Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 13: 890263, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36016955

RESUMO

Tembusu virus (TMUV) can result in a severe disease affecting domestic ducks. The role of T cells in protection from TMUV infection and the molecular basis of T cell-mediated protection against TMUV remain largely uncharacterized. Here, we used the high-virulence TMUV strain Y and the low-virulence TMUV strain PS to investigate the protective role for TMUV-specific CD4+ and CD8+ T cells. When tested in a 5-day-old Pekin duck model, Y and PS induced comparable levels of neutralizing antibody, whereas Y elicited significantly stronger cellular immune response relative to PS. Using a duck adoptive transfer model, we showed that both CD4+ and CD8+ T cells provided significant protection from TMUV-related disease, with CD8+ T cell conferring more robust protection to recipient ducklings. For TMUV, CD4+ T cells mainly provided help for neutralizing antibody response, whereas CD8+ T cells mainly mediated viral clearance from infected tissues. The difference in T cell immunity between Y and PS was primarily attributed to CD4+ T cells; adoptive transfer of Y-specific CD4+ T cells resulted in significantly enhanced protective ability, neutralizing antibody response, and viral clearance from the brain relative to PS-specific CD4+ T cells. Further investigations with chimeric viruses, mutant viruses, and their parental viruses identified two mutations (T151A and R304M) in the envelope (E) protein that contributed significantly to TMUV-specific CD4+ T cell-mediated protective ability and neutralizing antibody response, with more beneficial effects being conferred by R304M. These data indicate T cell-mediated immunity is important for protection from disease, for viral clearance from tissues, and for the production of neutralizing antibodies, and that the difference in CD4+T cell immunity between high- and low-virulence TMUV strains is primarily related to residues 151 and 304 in the E protein.


Assuntos
Infecções por Flavivirus , Flavivirus , Doenças das Aves Domésticas , Animais , Anticorpos Neutralizantes , Linfócitos T CD4-Positivos , Linfócitos T CD8-Positivos , Patos , Virulência
2.
J Immunol ; 209(4): 684-695, 2022 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-35879100

RESUMO

Previous studies identified three neutralizing epitopes on domains I, II, and III of the Tembusu virus (TMUV) envelope (E). More evidence is needed to understand the molecular basis of Ab-mediated neutralization and protection against TMUV. In this study, we observed a neutralizing mAb, 6C8, that neutralized TMUV infection primarily by inhibiting cell attachment. In immunofluorescence assays, 6C8 recognized the premembrane and E proteins coexpressed in HEK-293T cells, but failed to react with premembrane or E expressed individually. Epitope mapping identified nine E protein residues positioned on BC/EF loops and F/G strands in domain III and the first α-helical domain in the stem region. Further investigation with mutant viruses showed that 6C8 pressure resulted in mutations at residues 330 of BC loop and 409 of the first α-helical domain, although 6C8 only exhibited a moderate neutralizing activity in BHK-21 cells and a weak protective activity in BALB/c mice and Shaoxing duck models. Mutations A330S and T409M conferred high- and low-level 6C8 resistance, respectively, whereas the combination of A330S and T409M mutations conferred moderate-level 6C8 resistance. As a result, a quasispecies comprising three groups of antigenic variants appeared in BHK-21 cell-derived viral stocks after repeated passages of TMUV strain Y in the presence of 6C8 treatment. Taken together, these findings have raised a concern about Ab-induced antigenic variations in vivo, and they have revealed information concerning the conformational structure of the 6C8 epitope and its role in constraint on antigenic variations. The present work contributes to a better understanding of the complexity of the TMUV immunogen.


Assuntos
Infecções por Flavivirus , Flavivirus , Animais , Camundongos , Anticorpos Neutralizantes , Anticorpos Antivirais , Patos/virologia , Epitopos , Proteínas do Envelope Viral/genética , Humanos
3.
Arch Virol ; 167(8): 1687-1691, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35639191

RESUMO

Earlier studies have shown that Tembusu virus (TMUV) can elicit high levels of neutralizing antibodies, but the ability of antibodies to protect against TMUV-associated disease and to inhibit replication of TMUV in vivo remains to be investigated. Here, we tested the prophylactic efficacy of TMUV immune serum directly using a 2-day-old Pekin duck model. Passive administration of the immune serum prior to challenge protected ducklings against morbidity and mortality, substantially reduced TMUV-caused tissue injury, and significantly decreased TMUV levels in the periphery and central nervous system. These findings demonstrate that antibodies play a dominant protective role in controlling TMUV-associated disease.


Assuntos
Infecções por Flavivirus , Flavivirus , Doenças das Aves Domésticas , Animais , Patos , Soros Imunes
4.
Vet Microbiol ; 263: 109252, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34673357

RESUMO

Previous studies resulted in the isolation of a low-virulence plaque-purified variant from the third passage (P3) in BHK-21 cells of a Tembusu virus (TMUV) isolate, suggesting the presence of viral quasispecies in the P3 culture. To confirm this notion, the fourth passage virus (P4) was prepared by infecting BHK-21 cells with P3 for isolation of more variants. We isolated 10 plaque-purified viruses. Comparative genome sequence analysis identified six of the 10 viruses as genetically different variants, which harbored a total of eight amino acid differences in the envelope, NS1, NS3, and NS5 proteins. When tested in a 2-day-old Pekin duck model, P4 caused 80 % mortality, belonging to a high-virulence TMUV strain. Out of the six genetically different variants, two presented high-virulence, one exhibited moderate-virulence, and three displayed low-virulence, causing 60 %-70 %, 40 %, and 10 % mortalities, respectively. These results demonstrate that P4 contains at least three groups of variants with distinct virulence phenotypes. Analysis of links between the eight residues and virulence of the six variants identified NS1 protein residue 183 and NS5 protein residues 275 and/or 287 as novel determinants of TMUV virulence. The analysis also provided a new clue for future studies on the molecular basis of TMUV virulence in terms of genetic interaction of different proteins. Overall, our study provides direct evidence to suggest that TMUV exists in in vitro culture of a virulent isolate as a quasispecies, which may enhance our understanding of molecular mechanism of TMUV virulence.


Assuntos
Infecções por Flavivirus , Flavivirus , Doenças das Aves Domésticas , Animais , Linhagem Celular , Patos , Flavivirus/patogenicidade , Infecções por Flavivirus/veterinária , Infecções por Flavivirus/virologia , Fenótipo , Doenças das Aves Domésticas/virologia , Quase-Espécies , Virulência
5.
Virus Res ; 306: 198582, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34599934

RESUMO

We recently developed a Tembusu virus (TMUV)-specific monoclonal antibody (MAb) 12F11, which was found to recognize a long amino acid sequence between residues 8 and 77 of domain III of the envelope protein (EDIII). Here, the epitope recognized by MAb 12F11 was mapped using alanine substitutions combined with dissociation constant analysis. The findings, and prediction of tertiary structure of TMUV EDIII, showed that the MAb 12F11 epitope contained one critical residue and 13 peripheral residues. Moreover, the antigenic site was shown to span four loops (N-terminal region, AB, BC, and CD) and three ß-strands (A, B, and D). The present work contributes to the understanding of antigenic structure of TMUV envelope protein.


Assuntos
Anticorpos Neutralizantes , Flavivirus , Anticorpos Monoclonais , Anticorpos Antivirais , Mapeamento de Epitopos , Epitopos , Flavivirus/genética , Proteínas do Envelope Viral
6.
J Virol ; 95(6)2021 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-33328312

RESUMO

The Tembusu virus (TMUV) PS strain, derived by several passages and plaque purifications in BHK-21 cells, displays markedly lower virulence in Pekin ducklings relative to a natural isolate of TMUV, but the potential virulence determinants and the in vivo mechanisms for substantial virulence attenuation of the passage variant remain unknown. Here, we constructed a series of chimeric and mutant viruses and assessed their virulence using a 2-day-old Pekin duckling model. We showed that residue 304 in the envelope (E) protein is the molecular determinant of TMUV virulence. Further investigations with mutant and parental viruses demonstrated that acquisition of positive charges at E protein residue 304 plays a critical role in substantial attenuation of neurovirulence and neuroinvasiveness, which is linked to enhanced binding affinity for glycosaminoglycans (GAGs). In Pekin ducklings infected by subcutaneous inoculation, an Arg at residue 304 in the E protein was shown to contribute to more rapid virus clearance from the circulation, markedly reduced viremia, and significantly decreased viral growth in the extraneural tissues and the central nervous system, relative to a Met at the corresponding residue. These findings suggest that the in vivo mechanism of virulence attenuation of the TMUV passage variant closely resembles that proposed previously for GAG-binding variants of other flaviviruses. Overall, our study provides insight into the molecular basis of TMUV virulence and the in vivo consequences of acquisition of a GAG-binding determinant at residue 304 in the E protein of TMUV.IMPORTANCE TMUV-related disease emerged in 2010 and has a significant economic impact on the duck industry. Although the disease was originally recognized to affect adult ducks, increasing evidence has shown that TMUV also causes severe disease of young ducklings. It is, therefore, essential to investigate the pathogenesis of TMUV infection in a young duckling model. The significance of our studies is in identifying E protein residue Arg304 as the molecular determinant for TMUV virulence and in clarifying the crucial role of positive charges at E protein residue 304 in virulence attenuation of a TMUV passage variant. These data will greatly enhance our understanding of the pathogenesis of TMUV infection in ducklings and have implications for development of a safe and efficient vaccine.


Assuntos
Arginina/metabolismo , Infecções por Flavivirus/virologia , Flavivirus/patogenicidade , Proteínas do Envelope Viral/metabolismo , Animais , Arginina/genética , Linhagem Celular , Sistema Nervoso Central/virologia , Cricetinae , Patos , Glicosaminoglicanos/metabolismo , Mutação , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Viremia/virologia , Virulência/genética , Replicação Viral
7.
Front Microbiol ; 11: 585194, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33193231

RESUMO

Tembusu virus (TMUV) is a mosquito-borne flavivirus that most commonly affects adult breeder and layer ducks. However, a TMUV-caused neurological disease has also been found in ducklings below 7 weeks of age, highlighting the need to develop a safe vaccine for young ducklings. In this study, a plaque-purified PS TMUV strain was attenuated by serial passage in BHK-21 cells. Using 1-day-old Pekin ducklings as a model, the virus was confirmed to be attenuated sufficiently after 180 passages, whereas the neutralizing antibody response elicited by the 180th passage virus (PS180) was substantially impaired compared with PS. The findings suggest that sufficient attenuation results in loss of immunogenicity in the development of the live-attenuated TMUV vaccine. Comparative sequence analysis revealed that PS180 acquired one mutation (V41M) in prM and four mutations (T70A, Y176H, K313R, and F408L) in the envelope (E) protein. To identify the amino acid substitution(s) associated with loss of immunogenicity of PS180, we rescued parental viruses, rPS and rPS180, and produced mutant viruses, rPS180-M41V, rPS180-A70T, rPS180-H176Y, rPS180-R313K, rPS180-L408F, and rPS180-M5, which contained residue 41V in prM, residues 70T, 176Y, 313K, and 408F in E, and combination of the five residues, respectively, of PS in the backbone of the rPS180 genome. The neutralizing antibody response elicited by rPS180-L408F and rPS180-M5 was significantly higher than those by other mutant viruses and comparable to that by rPS. Furthermore, we produced mutant virus rPS-F408L, which contained residue 408L of PS180 in the backbone of the rPS genome. The F408L mutation conferred significantly decreased neutralizing antibody response to rPS-F408L, which was comparable to that elicited by rPS180. Based on homologous modeling, residue 408 was predicted to be located within the first helical domain of the stem region of the E protein (EH1). Together, these data demonstrate that a single mutation within the EH1 domain exerts a dramatical impact on the TMUV neutralizing antibody response. The present work may enhance our understanding of molecular basis of the TMUV neutralizing antibody response, and provides an important step for the development of a safe and efficient live-attenuated TMUV vaccine.

8.
Vet Microbiol ; 251: 108870, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33053451

RESUMO

Tembusu virus (TMUV) infection most commonly affects breeder and layer ducks during laying period, and can also affect young ducks below 7 weeks of age. Here, we report our investigation of a TMUV-caused fatal disease of Jingding ducklings (Anas platyrhynchos domesticus) in Northeast China. The disease resulted in mortalities of up to 40 % in 2 to 4-week-old ducks, up to 25 % in 5 to 6-week-old ducks, and less than 10 % in 7 to 8-week-old ducks. Using a TMUV-specific reverse transcription-PCR assay, all 44 ducks collected from 10 different farms were found positive for TMUV. Phylogenetic analysis of the E nucleotide sequence revealed that five of the six TMUV strains detected from three young ducks and three laying ducks were grouped within cluster 2.1. Inoculation of the liver sample of a 40-day-old sick duck in BHK-21 cells resulted in isolation of cluster 2.1 TMUV strain H. In experimental infections performed using 3-week-old Pekin ducklings (Anas platyrhynchos domesticus) (n = 30; 10 birds/group), high mortality (60 %) was caused by strain H, in sharp contrast with a very low mortality (10 %) caused by strain Y which was isolated during outbreaks of the TMUV-related disease of young Jinding ducks in 2014 in the same region. These findings clearly demonstrated that the cluster 2.1 TMUV strain H is more pathogenic for 3-week-old ducklings as compared to the cluster 2.2 TMUV strain Y. The present study may enhance our understanding of pathogenicity of TMUV in young ducks, and will stimulate further studies on the pathogenesis of TMUV infection.


Assuntos
Patos/virologia , Infecções por Flavivirus/veterinária , Flavivirus/patogenicidade , Doenças das Aves Domésticas/virologia , Fatores Etários , Animais , Linhagem Celular , China , Cricetinae , Surtos de Doenças , Flavivirus/classificação , Flavivirus/genética , Infecções por Flavivirus/mortalidade , Rim/citologia , Filogenia , Virulência
9.
Arch Virol ; 164(11): 2837-2841, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31494776

RESUMO

Since January 2019, abnormal molting has been observed frequently in approximately 40-day-old Pekin ducks in China. To investigate the possible involvement of a virus, we tested the prevalence of duck circovirus (DuCV), goose hemorrhagic polyomavirus (GHPyV), and goose parvovirus (GPV) in 11 molt cases in two provinces. GPV was detected in all cases, particularly in all samples collected from the feather area. The complete genome sequences of three GPV strains were determined and found to have 52 nucleotide changes relative to GPVs associated with short beak and dwarfism syndrome of Pekin ducks. These data will enhance our understanding of GPV diversity and outcomes of GPV infection in Pekin ducks.


Assuntos
Patos/virologia , Gansos/virologia , Muda/fisiologia , Parvovirinae/isolamento & purificação , Doenças das Aves Domésticas/virologia , Animais , China/epidemiologia , Circovirus/genética , Circovirus/isolamento & purificação , Genoma Viral/genética , Parvovirinae/genética , Polyomavirus/genética , Polyomavirus/isolamento & purificação , Doenças das Aves Domésticas/epidemiologia
10.
J Gen Virol ; 100(8): 1234-1240, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31282853

RESUMO

Using random amplification and reverse transcription-PCR, a novel RNA virus was detected in sera of domestic ducks. The full genome of the virus was determined for three strains, identifying the first hepacivirus-like flavivirus in birds. The virus, that we tentatively named duck hepacivirus-like virus (DuHV), possesses several unique molecular features, such as possession of the largest hepacivirus-like polyprotein gene and a Pegivirus A-like internal ribosome entry site. Sequence comparisons and phylogenetic and sliding-window analyses indicated that DuHV is most closely related to, but highly divergent from, the known hepaciviruses. DuHV was detected in 69.7 % of 185 serum samples from four duck species and in 31 of 33 flocks from five provinces of China, reflecting a high prevalence in duck populations and a wide geographical distribution. The detection of DuHV in the same flock in November 2018 and April 2019 suggested that persistent infection can be established in the infected ducks.


Assuntos
Patos , Infecções por Flavivirus/veterinária , Flavivirus/isolamento & purificação , Hepacivirus/isolamento & purificação , Doenças das Aves Domésticas/virologia , Animais , China , Flavivirus/classificação , Flavivirus/genética , Infecções por Flavivirus/virologia , Genoma Viral , Hepacivirus/química , Hepacivirus/classificação , Hepacivirus/genética , Conformação de Ácido Nucleico , Filogenia , RNA Viral/química , RNA Viral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA