Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29.653
Filtrar
1.
PLoS Negl Trop Dis ; 18(7): e0012008, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38949988

RESUMO

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.


Assuntos
Enterovirus Humano A , Doença de Mão, Pé e Boca , Vacinação , Doença de Mão, Pé e Boca/epidemiologia , Doença de Mão, Pé e Boca/prevenção & controle , Humanos , China/epidemiologia , Masculino , Feminino , Vacinação/estatística & dados numéricos , Lactente , Pré-Escolar , Enterovirus Humano A/imunologia , Incidência , Vacinas Virais/administração & dosagem , Umidade , Temperatura , Criança
2.
Biomed Res Int ; 2024: 4066641, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38962403

RESUMO

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.


Assuntos
Simulação por Computador , Infecções por Henipavirus , Vírus Nipah , Vírus Nipah/imunologia , Humanos , Infecções por Henipavirus/imunologia , Infecções por Henipavirus/prevenção & controle , Vacinas Virais/imunologia , Epitopos de Linfócito B/imunologia , Epitopos de Linfócito B/química , Biologia Computacional/métodos , Epitopos de Linfócito T/imunologia , Vacinação , Simulação de Acoplamento Molecular , Proteínas Virais/imunologia , Proteínas Virais/química , Proteínas Virais/genética , Animais
3.
Vet Med Sci ; 10(4): e1540, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38967727

RESUMO

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.


Assuntos
Síndrome Respiratória e Reprodutiva Suína , Vírus da Síndrome Respiratória e Reprodutiva Suína , Vacinas Atenuadas , Vacinas Virais , Animais , Vírus da Síndrome Respiratória e Reprodutiva Suína/imunologia , Síndrome Respiratória e Reprodutiva Suína/prevenção & controle , Suínos , Vacinas Virais/imunologia , Vacinas Virais/administração & dosagem , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/imunologia , Sus scrofa , Distribuição Aleatória
4.
Front Immunol ; 15: 1352018, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38989282

RESUMO

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.


Assuntos
Metaboloma , Polissacarídeos , Síndrome Respiratória e Reprodutiva Suína , Vírus da Síndrome Respiratória e Reprodutiva Suína , Vacinas de Produtos Inativados , Vacinas Virais , Animais , Suínos , Vírus da Síndrome Respiratória e Reprodutiva Suína/imunologia , Síndrome Respiratória e Reprodutiva Suína/imunologia , Síndrome Respiratória e Reprodutiva Suína/prevenção & controle , Vacinas Virais/imunologia , Feminino , Vacinas de Produtos Inativados/imunologia , Anticorpos Antivirais/sangue , Citocinas/metabolismo , Microbiota/efeitos dos fármacos , Microbiota/imunologia , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/imunologia , Adjuvantes Imunológicos
5.
BMC Vet Res ; 20(1): 301, 2024 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-38971791

RESUMO

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.


Assuntos
Epitopos , Ferritinas , Vírus da Febre Aftosa , Febre Aftosa , Nanopartículas , Vacinas Virais , Animais , Cobaias , Febre Aftosa/prevenção & controle , Febre Aftosa/imunologia , Vírus da Febre Aftosa/imunologia , Ferritinas/imunologia , Vacinas Virais/imunologia , Epitopos/imunologia , Camundongos , Feminino , Camundongos Endogâmicos BALB C , Proteínas Recombinantes/imunologia , Proteínas do Capsídeo
7.
Sheng Wu Gong Cheng Xue Bao ; 40(7): 2150-2161, 2024 Jul 25.
Artigo em Chinês | MEDLINE | ID: mdl-39044581

RESUMO

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.


Assuntos
Anticorpos Antivirais , Infecções por Coronavirus , Vírus da Diarreia Epidêmica Suína , Proteínas Recombinantes de Fusão , Vacinas Virais , Animais , Vírus da Diarreia Epidêmica Suína/imunologia , Vírus da Diarreia Epidêmica Suína/genética , Camundongos , Suínos , Vacinas Virais/imunologia , Vacinas Virais/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/genética , Infecções por Coronavirus/prevenção & controle , Infecções por Coronavirus/imunologia , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Deltacoronavirus/imunologia , Deltacoronavirus/genética , Doenças dos Suínos/prevenção & controle , Doenças dos Suínos/imunologia , Vacinas de Subunidades Antigênicas/imunologia , Vacinas de Subunidades Antigênicas/genética , Camundongos Endogâmicos BALB C , Feminino , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Domínios Proteicos , Imunogenicidade da Vacina , Imunidade Humoral
8.
Elife ; 122024 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-39046448

RESUMO

Canine distemper virus (CDV) belongs to morbillivirus, including measles virus (MeV) and rinderpest virus, which causes serious immunological and neurological disorders in carnivores, including dogs and rhesus monkeys, as recently reported, but their vaccines are highly effective. The attachment glycoprotein hemagglutinin (CDV-H) at the CDV surface utilizes signaling lymphocyte activation molecule (SLAM) and Nectin-4 (also called poliovirus-receptor-like-4; PVRL4) as entry receptors. Although fusion models have been proposed, the molecular mechanism of morbillivirus fusion entry is poorly understood. Here, we determined the crystal structure of the globular head domain of CDV-H vaccine strain at 3.2 Å resolution, revealing that CDV-H exhibits a highly tilted homodimeric form with a six-bladed ß-propeller fold. While the predicted Nectin-4-binding site is well conserved with that of MeV-H, that of SLAM is similar but partially different, which is expected to contribute to host specificity. Five N-linked sugars covered a broad area of the CDV-H surface to expose receptor-binding sites only, supporting the effective production of neutralizing antibodies. These features are common to MeV-H, although the glycosylation sites are completely different. Furthermore, real-time observation using high-speed atomic force microscopy revealed highly mobile features of the CDV-H dimeric head via the connector region. These results suggest that sugar-shielded tilted homodimeric structure and dynamic conformational changes are common characteristics of morbilliviruses and ensure effective fusion entry and vaccination.


Assuntos
Vírus da Cinomose Canina , Polissacarídeos , Internalização do Vírus , Vírus da Cinomose Canina/química , Vírus da Cinomose Canina/imunologia , Animais , Polissacarídeos/química , Polissacarídeos/metabolismo , Cães , Cinomose/virologia , Cinomose/prevenção & controle , Cristalografia por Raios X , Hemaglutininas Virais/química , Hemaglutininas Virais/metabolismo , Multimerização Proteica , Vacinação , Conformação Proteica , Vacinas Virais/imunologia , Vacinas Virais/química , Receptores Virais/metabolismo , Receptores Virais/química , Modelos Moleculares
9.
Sci Rep ; 14(1): 16798, 2024 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-39039173

RESUMO

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.


Assuntos
Proteínas do Capsídeo , Herpesvirus Humano 4 , Herpesvirus Humano 4/imunologia , Humanos , Proteínas do Capsídeo/imunologia , Proteínas do Capsídeo/química , Infecções por Vírus Epstein-Barr/imunologia , Infecções por Vírus Epstein-Barr/prevenção & controle , Epitopos de Linfócito B/imunologia , Biologia Computacional/métodos , Epitopos de Linfócito T/imunologia , Vacinas Virais/imunologia , Antígenos Virais/imunologia , Antígenos Virais/química , Modelos Moleculares , Simulação de Acoplamento Molecular
10.
PLoS One ; 19(7): e0305417, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39042625

RESUMO

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.


Assuntos
Orthohantavírus , Vacinas Virais , Orthohantavírus/imunologia , Vacinas Virais/imunologia , Humanos , Vacinologia/métodos , Simulação de Acoplamento Molecular , Simulação por Computador , Epitopos/imunologia , Epitopos/química , Modelos Moleculares , Infecções por Hantavirus/prevenção & controle , Infecções por Hantavirus/imunologia
11.
Methods Mol Biol ; 2824: 385-395, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39039425

RESUMO

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.


Assuntos
Vírus da Febre do Vale do Rift , Linfócitos T , Vacinas Virais , Animais , Camundongos , Linfócitos T/imunologia , Vírus da Febre do Vale do Rift/imunologia , Vacinas Virais/imunologia , Febre do Vale de Rift/imunologia , Febre do Vale de Rift/prevenção & controle , Imunização/métodos , Vacinação/métodos , Antígenos Virais/imunologia
12.
Microb Biotechnol ; 17(7): e14518, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38953907

RESUMO

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.


Assuntos
Anticorpos Antivirais , Bacillus subtilis , Vírus da Diarreia Epidêmica Suína , Esporos Bacterianos , Vírus da Diarreia Epidêmica Suína/genética , Vírus da Diarreia Epidêmica Suína/imunologia , Animais , Bacillus subtilis/genética , Bacillus subtilis/imunologia , Esporos Bacterianos/genética , Esporos Bacterianos/imunologia , Camundongos , Anticorpos Antivirais/sangue , Suínos , Vacinas Virais/imunologia , Vacinas Virais/genética , Vacinas Virais/administração & dosagem , Infecções por Coronavirus/veterinária , Infecções por Coronavirus/prevenção & controle , Doenças dos Suínos/prevenção & controle , Doenças dos Suínos/virologia , Doenças dos Suínos/microbiologia , Doenças dos Suínos/imunologia , Antígenos Virais/genética , Antígenos Virais/imunologia , Administração Oral , Citocinas/metabolismo , Imunoglobulina G/sangue , Camundongos Endogâmicos BALB C , Feminino , Técnicas de Visualização da Superfície Celular , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/imunologia
13.
Vet Microbiol ; 295: 110167, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38954881

RESUMO

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.


Assuntos
Anticorpos Neutralizantes , Anticorpos Antivirais , Vírus Hendra , Infecções por Henipavirus , Doenças dos Cavalos , Vacinação , Vacinas Virais , Animais , Cavalos , Vírus Hendra/imunologia , Doenças dos Cavalos/prevenção & controle , Doenças dos Cavalos/virologia , Doenças dos Cavalos/imunologia , Anticorpos Antivirais/sangue , Infecções por Henipavirus/prevenção & controle , Infecções por Henipavirus/veterinária , Infecções por Henipavirus/imunologia , Infecções por Henipavirus/virologia , Feminino , Vacinação/veterinária , Vacinas Virais/imunologia , Vacinas Virais/administração & dosagem , Anticorpos Neutralizantes/sangue , Imunidade Materno-Adquirida , Animais Recém-Nascidos/imunologia , Gravidez , Testes de Neutralização/veterinária , Austrália , Colostro/imunologia
14.
Emerg Microbes Infect ; 13(1): 2373313, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38946528

RESUMO

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.


Assuntos
Anticorpos Neutralizantes , Anticorpos Antivirais , Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Vacinas Atenuadas , Vacinas Virais , Animais , Vírus da Febre do Vale do Rift/imunologia , Vírus da Febre do Vale do Rift/genética , Febre do Vale de Rift/prevenção & controle , Febre do Vale de Rift/imunologia , Vacinas Virais/imunologia , Vacinas Virais/administração & dosagem , Camundongos , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Feminino , Vacinas Atenuadas/imunologia , Vacinas Atenuadas/administração & dosagem , Modelos Animais de Doenças , Imunidade Celular , Linfócitos T/imunologia , Imunidade Humoral , Camundongos Endogâmicos BALB C , Interferon gama/imunologia , Vacinação
15.
BMC Biotechnol ; 24(1): 45, 2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-38970027

RESUMO

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.


Assuntos
Biologia Computacional , Doença do Vírus de Marburg , Marburgvirus , Vacinas Virais , Marburgvirus/imunologia , Doença do Vírus de Marburg/prevenção & controle , Doença do Vírus de Marburg/imunologia , Vacinas Virais/imunologia , Biologia Computacional/métodos , Animais , Humanos , Epitopos de Linfócito T/imunologia , Epitopos de Linfócito T/genética , Epitopos/imunologia , Epitopos/genética , Epitopos/química , Escherichia coli/genética , Escherichia coli/metabolismo , Imunoinformática
16.
BMC Vet Res ; 20(1): 313, 2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-39010130

RESUMO

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.


Assuntos
Fazendeiros , Febre Aftosa , Vacinas Virais , Etiópia , Fazendeiros/psicologia , Febre Aftosa/prevenção & controle , Febre Aftosa/economia , Animais , Estudos Transversais , Vacinas Virais/economia , Humanos , Inquéritos e Questionários , Masculino , Feminino , Adulto , Pessoa de Meia-Idade , Bovinos , Vacinação/veterinária , Vacinação/economia
17.
Virology ; 597: 110152, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38968676

RESUMO

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.


Assuntos
Adjuvantes Imunológicos , Alphavirus , Anticorpos Antivirais , Antígenos Virais , Lipossomos , Proteínas do Envelope Viral , Vacinas Virais , Animais , Anticorpos Antivirais/imunologia , Camundongos , Lipossomos/imunologia , Alphavirus/imunologia , Antígenos Virais/imunologia , Proteínas do Envelope Viral/imunologia , Vacinas Virais/imunologia , Vacinas Virais/administração & dosagem , Adjuvantes Imunológicos/administração & dosagem , Vírus da Encefalite Equina Venezuelana/imunologia , Feminino , Anticorpos Neutralizantes/imunologia , Vírus Chikungunya/imunologia , Camundongos Endogâmicos BALB C , Imunoglobulina G/imunologia , Imunoglobulina G/sangue
18.
J Virol ; 98(7): e0062224, 2024 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-38953377

RESUMO

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.


Assuntos
Adenoviridae , Vírus da Febre Suína Africana , Febre Suína Africana , Vetores Genéticos , Genótipo , Vacinas Virais , Animais , Vírus da Febre Suína Africana/genética , Vírus da Febre Suína Africana/imunologia , Febre Suína Africana/prevenção & controle , Febre Suína Africana/virologia , Febre Suína Africana/imunologia , Suínos , Vacinas Virais/imunologia , Vacinas Virais/genética , Vacinas Virais/administração & dosagem , Vetores Genéticos/genética , Adenoviridae/genética , Adenoviridae/imunologia , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Vacinas de Subunidades Antigênicas/imunologia , Vacinas de Subunidades Antigênicas/genética , Antígenos Virais/imunologia , Antígenos Virais/genética
19.
Microb Pathog ; 193: 106786, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38971506

RESUMO

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.


Assuntos
Vírus da Cinomose Canina , Perfilação da Expressão Gênica , Vacinas Atenuadas , Animais , Células Vero , Chlorocebus aethiops , Vacinas Atenuadas/imunologia , Vacinas Atenuadas/genética , Vírus da Cinomose Canina/genética , Vírus da Cinomose Canina/imunologia , Transcriptoma , Transdução de Sinais , Biologia Computacional , Sequenciamento de Nucleotídeos em Larga Escala , Vacinas Virais/imunologia , Vacinas Virais/genética , Citocinas/metabolismo , Citocinas/genética , Cinomose/virologia , Cinomose/genética , Cinomose/imunologia , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , NF-kappa B/metabolismo , NF-kappa B/genética , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
20.
Arch Virol ; 169(8): 167, 2024 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-39020055

RESUMO

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.


Assuntos
Anticorpos Neutralizantes , Infecções por Vírus Epstein-Barr , Herpesvirus Humano 4 , Proteínas do Envelope Viral , Humanos , Infecções por Vírus Epstein-Barr/prevenção & controle , Infecções por Vírus Epstein-Barr/imunologia , Infecções por Vírus Epstein-Barr/virologia , Anticorpos Neutralizantes/imunologia , Herpesvirus Humano 4/imunologia , Herpesvirus Humano 4/genética , Proteínas do Envelope Viral/imunologia , Proteínas do Envelope Viral/genética , Anticorpos Antivirais/imunologia , Internalização do Vírus , Animais , Vacinas Virais/imunologia , Proteínas Virais/imunologia , Proteínas Virais/genética , Glicoproteínas de Membrana , Chaperonas Moleculares
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA