Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Vet Immunol Immunopathol ; 208: 34-43, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30712790

ABSTRACT

African swine fever virus (ASFV) causes serious disease in domestic pigs for which there is no vaccine currently available. ASFV is a large DNA virus that encodes for more than 150 proteins, thus making the identification of viral antigens that induce a protective immune response difficult. Based on the functional roles of several ASFV proteins found in previous studies, we selected combinations of ASFV recombinant proteins and pcDNAs-expressing ASFV genes, to analyze their ability to induce humoral and cellular immune responses in pigs. Pigs were immunized using a modified prime-boost approach with combinations of previously selected viral DNA and proteins, resulting in induction of antibodies and specific cell-mediated immune response, measured by IFN-γ ELISpots. The ability of antibodies from pigs immunized with various combinations of ASFV-specific antigens to neutralize infection in vitro, and antigen-specific activation of the cellular immune response were analyzed.


Subject(s)
African Swine Fever/prevention & control , DNA, Viral/immunology , Viral Proteins/immunology , Viral Vaccines/immunology , African Swine Fever/immunology , African Swine Fever Virus , Animals , Antibodies, Viral/blood , Antigens, Viral/immunology , DNA, Viral/administration & dosage , Enzyme-Linked Immunospot Assay , Immunity, Cellular , Interferon-gamma/immunology , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , Sus scrofa , Swine , Viral Proteins/administration & dosage , Viral Vaccines/administration & dosage
2.
Vaccines (Basel) ; 7(1)2019 Jan 28.
Article in English | MEDLINE | ID: mdl-30696015

ABSTRACT

African swine fever virus (ASFV) causes high morbidity and mortality in swine (Sus scrofa), for which there is no commercially available vaccine. Recent outbreaks of the virus in Trans-Caucasus countries, Eastern Europe, Belgium and China highlight the urgent need to develop effective vaccines against ASFV. Previously, we evaluated the immunogenicity of a vaccination strategy designed to test various combinations of ASFV antigens encoded by DNA plasmids and recombinant proteins with the aim to activate both humoral and cellular immunity. Based on our previous results, the objective of this study was to test the combined DNA-protein vaccine strategy using a cocktail of the most immunogenic antigens against virulent ASFV challenge. Pigs were vaccinated three times with a cocktail that included ASFV plasmid DNA (CD2v, p72, p32, +/-p17) and recombinant proteins (p15, p35, p54, +/-p17). Three weeks after the third immunization, all pigs were challenged with the virulent ASFV Armenia 2007 strain. The results showed that vaccinated pigs were not protected from ASFV infection or disease. Compared to the non-vaccinated controls, earlier onset of clinical signs, viremia, and death were observed for the vaccinated animals following virulent ASFV challenge. ASFV induced pathology was also enhanced in the vaccinated pigs. Furthermore, while the vaccinated pigs developed antigen-specific antibodies, immunized pig sera at the time of challenge lacked the capacity to neutralize virus, and instead was observed to enhance ASFV infection in vitro. The results of this work points to a putative immune enhancement mechanism involved in ASFV pathogenesis that warrants further investigation. This pilot study provides insight for the selection of appropriate combinations of ASFV antigens for the development of a rationally-designed, safe, and efficacious vaccine for ASF.

3.
Vaccine ; 36(19): 2694-2704, 2018 05 03.
Article in English | MEDLINE | ID: mdl-29609966

ABSTRACT

The risk of spread of African swine fever virus (ASFV) from Russia and Caucasian areas to several EU countries has recently emerged, making it imperative to improve our knowledge and defensive tools against this important pathogen. The ASFV genome encodes many genes which are not essential for virus replication but are known to control host immune evasion, such as NFκB and the NFAT regulator A238L, the apoptosis inhibitor A224L, the MHC-I antigen presenting modulator EP153R, and the A276R gene, involved in modulating type I IFN. These genes are hypothesized to be involved in virulence of the genotype I parental ASFV NH/P68. We here describe the generation of putative live attenuated vaccines (LAV) prototypes by constructing recombinant NH/P68 viruses lacking these specific genes and containing specific markers.


Subject(s)
African Swine Fever Virus/genetics , African Swine Fever/prevention & control , Vaccines, Attenuated/pharmacology , Viral Vaccines/pharmacology , African Swine Fever Virus/pathogenicity , Animals , COS Cells , Chlorocebus aethiops , Host-Pathogen Interactions/genetics , Mutation , Swine , Vaccines, Attenuated/immunology , Viral Proteins/genetics , Viral Proteins/immunology , Viral Vaccines/immunology , Virulence/genetics
4.
Sci Rep ; 7(1): 10369, 2017 09 04.
Article in English | MEDLINE | ID: mdl-28871180

ABSTRACT

African swine fever virus (ASFV) is a highly pathogenic, double-stranded DNA virus with a marked tropism for cells of the monocyte-macrophage lineage, affecting swine species and provoking severe economic losses and health threats. In the present study, four established porcine cell lines, IPAM-WT, IPAM-CD163, C∆2+ and WSL, were compared to porcine alveolar macrophage (PAM) in terms of surface marker phenotype, susceptibility to ASFV infection and virus production. The virulent ASFV Armenia/07, E70 or the naturally attenuated NHV/P68 strains were used as viral models. Cells expressed only low levels of specific receptors linked to the monocyte/macrophage lineage, with low levels of infection overall, with the exception of WSL, which showed more efficient production of strain NHV/P68 but not of strains E70 and Armenia/07.


Subject(s)
African Swine Fever Virus/physiology , African Swine Fever/virology , Phenotype , African Swine Fever/immunology , African Swine Fever/metabolism , Animals , Biomarkers , Cell Line , Gene Expression Regulation, Viral , Macrophages/immunology , Macrophages/metabolism , Monocytes/immunology , Monocytes/metabolism , Swine , Viral Load , Virus Replication
5.
Virus Res ; 173(1): 58-75, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23154157

ABSTRACT

Throughout a viral infection, the infected cell reprograms the gene expression pattern in order to establish a satisfactory antiviral response. African swine fever virus (ASFV), like other complex DNA viruses, sets up a number of strategies to evade the host's defense systems, such as apoptosis, inflammation and immune responses. The capability of the virus to persist in its natural hosts and in domestic pigs, which recover from infection with less virulent isolates, suggests that the virus displays effective mechanisms to escape host defense systems. ASFV has been described to regulate the activation of several transcription factors, thus regulating the activation of specific target genes during ASFV infection. Whereas some reports have concerned about anti-apoptotic ASFV genes and the molecular mechanisms by which ASFV interferes with inducible gene transcription and immune evasion, less is yet known regarding how ASFV regulates the translational machinery in infected cells, although a recent report has shown a mechanism for favored expression of viral genes based on compartmentalization of viral mRNA and ribosomes with cellular translation factors within the virus factory. The viral mechanisms involved both in the regulation of host genes transcription and in the control of cellular protein synthesis are summarized in this review.


Subject(s)
African Swine Fever Virus/physiology , Protein Biosynthesis , Transcription, Genetic , Viral Proteins/biosynthesis , Virus Replication , Animals , Host-Pathogen Interactions , Immune Evasion , Swine
6.
PLoS Pathog ; 8(6): e1002754, 2012.
Article in English | MEDLINE | ID: mdl-22719252

ABSTRACT

African swine fever (ASF) is caused by a large and highly pathogenic DNA virus, African swine fever virus (ASFV), which provokes severe economic losses and expansion threats. Presently, no specific protection or vaccine against ASF is available, despite the high hazard that the continued occurrence of the disease in sub-Saharan Africa, the recent outbreak in the Caucasus in 2007, and the potential dissemination to neighboring countries, represents. Although virus entry is a remarkable target for the development of protection tools, knowledge of the ASFV entry mechanism is still very limited. Whereas early studies have proposed that the virus enters cells through receptor-mediated endocytosis, the specific mechanism used by ASFV remains uncertain. Here we used the ASFV virulent isolate Ba71, adapted to grow in Vero cells (Ba71V), and the virulent strain E70 to demonstrate that entry and internalization of ASFV includes most of the features of macropinocytosis. By a combination of optical and electron microscopy, we show that the virus causes cytoplasm membrane perturbation, blebbing and ruffles. We have also found that internalization of the virions depends on actin reorganization, activity of Na(+)/H(+) exchangers, and signaling events typical of the macropinocytic mechanism of endocytosis. The entry of virus into cells appears to directly stimulate dextran uptake, actin polarization and EGFR, PI3K-Akt, Pak1 and Rac1 activation. Inhibition of these key regulators of macropinocytosis, as well as treatment with the drug EIPA, results in a considerable decrease in ASFV entry and infection. In conclusion, this study identifies for the first time the whole pathway for ASFV entry, including the key cellular factors required for the uptake of the virus and the cell signaling involved.


Subject(s)
African Swine Fever Virus/metabolism , African Swine Fever/virology , Pinocytosis/physiology , Virus Internalization , African Swine Fever/metabolism , Animals , Blotting, Western , Chlorocebus aethiops , Flow Cytometry , Host-Parasite Interactions/physiology , Microscopy, Confocal , Microscopy, Electron , Swine/virology , Vero Cells
7.
PLoS Pathog ; 5(8): e1000562, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19714237

ABSTRACT

African swine fever virus (ASFV), like other complex DNA viruses, deploys a variety of strategies to evade the host's defence systems, such as inflammatory and immune responses and cell death. Here, we analyse the modifications in the translational machinery induced by ASFV. During ASFV infection, eIF4G and eIF4E are phosphorylated (Ser1108 and Ser209, respectively), whereas 4E-BP1 is hyperphosphorylated at early times post infection and hypophosphorylated after 18 h. Indeed, a potent increase in eIF4F assembly is observed in ASFV-infected cells, which is prevented by rapamycin treatment. Phosphorylation of eIF4E, eIF4GI and 4E-BP1 is important to enhance viral protein production, but is not essential for ASFV infection as observed in rapamycin- or CGP57380-treated cells. Nevertheless, eIF4F components are indispensable for ASFV protein synthesis and virus spread, since eIF4E or eIF4G depletion in COS-7 or Vero cells strongly prevents accumulation of viral proteins and decreases virus titre. In addition, eIF4F is not only activated but also redistributed within the viral factories at early times of infection, while eIF4G and eIF4E are surrounding these areas at late times. In fact, other components of translational machinery such as eIF2alpha, eIF3b, eIF4E, eEF2 and ribosomal P protein are enriched in areas surrounding ASFV factories. Notably, the mitochondrial network is polarized in ASFV-infected cells co-localizing with ribosomes. Thus, translation and ATP synthesis seem to be coupled and compartmentalized at the periphery of viral factories. At later times after ASFV infection, polyadenylated mRNAs disappear from the cytoplasm of Vero cells, except within the viral factories. The distribution of these pools of mRNAs is similar to the localization of viral late mRNAs. Therefore, degradation of cellular polyadenylated mRNAs and recruitment of the translation machinery to viral factories may contribute to the inhibition of host protein synthesis, facilitating ASFV protein production in infected cells.


Subject(s)
African Swine Fever Virus/physiology , African Swine Fever/metabolism , African Swine Fever/virology , Eukaryotic Initiation Factors/metabolism , Animals , COS Cells , Caspase 3/metabolism , Chlorocebus aethiops , Immunohistochemistry , Mitochondria/metabolism , Phosphoproteins/metabolism , Phosphorylation , RNA Cap-Binding Proteins/metabolism , RNA, Messenger/metabolism , RNA, Viral/metabolism , Ribosomal Proteins/metabolism , Vero Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...