Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 878
Filter
1.
J Biosci ; 492024.
Article in English | MEDLINE | ID: mdl-38952078

ABSTRACT

Owing to the lack of effective vaccines, current control measures and eradication strategies for the African swine fever virus (ASFV) rely on early detection and stringent stamping-out procedures. In the present study, we developed two independent isothermal amplification assays, namely, loop-mediated isothermal amplification (LAMP) and polymerase spiral reaction (PSR), for quick visualization of the ASFV genome in clinical samples. Additionally, a quantitative real-time PCR (qRT-PCR)-based hydrolysis probe assay was developed for comparative assessment of sensitivity with the developed isothermal assays. The analytical sensitivity of the LAMP, PSR, and qRT-PCR was found to be 2.64 ×105 copies/µL, 2.64 ×102 copies/µL, and 2.64 ×101 copies/µL, respectively. A total of 165 clinical samples was tested using the developed visual assays. The relative accuracy, relative specificity, and relative diagnostic sensitivity for LAMP vs PSR were found to be 95.37% vs 102.48%, 97.46% vs 101.36%, and 73.33% vs 113.33%, respectively.


Subject(s)
African Swine Fever Virus , African Swine Fever , Nucleic Acid Amplification Techniques , Sensitivity and Specificity , African Swine Fever Virus/genetics , African Swine Fever Virus/isolation & purification , Animals , Nucleic Acid Amplification Techniques/methods , Swine , African Swine Fever/diagnosis , African Swine Fever/virology , Real-Time Polymerase Chain Reaction/methods , Molecular Diagnostic Techniques/methods , Genome, Viral/genetics
2.
Parasit Vectors ; 17(1): 278, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38943218

ABSTRACT

BACKGROUND: African swine fever (ASF) is a highly contagious and severe haemorrhagic disease of Suidae, with mortalities that approach 100 percent. Several studies suggested the potential implication of non-biting dipterans in the spread of ASFV in pig farms due to the identification of the ASFV DNA. However, to our knowledge, no study has evaluated the viral DNA load in non-biting dipterans collected in outbreak farms and no risk factors have been analysed. In this context, our study aimed to analyse the risk factors associated with the presence of non-biting dipterans collected from ASF outbreaks in relation to the presence and load of viral DNA. METHODS: Backyard farms (BF), type A farms (TAF), and commercial farms (CF), were targeted for sampling in 2020. In 2021, no BF were sampled. Each farm was sampled only once. The identification of the collected flies to family, genus, or species level was performed based on morphological characteristics using specific keys and descriptions. Pools were made prior to DNA extraction. All extracted DNA was tested for the presence of the ASFV using a real-time PCR protocol. For this study, we considered every sample with a CT value of 40 as positive. The statistical analysis was performed using Epi Info 7 software (CDC, USA). RESULTS: All collected non-biting flies belonged to five families: Calliphoridae, Sarcophagidae, Fanniidae, Drosophilidae, and Muscidae. Of the 361 pools, 201 were positive for the presence of ASFV DNA. The obtained CT values of the positive samples ranged from 21.54 to 39.63, with a median value of 33.59 and a mean value of 33.56. Significantly lower CT values (corresponding to higher viral DNA load) were obtained in Sarcophagidae, with a mean value of 32.56; a significantly higher number of positive pools were noticed in August, mean value = 33.12. CONCLUSIONS: Our study brings compelling evidence of the presence of the most common synanthropic flies near domestic pig farms carrying ASFV DNA, highlighting the importance of strengthening the biosecurity measures and protocols for prevention of the insect life cycle and distribution.


Subject(s)
African Swine Fever Virus , African Swine Fever , DNA, Viral , Diptera , Disease Outbreaks , Farms , Animals , African Swine Fever Virus/genetics , African Swine Fever Virus/isolation & purification , African Swine Fever Virus/classification , African Swine Fever/epidemiology , African Swine Fever/virology , African Swine Fever/transmission , Swine , Disease Outbreaks/veterinary , DNA, Viral/genetics , Romania/epidemiology , Diptera/virology , Diptera/classification , Diptera/genetics , Insect Vectors/virology , Insect Vectors/classification
3.
Emerg Microbes Infect ; 13(1): 2366406, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38847223

ABSTRACT

African swine fever, caused by African swine fever virus (ASFV), is a highly contagious and fatal disease that poses a significant threat to the global pig industry. The limited information on ASFV pathogenesis and ASFV-host interactions has recently prompted numerous transcriptomic studies. However, most of these studies have focused on elucidating the transcriptome profiles of ASFV-infected porcine alveolar macrophages in vitro. Here, we analyzed dynamic transcriptional patterns in vivo in nine organ tissues (spleen, submandibular lymph node, mesenteric lymph node, inguinal lymph node, tonsils, lungs, liver, kidneys, and heart) obtained from pigs in the early stages of ASFV infection (1 and 3 d after viremia). We observed rapid spread of ASFV to the spleen after viremia, followed by broad transmission to the liver and lungs and subsequently, the submandibular and inguinal lymph nodes. Profound variations in gene expression patterns were observed across all organs and at all time-points, providing an understanding of the distinct defence strategies employed by each organ against ASFV infection. All ASFV-infected organs exhibited a collaborative response, activating immune-associated genes such as S100A8, thereby triggering a pro-inflammatory cytokine storm and interferon activation. Functional analysis suggested that ASFV exploits the PI3K-Akt signalling pathway to evade the host immune system. Overall, our findings provide leads into the mechanisms underlying pathogenesis and host immune responses in different organs during the early stages of infection, which can guide further explorations, aid the development of efficacious antiviral strategies against ASFV, and identify valuable candidate gene targets for vaccine development.


Subject(s)
African Swine Fever Virus , African Swine Fever , Transcriptome , Animals , African Swine Fever Virus/genetics , African Swine Fever Virus/physiology , Swine , African Swine Fever/virology , Gene Expression Profiling , Lymph Nodes/virology , Spleen/virology , Spleen/metabolism , Viremia , Lung/virology , Liver/virology , Liver/metabolism
4.
Virus Res ; 346: 199412, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38838820

ABSTRACT

African swine fever virus (ASFV) is a large double-stranded DNA virus with a complex structural architecture and encodes more than 150 proteins, where many are with unknown functions. E184L has been reported as one of the immunogenic ASFV proteins that may contribute to ASFV pathogenesis and immune evasion. However, the antigenic epitopes of E184L are not yet characterized. In this study, recombinant E184L protein was expressed in prokaryotic expression system and four monoclonal antibodies (mAbs), designated as 1A10, 2D2, 3H6, and 4C10 were generated. All four mAbs reacted specifically with ASFV infected cells. To identify the epitopes of the mAbs, a series of overlapped peptides of E184L were designed and expressed as maltose binding fusion proteins. Accordingly, the expressed fusion proteins were probed with each E184L mAb separately by using Western blot. Following a fine mapping, the minimal linear epitope recognized by mAb 1A10 was identified as 119IQRQGFL125, and mAbs 2D2, 3H6, and 4C10 recognized a region located between 153DPTEFF158. Alignment of amino acids of E184L revealed that the two linear epitopes are highly conserved among different ASFV isolates. Furthermore, the potential application of the two epitopes in ASFV diagnosis was assessed through epitope-based ELISA using 24 ASFV positive and 18 negative pig serum and the method were able to distinguish positive and negative samples, indicating the two epitopes are dominant antigenic sites. To our knowledge, this is the first study to characterize the B cell epitopes of the antigenic E184L protein of ASFV, offering valuable tools for future research, as well as laying a foundation for serological diagnosis and epitope-based marker vaccine development.


Subject(s)
African Swine Fever Virus , Antibodies, Monoclonal , Antibodies, Viral , Epitope Mapping , Epitopes, B-Lymphocyte , African Swine Fever Virus/immunology , African Swine Fever Virus/genetics , Antibodies, Monoclonal/immunology , Epitopes, B-Lymphocyte/immunology , Animals , Antibodies, Viral/immunology , Swine , African Swine Fever/immunology , African Swine Fever/virology , Mice , Viral Proteins/immunology , Viral Proteins/genetics , Viral Proteins/chemistry , Antigens, Viral/immunology , Antigens, Viral/genetics , Antigens, Viral/chemistry , Mice, Inbred BALB C
5.
Vet Res ; 55(1): 73, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38849962

ABSTRACT

African swine fever virus (ASFV) causes a devastating disease affecting domestic and wild pigs. ASF was first introduced in Sardinia in 1978 and until 2019 only genotype I isolates were identified. A remarkable genetic stability of Sardinian ASFV isolates was described, nevertheless in 2019 two wild boar isolates with a sustained genomic deletion (4342 base pairs) were identified (7303WB/19, 7212WB/19). In this study, we therefore performed in vitro experiments with monocyte-derived macrophages (moMФ) to unravel the phenotypic characteristics of these deleted viruses. Both 7303WB/19 and 7212WB/19 presented a lower growth kinetic in moMФ compared to virulent Sardinian 26544/OG10, using either a high (1) or a low (0.01) multiplicity of infection (MOI). In addition, flow cytometric analysis showed that both 7303WB/19 and 7212WB/19 presented lower intracellular levels of both early and late ASFV proteins. We subsequently investigated whether deleted virus variants were previously circulating in wild boars in Sardinia. In the four years preceding the last genotype I isolation (February 2015-January 2019), other eight wild boar isolates were collected, all belonging to p72 genotype I, B602L subgroup X, but none of them presented a sustained genomic deletion. Overall, we observed the deleted virus isolates in Sardinia only in 2019, at the end of a strong eradication campaign, and our data suggest that it might possess an attenuated phenotype in vivo. A better understanding of ASFV evolution in endemic territories might contribute to development of effective control measures against ASF.


Subject(s)
African Swine Fever Virus , African Swine Fever , Genotype , Sus scrofa , Animals , African Swine Fever Virus/genetics , African Swine Fever Virus/physiology , Swine , Italy , African Swine Fever/virology , Genome, Viral , Phenotype , Sequence Deletion , Macrophages/virology
6.
Arch Virol ; 169(7): 147, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38879716

ABSTRACT

African swine fever virus (ASFV) isolates are grouped and tracked through analysis of their central variable region (CVR) sequences. In this study, sequences of 70 ASFV isolates collected from different regions of Russia between 2018 and 2022 were analyzed. The analysis based on the CVR sequences indicated that the isolates belonged to three distinct groups. Group 1 shared 100% sequence identity to the isolate Georgia 2007/1. Group 5 had a C > A single-nucleotide polymorphism (SNP) at position 601, while group 13 is new and unique to the Far East of Russia, with five isolates from the Amur, Khabarovsk, and Primorsky regions. These findings demonstrate a new approach to phylogenomics and cladistics of ASFV isolates within genotype II on the basis of the CVR.


Subject(s)
African Swine Fever Virus , African Swine Fever , Genotype , Phylogeny , African Swine Fever Virus/genetics , African Swine Fever Virus/classification , African Swine Fever Virus/isolation & purification , Animals , Russia , African Swine Fever/virology , Swine , Polymorphism, Single Nucleotide
7.
Arch Virol ; 169(7): 137, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38847873

ABSTRACT

The present study focuses on the pathological and molecular characterization of African swine fever virus (ASFV) associated with an outbreak in wild boars in two national parks in southern India in 2022-2023. Significant mortality was observed among free-ranging wild boars at Bandipur National Park, Karnataka, and Mudumalai National Park, Tamil Nadu. Extensive combing operations were undertaken in both national parks, spanning an area of around 100 km2, originating from the reported epicenter, to estimate the mortality rate. Recovered carcasses were pathologically examined, and ASFV isolates was genetically characterized. Our findings suggested spillover infection of ASFV from nearby domestic pigs, and the virus was equally pathogenic in wild boars and domestic pigs. ASFV intrusion was reported in the Northeastern region of the country, which borders China and Myanmar, whereas the current outbreak is very distantly located, in southern India. Molecular data will help in tracing the spread of the virus in the country.


Subject(s)
African Swine Fever Virus , African Swine Fever , Disease Outbreaks , Sus scrofa , Animals , African Swine Fever Virus/genetics , African Swine Fever Virus/isolation & purification , India/epidemiology , Swine , African Swine Fever/virology , African Swine Fever/epidemiology , African Swine Fever/mortality , Sus scrofa/virology , Disease Outbreaks/veterinary , Phylogeny , Animals, Wild/virology
8.
Arch Virol ; 169(7): 145, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38864875

ABSTRACT

Since 2020, African swine fever (ASF) has affected all pig breeds in Northeast India except Doom pigs, a unique indigenous breed from Assam and the closest relatives of Indian wild pigs. ASF outbreaks result in significant economic losses for pig farmers in the region. Based on sequencing and phylogenetic analysis of the B646L (p72) gene, it has been determined that ASFV genotype II is responsible for outbreaks in this region. Recent studies have shown that MYD88, LDHB, and IFIT1, which are important genes of the immune system, are involved in the pathogenesis of ASFV. The differential expression patterns of these genes in surviving ASFV-infected and healthy Doom breed pigs were compared to healthy controls at different stages of infection. The ability of Doom pigs to withstand common pig diseases, along with their genetic resemblance to wild pigs, make them ideal candidates for studying tolerance to ASFV infection. In the present study, we investigated the natural resistance to ASF in Doom pigs from an endemic area in Northeast India. The results of this study provide important molecular insights into the regulation of ASFV tolerance genes.


Subject(s)
African Swine Fever Virus , African Swine Fever , Disease Outbreaks , Phylogeny , Animals , African Swine Fever/virology , African Swine Fever/epidemiology , African Swine Fever/immunology , African Swine Fever Virus/genetics , African Swine Fever Virus/immunology , India/epidemiology , Swine , Disease Outbreaks/veterinary , Genotype , Myeloid Differentiation Factor 88/genetics , Disease Resistance/genetics
9.
Infect Genet Evol ; 122: 105612, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38824981

ABSTRACT

African swine fever (ASF) is a serious animal disease, and has spread to Africa, Europe and Asia, causing massive economic losses. African swine fever virus (ASFV) is transmitted from a reservoir host (warthog) to domestic pigs via a sylvatic cycle (transmission between warthogs and soft ticks) and a domestic cycle (transmission between domestic pigs) and survives by expressing a variety of genes related to virus-host interactions. We evaluated differences in codon usage patterns among ASFV genotypes and clades and explored the common and specific evolutionary and genetic characteristics of ASFV sequences. We analysed the evolutionary relationships, nucleotide compositions, codon usage patterns, selection pressures (mutational pressure and natural selection) and viral adaptation to host codon usage based on the coding sequences (CDS) of key functional genes of ASFV. AT bias was detected in the six genes analysed, irrespective of clade. The AT bias of genes (A224L, A179L, EP153R) encoding proteins involved in interaction with host cells after infection was high; among them, the AT bias of EP153R was the greatest at 78.3%. A large number of overrepresented codons were identified in EP153R, whereas there were no overrepresented codons with a relative synonymous codon usage (RSCU) value of ≥3 in B646L. In most genes, the pattern of selection pressure was similar for each clade, but in EP153R, diverse patterns of selection pressure were captured within the same clade and genotype. As a result of evaluating host adaptation based on the codon adaptation index (CAI), for B646L, E183L, CP204L and A179L, the codon usage patterns in all sequences were more similar to tick than domestic pig or wild boar. However, EP153R showed the lowest average CAI value of 0.52 when selecting tick as a reference set. The genes analysed in this study showed different magnitudes of selection pressure at the clade and genotype levels, which is likely to be related to the function of the encoded proteins and may determine key evolutionary traits of viruses, such as the level of genetic variation and host range. The diversity of codon adaptations at the genetic level in ASFV may account for differences in translational selection in ASFV hosts and provides insight into viral host adaptation and co-evolution.


Subject(s)
African Swine Fever Virus , African Swine Fever , Codon Usage , Evolution, Molecular , Selection, Genetic , African Swine Fever Virus/genetics , African Swine Fever Virus/classification , Animals , Swine , African Swine Fever/virology , African Swine Fever/genetics , Phylogeny , Genotype
10.
Viruses ; 16(6)2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38932205

ABSTRACT

African swine fever (ASF) is a contagious viral disease affecting pigs and wild boars. It typically presents as a hemorrhagic fever but can also manifest in various forms, ranging from acute to asymptomatic. ASF has spread extensively globally, significantly impacting the swine industry. The complex and highly variable character of the ASFV genome makes vaccine development and disease surveillance extremely difficult. The overall trend in ASFV evolution is towards decreased virulence and increased transmissibility. Factors such as gene mutation, viral recombination, and the strain-specificity of virulence-associated genes facilitate viral variations. This review deeply discusses the influence of these factors on viral immune evasion, pathogenicity, and the ensuing complexities encountered in vaccine development, disease detection, and surveillance. The ultimate goal of this review is to thoroughly explore the genetic evolution patterns and variation mechanisms of ASFV, providing a theoretical foundation for advancement in vaccine and diagnostic technologies.


Subject(s)
African Swine Fever Virus , African Swine Fever , Genetic Variation , Genome, Viral , African Swine Fever Virus/genetics , Animals , Swine , African Swine Fever/virology , Virulence , Viral Vaccines/immunology , Viral Vaccines/genetics , Evolution, Molecular , Immune Evasion/genetics , Mutation , Vaccine Development
11.
Viruses ; 16(6)2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38932241

ABSTRACT

African swine fever (ASF) is an acute, hemorrhagic, highly contagious disease in pigs caused by African swine fever virus (ASFV). Our previous study identified that the ASFV MGF300-2R protein functions as a virulence factor and found that MGF300-2R degrades IKKß via selective autophagy. However, the E3 ubiquitin ligase responsible for IKKß ubiquitination during autophagic degradation still remains unknown. In order to solve this problem, we first pulled down 328 proteins interacting with MGF300-2R through immunoprecipitation-mass spectrometry. Next, we analyzed and confirmed the interaction between the E3 ubiquitin ligase TRIM21 and MGF300-2R and demonstrated the catalytic role of TRIM21 in IKKß ubiquitination. Finally, we indicated that the degradation of IKKß by MGF300-2R was dependent on TRIM21. In summary, our results indicate TRIM21 is the E3 ubiquitin ligase involved in the degradation of IKKß by MGF300-2R, thereby augmenting our understanding of the functions of MGF300-2R and offering insights into the rational design of live attenuated vaccines and antiviral strategies against ASF.


Subject(s)
African Swine Fever Virus , I-kappa B Kinase , Ribonucleoproteins , Ubiquitin-Protein Ligases , Ubiquitination , Viral Proteins , Animals , African Swine Fever Virus/metabolism , African Swine Fever Virus/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Swine , I-kappa B Kinase/metabolism , Ribonucleoproteins/metabolism , Ribonucleoproteins/genetics , Viral Proteins/metabolism , Viral Proteins/genetics , African Swine Fever/virology , African Swine Fever/metabolism , Humans , HEK293 Cells , Host-Pathogen Interactions , Virulence Factors/metabolism , Autophagy , Protein Binding
13.
Virol Sin ; 39(3): 469-477, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38789040

ABSTRACT

Virus replication relies on complex interactions between viral proteins. In the case of African swine fever virus (ASFV), only a few such interactions have been identified so far. In this study, we demonstrate that ASFV protein p72 interacts with p11.5 using co-immunoprecipitation and liquid chromatography-mass spectrometry (LC-MS). It was found that protein p72 interacts specifically with p11.5 â€‹at sites amino acids (aa) 1-216 of p72 and aa 1-68 of p11.5. To assess the importance of p11.5 in ASFV infection, we developed a recombinant virus (ASFVGZΔA137R) by deleting the A137R gene from the ASFVGZ genome. Compared with ASFVGZ, the infectious progeny virus titers of ASFVGZΔA137R were reduced by approximately 1.0 logs. In addition, we demonstrated that the growth defect was partially attributable to a higher genome copies-to-infectious virus titer ratios produced in ASFVGZΔA137R-infected MA104 â€‹cells than in those infected with ASFVGZ. This finding suggests that MA104 â€‹cells infected with ASFVGZΔA137R may generate larger quantities of noninfectious particles. Importantly, we found that p11.5 did not affect virus-cell binding or endocytosis. Collectively, we show for the first time the interaction between ASFV p72 and p11.5. Our results effectively provide the relevant information of the p11.5 protein. These results extend our understanding of complex interactions between viral proteins, paving the way for further studies of the potential mechanisms and pathogenesis of ASFV infection.


Subject(s)
African Swine Fever Virus , Viral Proteins , Virus Replication , African Swine Fever Virus/genetics , African Swine Fever Virus/physiology , Animals , Swine , Viral Proteins/metabolism , Viral Proteins/genetics , Chlorocebus aethiops , African Swine Fever/virology , African Swine Fever/metabolism , Cell Line , Protein Binding , Chromatography, Liquid , Vero Cells , Mass Spectrometry
14.
Microbes Infect ; 26(5-6): 105348, 2024.
Article in English | MEDLINE | ID: mdl-38697277

ABSTRACT

African swine fever virus (ASFV) infection causes African swine fever (ASF), a highly contagious and fatal disease that poses severe threat to swine production. To gain insights into the host responses to ASFV, we generated recombinant adenovirus Ad5 expressing viral membrane proteins p54, p17, and pB117L individually and infected an alveolar cell line, 3D4/21, with these recombinant viruses. Then, the cell lysates were analyzed using label-free quantification proteomic analysis method. A total of 2158 differentially expressed proteins (DEPs) were identified, of which 817, 466, and 875 proteins were from Ad5-p54-, Ad5-p17-, Ad5-pB117L-infected 3D4/21 cells, respectively. Gene Ontology (GO) classification and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis revealed distinct yet interconnecting patterns of protein interaction networks. Specifically, the Ad5-p54 virus infection enriched the DEPs primarily involved in the metabolic pathways, endocytosis, adherens junction, and SNARE interactions in vesicular transport. The Ad5-p17 virus infection enriched the DEPs in endocytosis, ubiquitin-mediated proteolysis, N-Glycan biosynthesis, and apoptosis, while the Ad5-pB117L virus infection enriched the DEPs in metabolic pathways, endocytosis, oxidative phosphorylation, and focal adhesion. In summary, these results provide a comprehensive proteinomics analysis of the cellular responses to three ASFV membrane proteins, thus facilitating our understanding of ASFV pathogenesis.


Subject(s)
African Swine Fever Virus , African Swine Fever , Proteomics , African Swine Fever Virus/genetics , African Swine Fever Virus/physiology , African Swine Fever Virus/metabolism , Animals , Swine , Proteomics/methods , Cell Line , African Swine Fever/virology , African Swine Fever/metabolism , Host-Pathogen Interactions , Protein Interaction Maps , Membrane Proteins/metabolism , Membrane Proteins/genetics , Viral Matrix Proteins/metabolism , Viral Matrix Proteins/genetics
15.
Front Immunol ; 15: 1380220, 2024.
Article in English | MEDLINE | ID: mdl-38799458

ABSTRACT

African swine fever (ASF) is an acute hemorrhagic and devastating infectious disease affecting domestic pigs and wild boars. It is caused by the African swine fever virus (ASFV), which is characterized by genetic diversity and sophisticated immune evasion strategies. To facilitate infection, ASFV encodes multiple proteins to antagonize host innate immune responses, thereby contributing to viral virulence and pathogenicity. The molecular mechanisms employed by ASFV-encoded proteins to modulate host antiviral responses have not been comprehensively elucidated. In this study, it was observed that the ASFV MGF505-6R protein, a member of the multigene family 505 (MGF505), effectively suppressed the activation of the interferon-beta (IFN-ß) promoter, leading to reduced mRNA levels of antiviral genes. Additional evidence has revealed that MGF505-6R antagonizes the cGAS-STING signaling pathway by interacting with the stimulator of interferon genes (STING) for degradation in the autophagy-lysosomal pathway. The domain mapping revealed that the N-terminal region (1-260aa) of MGF505-6R is the primary domain responsible for interacting with STING, while the CTT domain of STING is crucial for its interaction with MGF505-6R. Furthermore, MGF505-6R also inhibits the activation of STING by reducing the K63-linked polyubiquitination of STING, leading to the disruption of STING oligomerization and TANK binding kinase 1 (TBK1) recruitment, thereby impairing the phosphorylation and nuclear translocation of interferon regulatory factor 3 (IRF3). Collectively, our study elucidates a novel strategy developed by ASFV MGF505-6R to counteract host innate immune responses. This discovery may offer valuable insights for further exploration of ASFV immune evasion mechanisms and antiviral strategies.


Subject(s)
African Swine Fever Virus , African Swine Fever , Membrane Proteins , Viral Proteins , Animals , African Swine Fever Virus/immunology , African Swine Fever Virus/genetics , Swine , Membrane Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/immunology , African Swine Fever/immunology , African Swine Fever/virology , African Swine Fever/metabolism , Viral Proteins/immunology , Viral Proteins/metabolism , Viral Proteins/genetics , Humans , Immunity, Innate , Interferon Type I/metabolism , Interferon Type I/immunology , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-3/immunology , Signal Transduction , Proteolysis , HEK293 Cells , Host-Pathogen Interactions/immunology , Immune Evasion , Interferon-beta/metabolism , Interferon-beta/immunology , Interferon-beta/genetics
16.
Int J Biol Macromol ; 270(Pt 1): 132432, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38761609

ABSTRACT

The African swine fever virus (ASFV) continues to pose significant economic and pandemic risks. Consequently, discovering new, efficient vaccines is crucial. Messenger RNA (mRNA) vaccines have emerged as promising candidates, providing minimal risk of insertional mutagenesis, high safety profiles, effectiveness, rapid scalability in production, and cost-effectiveness. In this study, we have developed an ASF p30 mRNA vaccine candidate (mRNA/Man-LNP) employing mannose-modified lipid nanoparticles (LNPs). The mRNA/Man-LNP exhibited effective antigen presentation and facilitated dendritic cells (DCs) maturation. Notably, it elicited strong IgG titers and activated CD4+ and CD8+ T-cells in immunized mice, all while adhering to stringent biosafety standards. This investigation demonstrates that mRNA/Man-LNP can trigger both humoral and cellular immune responses, suggesting its potential as a potent and promising vaccine candidate for controlling African swine fever (ASF).


Subject(s)
African Swine Fever Virus , African Swine Fever , Mannose , Nanoparticles , Viral Vaccines , Animals , Nanoparticles/chemistry , African Swine Fever Virus/immunology , African Swine Fever Virus/genetics , African Swine Fever/prevention & control , African Swine Fever/immunology , Mice , Viral Vaccines/immunology , Swine , Mannose/chemistry , Dendritic Cells/immunology , Lipids/chemistry , Vaccine Development , RNA, Messenger/genetics , RNA, Messenger/immunology , mRNA Vaccines , Female , Antibodies, Viral/immunology , Antibodies, Viral/blood , Liposomes
17.
Appl Microbiol Biotechnol ; 108(1): 350, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38809284

ABSTRACT

The African swine fever virus (ASFV) has the ability to infect pigs and cause a highly contagious acute fever that can result in a mortality rate as high as 100%. Due to the viral epidemic, the pig industry worldwide has suffered significant financial setbacks. The absence of a proven vaccine for ASFV necessitates the development of a sensitive and reliable serological diagnostic method, enabling laboratories to effectively and expeditiously detect ASFV infection. In this study, four strains of monoclonal antibodies (mAbs) against p72, namely, 5A1, 4C4, 8A9, and 5E10, were generated through recombinant expression of p72, the main capsid protein of ASFV, and immunized mice with it. Epitope localization was performed by truncated overlapping polypeptides. The results indicate that 5A1 and 4C4 recognized the amino acid 20-39 aa, 8A9 and 5E10 are recognized at 263-282 aa, which is consistent with the reported 265-280 aa epitopes. Conserved analysis revealed 20-39 aa is a high conservation of the epitopes in the ASFV genotypes. Moreover, a blocking ELISA assay for detection ASFV antibody based on 4C4 monoclonal antibody was developed and assessed. The receiver-operating characteristic (ROC) was performed to identify the best threshold value using 87 negative and 67 positive samples. The established test exhibited an area under the curve (AUC) of 0.9997, with a 95% confidence interval ranging from 99.87 to 100%. Furthermore, the test achieved a diagnostic sensitivity of 100% (with a 95% confidence interval of 95.72 to 100%) and a specificity of 98.51% (with a 95% confidence interval of 92.02 to 99.92%) when the threshold was set at 41.97%. The inter- and intra-batch coefficient of variation were below 10%, demonstrating the exceptional repeatability of the method. This method can detect the positive standard serum at a dilution as high as 1:512. Subsequently, an exceptional blocking ELISA assay was established with high diagnostic sensitivity and specificity, providing a novel tool for detecting ASFV antibodies. KEY POINTS: • Four strains of ASFV monoclonal antibodies against p72 were prepared and their epitopes were identified. • Blocking ELISA method was established based on monoclonal antibody 4C4 with an identified conservative epitope. • The established blocking ELISA method has a good effect on the detection of ASFV antibody.


Subject(s)
African Swine Fever Virus , African Swine Fever , Antibodies, Monoclonal , Antibodies, Viral , Capsid Proteins , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Animals , Antibodies, Monoclonal/immunology , African Swine Fever Virus/immunology , African Swine Fever Virus/genetics , Enzyme-Linked Immunosorbent Assay/methods , Antibodies, Viral/blood , Antibodies, Viral/immunology , Swine , African Swine Fever/diagnosis , African Swine Fever/immunology , African Swine Fever/virology , Mice , Capsid Proteins/immunology , Capsid Proteins/genetics , Mice, Inbred BALB C , Sensitivity and Specificity , Epitopes/immunology
18.
Front Immunol ; 15: 1361531, 2024.
Article in English | MEDLINE | ID: mdl-38698849

ABSTRACT

The whole-genome sequence of an African swine fever virus (ASFV) strain (HuB/HH/2019) isolated from Hubei, China, was highly similar to that of the Georgia 2007/1 strain ASFV. After infection with strong strains, domestic pigs show typical symptoms of infection, including fever, depression, reddening of the skin, hemorrhagic swelling of various tissues, and dysfunction. The earliest detoxification occurred in pharyngeal swabs at 4 days post-infection. The viral load in the blood was extremely high, and ASFV was detected in multiple tissues, with the highest viral loads in the spleen and lungs. An imbalance between pro- and anti-inflammatory factors in the serum leads to an excessive inflammatory response in the body. Immune factor expression is suppressed without effectively eliciting an immune defense. Antibodies against p30 were not detected in acutely dead domestic pigs. Sequencing of the peripheral blood mononuclear cell transcriptome revealed elevated transcription of genes associated with immunity, defense, and stress. The massive reduction in lymphocyte counts in the blood collapses the body's immune system. An excessive inflammatory response with a massive reduction in the lymphocyte count may be an important cause of mortality in domestic pigs. These two reasons have inspired researchers to reduce excessive inflammatory responses and stimulate effective immune responses for future vaccine development.


Subject(s)
African Swine Fever Virus , African Swine Fever , Animals , Swine , African Swine Fever/virology , African Swine Fever/immunology , African Swine Fever Virus/immunology , African Swine Fever Virus/genetics , Cytokines , Lymphocytes/immunology , Lymphocytes/metabolism , Genotype , Viral Load , Sus scrofa , Lymphocyte Count
19.
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
20.
Microb Pathog ; 191: 106669, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38697231

ABSTRACT

African swine fever (ASF) is a lethal disease caused by ASF virus (ASFV), severely impacting the global swine industry. Though nuclear acid-based detection methods are reliable, they are laboratory-dependent. In this study, we developed a device-independent, user friendly and cost-effective quantum dots based immunochromatographic strip (QDs-ICS) with high specificity and sensitivity for the rapid and on-site detection of ASFV antigen. For the preparation of the QDs-ICS, we generated a monoclonal antibody (mAb) mAb-8G8 and polyclonal antibody (pAb) against ASFV-p72 protein. The pAb was labelled with QDs to be used as the detection probe and the mAb-8G8 was coated on the nitrocellulose membrane as the test line. Our results proved that the strip displayed no cross-reactivity with other swine viruses and detection limit of the QDs-ICS was down to 1 ng/mL for the ASFV-p72 protein with great reproducibility. The strip also exhibited high stability with a storage period up to 12 months under room temperature. Twenty blind samples and one hundred clinical samples were examined by the QDs-ICS, conventional PCR and real-time PCR method, respectively. Results showed that the agreement rate between the QDs-ICS and PCR method was 100%, and the agreement rate between the strip and real-time PCR was 94%. The novel QDs-ICS developed here would be an effective tool for on-site detection of ASFV.


Subject(s)
African Swine Fever Virus , African Swine Fever , Antibodies, Monoclonal , Antibodies, Viral , Antigens, Viral , Chromatography, Affinity , Quantum Dots , Sensitivity and Specificity , African Swine Fever Virus/isolation & purification , African Swine Fever Virus/immunology , African Swine Fever Virus/genetics , Animals , African Swine Fever/diagnosis , African Swine Fever/virology , African Swine Fever/immunology , Swine , Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Chromatography, Affinity/methods , Antigens, Viral/analysis , Antigens, Viral/immunology , Reproducibility of Results , Reagent Strips
SELECTION OF CITATIONS
SEARCH DETAIL
...