Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Acta Biomater ; 183: 330-340, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38838909

ABSTRACT

Although vaccination with inactivated vaccines is a popular preventive method against pseudorabies virus (PRV) infection, inactivated vaccines have poor protection efficiency because of their weak immunogenicity. The development of an effective adjuvant is urgently needed to improve the efficacy of inactivated PRV vaccines. In this study, a promising nanocomposite adjuvant named as MIL@A-SW01-C was developed by combining polyacrylic acid-coated metal-organic framework MIL-53(Al) (MIL@A) and squalene (oil)-in-water emulsion (SW01) and then mixing it with a carbomer solution. One part of the MIL@A was loaded onto the oil/water interface of SW01 emulsion via hydrophobic interaction and coordination, while another part was dispersed in the continuous water phase using carbomer. MIL@A-SW01-C showed good biocompatibility, high PRV (antigen)-loading capability, and sustained antigen release. Furthermore, the MIL@A-SW01-C adjuvanted PRV vaccine induced high specific serum antibody titers, increased splenocyte proliferation and cytokine secretion, and a more balanced Th1/Th2 immune response compared with commercial adjuvants, such as alum and biphasic 201. In the mouse challenge experiment, two- and one-shot vaccinations resulted in survival rates of 73.3 % and 86.7 %, respectively. After one-shot vaccination, the host animal pigs were also challenged with wild PRV. A protection rate of 100 % was achieved, which was much higher than that observed with commercial adjuvants. This study not only establishes the superiority of MIL@A-SW01-C composite nanoadjuvant for inactivated PRV vaccine in mice and pigs but also presents an effective method for developing promising nanoadjuvants. STATEMENT OF SIGNIFICANCE: We have developed a nanocomposite of MIL-53(Al) and oil-in-water emulsion (MIL@A-SW01-C) as a promising adjuvant for the inactivated PRV vaccines. MIL@A-SW01-C has good biocompatibility, high PRV (antigen) loading capability, and prolonged antigen release. The developed nanoadjuvant induced much higher specific IgG antibody titers, increased splenocyte proliferation and cytokine secretion, and a more balanced Th1/Th2 immune response than commercial adjuvants alum and biphasic 201. In mouse challenge experiments, survival rates of 73.3 % and 86.7 % were achieved from two-shot and one-shot vaccinations, respectively. At the same time, a protection rate of 100 % was achieved with the host animal pigs challenged with wild PRV.


Subject(s)
Adjuvants, Immunologic , Emulsions , Animals , Adjuvants, Immunologic/pharmacology , Emulsions/chemistry , Mice , Swine , Herpesvirus 1, Suid/immunology , Pseudorabies Vaccines/immunology , Mice, Inbred BALB C , Oils/chemistry , Female , Water/chemistry , Vaccines, Inactivated/immunology , Pseudorabies/prevention & control , Pseudorabies/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Cytokines/metabolism
2.
Int J Biol Macromol ; 269(Pt 2): 132172, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38719009

ABSTRACT

Adjuvants including aluminum adjuvant (Alum) and oil-water emulsion have been widely used in inactivated pseudorabies virus (PRV) vaccines to improve their performance, however, they are not sufficient to protect from PRV infection because of the weak immune response and poor Th1-type immune response. Divalent manganese ion (Mn2+) has been reported to increase the cellular immune response significantly. In this work, a xanthan gum and carbomer-dispersed Mn2+-loaded tannic acid-polyethylene glycol (TPMnXC) nanoparticle colloid is developed and used as an adjuvant to improve the performance of the inactivated PRV vaccine. The good in vitro and in vivo biocompatibility of the developed TPMnXC colloid has been confirmed by the cell viability assay, erythrocyte hemolysis, blood routine analysis, and histological analysis of mouse organs and injection site. The TPMnXC-adjuvanted inactivated PRV vaccine (TPMnXC@PRV) significantly promotes higher and more balanced immune responses indicating with an increased specific total IgG antibody and IgG2a/IgG1 ratio, efficient splenocytes proliferation, and elevated Th1- and Th2-type cytokine secretion than those of control groups. Wild PRV challenge experiment is performed using mice as a model animal, achieving a protection rate of up to 86.67 %, which is much higher than those observed from the commercial Alum. This work not only demonstrates the high potentiality of TPMnXC in practical applications but also provides a new way to develop the Mn2+-loaded nanoadjuvant for veterinary vaccines.


Subject(s)
Adjuvants, Immunologic , Herpesvirus 1, Suid , Immunity, Cellular , Immunity, Humoral , Manganese , Nanoparticles , Polysaccharides, Bacterial , Tannins , Animals , Mice , Adjuvants, Immunologic/pharmacology , Nanoparticles/chemistry , Immunity, Cellular/drug effects , Immunity, Humoral/drug effects , Tannins/chemistry , Tannins/pharmacology , Manganese/chemistry , Polysaccharides, Bacterial/chemistry , Polysaccharides, Bacterial/pharmacology , Polysaccharides, Bacterial/immunology , Herpesvirus 1, Suid/immunology , Pseudorabies Vaccines/immunology , Vaccines, Inactivated/immunology , Pseudorabies/prevention & control , Pseudorabies/immunology , Female , Cytokines/metabolism , Mice, Inbred BALB C , Antibodies, Viral/blood , Antibodies, Viral/immunology , Polyphenols
3.
Viruses ; 16(5)2024 04 29.
Article in English | MEDLINE | ID: mdl-38793591

ABSTRACT

In recent years, pseudorabies virus (PRV) variants have resulted in an epidemic in swine herds and huge economic losses in China. Therefore, it is essential to develop an efficacious vaccine against the spread of PRV variants. Here, the triple-gene-deletion virus and the triple-gene-deletion plus gC virus were constructed by homologous recombination (HR). And then, their growth capacity, proliferation ability, and immune efficacy were evaluated. The results showed that the growth kinetics of the recombinant viruses were similar to those of the parental strain PRV-AH. Compared with the triple-gene-deletion virus group, the more dominant level of neutralizing antibody (NA) can be induced in the triple-gene-deletion plus gC virus group with the same 106.0 TCID50 dose after 4 and 6 weeks post-initial immunization (PII) (p < 0.0001). In addition, the antibody titers in mice immunized with the triple-gene-deletion plus gC virus were significantly higher than those immunized with triple-gene deletion virus with the same 105.0 TCID50 dose after 6 weeks PII (p < 0.001). More importantly, in the triple-gene-deletion plus gC virus group with 105.0 TCID50, the level of NA was close to that in the triple-gene deletion virus group with 106.0 TCID50 at 6 weeks PII. Meanwhile, the cytokines IL-4 and IFN-γ in sera were tested by enzyme-linked immunosorbent assay (ELISA) in each group. The highest level of IL-4 or IFN-γ was also elicited in the triple-gene deletion plus gC virus group at a dose of 106.0 TCID50. After challenge with PRV-AH, the survival rates of the triple-gene deletion plus gC virus immunized groups were higher than those of other groups. In immunized groups with 105.0 TCID50, the survival rate shows a significant difference between the triple-gene deletion plus gC virus group (75%, 6/8) and the triple-gene deletion virus group (12.5%, 1/8). In general, the immune efficacy of the PRV TK/gI/gE-deleted virus can be increased with additional gC insertion in mice, which has potential for developing an attenuated vaccine candidate for PRV control.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Gene Deletion , Herpesvirus 1, Suid , Pseudorabies Vaccines , Pseudorabies , Animals , Herpesvirus 1, Suid/genetics , Herpesvirus 1, Suid/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Mice , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Pseudorabies/prevention & control , Pseudorabies/immunology , Pseudorabies/virology , Pseudorabies Vaccines/immunology , Pseudorabies Vaccines/genetics , Pseudorabies Vaccines/administration & dosage , Mice, Inbred BALB C , Swine , Female , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Homologous Recombination , Cytokines/metabolism , China
4.
Mater Horiz ; 11(9): 2153-2168, 2024 05 07.
Article in English | MEDLINE | ID: mdl-38376908

ABSTRACT

Pseudorabies virus (PRV) is a highly contagious viral disease, which leads to severe financial losses in the breeding industry worldwide. Presently, PRV is mainly controlled using live attenuated and inactivated vaccines. However, these vaccines have an innate tendency to lose their structural conformation upon exposure to environmental and chemical stressors and cannot provide full protection against the emerging prevalent PRV variants. In this work, first, we synthesized aminated ZIF-7/8 nanoparticles (NPs), and then chemical bond-coated alginate dialdehyde (ADA, a type of dioxide alginate saccharide) on their surface via Schiff base reaction to obtain ZIF-7/8-ADA NPs. The as-fabricated ZIF-7/8-ADA NPs exhibited high stability, monodispersity and a high loading ratio of antigen. Furthermore, the ZIF-7/8-ADA NPs showed good biocompatibility in vitro and in vivo. Using ZIF-7/8-ADA NPs as an adjuvant and inactivated PRV as a model antigen, we constructed a PR vaccine through a simple mixture. The immunity studies indicated that ZIF-7/8-ADA induced an enhancement in the Th1/Th2 immune response, which was superior to that of the commercial ISA201, alum adjuvant and ZIF-7/8. Due to the pH-sensitive release of the antigen in lysosomes, the as-prepared PR vaccine subsequently accelerated the antigen presentation and improved the immune responses in vitro and in vivo. The results of PRV challenge using mice as the model demonstrated that ZIF-7/8-ADA achieved the same preventive effect as the commercial ISA201 and was much better than the alum adjuvant, and thus can serve as a promising delivery system and adjuvant to enhance humoral and cellular responses against PRV infection.


Subject(s)
Adjuvants, Immunologic , Alginates , Metal-Organic Frameworks , Nanoparticles , Animals , Alginates/chemistry , Alginates/pharmacology , Mice , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Pseudorabies/prevention & control , Pseudorabies/immunology , Herpesvirus 1, Suid/immunology , Pseudorabies Vaccines/immunology , Pseudorabies Vaccines/administration & dosage , Vaccines, Inactivated/immunology , Vaccines, Inactivated/administration & dosage , Drug Carriers/chemistry , Vaccination/methods , Mice, Inbred BALB C , Female
5.
Vet Microbiol ; 290: 110011, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38310713

ABSTRACT

Senecavirus A (SVA)-associated porcine idiopathic vesicular disease (PIVD) and Pseudorabies (PR) are highly contagious swine disease that pose a significant threat to the global pig industry. In the absence of an effective commercial vaccine, outbreaks caused by SVA have occurred in many parts of the world. In this study, the PRV variant strain PRV-XJ was used as the parental strain to construct a recombinant PRV strain with the TK/gE/gI proteins deletion and the VP3 protein co-expression, named rPRV-XJ-ΔTK/gE/gI-VP3. The results revealed that PRV is a suitable viral live vector for VP3 protein expressing. As a vaccine, rPRV-XJ-ΔTK/gE/gI-VP3 is safe for mice, vaccination with it did not cause any clinical symptoms of PRV. Intranasal immunization with rPRV-XJ-ΔTK/gE/gI-VP3 induced strong cellular immune response and high levels of specific antibody against VP3 and gB and neutralizing antibodies against both PRV and SVA in mice. It provided 100% protection to mice against the challenge of virulent strain PRV-XJ, and alleviated the pathological lesion of heart and liver tissue in SVA infected mice. rPRV-XJ-ΔTK/gE/gI-VP3 appears to be a promising vaccine candidate against PRV and SVA for the control of the PRV variant and SVA.


Subject(s)
Herpesvirus 1, Suid , Picornaviridae , Pseudorabies , Rodent Diseases , Swine Diseases , Viral Vaccines , Swine , Animals , Mice , Viral Envelope Proteins , Antibodies, Viral , Pseudorabies Vaccines
6.
Microb Pathog ; 187: 106513, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38147968

ABSTRACT

Since pseudorabies (PR) re-emerged and rapidly spread in China at the end of 2011, researchers have focused on effective vaccine strategies to prevent and control pseudorabies virus (PRV) infection in pig herds. Due to the extensive application of an attenuated vaccine based on the Bartha-K61 strain isolated in Hungary in 1961 and the variation of the PRV strain, it has been suggested that traditional vaccines based on the Bartha-K61 strain offer only partial protection against variant strains. It was therefore evaluated whether the Porcilis® Begonia vaccine, which is based on the NIA-3 strain with deletions in the gE and TK genes, is efficacious against experimental infection with the virulent, contemporary Chinese PRV strain ZJ01. In this study, piglets were vaccinated with Porcilis® Begonia through either the intradermal (ID) route or the intramuscular (IM) route and subsequently challenged intranasally with strain ZJ01 at 4 weeks post-vaccination. An unvaccinated challenge group and an unvaccinated/nonchallenged group were also included in the study. All animals were monitored for 14 days after challenge. Vaccinated and negative control pigs stayed healthy during the study, while the unvaccinated control animals developed lesions associated with PRV ZJ01 challenge, and 44% of these pigs died before the end of the experiment. This study demonstrated that ID or IM vaccination of pigs with a vaccine based on the NIA-3 strain Porcilis® Begonia clinically protects against fatal PRV challenge with the ZJ01 strain.


Subject(s)
Begoniaceae , Herpesvirus 1, Suid , Swine Diseases , Viral Vaccines , Swine , Animals , Herpesvirus 1, Suid/genetics , Pseudorabies Vaccines , Antibodies, Viral , Vaccination/veterinary , Viral Vaccines/genetics
7.
Chinese Journal of Biotechnology ; (12): 987-992, 2009.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-286612

ABSTRACT

We studied the immunogenicity of pseudorabies virus gC DNA vaccination by fusing the murine complement C3d receptor binding domain. First, pseudorabies virus gC gene was linked to four copies of C3d receptor binding domain (M284), and then cloned into the vector pcDNA3.1 to construct the recombinant plasmid sgC-M284. Through the experiment of immunized BALB/c mice, we found that the enzyme linked immunosorbent assay (ELISA) antibody titer for sgC-M284 was 17-fold higher than that for sgC alone, and protective rate of mice was augmented from 25% to 88% after lethal dose PrV (316 LD50) challenge. In addition, the IL-4 levels for sgC-M284 immunization approached that for the pseudorabies virus inactivated vaccine. In conclusion, we demonstrated murine C3d receptor binding domain fusion significantly increased Th2-biased immune response by inducing IL-4 production.


Subject(s)
Animals , Mice , Adjuvants, Immunologic , Physiology , Antibody Formation , Allergy and Immunology , Binding Sites , Cloning, Molecular , Complement C3d , Genetics , Allergy and Immunology , Herpesvirus 1, Suid , Genetics , Allergy and Immunology , Interleukin-4 , Allergy and Immunology , Mice, Inbred BALB C , Pseudorabies Vaccines , Allergy and Immunology , Receptors, Complement 3d , Genetics , Recombinant Proteins , Genetics , Allergy and Immunology , Swine , Vaccines, DNA , Allergy and Immunology , Viral Envelope Proteins , Pharmacology , Viral Fusion Proteins , Allergy and Immunology
8.
Chinese Journal of Biotechnology ; (12): 1149-1154, 2008.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-275411

ABSTRACT

We designed two pairs of primers and their corresponding TaqMan probes according to gH, gE gene of PRV. By optimizing the probe's concentration, Mg2+ concentration, primers concentration and sample DNA extraction, real-time fluorescent quantitative PCR (FQ-PCR) which can quickly identity field virus and vaccine virus of PRV was established. According to our results, the dynamic range of the FQ-PCR assay is between 10 x 10(1) copies/microL and 10 x l0(8) copies/microL, and the detection limit of FQ-PCR is 1.0 x 10(1) copies/microL, which is 100 fold higher than that of conventional PCR. We detected 60 doubtful tissue samples using the FQ-PCR assay, serum neutralization and conventional PCR. In conclusion, the FQ-PCR method is rapid, sensitive, specific and accurate, and can be used to detect field strains of PRV rapidly. The closed-tube format of the assay minimized the risk of contamination of subsequent reaction and the assay can be performed in 2 h or less. Development of real-time quantitative PCR provides the basis for the early and rapid detection and analyzing quantitatively the infectious degree of PRV.


Subject(s)
Animals , Fluorescent Dyes , Herpesvirus 1, Suid , Genetics , Polymerase Chain Reaction , Methods , Pseudorabies , Diagnosis , Virology , Pseudorabies Vaccines , Allergy and Immunology , Swine
9.
Article in English | WPRIM (Western Pacific) | ID: wpr-59829

ABSTRACT

Replication-incompetent adenoviruses expressing three major glycoproteins (gB, gC, and gD) of pseudorabies virus (PrV) were constructed and used to examine the ability of these glycoproteins to induce protective immunity against a lethal challenge. Among three constructs, recombinant adenovirus expressing gB (rAd-gB) was found to induce the most potent immunity biased to Th1-type, as determined by the IgG isotype ratio and the profile of the Th1/Th2 cytokine production. Conversely, the gC-expressing adenovirus (rAd-gC) revealed Th2-type immunity and the gD-expressing adenovirus (rAd-gD) induced lower levels of IFN-gamma and IL-4 production than other constructs, except IL-2 production. Mucosal delivery of rAd-gB induced mucosal IgA and serum IgG responses and biased toward Th2-type immune responses. However, these effects were not observed in response to systemic delivery of rAd-gB. In addition, rAd-gB appeared to induce effective protective immunity against a virulent viral infection, regardless of whether it was administered via the muscular or systemic route. These results suggest that administration of replication-incompetent adenoviruses can induce different types of immunity depending on the expressed antigen and that recombinant adenoviruses expressing gB induced the most potent Th1-biased humoral and cellular immunity and provided effective protection against PrV infection.


Subject(s)
Animals , Female , Mice , Adenoviridae/genetics , Antibody Formation , Cell Line , Cytokines/immunology , Glycoproteins/biosynthesis , Herpesvirus 1, Suid/genetics , Immunity, Cellular , Immunoglobulin G/immunology , Mice, Inbred C57BL , Pseudorabies/immunology , Pseudorabies Vaccines/administration & dosage , Swine , Th1 Cells/immunology , Th2 Cells/immunology , Virus Replication
10.
Chinese Journal of Biotechnology ; (12): 532-535, 2004.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-270092

ABSTRACT

To construct a TK-/gG- mutant of pseudorabies virus, the gG-detected transfer vector pUSKKBB and genomic DNA of pseudorabies virus TK-/gG-/LacZ+ were co-transfected into IBRS-2 cells. Transfection progeny were plated onto PK-15 cells and incubated for 2 days under methylcellulose. Then the overlay was removed and replaced by 1% low melting point agarose in DMEM supplemented with 150 microg/mL X-gal. After 2 days, white plaques were screened for and purified 4 times. By PCR amplification of gG-deleted gene and LacZ gene, a recombinant virus with TK-/gG- phenotype was confirmed. Sequence of the PCR product revealed that there were 1,176 bp detection in gG gene of the PRV TK-/gG- mutant. Amplifying the gG-deleted gene of different generations of the TK-/gG- mutant showed that the mutant was stable within PK-15 cells. TCID50 assay indicated that the recombinant virus grows well on PK-15 cells. The mice immunized with the TK-/gG- virus showed no sign of abnormality. As a control, all mice inoculated with PRV strain died from the infection. All mice that received TK-/gG- survived after a lethal PRV challenge. However none of the mice injected with phosphate-buffer saline (PBS) survived from the challenge. The above results demonstrated that the recombinant virus could be a candidate marker vaccine strain for eradicating pseudorabies in pig herds.


Subject(s)
Animals , Mice , Herpesvirus 1, Suid , Genetics , Virulence , Mice, Inbred BALB C , Mutation , Pseudorabies Vaccines , Allergy and Immunology , Swine , Thymidine Kinase , Genetics , Vaccines, Synthetic , Viral Envelope Proteins , Genetics , Allergy and Immunology
11.
Chinese Journal of Biotechnology ; (12): 526-531, 2004.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-270093

ABSTRACT

With the application of gE gene deleted pseudarabies virus (PRV) vaccine worldwide, a corresponding differential diagnosis based on gE glycoprotein is needed in the project of PRV eradication. In this study, PRV gE gene without signal and transmembrane region was amplified by PCR and cloned into pGEX-6P-1, generated pGEX-gE. After transformation of BL21 with pGEX-gE, an expressed fusion protein(about 63kD) was identified by SDS-PAGE. The recombinant proteins are produced as inclusion bodies. By changing the inductive conditions, the formation of inclusion bodies was inhibited and tended to increase the percentage of soluble recombinant protein. The antigenic reactivity of the recombinant protein was confirmed by Western blotting with polyclonal antibodies against PRV. Using purified recombinant tgE as antigen, an ELISA was developed to detect sera of PRV infected pigs and sera of pigs immunized with gE-deleted PRV vaccine. The total of 400 serum samples collected from field were comparatively tested with the tgE-ELISA and a commercial competitive ELISA based on monoclonal antibody against gE, the results indicated that the coincidental rate between the two tests is about 94%.


Subject(s)
Animals , Diagnosis, Differential , Enzyme-Linked Immunosorbent Assay , Herpesvirus 1, Suid , Allergy and Immunology , Pseudorabies , Diagnosis , Pseudorabies Vaccines , Allergy and Immunology , Recombinant Proteins , Allergy and Immunology , Swine , Vaccination , Viral Envelope Proteins , Genetics , Allergy and Immunology
SELECTION OF CITATIONS
SEARCH DETAIL