Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
Más filtros

Base de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Poult Sci ; 103(10): 104087, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-39094497

RESUMEN

Duck hepatitis A virus type 3 (DHAV-3) is an infectious virus that is highly fatal to ducklings and causes significant economic losses in the duck industry worldwide. Biosecurity and vaccination are required to control the pathogen. In the present study, we attenuated a lowly pathogenic DHAV-3 clinical isolate, named as HB, by serial passaging in duck embryos, and followed by several adaptive proliferations in specific-pathogen-free (SPF) chicken embryos. The virulence of DHAV-3 at different passages was assessed by infecting 3-day-old ducklings. We found that the HB strain lost pathogenicity to ducklings from the 55th passage onwards. The 80th passage strain (HB80), which achieved good growth capacity in duck embryos with a viral titer of 108.17 50% egg lethal dose per milliliter (ELD50/mL), was selected as a live attenuated vaccine candidate. The HB80 strain did not induce clinical symptoms or pathological lesions in 3-day-old ducklings and showed no virulence reversion after 5 rounds of in vivo back-passage. The minimum effective dose of HB80 was determined to be 104.5 ELD50 by hypodermic inoculation of the neck. Importantly, a single dose of HB80 elicited good immune responses and provided complete protection against challenge with the lethal DHAV-3 strain. Compared with the genomic sequence of the parental HB strain, HB80 had 7 amino acid substitutions, two of them are in the hypervariable region of the VP1 and polymerase-encoding 3D regions, which may play a role in virulence attenuation. Our data suggest that the attenuated HB80 strain is a promising vaccine candidate for the prevention of DHAV-3 infections in China. HB80 has been registered as a New Veterinary Drug Registration Certificate by the Chinese Ministry of Agriculture and Rural Affairs (MARA), and is the first live attenuated DHAV-3 vaccine strain to be officially licensed in China.

2.
Artículo en Inglés | MEDLINE | ID: mdl-39002060

RESUMEN

Duck viral hepatitis, primarily caused by duck hepatitis A virus type 1 (DHAV-1), poses a significant threat to the global duck industry. Bacillus subtilis is commonly utilized as a safe probiotic in the development of mucosal vaccines. In this study, a recombinant strain of B. subtilis, designated as B. subtilis RV, was constructed to display the DHAV-1 capsid protein VP1 on its spore surface using the outer coat protein B as an anchoring agent. The immunogenicity of this recombinant strain was evaluated in a mouse model through mixed feeding immunization. The results indicated that B. subtilis RV could elicit specific systemic and mucosal immune responses in mice, as evidenced by the high levels of serum IgG, intestinal secretory IgA, and potent virus-neutralizing antibodies produced. Furthermore, the recombinant strain significantly upregulated the expression levels of IL-2, IL-6, IL-10, TNF-α, and IFN-γ in the intestinal mucosa. Thus, the recombinant strain maintained the balance of the Th1/Th2 immune response and demonstrated an excellent mucosal immune adjuvant function. In summary, this study suggests that B. subtilis RV can be a novel alternative for effectively controlling DHAV-1 infection as a vaccine-based feed additive.

3.
Int J Biol Macromol ; 274(Pt 1): 133246, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38908633

RESUMEN

The mixed infection of duck hepatitis A virus 3 (DHAV-3) and novel duck reovirus (NDRV) has caused significant losses to the global duck farming industry. On-site point-of-care testing of viruses plays a crucial role in the early diagnosis, prevention, and disease control. Here, we proposed an RPA-CRISPR Cas12a/Cas13a one-pot strategy (DRCFS) for rapid and simultaneous detection of DHAV-3 and NDRV. This method integrated the reaction of RPA and CRISPR Cas12a/Cas13a in a single tube, eliminating the need to open the lid during the intermediate processes and thereby avoiding aerosol contamination. On this basis, we proposed a dual RPA-CRISPR strategy coupled with a lateral flow analysis platform (DRC-LFA). This circumvented the necessity for complex instruments, enabling direct visual interpretation of results, making the test more accessible and user-friendly. Our findings demonstrated that the DRCFS method could detect DHAV-3 and NDRV at concentrations as low as 100 copy/µL, while DRC-LFA achieved limit of 101 copies/µL within 35 min. Furthermore, when DRCFS, DRC-LFA, and qPCR were employed collectively for clinical samples analysis, all three methods yielded consistent results. The specificity, sensitivity, and user-friendly of these methods rendered them invaluable for on-site virus detection.


Asunto(s)
Sistemas CRISPR-Cas , Patos , Animales , Sistemas CRISPR-Cas/genética , Patos/virología , Virus de la Hepatitis del Pato/genética , Virus de la Hepatitis del Pato/aislamiento & purificación , Orthoreovirus Aviar/genética , Orthoreovirus Aviar/aislamiento & purificación
4.
Vet Res ; 55(1): 40, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38532469

RESUMEN

The interaction between viral components and cellular proteins plays a crucial role in viral replication. In a previous study, we showed that the 3'-untranslated region (3'-UTR) is an essential element for the replication of duck hepatitis A virus type 1 (DHAV-1). However, the underlying mechanism is still unclear. To gain a deeper understanding of this mechanism, we used an RNA pull-down and a matrix-assisted laser desorption/ionization time-of-flight mass spectrometry assay to identify new host factors that interact with the 3'-UTR. We selected interleukin-2 enhancer binding factor 2 (ILF2) for further analysis. We showed that ILF2 interacts specifically with both the 3'-UTR and the 3D polymerase (3Dpol) of DHAV-1 through in vitro RNA pull-down and co-immunoprecipitation assays, respectively. We showed that ILF2 negatively regulates viral replication in duck embryo fibroblasts (DEFs), and that its overexpression in DEFs markedly suppresses DHAV-1 replication. Conversely, ILF2 silencing resulted in a significant increase in viral replication. In addition, the RNA-dependent RNA polymerase (RdRP) activity of 3Dpol facilitated viral replication by enhancing viral RNA translation efficiency, whereas ILF2 disrupted the role of RdRP in viral RNA translation efficiency to suppress DHAV-1 replication. At last, DHAV-1 replication markedly suppressed the expression of ILF2 in DEFs, duck embryo hepatocytes, and different tissues of 1 day-old ducklings. A negative correlation was observed between ILF2 expression and the viral load in primary cells and different organs of young ducklings, suggesting that ILF2 may affect the viral load both in vitro and in vivo.


Asunto(s)
Virus de la Hepatitis del Pato , Hepatitis Viral Animal , Infecciones por Picornaviridae , Enfermedades de las Aves de Corral , Animales , Interleucina-2/genética , ARN Polimerasa Dependiente del ARN/genética , Regulación de la Expresión Génica , ARN Viral/genética , Patos/genética , Infecciones por Picornaviridae/veterinaria
5.
Poult Sci ; 102(12): 103117, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37852056

RESUMEN

Adenovirus serves as an excellent viral vector and is employed in vector vaccine research. Duck hepatitis A virus type 1 (DHAV1) and duck adenovirus type 3 (DAdV3) cause significant economic losses in the Chinese duck industry. In this study, we found an excellent exogenous gene insertion site in DAdV3 genome of CH-GD-12-2014 strain, within 3 intergenic regions (IGR). Subsequently, we generated a recombinant duck adenovirus named rDAdV3-VP1-188, which exhibits excellent replication characteristics and immunogenicity of DAdV3 and DHAV1. Animal experiments showed that rDAdV3-VP1-188 can provide 100% protection against the DAdV3 and 80% protection against DHAV1. These results showed that rDAdV3-VP1-188 could induce protection against DAdV3 and DHAV1 in ducks, thus indicating the feasibility of DAdV3 as a vector for the development of avian vector vaccines. These insights contribute to the further development of DAdV3 vectors and other adenovirus vectors.


Asunto(s)
Virus de la Hepatitis B del Pato , Virus de la Hepatitis del Pato , Enfermedades de las Aves de Corral , Animales , Virus de la Hepatitis del Pato/genética , Patos , Proteínas de la Cápside/genética , Adenoviridae/genética , Pollos , Proteínas Recombinantes/genética , Proteínas Virales
6.
Virus Res ; 336: 199216, 2023 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-37657508

RESUMEN

Duck hepatitis A virus (DHAV) is one of key pathogens for duck viral hepatitis, especially in Asian duck industry. Currently, two main genotypes (DHAV-1 and -3) exist. To explore insightfully the evolutionary character, we assessed the available 141 full-length genome sequences of DHAV isolated in 1986-2020 globally and divided DHAV-1 and DHAV-3 into further seven (DHAV-1 a-g) and five (DHAV-3 a-e) sub-clades, respectively. Phylogenetic and phylogeographic network analyses indicated great genetic diversity of DHAV identified in China, where the DHAV-1 cluster and DHAV-3 cluster were linked by virus strain HDHV1-BJ (GenBank ID: FJ157172.1) and Du_CH_LSD_090612 (GenBank ID: JF828995.1) via a long mutational branch and intermediate strains. Several strains previously identified as DHAV-1 according to the partial gene sequences were actually clustered within DHAV-3 in full-length genome-based analysis. Furthermore, we identified 32 recombination events across virus genome with the recombination hotspot at the 5' end and upstream of the capsid coding region. The highest variability of DHAV polyprotein was shown at the upstream region of the N terminus P-loop region, e.g., amino acids 672-716, followed by the aa 334-359 in the Capsid encoding region. The results presented here provides a robust insight into the genetic exchange patterns of DHAV genomes during the past decades, which may be used to map the evolutionary history and facilitate preventive measures of DHAVs.

7.
BMC Vet Res ; 19(1): 179, 2023 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-37773135

RESUMEN

BACKGROUND: Duck hepatitis A virus (DHAV) is a single-stranded, positive-strand small RNA virus that causes a very high mortality rate in ducklings. The DHAV-3 subtype incidence rate has recently increased in China, causing great economic losses to the waterfowl breeding industry. We analyzed the protection rate of DHAV vaccines used in mainland China from 2009 to 2021 and evaluated the effectiveness of vaccine prevention and control to reduce the economic losses caused by DHAV to the waterfowl breeding industry. We screened five electronic research databases and obtained 14 studies and patents on the protection efficiency of DHAV-1 and DHAV-3 vaccines. RESULTS: Meta-analysis demonstrated that immunized ducklings produced higher antibody levels and had a significantly higher survival rate than non-immunized ducklings [relative risk (RR) = 12, 95% confidence interval (CI) 6-26, P < 0.01]. The age of the ducks and vaccine valence did not affect protection efficiency. Data source analysis of the vaccine protection rate demonstrated that the vaccines conferred immune protection for ducklings in both small-scale experiments and large-scale clinical conditions. The analysis results revealed that although the vaccines conferred protection, the immune protective effect differed between small-scale experimental conditions and large-scale clinical conditions. This might have been due to non-standard vaccination and environmental factors. CONCLUSIONS: Domestic DHAV vaccines can protect ducklings effectively. The subjects immunized (breeding ducks or ducklings) and vaccine valence had no effect on the protective effect. Both small-scale experiments and large-scale clinical conditions conferred immune protection on ducklings, but vaccine immunization under small-scale experimental conditions had slightly better protective effects than large-scale clinical immunization.


Asunto(s)
Virus de la Hepatitis del Pato , Hepatitis Viral Animal , Infecciones por Picornaviridae , Enfermedades de las Aves de Corral , Vacunas , Animales , Infecciones por Picornaviridae/veterinaria , Vacunación/veterinaria , China/epidemiología , Patos
8.
Poult Sci ; 102(7): 102724, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37207573

RESUMEN

The placenta-specific 8 (PLAC8) gene, also known as ONZIN or C15, codes for a cysteine-rich peptide originally identified in mouse placental tissue and subsequently identified in a variety of epithelial tissues and immune cells. PLAC8 is also expressed in birds, such as ducks, where its functional roles remain unknown. Here, we aimed to determine the mRNA and protein expression profiles and the functional role of duck PLAC8 during the infection of duck hepatitis A virus type 1 (DHAV-1). We found that the duck PLAC8 is also a cysteine-rich polypeptide composed of 114 amino acid residues, with no signal peptide. Duck PLAC8 is highly expressed in the immune organs of young cherry valley ducks, including the thymus, bursa fabricius, and spleen. However, it has negligible expression level in liver, brain, kidney, and heart. Additionally, PLAC8 expression was considerably induced after DHAV-1 infection both in vitro and in vivo, especially in the immune organs of ducklings. This tissue expression distribution and induction upon infection suggest that PLAC8 might play a critical role in innate immunity. Our data showed that PLAC8 significantly suppressed the expression of Toll-like receptor 7 (TLR7), leading to decreased expression of downstream signaling molecules including myeloid differentiation primary response gene 88 (MyD88) and nuclear factor kappa-B (NF-κB). This ultimately resulted in low levels of type I interferon and interleukin 6 (IL-6). Additionally, PLAC8 positively regulated DHAV-1 replication levels. RNAi against PLAC8 in duck embryo fibroblasts considerably inhibited DHAV-1 propagation, while PLAC8 overexpression significantly facilitated DHAV-1 replication.


Asunto(s)
Patos , Virus de la Hepatitis del Pato , Hepatitis Viral Animal , Infecciones por Picornaviridae , Enfermedades de las Aves de Corral , Animales , Femenino , Ratones , Embarazo , Cisteína , Patos/genética , Patos/virología , Factor 88 de Diferenciación Mieloide/genética , Infecciones por Picornaviridae/veterinaria , Placenta , Transducción de Señal , Receptor Toll-Like 7/genética , Células HEK293 , Humanos
9.
Vet Res Commun ; 47(3): 1231-1241, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-36595200

RESUMEN

Sudden death of ducklings was reported in a duck farm located at Tiruvallur district in Tamil Nadu, India. Disease investigation began with post mortem findings of dead birds revealing enlarged pale-pink / pale-yellow liver with multifocal petechiae and ecchymosis. A positive amplification with duck hepatitis A virus specific primers by reverse transcription-polymerase chain reaction (RT-PCR) on the tissue samples collected from dead birds indicated infection by duck hepatitis A virus (DHAV), an avian picornavirus, known to cause acute and high-mortality in ducklings. The virus isolation was successful in 9-days old embryonated chicken eggs, in primary chicken embryo fibroblast (CEF) cells and from experimentally infected ducklings. The embryonic death on day 5 to 7 post inoculation in chicken embryos with signs of cutaneous hemorrhage, edema and greenish yellow liver together with histopathology of embryonic liver and kidney further confirmed DHAV infection. TEM analysis of the infected allantoic fluid and infected CEF cell culture supernatant showed the presence of spherical shaped, non-enveloped virion particles of ~ 20-38 nm diameter, typical for DHAV. Experimental infection of ducklings with RT-PCR positive tissue supernatant caused 40% to 50% mortality with typical petechial hemorrhages on the surface of liver. Further, histopathological analysis and RT-PCR of the inoculated duckling's tissues confirmed the presence of DHAV. Nucleotide sequencing of the 5'UTR region and VP1 region confirmed duck hepatitis A virus genotype 2 (DHAV-2). To the best of our knowledge, this is the first report of laboratory confirmation of DHAV-2 in India. This study warrants the need for the extensive epidemiological surveillance to understand the prevalence of DHAV-2 in India and to take appropriate control measures to curtail the disease spread.


Asunto(s)
Virus de la Hepatitis del Pato , Infecciones por Picornaviridae , Enfermedades de las Aves de Corral , Embrión de Pollo , Animales , Virus de la Hepatitis del Pato/genética , India/epidemiología , Enfermedades de las Aves de Corral/epidemiología , Infecciones por Picornaviridae/epidemiología , Infecciones por Picornaviridae/veterinaria , Patos , Genotipo
10.
Vet Res ; 54(1): 5, 2023 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-36703166

RESUMEN

Duck hepatitis A virus type 1 (DHAV-1) is an acute, highly lethal infectious agent that infects ducklings and causes up to 95% mortality in ducklings up to 1 week of age, posing a significant economic threat to the duck farming industry. Previous studies have found that the proteolytic enzyme 3 C encoded by DHAV-1 can inhibit the IRF7 protein from blocking the upstream signaling pathway of the type I interferon to promote viral replication. However, there are still few studies on the mechanism of DHAV-1 in immune evasion. Here, we demonstrate that the DHAV-1 3CD protein can interact with IRF7 protein and reduce IRF7 protein expression without directly affecting IRF7 protein nuclear translocation. Further studies showed that the 3CD protein could reduce the expression of RIG-I protein without affecting its transcription level. Furthermore, we found that the 3CD protein interacted with the N-terminal structural domain of RIG-I protein, interfered with the interaction between RIG-I and MAVS, and degraded RIG-I protein through the proteasomal degradation pathway, thereby inhibiting its mediated antiviral innate immunity to promote DHAV-1 replication. These data suggest a novel immune evasion mechanism of DHAV-1 mediated by the 3CD protein, and the results of this experiment are expected to improve the understanding of the biological functions of the viral precursor protein and provide scientific data to elucidate the mechanism of DHAV-1 infection and pathogenesis.


Asunto(s)
Virus de la Hepatitis del Pato , Interferón Tipo I , Animales , Inmunidad Innata , Transducción de Señal , Proteínas Virales , Patos
11.
Avian Pathol ; 52(2): 89-99, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36571394

RESUMEN

Duck viral hepatitis (DVH), mainly caused by duck hepatitis A virus (DHAV), is a highly fatal and rapidly spreading infectious disease of young ducklings that seriously jeopardizes the duck industry worldwide. DHAV type 1 (DHAV-1) is the main genotype responsible for disease outbreaks since 1945, and the disease situation is complicated by the emergence and dissemination of a novel genotype (DHAV-3) in some countries in Asia and Africa. Live attenuated DHAV vaccines are widely used to induce a considerable degree of protection in ducklings. Breeder ducks are immunized with inactivated or/and live DHAV vaccines to achieve satisfactory levels of passive immunity in progeny. In addition, novel characteristics of virus transmission, pathogenicity and pathogenesis of DHAV were recently characterized, necessitating the development of new vaccines and effective vaccination programmes against DVH. Therefore, a systematic dissection of the profiles, strengths and shortcomings of the available DHAV vaccines is essential. Moreover, to further increase the efficiency of vaccine production and administration, the development of next-generation DHAV vaccines using cutting-edge technologies is also required. In this review, based on a comprehensive summary of the research advances in the epidemiology, pathogenicity, and genomic features of DHAV, we focus on reviewing and analysing the features of the commercial and experimental DHAV vaccines. We also propose perspectives for disease control based on the specific disease situations in different countries. This review provides essential information for vaccine development and disease control of DVH.


Asunto(s)
Virus de la Hepatitis del Pato , Hepatitis Viral Animal , Infecciones por Picornaviridae , Enfermedades de las Aves de Corral , Animales , Infecciones por Picornaviridae/veterinaria , Vacunas Atenuadas , Vacunación/veterinaria , Patos
12.
Animals (Basel) ; 14(1)2023 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-38200736

RESUMEN

To investigate the distribution and genetic variation in four vertically transmitted duck pathogens, including duck hepatitis B virus (DHBV), duck circovirus (DuCV), duck hepatitis A virus 3 (DHAV-3), and avian reoviruses (ARV), we conducted an epidemiology study using PCR and RT-PCR assays on a duck population. We found that DHBV was the most prevalent virus (69.74%), followed by DuCV (39.48%), and then ARV (19.92%) and DHAV-3 (8.49%). Among the 271 duck samples, two, three or four viruses were detected in the same samples, indicating that the coinfection of vertical transmission agents is common in ducks. The genetic analysis results showed that all four identified DuCV strains belonged to genotype 1, the DHAV-3 strain was closely clustered with previously identified strains from China, and the ARV stain was clustered under genotype 1. These indicate that different viral strains are circulating among the ducks. Our findings will improve the knowledge of the evolution of DuCV, DHAV-3, and ARV, and help choose suitable strains for vaccination.

13.
Front Microbiol ; 13: 1064612, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36578574

RESUMEN

DNAzymes effectively inhibit the expression of viral genes. Duck hepatitis A virus type-1 (DHAV-1) genomic RNA carries an internal ribosome entry site (IRES). The IRES initiates the translation of DHAV-1 via a mechanism that differs from that of cap-dependent translation. Therefore, it is an attractive target for the treatment of DHAV-1. In this study, we designed 6 DNAzymes (Dzs) specifically targeting 300-618 nt sequence in the DHAV-1 5'untranslated region (UTR; a predicted IRES-like element). In the presence of divalent metal ions, three designed DNAzymes (DZ369, DZ454, and DZ514) efficiently cleaved the 300-618 nt sequence of the DHAV-1 5'UTR RNA. The activity of the Dzs was particularly dependent on Mg2+ ions. Subsequently, the translation inhibitory activity of these Dzs was determined by western blotting experiments. The Dzs effectively inhibited the translation mediated by the 300-618 nt of DHAV-1 5'UTR in duck embryo fibroblasts (DEFs). Importantly, DZ454 showed the strongest inhibitory effect, and its inhibition was time and dose dependent. However, none of the Dzs showed significant inhibition of cap-dependent translation. These results suggest that these Dzs show specificity for target RNA. Moreover, DZ454 inhibited the replication of DHAV-1. In conclusion, the designed DNAzymes can be used as inhibitors of DHAV-1 RNA translation and replication, providing new insights useful for the development of anti-DHAV-1 drugs.

14.
Vet Res ; 53(1): 64, 2022 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-35978392

RESUMEN

Duck hepatitis A virus type 1 (DHAV-1) is one of the main pathogens responsible for death in ducklings. Autophagy is a catabolic process that maintains cellular homeostasis, and the PI3KC3 protein plays an important role in the initiation of autophagy. DHAV-1 infection induces autophagy in duck embryo fibroblasts (DEFs) but the molecular mechanism between it and autophagy has not been reported. First, we determined that DHAV-1 infection induces autophagy in DEFs and that autophagy induction is dependent on the integrity of viral proteins by infecting DEFs with UV-inactivated or heat-inactivated DHAV-1. Then, in experiments using the pharmacological autophagy inducer rapamycin and the autophagy inhibitor chloroquine, autophagy inhibition was shown to reduce intracellular and extracellular DHAV-1 genome copies and viral titres. These results suggest that autophagy activated by DHAV-1 infection in DEFs affects DHAV-1 proliferation and extracellular release. Next, we screened the autophagy-inducing effects of the DHAV-1 structural proteins VP0, VP3, and VP1 and found that all DHAV-1 structural proteins could induce autophagy in DEFs but not the full autophagic flux. Finally, we found that VP1 promotes protein expression of PI3KC3 and Beclin1 by western blot experiments and that VP1 interacts with PI3KC3 by co-immunoprecipitation experiments; moreover, 3-MA-induced knockdown of PI3KC3 inhibited VP1 protein-induced autophagy in DEFs. In conclusion, the DHAV-1 structural protein VP1 regulates the PI3KC3 complex by interacting with PI3KC3 to induce autophagy in DEFs.


Asunto(s)
Virus de la Hepatitis del Pato , Hepatitis Viral Animal , Infecciones por Picornaviridae , Enfermedades de las Aves de Corral , Animales , Autofagia , Beclina-1 , Patos , Virus de la Hepatitis del Pato/fisiología , Infecciones por Picornaviridae/veterinaria
15.
Front Immunol ; 13: 839677, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35757688

RESUMEN

Host translation is generally modulated by viral infection, including duck hepatitis A virus (DHAV) infection. Previously, we reported that cellular protein synthesis in a cell model of duck embryo fibroblasts is significantly inhibited by DHAV infection but not viral proteins, suggesting that an important viral-host interaction occurs at the translational level. In this study, we aim to further understand the impact of DHAV virulence on cellular N6-methyladenosine (m6A) modification, which is essential to a wide variety of RNA biological processes, such as mRNA stabilization and translation. Using m6A antibody-based immunoprecipitation, m6A-seq, and LC-MS/MS, we observed that m6A-modified mRNA exists in both virulent and attenuated DHAV-infected duckling livers. Importantly, m6A levels in mRNA were much higher in attenuated DHAV-infected livers compared with virulent DHAV-infected livers, suggesting virulence-dependent regulation of m6A modification. Analysis of modification motifs indicated that GAAGAAG is the most enriched motif. Combined m6A-seq and RNA-seq data analysis indicated a generally positive correlation between m6A and mRNA expression levels in DHAV-infected duckling livers. GO analysis of genes with decreased or increased m6A levels showed that these genes were enriched in various terms, including oxidation-reduction processes and antiviral immune responses. Collectively, our work reveals DHAV virulence-dependent coordination between m6A modification and mRNA expression in duckling livers.


Asunto(s)
Virus de la Hepatitis A , Virus de la Hepatitis del Pato , Hepatitis Viral Animal , Infecciones por Picornaviridae , Enfermedades de las Aves de Corral , Animales , Cromatografía Liquida , Patos , Virus de la Hepatitis A/genética , Virus de la Hepatitis del Pato/genética , ARN Mensajero/genética , Espectrometría de Masas en Tándem
16.
Virol J ; 19(1): 111, 2022 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-35761382

RESUMEN

BACKGROUND: Duck hepatitis A virus type 1 (DHAV-1) is one of the most serious pathogens endangering the duck industry. However, there are few studies on the regulation of the cell cycle by DHAV-1. METHODS: In this study, flow cytometry was applied to analyze the effect of DHAV-1 infection on the cell cycle of duck embryo fibroblasts (DEFs). Subsequently, we analyzed the effects of cell cycle phases on DHAV-1 replication by real-time reverse transcriptase quantitative PCR (real-time RT-qPCR). RESULTS: Flow cytometry data analysis found that DEFs in the S phase increased by 25.85% and 54.21% at 24 h and 48 h after DHAV-1 infection, respectively. The levels of viral RNA detected by real-time RT-qPCR were higher in the DEFs with synchronization in the S phase or G0/G1 phase than in the control group. However, there was no difference in viral copy number between the G2/M phase arrest and control groups. In addition, non-structural protein 3D of DHAV-1 significantly increased cells in the S phase, indicating that 3D protein is one of the reasons for the cell cycle arrest in the S phase. CONCLUSIONS: In summary, DHAV-1 infection induces the cell cycle arrest of DEFs in the S phase. Both S phase and G0/G1 phase synchronization facilitate the replication of DHAV-1, and 3D protein is one of the reasons for the S phase arrest.


Asunto(s)
Virus de la Hepatitis del Pato , Hepatitis Viral Animal , Animales , Puntos de Control del Ciclo Celular , Patos , Virus de la Hepatitis del Pato/genética , Fase S
17.
Vaccines (Basel) ; 10(4)2022 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-35455272

RESUMEN

In this study, we sought to develop a subunit vaccine against the increasingly prevalent Duck hepatitis A virus serotype 3 (DHAV-3). The VP1 protein of DHAV-3 and a truncated version containing the C-terminal region of VP1, termed VP1-C, were expressed recombinantly in Escherichia coli as vaccine antigens. For enhanced immune response, a truncated version of flagellin, nFliC, was included as vaccine adjuvant. Ducklings were vaccinated once for immune response analysis and challenge test. Results showed that VP1-C elicited a higher level of virus-specific antibody response and neutralization titer than VP1. The addition of nFliC further enhanced the antibody response. In terms of cellular immune response, the VP1-C + nFliC vaccine elicited the highest level of T cell proliferation among the vaccine formulations tested. Examination of the cytokine expression profile showed that peripheral blood mononuclear cells from the VP1-C + nFliC vaccine group expressed the highest levels of pro-inflammatory (IL-6) and TH-1 type (IL-12 and IFN-γ) cytokines. Finally, in a DHAV-3 challenge test, the VP1-C + nFliC vaccine provided a 75% protection rate (n = 8), in contrast to 25% for the VP1 vaccine. In conclusion, E. coli-expressed VP1-C has been shown to be a promising antigen when combined with nFliC and may be further developed as a single-dose subunit vaccine against DHAV-3.

18.
Front Microbiol ; 12: 700434, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34867836

RESUMEN

Duck hepatitis A virus (DHAV), which mainly infects 1- to 4-week-old ducklings, has a fatality rate of 95% and poses a huge economic threat to the duck industry. However, the mechanism by which DHAV-1 regulates the immune response of host cells is rarely reported. This study examined whether DHAV-1 contains a viral protein that can regulate the innate immunity of host cells and its specific regulatory mechanism, further exploring the mechanism by which DHAV-1 resists the host immune response. In the study, the dual-luciferase reporter gene system was used to screen the viral protein that regulates the host innate immunity and the target of this viral protein. The results indicate that the DHAV-1 3C protein inhibits the pathway upstream of interferon (IFN)-ß by targeting the interferon regulatory factor 7 (IRF7) protein. In addition, we found that the 3C protein inhibits the nuclear translocation of the IRF7 protein. Further experiments showed that the 3C protein interacts with the IRF7 protein through its N-terminus and that the 3C protein degrades the IRF7 protein in a caspase 3-dependent manner, thereby inhibiting the IFN-ß-mediated antiviral response to promote the replication of DHAV-1. The results of this study are expected to serve as a reference for elucidating the mechanisms of DHAV-1 infection and pathogenicity.

19.
Vaccines (Basel) ; 9(12)2021 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-34960225

RESUMEN

With the continuous development of duck farming and the increasing breeding density, the incidence of duck hepatitis A virus type 1 (DHAV-1) has been on the rise, seriously endangering the development of duck farming. To reduce the use of antibiotics in duck breeding, susceptibility risks and mortality, and avoid virulence recovery and immune failure risk, this study aims to develop a new type of mucosal immune probiotics and make full use of molecular biology techniques, on the level of genetic engineering, to modify Lactococcus lactis (L. lactis). In this study, a secretory recombinant L. lactis named MG1363-VP1 with an enhanced Green Fluorescent Protein (eGFP) and translation enhancer T7g10L was constructed, which could express the VP1-eGFP fusion protein of DHAV-1. The animal experiment in ducklings was performed to detect the immune response and protection effect of oral microecologics by recombinant L. lactis. The results showed that oral L. lactis MG1363-VP1 significantly induced the body's humoral immune system and mucosal immune system to produce specific anti-VP1 IgG antibodies and mucosal secretory immunoglobulin A (sIgA) for DHAV-1 in ducklings, and cytokines including interleukin-2 (IL-2), interleukin-4 (IL-4), interleukin-10 (IL-10), and interferon gamma (IFN-γ). The mortality rate was monitored simultaneously by the natural infestation in the process of production and breeding; notably, the ducklings vaccinated with L. lactis MG1363-VP1 were effectively protected against the nature infection of DHAV-1. The recombinant L. lactis MG1363-VP1 constructed in this study provides a new means of preventing and controlling DHAV-1 infection in the future.

20.
Avian Dis ; 65(1): 1-9, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-34339115

RESUMEN

Duck hepatitis A virus (DHAV) causes acute hepatitis and mortality, resulting in high economic losses in the duck farm industry. The current study describes the outbreak of DHAV in vaccinated duck farms in North Egypt during 2019 and molecular characterization of the 3' untranslated region (UTR) and viral protein VP1 genes. The 30 samples were collected from 7- to 28-day-old commercial Pekin ducks that showed a history of nervous signs and sudden deaths and were on farms in 6 governorates. DHAV was typed by reverse transcription-polymerase chain reaction (RT-PCR) for 3' UTR and VP1 genes and revealed 20 positive farms, with the first detection of DHAV genotype 3 (DHAV-3) in 18 samples and the classic DHAV-1 in 2 samples. The phylogenetic analysis of VP1 and 3' UTR genes of the nine selected strains representative of six governorates revealed that seven strains were clustered with DHAV-3 Chinese and Korean-Vietnamese strains within different subgroups with 92.4%-93.7% amino acid identity; such strains were distinguishable from the vaccine strain of DHAV-1 used in Egypt with 74.4% amino acid identity. The other strains were closely related to the DHAV-1 Asian strain and the vaccine strain used in Egypt with 98.7%-99.6% amino acid identity for the VP1 gene with different clustering than that of recently isolated DHAV-1 Egyptian strains. The VP1 gene of DHAV-3 had 1 hypervariable region (HVR) with 10 amino acid mutations compared with DHAV3/DN2/Vietnam/2011, but DHAV-1 had 3 HVRs with 1 amino acid mutation in HVRII compared with the DHAV-1 vaccine strain. In conclusion, a new introduction of DHAV-3 with the classical DHAV-1 was recorded in Pekin duck farms in North Egypt that is genetically distant from the vaccinal strain.


Artículo regular­Circulacíon dual de los genotipos 1 y 3 del virus de la hepatitis A del pato en Egipto. El virus de la hepatitis A del pato (con las siglas en inglés DHAV) causa hepatitis aguda y mortalidad, lo que genera grandes pérdidas económicas en la industria de la críanza de patos. El estudio actual describe un brote del virus de la hepatitis A del pato en una granja de patos vacunados en el norte de Egipto durante el año 2019 y la caracterización molecular de los genes de la región no traducida 3' (3' UTR) y la proteína viral VP1. Las 30 muestras se recolectaron de patos Pekin comerciales de 7 a 28 días de edad que presentaban antecedentes de signos nerviosos y muerte súbita y se encontraban en granjas de seis gobernaciones. El virus de la hepatitis A del pato se tipificó mediante la transcripción inversa y reacción en cadena de la polimerasa (RT-PCR) para los genes 3' UTR y VP1 y reveló 20 granjas positivas, con la primera detección del genotipo 3 del virus de la hepatitis A del pato (DHAV-3) en 18 muestras y la detección del virus clásico de la hepatitis A del pato tipo1 en dos muestras. El análisis filogenético de los genes VP1 y 3' UTR de las nueve cepas seleccionadas representativas de seis provincias reveló que siete cepas se agruparon con cepas del virus de la hepatitis A del pato 3 chinas y coreano-vietnamitas dentro de diferentes subgrupos con una identidad de aminoácidos del 92.4% al 93.7%; dichas cepas se distinguían de la cepa vacunal del virus de la hepatitis A del pato tipo 1 utilizada en Egipto con 74.4% de identidad de aminoácidos. Las otras cepas estaban estrechamente relacionadas con la cepa asiática del virus de la hepatitis A del pato tipo 1 y la cepa de vacuna utilizada en Egipto con 98.7% -99.6% de identidad de aminoácidos para el gene VP1 con agrupaciones diferentes a las de las cepas egipcias de virus de la hepatitis A del pato tipo 1 aisladas recientemente. El gene VP1 del virus de la hepatitis A del pato tipo 3 tenía una región hipervariable (HVR) con 10 mutaciones en la secuencia de aminoácidos en comparación con la cepa DHAV3/ DN2/Vietnam/2011, pero el virus de la hepatitis A del pato tipo 1 tenía tres regiones hipervariables con una mutación de aminoácidos en la zona hipervariable II en comparación con la cepa de vacuna virus de la hepatitis A del pato tipo 1. En conclusión, se registró una nueva introducción del virus de la hepatitis A del pato tipo 3 con el virus de la hepatitis A del pato clásico tipo 1 en granjas de patos Pekín en el norte de Egipto, que está genéticamente distante de la cepa vacunal.


Asunto(s)
Patos , Virus de la Hepatitis del Pato/genética , Hepatitis Viral Animal/epidemiología , Infecciones por Picornaviridae/veterinaria , Enfermedades de las Aves de Corral/epidemiología , Secuencia de Aminoácidos , Animales , Egipto/epidemiología , Genotipo , Hepatitis Viral Animal/virología , Filogenia , Infecciones por Picornaviridae/epidemiología , Infecciones por Picornaviridae/virología , Enfermedades de las Aves de Corral/virología , Prevalencia , Alineación de Secuencia/veterinaria
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA