Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 12.855
Filtrer
1.
Sheng Wu Gong Cheng Xue Bao ; 40(7): 2150-2161, 2024 Jul 25.
Article de Chinois | MEDLINE | ID: mdl-39044581

RÉSUMÉ

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.


Sujet(s)
Anticorps antiviraux , Infections à coronavirus , Virus de la diarrhée porcine épidémique , Protéines de fusion recombinantes , Vaccins antiviraux , Animaux , Virus de la diarrhée porcine épidémique/immunologie , Virus de la diarrhée porcine épidémique/génétique , Souris , Suidae , Vaccins antiviraux/immunologie , Vaccins antiviraux/génétique , Protéines de fusion recombinantes/immunologie , Protéines de fusion recombinantes/génétique , Infections à coronavirus/prévention et contrôle , Infections à coronavirus/immunologie , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Deltacoronavirus (genre)/immunologie , Deltacoronavirus (genre)/génétique , Maladies des porcs/prévention et contrôle , Maladies des porcs/immunologie , Vaccins sous-unitaires/immunologie , Vaccins sous-unitaires/génétique , Souris de lignée BALB C , Femelle , Anticorps neutralisants/sang , Anticorps neutralisants/immunologie , Domaines protéiques , Immunogénicité des vaccins , Immunité humorale
2.
Sci Rep ; 14(1): 16798, 2024 Jul 22.
Article de Anglais | MEDLINE | ID: mdl-39039173

RÉSUMÉ

The available Epstein Barr virus vaccine has tirelessly harnessed the gp350 glycoprotein as its target epitope, but the result has not been preventive. Right here, we designed a global multi-epitope vaccine for EBV; with special attention to making sure all strains and preventive antigens are covered. Using a robust computational vaccine design approach, our proposed vaccine is armed with 6-16 mers linear B-cell epitopes, 4-9 mer CTL epitopes, and 8-15 mer HTL epitopes which are verified to induce interleukin 4, 10 & IFN-gamma. We employed deep computational mining coupled with expert intelligence in designing the vaccine, using human Beta defensin-3-which has been reported to induce the same TLRs as EBV-as the adjuvant. The tendency of the vaccine to cause autoimmune disorder is quenched by the assurance that the construct contains no EBNA-1 homolog. The protein vaccine construct exhibited excellent physicochemical attributes such as Aliphatic index 59.55 and GRAVY - 0.710; and a ProsaWeb Z score of - 3.04. Further computational analysis revealed the vaccine docked favorably with EBV indicted TLR 1, 2, 4 & 9 with satisfactory interaction patterns. With global coverage of 85.75% and the stable molecular dynamics result obtained for the best two interactions, we are optimistic that our nontoxic, non-allergenic multi-epitope vaccine will help to ameliorate the EBV-associated diseases-which include various malignancies, tumors, and cancers-preventively.


Sujet(s)
Protéines de capside , Herpèsvirus humain de type 4 , Herpèsvirus humain de type 4/immunologie , Humains , Protéines de capside/immunologie , Protéines de capside/composition chimique , Infections à virus Epstein-Barr/immunologie , Infections à virus Epstein-Barr/prévention et contrôle , Déterminants antigéniques des lymphocytes B/immunologie , Biologie informatique/méthodes , Déterminants antigéniques des lymphocytes T/immunologie , Vaccins antiviraux/immunologie , Antigènes viraux/immunologie , Antigènes viraux/composition chimique , Modèles moléculaires , Simulation de docking moléculaire
3.
PLoS One ; 19(7): e0305417, 2024.
Article de Anglais | MEDLINE | ID: mdl-39042625

RÉSUMÉ

Hantaviruses are single-stranded RNA viruses belonging to the family Bunyaviridae that causes hantavirus cardiopulmonary syndrome (HCPS) and hemorrhagic fever with renal syndrome (HFRS) worldwide. Currently, there is no effective vaccination or therapy available for the treatment of hantavirus, hence there is a dire need for research to formulate therapeutics for the disease. Computational vaccine designing is currently a highly accurate, time and cost-effective approach for designing effective vaccines against different diseases. In the current study, we shortlisted highly antigenic proteins i.e., envelope, and nucleoprotein from the proteome of hantavirus and subjected to the selection of highly antigenic epitopes to design of next-generation multi-epitope vaccine constructs. A highly antigenic and stable adjuvant was attached to the immune epitopes (T-cell, B-cell, and HTL) to design Env-Vac, NP-Vac, and Com-Vac constructs, which exhibit stronger antigenic, non-allergenic, and favorable physiochemical properties. Moreover, the 3D structures were predicted and docking analysis revealed robust interactions with the human Toll-like receptor 3 (TLR3) to initiate the immune cascade. The total free energy calculated for Env-Vac, NP-Vac, and Com-Vac was -50.02 kcal/mol, -24.13 kcal/mol, and -62.30 kcal/mol, respectively. In silico cloning, results demonstrated a CAI value for the Env-Vac, NP-Vac, and Com-Vac of 0.957, 0.954, and 0.956, respectively, while their corresponding GC contents were 65.1%, 64.0%, and 63.6%. In addition, the immune simulation results from three doses of shots released significant levels of IgG, IgM, interleukins, and cytokines, as well as antigen clearance over time, after receiving the vaccine and two booster doses. Our vaccines against Hantavirus were found to be highly immunogenic, inducing a robust immune response that demands experimental validation for clinical usage.


Sujet(s)
Orthohantavirus , Vaccins antiviraux , Orthohantavirus/immunologie , Vaccins antiviraux/immunologie , Humains , Vaccinologie/méthodes , Simulation de docking moléculaire , Simulation numérique , Épitopes/immunologie , Épitopes/composition chimique , Modèles moléculaires , Infections à hantavirus/prévention et contrôle , Infections à hantavirus/immunologie
4.
J Virol ; 98(7): e0062224, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-38953377

RÉSUMÉ

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.


Sujet(s)
Adenoviridae , Virus de la peste porcine africaine , Peste porcine africaine , Vecteurs génétiques , Génotype , Vaccins antiviraux , Animaux , Virus de la peste porcine africaine/génétique , Virus de la peste porcine africaine/immunologie , Peste porcine africaine/prévention et contrôle , Peste porcine africaine/virologie , Peste porcine africaine/immunologie , Suidae , Vaccins antiviraux/immunologie , Vaccins antiviraux/génétique , Vaccins antiviraux/administration et posologie , Vecteurs génétiques/génétique , Adenoviridae/génétique , Adenoviridae/immunologie , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Vaccins sous-unitaires/immunologie , Vaccins sous-unitaires/génétique , Antigènes viraux/immunologie , Antigènes viraux/génétique
5.
Nat Commun ; 15(1): 6270, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39054318

RÉSUMÉ

The prefusion conformation of human metapneumovirus fusion protein (hMPV Pre-F) is critical for eliciting the most potent neutralizing antibodies and is the preferred immunogen for an efficacious vaccine against hMPV respiratory infections. Here we show that an additional cleavage event in the F protein allows closure and correct folding of the trimer. We therefore engineered the F protein to undergo double cleavage, which enabled screening for Pre-F stabilizing substitutions at the natively folded protomer interfaces. To identify these substitutions, we developed an AI convolutional classifier that successfully predicts complex polar interactions often overlooked by physics-based methods and visual inspection. The combination of additional processing, stabilization of interface regions and stabilization of the membrane-proximal stem, resulted in a Pre-F protein vaccine candidate without the need for a heterologous trimerization domain that exhibited high expression yields and thermostability. Cryo-EM analysis shows the complete ectodomain structure, including the stem, and a specific interaction of the newly identified cleaved C-terminus with the adjacent protomer. Importantly, the protein induces high and cross-neutralizing antibody responses resulting in near complete protection against hMPV challenge in cotton rats, making the highly stable, double-cleaved hMPV Pre-F trimer an attractive vaccine candidate.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , Metapneumovirus , Protéines de fusion virale , Vaccins antiviraux , Metapneumovirus/immunologie , Metapneumovirus/génétique , Animaux , Anticorps neutralisants/immunologie , Humains , Anticorps antiviraux/immunologie , Protéines de fusion virale/immunologie , Protéines de fusion virale/composition chimique , Protéines de fusion virale/génétique , Vaccins antiviraux/immunologie , Infections à Paramyxoviridae/prévention et contrôle , Infections à Paramyxoviridae/immunologie , Cryomicroscopie électronique , Ingénierie des protéines/méthodes , Sigmodontinae , Femelle , Multimérisation de protéines , Modèles moléculaires
6.
Front Immunol ; 15: 1408510, 2024.
Article de Anglais | MEDLINE | ID: mdl-39021566

RÉSUMÉ

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.


Sujet(s)
Infections à Herpesviridae , Herpèsvirus équin de type 1 , Vaccins atténués , Animaux , Vaccins atténués/immunologie , Infections à Herpesviridae/prévention et contrôle , Infections à Herpesviridae/immunologie , Infections à Herpesviridae/virologie , Infections à Herpesviridae/médecine vétérinaire , Herpèsvirus équin de type 1/immunologie , Herpèsvirus équin de type 1/génétique , Equus caballus , Mesocricetus , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Protéines de l'enveloppe virale/immunologie , Protéines de l'enveloppe virale/génétique , Cricetinae , Maladies des chevaux/prévention et contrôle , Maladies des chevaux/immunologie , Maladies des chevaux/virologie , Vaccins antiviraux/immunologie , Vaccins antiviraux/génétique , Lignée cellulaire , Mutation
7.
Microb Pathog ; 193: 106786, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38971506

RÉSUMÉ

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.


Sujet(s)
Virus de la maladie de Carré , Analyse de profil d'expression de gènes , Vaccins atténués , Animaux , Cellules Vero , Chlorocebus aethiops , Vaccins atténués/immunologie , Vaccins atténués/génétique , Virus de la maladie de Carré/génétique , Virus de la maladie de Carré/immunologie , Transcriptome , Transduction du signal , Biologie informatique , Séquençage nucléotidique à haut débit , Vaccins antiviraux/immunologie , Vaccins antiviraux/génétique , Cytokines/métabolisme , Cytokines/génétique , Maladie de Carré/virologie , Maladie de Carré/génétique , Maladie de Carré/immunologie , Interactions hôte-pathogène/génétique , Interactions hôte-pathogène/immunologie , Facteur de transcription NF-kappa B/métabolisme , Facteur de transcription NF-kappa B/génétique , Récepteurs de type Toll/génétique , Récepteurs de type Toll/métabolisme
8.
Biomed Res Int ; 2024: 4066641, 2024.
Article de Anglais | MEDLINE | ID: mdl-38962403

RÉSUMÉ

The zoonotic viruses pose significant threats to public health. Nipah virus (NiV) is an emerging virus transmitted from bats to humans. The NiV causes severe encephalitis and acute respiratory distress syndrome, leading to high mortality rates, with fatality rates ranging from 40% to 75%. The first emergence of the disease was found in Malaysia in 1998-1999 and later in Bangladesh, Cambodia, Timor-Leste, Indonesia, Singapore, Papua New Guinea, Vietnam, Thailand, India, and other South and Southeast Asian nations. Currently, no specific vaccines or antiviral drugs are available. The potential advantages of epitope-based vaccines include their ability to elicit specific immune responses while minimizing potential side effects. The epitopes have been identified from the conserved region of viral proteins obtained from the UniProt database. The selection of conserved epitopes involves analyzing the genetic sequences of various viral strains. The present study identified two B cell epitopes, seven cytotoxic T lymphocyte (CTL) epitopes, and seven helper T lymphocyte (HTL) epitope interactions from the NiV proteomic inventory. The antigenic and physiological properties of retrieved protein were analyzed using online servers ToxinPred, VaxiJen v2.0, and AllerTOP. The final vaccine candidate has a total combined coverage range of 80.53%. The tertiary structure of the constructed vaccine was optimized, and its stability was confirmed with the help of molecular simulation. Molecular docking was performed to check the binding affinity and binding energy of the constructed vaccine with TLR-3 and TLR-5. Codon optimization was performed in the constructed vaccine within the Escherichia coli K12 strain, to eliminate the danger of codon bias. However, these findings must require further validation to assess their effectiveness and safety. The development of vaccines and therapeutic approaches for virus infection is an ongoing area of research, and it may take time before effective interventions are available for clinical use.


Sujet(s)
Simulation numérique , Infections à hénipavirus , Virus Nipah , Virus Nipah/immunologie , Humains , Infections à hénipavirus/immunologie , Infections à hénipavirus/prévention et contrôle , Vaccins antiviraux/immunologie , Déterminants antigéniques des lymphocytes B/immunologie , Déterminants antigéniques des lymphocytes B/composition chimique , Biologie informatique/méthodes , Déterminants antigéniques des lymphocytes T/immunologie , Vaccination , Simulation de docking moléculaire , Protéines virales/immunologie , Protéines virales/composition chimique , Protéines virales/génétique , Animaux
9.
BMC Biotechnol ; 24(1): 45, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38970027

RÉSUMÉ

Marburg virus (MARV) is a highly contagious and virulent agent belonging to Filoviridae family. MARV causes severe hemorrhagic fever in humans and non-human primates. Owing to its highly virulent nature, preventive approaches are promising for its control. There is currently no approved drug or vaccine against MARV, and management mainly involves supportive care to treat symptoms and prevent complications. Our aim was to design a novel multi-epitope vaccine (MEV) against MARV using immunoinformatics studies. In this study, various proteins (VP35, VP40 and glycoprotein precursor) were used and potential epitopes were selected. CTL and HTL epitopes covered 79.44% and 70.55% of the global population, respectively. The designed MEV construct was stable and expressed in Escherichia coli (E. coli) host. The physicochemical properties were also acceptable. MARV MEV candidate could predict comprehensive immune responses such as those of humoral and cellular in silico. Additionally, efficient interaction to toll-like receptor 3 (TLR3) and its agonist (ß-defensin) was predicted. There is a need for validation of these results using further in vitro and in vivo studies.


Sujet(s)
Biologie informatique , Maladie de Marbourg , Marburgvirus , Vaccins antiviraux , Marburgvirus/immunologie , Maladie de Marbourg/prévention et contrôle , Maladie de Marbourg/immunologie , Vaccins antiviraux/immunologie , Biologie informatique/méthodes , Animaux , Humains , Déterminants antigéniques des lymphocytes T/immunologie , Déterminants antigéniques des lymphocytes T/génétique , Épitopes/immunologie , Épitopes/génétique , Épitopes/composition chimique , Escherichia coli/génétique , Escherichia coli/métabolisme ,
10.
Vet Med Sci ; 10(4): e1540, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38967727

RÉSUMÉ

BACKGROUND: This field evaluation was designed to evaluate the efficacy of a new porcine reproductive and respiratory syndrome virus-2 (PRRSV-2) modified live virus vaccine at three independent pig farms. METHODS: Three farms were selected for this study based on their respiratory disease status caused by PRRSV-2 infection in post-weaning and growing pigs. Each farm housed a total of 40, 18-day-old pigs that were randomly allocated to one of two treatment groups. Pigs were administered a 1.0 mL dose of the bivalent vaccine intramuscularly at 21 days of age in accordance with the manufacturer's recommendations, whereas unvaccinated pigs were administered a single dose of phosphate buffered saline at the same age. RESULTS: Vaccinated groups were measured and calculated significantly (p < 0.05) higher in body weight and average daily weight gain on all three farms compared with unvaccinated groups. Vaccinated groups elicited PRRS antibodies and PRRSV-2-specific interferon-γ secreting cells, which reduced the amount of PRRSV-2 genomic copies in the blood and reduced macroscopic and microscopic lung lesions severity when compared with unvaccinated groups. CONCLUSIONS: The field evaluation data demonstrated that a new PRRSV-2 modified live virus vaccine was efficacious in swine herds suffering from respiratory diseases caused by PRRSV-2 infection.


Sujet(s)
Syndrome dysgénésique et respiratoire porcin , Virus du syndrome respiratoire et reproducteur porcin , Vaccins atténués , Vaccins antiviraux , Animaux , Virus du syndrome respiratoire et reproducteur porcin/immunologie , Syndrome dysgénésique et respiratoire porcin/prévention et contrôle , Suidae , Vaccins antiviraux/immunologie , Vaccins antiviraux/administration et posologie , Vaccins atténués/administration et posologie , Vaccins atténués/immunologie , Sus scrofa , Répartition aléatoire
11.
BMC Vet Res ; 20(1): 301, 2024 Jul 06.
Article de Anglais | MEDLINE | ID: mdl-38971791

RÉSUMÉ

BACKGROUND: Foot-and-mouth disease (FMD) is a devastating disease affecting cloven-hoofed animals, that leads to significant economic losses in affected countries and regions. Currently, there is an evident inclination towards the utilization of nanoparticles as powerful platforms for innovative vaccine development. Therefore, this study developed a ferritin-based nanoparticle (FNP) vaccine that displays a neutralizing epitope of foot-and-mouth disease virus (FMDV) VP1 (aa 140-158) on the surface of FNP, and evaluated the immunogenicity and protective efficacy of these FNPs in mouse and guinea pig models to provide a strategy for developing potential FMD vaccines. RESULTS: This study expressed the recombinant proteins Hpf, HPF-NE and HPF-T34E via an E. coli expression system. The results showed that the recombinant proteins Hpf, Hpf-NE and Hpf-T34E could be effectively assembled into nanoparticles. Subsequently, we evaluated the immunogenicity of the Hpf, Hpf-NE and Hpf-T34E proteins in mice, as well as the immunogenicity and protectiveness of the Hpf-T34E protein in guinea pigs. The results of the mouse experiment showed that the immune efficacy in the Hpf-T34E group was greater than the Hpf-NE group. The results from guinea pigs immunized with Hpf-T34E showed that the immune efficacy was largely consistent with the immunogenicity of the FMD inactivated vaccine (IV) and could confer partial protection against FMDV challenge in guinea pigs. CONCLUSIONS: The Hpf-T34E nanoparticles stand out as a superior choice for a subunit vaccine candidate against FMD, offering effective protection in FMDV-infected model animals. FNP-based vaccines exhibit excellent safety and immunogenicity, thus representing a promising strategy for the continued development of highly efficient and safe FMD vaccines.


Sujet(s)
Épitopes , Ferritines , Virus de la fièvre aphteuse , Fièvre aphteuse , Nanoparticules , Vaccins antiviraux , Animaux , Cochons d'Inde , Fièvre aphteuse/prévention et contrôle , Fièvre aphteuse/immunologie , Virus de la fièvre aphteuse/immunologie , Ferritines/immunologie , Vaccins antiviraux/immunologie , Épitopes/immunologie , Souris , Femelle , Souris de lignée BALB C , Protéines recombinantes/immunologie , Protéines de capside
12.
Sci Rep ; 14(1): 16605, 2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-39026076

RÉSUMÉ

Canine distemper virus (CDV) affects many domestic and wild animals. Variations among CDV genome linages could lead to vaccination failure. To date, there are several vaccine alternatives, such as a modified live virus and a recombinant vaccine; however, most of these alternatives are based on the ancestral strain Onderstepoort, which has not been circulating for years. Vaccine failures and the need to update vaccines have been widely discussed, and the development of new vaccine candidates is necessary to reduce circulation and mortality. Current vaccination alternatives cannot be used in wildlife animals due to the lack of safety data for most of the species, in addition to the insufficient immune response against circulating strains worldwide in domestic species. Computational tools, including peptide-based therapies, have become essential for developing new-generation vaccines for diverse models. In this work, a peptide-based vaccine candidate with a peptide library derived from CDV H and F protein consensus sequences was constructed employing computational tools. The molecular docking and dynamics of the selected peptides with canine MHC-I and MHC-II and with TLR-2 and TLR-4 were evaluated. In silico safety was assayed through determination of antigenicity, allergenicity, toxicity potential, and homologous canine peptides. Additionally, in vitro safety was also evaluated through cytotoxicity in cell lines and canine peripheral blood mononuclear cells (cPBMCs) and through a hemolysis potential assay using canine red blood cells. A multiepitope CDV polypeptide was constructed, synthetized, and evaluated in silico and in vitro by employing the most promising peptides for comparison with single CDV immunogenic peptides. Our findings suggest that predicting immunogenic CDV peptides derived from most antigenic CDV proteins could aid in the development of new vaccine candidates, such as multiple single CDV peptides and multiepitope CDV polypeptides, that are safe in vitro and optimized in silico. In vivo studies are being conducted to validate potential vaccines that may be effective in preventing CDV infection in domestic and wild animals.


Sujet(s)
Virus de la maladie de Carré , Maladie de Carré , Vaccins antiviraux , Virus de la maladie de Carré/immunologie , Animaux , Chiens , Vaccins antiviraux/immunologie , Maladie de Carré/prévention et contrôle , Maladie de Carré/immunologie , Simulation de docking moléculaire , Peptides/immunologie , Peptides/composition chimique , Vaccins sous-unitaires/immunologie , Protéines de fusion virale/immunologie
13.
Virology ; 597: 110152, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38968676

RÉSUMÉ

Cobalt-porphyrin phospholipid displays recombinant protein antigens on liposome surfaces via antigen polyhistidine-tag (His-tag), and when combined with monophosphorylated lipid A and QS-21 yields the "CPQ" vaccine adjuvant system. In this proof of principle study, CPQ was used to generate vaccine prototypes that elicited antibodies for two different alphaviruses (AV). Mice were immunized with computationally designed, His-tagged, physicochemical property consensus (PCPcon) protein antigens representing the variable B-domain of the envelope protein 2 (E2) from the serotype specific Venezuelan Equine Encephalitis Virus (VEEVcon) or a broad-spectrum AV-antigen termed EVCcon. The CPQ adjuvant enhanced the antigenicity of both proteins without eliciting detectable anti-His-tag antibodies. Antibodies elicited from mice immunized with antigens admixed with CPQ showed orders-of-magnitude higher levels of antigen-specific IgG compared to alternative control adjuvants. The ELISA results correlated with antiviral activity against VEEV strain TC83 and more weakly to Chikungunya virus 118/25. Thus, display of E.coli-produced His-tagged E2 protein segments on the surface of immunogenic liposomes elicits high levels of antigen-specific and AV neutralizing antibodies in mice with vaccination, while facilitating vaccine preparation and providing dose-sparing potential.


Sujet(s)
Adjuvants immunologiques , Alphavirus , Anticorps antiviraux , Antigènes viraux , Liposomes , Protéines de l'enveloppe virale , Vaccins antiviraux , Animaux , Anticorps antiviraux/immunologie , Souris , Liposomes/immunologie , Alphavirus/immunologie , Antigènes viraux/immunologie , Protéines de l'enveloppe virale/immunologie , Vaccins antiviraux/immunologie , Vaccins antiviraux/administration et posologie , Adjuvants immunologiques/administration et posologie , Virus de l'encéphalite équine du Venezuela/immunologie , Femelle , Anticorps neutralisants/immunologie , Virus du chikungunya/immunologie , Souris de lignée BALB C , Immunoglobuline G/immunologie , Immunoglobuline G/sang
14.
Emerg Microbes Infect ; 13(1): 2373313, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38946528

RÉSUMÉ

Rift Valley fever (RVF) is a mosquito-borne zoonotic disease caused by RVF virus (RVFV). RVFV infections in humans are usually asymptomatic or associated with mild febrile illness, although more severe cases of haemorrhagic disease and encephalitis with high mortality also occur. Currently, there are no licensed human vaccines available. The safety and efficacy of a genetically engineered four-segmented RVFV variant (hRVFV-4s) as a potential live-attenuated human vaccine has been tested successfully in mice, ruminants, and marmosets though the correlates of protection of this vaccine are still largely unknown. In the present study, we have assessed hRVFV-4s-induced humoral and cellular immunity in a mouse model of RVFV infection. Our results confirm that a single dose of hRVFV-4s is highly efficient in protecting naïve mice from developing severe disease following intraperitoneal challenge with a highly virulent RVFV strain and data show that virus neutralizing (VN) serum antibody titres in a prime-boost regimen are significantly higher compared to the single dose. Subsequently, VN antibodies from prime-boost-vaccinated recipients were shown to be protective when transferred to naïve mice. In addition, hRVFV-4s vaccination induced a significant virus-specific T cell response as shown by IFN-γ ELISpot assay, though these T cells did not provide significant protection upon passive transfer to naïve recipient mice. Collectively, this study highlights hRVFV-4s-induced VN antibodies as a major correlate of protection against lethal RVFV infection.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , Fièvre de la Vallée du Rift , Virus de la fièvre de la vallée du Rift , Vaccins atténués , Vaccins antiviraux , Animaux , Virus de la fièvre de la vallée du Rift/immunologie , Virus de la fièvre de la vallée du Rift/génétique , Fièvre de la Vallée du Rift/prévention et contrôle , Fièvre de la Vallée du Rift/immunologie , Vaccins antiviraux/immunologie , Vaccins antiviraux/administration et posologie , Souris , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Anticorps neutralisants/sang , Anticorps neutralisants/immunologie , Femelle , Vaccins atténués/immunologie , Vaccins atténués/administration et posologie , Modèles animaux de maladie humaine , Immunité cellulaire , Lymphocytes T/immunologie , Immunité humorale , Souris de lignée BALB C , Interféron gamma/immunologie , Vaccination
15.
Microb Biotechnol ; 17(7): e14518, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38953907

RÉSUMÉ

Porcine epidemic diarrhoea virus (PEDV) infects pigs of all ages by invading small intestine, causing acute diarrhoea, vomiting, and dehydration with high morbidity and mortality among newborn piglets. However, current PEDV vaccines are not effective to protect the pigs from field epidemic strains because of poor mucosal immune response and strain variation. Therefore, it is indispensable to develop a novel oral vaccine based on epidemic strains. Bacillus subtilis spores are attractive delivery vehicles for oral vaccination on account of the safety, high stability, and low cost. In this study, a chimeric gene CotC-Linker-COE (CLE), comprising of the B. subtilis spore coat gene cotC fused to the core neutralizing epitope CO-26 K equivalent (COE) of the epidemic strain PEDV-AJ1102 spike protein gene, was constructed. Then recombinant B. subtilis displaying the CLE on the spore surface was developed by homologous recombination. Mice were immunized by oral route with B. subtilis 168-CLE, B. subtilis 168, or phosphate-buffered saline (PBS) as control. Results showed that the IgG antibodies and cytokine (IL-4, IFN-γ) levels in the B. subtilis 168-CLE group were significantly higher than the control groups. This study demonstrates that B. subtilis 168-CLE can generate specific systemic immune and mucosal immune responses and is a potential vaccine candidate against PEDV infection.


Sujet(s)
Anticorps antiviraux , Bacillus subtilis , Virus de la diarrhée porcine épidémique , Spores bactériens , Virus de la diarrhée porcine épidémique/génétique , Virus de la diarrhée porcine épidémique/immunologie , Animaux , Bacillus subtilis/génétique , Bacillus subtilis/immunologie , Spores bactériens/génétique , Spores bactériens/immunologie , Souris , Anticorps antiviraux/sang , Suidae , Vaccins antiviraux/immunologie , Vaccins antiviraux/génétique , Vaccins antiviraux/administration et posologie , Infections à coronavirus/médecine vétérinaire , Infections à coronavirus/prévention et contrôle , Maladies des porcs/prévention et contrôle , Maladies des porcs/virologie , Maladies des porcs/microbiologie , Maladies des porcs/immunologie , Antigènes viraux/génétique , Antigènes viraux/immunologie , Administration par voie orale , Cytokines/métabolisme , Immunoglobuline G/sang , Souris de lignée BALB C , Femelle , Techniques d'exposition à la surface cellulaire , Glycoprotéine de spicule des coronavirus/génétique , Glycoprotéine de spicule des coronavirus/immunologie
16.
Vet Microbiol ; 295: 110167, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38954881

RÉSUMÉ

Hendra virus (HeV) is lethal to horses and a zoonotic threat to humans in Australia, causing severe neurological and/or respiratory disease with high mortality. An equine vaccine has been available since 2012. Foals acquire antibodies from their dams by ingesting colostrum after parturition, therefore it is assumed that foals of mares vaccinated against HeV will have passive HeV antibodies circulating during the first several months of life until they are actively vaccinated. However, no studies have yet examined passive or active immunity against HeV in foals. Here, we investigated anti-HeV antibody levels in vaccinated mares and their foals. Testing for HeV neutralising antibodies is cumbersome due to the requirement for Biosafety level 4 (BSL-4) containment to conduct virus neutralisation tests (VNT). For this study, a subset of samples was tested for HeV G-specific antibodies by both an authentic VNT with infectious HeV and a microsphere-based immunoassay (MIA), revealing a strong correlation. An indicative neutralising level was then applied to the results of a larger sample set tested using the MIA. Mares had high levels of HeV-specific neutralising antibodies at the time of parturition. Foals acquired high levels of maternal antibodies which then waned to below predictive protective levels in most foals by 6 months old when vaccination commenced. Foals showed a suboptimal response to vaccination, suggesting maternal antibodies may interfere with active vaccination. The correlation analysis between the authentic HeV VNT and HeV MIA will enable further high throughput serological studies to inform optimal vaccination protocols for both broodmares and foals.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , Virus Hendra , Infections à hénipavirus , Maladies des chevaux , Vaccination , Vaccins antiviraux , Animaux , Equus caballus , Virus Hendra/immunologie , Maladies des chevaux/prévention et contrôle , Maladies des chevaux/virologie , Maladies des chevaux/immunologie , Anticorps antiviraux/sang , Infections à hénipavirus/prévention et contrôle , Infections à hénipavirus/médecine vétérinaire , Infections à hénipavirus/immunologie , Infections à hénipavirus/virologie , Femelle , Vaccination/médecine vétérinaire , Vaccins antiviraux/immunologie , Vaccins antiviraux/administration et posologie , Anticorps neutralisants/sang , Immunité acquise d'origine maternelle , Animaux nouveau-nés/immunologie , Grossesse , Tests de neutralisation/médecine vétérinaire , Australie , Colostrum/immunologie
17.
Methods Mol Biol ; 2824: 385-395, 2024.
Article de Anglais | MEDLINE | ID: mdl-39039425

RÉSUMÉ

Rift Valley fever (RVF) caused by Rift Valley fever virus (RVFV) is a major health concern for both domesticated animals and humans in certain endemic areas of Africa. With changing environmental conditions and identification of vectors capable of transmitting the virus, there is high risk of RVFV spreading into other parts of the world. Furthermore, unavailability of effective vaccines in the event of an outbreak can be a major challenge as witnessed recently in case of SARS-CoV2 pandemic. Hence, identifying potential vaccines and testing their protective efficacy in preclinical models before clinical testing is the absolute need of the hour. Here, we describe methods used to quantify virus-specific T cell responses in mice that were immunized with RVFV strains or antigens.


Sujet(s)
Virus de la fièvre de la vallée du Rift , Lymphocytes T , Vaccins antiviraux , Animaux , Souris , Lymphocytes T/immunologie , Virus de la fièvre de la vallée du Rift/immunologie , Vaccins antiviraux/immunologie , Fièvre de la Vallée du Rift/immunologie , Fièvre de la Vallée du Rift/prévention et contrôle , Immunisation/méthodes , Vaccination/méthodes , Antigènes viraux/immunologie
18.
Front Immunol ; 15: 1352018, 2024.
Article de Anglais | MEDLINE | ID: mdl-38989282

RÉSUMÉ

In this study, we investigated how Radix pseudostellariae polysaccharide (RPP) enhances the immune response of the inactivated porcine reproductive and respiratory syndrome virus (PRRSV) vaccine through interactions with the microbiome and metabolome. We pretreated sows with 10 mg/kg body weight of RPP via drinking water for 7 days prior to intramuscular injection of the PRRSV vaccine. This significantly increased the concentrations of PRRSV GP5 protein antibody, interleukin (IL)-2, IL-4, IL-10, and interferon (IFN)-γ. Oral administration of RPP also significantly improved the abundance of beneficial bacteria in the stool, such as Parabacteroides distasonis, Prevotella_copri, Eubacterium_sp., and Clostridium_sp._CAG:226, and decreased the levels of potentially pathogenic bacteria, such as Paraeggerthella and [Clostridium] innocuum, compared to the vaccine alone. These bacterial changes were confirmed using quantitative real-time polymerase chain reaction (Q-PCR). Moreover, RPP treatment significantly increased the blood concentrations of L-theanine, taurodeoxycholic acid (TDCA), and N-arachidonoyl proline, and decreased the levels of L-glutamine, oclacitinib, lipoxin C4, and leukotriene C5 in sows after immunization (p< 0.05). The concentrations of various blood metabolites were validated using sandwich enzyme-linked immunosorbent assay (ELISA), confirming the accuracy of the metabolomics data. Intriguingly, the integration of microbiome and metabolome analyses highlighted the significance of Prevotella_copri and TDCA. We consequently developed a mouse immunity model using GP5 protein and discovered that oral administration of RPP significantly enhanced the levels of GP5 protein antibodies, IL-2, IL-4, IL-10, and IFN-γ in mouse serum. It also increased the number of CD3+ and CD3+CD4+ cells in the spleen. Additionally, Prevotella_copri was administered into the large intestine via the anus for 7 days prior to the intramuscular injection of the PRRSV GP5 protein. The results demonstrated a significant increase in TDCA and GP5 antibody concentration in the mouse serum, indicating that RPP modulates Prevotella_copri to elevate its metabolite TDCA, thereby enhancing the GP5 antibody level. In conclusion, oral administration of 10 mg/kg RPP optimizes gut flora diversity and blood metabolites, particularly Prevotella_copri and TDCA, thereby improving the immune response to the inactivated PRRSV vaccine.


Sujet(s)
Métabolome , Polyosides , Syndrome dysgénésique et respiratoire porcin , Virus du syndrome respiratoire et reproducteur porcin , Vaccins inactivés , Vaccins antiviraux , Animaux , Suidae , Virus du syndrome respiratoire et reproducteur porcin/immunologie , Syndrome dysgénésique et respiratoire porcin/immunologie , Syndrome dysgénésique et respiratoire porcin/prévention et contrôle , Vaccins antiviraux/immunologie , Femelle , Vaccins inactivés/immunologie , Anticorps antiviraux/sang , Cytokines/métabolisme , Microbiote/effets des médicaments et des substances chimiques , Microbiote/immunologie , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Microbiome gastro-intestinal/immunologie , Adjuvants immunologiques
19.
Arch Virol ; 169(8): 167, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-39020055

RÉSUMÉ

Epstein‒Barr virus (EBV) is a double-stranded DNA virus belonging to the family Orthoherpesviridae that is associated with the development of various tumors, such as lymphoma, nasopharyngeal carcinoma, and gastric cancer. There are no uniformly effective treatments for human EBV infection, and vaccines and immunotherapies are currently the main research directions. The glycoproteins gB and gH/gL are surface glycoproteins that are common to all herpesviruses, with subtle differences in structure and function between different viruses. The core membrane fusion machinery constituted by EBV gB and gH/gL is an important target of neutralizing antibodies in epithelial EBV infection due to its essential role in the fusion of viral and target cell membranes. In this article, we review the main modes of EBV infection, the structure and function of the core fusion machinery gB and gH/gL, and the development of neutralizing antibodies and prophylactic vaccines based on this target.


Sujet(s)
Anticorps neutralisants , Infections à virus Epstein-Barr , Herpèsvirus humain de type 4 , Protéines de l'enveloppe virale , Humains , Infections à virus Epstein-Barr/prévention et contrôle , Infections à virus Epstein-Barr/immunologie , Infections à virus Epstein-Barr/virologie , Anticorps neutralisants/immunologie , Herpèsvirus humain de type 4/immunologie , Herpèsvirus humain de type 4/génétique , Protéines de l'enveloppe virale/immunologie , Protéines de l'enveloppe virale/génétique , Anticorps antiviraux/immunologie , Pénétration virale , Animaux , Vaccins antiviraux/immunologie , Protéines virales/immunologie , Protéines virales/génétique , Glycoprotéines membranaires , Chaperons moléculaires
20.
J Biomed Sci ; 31(1): 73, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39010093

RÉSUMÉ

Enteroviruses (EVs) are the most prevalent viruses in humans. EVs can cause a range of acute symptoms, from mild common colds to severe systemic infections such as meningitis, myocarditis, and flaccid paralysis. They can also lead to chronic diseases such as cardiomyopathy. Although more than 280 human EV serotypes exist, only four serotypes have licenced vaccines. No antiviral drugs are available to treat EV infections, and global surveillance of EVs has not been effectively coordinated. Therefore, poliovirus still circulates, and there have been alarming epidemics of non-polio enteroviruses. Thus, there is a pressing need for coordinated preparedness efforts against EVs.This review provides a perspective on recent enterovirus outbreaks and global poliovirus eradication efforts with continuous vaccine development initiatives. It also provides insights into the challenges and opportunities in EV vaccine development. Given that traditional whole-virus vaccine technologies are not suitable for many clinically relevant EVs and considering the ongoing risk of enterovirus outbreaks and the potential for new emerging pathogenic strains, the need for new effective and adaptable enterovirus vaccines is emphasized.This review also explores the difficulties in translating promising vaccine candidates for clinical use and summarizes information from published literature and clinical trial databases focusing on existing enterovirus vaccines, ongoing clinical trials, the obstacles faced in vaccine development as well as the emergence of new vaccine technologies. Overall, this review contributes to the understanding of enterovirus vaccines, their role in public health, and their significance as a tool for future preparedness.


Sujet(s)
Infections à entérovirus , Enterovirus , Vaccins antiviraux , Humains , Infections à entérovirus/épidémiologie , Infections à entérovirus/prévention et contrôle , Infections à entérovirus/virologie , Enterovirus/immunologie , Vaccins antiviraux/immunologie , Développement de vaccin , Épidémies de maladies/prévention et contrôle , Épidémies/prévention et contrôle
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE