Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
J Gen Virol ; 104(11)2023 11.
Article in English | MEDLINE | ID: mdl-37976092

ABSTRACT

Virus vectored vaccines are not available commercially for cattle even though compelling potential applications exist. Bovine papular stomatitis virus (BPSV), a highly prevalent parapoxvirus, causes self-limited oral lesions in cattle. Ability of virus to accommodate large amounts of foreign DNA, induce low level of antiviral immunity, and circulate and likely persist in cattle populations, make BPSV an attractive candidate viral vector. Here, recombinant BPSV were constructed expressing either Bovine herpesvirus 1 (BoHV-1) glycoprotein gD (BPSVgD), or gD and gB (BPSVgD/gB). Immunization of BPSV serologically-positive calves with BPSVgD or BPSVgD/gB induced BoHV-1 neutralization antibodies and provided protection for three of four animals following a high dose BoHV-1 challenge at day 70 pi. Results indicate BPSV suitability as a candidate virus vector for cattle vaccines.


Subject(s)
Cattle Diseases , Herpesvirus 1, Bovine , Parapoxvirus , Stomatitis , Vaccines , Viral Vaccines , Cattle , Animals , Parapoxvirus/genetics , Antibodies, Viral , Herpesvirus 1, Bovine/genetics , Viral Vaccines/genetics , Cattle Diseases/prevention & control
2.
PLoS Pathog ; 17(10): e1009971, 2021 10.
Article in English | MEDLINE | ID: mdl-34614034

ABSTRACT

Viruses have evolved mechanisms to subvert critical cellular signaling pathways that regulate a wide range of cellular functions, including cell differentiation, proliferation and chemotaxis, and innate immune responses. Here, we describe a novel ORFV protein, ORFV113, that interacts with the G protein-coupled receptor Lysophosphatidic acid receptor 1 (LPA1). Consistent with its interaction with LPA1, ORFV113 enhances p38 kinase phosphorylation in ORFV infected cells in vitro and in vivo, and in cells transiently expressing ORFV113 or treated with soluble ORFV113. Infection of cells with virus lacking ORFV113 (OV-IA82Δ113) significantly decreased p38 phosphorylation and viral plaque size. Infection of cells with ORFV in the presence of a p38 kinase inhibitor markedly diminished ORFV replication, highlighting importance of p38 signaling during ORFV infection. ORFV113 enhancement of p38 activation was prevented in cells in which LPA1 expression was knocked down and in cells treated with LPA1 inhibitor. Infection of sheep with OV-IA82Δ113 led to a strikingly attenuated disease phenotype, indicating that ORFV113 is a major virulence determinant in the natural host. Notably, ORFV113 represents the first viral protein that modulates p38 signaling via interaction with LPA1 receptor.


Subject(s)
MAP Kinase Signaling System/physiology , Poxviridae Infections/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Viral Proteins/metabolism , Animals , Parapoxvirus , Sheep
3.
Viruses ; 13(8)2021 07 28.
Article in English | MEDLINE | ID: mdl-34452346

ABSTRACT

African swine fever (ASF) is a hemorrhagic disease of swine characterized by massive lymphocyte depletion in lymphoid tissues due to the apoptosis of B and T cells, a process likely triggered by factors released or secreted by infected macrophages. ASFV CD2v (EP402R) has been implicated in viral virulence and immunomodulation in vitro; however, its actual function(s) remains unknown. We found that CD2v expression in swine PK15 cells induces NF-κB-dependent IFN-ß and ISGs transcription and an antiviral state. Similar results were observed for CD2v protein treated swine PBMCs and macrophages, the major ASFV target cell. Notably, treatment of swine PBMCs and macrophages with CD2v protein induced apoptosis. Immunoprecipitation and colocalization studies revealed that CD2v interacts with CD58, the natural host CD2 ligand. Additionally, CD58 knockdown in cells or treatment of cells with an NF-κB inhibitor significantly reduced CD2v-mediated NF-κB activation and IFN-ß induction. Further, antibodies directed against CD2v inhibited CD2v-induced NF-κB activation and IFN-ß transcription in cells. Overall, results indicate that ASFV CD2v activates NF-κB, which induces IFN signaling and apoptosis in swine lymphocytes/macrophages. We propose that CD2v released from infected macrophages may be a significant factor in lymphocyte apoptosis observed in lymphoid tissue during ASFV infection in pigs.


Subject(s)
African Swine Fever Virus/metabolism , African Swine Fever/genetics , African Swine Fever/physiopathology , Interferon-beta/genetics , Leukocytes, Mononuclear/cytology , Viral Proteins/metabolism , African Swine Fever/immunology , African Swine Fever/virology , African Swine Fever Virus/genetics , Animals , Apoptosis , Interferon-beta/immunology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/virology , Macrophages/immunology , Macrophages/virology , NF-kappa B/genetics , NF-kappa B/immunology , Swine , Viral Proteins/genetics
4.
PLoS Pathog ; 13(12): e1006779, 2017 12.
Article in English | MEDLINE | ID: mdl-29244863

ABSTRACT

Poxviruses have evolved multiple strategies to subvert signaling by Nuclear Factor κB (NF-κB), a crucial regulator of host innate immune responses. Here, we describe an orf virus (ORFV) virion-associated protein, ORFV119, which inhibits NF-κB signaling very early in infection (≤ 30 min post infection). ORFV119 NF-κB inhibitory activity was found unimpaired upon translation inhibition, suggesting that virion ORFV119 alone is responsible for early interference in signaling. A C-terminal LxCxE motif in ORFV119 enabled the protein to interact with the retinoblastoma protein (pRb) a multifunctional protein best known for its tumor suppressor activity. Notably, experiments using a recombinant virus containing an ORFV119 mutation which abrogates its interaction with pRb together with experiments performed in cells lacking or with reduced pRb levels indicate that ORFV119 mediated inhibition of NF-κB signaling is largely pRb dependent. ORFV119 was shown to inhibit IKK complex activation early in infection. Consistent with IKK inhibition, ORFV119 also interacted with TNF receptor associated factor 2 (TRAF2), an adaptor protein recruited to signaling complexes upstream of IKK in infected cells. ORFV119-TRAF2 interaction was enhanced in the presence of pRb, suggesting that ORFV119-pRb complex is required for efficient interaction with TRAF2. Additionally, transient expression of ORFV119 in uninfected cells was sufficient to inhibit TNFα-induced IKK activation and NF-κB signaling, indicating that no other viral proteins are required for the effect. Infection of sheep with ORFV lacking the ORFV119 gene led to attenuated disease phenotype, indicating that ORFV119 contributes to virulence in the natural host. ORFV119 represents the first poxviral protein to interfere with NF-κB signaling through interaction with pRb.


Subject(s)
NF-kappa B/physiology , Orf virus/physiology , Orf virus/pathogenicity , Retinoblastoma Protein/physiology , Viral Proteins/physiology , Amino Acid Sequence , Animals , Cell Line , Cells, Cultured , Ecthyma, Contagious/etiology , Gene Knockdown Techniques , Genes, Viral , HEK293 Cells , HeLa Cells , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/physiology , Humans , I-kappa B Kinase/metabolism , Immunity, Innate , Mutation , NF-kappa B/antagonists & inhibitors , Orf virus/genetics , Retinoblastoma Protein/antagonists & inhibitors , Retinoblastoma Protein/genetics , Sheep , Signal Transduction , TNF Receptor-Associated Factor 2/metabolism , Viral Proteins/genetics , Viral Proteins/immunology , Virulence/genetics , Virulence/immunology , Virulence/physiology
5.
PLoS Pathog ; 13(8): e1006561, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28787456

ABSTRACT

Poxviruses have evolved unique proteins and mechanisms to counteract the nuclear factor κB (NF-κB) signaling pathway, which is an essential regulatory pathway of host innate immune responses. Here, we describe a NF-κB inhibitory virion protein of orf virus (ORFV), ORFV073, which functions very early in infected cells. Infection with ORFV073 gene deletion virus (OV-IA82Δ073) led to increased accumulation of NF-κB essential modulator (NEMO), marked phosphorylation of IκB kinase (IKK) subunits IKKα and IKKß, IκBα and NF-κB subunit p65 (NF-κB-p65), and to early nuclear translocation of NF-κB-p65 in virus-infected cells (≤ 30 min post infection). Expression of ORFV073 alone was sufficient to inhibit TNFα induced activation of the NF-κB signaling in uninfected cells. Consistent with observed inhibition of IKK complex activation, ORFV073 interacted with the regulatory subunit of the IKK complex NEMO. Infection of sheep with OV-IA82Δ073 led to virus attenuation, indicating that ORFV073 is a virulence determinant in the natural host. Notably, ORFV073 represents the first poxviral virion-associated NF-κB inhibitor described, highlighting the significance of viral inhibition of NF-κB signaling very early in infection.


Subject(s)
Ecthyma, Contagious/virology , Immune Evasion/physiology , NF-kappa B/immunology , Orf virus/pathogenicity , Virion/immunology , Animals , Ecthyma, Contagious/immunology , HeLa Cells , Humans , Immunoprecipitation , Orf virus/immunology , Orf virus/metabolism , Real-Time Polymerase Chain Reaction , Sheep , Signal Transduction/immunology , Viral Proteins/immunology , Virulence/physiology
6.
Arch Virol ; 160(6): 1527-32, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25804193

ABSTRACT

Bovine papular stomatitis virus (BPSV) infects cattle and, occupationally, humans. Prevalent subclinical infections, frequent reinfections, and virus persistence in healthy animals compound a poorly understood, but likely complex, scenario of BPSV perpetuation and transmission in nature. Here, we report the isolation of multiple BPSV strains coinfecting a single animal. Whole-genome analysis of isolated BPSV strains revealed genomic variability likely affecting virus virulence and infectivity. Further, incongruent phylogenetic relationships between viruses suggested genomic recombination. These results have significant implications for parapoxvirus infection biology and virus evolution in nature.


Subject(s)
Cattle Diseases/virology , Coinfection/veterinary , Parapoxvirus/genetics , Poxviridae Infections/veterinary , Animals , Base Sequence , Cattle/virology , Coinfection/virology , Molecular Sequence Data , Open Reading Frames/genetics , Phylogeny , Poxviridae Infections/genetics , Poxviridae Infections/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...