Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 29.669
Filtrer
1.
Front Immunol ; 15: 1440407, 2024.
Article de Anglais | MEDLINE | ID: mdl-39072326

RÉSUMÉ

Introduction: Bluetongue (BT), caused by bluetongue virus (BTV), is an important arthropod-borne livestock disease listed by the World Organization for Animal Health. Live-attenuated and inactivated vaccines have permitted to control BT but they do not simultaneously protect against the myriad of BTV serotypes. Recently, we identified the highly conserved BTV nonstructural protein NS1 and the N-terminal region of NS2 as antigens capable of conferring multiserotype protection against BTV. Methods: Here, we designed Modified Vaccinia Ankara (MVA) viral vectors that expressed BTV-4 proteins VP2 or VP7 along with NS1 and NS2-Nt as well as MVAs that expressed proteins VP2, VP7 or NS1 and NS2-Nt. Results: Immunization of IFNAR(-/-) mice with two doses of MVA-NS1-2A-NS2-Nt protected mice from BTV-4M infection by the induction of an antigen-specific T cell immune response. Despite rMVA expressing VP7 alone were not protective in the IFNAR(-/-) mouse model, inclusion of VP7 in the vaccine formulation amplified the cell-mediated response induced by NS1 and NS2-Nt. Expression of VP2 elicited protective non-cross-reactive neutralizing antibodies (nAbs) in immunized animals and improved the protection observed in the MVA-NS1-2A-NS2-Nt immunized mice when these three BTV antigens were co-expressed. Moreover, vaccines candidates co-expressing VP2 or VP7 along with NS1 and NS2-Nt provided multiserotype protection. We assessed protective efficacy of both vaccine candidates in sheep against virulent challenge with BTV-4M. Discussion: Immunization with MVA-VP7-NS1-2A-NS2-Nt partially dumped viral replication and clinical disease whereas administration of MVA-VP2-NS1-2A-NS2-Nt promoted a complete protection, preventing viraemia and the pathology produced by BTV infection.


Sujet(s)
Virus de la langue bleue , Fièvre catarrhale du mouton , Protéines de capside , Vecteurs génétiques , Récepteur à l'interféron alpha-bêta , Virus de la vaccine , Protéines virales non structurales , Vaccins antiviraux , Animaux , Virus de la langue bleue/immunologie , Virus de la langue bleue/génétique , Protéines virales non structurales/immunologie , Protéines virales non structurales/génétique , Fièvre catarrhale du mouton/prévention et contrôle , Fièvre catarrhale du mouton/immunologie , Fièvre catarrhale du mouton/virologie , Souris , Vaccins antiviraux/immunologie , Vaccins antiviraux/génétique , Virus de la vaccine/génétique , Virus de la vaccine/immunologie , Récepteur à l'interféron alpha-bêta/génétique , Protéines de capside/immunologie , Protéines de capside/génétique , Souris knockout , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Anticorps neutralisants/immunologie , Anticorps neutralisants/sang , Femelle
2.
J Med Virol ; 96(8): e29831, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39072815

RÉSUMÉ

Coxsackievirus B3 (CVB3) triggers viral myocarditis, with no effective vaccine yet. This fecal-oral transmitted pathogen has prompted interest in mucosal immunization strategies to impede CVB3 spread. We developed a new attenuated vaccine strain, named CVB3(mu). The potential of CVB3(mu) to stimulate mucosal immune protection remains to be elucidated. This study evaluates the attenuation characteristics of CVB3(mu) via a rapid evolution cellular model and RNA sequencing. Its temperature sensitivity and safety were evaluated through in vitro and in vivo experiments. The mucosal immunity protection of CVB3(mu) was assessed via intranasal immunization in Balb/c mice. The results indicate that CVB3(mu) exhibits temperature sensitivity and forms smaller plaques. It sustains fewer genetic mutations and still possesses certain attenuated traits up to the 25th passage, in comparison to CVB3(WT). Intranasal immunization elicited a significant serum neutralizing antibodies, and a substantial sIgA response in nasal washes. In vivo trials revealed CVB3(mu) protection in adult mice and passive protection in suckling mice against lethal CVB3(WT) challenges. In conclusion, CVB3(mu), a live attenuated intranasal vaccine, provides protection involving humoral and mucosal immunity, making it a promising candidate to control CVB3 spread and infection.


Sujet(s)
Administration par voie nasale , Anticorps neutralisants , Anticorps antiviraux , Infections à virus coxsackie , Entérovirus humain B , Immunité muqueuse , Souris de lignée BALB C , Vaccins atténués , Vaccins antiviraux , Animaux , Entérovirus humain B/immunologie , Vaccins atténués/immunologie , Vaccins atténués/administration et posologie , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Anticorps neutralisants/sang , Anticorps neutralisants/immunologie , Infections à virus coxsackie/immunologie , Infections à virus coxsackie/prévention et contrôle , Vaccins antiviraux/immunologie , Vaccins antiviraux/administration et posologie , Souris , Immunoglobuline A sécrétoire/immunologie , Humains , Femelle , Modèles animaux de maladie humaine
3.
Viruses ; 16(7)2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-39066282

RÉSUMÉ

Chimeric marker vaccine candidates, vGPE-/PAPeV Erns and vGPE-/PhoPeV Erns, have been generated and their efficacy and capability to differentiate infected from vaccinated animals were confirmed in previous studies. The safety profile of the two chimeric marker vaccine candidates, particularly in the potential reversion to virulence, was evaluated. Each virus was administered to pigs with a dose equivalent to the vaccination dose, and pooled tonsil homogenates were subsequently inoculated into further pigs. Chimeric virus vGPE-/PAPeV Erns displayed the most substantial attenuation, achieving this within only two passages, whereas vGPE-/PhoPeV Erns was detectable until the third passage and disappeared entirely by the fourth passage. The vGPE- strain, assessed alongside, consistently exhibited stable virus recovery across each passage without any signs of increased virulence in pigs. In vitro assays revealed that the type I interferon-inducing capacity of vGPE-/PAPeV Erns was significantly higher than that of vGPE-/PhoPeV Erns and vGPE-. In conclusion, the safety profile of the two chimeric marker vaccine candidates was affirmed. Further research is essential to ensure the stability of their attenuation and safety in diverse pig populations.


Sujet(s)
Virus de la peste porcine classique , Peste porcine classique , Vaccins atténués , Vaccins antiviraux , Animaux , Suidae , Virulence , Peste porcine classique/prévention et contrôle , Peste porcine classique/virologie , Peste porcine classique/immunologie , Vaccins antiviraux/immunologie , Vaccins antiviraux/effets indésirables , Vaccins antiviraux/administration et posologie , Virus de la peste porcine classique/immunologie , Virus de la peste porcine classique/génétique , Virus de la peste porcine classique/pathogénicité , Vaccins atténués/immunologie , Vaccins atténués/effets indésirables , Vaccins atténués/administration et posologie , Vaccins marqueurs/immunologie , Vaccins marqueurs/génétique , Vaccins marqueurs/administration et posologie , Vaccination
4.
Viruses ; 16(7)2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-39066240

RÉSUMÉ

Morbillivirus canis (canine distemper virus (CDV)) is recognized as a multihost pathogen responsible for a transmissible disease affecting both domestic and wild animals. A considerable portion of wildlife populations remain unvaccinated due to a lack of safety and immunogenicity data on existing vaccines for the prevention of CDV infection in these species. This review aimed to assess the current state of CDV vaccination research for both domestic and wild animals and to explore novel vaccine candidates through in vivo studies. It also sought to synthesize the scattered information from the extensive scientific literature on CDV vaccine research, identify key researchers in the field, and highlight areas where research on CDV vaccination is lacking. A scoping review was conducted across four databases following the PRISMA-ScR protocol, with information analyzed using absolute and relative frequencies and 95% confidence intervals (CIs) for study number proportions. Among the 2321 articles retrieved, 68 met the inclusion criteria and focused on CDV vaccines in various animal species, such as dogs, ferrets, minks, and mice. Most of the scientific community involved in this research was in the USA, Canada, France, and Denmark. Various vaccine types, including MLV CDV, recombinant virus, DNA plasmids, inactivated CDV, and MLV measles virus (MeV), were identified, along with diverse immunization routes and schedules employed in experimental and commercial vaccines. Safety and efficacy data were summarized. Notably, 37 studies reported postimmunization CDV challenge, primarily in dogs, revealing the survival rates of vaccinated animals. In summary, CDV vaccines generally demonstrate an acceptable safety profile in dogs and show promise as a means of controlling CDV. However, significant gaps in vaccine research persist, particularly concerning wildlife reservoirs, indicating the need for further investigation.


Sujet(s)
Animaux domestiques , Animaux sauvages , Virus de la maladie de Carré , Maladie de Carré , Vaccination , Vaccins antiviraux , Animaux , Animaux sauvages/virologie , Virus de la maladie de Carré/immunologie , Virus de la maladie de Carré/génétique , Vaccins antiviraux/immunologie , Vaccins antiviraux/effets indésirables , Vaccins antiviraux/administration et posologie , Maladie de Carré/prévention et contrôle , Maladie de Carré/immunologie , Maladie de Carré/virologie , Vaccination/médecine vétérinaire , Chiens , Furets , Souris , Immunogénicité des vaccins , Visons/virologie , Visons/immunologie
5.
Viruses ; 16(7)2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-39066269

RÉSUMÉ

In addition to the rabies virus (RABV), 16 more lyssavirus species have been identified worldwide, causing a disease similar to RABV. Non-rabies-related human deaths have been described, but the number of cases is unknown, and the potential of such lyssaviruses causing human disease is unpredictable. The current rabies vaccine does not protect against divergent lyssaviruses such as Mokola virus (MOKV) or Lagos bat virus (LBV). Thus, a more broad pan-lyssavirus vaccine is needed. Here, we evaluate a novel lyssavirus vaccine with an attenuated RABV vector harboring a chimeric RABV glycoprotein (G) in which the antigenic site I of MOKV replaces the authentic site of rabies virus (RABVG-cAS1). The recombinant vaccine was utilized to immunize mice and analyze the immune response compared to homologous vaccines. Our findings indicate that the vaccine RABVG-cAS1 was immunogenic and induced high antibody titers against both RABVG and MOKVG. Challenge studies with different lyssaviruses showed that replacing a single antigenic site of RABV G with the corresponding site of MOKV G provides a significant improvement over the homologous RABV vaccine and protects against RABV, Irkut virus (IRKV), and MOKV. This strategy of epitope chimerization paves the way towards a pan-lyssavirus vaccine to safely combat the diseases caused by these viruses.


Sujet(s)
Anticorps antiviraux , Lyssavirus , Vaccins antirabiques , Virus de la rage , Rage (maladie) , Animaux , Lyssavirus/immunologie , Lyssavirus/génétique , Souris , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Virus de la rage/immunologie , Virus de la rage/génétique , Vaccins antirabiques/immunologie , Vaccins antirabiques/administration et posologie , Rage (maladie)/prévention et contrôle , Rage (maladie)/immunologie , Rage (maladie)/virologie , Infections à Rhabdoviridae/prévention et contrôle , Infections à Rhabdoviridae/immunologie , Infections à Rhabdoviridae/médecine vétérinaire , Infections à Rhabdoviridae/virologie , Vaccins synthétiques/immunologie , Vaccins synthétiques/administration et posologie , Femelle , Vaccins antiviraux/immunologie , Glycoprotéines/immunologie , Glycoprotéines/génétique , Anticorps neutralisants/immunologie , Anticorps neutralisants/sang , Développement de vaccin , Humains , Antigènes viraux/immunologie , Souris de lignée BALB C
6.
Viruses ; 16(7)2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-39066162

RÉSUMÉ

Rift Valley fever (RVF) is a mosquito-borne zoonotic viral disease endemic to Africa and the Middle East. Live-attenuated RVF vaccines have been studied for both veterinary and human use due to their strong immunogenicity and cost-effective manufacturing. The live-attenuated MP-12 vaccine has been conditionally approved for veterinary use in the U.S.A., and next-generation live-attenuated RVF vaccine candidates are being actively researched. Assessing the virulence phenotype of vaccine seeds or lots is crucial for managing vaccine safety. Previously, preweaning 19-day-old outbred CD1 mice have been used to evaluate the MP-12 strain. This study aimed to characterize the relative virulence of three live-attenuated RVF vaccine strains in 19-day-old inbred C57BL/6 mice: the recombinant MP-12 (rMP-12), the RVax-1, and the ∆NSs-∆NSm-rZH501 strains. Although this mouse model did not show dose-dependent pathogenesis, mice that succumbed to the infection exhibited distinct brain pathology. Mice infected with ∆NSs-∆NSm-rZH501 showed an infiltration of inflammatory cells associated with infected neurons, and focal lesions formed around virus-infected cells. In contrast, mice infected with rMP-12 or RVax-1 showed a minimal association of inflammatory cells in the brain, yet the virus spread diffusely. The preweaning model is likely useful for evaluating host responses to attenuated RVFV strains, although further refinement may be necessary to quantitate the virulence among different RVFV strains or vaccine lots.


Sujet(s)
Modèles animaux de maladie humaine , Souris de lignée C57BL , 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/pathogénicité , 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/virologie , Fièvre de la Vallée du Rift/anatomopathologie , Fièvre de la Vallée du Rift/prévention et contrôle , Fièvre de la Vallée du Rift/immunologie , Souris , Vaccins atténués/immunologie , Vaccins atténués/administration et posologie , Vaccins antiviraux/immunologie , Vaccins antiviraux/administration et posologie , Virulence , Femelle
7.
Viruses ; 16(7)2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-39066207

RÉSUMÉ

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.


Sujet(s)
Anticorps antiviraux , Virus de la peste porcine classique , Peste porcine classique , Vaccins atténués , Vaccins antiviraux , Animaux , Peste porcine classique/prévention et contrôle , Peste porcine classique/virologie , Peste porcine classique/immunologie , Suidae , Femelle , Vaccins atténués/immunologie , Vaccins atténués/administration et posologie , Vaccins atténués/génétique , Vaccins atténués/effets indésirables , Virus de la peste porcine classique/immunologie , Virus de la peste porcine classique/génétique , Vaccins antiviraux/immunologie , Vaccins antiviraux/génétique , Vaccins antiviraux/administration et posologie , Vaccins antiviraux/effets indésirables , Grossesse , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Vaccins marqueurs/immunologie , Vaccins marqueurs/administration et posologie , Vaccins marqueurs/génétique , Vaccination/médecine vétérinaire , Anticorps neutralisants/sang , Anticorps neutralisants/immunologie
8.
Vet Immunol Immunopathol ; 274: 110803, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39003921

RÉSUMÉ

Vaccination has proven to be an effective means of controlling pathogens in animals. Since the introduction of veterinary vaccines in the 19th century, several generations of vaccines have been introduced. These vaccines have had a positive impact on global animal health and production. Despite, the success of veterinary vaccines, there are still some pathogens for which there are no effective vaccines available, such as African swine fever. Further, animal health is under the constant threat of emerging and re-emerging pathogens, some of which are zoonotic and can pose a threat to human health. The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has highlighted the need for new vaccine platforms that are safe and efficacious, but also importantly, are adaptable and can be modified rapidly to match the circulating pathogens. mRNA vaccines have been shown to be an effective vaccine platform against various viral and bacterial pathogens. This review will cover some of the recent advances in the field of mRNA vaccines for veterinary species. Moreover, various mRNA vaccines and their delivery methods, as well as their reported efficacy, will be discussed. Current limitations and future prospects of this vaccine platform in veterinary medicine will also be discussed.


Sujet(s)
Vaccins à ARNm , Animaux , Vaccins synthétiques/immunologie , Vaccins antiviraux/immunologie , Vaccination/médecine vétérinaire , ARN messager/génétique
9.
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
10.
PLoS Negl Trop Dis ; 18(7): e0012008, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38949988

RÉSUMÉ

BACKGROUND: Hand, foot, and mouth disease (HFMD) is a major public health issue in China while temperature and humidity are well-documented predictors. However, evidence on the combined effect of temperature and humidity is still limited. It also remains unclear whether such an effect could be modified by the enterovirus 71 (EV71) vaccination. METHODS: Based on 320,042 reported HFMD cases during the summer months between 2012 and 2019, we conducted a study utilizing Distributed Lag Non-Linear Models (DLNM) and time-varying DLNM to examine how China's HFMD EV71 vaccine strategy would affect the correlation between meteorological conditions and HFMD risk. RESULTS: The incidence of HFMD changed with the Discomfort Index in an arm-shaped form. The 14-day cumulative risk of HFMD exhibited a statistically significant increase during the period of 2017-2019 (following the implementation of the EV71 vaccine policy) compared to 2012-2016 (prior to the vaccine implementation). For the total population, the range of relative risk (RR) values for HFMD at the 75th, 90th, and 99th percentiles increased from 1.082-1.303 in 2012-2016 to 1.836-2.022 in 2017-2019. In the stratified analyses, Han Chinese areas show stronger relative growth, with RR values at the 75th, 90th, and 99th percentiles increased by 14.3%, 39.1%, and 134.4% post-vaccination, compared to increases of 22.7%, 41.6%, and 38.8% in minority areas. Similarly, boys showed greater increases (24.4%, 47.7%, 121.5%) compared to girls (8.1%, 28.1%, 58.3%). Additionally, the central Guizhou urban agglomeration displayed a tendency for stronger relative growth compared to other counties. CONCLUSIONS: Although the EV71 vaccine policy has been implemented, it hasn't effectively controlled the overall risk of HFMD. There's been a shift in the main viral subtypes, potentially altering population susceptibility and influencing HFMD occurrences. The modulating effects of vaccine intervention may also be influenced by factors such as race, sex, and economic level.


Sujet(s)
Entérovirus humain A , Syndrome mains-pieds-bouche , Vaccination , Syndrome mains-pieds-bouche/épidémiologie , Syndrome mains-pieds-bouche/prévention et contrôle , Humains , Chine/épidémiologie , Mâle , Femelle , Vaccination/statistiques et données numériques , Nourrisson , Enfant d'âge préscolaire , Entérovirus humain A/immunologie , Incidence , Vaccins antiviraux/administration et posologie , Humidité , Température , Enfant
12.
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
13.
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
14.
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 ,
15.
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
16.
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
18.
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
19.
BMC Vet Res ; 20(1): 313, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39010130

RÉSUMÉ

BACKGROUND: Foot and mouth disease is a contagious, transboundary, and economically devastating viral disease of cloven-hoofed animals. The disease can cause many consequences, including decreased productivity, limited market access, and elimination of flocks or herds. This study aimed to assess farmers' willingness to pay (WTP) for foot and mouth disease (FMD) vaccines and identify factors influencing their WTP. A cross-sectional questionnaire survey was conducted on 396 randomly selected livestock-owning farmers from three districts in the central Oromia region (Ambo, Dendi, and Holeta districts. The study utilized the contingent valuation method, specifically employing dichotomous choice bids with double bounds, to evaluate the willingness to pay (WTP) for the FMD vaccine. Mean WTP was assessed using interval regression, and influential factors were identified. RESULTS: The study revealed that the farmer's mean willingness to pay for a hypothetical foot and mouth disease vaccine was 37.5 Ethiopian Birr (ETB) [95% confidence interval [CI]: 34.5 40.58] in all data, while it was 23.84 (95% CI: 21.47-26.28) in the mixed farming system and 64.87 Ethiopian Birr (95% CI: 58.68 71.15) in the market-oriented farming system. We identified main livelihood, management system, sales income, breed, keeping animals for profit, and foot and mouth disease impact perception score as significant variables (p ≤ 0.05) determining the farmers' WTP for the FMD vaccine. CONCLUSION: Farmers demonstrated a high computed willingness to pay, which can be considered an advantage in the foot and mouth disease vaccination program in central Oromia. Therefore, it is necessary to ensure sufficient vaccine supply services to meet the high demand revealed.


Sujet(s)
Agriculteurs , Fièvre aphteuse , Vaccins antiviraux , Éthiopie , Agriculteurs/psychologie , Fièvre aphteuse/prévention et contrôle , Fièvre aphteuse/économie , Animaux , Études transversales , Vaccins antiviraux/économie , Humains , Enquêtes et questionnaires , Mâle , Femelle , Adulte , Adulte d'âge moyen , Bovins , Vaccination/médecine vétérinaire , Vaccination/économie
20.
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
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE