Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Front Microbiol ; 11: 1123, 2020.
Article in English | MEDLINE | ID: mdl-32587580

ABSTRACT

In this study, we explore the virulence of vesicular stomatitis New Jersey virus (VSNJV) in pigs and its potential relationship with the virus's ability to modulate innate responses. For this purpose, we developed a mutant of the highly virulent strain NJ0612NME6, containing a single amino acid substitution in the matrix protein (M51R). The M51R mutant of NJ0612NME6 was unable to suppress the transcription of genes associated with the innate immune response both in primary fetal porcine kidney cells and porcine primary macrophage cultures. Impaired viral growth was observed only in porcine macrophage cultures, indicating that the M51 residue is required for efficient replication of VSNJV in these cells. Furthermore, when inoculated in pigs by intradermal scarification of the snout, M51R infection was characterized by decreased clinical signs including reduced fever and development of less and smaller secondary vesicular lesions. Pigs infected with M51R had decreased levels of viral shedding and absence of RNAemia compared to the parental virus. The ability of the mutant virus to infect pigs by direct contact remained intact, indicating that the M51R mutation resulted in a partially attenuated phenotype capable of causing primary lesions and transmitting to sentinel pigs. Collectively, our results show a positive correlation between the ability of VSNJV to counteract the innate immune response in swine macrophage cultures and the level of virulence in pigs, a natural host of this virus. More studies are encouraged to evaluate the interaction of VSNJV with macrophages and other components of the immune response in pigs.

2.
J Virol ; 94(7)2020 03 17.
Article in English | MEDLINE | ID: mdl-31969432

ABSTRACT

African swine fever virus (ASFV) is the etiological agent of a contagious and often lethal disease of domestic pigs that has significant economic consequences for the swine industry. The disease is devastating the swine industry in Central Europe and East Asia, with current outbreaks caused by circulating strains of ASFV derived from the 2007 Georgia isolate (ASFV-G), a genotype II ASFV. In the absence of any available vaccines, African swine fever (ASF) outbreak containment relies on the control and culling of infected animals. Limited cross-protection studies suggest that in order to ensure a vaccine is effective, it must be derived from the current outbreak strain or at the very least from an isolate with the same genotype. Here, we report the discovery that the deletion of a previously uncharacterized gene, I177L, from the highly virulent ASFV-G produces complete virus attenuation in swine. Animals inoculated intramuscularly with the virus lacking the I177L gene, ASFV-G-ΔI177L, at a dose range of 102 to 106 50% hemadsorbing doses (HAD50), remained clinically normal during the 28-day observational period. All ASFV-G-ΔI177L-infected animals had low viremia titers, showed no virus shedding, and developed a strong virus-specific antibody response; importantly, they were protected when challenged with the virulent parental strain ASFV-G. ASFV-G-ΔI177L is one of the few experimental vaccine candidate virus strains reported to be able to induce protection against the ASFV Georgia isolate, and it is the first vaccine capable of inducing sterile immunity against the current ASFV strain responsible for recent outbreaks.IMPORTANCE Currently, there is no commercially available vaccine against African swine fever. Outbreaks of this disease are devastating the swine industry from Central Europe to East Asia, and they are being caused by circulating strains of African swine fever virus derived from the Georgia 2007 isolate. Here, we report the discovery of a previously uncharacterized virus gene, which when deleted completely attenuates the Georgia isolate. Importantly, animals infected with this genetically modified virus were protected from developing ASF after challenge with the virulent parental virus. Interestingly, ASFV-G-ΔI177L confers protection even at low doses (102 HAD50) and remains completely attenuated when inoculated at high doses (106 HAD50), demonstrating its potential as a safe vaccine candidate. At medium or higher doses (104 HAD50), sterile immunity is achieved. Therefore, ASFV-G-ΔI177L is a novel efficacious experimental ASF vaccine protecting pigs from the epidemiologically relevant ASFV Georgia isolate.


Subject(s)
African Swine Fever Virus/genetics , African Swine Fever/immunology , Viral Vaccines/immunology , African Swine Fever/prevention & control , Animals , Antibody Formation , Body Temperature , Cells, Cultured , Epidemics , Gene Deletion , Genotype , Macrophages/virology , Mutation , Swine , Viral Proteins/genetics , Viremia/virology , Virulence , Virus Replication
3.
Sci Rep ; 10(1): 494, 2020 01 16.
Article in English | MEDLINE | ID: mdl-31949276

ABSTRACT

The CD2-like African swine fever virus (ASFV) gene 8DR, (also known as EP402R) encodes for a structural transmembrane glycoprotein that has been shown to mediate hemadsorption and be involved in host immunomodulation as well as the induction of protective immune response. In addition, several natural ASFV isolates showing decreased virulence in swine has been shown to be non-hemadsorbing suggesting an association between altered or deleted forms of 8DR and virus attenuation. Here we demonstrate that deletion of 8DR gene from the genome of ASFV Georgia2010 isolate (ASFV-G-Δ8DR) does not significantly alter the virulence of the virus. ASFV-G-Δ8DR inoculated intramuscularly or intranasally (in a range of 102 to 104 TCID50) produced a clinical disease in domestic pigs indistinguishable from that induced by the same doses of the virulent parental ASFV Georgia2010 isolate. In addition, viremia values in ASFV-G-Δ8DR do not differ from those detected in animals infected with parental virus. Therefore, deletion of 8DR gene is not associated with a noticeable decrease in virulence of the ASFV Georgia isolate.


Subject(s)
African Swine Fever Virus/pathogenicity , African Swine Fever/virology , Gene Deletion , Glycoproteins/genetics , Viremia/virology , African Swine Fever Virus/genetics , Animals , Cells, Cultured , Genome, Viral , High-Throughput Nucleotide Sequencing , Macrophages/cytology , Macrophages/virology , Swine , Viral Proteins/genetics , Virulence Factors/genetics , Whole Genome Sequencing/methods
4.
Viruses ; 11(7)2019 07 02.
Article in English | MEDLINE | ID: mdl-31269702

ABSTRACT

African swine fever virus (ASFV) is the etiological agent of an often lethal disease of domestic pigs, African swine fever (ASF). The ASFV Georgia 2007 isolate (ASFV-G) is responsible for the current epidemic situation in Europe and Asia. Genetically modified ASFVs containing deletions of virulence-associated genes have produced attenuated phenotypes and induced protective immunity in swine. Here we describe the differential behavior of two viral genes, NL (DP71L) and UK (DP96R), both originally described as being involved in virus virulence. Deletion of either of these genes efficiently attenuated ASFV strain E70. We demonstrated that deletion of the UK gene from the ASFV-G genome did not decrease virulence when compared to the parental virus. Conversely, deletion of the NL gene produced a heterogeneous response, with early death in one of the animals and transient fever in the other animals. With this knowledge, we attempted to increase the safety profile of the previously reported experimental vaccine ASFV-GΔ9GL/ΔUK by deleting the NL gene. A triple gene-deletion virus was produced, ASFV-GΔ9GL/ΔNL/ΔUK. Although ASFV-GΔ9GL/ΔNL/ΔUK replicated in primary cell cultures of swine macrophages, it demonstrated a severe replication deficiency in pigs, failing to induce protection against challenge with parental ASFV-G.


Subject(s)
African Swine Fever Virus/genetics , African Swine Fever/prevention & control , Genes, Viral/genetics , Sequence Deletion , Vaccines, Attenuated/immunology , Viral Vaccines/immunology , Virulence Factors/genetics , African Swine Fever/immunology , African Swine Fever/transmission , African Swine Fever/virology , African Swine Fever Virus/growth & development , Amino Acid Sequence , Animals , Antibodies, Viral , Disease Models, Animal , Female , High-Throughput Nucleotide Sequencing , Macrophages/virology , Phenotype , Sequence Alignment , Survival Rate , Sus scrofa , Swine , Vaccines, Attenuated/genetics , Viral Vaccines/genetics , Virulence/genetics , Virus Replication
5.
Viruses ; 11(4)2019 03 29.
Article in English | MEDLINE | ID: mdl-30934875

ABSTRACT

Classical swine fever virus (CSFV) E2 protein, the major virus structural glycoprotein, is an essential component of the viral envelope. E2 is involved in virus absorption, induction of a protective immune response and is critical for virulence in swine. Using the yeast two-hybrid system, we identified protein phosphatase 1 catalytic subunit beta (PPP1CB), which is part of the Protein Phosphatase 1 (PP1) complex, as a specific binding host partner for E2. We further confirmed the occurrence of this interaction in CSFV-infected swine cells by using two independent methodologies: Co-immunoprecipitation and Proximity Ligation Assay. In addition, we demonstrated that pharmacological activation of the PP1 pathway has a negative effect on CSFV replication while inhibition of the PP1 pathway or knockdown of PPP1CB by siRNA had no observed effect. Overall, our data suggests that the CSFV E2 and PPP1CB protein interact in infected cells, and that activation of the PP1 pathway decreases virus replication.


Subject(s)
Catalytic Domain , Classical Swine Fever Virus/growth & development , Host-Pathogen Interactions , Protein Phosphatase 1/metabolism , Viral Envelope Proteins/metabolism , Animals , Classical Swine Fever Virus/immunology , Protein Binding , Protein Interaction Mapping , Swine , Two-Hybrid System Techniques , Virus Replication
6.
Viruses ; 10(9)2018 08 28.
Article in English | MEDLINE | ID: mdl-30154321

ABSTRACT

We have previously shown that Classical Swine Fever Virus (CSFV) p7 is an essential nonstructural protein with a viroporin activity, a critical function in the progression of virus infection. We also identified p7 domains and amino acid residues critical for pore formation. Here, we describe how p7 specifically interacts with host protein CAMLG, an integral ER transmembrane protein involved in intracellular calcium release regulation and signal response generation. Detection of interaction as well as the identification of p7 areas mediating interaction with CAMLG was performed by yeast two-hybrid. p7-CAMLG interaction was further confirmed by confocal microscopy in eukaryotic cells, co-expressing both proteins. Mutant forms of p7 having substituted native residues identified as mediating interaction with CAMLG showed a decreased co-localization compared with the native forms of p7. Furthermore, it is shown that native p7, but not the mutated forms of p7 that fail to interact with CAMLG, efficiently mediates calcium permeability in the ER. Interestingly, viruses harboring some of those mutated forms of p7 have been previously shown to have a significantly decreased virulence in swine.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Calcium/metabolism , Classical Swine Fever Virus/physiology , Endoplasmic Reticulum/metabolism , Host-Pathogen Interactions , Viral Regulatory and Accessory Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Line , HEK293 Cells , Humans , Protein Interaction Maps/physiology , Saccharomyces cerevisiae/genetics , Swine , Viral Regulatory and Accessory Proteins/genetics , Virulence/genetics
7.
Virus Res ; 249: 116-123, 2018 04 02.
Article in English | MEDLINE | ID: mdl-29605728

ABSTRACT

African swine fever virus (ASFV) causes a contagious and frequently lethal disease of pigs causing significant economic consequences to the swine industry. The ASFV genome encodes for more than 150 genes, but only a few of them have been studied in detail. Here we report the characterization of open reading frame L83L which encodes a highly conserved protein across all ASFV isolates. A recombinant ASFV harboring a HA tagged L83L protein was developed (ASFV-G-L83L-HA) and used to demonstrate that L83L is a transiently expressed early virus protein. A recombinant ASFV lacking the L83L gene (ASFV-G-ΔL83L) was developed from the highly virulent field isolate Georgia2007 (ASFV-G) and was used to show that L83L is a non-essential gene. ASFV-G-ΔL83L had similar replication in primary swine macrophage cells when compared to its parental virus ASFV-G. Analysis of host-protein interactions for L83L identified IL-1ß as its host ligand. Experimental infection of domestic pigs showed that ASFV-G-ΔL83L is as virulent as the parental virus ASFV-G.


Subject(s)
African Swine Fever Virus/physiology , Host-Pathogen Interactions , Interleukin-1beta/metabolism , Viral Proteins/metabolism , African Swine Fever Virus/genetics , Animals , Cells, Cultured , Gene Expression Profiling , Gene Knockout Techniques , Macrophages/virology , Swine , Viral Proteins/genetics , Virus Replication
8.
Sci Rep ; 8(1): 3154, 2018 02 16.
Article in English | MEDLINE | ID: mdl-29453406

ABSTRACT

African swine fever virus (ASFV) causes a highly contagious disease called African swine fever. This disease is often lethal for domestic pigs, causing extensive losses for the swine industry. ASFV is a large and complex double stranded DNA virus. Currently there is no commercially available treatment or vaccine to prevent this devastating disease. Development of recombinant ASFV for producing live-attenuated vaccines or studying the involvement of specific genes in virus virulence has relied on the relatively rare event of homologous recombination in primary swine macrophages, causing difficulty to purify the recombinant virus from the wild-type parental ASFV. Here we present the use of the CRISPR-Cas9 gene editing system as a more robust and efficient system to produce recombinant ASFVs. Using CRISPR-Cas9 a recombinant virus was efficiently developed by deleting the non-essential gene 8-DR from the genome of the highly virulent field strain Georgia07 using swine macrophages as cell substrate.


Subject(s)
African Swine Fever Virus/genetics , CRISPR-Cas Systems/genetics , DNA, Recombinant/genetics , Genetic Engineering/methods , High-Throughput Nucleotide Sequencing
9.
PLoS One ; 12(5): e0177433, 2017.
Article in English | MEDLINE | ID: mdl-28542321

ABSTRACT

Prophylactic vaccination using live attenuated classical swine fever (CSF) vaccines has been a very effective method to control the disease in endemic regions and during outbreaks in previously disease-free areas. These vaccines confer effective protection against the disease at early times post-vaccination although the mechanisms mediating the protection are poorly characterized. Here we present the events occurring after the administration of our in-house developed live attenuated marker vaccine, FlagT4Gv. We previously reported that FlagT4Gv intramuscular (IM) administered conferred effective protection against intranasal challenge with virulent CSFV (BICv) as early as 7 days post-vaccination. Here we report that FlagT4Gv is able to induce protection against disease as early as three days post-vaccination. Immunohistochemical testing of tissues from FlagT4Gv-inoculated animals showed that tonsils were colonized by the vaccine virus by day 3 post-inoculation. There was a complete absence of BICv in tonsils of FlagT4Gv-inoculated animals which had been intranasal (IN) challenged with BICv 3 days after FlagT4Gv infection, confirming that FlagT4Gv inoculation confers sterile immunity. Analysis of systemic levels of 19 different cytokines in vaccinated animals demonstrated an increase of IFN-α three days after FlagT4Gv inoculation compared with mock infected controls.


Subject(s)
Classical Swine Fever Virus/immunology , Classical Swine Fever/immunology , Classical Swine Fever/prevention & control , Viral Vaccines/pharmacology , Animals , Classical Swine Fever/virology , Classical Swine Fever Virus/pathogenicity , Classical Swine Fever Virus/physiology , Cytokines/blood , Female , Interferon-alpha/blood , Palatine Tonsil/immunology , Palatine Tonsil/virology , Sus scrofa , Swine , Time Factors , Vaccines, Attenuated/pharmacology , Vaccines, Marker/pharmacology , Virus Replication
10.
Sci Rep ; 7: 46747, 2017 04 24.
Article in English | MEDLINE | ID: mdl-28436458

ABSTRACT

African swine fever is a contagious and often lethal disease for domestic pigs with a significant economic impact for the swine industry. The etiological agent, African swine fever virus (ASFV), is a highly structurally complex double stranded DNA virus. No effective vaccines or antiviral treatment are currently commercially available. We present here the development of a strain of ASFV that has been shown to retain its ability to cause disease in swine, efficiently replicate in swine macrophage and that is fluorescently tagged. The insertion of an EGFP cassette replacing the reading frames for two neighboring genes, MGF360-13L and MGF360-14L, in highly virulent field isolate Georgia/2007, did not affect virus replication in cell cultures and did not affect disease progression in swine, the natural host for ASFV. A virulent fluorescently tagged ASFV is a suitable tool to conduct pathogenesis studies in swine, study on virus-macrophage interaction and to run large scale screens that require a sensitive high throughput output. Utilizing an EGFP reporter system for observing ASFV replication and infectivity can circumvent the time and labor-intensive steps associated with viral antigen-based assays such as the observation of hemadsorption or cytopathic effect.


Subject(s)
African Swine Fever Virus/metabolism , Green Fluorescent Proteins/metabolism , Macrophages/metabolism , African Swine Fever/virology , African Swine Fever Virus/genetics , African Swine Fever Virus/pathogenicity , Animals , Cells, Cultured , Fluorescence , Green Fluorescent Proteins/genetics , Host-Pathogen Interactions , Macrophages/virology , Sus scrofa , Swine , Swine Diseases/virology , Virulence/genetics
11.
J Virol ; 91(1)2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27795430

ABSTRACT

African swine fever virus (ASFV) is the etiological agent of a contagious and often lethal viral disease of domestic pigs that has significant economic consequences for the swine industry. The control of African swine fever (ASF) has been hampered by the unavailability of vaccines. Successful experimental vaccines have been derived from naturally occurring, cell culture-adapted, or genetically modified live attenuated ASFV. Recombinant viruses harboring engineered deletions of specific virulence-associated genes induce solid protection against challenge with parental viruses. Deletion of the 9GL (B119L) gene in the highly virulent ASFV isolates Malawi Lil-20/1 (Mal) and Pretoriuskop/96/4 (Δ9GL viruses) resulted in complete protection when challenged with parental isolates. When similar deletions were created within the ASFV Georgia 2007 (ASFV-G) genome, attenuation was achieved but the protective and lethal doses were too similar. To enhance attenuation of ASFV-G, we deleted another gene, UK (DP96R), which was previously shown to be involved in attenuation of the ASFV E70 isolate. Here, we report the construction of a double-gene-deletion recombinant virus, ASFV-G-Δ9GL/ΔUK. When administered intramuscularly (i.m.) to swine, there was no induction of disease, even at high doses (106 HAD50). Importantly, animals infected with 104 50% hemadsorbing doses (HAD50) of ASFV-G-Δ9GL/ΔUK were protected as early as 14 days postinoculation when challenged with ASFV-G. The presence of protection correlates with the appearance of serum anti-ASFV antibodies, but not with virus-specific circulating ASFV-specific gamma interferon (IFN-γ)-producing cells. ASFV-G-Δ9GL/ΔUK is the first rationally designed experimental ASFV vaccine that protects against the highly virulent ASFV Georgia 2007 isolate as early as 2 weeks postvaccination. IMPORTANCE: Currently, there is no commercially available vaccine against African swine fever. Outbreaks of the disease are devastating to the swine industry and are caused by circulating strains of African swine fever virus. Here, we report a putative vaccine derived from a currently circulating strain but containing two deletions in two separate areas of the virus, allowing increased safety. Using this genetically modified virus, we were able to vaccinate swine and protect them from developing ASF. We were able to achieve protection from disease as early as 2 weeks after vaccination, even when the pigs were exposed to a higher than normal concentration of ASFV.


Subject(s)
African Swine Fever Virus/genetics , African Swine Fever Virus/pathogenicity , African Swine Fever/prevention & control , Antibodies, Viral/biosynthesis , Viral Proteins/immunology , Viral Vaccines/administration & dosage , African Swine Fever/immunology , African Swine Fever/virology , African Swine Fever Virus/drug effects , African Swine Fever Virus/immunology , Amino Acid Sequence , Animals , Antibodies, Neutralizing/biosynthesis , Cytokines/biosynthesis , Cytokines/immunology , Dose-Response Relationship, Immunologic , Gene Deletion , Gene Expression , Immunogenicity, Vaccine , Injections, Intramuscular , Sequence Alignment , Swine , Time Factors , Vaccines, Synthetic , Viral Proteins/genetics , Viral Vaccines/biosynthesis , Viral Vaccines/genetics , Virulence
12.
Viruses ; 8(10)2016 10 22.
Article in English | MEDLINE | ID: mdl-27782090

ABSTRACT

African swine fever (ASF) is a lethal hemorrhagic disease of swine caused by a double-stranded DNA virus, ASF virus (ASFV). There is no vaccine to prevent the disease and current control measures are limited to culling and restricting animal movement. Swine infected with attenuated strains are protected against challenge with a homologous virulent virus, but there is limited knowledge of the host immune mechanisms generating that protection. Swine infected with Pretoriuskop/96/4 (Pret4) virus develop a fatal severe disease, while a derivative strain lacking virulence-associated gene 9GL (Pret4Δ9GL virus) is completely attenuated. Swine infected with Pret4Δ9GL virus and challenged with the virulent parental virus at 7, 10, 14, 21, and 28 days post infection (dpi) showed a progressive acquisition of protection (from 40% at 7 dpi to 80% at 21 and 28 dpi). This animal model was used to associate the presence of host immune response (ASFV-specific antibody and interferon (IFN)-γ responses, or specific cytokine profiles) and protection against challenge. With the exception of ASFV-specific antibodies in survivors challenged at 21 and 28 dpi, no association between the parameters assessed and protection could be established. These results, encompassing data from 65 immunized swine, underscore the complexity of the system under study, suggesting that protection relies on the concurrence of different host immune mechanisms.


Subject(s)
African Swine Fever Virus/immunology , African Swine Fever/immunology , African Swine Fever/prevention & control , Animals , Antibodies, Viral/blood , Cytokines/metabolism , Leukocytes, Mononuclear/immunology , Swine
13.
Virus Res ; 223: 181-9, 2016 09 02.
Article in English | MEDLINE | ID: mdl-27497620

ABSTRACT

African swine fever virus (ASFV) is the etiological agent of a contagious and often lethal disease of domestic pigs that has significant economic consequences for the swine industry. The viral genome encodes for more than 150 genes, and only a select few of these genes have been studied in some detail. Here we report the characterization of open reading frame Ep152R that has a predicted complement control module/SCR domain. This domain is found in Vaccinia virus proteins that are involved in blocking the immune response during viral infection. A recombinant ASFV harboring a HA tagged version of the Ep152R protein was developed (ASFV-G-Ep152R-HA) and used to demonstrate that Ep152R is an early virus protein. Attempts to construct recombinant viruses having a deleted Ep152R gene were consistently unsuccessful indicating that Ep152R is an essential gene. Interestingly, analysis of host-protein interactions for Ep152R using a yeast two-hybrid screen, identified BAG6, a protein previously identified as being required for ASFV replication. Furthermore, fluorescent microscopy analysis confirms that Ep152R-BAG6 interaction actually occurs in cells infected with ASFV.


Subject(s)
African Swine Fever Virus/physiology , African Swine Fever/metabolism , African Swine Fever/virology , Genes, Essential , Molecular Chaperones/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Conserved Sequence , Host-Pathogen Interactions , Macrophages/metabolism , Macrophages/virology , Models, Molecular , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Open Reading Frames , Protein Binding , Protein Conformation , Protein Interaction Mapping/methods , Protein Transport , Sequence Deletion , Swine , Two-Hybrid System Techniques , Viral Proteins/chemistry , Virus Replication
14.
Virus Res ; 221: 8-14, 2016 08 02.
Article in English | MEDLINE | ID: mdl-27182007

ABSTRACT

African swine fever virus (ASFV) produces a contagious disease of domestic pigs that results in severe economic consequences to the swine industry. Control of the disease has been hampered by the unavailability of vaccines. We recently reported the development of two experimental vaccine strains (ASFV-G-Δ9GL and ASFV-G-ΔMGF) based on the attenuation of the highly virulent and epidemiologically relevant Georgia2007 isolate. Deletion of the 9GL gene or six genes of the MGF360/505 group produced two attenuated ASFV strains which were able to confer protection to animals when challenged with the virulent parental virus. Both viruses, although efficient in inducing protection, present concerns regarding their safety. In an attempt to solve this problem we developed a novel virus strain, ASFV-G-Δ9GL/ΔMGF, based on the deletion of all genes deleted in ASFV-G-Δ9GL and ASFV-G-ΔMGF. ASFV-G-Δ9GL/ΔMGF is the first derivative of a highly virulent ASFV field strain subjected to a double round of recombination events seeking to sequentially delete specific genes. ASFV-G-Δ9GL/ΔMGF showed a decreased ability to replicate in primary swine macrophage cultures relative to that of ASFV-G and ASFV-G-ΔMGF but similar to that of ASFV-G-Δ9GL. ASFV-G-Δ9GL/ΔMGF was attenuated when intramuscularly inoculated into swine, even at doses as high as 10(6) HAD50. Animals infected with doses ranging from 10(2) to 10(6) HAD50 did not present detectable levels of virus in blood at any time post-infection and they did not develop detectable levels of anti-ASFV antibodies. Importantly, ASFV-G-Δ9GL/ΔMGF does not induce protection against challenge with the virulent parental ASFV-G isolate. Results presented here suggest caution towards approaches involving genomic manipulations when developing rationally designed ASFV vaccine strains.


Subject(s)
African Swine Fever Virus/genetics , African Swine Fever Virus/pathogenicity , African Swine Fever/pathology , African Swine Fever/virology , Sequence Deletion , Viral Proteins/genetics , Viral Vaccines/immunology , African Swine Fever/prevention & control , African Swine Fever Virus/immunology , African Swine Fever Virus/physiology , Animals , Antibodies, Viral/blood , Cells, Cultured , Georgia , Injections, Intramuscular , Macrophages/virology , Recombination, Genetic , Swine , Treatment Outcome , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Virulence , Virus Replication
15.
Virology ; 494: 178-89, 2016 07.
Article in English | MEDLINE | ID: mdl-27110709

ABSTRACT

Controlling classical swine fever (CSF) mainly involves vaccination with live attenuated vaccines (LAV). Experimental CSFV LAVs has been lately developed through reverse genetics using several different approaches. Here we present that codon de-optimization in the major CSFV structural glycoprotein E2 coding region, causes virus attenuation in swine. Four different mutated constructs (pCSFm1-pCSFm4) were designed using various mutational approaches based on the genetic background of the highly virulent strain Brescia (BICv). Three of these constructs produced infectious viruses (CSFm2v, CSFm3v, and CSFm4v). Animals infected with CSFm2v presented a reduced and extended viremia but did not display any CSF-related clinical signs. Animals that were infected with CSFm2v were protected against challenge with virulent parental BICv. This is the first report describing the development of an attenuated CSFV experimental vaccine by codon usage de-optimization, and one of the few examples of virus attenuation using this methodology that is assessed in a natural host.


Subject(s)
Classical Swine Fever Virus/genetics , Classical Swine Fever Virus/immunology , Classical Swine Fever/prevention & control , Vaccines, Attenuated/immunology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viral Vaccines/immunology , Animals , Base Sequence , Cell Line , Cells, Cultured , Classical Swine Fever/immunology , Classical Swine Fever/mortality , Classical Swine Fever/virology , Codon , Computational Biology/methods , Gene Expression Profiling , Gene Expression Regulation , Host-Pathogen Interactions , Mutation , Swine , Vaccines, Attenuated/genetics , Viral Envelope Proteins/chemistry , Viral Vaccines/genetics , Virulence/genetics , Virus Replication
16.
J Virol ; 89(16): 8556-66, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26063424

ABSTRACT

UNLABELLED: African swine fever virus (ASFV) is the etiological agent of an often lethal disease of domestic pigs. Disease control strategies have been hampered by the unavailability of vaccines against ASFV. Since its introduction in the Republic of Georgia, a highly virulent virus, ASFV Georgia 2007 (ASFV-G), has caused an epizootic that spread rapidly into Eastern European countries. Currently no vaccines are available or under development to control ASFV-G. In the past, genetically modified ASFVs harboring deletions of virulence-associated genes have proven attenuated in swine, inducing protective immunity against challenge with homologous parental viruses. Deletion of the gene 9GL (open reading frame [ORF] B119L) in highly virulent ASFV Malawi-Lil-20/1 produced an attenuated phenotype even when administered to pigs at 10(6) 50% hemadsorption doses (HAD50). Here we report the construction of a genetically modified ASFV-G strain (ASFV-G-Δ9GLv) harboring a deletion of the 9GL (B119L) gene. Like Malawi-Lil-20/1-Δ9GL, ASFV-G-Δ9GL showed limited replication in primary swine macrophages. However, intramuscular inoculation of swine with 10(4) HAD50 of ASFV-G-Δ9GL produced a virulent phenotype that, unlike Malawi-Lil-20/1-Δ9GL, induced a lethal disease in swine like parental ASFV-G. Interestingly, lower doses (10(2) to 10(3) HAD50) of ASFV-G-Δ9GL did not induce a virulent phenotype in swine and when challenged protected pigs against disease. A dose of 10(2) HAD50 of ASFV-G-Δ9GLv conferred partial protection when pigs were challenged at either 21 or 28 days postinfection (dpi). An ASFV-G-Δ9GL HAD50 of 10(3) conferred partial and complete protection at 21 and 28 dpi, respectively. The information provided here adds to our recent report on the first attempts toward experimental vaccines against ASFV-G. IMPORTANCE: The main problem for controlling ASF is the lack of vaccines. Studies on ASFV virulence lead to the production of genetically modified attenuated viruses that induce protection in pigs but only against homologous virus challenges. Here we produced a recombinant ASFV lacking virulence-associated gene 9GL in an attempt to produce a vaccine against virulent ASFV-G, a highly virulent virus isolate detected in the Caucasus region in 2007 and now spreading though the Caucasus region and Eastern Europe. Deletion of 9GL, unlike with other ASFV isolates, did not attenuate completely ASFV-G. However, when delivered once at low dosages, recombinant ASFV-G-Δ9GL induces protection in swine against parental ASFV-G. The protection against ASFV-G is highly effective after 28 days postvaccination, whereas at 21 days postvaccination, animals survived the lethal challenge but showed signs of ASF. Here we report the design and development of an experimental vaccine that induces protection against virulent ASFV-G.


Subject(s)
African Swine Fever Virus/genetics , African Swine Fever Virus/immunology , African Swine Fever/prevention & control , Viral Proteins/genetics , Viral Vaccines/pharmacology , Virulence Factors/genetics , Animals , Base Sequence , DNA Primers/genetics , Gene Deletion , Genetic Engineering/methods , High-Throughput Nucleotide Sequencing , Molecular Sequence Data , Mutation, Missense/genetics , Polymerase Chain Reaction , Swine , Viral Vaccines/genetics
17.
J Virol ; 89(11): 6048-56, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25810553

ABSTRACT

UNLABELLED: African swine fever virus (ASFV) is the etiological agent of a contagious and often lethal disease of domestic pigs that has significant economic consequences for the swine industry. The control of African swine fever (ASF) has been hampered by the unavailability of vaccines. Experimental vaccines have been developed using genetically modified live attenuated ASFVs where viral genes involved in virus virulence were removed from the genome. Multigene family 360 (MGF360) and MGF505 represent a group of genes sharing partial sequence and structural identities that have been connected with ASFV host range specificity, blocking of the host innate response, and virus virulence. Here we report the construction of a recombinant virus (ASFV-G-ΔMGF) derived from the highly virulent ASFV Georgia 2007 isolate (ASFV-G) by specifically deleting six genes belonging to MGF360 or MGF505: MGF505-1R, MGF360-12L, MGF360-13L, MGF360-14L, MGF505-2R, and MGF505-3R. ASFV-G-ΔMGF replicates as efficiently in primary swine macrophage cell cultures as the parental virus. In vivo, ASFV-G-ΔMGF is completely attenuated in swine, since pigs inoculated intramuscularly (i.m.) with either 10(2) or 10(4) 50% hemadsorbing doses (HAD50) remained healthy, without signs of the disease. Importantly, when these animals were subsequently exposed to highly virulent parental ASFV-G, no signs of the disease were observed, although a proportion of these animals harbored the challenge virus. This is the first report demonstrating the role of MGF genes acting as independent determinants of ASFV virulence. Additionally, ASFV-G-ΔMGF is the first experimental vaccine reported to induce protection in pigs challenged with highly virulent and epidemiologically relevant ASFV-G. IMPORTANCE: The main problem for controlling ASF is the lack of vaccines. Studies focusing on understanding ASFV virulence led to the production of genetically modified recombinant viruses that, while attenuated, are able to confer protection in pigs challenged with homologous viruses. Here we have produced an attenuated recombinant ASFV derived from highly virulent ASFV strain Georgia (ASFV-G) lacking only six of the multigene family 360 (MGF360) and MGF505 genes (ASFV-G-ΔMGF). It is demonstrated, by first time, that deleting specific MGF genes alone can completely attenuate a highly virulent field ASFV isolate. Recombinant virus ASFV-G-ΔMGF effectively confers protection in pigs against challenge with ASFV-G when delivered once via the intramuscular (i.m.) route. The protection against ASFV-G is highly effective by 28 days postvaccination. This is the first report of an experimental vaccine that induces solid protection against virulent ASFV-G.


Subject(s)
African Swine Fever Virus/immunology , African Swine Fever Virus/physiology , Gene Deletion , Viral Proteins/metabolism , Viral Vaccines/immunology , Virulence Factors/metabolism , Virus Replication , African Swine Fever/prevention & control , African Swine Fever Virus/genetics , African Swine Fever Virus/isolation & purification , Animals , Georgia , Injections, Intramuscular , Macrophages/virology , Sus scrofa , Swine , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Proteins/genetics , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Virulence , Virulence Factors/genetics
18.
J Virol ; 89(4): 2324-32, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25505073

ABSTRACT

UNLABELLED: African swine fever virus (ASFV) causes a contagious and often lethal disease of feral and domestic swine. Experimental vaccines derived from naturally occurring, genetically modified, or cell culture-adapted ASFV have been evaluated, but no commercial vaccine is available to control African swine fever (ASF). We report here the genotypic and phenotypic analysis of viruses obtained at different passages during the process of adaptation of a virulent ASFV field isolate from the Republic of Georgia (ASFV-G) to grow in cultured cell lines. ASFV-G was successively passaged 110 times in Vero cells. Viruses obtained at passages 30, 60, 80, and 110 were evaluated in vitro for the ability to replicate in Vero cells and primary swine macrophages cultures and in vivo for assessing virulence in swine. Replication of ASFV-G in Vero cells increased with successive passages, corresponding to a decreased replication in primary swine macrophages cultures. In vivo, progressive loss of virus virulence was observed with increased passages in Vero cells, and complete attenuation of ASFV-G was observed at passage 110. Infection of swine with the fully attenuated virus did not confer protection against challenge with virulent parental ASFV-G. Full-length sequence analysis of each of these viruses revealed significant deletions that gradually accumulated in specific areas at the right and left variable ends of the genome. Mutations that result in amino acid substitutions and frameshift mutations were also observed, though in a rather limited number of genes. The potential importance of these genetic changes in virus adaptation/attenuation is discussed. IMPORTANCE: The main problem in controlling ASF is the lack of vaccines. Attempts to produce vaccines by adaptation of ASFV to cultured cell lines have been made. These attempts led to the production of attenuated viruses that conferred only homologous protection. Specifics regarding adaptation of these isolates to cell cultures have been insufficiently described. Details like the numbers of passages required to obtain attenuated viruses, genetic modifications introduced into the virus genomes along passages, and the extent of attenuation and induced protective efficacy are not readily available. In this study, we assessed the changes that lead to decreased growth in swine macrophages and to attenuation in swine. Loss of virulence, probably associated with limited replication in vivo, may lead to the lack of protective immunity in swine observed after challenge. This report provides valuable information that can be used to further the understanding of ASFV gene function, virus attenuation, and protection against infection.


Subject(s)
Adaptation, Biological , African Swine Fever Virus/growth & development , African Swine Fever Virus/genetics , Mutation , Sequence Deletion , Serial Passage , African Swine Fever Virus/isolation & purification , African Swine Fever Virus/physiology , Animals , Cells, Cultured , Chlorocebus aethiops , DNA, Viral/chemistry , DNA, Viral/genetics , Genome, Viral , Genotype , Georgia (Republic) , Phenotype , Sequence Analysis, DNA , Swine , Virulence
19.
PLoS One ; 9(1): e85324, 2014.
Article in English | MEDLINE | ID: mdl-24416391

ABSTRACT

E2 is one of the envelope glycoproteins of pestiviruses, including classical swine fever virus (CSFV) and bovine viral diarrhea virus (BVDV). E2 is involved in several critical functions, including virus entry into target cells, induction of a protective immune response and virulence in swine. However, there is no information regarding any host binding partners for the E2 proteins. Here, we utilized the yeast two-hybrid system and identified fifty-seven host proteins as positive binding partners which bound E2 from both CSFV and BVDV with the exception of two proteins that were found to be positive for binding only to CSFV E2. Alanine scanning of CSFV E2 demonstrated that the binding sites for these cellular proteins on E2 are likely non-linear binding sites. The possible roles of the identified host proteins are discussed as the results presented here will be important for future studies to elucidate mechanisms of host protein-virus interactions during pestivirus infection. However, due to the limitations of the yeast two hybrid system, the proteins identified is not exhaustive and each interaction identified needs to be confirmed by independent experimental approaches in the context of virus-infected cells before any definitive conclusion can be drawn on relevance for the virus life cycle.


Subject(s)
Classical Swine Fever Virus/metabolism , Diarrhea Viruses, Bovine Viral/metabolism , Host-Pathogen Interactions , Viral Envelope Proteins/metabolism , Amino Acid Sequence , Animals , Binding Sites , Bovine Virus Diarrhea-Mucosal Disease/virology , Cattle , Classical Swine Fever/virology , Classical Swine Fever Virus/chemistry , Classical Swine Fever Virus/genetics , Diarrhea Viruses, Bovine Viral/chemistry , Diarrhea Viruses, Bovine Viral/genetics , Gene Expression , Gene Library , Molecular Sequence Annotation , Molecular Sequence Data , Protein Binding , Sequence Alignment , Swine , Two-Hybrid System Techniques , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics
20.
Virology ; 446(1-2): 260-7, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24074589

ABSTRACT

The role of non-structural protein 3A of foot-and-mouth disease virus (FMDV) on the virulence in cattle has received significant attention. Particularly, a characteristic 10-20 amino acid deletion has been implicated as responsible for virus attenuation in cattle: a 10 amino acid deletion in the naturally occurring, porcinophilic FMDV O1 Taiwanese strain, and an approximately 20 amino acid deletion found in egg-adapted derivatives of FMDV serotypes O1 and C3. Previous reports using chimeric viruses linked the presence of these deletions to an attenuated phenotype in cattle although results were not conclusive. We report here the construction of a FMDV O1Campos variant differing exclusively from the highly virulent parental virus in a 20 amino acid deletion between 3A residues 87-106, and its characterization in vitro and in vivo. We describe a direct link between a deletion in the FMDV 3A protein and disease attenuation in cattle.


Subject(s)
Cattle Diseases/pathology , Foot-and-Mouth Disease Virus/pathogenicity , Foot-and-Mouth Disease/pathology , Sequence Deletion , Viral Nonstructural Proteins/metabolism , Amino Acid Sequence , Animals , Cattle , Cattle Diseases/virology , Foot-and-Mouth Disease/virology , Foot-and-Mouth Disease Virus/genetics , Molecular Sequence Data , Nasal Mucosa/virology , Sequence Alignment , Serum/virology , Viral Load , Viral Nonstructural Proteins/genetics , Viral Plaque Assay , Virulence , Virulence Factors/genetics , Virulence Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...