Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 125
Filtrar
1.
Microbiol Spectr ; : e0030924, 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38888361

RESUMO

The tumor suppressor p53, primarily functioning as a transcription factor, has exhibited antiviral capabilities against various viruses in chickens, including infectious bursal disease virus (IBDV), avian leukosis virus subgroup J (ALV-J), and avian infectious laryngotracheitis virus (ILTV). Nevertheless, the existence of a universal antiviral mechanism employed by chicken p53 (chp53) against these viruses remains uncertain. This study conducted a comprehensive comparison of molecular networks involved in chp53's antiviral function against IBDV, ALV-J, and ILTV. This was achieved through an integrated analysis of ChIP-seq data, examining chp53's genome-wide chromatin occupancy, and RNA-seq data from chicken cells infected with these viruses. The consistent observation of chp53 target gene enrichment in metabolic pathways, confirmed via ChIP-qPCR, suggests a ubiquitous regulation of host cellular metabolism by chp53 across different viruses. Further genome binding motif conservation analysis and transcriptional co-factor prediction suggest conserved transcriptional regulation mechanism by which chp53 regulates host cellular metabolism during viral infection. These findings offer novel insights into the antiviral role of chp53 and propose that targeting the virus-host metabolic interaction through regulating p53 could serve as a universal strategy for antiviral therapies in chickens.IMPORTANCEThe current study conducted a comprehensive analysis, comparing molecular networks underlying chp53's antiviral role against infectious bursal disease virus (IBDV), avian leukosis virus subgroup J (ALV-J), and avian infectious laryngotracheitis virus (ILTV). This was achieved through a combined assessment of ChIP-seq and RNA-seq data obtained from infected chicken cells. Notably, enrichment of chp53 target genes in metabolic pathways was consistently observed across viral infections, indicating a universal role of chp53 in regulating cellular metabolism during diverse viral infections. These findings offer novel insights into the antiviral capabilities of chicken p53, laying a foundation for the potential development of broad-spectrum antiviral therapies in chickens.

2.
Virology ; 597: 110159, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38943781

RESUMO

Therapies targeting virus-host interactions are seen as promising strategies for treating gallid alphaherpesvirus 1 (ILTV) infection. Our study revealed a biphasic activation of two MAPK cascade pathways, MEK/ERK and p38 MAPK, as a notably activated host molecular event in response to ILTV infection. It exhibits antiviral functions at different stages of infection. Initially, the MEK/ERK pathway is activated upon viral invasion, leading to a broad suppression of metabolic pathways crucial for ILTV replication, thereby inhibiting viral replication from the early stage of ILTV infection. As the viral replication progresses, the p38 MAPK pathway activates its downstream transcription factor, STAT1, further hindering viral replication. Interestingly, ILTV overcomes this biphasic antiviral barrier by hijacking host p38-AKT axis, which protects infected cells from the apoptosis induced by infection and establishes an intracellular equilibrium conducive to extensive ILTV replication. These insights could provide potential therapeutic targets for ILTV infection.


Assuntos
Infecções por Herpesviridae , Sistema de Sinalização das MAP Quinases , Replicação Viral , Proteínas Quinases p38 Ativadas por Mitógeno , Animais , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Infecções por Herpesviridae/virologia , Infecções por Herpesviridae/metabolismo , Alphaherpesvirinae/fisiologia , Alphaherpesvirinae/genética , Alphaherpesvirinae/metabolismo , Interações Hospedeiro-Patógeno , Linhagem Celular , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT1/genética
3.
J Virol ; 98(3): e0191523, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38334327

RESUMO

As an intrinsic cellular mechanism responsible for the internalization of extracellular ligands and membrane components, caveolae-mediated endocytosis (CavME) is also exploited by certain pathogens for endocytic entry [e.g., Newcastle disease virus (NDV) of paramyxovirus]. However, the molecular mechanisms of NDV-induced CavME remain poorly understood. Herein, we demonstrate that sialic acid-containing gangliosides, rather than glycoproteins, were utilized by NDV as receptors to initiate the endocytic entry of NDV into HD11 cells. The binding of NDV to gangliosides induced the activation of a non-receptor tyrosine kinase, Src, leading to the phosphorylation of caveolin-1 (Cav1) and dynamin-2 (Dyn2), which contributed to the endocytic entry of NDV. Moreover, an inoculation of cells with NDV-induced actin cytoskeletal rearrangement through Src to facilitate NDV entry via endocytosis and direct fusion with the plasma membrane. Subsequently, unique members of the Rho GTPases family, RhoA and Cdc42, were activated by NDV in a Src-dependent manner. Further analyses revealed that RhoA and Cdc42 regulated the activities of specific effectors, cofilin and myosin regulatory light chain 2, responsible for actin cytoskeleton rearrangement, through diverse intracellular signaling cascades. Taken together, our results suggest that an inoculation of NDV-induced Src-mediated cellular activation by binding to ganglioside receptors. This process orchestrated NDV endocytic entry by modulating the activities of caveolae-associated Cav1 and Dyn2, as well as specific Rho GTPases and downstream effectors. IMPORTANCE: In general, it is known that the paramyxovirus gains access to host cells through direct penetration at the plasma membrane; however, emerging evidence suggests more complex entry mechanisms for paramyxoviruses. The endocytic entry of Newcastle disease virus (NDV), a representative member of the paramyxovirus family, into multiple types of cells has been recently reported. Herein, we demonstrate the binding of NDV to induce ganglioside-activated Src signaling, which is responsible for the endocytic entry of NDV through caveolae-mediated endocytosis. This process involved Src-dependent activation of the caveolae-associated Cav1 and Dyn2, as well as specific Rho GTPase and downstream effectors, thereby orchestrating the endocytic entry process of NDV. Our findings uncover a novel molecular mechanism of endocytic entry of NDV into host cells and provide novel insight into paramyxovirus mechanisms of entry.


Assuntos
Macrófagos , Doença de Newcastle , Vírus da Doença de Newcastle , Transdução de Sinais , Internalização do Vírus , Animais , Endocitose , Gangliosídeos/metabolismo , Macrófagos/metabolismo , Macrófagos/virologia , Doença de Newcastle/virologia , Vírus da Doença de Newcastle/fisiologia , Proteínas rho de Ligação ao GTP/metabolismo
4.
Viral Immunol ; 36(10): 649-658, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37903239

RESUMO

Infectious bronchitis virus (IBV), a gammacoronavirus within the Coronaviridae family, is an economically important etiological disease agent in chickens. Both early diagnosis and determination of the immune status of chickens are important for controlling IBV outbreaks in chicken flocks. The N protein is the most abundantly expressed virus-derived protein during IBV infection and can induce a strong immune response by producing antibodies during early infection or immunization. In this study, we found that the amino acid sequences of the N protein between CK/CH/LJL/04I and the other 22 IBVs were conserved, especially in the 1-160 amino acid region. Based on the sequence similarities, the three recombinant proteins, rN160 (amino acid positions 1-160), rN266 (144-409), and rN409 (1-409), were expressed using the Escherichia coli system and subsequently purified. The results demonstrated that the antigenicity and reactivity of rN160 were better than those of rN266 and rN409. As a result, an indirect enzyme-linked immunosorbent assay (ELISA) (rN160 ELISA) was developed to detect the IBV antibody based on the rN160 protein. Using 1,500 clinical field serum samples, the relative sensitivity, specificity, and accuracy of the rN160 ELISA were 98.97%, 92.34%, and 97.93%, respectively, compared to those of a commercial ELISA kit (IDEXX), indicating a strong positive correlation between the two methods. Taken together, these results reveal that the rN160 ELISA is a rapid, simple, and sensitive method for detecting group-specific IBV antibodies for epidemiological investigation and antibody-level monitoring.


Assuntos
Infecções por Coronavirus , Vírus da Bronquite Infecciosa , Doenças das Aves Domésticas , Animais , Galinhas , Anticorpos Antivirais , Ensaio de Imunoadsorção Enzimática/métodos , Aminoácidos , Infecções por Coronavirus/diagnóstico , Infecções por Coronavirus/veterinária
5.
Genes (Basel) ; 14(8)2023 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-37628666

RESUMO

Treatment options for herpesvirus infections that target the interactions between the virus and the host have been identified as promising. Our previous studies have shown that transcription factors p53 and Fos are essential host determinants of gallid alpha herpesvirus 1 (ILTV) infection. The impact of p53 and Fos on ILTV replication has 'not been fully understood yet. Using the sole ILTV-permissive chicken cell line LMH as a model, we examined the effects of hosts p53 and Fos on all phases of ILTV replication, including viral gene transcription, viral genome replication, and infectious virion generation. We achieved this by manipulating the expression of p53 and Fos in LMH cells. Our results demonstrate that the overexpression of either p53 or Fos can promote viral gene transcription at all stages of the temporal cascade of ILTV gene expression, viral genome replication, and infectious virion production, as assessed through absolute quantitative real-time PCR, ILTV-specific RT-qPCR assays, and TCID50 assays. These findings are consistent with our previous analyses of the effects of Fos and p53 knockdowns on virus production and also suggest that both p53 and Fos may be dispensable for ILTV replication. Based on the synergistic effect of regulating ILTV, we further found that there is an interaction between p53 and Fos. Interestingly, we found that p53 also has targeted sites upstream of ICP4, and these sites are very close to the Fos sites. In conclusion, our research offers an in-depth understanding of how hosts p53 and Fos affect ILTV replication. Understanding the processes by which p53 and Fos regulate ILTV infection will be improved by this knowledge, potentially paving the way for the development of novel therapeutics targeting virus-host interactions as a means of treating herpesvirus infections.


Assuntos
Bioensaio , Proteína Supressora de Tumor p53 , Animais , Proteína Supressora de Tumor p53/genética , Linhagem Celular , Galinhas , Interações entre Hospedeiro e Microrganismos
6.
Poult Sci ; 102(7): 102701, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37150176

RESUMO

Vitamin A is a fat-soluble vitamin that is a crucial mediator of the immune system. In this study, we evaluated the effect of oral vitamin A supplementation on host immune responses to infectious bronchitis virus (IBV) infection in chickens. Forty 1-day-old specific pathogen-free (SPF) chickens were fed a basal diet and randomly divided into 2 groups (n = 20 birds per group). Chickens in the experimental group were treated orally with vitamin A (dissolved in 0.1 mL soybean oil, at a dose of 8,000 IU per kg diet) daily. Birds in the control group were orally administered 0.1 mL soybean oil without vitamin A until 21 d of age. On d 21 after birth, all chickens were infected with 0.1 mL of 106.5 50% median embryo infectious dose of a pathogenic IBV strain (CK/CH/LDL/091022) by intraocular and intranasal routes. The results demonstrated that oral vitamin A supplementation did not affect the clinical course of disease and growth performance of SPF chickens. However, vitamin A supplementation increased the IBV-specific IgG serum levels and decreased the viral load in some tissues of IBV-infected chickens. In addition, the results demonstrated that vitamin A supplementation decreased the expression levels of most immune-related molecules in some tissues of IBV-infected chickens. Vitamin A supplementation decreased the mRNA expression levels of some avian ß-defensins (AvBD2, 3, 6, 7, 11, and 13) and increased the expression levels of AvBD9 and AvBD12 in some tissues of IBV-infected chickens. Similarly, vitamin A supplementation decreased the mRNA expression levels of some cytokines (interferon-γ, interleukin-1ß [IL-1ß], and IL-6) and increased the mRNA expression levels of IL-2 in some tissues of IBV-infected chickens. Furthermore, vitamin A supplementation decreased the mRNA expression levels of myeloid differentiation primary response protein 88, nuclear factor-κB p65, toll-like receptor 3, toll-like receptor 7, and CD4. In summary, the present study suggests that vitamin A supplementation enhances the immune function of SPF chickens against IBV infection by inhibiting viral replication, increasing the IBV-specific antibody titer, and suppressing the excessive inflammatory responses to IBV infection.


Assuntos
Infecções por Coronavirus , Vírus da Bronquite Infecciosa , Doenças das Aves Domésticas , Animais , Galinhas/genética , Vitamina A , Óleo de Soja , Imunidade , Suplementos Nutricionais , RNA Mensageiro , Infecções por Coronavirus/veterinária , Organismos Livres de Patógenos Específicos
7.
Viruses ; 16(1)2023 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-38275950

RESUMO

Nanoparticle-assisted polymerase chain reaction (nanoPCR) is a novel method for the rapid detection of pathogens. A sensitive and specific multiple nanoPCR assay was developed for simultaneous detection of avian leucosis virus (ALV) subgroups A, B and J. In this study, three pairs of primers were designed, based on the conserved region of the gp85 gene. An exploration of the optimal primer concentration and annealing temperature were carried out, for better performance of the nanoPCR assay. According to the results, the multiple nanoPCR assay amplified 336 pb, 625 bp and 167 bp fragments of ALV-A, -B and -J, respectively, and showed no cross-reactivity with irrelevant pathogens, suggesting the excellent specificity of the assay. The constructed standard DNA templates were used to estimate the limit of detection. As shown by the results, the detection limit of the nanoPCR assay was nearly 10 copies/µL. To further evaluate the detection ability of the assay, 186 clinical samples were detected using the nanoPCR assay, among which, 14 samples were confirmed as ALV positive; the results were further confirmed by sequencing. In conclusion, a highly specific and sensitive nanoPCR assay was successfully developed, which could be a useful tool for clinical diagnosis as well as for the discrimination of ALV-A, -B and -J.


Assuntos
Vírus da Leucose Aviária , Leucose Aviária , Nanopartículas , Animais , Vírus da Leucose Aviária/genética , Sensibilidade e Especificidade , Temperatura , Reação em Cadeia da Polimerase/métodos , Leucose Aviária/diagnóstico , Galinhas
8.
Front Microbiol ; 13: 1044141, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36504811

RESUMO

P53, a well-known tumor suppressor, has been confirmed to regulate the infection of various viruses, including chicken viruses. Our previous study observed antiviral effect of p53 inhibitor Pifithrin-α (PFT-α) on the infection of avian infectious laryngotracheitis virus (ILTV), one of the major avian viruses economically significant to the poultry industry globally. However, the potential link between this antiviral effect of PFT-α and p53 remains unclear. Using chicken LMH cell line which is permissive for ILTV infection as model, we explore the effects of p53 on ILTV replication and its underlying molecular mechanism based on genome-wide transcriptome analysis of genes with p53 binding sites. The putative p53 target genes were validated by ChIP-qPCR and RT-qPCR. Results demonstrated that, consistent with the effects of PFT-α on ILTV replication we previously reported, knockdown of p53 repressed viral gene transcription and the genome replication of ILTV effectively. The production of infectious virions was also suppressed significantly by p53 knockdown. Further bioinformatic analysis of genes with p53 binding sites revealed extensive repression of these putative p53 target genes enriched in the metabolic processes, especially nucleotide metabolism and ATP synthesis, upon p53 repression by PFT-α in ILTV infected LMH cells. Among these genes, eighteen were involved in nucleotide metabolism and ATP synthesis. Then eight of the 18 genes were selected randomly for validations, all of which were successfully identified as p53 target genes. Our findings shed light on the mechanisms through which p53 controls ILTV infection, meanwhile expand our knowledge of chicken p53 target genes.

9.
Front Endocrinol (Lausanne) ; 13: 1032268, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36568097

RESUMO

Diabetic cardiomyopathy (DCM) is a pathophysiological condition of cardiac structure and function changes in diabetic patients without coronary artery disease, hypertension, and other types of heart diseases. DCM is not uncommon in people with diabetes, which increases the risk of heart failure. However, the treatment is scarce, and the prognosis is poor. Since 1972, one clinical study after another on DCM has been conducted. However, the complex phenotype of DCM still has not been fully revealed. This dilemma hinders the pace of understanding the essence of DCM and makes it difficult to carry out penetrating clinical or basic research. This review summarizes the literature on DCM over the last 40 years and discusses the overall perspective of DCM, phase of progression, potential clinical indicators, diagnostic and screening criteria, and related randomized controlled trials to understand DCM better.


Assuntos
Diabetes Mellitus , Cardiomiopatias Diabéticas , Insuficiência Cardíaca , Humanos , Cardiomiopatias Diabéticas/diagnóstico , Coração , Insuficiência Cardíaca/terapia , Fenótipo , Prognóstico
10.
Poult Sci ; 101(11): 102164, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36167023

RESUMO

The tumor suppressor p53, which acts primarily as a transcription factor, can regulate infections from various viruses in chickens. However, the underlying mechanisms of the antiviral functions of chicken p53 (chp53) remain unclear due to the lack of detailed information on its transcriptional regulation. Here, to gain comprehensive insights into chp53 transcriptional regulatory function in a global and unbiased manner, we determined the genome-wide chromatin occupancy of chp53 by chromatin immunoprecipitation, which was followed by sequencing and chp53-mediated gene expression profile by RNA sequencing using chemically immortalized leghorn male hepatoma (LMH) cells with ectopic expression of chp53 as the model. The integrated parallel genome-wide chromatin occupancy and gene expression analysis characterized chp53 chromatin occupancy and identified 754 direct target genes of chp53. Furthermore, functional annotation and cross-species comparative biological analyses revealed the conserved key biological functions and DNA binding motifs of p53 between chickens and humans, which may be due to the consensus amino acid sequence and structure of p53 DNA-binding domains. The present study, to our knowledge, provides the first comprehensive characterization of the chp53 transcriptional regulatory network, and can possibly help to improve our understanding of p53 transcriptional regulatory mechanisms and their antiviral functions in chickens.


Assuntos
Cromatina , Proteína Supressora de Tumor p53 , Masculino , Humanos , Animais , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Galinhas/genética , Galinhas/metabolismo , Sítios de Ligação , DNA/metabolismo , Antivirais , Expressão Gênica
11.
J Virol ; 96(10): e0024122, 2022 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-35510864

RESUMO

In this study, 232 class I Newcastle disease viruses (NDVs) were identified from multiple bird species at nationwide live bird markets (LBMs) from 2017 to 2019 in China. Phylogenetic analysis indicated that all 232 isolates were clustered into genotype 1.1.2 of class I on the basis of the fusion (F) gene sequences, which were distinct from the genotypes identified in other countries. Most of the isolates (212/232) were shown to have the typical F gene molecular characteristics of class I NDVs, while a few (20/232) contained mutations at the site of the conventional start codon of the F gene, which resulted in open reading frames (ORFs) altered in length. The isolates with ACG, CTA, and ATA mutations showed different levels of increased virulence and replication capacity, suggesting that these viruses may be transitional types during the evolution of class I NDVs from avirulent to virulent. Further evaluation of biological characteristics with recombinant viruses obtained by reverse genetics demonstrated that the ATG located at genomic positions 4523 to 4525 was the authentic start codon in the F gene of class I NDV, and the specific ATA mutations which contributed to the expression of F protein on the surface of infected cells were the key determinants of increased replication capacity and virulence. Interestingly, the mutation at the corresponding site of genotype II LaSota of class II had no effects on the virulence and replication capacity in chickens. Our results suggest that the alteration of virulence and replication capacity caused by specific mutations in the F gene could be a specific characteristic of class I NDVs and indicate the possibility of the emergence of virulent NDVs due to the persistent circulation of class I NDVs. IMPORTANCE The available information on the distribution, genetic diversity, evolution, and biological characteristics of class I Newcastle disease viruses (NDVs) in domestic poultry is currently very limited. Here, identification of class I NDVs at nationwide live bird markets (LBMs) in China was performed and representative isolates were characterized. A widespread distribution of genotype 1.1.2 of class I NDVs was found in multiple bird species at LBMs in China. Though most isolates demonstrated typical molecular characteristics of class I NDVs, a few that contained specific mutations at the site of the conventional start codon of the fusion gene with increased virulence and replication capacity were identified for the first time. Our findings indicate that the virulence of class I NDVs could have evolved, and the widespread transmission and circulation of class I NDVs may represent a potential threat for disease outbreaks in poultry.


Assuntos
Doença de Newcastle , Doenças das Aves Domésticas , Animais , Galinhas/virologia , China/epidemiologia , Códon de Iniciação , Comércio , Monitoramento Epidemiológico/veterinária , Genótipo , Doença de Newcastle/epidemiologia , Vírus da Doença de Newcastle/genética , Filogenia , Aves Domésticas/virologia , Doenças das Aves Domésticas/epidemiologia , Virulência/genética
12.
Vet Microbiol ; 269: 109435, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35462119

RESUMO

Therapeutics targeting virus-host interactions have been considered promising strategies for treating herpesvirus infection. Our previous study on avian infectious laryngotracheitis virus (ILTV), an avian herpesvirus economically important to the poultry industry worldwide, identified the small molecule Pifithrin-α (PFT-α) as a potential therapeutic agent. However, the underlying mechanisms of its antiviral function remain largely unknown. Using the ILTV-permissive chicken cell line LMH as the model, we found that PFT-α effectively suppressed the transcription and genome replication of ILTV and greatly reduced the level of infectious virions. Genome-wide transcriptome analysis revealed extensive repression of the metabolic processes of infected cells by PFT-α administration. Further metabolome assays of ILTV-infected cells using liquid chromatography coupled with mass spectrometry suggest host nucleotide metabolism and ATP synthesis as the key targets of PFT-α treatment during its repression of ILTV replication, which was experimentally supported by the reduced transcription of many key enzymes essential to nucleotide metabolism and ATP synthesis. The present study provides insights into the mechanisms by which PFT-α inhibits ILTV infection, which may increase the probability of successful clinical application of this molecule.


Assuntos
Infecções por Herpesviridae , Herpesvirus Galináceo 1 , Doenças das Aves Domésticas , Trifosfato de Adenosina , Animais , Benzotiazóis , Galinhas , Infecções por Herpesviridae/veterinária , Herpesvirus Galináceo 1/genética , Nucleotídeos , Tolueno/análogos & derivados
13.
J Immunol ; 207(11): 2878-2891, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34697228

RESUMO

Ducks are an economically important waterfowl but a natural reservoir for some zoonotic pathogens, such as influenza virus and flaviviruses. Our understanding of the duck immune system and its interaction with viruses remains incomplete. In this study, we constructed the transcriptomic landscape of duck circulating immune cells, the first line of defense in the arthropod-borne transmission of arboviruses, using high-throughput single-cell transcriptome sequencing, which defined 14 populations of peripheral blood leukocytes (PBLks) based on distinct molecular signatures and revealed differences in the clustering of PBLks between ducks and humans. Taking advantage of in vivo sex differences in the susceptibility of duck PBLks to avian tembusu virus (TMUV) infection, a mosquito-borne flavivirus newly emerged from ducks with a broad host range from mosquitos to mammals, a comprehensive comparison of the in vivo dynamics of duck PBLks upon TMUV infection between sexes was performed at the single-cell level. Using this in vivo model, we discovered that TMUV infection reprogrammed duck PBLks differently between sexes, driving the expansion of granulocytes and priming granulocytes and monocytes for antiviral immune activation in males but decreasing the antiviral immune activity of granulocytes and monocytes by restricting their dynamic transitions from steady states to antiviral states with a decrease in the abundance of circulating monocytes in females. This study provides insights into the initial immune responses of ducks to arthropod-borne flaviviral infection and provides a framework for studying duck antiviral immunity.


Assuntos
Infecções por Flavivirus/imunologia , Células Mieloides/imunologia , Análise de Célula Única , Animais , Patos/virologia , Feminino , Masculino , Células Mieloides/patologia , Células Mieloides/virologia
14.
Viruses ; 13(6)2021 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-34207926

RESUMO

Gallid alpha-herpesvirus 1, also known as avian infectious laryngotracheitis virus (ILTV), continues to cause huge economic losses to the poultry industry worldwide. Similar to that of other herpesvirus-encoded proteins, the expression of viral genes encoded by ILTV is regulated by a cascade, and the underlying regulatory mechanism remains largely unclear. The viral immediate-early (IE) gene ICP4 plays a prominent role in the initiation of the transcription of early and late genes during ILTV replication. In this study, we identified AP-1 as the key regulator of the transcription of ILTV genes by bioinformatics analysis of genome-wide transcriptome data. Subsequent functional studies of the key members of the AP-1 family revealed that Fos, but not Jun, regulates ILTV infection through AP-1 since knockdown of Fos, but not Jun, by gene silencing significantly reduced ICP4 transcription and subsequent viral genome replication and virion production. Using several approaches, we identified ICP4 as a bona fide target gene of Fos that regulated Fos and has Fos response elements within its promoter. Neither the physical binding of Jun to the promoter of ICP4 nor the transcriptional activity of Jun was observed. In addition, knockdown of Fos reduced the transcription of MDH1 and ATP5A1, genes encoding two host rate-limiting enzymes essential for the production of the TCA intermediates OAA and ATP. The biological significance of the transcriptional regulation of MDH1 and ATP5A1 by Fos in ILTV infection was supported by the fact that anaplerosis of OAA and ATP rescued both ICP4 transcription and virion production in infected cells under when Fos was silenced. Our study identified the transcription factor Fos as a key regulator of ILTV infection through its transcription factor function on both the virus and host sides, improving the current understanding of both avian herpesvirus-host interactions and the roles of AP-1 in viral infection.


Assuntos
Regulação da Expressão Gênica , Infecções por Herpesviridae/veterinária , Herpesvirus Galináceo 1/fisiologia , Interações Hospedeiro-Patógeno , Doenças das Aves Domésticas/genética , Doenças das Aves Domésticas/virologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Animais , Linhagem Celular , Galinhas , Biologia Computacional , Metabolismo Energético , Perfilação da Expressão Gênica , Genes Precoces , Interações Hospedeiro-Patógeno/genética , Modelos Biológicos , Doenças das Aves Domésticas/diagnóstico , Doenças das Aves Domésticas/metabolismo , Replicação Viral
15.
Infect Genet Evol ; 93: 104980, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34182190

RESUMO

This study demonstrates that infectious bronchitis virus (IBV) strain M41, which is pathogenic for chickens, is nonpathogenic for pheasants. However, M41 replicated in the respiratory tracts of most inoculated pheasants and the virus was shed from their respiratory tracts in the early stages of infection (4 and 8 dpc). Similarly, the attenuated IBV H120 vaccine strain also replicated and the virus was shed from their respiratory tracts of most inoculated pheasants, whereas the pheasant coronavirus (PhCoV) I0623/17 replicated in the respiratory tracts of all challenged pheasants, which then shed virus for a long period of time. Strain M41 also replicated in selected tissues of the inoculated pheasants, including the lung, kidney, proventriculus, and cecal tonsil, although the viral titers were very low. Therefore, it was important to establish whether the H120 vaccine, which has a limited replication capacity in pheasants, induces a protective immune response to both "homologous" M41 and "heterologous" I0623/17 challenge. Vaccination with H120 induced humoral responses, and the replication of M41 was reduced or restricted in the tissues of the H120-vaccinated pheasants compared with its replication in unvaccinated birds. This implies that partial protection was conferred on pheasants by vaccination with the H120 vaccine. Prolonged viral replication and a large number of birds shedding virus into the respiratory tract were also observed in the unvaccinated pheasants after inoculation with M41. However, only limited protection against challenge with PhCoV I0623/17 was conferred on pheasants vaccinated with H120, largely because the replication of H120 in pheasants was limited, thus, limiting the immune responses induced by it. The low amino acid identity of the S1 subunit of the S proteins of H120 and I0623/17 might also account, at least in part, for the poor cross-protective immunity induced by H120. These results suggest that further work is required to rationally design vaccines that confer effective protection against PhCoV infection in commercial pheasant stocks.


Assuntos
Infecções por Coronavirus/veterinária , Galliformes , Vírus da Bronquite Infecciosa/fisiologia , Vacinas Virais/farmacologia , Animais , Anticorpos Neutralizantes/imunologia , Infecções por Coronavirus/prevenção & controle , Infecções por Coronavirus/virologia , Vírus da Bronquite Infecciosa/imunologia , Vírus da Bronquite Infecciosa/patogenicidade , Doenças das Aves Domésticas/prevenção & controle , Doenças das Aves Domésticas/virologia , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/metabolismo , Vacinas Atenuadas/farmacologia
16.
Vet Microbiol ; 256: 109044, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33836389

RESUMO

Avian Tembusu virus (TMUV) is a newly emerging avian pathogenic flavivirus that spreads rapidly, has an expanding host range and undergoes cross-species transmission. Our previous study identified avian monocytes/macrophages as the key targets of TMUV infection, since the infection of host monocytes/macrophages was crucial for the replication, transmission, and pathogenesis of TMUV. The polarization of host macrophages determines the functional phenotypes of macrophages; however, the effect of TMUV infection on macrophage polarization remains unclear. Here, we analysed the expression spectra of the marker genes of macrophage polarization upon TMUV infection in the HD11 chicken macrophage cell line and primary monocytes/macrophages isolated from the peripheral blood of specific pathogen-free (SPF) chickens and ducks. We found that viral replication mainly induced M1 marker genes and triggered nitric oxide (NO) release at different levels, suggesting that TMUV infection led mainly to host macrophages polarizing into the classically activated (M1) type. The NO that was increased upon infection did not function as an antiviral agent against TMUV, since the replication of TMUV in HD11 cells was not affected by the addition of an organic NO donor. Furthermore, upon TMUV infection, polarized HD11 cells exhibited increased migration but reduced phagocytosis, as evidenced by scratch assay and neutral red uptake assay, respectively. Our present study characterized the polarization of host monocytes/macrophages upon TMUV infection, which may lay a foundation for further research on the immune escape mechanism and pathogenic mechanism of TMUV.


Assuntos
Galinhas/fisiologia , Patos/fisiologia , Infecções por Flavivirus/veterinária , Flavivirus/imunologia , Doenças das Aves Domésticas/virologia , Animais , Antivirais , Linhagem Celular , Movimento Celular , Polaridade Celular , Galinhas/virologia , Patos/virologia , Infecções por Flavivirus/virologia , Especificidade de Hospedeiro , Macrófagos/fisiologia , Macrófagos/virologia , Monócitos/fisiologia , Monócitos/virologia , Óxido Nítrico/metabolismo , Fagocitose , Organismos Livres de Patógenos Específicos , Replicação Viral
17.
J Virol ; 95(13): e0228820, 2021 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-33762417

RESUMO

The cellular entry pathways and the mechanisms of Newcastle disease virus (NDV) entry into cells are poorly characterized. In this study, we demonstrated that chicken interferon-induced transmembrane protein 1 (chIFITM1), which is located in the early endosomes, could limit the replication of NDV in chicken macrophage cell line HD11, suggesting the endocytic entry of NDV into chicken macrophages. Then, we presented a systematic study about the entry mechanism of NDV into chicken macrophages. First, we demonstrated that a low-pH condition and dynamin were required during NDV entry. However, NDV entry into chicken macrophages was independent of clathrin-mediated endocytosis. We also found that NDV entry was dependent on membrane cholesterol. The NDV entry and replication were significantly reduced by nystatin and phorbol 12-myristate 13-acetate treatment, overexpression of dominant-negative (DN) caveolin-1, or knockdown of caveolin-1, suggesting that NDV entry depends on caveola-mediated endocytosis. However, macropinocytosis did not play a role in NDV entry into chicken macrophages. In addition, we found that Rab5, rather than Rab7, was involved in the entry and traffic of NDV. The colocalization of NDV with Rab5 and early endosome suggested that NDV virion was transported to early endosomes in a Rab5-dependent manner after internalization. Of particular note, the caveola-mediated endocytosis was also utilized by NDV to enter primary chicken macrophages. Moreover, NDV entered different cell types using different pathways. Collectively, our findings demonstrate for the first time that NDV virion enters chicken macrophages via a pH-dependent, dynamin and caveola-mediated endocytosis pathway and that Rab5 is involved in the traffic and location of NDV. IMPORTANCE Although the pathogenesis of Newcastle disease virus (NDV) has been extensively studied, the detailed mechanism of NDV entry into host cells is largely unknown. Macrophages are the first-line defenders of host defense against infection of pathogens. Chicken macrophages are considered one of the main types of target cells during NDV infection. Here, we comprehensively investigated the entry mechanism of NDV in chicken macrophages. This is the first report to demonstrate that NDV enters chicken macrophages via a pH-dependent, dynamin and caveola-mediated endocytosis pathway that requires Rab5. The result is important for our understanding of the entry of NDV in chicken macrophages, which will further advance the knowledge of NDV pathogenesis and provide useful clues for the development of novel preventive or therapeutic strategies against NDV infection. In addition, this information will contribute to our further understanding of pathogenesis with regard to other members of the Avulavirus genus in the Paramyxoviridae family.


Assuntos
Endocitose/fisiologia , Macrófagos/virologia , Doença de Newcastle/transmissão , Internalização do Vírus , Proteínas rab5 de Ligação ao GTP/metabolismo , Animais , Antígenos de Diferenciação/metabolismo , Cavéolas/metabolismo , Linhagem Celular , Embrião de Galinha , Galinhas , Dinaminas/metabolismo , Concentração de Íons de Hidrogênio , Vírus da Doença de Newcastle/crescimento & desenvolvimento , Interferência de RNA , RNA Interferente Pequeno/genética , Proteínas rab5 de Ligação ao GTP/genética
18.
Poult Sci ; 99(11): 5440-5451, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33142461

RESUMO

Four GI-1/Massachusetts-type (GI-1/Mass-type) infectious bronchitis virus (IBV) strains were isolated and the complete genomes of these isolates, coupled with the Mass-type live-attenuated vaccine H120 and the Mass-type pathogenic M41 strains, were sequenced in the present study. Our results show that isolates LJL/140820 and I0306/17 may be derived from the Ma5 (another Mass-type live-attenuated vaccine strain) and H120 vaccine strains, respectively. The I1124/16 strain was found to be a M41 variant that likely resulted from nucleotide accumulated mutations in the genome. Consistently, the results of the virus neutralization test showed that isolate I1124/16 was antigenically related but slight different from the M41. Our results from the protection experiments pointed out that chickens immunized with H120 failed to eliminate viral shedding after infection with the isolate I1124/16, which was different from that of M41; this result was consistent to the field observation and further implicated that the variant IBV isolate I1124/16 was antigenic different from the M41 strain. Furthermore, the I1124/16 was found to have comparable but slightly lower pathogenicity with the M41 strain. More studies based on the reverse genetic techniques are needed to elucidate the amino acids in the S1 subunit of spike protein contributing to the altered antigenicity of the isolate I1124/16. In addition, an IBV isolate, LJL/130609, was found to be originated from recombination events between the I1124/16- and Connecticut-like strains. Our results from the virus neutralization test also showed that isolates LJL/130609 and I1124/16 were antigenic closely related. Hence, there are at least 3 different genetic evolution patterns for the circulation of the GI-1/Mass-type IBV field strains in China. The differences of vaccines used, the field conditions and genetic pressures between different flocks, likely account for the emergence, evolution patterns, and characteristics of the Mass-type IBV strains.


Assuntos
Antígenos Virais , Infecções por Coronavirus , Heterogeneidade Genética , Vírus da Bronquite Infecciosa , Doenças das Aves Domésticas , Animais , Antígenos Virais/genética , Galinhas , China , Infecções por Coronavirus/veterinária , Infecções por Coronavirus/virologia , Vírus da Bronquite Infecciosa/genética , Doenças das Aves Domésticas/virologia
19.
Virus Genes ; 56(6): 734-748, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33009986

RESUMO

Fowlpox virus (FPV) is used as a vaccine vector to prevent diseases in poultry and mammals. The insertion site is considered as one of the main factors influencing foreign gene expression. Therefore, the identification of insertion sites that can stably and efficiently express foreign genes is crucial for the construction of recombinant vaccines. In this study, we found that the insertion of foreign genes into ORF054 and the ORF161/ORF162 intergenic region of the FPV genome did not affect replication, and that the foreign genes inserted into the intergenic region were more efficiently expressed than when they were inserted into a gene. Based on these results, the recombinant virus rFPVNX10-NDV F-E was constructed and immune protection against virulent FPV and Newcastle disease virus (NDV) was evaluated. Tests for anti-FPV antibodies in the vaccinated chickens were positive within 14 days post-vaccination. After challenge with FPV102, no clinical signs of FP were observed in vaccinated chickens, as compared to that in the control group (unvaccinated), which showed 100% morbidity. Low levels of NDV-specific neutralizing antibodies were detected in vaccinated chickens before challenge. After challenge with NDV ck/CH/LHLJ/01/06, all control chickens died within 4 days post-challenge, whereas 5/15 vaccinated chickens died between 4 and 12 days post-challenge. Vaccination provided an immune protection rate of 66.7%, whereas the control group showed 100% mortality. These results indicate that the ORF161/ORF162 intergenic region of FPVNX10 can be used as a recombination site for foreign gene expression in vivo and in vitro.


Assuntos
Vírus da Varíola das Aves Domésticas/genética , Varíola Aviária/prevenção & controle , Doença de Newcastle/prevenção & controle , Doenças das Aves Domésticas/prevenção & controle , Proteínas Virais de Fusão/genética , Vacinas Virais/genética , Animais , Linhagem Celular , Embrião de Galinha , Galinhas , DNA Intergênico , Fibroblastos , Vacinação/veterinária , Vacinas Sintéticas/genética
20.
Vet Microbiol ; 250: 108835, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33011664

RESUMO

To develop an alternative vectored vaccine against both Newcastle disease virus (NDV) and infectious laryngotracheitis virus (ILTV), the glycoprotein C (gC) gene was first deleted from an avirulent ILTV. Based on this gC-deleted ILTV mutant, a recombinant ILTV expressing the fusion protein (F) of a genotype VII NDV (designated ILTV-ΔgC-F) was then constructed. Expression of the NDV F protein in ILTV-ΔgC-F-infected LMH cells was examined with an immunofluorescence assay and western blotting. The F gene was stably maintained in the genome of ILTV-ΔgC-F and the F protein was stably expressed. Compared with the parental virus, ILTV-ΔgC-F demonstrated an increased penetration capacity in vitro, and an increased replication rate in vitro and in vivo. Both the parental virus and ILTV-ΔgC-F were avirulent in chickens. Vaccination of specific-pathogen-free chickens with ILTV-ΔgC-F induced ILTV-specific antibodies, detected with an enzyme-linked immunosorbent assay (ELISA), and provided complete clinical protection against virulent ILTV, although viral shedding and replication were detected in the respiratory tract in the early stage of infection in a very small number of birds. Vaccination with ILTV-ΔgC-F also provided significant protection against challenge with a virulent genotype VII NDV, although the level of NDV-specific antibodies detected with an ELISA was low. Notably, the numbers of birds that were positive for the virulent genotype VII NDV and the replication of the challenge virus NDV in selected target tissues were significantly lower in the ILTV-ΔgC-F-vaccinated chickens than in the control birds. Our results indicate that ILTV-ΔgC-F has potential utility as a bivalent candidate vaccine against both infectious laryngotracheitis and Newcastle disease.


Assuntos
Infecções por Herpesviridae/veterinária , Doença de Newcastle/prevenção & controle , Doenças das Aves Domésticas/prevenção & controle , Proteínas do Envelope Viral/genética , Proteínas Virais de Fusão/genética , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/sangue , Linhagem Celular , Galinhas , Deleção de Genes , Genótipo , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/prevenção & controle , Herpesvirus Galináceo 1/genética , Herpesvirus Galináceo 1/imunologia , Masculino , Doença de Newcastle/imunologia , Vírus da Doença de Newcastle/genética , Doenças das Aves Domésticas/imunologia , Doenças das Aves Domésticas/virologia , Proteínas Recombinantes/imunologia , Organismos Livres de Patógenos Específicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...