Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Immunol ; 2024 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-38905108

RESUMO

Hepatitis E virus (HEV) is a worldwide zoonotic and public health concern. The study of HEV biology is helpful for designing viral vaccines and drugs. Nanobodies have recently been considered appealing materials for viral biological research. In this study, a Bactrian camel was immunized with capsid proteins from different genotypes (1, 3, 4, and avian) of HEV. Then, a phage library (6.3 × 108 individual clones) was constructed using peripheral blood lymphocytes from the immunized camel, and 12 nanobodies against the truncated capsid protein of genotype 3 HEV (g3-p239) were screened. g3-p239-Nb55 can cross-react with different genotypes of HEV and block Kernow-C1/P6 HEV from infecting HepG2/C3A cells. To our knowledge, the epitope recognized by g3-p239-Nb55 was determined to be a novel conformational epitope located on the surface of viral particles and highly conserved among different mammalian HEV isolates. Next, to increase the affinity and half-life of the nanobody, it was displayed on the surface of ferritin, which can self-assemble into a 24-subunit nanocage, namely, fenobody-55. The affinities of fenobody-55 to g3-p239 were ∼20 times greater than those of g3-p239-Nb55. In addition, the half-life of fenobody-55 was nine times greater than that of g3-p239-Nb55. G3-p239-Nb55 and fenobody-55 can block p239 attachment and Kernow-C1/P6 infection of HepG2/C3A cells. Fenobody-55 can completely neutralize HEV infection in rabbits when it is preincubated with nonenveloped HEV particles. Our study reported a case in which a nanobody neutralized HEV infection by preincubation, identified a (to our knowledge) novel and conserved conformational epitope of HEV, and provided new material for researching HEV biology.

2.
J Virol ; 98(4): e0164923, 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38548704

RESUMO

Hepatitis E virus (HEV) is the most common cause of acute viral hepatitis worldwide, responsible for approximately 20 million infections annually. Among the three open reading frames (ORFs) of the HEV genome, the ORF3 protein is involved in virus release. However, the host proteins involved in HEV release need to be clarified. In this study, a host protein, thioredoxin domain-containing protein 5 (TXNDC5), interacted with the non-palmitoylated ORF3 protein by co-immunoprecipitation analysis. We determined that the overexpression or knockdown of TXNDC5 positively regulated HEV release from the host cells. The 17FCL19 mutation of the ORF3 protein lost the ability to interact with TXNDC5. The releasing amounts of HEV with the ORF3 mutation (FCL17-19SSP) were decreased compared with wild-type HEV. The overexpression of TXNDC5 can stabilize and increase ORF3 protein amounts, but not the TXNDC5 mutant with amino acids 1-88 deletion. Meanwhile, we determined that the function of TXNDC5 on the stabilization of ORF3 protein is independent of the Trx-like domains. Knockdown of TXNDC5 could lead to the degradation of ORF3 protein by the endoplasmic reticulum (ER)-associated protein degradation-proteasome system. However, the ORF3 protein cannot be degraded in the knockout-TXNDC5 stable cells, suggesting that it may hijack other proteins for its stabilization. Subsequently, we found that the other members of protein disulfide isomerase (PDI), including PDIA1, PDIA3, PDIA4, and PDIA6, can increase ORF3 protein amounts, and PDIA3 and PDIA6 interact with ORF3 protein. Collectively, our study suggested that HEV ORF3 protein can utilize TXNDC5 for its stability in ER to facilitate viral release. IMPORTANCE: Hepatitis E virus (HEV) infection is the leading cause of acute viral hepatitis worldwide. After the synthesis and modification in the cells, the mature ORF3 protein is essential for HEV release. However, the host protein involved in this process has yet to be determined. Here, we reported a novel host protein, thioredoxin domain-containing protein 5 (TXNDC5), as a chaperone, contributing to HEV release by facilitating ORF3 protein stability in the endoplasmic reticulum through interacting with non-palmitoylated ORF3 protein. However, we also found that in the knockout-TXNDC5 stable cell lines, the HEV ORF3 protein may hijack other proteins for its stabilization. For the first time, our study demonstrated the involvement of TXNDC5 in viral particle release. These findings provide some new insights into the process of the HEV life cycle, the interaction between HEV and host factors, and a new direction for antiviral design.


Assuntos
Vírus da Hepatite E , Hepatite E , Hepatite Viral Humana , Humanos , Vírus da Hepatite E/genética , Fatores Imunológicos , Isomerases de Dissulfetos de Proteínas/genética , Tiorredoxinas/genética , Vírion/metabolismo
3.
J Virol ; 98(2): e0165023, 2024 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-38271227

RESUMO

Vaccination is the most effective method to protect humans and animals from diseases. Anti-idiotype vaccines are safer due to their absence of pathogens. However, the commercial production of traditional anti-idiotype vaccines using monoclonal and polyclonal antibodies (mAb and pAb) is complex and has a high failure rate. The present study designed a novel, simple, low-cost strategy for developing anti-idiotype vaccines with nanobody technology. We used porcine circovirus type 2 (PCV2) as a viral model, which can result in serious economic loss in the pig industry. The neutralizing mAb-1E7 (Ab1) against PCV2 capsid protein (PCV2-Cap) was immunized in the camel. And 12 nanobodies against mAb-1E7 were screened. Among them, Nb61 (Ab2) targeted the idiotype epitope of mAb-1E7 and blocked mAb-1E7's binding to PCV2-Cap. Additionally, a high-dose Nb61 vaccination can also protect mice and pigs from PCV2 infection. Epitope mapping showed that mAb-1E7 recognized the 75NINDFL80 of PCV2-Cap and 101NYNDFLG107 of Nb61. Subsequently, the mAb-3G4 (Ab3) against Nb61 was produced and can neutralize PCV2 infection in the PK-15 cells. Structure analysis showed that the amino acids of mAb-1E7 and mAb-3G4 respective binding to PCV2-Cap and Nb61 were also similar on the amino acids sequences and spatial conformation. Collectively, our study first provided a strategy for producing nanobody-based anti-idiotype vaccines and identified that anti-idiotype nanobodies could mimic the antigen on amino acids and structures. Importantly, as more and more neutralization mAbs against different pathogens are prepared, anti-idiotype nanobody vaccines can be easily produced against the disease with our strategy, especially for dangerous pathogens.IMPORTANCEAnti-idiotype vaccines utilize idiotype-anti-idiotype network theory, eliminating the need for external antigens as vaccine candidates. Especially for dangerous pathogens, they were safer because they did not contact the live pathogenic microorganisms. However, developing anti-idiotype vaccines with traditional monoclonal and polyclonal antibodies is complex and has a high failure rate. We present a novel, universal, simple, low-cost strategy for producing anti-idiotype vaccines with nanobody technology. Using a neutralization antibody against PCV2-Cap, a nanobody (Ab2) was successfully produced and could mimic the neutralizing epitope of PCV2-Cap. The nanobody can induce protective immune responses against PCV2 infection in mice and pigs. It highlighted that the anti-idiotype vaccine using nanobody has a very good application in the future, especially for dangerous pathogens.


Assuntos
Infecções por Circoviridae , Circovirus , Anticorpos de Domínio Único , Vacinas Virais , Animais , Humanos , Camundongos , Proteínas do Capsídeo , Infecções por Circoviridae/prevenção & controle , Infecções por Circoviridae/veterinária , Epitopos , Suínos , Vacinas Virais/química , Vacinas Virais/imunologia
4.
J Nanobiotechnology ; 17(1): 35, 2019 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-30823927

RESUMO

BACKGROUND: Sensitive and specific antibodies can be used as essential probes to develop competitive enzyme-linked immunosorbent assay (cELISA). However, traditional antibodies are difficult to produce, only available in limited quantities, and ineffective as enzymatic labels. Nanobodies, which are single-domain antibodies (sdAbs), offer an alternative, more promising tool to circumvent these limitations. In the present work, a cELISA using nanobody-horseradish peroxidase (HRP) fusion protein firstly designed as a probe was developed for detecting anti-Newcastle disease virus (NDV) antibodies in chicken sera. RESULTS: In the study, a platform for the rapid and simple production of nanobody-HRP fusion protein was constructed. First, a total of 9 anti-NDV-NP protein nanobodies were screened from a immunised Bactrian camel. Then, the Nb5-HRP fusions were produced with the platform and used for the first time as sensitive reagents for developing cELISA to detect anti-NDV antibodies. The cut-off value of the cELISA was 18%, and the sensitivity and specificity were respectively 100% and 98.6%. The HI test and commercial ELISA kit (IDEXX) separately agreed 97.83% and 98.1% with cELISA when testing clinical chicken sera and both agreed 100% when testing egg yolks. However, for detecting anti-NDV antibodies in the sequential sera from the challenged chickens, cELISA demonstrated to be more sensitive than the HI test and commercial ELISA kit. Moreover, a close correlation (R2 = 0.914) was found between the percent competitive inhibition values of cELISA and HI titers. CONCLUSIONS: A platform was successfully designed to easily and rapidly produce the nanobody-HRP fusion protein, which was the first time to be used as reagents for establishing cELISA. Results suggest that the platform supports the development of a cELISA with high sensitivity, simplicity, and rapid detection of anti-NDV antibodies. Overall, we believe that the platform based on nanobody-HRP fusions can be widely used for future investigations and treatment other diseases and viruses.


Assuntos
Anticorpos Antivirais/sangue , Ensaio de Imunoadsorção Enzimática , Vírus da Doença de Newcastle/imunologia , Anticorpos de Domínio Único/imunologia , Animais , Anticorpos Antivirais/isolamento & purificação , Camelus , Galinhas , Escherichia coli , Biblioteca Gênica , Células HEK293 , Peroxidase do Rábano Silvestre/química , Humanos , Proteínas Recombinantes/química , Sensibilidade e Especificidade
5.
Transbound Emerg Dis ; 66(1): 72-82, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30043495

RESUMO

The zoonotic transmission of hepatitis E virus (HEV) is mainly mediated by HEV genotypes 3 and 4, with domestic pigs serving as an important reservoir for both genotypes. In China, genotype 4 HEV is the primary prevalent genotype on pig farms. In this study, the prevalence of HEV infection in pig herds of Shaanxi Province was investigated. Serological testing detected anti-HEV antibody-positive pigs in five selected cities, with 13 of 17 farms harbouring at least one positive pig (76.47%). Within positive farms, the proportion of positive pigs ranged from 1.6% to 37.5%. Genetic detection analyses of faecal samples revealed that pigs in four cities and on nine of 17 farms were positive for sequences homologous to a partial ORF2-coding region of HEV (306 bp), as were 6 of 53 bile and 1 of 26 semen samples. Meanwhile, DNA coding for partial HEV ORF1 (1,080 bp) and a longer gene segment coding for partial ORF2 (1,594 bp) were successfully amplified from RNA isolated from pig semen from one HEV-positive pig. Sequence comparisons of partial ORF2 gene sequences showed that HEV isolates from Shaanxi Province shared the highest identity (81.4%-96.1%) with genotype 4 HEV. Phylogenetic tree analysis grouped these isolates into three subgenotypes (4d, 4h and 4i), with subgenotype 4i the predominant subgenotype. In addition, the HEV isolate from pig semen belonged to subgenotype 4i HEV based on phylogenetic trees constructed both using partial ORF1 and ORF2 gene sequences. In conclusion, HEV infection is endemic on pig farms of Shaanxi Province, China, and 4i is the predominant HEV subgenotype. More important, this is the first study demonstrating detection of HEV RNA in pig semen, suggesting that artificial insemination can transmit HEV in pigs.


Assuntos
Vírus da Hepatite E/genética , Hepatite E/veterinária , Sêmen , Doenças dos Suínos/epidemiologia , Animais , China/epidemiologia , Fezes/virologia , Genótipo , Hepatite E/epidemiologia , Hepatite E/virologia , Masculino , Fases de Leitura Aberta/genética , Filogenia , Prevalência , RNA Viral/análise , Análise de Sequência de RNA/veterinária , Estudos Soroepidemiológicos , Suínos , Doenças dos Suínos/virologia
6.
Vet Microbiol ; 225: 48-52, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30322532

RESUMO

Rabbits are recognized as a zoonotic reservoir of hepatitis E virus (HEV) for transmission to humans and other zoonotic reservoirs such as swine. The purpose of this study was to assess the ability of rabbit HEV to cross the species barrier to infect mice and also the usefulness of this animal to study HEV transmission and pathogenesis. In this study, uninfected BALB/c mice were experimentally inoculated with rabbit HEV either via gavage or through contact-exposure with infected mice. Rabbit HEV propagation in mice was evaluated by studying fecal virus shedding, viremia, seroconversion and microscopic liver lesions. Rabbit HEV could be detected in all mice infected by gavage, but only in some contact-exposed mice, with some animals exhibiting fecal virus shedding, seroconversion or viremia (one mouse only). Compared with inoculated mice, anti-rabbit HEV antibody titers and viral copy numbers in fecal and serum samples were lower in contact-exposed mice. Infected mice mainly exhibited phlebitis, hepatocyte swelling and necrosis. Microscopic liver lesion scores for inoculated and contact-exposed infected mice were higher than scores for negative controls. This study therefore demonstrates that rabbit HEV could infect BALB/c mice both though inoculation via gavage and through contact-exposure.


Assuntos
Anticorpos Antivirais/sangue , Fezes/virologia , Vírus da Hepatite E/isolamento & purificação , Hepatite E/imunologia , Hepatite E/transmissão , Animais , Modelos Animais de Doenças , Hepatite E/epidemiologia , Hepatite E/virologia , Vírus da Hepatite E/imunologia , Hepatócitos/patologia , Humanos , Fígado/patologia , Fígado/ultraestrutura , Fígado/virologia , Camundongos , Camundongos Endogâmicos BALB C , Microscopia , RNA Viral/sangue , Coelhos , Soroconversão , Especificidade da Espécie , Organismos Livres de Patógenos Específicos , Viremia/virologia , Eliminação de Partículas Virais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...