Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 209
Filtrar
1.
Poult Sci ; 103(7): 103835, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38772092

RESUMO

Avian leukemia virus subgroup J (ALV-J) and chicken infectious anemia virus (CIAV) can be vertically transmitted; however, the pathogenicity of vertically transmitted coinfection with these 2 pathogens has not been studied. In this study, we created a model of chick morbidity in which chicks carried either ALV-J, CIAV, or both viruses via embryo inoculation. Thereafter, we analyzed the effects of vertically transmitted coinfection with CIAV and ALV-J on the pathogenicity of ALV-J and performed a purification assay based on hatching, mortality viremia positivity, and detection of fecal ALV-p27 antigen rates, and body weight. The hatching rate of the ALV-J+CIAV group was 68.57%, lower than those of the single infection and control groups. The survival curve showed that the mortality rates of the CIAV and ALV-J coinfection groups were higher than those of the single infection and control groups. Body weight statistics showed that coinfection aggravated the 7-d growth inhibition effect. The results of ALV-p27 antigen detection in cell culture supernatants showed that the positivity rates of the ALV-J and ALV-J+CIAV groups were 100% at all ages and 0% in the control group. The results of ALV-p27 antigen detection by anal swabs showed that the positivity rates of the ALV-J group were 92.86, 90.90, 88.89, and 93.33% at all ages, and that the ALV-J p27 positivity detection rate of anal swabs was lower than that of plasma virus isolation. The immune organ index of the ALV-J+CIAV group was significantly or very significantly lower than those of the single infection and control groups. The immune organ viral load showed that coinfection with CIAV and ALV-J promoted the proliferation of ALV-J and CIAV in immune organs. Coinfection with ALV-J and CIAV reduced chicken embryo hatchability and increased chick mortality and growth inhibition relative to their respective single infections. Additionally, coinfection with ALV-J + CIAV was even more detrimental in inducing immune organ atrophy (e.g., the thymus, spleen, and bursa), and promoted individual virus replication during coinfection.


Assuntos
Vírus da Leucose Aviária , Leucose Aviária , Vírus da Anemia da Galinha , Galinhas , Infecções por Circoviridae , Coinfecção , Transmissão Vertical de Doenças Infecciosas , Doenças das Aves Domésticas , Animais , Vírus da Leucose Aviária/fisiologia , Vírus da Leucose Aviária/patogenicidade , Galinhas/virologia , Leucose Aviária/virologia , Coinfecção/veterinária , Coinfecção/virologia , Doenças das Aves Domésticas/virologia , Vírus da Anemia da Galinha/fisiologia , Vírus da Anemia da Galinha/patogenicidade , Infecções por Circoviridae/veterinária , Infecções por Circoviridae/virologia , Transmissão Vertical de Doenças Infecciosas/veterinária , Virulência , Embrião de Galinha
2.
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
3.
Mediators Inflamm ; 2021: 6665871, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33628117

RESUMO

The aim of this study was to better understand the sequence characteristics and immune responses in avian leukosis virus subgroup J (ALV-J) infected yellow chicken flocks in South China. We isolated four strains of ALV-J virus from these flocks, which were then identified by several methods, including subtype-specific polymerase chain reaction (PCR), enzyme-linked immunosorbent assay (ELISA), and immunofluorescence assay (IFA). All four viruses were sequenced for their complete genomes and named GD19GZ01, GD19GZ02, GD19GZ03, and GD19GZ04. In comparison with the reference sequence, the homology analysis showed that the gag and pol genes were relatively conserved, whereas env contained much variation. Both GD19GZ01 and GD19GZ02 almost entirely lacked the rTM region and E element, while the latter was retained in GD19GZ03 and GD19GZ04. Moreover, the virus replication levels in GD19GZ03 and GD19GZ04were much higher than those in GD19GZ01 and GD19GZ02. And three virus recombination events in GD19GZ01 and GD19GZ02 were revealed by the results of PDR5 and SimPlot software analysis. Additionally, we found that some interferon-stimulating genes (CH25H, MX, PKR, OAS, and ZAP) and inflammatory mediators (IL-4, IL-6, IL-10, IL-12, 1L-18, and TNF-α) were significantly upregulated in the immune system organs of clinical chickens. Taken together, these findings clarify and reveal the sequence characteristics and trends in the variation of ALV-J infection in yellow chicken flocks of South China.


Assuntos
Vírus da Leucose Aviária/patogenicidade , Galinhas/imunologia , Galinhas/virologia , Animais , Vírus da Leucose Aviária/classificação , China , Ensaio de Imunoadsorção Enzimática , Interleucina-10/metabolismo , Interleucina-4/metabolismo , Interleucina-6/metabolismo , Filogenia
4.
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
5.
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
6.
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
7.
Vet Microbiol ; 244: 108683, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32402336

RESUMO

In order to evaluate the influence of the vertical transmission of avian leukosis virus (ALV) from J subgroup (ALV-J) positive parents on the vaccine efficacy of Marek's disease virus (MDV), ALV-J positive male breeders × female breeders of Three-yellow chickens and the ALV negative male breeder × the negative female breeders were used respectively for crossbreeding to produce eggs and the hatching offspring. The commercial CVI988/Rispens vaccine was used to vaccinate the crossbred offspring at 1-day-old. At 7-days-old, the birds were inoculated with the inactivated oil-emulsion vaccines (OEVs) AIV-H5 monovalent and NDV + AIV-H9 bivalent, respectively. Then the birds were challenged with a Chinese very virulent (vv) MDV field strain GXY2 at 14-day-old. The results showed that the viral load of the challenged GXY2 in the offspring from the ALV-J positive breeders was significantly higher than that from the ALV-negative breeders' (P < 0.05), and the mortality and tumor incidence of offspring from the ALV-J positive breeders were higher than those of the ALV-negative breeders. Also the offspring of the ALV-J positive breeders exhibited a significant negative effect on the development of the immune organs (P < 0.05) and lower antibody responses to the vaccinations with the commercial OEVs (P<0.05). The MD vaccine protective index in the offspring from the ALV-J positive breeders was lower than that from the ALV-negative breeders. The results of the study demonstrated that the vertical transmission of ALV from the ALV-J positive parents caused severe immunosuppression and significantly reduced the Marek's disease vaccine efficacy in Three-yellow chickens.


Assuntos
Leucose Aviária/imunologia , Leucose Aviária/transmissão , Terapia de Imunossupressão/veterinária , Transmissão Vertical de Doenças Infecciosas , Vacinas contra Doença de Marek/imunologia , Animais , Vírus da Leucose Aviária/patogenicidade , Cruzamento , Galinhas/imunologia , Galinhas/virologia , Feminino , Masculino , Doença de Marek/imunologia , Doença de Marek/prevenção & controle , Aves Domésticas/imunologia , Aves Domésticas/virologia , Doenças das Aves Domésticas/imunologia , Doenças das Aves Domésticas/prevenção & controle , Doenças das Aves Domésticas/virologia , Potência de Vacina , Carga Viral
8.
Avian Pathol ; 49(5): 440-447, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32301629

RESUMO

ABSTRACT The prototype fowl glioma-inducing virus (FGVp) causes fowl glioma and cerebellar hypoplasia in chickens. In this study, we investigated whether a strain of avian leukosis virus (ALV), associated with avian osteopetrosis and mesenchymal neoplasms, is able to induce fowl glioma. We encountered avian osteopetrosis and mesenchymal neoplasms, including myxosarcoma and rhabdomyosarcoma, in Japanese native chickens used for both egg-laying and meat production. These birds were also affected by non-suppurative encephalitis and glioma in their brains. Four ALV strains (GifN_001, GifN_002, GifN_004, GifN_005) were isolated, and a phylogenic analysis of envSU showed that these isolates were classified into different clusters from FGVp and the variants previously reported. Whereas the envSU shared a high identity (94.7%) with that of Rous sarcoma virus (strain Schmidt-Ruppin B) (RSV-SRB), the identity between envTM of GifN_001 and that of FGVp was high (94.5%), indicating that GifN_strains may emerge by recombination between FGVp and other exogenous ALVs. Specific-pathogen-free chickens inoculated in ovo with GifN_001 revealed fowl glioma and cerebellar hypoplasia. These results suggest that the newly isolated strains have acquired neuropathogenicity to chickens.


Assuntos
Vírus da Leucose Aviária/patogenicidade , Leucose Aviária/virologia , Galinhas/virologia , Glioma/veterinária , Osteopetrose/veterinária , Doenças das Aves Domésticas/virologia , Animais , Vírus da Leucose Aviária/classificação , Vírus da Leucose Aviária/genética , Cerebelo/anormalidades , Cerebelo/virologia , Embrião de Galinha , Deficiências do Desenvolvimento/virologia , Encefalite/veterinária , Encefalite/virologia , Feminino , Glioma/virologia , Mixossarcoma/veterinária , Mixossarcoma/virologia , Malformações do Sistema Nervoso/veterinária , Malformações do Sistema Nervoso/virologia , Osteopetrose/virologia , Filogenia , Recombinação Genética , Rabdomiossarcoma/veterinária , Rabdomiossarcoma/virologia , Organismos Livres de Patógenos Específicos
9.
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
10.
Vet Microbiol ; 242: 108589, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32122593

RESUMO

The CRISPR/CRISPR-associated protein 9 (Cas9) system is a powerful gene-editing tool originally discovered as an integral mediator of bacterial adaptive immunity. Recently, this technology has been explored for its potential utility in providing new and unique treatments for viral infection. Marek's disease virus (MDV) and avian leukosis virus subgroup J (ALV-J), major immunosuppressive viruses, cause significant economic losses to the chicken industry. Here, we evaluated the efficacy of using MDV as a CRISPR/Cas9-delivery system to directly target and disrupt the reverse-transcribed products of the ALV-J RNA genome during its infection cycle in vitro and in vivo. We first screened multiple potential guide RNA (gRNA) target sites in the ALV-J genome and identified several optimized targets capable of effectively disrupting the latently integrated viral genome and providing efficient defense against new infection by ALV-J in cells. The optimal single-gRNAs and Cas9-expression cassettes were inserted into the genome of an MDV vaccine strain. The results indicated that engineered MDV stably expressing ALV-J-targeting CRISPR/Cas9 efficiently resisted ALV-J challenge in host cells. These findings demonstrated the CRISPR/Cas9 system as an effective treatment strategy against ALV-J infection. Furthermore, the results highlighted the potential of MDV as an effective delivery system for CRISPR/Cas9 in chickens.


Assuntos
Leucose Aviária/prevenção & controle , Sistemas CRISPR-Cas , Herpesvirus Galináceo 2/genética , Vacinas contra Doença de Marek/genética , Doenças das Aves Domésticas/prevenção & controle , Animais , Leucose Aviária/virologia , Vírus da Leucose Aviária/genética , Vírus da Leucose Aviária/patogenicidade , Galinhas/virologia , Fibroblastos/virologia , Edição de Genes/métodos , Vetores Genéticos , Genoma Viral , Doenças das Aves Domésticas/virologia , RNA Guia de Cinetoplastídeos/genética , RNA Viral/genética , Organismos Livres de Patógenos Específicos , Integração Viral
11.
Avian Pathol ; 49(1): 29-35, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31429308

RESUMO

To determine whether geese are susceptible to infection by avian leukosis virus (ALV), 702 serum samples from domestic and foreign goose breeds were screened for p27 antigen as well as being inoculated into DF-1 cell cultures to isolate ALV. Although 5.7% of samples were positive for p27 antigen, reactivity appeared to be non-specific because no ALV was detected in the corresponding DF-1 cultures. To further determine whether geese are susceptible to ALV-J isolated from chickens, ALV-J strain JS09GY7 was artificially inoculated into 10-day-old goose embryos, with one-day-old hatched goslings then screened for p27 antigen and the presence of ALV. In all cases, the results of both tests were negative. Liver tissues from the 1-day-old goslings were screened using a polymerase chain reaction-based assay, which failed to amplify ALV-J gene fragments from any of the samples. Further, no histopathological damage was observed in the liver tissues. ALV-J was further inoculated intraperitoneally into one-day-old goslings, with cloacal swabs samples and plasma samples then collected every 5 days for 30 days. All samples were again negative for the presence of p27 antigen and ALV, and liver tissues from the challenged geese showed no histopathological damage and were negative for the presence of ALV-J gene fragments. Furthermore, p27 antigen detection, PCR-based screening, and indirect immunofluorescence assays were all negative following the infection of goose embryo fibroblasts with ALV-J. Together, these results confirm that virulent chicken-derived ALV-J strains cannot infect geese, and that p27 antigen detection in goose serum is susceptible to non-specific interference.


Assuntos
Vírus da Leucose Aviária/patogenicidade , Leucose Aviária/virologia , Galinhas , Gansos , Animais , Leucose Aviária/imunologia , Vírus da Leucose Aviária/genética , Vírus da Leucose Aviária/imunologia , Vírus da Leucose Aviária/isolamento & purificação , Galinhas/virologia , Cloaca/virologia , DNA Viral/química , DNA Viral/isolamento & purificação , Fibroblastos/virologia , Imunofluorescência/veterinária , Gansos/embriologia , Gansos/virologia , Fígado/patologia , Fígado/virologia , Antígeno Nuclear de Célula em Proliferação/sangue , Antígeno Nuclear de Célula em Proliferação/isolamento & purificação , Virulência
12.
Poult Sci ; 98(12): 6497-6504, 2019 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-31549168

RESUMO

One natural recombinant avian leukosis virus (ALV) strain GX14DJ3-18 was isolated from a native gamecock by DF-1 cell culture and identified with Polymerase Chain Reaction (PCR), immunofluorescence assay and the viral genome's nucleotide sequencing. This strain was revealed as a novel recombinant virus with nucleotide sequence similarities of 95.4% Long Terminal Repeated (LTR), 95.8% 5', UTR, 97.9% gag, and 92.9% 3'untranslated regions (UTR) in ALV-J. Also we found sequence similarities of 99.3% pol and 99.0% gp37 in ALV-E, and 89.9% gp85 in ALV-A. The simulated congenital infection with GX14DJ3-18 in Three-Yellow chickens exhibited a significant negative effect on the development of immune organs (P < 0.05). Also, lower antibody responses were found to vaccinations with the commercial vaccines of Newcastle disease virus and with subtypes H5 and H9 of avian influenza virus (P < 0.05). The incidence of tumor or tumor-like lesions in the challenged birds was 14.28% (5/35), while none were observed in the un-challenged control group (0/35). These results suggested that GX14DJ3-18 is a novel recombinant ALV that can induce pathogenicity in the commercial Three-Yellow chickens. We speculated that cross-provincial sales of gamecocks in which ALVs have not been eradicated thoroughly might be a potential route for the transmission of ALVs to commercial chickens.


Assuntos
Vírus da Leucose Aviária/isolamento & purificação , Vírus da Leucose Aviária/patogenicidade , Leucose Aviária/virologia , Galinhas , Genoma Viral , Doenças das Aves Domésticas/virologia , Animais , Sequência de Bases , China , Alinhamento de Sequência , Virulência
13.
J Virol ; 93(17)2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31217247

RESUMO

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


Assuntos
Vírus da Leucose Aviária/patogenicidade , Leucose Aviária/genética , Proteínas Aviárias/genética , Proteínas Aviárias/metabolismo , Receptores Virais/genética , Receptores Virais/metabolismo , Animais , Leucose Aviária/metabolismo , Leucose Aviária/virologia , Vírus da Leucose Aviária/classificação , Vírus da Leucose Aviária/fisiologia , Linhagem Celular , Galinhas , Suscetibilidade a Doenças , Fibroblastos/citologia , Fibroblastos/metabolismo , Fibroblastos/virologia , Mesocricetus , Especificidade da Espécie , Internalização do Vírus
14.
Vet Microbiol ; 231: 214-217, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30955812

RESUMO

In this study, a recombinant ALV with ALV-K env and ALV-J backbone was generated (designated ALV-K-env-J) and tested in vitro and in vivo. The growth curve in DF1 cells showed that the recombinant virus replicated more efficiently in comparison with the ALV-J and ALV-K. Although all the infected chickens showed growth retardation compared with the non-infected chickens, the viral and serological detection showed that the positive rate and virus load detected in blood and cloaca, and the positive rate and titer of antibody against p27 from the chickens infected with ALV-K-env-J were higher than those from the chickens infected with the ALV-K, but less than those from the chickens infected with the ALV-J. All these data clearly demonstrated that the recombination event in this study increased the pathogenesis of ALV-K, and the potential recombination between different ALV subgroups should be worried when the clinical co-infections occur.


Assuntos
Vírus da Leucose Aviária/genética , Vírus da Leucose Aviária/patogenicidade , Leucose Aviária/virologia , Doenças das Aves Domésticas/virologia , Recombinação Genética , Animais , Anticorpos Antivirais/sangue , Leucose Aviária/imunologia , Galinhas/virologia , Cloaca/virologia , Doenças das Aves Domésticas/imunologia , Testes Sorológicos , Carga Viral
15.
Poult Sci ; 98(7): 2772-2780, 2019 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-30768138

RESUMO

Avian leukosis virus subgroup J has been found to infect many types of chickens with various genetic backgrounds. The ALV-J strain NX0101, which was isolated from broiler breeders in 2001, mainly induces the formation of myeloid cell tumors. However, strain HN10PY01, which was recently isolated from laying hens, mainly induces the formation of myeloid cell tumors and hemangioma. In order to determine the difference in pathogenicity of the 2 strains in broiler chickens, 2 groups of chicken embryos were infected with NA0101 and HN10PY01 separately. A comparison was made of the mortality, oncogenicity, body weights, indexes for immune organs, levels of ALV group-specific antigen p27, and mRNA expression levels of the tumor-related gene, p53, in ALV-J-infected birds and immune organs of theses chickens in response to Newcastle Disease Virus (NDV) and avian influenza virus subtype H9 (AIV-H9) vaccination. The results indicated that strain NX0101 was highly pathogenic in broiler chickens and led to a 30% mortality rate and 45% oncogenicity, compared with the HN10PY01-infected birds. Weight of chickens was also significantly lower after 15 wk (P < 0.05). In addition, the mRNA expression levels of tumor-related p53 in medulla, liver, and lung in broilers infected with strain NX0101 were significantly higher than those infected with strain HN10PY01 (P < 0.05). These results indicated that strain NX0101 had a higher replication ability in broiler chickens. The findings of this study will contribute to further elucidating the mechanisms underlying host susceptibility and tumor classification in ALV-J-infected chickens.


Assuntos
Vírus da Leucose Aviária/patogenicidade , Neoplasias/virologia , Doenças das Aves Domésticas/virologia , Virulência , Animais , Leucose Aviária/mortalidade , Leucose Aviária/patologia , Peso Corporal , Embrião de Galinha , Galinhas , Vírus da Influenza A/imunologia , Vírus da Doença de Newcastle/imunologia , Vacinas/administração & dosagem
16.
Retrovirology ; 16(1): 1, 2019 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-30602379

RESUMO

BACKGROUND: The pathogenesis of immunological tolerance caused by avian leukosis virus subgroup J (ALV-J), an oncogenic retrovirus, is largely unknown. RESULTS: In this study, the development, differentiation, and immunological capability of B cells and their progenitors infected with ALV-J were studied both morphologically and functionally by using a model of ALV-J congenital infection. Compared with posthatch infection, congenital infection of ALV-J resulted in severe immunological tolerance, which was identified as the absence of detectable specific antivirus antibodies. In congenitally infected chickens, immune organs, particularly the bursa of Fabricius, were poorly developed. Moreover, IgM-and IgG-positive cells and total immunoglobulin levels were significantly decreased in these chickens. Large numbers of bursa follicles with no differentiation into cortex and medulla indicated that B cell development was arrested at the early stage. Flow cytometry analysis further confirmed that ALV-J blocked the differentiation of CD117+chB6+ B cell progenitors in the bursa of Fabricius. Furthermore, both the humoral immunity and the immunological capability of B cells and their progenitors were significantly suppressed, as assessed by (a) the antibody titres against sheep red blood cells and the Marek's disease virus attenuated serotype 1 vaccine; (b) the proliferative response of B cells against thymus-independent antigen lipopolysaccharide (LPS) in the spleen germinal centres; and (c) the capacities for proliferation, differentiation and immunoglobulin gene class-switch recombination of B cell progenitors in response to LPS and interleukin-4(IL-4) in vitro. CONCLUSIONS: These findings suggested that the anergy of B cells in congenitally infected chickens is caused by the developmental arrest and dysfunction of B cell progenitors, which is an important factor for the immunological tolerance induced by ALV-J.


Assuntos
Vírus da Leucose Aviária/imunologia , Leucose Aviária/congênito , Subpopulações de Linfócitos B/patologia , Anergia Clonal , Doenças das Aves Domésticas/congênito , Células-Tronco/patologia , Animais , Anticorpos Antivirais/sangue , Leucose Aviária/patologia , Vírus da Leucose Aviária/patogenicidade , Subpopulações de Linfócitos B/química , Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/virologia , Bolsa de Fabricius/patologia , Diferenciação Celular , Proliferação de Células , Galinhas , Citometria de Fluxo , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Doenças das Aves Domésticas/patologia , Proteínas Proto-Oncogênicas c-kit/análise , Células-Tronco/química , Células-Tronco/imunologia , Células-Tronco/virologia
17.
Appl Microbiol Biotechnol ; 103(1): 427-435, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30349931

RESUMO

Avian leukosis virus (ALV) is an avian oncogenic retrovirus that induces leukemia-like proliferative diseases in chickens. ALV infection can result in the development of immunological tolerance and persistent viremia. Since effective vaccines against ALV are not yet available, its current prevention primarily depends on detection and eradication to establish exogenous ALV-free poultry flocks. In this study, a rapid and simple colloidal gold test strip method, specific for the group-specific antigen, p27 protein, was developed and systematically evaluated for the detection of ALV from different samples. The detection limit of this assay was as low as 6.25 ng/ml for p27 protein and 80 TCID50/ml for different subgroups of ALV. Besides, the test strip showed high specificity in the detection of different subgroups of ALV, including ALV-A, ALV-B, ALV-J, and ALV-K, with no cross-reaction with other avian pathogens. Furthermore, we artificially infected specific pathogen-free (SPF) chickens with ALV-J, collected cloacal swabs, and examined viral shedding using both test strips and ELISA. Results from the test strip were highly consistent with that from ELISA. In addition, 1104 virus isolates from anti-coagulant blood samples, 645 albumen samples, and 4312 meconium samples were tested, and the test strip results agreed with those of ELISA kit up to 97.1%. All the results indicated that the colloidal gold test strip could serve as a simple, rapid, sensitive, and specific diagnostic method for eradication of ALV in poultry farms.


Assuntos
Vírus da Leucose Aviária/isolamento & purificação , Leucose Aviária/diagnóstico , Coloide de Ouro , Imunoensaio/métodos , Animais , Anticorpos Monoclonais/imunologia , Antígenos Virais/imunologia , Leucose Aviária/virologia , Vírus da Leucose Aviária/patogenicidade , Galinhas , Cabras , Imunoensaio/instrumentação , Imunoglobulina G/imunologia , Limite de Detecção , Camundongos , Doenças das Aves Domésticas/virologia , Sensibilidade e Especificidade , Fatores de Tempo
18.
Biosens Bioelectron ; 124-125: 1-7, 2019 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-30339973

RESUMO

A sensitive and specific photoelectrochemical (PEC) immunosensor was fabricated for subgroup J avian leukosis viruses (ALV-J) analysis based on a dual signal-on strategy. Gold nanoparticles (AuNPs) decorated graphitic carbon nitride (AuNPs/g-C3N4) as photoelectrochemical species and primary antibody (Ab1) against ALV-J were immobilized onto ITO electrode in turn. An ALP-CdTe-Ab2 bio-conjugant was fabricated by assembling second antibody (Ab2) and alkaline phosphatase (ALP) to CdTe quantum dots (QDs) surface. The PEC immunosensor was fabricated by successively anchoring the target ALV-J and ALP-CdTe-Ab2 bio-conjugants onto electrode surface via the immune recognition. By virtue of the matched energy levels between CdTe QDs and AuNPs/g-C3N4, ALP-CdTe-Ab2 bio-conjugants could serve as the PEC active probes for photocurrent enhancement. Moreover, the photocurrent response could be further enhanced attributed to the ALP catalytic chemistry to in situ produce ascorbic acid for electron donating, achieving an effective dual signal-on mode for PEC assay. On the basis of the ALV-J titers-dependent photocurrent increment, the fabricated PEC immunosensor showed high sensitivity, specificity and stability for ALV-J assay in a wide linear range with a low detection limit of 85 TCID50/mL. This PEC immunosensor with the dual signal-on strategy may open up a promising platform for more target analytes in novel immune analysis and clinical diagnostics.


Assuntos
Vírus da Leucose Aviária/isolamento & purificação , Leucose Aviária/diagnóstico , Técnicas Biossensoriais , Técnicas Eletroquímicas , Fosfatase Alcalina/química , Fosfatase Alcalina/imunologia , Animais , Leucose Aviária/virologia , Vírus da Leucose Aviária/patogenicidade , Compostos de Cádmio/química , Elétrons , Ouro/química , Grafite/química , Imunoensaio , Limite de Detecção , Nanopartículas Metálicas/química , Compostos de Nitrogênio/química , Pontos Quânticos/química , Telúrio/química
19.
Microb Pathog ; 126: 40-44, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30366127

RESUMO

Aberrant expression of microRNAs (miRNAs) is known to be involved in cancer progression caused by subgroup J avian leukosis virus (ALV-J) in liver tissues. To advance our understanding of the related pathological mechanisms and virus-host interactions, seven previously reported miRNAs were selected for a comparative analysis of miRNA expression between infected and uninfected DF-1 cells, including six miRNAs related to tumorigenesis (let-7b/7i, miR-221/222, miR-125b, miR-375 and miR-2127. The results showed that six of the seven miRNAs except gga-miR-375 were upregulated in cells infected with NX0101 (caused myeloma (ML)) and GD1109 (caused hemangioma (HE)) at 1 h post infection. On day 2 post-infection, all seven miRNAs were upregulated in infected DF-1 cells. On day 6 post-infection, gga-let-7b, gga-miR-125b, and gga-miR-375 were downregulated whereas gga-miR-221 and gga-miR-222 were upregulated in DF-1 cells infected with the two ALV-J strains of different phenotypes. However, expression of gga-let-7i was reduced in DF-1 cells infected with NX0101 and was increased in those infected with GD1109; gga-miR-2127 expression showed no significant difference between infected and uninfected cells. This study is the first to report the changes in the miRNA expression levels in DF-1 cells during the course of ALV-J infection, and suggests a relationship between its pathological mechanisms and miRNAs.


Assuntos
Vírus da Leucose Aviária/genética , Vírus da Leucose Aviária/patogenicidade , Interações Hospedeiro-Patógeno/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Animais , Leucose Aviária/virologia , Carcinogênese , Linhagem Celular , Embrião de Galinha , Galinhas , Regulação para Baixo , Fibroblastos/virologia , Regulação da Expressão Gênica , Genes Virais , Doenças das Aves Domésticas/virologia
20.
PLoS One ; 13(10): e0204931, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30286182

RESUMO

Avian leukosis virus subgroup J (ALV-J) can induce myeloid tumors and hemangiomas in chickens and causes severe economic losses with commercial layer chickens and meat-type chickens. Here, we generated ribominus RNA sequencing data from three normal chicken spleen tissues and three ALV-J-infected chicken spleen tissues. Structure analysis of transcripts showed that, compared to mRNAs and lncRNAs, chicken circRNAs shared relatively shorter transcripts and similar GC content. Differentially expression analysis showed 152 differentially expressed circRNAs with 106 circRNAs up regulated and 46 circRNAs down regulated. Through comparing differentially expressed circRNA host genes and mRNAs and performed ceRNA network analysis, we found several tumor or immune-related genes, in which, there were four genes existed in both differentially expressed mRNAs and circRNA host genes (Dock4, Fmr1, Zfhx3, Ralb) and two genes (Mll, Aoc3) involved in ceRNA network. We further characterized one exon-intron circRNA derived from HRH4 gene in the ceRNA network, termed circHRH4, which is an abundant and stable circRNA expressed in various tissues and cells in chicken and localizes in cytoplasm. Our results provide new insight into the pathology of ALV-J infection and circRNAs may also mediate tumorigenesis in chicken.


Assuntos
Vírus da Leucose Aviária/patogenicidade , Carcinogênese/genética , Redes Reguladoras de Genes , Doenças das Aves Domésticas/virologia , RNA/genética , Animais , Galinhas , Feminino , Regulação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Doenças das Aves Domésticas/genética , RNA Circular , RNA Mensageiro/genética , Análise de Sequência de RNA/veterinária , Baço/patologia , Baço/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...