Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
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
2.
J Biol Chem ; 291(28): 14815-25, 2016 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-27226547

RESUMO

Avian metapneumovirus (aMPV) fusion (F) protein mediates virus-cell membrane fusion to initiate viral infection, which requires F protein binding to its receptor(s) on the host cell surface. However, the receptor(s) for aMPV F protein is still not identified. All known subtype B aMPV (aMPV/B) F proteins contain a conserved Arg-Asp-Asp (RDD) motif, suggesting that the aMPV/B F protein may mediate membrane fusion via the binding of RDD to integrin. When blocked with integrin-specific peptides, aMPV/B F protein fusogenicity and viral replication were significantly reduced. Specifically we identified integrin αv and/or ß1-mediated F protein fusogenicity and viral replication using antibody blocking, small interfering RNAs (siRNAs) knockdown, and overexpression. Additionally, overexpression of integrin αv and ß1 in aMPV/B non-permissive cells conferred aMPV/B F protein binding and aMPV/B infection. When RDD was altered to RAE (Arg-Ala-Glu), aMPV/B F protein binding and fusogenic activity were profoundly impaired. These results suggest that integrin αvß1 is a functional receptor for aMPV/B F protein-mediated membrane fusion and virus infection, which will provide new insights on the fusogenic mechanism and pathogenesis of aMPV.


Assuntos
Fusão Celular , Metapneumovirus/fisiologia , Infecções por Paramyxoviridae/fisiopatologia , Receptores de Vitronectina/fisiologia , Proteínas Virais de Fusão/fisiologia , Animais , Linhagem Celular , Infecções por Paramyxoviridae/virologia , Replicação Viral
3.
J Virol ; 90(24): 11231-11246, 2016 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-27707927

RESUMO

The entry of avian metapneumovirus (aMPV) into host cells initially requires the fusion of viral and cell membranes, which is exclusively mediated by fusion (F) protein. Proteolysis of aMPV F protein by endogenous proteases of host cells allows F protein to induce membrane fusion; however, these proteases have not been identified. Here, we provide the first evidence that the transmembrane serine protease TMPRSS12 facilitates the cleavage of subtype B aMPV (aMPV/B) F protein. We found that overexpression of TMPRSS12 enhanced aMPV/B F protein cleavage, F protein fusogenicity, and viral replication. Subsequently, knockdown of TMPRSS12 with specific small interfering RNAs (siRNAs) reduced aMPV/B F protein cleavage, F protein fusogenicity, and viral replication. We also found a cleavage motif in the aMPV/B F protein (amino acids 100 and 101) that was recognized by TMPRSS12. The histidine, aspartic acid, and serine residue (HDS) triad of TMPRSS12 was shown to be essential for the proteolysis of aMPV/B F protein via mutation analysis. Notably, we observed TMPRSS12 mRNA expression in target organs of aMPV/B in chickens. Overall, our results indicate that TMPRSS12 is crucial for aMPV/B F protein proteolysis and aMPV/B infectivity and that TMPRSS12 may serve as a target for novel therapeutics and prophylactics for aMPV. IMPORTANCE: Proteolysis of the aMPV F protein is a prerequisite for F protein-mediated membrane fusion of virus and cell and for aMPV infection; however, the proteases used in vitro and vivo are not clear. A combination of analyses, including overexpression, knockdown, and mutation methods, demonstrated that the transmembrane serine protease TMPRSS12 facilitated cleavage of subtype B aMPV (aMPV/B) F protein. Importantly, we located the motif in the aMPV/B F protein recognized by TMPRSS12 and the catalytic triad in TMPRSS12 that facilitated proteolysis of the aMPV/B F protein. This is the first report on TMPRSS12 as a protease for proteolysis of viral envelope glycoproteins. Our study will shed light on the mechanism of proteolysis of aMPV F protein and pathogenesis of aMPV.


Assuntos
Interações Hospedeiro-Patógeno , Metapneumovirus/genética , Infecções por Paramyxoviridae/enzimologia , Doenças das Aves Domésticas/enzimologia , Proteínas Virais de Fusão/química , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Linhagem Celular , Galinhas , Chlorocebus aethiops , Cricetulus , Células Epiteliais/enzimologia , Células Epiteliais/imunologia , Células Epiteliais/virologia , Fibroblastos/enzimologia , Fibroblastos/imunologia , Fibroblastos/virologia , Regulação da Expressão Gênica , Metapneumovirus/crescimento & desenvolvimento , Metapneumovirus/imunologia , Modelos Moleculares , Infecções por Paramyxoviridae/imunologia , Infecções por Paramyxoviridae/virologia , Doenças das Aves Domésticas/imunologia , Doenças das Aves Domésticas/virologia , Domínios Proteicos , Estrutura Secundária de Proteína , Proteólise , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Células Vero , Proteínas Virais de Fusão/antagonistas & inibidores , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/metabolismo , Internalização do Vírus , Replicação Viral
4.
J Gen Virol ; 96(Pt 1): 150-158, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25274857

RESUMO

Avian leukosis virus subgroup J (ALV-J) was first isolated from meat-producing chickens that had developed myeloid leukosis. However, ALV-J infections associated with hemangiomas have occurred in egg-producing (layer) flocks in China. In this study, we identified an ALV-J layer isolate (HLJ13SH01) as a recombinant of ALV-J and a Rous sarcoma virus Schmidt-Ruppin B strain (RSV-SRB), which contained the RSV-SRB 5'-LTR and the other genes of ALV-J. Replication kinetic testing indicated that the HLJ13SH01 strain replicated faster than other ALV-J layer isolates in vitro. Sequence analysis indicated that the main difference between the two isolates was the 5'-LTR sequences, particularly the U3 sequences. A 19 nt insertion was uniquely found in the U3 region of the HLJ13SH01 strain. The results of a Dual-Glo luciferase assay revealed that the 19 nt insertion in the HLJ13SH01 strain increased the enhancer activity of the U3 region. Moreover, an additional CCAAT/enhancer element was found in the 19 nt insertion and the luciferase assay indicated that this element played a key role in increasing the enhancer activity of the 5'-U3 region. To confirm the potentiation effect of the 19 nt insertion and the CCAAT/enhancer element on virus replication, three infectious clones with 5'-U3 region variations were constructed and rescued. Replication kinetic testing of the rescued viruses demonstrated that the CCAAT/enhancer element in the 19 nt insertion enhanced the replication capacity of the ALV-J recombinant in vitro.


Assuntos
Vírus da Leucose Aviária/genética , Leucose Aviária/virologia , Galinhas/virologia , Replicação do DNA/genética , Doenças das Aves Domésticas/virologia , Vírus do Sarcoma de Rous/genética , Replicação Viral/genética , Animais , China
5.
Arch Virol ; 160(10): 2445-53, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26175070

RESUMO

The entry of enveloped viruses into host cells requires the fusion of viral and cell membranes. These membrane fusion reactions are mediated by virus-encoded glycoproteins. In the case of avian metapneumovirus (aMPV), the fusion (F) protein alone can mediate virus entry and induce syncytium formation in vitro. To investigate the fusogenic activity of the aMPV F protein, we compared the fusogenic activities of three subtypes of aMPV F proteins using a TCSD50 assay developed in this study. Interestingly, we found that the F protein of aMPV subtype B (aMPV/B) strain VCO3/60616 (aMPV/vB) was hyperfusogenic when compared with F proteins of aMPV/B strain aMPV/f (aMPV/fB), aMPV subtype A (aMPV/A), and aMPV subtype C (aMPV/C). We then further demonstrated that the amino acid (aa) residue 149F contributed to the hyperfusogenic activity of the aMPV/vB F protein. Moreover, we revealed that residue 149F had no effect on the fusogenic activities of aMPV/A, aMPV/C, and human metapneumovirus (hMPV) F proteins. Collectively, we provide the first evidence that the amino acid at position 149 affects the fusogenic activity of the aMPV/B F protein, and our findings will provide new insights into the fusogenic mechanism of this protein.


Assuntos
Variação Genética , Metapneumovirus/genética , Infecções por Paramyxoviridae/veterinária , Doenças das Aves Domésticas/virologia , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Chlorocebus aethiops , Humanos , Metapneumovirus/química , Metapneumovirus/classificação , Metapneumovirus/metabolismo , Dados de Sequência Molecular , Infecções por Paramyxoviridae/virologia , Alinhamento de Sequência , Perus/virologia , Células Vero , Proteínas Virais de Fusão/genética
6.
J Clin Microbiol ; 52(1): 37-44, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24131697

RESUMO

Avian leukosis virus (ALV) subgroups A, B, and J are very common in poultry flocks and have caused serious economic losses in recent years. A multiplex PCR (mPCR) method for the detection of these three subgroups was developed and optimized in this study. We first designed a common forward primer, PF, and three downstream primers, AR, BR, and JR, which can amplify 715 bp for subgroup A, 515 bp for subgroup B, and 422 bp for subgroup J simultaneously in one reaction. The mPCR method produced neither cross-reactions with other subgroups of ALVs nor nonspecific reactions with other common avian viruses. The detection limit of the mPCR was as low as 1 × 10(3) viral DNA copies of each of the three subgroups. In animal experiments, the mPCR detected ALVs 2 to 4 days earlier than did virus isolation from whole-blood samples and cloaca swabs. Furthermore, a total of 346 clinical samples (including 127 tissue samples, 86 cloaca swabs, 59 albumen samples, and 74 whole-blood samples) from poultry flocks with suspected ALV infection were examined by mPCR, routine PCR, and virus isolation. The positive sample/total sample ratios for ALV-A, ALV-B, and ALV-J were 48% (166/346) as detected by mPCR and 48% (166/346) as detected by routine PCR. However, the positive sample/total sample ratio detected by virus isolation was 40% (138/346). The results of the mPCR and routine PCR were confirmed by sequencing the specific fragments. These results indicate that the mPCR method is rapid, specific, sensitive, and convenient for use in epidemiological studies of ALV, clinical detection of ALV, and ALV eradication programs.


Assuntos
Vírus da Leucose Aviária/classificação , Vírus da Leucose Aviária/isolamento & purificação , Leucose Aviária/diagnóstico , Leucose Aviária/virologia , Técnicas de Diagnóstico Molecular/métodos , Reação em Cadeia da Polimerase Multiplex/métodos , Medicina Veterinária/métodos , Animais , Vírus da Leucose Aviária/genética , Epidemiologia Molecular/métodos , Aves Domésticas , Sensibilidade e Especificidade , Fatores de Tempo
7.
Arch Virol ; 159(8): 2051-7, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24643331

RESUMO

Reticuloendotheliosis virus (REV) causes an oncogenic, immunosuppressive and runting syndrome in many avian hosts worldwide. REV infection has never been reported in mallard ducks, however. To identify REV infection in mallards, we collected 40 mallard duck samples from Jilin Province of China. In this study, the REV strain, DBYR1102, was first isolated from a mallard in China and identified by PCR, indirect immunofluorescence assay and electron microscopy. The gp90 gene and complete LTR of DBYR1102 were amplified and sequenced. Phylogenetic analysis based on gp90 genes of REV indicated that the REV strain DBYR1102 is closely related to strain HLJR0901 from northeastern China, the prairie chicken isolate APC-566, and REV subtype III, represented by chick syncytial virus. This new strain is distantly related to two other subtypes of REV, 170A and SNV. Phylogenetic analysis based on the LTR yielded information similar to that obtained with the gp90 genes. The results of this study not only expand our epidemiological understanding of REV in the wild birds of China but also demonstrate the potential role of wild waterfowl in REV transmission.


Assuntos
Doenças das Aves/virologia , Vírus da Reticuloendoteliose/isolamento & purificação , Infecções por Retroviridae/veterinária , Animais , Anseriformes/virologia , Feminino , Masculino , Dados de Sequência Molecular , Filogenia , Vírus da Reticuloendoteliose/classificação , Vírus da Reticuloendoteliose/genética , Infecções por Retroviridae/virologia
8.
J Clin Microbiol ; 50(3): 953-60, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22205787

RESUMO

Avian leukosis virus subgroup J (ALV-J) was first isolated from meat-type chickens in 1988. No field cases of ALV-J infection or tumors in layer chickens were observed worldwide until 2004. However, layer flocks in China have experienced outbreaks of this virus in recent years. The molecular epidemiology of ALV-J strains isolated from layer flocks was investigated. The env genes of 77.8% (21/27) of the ALV-J layer isolates with a high degree of genetic variation were significantly different from the env genes of the prototype strain of ALV-J (HPRS-103) and American and Chinese strains from meat-type chickens (designated ALV-J broiler isolates). A total of 205 nucleotides were deleted from the 3' untranslated region of 89.5% (17/19) of the ALV-J layer isolates. Approximately 94.7% (16/17) of the layer isolates contained a complete E element of 146 to 149 residues. The U3 sequences of 84.2% (16/19) of the ALV-J layer isolates displayed less than 92.5% sequence homology to those of the ALV-J broiler isolates, although the transcriptional regulatory elements that are typical of avian retroviruses were highly conserved. Several unique nucleotide substitutions in the env gene, the U3 region, and the E element of most of the ALV-J layer isolates were detected. These results suggested that the env gene, E element, and U3 region in the ALV-J layer isolates have evolved rapidly and were significantly different from those of the ALV-J broiler isolates. These findings will contribute to a better understanding of the pathogenic mechanism of layer tumor diseases induced by ALV-J.


Assuntos
Vírus da Leucose Aviária/classificação , Vírus da Leucose Aviária/genética , Leucose Aviária/epidemiologia , Leucose Aviária/virologia , Surtos de Doenças , Doenças das Aves Domésticas/epidemiologia , Doenças das Aves Domésticas/virologia , Animais , Vírus da Leucose Aviária/isolamento & purificação , Galinhas , China/epidemiologia , Análise por Conglomerados , Variação Genética , Genótipo , Epidemiologia Molecular , Dados de Sequência Molecular , Mutação , RNA Viral/genética , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Proteínas do Envelope Viral/genética
9.
Virol J ; 8: 552, 2011 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-22185463

RESUMO

BACKGROUND: Avian leukosis virus subgroup J (ALV-J) preferentially induces myeloid leukosis (ML) in meat-type birds. Since 2008, many clinical cases of hemangioma rather than ML have frequently been reported in association with ALV-J infection in Chinese layer flocks. RESULTS: Three ALV-J strains associated with hemangioma were isolated and their proviral genomic sequences were determined. The three isolates, JL093-1, SD09DP03 and HLJ09MDJ-1, were 7,670, 7,670, and 7,633 nt in length. Their gag and pol genes were well conserved, with identities of 94.5-98.6% and 97.1-99.5%, respectively, with other ALV-J strains at the amino acid level (aa), while the env genes of the three isolates shared a higher aa identity with the env genes of other hemangioma strains than with those of ML strains. Interestingly, two novel 19-bp insertions in the U3 region in the LTR and 5' UTR, most likely derived from other retroviruses, were found in all the three isolates, thereby separately introducing one E2BP binding site in the U3 region in the LTR and RNA polymerase II transcription factor IIB and core promoter motif ten elements in the 5' UTR. Meanwhile, two binding sites in the U3 LTRs of the three isolates for NFAP-1 and AIB REP1 were lost, and a 1-base deletion in the E element of the 3' UTR of JL093-1 and SD09DP03 introduced a binding site for c-Ets-1. In addition to the changes listed above, the rTM of the 3' UTR was deleted in each of the three isolates. CONCLUSION: Our study is the first to discovery the coexistence of two novel insertions in the U3 region in the LTR and the 5' UTR of ALV-J associated with hemangioma symptoms, and the transcriptional regulatory elements introduced should be taken into consideration in the occurrence of hemangioma.


Assuntos
Vírus da Leucose Aviária/genética , Vírus da Leucose Aviária/patogenicidade , Genoma Viral/genética , Hemangioma/veterinária , Mutagênese Insercional/genética , Doenças das Aves Domésticas/patologia , Regiões 5' não Traduzidas/genética , Animais , Vírus da Leucose Aviária/classificação , Galinhas , China , Produtos do Gene env/química , Produtos do Gene env/genética , Hemangioma/patologia , Hemangioma/virologia , Dados de Sequência Molecular , Doenças das Aves Domésticas/virologia , Análise de Sequência de DNA , Sequências Repetidas Terminais/genética
10.
Mol Immunol ; 64(1): 106-11, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25438822

RESUMO

Infection with J group avian leukosis virus (ALV-J) can result in immunosuppression and subsequently increased susceptibility to secondary infection. The innate immune system is the first line defense system in prevention of further bacterial and viral infections. Cytokines play key roles in the innate immune system. In this study, we used RT-qPCR technology to test the cytokine mRNA expression levels in various immune tissues, including the spleen, bursa of fabricius and cecal tonsil, in the days following ALV-J infection. The results indicated that in the infected group, the expression levels of interleukin-6 (IL-6), IL-18, interferon-α (IFN-α) and IFN-γ significantly increased in the spleen and reached peak levels that were thousandfolds higher than baselines at 9-12 days post-infection (d.p.i.). The levels in the bursa of fabricius slightly increased, and the levels in the cecal tonsil were not significantly altered. Moreover, the pattern of the expression of these three cytokines in the spleens of the infected group was similar to the pattern of viremia of this group. These results suggest that the spleen plays an important role in the interaction between ALV-J infection and the innate immune system. This study contributes to the understanding of innate immune responses to ALV-J infection and also elucidates the mechanisms of the pathogenicity of ALV-J in chickens.


Assuntos
Vírus da Leucose Aviária/fisiologia , Leucose Aviária/genética , Leucose Aviária/virologia , Galinhas/imunologia , Citocinas/genética , Perfilação da Expressão Gênica , Imunidade/genética , Animais , Leucose Aviária/imunologia , Bolsa de Fabricius/metabolismo , Bolsa de Fabricius/patologia , Bolsa de Fabricius/virologia , Ceco/metabolismo , Ceco/patologia , Ceco/virologia , Galinhas/genética , Galinhas/virologia , Citocinas/metabolismo , Regulação da Expressão Gênica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Padrões de Referência , Baço/metabolismo , Baço/virologia , Fatores de Tempo , Viremia/genética , Viremia/virologia
11.
Sci Rep ; 5: 15584, 2015 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-26498473

RESUMO

Avian metapneumovirus (aMPV) and human metapneumovirus (hMPV) are members of the genus Metapneumovirus in the subfamily Pneumovirinae. Metapneumovirus fusion (F) protein mediates the fusion of host cells with the virus membrane for infection. Trypsin- and/or low pH-induced membrane fusion is a strain-dependent phenomenon for hMPV. Here, we demonstrated that three subtypes of aMPV (aMPV/A, aMPV/B, and aMPV/C) F proteins promoted cell-cell fusion in the absence of trypsin. Indeed, in the presence of trypsin, only aMPV/C F protein fusogenicity was enhanced. Mutagenesis of the amino acids at position 100 and/or 101, located at a putative cleavage region in aMPV F proteins, revealed that the trypsin-mediated fusogenicity of aMPV F proteins is regulated by the residues at positions 100 and 101. Moreover, we demonstrated that aMPV/A and aMPV/B F proteins mediated cell-cell fusion independent of low pH, whereas the aMPV/C F protein did not. Mutagenesis of the residue at position 294 in the aMPV/A, aMPV/B, and aMPV/C F proteins showed that 294G played a critical role in F protein-mediated fusion under low pH conditions. These findings on aMPV F protein-induced cell-cell fusion provide new insights into the molecular mechanisms underlying membrane fusion and pathogenesis of aMPV.


Assuntos
Metapneumovirus/metabolismo , Tripsina/metabolismo , Proteínas Virais de Fusão/química , Sequência de Aminoácidos , Animais , Chlorocebus aethiops , Cricetinae , Concentração de Íons de Hidrogênio , Fusão de Membrana , Metapneumovirus/genética , Mutagênese , Mutação , Células Vero
12.
J Virol Methods ; 187(2): 278-83, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23201286

RESUMO

An antigen-capture enzyme-linked immunosorbent assay (AC-ELISA) employing monoclonal and polyclonal antibodies against p27 was developed for the detection of the avian leukosis virus (ALV). The specificity of the optimized AC-ELISA was evaluated using avian leukosis virus subgroup J (ALV-J), avian leukosis virus subgroup A (ALV-A), avian leukosis virus subgroup B (ALV-B), avian infectious bronchitis virus (IBV), Marek's disease virus (MDV), avian infectious laryngotracheitis virus (ILTV), Fowlpox virus (FPV), infectious bursal disease virus (IBDV), Newcastle disease virus (NDV), avian reovirus (ARV), reticuloendotheliosis virus (REV), avian influenza virus (AIV) and Escherichia coli. The only specimens that yielded a strong signal were ALV-J, ALV-A and ALV-B, indicating that this assay is suitable for the detection of ALV. The limit of detection of this assay was 1.25 ng/ml of rp27 protein and 10(1.79)TCID(50) units of HLJ09MDJ-1 (ALV-J). Moreover, this AC-ELISA can detect ALV in cloacal swabs of chickens experimentally infected as early as 12 days post-infection. The AC-ELISA detected the virus in the albumin and cloacal swabs of naturally infected chickens, and the results were confirmed by PCR, indicating that the AC-ELISA was a suitable method for the detection of ALV. This test is rapid and sensitive and could be convenient for epidemiological studies and eradication programs.


Assuntos
Anticorpos Antivirais , Antígenos Virais/análise , Vírus da Leucose Aviária/imunologia , Leucose Aviária/diagnóstico , Técnicas de Laboratório Clínico/métodos , Medicina Veterinária/métodos , Virologia/métodos , Animais , Leucose Aviária/virologia , Galinhas , Ensaio de Imunoadsorção Enzimática/métodos , Sensibilidade e Especificidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA