Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 169
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Virol ; 97(8): e0026723, 2023 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-37582207

RESUMO

Avian leukemia virus subgroup J (ALV-J) causes various diseases associated with tumor formation and decreased fertility and induced immunosuppressive disease, resulting in significant economic losses in the poultry industry globally. Virus usually exploits the host cellular machinery for their replication. Although there are increasing evidences for the cellular proteins involving viral replication, the interaction between ALV-J and host proteins leading to the pivotal steps of viral life cycle are still unclear. Here, we reported that ribonucleoside-diphosphate reductase subunit M2 (RRM2) plays a critical role during ALV-J infection by interacting with capsid protein P27 and activating Wnt/ß-catenin signaling. We found that the expression of RRM2 is effectively increased during ALV-J infection, and that RRM2 facilitates ALV-J replication by interacting with viral capsid protein P27. Furthermore, ALV-J P27 activated Wnt/ß-catenin signaling by promoting ß-catenin entry into the nucleus, and RRM2 activated Wnt/ß-catenin signaling by enhancing its phosphorylation at Ser18 during ALV-J infection. These data suggest that the upregulation of RRM2 expression by ALV-J infection favors viral replication in host cells via activating Wnt/ß-catenin signaling. IMPORTANCE Our results revealed a novel mechanism by which RRM2 facilitates ALV-J growth. That is, the upregulation of RRM2 expression by ALV-J infection favors viral replication by interacting with capsid protein P27 and activating Wnt/ß-catenin pathway in host cells. Furthermore, the phosphorylation of serine at position 18 of RRM2 was verified to be the important factor regulating the activation of Wnt/ß-catenin signaling. This study provides insights for further studies of the molecular mechanism of ALV-J infection.


Assuntos
Vírus da Leucose Aviária , Leucose Aviária , Ribonucleosídeo Difosfato Redutase , Via de Sinalização Wnt , Animais , Vírus da Leucose Aviária/metabolismo , beta Catenina/metabolismo , Proteínas do Capsídeo/metabolismo , Galinhas , Ribonucleosídeo Difosfato Redutase/metabolismo
2.
J Virol ; 96(4): e0154921, 2022 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-34878920

RESUMO

Glycans on envelope glycoprotein (Env) of the subgroup J avian leukosis virus (ALV-J) play an essential role in the virion integrity and infection process. In this study, we found that, among the 13 predicted N-linked glycosylation sites (NGSs) in gp85 of Tibetan chicken strain TBC-J6, N17, and N193/N191 are pivotal for virus replication. Further research illustrated that a mutation at N193 weakened Env-receptor binding in a blocking assay of the viral entrance, coimmunoprecipitation, and ELISA. Our studies also showed that N17 was involved in Env protein processing and later virion incorporation based on the detection of p27 and Env protein in the supernatant and gp37 in the cell culture. This report is systematic research on clarifying the biological function of NGSs on ALV-J gp85, which would provide valuable insight into the role of gp85 in the ALV life cycle and anti-ALV-J strategies. IMPORTANCE ALV-J is a retrovirus that can cause multiple types of tumors in chickens. Among all the viral proteins, the heavily glycosylated envelope protein is especially crucial. Glycosylation plays a major role in Env protein function, including protein processing, receptor attachment, and immune evasion. Notably, viruses isolated recently seem to lose their 6th and 11th NGS, which proved to be important in receptor binding. In our study, the 1st (N17) and 8th (N193) NGS of gp85 of the strain TBC-J6 can largely influence the titer of this virus. Deglycosylation at N193 weakened Env-receptor binding while mutation at N17 influenced Env protein processing. This study systemically analyzed the function of NGSs in ALV-J in different aspects, which may help us to understand the life cycle of ALV-J and provide antiviral targets for the control of ALV-J.


Assuntos
Vírus da Leucose Aviária/metabolismo , Proteínas do Envelope Viral/metabolismo , Animais , Vírus da Leucose Aviária/crescimento & desenvolvimento , Linhagem Celular , Galinhas , Glicosilação , Mutação , Ligação Proteica , Processamento de Proteína Pós-Traducional , Receptores Virais/metabolismo , Proteínas do Envelope Viral/genética , Carga Viral/genética , Vírion/metabolismo
3.
J Virol ; 96(18): e0067822, 2022 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-36069550

RESUMO

The receptor of the subgroup A avian leukosis virus (ALV-A) in chicken is Tva, which is the homologous protein of human CD320 (huCD320), contains a low-density lipoprotein (LDL-A) module and is involved in the uptake of transcobalamin bound vitamin B12/cobalamin (Cbl). To map the functional determinants of Tva responsible for ALV-A receptor activity, a series of chimeric receptors were created by swapping the LDL-A module fragments between huCD320 and Tva. These chimeric receptors were then used for virus entry and binding assays to map the minimal ALV-A functional domain of Tva. The results showed that Tva residues 49 to 71 constituted the minimal functional domain that directly interacted with the ALV-A gp85 protein to mediate ALV-A entry. Single-residue substitution analysis revealed that L55 and W69, which were spatially adjacent on the surface of the Tva structure, were key residues that mediate ALV-A entry. Structural alignment results indicated that L55 and W69 substitutions did not affect the Tva protein structure but abolished the interaction force between Tva and gp85. Furthermore, substituting the corresponding residues of huCD320 with L55 and W69 of Tva converted huCD320 into a functional receptor of ALV-A. Importantly, soluble huCD320 harboring Tva L55 and W69 blocked ALV-A entry. Finally, we constructed a Tva gene-edited cell line with L55R and W69L substitutions that could fully resist ALV-A entry, while Cbl uptake was not affected. Collectively, our findings suggested that amino acids L55 and W69 of Tva were key for mediating virus entry. IMPORTANCE Retroviruses bind to cellular receptors through their envelope proteins, which is a crucial step in infection. While most retroviruses require two receptors for entry, ALV-A requires only one. Various Tva alleles conferring resistance to ALV-A, including Tvar1 (C40W substitution), Tvar2 (frame-shifting four-nucleotide insertion), Tvar3, Tvar4, Tvar5, and Tvar6 (deletion in the first intron), are known. However, the detailed entry mechanism of ALV-A in chickens remains to be explored. We demonstrated that Tva residues L55 and W69 were key for ALV-A entry and were important for correct interaction with ALV-A gp85. Soluble Tva and huCD320 harboring the Tva residues L55 and W69 effectively blocked ALV-A infection. Additionally, we constructed gene-edited cell lines targeting these two amino acids, which completely restricted ALV-A entry without affecting Cbl uptake. These findings contribute to a better understanding of the infection mechanism of ALV-A and provided novel insights into the prevention and control of ALV-A.


Assuntos
Aminoácidos , Vírus da Leucose Aviária , Aminoácidos/metabolismo , Animais , Leucose Aviária/virologia , Vírus da Leucose Aviária/metabolismo , Proteínas Aviárias/genética , Proteínas Aviárias/metabolismo , Galinhas/metabolismo , Humanos , Lipoproteínas LDL/metabolismo , Nucleotídeos/metabolismo , Receptores Virais/genética , Receptores Virais/metabolismo , Transcobalaminas/metabolismo , Vitamina B 12/metabolismo
4.
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
5.
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
6.
J Virol ; 92(1)2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29070685

RESUMO

Chicken Na+/H+ exchanger type I (chNHE1), a multispan transmembrane protein, is a cellular receptor of the subgroup J avian leukosis virus (ALV-J). To identify the functional determinants of chNHE1 responsible for the ALV-J receptor activity, a series of chimeric receptors was created by exchanging the extracellular loops (ECL) of human NHE1 (huNHE1) and chNHE1 and by ECL replacement with a hemagglutinin (HA) tag. These chimeric receptors then were used in binding and entry assays to map the minimal ALV-J gp85-binding domain of chNHE1. We show that ECL1 of chNHE1 (chECL1) is the critical functional ECL that interacts directly with ALV-J gp85; ECL3 is also involved in ALV-J gp85 binding. Amino acid residues 28 to 39 of the N-terminal membrane-proximal region of chECL1 constitute the minimal domain required for chNHE1 binding of ALV-J gp85. These residues are sufficient to mediate viral entry into ALV-J nonpermissive cells. Point mutation analysis revealed that A30, V33, W38, and E39 of chECL1 are the key residues mediating the binding between chNHE1 and ALV-J gp85. Further, the replacement of residues 28 to 39 of huNHE1 with the corresponding chNHE1 residues converted the nonfunctional ALV-J receptor huNHE1 to a functional one. Importantly, soluble chECL1 and huECL1 harboring chNHE1 residues 28 to 39 both could effectively block ALV-J infection. Collectively, our findings indicate that residues 28 to 39 of chNHE1 constitute a domain that is critical for receptor function and mediate ALV-J entry.IMPORTANCE chNHE1 is a cellular receptor of ALV-J, a retrovirus that causes infections in chickens and serious economic losses in the poultry industry. Until now, the domains determining the chNHE1 receptor function remained unknown. We demonstrate that chECL1 is critical for receptor function, with residues 28 to 39 constituting the minimal functional domain responsible for chNHE1 binding of ALV-J gp85 and efficiently mediating ALV-J cell entry. These residues are located in the membrane-proximal region of the N terminus of chECL1, suggesting that the binding site of ALV-J gp85 on chNHE1 is probably located on the apex of the molecule; the receptor-binding mode might be different from that of retroviruses. We also found that soluble chECL1, as well as huECL1 harboring chNHE1 residues 28 to 39, effectively blocked ALV-J infection. These findings contribute to a better understanding of the ALV-J infection mechanism and also provide new insights into the control strategies for ALV-J infection.


Assuntos
Aminoácidos/química , Vírus da Leucose Aviária/metabolismo , Receptores Virais/metabolismo , Trocadores de Sódio-Hidrogênio/química , Trocadores de Sódio-Hidrogênio/metabolismo , Ligação Viral , Internalização do Vírus , Aminoácidos/metabolismo , Animais , Leucose Aviária/virologia , Vírus da Leucose Aviária/química , Vírus da Leucose Aviária/genética , Galinhas , Humanos , Mutação Puntual , Receptores Virais/genética , Trocadores de Sódio-Hidrogênio/genética
7.
Curr Top Microbiol Immunol ; 407: 65-82, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28688086

RESUMO

The surface envelope protein of any virus is major determinant of the host cell that is infected and as a result a major determinant of viral pathogenesis. Retroviruses have a single surface protein named Env. It is a trimer of heterodimers and is responsible for binding to the host cell receptor and mediating fusion between the viral and host membranes. In this review we will discuss the history of the discovery of the avian leukosis virus (ALV) and human immunodeficiency virus type 1 (HIV-1) Env proteins and their receptor specificity, comparing the many differences but having some similarities. Much of the progress in these fields has relied on viral genetics and genetic polymorphisms in the host population. A special feature of HIV-1 is that its persistent infection in its human host, to the point of depleting its favorite target cells, allows the virus to evolve new entry phenotypes to expand its host range into several new cell types. This variety of entry phenotypes has led to confusion in the field leading to the major form of entry phenotype of HIV-1 being overlooked until recently. Thus an important part of this story is the description and naming of the most abundant entry form of the virus: R5 T cell-tropic HIV-1.


Assuntos
Vírus da Leucose Aviária/genética , Genes env/genética , HIV-1/genética , Animais , Vírus da Leucose Aviária/metabolismo , HIV-1/metabolismo , Humanos , Receptores Virais/metabolismo , Linfócitos T/metabolismo , Linfócitos T/virologia
8.
Proc Natl Acad Sci U S A ; 112(32): 9860-5, 2015 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-26216971

RESUMO

Antibody-based therapeutics have now had success in the clinic. The affinity and specificity of the antibody for the target ligand determines the specificity of therapeutic delivery and off-target side effects. The discovery and optimization of high-affinity antibodies to important therapeutic targets could be significantly improved by the availability of a robust, eukaryotic display technology comparable to phage display that would overcome the protein translation limitations of microorganisms. The use of eukaryotic cells would improve the diversity of the displayed antibodies that can be screened and optimized as well as more seamlessly transition into a large-scale mammalian expression system for clinical production. In this study, we demonstrate that the replication and polypeptide display characteristics of a eukaryotic retrovirus, avian leukosis virus (ALV), offers a robust, eukaryotic version of bacteriophage display. The binding affinity of a model single-chain Fv antibody was optimized by using ALV display, improving affinity >2,000-fold, from micromolar to picomolar levels. We believe ALV display provides an extension to antibody display on microorganisms and offers virus and cell display platforms in a eukaryotic expression system. ALV display should enable an improvement in the diversity of properly processed and functional antibody variants that can be screened and affinity-optimized to improve promising antibody candidates.


Assuntos
Anticorpos/metabolismo , Vírus da Leucose Aviária/metabolismo , Técnicas de Visualização da Superfície Celular/métodos , Células Eucarióticas/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Galinhas , Regiões Determinantes de Complementaridade , Glicoproteínas/metabolismo , Humanos , Cinética , Laminina/metabolismo , Dados de Sequência Molecular , Mutagênese , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Ligação Proteica , Proteínas Recombinantes de Fusão/metabolismo , Anticorpos de Cadeia Única/metabolismo , Proteínas do Envelope Viral/metabolismo , Vírion/metabolismo , Replicação Viral
9.
Poult Sci ; 94(4): 668-72, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25713393

RESUMO

The diagnosis of avian leukosis virus subgroup J (ALV-J) infection in Chinese Partridge Shank chickens was confirmed by necropsy, histopathological examinations, antibody tests, viral isolation, immunofluorescence assays, and sequence analysis. Myelocytoma, myeloma, and fibrosarcoma were simultaneously found in Partridge Shank flock with ALV-J infection. Sequence analysis of the env genes of ALV-J demonstrated that both gp85 and gp37 were highly homologous among the three strains from local chickens of those among ALV-J strains isolated from white meat-type chickens. The phylogenetic trees indicated that the three strains isolated in this study were closely related to reference strains isolated in so-called Chinese yellow chickens and some strains isolated from white meat-type chickens, both from the USA and China. The observed ALV-J infection was the first report on Partridge Shank chickens, and myelocytoma, myeloma, and fibrosarcoma were found at the same time in this batch of local chickens.


Assuntos
Vírus da Leucose Aviária/genética , Leucose Aviária/diagnóstico , Galinhas , Doenças das Aves Domésticas/diagnóstico , Animais , Leucose Aviária/epidemiologia , Leucose Aviária/virologia , Vírus da Leucose Aviária/metabolismo , China/epidemiologia , Fibrossarcoma/epidemiologia , Fibrossarcoma/veterinária , Fibrossarcoma/virologia , Incidência , Dados de Sequência Molecular , Mieloma Múltiplo/epidemiologia , Mieloma Múltiplo/veterinária , Mieloma Múltiplo/virologia , Filogenia , Doenças das Aves Domésticas/epidemiologia , Doenças das Aves Domésticas/virologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa/veterinária , Análise de Sequência de DNA/veterinária , Especificidade da Espécie
10.
Arch Virol ; 159(7): 1821-6, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24488027

RESUMO

Avian leukosis virus subgroup J (ALV-J), first isolated in 1989, preferentially infects meat-type birds. Chinese layer flocks have experienced outbreaks of this virus since 2008. To analyze the status of ALV-J infection in wild birds in China, 585 wild birds collected from three provinces of Northeast China from 2010 to 2012 were tested, and six ALV-J strains were isolated for the first time. Furthermore, the gp85 genes of the six strains were amplified, cloned, and sequenced. The results indicated that two different ALV-J strains coexisted in Chinese wild birds from 2010 to 2012. These results not only expand the epidemiological data available for ALV-J and provide necessary information for the further understanding of the evolution of ALV-J, but they also highlight the potential role of wild-bird migration in the spread of ALV-J.


Assuntos
Vírus da Leucose Aviária/genética , Vírus da Leucose Aviária/metabolismo , Leucose Aviária/virologia , Variação Genética , Proteínas do Envelope Viral/genética , Animais , Animais Selvagens , Leucose Aviária/epidemiologia , Aves , China/epidemiologia , Dados de Sequência Molecular , Filogenia
11.
Avian Dis ; 58(1): 34-8, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24758110

RESUMO

Endogenous retroviral elements (ERVs) are prolific components of the genomes of complex species, typically occupying more sequence space than do essential, protein-encoding genes. Much of what we know today about the structure and function, as well as the evolution and pathogenic potential, of ERVs was fleshed out over several decades during the last century using the avian leukosis virus subgroup E-related (ALVE) family of endogenous retroviruses of chickens as a model system. A critical enabling factor in the elucidation of ALVE structure and function is the ability to detect and unambiguously identify specific ALVE proviral elements and to develop accurate element profiles for individual chickens under study. Currently, the most common approach for ALVE locus detection involves element-specific PCR assays carried out using primers that target host DNA near the insertion site of the provirus (i.e., the upstream and downstream flanks of the unoccupied site). Here we describe a new approach for proviral detection that exploits restriction enzyme sites in flanking DNA to develop ALVE element profiles more rapidly than with assays currently in use. Moreover, unlike element-specific PCR tests, the "profiling" assay detects novel ALVEs for which insertion sites have not yet been identified as well as previously characterized elements.


Assuntos
Vírus da Leucose Aviária/isolamento & purificação , Leucose Aviária/virologia , Galinhas , Doenças das Aves Domésticas/virologia , Provírus/isolamento & purificação , Mapeamento por Restrição/métodos , Animais , Vírus da Leucose Aviária/genética , Vírus da Leucose Aviária/metabolismo , Enzimas de Restrição do DNA/genética , Enzimas de Restrição do DNA/isolamento & purificação , Enzimas de Restrição do DNA/metabolismo , DNA Viral/genética , DNA Viral/metabolismo , Dados de Sequência Molecular , Provírus/genética , Provírus/metabolismo , Mapeamento por Restrição/veterinária , Análise de Sequência de DNA/veterinária
12.
Vet Microbiol ; 291: 110012, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38387235

RESUMO

The ubiquitin-binding enzyme E2J1 is located on the endoplasmic reticulum membrane. It plays a role in transport throughout the process of ubiquitination. In mammals, UBE2J1 can promote RNA virus replication. However, the biological function of chicken UBE2J1 is unclear. In this study, chicken UBE2J1 was cloned for the first time, and UBE2J1 overexpression and shRNA knockdown plasmids were constructed. In chicken embryo fibroblasts, overexpression of UBE2J1 promoted the replication of subtype A avian leukosis virus, while knockdown of UBE2J1 inhibited the replication of ALV-A virus. In addition, we divided virus replication into virus adsorption and invasion into DF-1 cells, synthesis of proviral DNA, and release of viral particles. UBE2J1 promoted the replication of ALV-A virus by promoting the synthesis of proviral DNA. This result was caused by UBE2J1 inhibiting the production of interferon by inhibiting the STAT3/IRF1 pathway. We mutated ser at position 184 of UBE2J1 to Gly and found that this site plays a role as the phosphorylation site of UBE2J1. We confirmed that UBE2J1 promotes ALV-A replication in chicken embryo fibroblasts by inhibiting the STAT3/IRF1 pathway. This study provides new ideas and insights into ubiquitin-related proteins and antiviral immunity.


Assuntos
Vírus da Leucose Aviária , Leucose Aviária , Animais , Embrião de Galinha , Vírus da Leucose Aviária/genética , Vírus da Leucose Aviária/metabolismo , Galinhas , Mamíferos , Provírus , Transdução de Sinais , Ubiquitinas , Fator de Transcrição STAT3/metabolismo , Fatores Reguladores de Interferon/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo
13.
Avian Dis ; 57(2): 172-7, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24689170

RESUMO

Avian leukosis virus (ALV) infection in Taiwan Country chickens (TCCs) was investigated by using gene detection, virus isolation, and sequence analysis. The blood samples of 61 TCC flocks at market ages from a slaughter house were screened for exogenous ALVs using polymerase chain reaction to investigate the ALV infection status. The buffy coats from three breeder and four commercial chicken flocks were cocultured with DF-1 cells to isolate the virus. The full proviral DNA genomes of two ALV isolates were sequenced, analyzed, and compared with reference ALV strains. The gene detection results showed that 60 and 43 of the 61 flocks were infected with subgroup A of ALV (ALV-A) and subgroup J of ALV (ALV-J), respectively. Virus isolation results showed that five ALV-As and two ALV-Js were isolated from those seven TCC flocks. The full sequences of the isolates showed that isolate TW-3577 possessed a myeloblastosis-associated virus 1 gp85 coding region and an ALV-J 3'-untranslated region (3'UTR) and was similar to ordinary ALV-A. However, TW-3593 was unique. The 3'UTR of this isolate displayed high identity to endogenous counterpart sequence and its gp85 was different from all subgroups. This unique ALV is common in Taiwan.


Assuntos
Vírus da Leucose Aviária/genética , Leucose Aviária/epidemiologia , Galinhas , Doenças das Aves Domésticas/epidemiologia , Proteínas Virais/genética , Animais , Leucose Aviária/virologia , Vírus da Leucose Aviária/classificação , Vírus da Leucose Aviária/isolamento & purificação , Vírus da Leucose Aviária/metabolismo , Feminino , Humanos , Masculino , Dados de Sequência Molecular , Filogenia , Doenças das Aves Domésticas/virologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa/veterinária , Análise de Sequência de DNA/veterinária , Análise de Sequência de Proteína/veterinária , Taiwan/epidemiologia , Proteínas Virais/química , Proteínas Virais/metabolismo
14.
Avian Pathol ; 41(1): 11-9, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22845317

RESUMO

The present review is aimed at the non-specialist reader and is one of a number being written on important diseases of poultry to celebrate the 40th anniversary of the birth of Avian Pathology, the journal of the World Veterinary Poultry Association. The diseases of the avian leukosis complex have a number of features of distinction. They were the first neoplastic diseases in any species to be shown, 100 years ago, to be transmissible and caused by viruses, and have consequently been studied extensively by biomedical scientists as models for the role of viruses in cancer. They also became, from around the 1920s, the major cause of mortality and economic loss to the developed poultry industry, and were studied by agricultural scientists searching to understand and control them. The remit of the review is to cover research carried out over the 40 years since 1971, when the journal was founded. In this review on avian leukosis, an introductory summary is given of knowledge acquired over the preceding 60 years. Subsequently a selection is provided of discoveries, both fundamental and more applied, that seem to us to be of particular importance and interest. Much of the work was carried out by biomedical scientists interested in cancer. Probably the most significant was the discovery in the avian retroviruses of oncogenes that cause leukosis and other tumours and of their origin from proto-oncogenes in normal cells. These oncogenes are involved in cancer in many species, including chickens and humans. Other work was performed by agricultural scientists interested in poultry disease. Interests of the two groups have overlapped, particularly as a result of a shift of emphasis to molecular biology research.


Assuntos
Vírus da Leucose Aviária/genética , Leucose Aviária/patologia , Leucose Aviária/virologia , Pesquisa/história , Proteínas dos Retroviridae/genética , Animais , Vírus da Leucose Aviária/metabolismo , História do Século XX , História do Século XXI , Aves Domésticas
15.
Virol J ; 8: 556, 2011 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-22188662

RESUMO

BACKGROUND: Avian leukosis virus (ALV) is a major infectious disease that impacts the poultry industry worldwide. Despite intensive efforts, no effective vaccine has been developed against ALV because of mutations that lead to resistant forms. Therefore, there is a dire need to develop antiviral agents for the treatment of ALV infections and RNA interference (RNAi) is considered an effective antiviral strategy. RESULTS: In this study, the avian leukosis virus subgroup J (ALV-J) proviral genome, including the gag genes, were treated as targets for RNAi. Four pairs of miRNA sequences were designed and synthesized that targeted different regions of the gag gene. The screened target (i.e., the gag genes) was shown to effectively suppress the replication of ALV-J by 19.0-77.3%. To avoid the generation of escape variants during virus infection, expression vectors of multi-target miRNAs were constructed using the multi-target serial strategy (against different regions of the gag, pol, and env genes). Multi-target miRNAs were shown to play a synergistic role in the inhibition of ALV-J replication, with an inhibition efficiency of viral replication ranging from 85.0-91.2%. CONCLUSION: The strategy of multi-target miRNAs might be an effective method for inhibiting ALV replication and the acquisition of resistant mutations.


Assuntos
Vírus da Leucose Aviária/efeitos dos fármacos , Vírus da Leucose Aviária/metabolismo , MicroRNAs/farmacologia , Interferência de RNA , Replicação Viral/efeitos dos fármacos , Animais , Leucose Aviária/virologia , Vírus da Leucose Aviária/genética , Vírus da Leucose Aviária/fisiologia , Linhagem Celular , Produtos do Gene env/genética , Produtos do Gene env/metabolismo , Produtos do Gene gag/genética , Produtos do Gene gag/metabolismo , Produtos do Gene pol/genética , Produtos do Gene pol/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Doenças das Aves Domésticas/virologia , Transfecção
16.
Viruses ; 13(10)2021 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-34696398

RESUMO

Wnt/ß-catenin signaling is a highly conserved pathway related to a variety of biological processes in different cells. The regulation of replication of various viruses by Wnt/ß-catenin signaling pathway has been reported. However, the interaction between the Wnt/ß-catenin pathway and avian leukosis virus is unknown. In the present study, we investigated the effect of modulating the Wnt/ß-catenin pathway during avian leukosis virus subgroup J (ALV-J) infection. The activation of the Wnt/ß-catenin pathway by GSK-3 inhibitor increased ALV-J mRNA, viral protein expression, and virus production in CEF cells. This increase was suppressed by iCRT14, one of the specific inhibitors of the Wnt/ß-catenin signaling pathway. Moreover, treatment with iCRT14 reduced virus titer and viral gene expression significantly in CEF and LMH cells in a dose-dependent manner. Inhibition Wnt/ß-catenin signaling pathway by knockdown of ß-catenin reduced virus proliferation in CEF cells also. Collectively, these results suggested that the status of Wnt/ß-catenin signaling pathway modulated ALV-J replication. These studies extend our understanding of the role of Wnt/ß-catenin signaling pathway in ALV-J replication and make a new contribution to understanding the virus-host interactions of avian leukosis virus.


Assuntos
Vírus da Leucose Aviária/metabolismo , Replicação Viral/fisiologia , Via de Sinalização Wnt/fisiologia , Animais , Leucose Aviária/virologia , Linhagem Celular , Embrião de Galinha , Galinhas/virologia , China , Expressão Gênica/genética , Regulação Viral da Expressão Gênica/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Doenças das Aves Domésticas/virologia , Piridinas/farmacologia , Pirróis/farmacologia , RNA Mensageiro , Tiazolidinedionas/farmacologia
17.
Trop Anim Health Prod ; 42(8): 1829-36, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20640887

RESUMO

Avian leukosis viruses (ALVs) belong to Alpharetrovirus genus of the family Retroviridae that are widespread in nature. Different subgroups of ALV commonly infect egg-laying hens. They are responsible for economic losses due to both mortality and depressed performance in chickens. To investigate the presence of these viruses in chickens in Iran, 560 egg albumens were selected from different farms of Fars province, Iran. These eggs were obtained from flocks of two research centers of native fowl production (60 eggs), a broiler grandparent farm (100 eggs), three broiler breeder farms (300 eggs), and a commercial layer flock (100 eggs). Firstly, for primary screening a degenerative primer set (PU1 and PU2) were used in reverse transcriptase-polymerase chain reaction (RT-PCR). Positive cases were detected in 47 of 300 (15.7%) samples from three broiler breeders, 40 of 100 (40%) samples from commercial layer, 53 of 60 (88.3%) samples from flocks of two research centers of native fowl production, and none from the samples of broiler grandparent. Then RT-PCR was undertaken with primers PA1 and PA2 on the positive samples. RT-PCR analysis detected ALVs in two of 47 (4.3%) samples from three broiler breeders, 13 of 40 (32.5%) samples from commercial layer, and 19 of 53 (35.8%) samples from flocks of two research centers of native fowl production. The sequencing results showed that subgroup E of ALV was the most detected virus among chicken eggs and subgroup B was more prevalent in the eggs of native fowls. This is the first report of the ALV subgroup B and E in egg albumen in Iran.


Assuntos
Vírus da Leucose Aviária/genética , Leucose Aviária/diagnóstico , Leucose Aviária/epidemiologia , Leucose Aviária/virologia , Ovos/virologia , Filogenia , Albuminas/análise , Animais , Vírus da Leucose Aviária/classificação , Vírus da Leucose Aviária/metabolismo , Sequência de Bases , Análise por Conglomerados , Primers do DNA/genética , Irã (Geográfico)/epidemiologia , Dados de Sequência Molecular , Aves Domésticas , Reação em Cadeia da Polimerase Via Transcriptase Reversa/veterinária , Alinhamento de Sequência , Análise de Sequência de DNA/veterinária
18.
Cell Death Dis ; 11(8): 684, 2020 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-32826872

RESUMO

Autophagy and apoptosis, which are important processes for host immunity, are commonly exploited by viruses to facilitate their survival. However, to the best of our knowledge, very few studies have researched the mechanisms of action of the autophagic and apoptotic signaling pathways following viral infection. Thus, the present study aimed to investigate the mechanisms of action of growth arrest and DNA-damage-inducible ß (GADD45ß), an important resistance gene involved in the host resistance to ALV-J. Both ALV-J infection and the overexpression of GADD45ß inhibited autophagy during the early stages, which prevented the autophagosomes from binding to the lysosomes and resulted in an incomplete autophagic flux. Notably, GADD45ß was discovered to interact with MEKK4 in DF-1 cells. The genetic knockdown of GADD45ß and MEKK4 using small interfering RNA-affected ALV-J infection, which suggested that ALV-J may promote the binding of GADD45ß to MEKK4 to activate the p38MAPK signaling pathway, which subsequently inhibits autophagy. Furthermore, ALV-J was revealed to affect the autophagic pathway prior to affecting the apoptotic pathway. In conclusion, to the best of our knowledge, the present study was the first to investigate the combined effects of ALV-J infection on autophagy and apoptosis, and to suggest that ALV-J inhibits autophagy via the GADD45ß/MEKK4/p38MAPK signaling pathway.


Assuntos
Antígenos de Diferenciação/metabolismo , Autofagia/fisiologia , Vírus da Leucose Aviária/metabolismo , Animais , Apoptose/fisiologia , Vírus da Leucose Aviária/genética , Vírus da Leucose Aviária/patogenicidade , Linhagem Celular , Embrião de Galinha , Galinhas/genética , Interações Hospedeiro-Patógeno/fisiologia , MAP Quinase Quinase Quinase 4/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
19.
J Virol ; 82(16): 7818-27, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18550662

RESUMO

Unmethylated CpG islands are known to keep adjacent promoters transcriptionally active. In the CpG island adjacent to the adenosine phosphoribosyltransferase gene, the protection against transcriptional silencing can be attributed to the short CpG-rich core element containing Sp1 binding sites. We report here the insertion of this CpG island core element, IE, into the long terminal repeat of a retroviral vector derived from Rous sarcoma virus, which normally suffers from progressive transcriptional silencing in mammalian cells. IE insertion into a specific position between enhancer and promoter sequences led to efficient protection of the integrated vector from silencing and gradual CpG methylation in rodent and human cells. Individual cell clones with IE-modified reporter vectors display high levels of reporter expression for a sustained period and without substantial variegation in the cell culture. The presence of Sp1 binding sites is important for the protective effect of IE, but at least some part of the entire antisilencing capacity is maintained in IE with mutated Sp1 sites. We suggest that this strategy of antisilencing protection by the CpG island core element may prove generally useful in retroviral vectors.


Assuntos
Vírus da Leucose Aviária/metabolismo , Ilhas de CpG , Inativação Gênica , Sarcoma Aviário/genética , Sarcoma Aviário/virologia , Transcrição Gênica , Animais , Sítios de Ligação , Aves , Citometria de Fluxo , Genes Reporter , Humanos , Modelos Biológicos , Mutação , Vírus do Sarcoma de Rous/metabolismo , Fator de Transcrição Sp1/metabolismo , Sequências Repetidas Terminais
20.
Science ; 225(4660): 417-9, 1984 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-6330893

RESUMO

RAV-0, an endogenous avian leukosis virus, does not undergo congenital transmission in infected K28 chickens. In contrast, avian leukosis viruses of exogenous origin undergo highly efficient congenital transmission. The relative abilities of endogenous and exogenous viruses to undergo congenital transmission appear to be determined by the p27 capsid proteins of these viruses.


Assuntos
Leucose Aviária/congênito , Animais , Leucose Aviária/transmissão , Vírus da Leucose Aviária/metabolismo , Capsídeo/metabolismo , Galinhas , Ovos , Feminino , Ovalbumina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA