Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Poult Sci ; 103(2): 103370, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38150831

RESUMEN

The past few years have witnessed a rapid increase in cases of viral arthritis caused by avian reovirus (ARV) in chicken farms in China, attributed to the emergence of variant strains that render traditional vaccines ineffective, leading to substantial economic losses. In this study, we successfully isolated a novel ARV strain, designated as 2023ARV-GS-SDAU-1, from chickens in a broiler flock vaccinated with an ARV vaccine in Gansu province. We performed whole-genome sequencing and assessed its pathogenicity through 2 infection routes: oral administration and intraperitoneal injection. Our analysis revealed significant variations in the σA gene, associated with the inhibition of interferon secretion, compared to known ARV strains. The highest nucleotide identity observed was below 80%. Additionally, the σC gene exhibited notable variations compared to its homologous strains within the same group. Multiple alignment of the amino acid sequences classified the 2023ARV-GS-SDAU-1 strain under genotype I. Furthermore, our pathogenicity experiments indicated that the isolated strain exhibited more severe pathogenicity when administered via intraperitoneal injection in SPF chickens. In summary, our data suggest that the 2023ARV-GS-SDAU-1 strain represents a novel variant circulating in broiler flocks in China. These findings enrich currently available genetic information on ARV strains and provide a new complete genome sequence.


Asunto(s)
Orthoreovirus Aviar , Enfermedades de las Aves de Corral , Infecciones por Reoviridae , Animales , Orthoreovirus Aviar/genética , Virulencia , Pollos , Enfermedades de las Aves de Corral/epidemiología , Infecciones por Reoviridae/epidemiología , Infecciones por Reoviridae/veterinaria , Filogenia
2.
Vet Res ; 54(1): 117, 2023 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-38066626

RESUMEN

It is well-established that the genetic diversity, regional prevalence, and broad host range of astroviruses significantly impact the poultry industry. In July 2022, a small-scale commercial broiler farm in China reported cases of growth retardation and a 3% mortality rate. From chickens displaying proventriculitis and pancreatitis, three chicken astroviruses (CAstV) isolates were obtained and named SDAU2022-1-3. Complete genomic sequencing and analysis revealed the unique characteristics of these isolates from known CAstV strains in ORF1a, ORF1b, and ORF2 genes, characterized by an unusually high variability. Analysis of amino acid mutations in ORF1a, ORF1b, and ORF2 indicated that the accumulation of these mutations played a pivotal role in the emergence of the variant strain. Inoculation experiments demonstrated that affected chickens exhibited liver and kidney enlargement, localized proventricular hemorrhage, and a dark reddish-brown appearance in about two-thirds of the pancreas. Histopathological examination unveiled hepatic lymphocytic infiltration, renal tubular epithelial cell swelling, along with lymphocytic proventriculitis and pancreatitis. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) analysis indicated viremia and viral shedding at 3 days post-infection (dpi). The proventriculus displayed the highest viral loads, followed by the liver, kidney, duodenum, and pancreas. Liver parameters (AST and ALT) and kidney parameters (UA and UN) demonstrated mild damage consistent with earlier findings. While the possibility of new mutations in the ORF2 gene of CAstV causing proventriculitis and pancreatitis warrants further investigation, these findings deepen our comprehension of CAstV's pathogenicity in chickens. Additionally, they serve as valuable references for subsequent research endeavors.


Asunto(s)
Infecciones por Astroviridae , Avastrovirus , Pancreatitis , Enfermedades de las Aves de Corral , Animales , Avastrovirus/genética , Pollos , Virulencia , Infecciones por Astroviridae/veterinaria , Infecciones por Astroviridae/epidemiología , Pancreatitis/veterinaria , Filogenia
3.
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
4.
Vet Sci ; 10(5)2023 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-37235395

RESUMEN

Gyrovirus homsa1 (GyH1) is an emerging pathogenic single-stranded circular DNA virus that leads to immunosuppression, aplastic anemia, and multisystem damage in chickens. However, the prevalence of GyH1 infection in chickens and wild birds remains unknown. Here, we developed a double-antibody sandwich enzyme-linked immunosorbent assay (DAS-ELISA) to investigate GyH1 infection in 8 chicken species and 25 wild bird species. A total of 2258 serum samples from chickens (n = 2192) in 15 provinces, and wild birds (n = 66) in Jinan Wildlife Hospital were collected from 2017 to 2021 in China. The GyH1-positive rates in chickens and wild birds were 9.3% (203/2192) and 22.7% (15/66), respectively. GyH1 was present in all flocks in 15 provinces. From 2017 to 2021, the positive rate ranged from 7.93% (18/227) to 10.67% (56/525), and the highest positive rate was present in 2019. Upon chicken age, the highest positive rate (25.5%) was present in young chickens (14-35 days old). Moreover, the GyH1-positive rate in broiler breeders (12.6%, 21/167) was significantly higher than that in layer chickens (8.9%, 14/157). This study shows that GyH1 has spread in chicken flocks and wild birds, and the higher GyH1-positive rate in wild birds indicates the risk of spillover from wild birds to chickens. Our study expanded the GyH1 epidemiological aspects and provided a theoretical basis for GyH1 prevention.

5.
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
6.
Virus Res ; 326: 199044, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36652973

RESUMEN

Serum amyloid A (SAA), an acute response phase protein (APP), is crucial for the innate immune response during pathogenic microorganisms' invasion. Marek's disease virus (MDV) is a highly oncogenic alphaherpesvirus that activates multiple innate immune molecules, including SAA, in the host during infection. However, the pathway through which SAA participates in MDV-induced host innate immunity remains unknown. The present study aimed to elucidate the pathway through which SAA exerts its anti-MDV function. We observed that MDV infection in vivo and in vitro significantly elevated SAA expression. Furthermore, through SAA overexpression and knockdown experiments, we demonstrated that SAA could inhibit MDV replication. Subsequently, we found that SAA activated Toll-Like Receptor 2/4 (TLR2/4) -mediated Interferon Beta (IFN-ß) promoter activity and IFN regulatory factor 7 (IRF7) promoter activity. During MDV infection, SAA enhanced TLR2/4-mediated IFN-ß signal transduction and messenger RNAs (mRNAs) expression of type I IFN (IFN-I) and interferon-stimulated genes (ISGs). Finally, TLR2/4 inhibitor OxPAPC inhibits the anti-MDV activity of SAA. These results demonstrated that SAA inhibits MDV replication and enhancing TLR2/4-mediated IFN-ß signal transduction to promote IFNs and ISGs expression. This finding is the first to demonstrate the signaling pathway by which SAA exerts its anti-MDV function. It also provides new insights into the control of oncogenic herpesviruses from the perspective of acute response phase proteins.


Asunto(s)
Herpesvirus Gallináceo 2 , Interferón Tipo I , Enfermedad de Marek , Animales , Pollos , Herpesvirus Gallináceo 2/genética , Interferón Tipo I/metabolismo , Interferón beta/genética , Interferón beta/metabolismo , Enfermedad de Marek/genética , Proteína Amiloide A Sérica/genética , Proteína Amiloide A Sérica/metabolismo , Transducción de Señal , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo
7.
Cells ; 11(20)2022 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-36291177

RESUMEN

Synergism between avian leukosis virus subgroup J (ALV-J) and reticuloendotheliosis virus (REV) has been reported frequently in co-infected chicken flocks. Although significant progress has been made in understanding the tumorigenesis mechanisms of ALV and REV, how these two simple oncogenic retroviruses induce synergistic oncogenicity remains unclear. In this study, we found that ALV-J and REV synergistically promoted mutual replication, suppressed cellular senescence, and activated epithelial-mesenchymal transition (EMT) in vitro. Mechanistically, structural proteins from ALV-J and REV synergistically activated the expression of Musashi-1(MSI1), which directly targeted pri-miR-147 through its RNA binding site. This inhibited the maturation of miR-147, which relieved the inhibition of NF-κB/KIAA1199/EGFR signaling, thereby suppressing cellular senescence and activating EMT. We revealed a synergistic oncogenicity mechanism induced by ALV-J and REV in vitro. The elucidation of the synergistic oncogenicity of these two simple retroviruses could help in understanding the mechanism of tumorigenesis in ALV-J and REV co-infection and help identify promising molecular targets and key obstacles for the joint control of ALV-J and REV and the development of clinical technologies.


Asunto(s)
Virus de la Leucosis Aviar , Coinfección , MicroARNs , Enfermedades de las Aves de Corral , Animales , Enfermedades de las Aves de Corral/genética , FN-kappa B , Virus de la Leucosis Aviar/genética , Pollos/genética , MicroARNs/genética , Carcinogénesis/genética , Receptores ErbB
8.
Virulence ; 13(1): 1184-1198, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35795905

RESUMEN

Co-infection of Marek's disease virus (MDV) and reticuloendotheliosis virus (REV) synergistically drives disease progression, yet little is known about the mechanism of the synergism. Here, we found that co-infection of REV and MDV increased their replication via the RIOK3-Akt pathway. Initially, we noticed that the viral titres of MDV and REV significantly increased in REV and MDV co-infected cells compared with single-infected cells. Furthermore, tandem mass tag peptide labelling coupled with LC/MS analysis showed that Akt was upregulated in REV and MDV co-infected cells. Overexpression of Akt promoted synergistic replication of MDV and REV. Conversely, inhibition of Akt suppressed synergistic replication of MDV and REV. However, PI3K inhibition did not affect synergistic replication of MDV and REV, suggesting that the PI3K/Akt pathway is not involved in the synergism of MDV and REV. In addition, we revealed that RIOK3 was recruited to regulate Akt in REV and MDV co-infected cells. Moreover, wild-type RIOK3, but not kinase-dead RIOK3, mediated Akt phosphorylation and promoted synergistic replication of MDV and REV. Our results illustrate that MDV and REV activated a novel RIOK3-Akt signalling pathway to facilitate their synergistic replication.


Asunto(s)
Coinfección , Herpesvirus Gallináceo 2 , Enfermedad de Marek , Proteínas Serina-Treonina Quinasas/metabolismo , Virus de la Reticuloendoteliosis , Animales , Pollos , Enfermedades Genéticas Ligadas al Cromosoma X , Herpesvirus Gallináceo 2/metabolismo , Humanos , Enfermedad de Marek/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Virus de la Reticuloendoteliosis/genética , Virus de la Reticuloendoteliosis/metabolismo , Inmunodeficiencia Combinada Grave , Replicación Viral/fisiología
9.
BMC Vet Res ; 18(1): 253, 2022 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-35768837

RESUMEN

BACKGROUND: Transmissible viral proventriculitis (TVP) causes significant economic loss to the poultry industry. However, the exact causative agents are obscure. Here we examine the virome of proventriculus from specified pathogen free (SPF) chickens that reproduced by infection of proventricular homogenate from broiler chicken with TVP using long read sequencing of the Pacific Biosciences RSII platform. The normal SPF chickens were used as control. RESULTS: Our investigation reveals a virome of proventriculitis, including three Gyrovirus genera of the Aneloviridae: Gyrovirus homsa1 (GyH1) (also known as Gyrovirus 3, GyV3) (n = 2662), chicken anemia virus (CAV) (n = 482) and Gyrovirus galga1 (GyG1) (also known as avian Gyrovirus 2, AGV2) (n = 11); a plethora of novel CRESS viral genomes (n = 26) and a novel genomovirus. The 27 novel viruses were divided into three clusters. Phylogenetic analysis showed that the GyH1 strain was more closely related to the strains from chicken (MG366592) than mammalian (human and cat), the GyG1 strain was closely related to the strains from cat in China (MK089245) and from chicken in Brazil (HM590588), and the CAV strain was more closely related to the strains from Germany (AJ297684) and United Kingdom (U66304) than that previously found in China. CONCLUSION: In this study, we revealed that Gyrovirus virome showed high abundance in chickens with TVP, suggesting their potential role in TVP, especially GyH1. This study is expected to contribute to the knowledge of the etiology of TVP.


Asunto(s)
Virus de la Anemia del Pollo , Gyrovirus , Enfermedades de las Aves de Corral , Gastropatías , Animales , Virus de la Anemia del Pollo/genética , Pollos , Mamíferos , Filogenia , Proventrículo , Gastropatías/veterinaria , Viroma
10.
BMC Vet Res ; 18(1): 231, 2022 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-35717195

RESUMEN

BACKGROUND: Gyrovirus homsa1 (GyH1) (also known as Gyrovirus 3, GyV3) is a non-enveloped, small, single-stranded DNA virus, which was first identified in children with acute diarrhea, and was subsequently detected in marketed chickens, broilers with transmissible viral proventriculitis (TVP), and mammals. GyH1 is a pathogenic virus in chickens, causing aplastic anemia, immunosuppression, and multisystem damage. However, the seroepidemiology of GyH1 infection in chickens remains unclear. Here, we investigated the seroprevalence of GyH1 in chickens by ELISA to reveal the endemic status of GyH1 in China. RESULTS: An indirect ELISA with high sensitivity and specificity was developed for investigation of seroepidemiology of GyH1 in chickens in China. The seropositive rate of GyH1 ranged from 0.6% to 7.7% in thirteen provinces, and ranged from 4.1% to 8.1% in eight species chickens. The seropositive rate of GyH1 in broiler breeders was significantly higher than that of in layers. There was a negative correlation between seropositive rate and age of chickens. The highest and lowest seropositive rate were present in chickens at 30-60 days and over 180 days, respectively. CONCLUSIONS: The seroepidemiological investigation results demonstrated that natural GyH1 infection is widespread in chickens in China. Different species showed different susceptibility for GyH1. Aged chickens showed obvious age-resistance to GyH1. GyH1 has shown a high risk to the poultry industry and should be highly concerned.


Asunto(s)
Gyrovirus , Enfermedades de las Aves de Corral , Animales , Pollos , China/epidemiología , Gyrovirus/genética , Mamíferos , Aves de Corral , Estudios Seroepidemiológicos
11.
BMC Vet Res ; 18(1): 131, 2022 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-35379256

RESUMEN

BACKGROUND: Co-infection with the avian leukosis virus subgroup J (ALV-J) and the reticuloendotheliosis virus (REV) increases mutual viral replication, causing a more serious pathogenic effect by accelerating the progression of neoplasia and extending the tumor spectrum. However, the molecular mechanism underlying the synergistic replication of ALV-J and REV remains unclear. RESULTS: Here, we performed this study to compare the differentially expressed proteins among CEF cells infected with ALV-J, REV or both at the optimal synergistic infection time using TMT-based quantitative proteomics. We identified a total of 719 (292 upregulated and 427 downregulated) and 64 (35 upregulated and 29 downregulated) proteins by comparing co-infecting both viruses with monoinfecting ALV-J and REV, respectively. GO annotation and KEGG pathway analysis showed the differentially expressed proteins participated in virus-vector interaction, biological adhesion and immune response pathways in the synergistic actions of ALV-J and REV at the protein levels. Among the differentially expressed proteins, a large number of integrins were inhibited or increased in the co-infection group. Further, eight integrins, including ITGα1, ITGα3, ITGα5, ITGα6, ITGα8, ITGα9, ITGα11 and ITGß3, were validated in CEF cells by qRT-PCR or western blot. CONCLUSIONS: These findings proved that integrins may be key regulators in the mechanism of synergistic infection of REV and ALV-J, which will provide more insight into the pathogenesis of synergism of REV and ALV-J at protein level.


Asunto(s)
Virus de la Leucosis Aviar , Virus de la Reticuloendoteliosis , Animales , Virus de la Leucosis Aviar/fisiología , Pollos , Integrinas/genética , Proteómica , Virus de la Reticuloendoteliosis/genética
12.
Front Vet Sci ; 9: 854007, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35392111

RESUMEN

Coinfection with Marek's disease virus (MDV) and reticuloendotheliosis virus (REV) causes synergistic pathogenic effects and serious losses to the poultry industry. However, whether there is a synergism between the two viruses in viral replication and the roles of host factors in regulating MDV and REV coinfection remains elusive. In this study, we found that MDV and REV coinfection increased viral replication in coinfected cells as compared to a single infection in a limited period. Further, we explore the host cell responses to MDV and REV coinfection using tandem mass tag (TMT) peptide labeling coupled with liquid chromatography-tandem mass spectrometry (LC-MS/MS). Compared with MDV/REV-infected cells, 38 proteins increased (fold change > 1.2) and 60 decreased (fold change < 0.83) their abundance in MDV and REV coinfected cells. Differentially accumulated proteins (DAPs) were involved in important biological processes involved in the immune system process, cell adhesion and migration, cellular processes, and multicellular organismal systems. STRING analysis found that IRF7, MX1, TIMP3, and AKT1 may be associated with MDV and REV synergistic replication in chicken embryo fibroblasts (CEFs). Western blotting analysis showed that the selected DAPs were identical to the quantitative proteomics data. Taken together, we verified that MDV and REV can synergistically replicate in coinfected cells and revealed the host molecules involved in it. However, the synergistic pathogenesis of MDV and REV needs to be further studied.

13.
Vet Microbiol ; 267: 109389, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35259599

RESUMEN

Avian leukosis virus subgroup J (ALV-J), an oncogenic retrovirus, induces myelocytoma and various other tumors in broilers and layers. Many recent studies have shown that ALV-J can hijack host molecules to facilitate infection. However, the molecular mechanisms of this process are not clear. Here, we aimed to elucidate the molecular mechanisms contributing to ALV-J infection. ALV-J hijacked MIF via p10 and p27 to facilitate ALV-J infection. ALV-J persistently activated MIF expression in DF-1 cells, and MIF significantly facilitated ALV-J internalization and replication, which demonstrated by MIF overexpression and knockdown experiments and treatment with the MIF antagonist ISO-1. Furthermore, we found that the two subunit proteins of Gag, p10 and p27, interacted with MIF in the cytoplasm, respectively. These results suggested that the p10 and p27 subunit in Gag protein recruited MIF to promote ALV-J infection, providing insights into the roles of the p10/p27 and the host factor MIF in ALV-J infection. The finding may facilitate the development of new strategies for controlling ALV-J or retrovirus infections.


Asunto(s)
Virus de la Leucosis Aviar , Leucosis Aviar , Factores Inhibidores de la Migración de Macrófagos , Enfermedades de las Aves de Corral , Animales , Virus de la Leucosis Aviar/genética , Carcinogénesis , Pollos , Factores Inhibidores de la Migración de Macrófagos/genética
14.
Vet Microbiol ; 266: 109364, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35144044

RESUMEN

Marek's disease virus (MDV) is a highly oncogenic alphaherpesvirus that induces malignant T-cell lymphomas in chickens, leading to great economic loss in poultry industry. The unique-short kinase 3 (Us3), a serine/threonine kinase encoded by three MDV types (MDV-1, MDV-2 and HVT), is important for MDV replication. However, the mechanism of Us3 facilitating MDV replication has not been completely elucidated. In the present study, we report that Us3 significantly facilities MDV replication via inhibition of ß interferon (IFN-ß) production at the late phase of MDV infection. Overexpression or interference of Us3 significantly promoted or inhibited the replication of MDV, and accordingly inhibited or promoted the expression of IFN-ß. Further, Us3 was shown to suppresses interferon stimulatory DNA (ISD)-triggered IFN-ß production by targeting IFN regulatory factor 7 (IRF7) rather than NF-κB signaling. Moreover, Us3 but not kinase-dead (KD) Us3 mutant K220A blocked the nuclear translocation of IRF7 by inhibiting dimerization. Importantly, Us3 phosphorylated and interacted with IRF7. Furthermore, Us3-deficient MDV weakened viral replication and increased IFN-ß production in infected cells or chickens. These results indicated that Us3 interrupts the cytosolic DNA sensing pathway, thereby leading to inhibition of IFN-ß production by targeting IRF7, promoting MDV replication. Our finding expands the knowledge about the role of Us3 in MDV replication.


Asunto(s)
Herpesvirus Gallináceo 2 , Enfermedad de Marek , Animales , Pollos , Factor VII/metabolismo , Herpesvirus Gallináceo 2/genética , Proteínas Serina-Treonina Quinasas/genética , Serina/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral/genética
15.
J Virol ; 96(6): e0165721, 2022 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-35080427

RESUMEN

Avian leukosis virus subgroup J (ALV-J) induces myelocytomas, which can metastasize to multiple organs in diseased chickens. Although metastasis is the primary cause of death in such cases, the mechanism for it remains unclear. Here, we found that interaction between ALV-J surface protein (SU) and doublecortin-like kinase 1 (DCLK1) promotes epithelial-mesenchymal transition (EMT) and cell proliferation. We found that ALV-J can activate EMT in infected cells. Subsequently, proteomics analysis revealed that DCLK1, a well-established putative tumor stem cell marker, which is highly expressed in ALV-J-infected DF-1 cells and chickens, might be a potential factor mediating EMT. Furthermore, using immunofluorescence and immunoprecipitation, we verified that SU interacts with DCLK1. Functional studies suggested that overexpression of DCLK1 increased viral replication and promoted cell proliferation by accelerating the progression of cells from the G0/G1 phase to the S phase of cell cycle, whereas RNA interference of DCLK1 reduced viral replication and arrested cell proliferation by retarding cell cycle progression from the late G1 phase into the S phase in ALV-J-infected cells. Moreover, we demonstrate that the increased accumulation of DCLK1 promotes EMT by increasing the expression of N-cadherin, vimentin, MMP2, and transcription factor Snail1 and decreasing the expression of epithelial marker E-cadherin. These results suggest that ALV-J SU interacts with DCLK1, and accelerates cell proliferation, leading to increased viral replication and ultimately activating EMT, which paves the way for tumor metastasis. IMPORTANCE Tumor metastasis is a major challenge in cancer research, because of its systemic nature and the resistance of disseminated tumor cells to existing therapeutic agents. It is estimated that >90% of mortality from cancer is attributable to metastases. We found that ALV-J can activate EMT, which plays a critical role in cancer metastasis. Subsequently, we identified a tumor stem cell marker, DCLK1, in ALV-J infected cells, which interacts with surface protein (SU) of ALV-J to promote virus replication, activate EMT, and accelerate cell proliferation enabling ALV-J to obtain metastatic ability. Understanding the process of participation of ALV-J in EMT and the route of metastasis will help elucidate the mechanism of virus-induced tumor metastasis and help identify promising molecular targets and key obstacles for ALV-J control and clinical technology development.


Asunto(s)
Virus de la Leucosis Aviar , Leucosis Aviar , Quinasas Similares a Doblecortina , Transición Epitelial-Mesenquimal , Proteínas de la Membrana , Animales , Leucosis Aviar/fisiopatología , Virus de la Leucosis Aviar/genética , Proliferación Celular , Pollos , Quinasas Similares a Doblecortina/metabolismo , Proteínas de la Membrana/metabolismo
16.
J Virol ; 96(2): e0134421, 2022 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-34705559

RESUMEN

The CCCH-type zinc finger antiviral protein (ZAP) can recognize and induce the degradation of mRNAs and proteins of certain viruses, as well as exerting its antiviral activity by activating T cells. However, the mechanism of ZAP that mediates T cell activation during virus infection remains unclear. Here, we found a potential function of ZAP that relieves immunosuppression of T cell induced by avian leukosis virus subgroup J (ALV-J) via a novel signaling pathway that involves norbin-like protein (NLP), protein kinase C delta (PKC-δ), and nuclear factor of activated T cell (NFAT). Specifically, ZAP expression activated T cells by promoting the dephosphorylation and nuclear translocation of NFAT. Furthermore, knockdown of ZAP weakened the reactivity and antiviral response of T cells. Mechanistically, ZAP reduced PKC-δ activity by upregulating and reactivating NLP by competitively binding with viral protein. Knockdown of NLP decreased the dephosphorylation of PKC-δ by ZAP expression. Moreover, we show that knockdown of PKC-δ reduced the phosphorylation levels of NFAT and enhanced its nuclear translocation. Taken together, these data revealed that ZAP relieves immunosuppression caused by ALV-J and mediates T cell activation through the NLP-PKC-δ-NFAT pathway. IMPORTANCE The evolution of the host defense system is driven synchronously in the process of resisting virus invasion. Accordingly, host innate defense factors effectively work to suppress virus replication. However, it remains unclear whether the host innate defense factors are involved in antiviral immune responses against the invasion of immunosuppressive viruses. Here, we found that CCCH-type zinc finger antiviral protein (ZAP) effectively worked in resistance to immunosuppression caused by avian leukosis virus subgroup J (ALV-J), a classic immunosuppressive virus. Evidence showed that ZAP released the phosphatase activity of NLP inhibited by ALV-J and further activated NFAT by inactivating PKC-δ. This novel molecular mechanism, i.e., ZAP regulation of the antiviral immune response by mediating the NLP-PKC-δ-NFAT pathway, has greatly enriched the understanding of the functions of host innate defense factors and provided important scientific ideas and a theoretical basis for research on immunosuppressive viruses and antiviral immunity.


Asunto(s)
Virus de la Leucosis Aviar/inmunología , Factores de Transcripción NFATC/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteína Quinasa C-delta/metabolismo , Proteínas de Unión al ARN/metabolismo , Linfocitos T/inmunología , Animales , Pollos , Interacciones Huésped-Patógeno , Tolerancia Inmunológica , Activación de Linfocitos , Fosforilación , Unión Proteica , Proteínas de Unión al ARN/genética , Transducción de Señal , Linfocitos T/metabolismo , Linfocitos T/virología , Proteínas Virales/metabolismo
17.
Vet Res ; 52(1): 119, 2021 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-34526112

RESUMEN

Congenital avian leukosis virus subgroup J (ALV-J) infection can induce persistent immunotolerance in chicken, however, the underlying mechanism remains unclear. Here, we demonstrate that congenital ALV-J infection induces the production of high-frequency and activated CD4+CD25+ Tregs that maintain persistent immunotolerance. A model of congenital infection by ALV-J was established in fertilized eggs, and hatched chicks showed persistent immunotolerance characterized by persistent viremia, immune organ dysplasia, severe imbalance of the ratio of CD4+/CD8+ T cells in blood and immune organs, and significant decrease in CD3+ T cells and Bu-1+ B cells in the spleen. Concurrently, the mRNA levels of IL-2, IL-10, and IFN-γ showed significant fluctuations in immune organs. Moreover, the frequency of CD4+CD25+ Tregs in blood and immune organs significantly increased, and the frequency of CD4+CD25+ Tregs was positively correlated with changes in ALV-J load in immune organs. Interestingly, CD4+CD25+ Tregs increased in the marginal zone of splenic nodules in ALV-J-infected chickens and dispersed to the germinal center. In addition, the proliferation and activation of B cells in splenic nodules was inhibited, and the number of IgM+ and IgG+ cells in the marginal zone significantly decreased. We further found that the mRNA levels of TGF- ß and CTLA-4 in CD4+CD25+ Tregs of ALV-J-infected chickens significantly increased. Together, high-frequency and activated CD4+CD25+ Tregs inhibited B cells functions by expressing the inhibitory cytokine TGF-ß and inhibitory surface receptor CTLA-4, thereby maintaining persistent immunotolerance in congenital ALV-J-infected chickens.


Asunto(s)
Virus de la Leucosis Aviar/inmunología , Leucosis Aviar/inmunología , Pollos , Tolerancia Inmunológica , Enfermedades de las Aves de Corral/inmunología , Linfocitos T/inmunología , Animales , Linfocitos T CD4-Positivos , Embrión de Pollo , Organismos Libres de Patógenos Específicos
18.
Vet Res ; 52(1): 120, 2021 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-34526128

RESUMEN

Gyrovirus 3 (GyV3), the third novel emerging species of the genus Gyrovirus of the Anelloviridae family, has been described in multiple hosts. Epidemiologically, there are suggestions that GyV3 is associated with diarrhea/proventriculitis, however, no direct causal evidence exists between GyV3 infection and specific clinical diseases. Herein, we infected special pathogen-free (SPF) chickens with GyV3, and then assessed the pathogenicity and tissue tropism. The results revealed that GyV3 induced persistent infection characterized by diarrhea, aplastic anemia, immunosuppression, and persistent systemic lymphocytic inflammation. Clinically, the infected chickens presented ruffled feathers, diarrhea, anemia, and weight loss. Aplastic anemia was characterized by progressive depletion of hematopoietic cells in the bone marrow, immunosuppression was associated with atrophy of the thymus, spleen, and bursa of Fabricious, progressive lymphocytic inflammations were characterized by proventriculitis, adrenalitis, pancreatitis, hepatitis, nephritis, and bronchitis. Viral loads of GyV3 in tissues exhibited "M", "N", "W" or "V" type dynamic changes. The highest level of viral loads was reported in bone marrow at 7dpi, followed by the adrenal gland at 2 dpi, the sciatic nerve at 7 dpi, and bile at 35 dpi. The bone marrow and kidney demonstrate the strongest immunostaining of GyV3-VP1 antigen and were suggested as the target tissues of GyV3. Collectively, GyV3 is an immunosuppressive pathogenic virus that targets the bone marrow and kidney in chickens. Exploring the pathogenicity and tissue tropism of GyV3 will guide the basic understanding of the biology of GyV3 and its pathogenesis in chickens.


Asunto(s)
Pollos , Infecciones por Circoviridae/veterinaria , Gyrovirus/fisiología , Gyrovirus/patogenicidad , Enfermedades de las Aves de Corral/virología , Tropismo Viral , Anemia Aplásica/inmunología , Anemia Aplásica/veterinaria , Anemia Aplásica/virología , Animales , Infecciones por Circoviridae/virología , Diarrea/inmunología , Diarrea/veterinaria , Diarrea/virología , Tolerancia Inmunológica , Inflamación/inmunología , Inflamación/veterinaria , Inflamación/virología , Cinética , Linfocitos/inmunología , Virulencia
19.
Vet Microbiol ; 261: 109191, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34385005

RESUMEN

Gyrovirus 3 (GyV3) has been identified in humans and other hosts, suggesting its cross-species pathogenicity, which poses an increased public health risk. In the current study, we established chicken and mouse models of GyV3 infection. We found that GyV3 induced persistent infections, characterized by viremia, aplastic anemia, immunosuppression, and systematic lymphocytic inflammation, in both species. Kinetic viral loads and antigen expression demonstrated rapid viral replication and broad tissue tropism of GyV3 in both models. The highest viral loads and the strongest antigen immunostaining were present in bone marrow and cerebrum in both chickens and mice, indicating that these are target tissues for GyV3. Genetic diversity analysis of VP1 in infected chickens and mice showed that GyV3 adapts to new hosts via rapid evolution of the hypervariable region of the gene encoding the structural protein VP1. Overall, our results indicate that GyV3 is a cross-species pathogenic virus; therefore, more attention needs to be paid to high levels of GyV3-induced neurotropism and aplastic anemia as a public health risk.


Asunto(s)
Infecciones por Circoviridae/virología , Gyrovirus/patogenicidad , Especificidad del Huésped , Anemia Aplásica/etiología , Anemia Aplásica/virología , Animales , Antígenos Virales/inmunología , Proteínas de la Cápside/genética , Pollos , Infecciones por Circoviridae/complicaciones , Modelos Animales de Enfermedad , Variación Genética , Gyrovirus/genética , Ratones , Carga Viral
20.
Front Vet Sci ; 7: 569098, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33134354

RESUMEN

In October 2018, an outbreak of transmissible viral proventriculitis (TVP) occurred in 30-day-old commercial broiler chickens on a farm in Weifang, China. TVP, an infectious viral disease characterized by runting and stunting, is associated with many viruses, and has a significant economic impact on the global poultry industry. TVP is diagnosed according to clinical symptoms, gross and histological lesions, and negative PCR results for pathogenic bacteria, avian leukosis virus subgroup J, Marek's disease virus, reticuloendotheliosis virus, infectious bursa disease virus, avian reovirus, chicken anemia virus, infectious bronchitis virus, chicken proventricular necrosis virus, gyrovirus 3 and chicken circovirus. To further detect the possible causative pathogens of TVP, we used PacBio third-generation sequencing to examine proventricular samples. A dominant abundance of the novel cyclovirus (CyCV), chCyCV-SDAU-1, was identified in broilers with TVP. The complete chCyCV-SDAU-1 genome was verified via inverse PCR, was 1936 bp long, and consisted of Rep, Cp, and two intergenic regions. Phylogenetic tree analysis showed that chCyCV-SDAU-1 formed an independent branch with other cycloviruses. The homology of chCyCV-SDAU-1 with 20 others known cycloviruses was < 40%. Retrospective investigation showed that the CyCV infection rate in the broilers with TVP was 80% (16/20), while no CyCV was found in healthy chickens. In conclusion, a novel CyCV was identified in chickens with TVP, though its role in this disease is unclear.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...