Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Virol ; 98(2): e0200223, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38289108

ABSTRACT

Foot-and-mouth disease virus (FMDV) remains a challenge for cloven-hooved animals. The currently licensed FMDV vaccines induce neutralizing antibody (NAb)-mediated protection but show defects in the early protection. Dendritic cell (DC) vaccines have shown great potency in inducing rapid T-cell immunity in humans and mice. Whether DC vaccination could enhance early protection against FMDV has not been elaborately explored in domestic pigs. In this study, we employed DC vaccination as an experimental approach to study the roles of cellular immunity in the early protection against FMDV in pigs. Autologous DCs were differentiated from the periphery blood mononuclear cells of each pig, pulsed with inactivated FMDV (iFMDV-DC) and treated with LPS, and then injected into the original pigs. The cellular immune responses and protective efficacy elicited by the iFMDV-DC were examined by multicolor flow cytometry and tested by FMDV challenge. The results showed that autologous iFMDV-DC immunization induced predominantly FMDV-specific IFN-γ-producing CD4+ T cells and cytotoxic CD8+ T cells (CTLs), high NAb titers, compared to the inactivated FMDV vaccine, and accelerated the development of memory CD4 and CD8 T cells, which was concomitantly associated with early protection against FMDV virulent strain in pigs. Such early protection was associated with the rapid proliferation of secondary T-cell response after challenge and significantly contributed by secondary CD8 effector memory T cells. These results demonstrated that rapid induction of cellular immunity through DC immunization is important for improving early protection against FMDV. Enhancing cytotoxic CD8+ T cells may facilitate the development of more effective FMDV vaccines.IMPORTANCEAlthough the currently licensed FMDV vaccines provide NAb-mediated protection, they have defects in early immune protection, especially in pigs. In this study, we demonstrated that autologous swine DC immunization augmented the cellular immune response and induced an early protective response against FMDV in pigs. This approach induced predominantly FMDV-specific IFN-γ-producing CD4+ T cells and cytotoxic CD8+ T cells, high NAb titers, and rapid development of memory CD4 and CD8 T cells. Importantly, the early protection conferred by this DC immunization is more associated with secondary CD8+ T response rather than NAbs. Our findings highlighted the importance of enhancing cytotoxic CD8+ T cells in early protection to FMDV in addition to Th1 response and identifying a strategy or adjuvant comparable to the DC vaccine might be a future direction for improving the current FMDV vaccines.


Subject(s)
Foot-and-Mouth Disease Virus , Foot-and-Mouth Disease , Viral Vaccines , Animals , Humans , Mice , Antibodies, Neutralizing , Antibodies, Viral , CD8-Positive T-Lymphocytes , Foot-and-Mouth Disease/immunology , Foot-and-Mouth Disease/prevention & control , Foot-and-Mouth Disease Virus/physiology , Swine , Vaccination
2.
mBio ; 13(5): e0143422, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36106732

ABSTRACT

The maintenance of viral protein homeostasis depends on the machinery of the infected host cells, giving us an insight into the interplay between host and virus. Accumulating evidence suggests that heat shock protein 60 (HSP60), as one molecular chaperone, is involved in regulating virus infection. However, the role of HSP60 during foot-and-mouth disease virus (FMDV) replication and its specific mechanisms have not been reported. We demonstrate that HSP60 modulates the FMDV life cycle. HSP60 plays a role at the postentry stage of the viral life cycle, including RNA replication and mRNA translation; however, HSP60 does not affect viral replication of Seneca Valley virus (SVA) or encephalomyocarditis virus (EMCV). We found that HSP60 is involved in FMDV replication complex (RC) formation. Furthermore, our results indicate that HSP60 interacts with FMDV nonstructural proteins 3A and 2C, key elements of the viral replication complex. We also show that HSP60 regulates the stability of 3A and 2C via caspase-dependent and autophagy-lysosome-dependent degradation, thereby promoting FMDV RNA synthesis and mRNA translation mediated by the RC. Additionally, we determined that the apical domain of HSP60 is responsible for interacting with 3A and 2C. The N terminus of 3A and ATPase domain of 2C are involved in binding to HSP60. Importantly, HSP60 depletion potently reduced FMDV pathogenicity in infected mice. Altogether, this study demonstrates a specific role of HSP60 in promoting FMDV replication. Furthermore, targeting host HSP60 will help us design the FMDV-specific antiviral drugs. IMPORTANCE FMDV is the leading cause of the foot-and-mouth disease (FMD), affecting cloven-hoofed animals with high morbidity and mortality. We determined that HSP60 is required for efficient viral RNA replication and mRNA translation during FMDV infection. Furthermore, we demonstrate that HSP60 interacts with FMDV nonstructural proteins 3A and 2C, the elements of the RC; HSP60 contributes to the stability of 3A and 2C to affect the formation and function of the RC. We also validated the potential role of HSP60 as the antiviral target in vivo using small interfering RNA. These findings deepen the understanding of the host-virus interaction and provide information supporting the design of novel therapeutics for FMDV infection.


Subject(s)
Foot-and-Mouth Disease Virus , Foot-and-Mouth Disease , Animals , Mice , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Chaperonin 60/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , RNA, Small Interfering/metabolism , Cell Line , Foot-and-Mouth Disease Virus/genetics , Virus Replication/physiology , Antiviral Agents/metabolism , Caspases/metabolism
3.
J Virol Methods ; 300: 114406, 2022 02.
Article in English | MEDLINE | ID: mdl-34915087

ABSTRACT

Foot-and-mouth disease (FMD) is the highly contagious disease of cloven-hoofed animal that brings considerable economic losses to the animal husbandry. So FMD surveillance which relying on accurate diagnosis is important. Most producing the diagnostic antigen of inactivated FMD virus (FMDV) requires facilities with high biosafety. In our previous studies, virus-like particles(VLPs) resembled the structures of natural virus particles. Here, we established a competitive ELISA (cELISA) method for the detection of antibodies against serotype A FMDV based on serotype A FMDV-VLPs. Via detecting different positive serum and negative serum with different titers, and comparing with different commercial ELISA kits. The specificity and sensitivity of the assay were 100 % and 98 %, respectively. The coincidence rate using the PrioCHECK® FMDV Type A antibody ELISA kit and Liquid-phase blocking (LPB) ELISA were 95.30 % and 92.2 %. Repetitive experiments showed that variation coefficient of intra-batch and inter-batch were less than 9 % and 13 %. The result demonstrated that cELISA based on VLPs from prokaryotic system is highly specific, sensitive and reproducible. The cELISA could also be used to assess the immune responses of serotype A FMDV, especially in developing countries.


Subject(s)
Foot-and-Mouth Disease Virus , Foot-and-Mouth Disease , Animals , Antibodies, Viral , Enzyme-Linked Immunosorbent Assay/methods , Foot-and-Mouth Disease/diagnosis , Serogroup
5.
J Virol ; 95(16): e0017721, 2021 07 26.
Article in English | MEDLINE | ID: mdl-34011545

ABSTRACT

Foot-and-mouth disease (FMD) is a highly contagious viral disease affecting cloven-hoofed animals that causes a significant economic burden globally. Vaccination is the most effective FMD control strategy. However, FMD virus (FMDV) particles are prone to dissociate when appropriate physical or chemical conditions are unavailable, such as an incomplete cold chain. Such degraded vaccines result in compromised herd vaccination. Therefore, thermostable FMD particles are needed for use in vaccines. This study generated thermostable FMDV mutants (M3 and M10) by serial passages at high temperature, subsequent amplification, and purification. Both mutants contained an alanine-to-threonine mutation at position 13 in VP1 (A1013T), although M3 contained 3 additional mutations. The selected mutants showed improved stability and immunogenicity in neutralizing antibody titers, compared with the wild-type (wt) virus. The sequencing analysis and cryo-electron microscopy showed that the mutation of alanine to threonine at the 13th amino acid in the VP1 protein (A1013T) is critical for the capsid stability of FMDV. Virus-like particles containing A1013T (VLPA1013T) also showed significantly improved stability to heat treatment. This study demonstrated that Thr at the 13th amino acid of VP1 could stabilize the capsid of FMDV. Our findings will facilitate the development of a stable vaccine against FMDV serotype O. IMPORTANCE Foot-and-mouth disease (FMD) serotype O is one of the global epidemic serotypes and causes significant economic loss. Vaccination plays a key role in the prevention and control of FMD. However, the success of vaccination mainly depends on the quality of the vaccine. Here, the thermostable FMD virus (FMDV) mutants (M3 and M10) were selected through thermal screening at high temperatures with improved stability and immunogenicity compared with the wild-type virus. The results of multisequence alignment and cryo-electron microscopy (cryo-EM) analysis showed that the Thr substitution at the 13th amino acid in the VP1 protein is critical for the capsid stability of FMDV. For thermolabile type O FMDV, this major discovery will aid the development of its thermostable vaccine.


Subject(s)
Capsid Proteins/immunology , Capsid/immunology , Foot-and-Mouth Disease Virus/immunology , Viral Vaccines/immunology , Amino Acid Substitution , Animals , Capsid/chemistry , Capsid Proteins/chemistry , Capsid Proteins/genetics , Cryoelectron Microscopy , Foot-and-Mouth Disease/prevention & control , Foot-and-Mouth Disease Virus/genetics , Foot-and-Mouth Disease Virus/metabolism , Guinea Pigs , Hot Temperature , Immunogenicity, Vaccine , Mutation , Protein Stability , Serogroup , Vaccines, Virus-Like Particle/administration & dosage , Vaccines, Virus-Like Particle/genetics , Vaccines, Virus-Like Particle/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Virology
6.
Res Vet Sci ; 136: 89-96, 2021 May.
Article in English | MEDLINE | ID: mdl-33592449

ABSTRACT

Although the immunogenicity of DNA vaccines is nonideal, they are still considered as potential alternative vaccine candidates to conventional vaccines. Various DNA delivery systems, including nanoparticles, have been extensively explored and validated to further enhance the immunogenicity of DNA vaccines. DNA vaccines are considered as alternative vaccine candidates. Various DNA delivery systems, including nanoparticles, have been extensively explored to enhance the immunogenicity of DNA vaccines. In this study, positively charged Poly (D, l-lactide-co-glycolic acid) (PLGA) nanoparticles were generated and characterized as a delivery system for O-serotype foot-and-mouth DNA vaccine. A recombinant plasmid encoding swine interleukin (IL)-18, IL-2, or granulocyte-macrophage colony-stimulating factor (GM-CSF) gene was introduced into the DNA vaccine to further improve its immunogenicity, which was evaluated in a guinea pig model. PLGA-pVAX-VP013/IL-18 elicited significantly (P = 0.0149) higher FMDV-specific antibody levels than naked DNA before the challenge. The level of neutralizing antibodies induced by PLGA-pVAX-VP013/IL-18, PLGA-pVAX-VP013/IL-2, and PLGA-pVAX-VP013/GM-CSF significantly increased compared with that induced by naked DNA (P < 0.0001). The lymphocyte proliferation assay showed that cellular immunity induced by PLGA-pVAX-VP013/IL-18 and PLGA-pVAX-VP013/GM-CSF was dramatically enhanced compared with that induced by the inactivated vaccine. The protection by PLGA-pVAX-VP013/IL-18 was consistent with that by the inactivated vaccine post-challenge and was followed by PLGA-pVAX-VP013/GM-CSF. Therefore, cationic PLGA nanoparticles can deliver DNA vaccines and induce humoral and cellular immune responses. The co-administration of FMD DNA vaccine with IL-18 formulated with PLGA nanoparticles was the optimal strategy to improve the immunogenicity of FMD DNA vaccines.


Subject(s)
Foot-and-Mouth Disease Virus/immunology , Immunogenicity, Vaccine , Nanoparticles , Polylactic Acid-Polyglycolic Acid Copolymer/immunology , Vaccines, DNA/immunology , Viral Vaccines/immunology , Animals , Foot-and-Mouth Disease/immunology , Foot-and-Mouth Disease/prevention & control , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Guinea Pigs , Interleukin-18/immunology , Interleukin-2/immunology , Nanoparticles/administration & dosage , Polylactic Acid-Polyglycolic Acid Copolymer/administration & dosage , Serogroup
7.
Nanomedicine ; 33: 102358, 2021 04.
Article in English | MEDLINE | ID: mdl-33484882

ABSTRACT

Virus-like particle (VLP) vaccines have become one of the dominant vaccine candidates for foot-and-mouth disease (FMD). To further enhance the immunogenicity of VLP vaccines, gold nanocages (AuNCs) were selected as an adjuvant for the vaccine. Our experiments demonstrated that AuNCs had little biotoxicity in vivo and in vitro and improved the uptake of VLP in BHK-21 and RAW264.7 cell lines. The VLP-AuNCs activated DCs mainly through toll-like receptor 4 (TLR4) and promoted the secretion of IL-6, IL-1ß, and TNF-α. The conjugation of VLP and AuNCs triggered a strong immune response against FMD virus (FMDV) in mice and guinea pigs. The VLP-AuNCs significantly enhanced the proliferation of CD8+ T cells (P < 0.05) and the secretion of cellular immune-related cytokines (IFN-γ, P < 0.05; IL-12p70, P < 0.01) compared with VLP. The present study demonstrated that AuNCs, as a great potential adjuvant for FMDV VLP vaccines, significantly enhance the immune response.


Subject(s)
Adjuvants, Immunologic/chemistry , Drug Carriers/chemistry , Foot-and-Mouth Disease/prevention & control , Gold/chemistry , Metal Nanoparticles/chemistry , Vaccines, Virus-Like Particle/chemistry , Viral Vaccines/chemistry , Adjuvants, Immunologic/pharmacology , Animals , Biomedical Enhancement , CD8-Positive T-Lymphocytes , Cell Membrane Permeability , Cell Proliferation , Cytokines/metabolism , Drug Compounding , Drug Liberation , Female , Foot-and-Mouth Disease Virus , Guinea Pigs , Mice , Mice, Inbred BALB C , Neutralization Tests , RAW 264.7 Cells , Vaccines, Virus-Like Particle/pharmacology , Viral Vaccines/pharmacology
8.
Vaccines (Basel) ; 8(3)2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32942760

ABSTRACT

Senecavirus A (SVA) is the pathogen that has recently caused porcine idiopathic vesicular disease (PIVD). The clinical symptoms of PIVD are similar to those of acute foot-and-mouth disease and also can result in the death of newborn piglets, thus entailing economic losses. Vaccine immunization is the most effective way to prevent and control SVA. Among all SVA vaccines reported, only the SVA inactivated vaccine has been successfully developed. However, to ensure the elimination of this pathogen, safer and more effective vaccines are urgently required. A virus-like particles (VLPs)-based vaccine is probably the best alternative to inactivated vaccine. To develop an SVA VLPs vaccine and evaluate its immune effect, a prokaryotic expression system was used to produce SVA capsid protein and assemble VLPs. The VLPs were characterized by affinity chromatography, sucrose density gradient centrifugation, ZetaSizer and transmission electron microscopy. Meanwhile, the SVA CH-HB-2017 strain was used to infect pigs and to determine infection routes and dose. Experimental pigs were then immunized with the SVA VLPs vaccine emulsified in an ISA 201 adjuvant. The results showed that the VLPs vaccine induced neutralizing and specific antibodies at similar levels as an inactivated SVA vaccine after immunization. The level of INF-γ induced by the VLPs vaccine gradually decreased-similar to that of inactivated vaccine. These results indicated that VLPs vaccine may simultaneously cause both cellular and humoral immune responses. Importantly, after the challenge, the VLPs vaccine provided similar levels of protection as the inactivated SVA vaccine. In this study, we successfully obtained novel SVA VLPs and confirmed their highly immunogenicity, thus providing a superior candidate vaccine for defense and elimination of SVA, compared to the inactivated vaccine.

9.
Mol Cell Probes ; 53: 101643, 2020 10.
Article in English | MEDLINE | ID: mdl-32768439

ABSTRACT

Porcine vesicular disease caused by Senecavirus A (SVA) is a newly emerging disease in many countries. Based on clinical signs only, it is very challenging to distinguish SVA infection from other similar diseases, such as foot and mouth disease, swine vesicular disease, and vesicular stomatitis. Therefore, it is crucial to establish a detection assay for the clinical diagnosis of SVA infection. In this study, a pair of specific primers were designed based on the highly conserved L/VP4 gene sequence of SVA. The established SYBR green I-based quantitative reverse transcription polymerase chain reaction (qRT-PCR) method was used to detect SVA nucleic acids in clinical samples. The limit of detection SVA nucleic acids by qRT-PCR was 6.4 × 101 copies/µL, which was significantly more sensitive than that by gel electrophoresis of 6.4 × 103 copes/µL. This assay was specific and had no cross-reaction with other seven swine viruses. Using SYBR green I-based qRT-PCR, the SVA positive rates in experimental animal samples and field samples were 67.60% (96/142) and 80% (24/30) respectively. The results demonstrate that SYBR green I-based qRT-PCR is a rapid and specific method for the clinical diagnosis and epidemiological investigation of related vesicular diseases caused by SVA.


Subject(s)
Benzothiazoles/chemistry , Capsid Proteins/genetics , Diamines/chemistry , Picornaviridae/isolation & purification , Quinolines/chemistry , Swine Vesicular Disease/diagnosis , Animals , Limit of Detection , Picornaviridae/genetics , Picornaviridae Infections/diagnosis , Picornaviridae Infections/veterinary , Reverse Transcriptase Polymerase Chain Reaction , Swine , Swine Diseases/virology , Swine Vesicular Disease/virology
10.
Vaccine ; 36(45): 6752-6760, 2018 10 29.
Article in English | MEDLINE | ID: mdl-30268733

ABSTRACT

Virus-like particles (VLPs) have become a hot topic in modern vaccine research because of its safety, facile production, and immune properties. To further enhance the immune effect of VLPs, we synthesized and used gold-star nanoparticles (AuSNs) as adjuvant for vaccine. Foot-and-mouth disease (FMD) VLPs as target antigen were combined with AuSNs. The FMD VLPs-AuSNs complex was characterized through sodium dodecyl sulfate-polyacrylamide gel electrophoresis, Western blot, ultraviolet light absorption, and morphological measurement analyses. Result indicated that the FMD VLPs-AuSNs complex is non-toxic in different cell lines. AuSNs can effectively promote the entry of FMD VLPs into cells and improve macrophages activation when combined with FMD VLPs compared with FMD VLPs alone. Further animal vaccination and challenge tests revealed that the specific immune response and protection rate of AuSNs adjuvant group is higher than that of conventional mineral oil (ISA206) adjuvant group. AuSNs can effectively improve the immune protection effects of FMD VLPs vaccines, and exhibit potential as a new adjuvant for other vaccines.


Subject(s)
Foot-and-Mouth Disease Virus/immunology , Foot-and-Mouth Disease/immunology , Foot-and-Mouth Disease/prevention & control , Vaccines, Virus-Like Particle/immunology , Adjuvants, Immunologic/therapeutic use , Animals , Antibodies, Neutralizing/metabolism , Female , Foot-and-Mouth Disease Virus/pathogenicity , Guinea Pigs , Mice , Neutralization Tests , RAW 264.7 Cells , T-Lymphocytes/metabolism , Vaccination/methods , Vaccines, Virus-Like Particle/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL