Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sheng Wu Gong Cheng Xue Bao ; 40(5): 1548-1558, 2024 May 25.
Artigo em Chinês | MEDLINE | ID: mdl-38783815

RESUMO

In order to generate monoclonal antibodies against the akabane virus (AKAV) N protein, this study employed a prokaryotic expression system to express the AKAV N protein. Following purification, BALB/c mice were immunized, and their splenocytes were fused with mouse myeloma cells (SP2/0) to produce hybridoma cells. The indirect ELISA method was used to screen for positive hybridoma cells. Two specific hybridoma cell lines targeting AKAV N protein, designated as 2C9 and 5E9, were isolated after three rounds of subcloning. Further characterization was conducted through ELISA, Western blotting, and indirect immunofluorescence assay (IFA). The results confirmed that the monoclonal antibodies specifically target AKAV N protein, exhibiting strong reactivity in IFA. Subtype analysis identified the heavy chain of the 2C9 mAb's as IgG2b and its light chain as κ-type; the 5E9 mAb's heavy chain was determined to be IgG1, with a κ-type light chain. Their ELISA titers reached 1:4 096 000. This study successfully developed two monoclonal antibodies targeting AKAV N protein, which lays a crucial foundation for advancing diagnostic methods for akabane disease prevention and control, as well as for studying the function of the AKAV N protein.


Assuntos
Anticorpos Monoclonais , Animais , Feminino , Camundongos , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/imunologia , Escherichia coli/genética , Escherichia coli/metabolismo , Hibridomas/imunologia , Hibridomas/metabolismo , Camundongos Endogâmicos BALB C , Proteínas do Nucleocapsídeo/imunologia , Proteínas do Nucleocapsídeo/genética , Orthobunyavirus/imunologia , Orthobunyavirus/genética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia
2.
Appl Microbiol Biotechnol ; 108(1): 350, 2024 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-38809284

RESUMO

The African swine fever virus (ASFV) has the ability to infect pigs and cause a highly contagious acute fever that can result in a mortality rate as high as 100%. Due to the viral epidemic, the pig industry worldwide has suffered significant financial setbacks. The absence of a proven vaccine for ASFV necessitates the development of a sensitive and reliable serological diagnostic method, enabling laboratories to effectively and expeditiously detect ASFV infection. In this study, four strains of monoclonal antibodies (mAbs) against p72, namely, 5A1, 4C4, 8A9, and 5E10, were generated through recombinant expression of p72, the main capsid protein of ASFV, and immunized mice with it. Epitope localization was performed by truncated overlapping polypeptides. The results indicate that 5A1 and 4C4 recognized the amino acid 20-39 aa, 8A9 and 5E10 are recognized at 263-282 aa, which is consistent with the reported 265-280 aa epitopes. Conserved analysis revealed 20-39 aa is a high conservation of the epitopes in the ASFV genotypes. Moreover, a blocking ELISA assay for detection ASFV antibody based on 4C4 monoclonal antibody was developed and assessed. The receiver-operating characteristic (ROC) was performed to identify the best threshold value using 87 negative and 67 positive samples. The established test exhibited an area under the curve (AUC) of 0.9997, with a 95% confidence interval ranging from 99.87 to 100%. Furthermore, the test achieved a diagnostic sensitivity of 100% (with a 95% confidence interval of 95.72 to 100%) and a specificity of 98.51% (with a 95% confidence interval of 92.02 to 99.92%) when the threshold was set at 41.97%. The inter- and intra-batch coefficient of variation were below 10%, demonstrating the exceptional repeatability of the method. This method can detect the positive standard serum at a dilution as high as 1:512. Subsequently, an exceptional blocking ELISA assay was established with high diagnostic sensitivity and specificity, providing a novel tool for detecting ASFV antibodies. KEY POINTS: • Four strains of ASFV monoclonal antibodies against p72 were prepared and their epitopes were identified. • Blocking ELISA method was established based on monoclonal antibody 4C4 with an identified conservative epitope. • The established blocking ELISA method has a good effect on the detection of ASFV antibody.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Anticorpos Monoclonais , Anticorpos Antivirais , Proteínas do Capsídeo , Ensaio de Imunoadsorção Enzimática , Mapeamento de Epitopos , Animais , Anticorpos Monoclonais/imunologia , Vírus da Febre Suína Africana/imunologia , Vírus da Febre Suína Africana/genética , Ensaio de Imunoadsorção Enzimática/métodos , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Suínos , Febre Suína Africana/diagnóstico , Febre Suína Africana/imunologia , Febre Suína Africana/virologia , Camundongos , Proteínas do Capsídeo/imunologia , Proteínas do Capsídeo/genética , Camundongos Endogâmicos BALB C , Sensibilidade e Especificidade , Epitopos/imunologia
3.
Proc Natl Acad Sci U S A ; 121(10): e2312150121, 2024 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-38412127

RESUMO

African swine fever, one of the major viral diseases of swine, poses an imminent threat to the global pig industry. The high-efficient replication of the causative agent African swine fever virus (ASFV) in various organs in pigs greatly contributes to the disease. However, how ASFV manipulates the cell population to drive high-efficient replication of the virus in vivo remains unclear. Here, we found that the spleen reveals the most severe pathological manifestation with the highest viral loads among various organs in pigs during ASFV infection. By using single-cell-RNA-sequencing technology and multiple methods, we determined that macrophages and monocytes are the major cell types infected by ASFV in the spleen, showing high viral-load heterogeneity. A rare subpopulation of immature monocytes represents the major population infected at late infection stage. ASFV causes massive death of macrophages, but shifts its infection into these monocytes which significantly arise after the infection. The apoptosis, interferon response, and antigen-presentation capacity are inhibited in these monocytes which benefits prolonged infection of ASFV in vivo. Until now, the role of immature monocytes as an important target by ASFV has been overlooked due to that they do not express classical monocyte marker CD14. The present study indicates that the shift of viral infection from macrophages to the immature monocytes is critical for maintaining prolonged ASFV infection in vivo. This study sheds light on ASFV tropism, replication, and infection dynamics, and elicited immune response, which may instruct future research on antiviral strategies.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Suínos , Animais , Vírus da Febre Suína Africana/fisiologia , Baço/patologia , Replicação Viral , Macrófagos/patologia
4.
Int J Biol Macromol ; 254(Pt 3): 127724, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37898252

RESUMO

This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/policies/article-withdrawal.

5.
Microbiol Spectr ; 11(4): e0447222, 2023 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-37428080

RESUMO

Senecavirus A (SVA) is a type of nonenveloped single-stranded, positive-sense RNA virus. The VP2 protein is a structural protein that plays an important role in inducing early and late immune responses of the host. However, its antigenic epitopes have not been fully elucidated. Therefore, defining the B epitopes of the VP2 protein is of great importance to revealing its antigenic characterization. In this study, we analyzed B-cell immunodominant epitopes (IDEs) of the VP2 protein from the SVA strain CH/FJ/2017 using the Pepscan approach and a bioinformatics-based computational prediction method. The following four novel IDEs of VP2 were identified: IDE1, 41TKSDPPSSSTDQPTTT56; IDE2, 145PDGKAKSLQELNEEQW160; IDE3, 161VEMSDDYRTGKNMPF175; and IDE4, 267PYFNGLRNRFTTGT280. Most of the IDEs were highly conserved among the different strains. To our knowledge, the VP2 protein is a major protective antigen of SVA that can induce neutralizing antibodies in animals. Here, we analyzed the immunogenicity and neutralization activity of four IDEs of VP2. Consequently, all four IDEs showed good immunogenicity that could elicit specific antibodies in guinea pigs. A neutralization test in vitro showed that the peptide-specific guinea pig antisera of IDE2 could neutralize SVA strain CH/FJ/2017, and IDE2 was identified as a novel potential neutralizing linear epitope. This is the first time VP2 IDEs have been identified by using the Pepscan method and a bioinformatics-based computational prediction method. These results will help elucidate the antigenic epitopes of VP2 and clarify the basis for immune responses against SVA. IMPORTANCE The clinical symptoms and lesions caused by SVA are indistinguishable from those of other vesicular diseases in pigs. SVA has been associated with recent outbreaks of vesicular disease and epidemic transient neonatal losses in several swine-producing countries. Due to the continuing spread of SVA and the lack of commercial vaccines, the development of improved control strategies is urgently needed. The VP2 protein is a crucial antigen on the capsids of SVA particles. Furthermore, the latest research showed that VP2 could be a promising candidate for the development of novel vaccines and diagnostic tools. Hence, a detailed exploration of epitopes in the VP2 protein is necessary. In this study, four novel B-cell IDEs were identified using two different antisera with two different methods. IDE2 was identified as a new neutralizing linear epitope. Our findings will help in the rational design of epitope vaccines and further understanding of the antigenic structure of VP2.


Assuntos
Proteínas do Capsídeo , Epitopos de Linfócito B , Animais , Cobaias , Proteínas do Capsídeo/genética , Epitopos de Linfócito B/genética , Anticorpos Antivirais , Soros Imunes
6.
Appl Microbiol Biotechnol ; 107(15): 4903-4915, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37314455

RESUMO

Canine parvovirus (CPV) is an acute and highly infectious virus causing disease in puppies and, thus, affecting the global dog industry. The current CPV detection methods are limited by their sensitivity and specificity. Hence, the current study sought to develop a rapid, sensitive, simple, and accurate immunochromatographic (ICS) test to detect and control the spread and prevalence of CPV infection. More specifically, 6A8, a monoclonal antibody (mAb) with high specificity and sensitivity, was obtained by preliminary screening. The 6A8 antibody was labelled with colloidal gold particles. Subsequently, 6A8 and goat anti-mouse antibodies were coated onto a nitrocellulose membrane (NC) as the test and control lines, respectively. Furthermore, 6A8 and rabbit IgG antibodies were labelled with fluorescent microspheres and evenly sprayed onto a glass fibre membrane. Both strips could be prepared in 15 min with no noticeable cross-reactivity with other common canine intestinal pathogens. The strips were simultaneously used to detect CPV in 60 clinical samples using real-time quantitative PCR, hemagglutination, and hemagglutination inhibition assays. The colloidal gold (fluorescent) ICS test strip was stable for 6 (7) and 4 (5) months at 4 °C and room temperature (18-25 °C). Both test strips were easy to prepare and rapidly detected CPV with high sensitivity and specificity. Moreover, the results were easily interpretable. This study establishes a simple method for two CPV diseases, colloidal gold and fluorescent immunochromatographic (ICS) test strips. KEY POINTS: • CPV test strips do not exhibit cross-reactivity with other canine intestinal pathogens. • The strips are stable for months at 4 °C and at room temperature (18-25 °C). • These strips are a promising approach for the timely diagnosis and treatment of CPV.


Assuntos
Parvovirus Canino , Coelhos , Animais , Cães , Coloide de Ouro/química , Sensibilidade e Especificidade , Testes Imunológicos , Corantes , Cromatografia de Afinidade/métodos
7.
J Virol ; 97(5): e0022823, 2023 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-37162350

RESUMO

African swine fever (ASF), caused by the African swine fever virus (ASFV), is a transboundary infectious disease of domestic pigs and wild boars, resulting in significant swine production losses. Currently, no effective commercial ASF vaccines or therapeutic options are available. A previous study has shown that deletions of ASFV MGF110-9L and MGF505-7R genes (ASFV-Δ110-9L/505-7R) attenuated virulence in pigs and provided complete protection against parental lethal ASFV CN/GS/2018 (wild-type ASFV [ASFV-WT]) challenge, but the underlying mechanism is unclear. This study found that ASFV-Δ110-9L/505-7R weakened TBK1 degradation compared with ASFV-WT through RNA sequencing (RNA-seq) and Western blotting analyses. Furthermore, we confirmed that ASFV-Δ110-9L/505-7R blocked the degradation of TBK1 through the autophagy pathway. We also identified that the downregulation of an autophagy-related protein PIK3C2B was involved in the inhibition of TBK1 degradation induced by ASFV-Δ110-9L/505-7R. Additionally, we also confirmed that PIK3C2B promoted ASFV-Δ110-9L/505-7R replication in vitro. Together, this study elucidated a novel mechanism of virulence change of ASFV-Δ110-9L/505-7R, revealing a new mechanism of ASF live attenuated vaccines (LAVs) and providing theoretical guidance for the development of ASF vaccines. IMPORTANCE African swine fever (ASF) is a contagious and lethal hemorrhagic disease of pigs caused by the African swine fever virus (ASFV), leading to significant economic consequences for the global pig industry. The development of an effective and safe ASF vaccine has been unsuccessful. Previous studies have shown that live attenuated vaccines (LAVs) of ASFV are the most effective vaccine candidates to prevent ASF. Understanding the host responses caused by LAVs of ASFV is important in optimizing vaccine design and diversifying the resources available to control ASF. Recently, our laboratory found that the live attenuated ASFV-Δ110-9L/505-7R provided complete protection against parental ASFV-WT challenge. This study further demonstrated that ASFV-Δ110-9L/505-7R inhibits TBK1 degradation mediated by an autophagy activator PIK3C2B to increase type I interferon production. These results revealed an important mechanism for candidate vaccine ASFV-Δ110-9L/505-7R, providing strategies for exploring the virulence of multigene-deleted live attenuated ASFV strains and the development of vaccines.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Interferon Tipo I , Vacinas Virais , Animais , Febre Suína Africana/prevenção & controle , Vírus da Febre Suína Africana/genética , Interferon Tipo I/metabolismo , Sus scrofa , Suínos , Vacinas Atenuadas , Genes Virais
8.
J Virol ; 97(4): e0024723, 2023 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-37017515

RESUMO

The African swine fever virus (ASFV) has caused a devastating pandemic in domestic and wild swine, causing economic losses to the global swine industry. Recombinant live attenuated vaccines are an attractive option for ASFV treatment. However, safe and effective vaccines against ASFV are still scarce, and more high-quality experimental vaccine strains need to be developed. In this study, we revealed that deletion of the ASFV genes DP148R, DP71L, and DP96R from the highly virulent isolate ASFV CN/GS/2018 (ASFV-GS) substantially attenuated virulence in swine. Pigs infected with 104 50% hemadsorbing doses of the virus with these gene deletions remained healthy during the 19-day observation period. No ASFV infection was detected in contact pigs under the experimental conditions. Importantly, the inoculated pigs were protected against homologous challenges. Additionally, RNA sequence analysis showed that deletion of these viral genes induced significant upregulation of the host histone H3.1 gene (H3.1) and downregulation of the ASFV MGF110-7L gene. Knocking down the expression of H3.1 resulted in high levels of ASFV replication in primary porcine macrophages in vitro. These findings indicate that the deletion mutant virus ASFV-GS-Δ18R/NL/UK is a novel potential live attenuated vaccine candidate and one of the few experimental vaccine strains reported to induce full protection against the highly virulent ASFV-GS virus strain. IMPORTANCE Ongoing outbreaks of African swine fever (ASF) have considerably damaged the pig industry in affected countries. Thus, a safe and effective vaccine is important to control African swine fever spread. Here, an ASFV strain with three gene deletions was developed by knocking out the viral genes DP148R (MGF360-18R), NL (DP71L), and UK (DP96R). The results showed that the recombinant virus was completely attenuated in pigs and provided strong protection against parental virus challenge. Additionally, no viral genomes were detected in the sera of pigs housed with animals infected with the deletion mutant. Furthermore, transcriptome sequencing (RNA-seq) analysis revealed significant upregulation of histone H3.1 in virus-infected macrophage cultures and downregulation of the ASFV MGF110-7L gene after viral DP148R, UK, and NL deletion. Our study provides a valuable live attenuated vaccine candidate and potential gene targets for developing strategies for anti-ASFV treatment.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Deleção de Genes , Genes Virais , Vacinas Virais , Fatores de Virulência , Animais , Febre Suína Africana/imunologia , Febre Suína Africana/virologia , Vírus da Febre Suína Africana/genética , Vírus da Febre Suína Africana/imunologia , Vírus da Febre Suína Africana/patogenicidade , Células Cultivadas , Genes Virais/genética , Histonas/genética , Suínos , Vacinas Atenuadas/imunologia , Vacinas Virais/imunologia , Fatores de Virulência/genética
9.
Appl Microbiol Biotechnol ; 107(11): 3779-3788, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37099055

RESUMO

The p30 protein is abundantly expressed in the early stage of African swine fever virus (ASFV) infection. Thus, it is an ideal antigen candidate for serodiagnosis with the use of an immunoassay. In this study, a chemiluminescent magnetic microparticle immunoassay (CMIA) was developed for the detection of antibodies (Abs) against ASFV p30 protein in porcine serum. Purified p30 protein was coupled to magnetic beads, and the experimental conditions including concentration, temperature, incubation time, dilution ratio, buffers, and other relevant variables were evaluated and optimized. To evaluate the performance of the assay, a total of 178 pig serum samples (117 negative and 61 positive samples) were tested. According to receiver operator characteristic curve analysis, the cut-off value of the CMIA was 104,315 (area under the curve, 0.998; Youden's index, 0.974; 95% confidence interval: 99.45 to 100%). Sensitivity results showed that the dilution ratio of p30 Abs in ASFV-positive sera detected by the CMIA is much higher when compared to commercial blocking ELISA kit. Specificity testing showed that no cross-reactivity was observed with sera positive for other porcine disease viruses. The intraassay coefficient of variation (CV) was < 5%, and the interassay CV was < 10%. The p30-magnetic beads could be stored at 4 °C for more than 15 months without loss of activity. The kappa coefficient between CMIA and INGENASA blocking ELISA kit was 0.946, showing strong agreement. In conclusion, our method showed superiority with high sensitivity, specificity, reproducibility, and stability and potentialized its application in the development of a diagnostic kit for the detection of ASF in clinical samples. KEY POINTS: • ASFV tag-free p30 was successfully purified. • High sensitivity, specificity, relatively simple, and time-saving to detect antibody against ASFV were developed. • The development of CMIA will help the clinical diagnosis of ASFV and will be useful for large-scale serological test.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Suínos , Animais , Reprodutibilidade dos Testes , Febre Suína Africana/diagnóstico , Imunoensaio/métodos , Anticorpos Antivirais , Fenômenos Magnéticos
10.
J Immunol ; 210(4): 442-458, 2023 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-36602826

RESUMO

African swine fever is one of the most serious viral diseases that affects domestic and wild pigs. The causative agent, African swine fever virus (ASFV), has evolved sophisticated immune evasion mechanisms that target both innate and adaptive immune responses. However, the underlying molecular mechanisms have not been fully understood. Here, we report that ASFV E184L protein inhibits host innate immune response via targeting the stimulator of IFN genes (STING)-mediated signaling pathway in both human embryonic kidney HEK-293T cells and porcine pulmonary alveolar macrophages. E184L interacts with STING, impairing dimerization and oligomerization of STING but not affecting its puncta formation at the perinuclear region. Furthermore, E184L disrupts STING-TBK1-IRF3 complex formation, leading to inhibition of STING phosphorylation, and IRF3 dimerization and nuclear translocation. The 1-20 aa region in E184L is essential for E184L-STING interaction and blocking IL-1ß and type I IFN production. Deletion of E184L in ASFV considerably impairs antagonistic function of the virus in suppression of the STING-mediated antiviral response, an effect that is reversible by introduction of E184L. Importantly, the virulence of mutant ASFV lacking E184L is reduced in pigs compared with its parental virus due to induction of higher IFN production in vivo. Our findings indicate that ASFV E184L is an important antagonist of IFN signaling to evade host innate immune antiviral responses, which improves our understanding of immune evasion mechanisms of ASFV.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Animais , Humanos , Antivirais/metabolismo , Imunidade Inata , Suínos , Proteínas Virais , Replicação Viral , Proteínas de Membrana/metabolismo , Interferons/biossíntese
11.
Virol Sin ; 38(1): 84-95, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36442611

RESUMO

African swine fever (ASF) is etiologically an acute, highly contagious and hemorrhagic disease caused by African swine fever virus (ASFV). Due to its genetic variation and phenotypic diversity, until now, no efficient commercial vaccines or therapeutic options are available. The ASFV genome contains a conserved middle region and two flexible ends that code for five multigene families (MGFs), while the biological functions of the MGFs are not fully characterized. Here, ASFV MGF505-2R-deficient mutant ASFV-Δ2R was constructed based on a highly virulent genotype II field isolate ASFV CN/GS/2018 currently circulating in China. Transcriptomic profiling demonstrated that ASFV-Δ2R was capable of inducing a larger number of differentially expressed genes (DEGs) compared with ASFV CN/GS/2018. Hierarchical clustering of up-regulated DEGs revealed that ASFV-Δ2R induced the most dramatic expression of interferon-related genes and inflammatory and innate immune genes, as further validated by RT-qPCR. The GO and KEGG pathway analysis identified significantly enriched pathways involved in pathogen recognition and innate antiviral immunity. Conversely, pharmacological activation of those antiviral immune responses by exogenous cytokines, including type I/II IFNs, TNF-α and IL-1ß, exerted combinatory effects and synergized in antiviral capacity against ASFV replication. Collectively, MGF505-2R is a newly identified inhibitor of innate immunity potentially implicated in immune evasion.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Interferon Tipo I , Suínos , Animais , Vírus da Febre Suína Africana/genética , Febre Suína Africana/prevenção & controle , Imunidade Inata/genética , Antivirais
12.
J Virol ; 96(20): e0119222, 2022 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-36197109

RESUMO

African swine fever virus (ASFV) causes significant morbidity and mortality in pigs worldwide. The lack of vaccines or therapeutic options warrants urgent further investigation. To this aim, we developed a rationally designed live attenuated ASFV-Δ110-9L/505-7R mutant based on the highly pathogenic Genotype II ASFV CN/GS/2018 backbone by deleting 2 well-characterized interferon inhibitors MGF110-9L and MGF505-7R. The mutant was slightly attenuated in vitro compared to parental ASFV but highly tolerant to genetic modifications even after 30 successive passages in vitro. Groups of 5 pigs were intramuscularly inoculated with increasing doses of the mutant, ranging from 103 to 106 hemadsorption units (HAD50). Thirty-five days later, all groups were challenged with 102 HAD50 of virulent parental ASFV. All the animals were clinically normal and devoid of clinical signs consistent with ASFV at the period of inoculation. In the virulent challenge, 2 animals from 103 HAD50-inoculated group and 1 animal from 104 HAD50-inoculated group were unprotected with severe postmortem and histological lesions. The rest of animals survived and manifested with relatively normal clinical appearance accompanied by tangible histological improvements in the extent of tissue damage. Meanwhile, antibody response, as represented by p30-specific antibody titers was positively correlated to protective efficacy, potentializing its usage as an indicator of protection. Moreover, compared to 1 dose, 2 doses provided additional protection, proving that 2 doses were better than 1 dose. The sufficiency in effectiveness supports the claim that our attenuated mutant may be a viable vaccine option with which to fight ASF. IMPORTANCE African swine fever virus (ASFV) is a causative agent of acute viral hemorrhagic disease of domestic swine which is associated with significant economic losses in the pig industry. The lack of vaccines or treatment options requires urgent further investigation. ASFV MGF110-9L and MGF505-7R, 2 well-characterized interferon inhibitors, were associated with viral virulence, host range, and immune modulation. In this study, a recombinant two-gene deletion ASFV mutant with deletion of MGF110-9L and MGF505-7R was constructed. The result showed that the mutant was safe, and also highly resistant to genetic modification even after 30 successive passages. High doses of our mutant (105 and 106 HAD50) provided sterile immunity and complete protection in a virulent challenge. Two doses were superior to 1 dose and provided additional protection. This study develops a new ASFV-specific live attenuated vaccine and may be a viable vaccine option against ASF.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Peste Suína Clássica , Vacinas Virais , Suínos , Animais , Vacinas Atenuadas , Interferons/genética , Proteínas Virais/genética , Antivirais , África
13.
Microbiol Spectr ; 10(2): e0209721, 2022 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-35254168

RESUMO

Senecavirus A (SVA) infection induces inflammation in animals, such as fever, diarrhea, vesicles and erosions, and even death. The inflammatory cytokine interleukin-1ß (IL-1ß) plays a pivotal role in inflammatory responses to combat microbes. Although SVA infection can produce inflammatory clinical symptoms, the modulation of IL-1ß production by SVA infection remains unknown at present. Here, both in vitro and in vivo, SVA robustly induced IL-1ß production in macrophages and pigs. Infection performed in NOD-, LRR-, and pyrin domain-containing three (NLRP3) knockdown cells indicated that NLRP3 is essential for SVA-induced IL-1ß secretion. Importantly, we identified that the 1 to 154 amino acid (aa) portion of SVA 3D binds to the NLRP3 NACHT domain to activate NLRP3 inflammasome assembly and IL-1ß secretion. In addition, the SVA 3D protein interacts with IKKα and IKKß to induce NF-κB activation, which facilitates pro-IL-1ß transcription. Meanwhile, 3D induces p65 nucleus entry. Moreover, SVA 3D induces calcium influx and potassium efflux, which triggers IL-1ß secretion. Ion channels might be related to 3D binding with NLRP3, resulting in NLRP3-ASC complex assembly. We found that 3D protein expression induced tissue hemorrhage and swelling in the mice model. Consistently, expression of 3D in mice caused IL-1ß maturation and secretion. In the natural host of pigs, we confirmed that 3D also induced IL-1ß production. Our data reveal a novel mechanism underlying the activation of the NLRP3 inflammasome after SVA 3D expression, which provides clues for controlling pig's inflammation during the SVA infection. IMPORTANCE Inflammation refers to the response of the immune system to viral, bacterial, and fungal infections or other foreign particles in the body, which can involve the production of a wide array of soluble inflammatory mediators. The NLRP3 inflammasome is one of the best-characterized inflammasome leading to IL-1ß production and maturation. Senecavirus A (SVA) is an oncolytic virus that can cause fever, vesicles and erosions, severe fatal diarrhea, and even the sudden death of piglets. In this study, we demonstrated that 1 to 154 aa of SVA polymerase protein 3D interacts with the NACHT domain of NLRP3 to induce IL-1ß production via the NF-κB signaling pathway and ion channel signal. Our study unveils the mechanism underlying the regulation of inflammasome assembly and production of IL-1ß in response to SVA infection that will help better understand the modulation of host inflammation in pathogens invasion and development of the vaccine.


Assuntos
Inflamassomos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Animais , Diarreia , Inflamassomos/metabolismo , Inflamação , Canais Iônicos , Camundongos , Camundongos Endogâmicos NOD , NF-kappa B/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Picornaviridae , Suínos
14.
Appl Microbiol Biotechnol ; 106(3): 1199-1210, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35089400

RESUMO

African swine fever virus (ASFV) causes acute, febrile, and highly contagious diseases in swine. Early diagnosis is critically important for African swine fever (ASF) prevention and control in the absence of an effective vaccine. P30 is one of the most immunogenic proteins that are produced during the early stage of an ASFV infection. This makes P30 a good serological target for ASF detection and surveillance. In this study, two P30-reactive monoclonal antibodies (mAbs), 2H2 and 5E8, were generated from mice immunized with recombinant P30 protein (rP30). Epitope mapping was performed with overlapping polypeptides, alanine mutants, and synthetic peptides. The mapping results revealed that 2H2 recognized a region located in the N-terminal, 16-48 aa. In contrast, 5E8 recognized a linear epitope in the C-terminal, 122-128 aa. Further analysis indicated that the epitope recognized by 2H2 was highly conserved in genotypes I and II, while the 5E8 epitope was conserved in most genotypes and the Ser to Pro change at position 128 in genotypes IV, V, and VI did not affect recognition. Overall, the results of this study provide valuable information on the antigenic regions of ASFV P30 and lay the foundation for the serological diagnosis of ASF and vaccine research. KEY POINTS: • Two specific and reactive mAbs were prepared and their epitopes were identified. • 2H2 recognized a novel epitope highly conserved in genotypes I and II. • 5E8 recognized a seven-amino acid linear epitope highly conserved in most genotypes.


Assuntos
Vírus da Febre Suína Africana , Anticorpos Monoclonais/imunologia , Mapeamento de Epitopos , Proteínas Virais/imunologia , Febre Suína Africana , Vírus da Febre Suína Africana/imunologia , Animais , Anticorpos Antivirais , Epitopos/genética , Camundongos , Suínos
15.
PLoS Pathog ; 18(1): e1010270, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35089988

RESUMO

ASFV is a large DNA virus that is highly pathogenic in domestic pigs. How this virus is sensed by the innate immune system as well as why it is so virulent remains enigmatic. In this study, we show that the ASFV genome contains AT-rich regions that are recognized by the DNA-directed RNA polymerase III (Pol-III), leading to viral RNA sensor RIG-I-mediated innate immune responses. We further show that ASFV protein I267L inhibits RNA Pol-III-RIG-I-mediated innate antiviral responses. I267L interacts with the E3 ubiquitin ligase Riplet, disrupts Riplet-RIG-I interaction and impairs Riplet-mediated K63-polyubiquitination and activation of RIG-I. I267L-deficient ASFV induces higher levels of interferon-ß, and displays compromised replication both in primary macrophages and pigs compared with wild-type ASFV. Furthermore, I267L-deficiency attenuates the virulence and pathogenesis of ASFV in pigs. These findings suggest that ASFV I267L is an important virulence factor by impairing innate immune responses mediated by the RNA Pol-III-RIG-I axis.


Assuntos
Vírus da Febre Suína Africana/patogenicidade , Imunidade Inata/imunologia , Fatores de Virulência/imunologia , Virulência/imunologia , Febre Suína Africana/imunologia , Vírus da Febre Suína Africana/imunologia , Animais , RNA Polimerase III/imunologia , Receptores de Superfície Celular/imunologia , Suínos
16.
Transbound Emerg Dis ; 69(4): e216-e223, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34370390

RESUMO

African swine fever (ASF) is one of the most severe infectious diseases of pigs. In this study, a loop-mediated isothermal amplification (LAMP) assay coupled with the clustered regularly interspaced short palindromic repeats (CRISPR)/Cas12a system was established in one tube for the detection of the African swine fever virus (ASFV) p72 gene. The single-stranded DNA-fluorophore quencher reporter and CRISPR-derived RNA were screened and selected for the CRISPR detection system. In combination with LAMP amplification assay, the detection limit for the LAMP-CRISPR assay can reach 7 copies/µl of p72 gene per reaction. Furthermore, this method displays no cross-reactivity with other porcine DNA or RNA viruses. The performance of the LAMP-CRISPR assay was compared with real-time qPCR tests for clinical samples; a good consistency between the LAMP-CRISPR assay and real-time qPCR was observed. The method shed a light on the convenient, portable, low cost, highly sensitive and specific detection of ASFV, demonstrating a great application potential for monitoring on-site ASFV in the field.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Doenças dos Suínos , Febre Suína Africana/diagnóstico , Vírus da Febre Suína Africana/genética , Animais , Sistemas CRISPR-Cas , Técnicas de Diagnóstico Molecular , Técnicas de Amplificação de Ácido Nucleico/métodos , Técnicas de Amplificação de Ácido Nucleico/veterinária , Sensibilidade e Especificidade , Suínos , Doenças dos Suínos/genética
17.
Appl Microbiol Biotechnol ; 106(2): 799-810, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34939134

RESUMO

African swine fever (ASF) is an acute and highly contagious infectious disease caused by the African swine fever virus (ASFV). Currently, there is no vaccine against ASF worldwide, and no effective treatment measures are available. For this reason, developing a simple, rapid, specific, and sensitive serological detection method for ASFV antibodies is crucial for the prevention and control of ASF. In this study, a 1:1 mixture of gold-labeled p30 and p72 probes was used as the gold-labeled antigen. The p30 and p72 proteins and their monoclonal antibodies were coated on a nitrocellulose membrane (NC) as a test (T) line and control (C) line, respectively. A colloidal-gold dual immunochromatography strip (ICS) for ASFV p30 and p72 protein antibodies was established. The results showed that the colloidal-gold dual ICS could specifically detect ASFV antibodies within 5-10 min. There was no cross-reaction after testing healthy pig serum; porcine reproductive and respiratory syndrome virus (PRRSV), foot-and-mouth disease type A virus (FMDV-A), foot-and-mouth disease type O virus (FMDV-O), porcine circovirus type 2 (PCV-2), and classical swine fever virus (CSFV) positive sera. A positive result was obtained only for the positive control P1. The sensitivity of the test strips was 1:256, which was equivalent to that of commercially ELISA kits. Their coincidence rate with the two commercial ASFV ELISA antibodies detection kits was higher than 98%. The test strips were stably stored at 18-25 °C and 4 °C for 4 and 6 months, respectively. The colloidal-gold dual ICS prepared in this study had high sensitivity and specificity and were characterized by rapid detection, simple operation, and easy interpretation of results. Therefore, they are of great significance to diagnose, prevent, and control African swine fever. KEY POINTS: • We establish an antibody detection that is quick and can monitor an ASF infection. • We observe changes in two protein antibodies to dynamically monitor ASF infection. • We use diversified detection on a single test strip to detect both antibodies.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Febre Suína Africana/diagnóstico , Animais , Cromatografia de Afinidade , Ensaio de Imunoadsorção Enzimática , Coloide de Ouro , Suínos
18.
J Virol ; 96(1): e0150021, 2022 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-34613824

RESUMO

African swine fever (ASF), a devastating infectious disease in swine, severely threatens the global pig farming industry. Disease control has been hampered by the unavailability of vaccines. Here, we report that deletion of the QP509L and QP383R genes (ASFV-ΔQP509L/QP383R) from the highly virulent ASF virus (ASFV) CN/GS/2018 strain results in complete viral attenuation in swine. Animals inoculated with ASFV-ΔQP509L/QP383R at a 104 50% hemadsorbing dose (HAD50) remained clinically normal during the 17-day observational period. All ASFV-ΔQP509L/QP383R-infected animals had low viremia titers and developed a low-level p30-specific antibody response. However, ASFV-ΔQP509L/QP383R did not induce protection against challenge with the virulent parental ASFV CN/GS/2018 isolate. RNA-sequencing analysis revealed that innate immune-related genes (Ifnb, Traf2, Cxcl10, Isg15, Rantes, and Mx1) were significantly lower in ASFV-ΔQP509L/QP383R-infected than in ASFV-infected porcine alveolar macrophages. In addition, ASFV-ΔQP509L/QP383R-infected pigs had low levels of interferon-ß (IFN-ß) based on enzyme-linked immunosorbent assay (ELISA). These data suggest that deletion of ASFV QP509L/383R reduces virulence but does not induce protection against lethal ASFV challenge. IMPORTANCE African swine fever (ASF) is endemic to several parts of the word, with outbreaks of the disease devastating the swine farming industry; currently, no commercially available vaccine exists. Here, we report that deletion of the previously uncharacterized QP509L and QP383R viral genes completely attenuates virulence in the ASF virus (ASFV) CN/GS/2018 isolate. However, ASFV-ΔQP509L/QP383R-infected animals were not protected from developing an ASF infection after challenge with the virulent parental virus. ASFV-ΔQP509L/QP383R induced lower levels of innate immune-related genes and IFN-ß than the parental virus. Our results increase our knowledge of developing an effective and live ASF attenuated vaccine.


Assuntos
Vírus da Febre Suína Africana/genética , Febre Suína Africana/virologia , Interações Hospedeiro-Patógeno , Deleção de Sequência , Proteínas Virais/genética , Febre Suína Africana/imunologia , Vírus da Febre Suína Africana/imunologia , Animais , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Células Cultivadas , Resistência à Doença , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Imunização , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/virologia , Mutagênese , Suínos , Transcriptoma , Virulência/genética , Fatores de Virulência/genética , Replicação Viral
19.
Sheng Wu Gong Cheng Xue Bao ; 37(9): 3211-3220, 2021 Sep 25.
Artigo em Chinês | MEDLINE | ID: mdl-34622629

RESUMO

To develop Senecavirus A (SVA) virus-like particles (VLPs), a recombinant prokaryotic expression plasmid pET28a-SVA-VP031 was constructed to co-express SVA structural proteins VP0, VP3 and VP1, according to the genomic sequence of the field isolate CH-FJ-2017 after the recombinant proteins were expressed in E .coli system, and purified by Ni+ ion chromatographic method. The SVA VLPs self-assemble with a high yield in vitro buffer. A typical VLPs with an average diameter of 25-30 nm which is similar to native virions by using TEM detection. Animals immunized by SVA VLPs shown that the VLPs induced high titers neutralizing antibodies in Guinea pigs. This study indicated that the VLPs produced with co-expressing SVA structural proteins VP0, VP3 and VP1 in prokaryotic system is a promising candidate and laid an important foundation for the development of a novel SVA VLPs vaccine.


Assuntos
Picornaviridae , Animais , Anticorpos Neutralizantes , Escherichia coli/genética , Genômica , Cobaias , Picornaviridae/genética
20.
Virol J ; 15(1): 162, 2018 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-30352599

RESUMO

BACKGROUND: Retinoic acid-inducible gene I (RIG-I) is a key cytosolic receptor of the innate immune system. Seneca valley virus (SVV) is a newly emerging RNA virus that infects pigs causing significant economic losses in pig industry. RIG-I plays different roles during different viruses infections. The role of RIG-I in SVV-infected cells remains unknown. Understanding of the role of RIG-I during SVV infection will help to clarify the infection process of SVV in the infected cells. METHODS: In this study, we generated a RIG-I knockout (KO) porcine kidney PK-15 cell line using the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/CRISPR-associated protein-9 nuclease (Cas9) genome editing tool. The RIG-I gene sequence of RIG-I KO cells were determined by Sanger sequencing method, and the expression of RIG-I protein in the RIG-I KO cells were detected by Western bloting. The activation status of type I interferon pathway in Sendai virus (SeV)- or SVV-infected RIG-I KO cells was investigated by measuring the mRNA expression levels of interferon (IFN)-ß and IFN-stimulated genes (ISGs). The replicative state of SVV in the RIG-I KO cells was evaluated by qPCR, Western bloting, TCID50 assay and indirect immunofluorescence assay. RESULTS: Gene editing of RIG-I in PK-15 cells successfully resulted in the destruction of RIG-I expression. RIG-I KO PK-15 cells had a lower expression of IFN-ß and ISGs compared with wildtype (WT) PK-15 cells when stimulated by the model RNA virus SeV. The amounts of viral RNA and viral protein as well as viral yields in SVV-infected RIG-I WT and KO cells were determined and compared, which showed that knockout of RIG-I significantly increased SVV replication and propagation. Meanwhile, the expression of IFN-ß and ISGs were considerably decreased in RIG-I KO cells compared with that in RIG-I WT cells during SVV infection. CONCLUSION: Altogether, this study indicated that RIG-I showed an antiviral role against SVV and was essential for activation of type I IFN signaling during SVV infection. In addition, this study suggested that the CRISPR/Cas9 system can be used as an effective tool to modify cell lines to increase viral yields during SVV vaccine development.


Assuntos
Proteína DEAD-box 58/metabolismo , Interferon beta/metabolismo , Infecções por Picornaviridae/imunologia , Infecções por Picornaviridae/veterinária , Picornaviridae/imunologia , Doenças dos Suínos/virologia , Replicação Viral/genética , Animais , Linhagem Celular , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Proteína DEAD-box 58/genética , Edição de Genes , Técnicas de Inativação de Genes , Imunidade Inata/genética , Interferon beta/genética , Suínos , Doenças dos Suínos/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...