Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.336
Filtrar
1.
Viruses ; 16(7)2024 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-39066330

RESUMEN

Avian leukosis viruses (ALVs) include a group of avian retroviruses primarily associated with neoplastic diseases in poultry, commonly referred to as avian leukosis. Belonging to different subgroups based on their envelope properties, ALV subgroups A, B, and J (ALV-A, ALV-B, and ALV-J) are the most widespread in poultry populations. Early identification and removal of virus-shedding birds from infected flocks are essential for the ALVs' eradication. Therefore, the development of rapid, accurate, simple-to-use, and cost effective on-site diagnostic methods for the detection of ALV subgroups is very important. Cas13a, an RNA-guided RNA endonuclease that cleaves target single-stranded RNA, also exhibits non-specific endonuclease activity on any bystander RNA in close proximity. The distinct trans-cleavage activity of Cas13 has been exploited in the molecular diagnosis of multiple pathogens including several viruses. Here, we describe the development and application of a highly sensitive Cas13a-based molecular test for the specific detection of proviral DNA of ALV-A, B, and J subgroups. Prokaryotically expressed LwaCas13a, purified through ion exchange and size-exclusion chromatography, was combined with recombinase polymerase amplification (RPA) and T7 transcription to establish the SHERLOCK (specific high-sensitivity enzymatic reporter unlocking) molecular detection system for the detection of proviral DNA of ALV-A/B/J subgroups. This novel method that needs less sample input with a short turnaround time is based on isothermal detection at 37 °C with a color-based lateral flow readout. The detection limit of the assay for ALV-A/B/J subgroups was 50 copies with no cross reactivity with ALV-C/D/E subgroups and other avian oncogenic viruses such as reticuloendotheliosis virus (REV) and Marek's disease virus (MDV). The development and evaluation of a highly sensitive and specific visual method of detection of ALV-A/B/J nucleic acids using CRISPR-Cas13a described here will help in ALV detection in eradication programs.


Asunto(s)
Virus de la Leucosis Aviar , Leucosis Aviar , Sistemas CRISPR-Cas , ADN Viral , Provirus , Virus de la Leucosis Aviar/genética , Virus de la Leucosis Aviar/aislamiento & purificación , Virus de la Leucosis Aviar/clasificación , Animales , Provirus/genética , Provirus/aislamiento & purificación , Leucosis Aviar/virología , Leucosis Aviar/diagnóstico , ADN Viral/genética , Enfermedades de las Aves de Corral/virología , Enfermedades de las Aves de Corral/diagnóstico , Pollos/virología , Sensibilidad y Especificidad , Proteínas Asociadas a CRISPR/genética , Proteínas Asociadas a CRISPR/metabolismo
2.
Arch Virol ; 169(7): 155, 2024 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-38951272

RESUMEN

Given the high prevalence of avian leukosis virus subgroup K (ALV-K) in chickens in China, the positive rate of ALV-K in local chickens in Henan province was investigated, and the genetic region encoding the glycoprotein gp85 of isolates from positive chickens was analyzed. The positive rate of ALV-K in local chickens in Henan was found to be 87.2% (41/47). Phylogenetic analysis of gp85 sequences revealed six clusters that differed in their host range regions (hr1 and hr2) and variable regions (vr1, vr2, and vr3). Evidence of recombination of hr1, hr2, vr1, vr2, and vr3 was observed between the different clusters. The isolate HN23LS02 appears to have obtained its hr1 and hr2 regions from separate lineages via recombination but without having a significant affect on the replication capacity of the virus.


Asunto(s)
Virus de la Leucosis Aviar , Leucosis Aviar , Pollos , Especificidad del Huésped , Filogenia , Enfermedades de las Aves de Corral , Recombinación Genética , Proteínas del Envoltorio Viral , Animales , Virus de la Leucosis Aviar/genética , Virus de la Leucosis Aviar/clasificación , Virus de la Leucosis Aviar/aislamiento & purificación , Pollos/virología , Leucosis Aviar/virología , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Enfermedades de las Aves de Corral/virología , China
3.
Int J Biol Macromol ; 275(Pt 1): 133644, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38964687

RESUMEN

Apoptosis plays a crucial role in host antiviral defense. The avian leukosis virus subgroup J (ALV-J), an avian oncogenic retrovirus, has been shown to suppress apoptosis while promoting its own replication. ALV-J induces myeloid tumors and hemangiomas in chickens resulting in significant economic losses for commercial layer and meat-type chicken production. B-cell lymphoma/leukemia 11B (Bcl11b) encodes a C2H2-type zinc finger protein-BCL11B, that exerts critical functions in cell proliferation, differentiation, and plays an essential role in the immune system. Previous study has been shown that Bcl11b is associated with ALV-J infection. In this study, we further investigated the pathological changes in ALV-J infected cells and examined the role and expression regulation of chicken Bcl11b. Our results demonstrate that Bcl11b, as an interferon-stimulated gene (ISG), encodes C2H2-type zinc finger protein BCL11B that promotes apoptosis to inhibit ALV-J infection. Additionally, gga-miR-1612 and gga-miR-6701-3p regulate apoptosis and are involved in ALV-J infection by targeting Bcl11b, thus revealing immune response strategies between the host and ALV-J. Although the underlying mechanisms require further validation, Bcl11b and its regulatory miRNAs are the first to demonstrate inhibition of ALV-J replication via apoptosis. BCL11B can a valuable target for treating diseases triggered by ALV-J infection.


Asunto(s)
Apoptosis , Virus de la Leucosis Aviar , Leucosis Aviar , Pollos , Replicación Viral , Virus de la Leucosis Aviar/fisiología , Animales , Leucosis Aviar/virología , MicroARNs/genética , MicroARNs/metabolismo , Dedos de Zinc , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Regulación de la Expresión Génica
4.
Poult Sci ; 103(8): 103898, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38936216

RESUMEN

Exosome-mediated horizontal and vertical transmission of subgroup J avian leukosis virus (ALV-J) in poultry flocks can lead to growth inhibition and severe immunosuppression. However, there are few reports on the early infection of chicken embryonic stem cells (cESCs) with ALV-J. In this study, we confirmed that early infection with ALV-J can accelerate the differentiation of cESCs and promote the secretion of exosomes. To investigate the modulation strategy of ALV-J in cESCs, circRNA sequencing was performed for further analysis. A total of 305 differentially expressed circRNAs (DECs) were obtained, including 71 upregulated DECs. Circ-CCDC7 was found to be the most upregulated DEC and was assessed by qRT-PCR, with the result consistent with the result of circRNA-seq. Based on qRT-PCR, gga-miR-6568-3p was found to be the target of the top 3 DECs, including circ-CCDC7, and the stem cell marker gene Pax7 was identified as the target gene of gga-miR-6568-3p. This study demonstrated that exosomal circ-CCDC7/gga-miR-6568-3p/Pax7 accelerates the differentiation of cESCs after early infection with ALV-J.


Asunto(s)
Virus de la Leucosis Aviar , Leucosis Aviar , Diferenciación Celular , Pollos , Exosomas , MicroARNs , ARN Circular , Animales , Virus de la Leucosis Aviar/fisiología , Exosomas/metabolismo , Exosomas/virología , Exosomas/genética , ARN Circular/genética , ARN Circular/metabolismo , Leucosis Aviar/virología , MicroARNs/genética , MicroARNs/metabolismo , Enfermedades de las Aves de Corral/virología , Enfermedades de las Aves de Corral/genética , Células Madre Embrionarias/virología , Células Madre Embrionarias/fisiología , Embrión de Pollo , Proteínas Aviares/genética , Proteínas Aviares/metabolismo
5.
Poult Sci ; 103(7): 103835, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38772092

RESUMEN

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.


Asunto(s)
Virus de la Leucosis Aviar , Leucosis Aviar , Virus de la Anemia del Pollo , Pollos , Infecciones por Circoviridae , Coinfección , Transmisión Vertical de Enfermedad Infecciosa , Enfermedades de las Aves de Corral , Animales , Virus de la Leucosis Aviar/fisiología , Virus de la Leucosis Aviar/patogenicidad , Pollos/virología , Leucosis Aviar/virología , Coinfección/veterinaria , Coinfección/virología , Enfermedades de las Aves de Corral/virología , Virus de la Anemia del Pollo/fisiología , Virus de la Anemia del Pollo/patogenicidad , Infecciones por Circoviridae/veterinaria , Infecciones por Circoviridae/virología , Transmisión Vertical de Enfermedad Infecciosa/veterinaria , Virulencia , Embrión de Pollo
6.
Poult Sci ; 103(6): 103755, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38663206

RESUMEN

Avian leukosis virus subgroup K (ALV-K) is composed of newly emerging isolates, which cluster separately from the well-characterized subgroups A, B, C, D, E, and J in sequence analysis, and exhibits a specific host range and a unique pattern of superinfection interference. Avian leukosis virus subgroup K replicate more slowly in avian cells than other ALV strains, leading to escaped detection during ALV eradication, but the underlying mechanism are largely unknown. In our previous study, we have reported that JS11C1 and most of other suspected ALV-K strains possessed unique mutations in the U3 region. Here, we selected 5 mutations in some important transcriptional regulation elements to explore the possible factor contributing for the lower activity of LTR, including CA-TG mutation in the CAAT box, 21 nt deletion in the CAAT box, A-G and A-T mutations in the CArG boxes, 11 nt insertion in the PRE boxes, and C-T mutation in the TATA box. On the basis of infectious clone of JS11C1, we demonstrated that the 11 nt fragment in the PRE boxes was associated with the transcription activity of LTR, the enhancer ability of U3, and the replication capacity of the virus. Notably, we determined the differential U3-protein interaction profile of ALVs and found that the 11 nt fragment specifically binds to cellular SERPINE1 mRNA binding protein 1 (SERBP1) to increase the LTR activity and enhance virus replication. Collectively, these findings reveal that a 11 nt fragment in the U3 gene contributed to its binding ability to the cellular SERBP1 to enhance its transcription and the infectious virus productions in avian cells. This study highlighted the vital role of host factor in retrovirus replication and thus provides a new perspective to elucidate the interaction between retrovirus and its host and a molecular basis to develop efficient strategies against retroviruses.


Asunto(s)
Virus de la Leucosis Aviar , Leucosis Aviar , Pollos , Virus de la Leucosis Aviar/fisiología , Virus de la Leucosis Aviar/genética , Animales , Leucosis Aviar/virología , Enfermedades de las Aves de Corral/virología , Transcripción Genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Replicación Viral , Línea Celular , Mutación
7.
Virol J ; 21(1): 83, 2024 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-38600532

RESUMEN

BACKGROUND: Avian leukosis virus Subgroup-J (ALV-J) is a rapidly oncogenic evolving retrovirus infecting a variety of avian species; causing severe economic losses to the local poultry industry. METHODS: To investigate ALV-J, a total of 117 blood samples and 57 tissue specimens of different organs were collected for virological, and pathological identification, serological examinations, molecular characterization, and sequencing analysis. To the best of our knowledge, this is the first detailed report recorded in broiler flocks in Egypt. The present study targets the prevalence of a viral tumor disease circulating in broiler flocks in the El-Sharqia, El-Dakahliya, and Al-Qalyubiyya Egyptian governorates from 2021 to 2023 using different diagnostic techniques besides ALV-J gp85 genetic diversity determination. RESULT: We first isolated ALV-J on chicken embryo rough cell culture; showing aggregation, rounding, and degeneration. Concerning egg inoculation, embryonic death, stunting, and curling were observed. Only 79 serum samples were positive for ALV-J (67.52%) based on the ELISA test. Histopathological investigation showed tumors consist of uniform masses, usually well-differentiated myelocytes, lymphoid cells, or both in the liver, spleen, and kidneys. Immunohistochemical examination showed that the myelocytomatosis-positive signals were in the spleen, liver, and kidney. The PCR assay of ALV-J gp85 confirmed 545 base pairs with only 43 positive samples (75.4%). Two positive samples were sequenced and submitted to the Genbank with accession numbers (OR509852-OR509853). Phylogenetic analysis based on the gp85 gene showed that the ALV-J Dakahlia-2 isolate is genetically related to ALV-EGY/YA 2021.3, ALV-EGY/YA 2021.4, ALV-EGY/YA 2021.14, and ALV-EGY/YA 2021.9 with amino acid identity percentage 96%, 97%; 96%, 96%; respectively. Furthermore, ALV-J Sharqia-1 isolate is highly genetically correlated to ALV-EGY/YA 2021.14, and ALV-EGY/YA 2021.9, ALV-J isolate QL1, ALV-J isolate QL4, ALV-J isolate QL3, ALV-EGY/YA 2021.4 with amino acid identity percentage 97%, 97%; 98%, 97%, 97%, 95%; respectively. CONCLUSIONS: This study confirmed that ALV-J infection had still been prevalent in broilers in Egypt, and the genetic characteristics of the isolates are diverse.


Asunto(s)
Virus de la Leucosis Aviar , Leucosis Aviar , Enfermedades de las Aves de Corral , Embrión de Pollo , Animales , Pollos , Leucosis Aviar/patología , Virus de la Leucosis Aviar/genética , Egipto/epidemiología , Filogenia , Evolución Molecular , Aminoácidos/genética
8.
Poult Sci ; 103(6): 103671, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38569240

RESUMEN

N6-methyladenosine (m6A) methylation in transcripts has been suggested to influence tumorigenesis in liver tumors caused by the avian leukosis virus subgroup J (ALV-J). However, m6A modifications during ALV-J infection in vitro remain unclear. Herein, we performed m6A and RNA sequencing in ALV-J-infected chicken fibroblasts (DF-1). A total of 51 differentially expressed genes containing differentially methylated peaks were identified, which were markedly enriched in microRNAs (miRNAs) in cancer cells as well as apoptosis, mitophagy and autophagy, RNA degradation, and Hippo and MAPK signaling pathways. Correlation analysis indicated that YTHDC1 (m6A-reader gene) plays a key role in m6A modulation during ALV-J infection. The env gene of ALV-J harbored the strongest peak, and untranslated regions and long terminal repeats also contained peaks of different degrees. To the best of our knowledge, this is the first thorough analysis of m6A patterns in ALV-J-infected DF-1 cells. Combined with miRNA profiles, this study provides a useful basis for future research into the key pathways of ALV-J infection associated with m6A alteration.


Asunto(s)
Adenosina , Virus de la Leucosis Aviar , Leucosis Aviar , Pollos , MicroARNs , Enfermedades de las Aves de Corral , Transcriptoma , Animales , Virus de la Leucosis Aviar/fisiología , MicroARNs/genética , MicroARNs/metabolismo , Leucosis Aviar/virología , Enfermedades de las Aves de Corral/virología , Enfermedades de las Aves de Corral/genética , Adenosina/análogos & derivados , Adenosina/metabolismo , Fibroblastos/virología
9.
Poult Sci ; 103(6): 103693, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38598912

RESUMEN

Avian leukosis virus subgroup J (ALV-J) is a retrovirus that can cause immunosuppression and tumors in chicken. However, relative pathogenesis is still not clear. At present, metabolomics has shown great potential in the screening of tumor metabolic markers, prognostic evaluation, and drug target design. In this study, we utilize an untargeted metabolomics approach based on ultrahigh-performance liquid chromatography-quadrupole time-of-flight tandem mass spectrometry (UHPLC-QTOF-MS) to analyze the metabolic changes in chicken embryo fibroblast (CEF) cells infected by ALV-J. We found that ALV-J infection significantly altered a wealth of metabolites compared with control group. Additionally, most of the differentially expressed metabolites belonged to lipid metabolism, purine nucleotide metabolism and amino acid metabolism. Among them, the proportion of lipid metabolites account for the highest proportion (around 31%). Results suggest that these changes may be conductive to the formation of virion, thereby promoting the replication of ALV-J. These data provided metabolic evidence and potential biomarkers for the cellular metabolic changes induced by ALV-J, and provided important insight for further understanding the replication needs and pathogenesis of ALV-J.


Asunto(s)
Virus de la Leucosis Aviar , Fibroblastos , Metabolómica , Enfermedades de las Aves de Corral , Animales , Virus de la Leucosis Aviar/fisiología , Metabolómica/métodos , Embrión de Pollo , Fibroblastos/virología , Cromatografía Líquida de Alta Presión/veterinaria , Enfermedades de las Aves de Corral/virología , Espectrometría de Masas en Tándem/veterinaria , Leucosis Aviar/virología , Pollos , Metaboloma
10.
Poult Sci ; 103(6): 103617, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38547674

RESUMEN

Avian leukosis virus Subgroup J (ALV-J) exhibits high morbidity and pathogenicity, affecting approximately 20% of poultry farms. It induces neoplastic diseases and immunosuppression. Phorbol-12-myristate-13-acetate-induced protein 1 (PMAIP1), a proapoptotic mitochondrial protein in the B-cell lymphoma-2 (Bcl-2) family, plays a role in apoptosis in cancer cells. However, the connection between the PMAIP1 gene and ALV-J pathogenicity remains unexplored. This study investigates the potential impact of the PMAIP1 gene on ALV-J replication and its regulatory mechanisms. Initially, we examined PMAIP1 expression using quantitative real-time PCR (qRT-PCR) in vitro and in vivo. Furthermore, we manipulated PMAIP1 expression in chicken fibroblast cells (DF-1) and assessed its effects on ALV-J infection through qRT-PCR, immunofluorescence assay (IFA), and western blotting (WB). Our findings reveal a significant down-regulation of PMAIP1 in the spleen, lung, and kidney, coupled with an up-regulation in the bursa and liver of ALV-J infected chickens compared to uninfected ones. Additionally, DF-1 cells infected with ALV-J displayed a notable up-regulation of PMAIP1 at 6, 12, 24, 48, 74, and 108 h. Over-expression of PMAIP1 enhanced ALV-J replication, interferon expression, and proinflammatory factors. Conversely, interference led to contrasting results. Furthermore, we observed that PMAIP1 promotes virus replication by modulating mitochondrial function. In conclusion, the PMAIP1 gene facilitates virus replication by regulating mitochondrial function, thereby enriching our understanding of mitochondria-related genes and their involvement in ALV-J infection, offering valuable insights for avian leukosis disease resistance strategies.


Asunto(s)
Virus de la Leucosis Aviar , Leucosis Aviar , Pollos , Mitocondrias , Enfermedades de las Aves de Corral , Replicación Viral , Animales , Virus de la Leucosis Aviar/fisiología , Enfermedades de las Aves de Corral/virología , Enfermedades de las Aves de Corral/genética , Mitocondrias/metabolismo , Leucosis Aviar/virología , Proteínas Aviares/genética , Proteínas Aviares/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo
11.
Vet Microbiol ; 291: 110012, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38387235

RESUMEN

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


Asunto(s)
Virus de la Leucosis Aviar , Leucosis Aviar , Animales , Embrión de Pollo , Virus de la Leucosis Aviar/genética , Virus de la Leucosis Aviar/metabolismo , Pollos , Mamíferos , Provirus , Transducción de Señal , Ubiquitinas , Factor de Transcripción STAT3/metabolismo , Factores Reguladores del Interferón/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo
12.
PLoS Pathog ; 20(2): e1011928, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38324558

RESUMEN

The subgroup J avian leukosis virus (ALV-J), a retrovirus, uses its gp85 protein to bind to the receptor, the chicken sodium hydrogen exchanger isoform 1 (chNHE1), facilitating viral invasion. ALV-J is the main epidemic subgroup and shows noteworthy mutations within the receptor-binding domain (RBD) region of gp85, especially in ALV-J layer strains in China. However, the implications of these mutations on viral replication and transmission remain elusive. In this study, the ALV-J layer strain JL08CH3-1 exhibited a more robust replication ability than the prototype strain HPRS103, which is related to variations in the gp85 protein. Notably, the gp85 of JL08CH3-1 demonstrated a heightened binding capacity to chNHE1 compared to HPRS103-gp85 binding. Furthermore, we showed that the specific N123I mutation within gp85 contributed to the enhanced binding capacity of the gp85 protein to chNHE1. Structural analysis indicated that the N123I mutation primarily enhanced the stability of gp85, expanded the interaction interface, and increased the number of hydrogen bonds at the interaction interface to increase the binding capacity between gp85 and chNHE1. We found that the N123I mutation not only improved the viral replication ability of ALV-J but also promoted viral shedding in vivo. These comprehensive data underscore the notion that the N123I mutation increases receptor binding and intensifies viral replication.


Asunto(s)
Virus de la Leucosis Aviar , Leucosis Aviar , Enfermedades de las Aves de Corral , Animales , Virus de la Leucosis Aviar/genética , Virus de la Leucosis Aviar/química , Mutación , Pollos , Isoformas de Proteínas/genética , Proteínas del Envoltorio Viral/genética
13.
BMC Vet Res ; 20(1): 41, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38302973

RESUMEN

The coinfection of ALVs (ALV-J plus ALV-A or/and ALV-B) has played an important role in the incidence of tumors recently found in China in local breeds of yellow chickens. The study aims to obtain a better knowledge of the function and relevance of ALV coinfection in the clinical disease of avian leukosis, as well as its unique effect on the pathogenicity in Three-yellow chickens. One-day-old Three-yellow chicks (one day old) were infected with ALV-A, ALV-B, and ALV-J mono-infections, as well as ALV-A + J, ALV-B + J, and ALV-A + B + J coinfections, via intraperitoneal injection, and the chicks were then grown in isolators until they were 15 weeks old. The parameters, including the suppression of body weight gain, immune organ weight, viremia, histopathological changes and tumor incidence, were observed and compared with those of the uninfected control birds. The results demonstrated that coinfection with ALVs could induce more serious suppression of body weight gain (P < 0.05), damage to immune organs (P < 0.05) and higher tumor incidences than monoinfection, with triple infection producing the highest pathogenicity. The emergence of visible tumors and viremia occurred faster in the coinfected birds than in the monoinfected birds. These findings demonstrated that ALV coinfection resulted in considerably severe pathogenic and immunosuppressive consequences.


Asunto(s)
Virus de la Leucosis Aviar , Leucosis Aviar , Coinfección , Neoplasias , Enfermedades de las Aves de Corral , Animales , Pollos , Coinfección/veterinaria , Virulencia , Viremia/veterinaria , Leucosis Aviar/epidemiología , Neoplasias/veterinaria , Peso Corporal , Enfermedades de las Aves de Corral/epidemiología
14.
J Virol ; 97(11): e0093723, 2023 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-37909729

RESUMEN

IMPORTANCE: The synergy of two oncogenic retroviruses is an essential phenomenon in nature. The synergistic replication of ALV-J and REV in poultry flocks increases immunosuppression and pathogenicity, extends the tumor spectrum, and accelerates viral evolution, causing substantial economic losses to the poultry industry. However, the mechanism of synergistic replication between ALV-J and REV is still incompletely elusive. We observed that microRNA-155 targets a dual pathway, PRKCI-MAPK8 and TIMP3-MMP2, interacting with the U3 region of ALV-J and REV, enabling synergistic replication. This work gives us new targets to modulate ALV-J and REV's synergistic replication, guiding future research on the mechanism.


Asunto(s)
Virus de la Leucosis Aviar , Leucosis Aviar , MicroARNs , Enfermedades de las Aves de Corral , Virus de la Reticuloendoteliosis , Animales , Virus de la Reticuloendoteliosis/genética , Virus de la Leucosis Aviar/genética , Pollos , MicroARNs/genética , Replicación Viral
15.
Viruses ; 15(9)2023 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-37766196

RESUMEN

Avian leukosis (AL), caused by avian leukosis virus (ALV), is a contagious tumor disease that results in significant economic losses for the poultry industry. Currently, ALV-A, B, J, and K subgroups are the most common in commercial poultry and cause possible coinfections. Therefore, close monitoring is necessary to avoid greater economic losses. In this study, a novel multiplex quantitative polymerase chain reaction (qPCR) assay was developed to detect ALV-A, ALV-B, ALV-J, and ALV-K with limits of detection of 40, 11, 13.7, and 96 copies/µL, respectively, and no cross-reactivity with other ALV subtypes and avian pathogens. We detected 852 cell cultures inoculated with clinical samples using this method, showing good consistency with conventional PCR and ELISA. The most prevalent ALV strain in Hubei Province, China, was still ALV-J (11.74%). Although single infections with ALV-A, ALV-B, and ALV-K were not found, coinfections with different subgroup strains were identified: 0.7% for ALV-A/J, 0.35% for ALV-B/J, 0.25% for ALV-J/K, and 0.12% for ALV-A/B/K and ALV-A/B/J. Therefore, our novel multiplex qPCR may be a useful tool for molecular epidemiology, clinical detection of ALV, and ALV eradication programs.


Asunto(s)
Virus de la Leucosis Aviar , Leucosis Aviar , Coinfección , Animales , Virus de la Leucosis Aviar/genética , Coinfección/diagnóstico , Coinfección/veterinaria , Leucosis Aviar/diagnóstico , Técnicas de Cultivo de Célula , Reacción en Cadena de la Polimerasa Multiplex
16.
Vet Microbiol ; 284: 109821, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37536160

RESUMEN

While the presence of host cell proteins in virions and their role in viral life cycles have been demonstrated in various viruses, such characteristics have remained largely unknown in avian leukosis virus (ALV). To investigate whether this is the case in ALV, we purified high-integrity and high-purity virions from the avian leukosis virus subgroup J (ALV-J) and subjected them to proteome analysis using nano LC-MS/MS. This analysis identified 53 cellular proteins that are incorporated into mature ALV-J virions, and we verified the reliability of the packaged cellular proteins through subtilisin digestion and immunoblot analysis. Functional annotation revealed the potential functions of these proteins in the viral life cycle and tumorigenesis. Overall, our findings have important implications for understanding the interaction between ALV-J and its host, and provide new insights into the cellular requirements that define ALV-J infection.


Asunto(s)
Virus de la Leucosis Aviar , Leucosis Aviar , Animales , Pollos , Virus de la Leucosis Aviar/genética , Espectrometría de Masas en Tándem/veterinaria , Proteómica , Reproducibilidad de los Resultados
17.
J Virol ; 97(8): e0026723, 2023 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-37582207

RESUMEN

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


Asunto(s)
Virus de la Leucosis Aviar , Leucosis Aviar , Ribonucleósido Difosfato Reductasa , Vía de Señalización Wnt , Animales , Virus de la Leucosis Aviar/metabolismo , beta Catenina/metabolismo , Proteínas de la Cápside/metabolismo , Pollos , Ribonucleósido Difosfato Reductasa/metabolismo
18.
Avian Pathol ; 52(4): 264-276, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37194644

RESUMEN

We previously described cardiomyocyte abnormality caused by Km_5666 strain, a variant of fowl glioma-inducing virus (FGV) prototype, which is an avian leukosis virus (ALV). However, the cardiac involvement appeared to be eradicated from the flock after a few years. An epidemiological survey from 2017 to 2020 was performed to elucidate the current prevalence of the cardiopathogenic strains in this flock. Four of the 71 bantams pathologically examined showed both glioma and cardiomyocyte abnormality, from which three ALV strains were detected. DNA sequencing revealed that several different ALV strains coexisted in each bantam and that the conserved Km_5666 virus fluid also contained at least two different ALV strains. We generated three infectious molecular clones from these samples, named KmN_77_clone_A, KmN_77_clone_B, and Km_5666_clone. The envSU of KmN_77_clone_A shared high sequence identity with that of Km_5666 (94.1%). In contrast, the envSU of KmN_77_clone_B showed >99.2% nucleotide similarity with that of an FGV variant without cardiopathogenicity. Furthermore, Km_5666_clone experimentally reproduced both gliomas and cardiomyocyte abnormality in chickens. From these results, it is suggested that the pathogenic determinant of cardiomyocyte abnormality is located in envSU similar to that of Km_5666. The cloning technique described here is beneficial for evaluating the viral pathogenicity in cases where affected birds are coinfected with several different ALV strains.


Asunto(s)
Virus de la Leucosis Aviar , Leucosis Aviar , Glioma , Enfermedades de las Aves de Corral , Animales , Virus de la Leucosis Aviar/genética , Pollos , Glioma/veterinaria , Células Clonales/patología
19.
Avian Dis ; 67(1): 102-107, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-37140118

RESUMEN

The U.S. Department of Agriculture Avian Disease and Oncology Laboratory currently relies on live birds of specific genetic backgrounds for producing chicken-embryo fibroblasts that are used for the diagnosis and subtyping of field isolates associated with avian leukosis virus (ALV) outbreaks. As an alternative to maintaining live animals for this purpose, we are currently developing cell lines capable of achieving the same result by ablation of the entry receptors utilized by ALV strains. We used CRISPR-Cas9 on the cell fibroblast-derived cell line DF-1 to disrupt the tva gene, which encodes the receptor required for binding and entry of ALV-A into cells. We ultimately identified seven DF-1 clones that had biallelic and homozygous indels at the Cas9 target site, exon 2 of tva. When tested in vitro for their ability to host ALV-A, the five clones that had frameshift mutations that disrupted the Tva protein were unable to support ALV-A replication. This result clearly demonstrates that modified cell lines can be used as part of a battery of tests to determine ALV subtype for isolate characterization, thus eliminating the need for live birds.


Nota de investigación- La ablación dirigida del exón 2 del gene del receptor del virus de la leucosis aviar A (ALV-A) en una línea celular de fibroblastos de pollo mediante CRISPR anula la infección por ALV-A. El Laboratorio de Oncología y Enfermedades Aviares del Departamento de Agricultura de los Estados Unidos. actualmente depende de aves vivas con antecedentes genéticos específicos para producir fibroblastos de embrión de pollo que se utilizan para el diagnóstico y la subtipificación de aislamientos de campo asociados con brotes del virus de la leucosis aviar (ALV). Como alternativa al mantenimiento de animales vivos para este propósito, actualmente se están desarrollando líneas celulares capaces de lograr el mismo resultado mediante la ablación de los receptores de entrada utilizados por las cepas ALV. Se utilizó el método repeticiones palindrómicas cortas agrupadas y regularmente interespaciadas o CRISPR-Cas9 en la línea celular DF-1 derivada de fibroblastos para interrumpir el gene Tva, que codifica el receptor requerido para la unión y entrada de ALV-A en las células. Finalmente, se identificaron siete clones de DF-1 que tenían inserciones y deleciones (indeles) bialélicos y homocigóticos en el sitio blanco Cas9, exón 2 del gene tva. Cuando se probó in vitro su capacidad para albergar ALV-A, los cinco clones que tenían mutaciones que involucraban al marco de lectura y que interrumpieron la proteína Tva no pudieron admitir la replicación de ALVA. Este resultado demuestra claramente que las líneas celulares modificadas se pueden utilizar como parte de una batería de pruebas para determinar el subtipo de ALV para la caracterización de los aislamientos, eliminando así la necesidad de aves vivas.


Asunto(s)
Virus de la Leucosis Aviar , Leucosis Aviar , Enfermedades de las Aves de Corral , Animales , Pollos , Virus de la Leucosis Aviar/genética , Línea Celular , Exones , Fibroblastos
20.
Infect Genet Evol ; 109: 105415, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36775048

RESUMEN

Tibetan chicken is found in China Tibet (average altitude; ˃4500 m). However, little is known about avian leukosis virus subgroup J (ALV-J) found in Tibetan chickens. ALV-J is a typical alpharetrovirus that causes immunosuppression and myelocytomatosis and thus seriously affects the development of the poultry industry. In this study, Tibet-origin mutant ALV-J was isolated from Tibetan chickens and named RKZ-1-RKZ-5. A Myelocytomatosis outbreak occurred in a commercial Tibetan chicken farm in Shigatse of Rikaze, Tibet, China, in March 2022. About 20% of Tibetan chickens in the farm showed severe immunosuppression, and mortality increased to 5.6%. Histopathological examination showed typical myelocytomas in various tissues. Virus isolation and phylogenetic analysis demonstrated that ALV-J caused the disease. Gene-wide phylogenetic analysis showed the RKZ isolates were the original strains of the previously reported Tibetan isolates (TBC-J4 and TBC-J6) (identity; 94.5% to 94.9%). Furthermore, significant nucleotide mutations and deletions occurred in the hr1 and hr2 hypervariable regions of gp85 gene, 3'UTR, Y Box, and TATA Box of 3'LTR. Pathogenicity experiments demonstrated that the viral load, viremia, and viral shedding level were significantly higher in RKZ-1-infected chickens than in NX0101-infected chickens. Notably, RKZ-1 caused more severe cardiopulmonary damage in SPF chickens. These findings prove the origin of Tibet ALV-J and provide insights into the molecular characteristics and pathogenic ability of ALV-J in the plateau area. Therefore, this study may provide a basis for ALV-J prevention and eradication in Tibet.


Asunto(s)
Virus de la Leucosis Aviar , Leucosis Aviar , Enfermedades de las Aves de Corral , Animales , Pollos , Tibet/epidemiología , Filogenia , Virulencia/genética , China/epidemiología , Leucosis Aviar/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...