Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Front Vet Sci ; 11: 1390486, 2024.
Article in English | MEDLINE | ID: mdl-38868498

ABSTRACT

Activation-induced markers (AIMs) are frequently analyzed to identify re-activated human memory T cells. However, in pigs the analysis of AIMs is still not very common. Based on available antibodies, we designed a multi-color flow cytometry panel comprising pig-specific or cross-reactive antibodies against CD25, CD69, CD40L (CD154), and ICOS (CD278) combined with lineage/surface markers against CD3, CD4, and CD8α. In addition, we included an antibody against tumor necrosis factor alpha (TNF-α), to study the correlation of AIM expression with the production of this abundant T cell cytokine. The panel was tested on peripheral blood mononuclear cells (PBMCs) stimulated with phorbol 12-myristate 13-acetate (PMA)/ionomycin, Staphylococcus enterotoxin B (SEB) or PBMCs from African swine fever virus (ASFV) convalescent pigs, restimulated with homologous virus. PMA/ionomycin resulted in a massive increase of CD25/CD69 co-expressing T cells of which only a subset produced TNF-α, whereas CD40L expression was largely associated with TNF-α production. SEB stimulation triggered substantially less AIM expression than PMA/ionomycin but also here CD25/CD69 expressing T cells were identified which did not produce TNF-α. In addition, CD40L-single positive and CD25+CD69+CD40L+TNF-α- T cells were identified. In ASFV restimulated T cells TNF-α production was associated with a substantial proportion of AIM expressing T cells but also here ASFV-reactive CD25+CD69+TNF-α- T cells were identified. Within CD8α+ CD4 T cells, several CD25/CD40L/CD69/ICOS defined phenotypes expanded significantly after ASFV restimulation. Hence, the combination of AIMs tested will allow the identification of primed T cells beyond the commonly used cytokine panels, improving capabilities to identify the full breadth of antigen-specific T cells in pigs.

2.
Sci Rep ; 14(1): 5944, 2024 03 11.
Article in English | MEDLINE | ID: mdl-38467747

ABSTRACT

African swine fever (ASF) is a global threat to animal health and food security. ASF is typically controlled by strict biosecurity, rapid diagnosis, and culling of affected herds. Much progress has been made in developing modified live virus vaccines against ASF. There is host variation in response to ASF infection in the field and under controlled conditions. To better understand the dynamics underlying this host differential morbidity, whole transcriptome profiling was carried out in twelve immunized and five sham immunized pigs. Seventeen MHC homozygous inbred Large white Babraham pigs were sampled at three time points before and after the challenge. The changes in the transcriptome profiles of infected animals were surveyed over time. In addition, the immunization effect on the host response was studied as well among the contrasts of all protection subgroups. The results showed two promising candidate genes to distinguish between recovered and non-recovered pigs after infection with a virulent African swine fever virus (ASFV) pre-infection: HTRA3 and GFPT2 (padj < 0.05). Variant calling on the transcriptome assemblies showed a two-base pair insertion into the ACOX3 gene closely located to HTRA3 that may regulate its expression as a putative genomic variant for ASF. Several significant DGEs, enriched gene ontology (GO) terms, and KEGG pathways at 1 day and 7 days post-infection, compared to the pre-infection, indicate a significant inflammation response immediately after ASF infection. The presence of the virus was confirmed by the mapping of RNA-Seq reads on two whole viral genome sequences. This was concordant with a higher virus load in the non-recovered animals 7 days post-infection. There was no transcriptome signature on the immunization at pre-infection and 1 day post-infection. More samples and data from additional clinical trials may support these findings.


Subject(s)
African Swine Fever Virus , African Swine Fever , Swine , Animals , African Swine Fever Virus/genetics , Transcriptome , Gene Expression Profiling , Immunization
3.
Microbiol Resour Announc ; 13(4): e0006724, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38526091

ABSTRACT

African swine fever virus causes a lethal hemorrhagic disease of domestic pigs. The NAM P1/1995 isolate was originally described as B646L genotype XVIII; however, full genome sequencing revealed that this assignment was incorrect.

4.
Subcell Biochem ; 106: 283-331, 2023.
Article in English | MEDLINE | ID: mdl-38159232

ABSTRACT

African swine fever virus is a complex double-stranded DNA virus that exhibits tropism for cells of the mononuclear phagocytic system. Virus replication is a multi-step process that involves the nucleus of the host cell as well the formation of large perinuclear sites where progeny virions are assembled prior to transport to, and budding through, the plasma membrane. Like many viruses, African swine fever virus reorganises the cellular architecture to facilitate its replication and has evolved multiple mechanisms to avoid the potential deleterious effects of host cell stress response pathways. However, how viral proteins and virus-induced structures trigger cellular stress pathways and manipulate the subsequent responses is still relatively poorly understood. African swine fever virus alters nuclear substructures, modulates autophagy, apoptosis and the endoplasmic reticulum stress response pathways. The viral genome encodes for at least 150 genes, of which approximately 70 are incorporated into the virion. Many of the non-structural genes have not been fully characterised and likely play a role in host range and modifying immune responses. As the field moves towards approaches that take a broader view of the effect of expression of individual African swine fever genes, we summarise how the different steps in virus replication interact with the host cell and the current state of knowledge on how it modulates the resulting stress responses.


Subject(s)
African Swine Fever Virus , African Swine Fever , Swine , Animals , African Swine Fever Virus/genetics , African Swine Fever/metabolism , Viral Proteins/genetics , Host-Pathogen Interactions , Virus Replication
5.
Vaccines (Basel) ; 11(10)2023 Oct 10.
Article in English | MEDLINE | ID: mdl-37896980

ABSTRACT

African swine fever (ASF) is a lethal disease in pigs that has grave socio-economic implications worldwide. For the development of vaccines against the African swine fever virus (ASFV), immunogenic antigens that generate protective immune responses need to be identified. There are over 150 viral proteins-many of which are uncharacterized-and humoral immunity to ASFV has not been closely examined. To profile antigen-specific antibody responses, we developed luciferase-linked antibody capture assays (LACAs) for a panel of ASFV capsid proteins and screened sera from inbred and outbred animals that were previously immunized with low-virulent ASFV before challenge with virulent ASFV. Antibodies to B646L/p72, D117L/p17, M1249L, and E120R/p14.5 were detected in this study; however, we were unable to detect B438L-specific antibodies. Anti-B646L/p72 and B602L antibodies were associated with recovery from disease after challenges with genotype I OUR T88/1 but not genotype II Georgia 2007/1. Antibody responses against M1249L and E120R/p14.5 were observed in animals with reduced clinical signs and viremia. Here, we present LACAs as a tool for the targeted profiling of antigen-specific antibody responses to inform vaccine development.

6.
Pathogens ; 12(8)2023 Aug 08.
Article in English | MEDLINE | ID: mdl-37623982

ABSTRACT

African swine fever virus (ASFV) is known to be very stable and can remain infectious over long periods of time especially at low temperatures and within different matrices, particularly those containing animal-derived organic material. However, there are some gaps in our knowledge pertaining to the survivability and infectivity of ASFV in groundwater. This study aims to determine the stability and infectivity of the cell culture-adapted ASFV strain BA71V by plaque assay after incubation of the virus within river water samples at three different environmentally relevant temperatures (4 °C, 15 °C, and 21 °C) over the course of 42 days. The results from this study indicate that ASFV can remain stable and infectious when maintained at 4 °C in river water for more than 42 days, but as incubation temperatures are increased, the stability is reduced, and the virus is no longer able to form plaques after 28 days and 14 days, respectively, when stored at 15 °C and 21 °C. Characterizing the survivability of ASFV in groundwater can allow us to develop more appropriate inactivation and disinfection methods to support disease control and mitigate ASFV outbreaks.

7.
Viruses ; 15(8)2023 07 27.
Article in English | MEDLINE | ID: mdl-37631977

ABSTRACT

Viral replication fully relies on the host cell machinery, and physical interactions between viral and host proteins mediate key steps of the viral life cycle. Therefore, identifying virus-host protein-protein interactions (PPIs) provides insights into the molecular mechanisms governing virus infection and is crucial for designing novel antiviral strategies. In the case of the African swine fever virus (ASFV), a large DNA virus that causes a deadly panzootic disease in pigs, the limited understanding of host and viral targets hinders the development of effective vaccines and treatments. This review summarizes the current knowledge of virus-host and virus-virus PPIs by collecting and analyzing studies of individual viral proteins. We have compiled a dataset of experimentally determined host and virus protein targets, the molecular mechanisms involved, and the biological functions of the identified virus-host and virus-virus protein interactions during infection. Ultimately, this work provides a comprehensive and systematic overview of ASFV interactome, identifies knowledge gaps, and proposes future research directions.


Subject(s)
African Swine Fever Virus , Animals , Swine , Antiviral Agents , Microbial Interactions , Virus Replication
8.
Sci Rep ; 13(1): 5318, 2023 03 31.
Article in English | MEDLINE | ID: mdl-37002287

ABSTRACT

African swine fever (ASF) caused by ASF virus (ASFV) is an infectious transboundary animal disease notifiable to the World Organization for Animal Health causing high mortality in domestic pigs and wild boars threatening the global domestic pig industry. To date, twenty-four ASFV genotypes have been described and currently genotypes II, IX, X, XV and XVI are known to be circulating in Tanzania. Despite the endemic status of ASF in Tanzania, only one complete genome of ASFV from the country has been described. This study describes the first complete genome sequence of ASFV genotype XV. In addition, the first Tanzanian complete genome of ASFV genotype IX and three ASFV strains belonging to genotype II collected during ASF outbreaks in domestic pigs in Tanzania were determined in this study using Illumina sequencing and comparative genomics analysis. The generated ASFV complete genome sequences ranged from 171,004 to 184,521 base pairs in length with an average GC content of 38.53% and encoded 152 to 187 open reading frames. The results of this study provide insights into the genomic structure of ASFV and can be used to monitor changes within the ASFV genome and improve our understanding of ASF transmission dynamics.


Subject(s)
African Swine Fever Virus , African Swine Fever , Swine , Animals , Sus scrofa , African Swine Fever Virus/genetics , Tanzania/epidemiology , Genotype
9.
Viruses ; 16(1)2023 12 19.
Article in English | MEDLINE | ID: mdl-38275939

ABSTRACT

The 2023 International African Swine Fever Workshop (IASFW) took place in Beijing, China, on 18-20 September 2023. It was jointly organized by the U.S.-China Center for Animal Health (USCCAH) at Kansas State University (KSU) and the Chinese Veterinary Drug Association (CVDA) and sponsored by the United States Department of Agriculture Foreign Agricultural Service (USDA-FAS), Harbin Veterinary Research Institute, and Zoetis Inc. The objective of this workshop was to provide a platform for ASF researchers around the world to unite and share their knowledge and expertise on ASF control and prevention. A total of 24 outstanding ASF research scientists and experts from 10 countries attended this meeting. The workshop included presentations on current ASF research, opportunities for scientific collaboration, and discussions of lessons and experiences learned from China/Asia, Africa, and Europe. This article summarizes the meeting highlights and presents some critical issues that need to be addressed for ASF control and prevention in the future.


Subject(s)
African Swine Fever Virus , African Swine Fever , Swine , Animals , Humans , African Swine Fever/prevention & control , African Swine Fever/epidemiology , Asia , China/epidemiology , Africa/epidemiology , Sus scrofa , Disease Outbreaks/veterinary
10.
Viruses ; 14(7)2022 07 07.
Article in English | MEDLINE | ID: mdl-35891467

ABSTRACT

African swine fever virus is currently present in all of the world's continents apart from Antarctica, and efforts to control the disease are hampered by the lack of a commercially available vaccine. The Babraham large white pig is a highly inbred line that could represent a powerful tool to improve our understanding of the protective immune responses to this complex pathogen; however, previous studies indicated differential vaccine responses after the African swine fever virus challenge of inbred minipigs with different swine leukocyte antigen haplotypes. Lymphocyte numbers and African swine fever virus-specific antibody and T-cell responses were measured in inbred and outbred animals after inoculation with a low virulent African swine fever virus isolate and subsequent challenge with a related virulent virus. Surprisingly, diminished immune responses were observed in the Babraham pigs when compared to the outbred animals, and the inbred pigs were not protected after challenge. Recovery of Babraham pigs after challenge weakly correlated with antibody responses, whereas protective responses in outbred animals more closely correlated with the T-cell response. The Babraham pig may, therefore, represent a useful model for studying the role of antibodies in protection against the African swine fever virus.


Subject(s)
African Swine Fever Virus , African Swine Fever , Viral Vaccines , Animals , Immunity, Humoral , Immunization , Swine , Swine, Miniature
11.
Methods Mol Biol ; 2503: 63-72, 2022.
Article in English | MEDLINE | ID: mdl-35575886

ABSTRACT

Primary cultures represent the most reliable method to isolate and propagate field isolates of African swine fever virus (ASFV ). Within the pig ASFV predominantly targets the reticuloendothelial system for replication; therefore, primary macrophage cell cultures are commonly used to isolate, propagate, and study the virus life cycle in the laboratory. In this chapter we will describe methods for the direct isolation of pulmonary alveolar macrophages by lung lavage and the culture of monocyte-derived macrophages from pig blood. We also include a method for the positive selection of CD14+ monocytes as a source for monocyte-derived macrophages from pig blood using microbeads.


Subject(s)
African Swine Fever Virus , African Swine Fever , Animals , Cells, Cultured , Lung , Macrophages , Swine
12.
Methods Mol Biol ; 2503: 95-104, 2022.
Article in English | MEDLINE | ID: mdl-35575888

ABSTRACT

Real-time polymerase chain reaction (PCR) for the detection of African swine fever virus (ASFV) is the tool of choice for the diagnostic laboratory and is a robust and easily scalable method for the researcher analyzing viral replication both in vitro and in vivo. In this chapter, we describe protocols for both quantitative real-time polymerase chain reactions (qPCR) and non-quantitative real-time polymerase chain reactions (real-time PCR) for the detection of African swine fever virus genome in a range of samples.


Subject(s)
African Swine Fever Virus , African Swine Fever , African Swine Fever/diagnosis , African Swine Fever Virus/genetics , Animals , Clinical Laboratory Techniques , Real-Time Polymerase Chain Reaction/methods , Sensitivity and Specificity , Swine
13.
Methods Mol Biol ; 2503: 179-186, 2022.
Article in English | MEDLINE | ID: mdl-35575895

ABSTRACT

African swine fever virus is a cytolytic virus that leads to the apoptosis of both cultured cells and primary macrophages. Cell culture supernatants of virus-infected cells are routinely used for virological and immunological studies, despite differences in the biological behavior between such preparations and highly purified virus. In addition, more recent data suggests that exosomes containing viral proteins may be secreted from infected cells. While African swine fever virus can be purified through a number of methods, in our hands Percoll provides the most robust method of separating virus from cellular contaminants.


Subject(s)
African Swine Fever Virus , African Swine Fever , Animals , Cell Line , Cells, Cultured , DNA Viruses , Swine , Viral Proteins
14.
Pathogens ; 11(2)2022 Feb 20.
Article in English | MEDLINE | ID: mdl-35215216

ABSTRACT

African swine fever virus (ASFV) remains a threat to global pig populations. Infections with ASFV lead to a hemorrhagic disease with up to 100% lethality in Eurasian domestic and wild pigs. Although myeloid cells are the main target cells for ASFV, T cell responses are impacted by the infection as well. The complex responses remain not well understood, and, consequently, there is no commercially available vaccine. Here, we review the current knowledge about the induction of antiviral T cell responses by cells of the myeloid lineage, as well as T cell responses in infected animals, recent efforts in vaccine research, and T cell epitopes present in ASFV.

15.
Vet Med Sci ; 8(2): 607-609, 2022 03.
Article in English | MEDLINE | ID: mdl-34967133

ABSTRACT

BACKGROUND: African swine fever (ASF) is a highly contagious and deadly viral disease affecting domestic and wild pigs of all ages. African swine fever virus (ASFV) has spread rapidly through Eastern and Southeastern Asia first appearing in Vietnam in 2019. OBJECTIVES: Molecular typing of African swine fever virus (ASFV) in Vietnam has identified two principal variants circulating based on the sequencing of the intergenic region (IRG) between the I73R and I329L genes. Identification of additional genetic markers would enable higher resolution tracing of outbreaks within the country. METHODS: Sequence analysis suggested the IRG between the A179L and A137R genes may also exhibit variability, PCR primers were designed and samples from Vietnam were subject to Sanger sequencing. RESULTS: We developed a novel method for sub-grouping of ASFV based on the IRG between the A179L and A137R genes of ASFV. Our results demonstrated that the finding of the insertion or deletion of an 11- nucleotide sequence (GATACAATTGT) between the A179L-A137R genes. CONCLUSIONS: The sub-grouping method may provide useful insights into the evolution of genotype II ASFV as well as providing evidence of a relationship between geographically separated outbreaks.


Subject(s)
African Swine Fever Virus , African Swine Fever , Swine Diseases , African Swine Fever/epidemiology , African Swine Fever Virus/genetics , Animals , DNA, Intergenic/genetics , Genotype , Phylogeny , Sequence Analysis, DNA/veterinary , Sus scrofa/genetics , Swine
16.
Viruses ; 13(11)2021 10 28.
Article in English | MEDLINE | ID: mdl-34834981

ABSTRACT

Deoxyuridine 5'-triphosphate nucleotidohydrolase (dUTPase) of African swine fever virus (ASFV) is an essential enzyme required for efficient virus replication. Previous crystallography data have indicated that dUTPase (E165R) may serve as a therapeutic target for inhibiting ASFV replication; however, the specificity of the targeting site(s) in ASFV dUTPase remains unclear. In this study, 19 mouse monoclonal antibodies (mAbs) were produced, in which four mAbs showed inhibitory reactivity against E165R recombinant protein. Epitope mapping studies indicated that E165R has three major antigenic regions: 100-120 aa, 120-140 aa, and 140-165 aa. Three mAbs inhibited the dUTPase activity of E165R by binding to the highly conserved 149-RGEGRFGSTG-158 amino acid sequence. Interestingly, 8F6 mAb specifically recognized ASFV dUTPase but not Sus scrofa dUTPase, which may be due to structural differences in the amino acids of F151, R153, and F154 in the motif V region. In summary, we developed anti-E165R-specific mAbs, and identified an important antibody-binding antigenic epitope in the motif V of ASFV dUTPase. Our study provides a comprehensive analysis of mAbs that target the antigenic epitope of ASFV dUTPase, which may contribute to the development of novel antibody-based ASFV therapeutics.


Subject(s)
African Swine Fever Virus/genetics , African Swine Fever Virus/immunology , Antibodies, Monoclonal/immunology , Epitopes/isolation & purification , Pyrophosphatases/genetics , African Swine Fever/virology , Amino Acid Sequence , Animals , Antibodies, Viral/immunology , Antigens, Viral/immunology , Epitope Mapping , Epitopes/genetics , Recombinant Proteins , Swine , Virus Replication
17.
J Gen Virol ; 102(8)2021 08.
Article in English | MEDLINE | ID: mdl-34406116

ABSTRACT

African swine fever is a devastating disease of domestic swine and wild boar caused by a large double-stranded DNA virus that encodes for more than 150 open reading frames. There is no licensed vaccine for the disease and the most promising current candidates are modified live viruses that have been attenuated by deletion of virulence factors. Like many viruses African swine fever virus significantly alters the host cell machinery to benefit its replication and viral genes that modify host pathways represent promising targets for development of gene deleted vaccines. Autophagy is an important cellular pathway that is involved in cellular homeostasis, innate and adaptive immunity and therefore is manipulated by a number of different viruses. Autophagy is regulated by a complex protein cascade and here we show that African swine fever virus can block formation of autophagosomes, a critical functional step of the autophagy pathway through at least two different mechanisms. Interestingly this does not require the A179L gene that has been shown to interact with Beclin-1, an important autophagy regulator.


Subject(s)
African Swine Fever Virus/pathogenicity , African Swine Fever/virology , Viral Proteins/metabolism , Animals , Autophagy , Chlorocebus aethiops , Swine , Vero Cells , Virulence
18.
Viruses ; 13(1)2021 Jan 08.
Article in English | MEDLINE | ID: mdl-33429879

ABSTRACT

African swine fever virus (ASFV) is a highly contagious pathogen which causes a lethal haemorrhagic fever in domestic pigs and wild boar. The large, double-stranded DNA virus replicates in perinuclear cytoplasmic replication sites known as viral factories. These factories are complex, multi-dimensional structures. Here we investigated the protein and membrane compartments of the factory using super-resolution and electron tomography. Click IT chemistry in combination with stimulated emission depletion (STED) microscopy revealed a reticular network of newly synthesized viral proteins, including the structural proteins p54 and p34, previously seen as a pleomorphic ribbon by confocal microscopy. Electron microscopy and tomography confirmed that this network is an accumulation of membrane assembly intermediates which take several forms. At early time points in the factory formation, these intermediates present as small, individual membrane fragments which appear to grow and link together, in a continuous progression towards new, icosahedral virions. It remains unknown how these membranes form and how they traffic to the factory during virus morphogenesis.


Subject(s)
African Swine Fever Virus/physiology , African Swine Fever/virology , Virus Replication , African Swine Fever Virus/ultrastructure , Animals , Cell Culture Techniques , Chlorocebus aethiops , Fluorescent Antibody Technique , Swine , Vero Cells , Virus Assembly
19.
J Vet Diagn Invest ; 32(6): 961-963, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32964810

ABSTRACT

African swine fever (ASF) is a devastating viral disease of pigs and wild boar, and it threatens global food security. We aimed to identify suitable sample matrices for use in ASF surveillance programs. Six pigs inoculated with ASFV were sampled at postmortem. Blood, bone marrow, ear biopsies, and oral, nasal, and rectal swabs were taken from all pigs. All samples were analyzed using 3 real-time PCR (rtPCR) assays and a LAMP assay. ASFV was detected at > 107 genome copies/mL in blood; bone marrow was found to provide the highest viral load. Ct values provided by the rtPCR assays were correlated, and ASFV was detected in all oral, nasal, and rectal swabs and in all ear biopsy samples irrespective of the location from which they were taken. The LAMP assay had lower sensitivity, and detected ASFV in 54 of 66 positive samples, but delivered positive results within 17 min. We identified additional sample matrices that can be considered depending on the sampling situation: bone marrow had a high probability of detection, which could be useful for decomposed carcasses. However, ear biopsies provide an appropriate, high-throughput sample matrix to detect ASFV and may be useful during surveillance programs.


Subject(s)
African Swine Fever/diagnosis , Population Surveillance/methods , African Swine Fever Virus/genetics , Animals , DNA, Viral/genetics , Molecular Diagnostic Techniques/standards , Molecular Diagnostic Techniques/veterinary , Nucleic Acid Amplification Techniques/standards , Nucleic Acid Amplification Techniques/veterinary , Real-Time Polymerase Chain Reaction/standards , Real-Time Polymerase Chain Reaction/veterinary , Sensitivity and Specificity , Swine
20.
J Virol ; 94(21)2020 10 14.
Article in English | MEDLINE | ID: mdl-32796064

ABSTRACT

African swine fever virus (ASFV) causes a lethal hemorrhagic disease of domestic pigs, against which no vaccine is available. ASFV has a large, double-stranded DNA genome that encodes over 150 proteins. Replication takes place predominantly in the cytoplasm of the cell and involves complex interactions with host cellular components, including small noncoding RNAs (sncRNAs). A number of DNA viruses are known to manipulate sncRNA either by encoding their own or disrupting host sncRNA. To investigate the interplay between ASFV and sncRNAs, a study of host and viral small RNAs extracted from ASFV-infected primary porcine macrophages (PAMs) was undertaken. We discovered that ASFV infection had only a modest effect on host miRNAs, with only 6 miRNAs differentially expressed during infection. The data also revealed 3 potential novel small RNAs encoded by ASFV, ASFVsRNA1-3. Further investigation of ASFVsRNA2 detected it in lymphoid tissue from pigs with ASF. Overexpression of ASFVsRNA2 led to an up to 1-log reduction in ASFV growth, indicating that ASFV utilizes a virus-encoded small RNA to disrupt its own replication.IMPORTANCE African swine fever (ASF) poses a major threat to pig populations and food security worldwide. The disease is endemic to Africa and Eastern Europe and is rapidly emerging into Asia, where it has led to the deaths of millions of pigs in the last 12 months. The development of safe and effective vaccines to protect pigs against ASF has been hindered by lack of understanding of the complex interactions between ASFV and the host cell. We focused our work on characterizing the interactions between ASFV and sncRNAs. Although comparatively modest changes to host sncRNA abundances were observed upon ASFV infection, we discovered and characterized a novel functional ASFV-encoded sncRNA. The results from this study add important insights into ASFV host-pathogen interactions. This knowledge may be exploited to develop more effective ASFV vaccines that take advantage of the sncRNA system.


Subject(s)
African Swine Fever Virus/genetics , African Swine Fever/genetics , Genome, Viral , Host-Pathogen Interactions/genetics , MicroRNAs/genetics , RNA, Small Untranslated/genetics , RNA, Viral/genetics , African Swine Fever/metabolism , African Swine Fever/virology , African Swine Fever Virus/metabolism , Animals , Gene Expression Regulation , Genome Size , Lymphoid Tissue , Macrophages , MicroRNAs/classification , MicroRNAs/metabolism , Primary Cell Culture , RNA, Small Untranslated/classification , RNA, Small Untranslated/metabolism , RNA, Viral/classification , RNA, Viral/metabolism , Signal Transduction , Sus scrofa , Swine , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...