Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters











Publication year range
1.
Analyst ; 149(9): 2586-2593, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38497408

ABSTRACT

Nipah virus (NiV), a bat-borne zoonotic viral pathogen with high infectivity and lethality to humans, has caused severe outbreaks in several countries of Asia during the past two decades. Because of the worldwide distribution of the NiV natural reservoir, fruit bats, and lack of effective treatments or vaccines for NiV, routine surveillance and early detection are the key measures for containing NiV outbreaks and reducing its influence. In this study, we developed two rapid, sensitive and easy-to-conduct methods, RAA-CRISPR/Cas12a-FQ and RAA-CRISPR/Cas12a-FB, for NiV detection based on a recombinase-aided amplification (RAA) assay and a CRISPR/Cas12a system by utilizing dual-labeled fluorophore-quencher or fluorophore-biotin ssDNA probes. These two methods can be completed in 45 min and 55 min and achieve a limit of detection of 10 copies per µL and 100 copies per µL of NiV N DNA, respectively. In addition, they do not cross-react with nontarget nucleic acids extracted from the pathogens causing similar symptoms to NiV, showing high specificity for NiV N DNA detection. Meanwhile, they show satisfactory performance in the detection of spiked samples from pigs and humans. Collectively, the RAA-CRISPR/Cas12a-FQ and RAA-CRISPR/Cas12a-FB methods developed by us would be promising candidates for the early detection and routine surveillance of NiV in resource-poor areas and outdoors.


Subject(s)
CRISPR-Cas Systems , Nipah Virus , Virology , Animals , Humans , CRISPR-Cas Systems/genetics , DNA, Viral/genetics , DNA, Viral/analysis , Fluorescent Dyes/chemistry , Limit of Detection , Nipah Virus/genetics , Nipah Virus/isolation & purification , Nucleic Acid Amplification Techniques/methods , Virology/methods
2.
J Nanobiotechnology ; 22(1): 11, 2024 Jan 03.
Article in English | MEDLINE | ID: mdl-38167103

ABSTRACT

The pandemic caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has had a profound impact on the global health and economy. While mass vaccination for herd immunity is effective, emerging SARS-CoV-2 variants can evade spike protein-based COVID-19 vaccines. In this study, we develop a new immunization strategy by utilizing a nanocarrier, dendritic mesoporous silica nanoparticle (DMSN), to deliver the receptor-binding domain (RBD) and conserved T-cell epitope peptides (DMSN-P-R), aiming to activate both humoral and cellular immune responses in the host. The synthesized DMSN had good uniformity and dispersion and showed a strong ability to load the RBD and peptide antigens, enhancing their uptake by antigen-presenting cells (APCs) and promoting antigen delivery to lymph nodes. The DMSN-P-R vaccine elicited potent humoral immunity, characterized by highly specific RBD antibodies. Neutralization tests demonstrated significant antibody-mediated neutralizing activity against live SARS-CoV-2. Crucially, the DMSN-P-R vaccine also induced robust T-cell responses that were specifically stimulated by the RBD and conserved T-cell epitope peptides of SARS-CoV-2. The DMSN demonstrated excellent biocompatibility and biosafety in vitro and in vivo, along with degradability. Our study introduces a promising vaccine strategy that utilizes nanocarriers to deliver a range of antigens, effectively enhancing both humoral and cellular immune responses to prevent virus transmission.


Subject(s)
COVID-19 , Nanoparticles , Humans , SARS-CoV-2 , COVID-19 Vaccines , Epitopes, T-Lymphocyte , Vaccination , Antibodies, Neutralizing , Peptides , Antibodies, Viral
3.
Front Microbiol ; 11: 1449, 2020.
Article in English | MEDLINE | ID: mdl-32733405

ABSTRACT

Type A foot-and-mouth disease virus (FMDV) has been detected on China's pig farms since 2015, and all suspected samples have been strain A/GDMM/CHA/2013. To overcome the shortcomings of inactive FMDV vaccines, we expressed the capsid protein precursor P1-2A and mutated viral 3C protease of FMDV strain A/GDMM/CHA/2013 in a replication-deficient human adenovirus type 5 vector in this study. A significant humoral immune response, T-cell-mediated antiviral response, and mucosa-mediated antiviral response were induced by the adenovirus-vectored FMDV vaccines in BALB/c mice. Immunization of guinea pigs with the adenovirus-vectored FMD vaccines induced significant neutralizing antibodies and anti-FMDV immunoglobulin A antibodies. The recombinant adenovirus rAdv-P12A3CG38SF48S-GD protected 100% of guinea pigs against challenge when administered intramuscularly. Our study demonstrated the potential utility of rAdv-P12A3CG38SF48S-GD as a vaccine against type A FMDV.

4.
Antiviral Res ; 182: 104827, 2020 10.
Article in English | MEDLINE | ID: mdl-32579897

ABSTRACT

Feline calicivirus (FCV) is a highly contagious pathogen that causes acute upper respiratory infections and oral disease in cats, thus seriously endangering feline health. Recently, there have been outbreaks of particularly virulent variant strains of FCV, which can cause both acute symptoms and fatal systemic disease. The discovery of effective antiviral agents to treat FCV infection is, therefore, gradually assuming increased importance. In this study, we showed that both nitazoxanide and mizoribine had antiviral activity in F81 cells infected with different strains of FCV and also demonstrated a synergistic effect between the two drugs. Experiments in cats challenged with FCV showed that nitazoxanide significantly reduced the clinical symptoms of FCV infection, reduced viral load in the trachea and lungs, and reduced viral shedding. Our results showed that nitazoxanide and mizoribine could potentially be used as therapeutic agents to treat FCV infection.


Subject(s)
Antiviral Agents/pharmacology , Caliciviridae Infections/veterinary , Calicivirus, Feline/drug effects , Nitro Compounds/pharmacology , Ribonucleosides/pharmacology , Thiazoles/pharmacology , Viral Load/drug effects , Animals , Antiviral Agents/therapeutic use , Caliciviridae Infections/drug therapy , Calicivirus, Feline/pathogenicity , Cat Diseases/drug therapy , Cat Diseases/virology , Cats , Cell Line , Drug Synergism , Female , Lung/drug effects , Lung/virology , Nitro Compounds/therapeutic use , Ribonucleosides/therapeutic use , Thiazoles/therapeutic use , Trachea/drug effects , Trachea/virology , Virus Shedding/drug effects
5.
J Med Virol ; 91(9): 1595-1601, 2019 09.
Article in English | MEDLINE | ID: mdl-31032977

ABSTRACT

Foot-and-mouth disease (FMD) is a highly contagious disease that affects cloven-hoof animals including cattle, swine, sheep, goats, and lots of wild species. Effectively control measures are urged needed. Here, we showed that homoharringtonine treatment exhibited a strong inhibitory effect against two different strains of FMDVs (O/MYA98/BY/2010 and A/GD/MM/2013) in swine kidney (IBRS-2) cells. Further experiments demonstrated that homoharringtonine did not affect virus attachment or entry. Using time-of-addition assays, we found that the antiviral activity of homoharringtonine occurred primarily during the early stage of infection. These results demonstrated that homoharringtonine might be an effective anti-FMDV drug. Further studies are required to explore the antiviral activity of homoharringtonine against FMDV replication in vivo.


Subject(s)
Antiviral Agents/pharmacology , Foot-and-Mouth Disease Virus/drug effects , Foot-and-Mouth Disease/virology , Homoharringtonine/pharmacology , Animals , Antiviral Agents/chemistry , Cell Line , Foot-and-Mouth Disease Virus/physiology , Homoharringtonine/chemistry , Humans , Molecular Structure , Virus Internalization , Virus Replication/drug effects
6.
Vaccine ; 37(14): 2016-2025, 2019 03 28.
Article in English | MEDLINE | ID: mdl-30808570

ABSTRACT

Foot-and-mouth disease virus (FMDV) is the etiological agent of a highly contagious disease that affects cloven-hoofed animal species. The FMDV capsid is highly acid labile and viral particles lose their immunogenicity when they disassemble at mildly acidic pHs. The viral capsid of FMDV serotype O is more sensitive than those of other serotypes, making it more difficult to acquire enough empty-capsid-like particles in the acidic insect cell environment for research. In this study, novel FMDV mutants with increased acid resistance were isolated using BHK-21 cell cultured under low-pH conditions. Amino acid substitutions Q25R, K41E, and N85A in the VP1 capsid protein and K154Q in the VP3 capsid protein were detected in all six mutants. Based on these amino acid replacements, empty-capsid-like particles of FMDV serotype O, which were resistant to the acid-induced dissociation of the capsid into pentameric subunits, were produced in insect cells. We characterized the protective immunity induced by these acid-resistant empty capsid particles. Significant humoral and cellular immune responses were elicited in mice after immunization with the acid-resistant empty capsid particles. The acid-resistant empty-capsid-like particles also induced strong neutralizing antibodies in guinea pigs and protected all the guinea pigs from FMDV challenge. Our results suggest that these acid-resistant empty-capsid-like particles have potential utility as a vaccine against serotype O FMDV infection.


Subject(s)
Capsid Proteins/immunology , Foot-and-Mouth Disease Virus/immunology , Foot-and-Mouth Disease/prevention & control , Immunogenicity, Vaccine , Vaccines, Virus-Like Particle/immunology , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Capsid Proteins/chemistry , Guinea Pigs , Immunity, Cellular , Mice , Outcome Assessment, Health Care , Serogroup , Vaccines, Virus-Like Particle/administration & dosage , Viral Vaccines/administration & dosage
7.
J Med Virol ; 91(2): 208-214, 2019 02.
Article in English | MEDLINE | ID: mdl-30039874

ABSTRACT

Foot-and-mouth disease (FMD) is a disease of worldwide economic importance, and vaccines play an important role in preventing FMDV outbreaks. However, new control strategies are still needed since FMDV outbreaks still occur in some disease-free countries. Currently, interferon (IFN)-based strategies have been demonstrated to be an efficient biotherapeutic option against FMDV; however, interferon omega (IFN-ω) has not yet been assessed in this capacity. Thus, this study evaluated the antiviral activity of porcine IFN omega 7 (PoIFN-ω7) against FMDV. After the PoIFN-ω7 was expressed and purified, cell proliferation assays and quantitative real-time reverse transciption-polymerase chain reaction were used to evaluate the effective anti-cytopathic concentration of PoIFN-ω7 and its effectiveness pre- and post-infection with FMDV in swine kidney cells (IBRS-2). Results showed the rHis-PoIFN-ω7 fusion protein was considerably expressed using Escherichia coli BL21 (DE3) strain, and the recombinant protein exhibited significant in vitro protection against FMDV, including two strains belonging to type O and A FMDV, respectively. In addition, PoIFN-ω7 upregulated the transcription of Mx1, ISG15, OAS1, and PKR genes. These findings indicated that IFN-ω has the potential for serving as a useful therapeutic agent to prevent FMDV or other viral outbreaks in pigs.


Subject(s)
Antiviral Agents/pharmacology , Foot-and-Mouth Disease Virus/drug effects , Foot-and-Mouth Disease Virus/growth & development , Interferon Type I/pharmacology , Recombinant Fusion Proteins/pharmacology , Animals , Cytopathogenic Effect, Viral , Interferon Type I/genetics , Recombinant Fusion Proteins/genetics , Swine
8.
Microb Pathog ; 127: 79-84, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30500407

ABSTRACT

Recently, a novel type I interferon alphaomega (IFN-αω), also known as IFN-µ, was identified. However, the biological activity of IFN-αω remain poorly understood. In this study, the porcine IFN-αω (PoIFN-αω) was expressed, purified, and its antiviral activities assessed by its ability to inhibit the cytopathic effect caused by FMDV on IBRS-2 cells. In addition, q-PCR was used to evaluate the expression of IFN-stimulated genes induced by PoIFN-αω. It was found that PoIFN-αω exerted effective antiviral activity against FMDV pre- and post-infection. Additionally, PoIFN-αω induced the transcription of IFN-stimulated genes, including Mx1, ISG15, OAS1, and PKR genes. Our study reported a new indication of PoIFN-αω as an effective anti-FMDV agent for the first time.


Subject(s)
Antiviral Agents/pharmacology , Foot-and-Mouth Disease Virus/drug effects , Interferon Type I/pharmacology , Recombinant Proteins/pharmacology , Animals , Antiviral Agents/isolation & purification , Antiviral Agents/metabolism , Cell Line , Cytopathogenic Effect, Viral , Gene Expression Profiling , Immunologic Factors/biosynthesis , Interferon Type I/genetics , Interferon Type I/isolation & purification , Interferon Type I/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Swine
9.
Cell Physiol Biochem ; 51(5): 2377-2396, 2018.
Article in English | MEDLINE | ID: mdl-30537741

ABSTRACT

The interferons (IFNs) are a primary defense against pathogens because of the strong antiviral activities they induce. IFNs can be classified into three groups: type I, type II and type III, according to their genetic, structural, and functional characteristics and their receptors on the cell surface. The type I IFNs are the largest group and include IFN-α, IFN-ß, IFN-ε, IFN-ω, IFN-κ, IFN-δ, IFN-τ and IFN-ζ. The use of IFNs for the treatment of viral infectious diseases on their antiviral activity may become an important therapeutic option, for example, IFN-α is well known for the successful treatment of hepatitis B and C virus infections, and interest is increasing in the antiviral efficacy of other novel IFN classes and their potential applications. Therefore, in this review, we summarize the recent progress in the study of the biological activities of all the type I IFN classes and their potential applications in the treatment of infections with immunodeficiency virus, hepatitis viruses, and influenza viruses.


Subject(s)
Antiviral Agents/therapeutic use , HIV Infections/drug therapy , Hepatitis, Viral, Human/drug therapy , Influenza, Human/drug therapy , Interferon Type I/therapeutic use , Animals , Antiviral Agents/pharmacology , HIV/drug effects , Hepatitis Viruses/drug effects , Humans , Interferon Type I/pharmacology , Orthomyxoviridae/drug effects , Orthomyxoviridae Infections/drug therapy
10.
Int Immunopharmacol ; 59: 47-52, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29621736

ABSTRACT

Foot-and-mouth disease (FMD) is one of the most devastating diseases affecting livestock. Since vaccines fail to provide protection until seven days post-vaccination, the application of anti-viral molecules is imperative for suppressing the spread of FMDV prior to development of an adaptive immune response. Interferons (IFNs) are effective for the host to fight FMDV infections; however, a novel type I IFNs, interferon delta (IFN-δ), has not been investigated for their antiviral effects against this virus. Thus, this study investigated FMDV infection, upon pre- and post-treatment with PoIFN-δ8. Real-time quantitative PCR was used to quantify the expression levels of IFN-stimulated genes (ISGs), including ISG15, OAS1, PKR, and Mx1. Results showed the PoIFN-δ8 lacking its signal sequence was efficiently expressed in Escherichia coli, and the purified recombinant PoIFN-δ8 exerted a significantly protective effect against two different serotypes of FMDV in IBRS-2 cells. In addition, PoIFN-δ8 induced the expression of IFN-stimulated genes. These findings highlight the significance of PoIFN-δ might serve as an antiviral agent for the prevention of FMDV in pigs and will stimulate the study of exploiting the potential biological functions of IFN-δ in the future.


Subject(s)
Antiviral Agents/pharmacology , Foot-and-Mouth Disease Virus/drug effects , Interferon Type I/pharmacology , Animals , Cell Line , Swine
11.
Int Immunopharmacol ; 52: 253-260, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28957693

ABSTRACT

Since 1985, interferon (IFN)-ω, a type I IFN, has been identified in many animals, but not canines and mice. It has been demonstrated to have antiviral, anti-proliferation, and antitumor activities that are similar to those of IFN-α. To date, IFN-ω has been explored as a treatment option for some diseases or viral infections in humans and other animals. Studies have revealed that human IFN-ω displays antitumor activities in some models of human cancer cells and that it can be used to diagnose some diseases. While recombinant feline IFN-ω has been licensed in several countries for treating canine parvovirus, feline leukemia virus, and feline immunodeficiency virus infections, it also exhibits a certain efficacy when used to treat other viral infections or diseases. This review examines the known biological activity of IFN-ω and its clinical applications. We expect that the information provided in this review will stimulate further studies of IFN-ω as a therapeutic agent.


Subject(s)
Antiviral Agents/therapeutic use , Immunotherapy/methods , Interferon Type I/metabolism , Virus Diseases/therapy , Animals , Cats , Dogs , Humans , Interferon Type I/immunology , Interferon Type I/therapeutic use , Mice , Recombinant Proteins/therapeutic use , Virus Diseases/immunology
12.
Acta Trop ; 173: 69-75, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28495401

ABSTRACT

Foot-and-mouth disease (FMD) is a significant zoonotic infectious disease. It has an important economic impact throughout the world. As well, it is a considerable threat to food security. At present, the molecular mechanism of FMDV infection is not clear to a large extent. Innate immune response is the first line of defense against infectious diseases. The systematic analysis of the host immune response to infection has an important role in understanding the pathogenesis of infection. However, there are few reports about effect of immune regulation on virus replication in the interaction of virus and host cellular. High-throughput RNA-seq technology as a powerful and efficient means for transcript analysis provides a new insight into FMDV study. In this study, RNA extracted from pig PBMCs infected with O subtype FMDV at 4 dpi. A total of 29942658 and 31452917 Illumina read pairs were obtained from the non-infected (NI) group and infected (I) group, respectively. The clean bases for all samples are 3.61G (NI group) and 3.79G (I group), respectively. The clean reads of the NI and I group that mapped to pig genome data were 47195073 (81.82%) and 46556714 (76.85%), respectively. Most of the clean reads were distributed in the exon region, followed by intron region and intergenic region. Differently expressed (DE) genes were analyzed using edgeR software. 451 genes were differentially expressed between the infected and the non-infected groups. According to the comparison analysis, more genes were down-regulated in the non-infected samples than in those infected with FMDV.66 out of 451 genes were down-regulated, 385 out of 451 genes were up-regulated following FMDV infection. For function classification and pathway analysis, among 17741 assembled unigenes, there are 349 genes which are different genes of GO notes. Moreover, 49 genes were down-regulated, 300 genes were up-regulated associate with GO term. 1621 were successfully annotated by GO assignments, belonging to one or more of the three categories: biological process, cellular component, and molecular function. According to KEGG analysis,the main pathway was represented including protein processing in endoplasmic reticulum, phagosome, cell cycle and cytokine-cytokine receptor interaction. Some key DE genes related to immune process and signaling pathways were analyzed and quantified by RT-PCR. This is the first systematical transcriptome analysis of pig PBMCs infected by FMDV. These findings will help us better understand the host Cell-FMDV interaction and its relationship to pathogenesis, as well as contribute to the prevention and control of FMDV.


Subject(s)
Foot-and-Mouth Disease Virus/immunology , Gene Expression Profiling , Leukocytes, Mononuclear/physiology , Swine , Transcriptome , Animals , Cells, Cultured , Gene Expression Regulation/immunology , Immunity, Innate , RNA , Sequence Analysis, RNA , Virus Replication
13.
J Med Virol ; 89(11): 2041-2046, 2017 11.
Article in English | MEDLINE | ID: mdl-28390158

ABSTRACT

Foot-and-mouth disease virus (FMDV) causes an economically important and highly contagious disease of cloven-hoofed animals such as cattle, swine, and sheep. FMD vaccine is the traditional way to protect against the disease, which can greatly reduce its occurrence. However, the use of FMD vaccines to protect early infection is limited. Therefore, the alternative strategy of applying antiviral agents is required to control the spread of FMDV in outbreak situations. As previously reported, LiCl has obviously inhibition effects on a variety of viruses such as transmissible gastroenteritis virus (TGEV), infectious bronchitis coronavirus (IBV), and pseudorabies herpesvirus and EV-A71 virus. In this study, our findings were the first to demonstrate that LiCl inhibition of the FMDV replication. In this study, BHK-21 cell was dose-dependent with LiCl at various stages of FMDV. Virus titration assay was calculated by the 50% tissue culture infected dose (TCID50 ) with the Reed and Muench method. The cytotoxicity assay of LiCl was performed by the CCK8 kit. The expression level of viral mRNA was measured by RT-qPCR. The results revealed LiCl can inhibit FMDV replication, but it cannot affect FMDV attachment stage and entry stage in the course of FMDV life cycle. Further studies confirmed that the LiCl affect the replication stage of FMDV, especially the early stages of FMDV replication. So LiCl has potential as an effective anti-FMDV drug. Therefore, LiCl may be an effective drug for the control of FMDV. Based on that, the mechanism of the antiviral effect of LiCl on FMDV infection is need to in-depth research in vivo.


Subject(s)
Antiviral Agents/pharmacology , Foot-and-Mouth Disease Virus/drug effects , Lithium Chloride/pharmacology , Virus Replication/drug effects , Animals , Cattle , Cell Line , DNA Replication/drug effects , Foot-and-Mouth Disease/drug therapy , Foot-and-Mouth Disease/virology , Foot-and-Mouth Disease Virus/genetics , Foot-and-Mouth Disease Virus/physiology , Polymerase Chain Reaction , RNA, Viral/genetics , Sheep , Swine , Time Factors
14.
Biomed Res Int ; 2016: 7849203, 2016.
Article in English | MEDLINE | ID: mdl-27478836

ABSTRACT

Foot-and-mouth disease (FMD) is a highly contagious livestock disease of cloven-hoofed animals which causes severe economic losses. The replication-deficient, human adenovirus-vectored FMD vaccine has been proven effective against FMD. However, the role of T-cell-mediated antiviral responses and the mucosae-mediated antiviral responses induced by the adenovirus-vectored FMD vaccine was rarely examined. Here, the capsid protein precursor P1-2A and viral protease 3C of the type O FMDV were expressed in replicative-deficient human adenovirus type 5 vector. BALB/c mice immunized intramuscularly and intraperitoneally with recombinant adenovirus rAdv-P12A3C elicited higher FMDV-specific IgG antibodies, IFN-γ, and IL-4 cytokines than those in mice immunized with inactivated FMDV vaccine. Moreover, BALB/c mice immunized with recombinant adenovirus rAdv-P12A3C by oral and intraocular-nasal immunization induced high FMDV-specific IgA antibodies. These results show that the recombinant adenovirus rAdv-P12A3C could resist FMDV comprehensively. This study highlights the potential of rAdv-P12A3C to serve as a type O FMDV vaccine.


Subject(s)
Adenoviridae/genetics , Antibodies, Viral/immunology , Capsid Proteins/immunology , Cysteine Endopeptidases/immunology , Foot-and-Mouth Disease Virus/genetics , Recombinant Proteins/immunology , Viral Proteins/immunology , 3C Viral Proteases , Animals , Antibodies, Viral/blood , Capsid Proteins/genetics , Capsid Proteins/metabolism , Cell Proliferation/drug effects , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Female , HEK293 Cells , Humans , Immunoglobulin A/analysis , Immunoglobulin A/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Mice , Mice, Inbred BALB C , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , T-Lymphocytes , Viral Proteins/genetics , Viral Proteins/metabolism , Viral Vaccines/immunology
15.
Vet Microbiol ; 183: 92-6, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26790940

ABSTRACT

Foot-and-mouth disease is a highly contagious, acute viral disease of cloven-hoofed animal species causing severe economic losses worldwide. Among the seven serotypes of foot-and-mouth disease virus (FMDV), serotype O is predominant, but its viral capsid is more acid sensitive than other serotypes, making it more difficult to produce empty serotype O VLPs in the low pH insect hemolymph. Therefore, a novel chimeric virus-like particle (VLP)-based candidate vaccine for serotype O FMDV was developed and characterized in the present study. The chimeric VLPs were composed of antigenic VP1 from serotype O and segments of viral capsid proteins from serotype Asia1. These VLPs elicited significantly higher FMDV-specific antibody levels in immunized mice than did the inactivated vaccine. Furthermore, the chimeric VLPs protected guinea pigs from FMDV challenge with an efficacy similar to that of the inactivated vaccine. These results suggest that chimeric VLPs have the potential for use in vaccines against serotype O FMDV infection.


Subject(s)
Capsid Proteins/immunology , Foot-and-Mouth Disease Virus/immunology , Foot-and-Mouth Disease/prevention & control , Foot-and-Mouth Disease/virology , Vaccines, Virus-Like Particle/immunology , Animals , Antigens, Viral/immunology , Capsid Proteins/genetics , Foot-and-Mouth Disease Virus/genetics , Foot-and-Mouth Disease Virus/ultrastructure , Gene Expression Regulation, Viral , Guinea Pigs , Mice , Microscopy, Electron, Transmission , Random Allocation , Sf9 Cells
SELECTION OF CITATIONS
SEARCH DETAIL