Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.977
Filter
1.
Viruses ; 16(9)2024 Sep 21.
Article in English | MEDLINE | ID: mdl-39339973

ABSTRACT

Bovine herpesvirus type 1 (BoHV-1) establishes lifelong latency in trigeminal ganglionic (TG) neurons following intranasal and ocular infection in cattle. Periodically, the latent virus reactivates in the TG due to stress and is transported anterogradely to nerve endings in the nasal epithelium, where the virus replicates and sheds. Consequently, BoHV-1 is transmitted to susceptible animals and maintained in the cattle population. Modified live BoHV-1 vaccine strains (BoHV-1 MLV) also have a similar latency reactivation. Therefore, they circulate and are maintained in cattle herds. Additionally, they can regain virulence and cause vaccine outbreaks because they mutate and recombine with other circulating field wild-type (wt) strains. Recently, we constructed a BoHV-1 quadruple mutant virus (BoHV-1qmv) that lacks immune evasive properties due to UL49.5 and glycoprotein G (gG) deletions. In addition, it also lacks the gE cytoplasmic tail (gE CT) and Us9 gene sequences designed to make it safe, increase its vaccine efficacy against BoHV-1, and restrict its anterograde neuronal transport noted above. Further, we engineered the BoHV-1qmv-vector to serve as a subunit vaccine against the Rift Valley fever virus (BoHV-1qmv Sub-RVFV) (doi: 10.3390/v15112183). In this study, we determined the latency reactivation and nasal virus shedding properties of BoHV-1qmv (vector) and BoHV-1qmv-vectored subunit RVFV (BoHV-1qmv sub-RVFV) vaccine virus in calves in comparison to the BoHV-1 wild-type (wt) following intranasal inoculation. The real-time PCR results showed that BoHV-1 wt- but not the BoHV-1qmv vector- and BoHV-1qmv Sub-RVFV-inoculated calves shed virus in the nose following dexamethasone-induced latency reactivation; however, like the BoHV-1 wt, both the BoHV-1qmv vector and BoHV-1qmv Sub-RVFV viruses established latency, were reactivated, and replicated in the TG neurons. These results are consistent with the anterograde neurotransport function of the gE CT and Us9 sequences, which are deleted in the BoHV-1qmv and BoHV-1qmv Sub-RVFV.


Subject(s)
Herpesvirus 1, Bovine , Nasal Mucosa , Neurons , Trigeminal Ganglion , Virus Activation , Virus Latency , Virus Shedding , Animals , Herpesvirus 1, Bovine/genetics , Herpesvirus 1, Bovine/physiology , Herpesvirus 1, Bovine/immunology , Cattle , Nasal Mucosa/virology , Trigeminal Ganglion/virology , Neurons/virology , Gene Deletion , Vaccines, Attenuated/immunology , Vaccines, Attenuated/genetics , Vaccines, Attenuated/administration & dosage , Virus Replication , Cattle Diseases/virology , Cattle Diseases/prevention & control , Cattle Diseases/immunology , Vaccines, Subunit/immunology , Vaccines, Subunit/genetics , Herpesviridae Infections/veterinary , Herpesviridae Infections/virology , Herpesviridae Infections/prevention & control , Herpesviridae Infections/immunology , Viral Vaccines/immunology , Viral Vaccines/genetics , Genetic Vectors/genetics , Infectious Bovine Rhinotracheitis/virology , Infectious Bovine Rhinotracheitis/prevention & control , Infectious Bovine Rhinotracheitis/immunology , Herpesvirus Vaccines/genetics , Herpesvirus Vaccines/immunology
2.
Virulence ; 15(1): 2398171, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39258802

ABSTRACT

Orf virus (ORFV) is an acute contact, epitheliotropic, zoonotic, and double-stranded DNA virus that causes significant economic losses in the livestock industry. The objective of this study is to design an immunoinformatics-based multi-epitope subunit vaccine against ORFV. Various immunodominant cytotoxic T lymphocytes (CTL), helper T lymphocytes (HTL), and B-cell epitopes from the B2L, F1L, and 080 protein of ORFV were selected and linked by short connectors to construct a multi-epitope subunit vaccine. Immunogenicity was enhanced by adding an adjuvant ß-defensin to the N-terminal of the vaccine using the EAAAK linker. The vaccine exhibited a significant degree of antigenicity and solubility, without allergenicity or toxicity. The 3D formation of the vaccine was subsequently anticipated, improved, and verified. The optimized model exhibited a lower Z-score of -4.33, indicating higher quality. Molecular docking results demonstrated that the vaccine strongly binds to TLR2 and TLR4. Molecular dynamics results indicated that the docked vaccine-TLR complexes were stable. Immune simulation analyses further confirmed that the vaccine can induce a marked increase in IgG and IgM antibody titers, and elevated levels of IFN-γ and IL-2. Finally, the optimized DNA sequence of the vaccine was cloned into the vector pET28a (+) for high expression in the E.coli expression system. Overall, the designed multi-epitope subunit vaccine is highly stable and can induce robust humoral and cellular immunity, making it a promising vaccine candidate against ORFV.


Subject(s)
Epitopes, B-Lymphocyte , Molecular Docking Simulation , Molecular Dynamics Simulation , Orf virus , Vaccines, Subunit , Viral Vaccines , Vaccines, Subunit/immunology , Vaccines, Subunit/genetics , Vaccines, Subunit/chemistry , Animals , Orf virus/immunology , Orf virus/genetics , Viral Vaccines/immunology , Viral Vaccines/chemistry , Viral Vaccines/genetics , Mice , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/genetics , Epitopes, B-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/chemistry , Antibodies, Viral/immunology , Antibodies, Viral/blood , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/chemistry , Ecthyma, Contagious/prevention & control , Ecthyma, Contagious/immunology , Ecthyma, Contagious/virology , Mice, Inbred BALB C , Female , T-Lymphocytes, Cytotoxic/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology
3.
Viruses ; 16(9)2024 Sep 14.
Article in English | MEDLINE | ID: mdl-39339941

ABSTRACT

African swine fever (ASF) is an acute infectious disease with a high mortality rate in both domestic and wild boars. Commercial vaccines or antiviral drugs for ASF were not available due to the complex diversity of the structure and genome of its pathogen African swine fever virus (ASFV). In recent years, there have been many reports on candidate strains of attenuated vaccines for ASFV. In this study, we obtained a recombinant virus named SY18ΔL60LΔCD2v by simultaneously deleting the L60L gene and CD2v gene from highly virulent strain SY18. In vitro, SY18ΔL60LΔCD2v displayed a decreased growth kinetic compared to that of parental SY18. In vivo, high doses (105 TCID50) of SY18ΔL60LΔCD2v can protect pigs (5/5) from attacks by the parental SY18 strain (102 TCID50). Low doses (102 TCID50) of SY18ΔL60LΔCD2v only protected 20% of pigs (1/5) from attacks by the parental SY18 strain (102 TCID50). The results indicated that the absence of these two genes in SY18 could induce protection against the homologous parental strain, and there were no obvious clinical symptoms or viremia. These results indicate that the SY18ΔL60LΔCD2v strain can serve as a new live attenuated vaccine candidate for the prevention and control of ASFV infection.


Subject(s)
African Swine Fever Virus , African Swine Fever , Gene Deletion , Vaccines, Attenuated , Viral Vaccines , African Swine Fever Virus/genetics , African Swine Fever Virus/immunology , African Swine Fever Virus/pathogenicity , Animals , Swine , African Swine Fever/prevention & control , African Swine Fever/virology , African Swine Fever/immunology , Vaccines, Attenuated/immunology , Vaccines, Attenuated/genetics , Vaccines, Attenuated/administration & dosage , Viral Vaccines/immunology , Viral Vaccines/genetics , Viral Proteins/genetics , Viral Proteins/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood , Viremia/prevention & control
4.
Microb Genom ; 10(9)2024 Sep.
Article in English | MEDLINE | ID: mdl-39222347

ABSTRACT

The chicken immune system and microbiota play vital roles in maintaining gut homeostasis and protecting against pathogens. In mammals, XCR1+ conventional dendritic cells (cDCs) are located in the gut-draining lymph nodes and play a major role in gut homeostasis. These cDCs sample antigens in the gut luminal contents and limit the inflammatory response to gut commensal microbes by generating appropriate regulatory and effector T-cell responses. We hypothesized that these cells play similar roles in sustaining gut homeostasis in chickens, and that chickens lacking XCR1 were likely to contain a dysbiotic caecal microbiota. Here we compare the caecal microbiota of chickens that were either heterozygous or homozygous XCR1 knockouts, that had or had not been vaccinated for infectious bronchitis virus (IBV). We used short-read (Illumina) and long-read (PacBio HiFi) metagenomic sequencing to reconstruct 670 high-quality, strain-level metagenome assembled genomes. We found no significant differences between alpha diversity or the abundance of specific microbial taxa between genotypes. However, IBV vaccination was found to correlate with significant differences in the richness and beta diversity of the microbiota, and to the abundance of 40 bacterial genera. In conclusion, we found that a lack of XCR1 was not correlated with significant changes in the chicken microbiota, but IBV vaccination was.


Subject(s)
Cecum , Chickens , Gastrointestinal Microbiome , Infectious bronchitis virus , Animals , Chickens/microbiology , Infectious bronchitis virus/immunology , Infectious bronchitis virus/genetics , Cecum/microbiology , Vaccination , Poultry Diseases/microbiology , Poultry Diseases/virology , Poultry Diseases/immunology , Coronavirus Infections/veterinary , Coronavirus Infections/prevention & control , Coronavirus Infections/immunology , Viral Vaccines/immunology , Viral Vaccines/genetics , Receptors, G-Protein-Coupled/genetics , Metagenome , Dendritic Cells/immunology , Bacteria/classification , Bacteria/genetics , Metagenomics
5.
Arch Microbiol ; 206(10): 393, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39240318

ABSTRACT

Middle East respiratory syndrome coronavirus (MERS-CoV) is a zoonotic disease affecting camels and humans. The live attenuated vaccine represents a candidate human vaccine because it can induce strong immune responses in immunized hosts. The attenuated vaccine strain of the highly pathogenic virus can also be used to produce a cell-based vaccine in the BSL2 GMP facility. In this study, we evaluated the reversion potential of pathogenicity to pathogenic wild-type virus to ensure the safety of the live attenuated vaccine strain. We passaged our previously developed cold-adapted live attenuated MERS-CoV vaccine strain at 22 °C (EMC2012-CA22°C) in Vero cells at 37 °C as often as 15 times to determine the potential of pathogenicity reversion in hDPP4 (human dipeptidyl peptidase 4)-transgenic mice, K18-hDPP4. The serial passage of EMC2012-CA22°C in Vero cells at 37 °C up to 15 times did not result in pathogenicity reversion to wild-type MERS-CoV. In K18-hDPP4 mice infected with this virus, no weight loss or mortality was observed, and no virus was detected in tissues such as the lung, kidney, brain, and nasal turbinate. In addition, mice immunized with this virus produced a robust neutralizing antibody response and were fully protected from lethal challenge with wild-type MERS-CoV. The cold-adapted attenuated MERS-CoV vaccine strain (EMC2012-CA22°C) was not reverted to wild-type pathogenic virus after 15 passages in Vero cells at 37 °C.


Subject(s)
Cold Temperature , Middle East Respiratory Syndrome Coronavirus , Vaccines, Attenuated , Viral Vaccines , Animals , Chlorocebus aethiops , Vero Cells , Middle East Respiratory Syndrome Coronavirus/genetics , Middle East Respiratory Syndrome Coronavirus/immunology , Vaccines, Attenuated/immunology , Mice , Viral Vaccines/immunology , Viral Vaccines/genetics , Coronavirus Infections/prevention & control , Coronavirus Infections/virology , Coronavirus Infections/immunology , Mice, Transgenic , Humans , Antibodies, Viral/immunology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Serial Passage , Dipeptidyl Peptidase 4/genetics , Female
6.
Viruses ; 16(8)2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39205155

ABSTRACT

Filoviruses, like the Marburg (MARV) and Ebola (EBOV) viruses, have caused outbreaks associated with significant hemorrhagic morbidity and high fatality rates. Vaccines offer one of the best countermeasures for fatal infection, but to date only the EBOV vaccine has received FDA licensure. Given the limited cross protection between the EBOV vaccine and Marburg hemorrhagic fever (MHF), we analyzed the protective efficacy of a similar vaccine, rVSV-MARV, in the lethal cynomolgus macaque model. NHPs vaccinated with a single dose (as little as 1.6 × 107 pfu) of rVSV-MARV seroconverted to MARV G-protein prior to challenge on day 42. Vaccinemia was measured in all vaccinated primates, self-resolved by day 14 post vaccination. Importantly, all vaccinated NHPs survived lethal MARV challenge, and showed no significant alterations in key markers of morbid disease, including clinical signs, and certain hematological and clinical chemistry parameters. Further, apart from one primate (from which tissues were not collected and no causal link was established), no pathology associated with Marburg disease was observed in vaccinated animals. Taken together, rVSV-MARV is a safe and efficacious vaccine against MHF in cynomolgus macaques.


Subject(s)
Macaca fascicularis , Marburg Virus Disease , Marburgvirus , Vesiculovirus , Viral Vaccines , Animals , Marburg Virus Disease/prevention & control , Marburg Virus Disease/immunology , Marburg Virus Disease/virology , Marburgvirus/immunology , Marburgvirus/genetics , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Vesiculovirus/genetics , Vesiculovirus/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Disease Models, Animal , Vaccination , Male , Female , Vaccine Efficacy , Genetic Vectors , Immunogenicity, Vaccine
7.
Viruses ; 16(8)2024 Aug 03.
Article in English | MEDLINE | ID: mdl-39205223

ABSTRACT

Newcastle disease (ND) is caused by virulent strains of avian paramyxovirus type 1, also known as Newcastle disease virus (NDV). Despite vaccination, the frequency of reported outbreaks in Ethiopia has increased. From January to June 2022, an active outbreak investigation was conducted in six commercial chicken farms across areas of central Ethiopia to identify the circulating NDV strains. Thirty pooled tissue specimens were collected from chickens suspected of being infected with NDV. A questionnaire survey of farm owners and veterinarians was also carried out to collect information on the farms and the outbreak status. NDV was isolated using specific-pathogen-free (SPF)-embryonated chicken eggs and detected using haemagglutination and the reverse transcriptase-polymerase chain reaction (RT-PCR). The genotype and virulence of field NDV isolates were determined using phylogenetic analysis of fusion (F) protein gene sequences and the mean death time (MDT) test in SPF-embryonated chicken eggs. The questionnaire results revealed that ND caused morbidity (23.1%), mortality (16.3%), case fatality (70.8%), and significant economic losses. Eleven of thirty tissue specimens tested positive for NDV using haemagglutination and RT-PCR. The MDT testing and sequence analysis revealed the presence of virulent NDV classified as genotype VII of class II velogenic pathotype and distinct from locally used vaccine strains (genotype II). The amino acid sequences of the current virulent NDV fusion protein cleavage site motif revealed 112RRQKR↓F117, unlike the locally used avirulent vaccine strains (112GRQGR↓L117). The epidemiological data, MDT results, cleavage site sequence, and phylogenetic analysis all indicated that the present NDV isolates were virulent. The four NDV sequences were deposited in GenBank with accession numbers F gene (PP726912-15) and M gene (PP726916-19). The genetic difference between avirulent vaccine strains and circulating virulent NDV could explain the low level of protection provided by locally used vaccines. Further studies are needed to better understand the circulating NDV genotypes in different production systems.


Subject(s)
Chickens , Disease Outbreaks , Genotype , Newcastle Disease , Newcastle disease virus , Phylogeny , Poultry Diseases , Viral Vaccines , Animals , Newcastle disease virus/genetics , Newcastle disease virus/isolation & purification , Newcastle disease virus/classification , Newcastle disease virus/pathogenicity , Chickens/virology , Ethiopia/epidemiology , Newcastle Disease/virology , Newcastle Disease/epidemiology , Poultry Diseases/virology , Poultry Diseases/epidemiology , Viral Vaccines/immunology , Viral Vaccines/genetics , Viral Vaccines/administration & dosage , Virulence , Farms , Viral Fusion Proteins/genetics
8.
Viruses ; 16(8)2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39205300

ABSTRACT

African swine fever (ASF) is a highly contagious and severe hemorrhagic transboundary swine viral disease with up to a 100% mortality rate, which leads to a tremendous socio-economic loss worldwide. The lack of safe and efficacious ASF vaccines is the greatest challenge in the prevention and control of ASF. In this study, we generated a safe and effective live-attenuated virus (LAV) vaccine candidate VNUA-ASFV-LAVL3 by serially passaging a virulent genotype II strain (VNUA-ASFV-L2) in an immortalized porcine alveolar macrophage cell line (3D4/21, 50 passages). VNUA-ASFV-LAVL3 lost its hemadsorption ability but maintained comparable growth kinetics in 3D4/21 cells to that of the parental strain. Notably, it exhibited significant attenuation of virulence in pigs across different doses (103, 104, and 105 TCID50). All vaccinated pigs remained healthy with no clinical signs of African swine fever virus (ASFV) infection throughout the 28-day observation period of immunization. VNUA-ASFV-LAVL3 was efficiently cleared from the blood at 14-17 days post-infection, even at the highest dose (105 TCID50). Importantly, the attenuation observed in vivo did not compromise the ability of VNUA-ASFV-LAVL3 to induce protective immunity. Vaccination with VNUA-ASFV-LAVL3 elicited robust humoral and cellular immune responses in pigs, achieving 100% protection against a lethal wild-type ASFV (genotype II) challenge at all tested doses (103, 104, and 105 TCID50). Furthermore, a single vaccination (104 TCID50) provided protection for up to 2 months. These findings suggest that VNUA-ASFV-LAVL3 can be utilized as a promising safe and efficacious LAV candidate against the contemporary pandemic genotype II ASFV.


Subject(s)
African Swine Fever Virus , African Swine Fever , Genotype , Vaccines, Attenuated , Viral Vaccines , Animals , African Swine Fever Virus/genetics , African Swine Fever Virus/immunology , Vaccines, Attenuated/immunology , Vaccines, Attenuated/genetics , Vaccines, Attenuated/administration & dosage , Swine , African Swine Fever/prevention & control , African Swine Fever/immunology , African Swine Fever/virology , Viral Vaccines/immunology , Viral Vaccines/genetics , Viral Vaccines/administration & dosage , Antibodies, Viral/blood , Antibodies, Viral/immunology , Cell Line , Virulence , Vaccination/veterinary
9.
Int J Biol Macromol ; 277(Pt 4): 134366, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39098702

ABSTRACT

Intact capsids of foot-and-mouth disease virus (FMDV) play a vital role in eliciting a protective immune response. Any change in the physico-chemical environment of the capsids results in dissociation and poor immunogenicity. Structural bioinfomatics studies have been carried out to predict the amino acids at the interpentameric region that resulted in the identification of mutant virus-like particles(VLPs) of FMDV serotype Asia1/IND/63/1972. The insect cell expressed VLPs were evaluated for their stability by sandwich ELISA. Among 10 mutants, S93H showed maximum retention of antigenicity at different temperatures, indicating its higher thermal stability as revealed by the in-silico analysis and retained the antigenic sites of the virus demonstrated by Sandwich ELISA. The concordant results of the liquid phase blocking ELISA for estimation of antibody titre of known sera with stable mutant VLP as antigen in place of virus antigen demonstrate its diagnostic potential. The stable mutant VLP elicited a robust immune response with 85.6 % protection in guinea pigs against virus challenge. The stabilized VLP based antigen requires minimum biosafety and cold storage for production and transit besides, complying with differentiation of infected from vaccinated animals. It can effectively replace the conventional virus handling during antigen production for prophylactic and diagnostic use.


Subject(s)
Foot-and-Mouth Disease Virus , Foot-and-Mouth Disease , Serogroup , Foot-and-Mouth Disease Virus/immunology , Foot-and-Mouth Disease Virus/genetics , Animals , Foot-and-Mouth Disease/prevention & control , Foot-and-Mouth Disease/diagnosis , Foot-and-Mouth Disease/immunology , Guinea Pigs , Vaccines, Virus-Like Particle/immunology , Vaccines, Virus-Like Particle/genetics , Antibodies, Viral/immunology , Antibodies, Viral/blood , Antigens, Viral/immunology , Antigens, Viral/genetics , Capsid Proteins/immunology , Capsid Proteins/genetics , Capsid Proteins/chemistry , Viral Vaccines/immunology , Viral Vaccines/genetics , Mutation
10.
Gene ; 930: 148859, 2024 Dec 20.
Article in English | MEDLINE | ID: mdl-39151673

ABSTRACT

Newcastle disease was suspected in 37 commercial poultry farms, including 12 layer and 25 broiler farms in four districts of Gujarat, India. Vaccination had been done in 32 (20 broilers and 12 layers) farms. Tissue samples from each farm were pooled as one sample. In egg embryo inoculation, HA-HI and PCR, respectively, 32/37, 29/37, and 24/37 samples were found positive. Pathotyping by mean death time calculation and primer combination PCR revealed velogenic NDV, which was later confirmed with the presence of the 112-RRQKR*F-117 sequence at the F protein cleavage site. Phylogenetic analysis of full F gene sequences (N=10) confirmed the presence of sub-genotype VII.2 in 9/10 sequences, and genotype II in one sample. These 9 sequences were only 0.7 to 2.6 % divergent with two VII.2 (=VIIi) sequences (HQ697254.1 chicken/Banjarmas/Indonesia and KU862293.1 Parakeet/Karachi/Pakistan) but had 2.2 to 3.6 % diversion from two VII.2 sequences (OR185447 and MZ546197) from India. Then branching was found from sequences of VIIh, VIIk (VII.2), and VIIa (VII.1.2), and then from sub-genotypes VII.1.1 and VII.1.2. Due to less than 5 % diversion, these sequences could not be qualified as new sub-genotype in evolutionary distance analysis. At the amino acid level, our sequences had aa N-T-I-A-L-T at 24-79-125-385-445-482. Whereas at the same positions, in most of the retrieved VII.2 sequences and vaccines, the sequence was S-A-V-T-Q/I- E/A. Two sequences revealed additional six and four amino acid differences,respectively.This indicates rapid continuous genetic evolution of sub-genotype VII.2 and partially explains vaccinal immunity escape.


Subject(s)
Chickens , Evolution, Molecular , Genotype , Newcastle Disease , Newcastle disease virus , Phylogeny , Poultry Diseases , Animals , Newcastle disease virus/genetics , India/epidemiology , Newcastle Disease/virology , Chickens/virology , Poultry Diseases/virology , Viral Vaccines/genetics , Viral Vaccines/immunology , Vaccination/veterinary , Farms
11.
Microbiol Spectr ; 12(10): e0069224, 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39145626

ABSTRACT

Porcine epidemic diarrhea virus (PEDV) is an enteropathogenic coronavirus that causes substantial economic loss to the global pig industry. The emergence of PEDV variants has increased the need for new vaccines, as commercial vaccines confer inferior protection against currently circulating strains. It is well established that the induction of mucosal immunity is crucial for PEDV vaccines to provide better protection against PEDV infection. In this study, we constructed a recombinant adenovirus expressing the core neutralization epitope (COE) of G2b PEDV based on human adenovirus serotype 5 (Ad5). We evaluated the effects of different administration routes and doses of vaccine immunogenicity in Balb/c mice. Both intramuscular (IM) and intranasal (IN) administration elicited significant humoral responses, including COE-specific IgG in serum and mucosal secretions, along with serum-neutralizing antibodies. Moreover, IN delivery was more potent than IM in stimulating IgA in serum and mucosal samples and in dampening the immune response to the Ad5 vector. The immune response was stronger after high versus low dose IM injection, whereas no significant difference was observed between high and low IN doses. In summary, our findings provide important insights for developing novel PEDV vaccines.IMPORTANCEPorcine epidemic diarrhea (PED) is a highly contagious disease that has severe economic implications for the pork industry. Developing an effective vaccine against PEDV remains a necessity. Here, we generated a recombinant adenovirus vaccine based on Ad5 to express the COE protein of PEDV (rAd5-PEDV-COE) and systematically evaluated the immunogenicity of the adenovirus-vectored vaccine using different administration routes (intramuscular and intranasal) and doses in a mouse model. Our results show that rAd5-PEDV-COE induced potent systemic humoral response regardless of the dose or immunization route. Notably, intranasal delivery was superior to induce peripheral and mucosal IgA antibodies compared with intramuscular injection. Our data provide valuable insights into designing novel PEDV vaccines.


Subject(s)
Administration, Intranasal , Antibodies, Neutralizing , Antibodies, Viral , Immunity, Mucosal , Mice, Inbred BALB C , Porcine epidemic diarrhea virus , Vaccines, Synthetic , Animals , Mice , Porcine epidemic diarrhea virus/immunology , Porcine epidemic diarrhea virus/genetics , Antibodies, Viral/blood , Antibodies, Viral/immunology , Swine , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Female , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Coronavirus Infections/prevention & control , Coronavirus Infections/immunology , Coronavirus Infections/veterinary , Adenoviridae/genetics , Adenoviridae/immunology , Humans , Swine Diseases/prevention & control , Swine Diseases/immunology , Swine Diseases/virology , Antibody Formation/immunology , Immunoglobulin A , Genetic Vectors/genetics
12.
Virus Genes ; 60(5): 528-536, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38990486

ABSTRACT

Myxoma virus (MYXV) is a double-stranded DNA-containing virus of the family Poxviridae, genus Leporipoxvirus. MYXV is an important model virus for evolutionary and immunological research and a promising oncolytic. In this study, we sequenced and analyzed two complete genomes of MYXV virus vaccine strains B-82 and Rabbivac-B, which are widely used for vaccine production in Russia. Here, we first show that MYXV vaccine strains B-82 and Rabbivac-B share a common origin with the American recombinant MYXV MAV vaccine strain. In addition, our data suggest that the MYXV B-82 and Rabbivac-B strains contain a number of genes at the 5' and 3' ends that are identical to the virulent MYXV Lausanne strain. Several unique genetic signatures were identified in the M013L, M017L, M023, and M121R genes, helping to achieve high genetic resolution between vaccine strains. Overall, these findings highlight the evolutionary flexibility of certain genes in the MYXV genome and provide insights into the molecular epidemiology of the virus and subsequent vaccine development.


Subject(s)
Genome, Viral , Myxoma virus , Phylogeny , Viral Vaccines , Genome, Viral/genetics , Myxoma virus/genetics , Viral Vaccines/genetics , Viral Vaccines/immunology , Animals , Evolution, Molecular , Russia
13.
Braz J Microbiol ; 55(3): 2953-2968, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39060911

ABSTRACT

Canine parvovirus (CPV-2) is a highly contagious virus affecting dogs worldwide, posing a significant threat. The VP2 protein stands out as the predominant and highly immunogenic structural component of CPV-2. Soon after its emergence, CPV-2 was replaced by variants known as CPV-2a, 2b and 2c, marked by changes in amino acid residue 426 of VP2. Additional amino acid alterations have been identified within VP2, with certain modifications serving as signatures of emerging variants. In Brazil, CPV-2 outbreaks persist with diverse VP2 profiles. Vaccination is the main preventive measure against the virus. However, the emergence of substitutions presents challenges to conventional vaccine methods. Commercial vaccines are formulated with strains that usually do not match those currently circulating in the field. To address this, the study aimed to investigate CPV-2 variants in Brazil, predict epitopes, and design an in silico vaccine tailored to local variants employing reverse vaccinology. The methodology involved data collection, genetic sequence analysis, and amino acid comparison between field strains and vaccines, followed by the prediction of B and T cell epitope regions. The predicted epitopes were evaluated for antigenicity, allergenicity and toxicity. The final vaccine construct consisted of selected epitopes linked to an adjuvant and optimized for expression in Escherichia coli. Structural predictions confirmed the stability and antigenicity of the vaccine, while molecular docking demonstrated interaction with the canine toll-like receptor 4. Molecular dynamics simulations indicated a stable complex formation. In silico immune simulations demonstrated a progressive immune response post-vaccination, including increased antibody production and T-helper cell activity. The multi-epitope vaccine design targeted prevalent CPV-2 variants in Brazil and potentially other regions globally. However, experimental validation is essential to confirm our in silico findings.


Subject(s)
Computer Simulation , Dog Diseases , Parvoviridae Infections , Parvovirus, Canine , Viral Vaccines , Parvovirus, Canine/immunology , Parvovirus, Canine/genetics , Parvovirus, Canine/chemistry , Animals , Dogs , Dog Diseases/prevention & control , Dog Diseases/immunology , Dog Diseases/virology , Parvoviridae Infections/prevention & control , Parvoviridae Infections/veterinary , Parvoviridae Infections/immunology , Brazil , Viral Vaccines/immunology , Viral Vaccines/genetics , Viral Vaccines/chemistry , Vaccinology/methods , Capsid Proteins/immunology , Capsid Proteins/genetics , Capsid Proteins/chemistry , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/genetics , Epitopes/immunology , Epitopes/genetics , Epitopes/chemistry , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/chemistry
14.
Vet Microbiol ; 296: 110192, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39032444

ABSTRACT

VP1, a major immunogenic protein of foot-and-mouth disease virus (FMDV), facilitates viral attachment and entry into host cells. VP1 possesses critical epitope sequences responsible for inducing neutralizing antibodies but its expression using Saccharomyces cerevisiae has been hampered despite evidence that the presence of VP1 does not negatively impact the yeast's biology. In this study, we fused proteins to enhance VP1 expression using S. cerevisiae. Among short P1 chimeras containing VP1 including VP3-VP1 and VP2-VP1, VP3-VP1 fusion proteins showed higher expression levels than VP2-VP1. We subsequently designed new fusion proteins, of which 20 amino acids of N-terminal VP3 fused with VP1-Co1 (referred to 20aaVP3-VP1-Co1) showed the highest expression level. Lowering the culture temperature from 30 °C to 20 °C further enhanced fusion protein production. The highest expression level of 20aaVP3-VP1-Co1 was estimated to be 7.7 mg/L, which is comparable to other heterologous proteins produced using our S. cerevisiae expression system. Oral administration of the cell expressing 20aaVP3-VP1-Co1 induced VP1-specific IgG and IgA responses in mice. The S. cerevisiae-expressed 20aaVP3-VP1-Co1 fusion protein induced a significant immune response to the FMDV structural epitope protein, which opens the possibility of an oral FMDV vaccine.


Subject(s)
Antibodies, Viral , Capsid Proteins , Foot-and-Mouth Disease Virus , Foot-and-Mouth Disease , Recombinant Fusion Proteins , Saccharomyces cerevisiae , Viral Vaccines , Animals , Foot-and-Mouth Disease Virus/immunology , Foot-and-Mouth Disease Virus/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/immunology , Capsid Proteins/genetics , Capsid Proteins/immunology , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/genetics , Mice , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Antibodies, Viral/blood , Foot-and-Mouth Disease/prevention & control , Foot-and-Mouth Disease/immunology , Administration, Oral , Immunization , Female , Codon , Antibodies, Neutralizing/immunology , Mice, Inbred BALB C , Immunoglobulin A
15.
Virol Sin ; 39(4): 675-684, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38997087

ABSTRACT

Norovirus (NoV) infection is a major cause of gastroenteritis worldwide. The virus poses great challenges in developing vaccines with broad immune protection due to its genetic and antigenic diversity. To date, there are no approved NoV vaccines for clinical use. Here, we aimed to develop a broad-acting quadrivalent NoV vaccine based on a chimpanzee adenovirus vector, AdC68, carrying the major capsid protein (VP1) of noroviral GI and GII genotypes. Compared to intramuscular (i.m.), intranasal (i.n.), or other prime-boost immunization regimens (i.m. â€‹+ â€‹i.m., i.m. â€‹+ â€‹i.n., i.n. â€‹+ â€‹i.m.), AdC68-GI.1-GII.3 (E1)-GII.4-GII.17 (E3), administered via i.n. â€‹+ â€‹i.n. induced higher titers of serum IgG antibodies and higher IgA antibodies in bronchoalveolar lavage fluid (BALF) and saliva against the four homologous VP1s in mice. It also significantly stimulated the production of blocking antibodies against the four genotypes. In response to re-stimulation with virus-like particles (VLP)-GI.1, VLP-GII.3, VLP-GII.4, and VLP-GII.17, the quadrivalent vaccine administered according to the i.n. â€‹+ â€‹i.n. regimen effectively triggered specific cell-mediated immune responses, primarily characterized by IFN-γ secretion. Furthermore, the preparation of this novel quadrivalent NoV vaccine requires only a single recombinant adenovirus to provide broad preventive immunity against the major GI/GII epidemic strains, making it a promising vaccine candidate for further development.


Subject(s)
Adenoviridae , Antibodies, Viral , Caliciviridae Infections , Genetic Vectors , Mice, Inbred BALB C , Norovirus , Pan troglodytes , Viral Vaccines , Animals , Norovirus/immunology , Norovirus/genetics , Antibodies, Viral/blood , Antibodies, Viral/immunology , Mice , Caliciviridae Infections/prevention & control , Caliciviridae Infections/immunology , Viral Vaccines/immunology , Viral Vaccines/genetics , Viral Vaccines/administration & dosage , Genetic Vectors/genetics , Genetic Vectors/immunology , Adenoviridae/genetics , Adenoviridae/immunology , Female , Capsid Proteins/genetics , Capsid Proteins/immunology , Immunoglobulin G/blood , Gastroenteritis/prevention & control , Gastroenteritis/virology , Gastroenteritis/immunology , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Immunoglobulin A/blood , Genotype , Saliva/immunology , Saliva/virology , Bronchoalveolar Lavage Fluid/immunology , Bronchoalveolar Lavage Fluid/virology
16.
Front Immunol ; 15: 1408510, 2024.
Article in English | MEDLINE | ID: mdl-39021566

ABSTRACT

Equid alphaherpesvirus 1 (EqAHV1) is a viral pathogen known to cause respiratory disease, neurologic syndromes, and abortion storms in horses. Currently, there are no vaccines that provide complete protection against EqAHV1. Marker vaccines and the differentiation of infected and vaccinated animals (DIVA) strategy are effective for preventing and controlling outbreaks but have not been used for the prevention of EqAHV1 infection. Glycoprotein 2 (gp2), located on the envelope of viruses (EqAHV1), exhibits high antigenicity and functions as a molecular marker for DIVA. In this study, a series of EqAHV1 mutants with deletion of gp2 along with other virulence genes (TK, UL24/TK, gI/gE) were engineered. The mutant viruses were studied in vitro and then in an in vivo experiment using Golden Syrian hamsters to assess the extent of viral attenuation and the immune response elicited by the mutant viruses in comparison to the wild-type (WT) virus. Compared with the WT strain, the YM2019 Δgp2, ΔTK/gp2, and ΔUL24/TK/gp2 strains exhibited reduced growth in RK-13 cells, while the ΔgI/gE/gp2 strain exhibited significantly impaired proliferation. The YM2019 Δgp2 strain induced clinical signs and mortality in hamsters. In contrast, the YM2019 ΔTK/gp2 and ΔUL24/TK/gp2 variants displayed diminished pathogenicity, causing no observable clinical signs or fatalities. Immunization with nasal vaccines containing YM2019 ΔTK/gp2 and ΔUL24/TK/gp2 elicited a robust immune response in hamsters. In particular, compared with the vaccine containing the ΔTK/gp2 strain, the vaccine containing the ΔUL24/TK/gp2 strain demonstrated enhanced immune protection upon challenge with the WT virus. Furthermore, an ELISA for gp2 was established and refined to accurately differentiate between infected and vaccinated animals. These results confirm that the ΔUL24/TK/gp2 strain is a safe and effective live attenuated vaccine candidate for controlling EqAHV1 infection.


Subject(s)
Herpesviridae Infections , Herpesvirus 1, Equid , Vaccines, Attenuated , Animals , Vaccines, Attenuated/immunology , Herpesviridae Infections/prevention & control , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Herpesviridae Infections/veterinary , Herpesvirus 1, Equid/immunology , Herpesvirus 1, Equid/genetics , Horses , Mesocricetus , Antibodies, Viral/blood , Antibodies, Viral/immunology , Viral Envelope Proteins/immunology , Viral Envelope Proteins/genetics , Cricetinae , Horse Diseases/prevention & control , Horse Diseases/immunology , Horse Diseases/virology , Viral Vaccines/immunology , Viral Vaccines/genetics , Cell Line , Mutation
17.
Viruses ; 16(7)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-39066207

ABSTRACT

Classical Swine Fever (CSF), a highly contagious viral disease affecting pigs and wild boar, results in significant economic losses in the swine industry. In endemic regions, prophylactic vaccination and stamping-out strategies are used to control CSF outbreaks. However, sporadic outbreaks and persistent infections continue to be reported. Although the conventional attenuated CSF vaccines protect pigs against the disease, they do not allow for the differentiation of infected from vaccinated animals (DIVA), limiting their use as an eradication tool. In this study, three targeted attenuation strategies were employed to generate vaccine candidates based on the current prevalent CSFV group 2 strains GD18 and QZ07: a single deletion of H79 in Erns (QZ07-sdErnsH-KARD), double deletion of H79 and C171 in Erns (GD18-ddErnsHC-KARD and QZ07-ddErnsHC-KARD), and deletion of H79 in Erns combined with a 5-168 amino acids deletion of Npro (GD18-ddNpro-ErnsH-KARD). Additionally, a negative serological marker with four substitutions in a highly conserved epitope in E2 recognized by the monoclonal antibody 6B8 was introduced in each candidate for DIVA purposes. The safety of these four resulting vaccine candidates was evaluated in pregnant sows. Two candidates, GD18-ddErnsHC-KARD and QZ07-sdErnsH-KARD were found to be safe for pregnant sows and unlikely to cause vertical transmission. Both candidates also demonstrated potential to be used as DIVA vaccines, as was shown using a proprietary blocking ELISA based on the 6B8 monoclonal antibody. These results, together with our previous work, constitute a proof-of-concept for the rational design of CSF antigenically marked modified live virus vaccine candidates.


Subject(s)
Antibodies, Viral , Classical Swine Fever Virus , Classical Swine Fever , Vaccines, Attenuated , Viral Vaccines , Animals , Classical Swine Fever/prevention & control , Classical Swine Fever/virology , Classical Swine Fever/immunology , Swine , Female , Vaccines, Attenuated/immunology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/adverse effects , Classical Swine Fever Virus/immunology , Classical Swine Fever Virus/genetics , Viral Vaccines/immunology , Viral Vaccines/genetics , Viral Vaccines/administration & dosage , Viral Vaccines/adverse effects , Pregnancy , Antibodies, Viral/blood , Antibodies, Viral/immunology , Vaccines, Marker/immunology , Vaccines, Marker/administration & dosage , Vaccines, Marker/genetics , Vaccination/veterinary , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology
18.
Sheng Wu Gong Cheng Xue Bao ; 40(7): 2150-2161, 2024 Jul 25.
Article in Chinese | MEDLINE | ID: mdl-39044581

ABSTRACT

This study aims to develop an effective bivalent subunit vaccine that is promising to prevent both porcine deltacoronavirus (PDCoV) and porcine epidemic diarrhea virus (PEDV). The receptor-binding domains (RBDs) of PDCoV and PEDV were fused and cloned into the eukaryotic expression vector pCDNA3.1(+). The fusion protein PDCoV-RBD-PEDV-RBD (pdRBD-peRBD) was expressed by the ExpiCHOTM expression system and purified. Mice were immunized with the fusion protein at three different doses (10, 20, and 30 µg). The humoral immune response and cellular immune response induced by the fusion protein were evaluated by ELISA and flow cytometry. The neutralization titers of the serum of immunized mice against PDCoV and PEDV were determined by the microneutralization test. The results showed that high levels of IgG antibodies were induced in the three different dose groups after booster immunization, and there was no significant difference in the antibody level between different dose groups, indicating that the immunization dose of 10 µg could achieve the fine immune effect. The results of flow cytometry showed that the immunization groups demonstrated increased proportion of CD3+CD4+ T cells and decreased proportion of CD3+CD8+ T cells, which was consistent with the expectation about the humoral immune response induced by the subunit vaccine. At the same time, the levels of interleukin (IL)-2, IL-4, and interferon (IFN)-γ in the serum were determined. The results showed that the fusion protein induced both humoral immune effect and cellular immune response. The results of the neutralization test showed that the antibody induced by 10 µg fusion protein neutralized both PDCoV and PEDV in vitro, with the titers of 1:179.25 and 1:141.21, respectively. The above results suggested that the pdRBD-peRBD could induce a high level of humoral immune response at a dose of 10 µg, and the induced antibody could neutralize both PDCoV and PEDV. Therefore, the fusion protein pdRBD-peRBD is expected to be an effective subunit vaccine that can simultaneously prevent PDCoV and PEDV.


Subject(s)
Antibodies, Viral , Coronavirus Infections , Porcine epidemic diarrhea virus , Recombinant Fusion Proteins , Viral Vaccines , Animals , Porcine epidemic diarrhea virus/immunology , Porcine epidemic diarrhea virus/genetics , Mice , Swine , Viral Vaccines/immunology , Viral Vaccines/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/genetics , Coronavirus Infections/prevention & control , Coronavirus Infections/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Deltacoronavirus/immunology , Deltacoronavirus/genetics , Swine Diseases/prevention & control , Swine Diseases/immunology , Vaccines, Subunit/immunology , Vaccines, Subunit/genetics , Mice, Inbred BALB C , Female , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Protein Domains , Immunogenicity, Vaccine , Immunity, Humoral
19.
Microb Pathog ; 193: 106786, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38971506

ABSTRACT

To better understand the interaction between attenuated vaccines and host antiviral responses, we used bioinformatics and public transcriptomics data to analyze the immune response mechanisms of host cells after canine distemper virus (CDV) infection in Vero cells and screened for potential key effector factors. In this study, CDV-QN-1 infect with Vero cells at an MOI of 0.5, and total RNA was extracted from the cells 24 h later and reverse transcribed into cDNA. Transcriptome high-throughput sequencing perform using Illumina. The results showed that 438 differentially expressed genes were screened, of which 409 were significantly up-regulated and 29 were significantly down-regulated. Eight differentially expressed genes were randomly selected for RT-qPCR validation, and the change trend was consistent with the transcriptomics data. GO and KEGG analysis of differentially expressed genes revealed that most of the differentially expressed genes in CDV-QN-1 infection in the early stage were related to immune response and antiviral activity. The enriched signaling pathways mainly included the interaction between cytokines and cytokine receptors, the NF-kappa B signaling pathway, the Toll-like receptor signaling pathway, and the NOD-like receptor signaling pathway. This study provides a foundation for further exploring the pathogenesis of CDV and the innate immune response of host cells in the early stage of infection.


Subject(s)
Distemper Virus, Canine , Gene Expression Profiling , Vaccines, Attenuated , Animals , Vero Cells , Chlorocebus aethiops , Vaccines, Attenuated/immunology , Vaccines, Attenuated/genetics , Distemper Virus, Canine/genetics , Distemper Virus, Canine/immunology , Transcriptome , Signal Transduction , Computational Biology , High-Throughput Nucleotide Sequencing , Viral Vaccines/immunology , Viral Vaccines/genetics , Cytokines/metabolism , Cytokines/genetics , Distemper/virology , Distemper/genetics , Distemper/immunology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , NF-kappa B/metabolism , NF-kappa B/genetics , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism
20.
J Virol ; 98(7): e0062224, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38953377

ABSTRACT

African swine fever virus causes a lethal hemorrhagic disease in domestic swine and wild boar for which currently licensed commercial vaccines are only available in Vietnam. Development of subunit vaccines is complicated by the lack of information on protective antigens as well as suitable delivery systems. Our previous work showed that a pool of eight African swine fever virus genes vectored using an adenovirus prime and modified vaccinia virus boost could prevent fatal disease after challenge with a virulent genotype I isolate of the virus. Here, we identify antigens within this pool of eight that are essential for the observed protection and demonstrate that adenovirus-prime followed by adenovirus-boost can also induce protective immune responses against genotype I African swine fever virus. Immunization with a pool of adenoviruses expressing individual African swine fever virus genes partially tailored to genotype II virus did not protect against challenge with genotype II Georgia 2007/1 strain, suggesting that different antigens may be required to induce cross-protection for genetically distinct viruses. IMPORTANCE: African swine fever virus causes a lethal hemorrhagic disease in domestic pigs and has killed millions of animals across Europe and Asia since 2007. Development of safe and effective subunit vaccines against African swine fever has been problematic due to the complexity of the virus and a poor understanding of protective immunity. In a previous study, we demonstrated that a complex combination of eight different virus genes delivered using two different viral vector vaccine platforms protected domestic pigs from fatal disease. In this study, we show that three of the eight genes are required for protection and that one viral vector is sufficient, significantly reducing the complexity of the vaccine. Unfortunately, this combination did not protect against the current outbreak strain of African swine fever virus, suggesting that more work to identify immunogenic and protective viral proteins is required to develop a truly effective African swine fever vaccine.


Subject(s)
Adenoviridae , African Swine Fever Virus , African Swine Fever , Genetic Vectors , Genotype , Viral Vaccines , Animals , African Swine Fever Virus/genetics , African Swine Fever Virus/immunology , African Swine Fever/prevention & control , African Swine Fever/virology , African Swine Fever/immunology , Swine , Viral Vaccines/immunology , Viral Vaccines/genetics , Viral Vaccines/administration & dosage , Genetic Vectors/genetics , Adenoviridae/genetics , Adenoviridae/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Vaccines, Subunit/immunology , Vaccines, Subunit/genetics , Antigens, Viral/immunology , Antigens, Viral/genetics
SELECTION OF CITATIONS
SEARCH DETAIL