Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Retrovirology ; 19(1): 19, 2022 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-36002842

RESUMO

BACKGROUND: Avian leukosis virus (ALV) is an infectious retrovirus, that mainly causes various forms of tumours, immunosuppression, a decreased egg production rate and slow weight gain in poultry. ALV consists of 11 subgroups, A-K, among which ALV-K is an emerging subgroup that has become prevalent in the past 10 years. Most ALV-K isolates showed weak replication ability and pathogenicity. In this study, the weak replication ability of ALV-K was explored from the perspective of the interaction between ALV-K gp85 and the Tva receptor. METHODS: Fourteen soluble recombinant ALV-A/K gp85 chimeric proteins were constructed by substituting the sequence difference regions (hr1, hr2 and vr3) of the ALV-A gp85 protein with the skeleton ALV-K gp85 protein for co-IP and competitive blocking tests. RESULTS: The binding capacity of ALV-K gp85 to Tva was significantly weaker than that of ALV-A gp85 (P < 0.05) and the key amino acid sites 199-205, 269, 319, 321 and 324 of ALV-K env contributed to the weaker replication capacity of ALV-K than ALV-A. CONCLUSIONS: This is the first study to reveal the molecular factors of the weak replication ability of ALV-K from the perspective of the interaction of ALV-K gp85 to Tva, providing a basis for further elucidation of the infection mechanism of ALV-K.


Assuntos
Vírus da Leucose Aviária , Leucose Aviária , Doenças das Aves Domésticas , Aminoácidos/metabolismo , Animais , Leucose Aviária/metabolismo , Vírus da Leucose Aviária/genética , Galinhas , Humanos , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo
2.
Front Immunol ; 12: 774323, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34777393

RESUMO

J subgroup avian leukosis virus (ALV-J) infection causes serious immunosuppression problems, leading to hematopoietic malignancy tumors in chicken. It has been demonstrated that interferon-stimulated genes (ISGs) could limit ALV-J replication; nevertheless, the underlying mechanisms remain obscure. Here, we demonstrate that Long-chain Acyl-CoA synthetase 1 (ACSL1) is an interferon (IFN)-stimulated gene that specifically restricts the replication of ALV-J due to the higher IFN-I production. More importantly, ACSL1 induces primary monocyte-derived macrophages (MDMs) to pro-inflammatory phenotypic states during ALV-J infection, and ACSL1 mediates apoptosis through the PI3K/Akt signaling pathway in ALV-J-infected primary monocyte-derived macrophages (MDMs). Overall, these results provide evidence that ACSL1 contributes to the antiviral response against ALV-J.


Assuntos
Vírus da Leucose Aviária/fisiologia , Leucose Aviária/metabolismo , Leucose Aviária/virologia , Coenzima A Ligases/metabolismo , Interferon Tipo I/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Replicação Viral , Animais , Apoptose , Biomarcadores , Galinhas , Coenzima A Ligases/genética , Suscetibilidade a Doenças , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Modelos Biológicos , Regiões Promotoras Genéticas
3.
J Virol ; 94(11)2020 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-32213616

RESUMO

Different from other subgroups of avian leukosis viruses (ALVs), ALV-J is highly pathogenic. It is the main culprit causing myeloid leukemia and hemangioma in chickens. The distinctiveness of the env gene of ALV-J, with low homology to those of other ALVs, is linked to its unique pathogenesis, but the underlying mechanism remains unclear. Previous studies show that env of ALV-J can be grouped into three species based on the tyrosine motifs in the cytoplasmic domain (CTD) of Gp37, i.e., the inhibitory, bifunctional, and active groups. To explore whether the C terminus or the tyrosine motifs in the CTD of Gp37 affect the pathogenicity of ALV-J, a set of ALV-J infectious clones containing different C termini of Gp37 or the mutants at the tyrosine sites were tested in vitro and in vivo Viral growth kinetics indicated not only that ALV-J with active env is the fastest in replication and ALV-J with inhibitory env is the lowest but also that the tyrosine sites essentially affected the replication of ALV-J. Moreover, in vivo studies demonstrated that chickens infected by ALV-J with active or bifunctional env showed higher viremia, cloacal viral shedding, and viral tissue load than those infected by ALV-J with inhibitory env Notably, the chickens infected by ALV-J with active or bifunctional env showed significant loss of body weight compared with the control chickens. Taken together, these findings reveal that the C terminus of Gp37 plays a vital role in ALV-J pathogenesis, and change from inhibitory env to bifunctional or active env increases the pathogenesis of ALV-J.IMPORTANCE ALV-J can cause severe immunosuppression and myeloid leukemia in infected chickens. However, no vaccine or antiviral drug is available against ALV-J, and the mechanism for ALV-J pathogenesis needs to be elucidated. It is generally believed that gp85 and LTR of ALV contribute to its pathogenesis. Here, we found that the C terminus and the tyrosine motifs (YxxM, ITIM, and ITAM-like) in the CTD of Gp37 of ALV-J could affect the pathogenicity of ALV-J in vitro and in vivo The pathogenicity of ALV-J with Gp37 containing ITIM only was significantly less than ALV-J with Gp37 containing both YxxM and ITIM and ALV-J with Gp37 containing both YxxM and ITAM-like. This study highlights the vital role of the C terminus of Gp37 in the pathogenesis of ALV-J and thus provides a new perspective to elucidate the interaction between ALV-J and its host and a molecular basis to develop efficient strategies against ALV-J.


Assuntos
Vírus da Leucose Aviária/metabolismo , Vírus da Leucose Aviária/patogenicidade , Leucose Aviária/metabolismo , Doenças das Aves Domésticas/metabolismo , Proteínas do Envelope Viral/metabolismo , Motivos de Aminoácidos , Animais , Leucose Aviária/genética , Leucose Aviária/patologia , Vírus da Leucose Aviária/genética , Linhagem Celular , Galinhas , Mutação , Doenças das Aves Domésticas/genética , Doenças das Aves Domésticas/patologia , Domínios Proteicos , Proteínas do Envelope Viral/genética
4.
J Leukoc Biol ; 107(2): 299-307, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31945209

RESUMO

The zinc finger antiviral protein (ZAP), as a host restriction factor, inhibits the replication of certain viruses by binding viral mRNA or proteins for degradation. However, little is known about the role of ZAP in the antiviral immune response. We now show that ZAP participates in the antiviral immune response by activating T cells. Overexpression of ZAP significantly inhibited avian leukosis virus subgroup J (ALV-J) replication and reduced the associated inflammatory damage in vivo. In this study, we found that ZAP tended to be expressed in T lymphocytes, especially after ALV-J infection. T lymphocyte proliferation proceeded as usual in response to ALV-J infection in the presence of ZAP, indicating that ZAP endows T lymphocytes with resistance to the immunosuppression caused by ALV-J. Furthermore, ZAP activated cytokine secretion by T lymphocytes by contributing to nuclear translocation of nuclear factors of activated T cells and indirectly promoted anti-ALV-J antibody generation. Together, our findings show that ZAP, acting as an immunomodulatory factor, is involved in the antiviral immune response via T lymphocyte activation.


Assuntos
Antivirais/metabolismo , Leucose Aviária/imunologia , Ativação Linfocitária/imunologia , Linfoma/imunologia , Doença de Marek/imunologia , Proteínas de Ligação a RNA/metabolismo , Linfócitos T/imunologia , Animais , Leucose Aviária/metabolismo , Leucose Aviária/virologia , Vírus da Leucose Aviária/imunologia , Proteínas Aviárias/genética , Proteínas Aviárias/metabolismo , Galinhas , Citocinas/metabolismo , Linfoma/metabolismo , Linfoma/virologia , Doença de Marek/metabolismo , Doença de Marek/virologia , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/imunologia , Linfócitos T/metabolismo , Células Tumorais Cultivadas , Proteínas Virais/genética , Proteínas Virais/metabolismo , Replicação Viral , Dedos de Zinco
5.
J Virol ; 93(17)2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31217247

RESUMO

Avian leukosis virus subgroup K (ALV-K) is composed of newly emerging isolates, which, in sequence analyses, cluster separately from the well-characterized subgroups A, B, C, D, E, and J. However, it remains unclear whether ALV-K represents an independent ALV subgroup with regard to receptor usage, host range, and superinfection interference. In the present study, we examined the host range of the Chinese infectious isolate JS11C1, an ALV-K prototype, and we found substantial overlap of species that were either resistant or susceptible to ALV-A and JS11C1. Ectopic expression of the chicken tva gene in mammalian cells conferred susceptibility to JS11C1, while genetic ablation of the tva gene rendered chicken DF-1 cells resistant to infection by JS11C1. Thus, tva expression is both sufficient and necessary for JS11C1 entry. Receptor sharing was also manifested in superinfection interference, with preinfection of cells with ALV-A, but not ALV-B or ALV-J, blocking subsequent JS11C1 infection. Finally, direct binding of JS11C1 and Tva was demonstrated by preincubation of the virus with soluble Tva, which substantially decreased viral infectivity in susceptible chicken cells. Collectively, these findings indicate that JS11C1 represents a new and bona fide ALV subgroup that utilizes Tva for cell entry and binds to a site other than that for ALV-A.IMPORTANCE ALV consists of several subgroups that are particularly characterized by their receptor usage, which subsequently dictates the host range and tropism of the virus. A few newly emerging and highly pathogenic Chinese ALV strains have recently been suggested to be an independent subgroup, ALV-K, based solely on their genomic sequences. Here, we performed a series of experiments with the ALV-K strain JS11C1, which showed its dependence on the Tva cell surface receptor. Due to the sharing of this receptor with ALV-A, both subgroups were able to interfere with superinfection. Because ALV-K could become an important pathogen and a significant threat to the poultry industry in Asia, the identification of a specific receptor could help in the breeding of resistant chicken lines with receptor variants with decreased susceptibility to the virus.


Assuntos
Vírus da Leucose Aviária/patogenicidade , Leucose Aviária/genética , Proteínas Aviárias/genética , Proteínas Aviárias/metabolismo , Receptores Virais/genética , Receptores Virais/metabolismo , Animais , Leucose Aviária/metabolismo , Leucose Aviária/virologia , Vírus da Leucose Aviária/classificação , Vírus da Leucose Aviária/fisiologia , Linhagem Celular , Galinhas , Suscetibilidade a Doenças , Fibroblastos/citologia , Fibroblastos/metabolismo , Fibroblastos/virologia , Mesocricetus , Especificidade da Espécie , Internalização do Vírus
6.
Poult Sci ; 98(9): 3533-3538, 2019 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-31002116

RESUMO

Avian leukosis virus subgroup J (ALV-J) belongs to α-retrovirus genus of retroviridae, causing neoplastic disease, immunosuppression, and other problems in the poultry industry worldwide. The aim of this study was to determine whether the reproductive hormone fluctuation affect the ALV-J viremia at the early egg-laying phase in chicken. Total 8 suspected ALV-J-infected Chinese yellow chickens in the early egg-laying phase were collected from 2 different farms, and further confirmed by PCR and immunofluorescence assay. Plasma samples were collected from experimental chickens for 5 to 10 consecutive weeks at a settled time in each week. ALV-J viremia and reproductive hormone levels were monitored by ELISA or radioimmunoassay. The results showed that fluctuations in luteinizing hormone (LH), follicle stimulating hormone (FSH), and progesterone might have an impact on the disappearance of ALV-J viremia in chickens with intermittent viremia, but not in chickens with persistent viremia. These results suggest that reproductive hormone changes during the early egg-laying phase will affect the detection of positive ALV-J-infected chickens in the ALV-J eradication procedures.


Assuntos
Leucose Aviária/virologia , Galinhas/metabolismo , Hormônio Foliculoestimulante/sangue , Hormônio Luteinizante/sangue , Doenças das Aves Domésticas/virologia , Progesterona/sangue , Viremia/veterinária , Animais , Leucose Aviária/metabolismo , Vírus da Leucose Aviária/fisiologia , Feminino , Doenças das Aves Domésticas/metabolismo , Viremia/metabolismo , Viremia/virologia
7.
Gene ; 701: 72-81, 2019 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-30898701

RESUMO

Avian leukosis virus subgroup J (ALV-J) is an oncogenic retrovirus that causes severe economic losses to the poultry industry worldwide. Circular RNAs (circRNAs) are a class of non-coding RNAs that has been described in various biological systems and pathogenic processes. However, the immune mechanisms in response to circRNAs remain unknown. In this study, high-throughput transcriptome sequencing was used to detect circRNAs present in chicken macrophage (HD11) and chick embryo fibroblast (CEF) cells infected with ALV-J. We identified 7684 circRNAs from diverse genomic locations in CEF and HD11 after ALV-J infection, these RNAs showed complex expression patterns that differed based on the cells type and infection time. In total, 302 differentially expressed (DE) circRNAs and 164 DE circRNAs were identified in CEF and HD11 after ALV-J infection, respectively. CircRNA7419-associated with KDM4C- and circRNA6679 and circRNA6680-associated with TNFAIP6- were involved in the immune response upon ALV-J infection in CEF. Host genes were analyzed through further bioinformatics analysis. The result confirmed that a large number of DE circRNAs corresponded to several immune-associated or tumor-associated terms and pathways, such as Mucin type O-Glycan biosynthesis, MAPK signaling pathway, B cell receptor signaling, and Wnt signaling pathway in CEF, as well as Jak-STAT signaling pathway, apoptosis, and MAPK signaling pathway in HD11. CircRNAs related to the B cell receptor signaling pathway in CEF, and the Jak-STAT signaling pathway in HD11, were selected for circRNA-miRNA interaction network analyses. Our study indicates that circRNAs expression was altered by ALV-J infection in both CEF and HD11, and may play a key role in the progression of ALV-J infection.


Assuntos
Vírus da Leucose Aviária , Leucose Aviária , Galinhas , Sistema de Sinalização das MAP Quinases , Doenças das Aves Domésticas , RNA , Via de Sinalização Wnt , Animais , Leucose Aviária/genética , Leucose Aviária/metabolismo , Leucose Aviária/patologia , Vírus da Leucose Aviária/genética , Vírus da Leucose Aviária/metabolismo , Proteínas Aviárias/genética , Proteínas Aviárias/metabolismo , Linhagem Celular , Embrião de Galinha , Galinhas/genética , Galinhas/metabolismo , Galinhas/virologia , Doenças das Aves Domésticas/genética , Doenças das Aves Domésticas/metabolismo , Doenças das Aves Domésticas/patologia , Doenças das Aves Domésticas/virologia , RNA/genética , RNA/metabolismo
8.
Res Vet Sci ; 123: 65-70, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30592994

RESUMO

The CCCH-type zinc finger antiviral protein (ZAP), a host antiviral factor, plays an important role in innate defenses. Although the anti-viral mechanism of ZAP has been elucidated, however, the tissue specificity and the viral infection correlativity have not been fully understood. Here, we tested the dynamic distribution and localization of chicken ZAP (chZAP) before and after avian leukosis virus subgroup J (ALV-J) infection. The results showed that chZAP was highly expressed in adrenal gland and testis before ALV-J infection, and significantly upregulated in liver, kidney and bursa of Fabricius, and extremely overexpressed in spleen after ALV-J infection. The results indicated that chZAP is an inducible protein and showed specific overexpression in spleen after ALV-J infection. Furthermore, we demonstrated that chZAP, as a host intracytoplasmic factor, accumulated and migrated to the periphery of nucleus in DF-1 cells post-infection with ALV-J. Taken together, chZAP characterized as an inducible antiviral protein and specifically overexpressed in spleen after ALV-J infection.


Assuntos
Vírus da Leucose Aviária , Leucose Aviária/metabolismo , Proteínas de Transporte/metabolismo , Galinhas , Baço/metabolismo , Animais , Leucose Aviária/imunologia , Leucose Aviária/virologia , Bolsa de Fabricius , Galinhas/metabolismo , Regulação da Expressão Gênica/imunologia , Baço/virologia , Regulação para Cima , Dedos de Zinco
9.
Retrovirology ; 15(1): 45, 2018 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-29970099

RESUMO

BACKGROUND: Co-infection with avian leukosis virus subgroup J and reticuloendotheliosis virus induces synergistic pathogenic effects and increases mortality. However, the role of exosomal miRNAs in the molecular mechanism of the synergistic infection of the two viruses remains unknown. RESULTS: In this study, exosomal RNAs from CEF cells infected with ALV-J, REV or both at the optimal synergistic infection time were analysed by Illumina RNA deep sequencing. A total of 54 (23 upregulated and 31 downregulated) and 16 (7 upregulated and 9 downregulated) miRNAs were identified by comparing co-infection with two viruses, single-infected ALV-J and REV, respectively. Moreover, five key miRNAs, including miR-184-3p, miR-146a-3p, miR-146a-5p, miR-3538 and miR-155, were validated in both exosomes and CEF cells by qRT-PCR. GO annotation and KEGG pathway analysis of the miRNA target genes showed that the five differentially expressed miRNAs participated in virus-vector interaction, oxidative phosphorylation, energy metabolism and cell growth. CONCLUSIONS: We demonstrated that REV and ALV-J synergistically increased the accumulation of exosomal miRNAs, which sheds light on the synergistic molecular mechanism of ALV-J and REV.


Assuntos
Vírus da Leucose Aviária/fisiologia , Coinfecção , Exossomos/genética , MicroRNAs/genética , Interações Microbianas , Vírus da Reticuloendoteliose/fisiologia , Infecções por Retroviridae/genética , Infecções por Retroviridae/virologia , Animais , Leucose Aviária/genética , Leucose Aviária/metabolismo , Leucose Aviária/virologia , Linhagem Celular , Exossomos/metabolismo , Exossomos/ultraestrutura , Regulação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Interações Hospedeiro-Patógeno , Interferência de RNA , Reprodutibilidade dos Testes , Infecções por Retroviridae/metabolismo , Replicação Viral
10.
Immunol Invest ; 47(5): 443-456, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29768058

RESUMO

Immunosuppressive virus, which can cause suppressed immunity and vaccination failure, frequently occurs in chicken flocks and seriously destroys the poultry industry. Our previous studies have reported that Taishan Pinus massoniana pollen polysaccharide (TPPPS) possess immunomodulatory effects and improve the immune effects of vaccines. In this study, avian leukosis virus subgroup B (ALV-B) was chosen as immunosuppressive virus to artificially establish immunosuppressive models in chickens, and the immune modulatory ability of TPPPS on the immune response of chickens was evaluated. Four randomly assigned groups (Group I-IV) of these immunosuppressed chickens were administered with TPPPS at doses of 0, 100, 200, and 400 mg/kg (every kilogram chick), respectively. Group V was administered with saline as control. At seven day old, 10 chickens randomly selected from Group I-V were inoculated with the attenuated Newcastle disease (ND) vaccine. The results showed that during the monitoring period, TPPPS significantly enhanced weight of immune organs, peripheral lymphocyte proliferation, the percentage of CD4+ and the ratio of CD4+/CD8+, IL-2 and IFN-γ production, and ALV-B antibody positive rate of chickens in a dose-dependent manner, with 400 mg/kg TPPPS being the most effective. In addition, the antibody titer against Newcastle disease virus (NDV) in Group IV with 400 mg/kg was significantly higher than those in other groups. We observed the stronger immunity in the TPPPS group, which indicates that TPPPS could be used as an immunoenhancer to relieve immunosuppression caused by ALV-B in the poultry industry.


Assuntos
Vírus da Leucose Aviária/imunologia , Leucose Aviária/imunologia , Leucose Aviária/virologia , Galinhas/imunologia , Galinhas/virologia , Imunomodulação , Pólen/imunologia , Polissacarídeos/imunologia , Adjuvantes Imunológicos , Animais , Anticorpos Antivirais/imunologia , Antígenos de Plantas/imunologia , Leucose Aviária/metabolismo , Citocinas/metabolismo , Sistema Imunitário/citologia , Sistema Imunitário/imunologia , Sistema Imunitário/metabolismo , Ativação Linfocitária , Linfócitos/imunologia , Linfócitos/metabolismo , Pinus , Carga Viral , Vacinas Virais/administração & dosagem , Replicação Viral/imunologia
11.
J Virol ; 92(8)2018 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-29263268

RESUMO

The group of highly related avian leukosis viruses (ALVs) in chickens are thought to have evolved from a common retroviral ancestor into six subgroups, A to E and J. These ALV subgroups use diverse cellular proteins encoded by four genetic loci in chickens as receptors to gain entry into host cells. Hosts exposed to ALVs might be under selective pressure to develop resistance to ALV infection. Indeed, resistance alleles have previously been identified in all four receptor loci in chickens. The tvb gene encodes a receptor, which determines the susceptibility of host cells to ALV subgroup B (ALV-B), ALV-D, and ALV-E. Here we describe the identification of two novel alleles of the tvb receptor gene, which possess independent insertions each within exon 4. The insertions resulted in frameshift mutations that reveal a premature stop codon that causes nonsense-mediated decay of the mutant mRNA and the production of truncated Tvb protein. As a result, we observed that the frameshift mutations in the tvb gene significantly lower the binding affinity of the truncated Tvb receptors for the ALV-B, ALV-D, and ALV-E envelope glycoproteins and significantly reduce susceptibility to infection by ALV-B, ALV-D and ALV-E in vitro and in vivo Taken together, these findings suggest that frameshift mutation can be a molecular mechanism of reducing susceptibility to ALV and enhance our understanding of virus-host coevolution.IMPORTANCE Avian leukosis virus (ALV) once caused devastating economic loss to the U.S. poultry industry prior the current eradication schemes in place, and it continues to cause severe calamity to the poultry industry in China and Southeast Asia, where deployment of a complete eradication scheme remains a challenge. The tvb gene encodes the cellular receptor necessary for subgroup B, D, and E ALV infection. Two tvb allelic variants that resulted from frameshift mutations have been identified in this study, which have been shown to have significantly reduced functionality in mediating subgroup B, D, and E ALV infection. Unlike the control of herpesvirus-induced diseases by vaccination, the control of avian leukosis in chickens has relied totally on virus eradication measures and host genetic resistance. This finding enriches the allelic pool of the tvb gene and expands the potential for genetic improvement of ALV resistance in varied chicken populations by selection.


Assuntos
Vírus da Leucose Aviária/metabolismo , Leucose Aviária , Proteínas Aviárias , Galinhas , Mutação da Fase de Leitura , Predisposição Genética para Doença , Receptores Virais , Animais , Leucose Aviária/genética , Leucose Aviária/metabolismo , Vírus da Leucose Aviária/genética , Proteínas Aviárias/genética , Proteínas Aviárias/metabolismo , Linhagem Celular , Galinhas/genética , Galinhas/metabolismo , Galinhas/virologia , Receptores Virais/genética , Receptores Virais/metabolismo
12.
Vet Res Commun ; 41(3): 219-226, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28190219

RESUMO

Two important microRNAs, gga-let-7b and gga-let-7i were examined for the relative expression in liver and bone marrow tissues from specific pathogen free chickens that were challenged either with GD1109 or NX0101 strain of subgroup J avian leukosis virus (ALV-J). The GD1109 strain of ALV-J reportedly causes hemangioma (HE) and NX0101 reportedly causes myeloma (ML) in susceptible chickens. Temporal changes of both gga-let-7b and gga-let-7i expression in ALV-J infected chickens were observed in contrast to its counterpart of a non-infected negative control group of chickens (P < 0.05 or P < 0.01) during the first 120 days post infection. Use of the web-based computational DIANA-mirPath software (available at http://microrna.gr/mirpath ), it was predicted that both gga-let-7b and gga-let-7i were involved in multiple pathways including signaling pathways, such as MAPK, TGF-beta, Notch, Wnt, mTOR, Cell cycle, P53 and Jak-STAT. Combining our experimental data with reports on the microRNAs, we suggest that both gga-let-7i and gga-let-7b may also act as tumor suppressors in chicken, especially play a critical role in tumorigenesis induced by ALV-J.


Assuntos
Vírus da Leucose Aviária , Leucose Aviária/metabolismo , MicroRNAs/metabolismo , Animais , Leucose Aviária/virologia , Galinhas/metabolismo , Galinhas/virologia , Regulação Viral da Expressão Gênica , Reação em Cadeia da Polimerase/veterinária , Fatores de Tempo
13.
Microb Pathog ; 104: 48-55, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28065818

RESUMO

Avian leukosis virus subgroup J (ALV-J), an oncogenic retrovirus, induces growth retardation and neoplasia in chickens, leading to enormous economic losses in poultry industry. Increasing evidences showed several signal pathways involved in ALV-J infection. However, what signaling pathway involved in growth retardation is largely unknown. To explore the possible signaling pathway, we tested the cell proliferation and associated miRNAs in ALV-J infected CEF cells by CCK-8 and Hiseq, respectively. The results showed that cell proliferation was significantly inhibited by ALV-J and three associated miRNAs were identified to target Wnt/ß-catenin pathway. To verify the Wnt/ß-catenin pathway involved in cell growth retardation, we analyzed the key molecules of Wnt pathway in ALV-J infected CEF cells. Our data demonstrated that protein expression of ß-catenin was decreased significantly post ALV-J infection compared with the normal (P < 0.05). The impact of this down-regulation caused low expression of known target genes (Axin2, CyclinD1, Tcf4 and Lef1). Further, to obtain in vivo evidence, we set up an ALV-J infection model. Post 7 weeks infection, ALV-J infected chickens showed significant growth retardation. Subsequent tests showed that the expression of ß-catenin, Tcf1, Tcf4, Lef1, Axin2 and CyclinD1 were down-regulated in muscles of growth retardation chickens. Taken together, all data demonstrated that chicken growth retardation caused by ALV-J associated with down-regulated Wnt/ß-catenin signaling pathway.


Assuntos
Vírus da Leucose Aviária/fisiologia , Leucose Aviária/metabolismo , Leucose Aviária/virologia , Galinhas , Fenótipo , Via de Sinalização Wnt , Animais , Leucose Aviária/complicações , Leucose Aviária/genética , Vírus da Leucose Aviária/classificação , Linhagem Celular , Proliferação de Células , Análise por Conglomerados , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , MicroRNAs/genética , Fatores de Transcrição/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
14.
J Virol ; 91(3)2017 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-27881654

RESUMO

The J subgroup of avian leukosis virus (ALV-J) infects domestic chickens, jungle fowl, and turkeys. This virus enters the host cell through a receptor encoded by the tvj locus and identified as Na+/H+ exchanger 1. The resistance to avian leukosis virus subgroup J in a great majority of galliform species has been explained by deletions or substitutions of the critical tryptophan 38 in the first extracellular loop of Na+/H+ exchanger 1. Because there are concerns of transspecies virus transmission, we studied natural polymorphisms and susceptibility/resistance in wild galliforms and found the presence of tryptophan 38 in four species of New World quails. The embryo fibroblasts of New World quails are susceptible to infection with avian leukosis virus subgroup J, and the cloned Na+/H+ exchanger 1 confers susceptibility on the otherwise resistant host. New World quails are also susceptible to new avian leukosis virus subgroup J variants but resistant to subgroups A and B and weakly susceptible to subgroups C and D of avian sarcoma/leukosis virus due to obvious defects of the respective receptors. Our results suggest that the avian leukosis virus subgroup J could be transmitted to New World quails and establish a natural reservoir of circulating virus with a potential for further evolution. IMPORTANCE: Since its spread in broiler chickens in China and Southeast Asia in 2000, ALV-J remains a major enzootic challenge for the poultry industry. Although the virus diversifies rapidly in the poultry, its spillover and circulation in wild bird species has been prevented by the resistance of most species to ALV-J. It is, nevertheless, important to understand the evolution of the virus and its potential host range in wild birds. Because resistance to avian retroviruses is due particularly to receptor incompatibility, we studied Na+/H+ exchanger 1, the receptor for ALV-J. In New World quails, we found a receptor compatible with virus entry, and we confirmed the susceptibilities of four New World quail species in vitro We propose that a prospective molecular epidemiology study be conducted to identify species with the potential to become reservoirs for ALV-J.


Assuntos
Vírus da Leucose Aviária/fisiologia , Leucose Aviária/genética , Leucose Aviária/virologia , Suscetibilidade a Doenças , Codorniz , Sequência de Aminoácidos , Aminoácidos , Animais , Leucose Aviária/metabolismo , Vírus da Leucose Aviária/classificação , Células Cultivadas , Resistência à Doença/genética , Evolução Molecular , Expressão Gênica , Loci Gênicos , Especificidade de Hospedeiro , Interações Hospedeiro-Patógeno , Filogenia , Polimorfismo Genético , Domínios e Motivos de Interação entre Proteínas , Trocadores de Sódio-Hidrogênio/química , Trocadores de Sódio-Hidrogênio/genética , Trocadores de Sódio-Hidrogênio/metabolismo , Replicação Viral
15.
Microb Pathog ; 102: 29-35, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27890652

RESUMO

We have previously shown that the extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK/MAPK) pathway contributes to subgroup J avian leukosis virus (ALV-J) replication and tumorigenicity. However, a role for ERK/MAPK signaling in ALV-A and ALV-B replication is unknown. In this study we successfully constructed and recovered a recombinant form of ALV-A strain GD13-1 which showed similarities in growth to the parental wild type virus in vitro. ALV subgroups J, A or B all triggered ERK2 activation in primary CEF cells. ERK/MAPK inhibition markedly suppressed ALV-A and ALV-B replication as shown by extremely low levels of viral transcription and virus protein production. This finding provides evidence that ERK/MAPK signaling responses play important roles in ALV replication and may represent novel drug targets for therapeutic intervention strategies.


Assuntos
Vírus da Leucose Aviária/efeitos dos fármacos , Vírus da Leucose Aviária/fisiologia , Leucose Aviária/metabolismo , Leucose Aviária/virologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Replicação Viral/efeitos dos fármacos , Animais , Linhagem Celular , Células Cultivadas , Galinhas , Fibroblastos/metabolismo , Fibroblastos/virologia , Flavonoides , Ordem dos Genes , Vetores Genéticos/genética , Genoma Viral , Proteína Quinase 1 Ativada por Mitógeno/metabolismo
16.
Genet Mol Res ; 15(1)2016 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-27050957

RESUMO

Nucleotide-binding oligomerization domain-like receptors (NLRs) play a key role in the innate immune response as pattern-recognition receptors. However, the role of NLRC5, which is a member of the NLR family, in NF-κB activation and MHC-I expression remains debatable. Infection with the J group avian leukosis virus (ALV-J) can result in immunosuppression and a subsequent increase in susceptibility to secondary infection. This results in huge economic losses to the poultry industry worldwide. Using quantitative real-time polymerase chain reaction (qRT-PCR), we investigated the mRNA expression levels of NLRC5 signal pathway-related genes in secondary chicken embryo fibroblasts 7 days after infection with ALV-J. The results indicated that, compared with the control groups, the expression levels of TLR7, MHC-I, and IL-18 increased significantly in the infected groups at 7 days post-infection (d.p.i.). The expression levels of NLRC5 and IL-6 were conspicuously downregulated at 7 d.p.i., but the expression levels of NF-κB, STAT1, and STAT3 were not significantly altered. These results suggest that NLRC5 and some genes involved in the NLRC5 pathway play a key role in antiviral immunity, typically the response to ALV-J infection. Moreover, MHC-I expression levels vary between different cell types.


Assuntos
Leucose Aviária/metabolismo , Proteínas NLR/metabolismo , Transdução de Sinais , Regulação para Cima , Animais , Leucose Aviária/genética , Células Cultivadas , Embrião de Galinha , Citocinas , Fibroblastos/metabolismo , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , NF-kappa B/genética , NF-kappa B/metabolismo , Proteínas NLR/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT3/metabolismo , Receptor 7 Toll-Like/genética , Receptor 7 Toll-Like/metabolismo
17.
Biomed Res Int ; 2015: 395307, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25632391

RESUMO

Avian leukosis virus subgroup J (ALV-J) is an avian oncogenic retrovirus that can induce various clinical tumors and has caused severe economic losses in China. To improve our understanding of the host cellular responses to virus infection and the pathogenesis of ALV-J infection, we applied isobaric tags for relative and absolute quantification (iTRAQ) labeling coupled with multidimensional liquid chromatography-tandem mass spectrometry to detect the protein changes in DF-1 cells infected and mock-infected with ALV-J. A total of 75 cellular proteins were significantly changed, including 33 upregulated proteins and 42 downregulated proteins. The reliability of iTRAQ-LC MS/MS was confirmed via real-time PCR. Most of these proteins were related to the physiological functions of metabolic processes, biosynthetic processes, responses to stimuli, protein binding, signal transduction, cell cytoskeleton, and so forth. We also found some proteins that play important roles in apoptosis and oncogenicity. The differentially expressed proteins identified may provide valuable information to elucidate the pathogenesis of virus infection and virus-host interactions.


Assuntos
Vírus da Leucose Aviária/fisiologia , Leucose Aviária/metabolismo , Cromatografia Líquida/métodos , Proteoma/metabolismo , Proteômica/métodos , Espectrometria de Massas em Tandem/métodos , Animais , Linhagem Celular , Galinhas , Técnica Indireta de Fluorescência para Anticorpo , Marcação por Isótopo , Anotação de Sequência Molecular , Mapeamento de Interação de Proteínas , Reação em Cadeia da Polimerase em Tempo Real , Reprodutibilidade dos Testes , Software , Transcrição Gênica
18.
Avian Pathol ; 43(5): 437-42, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25117822

RESUMO

Fowl glioma is characterized morphologically by multiple nodular astrocytic growth with disseminated non-suppurative encephalitis. The disease is caused by fowl glioma-inducing virus (FGV) and its variants, belonging to subgroup A of avian leukosis virus (ALV-A). Fifty-seven FGV variants have so far been isolated from Japanese fowls and these variants have a variable degree of glioma inducibility. However, how these ALVs induce glioma with different degrees and frequencies has not been fully elucidated. In this study, we investigated the relationship between intracerebral viral replication and astrocytic growth in the early infectious phase. Replication abilities of two ALV strains, Sp-53 (a FGV variant) and ALV-based replication-competent vector RCAS(A) without glioma inducibility, were compared in the brains of C/O specific pathogen free chickens at 35 days of age. Sp-53 replicated faster than RCAS(A), and the histological score and the level of interleukin (IL)-1ß in brains increased depending on the level of intracerebral viral RNA. Up-regulation of IL-1ß was also demonstrated in primary cultured astrocytes. These results suggest that the astrocytic growth in this phase is enhanced through the autocrine/paracrine production of IL-1ß in the FGV-infected astrocytes.


Assuntos
Astrócitos/fisiologia , Comunicação Autócrina/fisiologia , Vírus da Leucose Aviária/classificação , Glioma/veterinária , Interleucina-1beta/metabolismo , Comunicação Parácrina/fisiologia , Animais , Astrócitos/citologia , Leucose Aviária/metabolismo , Leucose Aviária/virologia , Linhagem Celular , Galinhas , Regulação da Expressão Gênica , Glioma/virologia , Interleucina-1beta/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Organismos Livres de Patógenos Específicos , Carga Viral , Replicação Viral
19.
PLoS One ; 9(4): e90878, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24694742

RESUMO

Avian leukosis is a neoplastic disease caused in part by subgroup J avian leukosis virus J (ALV-J). Micro ribonucleic acids (miRNAs) play pivotal oncogenic and tumour-suppressor roles in tumour development and progression. However, little is known about the potential role of miRNAs in avian leukosis tumours. We have found a novel tumour-suppressor miRNA, gga-miR-375, associated with avian leukosis tumorigenesis by miRNA microarray in a previous report. We have also previously studied the biological function of gga-miR-375; Overexpression of gga-miR-375 significantly inhibited DF-1 cell proliferation, and significantly reduced the expression of yes-associated protein 1 (YAP1) by repressing the activity of a luciferase reporter carrying the 3'-untranslated region of YAP1. This indicates that gga-miR-375 is frequently downregulated in avian leukosis by inhibiting cell proliferation through YAP1 oncogene targeting. Overexpression of gga-miR-375 markedly promoted serum starvation induced apoptosis, and there may be the reason why the tumour cycle is so long in the infected chickens. In vivo assays, gga-miR-375 was significantly downregulated in chicken livers 20 days after infection with ALV-J, and YAP1 was significantly upregulated 20 days after ALV-J infection (P<0.05). We also found that expression of cyclin E, an important regulator of cell cycle progression, was significantly upregulated (P<0.05). Drosophila inhibitor of apoptosis protein 1 (DIAP1), which is related to caspase-dependent apoptosis, was also significantly upregulated after infection. Our data suggests that gga-miR-375 may function as a tumour suppressor thereby regulating cancer cell proliferation and it plays a key role in avian leukosis tumorigenesis.


Assuntos
Vírus da Leucose Aviária/metabolismo , Leucose Aviária/metabolismo , Transformação Celular Viral , Genes Supressores de Tumor , MicroRNAs/biossíntese , RNA Neoplásico/metabolismo , Regiões 3' não Traduzidas/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Apoptose/genética , Leucose Aviária/genética , Vírus da Leucose Aviária/genética , Proteínas Aviárias/genética , Proteínas Aviárias/metabolismo , Células CHO , Embrião de Galinha , Galinhas , Cricetinae , Cricetulus , Fibroblastos , MicroRNAs/genética , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , RNA Neoplásico/genética
20.
J Virol ; 88(6): 3182-91, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24371071

RESUMO

UNLABELLED: The integration of retroviruses into the host genome following nonrandom genome-wide patterns may lead to the deregulation of gene expression and oncogene activation near the integration sites. Slow-transforming retroviruses have been widely used to perform genetic screens for the identification of genes involved in cancer. To investigate the involvement of avian leukosis virus subgroup J (ALV-J) integration in myeloid leukosis (ML) in chickens, we utilized an ALV-J insertional identification platform based on hybrid capture target enrichment and next-generation sequencing (NGS). Using high-definition mapping of the viral integration sites in the chicken genome, 241 unique insertion sites were obtained from six different ALV-J-induced ML samples. On the basis of previous statistical definitions, MYC, TERT, and ZIC1 genes were identified as common insertion sites (CIS) of provirus integration in tumor cells; these three genes have previously been shown to be involved in the malignant transformation of different human cell types. Compared to control samples, the expression levels of all three CIS genes were significantly upregulated in chicken ML samples. Furthermore, they were frequently, but not in all field ML cases, deregulated at the mRNA level as a result of ALV-J infection. Our findings contribute to the understanding of the relationship between multipathotypes associated with ALV-J infection and the molecular background of tumorigenesis. IMPORTANCE: ALV-Js have been successfully eradicated from chicken breeding flocks in the poultry industries of developed countries, and the control and eradication of ALV-J in China are now progressing steadily. To further study the pathogenesis of ALV-J infections, it will be necessary to elucidate the in vivo viral integration and tumorigenesis mechanism. In this study, 241 unique insertion sites were obtained from six different ALV-J-induced ML samples. In addition, MYC, TERT, and ZIC1 genes were identified as the CIS of ALV-J in tumor cells, which might be a putative "driver" for the activation of the oncogene. In addition, the CIS genes showed deregulated expression compared to nontumor samples. These results have potentially important implications for the mechanism of viral carcinogenesis.


Assuntos
Vírus da Leucose Aviária/fisiologia , Leucose Aviária/genética , Regulação da Expressão Gênica , Doenças das Aves Domésticas/genética , Proteínas Proto-Oncogênicas c-myc/genética , Telomerase/genética , Fatores de Transcrição/genética , Integração Viral , Animais , Leucose Aviária/metabolismo , Leucose Aviária/virologia , Vírus da Leucose Aviária/genética , Proteínas Aviárias/genética , Proteínas Aviárias/metabolismo , Galinhas , Dados de Sequência Molecular , Doenças das Aves Domésticas/metabolismo , Doenças das Aves Domésticas/virologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Telomerase/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...