Infections/diagnosis , Picornaviridae Infections/epidemiology , High-Throughput Nucleotide Sequencing/veterinary , RNA, Viral/genetics , Male , Female
14.
Vet Microbiol ; 296: 110198, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39067145

ABSTRACT

Senecavirus A (SVA) is a causative agent that can cause vesicular disease in swine, which causes a great threat to the swine husbandry in the world. Therefore, it is necessary to develop a vaccine that can effectively prevent the spread of SVA. In this study, we developed a 24-polymeric nano-scaffold using ß-annulus peptide from tomato bushy effect virus (TBSV) by coupling this antigen to SVA B cell epitope VP121-26 and VP2 proteins via linkers, respectively. The SVA-based nanoparticle protein of the VP1(B)-ß-VP2 was expressed and purified by low-cost prokaryotic system to prepare a SVA nanoparticle vaccine. The immunological protective effect of SVA nanoparticle vaccine was evaluated in mouse and swine models, respectively. The results suggested that both mice and swine could induce high levels SVA neutralizing antibodies and IgG antibodies after two doses immunization. In addition, the swine challenge protection experiment showed that the protection rate of immune SVA nanoparticle vaccine and SVA inactivated vaccine both were 80 %, while the negative control had no protection effect. It demonstrated that SVA nanoparticle vaccine effectively prevented SVA infection in swine. In summary, the preparation of SVA vaccine by using ß-annulus peptide is a promising candidate vaccine for prevent SVA transmission, and provides a new idea for the development of novel SVA vaccines.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Nanovaccines , Picornaviridae Infections , Picornaviridae , Swine Diseases , Viral Vaccines , Animals , Female , Mice , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Capsid Proteins/immunology , Mice, Inbred BALB C , Nanovaccines/administration & dosage , Nanovaccines/immunology , Picornaviridae/immunology , Picornaviridae Infections/veterinary , Picornaviridae Infections/prevention & control , Picornaviridae Infections/immunology , Picornaviridae Infections/virology , Swine , Swine Diseases/prevention & control , Swine Diseases/virology , Swine Diseases/immunology , Viral Structural Proteins/immunology , Viral Vaccines/immunology , Viral Vaccines/administration & dosage
15.
Viruses ; 16(7)2024 Jun 25.
Article in English | MEDLINE | ID: mdl-39066186

ABSTRACT

A diagnostic investigation into an outbreak of fatal respiratory disease among young goats in Iowa, USA revealed bronchitis lesions of unknown etiology and secondary bacterial bronchopneumonia. Hypothesis-free metagenomics identified a previously unreported picornavirus (USA/IA26017/2023), and further phylogenetic analysis classified USA/IA26017/2023 as an aphthovirus related to bovine rhinitis B virus. Viral nucleic acid was localized to lesions of bronchitis using in situ hybridization. This marks the first report of a picornavirus putatively causing respiratory disease in goats and highlights the potential for cross-species transmission of aphthoviruses.


Subject(s)
Bronchitis , Goat Diseases , Goats , Phylogeny , Animals , Goat Diseases/virology , Bronchitis/virology , Bronchitis/veterinary , Picornaviridae Infections/veterinary , Picornaviridae Infections/virology , Iowa , Disease Outbreaks/veterinary , Metagenomics , Cattle
16.
Mol Biol Rep ; 51(1): 777, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38904698

ABSTRACT

BACKGROUND: Senecavirus A (SV-A) is an RNA virus that belongs to the genus Senecavirus within the family Picornaviridae. This study aimed to analyze factors that can influence the molecular diagnosis of Senecavirus A, such as oligonucleotides, RNA extraction methods, and RT-qPCR kits. METHODS: Samples from suspected cases of vesicular disease in Brazilian pigs were analyzed for foot-and-mouth disease, swine vesicular disease, and vesicular stomatitis. All tested negative for these diseases but positive for SV-A. RT-qPCR tests were used, comparing different reagent kits and RNA extraction methods. Sensitivity and repeatability were evaluated, demonstrating efficacy in detecting SV-A in clinical samples. RESULTS: In RNA extraction, significant reduction in Cq values was observed with initial dilutions, particularly with larger supernatant volumes. Trizol and Maxwell showed greater sensitivity in automated equipment protocols, though results varied in tissue tests. RT-qPCR kit comparison revealed differences in amplification using viral RNA but minimal differences with plasmid DNA. Sensitivity among methods was comparable, with slight variations in non-amplified samples. Repeatability tests showed consistent results among RT-qPCRs, demonstrating similarity between methods despite minor discrepancies in Cq values. CONCLUSIONS: Trizol, silica columns, and semi-automated extraction were compared, as well as different RT-qPCR kits. The study found significant variations that could impact the final diagnosis.


Subject(s)
Picornaviridae Infections , Picornaviridae , RNA, Viral , Swine Diseases , Animals , Picornaviridae/genetics , Picornaviridae/isolation & purification , Swine , Picornaviridae Infections/diagnosis , Picornaviridae Infections/veterinary , Picornaviridae Infections/virology , RNA, Viral/genetics , Swine Diseases/virology , Swine Diseases/diagnosis , Sensitivity and Specificity , Real-Time Polymerase Chain Reaction/methods , Swine Vesicular Disease/diagnosis , Swine Vesicular Disease/virology , Foot-and-Mouth Disease/diagnosis , Foot-and-Mouth Disease/virology , Brazil , Reproducibility of Results
17.
Viruses ; 16(6)2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38932208

ABSTRACT

Viruses from Picornaviridae family are known pathogens of poultry, although the information on their occurrence and pathogenicity in pigeons is scarce. In this research, efforts are made to broaden the knowledge on Megrivirus B and Pigeon picornavirus B prevalence, phylogenetic relationship with other avian picornaviruses and their possible connection with enteric disease in racing pigeons. As a result of Oxford Nanopore Sequencing, five Megrivirus and two pigeon picornavirus B-like genome sequences were recovered, among which three recombinant strains were detected. The recombinant fragments represented an average of 10.9% and 25.5% of the genome length of the Pigeon picornavirus B and Megrivirus B reference strains, respectively. The phylogenetic analysis revealed that pigeons are carriers of species-specific picornaviruses. TaqMan qPCR assays revealed 7.8% and 19.0% prevalence of Megrivirus B and 32.2% and 39.7% prevalence of Pigeon picornavirus B in the group of pigeons exhibiting signs of enteropathy and in the group of asymptomatic pigeons, respectively. In turn, digital droplet PCR showed a considerably higher number of genome copies of both viruses in sick than in asymptomatic pigeons. The results of quantitative analysis leave the role of picornaviruses in enteropathies of pigeons unclear.


Subject(s)
Bird Diseases , Columbidae , Genome, Viral , Phylogeny , Picornaviridae Infections , Picornaviridae , Animals , Columbidae/virology , Picornaviridae/genetics , Picornaviridae/classification , Picornaviridae/isolation & purification , Bird Diseases/virology , Picornaviridae Infections/veterinary , Picornaviridae Infections/virology , Recombination, Genetic
18.
Viruses ; 16(6)2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38932262

ABSTRACT

Hepatitis A virus (HAV), a member of the genus Hepatovirus (Picornaviridae HepV), remains a significant viral pathogen, frequently causing enterically transmitted hepatitis worldwide. In this study, we conducted an epidemiological survey of HepVs carried by small terrestrial mammals in the wild in Yunnan Province, China. Utilizing HepV-specific broad-spectrum RT-PCR, next-generation sequencing (NGS), and QNome nanopore sequencing (QNS) techniques, we identified and characterized two novel HepVs provisionally named EpMa-HAV and EpLe-HAV, discovered in the long-tailed mountain shrew (Episoriculus macrurus) and long-tailed brown-toothed shrew (Episoriculus leucops), respectively. Our sequence and phylogenetic analyses of EpMa-HAV and EpLe-HAV indicated that they belong to the species Hepatovirus I (HepV-I) clade II, also known as the Chinese shrew HepV clade. Notably, the codon usage bias pattern of novel shrew HepVs is consistent with that of previously identified Chinese shrew HepV. Furthermore, our structural analysis demonstrated that shrew HepVs differ from other mammalian HepVs in RNA secondary structure and exhibit variances in key protein sites. Overall, the discovery of two novel HepVs in shrews expands the host range of HepV and underscores the existence of genetically diverse animal homologs of human HAV within the genus HepV.


Subject(s)
Genome, Viral , Phylogeny , Shrews , Animals , Shrews/virology , China/epidemiology , RNA, Viral/genetics , Genomics/methods , High-Throughput Nucleotide Sequencing , Picornaviridae Infections/veterinary , Picornaviridae Infections/virology , Picornaviridae Infections/epidemiology
19.
Fish Shellfish Immunol ; 151: 109664, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38844186

ABSTRACT

Mass Mortality Events (MMEs) affecting the noble pen shell Pinna nobilis have been reported since 2016. In this work, we used an in vitro flow cytometric assay to evaluate phagocytosis, coupled with cytology and Electron Microscopy (TEM), to define animal immunocompetence following infection by P. nobilis Picornavirus (PnPV). The study was performed on 27 animals in July 2021 and May 2022 on two natural population from the Ebro Delta (Catalonia, Spain) and animals maintained in captivity at facilities in Valencia and Murcia Aquarium. Hemolymph was collected in the field and in captivity as a non-destructive sampling method. Based on dimension and internal complexity, flow cytometry identified three haemocyte types, distinguished in granulocytes, hyalinocytes and a third type, biggest in size and with high internal complexity and granularity. Those cells corresponded at ultrastructure to hemocytes with advanced phases of PnPV infection and related to cytopathic effect of the replicating virus displaying numerous Double Membrane Vesicles (DMVs) and cells corpse fusion. The results showed that pen shell in captivity had significantly lower Total Hemocyte Count (THC) compared with natural population of Alfacs Bay (mean number of 7-9 x 104 vs 2-5 x 105 cells/mL, respectively). FACS (Fluorescence-activated cell sorting) based phagocytosis analysis demonstrate that animals in captivity at IMEDMAR-UCV and Murcia Aquarium, had scarce or absent ability to phagocyte the two stimuli (Staphylococcus aureus and Zymosan A) (10,2 % ± 1,7 of positives) if compared with the natural population in Alfacs Bay (28,5 % ± 5,6 of positive). Ultrastructure images showed that PnPV itself can lead to an alteration of the hemocyte cytoskeleton, impairing the capabilities to perform an active phagocytosis and an efficient phagolysosome fusion.


Subject(s)
Hemocytes , Picornaviridae , Animals , Picornaviridae/immunology , Hemocytes/immunology , Hemocytes/ultrastructure , Immunocompetence , Bivalvia/immunology , Bivalvia/virology , Phagocytosis , Spain , Flow Cytometry/veterinary , Microscopy, Electron, Transmission/veterinary , Picornaviridae Infections/veterinary , Picornaviridae Infections/immunology , Picornaviridae Infections/virology
20.
BMC Vet Res ; 20(1): 258, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38877537

ABSTRACT

BACKGROUND: Senecavirus A (SVA), identified in 2002, is known to cause porcine idiopathic vesicular disease (PIVD), which presents with symptoms resembling other vesicular diseases. This similarity complicates field diagnosis. Conventional molecular diagnostic techniques are limited by their cost, sensitivity, and requirement for complicated instrumentation. Therefore, developing an effective and accurate diagnostic method is crucial for timely identification and isolation of affected pigs, thereby preventing further disease spread. METHODS: In this study, we developed a highly-specific and ultra-sensitive SVA detection method powered by CRISPR/Cas12a. To enhance the availability in laboratories with varied equipment conditions, microplate reader and ultraviolet light transilluminator were introduced. Moreover, PCR amplification has also been incorporated into this method to improve sensitivity. The specificity and sensitivity of this method were determined following the preparation of the recombinant Cas12a protein and optimization of the CRISPR/Cas12a-based trans-cleavage system. RESULTS: The method demonstrated no cross-reactivity with ten kinds of viruses of swine. The minimum template concentration required to activate substantial trans-cleavage activity was determined to be 106 copies/µL of SVA templates. However, when PCR amplification was incorporated, the method achieved a detection limit of one copy of SVA templates per reaction. It also exhibited 100% accuracy in simulated sample testing. The complete testing process does not exceed three hours. CONCLUSIONS: Importantly, this method utilizes standard laboratory equipment, making it accessible for use in resource-limited settings and facilitating widespread and ultra-sensitive screening during epidemics. Overall, the development of this method not only broadens the array of tools available for detecting SVA but also holds significant promise for controlling the spread of PIVD.


Subject(s)
CRISPR-Cas Systems , Picornaviridae , Sensitivity and Specificity , Swine Diseases , Animals , Swine , Picornaviridae/isolation & purification , Picornaviridae/genetics , Swine Diseases/virology , Swine Diseases/diagnosis , Picornaviridae Infections/veterinary , Picornaviridae Infections/diagnosis , Picornaviridae Infections/virology , Polymerase Chain Reaction/veterinary , Polymerase Chain Reaction/methods , CRISPR-Associated Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL