Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
1.
BMC Vet Res ; 17(1): 164, 2021 Apr 14.
Article in English | MEDLINE | ID: mdl-33853597

ABSTRACT

BACKGROUND: Since 2011, numerous highly virulent and antigenic variant viral strains have been reported in pigs that were vaccinated against the swine pseudorabies virus. These infections have led to substantial economic losses in the Chinese swine industry. RESULTS: This study, constructed a novel recombinant vaccine strain with gI/gE deletion (PRV-GD2013-ΔgI/gE) by overlapping PCR and homologous recombination technology. The growth curves and plaque morphology of the recombinant virus were similar to those of the parental strain. However, PRV-GD2013-ΔgI/gE infection was significantly attenuated in mice compared with that of PRV-GD2013. Two-week-old piglets had normal rectal temperatures and displayed no clinical symptoms after being inoculated with 105 TCID50 PRV-GD2013-ΔgI/gE, indicating that the recombinant virus was avirulent in piglets. Piglets were immunized with different doses of PRV-GD2013-ΔgI/gE, or a single dose of Bartha-K61 or DMEM, and infected with PRV-GD2013 at 14 days post-vaccination. Piglets given high doses of PRV-GD2013-ΔgI/gE showed no obvious clinical symptoms, and their antibody levels were higher than those of other groups, indicating that the piglets were completely protected from PRV-GD2013. CONCLUSIONS: The PRV-GD2013-ΔgI/gE vaccine strain could be effective for immunizing Chinese swine herds against the pseudorabies virus (PRV) strain.


Subject(s)
Pseudorabies Vaccines/immunology , Pseudorabies/prevention & control , Swine Diseases/virology , Animals , Cell Line , Cricetinae , Female , Gene Deletion , Herpesvirus 1, Suid/genetics , Herpesvirus 1, Suid/immunology , Homologous Recombination , Mice, Inbred BALB C , Polymerase Chain Reaction , Pseudorabies/immunology , Swine , Swine Diseases/immunology , Swine Diseases/prevention & control , Vaccines, Synthetic/virology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology
2.
Microbiol Immunol ; 63(7): 269-279, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31141221

ABSTRACT

Pseudorabies, a herpesvirus infection, is mainly controlled by using attenuated live vaccines. In this study, the effect of ginseng stem and leaf saponins (GSLS) in combination with selenium (Se; in the form of sodium selenite) on vaccination against attenuated pseudorabies virus (aPrV) was evaluated. It was found that GSLS and Se have an adjuvant effect and that a combination of GSLS and Se stimulates significantly enhanced immune responses than does GSLS or Se alone. Following oral administration of GSLS, mice immunized with an attenuated PrV vaccine diluted in Se-containing physiological saline solution (PSS) provoked a significantly stronger gB-specific serum antibodies response (IgG, IgG1 and IgG2a), enhanced lymphocyte proliferation and cytolytic activity of NK cells, along with higher production of cytokines (IFN-γ, IL-12, IL-5 and IL-10) by splenocytes. Notably, the combination of GSLS and Se conferred a much higher resistance to fPrV challenge after immunization of the mice with aPrV vaccine. This study offers convincing experimental evidence that an injection of Se with oral GSLS is a promising adjuvant combination that improves the efficacy of vaccination against PrV and deserves further study regarding improvement of responses to other animal vaccines.


Subject(s)
Adaptive Immunity/drug effects , Herpesvirus 1, Suid/immunology , Panax/chemistry , Plant Leaves/chemistry , Pseudorabies Vaccines/immunology , Saponins/pharmacology , Selenium/pharmacology , Vaccines, Attenuated/immunology , Adjuvants, Immunologic/administration & dosage , Administration, Oral , Animals , Antibody Formation , Cell Proliferation/drug effects , Cytokines/metabolism , Drug Combinations , Female , Foot-and-Mouth Disease/prevention & control , Immunization , Immunoglobulin G/blood , Killer Cells, Natural/drug effects , Lymphocytes/drug effects , Mice , Mice, Inbred ICR , Newcastle Disease/prevention & control , Plant Extracts/pharmacokinetics , Pseudorabies/prevention & control , Saponins/administration & dosage , Selenium/administration & dosage , Vaccination , Vaccines, Attenuated/administration & dosage
3.
BMC Vet Res ; 15(1): 2, 2019 Jan 03.
Article in English | MEDLINE | ID: mdl-30606159

ABSTRACT

BACKGROUND: Since 2011, pseudorabies caused by a variant PRV has re-emerged in many Chinese Bartha-K61-vaccinated pig farms. An efficacious vaccine is necessary to control this disease. We described the construction of a gD&gC-substituted pseudorabies virus (PRV B-gD&gCS) from the Bartha-K61 (as backbone) and AH02LA strain (as template for gD and gC genes) through bacterial artificial chromosome (BAC) technology using homologous recombination. The growth kinetics of PRV B-gD&gCS was compared with Bartha-K61. Its safety was evaluated in 28-day-old piglets. Protection efficacy was tested in piglets by lethal challenge with AH02LA at 7 days post vaccination, including body temperature, clinical symptoms, virus shedding, mortality rate, and lung lesions. RESULTS: The results showed that a BAC clone of Bartha-K61 and a B-gD&gCS clone were successfully generated. The growth kinetics of PRV B-gD&gCS strain on ST (Swine testicular) cells was similar to that of the Bartha-K61 strain. No piglets inoculated intramuscularly with PRV B-gD&gCS strain exhibited any clinical symptoms or virus shedding. After AH02LA challenge, all piglets in PRV B-gD&gCS and Bartha-K61 groups (n = 5 each) survived without exhibiting any clinical symptoms and high body temperature. More importantly, PRV B-gD&gCS strain completely prevented virus shedding in 2 piglets and reduced virus shedding post challenge in the other 3 piglets as compared with Bartha-K61 group. CONCLUSIONS: Our results suggest that PRV B-gD&gCS strain is a promising vaccine candidate for the effective control of current severe epidemic pseudorabies in China.


Subject(s)
Herpesvirus 1, Suid/immunology , Pseudorabies Vaccines/immunology , Pseudorabies/prevention & control , Swine Diseases/prevention & control , Animals , Animals, Newborn/immunology , Animals, Newborn/virology , China , Genetic Variation/genetics , Herpesvirus 1, Suid/genetics , Herpesvirus 1, Suid/physiology , Pseudorabies/immunology , Swine , Swine Diseases/immunology , Swine Diseases/virology , Vaccines, Synthetic , Virus Shedding
4.
J Cell Physiol ; 233(4): 2681-2692, 2018 04.
Article in English | MEDLINE | ID: mdl-28833090

ABSTRACT

CRSBP-1 (mammalian LYVE-1) is a membrane glycoprotein highly expressed in lymphatic endothelial cells (LECs). It has multiple ligands, including hyaluronic acid (HA) and growth factors/cytokines (e.g., PDGF-BB and VEGF-A) containing CRS motifs (clusters of basic amino-acid residues). The ligand binding activities are mediated by Link module and acidic-amino-acid-rich (AAAR) domains, respectively. These CRSBP-1/LYVE-1 ligands have been shown to induce opening of lymphatic intercellular junctions in LEC monolayers and in lymphatic vessels in wild-type mice. We hypothesize that CRSBP-1/LYVE-1 ligands, particularly CRS-containing growth factors/cytokines, are secreted by immune and cancer cells for lymphatic entry during adaptive immune responses and lymphatic metastasis. We have looked into the origin of the Link module and AAAR domain of LYVE-1 in evolution and its association with the development of lymph nodes and efficient adaptive immunity. Lymph nodes represent the only major recent innovation of the adaptive immune systems in evolution particularly to mammals and bird. Here we demonstrate that the development of the LYVE-1 gene with the AAAR domain in evolution is associated with acquisition of lymph nodes and adaptive immunity. LYVE-1 from other species, which have no lymph nodes, lack the AAAR domain and efficient adaptive immunity. Synthetic CRSBP-1 ligands PDGF and VEGF peptides, which contain the CRS motifs of PDGF-BB and VEGF-A, respectively, specifically bind to CRSBP-1 but do not interact with either PDGFßR or VEGFR2. These peptides function as adjuvants by enhancing adaptive immunity of pseudorabies virus (PRV) vaccine in pigs. These results support the notion that LYVE-1 is involved in adaptive immunity in mammals.


Subject(s)
Adaptive Immunity , Amino Acids, Acidic/metabolism , Evolution, Molecular , Lymph Nodes/immunology , Membrane Proteins/chemistry , Membrane Proteins/genetics , Adaptive Immunity/drug effects , Adjuvants, Immunologic/pharmacology , Amino Acid Sequence , Animals , Base Sequence , Female , Ligands , Lymph Nodes/drug effects , Membrane Proteins/metabolism , Peptides/pharmacology , Phylogeny , Platelet-Derived Growth Factor/pharmacology , Protein Domains , Pseudorabies Vaccines/immunology , Sequence Alignment , Sharks , Structural Homology, Protein , Structure-Activity Relationship , Sus scrofa , Vascular Endothelial Growth Factor A/pharmacology , Zebrafish
5.
BMC Vet Res ; 12(1): 277, 2016 Dec 07.
Article in English | MEDLINE | ID: mdl-27923365

ABSTRACT

BACKGROUND: Since the end of 2011 an outbreak of pseudorabies affected Chinese pig herds that had been vaccinated with the commercial vaccine made of Bartha K61 strain. It is now clear that the outbreak was caused by an emergent PRV variant. Even though vaccines made of PRV Bartha K61 strain can confer certain cross protection against PRV variants based on experimental data, less than optimal clinical protection and virus shedding reduction were observed, making the control or eradication of this disease difficult. RESULTS: An infectious clone of PRV AH02LA strain was constructed to generate a gE deletion mutant PRV(LA-AB) strain. PRV(LA-AB) strain can reach a titer of 108.43 TCID50 /mL (50% tissue culture infectious dose) on BHK-21 cells. To evaluate the efficiency of the inactivated vaccine made of PRV(LA-AB) strain, thirty 3-week-old PRV-negative piglets were divided randomly into six groups for vaccination and challenge test. All five piglets in the challenge control showed typical clinical symptoms of pseudorabies post challenge. Sneezing and nasal discharge were observed in four and three piglets in groups C(vaccinated with inactivated PRV Bartha K61 strain vaccine) and D(vaccinated with live PRV Bartha K61 strain vaccine) respectively. In contrast, piglets in both groups A(vaccinated with inactivated PRV LA-AB strain vaccine) and B(vaccinated with inactivated PRV LA-AB strain vaccine with adjuvant) presented mild or no clinical symptoms. Moreover, viral titers detected via nasal swabs were approximately 100 times lower in group B than in the challenge control, and the duration of virus shedding (3-4 days) was shorter than in either the challenge control (5-10 days) or groups C and D (5-6 days). CONCLUSIONS: The infectious clone constructed in this study harbors the whole genome of the PRV variant AH02LA strain. The gE deletion mutant PRV(LA-AB)strain generated from PRV AH02LA strain can reach a high titer on BHK-21 cells. An inactivated vaccine of PRV LA-AB provides clinical protection and significantly reduces virus shedding post challenge, especially if accompanied by the adjuvant CVC1302. While Inactivated or live vaccines made of PRV Barth K61 strain can provide only partial protection in this test.


Subject(s)
Pseudorabies Vaccines/standards , Pseudorabies/prevention & control , Swine Diseases/prevention & control , Viral Vaccines/standards , Virus Shedding/immunology , Animals , China , Gene Deletion , Herpesviridae/genetics , Herpesviridae/immunology , Nose/virology , Pseudorabies/pathology , Pseudorabies/virology , Pseudorabies Vaccines/immunology , Random Allocation , Swine , Swine Diseases/pathology , Swine Diseases/virology , Vaccines, Inactivated/immunology , Vaccines, Inactivated/standards , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viral Vaccines/immunology
6.
Toxicol Appl Pharmacol ; 278(1): 31-8, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24735996

ABSTRACT

The effect of a seven-day antibiotic therapy with doxycycline was investigated on the postvaccinal humoral and cellular immune response in pigs. The selected parameters of non-specific immunity were also studied. Fifty pigs were used (control not vaccinated (C, n=10), control vaccinated (CV, n=20), and experimental - received doxycycline (DOXY, n=20)). For vaccination live-attenuated vaccine against pseudorabies (PR) was used. From day -1 to day 5 pigs from DOXY group received doxycycline orally with drinking water, at the recommended dose. Pigs from DOXY and CV groups were vaccinated at 8 and 10 weeks of age. The results of the present study showed that cell-mediated postvaccinal immune response can be modulated by oral treatment with doxycycline. Significantly lower values of stimulation index were observed after PRV restimulation in doxycycline-treated pigs. Moreover, in the DOXY group a significant decrease in IFN-γ production after PRV restimulation was noted. The significantly lower number of CD4+CD8+ cells was also observed in doxy-treated, vaccinated pigs, 2 weeks after final vaccination. Simultaneously, specific humoral response was not disturbed. This study demonstrated the importance of defining the immune modulatory activity of doxycycline because it may alter the immune responses to vaccines. The exact mechanism of T-cell response suppression by doxycycline remains to be elucidated, however the influence of doxycycline on the secretion of various cytokines, including IFN-γ, may be considered as a possible cause. The present observations should prompt further studies on the practical significance of such phenomena in terms of clinical implications.


Subject(s)
Anti-Bacterial Agents/pharmacology , Doxycycline/pharmacology , Immunity, Cellular/drug effects , Immunity, Humoral/drug effects , Pseudorabies Vaccines/immunology , Acute-Phase Proteins/metabolism , Animals , Anti-Bacterial Agents/administration & dosage , Antibodies, Viral/blood , CD4 Lymphocyte Count , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Cell Proliferation/drug effects , Cells, Cultured , Doxycycline/administration & dosage , Drug Administration Schedule , Drug Interactions , Female , Interferon-gamma/metabolism , Interleukin-4/metabolism , Lymphocyte Activation/drug effects , Male , Models, Animal , Pseudorabies Vaccines/administration & dosage , Sus scrofa , Th1 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/virology , Th2 Cells/drug effects , Th2 Cells/immunology , Th2 Cells/virology , Time Factors , Vaccination , Vaccines, Attenuated/immunology
7.
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
8.
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
9.
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
10.
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
11.
Emerg Infect Dis ; 19(11): 1749-55, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24188614

ABSTRACT

The widely used pseudorabies virus (PRV) Bartha-K61 vaccine has played a key role in the eradication of PRV. Since late 2011, however, a disease characterized by neurologic symptoms and a high number of deaths among newborn piglets has occurred among Bartha-K61-vaccinated pigs on many farms in China. Clinical samples from pigs on 15 farms in 6 provinces were examined. The PRV gE gene was detectable by PCR in all samples, and sequence analysis of the gE gene showed that all isolates belonged to a relatively independent cluster and contained 2 amino acid insertions. A PRV (named HeN1) was isolated and caused transitional fever in pigs. In protection assays, Bartha-K61 vaccine provided 100% protection against lethal challenge with SC (a classical PRV) but only 50% protection against 4 challenges with strain HeN1. The findings suggest that Bartha-K61 vaccine does not provide effective protection against PRV HeN1 infection.


Subject(s)
Herpesvirus 1, Suid/genetics , Herpesvirus 1, Suid/immunology , Pseudorabies Vaccines/immunology , Pseudorabies/immunology , Pseudorabies/prevention & control , Swine Diseases , Animals , Antibodies, Viral/immunology , China , Herpesvirus 1, Suid/isolation & purification , Neutralization Tests , Phylogeny , Swine , Vaccination , Viral Envelope Proteins/genetics
12.
Microbiol Immunol ; 56(8): 529-40, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22551253

ABSTRACT

The co-administration of two or more cytokines may generate additive or synergistic effects for controlling infectious diseases. However, the practical use of cytokine combinations for the modulation of immune responses against inactivated vaccine has not been demonstrated in livestock yet, primarily due to protein stability, production, and costs associated with mass administration. In light of the current situation, we evaluated the immunomodulatory functions of the combined administration of swine interleukin-18 (swIL-18) and interferon-α (swIFN-α) against an inactivated PrV vaccine using attenuated Salmonella enterica serovar Typhimurium as a cytokine delivery system. Co-administration of S. enterica serovar Typhimurium expressing swIL-18 and swIFN-α produced enhanced Th1-biased humoral and cellular immune responses against the inactivated PrV vaccine, when compared to single administration of S. enterica serovar Typhimurium expressing either swIL-18 or swIFN-α. Also, enhanced immune responses in co-administered piglets occurred rapidly after virulent PrV challenge, and piglets that received co-administration of S. enterica serovar Typhimurium expressing swIL-18 and swIFN-α displayed a greater alleviation of clinical severity following the virulent PrV challenge, as determined by clinical scores and cumulative daily weight gain. Furthermore, this enhancement was confirmed by reduced nasal shedding of PrV following viral challenge. Therefore, these results suggest that oral co-administration of S. enterica serovar Typhimurium expressing swIL-18 and swIFN-α provide enhanced Th1-biased immunity against inactivated PrV vaccine to alleviate clinical signs caused by PrV challenge.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Genetic Vectors/administration & dosage , Herpesvirus 1, Suid/immunology , Interferon-alpha/administration & dosage , Interleukin-18/administration & dosage , Pseudorabies Vaccines/immunology , Th1 Cells/immunology , Animals , Body Weight , Disease Models, Animal , Immunity, Cellular , Immunity, Humoral , Interferon-alpha/genetics , Interleukin-18/genetics , Pseudorabies/immunology , Pseudorabies/pathology , Pseudorabies/prevention & control , Pseudorabies Vaccines/administration & dosage , Salmonella typhimurium/genetics , Severity of Illness Index , Swine , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology
13.
Vet Microbiol ; 258: 109104, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34004569

ABSTRACT

Pseudorabies is a highly infectious disease with severe clinical symptoms, causing acute death in infected pigs and leading to substantial economic losses among swine producers. In this study, a vaccine candidate strain in which the protein kinase UL13 gene was deleted was constructed with the CRISPR/Cas9 system based on the recombinant pseudorabies virus (PRV) ZJ01-ΔgI/gE/TK. Pigs immunized with ZJ01-ΔgI/gE/TK or ZJ01-ΔgI/gE/TK/UL13 produced high levels of anti-gB antibodies and virus-neutralizing antibodies. ZJ01-ΔgI/gE/TK/UL13 provided greater protective efficacy against challenge with PRV variant strain ZJ01 than did Bartha-K61 or ZJ01-ΔgI/gE/TK. The pigs vaccinated with ZJ01-ΔgI/gE/TK/UL13 excreted significantly less virus than those vaccinated with Bartha-K61 or ZJ01-ΔgI/gE/TK. The viral loads in the lungs of pigs treated with ZJ01-ΔgI/gE/TK/UL13 were lower than those in pigs treated with ZJ01-ΔgI/gE/TK after challenge with PRV variant strain ZJ01. These data indicated that ZJ01-ΔgI/gE/TK/UL13 had greater protective efficacy and safety than the commercial ZJ01-ΔgI/gE/TK and Bartha-K61 vaccines, and could be developed as a promising vaccine candidate for the prevention and control of this disease.


Subject(s)
Herpesvirus 1, Suid/genetics , Pseudorabies Vaccines/immunology , Pseudorabies/virology , Swine Diseases/prevention & control , Animals , Cell Line , Interferon-beta/genetics , Interferon-beta/metabolism , Pseudorabies/immunology , Serologic Tests , Swine , Swine Diseases/virology
14.
Viruses ; 13(4)2021 04 16.
Article in English | MEDLINE | ID: mdl-33923590

ABSTRACT

Owing to viral evolution and recombination, emerging pseudorabies virus (PRV) strains have caused unprecedented outbreaks in swine farms even when the pigs were previously vaccinated, which might indicate that traditional vaccines were unable to provide effective protection. The development of safe and efficacious vaccines presents prospects to minimize the clinical signs and eventually eradicate the infection. In this study, we used an emerging PRV strain, HNX, as the parental strain to construct a recombinant PRV with TK/gE gene deletion and Fms-related tyrosine kinase 3 ligand (Flt3L) expression, named HNX-TK-/gE--Flt3L. HNX-TK-/gE--Flt3L enhanced the maturation of bone marrow derived dendritic cells (DCs) in vitro. Significantly more activated DCs were detected in HNX-TK-/gE--Flt3L-immunized mice compared with those immunized with HNX-TK-/gE-. Subsequently, a remarkable increase of neutralizing antibodies, gB-specific IgG antibodies, and interferon-gamma (IFN-γ) was observed in mice vaccinated with HNX-TK-/gE--Flt3L. In addition, a lower mortality and less histopathological damage were observed in HNX-TK-/gE--Flt3L vaccinated mice with upon PRV lethal challenge infection. Taken together, our results revealed the potential of Flt3L as an ideal adjuvant that can activate DCs and enhance protective immune responses and support the further evaluation of HNX-TK-/gE--Flt3L as a promising PRV vaccine candidate.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Dendritic Cells/immunology , Pseudorabies Vaccines , Pseudorabies/prevention & control , Swine Diseases/prevention & control , Adaptive Immunity , Animals , Cells, Cultured , Female , Gene Deletion , HEK293 Cells , Humans , Immunity, Innate , Mice , Mice, Inbred BALB C , Pseudorabies Vaccines/genetics , Pseudorabies Vaccines/immunology , Swine
15.
Vet Microbiol ; 255: 109022, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33711567

ABSTRACT

Herpesvirus based multivalent vaccines have been extensively studied, whereas few of them have been successfully used in clinic and animal husbandry industry due to the low expression of foreign immunogens in herpesvirus. In this study, we developed a new strategy to construct herpesvirus based bivalent vaccine with high-level expression of foreign immunogen, by which the ORF2 gene encoding the major antigen protein Cap of porcine circovirus type 2 (PCV2), was highly expressed in pseudorabies virus (PRV). To obtain the high expression of PCV2 immunogen, tandem repeats of PCV2 ORF2 gene were firstly linked by protein quantitation ratioing (PQR) linker to reach equal expression of each ORF2 gene. Then, the multiple copies of ORF2 gene were respectively inserted into the gE and gG sites of PRV using CRISPR/Cas9 system, in which the expression of ORF2 gene was driven by endogenous strong promoters of PRV. Through this way, the highest yield of Cap protein was achieved in two copies of quadruple ORF2 gene insertion. Finally, in mice and pigs immunized with the bivalent vaccine candidate, we detected high titer of specific antibodies for PRV and neutralized antibodies for PCV2, and observed protective effect of the bivalent vaccine candidate against PRV challenge in immunized pigs, suggesting a potential clinical application of the bivalent vaccine candidate we constructed. Together, our strategy could be extensively applied to the generation of other multivalent vaccines, and will pave the way to construct herpesvirus based multivalent vaccines to effectively reduce the cost of vaccine.


Subject(s)
Circovirus/immunology , Herpesvirus 1, Suid , Pseudorabies Vaccines/immunology , Pseudorabies/prevention & control , Swine Diseases/prevention & control , Animals , CRISPR-Cas Systems , Cell Line , Female , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Plasmids , Swine , Swine Diseases/blood , Viral Proteins/immunology
16.
J Vet Med Sci ; 82(6): 846-855, 2020 Jun 30.
Article in English | MEDLINE | ID: mdl-32336701

ABSTRACT

The prevalence of an emerging variant of the pseudorabies virus (PRV) has been causing serious losses to farmers in China. Moreover, the commercially available PRV vaccine often fails to provide thorough protection. Therefore, in this study, we generated a PRV-∆gC\gE∆TK strain with defects in gC, gE, and TK of PRV. Compared to the parental PRV strain and the single gene deletion strains (PRV-∆gC, PRV-∆gE, and PRV-∆TK), PRV-∆gC\gE∆TK grew slowly, and exhibited fewer and smaller plaques on swine testis (ST) cells. Furthermore, animal experiment results showed that mice that were immunized intramuscularly with PRV-∆gC\gE∆TK, survived throughout the experiment with no observed clinical symptoms, and were completely protected against PRV challenge. Additionally, deletion of the gC, gE, and TK genes significantly alleviated viral damage in the brain. Furthermore, one-day-old weaned piglets immunized intramuscularly with PRV-∆gC\gE∆TK elicited higher levels of gB antibodies against both the emerging PRV variant and the parental PRV, exhibited full protection against challenge with both variants, and showed neutralization capacity against PRV. These data suggest that PRV-∆gC\gE∆TK is a promising vaccine candidate for the control of pseudorabies.


Subject(s)
Herpesvirus 1, Suid/genetics , Herpesvirus 1, Suid/immunology , Pseudorabies Vaccines/administration & dosage , Pseudorabies/prevention & control , Swine Diseases/prevention & control , Animals , CRISPR-Cas Systems , Gene Deletion , Genes, Viral , HEK293 Cells , Humans , Immunization , Injections, Intramuscular , Mice, Inbred BALB C , Pseudorabies/virology , Pseudorabies Vaccines/immunology , Swine , Swine Diseases/virology , Vaccines, Attenuated
17.
Viruses ; 12(4)2020 03 27.
Article in English | MEDLINE | ID: mdl-32230737

ABSTRACT

Pseudorabies (PR), caused by pseudorabies virus (PRV), is an acute and febrile infectious disease in swine. To eradicate PR, a more efficacious vaccine needs to be developed. Here, the gE/gI- and TK/gE/gI-gene-deleted recombinant PRV (rGXΔgE/gI and rGXΔTK/gE/gI) are constructed through CRISPR/Cas9 and Cre/Lox systems. We found that the rGXΔTK/gE/gI was safer than rGXΔgE/gI in mice. Additionally, the effects of rGXΔgE/gI and rGXΔTK/gE/gI were further evaluated in swine. The rGXΔgE/gI and rGXΔTK/gE/gI significantly increased numbers of IFN-γ-producing CD4+ and CD8+ T-cells in swine, whereas there was no difference between rGXΔgE/gI and rGXΔTK/gE/gI. Moreover, rGXΔgE/gI and rGXΔTK/gE/gI promoted a PRV-specific humoral immune response. The PRV-specific humoral immune response induced by rGXΔgE/gI was consistent with that caused by rGXΔTK/gE/gI. After the challenge, swine vaccinated with rGXΔgE/gI and rGXΔTK/gE/gI showed no clinical signs and viral shedding. However, histopathological detection revealed that rGXΔgE/gI, not rGXΔTK/gE/gI, caused pathological lesions in brain and lung tissues. In summary, these results demonstrate that the TK/gE/gI-gene-deleted recombinant PRV was safer compared with rGXΔgE/gI in swine. The data imply that the TK/gE/gI-gene-deleted recombinant PRV may be a more efficacious vaccine candidate for the prevention of PR.


Subject(s)
CRISPR-Cas Systems , Gene Deletion , Herpesvirus 1, Suid/genetics , Herpesvirus 1, Suid/immunology , Homologous Recombination , Integrases/metabolism , Pseudorabies Vaccines/genetics , Pseudorabies Vaccines/immunology , Amino Acid Sequence , Animals , Antibodies, Viral/immunology , Female , Gene Targeting , Genetic Engineering , Genome, Viral , HEK293 Cells , Herpesvirus 1, Suid/isolation & purification , Humans , Mice , Swine , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Virulence
18.
Vet Microbiol ; 251: 108886, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33129042

ABSTRACT

Messenger RNA-based vaccines represent new tools with prophylactic and therapeutic potential characterized by high flexibility of application for infectious diseases. Pseudorabies virus (PRV) is one of the major viruses affecting the pig industry. PRV has serious effects in piglets, sows, and growing-fattening pigs and can lead to huge economic losses. In this study, an envelope glycoprotein D (gD) gene-based specific mRNA vaccine was generated, and a mouse model was used to investigate the protective efficacy of the vaccine. The gD mRNA vaccine and the recombinant plasmid pVAX-gD were transfected into BHK21 cells, and the antigenicity of the expressed proteins was detected by Western blot analysis. Groups of mice were vaccinated with the gD mRNA vaccine, pVAX-gD, and PBS. T cell immune responses were measured by flow cytometry or ELISA and serum neutralization tests every two weeks. The challenge with the PRV-XJ strain was performed eight weeks after the primary immunization, and the response was monitored for 15 days. The levels of specific and neutralizing antibodies in the gD mRNA vaccine group were significantly increased in 8 weeks compared to those in the control group, and cytokine levels, including that of IFN-γ/IL-2, were considerably higher than those in the control animal. Additionally, the proportion of CD4+/CD8+ cells in peripheral lymphocytes was remarkably increased. Our data demonstrate that mRNA is a promising and effective tool for the development of vaccines. The PRV-gD-based mRNA vaccine can elicit an efficient neutralizing antibody response and induce effective protection in mice in defense against PRV infection.


Subject(s)
Antibodies, Viral/blood , Herpesvirus 1, Suid/immunology , Pseudorabies Vaccines/immunology , Pseudorabies/prevention & control , Vaccines, Synthetic/immunology , Viral Envelope Proteins/immunology , Animals , Antibodies, Neutralizing/blood , Cell Line , Cricetinae , Female , Kidney/cytology , Mice , Mice, Inbred BALB C , Pseudorabies/immunology , Pseudorabies Vaccines/administration & dosage , Swine , Swine Diseases/immunology , Swine Diseases/prevention & control , Swine Diseases/virology , Transfection , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Viral Envelope Proteins/genetics , mRNA Vaccines
19.
J Immunol Res ; 2020: 2714257, 2020.
Article in English | MEDLINE | ID: mdl-32149156

ABSTRACT

Pseudorabies is an important infectious disease of swine, and immunization using attenuated pseudorabies virus (aPrV) vaccine is a routine practice to control this disease in swine herds. This study was to evaluate a saline solution containing ginseng stem-leaf saponins (GSLS) and sodium selenite (Se) as a vaccine adjuvant for its enhancement of immune response to aPrV vaccine. The results showed that aPrV vaccine diluted with saline containing GSLS-Se (aP-GSe) induced significantly higher immune responses than that of the vaccine diluted with saline alone (aP-S). The aP-GSe promoted higher production of gB-specific IgG, IgG1, and IgG2a, neutralizing antibody titers, secretion of Th1-type (IFN-γ, IL-2, IL-12), and Th2-type (IL-4, IL-6, IL-10) cytokines, and upregulated the T-bet/GATA-3 mRNA expression when compared to aP-S. In addition, cytolytic activity of NK cells, lymphocyte proliferation, and CD4+/CD8+ ratio was also significantly increased by aP-GSe. More importantly, aP-GSe conferred a much higher resistance of mice to a field virulent pseudorabies virus (fPrV) challenge. As the present study was conducted in mice, further study is required to evaluate the aP-GSe to improve the vaccination against PrV in swine.


Subject(s)
Adjuvants, Immunologic , Panax/chemistry , Saponins/pharmacology , Selenium/pharmacology , Th1 Cells/drug effects , Th1 Cells/immunology , Th2 Cells/drug effects , Th2 Cells/immunology , Vaccines/immunology , Animals , Antibodies, Neutralizing/immunology , Biomarkers , CD4-CD8 Ratio , Cytokines/metabolism , Female , Gene Expression , Immunoglobulin G/immunology , Mice , Pseudorabies Vaccines/immunology , Saponins/chemistry , Selenium/chemistry , Solutions , Spleen/drug effects , Spleen/immunology , Spleen/metabolism , Swine , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Th1 Cells/metabolism , Th2 Cells/metabolism
20.
Pol J Vet Sci ; 22(4): 639-645, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31867931

ABSTRACT

Since late 2011, porcine infections with highly virulent and antigenic variant of pseudorabies virus (PRV) cause great economic loss in the swine industry in China, and its emergence leads to variable protection efficacy of the commercially available PRV vaccine. In the present study, the potential cross-protective efficacy of two live virus vaccines, includ- ing a commercial vaccine, and an attenuated low pathogenic PRV variant (rPRVTJ-delTK/gE/gI) against a PRV variant Tianjing (TJ) was evaluated in piglets. Vaccination of piglets with the live vaccine Bartha-K61 could not reduce the clinical signs, and was partially efficacious in the reduc- tion of viral loads upon PRV variant TJ challenge, indicating that this live vaccine provided limited cross-protection efficacy against the PRV variant infection. Additionally, rPRVTJ-delTK/gE/gI appeared to exert some beneficial efficiency in shortening the period of clinical fever and improv- ing the growth performance of the challenged pigs. Our findings give a valuable guidance for the choice and use of PRV vaccines to control PRV variant infection in the field.


Subject(s)
Herpesvirus 1, Suid/classification , Pseudorabies Vaccines/immunology , Pseudorabies/prevention & control , Swine Diseases/prevention & control , Animals , Cell Line , Pseudorabies/virology , Swine
SELECTION OF CITATIONS
SEARCH DETAIL