Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 287
Filtrar
1.
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
2.
J Gen Virol ; 103(2)2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35130137

RESUMO

Avian leukosis virus (ALV) is a retrovirus that induces tumours in infected birds; ALV is divided into different subgroups according to the env gene and cellular tropism. In general, ALV subgroup J (ALV-J) is considered to be the most pathogenic and prevalent subgroup while subgroup K (ALV-K), a newly identified subgroup, only causes mild symptoms. To illuminate the roles of the env viral gene and LTR sequence in pathogenic differences between ALV-J and ALV-K, rescued ALV-J strain rSDAU1005, rescued ALV-K strain rJS11C1, and recombinant strains rENV(J)-LTR(K) and rENV(K)-LTR(J) were characterized and investigated in this study. Among rescued viruses, rSDAU1005 had the highest replication efficiency while rJS11C1 replicated the slowest (replication efficiency rankings were rSDAU1005 >rENV(K)-LTR(J)>rENV(J)-LTR(K)>rJS11 C1). The luciferase reporter gene assay results showed that the promoter activity of ALV-K LTR was lower than that of the ALV-J LTR promoter, which may have accounted for the slower replication efficiency of ALV-K. Pathogenicity of the four rescued viruses was determined via inoculating the yolk sacs of specific-pathogen-free chickens. The results demonstrated that all four viruses were pathogenic; rSDAU1005 caused the most severe growth retardation and immunosuppression. rENV(J)-LTR(K) was more pathogenic when compared to rENV(K)-LTR(J), indicating that env and the LTR sequence play important roles in pathogenicity between ALV-K and ALV-J. Additionally, env seemed to especially play a role in ALV-K pathogenesis. This study provided scientific data and insight to improve detection methods and judgement criteria in ALV clearance and surveillance.


Assuntos
Vírus da Leucose Aviária/genética , Leucose Aviária/virologia , Genes env , Proteínas do Envelope Viral/genética , Animais , Aves
3.
Vet Microbiol ; 264: 109293, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34883334

RESUMO

Control of ALV-J in breed of chicken is still a serious issue that need more attention to be paid. Vertical transmission of ALV-J often give rise to more adverse pathogenicity. However, the way to elimination of ALV-J underlying vertical transmission remains not-well understood. In addition, effective vaccines or drugs have not been developed to prevent and control the transmission of ALV-J so far. CD81, a member of the tetraspanins superfamily, plays important roles in regulating membrane proteins, facilitating cells adhesion or fusion, and also participates in viral infection. The purpose of this study was to investigate whether antibodies against certain tetraspanins affect infection of ALV-J. Here, we showed that anti-CD81 antibody could inhibit viral RNA and protein level. We also found that anti-CD81 antibody interacts with viral protein p27, p32 and gp37. Moreover, treatment with antibody to CD81 can effectively prevent the vertical transmission of ALV-J in animal model. Collectively, current study provides new avenues for the control of ALV-J transmission.


Assuntos
Anticorpos , Vírus da Leucose Aviária , Leucose Aviária , Doenças das Aves Domésticas , Animais , Anticorpos/metabolismo , Leucose Aviária/prevenção & controle , Leucose Aviária/transmissão , Leucose Aviária/virologia , Vírus da Leucose Aviária/classificação , Galinhas , Transmissão Vertical de Doenças Infecciosas , Doenças das Aves Domésticas/prevenção & controle , Doenças das Aves Domésticas/transmissão , Doenças das Aves Domésticas/virologia , Tetraspanina 28/imunologia
4.
Vet Microbiol ; 264: 109278, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34808431

RESUMO

MicroRNAs (miRNAs) involved host-virus interaction, affecting the replication or pathogenesis of several viruses. Although avian leukosis virus subgroup J (ALV-J) has been one of the most studied avian viruses, the effects of various host miRNAs on ALV-J infection and its underlying molecular mechanisms are still unclear. Here, we reported that gga-miR-200b-3p acts as a positive host factor enhancing ALV-J replication. We found that gga-miR-200b-3p was increased in response to ALV-J infection in host cells, and that gga-miR-200b-3p effectively enhanced ALV-J replication via targeting host protein dual-specificity phosphatase 1 (DUSP1). Collectively, these findings highlight a crucial role of gga-miR-200b-3p in ALV-J replication.


Assuntos
Vírus da Leucose Aviária , Leucose Aviária , Fosfatases de Especificidade Dupla , Interações entre Hospedeiro e Microrganismos , MicroRNAs , Replicação Viral , Animais , Leucose Aviária/patologia , Leucose Aviária/virologia , Vírus da Leucose Aviária/enzimologia , Vírus da Leucose Aviária/genética , Galinhas , Fosfatases de Especificidade Dupla/metabolismo , Interações entre Hospedeiro e Microrganismos/fisiologia , MicroRNAs/genética , MicroRNAs/metabolismo , Replicação Viral/genética
5.
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
6.
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
8.
Vet Microbiol ; 261: 109205, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34391195

RESUMO

Based on the RNA-seq data of chicken spleen tissues infected with J subgroup avian leukosis virus (ALV-J), we found that prolactin (PRL) gene was one of differentially expressed gene. We measured ALV-J viremia and PRL levels in the plasma of two groups of ALV-J-infected adult chickens. Furthermore, recombinant chicken PRL (cPRL) was used to assess how cPRL affects ALV-J virus replication both in vivo and in vitro. The results showed that PRL levels in the plasma of adult chickens infected with ALV-J were lower than those of uninfected chickens, and that the difference was more significant in the avian leukemia pathological apparent changes. Notably, the fluctuations in PRL levels might influence the disappearance of ALV-J viremia in chickens. The in vitro results showed that preincubating DF-1 cells with cPRL before ALV-J infection elicited the best antiviral effects. Moreover, these effects were not dose-dependent. in vivo, injection of cPRL into ALV-J-infected chicks could reduce the levels of viremia at the 14 days post infection (dpi). Additionally, the expression of the interferon-stimulated genes oligoadenylate synthetase-like (OSAL) and vasoactive intestinal peptide (VIP) increased, and that of the proinflammatory cytokine-encoding TNTα, IL-1ß, and IL-6 genes decreased in the spleens of ALV-J-infected chicks injected with cPRL, leading to inhibition of viral replication at the 7 dpi. Collectively, our data demonstrated that PRL plays an important antiviral role in the immune response to ALV-J infection. This is the first report of the relationship between ALV-J infection and PRL. It is of great significance for the prevention and control of ALV-J.


Assuntos
Leucose Aviária/virologia , Prolactina/genética , Baço/imunologia , Baço/virologia , Viremia/veterinária , Animais , Leucose Aviária/sangue , Leucose Aviária/prevenção & controle , Vírus da Leucose Aviária/imunologia , Linhagem Celular , Galinhas , Regulação da Expressão Gênica , Prolactina/sangue , Transcriptoma
9.
Vet Microbiol ; 260: 109184, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34311270

RESUMO

Avian leukosis virus (ALV) can induce various tumors and cause serious production problems. ALVs isolated from chickens were divided into six subgroups (A-J). In 2012, a strain of a putative novel subgroup of ALVs was isolated from Chinese native chickens in Jiangsu Province and named as ALV-K. In this study, three ALV-K strains (JS14LH01, JS13LH14, and JS15SG01) were isolated from chickens with suspected ALV infection in Jiangsu Province. Their complete genomes were amplified, sequenced, and analyzed systematically. The results showed that JS14LH01 and JS13LH14 were ALV-K and ALV-E recombinant strains. Whereas JS15SG01 is an ALV-K, ALV-E, and ALV-J multiple recombinant strain containing the U3 region of ALV-J. The pathogenicity test of JS15SG01 revealed that, compared with previous ALV-K strains, the viremia and viral shedding level of JS15SG01-infected chickens were significantly increased, reaching 100 % and 59 %, respectively. More important, JS15SG01 induced significant proliferation of gliocytes in the cerebral cortex of infected chickens, accompanied by the neurotropic phenomenon. This is the first report about a multiple recombinant ALV-K strain that could invade and injure the brain tissue of chickens in China. Our findings enriched the epidemiologic data of ALV and helped to reveal the evolution of ALV strains prevalent in chicken fields.


Assuntos
Vírus da Leucose Aviária/genética , Leucose Aviária/virologia , Galinhas/virologia , Recombinação Genética , Animais , Leucose Aviária/epidemiologia , Vírus da Leucose Aviária/isolamento & purificação , Vírus da Leucose Aviária/patogenicidade , China
10.
FEMS Microbiol Lett ; 368(10)2021 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-34048535

RESUMO

Chicken is one of the economically important poultry species. Avian leucosis virus subgroup J (ALV-J) has emerged as a serious cause of mortality and suboptimal performance of domestic chickens. Changes in virome may contribute to pathogenesis. Thus, it is important to investigate the effects of ALV-J infection on the composition of the virome in chicken. In the study metagenomic sequencing was used to characterize the virome of feces collected from the AVL-J infected chickens and the controls. Our results indicated that the chicken gut virome contained a diverse range of viruses that can be found in mammal, reptile, fish, and frogs. Furthermore, at the order, family and genus levels, AVL-J infection significantly altered the chicken gut virome composition. The predominant order was Herpesvirales, accounting for more than 96% of the chicken gut virome. Furthermore, the relative abundance of Caudovirales in the controls was higher than that in the AVL-J-infected chickens. At the family level, the relative abundance of Herpesviridae, Myoviridae, Alloherpesviridae, and Genomoviridae was significantly altered in the AVL-J-infected chickens compared with that in the controls. Additionally, the relative abundance of 15 genera showed a significant difference between the AVL-J-infected chickens and controls. These results will increase our understanding of the viral diversity and changes in the virome of chicken gut, with implications in chicken health.


Assuntos
Vírus da Leucose Aviária/fisiologia , Leucose Aviária/virologia , Trato Gastrointestinal/virologia , Doenças das Aves Domésticas/virologia , Animais , Vírus da Leucose Aviária/classificação , Vírus da Leucose Aviária/genética , Vírus da Leucose Aviária/isolamento & purificação , Galinhas/virologia , Fezes/virologia , Feminino , Masculino , Viroma , Vírus/classificação , Vírus/genética , Vírus/isolamento & purificação
11.
Sci Rep ; 11(1): 4797, 2021 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-33637946

RESUMO

Avian leukosis caused by avian leukosis virus (ALV) is one of the most severe diseases endangering the poultry industry. When the eradication measures performed in commercial broilers and layers have achieved excellent results, ALV in some local chickens has gradually attracted attention. Since late 2018, following the re-outbreak of ALV-J in white feather broilers in China, AL-like symptoms also suddenly broke out in some local flocks, leading to great economic losses. In this study, a systematic epidemiological survey was carried out in eight local chicken flocks in Jiangxi Province, China, and 71 strains were finally isolated from 560 samples, with the env sequences of them being successfully sequenced. All of those new isolates belong to subgroup J but they have different molecular features and were very different from the strains that emerged in white feature broilers recently, with some strains being highly consistent with those previously isolated from commercial broilers, layers and other flocks or even isolated from USA and Russian, suggesting these local chickens have been acted as reservoirs to accumulate various ALV-J strains for a long time. More seriously, phylogenetic analysis shows that there were also many novel strains emerging and in a separate evolutionary branch, indicating several new mutated ALVs are being bred in local chickens. Besides, ALV-J strains isolated in this study can be further divided into ten groups, while there were more or fewer groups in different chickens, revealing that ALV may cross propagate in those flocks. The above analyses explain the complex background and future evolution trend of ALV-J in Chinese local chickens, providing theoretical support for the establishment of corresponding prevention and control measures.


Assuntos
Vírus da Leucose Aviária/genética , Leucose Aviária/virologia , Galinhas/virologia , Doenças das Aves Domésticas/virologia , Animais , Leucose Aviária/epidemiologia , Leucose Aviária/patologia , Vírus da Leucose Aviária/isolamento & purificação , China/epidemiologia , Variação Genética , Filogenia , Doenças das Aves Domésticas/epidemiologia , Doenças das Aves Domésticas/patologia
12.
Vet Microbiol ; 252: 108908, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33254056

RESUMO

The objective was to identify the active fractions of polysaccharide against replication of ALV-J and elucidate their structure activity relationship. The optimal extraction conditions were extracting temperature 90℃, pH 9 and the ratio of liquid to solid 30:1. Under these conditions, extraction yield of total polysaccharide was 6.5 % ± 0.19 %. Total polysaccharide was then purified by DEAE-52 cellulose and Sephadex G-200 gel. Three fractions, PPP-1, PPP-2, and PPP-3, were identified with molecular weight of 463.70, 99.41, and 26.97 kDa, respectively. Three polysaccharide fractions were all composed of 10 monosaccharides in different proportions. Compared with PPP-1, which was mainly composed of glucose, PPP-2 and PPP-3 contained a higher proportion of galactose, glucuronic acid and galacturonic acid. The Congo red assay indicated that the PPP-2 may have a triple helical structure, while PPP-1 and PPP-3 were absent. In vitro assay showed that there was no significant cytotoxicity among the polysaccharide fractions under the concentration of 800 µg mL-1 (P > 0.05). The antiviral test showed that PPP-2 had the strongest activity, indicating PPP-2 was the major antiviral component. The structure-activity relationship showed that the antiviral activities of polysaccharide fractions were affected by their monosaccharide composition, molecular weight, and triple helical structure, which was a result of a combination of multiple molecular structural factors. These results showed that the PPP-2 could be exploited as a valued product for replacing synthetic antiviral drugs, and provided support for future applications of polysaccharide from Pinus massoniana pollen as a useful source for antiviral agent.


Assuntos
Antivirais/farmacologia , Vírus da Leucose Aviária/efeitos dos fármacos , Leucose Aviária/tratamento farmacológico , Pinus/química , Polissacarídeos/farmacologia , Replicação Viral/efeitos dos fármacos , Animais , Antivirais/química , Antivirais/isolamento & purificação , Leucose Aviária/virologia , Vírus da Leucose Aviária/fisiologia , Linhagem Celular , Embrião de Galinha , Monossacarídeos/química , Monossacarídeos/isolamento & purificação , Monossacarídeos/farmacologia , Pólen/química , Polissacarídeos/química , Polissacarídeos/isolamento & purificação , Relação Estrutura-Atividade
13.
Vet Microbiol ; 250: 108862, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33007608

RESUMO

The differences among different sub-groups of the avian leukosis virus (ALV) genome are mainly concentrated in the env gene, which binds to cell-specific receptors and determines the characteristics of viral tropism and pathogenicity. In this study, two rescued viruses rGX15MM6-2 (ALV of subgroup J, ALV-J) and rGX14FF03 (ALV of subgroup B, ALV-B) and a recombinant virus rALV-B-Jenv (ALV-B's backbone with ALV-J's env) were generated and tested utilizing both in vitro and in vivo experiments. The results showed that the replication ability of the viruses released in DF-1 cell cultures was listed in order as rGX15MM6-2 > rALV-B-Jenv > rGX14FF03. rGX15MM6-2 caused the most serious suppression of body weight gain, exhibited a significant negative effect on the development of immune organs (P < 0.05) and lower antibody responses to vaccinations with the commercial oil-emulsion vaccines (OEVs) (P<0.05) in the challenged chickens. The viral detection showed that the positive rate in blood from the birds infected with rALV-B-Jenv were respectively higher than those from the birds infected with rGX14FF03 (P < 0.05). At 25 wpi, similar tumors were found in the abdominal cavity of the birds in rGX15MM6-2 and rALV-B-Jenv groups. The results demonstrated that the ALV-J env gene significantly increases the pathogenicity of the recombinant ALV-B. With the increasing incidence of co-infections of different subgroups of ALV in the field, the possibility of viral recombination is increasing and demands further study.


Assuntos
Vírus da Leucose Aviária/genética , Vírus da Leucose Aviária/patogenicidade , Leucose Aviária/virologia , Genes env , Replicação Viral , Animais , Leucose Aviária/imunologia , Vírus da Leucose Aviária/classificação , Linhagem Celular , Galinhas/virologia , Fibroblastos/virologia , Doenças das Aves Domésticas/imunologia , Doenças das Aves Domésticas/virologia , Recombinação Genética , Tropismo Viral , Viremia , Virulência
14.
Arch Virol ; 165(12): 2877-2881, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32885326

RESUMO

Avian leukosis virus (ALV) is associated with immune suppression, neoplasia, and reduced performance in chickens. In this study, two strains of ALV were isolated from Luxi gamecocks by DF-1 cell culture and identified by PCR, immunofluorescence assay, and sequencing of the viral genome. These strains were found to be novel recombinant viruses with nucleotide sequence identity of over 93.0% in the LTR and 94.4% in U3 to ALV-J, over 95.0% in the 5'UTR to ALV-C, over 93.4% in gp85 to ALV-B, and over 96.0% in gp37 to ALV-E. These results indicate that these two isolates are recombinants between ALV-J, ALV-C, ALV-E and ALV-B.


Assuntos
Vírus da Leucose Aviária/isolamento & purificação , Leucose Aviária/virologia , Galinhas/virologia , Genoma Viral , Doenças das Aves Domésticas/virologia , Vírus Reordenados/isolamento & purificação , Animais , Vírus da Leucose Aviária/patogenicidade , Sequência de Bases , China , Filogenia , Vírus Reordenados/patogenicidade , Análise de Sequência , Proteínas do Envelope Viral/genética , Virulência
15.
J Virol ; 94(22)2020 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-32878894

RESUMO

Subgroup J avian leukemia virus (ALV-J), belonging to the genus Alpharetrovirus, enters cells through its envelope surface unit (gp85) via specifically recognizing the cellular receptor chicken Na+/H+ exchanger type I (chNHE1), the 28 to 39 N-terminal residues of which were characterized as the minimal receptor functional domain in our previous studies. In this study, to further clarify the precise organization and properties of the interaction between ALV-J gp85 and chNHE1, we identified the chNHE1-binding domain of ALV-J gp85 using a series of gp85 mutants with segment substitutions and evaluating their effects on chNHE1 binding in protein-cell binding assays. Our results showed that hemagglutinin (HA) substitutions of amino acids (aa) 38 to 131 (N terminus of gp85) and aa 159 to 283 (C terminus of gp85) significantly inhibited the interaction between gp85 and chNHE1/chNHE1 loop 1. In addition, these HA-substituted chimeric gp85 proteins could not effectively block the entry of ALV-J into chNHE1-expressing cells. Furthermore, analysis of various N-linked glycosylation sites and cysteine mutants in gp85 revealed that glycosylation sites (N6 and N11) and cysteines (C3 and C9) were directly involved in receptor-gp85 binding and important for the entry of ALV-J into cells. Taken together, our findings indicated that the bipartite sequence motif, spanning aa 38 to 131 and aa 159 to 283, of ALV-J gp85 was essential for binding to chNHE1, with its two N-linked glycosylation sites and two cysteines being important for its receptor-binding function and subsequent viral infection steps.IMPORTANCE Infection of a cell by retroviruses requires the attachment and fusion of the host and viral membranes. The specific adsorption of envelope (Env) surface proteins to cell receptors is a key step in triggering infections and has been the target of antiviral drug screening. ALV-J is an economically important avian pathogen that belongs to the genus Alpharetrovirus and has a wider host range than other ALV subgroups. Our results showed that the amino acids 38 to 131 of the N terminus and 159 to 283 of the C terminus of ALV-J gp85 controlled the efficiency of gp85 binding to chNHE1 and were critical for viral infection. In addition, the glycosylation sites (N6 and N11) and cysteines (C3 and C9) of gp85 played a crucial role in the receptor binding and viral entry. These findings might help elucidate the mechanism of the entry of ALV-J into host cells and provide antiviral targets for the control of ALV-J.


Assuntos
Vírus da Leucose Aviária/fisiologia , Leucose Aviária/virologia , Receptores Virais/metabolismo , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Animais , Vírus da Leucose Aviária/genética , Linhagem Celular , Galinhas/metabolismo , Especificidade de Hospedeiro , Proteínas de Membrana/metabolismo , Doenças das Aves Domésticas/virologia , Domínios Proteicos , Trocadores de Sódio-Hidrogênio/metabolismo , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética
16.
Arch Virol ; 165(11): 2615-2618, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32770484

RESUMO

In this study, an avian leukosis virus (ALV) strain (GX-2020-01) was isolated from a three-yellow chicken, and its complete genome was 7570 bp long with the typical organization "5'LTR-gag-pol-env-3'LTR." Phylogenetic analysis and sequence comparison revealed that it belongs to the ALV-J subgroup. However, the LTR region of GX-2020-01 is highly similar to that of reference strains of ALV-K/E (96.61%-97.10%), demonstrating that this novel isolate is a natural recombinant. The replication efficiency of GX-2020-01 was significantly lower than the previously isolated ALV-J strain (NX0101), indicating that the recombination event might have resulted in slower virus replication, making it harder for it to be detected through routine testing.


Assuntos
Vírus da Leucose Aviária/classificação , Leucose Aviária/virologia , Genoma Viral , Doenças das Aves Domésticas/virologia , Animais , Vírus da Leucose Aviária/isolamento & purificação , Galinhas , China , Filogenia , Vírus Reordenados/classificação , Vírus Reordenados/isolamento & purificação , Proteínas Virais/genética , Sequenciamento Completo do Genoma
17.
Vet Microbiol ; 247: 108781, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32768227

RESUMO

Immune tolerance induced by avian leukosis virus subgroup J (ALV-J) is a prerequisite for tumorigenesis. Although we had reported that B cell anergy induced by ALV-J was the main reason for immune tolerance, the molecular mechanism still remains unclear. Here, we found SU protein of ALV-J interacted with tyrosine kinase Lyn (a key protein in BCR signaling pathway) by confocal laser scanning microscopy and co-immunoprecipitation test, which suggested that Lyn might play an important role in B cell anergy induced by ALV-J. Correspondingly, the mRNA and protein level of Lyn was significantly up-regulated in B cells after ALV-J infection. Subsequently, the phosphorylated protein levels of Lyn at Tyr507 site were significantly up-regulated in ALV-J-infected B cells after BCR signal activation, but the phosphorylated protein level of Syk (a direct substrate of Lyn) at Tyr525/526 site, Ca2+ flux, and NF-κB p65 protein level were significantly down-regulated. Interestingly, the phosphorylated protein level of Syk at Tyr525/526 site, Ca2+ flux, and NF-κB p65 protein level were both significantly retrieved after the shLyn treatment in B cells infected by ALV-J. In summary, these results indicated that ALV-J activated the negative regulatory effect of phosphorylated Lyn protein at 507 site in BCR signal transduction pathway and then mediated B cell anergy, which will provide a new insight for revealing the pathogenesis of immune tolerance induced by ALV-J.


Assuntos
Vírus da Leucose Aviária/imunologia , Linfócitos B/imunologia , Anergia Clonal , Transdução de Sinais/imunologia , Quinases da Família src/genética , Animais , Leucose Aviária/imunologia , Leucose Aviária/virologia , Vírus da Leucose Aviária/classificação , Linfócitos B/virologia , Galinhas/imunologia , Galinhas/virologia , Regulação da Expressão Gênica , Fosforilação , Doenças das Aves Domésticas/virologia , Organismos Livres de Patógenos Específicos , Regulação para Cima
18.
Virus Res ; 286: 198083, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32621842

RESUMO

MicroRNAs(miRNAs) have been reported to regulate gene expression in many processes. MiRNA in extracellular vesicles (EVs) also have been widely investigated, while there is no studies of miRNAs in seminal EVs. Subgroup J of Avian leukosis virus (ALV-J) can be transmitted vertically, but the mechanism of it is not clear enough. This study was to examine the miRNA expression profile in seminal EVs and inquire into the relation between it and the vertical transmission by performing gene ontology (GO) and pathway enrichment analysis. Here, we first isolated and characterized seminal EVs by Nanoparticle Tracking Analysis、Western Blot and Transmission electron microscopy experiments. By deep sequencing of each EVs miRNA library, 9 typical differentially expressed miRNA, including 6 up-regulated and 3 down-regulated, were identified. Gene target prediction, GO annotation and KEGG pathway enrichment analysis showed possible function associated with these miRNAs. Overall, these findings will increase our understanding of the content and composition of miRNA in seminal EVs and provide new insights into the important role of the seminal EVs miRNAs regulation in ALV-J transmission.


Assuntos
Leucose Aviária/virologia , Vesículas Extracelulares/virologia , Perfilação da Expressão Gênica , MicroRNAs/genética , Sêmen/virologia , Animais , Vírus da Leucose Aviária , Galinhas/virologia , Regulação para Baixo , Masculino , Doenças das Aves Domésticas/virologia , Análise de Sequência de DNA , Regulação para Cima
19.
Poult Sci ; 99(7): 3469-3480, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32616241

RESUMO

During the process of transmission and spread of avian leukosis virus subgroup J (ALV-J) in chickens worldwide, the viral genome is constantly changing. A comprehensive and systematic study of the evolutionary process of ALV-J in China is needed. In this study, we amplified the full-length viral cDNA sequences of 16 ALV-J isolates of Yellow-chicken origin and analyzed and compared these sequences with another 69 ALV-J strains isolated during the years 1988-2018. These isolates were then sorted into 2 clusters: cluster I included isolates that mainly originated from the layers and White-feather broilers from northern China; cluster II included isolates mainly from the Yellow-chicken, most of them being from southern China. According to the sequence homologies of the whole genome and gag, pol, gp85, and gp37 genes, the ALV-J strains are more likely to randomly change in different directions from the original strain HPRS-103 as time passes. The results of entropy analysis of the sequences of gag, pol, and env revealed that the env gene had the largest variation, and the gag gene nonconserved sites are mainly concentrated in p19, p10, and p12. In addition, 84.71% (72/85) of the isolates had the 205-nucleotide (nt) deletion in the 3'UTR region, and 30.59% (26/85) of the isolates had the 125-nt to 127-nt deletion in the E element. Our study provides evidence for the coexistence of 2 extremely different clusters of ALV-J prevailing in China and in some other countries during the period of 1988-2018 and implies that the clusters are highly dependent on the host genetic background and the geographic location.


Assuntos
Vírus da Leucose Aviária/genética , Leucose Aviária/virologia , Galinhas , Doenças das Aves Domésticas/virologia , Análise de Sequência de DNA/veterinária , Animais , China , DNA Complementar/análise , DNA Viral/análise , Geografia , Interações Hospedeiro-Patógeno
20.
BMC Vet Res ; 16(1): 218, 2020 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-32600312

RESUMO

BACKGROUND: Studies have shown that some viral infections cause structural changes in the intestinal microflora, but little is known about the effects of tumorigenic viral infection on the intestinal microflora of chickens. RESULTS: A 29-week commercial layer flock positive for avian leukosis virus-J (ALV-J), Marek's disease virus (MDV) and avian reticuloendotheliosis virus (REV) was selected, and fresh fecal samples were collected and examined for the composition of the gut microflora by Illumina sequencing of the V3-V4 region of the 16S rRNA gene. The operational taxonomic units (OTUs) of the fecal microbiota differentiated the chickens infected with only ALV-J and those coinfected with ALV-J and MDV or REV from infection-negative chickens. The enrichment and diversity of cloacal microflora in chickens infected with ALV-J alone were slightly different from those in the infection-negative chickens. However, the diversity of cloacal microflora was significantly increased in chickens coinfected with both ALV-J and MDV or REV. CONCLUSIONS: The intestinal microbiota was more strongly disturbed in chickens after coinfection with ALV-J and MDV or REV than after infection with ALV-J alone, and there may be underlying mechanisms by which the capacity for the stabilization of the intestinal flora was impaired due to viral infection and tumorigenesis.


Assuntos
Bactérias/classificação , Coinfecção/veterinária , Microbioma Gastrointestinal , Doenças das Aves Domésticas/virologia , Animais , Leucose Aviária/virologia , Vírus da Leucose Aviária/isolamento & purificação , Bactérias/genética , Bactérias/isolamento & purificação , Galinhas , Fezes/microbiologia , Feminino , Herpesvirus Galináceo 2/isolamento & purificação , Doença de Marek/virologia , Doenças das Aves Domésticas/microbiologia , RNA Ribossômico 16S , Vírus da Reticuloendoteliose/isolamento & purificação , Infecções por Retroviridae/veterinária , Infecções Tumorais por Vírus/veterinária
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...