Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Virol J ; 20(1): 99, 2023 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-37226231

RESUMO

Several approaches have been developed to analyze the entry of highly pathogenic viruses. In this study, we report the implementation of a Bimolecular Multicellular Complementation (BiMuC) assay to safely and efficiently monitor SARS-CoV-2 S-mediated membrane fusion without the need for microscopy-based equipment. Using BiMuC, we screened a library of approved drugs and identified compounds that enhance S protein-mediated cell-cell membrane fusion. Among them, ethynylestradiol promotes the growth of SARS-CoV-2 and Influenza A virus in vitro. Our findings demonstrate the potential of BiMuC for identifying small molecules that modulate the life cycle of enveloped viruses, including SARS-CoV-2.


Assuntos
COVID-19 , Humanos , SARS-CoV-2 , Internalização do Vírus , Bioensaio , Biblioteca Gênica
2.
Mol Pharm ; 17(5): 1608-1620, 2020 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-32233501

RESUMO

This work analyzes the immunogenicity of six genetically engineered constructs based on elastin-like recombinamers (ELRs) fused to the Gn glycoprotein from Rift Valley fever virus (RVFV). Upon transfection, all constructs showed no effect on cell viability. While fusion constructs including ELR blocks containing hydrophobic amino acids (alanine or isoleucine) did not increase the expression of viral Gn in eukaryotic cells, glutamic acid- or valine-rich fusion proteins showed enhanced expression levels compared with the constructs encoding the viral antigen alone. However, in vivo DNA plasmid immunization assays determined that the more hydrophobic constructs reduced viremia levels after RVFV challenge to a higher extent than glutamic- or valine-rich encoding plasmids and were better inducers of cellular immunity as judged by in vitro restimulation experiments. Although the Gn-ELR fusion constructs did not surpass the protective efficacy of a plasmid vaccine expressing nonfused Gn, our results warrant further experiments directed to take advantage of the immunomodulatory potential of ELR biomaterials for improving vaccines against infectious diseases.


Assuntos
Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Doenças dos Ovinos , Vacinas de DNA , Vacinas Virais , Animais , Anticorpos Antivirais , Elastina/genética , Febre do Vale de Rift/prevenção & controle , Vírus da Febre do Vale do Rift/genética , Vírus da Febre do Vale do Rift/metabolismo , Ovinos , Doenças dos Ovinos/prevenção & controle , Valina , Vacinas Virais/genética
3.
Artigo em Inglês | MEDLINE | ID: mdl-31085519

RESUMO

Rift Valley fever virus (RVFV) is an emerging, mosquito-borne, zoonotic pathogen with recurrent outbreaks taking a considerable toll in human deaths in many African countries, for which no effective treatment is available. In cell culture studies and with laboratory animal models, the nucleoside analogue favipiravir (T-705) has demonstrated great potential for the treatment of several seasonal, chronic, and emerging RNA virus infections in humans, suggesting applicability to control some viral outbreaks. Treatment with favipiravir was shown to reduce the infectivity of Rift Valley fever virus both in cell cultures and in experimental animal models, but the mechanism of this protective effect is not understood. In this work, we show that favipiravir at concentrations well below the toxicity threshold estimated for cells is able to extinguish RVFV from infected cell cultures. Nucleotide sequence analysis has documented RVFV mutagenesis associated with virus extinction, with a significant increase in G to A and C to U transition frequencies and a decrease of specific infectivity, hallmarks of lethal mutagenesis.


Assuntos
Amidas/farmacologia , Mutagênese/genética , Pirazinas/farmacologia , Vírus da Febre do Vale do Rift/genética , Animais , Sequência de Bases , Linhagem Celular , Chlorocebus aethiops , Culicidae , Mutagênese/efeitos dos fármacos , RNA Viral/genética , Vírus da Febre do Vale do Rift/efeitos dos fármacos , Células Vero
4.
J Virol ; 92(16)2018 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-29875250

RESUMO

The development of vaccines against bluetongue, a prevalent livestock disease, has been focused on surface antigens that induce strong neutralizing antibody responses. Because of their antigenic variability, these vaccines are usually serotype restricted. We now show that a single highly conserved nonstructural protein, NS1, expressed in a modified vaccinia Ankara virus (MVA) vector can provide multiserotype protection in IFNAR-/- 129 mice against bluetongue virus (BTV) that is largely dependent on CD8 T cell responses. We found that the protective antigenic capacity of NS1 resides within the N terminus of the protein and is provided in the absence of neutralizing antibodies. The protective CD8 T cell response requires the presence of a specific peptide within the N terminus of NS1, since its deletion ablates the efficacy of the vaccine formulation. These data reveal the importance of the nonstructural protein NS1 in CD8 T cell-mediated protection against multiple BTV serotypes when vectorized as a recombinant MVA vaccine.IMPORTANCE Conventional vaccines have controlled or limited BTV expansion in the past, but they cannot address the need for cross-protection among serotypes and do not allow distinguishing between infected and vaccinated animals (DIVA strategy). There is a need to develop universal vaccines that induce effective protection against multiple BTV serotypes. In this work we have shown the importance of the nonstructural protein NS1, conserved among all the BTV serotypes, in CD8 T cell-mediated protection against multiple BTV serotypes when vectorized as a recombinant MVA vaccine.


Assuntos
Vírus Bluetongue/imunologia , Linfócitos T CD8-Positivos/imunologia , Epitopos Imunodominantes/imunologia , Receptor de Interferon alfa e beta/deficiência , Proteínas não Estruturais Virais/imunologia , Vacinas Virais/imunologia , Animais , Bluetongue/prevenção & controle , Células Cultivadas , Modelos Animais de Doenças , Portadores de Fármacos , ELISPOT , Vetores Genéticos , Interferon gama/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Knockout , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vaccinia virus/genética , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
5.
J Virol ; 91(10)2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28250124

RESUMO

Crimean-Congo hemorrhagic fever virus (CCHFV) is a bunyavirus causing severe hemorrhagic fever disease in humans, with high mortality rates. The requirement of a high-containment laboratory and the lack of an animal model hampered the study of the immune response and protection of vaccine candidates. Using the recently developed interferon alpha receptor knockout (IFNAR-/-) mouse model, which replicates human disease, we investigated the immunogenicity and protection of two novel CCHFV vaccine candidates: a DNA vaccine encoding a ubiquitin-linked version of CCHFV Gc, Gn, and N and one using transcriptionally competent virus-like particles (tc-VLPs). In contrast to most studies that focus on neutralizing antibodies, we measured both humoral and cellular immune responses. We demonstrated a clear and 100% efficient preventive immunity against lethal CCHFV challenge with the DNA vaccine. Interestingly, there was no correlation with the neutralizing antibody titers alone, which were higher in the tc-VLP-vaccinated mice. However, the animals with a lower neutralizing titer, but a dominant cell-mediated Th1 response and a balanced Th2 response, resisted the CCHFV challenge. Moreover, we found that in challenged mice with a Th1 response (immunized by DNA/DNA and boosted by tc-VLPs), the immune response changed to Th2 at day 9 postchallenge. In addition, we were able to identify new linear B-cell epitope regions that are highly conserved between CCHFV strains. Altogether, our results suggest that a predominantly Th1-type immune response provides the most efficient protective immunity against CCHFV challenge. However, we cannot exclude the importance of the neutralizing antibodies as the surviving immunized mice exhibited substantial amounts of them.IMPORTANCE Crimean-Congo hemorrhagic fever virus (CCHFV) is responsible for hemorrhagic diseases in humans, with a high mortality rate. There is no FDA-approved vaccine, and there are still gaps in our knowledge of the immune responses to infection. The recently developed mouse models mimic human CCHF disease and are useful to study the immunogenicity and the protection by vaccine candidates. Our study shows that mice vaccinated with a specific DNA vaccine were fully protected. Importantly, we show that neutralizing antibodies are not sufficient for protection against CCHFV challenge but that an extra Th1-specific cellular response is required. Moreover, we describe the identification of five conserved B-cell epitopes, of which only one was previously known, that could be of great importance for the development of diagnostics tools and the improvement of vaccine candidates.


Assuntos
Proteínas do Capsídeo/imunologia , Febre Hemorrágica da Crimeia/imunologia , Febre Hemorrágica da Crimeia/prevenção & controle , Plasmídeos/genética , Vacinas de DNA/imunologia , Vacinas de Partículas Semelhantes a Vírus/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Anticorpos Neutralizantes/sangue , Proteínas do Capsídeo/genética , Modelos Animais de Doenças , Epitopos de Linfócito B/imunologia , Vírus da Febre Hemorrágica da Crimeia-Congo/química , Vírus da Febre Hemorrágica da Crimeia-Congo/genética , Vírus da Febre Hemorrágica da Crimeia-Congo/imunologia , Febre Hemorrágica da Crimeia/virologia , Humanos , Imunidade Celular , Imunização , Imunogenicidade da Vacina , Interferon-alfa/deficiência , Interferon-alfa/genética , Camundongos , Camundongos Knockout , Plasmídeos/administração & dosagem , Células Th1 , Células Th2 , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Proteínas do Envelope Viral/genética
6.
Vet Res ; 49(1): 21, 2018 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-29467018

RESUMO

The aim of this work was to evaluate the immunogenicity and efficacy of DNA and MVA vaccines encoding the RVFV glycoproteins Gn and Gc in an ovine model of RVFV infection. Adult sheep of both sexes were challenged 12 weeks after the last immunization and clinical, virological, biochemical and immunological consequences, were analyzed. Strategies based on immunization with homologous DNA or heterologous DNA/MVA prime-boost were able to induce a rapid in vitro neutralizing antibody response as well as IFNγ production after in vitro virus specific re-stimulation. In these animals we observed reduced viremia levels and less clinical signs when compared with mock-immunized controls. In contrast, sheep inoculated with a homologous MVA prime-boost showed increased viremia correlating with the absence of detectable neutralizing antibody responses, despite of inducing cellular responses after the last immunization. However, faster induction of neutralizing antibodies and IFNγ production after challenge were found in this group when compared to the mock vaccinated group, indicative of a primed immune response. In conclusion, these results suggest that vaccination strategies based on DNA priming were able to mount and maintain specific anti-RVFV glycoprotein immune responses upon homologous or heterologous booster doses, warranting further optimization in large animal models of infection.


Assuntos
Febre do Vale de Rift/prevenção & controle , Vírus da Febre do Vale do Rift/imunologia , Doenças dos Ovinos/prevenção & controle , Vacinação/veterinária , Vacinas de DNA/farmacologia , Vacinas Virais/farmacologia , Animais , Feminino , Masculino , Febre do Vale de Rift/virologia , Ovinos , Doenças dos Ovinos/virologia
7.
Nanomedicine ; 12(5): 1185-92, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26970026

RESUMO

In this work we have tested the potential antiviral activity of silver nanoparticles formulated as Argovit™ against Rift Valley fever virus (RVFV). The antiviral activity of Argovit was tested on Vero cell cultures and in type-I interferon receptor deficient mice (IFNAR (-/-) mice) by two different approaches: (i) different dilutions of Argovit were added to previously infected cells or administrated to animals infected with a lethal dose of virus; (ii) virus was pre-incubated with different dilutions of Argovit before inoculation in mice or cells. Though the ability of silver nanoparticles to control an ongoing RVFV infection in the conditions tested was limited, the incubation of virus with Argovit before the infection led to a reduction of the infectivity titers both in vitro and in vivo. These results reveal the potential application of silver nanoparticles to control the infectivity of RVFV, which is an important zoonotic pathogen.


Assuntos
Antivirais/farmacologia , Nanopartículas/uso terapêutico , Vírus da Febre do Vale do Rift/efeitos dos fármacos , Prata/uso terapêutico , Animais , Camundongos , Febre do Vale de Rift/prevenção & controle , Vírus da Febre do Vale do Rift/patogenicidade
8.
Arch Virol ; 159(3): 535-46, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24100475

RESUMO

Rift Valley fever virus (RVFV) is a vector-borne virus that causes high neonatal mortality in livestock and deadly haemorrhagic fever in humans. In this paper, we describe the generation of monoclonal antibodies (mabs) against all three structural proteins of RVFV (glycoproteins Gn and Gc and nucleocapsid protein NP). After immunization of BALB/c mice with individual recombinant proteins, a total of 45 clones secreting ELISA-reactive monoclonal antibodies against NP, Gn and Gc epitopes were obtained. Twelve clones were directed to NP, 28 to Gn, and 5 to Gc. Western blot analysis revealed that most of the mabs were reactive to linearized epitopes on recombinant as well as native virus proteins. Six mabs against NP, 21 against Gn and all mabs against Gc also detected conformational epitopes, as shown by indirect immunofluorescence on RVFV-infected cells. All of the mabs were evaluated for their use in a competition enzyme-linked immunosorbent assay (ELISA) for the detection of a RVFV infection. Several mabs were identified that competed with polyclonal rabbit serum, and one of them - mab Gn123, raised against Gn protein - was selected for a proof-of-principle study with field sera from a recent Rift Valley fever outbreak. The novel Gn-based competition ELISA demonstrated high performance, offering a promising alternative and addition to serological assays based on nucleocapsid protein.


Assuntos
Anticorpos Monoclonais , Anticorpos Antivirais/sangue , Febre do Vale de Rift/diagnóstico , Vírus da Febre do Vale do Rift/imunologia , Proteínas Estruturais Virais/imunologia , Animais , Anticorpos Monoclonais/isolamento & purificação , Anticorpos Antivirais/isolamento & purificação , Antígenos Virais/genética , Antígenos Virais/imunologia , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Vírus da Febre do Vale do Rift/genética , Testes Sorológicos/métodos , Proteínas Estruturais Virais/genética
9.
Methods Mol Biol ; 2824: 281-318, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39039419

RESUMO

Rift Valley fever virus (RVFV; genus Phlebovirus, family Phenuiviridae, order Bunyavirales) is a mosquito-borne zoonotic pathogen endemic in Africa. Its negative-stranded genomic RNA (vRNA) is divided into three segments termed L, M, and S. Both vRNAs and antigenomic cRNAs are encapsidated by viral nucleoprotein (N) to form nucleocapsids, which constitute the template for genome transcription and replication. Based on a number of electron microscopy and structural studies, the viral RNAs of negative-strand RNA viruses, including phleboviruses, are commonly considered to be entirely and uniformly covered by N protein. However, high resolution data supporting this notion was missing to date.Here, we describe a method how to globally map all N-RNA interactions of RVFV by using iCLIP (individual-nucleotide resolution UV cross-linking and immunoprecipitation). The protocol is based on covalent cross-linking of direct protein-RNA interactions by UV irradiation. Following sample lysis, a selective isolation of N in complex with its RNA targets is achieved by immunoprecipitation. Then, N-RNA complexes are separated by SDS-PAGE, and after membrane transfer, RNA is isolated and subjected to library preparation and high-throughput sequencing. We explain how the standard iCLIP protocol can be adapted to RVFV N-RNA interaction studies. The protocol describes mapping of all N interactions with the vRNAs and cRNAs derived either from RVFV particles or from infected cells.


Assuntos
Genoma Viral , Nucleoproteínas , RNA Viral , Vírus da Febre do Vale do Rift , Vírus da Febre do Vale do Rift/genética , RNA Viral/genética , RNA Viral/metabolismo , Nucleoproteínas/metabolismo , Nucleoproteínas/genética , Mapeamento de Nucleotídeos/métodos , Imunoprecipitação/métodos , Humanos , Febre do Vale de Rift/virologia , Febre do Vale de Rift/metabolismo , Animais
10.
PLoS Negl Trop Dis ; 18(8): e0012011, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39159263

RESUMO

Rift Valley fever (RVF) is an important zoonotic viral disease affecting several species of domestic and wild ruminants, causing major economic losses and dozens of human deaths in various geographical areas of Africa, where it is endemic. Although it is not present in Europe, there is a risk of its introduction and spread linked to globalisation and climate change. At present, the only measure that could help to prevent the disease is vaccination of flocks in areas at risk of RVF. Available live attenuated vaccines are an effective means of controlling the disease, but their use is often questioned due to residual virulence, particularly in susceptible hosts such as pregnant sheep. On the other hand, no vaccine is currently licensed for use in humans. The development of safe and effective vaccines is therefore a major area of research. In previous studies, we selected under selective mutagenic pressure a highly attenuated RVFV 56/74 virus variant called 40Fp8. This virus showed an extremely attenuated phenotype in both wild-type and immunodeficient A129 (IFNARKO) mice, yet was still able to induce protective immunity after a single inoculation, thus supporting its use as a safe, live attenuated vaccine. To further investigate its safety, in this work we have analysed the attenuation level of 40Fp8 in immunosuppressed mice (A129) when administered by the intranasal route, and compared it with other attenuated RVF viruses that are the basis of vaccines in use or in development. Our results show that 40Fp8 has a much higher attenuated level than these other viruses and confirm its potential as a candidate for safe RVF vaccine development.


Assuntos
Administração Intranasal , Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Vacinas Atenuadas , Vacinas Virais , Animais , Febre do Vale de Rift/prevenção & controle , Febre do Vale de Rift/imunologia , Vacinas Atenuadas/imunologia , Vacinas Atenuadas/administração & dosagem , Vírus da Febre do Vale do Rift/imunologia , Camundongos , Vacinas Virais/imunologia , Vacinas Virais/administração & dosagem , Feminino , Vacinação/métodos , Anticorpos Antivirais/sangue
11.
Viruses ; 16(1)2024 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-38257787

RESUMO

The introduction of three single nucleotide mutations into the genome of the virulent RVFV ZH548 strain allows for the rescue of a fully attenuated virus in mice (ZH548-rA2). These mutations are located in the viral genes encoding the RdRp and the non-structural protein NSs. This paper shows the results obtained after the subcutaneous inoculation of ZH548-rA2 in adult sheep and the subsequent challenge with the parental virus (ZH548-rC1). Inoculation with the ZH548-rA2 virus caused no detectable clinical or pathological effect in sheep, whereas inoculation of the parental rC1 virus caused lesions compatible with viral infection characterised by the presence of scattered hepatic necrosis. Viral infection was confirmed via immunohistochemistry, with hepatocytes within the necrotic foci appearing as the main cells immunolabelled against viral antigen. Furthermore, the inoculation of sheep with the rA2 virus prevented the liver damage expected after rC1 virus inoculation, suggesting a protective efficacy in sheep which correlated with the induction of both humoral and cell-mediated immune responses.


Assuntos
Vírus da Febre do Vale do Rift , Viroses , Animais , Camundongos , Ovinos , Vírus da Febre do Vale do Rift/genética , Antígenos Virais , Genes Virais , Hepatócitos
12.
Nat Commun ; 15(1): 7602, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-39217162

RESUMO

Rift Valley fever virus (RVFV) is a mosquito-borne zoonotic pathogen. Its RNA genome consists of two negative-sense segments (L and M) with one gene each, and one ambisense segment (S) with two opposing genes separated by the noncoding "intergenic region" (IGR). These vRNAs and the complementary cRNAs are encapsidated by nucleoprotein (N). Using iCLIP2 (individual-nucleotide resolution UV crosslinking and immunoprecipitation) to map all N-vRNA and N-cRNA interactions, we detect N coverage along the L and M segments. However, the S segment vRNA and cRNA each contain approximately 100 non-encapsidated nucleotides stretching from the IGR into the 5'-adjacent reading frame. These exposed regions are RNase-sensitive and predicted to form stem-loop structures with the mRNA transcription termination motif positioned near the top. Moreover, optimal S segment transcription and replication requires the entire exposed region rather than only the IGR. Thus, the RVFV S segment contains a central, non-encapsidated RNA region with a functional role.


Assuntos
RNA Viral , Vírus da Febre do Vale do Rift , Vírus da Febre do Vale do Rift/genética , RNA Viral/genética , Animais , DNA Intergênico/genética , Genoma Viral , Replicação Viral/genética , Febre do Vale de Rift/virologia , Febre do Vale de Rift/transmissão , Conformação de Ácido Nucleico , Nucleoproteínas/genética , Nucleoproteínas/metabolismo , Humanos , Transcrição Gênica
13.
Virol J ; 10: 349, 2013 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-24304565

RESUMO

BACKGROUND: Rift Valley Fever (RVF) is a viral zoonosis that historically affects livestock production and human health in sub-Saharan Africa, though epizootics have also occurred in the Arabian Peninsula. Whilst an effective live-attenuated vaccine is available for livestock, there is currently no licensed human RVF vaccine. Replication-deficient chimpanzee adenovirus (ChAd) vectors are an ideal platform for development of a human RVF vaccine, given the low prevalence of neutralizing antibodies against them in the human population, and their excellent safety and immunogenicity profile in human clinical trials of vaccines against a wide range of pathogens. METHODS: Here, in BALB/c mice, we evaluated the immunogenicity and efficacy of a replication-deficient chimpanzee adenovirus vector, ChAdOx1, encoding the RVF virus envelope glycoproteins, Gn and Gc, which are targets of virus neutralizing antibodies. The ChAdOx1-GnGc vaccine was assessed in comparison to a replication-deficient human adenovirus type 5 vector encoding Gn and Gc (HAdV5-GnGc), a strategy previously shown to confer protective immunity against RVF in mice. RESULTS: A single immunization with either of the vaccines conferred protection against RVF virus challenge eight weeks post-immunization. Both vaccines elicited RVF virus neutralizing antibody and a robust CD8+ T cell response. CONCLUSIONS: Together the results support further development of RVF vaccines based on replication-deficient adenovirus vectors, with ChAdOx1-GnGc being a potential candidate for use in future human clinical trials.


Assuntos
Adenoviridae/genética , Portadores de Fármacos , Vetores Genéticos , Febre do Vale de Rift/prevenção & controle , Vírus da Febre do Vale do Rift/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Linfócitos T CD8-Positivos/imunologia , Modelos Animais de Doenças , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Febre do Vale de Rift/imunologia , Vírus da Febre do Vale do Rift/genética , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
14.
Vaccine ; 41(20): 3275-3284, 2023 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-37085455

RESUMO

Schmallenberg Virus (SBV), an arbovirus from the Peribunyaviridae family and Orthobunyavirus genus, was discovered in late 2011 in Germany and has been circulating in Europe, Asia and Africa ever since. The virus causes a disease associated with ruminants that includes fever, fetal malformation, drop in milk production, diarrhoea and stillbirths, becoming a burden for small and large farms. Building on previous studies on SBV nucleoprotein (SBV-N) as a promising vaccine candidate, we have investigated the possible protein regions responsible for protection. Based on selective truncation of domains designed from the available crystal structure of the SBV-N, we identified both the N-terminal domain (N-term; Met1 - Thr133) and a smaller fragment within (C4; Met1 - Ala58) as vaccine prototypes. Two injections of the N-term and C4 polypeptides protected mice knockout for type I interferon (IFN) receptors (IFNAR-/-) challenged with virulent SBV, opposite to control groups that presented severe signs of morbidity and weight loss. Viremia analyses along with the presence of IFN-γ secreted from splenocytes re-stimulated with the N-terminal region of the protein corroborate that these two portions of SBV-N can be employed as subunit vaccines. Apart from both proteinaceous fragments being easily produced in bacterial cells, the C4 polypeptide shares a high sequence homology (∼87.1 %) with the corresponding region of nucleoproteins of several viruses of the Simbu serogroup, a group of Orthobunyaviruses that comprises SBV and veterinary pathogens like Akabane virus and human infecting viruses like Oropouche. Thus, we propose that this smaller fragment is better suited for vaccine nanoparticle formulation, and it paves the way to further research with other related Orthobunyaviruses.


Assuntos
Infecções por Bunyaviridae , Doenças dos Bovinos , Orthobunyavirus , Vacinas , Humanos , Animais , Camundongos , Bovinos , Orthobunyavirus/genética , Infecções por Bunyaviridae/prevenção & controle , Infecções por Bunyaviridae/veterinária , Viremia/prevenção & controle , Nucleoproteínas/genética , Sorogrupo , Imunização , Ruminantes , Doenças dos Bovinos/prevenção & controle
15.
Front Vet Sci ; 10: 1121935, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36777670

RESUMO

Introduction: Coronavirus disease 2019 (COVID-19) is an infectious zoonotic disease caused by SARS-CoV-2. Monitoring the infection in pets is recommended for human disease surveillance, prevention, and control since the virus can spread from people to animals during close contact. Several diagnostic tests have been adapted from humans to animals, but limited data on the validation process are available. Methods: Herein, the first comparative study of six "in house" and two commercial serological tests developed to monitor SARS-CoV-2 infection in pets was performed with a well-coded panel of sera (61 cat sera and 74 dog sera) with a conservative criterion (viral seroneutralisation and/or RT-qPCR results) as a reference. Four "in house" tests based on either the RBD fragment of the spike protein (RBD-S) or the N-terminal fragment of the nucleoprotein (N) were developed for the first time. The analytical specificity (ASp) of those tests that showed the best diagnostic performance was assessed. The validation included the analysis of a panel of sera obtained pre-pandemic from cats and dogs infected with other coronaviruses to determine the analytical Sp (17 cat sera and 41 dog sera). Results and discussion: ELISAS based on the S protein are recommended in serosurveillance studies for cats (RBD-S SALUVET ELISA, ELISA COVID UNIZAR and INgezim® COVID 19 S VET) and dogs (INgezim® COVID 19 S VET and RBD-S SALUVET ELISA). These tests showed higher diagnostic sensitivity (Se) and DSp in cats (>90%) than in dogs. When sera obtained prior to the pandemic and from animals infected with other coronaviruses were analyzed by RBD-S and N SALUVET ELISAs and INgezim® COVID 19 S VET, a few cross reactors or no cross reactions were detected when dog and cat sera were analyzed by tests based on the S protein, respectively. In contrast, the number of cross reactions increased when the test was based on the N protein. Thus, the use of tests based on the N protein was discarded for serodiagnosis purposes. The results obtained revealed the most accurate serological tests for each species. Further studies should attempt to improve the diagnostic performance of serological tests developed for dogs.

16.
Vaccines (Basel) ; 11(5)2023 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-37243110

RESUMO

The COVID-19 pandemic has underscored the importance of swift responses and the necessity of dependable technologies for vaccine development. Our team previously developed a fast cloning system for the modified vaccinia virus Ankara (MVA) vaccine platform. In this study, we reported on the construction and preclinical testing of a recombinant MVA vaccine obtained using this system. We obtained recombinant MVA expressing the unmodified full-length SARS-CoV-2 spike (S) protein containing the D614G amino-acid substitution (MVA-Sdg) and a version expressing a modified S protein containing amino-acid substitutions designed to stabilize the protein a in a pre-fusion conformation (MVA-Spf). S protein expressed by MVA-Sdg was found to be expressed and was correctly processed and transported to the cell surface, where it efficiently produced cell-cell fusion. Version Spf, however, was not proteolytically processed, and despite being transported to the plasma membrane, it failed to induce cell-cell fusion. We assessed both vaccine candidates in prime-boost regimens in the susceptible transgenic K18-human angiotensin-converting enzyme 2 (K18-hACE2) in mice and in golden Syrian hamsters. Robust immunity and protection from disease was induced with either vaccine in both animal models. Remarkably, the MVA-Spf vaccine candidate produced higher levels of antibodies, a stronger T cell response, and a higher degree of protection from challenge. In addition, the level of SARS-CoV-2 in the brain of MVA-Spf inoculated mice was decreased to undetectable levels. Those results add to our current experience and range of vaccine vectors and technologies for developing a safe and effective COVID-19 vaccine.

17.
J Virol ; 85(13): 6098-105, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21450816

RESUMO

Rift Valley fever virus (RVFV) is a zoonotic pathogen that primarily affects ruminants but can also be lethal in humans. A negative-stranded RNA virus of the family Bunyaviridae, this pathogen is transmitted mainly via mosquito vectors. RVFV has shown the ability to inflict significant damage to livestock and is also a threat to public health. While outbreaks have traditionally occurred in sub-Saharan Africa, recent outbreaks in the Middle East have raised awareness of the potential of this virus to spread to Europe, Asia, and the Americas. Although the virus was initially characterized almost 80 years ago, the only vaccine approved for widespread veterinary use is an attenuated strain that has been associated with significant pathogenic side effects. However, increased understanding of the molecular biology of the virus over the last few years has led to recent advances in vaccine design and has enabled the development of more-potent prophylactic measures to combat infection. In this review, we discuss several aspects of RVFV, with particular emphasis on the molecular components of the virus and their respective roles in pathogenesis and an overview of current vaccine candidates. Progress in understanding the epidemiology of Rift Valley fever has also enabled prediction of potential outbreaks well in advance, thus providing another tool to combat the physical and economic impact of this disease.


Assuntos
Febre do Vale de Rift/prevenção & controle , Vírus da Febre do Vale do Rift/patogenicidade , Vacinas Virais/administração & dosagem , Animais , Culicidae/virologia , Surtos de Doenças/prevenção & controle , Saúde Global , Humanos , Insetos Vetores , Gado , Febre do Vale de Rift/epidemiologia , Febre do Vale de Rift/virologia , Vírus da Febre do Vale do Rift/genética , Vírus da Febre do Vale do Rift/imunologia , Proteínas Virais/genética , Proteínas Virais/metabolismo , Vacinas Virais/imunologia
18.
Methods Mol Biol ; 2465: 1-26, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35118613

RESUMO

Ensuring the maximum standards of quality and welfare in animal production requires developing effective tools to halt and prevent the spread of the high number of infectious diseases affecting animal husbandry. Many of these diseases are caused by pathogens of viral etiology. To date, one of the best strategies is to implement preventive vaccination policies whenever possible. However, many of the currently manufactured animal vaccines still rely in classical vaccine technologies (killed or attenuated vaccines). Under some circumstances, these vaccines may not be optimal in terms of safety and immunogenicity, nor adequate for widespread application in disease-free countries at risk of disease introduction. One step ahead is needed to improve and adapt vaccine manufacturing to the use of new generation vaccine technologies already tested in experimental settings. In the context of viral diseases of veterinary interest, we overview current vaccine technologies that can be approached, with a brief insight in the type of immunity elicited.


Assuntos
Vacinas Virais , Viroses , Animais , Vacinação/veterinária , Vacinas Atenuadas , Viroses/prevenção & controle , Viroses/veterinária
19.
Methods Mol Biol ; 2465: 209-225, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35118624

RESUMO

Live attenuated viruses remain as vaccine agents with unparalleled performance in terms of duration, magnitude, and breadth of induced immune responses. As the yellow fever-attenuated vaccine strain Y17D, attenuated Rift Valley fever virus shares features suitable to be used as a viral vector for heterologous antigen expression and bivalent vaccine development. Current reverse genetics technology showed the successful rescue of RVFV carrying foreign antigens with little immunogenicity loss in experimental animal models. We show here the basic experimental protocol to achieve the expression of candidate vaccine antigens from other important diseases of ruminants using RVFV as a vector platform as well as preliminary steps for the characterization of immunogenicity in vivo.


Assuntos
Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Vacinas Virais , Animais , Antígenos Virais/genética , Febre do Vale de Rift/prevenção & controle , Vírus da Febre do Vale do Rift/genética , Ruminantes , Vacinas Virais/genética
20.
Sci Rep ; 12(1): 263, 2022 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-34997166

RESUMO

Cold Atmospheric Plasma (CAP) and Plasma Activated Media (PAM) are effective against bacteria, fungi, cancer cells, and viruses because they can deliver Reactive Oxygen and Nitrogen Species (RONS) on a living tissue with negligible damage on health cells. The antiviral activity of CAP against SARS-CoV-2 is being investigated, however, the same but of PAM has not been explored despite its potential. In the present study, the capability of Plasma Activated Media (PAM) to inactivate SARS-CoV-2 and PR8 H1N1 influenza virus with negligible damage on healthy cells is demonstrated. PAM acted by both virus detaching and diminished replication. Furthermore, the treatment of A549 lung cells at different times with buffered PAM did not induce interleukin 8 expression, showing that PAM did not induce inflammation. These results open a new research field by using PAM to the development novel treatments for COVID-19, influenza, and other respiratory diseases.


Assuntos
Antivirais/farmacologia , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Gases em Plasma/farmacologia , SARS-CoV-2/efeitos dos fármacos , Células A549 , Descoberta de Drogas , Humanos , Influenza Humana/tratamento farmacológico , Espécies Reativas de Nitrogênio/farmacologia , Espécies Reativas de Oxigênio/farmacologia , Tratamento Farmacológico da COVID-19
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA