Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.901
Filter
1.
Brief Bioinform ; 25(3)2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38752857

ABSTRACT

Avian reoviruses continue to cause disease in turkeys with varied pathogenicity and tissue tropism. Turkey enteric reovirus has been identified as a causative agent of enteritis or inapparent infections in turkeys. The new emerging variants of turkey reovirus, tentatively named turkey arthritis reovirus (TARV) and turkey hepatitis reovirus (THRV), are linked to tenosynovitis/arthritis and hepatitis, respectively. Turkey arthritis and hepatitis reoviruses are causing significant economic losses to the turkey industry. These infections can lead to poor weight gain, uneven growth, poor feed conversion, increased morbidity and mortality and reduced marketability of commercial turkeys. To combat these issues, detecting and classifying the types of reoviruses in turkey populations is essential. This research aims to employ clustering methods, specifically K-means and Hierarchical clustering, to differentiate three types of turkey reoviruses and identify novel emerging variants. Additionally, it focuses on classifying variants of turkey reoviruses by leveraging various machine learning algorithms such as Support Vector Machines, Naive Bayes, Random Forest, Decision Tree, and deep learning algorithms, including convolutional neural networks (CNNs). The experiments use real turkey reovirus sequence data, allowing for robust analysis and evaluation of the proposed methods. The results indicate that machine learning methods achieve an average accuracy of 92%, F1-Macro of 93% and F1-Weighted of 92% scores in classifying reovirus types. In contrast, the CNN model demonstrates an average accuracy of 85%, F1-Macro of 71% and F1-Weighted of 84% scores in the same classification task. The superior performance of the machine learning classifiers provides valuable insights into reovirus evolution and mutation, aiding in detecting emerging variants of pathogenic TARVs and THRVs.


Subject(s)
Machine Learning , Orthoreovirus, Avian , Reoviridae Infections , Turkeys , Animals , Orthoreovirus, Avian/genetics , Orthoreovirus, Avian/classification , Orthoreovirus, Avian/pathogenicity , Turkeys/virology , Reoviridae Infections/virology , Poultry Diseases/virology , Phylogeny
2.
PLoS Pathog ; 20(8): e1012468, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39146367

ABSTRACT

Genetic editing of the germline using CRISPR/Cas9 technology has made it possible to alter livestock traits, including the creation of resistance to viral diseases. However, virus adaptability could present a major obstacle in this effort. Recently, chickens resistant to avian leukosis virus subgroup J (ALV-J) were developed by deleting a single amino acid, W38, within the ALV-J receptor NHE1 using CRISPR/Cas9 genome editing. This resistance was confirmed both in vitro and in vivo. In vitro resistance of W38-/- chicken embryonic fibroblasts to all tested ALV-J strains was shown. To investigate the capacity of ALV-J for further adaptation, we used a retrovirus reporter-based assay to select adapted ALV-J variants. We assumed that adaptive mutations overcoming the cellular resistance would occur within the envelope protein. In accordance with this assumption, we isolated and sequenced numerous adapted virus variants and found within their envelope genes eight independent single nucleotide substitutions. To confirm the adaptive capacity of these substitutions, we introduced them into the original retrovirus reporter. All eight variants replicated effectively in W38-/- chicken embryonic fibroblasts in vitro while in vivo, W38-/- chickens were sensitive to tumor induction by two of the variants. Importantly, receptor alleles with more extensive modifications have remained resistant to the virus. These results demonstrate an important strategy in livestock genome engineering towards antivirus resistance and illustrate that cellular resistance induced by minor receptor modifications can be overcome by adapted virus variants. We conclude that more complex editing will be necessary to attain robust resistance.


Subject(s)
Avian Leukosis Virus , Avian Leukosis , Chickens , Animals , Avian Leukosis Virus/genetics , Avian Leukosis Virus/physiology , Chickens/virology , Avian Leukosis/virology , Avian Leukosis/genetics , Poultry Diseases/virology , Poultry Diseases/genetics , Disease Resistance/genetics , CRISPR-Cas Systems , Gene Editing , Chick Embryo , Evolution, Molecular , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Fibroblasts/virology , Fibroblasts/metabolism
3.
PLoS Pathog ; 20(7): e1012415, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39078847

ABSTRACT

Like all coronaviruses, infectious bronchitis virus, the causative agent of infectious bronchitis in chickens, exhibits a high mutation rate. Adaptive mutations that arise during the production of live attenuated vaccines against IBV often decrease virulence. The specific impact of these mutations on viral pathogenicity, however, has not been fully elucidated. In this study, we identified a mutation at the 3' end of the S gene in an IBV strain that was serially passaged in chicken embryos, and showed that this mutation resulted in a 9-aa truncation of the cytoplasmic tail (CT) of the S protein. This phenomenon of CT truncation has previously been observed in the production of attenuated vaccines against other coronaviruses such as the porcine epidemic diarrhea virus. We next discovered that the 9-aa truncation in the S protein CT resulted in the loss of the endoplasmic-reticulum-retention signal (KKSV). Rescue experiments with recombinant viruses confirmed that the deletion of the KKSV motif impaired the localization of the S protein to the endoplasmic-reticulum-Golgi intermediate compartment (ERGIC) and increased its expression on the cell surface. This significantly reduced the incorporation of the S protein into viral particles, impaired early subgenomic RNA and protein synthesis, and ultimately reduced viral invasion efficiency in CEK cells. In vivo experiments in chickens confirmed the reduced pathogenicity of the mutant IBV strains. Additionally, we showed that the adaptive mutation altered the TRS-B of ORF3 and impacted the transcriptional regulation of this gene. Our findings underscore the significance of this adaptive mutation in the attenuation of IBV infection and provide a novel strategy for the development of live attenuated IBV vaccines.


Subject(s)
Chickens , Coronavirus Infections , Infectious bronchitis virus , Poultry Diseases , Spike Glycoprotein, Coronavirus , Vaccines, Attenuated , Animals , Infectious bronchitis virus/genetics , Infectious bronchitis virus/pathogenicity , Chick Embryo , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/metabolism , Coronavirus Infections/virology , Vaccines, Attenuated/genetics , Poultry Diseases/virology , Poultry Diseases/genetics , Virulence , Viral Vaccines/genetics , Viral Vaccines/immunology , Mutation
4.
PLoS Pathog ; 20(5): e1012261, 2024 May.
Article in English | MEDLINE | ID: mdl-38805555

ABSTRACT

Marek's disease virus (MDV) vaccines were the first vaccines that protected against cancer. The avirulent turkey herpesvirus (HVT) was widely employed and protected billions of chickens from a deadly MDV infection. It is also among the most common vaccine vectors providing protection against a plethora of pathogens. HVT establishes latency in T-cells, allowing the vaccine virus to persist in the host for life. Intriguingly, the HVT genome contains telomeric repeat arrays (TMRs) at both ends; however, their role in the HVT life cycle remains elusive. We have previously shown that similar TMRs in the MDV genome facilitate its integration into host telomeres, which ensures efficient maintenance of the virus genome during latency and tumorigenesis. In this study, we investigated the role of the TMRs in HVT genome integration, latency, and reactivation in vitro and in vivo. Additionally, we examined HVT infection of feather follicles. We generated an HVT mutant lacking both TMRs (vΔTMR) that efficiently replicated in cell culture. We could demonstrate that wild type HVT integrates at the ends of chromosomes containing the telomeres in T-cells, while integration was severely impaired in the absence of the TMRs. To assess the role of TMRs in vivo, we infected one-day-old chickens with HVT or vΔTMR. vΔTMR loads were significantly reduced in the blood and hardly any virus was transported to the feather follicle epithelium where the virus is commonly shed. Strikingly, latency in the spleen and reactivation of the virus were severely impaired in the absence of the TMRs, indicating that the TMRs are crucial for the establishment of latency and reactivation of HVT. Our findings revealed that the TMRs facilitate integration of the HVT genome into host chromosomes, which ensures efficient persistence in the host, reactivation, and transport of the virus to the skin.


Subject(s)
Chickens , Marek Disease , Telomere , Virus Integration , Virus Latency , Animals , Chickens/virology , Telomere/genetics , Telomere/virology , Marek Disease/virology , Marek Disease/immunology , Marek Disease/prevention & control , Genetic Vectors , Herpesvirus 1, Meleagrid/genetics , Herpesvirus 1, Meleagrid/immunology , Marek Disease Vaccines/immunology , Marek Disease Vaccines/genetics , Genome, Viral , Herpesvirus 2, Gallid/genetics , Herpesvirus 2, Gallid/immunology , Repetitive Sequences, Nucleic Acid , Poultry Diseases/virology , Poultry Diseases/immunology , Poultry Diseases/prevention & control
5.
PLoS Pathog ; 20(5): e1012232, 2024 May.
Article in English | MEDLINE | ID: mdl-38743760

ABSTRACT

Infectious bronchitis virus (IBV) is a coronavirus that infects chickens, which exhibits a broad tropism for epithelial cells, infecting the tracheal mucosal epithelium, intestinal mucosal epithelium, and renal tubular epithelial cells. Utilizing single-cell RNA sequencing (scRNA-seq), we systematically examined cells in renal, bursal, and tracheal tissues following IBV infection and identified tissue-specific molecular markers expressed in distinct cell types. We evaluated the expression of viral RNA in diverse cellular populations and subsequently ascertained that distal tubules and collecting ducts within the kidney, bursal mucosal epithelial cells, and follicle-associated epithelial cells exhibit susceptibility to IBV infection through immunofluorescence. Furthermore, our findings revealed an upregulation in the transcription of proinflammatory cytokines IL18 and IL1B in renal macrophages as well as increased expression of apoptosis-related gene STAT in distal tubules and collecting duct cells upon IBV infection leading to renal damage. Cell-to-cell communication unveiled potential interactions between diverse cell types, as well as upregulated signaling pathways and key sender-receiver cell populations after IBV infection. Integrating single-cell data from all tissues, we applied weighted gene co-expression network analysis (WGCNA) to identify gene modules that are specifically expressed in different cell populations. Based on the WGCNA results, we identified seven immune-related gene modules and determined the differential expression pattern of module genes, as well as the hub genes within these modules. Our comprehensive data provides valuable insights into the pathogenesis of IBV as well as avian antiviral immunology.


Subject(s)
Cell Communication , Chickens , Coronavirus Infections , Gene Regulatory Networks , Infectious bronchitis virus , Single-Cell Analysis , Animals , Infectious bronchitis virus/genetics , Infectious bronchitis virus/physiology , Coronavirus Infections/virology , Coronavirus Infections/genetics , Poultry Diseases/virology , Poultry Diseases/genetics , Poultry Diseases/immunology , Sequence Analysis, RNA , Epithelial Cells/virology , Epithelial Cells/metabolism
6.
J Virol ; 98(5): e0018124, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38639485

ABSTRACT

Infectious bursal disease (IBD) is an acute and fatal immunosuppressive disease caused by infectious bursal disease virus (IBDV). As an obligate intracellular parasite, IBDV infection is strictly regulated by host factors. Knowledge on the antiviral activity and possible mechanism of host factors might provide the theoretical basis for the prevention and control of IBD. In this study, RNA-sequencing results indicated that many host factors were induced by IBDV infection, among which the expression levels of OASL (2´,5´-oligadenylate synthetase-like protein) was significantly upregulated. OASL overexpression significantly inhibited IBDV replication, whereas OASL knockdown promoted IBDV replication. Interestingly, the antiviral ability of OASL was independent of its canonical enzymatic activity, i.e., OASL targeted viral protein VP2 for degradation, depending on the autophagy receptor p62/SQSTM1 in the autophagy pathway. Additionally, the 316 lysine (K) of VP2 was the key site for autophagy degradation, and its replacement with arginine disrupted VP2 degradation induced by OASL and enhanced IBDV replication. Importantly, our results for the first time indicate a unique and potent defense mechanism of OASL against double-stranded RNA virus by interaction with viral proteins, which leads to their degradation. IMPORTANCE: OASL (2´,5´-oligadenylate synthetase-like protein) exhibits broad-spectrum antiviral effects against single-stranded RNA viruses in mammals, potentially serving as a promising target for novel antiviral strategies. However, its role in inhibiting the replication of double-stranded RNA viruses (dsRNA viruses), such as infectious bursal disease virus (IBDV), in avian species remains unclear. Our findings indicated a unique and potent defense mechanism of OASL against dsRNA viruses. It has been previously shown in mammals that OASL inhibits virus replication through increasing interferon production. The groundbreaking aspect of our study is the finding that OASL has the ability to interact with IBDV viral protein VP2 and target it for degradation and thus exerts its antiviral effect. Our results reveal the interaction between avian natural antiviral immune response and IBDV infection. Our study not only enhances our understanding of bird defenses against viral infections but can also inform strategies for poultry disease management.


Subject(s)
2',5'-Oligoadenylate Synthetase , Autophagy , Birnaviridae Infections , Chickens , Infectious bursal disease virus , Viral Structural Proteins , Virus Replication , Infectious bursal disease virus/physiology , Animals , Birnaviridae Infections/virology , Birnaviridae Infections/metabolism , Viral Structural Proteins/metabolism , Viral Structural Proteins/genetics , 2',5'-Oligoadenylate Synthetase/metabolism , 2',5'-Oligoadenylate Synthetase/genetics , Poultry Diseases/virology , Poultry Diseases/metabolism , Host-Pathogen Interactions , HEK293 Cells , Humans , Cell Line
7.
J Virol ; 98(3): e0151223, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38415626

ABSTRACT

H9N2 avian influenza is a low-pathogenic avian influenza circulating in poultry and wild birds worldwide and frequently contributes to chicken salpingitis that is caused by avian pathogenic Escherichia coli (APEC), leading to huge economic losses and risks for food safety. Currently, how the H9N2 virus contributes to APEC infection and facilitates salpingitis remains elusive. In this study, in vitro chicken oviduct epithelial cell (COEC) model and in vivo studies were performed to investigate the role of H9N2 viruses on secondary APEC infection, and we identified that H9N2 virus enhances APEC infection both in vitro and in vivo. To understand the mechanisms behind this phenomenon, adhesive molecules on the cell surface facilitating APEC adhesion were checked, and we found that H9N2 virus could upregulate the expression of fibronectin, which promotes APEC adhesion onto COECs. We further investigated how fibronectin expression is regulated by H9N2 virus infection and revealed that transforming growth factor beta (TGF-ß) signaling pathway is activated by the NS1 protein of the virus, thus regulating the expression of adhesive molecules. These new findings revealed the role of H9N2 virus in salpingitis co-infected with APEC and discovered the molecular mechanisms by which the H9N2 virus facilitates APEC infection, offering new insights to the etiology of salpingitis with viral-bacterial co-infections.IMPORTANCEH9N2 avian influenza virus (AIV) widely infects poultry and is sporadically reported in human infections. The infection in birds frequently causes secondary bacterial infections, resulting in severe symptoms like pneumonia and salpingitis. Currently, the mechanism that influenza A virus contributes to secondary bacterial infection remains elusive. Here we discovered that H9N2 virus infection promotes APEC infection and further explored the underlying molecular mechanisms. We found that fibronectin protein on the cell surface is vital for APEC adhesion and also showed that H9N2 viral protein NS1 increased the expression of fibronectin by activating the TGF-ß signaling pathway. Our findings offer new information on how AIV infection promotes APEC secondary infection, providing potential targets for mitigating severe APEC infections induced by H9N2 avian influenza, and also give new insights on the mechanisms on how viruses promote secondary bacterial infections in animal and human diseases.


Subject(s)
Escherichia coli Infections , Influenza A Virus, H9N2 Subtype , Influenza in Birds , Poultry Diseases , Salpingitis , Animals , Female , Humans , Chickens , Escherichia coli , Fibronectins/metabolism , Influenza A Virus, H9N2 Subtype/physiology , Influenza in Birds/complications , Oviducts/metabolism , Poultry , Poultry Diseases/metabolism , Poultry Diseases/virology , Salpingitis/metabolism , Salpingitis/veterinary , Salpingitis/virology , Transforming Growth Factor beta/metabolism , Viral Proteins/metabolism , Escherichia coli Infections/complications , Escherichia coli Infections/veterinary
8.
J Virol ; 98(7): e0083024, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38940559

ABSTRACT

Viruses have evolved a range of strategies to utilize or manipulate the host's cellular translational machinery for efficient infection, although the mechanisms by which infectious bronchitis virus (IBV) manipulates the host translation machinery remain unclear. In this study, we firstly demonstrate that IBV infection causes host shutoff, although viral protein synthesis is not affected. We then screened 23 viral proteins, and identified that more than one viral protein is responsible for IBV-induced host shutoff, the inhibitory effects of proteins Nsp15 were particularly pronounced. Ribosome profiling was used to draw the landscape of viral mRNA and cellular genes expression model, and the results showed that IBV mRNAs gradually dominated the cellular mRNA pool, the translation efficiency of the viral mRNAs was lower than the median efficiency (about 1) of cellular mRNAs. In the analysis of viral transcription and translation, higher densities of RNA sequencing (RNA-seq) and ribosome profiling (Ribo-seq) reads were observed for structural proteins and 5' untranslated regions, which conformed to the typical transcriptional characteristics of nested viruses. Translational halt events and the number of host genes increased significantly after viral infection. The translationally paused genes were enriched in translation, unfolded-protein-related response, and activation of immune response pathways. Immune- and inflammation-related mRNAs were inefficiently translated in infected cells, and IBV infection delayed the production of IFN-ß and IFN-λ. Our results describe the translational landscape of IBV-infected cells and demonstrate new strategies by which IBV induces host gene shutoff to promote its replication. IMPORTANCE: Infectious bronchitis virus (IBV) is a γ-coronavirus that causes huge economic losses to the poultry industry. Understanding how the virus manipulates cellular biological processes to facilitate its replication is critical for controlling viral infections. Here, we used Ribo-seq to determine how IBV infection remodels the host's biological processes and identified multiple viral proteins involved in host gene shutoff. Immune- and inflammation-related mRNAs were inefficiently translated, the translation halt of unfolded proteins and immune activation-related genes increased significantly, benefitting IBV replication. These data provide new insights into how IBV modulates its host's antiviral responses.


Subject(s)
Chickens , Coronavirus Infections , Host-Pathogen Interactions , Infectious bronchitis virus , Protein Biosynthesis , Ribosomes , Virus Replication , Infectious bronchitis virus/physiology , Infectious bronchitis virus/genetics , Animals , Ribosomes/metabolism , Coronavirus Infections/virology , Coronavirus Infections/immunology , Coronavirus Infections/metabolism , Host-Pathogen Interactions/genetics , Chickens/virology , RNA, Viral/genetics , RNA, Viral/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Viral Proteins/metabolism , Viral Proteins/genetics , Poultry Diseases/virology , Poultry Diseases/immunology , Poultry Diseases/genetics , Cell Line , Humans
9.
Emerg Infect Dis ; 30(6): 1223-1227, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38703023

ABSTRACT

Highly pathogenic avian influenza H5N6 and H5N1 viruses of clade 2.3.4.4b were simultaneously introduced into South Korea at the end of 2023. An outbreak at a broiler duck farm consisted of concurrent infection by both viruses. Sharing genetic information and international surveillance of such viruses in wild birds and poultry is critical.


Subject(s)
Disease Outbreaks , Influenza A Virus, H5N1 Subtype , Influenza in Birds , Phylogeny , Influenza in Birds/virology , Influenza in Birds/epidemiology , Republic of Korea/epidemiology , Animals , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/pathogenicity , Ducks/virology , Influenza A virus/genetics , Influenza A virus/classification , Coinfection/virology , Coinfection/epidemiology , History, 21st Century , Poultry Diseases/virology , Poultry Diseases/epidemiology
10.
J Gen Virol ; 105(7)2024 Jul.
Article in English | MEDLINE | ID: mdl-38980150

ABSTRACT

Between 2013 and 2017, the A/Anhui/1/13-lineage (H7N9) low-pathogenicity avian influenza virus (LPAIV) was epizootic in chickens in China, causing mild disease, with 616 fatal human cases. Despite poultry vaccination, H7N9 has not been eradicated. Previously, we demonstrated increased pathogenesis in turkeys infected with H7N9, correlating with the emergence of the L217Q (L226Q H3 numbering) polymorphism in the haemagglutinin (HA) protein. A Q217-containing virus also arose and is now dominant in China following vaccination. We compared infection and transmission of this Q217-containing 'turkey-adapted' (ty-ad) isolate alongside the H7N9 (L217) wild-type (wt) virus in different poultry species and investigated the zoonotic potential in the ferret model. Both wt and ty-ad viruses demonstrated similar shedding and transmission in turkeys and chickens. However, the ty-ad virus was significantly more pathogenic than the wt virus in turkeys but not in chickens, causing 100 and 33% mortality in turkeys respectively. Expanded tissue tropism was seen for the ty-ad virus in turkeys but not in chickens, yet the viral cell receptor distribution was broadly similar in the visceral organs of both species. The ty-ad virus required exogenous trypsin for in vitro replication yet had increased replication in primary avian cells. Replication was comparable in mammalian cells, and the ty-ad virus replicated successfully in ferrets. The L217Q polymorphism also affected antigenicity. Therefore, H7N9 infection in turkeys can generate novel variants with increased risk through altered pathogenicity and potential HA antigenic escape. These findings emphasize the requirement for enhanced surveillance and understanding of A/Anhui/1/13-lineage viruses and their risk to different species.


Subject(s)
Chickens , Ferrets , Influenza A Virus, H7N9 Subtype , Influenza in Birds , Turkeys , Animals , Turkeys/virology , Influenza in Birds/virology , Influenza in Birds/transmission , Influenza A Virus, H7N9 Subtype/genetics , Influenza A Virus, H7N9 Subtype/pathogenicity , Chickens/virology , Virulence , China/epidemiology , Poultry Diseases/virology , Poultry Diseases/transmission , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Virus Shedding , Virus Replication , Zoonoses/virology , Influenza, Human/virology , Influenza, Human/transmission
11.
Proc Biol Sci ; 291(2031): 20241713, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39317317

ABSTRACT

High pathogenicity avian influenza virus (HPAIV) is a rapidly evolving virus causing significant economic and environmental harm. Wild birds are a key viral reservoir and an important source of viral incursions into animal populations, including poultry. However, we lack a thorough understanding of which species drive incursions and whether this changes over time. We explored associations between the abundances of 152 avian species and outbreaks of highly pathogenic avian influenza (HPAI) in poultry premises across Great Britain between October 2021 and January 2023. Spatial generalized additive models were used, with species abundance distributions sourced from eBird. Associations were investigated at the species-specific level and across species aggregations. During autumn/winter, associations were generally strongest with waterbirds such as ducks and geese; however, we also found significant associations in groups such as non-native gamebirds and rapid change in species-specific associations over time. Our results demonstrate the value of citizen science to rapidly explore wild species as potential facilitators of disease incursions into well-monitored populations, especially in regions where viral surveillance in wild species is limited. This can be a critical step towards prioritizing targeted surveillance that could inform species-specific biosecurity measures; particularly for HPAIV, which has undergone sudden shifts in host range and continues to rapidly evolve.


Subject(s)
Animals, Wild , Birds , Citizen Science , Disease Outbreaks , Influenza in Birds , Poultry , Animals , Influenza in Birds/epidemiology , Influenza in Birds/virology , Birds/virology , Poultry/virology , Disease Outbreaks/veterinary , United Kingdom/epidemiology , Poultry Diseases/epidemiology , Poultry Diseases/virology , Ducks/virology , Seasons
12.
J Virol ; 97(6): e0028423, 2023 06 29.
Article in English | MEDLINE | ID: mdl-37255472

ABSTRACT

Hepatitis-pericardial syndrome (HHS) is an acute highly infectious avian disease caused by fowl adenovirus serotype 4 (FAdV-4), characterized by fulminant hepatitis and hydropericardium in broilers. Since 2015, a widespread epidemic has occurred in China due to the emergence of hypervirulent FAdV-4 (HPFAdV-4), causing huge losses to the stakeholders. However, the pathogenesis of HPFAdV-4 and the host responses to its infection remain elusive. Here, we show that infection of leghorn male hepatocellular (LMH) cells by HPFAdV-4 induced complete autophagy in cells and that the autophagy induced by recombinant HPFAdV-4-ON1 (rHPFAdV-4-ON1), a viral strain generated by replacing the hexon gene of wild-type HPFAdV-4 (HPFAdV-4-WT) with the one of nonpathogenic strain FAdV-4-ON1, was remarkably mitigated compared to that of the rHPFAdV-4-WT control, suggesting that HPFAdV-4 hexon is responsible for virus-induced autophagy. Importantly, we found that hexon interacted with a cellular protein, BAG3, a host protein that initiates autophagy, and that BAG3 expression increased in cells infected with HPFAdV-4. Furthermore, knockdown of BAG3 by RNA interference (RNAi) significantly inhibited HPFAdV-4- or hexon-induced autophagy and suppressed viral replication. On the contrary, expression of hexon markedly upregulated the expression of BAG3 via activating the P38 signaling pathway, triggering autophagy. Thus, these findings reveal that HPFAdV-4 hexon interacts with the host protein BAG3 and promotes BAG3 expression by activating P38 signaling pathway, thereby inducing autophagy and enhancing viral proliferation, which immensely furthers our understanding of the pathogenesis of HPFAdV-4 infection. IMPORTANCE HHS, mainly caused by HPFAdV-4, has caused large economic losses to the stakeholders in recent years. Infection of leghorn male hepatocellular (LMH) cells by HPFAdV-4 induced complete autophagy that is essential for HPFAdV-4 replication. By a screening strategy, the viral protein hexon was found responsible for virus-induced autophagy in cells. Importantly, hexon was identified as a factor promoting viral replication by interaction with BAG3, an initiator of host cell autophagy. These findings will help us to better understand the host response to HPFAdV-4 infection, providing a novel insight into the pathogenesis of HPFAdV-4 infection.


Subject(s)
Adenoviridae Infections , Autophagy , Capsid Proteins , Poultry Diseases , Virus Replication , Animals , Male , Adenoviridae/genetics , Adenoviridae/physiology , Adenoviridae Infections/veterinary , Chickens , Poultry Diseases/virology , Serogroup
13.
J Virol ; 97(5): e0032423, 2023 05 31.
Article in English | MEDLINE | ID: mdl-37042750

ABSTRACT

In ovo vaccination is an attractive immunization approach for chickens. However, most live Newcastle disease virus (NDV) vaccine strains used safely after hatching are unsafe as in ovo vaccines due to their high pathogenicity for chicken embryos. The mechanism for viral pathogenicity in chicken embryos is poorly understood. Our previous studies reported that NDV strain TS09-C was a safe in ovo vaccine, and the F protein cleavage site (FCS) containing three basic amino acids (3B-FCS) was the crucial determinant of the attenuation of TS09-C in chicken embryos. Here, five trypsin-like proteases that activated NDV in chicken embryos were identified. The F protein with 3B-FCS was sensitive to the proteases Tmprss4, Tmprss9, and F7, was present in fewer tissue cells of chicken embryos, which limited the viral tropism, and was responsible for the attenuation of NDV with 3B-FCS, while the F protein with FCS containing two basic amino acids could be cleaved not only by Tmprss4, Tmprss9, and F7 but also by Prss23 and Cfd, was present in most tissue cells, and thereby was responsible for broad tissue tropism and high pathogenicity of virus in chicken embryos. Furthermore, when mixed with the protease inhibitors aprotinin and camostat, NDV with 2B-FCS exhibited greatly weakened pathogenicity in chicken embryos. Thus, our results extend the understanding of the molecular mechanism of NDV pathogenicity in chicken embryos and provide a novel molecular target for the rational design of in ovo vaccines, ensuring uniform and effective vaccine delivery and earlier induction of immune protection by the time of hatching. IMPORTANCE As an attractive immunization approach for chickens, in ovo vaccination can induce a considerable degree of protection by the time of hatching, provide support in closing the window in which birds are susceptible to infection, facilitate fast and uniform vaccine delivery, and reduce labor costs by the use of mechanized injectors. The commercial live Newcastle disease virus (NDV) vaccine strains are not safe for in ovo vaccination and cause the death of chicken embryos. The mechanism for viral pathogenicity in chicken embryos is poorly understood. In the present study, we identified five trypsin-like proteases that activate NDV in chicken embryos and elucidated their roles in the tissue tropism and pathogenicity of NDV used as in ovo vaccine. Finally, we revealed the molecular basis for the pathogenicity of NDV in chicken embryos and provided a novel strategy for the rational design of in ovo ND vaccines.


Subject(s)
Newcastle Disease , Peptide Hydrolases , Poultry Diseases , Viral Vaccines , Animals , Chick Embryo , Antibodies, Viral , Chickens , Newcastle Disease/immunology , Newcastle Disease/virology , Newcastle disease virus/physiology , Peptide Hydrolases/metabolism , Poultry Diseases/immunology , Poultry Diseases/virology , Vaccines, Attenuated , Viral Vaccines/administration & dosage , Virulence
14.
J Virol ; 97(10): e0071623, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37737586

ABSTRACT

IMPORTANCE: Marek's disease virus (MDV) is a ubiquitous chicken pathogen that inflicts a large economic burden on the poultry industry, despite worldwide vaccination programs. MDV is only partially controlled by available vaccines, and the virus retains the ability to replicate and spread between vaccinated birds. Following an initial infection, MDV enters a latent state and integrates into host telomeres and this may be a prerequisite for malignant transformation, which is usually fatal. To understand the mechanism that underlies the dynamic relationship between integrated-latent and reactivated MDV, we have characterized integrated MDV (iMDV) genomes and their associated telomeres. This revealed a single orientation among iMDV genomes and the loss of some terminal sequences that is consistent with integration by homology-directed recombination and excision via a telomere-loop-mediated process.


Subject(s)
Chickens , Genome, Viral , Herpesvirus 2, Gallid , Homologous Recombination , Marek Disease , Telomere , Virus Integration , Animals , Chickens/virology , Genome, Viral/genetics , Herpesvirus 2, Gallid/genetics , Marek Disease/genetics , Marek Disease/virology , Poultry Diseases/genetics , Poultry Diseases/virology , Telomere/genetics , Viral Vaccines/immunology , Virus Activation , Virus Latency , Virus Integration/genetics
15.
J Virol ; 97(4): e0140622, 2023 04 27.
Article in English | MEDLINE | ID: mdl-37022163

ABSTRACT

The genomes of numerous herpesviruses have been cloned as infectious bacterial artificial chromosomes. However, attempts to clone the complete genome of infectious laryngotracheitis virus (ILTV), formally known as Gallid alphaherpesvirus-1, have been met with limited success. In this study, we report the development of a cosmid/yeast centromeric plasmid (YCp) genetic system to reconstitute ILTV. Overlapping cosmid clones were generated that encompassed 90% of the 151-Kb ILTV genome. Viable virus was produced by cotransfecting leghorn male hepatoma (LMH) cells with these cosmids and a YCp recombinant containing the missing genomic sequences - spanning the TRS/UL junction. An expression cassette for green fluorescent protein (GFP) was inserted within the redundant inverted packaging site (ipac2), and the cosmid/YCp-based system was used to generate recombinant replication-competent ILTV. Viable virus was also reconstituted with a YCp clone containing a BamHI linker within the deleted ipac2 site, further demonstrating the nonessential nature of this site. Recombinants deleted in the ipac2 site formed plaques undistinguished from those viruses containing intact ipac2. The 3 reconstituted viruses replicated in chicken kidney cells with growth kinetics and titers similar to the USDA ILTV reference strain. Specific pathogen-free chickens inoculated with the reconstituted ILTV recombinants succumbed to levels of clinical disease similar to that observed in birds inoculated with wildtype viruses, demonstrating the reconstituted viruses were virulent. IMPORTANCE Infectious laryngotracheitis virus (ILTV) is an important pathogen of chicken with morbidity of 100% and mortality rates as high as 70%. Factoring in decreased production, mortality, vaccination, and medication, a single outbreak can cost producers over a million dollars. Current attenuated and vectored vaccines lack safety and efficacy, leaving a need for better vaccines. In addition, the lack of an infectious clone has also impeded understanding viral gene function. Since infectious bacterial artificial chromosome (BAC) clones of ILTV with intact replication origins are not feasible, we reconstituted ILTV from a collection of yeast centromeric plasmids and bacterial cosmids, and identified a nonessential insertion site within a redundant packaging site. These constructs and the methodology necessary to manipulate them will facilitate the development of improved live virus vaccines by modifying genes encoding virulence factors and establishing ILTV-based viral vectors for expressing immunogens of other avian pathogens.


Subject(s)
Cosmids , Herpesvirus 1, Gallid , Mutagenesis , Plasmids , Animals , Male , Chickens , Cosmids/genetics , Herpesviridae Infections/virology , Herpesvirus 1, Gallid/genetics , Herpesvirus 1, Gallid/pathogenicity , Plasmids/genetics , Poultry Diseases/virology , Saccharomyces cerevisiae/genetics , Cell Line , Genome, Viral/genetics , Viral Proteins/genetics , Viral Proteins/metabolism
16.
J Virol ; 97(3): e0003823, 2023 03 30.
Article in English | MEDLINE | ID: mdl-36779761

ABSTRACT

Coronaviruses infect a wide variety of host species, resulting in a range of diseases in both humans and animals. The coronavirus genome consists of a large positive-sense single-stranded molecule of RNA containing many RNA structures. One structure, denoted s2m and consisting of 41 nucleotides, is located within the 3' untranslated region (3' UTR) and is shared between some coronavirus species, including infectious bronchitis virus (IBV), severe acute respiratory syndrome coronavirus (SARS-CoV), and SARS-CoV-2, as well as other pathogens, including human astrovirus. Using a reverse genetic system to generate recombinant viruses, we investigated the requirement of the s2m structure in the replication of IBV, a globally distributed economically important Gammacoronavirus that infects poultry causing respiratory disease. Deletion of three nucleotides predicted to destabilize the canonical structure of the s2m or the deletion of the nucleotides corresponding to s2m impacted viral replication in vitro. In vitro passaging of the recombinant IBV with the s2m sequence deleted resulted in a 36-nucleotide insertion in place of the deletion, which was identified to be composed of a duplication of flanking sequences. A similar result was observed following serial passage of human astrovirus with a deleted s2m sequence. RNA modeling indicated that deletion of the nucleotides corresponding to the s2m impacted other RNA structures present in the IBV 3' UTR. Our results indicated for both IBV and human astrovirus a preference for nucleotide occupation in the genome location corresponding to the s2m, which is independent of the specific s2m sequence. IMPORTANCE Coronaviruses infect many species, including humans and animals, with substantial effects on livestock, particularly with respect to poultry. The coronavirus RNA genome consists of structural elements involved in viral replication whose roles are poorly understood. We investigated the requirement of the RNA structural element s2m in the replication of the Gammacoronavirus infectious bronchitis virus, an economically important viral pathogen of poultry. Using reverse genetics to generate recombinant IBVs with either a disrupted or deleted s2m, we showed that the s2m is not required for viral replication in cell culture; however, replication is decreased in tracheal tissue, suggesting a role for the s2m in the natural host. Passaging of these viruses as well as human astrovirus lacking the s2m sequence demonstrated a preference for nucleotide occupation, independent of the s2m sequence. RNA modeling suggested deletion of the s2m may negatively impact other essential RNA structures.


Subject(s)
Infectious bronchitis virus , Mamastrovirus , Mutagenesis, Insertional , Animals , Humans , 3' Untranslated Regions/genetics , Chickens/virology , Infectious bronchitis virus/genetics , Mamastrovirus/genetics , Mutagenesis, Insertional/genetics , Poultry Diseases/virology , RNA, Viral/genetics , Virus Replication/genetics , RNA Stability/genetics , Sequence Deletion/genetics
17.
J Virol ; 97(11): e0132223, 2023 Nov 30.
Article in English | MEDLINE | ID: mdl-37882519

ABSTRACT

IMPORTANCE: Chickens immunized with the infectious laryngotracheitis chicken embryo origin (CEO) vaccine (Medivac, PT Medion Farma Jaya) experience adverse reactions, hindering its safety and effective use in poultry flocks. To improve the effect of the vaccine, we sought to find a strategy to alleviate the respiratory reactions associated with the vaccine. Here, we confirmed that co-administering the CEO vaccine with chIL-2 by oral delivery led to significant alleviation of the vaccine reactions in chickens after immunization. Furthermore, we found that the co-administration of chIL-2 with the CEO vaccine reduced the clinical signs of the CEO vaccine while enhancing natural killer cells and cytotoxic T lymphocyte response to decrease viral loads in their tissues, particularly in the trachea and conjunctiva. Importantly, we demonstrated that the chIL-2 treatment can ameliorate the replication of the CEO vaccine without compromising its effectiveness. This study provides new insights into further applications of chIL-2 and a promising strategy for alleviating the adverse reaction of vaccines.


Subject(s)
Chickens , Herpesviridae Infections , Herpesvirus 1, Gallid , Interleukin-2 , Killer Cells, Natural , T-Lymphocytes, Cytotoxic , Viral Vaccines , Animals , Administration, Oral , Chickens/immunology , Chickens/virology , Conjunctiva/virology , Herpesviridae Infections/immunology , Herpesviridae Infections/prevention & control , Herpesviridae Infections/veterinary , Herpesviridae Infections/virology , Herpesvirus 1, Gallid/immunology , Interleukin-2/administration & dosage , Interleukin-2/immunology , Killer Cells, Natural/immunology , Poultry Diseases/immunology , Poultry Diseases/prevention & control , Poultry Diseases/virology , Respiratory Tract Diseases/immunology , Respiratory Tract Diseases/prevention & control , Respiratory Tract Diseases/veterinary , Respiratory Tract Diseases/virology , T-Lymphocytes, Cytotoxic/immunology , Trachea/virology , Viral Load , Viral Vaccines/administration & dosage , Viral Vaccines/adverse effects , Viral Vaccines/biosynthesis , Viral Vaccines/immunology
18.
BMC Microbiol ; 24(1): 338, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39261757

ABSTRACT

Currently, phage biocontrol is increasingly used as a green and natural technology for treating Salmonella and other infections, but phages exhibit instability and activity loss during storage. Therefore, in this study, the effects of lyophilization on the activity and stability of phage cocktails for the control of multidrug-resistant Salmonella in broiler chickens were determined. Eight serotypes of Salmonella were isolated and identified from broiler chicken farms, and bacteriophages against multidrug-resistant Salmonella enterica subsp. enterica serovar Kentucky, Salmonella enterica subsp. enterica serovar Typhimrium and Salmonella enterica subsp. enterica serovar Enteritidis were isolated. The bacteriophage cocktail was prepared and lyophilized, and it was subjected to in vitro and in vivo examinations. A reconstituted lyophilized bacteriophage cocktail was used for the oral treatment of chicks before and after challenge with multidrug-resistant S. Kentucky. The colonization of cecum by S. Kentucky was detected by using real-time PCR, and the serum levels of IgM, IgA and IL-4 and pathological changes in the different groups were detected. Three Caudovirales phages families were identified including Autographiviridae, Straboviridae and Drexlerviridae against multidrug-resistant S. Kentucky, S. Typhimrium and S. Enteritidis. The groups treated with the bacteriophage cocktail showed no clinical signs, no postmortem lesions, and a mortality rate of 0%, which improved the growth performance parameters. Additionally, the estimated serum levels of IgM, IgA and IL-4 were significantly greater in the bacteriophage cocktail-treated groups. Lyophilization effectively preserves the long-term storage stability of phages. Therefore, lyophilized bacteriophage cocktail therapy is a valuable approach for controlling multidrug-resistant Salmonella infections in broiler chickens.


Subject(s)
Chickens , Drug Resistance, Multiple, Bacterial , Freeze Drying , Poultry Diseases , Salmonella Infections, Animal , Salmonella Phages , Salmonella , Animals , Chickens/microbiology , Freeze Drying/methods , Poultry Diseases/microbiology , Poultry Diseases/therapy , Poultry Diseases/virology , Poultry Diseases/prevention & control , Salmonella Infections, Animal/microbiology , Salmonella Infections, Animal/therapy , Salmonella/virology , Salmonella Phages/physiology , Cecum/microbiology , Cecum/virology , Phage Therapy/methods , Bacteriophages/genetics , Bacteriophages/physiology , Bacteriophages/isolation & purification
19.
Microb Pathog ; 194: 106829, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39084310

ABSTRACT

Goose astroviruses (GAstVs) are important pathogens which can cause gout in goslings leading to huge economic losses for the goose farming industry in China. In 2023, an infectious disease characterized by visceral gout broke out in commercial goose farms in Guangxi and Guangdong provinces of China. In this study, two GAstV strains of GXNN and GDCS were successfully isolated from these two disease-ridden goose farms. The complete genomic lengths of these two strains were 7166 bp, and phylogenetic analysis showed that they were both GAstV-2 subtypes. The 3-dimensional structures of the capsid protein were predicted and six characteristic mutation sites at amino acid positions 60, 61, 228, 229, 456 and 523 were found within the strong antigenic regions. A recombination event occurred at 6833-7070 nt between the GAstV TZ03 and Turkey astrovirus CA/00 and this was detected in both the GXNN and GDCS strains. Another recombinant event occurred at 63-2747 nt between the GAstV XT1 and GAstV SDPY and this was detected in the GDCS strain. When 1-day-old goslings were infected with the novel GXNN and GDCS strains, they showed severe visceral gout. This was accompanied by enlarged spleens, liver hemorrhages and urate deposits in the kidneys and ureters and their blood urea nitrogen levels were significantly elevated. The mortality rates of the GXNN- and GDCS-infected groups were pathogenically high at 80 % and 60 %, respectively. These results will promote our understanding of the evolution and epidemic potential of GAstVs in China.


Subject(s)
Astroviridae Infections , Capsid Proteins , Geese , Genome, Viral , Gout , Phylogeny , Poultry Diseases , Animals , Geese/virology , China , Astroviridae Infections/veterinary , Astroviridae Infections/virology , Poultry Diseases/virology , Poultry Diseases/pathology , Gout/virology , Gout/veterinary , Gout/pathology , Capsid Proteins/genetics , Avastrovirus/genetics , Avastrovirus/pathogenicity , Avastrovirus/isolation & purification , Avastrovirus/classification , Virulence , Astroviridae/genetics , Astroviridae/isolation & purification , Astroviridae/pathogenicity
20.
Microb Pathog ; 190: 106638, 2024 May.
Article in English | MEDLINE | ID: mdl-38574829

ABSTRACT

Autophagy plays an important role in the lifecycle of viruses. However, there is currently a lack of systematic research on the relationship between Infectious Bronchitis Virus (IBV) and autophagy. This study aims to investigate the impact of IBV on autophagy and the role of autophagy in viral replication. We observed that IBV infection increased the expression of microtubule-associated protein 1 light chain 3, a marker of autophagy, decreased the expression of sequestosome 1, and led to elevated intracellular LC3 puncta levels. These findings suggest that IBV infection activates the autophagic process in cells. To investigate the impact of autophagy on the replication of IBV, we utilized rapamycin as an autophagy activator and 3-methyladenine as an autophagy inhibitor. Our results indicate that IBV promotes viral replication by inducing autophagy. Further investigation revealed that IBV induces autophagosome formation by inhibiting the mTOR-ULK1 pathway and activating the activity of vacuolar protein sorting 34 (VPS34), autophagy-related gene 14, and the Beclin-1 complex. VPS34 plays a crucial role in this process, as inhibiting VPS34 protein activity enhances cell proliferation after IBV infection. Additionally, inhibiting VPS34 significantly improves the survival rate of IBV-infected chicks, suppresses IBV replication in the kidney, and alleviates tracheal, lung, and kidney damage caused by IBV infection. In summary, IBV infection can induce autophagy by modulating the mTOR/ULK1 signaling pathway and activating the VPS34 complex, while autophagy serves to promote virus replication.


Subject(s)
Autophagy , Chickens , Class III Phosphatidylinositol 3-Kinases , Infectious bronchitis virus , Virus Replication , Infectious bronchitis virus/physiology , Animals , Class III Phosphatidylinositol 3-Kinases/metabolism , Chickens/virology , Coronavirus Infections/virology , Coronavirus Infections/metabolism , Sirolimus/pharmacology , Beclin-1/metabolism , Beclin-1/genetics , TOR Serine-Threonine Kinases/metabolism , Signal Transduction , Cell Line , Poultry Diseases/virology , Autophagosomes/metabolism , Autophagosomes/virology , Chlorocebus aethiops , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL