Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.219
Filter
2.
Antiviral Res ; 226: 105900, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38705200

ABSTRACT

BACKGROUND & AIMS: The spread of foot-and-mouth disease virus (FMDV) through aerosol droplets among cloven-hoofed ungulates in close contact is a major obstacle for successful animal husbandry. Therefore, the development of suitable mucosal vaccines, especially nasal vaccines, to block the virus at the initial site of infection is crucial. PATIENTS AND METHODS: Here, we constructed eukaryotic expression plasmids containing the T and B-cell epitopes (pTB) of FMDV in tandem with the molecular mucosal adjuvant Fms-like tyrosine kinase receptor 3 ligand (Flt3 ligand, FL) (pTB-FL). Then, the constructed plasmid was electrostatically attached to mannose-modified chitosan-coated poly(lactic-co-glycolic) acid (PLGA) nanospheres (MCS-PLGA-NPs) to obtain an active nasal vaccine targeting the mannose-receptor on the surface of antigen-presenting cells (APCs). RESULTS: The MCS-PLGA-NPs loaded with pTB-FL not only induced a local mucosal immune response, but also induced a systemic immune response in mice. More importantly, the nasal vaccine afforded an 80% protection rate against a highly virulent FMDV strain (AF72) when it was subcutaneously injected into the soles of the feet of guinea pigs. CONCLUSIONS: The nasal vaccine prepared in this study can effectively induce a cross-protective immune response against the challenge with FMDV of same serotype in animals and is promising as a potential FMDV vaccine.


Subject(s)
Administration, Intranasal , Chitosan , Foot-and-Mouth Disease Virus , Foot-and-Mouth Disease , Nanospheres , Polylactic Acid-Polyglycolic Acid Copolymer , Viral Vaccines , Animals , Chitosan/chemistry , Chitosan/administration & dosage , Foot-and-Mouth Disease Virus/immunology , Foot-and-Mouth Disease Virus/genetics , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Foot-and-Mouth Disease/prevention & control , Foot-and-Mouth Disease/immunology , Mice , Nanospheres/chemistry , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Mice, Inbred BALB C , Antibodies, Viral/blood , Antibodies, Viral/immunology , Female , Nucleic Acids/administration & dosage , Immunity, Mucosal , Drug Delivery Systems
3.
Emerg Microbes Infect ; 13(1): 2348526, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38683015

ABSTRACT

The foot-and-mouth disease virus (FMDV) Leader proteinase Lpro inhibits host mRNA translation and blocks the interferon response which promotes viral survival. Lpro is not required for viral replication in vitro but serotype A FMDV lacking Lpro has been shown to be attenuated in cattle and pigs. However, it is not known, whether leaderless viruses can cause persistent infection in vivo after simulated natural infection and whether the attenuated phenotype is the same in other serotypes. We have generated an FMDV O/FRA/1/2001 variant lacking most of the Lpro coding region (ΔLb). Cattle were inoculated intranasopharyngeally and observed for 35 days to determine if O FRA/1/2001 ΔLb is attenuated during the acute phase of infection and whether it can maintain a persistent infection in the upper respiratory tract. We found that although this leaderless virus can replicate in vitro in different cell lines, it is unable to establish an acute infection with vesicular lesions and viral shedding nor is it able to persistently infect bovine pharyngeal tissues.


Subject(s)
Cattle Diseases , Foot-and-Mouth Disease Virus , Foot-and-Mouth Disease , Persistent Infection , Serogroup , Virus Replication , Animals , Cattle , Foot-and-Mouth Disease Virus/genetics , Foot-and-Mouth Disease Virus/physiology , Foot-and-Mouth Disease Virus/classification , Foot-and-Mouth Disease Virus/pathogenicity , Foot-and-Mouth Disease Virus/isolation & purification , Foot-and-Mouth Disease/virology , Cattle Diseases/virology , Persistent Infection/virology , Cell Line , Endopeptidases/genetics , Endopeptidases/metabolism , Virus Shedding
4.
Viruses ; 16(4)2024 04 17.
Article in English | MEDLINE | ID: mdl-38675963

ABSTRACT

Southern Africa Territories 2 (SAT2) foot-and-mouth disease (FMD) has crossed long-standing regional boundaries in recent years and entered the Middle East. However, the existing vaccines offer poor cross-protection against the circulating strains in the field. Therefore, there is an urgent need for an alternative design approach for vaccines in anticipation of a pandemic of SAT2 Foot-and-mouth disease virus (FMDV). The porcine parvovirus (PPV) VP2 protein can embed exogenous epitopes into the four loops on its surface, assemble into virus-like particles (VLPs), and induce antibodies and cytokines to PPV and the exogenous epitope. In this study, chimeric porcine parvovirus VP2 VLPs (chimeric PPV-SAT2-VLPs) expressing the T-and/or B-cell epitopes of the structural protein VP1 of FMDV SAT2 were produced using the recombinant pFastBac™ Dual vector of baculoviruses in Sf9 and HF cells We used the Bac-to-Bac system to construct the recombinant baculoviruses. The VP2-VLP--SAT2 chimeras displayed chimeric T-cell epitope (amino acids 21-40 of VP1) and/or the B-cell epitope (amino acids 135-174) of SAT FMDV VP1 by substitution of the corresponding regions at the N terminus (amino acids 2-23) and/or loop 2 and/or loop 4 of the PPV VP2 protein, respectively. In mice, the chimeric PPV-SAT2-VLPs induced specific antibodies against PPV and the VP1 protein of SAT2 FMDV. The VP2-VLP-SAT2 chimeras induced specific antibodies to PPV and the VP1 protein specific epitopes of FMDV SAT2. In this study, as a proof-of-concept, successfully generated chimeric PPV-VP2 VLPs expressing epitopes of the structural protein VP1 of FMDV SAT2 that has a potential to prevent FMDV SAT2 and PPV infection in pigs.


Subject(s)
Antibodies, Viral , Antigens, Viral , Capsid Proteins , Foot-and-Mouth Disease Virus , Foot-and-Mouth Disease , Parvovirus, Porcine , Vaccines, Virus-Like Particle , Viral Vaccines , Animals , Foot-and-Mouth Disease Virus/immunology , Foot-and-Mouth Disease Virus/genetics , Mice , Foot-and-Mouth Disease/immunology , Foot-and-Mouth Disease/prevention & control , Foot-and-Mouth Disease/virology , Capsid Proteins/immunology , Capsid Proteins/genetics , Parvovirus, Porcine/immunology , Parvovirus, Porcine/genetics , Antibodies, Viral/immunology , Antibodies, Viral/blood , Viral Vaccines/immunology , Viral Vaccines/genetics , Vaccines, Virus-Like Particle/immunology , Vaccines, Virus-Like Particle/genetics , Swine , Immunity, Humoral , Immunity, Cellular , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/genetics , Serogroup , Mice, Inbred BALB C , Female , Epitopes/immunology , Epitopes/genetics , Sf9 Cells , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood
5.
Arch Virol ; 169(5): 101, 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38630189

ABSTRACT

Foot-and-mouth disease is a highly contagious disease affecting cloven-hoofed animals, resulting in considerable economic losses. Its causal agent is foot-and-mouth disease virus (FMDV), a picornavirus. Due to its error-prone replication and rapid evolution, the transmission and evolutionary dynamics of FMDV can be studied using genomic epidemiological approaches. To analyze FMDV evolution and identify possible transmission routes in an Argentinean region, field samples that tested positive for FMDV by PCR were obtained from 21 farms located in the Mar Chiquita district. Whole FMDV genome sequences were obtained by PCR amplification in seven fragments and sequencing using the Sanger technique. The genome sequences obtained from these samples were then analyzed using phylogenetic, phylogeographic, and evolutionary approaches. Three local transmission clusters were detected among the sampled viruses. The dataset was analyzed using Bayesian phylodynamic methods with appropriate coalescent and relaxed molecular clock models. The estimated mean viral evolutionary rate was 1.17 × 10- 2 substitutions/site/year. No significant differences in the rate of viral evolution were observed between farms with vaccinated animals and those with unvaccinated animals. The most recent common ancestor of the sampled sequences was dated to approximately one month before the first reported case in the outbreak. Virus transmission started in the south of the district and later dispersed to the west, and finally arrived in the east. Different transmission routes among the studied herds, such as non-replicating vectors and close contact contagion (i.e., aerosols), may be responsible for viral spread.


Subject(s)
Foot-and-Mouth Disease Virus , Picornaviridae , Animals , Foot-and-Mouth Disease Virus/genetics , Argentina/epidemiology , Bayes Theorem , Phylogeny
6.
Virulence ; 15(1): 2333562, 2024 12.
Article in English | MEDLINE | ID: mdl-38622757

ABSTRACT

The Picornaviridae are a large group of positive-sense, single-stranded RNA viruses, and most research has focused on the Enterovirus genus, given they present a severe health risk to humans. Other picornaviruses, such as foot-and-mouth disease virus (FMDV) and senecavirus A (SVA), affect agricultural production with high animal mortality to cause huge economic losses. The 3Dpol protein of picornaviruses is widely known to be used for genome replication; however, a growing number of studies have demonstrated its non-polymerase roles, including modulation of host cell biological processes, viral replication complex assembly and localization, autophagy, and innate immune responses. Currently, there is no effective vaccine to control picornavirus diseases widely, and clinical therapeutic strategies have limited efficiency in combating infections. Many efforts have been made to develop different types of drugs to prohibit virus survival; the most important target for drug development is the virus polymerase, a necessary element for virus replication. For picornaviruses, there are also active efforts in targeted 3Dpol drug development. This paper reviews the interaction of 3Dpol proteins with the host and the progress of drug development targeting 3Dpol.


Subject(s)
Enterovirus , Foot-and-Mouth Disease Virus , Picornaviridae Infections , Animals , Humans , Gene Products, pol/metabolism , Foot-and-Mouth Disease Virus/genetics , Foot-and-Mouth Disease Virus/metabolism , Virus Replication , RNA, Viral/genetics
7.
Sci Rep ; 14(1): 7929, 2024 04 04.
Article in English | MEDLINE | ID: mdl-38575673

ABSTRACT

Foot and mouth disease (FMD) is a highly contagious, endemic, and acute viral cattle ailment that causes major economic damage in Ethiopia. Although several serotypes of the FMD virus have been detected in Ethiopia, there is no documented information about the disease's current serostatus and serotypes circulating in the Wolaita zone. Thus, from March to December 2022, a cross-sectional study was conducted to evaluate FMDV seroprevalence, molecular detection, and serotype identification in three Wolaita Zone sites. A multistage sample procedure was used to choose three peasant associations from each study region, namely Wolaita Sodo, Offa district, and Boloso sore district. A systematic random sampling technique was employed to pick 384 cattle from the population for the seroprevalence research, and 10 epithelial tissue samples were purposefully taken from outbreak individuals for molecular detection of FMDV. The sera were examined using 3ABC FMD NSP Competition ELISA to find antibodies against FMDV non-structural proteins, whereas epithelial tissue samples were analyzed for molecular detection using real-time RT-PCR, and sandwich ELISA was used to determine the circulating serotypes. A multivariable logistic regression model was used to evaluate the associated risk variables. The total seroprevalence of FMD in cattle was 46.88% (95% CI 41.86-51.88), with Wolaita Sodo Town having the highest seroprevalence (63.28%). As a consequence, multivariable logistic regression analysis revealed that animal age, herd size, and interaction with wildlife were all substantially related to FMD seroprevalence (p < 0.05). During molecular detection, only SAT-2 serotypes were found in 10 tissue samples. Thus, investigating FMD outbreaks and identifying serotypes and risk factors for seropositivity are critical steps in developing effective control and prevention strategies based on the kind of circulating serotype. Moreover, further research for animal species other than cattle was encouraged.


Subject(s)
Cattle Diseases , Foot-and-Mouth Disease Virus , Foot-and-Mouth Disease , Humans , Cattle , Animals , Foot-and-Mouth Disease Virus/genetics , Seroepidemiologic Studies , Cross-Sectional Studies , Ethiopia/epidemiology , Cattle Diseases/diagnosis , Cattle Diseases/epidemiology , Foot-and-Mouth Disease/diagnosis , Foot-and-Mouth Disease/epidemiology , Serogroup , Disease Outbreaks/veterinary , Animals, Wild , Antibodies, Viral
8.
Virology ; 595: 110070, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38657363

ABSTRACT

Foot-and-mouth disease is a highly contagious and infectious disease affecting cloven-hoofed animals. Disease control is complicated by its highly contagious nature and antigenic diversity. Host microRNAs (miRNAs) are post-transcriptional regulators that either promote or repress viral replications in virus infection. In the present study, we found that ssc-miR-7139-3p (Sus scrofa miR-7139-3p) was significantly up-regulated in host cells during foot-and-mouth disease virus (FMDV) infection. Overexpression of miR-7139-3p attenuated FMDV replication, whereas inhibition promoted FMDV replication. In addition, the survival rate of FMDV infected suckling mice was increased through injection of miR-7139-3p agomiR. Further studies revealed that miR-7139-3p targets Bcl-2 to initiate the apoptotic pathway and caspase-3 cleaved 3Cpro behind the 174th aspartic acid (D174), which eventually promotes the degradation of 3Cpro. Overall, our findings demonstrate that miR-7139-3p suppresses FMDV replication by promoting degradation of 3Cpro through targeting the apoptosis-negative regulatory gene Bcl-2.


Subject(s)
Apoptosis , Foot-and-Mouth Disease Virus , Foot-and-Mouth Disease , MicroRNAs , Proto-Oncogene Proteins c-bcl-2 , Virus Replication , Animals , Foot-and-Mouth Disease Virus/genetics , Foot-and-Mouth Disease Virus/physiology , MicroRNAs/genetics , MicroRNAs/metabolism , Foot-and-Mouth Disease/virology , Mice , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Swine , Viral Proteins/genetics , Viral Proteins/metabolism , 3C Viral Proteases/metabolism , Cell Line , Sus scrofa , Host-Pathogen Interactions , Cysteine Endopeptidases/metabolism , Cysteine Endopeptidases/genetics , Proteolysis , Caspase 3/metabolism , Caspase 3/genetics
9.
Viruses ; 16(4)2024 03 27.
Article in English | MEDLINE | ID: mdl-38675855

ABSTRACT

The foot-and-mouth disease virus is a highly contagious and economically devastating virus of cloven-hooved animals, including cattle, buffalo, sheep, and goats, causing reduced animal productivity and posing international trade restrictions. For decades, chemically inactivated vaccines have been serving as the most effective strategy for the management of foot-and-mouth disease. Inactivated vaccines are commercially produced in cell culture systems, which require successful propagation and adaptation of field isolates, demanding a high cost and laborious time. Cell culture adaptation is chiefly indebted to amino acid substitutions in surface-exposed capsid proteins, altering the necessity of RGD-dependent receptors to heparan sulfate macromolecules for virus binding. Several amino acid substations in VP1, VP2, and VP3 capsid proteins of FMDV, both at structural and functional levels, have been characterized previously. This literature review combines frequently reported amino acid substitutions in virus capsid proteins, their critical roles in virus adaptation, and functional characterization of the substitutions. Furthermore, this data can facilitate molecular virologists to develop new vaccine strains against the foot-and-mouth disease virus, revolutionizing vaccinology via reverse genetic engineering and synthetic biology.


Subject(s)
Amino Acid Substitution , Capsid Proteins , Foot-and-Mouth Disease Virus , Foot-and-Mouth Disease , Viral Tropism , Foot-and-Mouth Disease Virus/genetics , Foot-and-Mouth Disease Virus/metabolism , Animals , Capsid Proteins/genetics , Capsid Proteins/metabolism , Capsid Proteins/chemistry , Foot-and-Mouth Disease/virology , Receptors, Virus/metabolism , Receptors, Virus/genetics , Viral Structural Proteins/genetics , Viral Structural Proteins/metabolism , Cell Culture Techniques
10.
PLoS Pathog ; 20(3): e1012104, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38512977

ABSTRACT

The interaction between foot-and-mouth disease virus (FMDV) and the host is extremely important for virus infection, but there are few researches on it, which is not conducive to vaccine development and FMD control. In this study, we designed a porcine genome-scale CRISPR/Cas9 knockout library containing 93,859 single guide RNAs targeting 16,886 protein-coding genes, 25 long ncRNAs, and 463 microRNAs. Using this library, several previously unreported genes required for FMDV infection are highly enriched post-FMDV selection in IBRS-2 cells. Follow-up studies confirmed the dependency of FMDV on these genes, and we identified a functional role for one of the FMDV-related host genes: TOB1 (Transducer of ERBB2.1). TOB1-knockout significantly inhibits FMDV infection by positively regulating the expression of RIG-I and MDA5. We further found that TOB1-knockout led to more accumulation of mRNA transcripts of transcription factor CEBPA, and thus its protein, which further enhanced transcription of RIG-I and MDA5 genes. In addition, TOB1-knockout was shown to inhibit FMDV adsorption and internalization mediated by EGFR/ERBB2 pathway. Finally, the FMDV lethal challenge on TOB1-knockout mice confirmed that the deletion of TOB1 inhibited FMDV infection in vivo. These results identify TOB1 as a key host factor involved in FMDV infection in pigs.


Subject(s)
Foot-and-Mouth Disease Virus , Foot-and-Mouth Disease , Animals , Mice , ErbB Receptors/metabolism , Foot-and-Mouth Disease/genetics , Foot-and-Mouth Disease Virus/genetics , Gene Expression Regulation , RNA, Guide, CRISPR-Cas Systems , Swine
11.
Microbiol Spectr ; 12(3): e0365823, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38323828

ABSTRACT

The internal ribosome entry site (IRES) element constitutes a cis-acting RNA regulatory sequence that recruits the ribosomal initiation complex in a cap-independent manner, assisted by various RNA-binding proteins and IRES trans-acting factors. Foot-and-mouth disease virus (FMDV) contains a functional IRES element and takes advantage of this element to subvert host translation machinery. Our study identified a novel mechanism wherein RALY, a member of the heterogeneous nuclear ribonucleoproteins (hnRNP) family belonging to RNA-binding proteins, binds to the domain 3 of FMDV IRES via its RNA recognition motif residue. This interaction results in the downregulation of FMDV replication by inhibiting IRES-driven translation. Furthermore, our findings reveal that the inhibitory effect exerted by RALY on FMDV replication is not attributed to the FMDV IRES-mediated assembly of translation initiation complexes but rather to the impediment of 80S ribosome complex formation after binding with 40S ribosomes. Conversely, 3Cpro of FMDV counteracts RALY-mediated inhibition by the ubiquitin-proteasome pathway. Therefore, these results indicate that RALY, as a novel critical IRES-binding protein, inhibits FMDV replication by blocking the formation of 80S ribosome, providing a deeper understanding of how viruses recruit and manipulate host factors. IMPORTANCE: The translation of FMDV genomic RNA driven by IRES element is a crucial step for virus infections. Many host proteins are hijacked to regulate FMDV IRES-dependent translation, but the regulatory mechanism remains unknown. Here, we report for the first time that cellular RALY specifically interacts with the IRES of FMDV and negatively regulates viral replication by blocking 80S ribosome assembly on FMDV IRES. Conversely, RALY-mediated inhibition is antagonized by the viral 3C protease by the ubiquitin-proteasome pathway. These results would facilitate further understanding of virus-host interactions and translational control during viral infection.


Subject(s)
Foot-and-Mouth Disease Virus , Animals , Foot-and-Mouth Disease Virus/genetics , Foot-and-Mouth Disease Virus/metabolism , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , RNA-Binding Proteins/genetics , Ribosomes/genetics , Endopeptidases/metabolism , Internal Ribosome Entry Sites , 3C Viral Proteases , Ubiquitins/genetics , Ubiquitins/metabolism
12.
Mol Biol Rep ; 51(1): 370, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38411732

ABSTRACT

BACKGROUND: Foot and mouth disease (FMD) is a highly contagious disease that impacts cloven-hoofed animals globally. The illegal trade of livestock between the border regions of Pakistan and Afghanistan can contribute to the spread of this disease. This study focuses on investigating the outbreaks of FMD that occurred in this area from June 2020 to May 2021. METHODS: RESULTS: A total of 233 epithelial tissue samples were collected, and 77% were found positive for FMDV through an antigen-detection by ELISA and molecular conformation through RT-PCR. The study found three serotypes of FMDV dominating in the border area of Pakistan with Afghanistan: O, A, and Asia-1. The outbreak activity was peaked between August/September followed by July/October 2020. Phylogenetic analysis conducted using the VP1 region sequence showed that serotype O isolates belonged to the Middle East-South Asia (ME-SA) topotype, PanAsia-2 lineage, and ANT-10 sub-lineage, while serotype Asia-1 isolates belonged to a novel lineage BD-18.The highest prevalence of serotype O of FMDV was found in cattle and buffalo of 1-2 year age group, while the highest outbreak ratio of serotype O was recorded in goats of 0-1 year age group and sheep of > 2 year age group. The serotype O was more prevalent in male than female sheep. The type A was more prevalent in females of sheep and goats than their corresponding males. The serotype Asia-1 was more prevalent in females of cattle and sheep than their corresponding males. The outbreak epidemiology of FMD varied significantly between various regions, months of study, animal species, age groups, and gender. CONCLUSIONS: The study found that FMD outbreaks in the border area of Pakistan and Afghanistan were diverse and complicated, and that different types of FMDV were circulating. The study recommended effective actions to stop FMD transmission in this area.


Subject(s)
Foot-and-Mouth Disease Virus , Female , Male , Cattle , Animals , Sheep , Foot-and-Mouth Disease Virus/genetics , Afghanistan/epidemiology , Pakistan/epidemiology , Phylogeny , Buffaloes , Goats
13.
Appl Microbiol Biotechnol ; 108(1): 81, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38194136

ABSTRACT

We engineered Saccharomyces cerevisiae to express structural proteins of foot-and-mouth disease virus (FMDV) and produce virus-like particles (VLPs). The gene, which encodes four structural capsid proteins (VP0 (VP4 and VP2), VP3, and VP1), followed by a translational "ribosomal skipping" sequence consisting of 2A and protease 3C, was codon-optimized and chemically synthesized. The cloned gene was used to transform S. cerevisiae 2805 strain. Western blot analysis revealed that the polyprotein consisting of VP0, VP3, and VP1 was processed into the discrete capsid proteins. Western blot analysis of 3C confirmed the presence of discrete 3C protein, suggesting that the 2A sequence functioned as a "ribosomal skipping" signal in the yeast for an internal re-initiation of 3C translation from a monocistronic transcript, thereby indicating polyprotein processing by the discrete 3C protease. Moreover, a band corresponding to only VP2, which was known to be non-enzymatically processed from VP0 to both VP4 and VP2 during viral assembly, further validated the assembly of processed capsid proteins into VLPs. Electron microscopy showed the presence of the characteristic icosahedral VLPs. Our results clearly demonstrate that S. cerevisiae processes the viral structural polyprotein using a viral 3C protease and the resulting viral capsid subunits are assembled into virion particles. KEY POINTS: • Ribosomal skipping by self-cleaving FMDV peptide in S. cerevisiae. • Proteolytic processing of a structural polyprotein from a monocistronic transcript. • Assembly of the processed viral capsid proteins into a virus-like particle.


Subject(s)
Foot-and-Mouth Disease Virus , Saccharomyces cerevisiae , Animals , Saccharomyces cerevisiae/genetics , Foot-and-Mouth Disease Virus/genetics , Capsid Proteins/genetics , Endopeptidases , Peptide Hydrolases , Polyproteins/genetics , 3C Viral Proteases
15.
Virology ; 590: 109950, 2024 02.
Article in English | MEDLINE | ID: mdl-38104361

ABSTRACT

Despite routine vaccination, Israel experiences recurrent outbreaks of foot and mouth disease (FMD). We analyzed VP1 coding sequences of viruses isolated during FMD outbreaks from 2001 to 2011 in Israel and neighboring nations. The Israeli strains were aligned with strains from neighboring countries in corresponding years, implying repeated FMD virus incursions. In 2007 a large FMD epidemic, caused by a serotype O virus, occurred in Israel. Bayesian analysis of whole-genome sequences of viruses isolated during this epidemic revealed predominant transmission among extensively farmed beef-cattle and small ruminants. Small ruminants were key in spreading to beef-cattle, which then transmitted the virus to feedlot-cattle. Wild gazelles had a minor role in transmission. The results may suggest probable transmission of FMD virus from the Palestinian Authority to Israel. Targeting extensive farms via enhanced surveillance and vaccination could improve FMDV control. Given cross-border transmission, a collaborative FMD mitigation strategy across the Middle-East is crucial.


Subject(s)
Cattle Diseases , Foot-and-Mouth Disease Virus , Foot-and-Mouth Disease , Cattle , Animals , Sheep , Foot-and-Mouth Disease Virus/genetics , Israel/epidemiology , Bayes Theorem , Phylogeny , Foot-and-Mouth Disease/epidemiology , Foot-and-Mouth Disease/prevention & control , Serogroup , Disease Outbreaks/veterinary , Cattle Diseases/epidemiology , Sequence Analysis , Ruminants
16.
Int J Mol Sci ; 24(23)2023 Dec 01.
Article in English | MEDLINE | ID: mdl-38069369

ABSTRACT

Mast cells can recognize foot-and-mouth disease virus-like particles (FMDV-VLPs) via mannose receptors (MRs) to produce differentially expressed cytokines. The regulatory role of chromatin accessibility in this process is unclear. Bone marrow-derived mast cells (BMMCs) were cultured, and an assay of transposase-accessible chromatin sequencing (ATAC-seq) was applied to demonstrate the regulation of chromatin accessibility in response to the BMMCs' recognition of FMDV-VLPs. A pathway enrichment analysis showed that peaks associated with the nuclear factor-κB (NF-κB), mitogen-activated protein kinase (MAPK), phosphatidylinositol 3 kinase-protein kinase B (PI3K-Akt), and other signaling pathways, especially the NF-κB pathway, were involved in the BMMCs' recognition of VLPs. Moreover, transcription factors including SP1, NRF1, AP1, GATA3, microphthalmia-associated transcription factor (MITF), and NF-κB-p65 may bind to the motifs with altered chromatin accessibility to regulate gene transcription. Furthermore, the expression of NF-κB, interleukin (IL)-9, tumor necrosis factor (TNF)-α, and interferon (IFN)-γ in the BMMCs of the VLP group increased compared with that of the BMMCs in the control group, whereas the expression of IL-10 did not differ significantly between groups. After inhibiting the MRs, the expression of NF-κB, IL-9, TNF-α, and IFN-γ decreased significantly, whereas the expression of IL-10 increased. The expression of MAPK and IL-6 showed no significant change after MR inhibition. This study demonstrated that MRs expressed on BMMCs can affect the NF-κB pathway by changing chromatin accessibility to regulate the transcription of specific cytokines, ultimately leading to the differential expression of cytokines. These data provide a theoretical basis and new ideas for the development of a novel vaccine for FMD.


Subject(s)
Foot-and-Mouth Disease Virus , NF-kappa B , Animals , NF-kappa B/metabolism , Interleukin-10 , Foot-and-Mouth Disease Virus/genetics , Phosphatidylinositol 3-Kinases/metabolism , Chromatin Immunoprecipitation Sequencing , Cytokines/metabolism , Mitogen-Activated Protein Kinases/metabolism , Tumor Necrosis Factor-alpha/metabolism , Chromatin/genetics
17.
Sheng Wu Gong Cheng Xue Bao ; 39(12): 4849-4860, 2023 Dec 25.
Article in Chinese | MEDLINE | ID: mdl-38147986

ABSTRACT

Transient expression is the major method to express foot-and-mouth disease virus (FMDV) capsid proteins in mammalian cells. To achieve stable expression of FMDV capsid proteins and efficient assembly of virus like particles (VLPs) in cells, the plasmids of piggyBac (PB) transposon-constitutive expression and PB transposon-tetracycline (Tet) inducible expression vectors were constructed. The function of the plasmids was tested by fluorescent proteins. By adding antibiotics, the constitutive cell pools (C-WT, C-L127P) expressing P12A3C (WT/L127P) genes and the inducible cell pools (I-WT, I-L127P) expressing P12A3C (WT/L127P) genes were generated. The genes of green fluorescent protein, 3C protease and reverse tetracycline transactivator (rtTA) were integrated into chromosome, which was confirmed by fluorescence observation and PCR testing. The cell pool I-L127P has a stronger production capacity of capsid proteins and VLPs, which was confirmed by Western blotting and enzyme linked immunosorbent assay (ELISA), respectively. In conclusion, inducing the chromosomal expression of FMDV capsid proteins was firstly reported, which may facilitate the technical process of mammalian production of FMDV VLPs vaccine and the construction of mammalian inducible expression systems for other proteins.


Subject(s)
Foot-and-Mouth Disease Virus , Foot-and-Mouth Disease , Viral Vaccines , Animals , Foot-and-Mouth Disease Virus/genetics , Capsid Proteins , Viral Proteins/metabolism , Foot-and-Mouth Disease/prevention & control , Tetracyclines/metabolism , Antibodies, Viral , Mammals/metabolism
18.
BMC Res Notes ; 16(1): 323, 2023 Nov 08.
Article in English | MEDLINE | ID: mdl-37941022

ABSTRACT

OBJECTIVE: Determining the serotype of circulating virus strains is important in implementing effective vaccination. In this study, Foot-and-Mouth Disease (FMD) Southern African territory 2 (SAT2) specific primers and TaqMan probe were designed towards rapid SAT2 detection and serotyping. The primers were tested by endpoint reverse transcription (RT) polymerase chain reaction (PCR) and quantitative PCR (RT-qPCR) using the vaccine strain SAT2035. The SAT2 serotype-specific RT-qPCR assay was compared with currently used ELISA and VP1 sequencing using Cohen's kappa statistics. RESULTS: The primers yielded amplicons of band size 190 bp during endpoint RT-PCR. When coupled with the probe, the primers reaction efficiency was determined to be 99% with an r2 value of 0.994. The results show that the SAT2 assay has comparable performance to VP1 sequencing (k = 1) and a moderate degree of agreement with ELISA (k = 0.571). The data shows that the newly designed assay could be considered for serotyping of SAT2 strains. However, for this assay to be complete there is a need to design effective SAT1 and SAT3 primers and probes that can be multiplexed to target other serotypes that co-circulate within relevant FMD endemic pools. For future implementation of the assay there is also a need to increase the number of field samples towards validation of the assay.


Subject(s)
Foot-and-Mouth Disease Virus , Foot-and-Mouth Disease , Animals , Foot-and-Mouth Disease Virus/genetics , Serotyping/methods , Foot-and-Mouth Disease/diagnosis , Foot-and-Mouth Disease/epidemiology , Foot-and-Mouth Disease/prevention & control , Serogroup , Africa, Southern
19.
J Gen Virol ; 104(7)2023 07.
Article in English | MEDLINE | ID: mdl-37436428

ABSTRACT

Foot-and-mouth-disease virus (FMDV), the aetiological agent responsible for foot-and-mouth disease (FMD), is a member of the genus Aphthovirus within the family Picornavirus. In common with all picornaviruses, replication of the single-stranded positive-sense RNA genome involves synthesis of a negative-sense complementary strand that serves as a template for the synthesis of multiple positive-sense progeny strands. We have previously employed FMDV replicons to examine viral RNA and protein elements essential to replication, but the factors affecting differential strand production remain unknown. Replicon-based systems require transfection of high levels of RNA, which can overload sensitive techniques such as quantitative PCR, preventing discrimination of specific strands. Here, we describe a method in which replicating RNA is labelled in vivo with 5-ethynyl uridine. The modified base is then linked to a biotin tag using click chemistry, facilitating purification of newly synthesised viral genomes or anti-genomes from input RNA. This selected RNA can then be amplified by strand-specific quantitative PCR, thus enabling investigation of the consequences of defined mutations on the relative synthesis of negative-sense intermediate and positive-strand progeny RNAs. We apply this new approach to investigate the consequence of mutation of viral cis-acting replication elements and provide direct evidence for their roles in negative-strand synthesis.


Subject(s)
Foot-and-Mouth Disease Virus , Foot-and-Mouth Disease , Picornaviridae , Animals , Foot-and-Mouth Disease Virus/genetics , Foot-and-Mouth Disease Virus/metabolism , Virus Replication/genetics , Picornaviridae/genetics , RNA, Viral/metabolism
20.
Virus Res ; 333: 199140, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37268276

ABSTRACT

Foot and mouth disease (FMD) has engendered large scale socioeconomic crises on numerous occasions owing to its extreme contagiousness, transboundary nature, complicated epidemiology, negative impact on productivity, trade embargo, and need for intensive surveillance and expensive control measures. Emerging FMD virus variants have been predicted to have originated and spread from endemic Pool 2, native to South Asia, to other parts of the globe. In this study, 26 Indian serotype A isolates sampled between the year 2015 and 2022 were sequenced for the VP1 region. BLAST and maximum likelihood phylogeny suggest emergence of a novel genetic group within genotype 18, named here as 'A/ASIA/G-18/2019' lineage, that is restricted so far only to India and its eastern neighbour, Bangladesh. The lineage subsequent to its first appearance in 2019 seems to have displaced all other prevalent strains, in support of the phenomenon of 'genotype/lineage turnover'. It has diversified into two distinct sub-clusters, reflecting a phase of active evolution. The rate of evolution of the VP1 region for the Indian serotype A dataset was estimated to be 6.747 × 10-3 substitutions/site/year. India is implementing a vaccination centric FMD control programme. The novel lineage showed good antigenic match with the proposed vaccine candidate A IND 27/2011 when tested in virus neutralization test, while the existing vaccine strain A IND 40/2000 showed homology with only 31% of the isolates. Therefore, in order to combat this challenge of antigenic divergence, A IND 27/2011 could be the preferred strain in the Indian vaccine formulations.


Subject(s)
Foot-and-Mouth Disease Virus , Foot-and-Mouth Disease , Animals , Foot-and-Mouth Disease Virus/genetics , Serogroup , Antigens, Viral , India/epidemiology , Phylogeny
SELECTION OF CITATIONS
SEARCH DETAIL
...