Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.322
Filter
1.
Front Cell Infect Microbiol ; 14: 1448480, 2024.
Article in English | MEDLINE | ID: mdl-39224701

ABSTRACT

Introduction: Muscovy duck parvovirus (MDPV), Goose parvovirus (GPV), Duck circovirus, (DuCV) and Duck adenovirus 3 (DAdV-3) are important pathogens that cause high morbidity and mortality in ducks, causing huge economic loss for the duck industry. Methods: The present study, a quadruplex one-step real time quantitative PCR method for the detection of MDPV, GPV, DuCV, and DAdV-3 was developed. Results: The results showed that assay had no cross-reactivity with other poultry pathogens [Duck plague virus (DPV), Duck tembusu virus (DTMUV), H6 avian influenza virus (H6 AIV), New duck reovirus (NDRV), Newcastle disease virus (NDV), H4 avian influenza virus (H4 AIV), Escherichia coli (E. coli), Muscovy duck reovirus (MDRV), Egg drop syndrome virus (EDSV), Pasteurella multocida (P. multocida)]. The sensitivity result showed that the limits of detection for MDPV, GPV, DuCV, and DAdV-3 were 10, 10, 1 and 10 copies/µl, respectively; The coefficients of variation intra- and inter-method was 1-2%; The range of linear (109 to 103 copies/µL) demonstrated the R2 values for MDPV, GPV, DuCV, and DAdV-3 as 0.9975, 0.998, 0.9964, and 0.996, respectively. The quadruplex real time quantitative PCR method efficiency was 90.30%, 101.10%, 90.72%, and 90.57% for MDPV, GPV, DuCV, and DAdV-3, respectively. 396 clinical specimens collected in some duck sausages from June 2022 to July 2023 were simultaneously detected using the established quadruplex real time quantitative PCR method and the reported assays. The detection rates for MDPV, GPV, DuCV, and DAdV-3 were 8.33% (33/396), 17.93% (71/396), 33.58% (133/396), and 29.04% (115/396), respectively. The agreement between these assays was greater than 99.56%. Discussion: The developed quadruplex real-time quantitative PCR assay can accurately detect these four viruses infecting ducks, providing a rapid, sensitive, specific and accurate technique for clinical testing.


Subject(s)
Ducks , Poultry Diseases , Real-Time Polymerase Chain Reaction , Animals , Ducks/virology , Real-Time Polymerase Chain Reaction/methods , Poultry Diseases/virology , Poultry Diseases/diagnosis , Sensitivity and Specificity , Parvovirinae/genetics , Parvovirinae/isolation & purification , Parvovirinae/classification , Aviadenovirus/genetics , Aviadenovirus/isolation & purification , Aviadenovirus/classification , Circovirus/genetics , Circovirus/isolation & purification , Circovirus/classification , Parvoviridae Infections/veterinary , Parvoviridae Infections/diagnosis , Parvoviridae Infections/virology
2.
Schweiz Arch Tierheilkd ; 166(9): 460-464, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39225507

ABSTRACT

INTRODUCTION: The novel Porcine circovirus 3 (PCV-3) has been associated in the past years to different porcine diseases, including reproductive failure. The potential occurrence of PCV-3 in abortions from Swiss pig herds has not been investigated so far. Thus, we conducted a retrospective study on pig aborted cases submitted to our laboratory in the University of Bern during the last 10 years with the main aim of investigating the possible presence of PCV-3 in foetal and/or placental tissue. Twelve out of the 53 studied cases showed mild histopathological changes as previously described in PCV-3 positive cases. However, in none of the cases, PCV-3 genetic material could be detected in the examined formalin-fixed, paraffin-embedded tissues. In only one third of the cases, a cause for the abortion was found, which is similar to other studies. Our survey suggests that PCV-3 was not involved in the porcine abortion cases submitted over the last decade at our institution in Switzerland.


INTRODUCTION: Le nouveau Circovirus porcin 3 (PCV-3) a été associé ces dernières années à différentes maladies porcines, y compris des troubles de la reproduction. La présence potentielle du PCV-3 dans les avortements de porcs en Suisse n'a pas été étudiée jusqu'à présent. Nous avons donc mené une étude rétrospective sur les cas d'avortements de porcs soumis à notre laboratoire de l'Université de Berne au cours des 10 dernières années, dans le but principal d'étudier la présence éventuelle du PCV-3 dans les tissus fœtaux et/ou placentaires. Douze des 53 cas étudiés présentaient des changements histopathologiques légers, tels que décrits précédemment dans les cas positifs au PCV-3. Cependant, dans aucun des cas, le matériel génétique du PCV-3 n'a pu être détecté dans les tissus examinés fixés au formol et inclus en paraffine. Dans un tiers des cas seulement, une cause d'avortement a été trouvée, ce qui est similaire à d'autres études. Notre étude suggère que le PCV-3 n'a pas été impliqué dans les cas d'avortements porcins soumis au cours de la dernière décennie dans notre institution en Suisse.


Subject(s)
Abortion, Veterinary , Circoviridae Infections , Circovirus , Swine Diseases , Animals , Female , Pregnancy , Abortion, Veterinary/virology , Circoviridae Infections/veterinary , Circoviridae Infections/virology , Circovirus/isolation & purification , Circovirus/genetics , Formaldehyde , Paraffin Embedding/veterinary , Placenta/virology , Placenta/pathology , Retrospective Studies , Swine , Swine Diseases/virology , Swine Diseases/pathology , Switzerland/epidemiology
3.
Front Immunol ; 15: 1332444, 2024.
Article in English | MEDLINE | ID: mdl-39156896

ABSTRACT

Introduction: Since their identification in 1974, circoviruses have caused clinicopathological diseases in various animal species, including humans. However, their origin, transmission, and genetic evolution remain poorly understood. Methods: In this study, the genome sequences of circovirus were obtained from GenBank, and the Bayesian stochastic search variable selection algorithm was employed to analyzed the evolution and origin of circovirus. Results: Here, the evolutionary origin, mode of transmission, and genetic recombination of the circovirus were determined based on the available circovirus genome sequences. The origin of circoviruses can be traced back to fish circovirus, which might derive from fish genome, and human contributes to transmission of fish circovirus to other species. Furthermore, mosquitos, ticks, bats, and/or rodents might play a role as intermediate hosts in circovirus intra- and inter-species transmission. Two major lineages (A and B) of circoviruses are identified, and frequent recombination events accelerate their variation and spread. The time to the most recent common ancestor of circoviruses can be traced back to around A.D. 600 and has been evolving at a rate of 10-4 substitutions site-1 year-1 for a long time. Discussion: These comprehensive findings shed light on the evolutionary origin, population dynamics, transmission model, and genetic recombination of the circovirus providing valuable insights for the development of prevention and control strategies against circovirus infections.


Subject(s)
Circoviridae Infections , Circovirus , Evolution, Molecular , Phylogeny , Recombination, Genetic , Animals , Humans , Circovirus/genetics , Circoviridae Infections/transmission , Circoviridae Infections/virology , Circoviridae Infections/veterinary , Genome, Viral , Bayes Theorem
4.
Virol J ; 21(1): 184, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39135096

ABSTRACT

Porcine circovirus 3 (PCV3) was first reported in the United States in 2016; this virus is considered to be involved in diverse pathologies, such as multisystem inflammation, porcine dermatitis and nephropathy syndrome, and reproductive disorders. However, successful isolation of PCV3 using cultured cells has been rare. In this study, we aimed to isolate PCV3 using primary porcine bone marrow-derived cells. Mononuclear cells were isolated from the femur bones of clinically healthy pigs. These primary cells were cultured for 6-10 days post-seeding and infected with PCV3-containing tissue homogenates. The cells were cultured for up to 37 days, and the culture medium was changed every 3-4 days. The growth curve of PCV3 in porcine bone marrow cells revealed a decline in growth during the first 10 days post-infection, followed by an increase leading to > 1010 genomic copies/mL of the cell culture supernatant; moreover, the virus was capable of passaging. The indirect fluorescent antibody assay for PCV3 infection revealed the presence of PCV3 capsid protein in the cytoplasm and nuclei of infected cells. Bone marrow cells were passaged for more than 20 generations (over 5 months), and PCV3 persistently infected the cells. PCV3-infected bone marrow cells expressed mesenchymal markers. These results reflect that primary porcine bone marrow-derived mesenchymal cells are permissive to PCV3 and continuously replicate a high copy number of the PCV3 genome. These findings regarding the high replication rate of PCV3 in bone marrow-derived mesenchymal cells could enhance our understanding of PCV3 pathogenicity.


Subject(s)
Bone Marrow Cells , Circovirus , Animals , Swine , Circovirus/physiology , Circovirus/isolation & purification , Circovirus/genetics , Bone Marrow Cells/virology , Cells, Cultured , Circoviridae Infections/virology , Circoviridae Infections/veterinary , Swine Diseases/virology , Capsid Proteins/genetics , Capsid Proteins/metabolism , Virus Cultivation/methods
5.
J Virol ; 98(8): e0022324, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39046246

ABSTRACT

Porcine circovirus type 3 (PCV3) is closely associated with various diseases, such as the porcine dermatitis, nephropathy syndrome, and multisystemic clinicopathological diseases. PCV3-associated diseases are increasingly recognized as severe diseases in the global swine industry. Ring finger protein 2 (RNF2), an E3 ubiquitin ligase exclusively located in the nucleus, contributes to various biological processes. This ligase interacts with the PCV3 Cap. However, its role in PCV3 replication remains unclear. This study confirmed that the nuclear localization signal domain of the Cap and the RNF2 N-terminal RING domain facilitate the interaction between the Cap and RNF2. Furthermore, RNF2 promoted the binding of K48-linked polyubiquitination chains to lysine at positions 139 and 140 (K139 and K140) of the PCV3 Cap, thereby degrading the Cap. RNF2 knockdown and overexpression increased or decreased PCV3 replication, respectively. Moreover, the RING domain-deleted RNF2 mutant eliminated the RNF2-induced degradation of the PCV3 Cap and RNF2-mediated inhibition of viral replication. This indicates that both processes were associated with its E3 ligase activity. Our findings demonstrate that RNF2 can interact with and degrade the PCV3 Cap via its N-terminal RING domain in a ubiquitination-dependent manner, thereby inhibiting PCV3 replication.IMPORTANCEPorcine circovirus type 3 is a recently described pathogen that is prevalent worldwide, causing substantial economic losses to the swine industry. However, the mechanisms through which host proteins regulate its replication remain unclear. Here, we demonstrate that ring finger protein 2 inhibits porcine circovirus type 3 replication by interacting with and degrading the Cap of this pathogen in a ubiquitination-dependent manner, requiring its N-terminal RING domain. Ring finger protein 2-mediated degradation of the Cap relies on its E3 ligase activity and the simultaneous existence of K139 and K140 within the Cap. These findings reveal the mechanism by which this protein interacts with and degrades the Cap to inhibit porcine circovirus type 3 replication. This consequently provides novel insights into porcine circovirus type 3 pathogenesis and facilitates the development of preventative measures against this pathogen.


Subject(s)
Capsid Proteins , Circovirus , Ubiquitin-Protein Ligases , Ubiquitination , Virus Replication , Circovirus/genetics , Circovirus/metabolism , Circovirus/physiology , Animals , Swine , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Capsid Proteins/metabolism , Capsid Proteins/genetics , Humans , HEK293 Cells , Proteolysis , Cell Line , Swine Diseases/virology , Swine Diseases/metabolism , Circoviridae Infections/virology , Circoviridae Infections/metabolism , Protein Binding
6.
BMC Vet Res ; 20(1): 312, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-38997779

ABSTRACT

BACKGROUND: Canine circovirus (CanineCV) is a single-stranded circular DNA virus that infects domestic and wild canids in many countries. CanineCV is associated with gastroenteritis and diarrhea, respiratory disease, and generalized vasculitis leading to a fatal event. The Capsid protein (Cap) is a structural protein of the virus which has high genetic variability and plays a role in the canine immune response. In this study, we cloned the full-length CanineCV Capsid gene (Cap). In-silico analyses were used to explore the genomic and amino acid variability and natural selection acting on the Cap gene. The immune relevance for T-cell and B-cell epitopes was predicted by the immunoinformatic approach. RESULTS: According to the Cap gene, our results showed that CanineCV was separated into five phylogenetic groups. The obtained CanineCV strain from this study was grouped with the previously discovered Thai strain (MG737385), as supported by a haplotype network. Entropy analyses revealed high nucleotide and amino acid variability of the Capsid region. Selection pressure analysis revealed four codons at positions 24, 50, 103, and 111 in the Cap protein evolved under diversifying selection. Prediction of B-cell epitopes exhibited four consensus sequences based on physiochemical properties, and eleven peptide sequences were predicted as T-cell epitopes. In addition, the positive selection sites were located within T-cell and B-cell epitopes, suggesting the role of the host immune system as a driving force in virus evolution. CONCLUSIONS: Our study provides knowledge of CanineCV genetic diversity, virus evolution, and potential epitopes for host cell immune response.


Subject(s)
Capsid Proteins , Circovirus , Phylogeny , Thailand , Circovirus/genetics , Capsid Proteins/genetics , Animals , Dogs , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Epitopes, B-Lymphocyte/genetics , Epitopes, B-Lymphocyte/immunology , Circoviridae Infections/veterinary , Circoviridae Infections/virology , Genetic Variation , Dog Diseases/virology , Amino Acid Sequence
7.
Sci Rep ; 14(1): 15535, 2024 07 05.
Article in English | MEDLINE | ID: mdl-38969759

ABSTRACT

Since PCV4 was first described in 2019, the virus has been identified in several countries in Southeast Asia and Europe. Most studies have been limited to detecting PCV4 by PCR. Thus, PCV4 has an unclear association with clinical disease. This study utilized 512 porcine clinical lung, feces, spleen, serum, lymphoid tissue, and fetus samples submitted to the ISU-VDL from June-September 2023. PCV4 was detected in 8.6% of samples with an average Ct value of 33. While detection rates among sample types were variable, lymphoid tissue had the highest detection rate (18.7%). Two ORF2 sequences were obtained from lymphoid tissue samples and had 96.36-98.98% nucleotide identity with reference sequences. Direct detection of PCV4 by RNAscope revealed viral replication in B lymphocytes and macrophages in lymph node germinal centers and histiocytic and T lymphocyte infiltration in the lamina propria of the small intestine. PCV4 detection was most commonly observed in nursery to finishing aged pigs displaying respiratory and enteric disease. Coinfection with PCV2, PCV3, and other endemic pathogens was frequently observed, highlighting the complex interplay between different PCVs and their potential roles in disease pathogenesis. This study provides insights into the frequency of detection, tissue distribution, and genetic characteristics of PCV4 in the US.


Subject(s)
Circoviridae Infections , Circovirus , Swine Diseases , Animals , Circovirus/genetics , Circovirus/isolation & purification , Swine , Circoviridae Infections/veterinary , Circoviridae Infections/virology , Swine Diseases/virology , United States/epidemiology , Lymphoid Tissue/virology , Coinfection/virology , Coinfection/veterinary , Lung/virology
8.
Sheng Wu Gong Cheng Xue Bao ; 40(7): 2333-2345, 2024 Jul 25.
Article in Chinese | MEDLINE | ID: mdl-39044595

ABSTRACT

The aim of this study was to establish a rapid method for constructing infectious clones of porcine circovirus type 2 (PCV2). In this study, we constructed circular infectious clones of PCV2 by seamless cloning technology, using the clinically isolated strain PCV2-LX as a template. Meanwhile, this method was compared with the conventional restriction-ligation approach, focusing on the in vitro circularization (self-ligation) process of the genome and the growth characteristics of rescued viruses. The results showed that this method eliminates the need to analyze and introduce restriction endonuclease sites, thus avoiding the complexities associated with traditional restriction enzyme-based cloning steps. It offers a simple and rapid operation, enabling more efficient editing of the PCV2 genome. The infectious clones constructed using this method could be successfully rescued through liposome transfection, resulting in the production of recombinant viruses that could be stably passaged. Moreover, the recombinant viruses rescued by this method exhibited enhanced proliferative capacity in PK-15 cells and 3D4/31 cells (immortalized porcine alveolar macrophages). In conclusion, this study has established a novel reverse genetics system for PCV2, providing a new strategy for the development of PCV2 genetic engineering vaccines. Additionally, it serves as a reference for the construction of infectious clones for other emerging circoviruses such as PCV3 and PCV4.


Subject(s)
Circovirus , DNA, Viral , Circovirus/genetics , Swine , Animals , DNA, Viral/genetics , Cloning, Molecular , Genome, Viral , Reverse Genetics/methods , Circoviridae Infections/virology , Cell Line
9.
BMC Vet Res ; 20(1): 328, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-39033103

ABSTRACT

BACKGROUND: Canine circovirus (CanineCV), a non-enveloped virus with a circular DNA genome, has been identified in various avian and mammalian species, including domestic and wild canids. This study aimed to comprehensively analyze the prevalence of CanineCV across diverse animal species in 11 provinces of China. RESULTS: A total of 1,666 serum samples were collected, revealing a 5.82% prevalence of CanineCV in dogs, with the highest rates being observed in southern and eastern China. Phylogenetic analysis of 266 global CanineCV genomes sourced from the NCBI identified six distinct genotypes, elucidating the complex dynamics of their evolution. Evidence suggested a potential bat origin for CanineCV, with positive selection and high rates of evolution being observed. Recombination analysis revealed dynamic genetic exchange, highlighting the intricate nature of CanineCV evolution. Mutational analysis identified key amino acid substitutions likely to influence the virus's adaptation. Additionally, glycosylation, palmitoylation, and SUMOylation sites were predicted, shedding light on crucial functional properties of the virus. CONCLUSIONS: This study provides a global perspective on the origin, genetic diversity, and evolutionary dynamics of CanineCV. Understanding these factors is crucial for elucidating its epidemiology and potential health risks.


Subject(s)
Circoviridae Infections , Circovirus , Dog Diseases , Phylogeny , Animals , Circovirus/genetics , Circovirus/classification , Dogs , Dog Diseases/virology , Dog Diseases/epidemiology , China/epidemiology , Circoviridae Infections/veterinary , Circoviridae Infections/epidemiology , Circoviridae Infections/virology , Evolution, Molecular , Genome, Viral , Genetic Variation , Prevalence , Genotype
10.
Front Immunol ; 15: 1438371, 2024.
Article in English | MEDLINE | ID: mdl-39081314

ABSTRACT

Introduction: Variant pseudorabies virus (PRV) is a newly emerged zoonotic pathogen that can cause human blindness. PRV can take advantage of its large genome and multiple non-essential genes to construct recombinant attenuated vaccines carrying foreign genes. However, a major problem is that the foreign genes in recombinant PRV are only integrated into the genome for independent expression, rather than assembled on the surface of virion. Methods: We reported a recombinant PRV with deleted gE/TK genes and an inserted porcine circovirus virus 2 (PCV2) Cap gene into the extracellular domain of the PRV gE gene using the Cre-loxP recombinant system combined with the CRISPR-Cas9 gene editing system. This recombinant PRV (PRV-Cap), with the envelope-embedded Cap protein, exhibits a similar replication ability to its parental virus. Results: An immunogenicity assay revealed that PRV-Cap immunized mice have 100% resistance to lethal PRV and PCV2 attacks. Neutralization antibody and ELISPOT detections indicated that PRV-Cap can enhance neutralizing antibodies to PRV and produce IFN-γ secreting T cells specific for both PRV and PCV2. Immunological mechanistic investigation revealed that initial immunization with PRV-Cap stimulates significantly early activation and expansion of CD69+ T cells, promoting the activation of CD4 Tfh cell dependent germinal B cells and producing effectively specific effector memory T and B cells. Booster immunization with PRV-Cap recalled the activation of PRV-specific IFN-γ+IL-2+CD4+ T cells and IFN-γ+TNF-α+CD8+ T cells, as well as PCV2-specific IFN-γ+TNF-α+CD8+ T cells. Conclusion: Collectively, our data suggested an immunological mechanism in that the recombinant PRV with envelope-assembled PCV2 Cap protein can serve as an excellent vaccine candidate for combined immunity against PRV and PCV2, and provided a cost-effective method for the production of PRV- PCV2 vaccine.


Subject(s)
Circoviridae Infections , Circovirus , Herpesvirus 1, Suid , Animals , Circovirus/immunology , Circovirus/genetics , Mice , Herpesvirus 1, Suid/immunology , Herpesvirus 1, Suid/genetics , Circoviridae Infections/immunology , Circoviridae Infections/prevention & control , Antibodies, Viral/immunology , Antibodies, Viral/blood , Viral Vaccines/immunology , Viral Envelope Proteins/immunology , Viral Envelope Proteins/genetics , Swine , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Capsid Proteins/immunology , Capsid Proteins/genetics , Vaccines, Synthetic/immunology , Pseudorabies/immunology , Pseudorabies/prevention & control , Female , Mice, Inbred BALB C
11.
Vet J ; 307: 106199, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39038778

ABSTRACT

Porcine circoviruses (PCVs) contain four types: PCV1, PCV2, PCV3, and PCV4, all of which can infect pigs. Among them, PCV1 is non-pathogenic, and PCV2 can cause porcine circovirus diseases (PCVD) or porcine circovirus-associated diseases (PCVAD). Although the pathogenicity of PCV3 and PCV4 is still controversial, increasing evidence shows that PCV3 and PCV4 can cause PCV-related disease. However, mixed infection of PCV2, PCV3, and PCV4 with other pathogens often occurs in large-scale pig breeding, bringing severe economic losses to the global pig industry. In this study, the soluble recombinant proteins of PCV2, PCV3, and PCV4 Cap were expressed by the prokaryotic expression system and biotinylated to combine with the Streptavidin magnetic beads, followed by immunogenicity evaluation of the recombinant proteins. Furthermore, we also assessed the efficacy and immunogenicity of trivalent recombinant proteins conjugated with different adjuvants in mice. The results showed that the highly effective anti-PCV serum was successfully prepared, and the recombinant proteins conjugated with different adjuvants produced various degrees of humoral and cellular immunity in mice. Three recombinant proteins are effective immunogens, and the trivalent proteins coupled with the aluminum adjuvant or GM-CSF-CpG for two-dose immunization can stimulate prominent humoral and cellular immunity against PCVs in vivo. The soluble recombinant proteins are the most promising candidate for developing a trivalent vaccine against PCVs (PCV2, PCV3, and PCV4) infection simultaneously.


Subject(s)
Capsid Proteins , Circoviridae Infections , Circovirus , Circovirus/immunology , Circovirus/genetics , Animals , Capsid Proteins/genetics , Capsid Proteins/immunology , Circoviridae Infections/veterinary , Circoviridae Infections/prevention & control , Circoviridae Infections/virology , Circoviridae Infections/immunology , Swine , Mice , Swine Diseases/virology , Swine Diseases/prevention & control , Swine Diseases/immunology , Recombinant Proteins/immunology , Female , Viral Vaccines/immunology , Mice, Inbred BALB C , Antibodies, Viral/blood
12.
Res Vet Sci ; 176: 105336, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38880017

ABSTRACT

Circoviruses cause severe disease in pigs and birds. Canine circovirus has thus far only been associated with respiratory and gastrointestinal disorders and systemic disease in dogs. The Iberian lynx (Lynx pardinus) is one of the most endangered carnivores in Europe and the most endangered felid worldwide. Exploring the virome of these animals may be important in terms of virus discovery and assessing the interspecies-circulation of viruses from related carnivores. In this study, 162 spleen samples from Iberian lynx were screened for CRESS DNA viruses. Overall, 11 (6.8%) of 162 samples tested positive using a consensus PCR. Partial rep sequences were tightly related to each other (96.6-100%). Specific molecular protocols were designed on the partial rep sequences of the novel virus, Iberian lynx-associated circovirus-1 (ILCV-1). By screening a subset of 45 spleen samples, the infection rate of ILCV-1 in Iberian lynxes was 57.8% (26/45). ILCV-1 strains formed a separate cluster intermingled with bat, rodent, mongoose, and felid circoviruses. The genome of the novel virus displayed the highest nucleotide identity (64.3-65.3%) to mongoose circoviruses, thus representing a novel candidate circovirus species. The detection of these viruses in the spleen tissues could suggest systemic infection in the animal host. Overall, these findings suggest that this novel circovirus is common in the Iberian lynx. Further studies are warranted to assess the possible health implications of ILCV-1 in this endangered species.


Subject(s)
Circoviridae Infections , Circovirus , Lynx , Phylogeny , Animals , Circovirus/genetics , Circovirus/isolation & purification , Circovirus/classification , Lynx/virology , Circoviridae Infections/veterinary , Circoviridae Infections/virology , Circoviridae Infections/epidemiology , Spain , Spleen/virology , Genome, Viral , Polymerase Chain Reaction/veterinary
13.
Vet Microbiol ; 295: 110151, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38870752

ABSTRACT

Porcine circovirus type 2 (PCV2) stands as a predominant etiological agent in porcine circovirus-associated diseases. To manage the spread of the disease, it is necessary to develop a next-generation vaccine expressing PCV2 antigens that target the prevailing genotype such as PCV2d. A bacterial-mediated vaccine delivery by live-attenuated Salmonella has attracted interest for its low-cost production and highly effective vaccine delivery. Thus, in this study, we utilized the advantages of the Salmonella-mediated vaccine delivery by cloning PCV2d cap and rep into a eukaryotic expression plasmid pJHL204 and electroporation into an engineered live-attenuated Salmonella Typhimurium JOL2500 (Δlon, ΔcpxR, ΔsifA, Δasd). The eukaryotic antigen expression by JOL2995 (p204:cap) and JOL2996 (p204:rep) was confirmed in vitro and in vivo which showed efficient antigen delivery. Furthermore, vaccination of mice model with the vaccine candidates elicited humoral and cell-mediated immune responses as depicted by high levels of PCV2-specific antibodies, CD4+ and CD8+ T cells, and neutralizing antibodies, especially by JOL2995 (p204:cap) which correlated with the significant decrease in the viral load in PCV2d-challenged mice. Interestingly, JOL2996 (p204:rep) may not have elicited high levels of neutralizing antibodies and protective efficacy, but it elicited considerably higher cell-mediated immune responses. This study demonstrated Salmonella-mediated vaccine delivery system coupled with the eukaryotic expression vector can efficiently deliver and express the target PCV2d antigens for strong induction of immune response and protective efficacy in mice model, further supporting the potential application of the Salmonella-mediated vaccine delivery system as an effective novel approach in vaccine strategies for PCV2d.


Subject(s)
Circoviridae Infections , Circovirus , Genetic Vectors , Salmonella typhimurium , Viral Vaccines , Animals , Circovirus/immunology , Circovirus/genetics , Mice , Salmonella typhimurium/immunology , Salmonella typhimurium/genetics , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Circoviridae Infections/prevention & control , Circoviridae Infections/veterinary , Circoviridae Infections/immunology , Swine , Antigens, Viral/immunology , Antigens, Viral/genetics , Mice, Inbred BALB C , Antibodies, Viral/blood , Female , Antibodies, Neutralizing/blood , Capsid Proteins/immunology , Capsid Proteins/genetics , Swine Diseases/prevention & control , Swine Diseases/immunology , Swine Diseases/virology
14.
Virus Res ; 347: 199428, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38942295

ABSTRACT

In China, a novel pathogen within the genus Circovirus has been identified as a causative agent of the 'novel acute hemorrhage syndrome' (NAHS) in aquacultured populations of turbot (Scophthalmus maximus L.). Histopathological examination using light microscopy revealed extensive necrosis within the cardiac, splenic, and renal tissues of the afflicted fish. Utilizing transmission electron microscopy (TEM), we detected the presence of circovirus particles within the cytoplasm of these cells, with the virions consistently exhibiting a spherical morphology of 20-40 nm in diameter. TEM inspections confirmed the predominance of these virions in the heart, spleen, and kidney. Subsequent molecular characterization through polymerase chain reaction (PCR) analysis corroborated the TEM findings, with positive signals in the aforementioned tissues, in stark contrast to the lack of detection in gill, fin, liver, and intestinal tissues. The TEM observations, supported by PCR electrophoresis data, strongly suggest that the spleen and kidney are the primary targets of the viral infection. Further characterization using biophysical, biochemical assays, and genomic sequencing confirmed the viral classification within the genus Circovirus, resulting in the nomenclature of turbot circovirus (TurCV). The current research endeavors to shed light on the pathogenesis of this pathogen, offering insights into the infection mechanisms of TurCV in this novel piscine host, thereby contributing to the broader understanding of its impact on turbot health and aquaculture.


Subject(s)
Circoviridae Infections , Circovirus , Fish Diseases , Flatfishes , Genome, Viral , Phylogeny , Animals , Circovirus/genetics , Circovirus/classification , Circovirus/isolation & purification , China , Circoviridae Infections/veterinary , Circoviridae Infections/virology , Circoviridae Infections/pathology , Fish Diseases/virology , Flatfishes/virology , Microscopy, Electron, Transmission , Genomics , Kidney/virology , Kidney/pathology , Spleen/virology , Spleen/pathology
15.
mSphere ; 9(7): e0022524, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-38926905

ABSTRACT

Porcine circovirus type 4 (PCV4), a recently identified circovirus, is prevalent in numerous provinces in China, as well as in South Korea, Thailand, and Europe. PCV4 virus rescued from an infectious clone showed pathogenicity, suggesting the economic impact of PCV4. However, there remains a lack of understanding regarding the immunogenicity and epitopes of PCV4. This study generated a monoclonal antibody (MAb) 1D8 by immunizing mice with PCV4 virus-like particles (VLPs). Subsequently, the epitope recognized by the MAb 1D8 was identified by truncated protein expression and alanine scanning mutagenesis analysis. Results showed that the 225PKQG228 located at the C-terminus of the PCV4 Cap protein is the minimal motif binding to the MAb. Homology modeling analysis and immunoelectron microscopy revealed that the epitope extends beyond the outer surface of the PCV4 VLP. Moreover, the epitope is highly conserved among PCV4 strains and does not react with other PCVs. Together, the MAb 1D8 recognized epitope shows potential for detecting PCV4. These findings significantly contribute to the design of antigens for PCV4 detection and control strategies. IMPORTANCE: Porcine circovirus type 4 (PCV4) is a novel circovirus. Although PCV4 has been identified in several countries, including China, Korea, Thailand, and Spain, no vaccine is available. Given the potential pathogenic effects of PCV4 on pigs, PCV4 could threaten the global pig farming industry, highlighting the urgency for further investigation. Thus, epitopes of PCV4 remain to be determined. Our finding of a conserved epitope significantly advances vaccine development and pathogen detection.


Subject(s)
Antibodies, Monoclonal , Antibodies, Viral , Capsid Proteins , Circovirus , Epitopes, B-Lymphocyte , Circovirus/immunology , Circovirus/genetics , Capsid Proteins/immunology , Capsid Proteins/genetics , Capsid Proteins/chemistry , Animals , Antibodies, Monoclonal/immunology , Mice , Swine , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/genetics , Antibodies, Viral/immunology , Mice, Inbred BALB C , Circoviridae Infections/veterinary , Circoviridae Infections/immunology , Circoviridae Infections/virology , Swine Diseases/virology , Swine Diseases/immunology , Female
16.
Poult Sci ; 103(8): 103936, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38909502

ABSTRACT

Goose circovirus (GoCV) is a common pathogen that causes immunosuppression and promotes secondary infections with other infectious agents in geese worldwide. In the present study, we identified GoCV in 2 out of 93 duck flocks from China and successfully sequenced the complete genomes of 2 strains (AH22du and HN20du). The whole genome of the two strains shared a high identity of 90.5 to 98.63% with China GoCV reference, and low identity of 58.98% with DuCV reference, respectively. Phylogenetic tree constructed on the two and other genome sequences of GoCV revealed three main branches. Both strains sequenced in this study were distributed on different sub-branches with most other Chinese GoCV strains, and AH22du clustered into an independent sub-branch within the cluster. Recombination analysis predicted that HN20du might potentially recombine from the major parent of yk4 (Zhejiang Province, China, 2007) and minor parent of GD/YJ/g2 (Guangdong Province, China, 2020). To the best of our knowledge, this is the first report of GoCV in ducks from China. This broadened host spectrum of GoCVs requires attention from the waterfowl industry and researchers.


Subject(s)
Circoviridae Infections , Circovirus , Ducks , Phylogeny , Poultry Diseases , Animals , Ducks/virology , Circovirus/genetics , Circovirus/isolation & purification , Circovirus/classification , China , Circoviridae Infections/veterinary , Circoviridae Infections/virology , Circoviridae Infections/epidemiology , Poultry Diseases/virology , Poultry Diseases/epidemiology , Genome, Viral , Geese/virology
17.
Sci Rep ; 14(1): 13815, 2024 06 15.
Article in English | MEDLINE | ID: mdl-38877168

ABSTRACT

This study was aimed to investigate the frequency of PiCV recombination, the kinetics of PiCV viremia and shedding and the correlation between viral replication and host immune response in young pigeons subclinically infected with various PiCV variants and kept under conditions mimicking the OLR system. Fifteen racing pigeons originating from five breeding facilities were housed together for six weeks. Blood and cloacal swab samples were collected from birds every seven days to recover complete PiCV genomes and determine PiCV genetic diversity and recombination dynamics, as well as to assess virus shedding rate, level of viremia, expression of selected genes and level of anti-PiCV antibodies. Three hundred and eighty-eight complete PiCV genomes were obtained and thirteen genotypes were distinguished. Twenty-five recombination events were detected. Recombinants emerged during the first three weeks of the experiment which was consistent with the peak level of viremia and viral shedding. A further decrease in viremia and shedding partially corresponded with IFN-γ and MX1 gene expression and antibody dynamics. Considering the role of OLR pigeon rearing system in spreading infectious agents and allowing their recombination, it would be reasonable to reflect on the relevance of pigeon racing from both an animal welfare and epidemiological perspective.


Subject(s)
Bird Diseases , Circoviridae Infections , Circovirus , Columbidae , Virus Shedding , Animals , Columbidae/virology , Circovirus/genetics , Circovirus/immunology , Circoviridae Infections/veterinary , Circoviridae Infections/virology , Circoviridae Infections/epidemiology , Circoviridae Infections/immunology , Bird Diseases/virology , Bird Diseases/epidemiology , Bird Diseases/immunology , Viremia/epidemiology , Viremia/virology , Viremia/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Genome, Viral , Recombination, Genetic , Genotype , Virus Replication , Phylogeny
18.
Viruses ; 16(6)2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38932224

ABSTRACT

Porcine parvoviruses (PPVs) are among the most important agents of reproductive failure in swine worldwide. PPVs comprise eight genetically different species ascribed to four genera: Protoparvovirus (PPV1, PPV8), Tetraparvovirus (PPV2-3), Copiparvovirus (PPV4-6), and Chaphamaparvovirus (PPV7). In 2016, PPV7 was firstly detected in the USA and afterwards in Europe, Asia, and South America. Recently, it was also identified in Italy in pig farms with reproductive failure. This study aimed to evaluate the circulation of PPV7 in domestic and wild pigs in Sardinia, Italy. In addition, its coinfection with Porcine Circovirus 2 (PCV2) and 3 (PCV3) was analysed, and PPV7 Italian strains were molecularly characterised. PPV7 was detected in domestic pigs and, for the first time, wild pigs in Italy. The PPV7 viral genome was detected in 20.59% of domestic and wild pig samples. PPV7 detection was significantly lower in domestic pigs, with higher PCV2/PCV3 co-infection rates observed in PPV7-positive than in PPV7-negative domestic pigs. Molecular characterisation of the NS1 gene showed a very high frequency of recombination that could presumably promote virus spreading.


Subject(s)
Coinfection , Parvoviridae Infections , Parvovirus, Porcine , Phylogeny , Swine Diseases , Animals , Parvovirus, Porcine/genetics , Parvovirus, Porcine/classification , Parvovirus, Porcine/isolation & purification , Italy/epidemiology , Parvoviridae Infections/veterinary , Parvoviridae Infections/epidemiology , Parvoviridae Infections/virology , Swine , Swine Diseases/virology , Swine Diseases/epidemiology , Coinfection/virology , Coinfection/veterinary , Coinfection/epidemiology , Genome, Viral , Circovirus/genetics , Circovirus/classification , Circovirus/isolation & purification , Circoviridae Infections/veterinary , Circoviridae Infections/virology , Circoviridae Infections/epidemiology , DNA, Viral/genetics
19.
Sci Rep ; 14(1): 10263, 2024 05 04.
Article in English | MEDLINE | ID: mdl-38704425

ABSTRACT

We report the first detection and prevalence of Beak and feather disease virus (BFDV) in Australia's Red Goshawk (Erythrotriorchis radiatus). This is a new host for this pervasive pathogen amongst a growing list of non-psittacine species including birds of prey from the orders Accipitriformes (hawks, eagles, kites), Falconiformes (falcons and caracas), and Strigiformes (owls). The Red Goshawk is the first non-psittacine species listed as Endangered to be diagnosed with BFDV. We report an initial case of infection discovered post-mortem in a dead nestling and subsequent surveillance of birds from across northern Australia. We reveal BFDV prevalence rates in a wild raptor population for the first time, with detections in 25% (n = 7/28) of Red Goshawks sampled. Prevalence appears higher in juveniles compared to adults, although not statistically significant, but is consistent with studies of wild psittacines. BFDV genotypes were associated with the Loriinae (lorikeets, budgerigar, and fig parrots), Cacatuini (Cockatoos), and Polytelini (long-tailed parrots) tribes; species which are preyed upon by Red Goshawks. A positive BFDV status may be associated with lower body mass but small sample sizes precluded robust statistical analysis. We postulate the possible impacts of the virus on Red Goshawks and discuss future research priorities given these preliminary observations.


Subject(s)
Bird Diseases , Circoviridae Infections , Circovirus , Endangered Species , Animals , Bird Diseases/virology , Bird Diseases/epidemiology , Circoviridae Infections/epidemiology , Circoviridae Infections/veterinary , Circoviridae Infections/virology , Circovirus/genetics , Circovirus/isolation & purification , Hawks/virology , Australia/epidemiology , Phylogeny , Prevalence , Genotype
20.
Arch Virol ; 169(5): 115, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38709425

ABSTRACT

Porcine circoviruses (PCVs) are a significant cause of concern for swine health, with four genotypes currently recognized. Two of these, PCV3 and PCV4, have been detected in pigs across all age groups, in both healthy and diseased animals. These viruses have been associated with various clinical manifestations, including porcine dermatitis and nephropathy syndrome (PDNS) and respiratory and enteric signs. In this study, we detected PCV3 and PCV4 in central China between January 2022 and February 2023. We tested fecal swabs and tissue samples from growing-finishing and suckling pigs with or without respiratory and systemic manifestations and found the prevalence of PCV3 to be 15.15% (15/99) and that of PCV3/PCV4 coinfection to be 4.04% (4/99). This relatively low prevalence might be attributed to the fact that most of the clinical samples were collected from pigs exhibiting respiratory signs, with only a few samples having been obtained from pigs with diarrhea. In some cases, PCV2 was also detected, and the coinfection rates of PCV2/3, PCV2/4, and PCV2/3/4 were 6.06% (6/99), 5.05% (5/99), and 3.03% (3/99), respectively. The complete genomic sequences of four PCV3 and two PCV4 isolates were determined. All four of the PCV3 isolates were of subtype PCV3b, and the two PCV4 isolates were of subtype PCV4b. Two mutations (A24V and R27K) were found in antibody recognition domains of PCV3, suggesting that they might be associated with immune escape. This study provides valuable insights into the molecular epidemiology and evolution of PCV3 and PCV4 that will be useful in future investigations of genotyping, immunogenicity, and immune evasion strategies.


Subject(s)
Circoviridae Infections , Circovirus , Genotype , Phylogeny , Swine Diseases , Circovirus/genetics , Circovirus/isolation & purification , Circovirus/classification , Animals , Swine , China/epidemiology , Swine Diseases/virology , Swine Diseases/epidemiology , Circoviridae Infections/veterinary , Circoviridae Infections/virology , Circoviridae Infections/epidemiology , Coinfection/virology , Coinfection/veterinary , Coinfection/epidemiology , Genome, Viral/genetics , Feces/virology
SELECTION OF CITATIONS
SEARCH DETAIL