Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Virol ; 97(8): e0018123, 2023 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-37565750

RESUMO

Vacuolar protein sorting 28 (Vps28), a component of the ESCRT-I (endosomal sorting complex required for transport I), plays an important role in the pathogen life cycle. Here, we investigated the reciprocal regulation between Vps28 and the foot-and-mouth disease virus (FMDV). Overexpression of Vps28 decreased FMDV replication. On the contrary, the knockdown of Vps28 increased viral replication. Subsequently, the mechanistic study showed that Vps28 destabilized the replication complex (RC) by associating with 3A rather than 2C protein. In addition, Vps28 targeted FMDV VP0, VP1, and VP3 for degradation to inhibit viral replication. To counteract this, FMDV utilized tactics to restrict Vps28 to promote viral replication. FMDV degraded Vps28 mainly through the ubiquitin-proteasome pathway. Additional data demonstrated that 2B and 3A proteins recruited E3 ubiquitin ligase tripartite motif-containing protein 21 to degrade Vps28 at Lys58 and Lys25, respectively, and FMDV 3Cpro degraded Vps28 through autophagy and its protease activity. Meantime, the 3Cpro-mediated Vps28 degradation principally alleviated the ability to inhibit viral propagation. Intriguingly, we also demonstrated that the N-terminal and C-terminal domains of Vps28 were responsible for the suppression of FMDV replication, which suggested the elaborated counteraction between FMDV and Vps28. Collectively, our results first investigate the role of ESCRTs in host defense against picornavirus and unveil underlying strategies utilized by FMDV to evade degradation machinery for triumphant propagation. IMPORTANCE ESCRT machinery plays positive roles in virus entry, replication, and budding. However, little has been reported on its negative regulation effects during viral infection. Here, we uncovered the novel roles of ESCRT-I subunit Vps28 on FMDV replication. The data indicated that Vps28 destabilized the RC and impaired viral structural proteins VP0, VP1, and VP3 to inhibit viral replication. To counteract this, FMDV hijacked intracellular protein degradation pathways to downregulate Vps28 expression and thus promoted viral replication. Our findings provide insights into how ESCRT regulates pathogen life cycles and elucidate additional information regarding FMDV counteraction of host antiviral activity.


Assuntos
Vírus da Febre Aftosa , Febre Aftosa , Animais , Vírus da Febre Aftosa/metabolismo , Proteínas Virais/metabolismo , Transdução de Sinais , Transporte Proteico , Replicação Viral/fisiologia
2.
J Med Virol ; 91(6): 941-948, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30701562

RESUMO

Foot-and-mouth disease (FMD) is an acute and febrile infectious disease, which can cause great economic losses. Virus-like particles (VLPs) as an advantageous antigen can induce significant specific immune response. To improve immunity of VLPs, especially, make it induce persistent immune response, the hollow mesoporous silica nanoparticles (HMSNs) as a potential nano-adjuvant were synthesized and loaded the FMD virus (FMDV) VLPs. They were injected into guinea pigs and the specific immune response was detected. The results confirmed that HMSNs/VLPs can induce persistent humoral immunity with high-level antibody titer for more than three months. HMSNs also improve the T-lymphocyte proliferation and IFN-γ induced by FMDV VLPs, and provides the ideal protection against FMDV challenge. These consequences indicated that HMSNs were good protein delivery vehicle and potential nano-adjuvant of vaccines.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Anticorpos Antivirais/sangue , Febre Aftosa/prevenção & controle , Nanopartículas/administração & dosagem , Dióxido de Silício/administração & dosagem , Vacinas Virais/imunologia , Adjuvantes Imunológicos/química , Animais , Anticorpos Neutralizantes/imunologia , Antígenos Virais/imunologia , Febre Aftosa/imunologia , Vírus da Febre Aftosa , Cobaias , Imunidade Humoral , Nanopartículas/química , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Vacinas de Partículas Semelhantes a Vírus/imunologia , Vacinas Virais/administração & dosagem
3.
Appl Microbiol Biotechnol ; 103(7): 3015-3024, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30783719

RESUMO

Foot-and-mouth disease (FMD) is a highly contagious disease that affects all susceptible cloven-hoofed animals, resulting in considerable economic losses to animal industries worldwide. Numerous categories of enzyme-linked immunosorbent assays (ELISA) have been developed and widely used to evaluate herd immunity. Manufacturing inactivated FMD virus (FMDV) as a diagnostic antigen requires a facility with a high level of biosafety, but this requirement raises concern on viral leakage. In our previous study, bacterium-original FMD virus-like particles (VLPs) resemble the authentic FMDV and induce protective immunity against homologous viral challenges, thereby demonstrating that they are sufficiently safe without limitations on biosafety facilities and easily prepared. Herein, we developed a competitive ELISA (cELISA) based on FMDV-VLPs as a diagnostic antigen to evaluate herd immunity. The criterion of this cELISA was determined by detecting panels of positive sera with different antibody titers and negative sera. The working parameter of cELISA was optimized, and samples with a percentage inhibition of ≥ 50% were considered positive. The specificity of cELISA to test 277 serum samples with various antibody titers was 100%, and the sensitivity reached 96%. The coincidence rates of cELISA with a VDPro® FMDV and a PrioCHECK® FMDV type O antibody ELISA kit were 97.8% and 98.2%, respectively. Repeatability tests demonstrated that the coefficients of variation within and between runs were less than 7% and 14%, respectively. Our data demonstrated that cELISA based on bacterium-original VLPs had high specificity, sensitivity, and reproducibility. The cELISA could also be used for evaluating vaccination herd immunity effects, especially in developing countries.


Assuntos
Anticorpos Antivirais/sangue , Ensaio de Imunoadsorção Enzimática/métodos , Vírus da Febre Aftosa/imunologia , Febre Aftosa/diagnóstico , Animais , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/imunologia , Bovinos , Imunidade Coletiva , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Sorogrupo , Ovinos , Suínos
4.
Vaccine ; 42(18): 3789-3801, 2024 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-38714448

RESUMO

Inactivated vaccines lack the capability to serologically differentiate between infected and vaccinated animals, thereby impeding the effective eradication of pathogen. Conversely, vaccines based on virus-like particles (VLPs) emulate natural viruses in both size and antigenic structure, presenting a promising alternative to overcome these limitations. As the complexity of swine infectious diseases increases, the increase of vaccine types and doses may intensify the stress response. This exacerbation can lead to diminished productivity, failure of immunization, and elevated costs. Given the critical dynamics of co-infection and the clinically indistinguishable symptoms associated with foot-and-mouth disease virus (FMDV) and senecavirus A (SVA), there is a dire need for an efficacious intervention. To address these challenges, we developed a combined vaccine composed of three distinct VLPs, specifically designed to target SVA and FMDV serotypes O and A. Our research demonstrates that this trivalent VLP vaccine induces antigen-specific and robust serum antibody responses, comparable to those produced by the respective monovalent vaccines. Moreover, the immune sera from the combined VLP vaccine strongly neutralized FMDV type A and O, and SVA, with neutralization titers comparable to those of the individual vaccines, indicating a high level of immunogenic compatibility among the three VLP components. Importantly, the combined VLPs vaccines-immunized sera conferred efficient protection against single or mixed infections with FMDV type A and O, and SVA viruses in pigs. In contrast, individual vaccines could only protect pigs against homologous virus infections and not against heterologous challenges. This study presents a novel combined vaccines candidate against FMD and SVA, and provides new insights for the development of combination vaccines for other viral swine diseases.


Assuntos
Anticorpos Neutralizantes , Anticorpos Antivirais , Vírus da Febre Aftosa , Febre Aftosa , Picornaviridae , Doenças dos Suínos , Vacinas de Partículas Semelhantes a Vírus , Vacinas Virais , Animais , Vacinas de Partículas Semelhantes a Vírus/imunologia , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Febre Aftosa/prevenção & controle , Febre Aftosa/imunologia , Vírus da Febre Aftosa/imunologia , Suínos , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/sangue , Vacinas Virais/imunologia , Vacinas Virais/administração & dosagem , Doenças dos Suínos/prevenção & controle , Doenças dos Suínos/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/sangue , Camundongos , Picornaviridae/imunologia , Infecções por Picornaviridae/prevenção & controle , Infecções por Picornaviridae/imunologia , Infecções por Picornaviridae/veterinária , Feminino , Vacinas Combinadas/imunologia , Vacinas Combinadas/administração & dosagem , Coinfecção/prevenção & controle , Coinfecção/imunologia
5.
Vet Microbiol ; 298: 110237, 2024 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-39217891

RESUMO

Foot-and-mouth disease virus (FMDV), a member of picornavirus, can enter into host cell via macropinocytosis. Although it is known that receptor tyrosine kinases (RTKs) play a crucial role in FMDV macropinocytic entry, the specific RTK responsible for regulating this process and the intricacies of RTK-mediated downstream signaling remain to be elucidated. Here, we conducted a screening of RTK inhibitors to assess their efficacy against FMDV. Our findings revealed that two compounds specifically targeting fibroblast growth factor receptor 1 (FGFR1) and FMS-like tyrosine kinase 3 (FLT3) significantly disrupted FMDV entry. Furthermore, additional evaluation through gene knockdown and overexpression confirmed the promotion effect of FGFR1 and FLT3 on FMDV entry. Interestingly, we discovered that the increasement of FMDV entry facilitated by FGFR1 and FLT3 can be ascribed to increased macropinocytic uptake. Additionally, in-depth mechanistic study demonstrated that FGFR1 interacts with FMDV VP3 and undergoes phosphorylation during FMDV entry. Furthermore, the FGFR1 inhibitor inhibited FMDV-induced activation of p21-activated kinase 1 (PAK1) on Thr212 and Thr423 sites. Consistent with these findings, the ectopic expression of FGFR1 resulted in a concomitant increase in phosphorylation level of PAK1 on Thr212 and Thr423 sites. Taken together, our findings represent the initial exploration of FGFR1's involvement in FMDV macropinocytic entry, providing novel insights with potential implications for the development of antiviral strategies.

6.
Front Microbiol ; 14: 1112641, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36819011

RESUMO

Introduction: Foot-and-mouth disease virus (FMDV) infects the host by invading mucosal epithelial cells of the respiratory or digestive tract. Therefore, establishing a specific antiviral mucosal immune barrier can effectively block viral invasion. Methods: We evaluated local mucosal and systemic immune responses elicited by intranasal immunization of mice with foot-and-mouth disease (FMD) calcium phosphate mineralized virus-like particles (CaP-VLPs) and tested whether three commercial mucosal adjuvants enhanced the immunogenicity of the antigen. The biosafety of the vaccine was verified through gross observation and pathological analysis of the lungs. Results: CaP-VLPs effectively induced secretion of IgA (sIgA) from multiple sites in mouse mucosa and produced anti-FMD-specific IgG in the serum. Splenic lymphocytes specifically proliferated and secreted IFN-γ following antigen stimulation, indicating the vaccine can induce a certain level of cellular immune response. Finally, the pathological examination confirmed that CaP-VLPs did not cause substantial damage to the lungs of animals after immunization via mucosal administration. Notably, the vaccine mixed with S adjuvant increased the content of sIgA and serum IgG, and the high level of IgG in serum was maintained at least 7 weeks. Discussion: Overall, this study reveals that FMD CaP-VLPs can induce good local mucosal immune and systemic immune response through intranasal immunization, and the immune response was specifically enhanced by S adjuvant. These data support that CaP-VLPs-S as a candidate mucosal vaccine for the prevention of FMD vaccine infection.

7.
J Mater Chem B ; 10(15): 2853-2864, 2022 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-35319039

RESUMO

Virus-like particle (VLPs) vaccines have been extensively studied due to their good immunogenicity and safety; however, they highly rely on cold-chain storage and transportation. Nanotechnology of bio-mineralization as a useful strategy has been employed to improve the thermal stability and immunogenicity of VLPs. A zeolitic imidazole framework (ZIF-8), a core-shell structured nanocomposite, was applied to encapsulate foot-and-mouth disease virus (FMDV) VLPs. It was found that the ZIF-8 shell enhanced the heat resistance of VLPs and promoted their ability to be taken up by cells and escape from lysosomes. The VLPs-ZIF-8 easily activated antigen-presenting cells (APCs), triggered higher secretion levels of cytokines, and elicited stronger immune responses than VLPs alone even after being treated at 37 °C for 7 days. This platform has good potential in the development of VLP-based vaccine products without transportation.


Assuntos
Vírus da Febre Aftosa , Estruturas Metalorgânicas , Nanopartículas , Vacinas de Partículas Semelhantes a Vírus , Animais , Imunidade
8.
J Virol Methods ; 300: 114406, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34915087

RESUMO

Foot-and-mouth disease (FMD) is the highly contagious disease of cloven-hoofed animal that brings considerable economic losses to the animal husbandry. So FMD surveillance which relying on accurate diagnosis is important. Most producing the diagnostic antigen of inactivated FMD virus (FMDV) requires facilities with high biosafety. In our previous studies, virus-like particles(VLPs) resembled the structures of natural virus particles. Here, we established a competitive ELISA (cELISA) method for the detection of antibodies against serotype A FMDV based on serotype A FMDV-VLPs. Via detecting different positive serum and negative serum with different titers, and comparing with different commercial ELISA kits. The specificity and sensitivity of the assay were 100 % and 98 %, respectively. The coincidence rate using the PrioCHECK® FMDV Type A antibody ELISA kit and Liquid-phase blocking (LPB) ELISA were 95.30 % and 92.2 %. Repetitive experiments showed that variation coefficient of intra-batch and inter-batch were less than 9 % and 13 %. The result demonstrated that cELISA based on VLPs from prokaryotic system is highly specific, sensitive and reproducible. The cELISA could also be used to assess the immune responses of serotype A FMDV, especially in developing countries.


Assuntos
Vírus da Febre Aftosa , Febre Aftosa , Animais , Anticorpos Antivirais , Ensaio de Imunoadsorção Enzimática/métodos , Febre Aftosa/diagnóstico , Sorogrupo
9.
AMB Express ; 11(1): 7, 2021 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-33409664

RESUMO

Virus-like particles (VLPs) are high-priority antigens with highly ordered repetitive structures, which are similar to natural viral particles. We have developed a competitive enzyme-linked immunosorbent assay (cELISA) for detecting antibodies directed against Senecavirus A (SVA). Our assay utilizes SVA VLPs that were expressed and assembled in an E. coli expression system as the coating antigens. VLPs have better safety and immunogenicity than intact viral particles or peptides. The VLPs-based cELISA was used to test 342 serum samples collected from different pig farms, and the results showed that its specificity and sensitivity were 100% and 94%, respectively. The consistency rates of cELISA with the BIOSTONE AsurDx™ Senecavirus A (SVA) Antibody Test Kit and an indirect immunofluorescent assay were 90.0% and 94.2%, respectively. Therefore, this VLPs-based cELISA can be effectively and reliably used for the detection and discrimination of SVA infection in serum samples.

10.
Viruses ; 13(9)2021 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-34578346

RESUMO

In cells, the contributions of DEAD-box helicases (DDXs), without which cellular life is impossible, are of utmost importance. The extremely diverse roles of the nucleolar helicase DDX21, ranging from fundamental cellular processes such as cell growth, ribosome biogenesis, protein translation, protein-protein interaction, mediating and sensing transcription, and gene regulation to viral manipulation, drew our attention. We designed this project to study virus-host interactions and viral pathogenesis. A pulldown assay was used to investigate the association between foot-and-mouth disease virus (FMDV) and DDX21. Further insight into the DDX21-FMDV interaction was obtained through dual-luciferase, knockdown, overexpression, qPCR, and confocal microscopy assays. Our results highlight the antagonistic feature of DDX21 against FMDV, as it progressively inhibited FMDV internal ribosome entry site (IRES) -dependent translation through association with FMDV IRES domains 2, 3, and 4. To subvert this host helicase antagonism, FMDV degraded DDX21 through its non-structural proteins 2B, 2C, and 3C protease (3Cpro). Our results suggest that DDX21 is degraded during 2B and 2C overexpression and FMDV infection through the caspase pathway; however, DDX21 is degraded through the lysosomal pathway during 3Cpro overexpression. Further investigation showed that DDX21 enhanced interferon-beta and interleukin-8 production to restrict viral replication. Together, our results demonstrate that DDX21 is a novel FMDV IRES trans-acting factor, which negatively regulates FMDV IRES-dependent translation and replication.


Assuntos
RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/metabolismo , Vírus da Febre Aftosa/genética , Vírus da Febre Aftosa/metabolismo , Animais , Linhagem Celular , Febre Aftosa/virologia , Regulação Viral da Expressão Gênica , Técnicas de Silenciamento de Genes , Ribonucleoproteínas Nucleares Heterogêneas , Interações entre Hospedeiro e Microrganismos , Interações Hospedeiro-Patógeno , Humanos , Interferon beta/genética , Sítios Internos de Entrada Ribossomal , Proteína de Ligação a Regiões Ricas em Polipirimidinas , Mapas de Interação de Proteínas , Proteínas Virais/genética , Replicação Viral/genética
11.
Front Immunol ; 12: 616402, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34093518

RESUMO

In addition to ribosomal protein synthesis and protein translation, ribosomal proteins also participate in tumorigenesis and tumor progression, immune responses, and viral replication. Here, we show that ribosomal protein L13 (RPL13) participates in the antiviral immune response induced by foot-and-mouth disease virus (FMDV), inhibiting FMDV replication. The overexpression of RPL13 promoted the induction and activation of the promoters of the nuclear factor-κB (NF-κB) and interferon-ß (IFN-ß) genes, and the expression and protein secretion of the antiviral factor IFN-ß and proinflammatory cytokine interleukin-6 (IL-6). The knockdown of RPL13 had the opposite effects. We also found that the FMDV 3Cpro protease interacts with RPL13, and that its activity reduces the expression of RPL13, thus antagonizing the RPL13-mediated antiviral activity. This study extends our knowledge of the extraribosomal functions of ribosomal proteins and provides new scientific information on cellular antiviral defenses and virus-antagonizing mechanisms.


Assuntos
Vírus da Febre Aftosa/imunologia , Febre Aftosa/imunologia , Febre Aftosa/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Imunidade Inata , Proteínas de Neoplasias/metabolismo , Proteínas Ribossômicas/metabolismo , Animais , Biomarcadores , Linhagem Celular , RNA Helicases DEAD-box/metabolismo , Febre Aftosa/virologia , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Humanos , Proteínas de Neoplasias/genética , Ligação Proteica , Interferência de RNA , RNA Interferente Pequeno/genética , Proteínas Ribossômicas/genética , Transdução de Sinais , Replicação Viral
12.
Vaccines (Basel) ; 8(3)2020 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-32942760

RESUMO

Senecavirus A (SVA) is the pathogen that has recently caused porcine idiopathic vesicular disease (PIVD). The clinical symptoms of PIVD are similar to those of acute foot-and-mouth disease and also can result in the death of newborn piglets, thus entailing economic losses. Vaccine immunization is the most effective way to prevent and control SVA. Among all SVA vaccines reported, only the SVA inactivated vaccine has been successfully developed. However, to ensure the elimination of this pathogen, safer and more effective vaccines are urgently required. A virus-like particles (VLPs)-based vaccine is probably the best alternative to inactivated vaccine. To develop an SVA VLPs vaccine and evaluate its immune effect, a prokaryotic expression system was used to produce SVA capsid protein and assemble VLPs. The VLPs were characterized by affinity chromatography, sucrose density gradient centrifugation, ZetaSizer and transmission electron microscopy. Meanwhile, the SVA CH-HB-2017 strain was used to infect pigs and to determine infection routes and dose. Experimental pigs were then immunized with the SVA VLPs vaccine emulsified in an ISA 201 adjuvant. The results showed that the VLPs vaccine induced neutralizing and specific antibodies at similar levels as an inactivated SVA vaccine after immunization. The level of INF-γ induced by the VLPs vaccine gradually decreased-similar to that of inactivated vaccine. These results indicated that VLPs vaccine may simultaneously cause both cellular and humoral immune responses. Importantly, after the challenge, the VLPs vaccine provided similar levels of protection as the inactivated SVA vaccine. In this study, we successfully obtained novel SVA VLPs and confirmed their highly immunogenicity, thus providing a superior candidate vaccine for defense and elimination of SVA, compared to the inactivated vaccine.

13.
Viruses ; 12(12)2020 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-33255534

RESUMO

DEAD-box helicase 23 (DDX23) is a host nuclear helicase, which is a part of the spliceosomal complex and involved in pre-mRNA splicing. To investigate whether DDX23, an internal ribosomal entry sites transacting factor (ITAF) affects foot-and-mouth disease virus (FMDV) replication and translation through internal ribosome entry site (IRES)-dependent manner. For this, we utilized a pull-down assay, Western blotting, quantitative real-time PCR, confocal microscopy, overexpression and small interfering RNA knockdown, as well as the median tissue culture infective dose. Our findings showed that FMDV infection inhibited DDX23 expression and the overexpression of DDX23 reduced viral replication, however, CRISPR Cas9 knockout/small interfering RNA knockdown increased FMDV replication. FMDV IRES domain III and IV interacted with DDX23, whereas DDX23 interacted with FMDV 3C proteinase and significantly degraded. The enzymatic activity of FMDV 3C proteinase degraded DDX23, whereas FMDV degraded DDX23 via the lysosomal pathway. Additionally, IRES-driven translation was suppressed in DDX23-overexpressing cells, and was enhanced in DDX23 knocked down. Collectively, our results demonstrated that DDX23 negatively affects FMDV IRES-dependent translation, which could be a useful target for the design of antiviral drugs.


Assuntos
Cisteína Endopeptidases/metabolismo , RNA Helicases DEAD-box/metabolismo , Vírus da Febre Aftosa/fisiologia , Febre Aftosa/metabolismo , Febre Aftosa/virologia , Regulação Viral da Expressão Gênica , Proteínas Virais/metabolismo , Replicação Viral , Proteases Virais 3C , Animais , Linhagem Celular , RNA Helicases DEAD-box/química , RNA Helicases DEAD-box/genética , Sítios Internos de Entrada Ribossomal , Lisossomos , Ligação Proteica , Biossíntese de Proteínas , Proteólise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA