Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
1.
Am Nat ; 204(2): 133-146, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39008835

RESUMO

AbstractInfectious disease dynamics operate across biological scales: pathogens replicate within hosts but transmit among populations. Functional changes in the pathogen-host interaction thus generate cascading effects across organizational scales. We investigated within-host dynamics and among-host transmission of three strains (SAT-1, -2, -3) of foot-and-mouth disease viruses (FMDVs) in their wildlife host, African buffalo. We combined data on viral dynamics and host immune responses with mathematical models to ask the following questions: How do viral and immune dynamics vary among strains? Which viral and immune parameters determine viral fitness within hosts? And how do within-host dynamics relate to virus transmission? Our data reveal contrasting within-host dynamics among viral strains, with SAT-2 eliciting more rapid and effective immune responses than SAT-1 and SAT-3. Within-host viral fitness was overwhelmingly determined by variation among hosts in immune response activation rates but not by variation among individual hosts in viral growth rate. Our analyses investigating across-scale linkages indicate that viral replication rate in the host correlates with transmission rates among buffalo and that adaptive immune activation rate determines the infectious period. These parameters define the virus's relative basic reproductive number (ℛ0), suggesting that viral invasion potential may be predictable from within-host dynamics.


Assuntos
Búfalos , Vírus da Febre Aftosa , Febre Aftosa , Animais , Búfalos/virologia , Vírus da Febre Aftosa/imunologia , Vírus da Febre Aftosa/crescimento & desenvolvimento , Febre Aftosa/transmissão , Febre Aftosa/virologia , Febre Aftosa/imunologia , Interações Hospedeiro-Patógeno/imunologia , Replicação Viral , Modelos Biológicos
2.
J Virol ; 95(13): e0023821, 2021 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-33853964

RESUMO

Nucleolin (NCL), a stress-responsive RNA-binding protein, has been implicated in the translation of internal ribosome entry site (IRES)-containing mRNAs, which encode proteins involved in cell proliferation, carcinogenesis, and viral infection (type I IRESs). However, the details of the mechanisms by which NCL participates in IRES-driven translation have not hitherto been described. Here, we identified NCL as a protein that interacts with the IRES of foot-and-mouth disease virus (FMDV), which is a type II IRES. We also mapped the interactive regions within FMDV IRES and NCL in vitro. We found that NCL serves as a substantial regulator of FMDV IRES-driven translation but not of bulk cellular or vesicular stomatitis virus cap-dependent translation. NCL also modulates the translation of and infection by Seneca Valley virus (type III-like IRES) and classical swine fever virus (type III IRES), which suggests that its function is conserved in unrelated IRES-containing viruses. We also show that NCL affects viral replication by directly regulating the production of viral proteins and indirectly regulating FMDV RNA synthesis. Importantly, we observed that the cytoplasmic relocalization of NCL during FMDV infection is a substantial step for viral IRES-driven translation and that NCL specifically promotes the initiation phase of the translation process by recruiting translation initiation complexes to viral IRES. Finally, the functional importance of NCL in FMDV pathogenicity was confirmed in vivo. Taken together, our findings demonstrate a specific function for NCL in selective mRNA translation and identify a target for the development of a broad-spectrum class of antiviral interventions. IMPORTANCE FMDV usurps the cellular translation machinery to initiate viral protein synthesis via a mechanism driven by IRES elements. It allows the virus to shut down bulk cellular translation, while providing an advantage for its own gene expression. With limited coding capacity in its own genome, FMDV has evolved a mechanism to hijack host proteins to promote the recruitment of the host translation machinery, a process that is still not well understood. Here, we identified nucleolin (NCL) as a positive regulator of the IRES-driven translation of FMDV. Our study supports a model in which NCL relocalizes from the nucleus to the cytoplasm during the course of FMDV infection, where the cytoplasmic NCL promotes FMDV IRES-driven translation by bridging the translation initiation complexes with viral IRES. Our study demonstrates a previously uncharacterized role of NCL in the translation initiation of IRES-containing viruses, with important implications for the development of broad antiviral interventions.


Assuntos
Vírus da Febre Aftosa/genética , Regulação Viral da Expressão Gênica/genética , Sítios Internos de Entrada Ribossomal/genética , Iniciação Traducional da Cadeia Peptídica/genética , Fosfoproteínas/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Linhagem Celular , Proliferação de Células/genética , Chlorocebus aethiops , Vírus da Febre Suína Clássica/genética , Cricetinae , Vírus da Febre Aftosa/crescimento & desenvolvimento , Camundongos , Camundongos Endogâmicos BALB C , Picornaviridae/genética , Interferência de RNA , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Suínos , Células Vero , Replicação Viral/genética , Nucleolina
3.
Virus Genes ; 56(1): 1-15, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31776851

RESUMO

Foot-and-mouth disease is endemic in livestock in large parts of Africa and Asia, where it is an important driver of food insecurity and a major obstacle to agricultural development and the international trade in animal products. Virtually all commercially available vaccines are inactivated whole-virus vaccines produced in cell culture, but the adaptation of a field isolate of the virus to growth in culture is laborious and time-consuming. This is of particular concern for the development of vaccines to newly emerging virus lineages, where long lead times from virus isolate to vaccine can delay the implementation of effective control programs. High antigen yields in production cells are also necessary to make vaccines affordable for less developed countries in endemic areas. Therefore, a rational approach to cell culture adaptation that combines prior knowledge of common adaptive mutations and reverse genetics techniques is urgently required. This review provides an overview of amino acid exchanges in the viral capsid proteins in the context of adaptation to cell culture.


Assuntos
Substituição de Aminoácidos , Proteínas do Capsídeo/genética , Vírus da Febre Aftosa/crescimento & desenvolvimento , Vírus da Febre Aftosa/genética , Febre Aftosa/virologia , Animais , Proteínas do Capsídeo/metabolismo , Técnicas de Cultura de Células , Febre Aftosa/prevenção & controle , Vírus da Febre Aftosa/metabolismo , Vacinas Virais/genética , Vacinas Virais/metabolismo , Cultura de Vírus
4.
J Appl Microbiol ; 128(5): 1524-1531, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-31883170

RESUMO

AIMS: As cell-adapted foot-and-mouth disease virus (FMDV) with H56R mutation in VP3 has reduced thermostability, this study aimed to investigate the effect of thermostabilizers on cell-adapted FMDV for vaccine production. METHODS AND RESULTS: We examined the effect of 3% sucrose, 10% (or 25%) glycerol or 10% FBS on cell-adapted FMDV O/SKR/JC/2014, containing H56R mutation in VP3, as vaccine seed virus at -80, 4, 25 or 37°C for 2, 4 or 7 days. The stabilizing effect of 3% sucrose on O/SKR/JC/2014 was observed at 25, 37°C, and after repeated freeze-thaw cycles. Additionally, we tested the effect of 3% sucrose on the growth of FMDV or cells and did not observe any decrease in either viral growth or cell viability. CONCLUSIONS: Our study showed the protective effect of 3% sucrose on FMDV infectivity at various temperatures; this virus stock in 3% sucrose could be used for infecting cells without the removal of sucrose. SIGNIFICANCE AND IMPACT OF THE STUDY: We suggest that 3% sucrose-containing medium could be beneficial for the stable storage and transport of cell-adapted FMDV.


Assuntos
Vírus da Febre Aftosa/crescimento & desenvolvimento , Sacarose/análise , Excipientes de Vacinas/análise , Vacinas Virais/química , Animais , Proteínas do Capsídeo/genética , Sobrevivência Celular/efeitos dos fármacos , Febre Aftosa/virologia , Vírus da Febre Aftosa/efeitos dos fármacos , Vírus da Febre Aftosa/genética , Vírus da Febre Aftosa/imunologia , Mutação , Sacarose/farmacologia , Temperatura , Excipientes de Vacinas/farmacologia , Potência de Vacina
5.
Vaccine ; 37(43): 6380-6389, 2019 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-31519448

RESUMO

The anticipated increasing demand for inactivated foot-and-mouth (FMD) disease vaccine calls for its larger production capacity, while development of a large-scale process typically requires high running cost and has very limited experimental throughput at manufacturing scale. Thus, an economic scale-down model of representing a large-scale process becomes necessary and essential. In this study, we used a systematic approach to establish a scale-down model representing a 4000-L culture process for FMD vaccine production by suspension BHK-21 cells. In detail, we firstly compared hydrodynamic properties of three bioreactors (14-L, 800-L and 4000-L) under three different conditions (equivalent mixing time, equivalent shear stress and equivalent volumetric power). We figured out equivalent volumetric power (P/V) potentially as an appropriate scale-down strategy, since it resulted in comparable calculated hydrodynamic parameters among three bioreactors. Next, we used computational fluid dynamics (CFD) simulation to provide more details about hydrodynamic environments inside the bioreactors, which supports the reliability of this scale-down strategy. Finally, we compared cell growth, metabolites, vaccine productivity and product quality attributes during FMD vaccine production by BHK-21 cells and observed very close performances among three bioreactors, which once again demonstrates the robustness of this scale-down model. This scale-down strategy can be applied to study variations and critical quality attributes (CQAs) in the resultant production process based on quality by design (QbD) principles, aiming at further more efficient optimization of vaccine production.


Assuntos
Reatores Biológicos/virologia , Técnicas de Cultura de Células/instrumentação , Técnicas de Cultura de Células/métodos , Vírus da Febre Aftosa/crescimento & desenvolvimento , Febre Aftosa/prevenção & controle , Vacinas Virais , Animais , Linhagem Celular , Cricetinae , Cricetulus , Vírus da Febre Aftosa/imunologia , Hidrodinâmica , Rim/citologia , Camundongos , Reprodutibilidade dos Testes , Vacinas de Produtos Inativados
6.
Virology ; 535: 162-170, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31306911

RESUMO

MicroRNAs (miRNAs) play important regulatory roles during interactions between virus pathogens and host cells, but whether and how they work in the case of foot-and-mouth disease virus (FMDV) is less understood. Based on a microarray-based miRNA profiling in the porcine kidney cell line PK-15, we identified 36 differentially expressed host miRNAs at the early stage of FMDV infection, among which miR-1307 was significantly induced. Functional characterization demonstrated that miR-1307 attenuated FMDV replication. Further experiments proved that miR-1307 specifically promoted the degradation of the viral structural protein VP3 indirectly through proteasome pathway. Moreover, innate immune signaling was activated and expression of immune responsive genes was significantly enhanced in the miR-1307-overexpressing clones. Together, our data demonstrated that miR-1307 suppresses FMDV replication by destabilizing VP3 and enhancing host immune response. Importantly, subcutaneous injection of miR-1307 agomir delayed the FMDV-induced lethality in suckling mice, exhibiting its therapeutic potential to control foot-and-mouth disease (FMD).


Assuntos
Proteínas do Capsídeo/metabolismo , Vírus da Febre Aftosa/imunologia , Febre Aftosa/imunologia , Imunidade Inata , Proteólise , Replicação Viral , Animais , Linhagem Celular , Modelos Animais de Doenças , Células Epiteliais/imunologia , Células Epiteliais/virologia , Vírus da Febre Aftosa/crescimento & desenvolvimento , Perfilação da Expressão Gênica , Interações entre Hospedeiro e Microrganismos , Camundongos , Modelos Teóricos , Complexo de Endopeptidases do Proteassoma/metabolismo , Análise de Sobrevida , Suínos
7.
Viruses ; 11(6)2019 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-31167384

RESUMO

Foot-and-mouth disease virus (FMDV) is endemic in many parts of the world. Vaccination is an important control measure, limits viral spread, and can help to eradicate the disease. However, vaccination programs are cost-intensive because of the short shelf life of vaccines and the need for frequent re-vaccination. Animal-component-free (ACF) or chemically defined media (CDM) at high cell densities are a promising approach for the production of inexpensive high-quality vaccines, but the occurrence of cell density effects has been reported for various virus-cell systems in vaccine production. For FMDV, the use of CDM or ACF media for vaccine production has not been studied and no information about cell density effects is available. This work describes the propagation of FMDV in ACF or in CDM. Cells were grown at increasing cell densities and either 100% media exchange or addition of 30% fresh media was performed before infection with FMDV. Increasing cell densities reduced the viral titer and increased yield variability in all media except BHK300G. This effect can be mitigated by performing a 100% media exchange before infection or when using the controlled environment of a bioreactor. The media composition and also a fragile relationship between virus and cell metabolism seem to be causal for that phenomenon.


Assuntos
Contagem de Células , Técnicas de Cultura de Células/métodos , Meios de Cultura , Vírus da Febre Aftosa/crescimento & desenvolvimento , Animais , Linhagem Celular , Vírus da Febre Aftosa/metabolismo , Replicação Viral
8.
J Virol ; 93(15)2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31092573

RESUMO

African buffaloes (Syncerus caffer) are the principal "carrier" hosts of foot-and-mouth disease virus (FMDV). Currently, the epithelia and lymphoid germinal centers of the oropharynx have been identified as sites for FMDV persistence. We carried out studies in FMDV SAT1 persistently infected buffaloes to characterize the diversity of viruses in oropharyngeal epithelia, germinal centers, probang samples (oropharyngeal scrapings), and tonsil swabs to determine if sufficient virus variation is generated during persistence for immune escape. Most sequencing reads of the VP1 coding region of the SAT1 virus inoculum clustered around 2 subpopulations differing by 22 single-nucleotide variants of intermediate frequency. Similarly, most sequences from oropharynx tissue clustered into two subpopulations, albeit with different proportions, depending on the day postinfection (dpi). There was a significant difference between the populations of viruses in the inoculum and in lymphoid tissue taken at 35 dpi. Thereafter, until 400 dpi, no significant variation was detected in the viral populations in samples from individual animals, germinal centers, and epithelial tissues. Deep sequencing of virus from probang or tonsil swab samples harvested prior to postmortem showed less within-sample variability of VP1 than that of tissue sample sequences analyzed at the same time. Importantly, there was no significant difference in the ability of sera collected between 14 and 400 dpi to neutralize the inoculum or viruses isolated at later time points in the study from the same animal. Therefore, based on this study, there is no evidence of escape from antibody neutralization contributing to FMDV persistent infection in African buffalo.IMPORTANCE Foot-and-mouth disease virus (FMDV) is a highly contagious virus of cloven-hoofed animals and is recognized as the most important constraint to international trade in animals and animal products. African buffaloes (Syncerus caffer) are efficient carriers of FMDV, and it has been proposed that new virus variants are produced in buffalo during the prolonged carriage after acute infection, which may spread to cause disease in livestock populations. Here, we show that despite an accumulation of low-frequency sequence variants over time, there is no evidence of significant antigenic variation leading to immune escape. Therefore, carrier buffalo are unlikely to be a major source of new virus variants.


Assuntos
Búfalos , Portador Sadio/veterinária , Evolução Molecular , Vírus da Febre Aftosa/crescimento & desenvolvimento , Febre Aftosa/imunologia , Febre Aftosa/virologia , Evasão da Resposta Imune , Animais , Proteínas do Capsídeo/genética , Portador Sadio/imunologia , Portador Sadio/virologia , Epitélio/virologia , Vírus da Febre Aftosa/genética , Vírus da Febre Aftosa/imunologia , Instabilidade Genômica , Centro Germinativo/virologia , Mutação , Orofaringe/virologia , Análise de Sequência de DNA
9.
J Med Virol ; 91(2): 208-214, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30039874

RESUMO

Foot-and-mouth disease (FMD) is a disease of worldwide economic importance, and vaccines play an important role in preventing FMDV outbreaks. However, new control strategies are still needed since FMDV outbreaks still occur in some disease-free countries. Currently, interferon (IFN)-based strategies have been demonstrated to be an efficient biotherapeutic option against FMDV; however, interferon omega (IFN-ω) has not yet been assessed in this capacity. Thus, this study evaluated the antiviral activity of porcine IFN omega 7 (PoIFN-ω7) against FMDV. After the PoIFN-ω7 was expressed and purified, cell proliferation assays and quantitative real-time reverse transciption-polymerase chain reaction were used to evaluate the effective anti-cytopathic concentration of PoIFN-ω7 and its effectiveness pre- and post-infection with FMDV in swine kidney cells (IBRS-2). Results showed the rHis-PoIFN-ω7 fusion protein was considerably expressed using Escherichia coli BL21 (DE3) strain, and the recombinant protein exhibited significant in vitro protection against FMDV, including two strains belonging to type O and A FMDV, respectively. In addition, PoIFN-ω7 upregulated the transcription of Mx1, ISG15, OAS1, and PKR genes. These findings indicated that IFN-ω has the potential for serving as a useful therapeutic agent to prevent FMDV or other viral outbreaks in pigs.


Assuntos
Antivirais/farmacologia , Vírus da Febre Aftosa/efeitos dos fármacos , Vírus da Febre Aftosa/crescimento & desenvolvimento , Interferon Tipo I/farmacologia , Proteínas Recombinantes de Fusão/farmacologia , Animais , Efeito Citopatogênico Viral , Interferon Tipo I/genética , Proteínas Recombinantes de Fusão/genética , Suínos
10.
J Theor Biol ; 460: 209-219, 2019 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-30273575

RESUMO

In mainland China, there are three prevalent serotypes of foot and mouth disease virus (FMDV) and they circulate in different susceptible animals, which respond to viral infection in various ways and present different prevalent features. Although powerful control measures are carried on regularly, the epidemic are still prevalent in livestock. Therefore it is essential to assess the disease trends of foot and mouth disease (FMD) in domestic animals in mainland China. The participation of contaminated environment in the transmission dynamics has been confirmed in laboratory research and it can alter the conditions for FMDV invasion and persistence. So environment transmission plays a key role in disease spreading process. In this paper, we establish a dynamical switching model with environment transmission to investigate the relevant internal force mechanism with respect to the threshold switching, effect of saturation of the quantity of FMDV in environment, and prevalent characteristics of the disease in mainland China. Through the dynamical analysis of the model, we understand that under different conditions, there may appear coexistence of one, two, three or even four steady states, and bistability might occur, showing that the development trend of epidemic not only depends on the model parameters, but also is associated with the initial condition. We further study the influence of key parameters on the dynamical behavior and classify the parameter space into several regions with different composition patterns. Applying the model to assess the development trend of FMD in livestock in mainland China, we find that under certain conditions, some of the serotypes persists and some may disappear, thus we can provide some suggestions for disease control of prevailing serotype.


Assuntos
Vírus da Febre Aftosa/genética , Febre Aftosa/transmissão , Modelos Biológicos , Animais , Animais Domésticos , China , Poluição Ambiental/efeitos adversos , Febre Aftosa/epidemiologia , Vírus da Febre Aftosa/crescimento & desenvolvimento , Sorogrupo
11.
mSphere ; 3(6)2018 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-30541776

RESUMO

The pathogenesis of foot-and-mouth disease virus (FMDV) in cattle was investigated through early and late stages of infection by use of an optimized experimental model for controlled contact exposure. Time-limited exposure of cattle to FMDV-infected pigs led to primary FMDV infection of the nasopharyngeal mucosa in both vaccinated and nonvaccinated cattle. In nonvaccinated cattle, the infection generalized rapidly to cause clinical disease, without apparent virus amplification in the lungs prior to establishment of viremia. Vaccinated cattle were protected against clinical disease and viremia; however, all vaccinated cattle were subclinically infected, and persistent infection occurred at similarly high prevalences in both animal cohorts. Infection dynamics in cattle were consistent and synchronous and comparable to those of simulated natural and needle inoculation systems. However, the current experimental model utilizes a natural route of virus exposure and is therefore superior for investigations of disease pathogenesis and host response. Deep sequencing of viruses obtained during early infection of pigs and cattle indicated that virus populations sampled from sites of primary infection were markedly more diverse than viruses from vesicular lesions of cattle, suggesting the occurrence of substantial bottlenecks associated with vesicle formation. These data expand previous knowledge of FMDV pathogenesis in cattle and provide novel insights for validation of inoculation models of bovine FMD studies.IMPORTANCE Foot-and-mouth disease virus (FMDV) is an important livestock pathogen that is often described as the greatest constraint to global trade in animal products. The present study utilized a standardized pig-to-cow contact exposure model to demonstrate that FMDV infection of cattle initiates in the nasopharyngeal mucosa following natural virus exposure. Furthermore, this work confirmed the role of the bovine nasopharyngeal mucosa as the site of persistent FMDV infection in vaccinated and nonvaccinated cattle. The critical output of this study validates previous studies that have used simulated natural inoculation models to characterize FMDV pathogenesis in cattle and emphasizes the importance of continued research of the unique virus-host interactions that occur within the bovine nasopharynx. Specifically, vaccines and biotherapeutic countermeasures designed to prevent nasopharyngeal infection of vaccinated animals could contribute to substantially improved control of FMDV.


Assuntos
Doenças dos Bovinos/virologia , Transmissão de Doença Infecciosa , Epitélio/virologia , Vírus da Febre Aftosa/crescimento & desenvolvimento , Vírus da Febre Aftosa/isolamento & purificação , Febre Aftosa/virologia , Nasofaringe/virologia , Animais , Bovinos , Febre Aftosa/transmissão , Suínos , Doenças dos Suínos/virologia
12.
Sci Rep ; 8(1): 5545, 2018 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-29615727

RESUMO

Internal ribosome entry site (IRES) elements are RNA regions that recruit the translation machinery internally. Here we investigated the conformational changes and RNA dynamics of a picornavirus IRES upon incubation with distinct ribosomal fractions. Differential SHAPE analysis of the free RNA showed that nucleotides reaching the final conformation on long timescales were placed at domains 4 and 5, while candidates for long-range interactions were located in domain 3. Salt-washed ribosomes induced a fast RNA local flexibility modification of domains 2 and 3, while ribosome-associated factors changed domains 4 and 5. Consistent with this, modeling of the three-dimensional RNA structure indicated that incubation of the IRES with native ribosomes induced a local rearrangement of the apical region of domain 3, and a reorientation of domains 4 and 5. Furthermore, specific motifs within domains 2 and 3 showed a decreased flexibility upon incubation with ribosomal subunits in vitro, and presence of the IRES enhanced mRNA association to the ribosomal subunits in whole cell lysates. The finding that RNA modules can provide direct IRES-ribosome interaction suggests that linking these motifs to additional sequences able to recruit trans-acting factors could be useful to design synthetic IRESs with novel activities.


Assuntos
Vírus da Febre Aftosa/genética , Sítios Internos de Entrada Ribossomal/genética , Biossíntese de Proteínas , RNA Viral/química , RNA Viral/genética , Ribossomos/metabolismo , Sequência de Bases , Vírus da Febre Aftosa/crescimento & desenvolvimento , Vírus da Febre Aftosa/metabolismo , Conformação de Ácido Nucleico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Subunidades Ribossômicas Maiores de Eucariotos/genética , Subunidades Ribossômicas Maiores de Eucariotos/metabolismo , Subunidades Ribossômicas Menores de Eucariotos/genética , Subunidades Ribossômicas Menores de Eucariotos/metabolismo , Ribossomos/genética
13.
J Virol ; 92(5)2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29212943

RESUMO

Productive picornavirus infection requires the hijacking of host cell pathways to aid with the different stages of virus entry, synthesis of the viral polyprotein, and viral genome replication. Many picornaviruses, including foot-and-mouth disease virus (FMDV), assemble capsids via the multimerization of several copies of a single capsid precursor protein into a pentameric subunit which further encapsidates the RNA. Pentamer formation is preceded by co- and posttranslational modification of the capsid precursor (P1-2A) by viral and cellular enzymes and the subsequent rearrangement of P1-2A into a structure amenable to pentamer formation. We have developed a cell-free system to study FMDV pentamer assembly using recombinantly expressed FMDV capsid precursor and 3C protease. Using this assay, we have shown that two structurally different inhibitors of the cellular chaperone heat shock protein 90 (hsp90) impeded FMDV capsid precursor processing and subsequent pentamer formation. Treatment of FMDV permissive cells with the hsp90 inhibitor prior to infection reduced the endpoint titer by more than 10-fold while not affecting the activity of a subgenomic replicon, indicating that translation and replication of viral RNA were unaffected by the drug.IMPORTANCE FMDV of the Picornaviridae family is a pathogen of huge economic importance to the livestock industry due to its effect on the restriction of livestock movement and necessary control measures required following an outbreak. The study of FMDV capsid assembly, and picornavirus capsid assembly more generally, has tended to be focused upon the formation of capsids from pentameric intermediates or the immediate cotranslational modification of the capsid precursor protein. Here, we describe a system to analyze the early stages of FMDV pentameric capsid intermediate assembly and demonstrate a novel requirement for the cellular chaperone hsp90 in the formation of these pentameric intermediates. We show the added complexity involved for this process to occur, which could be the basis for a novel antiviral control mechanism for FMDV.


Assuntos
Proteínas do Capsídeo/metabolismo , Vírus da Febre Aftosa/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Chaperonas Moleculares/metabolismo , Montagem de Vírus , Proteases Virais 3C , Animais , Benzoquinonas/farmacologia , Proteínas do Capsídeo/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular , Sistema Livre de Células , Cricetinae , Cisteína Endopeptidases/genética , Cisteína Endopeptidases/metabolismo , Febre Aftosa/metabolismo , Vírus da Febre Aftosa/genética , Vírus da Febre Aftosa/crescimento & desenvolvimento , Proteínas de Choque Térmico HSP90/efeitos dos fármacos , Isoxazóis/farmacologia , Lactamas Macrocíclicas/farmacologia , Precursores de Proteínas/efeitos dos fármacos , Precursores de Proteínas/metabolismo , Processamento de Proteína Pós-Traducional , RNA Viral/genética , RNA Viral/metabolismo , Resorcinóis/farmacologia , Proteínas Virais/efeitos dos fármacos , Proteínas Virais/genética , Proteínas Virais/metabolismo , Montagem de Vírus/genética , Montagem de Vírus/fisiologia , Replicação Viral
14.
J Leukoc Biol ; 102(5): 1237-1247, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28811319

RESUMO

Galectin-8 (Gal-8) is a mammalian ß-galactoside-binding lectin, endowed with proinflammatory properties. Given its capacity to enhance antigen-specific immune responses in vivo, we investigated whether Gal-8 was also able to promote APC activation to sustain T cell activation after priming. Both endogenous [dendritic cells (DCs)] and bone marrow-derived DCs (BMDCs) treated with exogenous Gal-8 exhibited a mature phenotype characterized by increased MHC class II (MHCII), CD80, and CD86 surface expression. Moreover, Gal-8-treated BMDCs (Gal-8-BMDCs) stimulated antigen-specific T cells more efficiently than immature BMDCs (iBMDCs). Proinflammatory cytokines IL-3, IL-2, IL-6, TNF, MCP-1, and MCP-5, as well as growth factor G-CSF, were augmented in Gal-8-BMDC conditioned media, with IL-6 as the most prominent. Remarkably, BMDCs from Gal-8-deficient mice (Lgals8-/- BMDC) displayed reduced CD86 and IL-6 expression and an impaired ability to promote antigen-specific CD4 T cell activation. To test if Gal-8-induced activation correlates with the elicitation of an effective immune response, soluble Gal-8 was coadministrated with antigen during immunization of BALB/cJ mice in the experimental foot-and-mouth disease virus (FMDV) model. When a single dose of Gal-8 was added to the antigen formulation, an increased specific and neutralizing humoral response was developed, sufficient to enhance animal protection upon viral challenge. IL-6 and IFN-γ, as well as lymphoproliferative responses, were also incremented in Gal-8/antigen-immunized animals only at 48 h after immunization, suggesting that Gal-8 induces the elicitation of an inflammatory response at an early stage. Taking together, these findings argue in favor of the use of Gal-8 as an immune-stimulator molecule to enhance the adaptive immune response.


Assuntos
Apresentação de Antígeno , Antígenos Virais/imunologia , Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/imunologia , Febre Aftosa/imunologia , Galectinas/imunologia , Imunidade Adaptativa , Animais , Antígenos Virais/administração & dosagem , Antígenos Virais/genética , Linfócitos T CD4-Positivos/virologia , Quimiocina CCL2/genética , Quimiocina CCL2/imunologia , Células Dendríticas/virologia , Febre Aftosa/genética , Febre Aftosa/prevenção & controle , Febre Aftosa/virologia , Vírus da Febre Aftosa/crescimento & desenvolvimento , Vírus da Febre Aftosa/imunologia , Galectinas/genética , Galectinas/farmacologia , Regulação da Expressão Gênica , Fator Estimulador de Colônias de Granulócitos/genética , Fator Estimulador de Colônias de Granulócitos/imunologia , Imunização , Interleucina-2/genética , Interleucina-2/imunologia , Interleucina-3/genética , Interleucina-3/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Ativação Linfocitária , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Quimioatraentes de Monócitos/genética , Proteínas Quimioatraentes de Monócitos/imunologia , Transdução de Sinais , Fatores de Tempo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
15.
Viruses ; 9(8)2017 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-28820470

RESUMO

Foot-and-mouth disease virus (FMDV) causes a highly contagious disease with catastrophic economic impact for affected countries. BHK21 suspension cells are preferred for the industrial production of FMDV vaccine antigen, but not all virus strains can be successfully propagated in these cells. Serotype Asia-1 is often affected by this phenomenon. In this study, the Asia-1 strain Shamir was used to examine viral, cellular and environmental factors that contribute to resistance to cell culture infection. Cell media composition, pH and ammonium chloride concentration did not affect Asia-1 differently than other serotypes. Virus replication after transfection of viral genome was not impaired, but the adhesion to the cells was markedly reduced for Asia-1 in comparison to serotype A. The Asia-1 Shamir virus was successfully adapted to grow in the resistant cells by using a closely related but susceptible cell line. Sequence analysis of the adapted virus revealed two distinct mutations in the capsid protein VP1 that might mediate cell attachment and entry.


Assuntos
Capsídeo/metabolismo , Vírus da Febre Aftosa/fisiologia , Febre Aftosa/virologia , Adaptação Biológica , Animais , Células CHO , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Cricetulus , Vírus da Febre Aftosa/genética , Vírus da Febre Aftosa/crescimento & desenvolvimento , Replicação Viral
16.
Infect Genet Evol ; 54: 324-329, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28734763

RESUMO

Foot-and-mouth disease virus (FMDV) capsid precursor protein P1-2A is cleaved by viral-encoded 3C protease (3Cpro) to generate VP0, VP3, VP1 and 2A proteins. It was reported earlier that substitution of a single amino acid residue within the 2A peptide sequence (L2P) blocked the 3Cpro mediated VP1/2A cleavage and produced 'self-tagged' FMDV particles containing uncleaved 2A-peptide. To determine whether the uncleaved 2A-peptide can function as a target structure to harbour and display exogenous epitope on FMDV particles, a full-length FMDV cDNA clone containing a HA-tag within the uncleaved 2A-peptide sequence was constructed. Subsequently, chimeric marker FMDV, displaying a HA-tag on the viral surface was rescued through reverse genetics approach. The 2A-HA epitope tag-inserted recombinant chimeric FMDV serotype O was genetically stable through up to ten serial passages in cell culture and exhibited growth properties similar to the parental virus. Furthermore the surface displayed HA-epitope tag was able to react with anti-HA antibodies as determined by various immuno-assays. The results from our study suggest that the uncleaved 2A-peptide of FMDV is suitable to present foreign antigenic epitopes on the surface of FMD virion.


Assuntos
Proteínas do Capsídeo/genética , Vírus da Febre Aftosa/crescimento & desenvolvimento , Febre Aftosa/virologia , Peptídeos/genética , Vírion/crescimento & desenvolvimento , Proteases Virais 3C , Substituição de Aminoácidos , Animais , Proteínas do Capsídeo/química , Linhagem Celular , Cisteína Endopeptidases/metabolismo , Epitopos/genética , Epitopos/imunologia , Vírus da Febre Aftosa/genética , Vírus da Febre Aftosa/imunologia , Peptídeos/imunologia , Inoculações Seriadas , Proteínas Virais/metabolismo , Vírion/genética
17.
J Virol ; 91(15)2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28515297

RESUMO

Foot-and-mouth disease virus (FMDV) RNA-dependent RNA polymerase (RdRp) (3Dpol) catalyzes viral RNA synthesis. Its characteristic low fidelity and absence of proofreading activity allow FMDV to rapidly mutate and adapt to dynamic environments. In this study, we used the structure of FMDV 3Dpol in combination with previously reported results from similar picornaviral polymerases to design point mutations that would alter replication fidelity. In particular, we targeted Trp237 within conserved polymerase motif A because of the low reversion potential inherent in the single UGG codon. Using biochemical and genetic tools, we show that the replacement of tryptophan 237 with phenylalanine imparts higher fidelity, but replacements with isoleucine and leucine resulted in lower-fidelity phenotypes. Viruses containing these W237 substitutions show in vitro growth kinetics and plaque morphologies similar to those of the wild-type (WT) A24 Cruzeiro strain in BHK cells, and both high- and low-fidelity variants retained fitness during coinfection with the wild-type virus. The higher-fidelity W237F (W237FHF) mutant virus was more resistant to the mutagenic nucleoside analogs ribavirin and 5-fluorouracil than the WT virus, whereas the lower-fidelity W237I (W237ILF) and W237LLF mutant viruses exhibited lower ribavirin resistance. Interestingly, the variant viruses showed heterogeneous and slightly delayed growth kinetics in primary porcine kidney cells, and they were significantly attenuated in mouse infection experiments. These data demonstrate, for a single virus, that either increased or decreased RdRp fidelity attenuates virus growth in animals, which is a desirable feature for the development of safer and genetically more stable vaccine candidates.IMPORTANCE Foot-and-mouth disease (FMD) is the most devastating disease affecting livestock worldwide. Here, using structural and biochemical analyses, we have identified FMDV 3Dpol mutations that affect polymerase fidelity. Recombinant FMDVs containing substitutions at 3Dpol tryptophan residue 237 were genetically stable and displayed plaque phenotypes and growth kinetics similar to those of the wild-type virus in cell culture. We further demonstrate that viruses harboring either a W237FHF substitution or W237ILF and W237LLF mutations were highly attenuated in animals. Our study shows that obtaining 3Dpol fidelity variants by protein engineering based on polymerase structure and function could be exploited for the development of attenuated FMDV vaccine candidates that are safer and more stable than strains obtained by selective pressure via mutagenic nucleotides or adaptation approaches.


Assuntos
Antígenos Virais/genética , Antígenos Virais/metabolismo , Vírus da Febre Aftosa/enzimologia , Vírus da Febre Aftosa/patogenicidade , Engenharia de Proteínas , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Substituição de Aminoácidos , Animais , Antivirais , Células Cultivadas , Cricetinae , Análise Mutacional de DNA , Modelos Animais de Doenças , Farmacorresistência Viral , Fluoruracila/farmacologia , Febre Aftosa/patologia , Febre Aftosa/virologia , Vírus da Febre Aftosa/genética , Vírus da Febre Aftosa/crescimento & desenvolvimento , Camundongos , Mutagênese Sítio-Dirigida , Mutação Puntual , Ribavirina/farmacologia , Suínos , Triptofano/genética , Triptofano/metabolismo , Ensaio de Placa Viral
18.
Virol J ; 14(1): 89, 2017 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-28464897

RESUMO

BACKGROUND: Understanding the mechanisms of attenuation and virulence of foot-and-mouth disease virus (FMDV) in the natural host species is critical for development of next-generation countermeasures such as live-attenuated vaccines. Functional genomics analyses of FMDV have identified few virulence factors of which the leader proteinase (Lpro) is the most thoroughly investigated. Previous work from our laboratory has characterized host factors in cattle inoculated with virulent FMDV and attenuated mutant strains with transposon insertions within Lpro. METHODS: In the current study, the characteristics defining virulence of FMDV in cattle were further investigated by comparing the pathogenesis of a mutant, attenuated strain (FMDV-Mut) to the parental, virulent virus from which the mutant was derived (FMDV-WT). The only difference between the two viruses was an insertion mutation in the inter-AUG region of the leader proteinase of FMDV-Mut. All cattle were infected by simulated-natural, aerosol inoculation. RESULTS: Both viruses were demonstrated to establish primary infection in the nasopharyngeal mucosa with subsequent dissemination to the lungs. Immunomicroscopic localization of FMDV antigens indicated that both viruses infected superficial epithelial cells of the nasopharynx and lungs. The critical differences between the two viruses were a more rapid establishment of infection by FMDV-WT and quantitatively greater virus loads in secretions and infected tissues compared to FMDV-Mut. The slower replicating FMDV-Mut established a subclinical infection that was limited to respiratory epithelial sites, whereas the faster replication of FMDV-WT facilitated establishment of viremia, systemic dissemination of infection, and clinical disease. CONCLUSION: The mutant FMDV was capable of achieving all the same early pathogenesis landmarks as FMDV-WT, but was unable to establish systemic infection. The precise mechanism of attenuation remains undetermined; but current data suggests that the impaired replication of the mutant is more responsible for attenuation than differences in host immunological factors. These results complement previous studies by providing data of high-granularity describing tissue-specific tropism of FMDV and by demonstrating microscopic localization of virulent and attenuated clones of the same field-strain FMDV.


Assuntos
Doenças dos Bovinos/virologia , Vírus da Febre Aftosa/patogenicidade , Febre Aftosa/virologia , Virulência , Aerossóis , Animais , Bovinos , Células Epiteliais/patologia , Células Epiteliais/virologia , Febre Aftosa/imunologia , Febre Aftosa/patologia , Vírus da Febre Aftosa/genética , Vírus da Febre Aftosa/crescimento & desenvolvimento , Vírus da Febre Aftosa/isolamento & purificação , Pulmão/virologia , Mutagênese Insercional , Nasofaringe/patologia , Nasofaringe/virologia , RNA Viral/isolamento & purificação , Vacinas Atenuadas/imunologia , Proteínas Estruturais Virais , Fatores de Virulência , Replicação Viral
19.
Virus Genes ; 53(3): 340-351, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28364140

RESUMO

The Jumonji C-domain containing protein 6 (JMJD6) has had a convoluted history, and recent reports indicating a multifactorial role in foot-and-mouth disease virus (FMDV) infection have further complicated the functionality of this protein. It was first identified as the phosphatidylserine receptor on the cell surface responsible for recognizing phosphatidylserine on the surface of apoptotic cells resulting in their engulfment by phagocytic cells. Subsequent study revealed a nuclear subcellular localization, where JMJD6 participated in lysine hydroxylation and arginine demethylation of histone proteins and other non-histone proteins. Interestingly, to date, JMDJ6 remains the only known arginine demethylase with a growing list of known substrate molecules. These conflicting associations rendered the subcellular localization of JMJD6 to be quite nebulous. Further muddying this area, two different groups illustrated that JMJD6 could be induced to redistribute from the cell surface to the nucleus of a cell. More recently, JMJD6 was demonstrated to be a host factor contributing to the FMDV life cycle, where it was not only exploited for its arginine demethylase activity, but also served as an alternative virus receptor. This review attempts to coalesce these divergent roles for a single protein into one cohesive account. Given the diverse functionalities already characterized for JMJD6, it is likely to continue to be a confounding protein resulting in much contention going into the near future.


Assuntos
Vírus da Febre Aftosa/metabolismo , Febre Aftosa/virologia , Histona Desmetilases com o Domínio Jumonji/fisiologia , Replicação Viral/fisiologia , Animais , Arginina/metabolismo , Membrana Celular/metabolismo , Núcleo Celular/enzimologia , Núcleo Celular/genética , Febre Aftosa/enzimologia , Febre Aftosa/genética , Vírus da Febre Aftosa/genética , Vírus da Febre Aftosa/crescimento & desenvolvimento , Histona Desmetilases/metabolismo , Histonas , Interações Hospedeiro-Patógeno , Humanos , Lisina/metabolismo , Fagócitos/virologia , Fosfatidilserinas/metabolismo , Pró-Colágeno-Lisina 2-Oxoglutarato 5-Dioxigenase/metabolismo , Ligação Proteica , Receptores de Superfície Celular/metabolismo
20.
Virus Res ; 233: 105-112, 2017 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-28322918

RESUMO

Antiviral agents are increasingly considered an option for veterinary medicine. An understanding of their mechanism of activity is important to plan their administration either as monotherapy or in combination with other agents. Previous studies have shown that the broad spectrum antiviral agent favipiravir (T-705) and its derivatives T-1105 and T-1106 are efficient inhibitors of foot-and-mouth disease virus (FMDV) replication in cell culture and in vivo. However, no mechanism for their activity against FMDV has been proposed. In the present study we show that favipiravir (T-705) can act as a lethal mutagen for FMDV in cell culture. Evidence includes virus extinction associated with increase in mutation frequency in the mutant spectrum of 860 residues of the 3D (polymerase)-coding region, and a decrease of specific infectivity while the consensus nucleotide sequence of the region analyzed remained invariant. The mutational spectrum evoked by favipiravir differs from that observed with other viruses in that no predominant transition type is observed, indicating that a movement towards A,U- or G,C-rich regions of sequence space is not a prerequisite for virus extinction. We discuss prospects for the use of favipiravir to assist in the control of FMDV, and its possible broader use in veterinary medicine as an extension of its current status as antiviral agent for human influenza virus.


Assuntos
Amidas/farmacologia , Antivirais/farmacologia , Vírus da Febre Aftosa/efeitos dos fármacos , Mutagênicos/farmacologia , Pirazinas/farmacologia , Replicação Viral/efeitos dos fármacos , Animais , Linhagem Celular , Cricetulus , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/virologia , Vírus da Febre Aftosa/genética , Vírus da Febre Aftosa/crescimento & desenvolvimento , Concentração Inibidora 50 , Mutagênese , Taxa de Mutação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA