Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Bio Protoc ; 14(12): e5017, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38948261

ABSTRACT

Foot-and-mouth disease (FMD) is a severe and extremely contagious viral disease of cloven-hoofed domestic and wild animals, which leads to serious economic losses to the livestock industry globally. FMD is caused by the FMD virus (FMDV), a positive-strand RNA virus that belongs to the genus Aphthovirus, within the family Picornaviridae. Early detection and characterization of FMDV strains are key factors to control new outbreaks and prevent the spread of the disease. Here, we describe a direct RNA sequencing method using Oxford Nanopore Technology (ONT) Flongle flow cells on MinION Mk1C (or GridION) to characterize FMDV. This is a rapid, low cost, and easily deployed point of care (POC) method for a near real-time characterization of FMDV in endemic areas or outbreak investigation sites. Key features • Saves ~35 min of the original protocol time by omitting the reverse transcription step and lowers the costs of reagents and consumables. • Replaces the GridION flow cell from the original protocol with the Flongle, which saves ~90% on the flow cell cost. • Combines the NGS benchwork with a modified version of our African swine fever virus (ASFV) fast analysis pipeline to achieve FMDV characterization within minutes. Graphical overview Schematic of direct RNA sequencing of foot-and-mouth disease virus (FMDV) process, which takes ~50 min from extracted RNA to final loading, modified from the ONT SQK-RNA002 protocol (Version: DRS_9080_v2_revO_14Aug2019).

2.
Viruses ; 16(5)2024 05 05.
Article in English | MEDLINE | ID: mdl-38793613

ABSTRACT

African swine fever virus (ASFV) is the causative agent of a severe and highly contagious viral disease affecting domestic and wild swine. The current ASFV pandemic strain has a high mortality rate, severely impacting pig production and, for countries suffering outbreaks, preventing the export of their pig products for international trade. Early detection and diagnosis of ASFV is necessary to control new outbreaks before the disease spreads rapidly. One of the rate-limiting steps to identify ASFV by next-generation sequencing platforms is library preparation. Here, we investigated the capability of the Oxford Nanopore Technologies' VolTRAX platform for automated DNA library preparation with downstream sequencing on Nanopore sequencing platforms as a proof-of-concept study to rapidly identify the strain of ASFV. Within minutes, DNA libraries prepared using VolTRAX generated near-full genome sequences of ASFV. Thus, our data highlight the use of the VolTRAX as a platform for automated library preparation, coupled with sequencing on the MinION Mk1C for field sequencing or GridION within a laboratory setting. These results suggest a proof-of-concept study that VolTRAX is an effective tool for library preparation that can be used for the rapid and real-time detection of ASFV.


Subject(s)
African Swine Fever Virus , African Swine Fever , Gene Library , Genome, Viral , High-Throughput Nucleotide Sequencing , African Swine Fever Virus/genetics , African Swine Fever Virus/isolation & purification , Animals , Swine , African Swine Fever/diagnosis , African Swine Fever/virology , High-Throughput Nucleotide Sequencing/methods , DNA, Viral/genetics , Sequence Analysis, DNA
3.
Microbiol Resour Announc ; : e0001224, 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38629846

ABSTRACT

We report the near-full genome sequence of a vesicular stomatitis Indiana virus (VSIV) originally collected from a naturally infected bovine in south-central Mexico. This sequence represents a coding-complete genome sequence of a VSIV from Mexico, a country where vesicular stomatitis is endemic.

4.
Viruses ; 15(8)2023 08 11.
Article in English | MEDLINE | ID: mdl-37632064

ABSTRACT

African swine fever (ASF) is a lethal disease of domestic pigs that has been causing outbreaks for over a century in Africa ever since its first discovery in 1921. Since 1957, there have been sporadic outbreaks outside of Africa; however, no outbreak has been as devastating and as far-reaching as the current pandemic that originated from a 2007 outbreak in the Republic of Georgia. Derivatives with a high degree of similarity to the progenitor strain, ASFV-Georgia/2007, have been sequenced from various countries in Europe and Asia. However, the current strains circulating in Africa are largely unknown, and 24 different genotypes have been implicated in different outbreaks. In this study, ASF isolates were collected from samples from swine suspected of dying from ASF on farms in Ghana in early 2022. While previous studies determined that the circulating strains in Ghana were p72 Genotype I, we demonstrate here that the strains circulating in 2022 were derivatives of the p72 Genotype II pandemic strain. Therefore, this study demonstrates for the first time the emergence of Genotype II ASFV in Ghana.


Subject(s)
African Swine Fever Virus , African Swine Fever , Animals , Swine , African Swine Fever Virus/genetics , Ghana/epidemiology , African Swine Fever/epidemiology , Genotype , Sus scrofa
5.
Sci Rep ; 13(1): 1024, 2023 01 19.
Article in English | MEDLINE | ID: mdl-36658154

ABSTRACT

African swine fever is a lethal disease of domestic pigs, geographically expanding as a pandemic, that is affecting countries across Eurasia and severely damaging their swine production industry. After more than 40 years of being absent in the Western hemisphere, in 2020 ASF reappeared in the Dominican Republic and Haiti. The recent outbreak strain in the Dominican Republic has been identified as a genotype II ASFV a derivative of the ASF strain circulating in Asia and Europe. However, to date no full-length genome sequence from either the 1978-1980 Here we report the complete genome sequence of an African swine fever virus (ASFV) (DR-1980) that was previously isolated from blood collected in 1980 from the Dominican Republic at the end of the last outbreak, before culling of all swine on the island of Hispaniola and stored in the Plum Island Animal Disease Center ASFV repository. A contig representing the full-length genome (183,687 base pairs) was de novo assembled into a single contig using both Nanopore and Illumina sequences. DR-1980 was determined to belong to genotype I and, as determined by full genome comparison, a close relative to the sequenced Sardinia viruses that were causing outbreaks at this time.


Subject(s)
African Swine Fever Virus , African Swine Fever , Swine , Animals , African Swine Fever Virus/genetics , African Swine Fever/epidemiology , Dominican Republic/epidemiology , Sus scrofa , Disease Outbreaks
6.
Viruses ; 14(5)2022 05 19.
Article in English | MEDLINE | ID: mdl-35632831

ABSTRACT

African swine fever virus (ASFV) is the etiological agent of African swine fever (ASF), a disease of domestic and wild swine that has spread throughout a large geographical area including Central Europe, East and Southeast Asia, and Southern Africa. Typically, the clinical presentation of the disease in affected swine heavily depends on the virulence of the ASFV strain. Very recently, ASFV was detected in the Dominican Republic (DR) and Haiti, constituting the first diagnosis of ASFV in more than 40 years in the Western hemisphere. In this report, the clinical presentation of the disease in domestic pigs inoculated with an ASFV field strain isolated from samples collected in the DR (ASFV-DR21) was observed. Two groups of domestic pigs were inoculated either intramuscularly (IM) or oronasally (ON) with ASFV-DR21 (104 hemadsorbing dose-50% (HAD50)). A group of naïve pigs (designated as the contact group) was co-housed with the ASFV-DR21 IM-inoculated animals to evaluate ASFV transmission and disease manifestation. Animals inoculated IM with ASFV-DR21 developed an acute disease leading to humane euthanasia at approximately day 7 post-inoculation (pi). Interestingly, animals inoculated via the ON route with ASFV-DR21 developed a heterogeneous pattern of disease kinetics. One animal developed an acute form of the disease and was euthanized on day 7 pi, another animal experienced a protracted presentation of the disease with euthanasia by day 16 pi, and the remaining two animals presented a milder form of the disease, surviving through the 28-day observational period. The contact animals also presented with a heterogenous presentation of the disease. Three of the animals presented protracted but severe forms of the disease being euthanized at days 14, 15 and 21 pi. The other two animals presented with a milder form of the disease, surviving the entire observational period. In general, virus titers in the blood of animals in all study groups closely followed the clinical presentation of the disease, both in length and extent. Importantly, all animals presenting with a prolonged form of the disease, as well as those surviving throughout the observational period, developed a strong ASFV-specific antibody response. These results suggest that ASFV-DR21, unless inoculated parenterally, produces a spectrum of clinical disease, with some animals experiencing an acute fatal form while others presented with a mild transient disease accompanied by the induction of a strong antibody response. At the time of publication, this is the first report characterizing the virulent phenotype of an ASFV field strain isolated from samples collected in the DR during the 2021 outbreak and provides information that may be used in developing epidemiological management measures to control ASF on the island of Hispaniola.


Subject(s)
African Swine Fever Virus , African Swine Fever , African Swine Fever Virus/genetics , Animals , Dominican Republic , Sus scrofa , Swine , Virulence/genetics
7.
Viruses ; 12(10)2020 10 20.
Article in English | MEDLINE | ID: mdl-33092057

ABSTRACT

African swine fever virus (ASFV) is currently the most dreaded infectious disease affecting the global swine production industry. There is no commercial vaccine available, making the culling of infected animals the current solution to control outbreaks. Effective experimental vaccines have been developed by deleting virus genes associated with virulence. Deletion of the ASFV 9GL gene (∆9GL) has resulted in the attenuation of different ASFV strains, although the degree of attenuation varies across isolates. Here, we investigated the possibility of the increased safety of the experimental vaccine strain ASFV-G-Δ9GL by deleting two additional virus genes involved in pathogenesis, CD2v, a CD2 like viral encoded gene from the EP402R open reading frame (ORF), and C-type lectin-like viral gene, encoded from the EP153R ORF. Two new recombinant viruses were developed, ASFV-G-Δ9GL/ΔCD2v and ASFV-G-Δ9GL/ΔCD2v/ΔEP153R, harboring two and three gene deletions, respectively. ASFV-G-Δ9GL/ΔCD2v/ΔEP153R, but not ASFV-G-Δ9GL/ΔCD2v, had a decreased ability to replicate in vitro in swine macrophage cultures when compared with parental ASFV-G-Δ9GL. Importantly, ASFV-G-Δ9GL/ΔCD2v and ASFV-G-Δ9GL/ΔCD2v/ΔEP153R induced almost undetectable viremia levels when inoculated into domestic pigs and failed to protect them against challenge with parental virulent ASFV-Georgia, while ASFV-G-Δ9GL offered robust protection during challenge. Therefore, the deletion of CD2-like and C-type lectin-like genes significantly decreased the protective potential of ASFV-G-Δ9GL as a vaccine candidate. This study constitutes an example of the unpredictability of genetic manipulation involving the simultaneous deletion of multiple genes from the ASFV genome.


Subject(s)
African Swine Fever Virus , Viral Vaccines/immunology , Virulence Factors/genetics , African Swine Fever/immunology , African Swine Fever/prevention & control , African Swine Fever Virus/genetics , African Swine Fever Virus/immunology , Animals , Antibodies, Viral , Antigens, Viral/genetics , Gene Deletion , Genes, Viral , Genome, Viral , Lectins/genetics , Swine , Vaccination/veterinary , Vaccines, Attenuated , Viral Proteins/genetics
8.
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
9.
J Clin Microbiol ; 58(1)2019 12 23.
Article in English | MEDLINE | ID: mdl-31694969

ABSTRACT

African swine fever virus (ASFV) is the causative agent of a severe and highly contagious viral disease of pigs that poses serious economic consequences to the swine industry due to the high mortality rate and impact on international trade. There is no effective vaccine to control African swine fever (ASF), and therefore, efficient disease control is dependent on early detection and diagnosis of ASFV. The large size of the ASFV genome (∼180 kb) has historically hindered efforts to rapidly obtain a full-genome sequence. Rapid acquisition of data is critical for characterization of the isolate and to support epidemiological efforts. Here, we investigated the capacity of the Oxford Nanopore MinION sequence sensing device to act as a rapid sequencing tool. When coupled with our novel companion software script, the African swine fever fast analysis sequencing tool (ASF-FAST), the analysis of output data was performed in real time. Complete ASFV genome sequences were generated from cell culture isolates and blood samples obtained from experimentally infected pigs. Removal of the host-methylated DNA from the extracted nucleic acid facilitated rapid ASFV sequence identification, with reads specific to ASFV detected within 6 min after the initiation of sequencing. Regardless of the starting material, sufficient sequence was available for complete genome resolution (up to 100%) within 10 min. Overall, this paper highlights the use of Nanopore sequencing technology in combination with the ASF-FAST software for the purpose of rapid and real-time resolution of the full ASFV genome from a diagnostic sample.


Subject(s)
African Swine Fever Virus/classification , African Swine Fever Virus/genetics , African Swine Fever/diagnosis , African Swine Fever/virology , Computational Biology/methods , Nanopore Sequencing , Software , African Swine Fever Virus/isolation & purification , Animals , Genome, Viral , High-Throughput Nucleotide Sequencing , Nanopore Sequencing/methods , Reagent Kits, Diagnostic , Reproducibility of Results , Sequence Analysis, DNA , Swine
10.
PLoS One ; 14(11): e0223955, 2019.
Article in English | MEDLINE | ID: mdl-31725732

ABSTRACT

African swine fever (ASF) is a swine disease caused by a large, structurally complex, double-stranded DNA virus, African swine fever virus (ASFV). In domestic pigs, acute infection by highly virulent ASF viruses causes hemorrhagic fever and death. Previous work has suggested that ASFV pathogenesis is primarily mediated by host cytokines produced by infected monocytes and macrophages. To better understand molecular mechanisms mediating virus pathogenesis and immune evasion, we used transcriptome analysis to identify gene expression changes after ASFV infection in ex vivo swine macrophages. Our results suggest that the cytokines of TNF family including FASLG, LTA, LTB, TNF, TNFSF4, TNFSF10, TNFSF13B and TNFSF18 are the major causative cytokine factors in ASF pathogenesis via inducing apoptosis. Other up-regulated proinflammatory cytokines (IL17F and interferons) and down-regulated anti-inflammatory cytokine (IL10) may also significantly contribute to ASF pathogenesis and cause excessive tissue inflammatory responses. The differential expression of genes also indicates that ASFV could evade both the innate and adaptive immune responses by (i) inhibiting MHC Class II antigen processing and presentation, (ii) avoiding CD8+ T effector cells and neutrophil extracellular traps via decreasing expression of neutrophil/CD8+ T effector cell-recruiting chemokines, (iii) suppressing M1 activation of macrophages, (iv) inducing immune suppressive cytokines, and (v) inhibiting the processes of macrophage autophagy and apoptosis. These results provide novel information to further investigate and better understand the mechanism of pathogenesis and immune evasion of this devastating swine disease.


Subject(s)
African Swine Fever Virus/pathogenicity , African Swine Fever/immunology , Immune Evasion , Macrophages/immunology , Macrophages/virology , Adaptive Immunity/genetics , African Swine Fever/genetics , African Swine Fever/virology , African Swine Fever Virus/immunology , Animals , Antigen Presentation/genetics , Autophagy-Related Proteins/genetics , Cells, Cultured , Chemokines/genetics , Cytokines/genetics , Down-Regulation , Gene Expression Profiling , Immune Evasion/genetics , Immunity, Innate/genetics , Macrophage Activation/genetics , Macrophage Activation/immunology , Receptors, Cytokine/genetics , Signal Transduction/genetics , Sus scrofa , Swine , Up-Regulation
11.
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
12.
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
13.
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
14.
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
15.
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
16.
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
17.
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
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...