Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
1.
J Biol Chem ; 300(6): 107395, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38768812

RESUMEN

B2 haplotype major histocompatibility complex (MHC) has been extensively reported to confer resistance to various avian diseases. But its peptide-binding motif is unknown, and the presenting peptide is rarely identified. Here, we identified its peptide-binding motif (X-A/V/I/L/P/S/G-X-X-X-X-X-X-V/I/L) in vitro using Random Peptide Library-based MHC I LC-MS/MS analysis. To further clarify the structure basis of motif, we determined the crystal structure of the BF2∗02:01-PB2552-560 complex at 1.9 Å resolution. We found that BF2∗02:01 had a relatively wide antigen-binding groove, and the structural characterization of pockets was consistent with the characterization of peptide-binding motif. The wider features of the peptide-binding motif and increased number of peptides bound by BF2∗02:01 than BF2∗04:01 might resolve the puzzles for the presence of potential H9N2 resistance in B2 chickens. Afterward, we explored the H9N2 avian influenza virus (AIV)-induced cellular immune response in B2 haplotype chickens in vivo. We found that ratio of CD8+ T cell and kinetic expression of cytotoxicity genes including Granzyme K, interferon-γ, NK lysin, and poly-(ADP-ribose) polymerase in peripheral blood mononuclear cells were significantly increased in defending against H9N2 AIV infection. Especially, we selected 425 epitopes as candidate epitopes based on the peptide-binding motif and further identified four CD8+ T-cell epitopes on H9N2 AIV including NS198-106, PB2552-560, NP182-190, and NP455-463 via ELI-spot interferon-γ detections after stimulating memory lymphocytes with peptides. More importantly, these epitopes were found to be conserved in H7N9 AIV and H9N2 AIV. These findings provide direction for developing effective T cell epitope vaccines using well-conserved internal viral antigens in chickens.


Asunto(s)
Pollos , Epítopos de Linfocito T , Subtipo H9N2 del Virus de la Influenza A , Gripe Aviar , Subtipo H9N2 del Virus de la Influenza A/inmunología , Animales , Epítopos de Linfocito T/inmunología , Gripe Aviar/inmunología , Gripe Aviar/virología , Linfocitos T CD8-positivos/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo
2.
PLoS Pathog ; 19(10): e1011685, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37819993

RESUMEN

Chicken lung is an important target organ of avian influenza virus (AIV) infection, and different pathogenic virus strains lead to opposite prognosis. Using a single-cell RNA sequencing (scRNA-seq) assay, we systematically and sequentially analyzed the transcriptome of 16 cell types (19 clusters) in the lung tissue of chickens infected with H5N1 highly pathogenic avian influenza virus (HPAIV) and H9N2 low pathogenic avian influenza virus (LPAIV), respectively. Notably, we developed a valuable catalog of marker genes for these cell types. Compared to H9N2 AIV infection, H5N1 AIV infection induced extensive virus replication and the immune reaction across most cell types simultaneously. More importantly, we propose that infiltrating inflammatory macrophages (clusters 0, 1, and 14) with massive viral replication, pro-inflammatory cytokines (IFN-ß, IL1ß, IL6 and IL8), and emerging interaction of various cell populations through CCL4, CCL19 and CXCL13, potentially contributed to the H5N1 AIV driven inflammatory lung injury. Our data revealed complex but distinct immune response landscapes in the lung tissue of chickens after H5N1 and H9N2 AIV infection, and deciphered the potential mechanisms underlying AIV-driven inflammatory reactions in chicken. Furthermore, this article provides a rich database for the molecular basis of different cell-type responses to AIV infection.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A , Subtipo H9N2 del Virus de la Influenza A , Gripe Aviar , Lesión Pulmonar , Animales , Pollos/metabolismo , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H9N2 del Virus de la Influenza A/genética , Análisis de la Célula Individual
3.
J Virol ; 97(11): e0115223, 2023 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-37902396

RESUMEN

IMPORTANCE: 3'UTRs can affect gene transcription and post-transcriptional regulation in multiple ways, further influencing the function of proteins in a unique manner. Recently, ALV-J has been mutating and evolving rapidly, especially the 3'UTR of viral genome. Meanwhile, clinical symptoms caused by ALV-J have changed significantly. In this study, we found that the ALV-J strains containing △-r-TM-type 3'UTR are the most abundant. By constructing ALV-J infectious clones and subgenomic vectors containing different 3'UTRs, we prove that 3'UTRs directly affect viral tissue preference and can promote virus replication as an enhancer. ALV-J strain containing 3'UTR of △-r-TM proliferated fastest in primary cells. All five forms of 3'UTRs can assist intron-containing viral mRNA nuclear export, with similar efficiency. ALV-J mRNA half-life is not influenced by different 3'UTRs. Our results dissect the roles of 3'UTR on regulating viral replication and pathogenicity, providing novel insights into potential anti-viral strategies.


Asunto(s)
Regiones no Traducidas 3' , Transporte Activo de Núcleo Celular , Virus de la Leucosis Aviar , Replicación Viral , Expresión Génica , Regulación de la Expresión Génica , Virus de la Leucosis Aviar/genética , Virus de la Leucosis Aviar/fisiología
4.
J Immunol ; 209(5): 979-990, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35940633

RESUMEN

Domestic ducks are the important host for H5N1 highly pathogenic avian influenza virus (HPAIV) infection and epidemiology, but little is known about the duck T cell response to H5N1 AIV infection. In infection experiments of mallard ducks, we detected significantly increased CD8+ cells and augmented expression of cytotoxicity-associated genes, including granzyme A and IFN-γ, in PBMCs from 5 to 9 d postinfection when the virus shedding was clearly decreased, which suggested the importance of the duck cytotoxic T cell response in eliminating H5N1 infection in vivo. Intriguingly, we found that a CD8high+ population of PBMCs was clearly upregulated in infected ducks from 7 to 9 d postinfection compared with uninfected ducks. Next, we used Smart-Seq2 technology to investigate the heterogeneity and transcriptional differences of the duck CD8+ cells. Thus, CD8high+ cells were likely to be more responsive to H5N1 AIV infection, based on the high level of expression of genes involved in T cell responses, activation, and proliferation, including MALT1, ITK, LCK, CD3E, CD247, CFLAR, IL-18R1, and IL-18RAP. More importantly, we have also successfully cultured H5N1 AIV-specific duck T cells in vitro, to our knowledge, for the first time, and demonstrated that the CD8high+ population was increased with the duck T cell activation and response in vitro, which was consistent with results in vivo. Thus, the duck CD8high+ cells represent a potentially effective immune response to H5N1 AIV infection in vivo and in vitro. These findings provide novel insights and direction for developing effective H5N1 AIV vaccines.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A , Gripe Aviar , Animales , Linfocitos T CD8-positivos/patología , Patos , Granzimas
5.
J Virol ; 96(6): e0202721, 2022 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-35107377

RESUMEN

Marek's disease virus (MDV) is a member of the genus Mardivirus in the subfamily Alphaherpesvirinae. There are three different serotypes of MDV designated as MDV-1 (Gallid herpesvirus type 2), MDV-2 (Gallid herpesvirus type 3), and MDV-3 (Meleagrid herpesvirus 1, herpesvirus of turkeys, HVT). MDV-1 is the only serotype that induces Marek's disease (MD), a lymphoproliferative disorder resulting in aggressive T-cell lymphomas and paralytic symptoms. In the lymphomas and lymphoblastoid cell lines (LCL) derived from them, MDV establishes latent infection with limited viral gene expression. The latent viral genome in LCL can be activated by co-cultivation with chicken embryo fibroblast (CEF) monolayers. MSB-1, one of the first MDV-transformed LCL established from the splenic lymphoma, is distinct in harboring both the oncogenic MDV-1 and non-oncogenic MDV-2 viruses. Following the successful application of CRISPR/Cas9 editing approach for precise knockdown of the MDV-1 genes in LCL, we describe here the targeted deletion of MDV-2 glycoprotein B (gB) in MSB-1 cells. Due to the essential nature of gB for infectivity, the production of MDV-2 plaques on CEF was completely abolished in the MDV-2-gB-deleted MSB-1 cells. Our study has demonstrated that the CRISPR/Cas9 system can be used for targeted inactivation of the co-infecting MDV-2 without affecting the MDV-1 in the MSB-1 cell line. Successful inactivation of MDV-2 demonstrated here also points toward the possibility of using targeted gene editing as an antiviral strategy against pathogenic MDV-1 and other viruses infecting chickens. IMPORTANCE Marek's disease (MD) is a lymphoproliferative disease of chickens characterized by rapid-onset lymphomas in multiple organs and by infiltration into peripheral nerves, causing paralysis. Lymphoblastoid cell lines (LCL) derived from MD lymphomas have served as valuable resources to improve understanding of distinct aspects of virus-host interactions in transformed cells including transformation, latency, and reactivation. MDV-transformed LCL MSB-1, derived from spleen lymphoma induced by the BC-1 strain of MDV, has a unique feature of harboring an additional non-pathogenic MDV-2 strain HPRS-24. By targeted deletion of essential gene glycoprotein B from the MDV-2 genome within the MSB-1 cells, we demonstrated the total inhibition of MDV-2 virus replication on co-cultivated CEF, with no effect on MDV-1 replication. The identified viral genes critical for reactivation/inhibition of viruses will be useful as targets for development of de novo disease resistance in chickens to avian pathogens.


Asunto(s)
Herpesvirus Gallináceo 3 , Linfoma , Enfermedad de Marek , Proteínas del Envoltorio Viral , Animales , Sistemas CRISPR-Cas , Línea Celular , Embrión de Pollo , Pollos , Herpesvirus Gallináceo 3/genética , Linfoma/veterinaria , Linfoma/virología , Proteínas del Envoltorio Viral/genética
6.
J Virol ; 96(4): e0154921, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-34878920

RESUMEN

Glycans on envelope glycoprotein (Env) of the subgroup J avian leukosis virus (ALV-J) play an essential role in the virion integrity and infection process. In this study, we found that, among the 13 predicted N-linked glycosylation sites (NGSs) in gp85 of Tibetan chicken strain TBC-J6, N17, and N193/N191 are pivotal for virus replication. Further research illustrated that a mutation at N193 weakened Env-receptor binding in a blocking assay of the viral entrance, coimmunoprecipitation, and ELISA. Our studies also showed that N17 was involved in Env protein processing and later virion incorporation based on the detection of p27 and Env protein in the supernatant and gp37 in the cell culture. This report is systematic research on clarifying the biological function of NGSs on ALV-J gp85, which would provide valuable insight into the role of gp85 in the ALV life cycle and anti-ALV-J strategies. IMPORTANCE ALV-J is a retrovirus that can cause multiple types of tumors in chickens. Among all the viral proteins, the heavily glycosylated envelope protein is especially crucial. Glycosylation plays a major role in Env protein function, including protein processing, receptor attachment, and immune evasion. Notably, viruses isolated recently seem to lose their 6th and 11th NGS, which proved to be important in receptor binding. In our study, the 1st (N17) and 8th (N193) NGS of gp85 of the strain TBC-J6 can largely influence the titer of this virus. Deglycosylation at N193 weakened Env-receptor binding while mutation at N17 influenced Env protein processing. This study systemically analyzed the function of NGSs in ALV-J in different aspects, which may help us to understand the life cycle of ALV-J and provide antiviral targets for the control of ALV-J.


Asunto(s)
Virus de la Leucosis Aviar/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Animales , Virus de la Leucosis Aviar/crecimiento & desarrollo , Línea Celular , Pollos , Glicosilación , Mutación , Unión Proteica , Procesamiento Proteico-Postraduccional , Receptores Virales/metabolismo , Proteínas del Envoltorio Viral/genética , Carga Viral/genética , Virión/metabolismo
7.
J Med Virol ; 95(1): e28324, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36401345

RESUMEN

Dynamic alteration of the epitranscriptome exerts regulatory effects on the lifecycle of oncogenic viruses in vitro. However, little is known about these effects in vivo because of the general lack of suitable animal infection models of these viruses. Using a model of rapid-onset Marek's disease lymphoma in chickens, we investigated changes in viral and host messenger RNA (mRNA) N6-methyladenosine (m6 A) modification during Marek's disease virus (MDV) infection in vivo. We found that the expression of major epitranscriptomic proteins varies among viral infection phases, reprogramming both the viral and the host epitranscriptomes. Specifically, the methyltransferase-like 3 (METTL3)/14 complex was suppressed during the lytic and reactivation phases of the MDV lifecycle, whereas its expression was increased during the latent phase and in MDV-induced tumors. METTL3/14 overexpression inhibits, whereas METTL3/14 knockdown enhances, MDV gene expression and replication. These findings reveal the dynamic features of the mRNA m6 A modification program during viral replication in vivo, especially in relation to key pathways involved in tumorigenesis.


Asunto(s)
Enfermedad de Marek , Animales , Enfermedad de Marek/genética , Virus Oncogénicos/genética , Pollos , ARN Mensajero/genética , ARN Mensajero/metabolismo
8.
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
9.
Int J Mol Sci ; 23(19)2022 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-36232572

RESUMEN

Avian leukosis virus (ALV) induces B-cell lymphomas and other malignancies in chickens through insertional activation of oncogenes, and c-myc activation has been commonly identified in ALV-induced tumors. Using ALV-transformed B-lymphoma-derived HP45 cell line, we applied in situ CRISPR-Cas9 editing of integrated proviral long terminal repeat (LTR) to examine the effects on gene expression and cell proliferation. Targeted deletion of LTR resulted in significant reduction in expression of a number of LTR-regulated genes including c-myc. LTR deletion also induced apoptosis of HP45 cells, affecting their proliferation, demonstrating the significance of LTR-mediated regulation of critical genes. Compared to the global effects on expression and functions of multiple genes in LTR-deleted cells, deletion of c-myc had a major effect on the HP45 cells proliferation with the phenotype similar to the LTR deletion, demonstrating the significance of c-myc expression in ALV-induced lymphomagenesis. Overall, our studies have not only shown the potential of targeted editing of the LTR for the global inhibition of retrovirus-induced transformation, but also have provided insights into the roles of LTR-regulated genes in ALV-induced neoplastic transformation.


Asunto(s)
Virus de la Leucosis Aviar , Animales , Virus de la Leucosis Aviar/genética , Línea Celular , Proliferación Celular/genética , Pollos/genética , Provirus/genética , Secuencias Repetidas Terminales/genética
10.
J Virol ; 94(10)2020 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-32161176

RESUMEN

The Bcl-2 (B cell lymphoma 2)-related protein Nr-13 plays a major role in the regulation of cell death in developing avian B cells. With over 65% sequence similarity to the chicken Nr-13, herpesvirus of turkeys (HVT) vNr-13, encoded by the HVT079 and HVT096 genes, is the first known alphaherpesvirus-encoded Bcl-2 homolog. HVT-infected cells were reported to be relatively more resistant to serum starvation, suggested that vNr-13 could be involved in protecting the cells. Here, we describe CRISPR/Cas9-based editing of exon 1 of the HVT079 and HVT096 genes from the HVT genome to generate the mutant HVT-ΔvNr-13 to gain insights into its functional roles. Overall, wild-type HVT and HVT-ΔvNr-13 showed similar growth kinetics; however, at early time points, HVT-ΔvNr-13 showed 1.3- to 1.7-fold-lower growth of cell-associated virus and 3- to 6.2-fold-lower growth of cell-free virus. In transfected cells, HVT vNr-13 showed a mainly diffuse cytoplasmic distribution with faint nuclear staining. Further, vNr-13 localized to the mitochondria and endoplasmic reticulum (ER) and disrupted mitochondrial network morphology in the transfected cells. In the wild-type HVT-infected cells, vNr-13 expression appeared to be directly involved in the disruption of the mitochondrial network, as the mitochondrial network morphology was substantially restored in the HVT-ΔvNr-13-infected cells. IncuCyte S3 real-time apoptosis monitoring demonstrated that vNr-13 is unequivocally involved in the apoptosis inhibition, and it is associated with an increase of PFU, especially under serum-free conditions in the later stages of the viral replication cycle. Furthermore, HVT blocks apoptosis in infected cells but activates apoptosis in noninfected bystander cells.IMPORTANCE B cell lymphoma 2 (Bcl-2) family proteins play important roles in regulating apoptosis during homeostasis, tissue development, and infectious diseases. Several viruses encode homologs of cellular Bcl-2-proteins (vBcl-2) to inhibit apoptosis, which enable them to replicate and persist in the infected cells and to evade/modulate the immune response of the host. Herpesvirus of turkeys (HVT) is a nonpathogenic alphaherpesvirus of turkeys and chickens that is widely used as a live vaccine against Marek's disease and as recombinant vaccine viral vectors for protecting against multiple avian diseases. Identical copies of the HVT genes HVT079 and HVT096 encode the Bcl-2 homolog vNr-13. While previous studies have identified the potential ability of vNr-13 in inhibiting apoptosis induced by serum deprivation, there have been no detailed investigations on the functions of vNr-13. Using CRISPR/Cas9-based ablation of the vNr-13 gene, we demonstrated the roles of HVT vNr-13 in early stages of the viral replication cycle, mitochondrial morphology disruption, and apoptosis inhibition in later stages of viral replication.


Asunto(s)
Apoptosis/fisiología , Proteínas Aviares/metabolismo , Herpesviridae/genética , Proteínas de la Membrana/metabolismo , Mitocondrias/metabolismo , Pavos/virología , Proteínas Virales/metabolismo , Replicación Viral/fisiología , Animales , Proteínas Aviares/genética , Sistemas CRISPR-Cas , Pollos/metabolismo , Retículo Endoplásmico/metabolismo , Infecciones por Herpesviridae/virología , Linfoma de Células B/inmunología , Proteínas de la Membrana/genética , Enfermedades de las Aves de Corral/virología , Alineación de Secuencia , Análisis de Secuencia , Proteínas Virales/genética , Vacunas Virales/inmunología
11.
Vet Res ; 52(1): 20, 2021 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-33579382

RESUMEN

Processing and packaging of herpesvirus genomic DNA is regulated by a packaging-associated terminase complex comprising of viral proteins pUL15, pUL28 and pUL33. Marek's disease virus (MDV) homologs UL28 and UL33 showed conserved functional features with high sequence identity with the corresponding Herpes simplex virus 1 (HSV-1) homologs. As part of the investigations into the role of the UL28 and UL33 homologs of oncogenic MDV for DNA packaging and replication in cultured cells, we generated MDV mutant clones deficient in UL28 or UL33 of full-length MDV genomes. Transfection of UL28- or UL33-deleted BAC DNA into chicken embryo fibroblast (CEF) did not result either in the production of visible virus plaques, or detectable single cell infection after passaging onto fresh CEF cells. However, typical MDV plaques were detectable in CEF transfected with the DNA of revertant mutants where the deleted genes were precisely reinserted. Moreover, the replication defect of the UL28-deficient mutant was completely restored when fragment encoding the full UL28 gene was co-transfected into CEF cells. Viruses recovered from the revertant construct, as well as by the UL28 co-transfection, showed replication ability comparable with parental virus. Furthermore, the transmission electron microscopy study indicated that immature capsids were assembled without the UL28 expression, but with the loss of infectivity. Importantly, predicted three-dimensional structures of UL28 between MDV and HSV-1 suggests conserved function in virus replication. For the first time, these results revealed that both UL28 and UL33 are essential for MDV replication through regulating DNA cleavage and packaging.


Asunto(s)
ADN Viral/química , Endodesoxirribonucleasas/genética , Mardivirus/fisiología , Receptores de Quimiocina/genética , Proteínas Virales/genética , Replicación Viral , Secuencia de Aminoácidos , Animales , Embrión de Pollo , Endodesoxirribonucleasas/química , Endodesoxirribonucleasas/metabolismo , Mardivirus/enzimología , Mardivirus/genética , División del ARN , Receptores de Quimiocina/química , Receptores de Quimiocina/metabolismo , Alineación de Secuencia , Organismos Libres de Patógenos Específicos , Proteínas Virales/química , Proteínas Virales/metabolismo
12.
Int J Mol Sci ; 22(22)2021 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-34830257

RESUMEN

To investigate the antiviral effect of lithium chloride (LiCl) on the replication of Marek's disease virus (MDV) in chicken embryonic fibroblast (CEF) cells, real-time PCR, Western blotting, plaque counting, and indirect immunofluorescence experiments were performed at different time points of LiCl treated CEF cells with virus infection. The results demonstrated that LiCl could affect multiple steps of virus replication and inhibit viral gene expression and protein synthesis in a dose- and time-dependent manner. Moreover, LiCl could directly affect viral infectivity as well. In addition, LiCl significantly affected the gene expression of IFN-ß related genes in virus-infected cells. These results indicate that LiCl significantly inhibits MDV replication and proliferation in CEF cells and it has the potential to be used as an antiviral agent against MDV.


Asunto(s)
Antivirales/farmacología , Herpesvirus Gallináceo 2/efectos de los fármacos , Cloruro de Litio/farmacología , Proteínas Oncogénicas Virales/genética , Replicación Viral/efectos de los fármacos , 2',5'-Oligoadenilato Sintetasa/genética , 2',5'-Oligoadenilato Sintetasa/metabolismo , Animales , Línea Celular , Embrión de Pollo , Pollos , Relación Dosis-Respuesta a Droga , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/virología , Regulación de la Expresión Génica , Herpesvirus Gallináceo 2/genética , Herpesvirus Gallináceo 2/metabolismo , Interacciones Huésped-Patógeno/efectos de los fármacos , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/metabolismo , Interferón beta/genética , Interferón beta/metabolismo , Proteínas Oncogénicas Virales/antagonistas & inhibidores , Proteínas Oncogénicas Virales/metabolismo , Carga Viral/efectos de los fármacos
13.
J Virol ; 93(17)2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31189706

RESUMEN

MicroRNAs (miRNAs) are small noncoding RNAs with profound regulatory roles in many areas of biology, including cancer. MicroRNA 155 (miR-155), one of the extensively studied multifunctional miRNAs, is important in several human malignancies such as diffuse large B cell lymphoma and chronic lymphocytic leukemia. Moreover, miR-155 orthologs KSHV-miR-K12-11 and MDV-miR-M4, encoded by Kaposi's sarcoma-associated herpesvirus (KSHV) and Marek's disease virus (MDV), respectively, are also involved in oncogenesis. In MDV-induced T-cell lymphomas and in lymphoblastoid cell lines derived from them, MDV-miR-M4 is highly expressed. Using excellent disease models of infection in natural avian hosts, we showed previously that MDV-miR-M4 is critical for the induction of T-cell lymphomas as mutant viruses with precise deletions were significantly compromised in their oncogenicity. However, those studies did not elucidate whether continued expression of MDV-miR-M4 is essential for maintaining the transformed phenotype of tumor cells. Here using an in situ CRISPR/Cas9 editing approach, we deleted MDV-miR-M4 from the MDV-induced lymphoma-derived lymphoblastoid cell line MDCC-HP8. Precise deletion of MDV-miR-M4 was confirmed by PCR, sequencing, quantitative reverse transcription-PCR (qRT-PCR), and functional analysis. Continued proliferation of the MDV-miR-M4-deleted cell lines demonstrated that MDV-miR-M4 expression is not essential for maintaining the transformed phenotype, despite its initial critical role in the induction of lymphomas. Ability to examine the direct role of oncogenic miRNAs in situ in tumor cell lines is valuable in delineating distinct determinants and pathways associated with the induction or maintenance of transformation in cancer cells and will also contribute significantly to gaining further insights into the biology of oncogenic herpesviruses.IMPORTANCE Marek's disease virus (MDV) is an alphaherpesvirus associated with Marek's disease (MD), a highly contagious neoplastic disease of chickens. MD serves as an excellent model for studying virus-induced T-cell lymphomas in the natural chicken hosts. Among the limited set of genes associated with MD oncogenicity, MDV-miR-M4, a highly expressed viral ortholog of the oncogenic miR-155, has received extensive attention due to its direct role in the induction of lymphomas. Using a targeted CRISPR-Cas9-based gene editing approach in MDV-transformed lymphoblastoid cell lines, we show that MDV-miR-M4, despite its critical role in the induction of tumors, is not essential for maintaining the transformed phenotype and continuous proliferation. As far as we know, this was the first study in which precise editing of an oncogenic miRNA was carried out in situ in MD lymphoma-derived cell lines to demonstrate that it is not essential in maintaining the transformed phenotype.


Asunto(s)
Transformación Celular Viral/genética , Linfoma/virología , Mardivirus/patogenicidad , MicroARNs/genética , Animales , Sistemas CRISPR-Cas , Línea Celular Transformada , Línea Celular Tumoral , Proliferación Celular , Humanos , Mardivirus/genética , ARN Viral/genética
14.
BMC Vet Res ; 16(1): 371, 2020 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-33008383

RESUMEN

BACKGROUND: Baicalin, the main metabolic component of Scutellaria baicalensis Georgi, has various pharmacological properties including anti-inflammatory, anti-oxidant, anti-apoptotic, anti-bactericidal and anti-viral. The purpose of this study was to investigate the anti-Marek's disease virus (MDV) activities of baicalin in CEF cells. RESULTS: Here, we showed that baicalin could inhibit viral mRNA, protein levels and overall plaque formation in a time-dependent manner. We also found that baicalin could consistently inhibit MDV replication and directly affect the virus infectivity. Moreover, baicalin treatment has no effect on expression level of antiviral cytokine and inflammatory cytokines in MDV infected CEFs. CONCLUSIONS: These results demonstrate that baicalin could be a potential drug against MDV infection.


Asunto(s)
Flavonoides/farmacología , Mardivirus/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Animales , Antivirales/farmacología , Células Cultivadas , Embrión de Pollo , Citocinas/metabolismo , Fibroblastos , Flavonoides/toxicidad , Enfermedad de Marek/virología , ARN Mensajero
15.
BMC Vet Res ; 15(1): 383, 2019 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-31666067

RESUMEN

BACKGROUND: Avian leukosis (AL), which is caused by avian leukosis virus (ALV), has led to substantial economic losses in the poultry industry. The kit used to detect all ALV-positive chickens in breeder flocks is very important for efficiently controlling AL. However, a new emerging ALV subtype is currently a severe challenge in the poultry industry. RESULTS: In this paper, we compared different enzyme-linked immunosorbent assay (ELISA) kits for detecting p27 of ALV in the same batch of meconium samples. Different positive samples were further analyzed by PCR or virus isolation. The results showed that 36 positive samples among the 1812 chicken meconium samples could be detected by a sandwich ELISA (sELISA) kit, but only 17 positive samples could be identified by a commercial kit. To verify this result, cloacal swabs and viruses isolated from the positive chickens (2 days old) were used to detect the presence of p27. The results showed that the positive rate of p27 was 100% for the swabs and 40% for virus isolation. Surprisingly, PCR and sequence analysis revealed that the env gene of ALV in these positive samples belonged to the novel subgroup K (ALV-K). CONCLUSION: These data not only demonstrate the relatively high sensitivity of the sELISA kit but also highlight the challenge of controlling ALV-K.


Asunto(s)
Virus de la Leucosis Aviar/aislamiento & purificación , Pollos/virología , Cloaca/virología , Antígeno Nuclear de Célula en Proliferación/aislamiento & purificación , Animales , Virus de la Leucosis Aviar/genética , Ensayo de Inmunoadsorción Enzimática/veterinaria , Juego de Reactivos para Diagnóstico , Sensibilidad y Especificidad
16.
Retrovirology ; 15(1): 45, 2018 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-29970099

RESUMEN

BACKGROUND: Co-infection with avian leukosis virus subgroup J and reticuloendotheliosis virus induces synergistic pathogenic effects and increases mortality. However, the role of exosomal miRNAs in the molecular mechanism of the synergistic infection of the two viruses remains unknown. RESULTS: In this study, exosomal RNAs from CEF cells infected with ALV-J, REV or both at the optimal synergistic infection time were analysed by Illumina RNA deep sequencing. A total of 54 (23 upregulated and 31 downregulated) and 16 (7 upregulated and 9 downregulated) miRNAs were identified by comparing co-infection with two viruses, single-infected ALV-J and REV, respectively. Moreover, five key miRNAs, including miR-184-3p, miR-146a-3p, miR-146a-5p, miR-3538 and miR-155, were validated in both exosomes and CEF cells by qRT-PCR. GO annotation and KEGG pathway analysis of the miRNA target genes showed that the five differentially expressed miRNAs participated in virus-vector interaction, oxidative phosphorylation, energy metabolism and cell growth. CONCLUSIONS: We demonstrated that REV and ALV-J synergistically increased the accumulation of exosomal miRNAs, which sheds light on the synergistic molecular mechanism of ALV-J and REV.


Asunto(s)
Virus de la Leucosis Aviar/fisiología , Coinfección , Exosomas/genética , MicroARNs/genética , Interacciones Microbianas , Virus de la Reticuloendoteliosis/fisiología , Infecciones por Retroviridae/genética , Infecciones por Retroviridae/virología , Animales , Leucosis Aviar/genética , Leucosis Aviar/metabolismo , Leucosis Aviar/virología , Línea Celular , Exosomas/metabolismo , Exosomas/ultraestructura , Regulación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Interacciones Huésped-Patógeno , Interferencia de ARN , Reproducibilidad de los Resultados , Infecciones por Retroviridae/metabolismo , Replicación Viral
17.
Arch Virol ; 163(4): 987-995, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29327234

RESUMEN

Lithium chloride (LiCl) has been reported to possess antiviral activity against several viruses. In the current study, we assessed the antiviral activity effect of LiCl on ALV-J infection in CEF cells by using real-time PCR, Western blot analysis, IFA and p27 ELISA analysis. Our results showed that both viral RNA copy number and protein level decreased significantly in a dose and time dependent manner. Time-course analysis revealed that the antiviral effect was more pronounced when CEFs were treated at the post infection stage rather than at early absorption or pre-absorption stages. Further experiments demonstrated that LiCl did not affect virus attachment or entry, but rather affected early virus replication. We also found that inhibition of viral replication after LiCl treatment was associated with reduced mRNA levels of pro-inflammatory cytokines. These results demonstrate that LiCl effectively blocked ALV-J replication in CEF cells and may be used as an antiviral agent against ALV-J.


Asunto(s)
Antivirales/farmacología , Virus de la Leucosis Aviar/efectos de los fármacos , Interacciones Huésped-Patógeno , Cloruro de Litio/farmacología , Proteínas del Envoltorio Viral/antagonistas & inhibidores , Replicación Viral/efectos de los fármacos , Animales , Virus de la Leucosis Aviar/genética , Virus de la Leucosis Aviar/crecimiento & desarrollo , Supervivencia Celular/efectos de los fármacos , Embrión de Pollo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/antagonistas & inhibidores , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Relación Dosis-Respuesta a Droga , Fibroblastos/efectos de los fármacos , Fibroblastos/virología , Regulación de la Expresión Génica , Interleucinas/antagonistas & inhibidores , Interleucinas/genética , Interleucinas/metabolismo , Cultivo Primario de Células , ARN Viral/antagonistas & inhibidores , ARN Viral/genética , ARN Viral/metabolismo , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Acoplamiento Viral/efectos de los fármacos
18.
BMC Vet Res ; 14(1): 295, 2018 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-30257680

RESUMEN

BACKGROUND: The gp85 is the main envelope protein of avian leukosis subgroup J (ALV-J) involved in virus neutralization. Here, we mapped the epitope in ALV-J gp85 by ELISA using synthetic peptides and developed epitope based diagnostic methods for ALV-J infection. RESULTS: The results revealed that monoclonal antibody (mAb) JE9 recognized 83WDPQEL88 motif, which was highly conserved in gp85 among different ALV-J strains by homology analysis. Moreover, after evaluation with two hundred and forty sera samples obtained from different chicken farms, the epitope-based peptide ELISA had much higher sensitivity than commercial ELISA kit for antibody detection of ALV-J. CONCLUSIONS: A novel B-cell epitope recognized by the mAb JE9 was identified. The developed peptide-ELISA based on this novel B-cell epitope could be useful in laboratory viral diagnosis, routine surveillance in chicken farms, and also in understanding the pathogenesis of ALV-J.


Asunto(s)
Virus de la Leucosis Aviar/inmunología , Pollos , Epítopos de Linfocito B/inmunología , Enfermedades de las Aves de Corral/virología , Proteínas del Envoltorio Viral/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/inmunología , Leucosis Aviar/inmunología , Leucosis Aviar/virología , Virus de la Leucosis Aviar/clasificación , Virus de la Leucosis Aviar/genética , Mapeo Epitopo , Epítopos de Linfocito B/química , Enfermedades de las Aves de Corral/diagnóstico , Enfermedades de las Aves de Corral/inmunología , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/genética
19.
J Gen Virol ; 98(4): 810-820, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28113043

RESUMEN

The v-rel oncoprotein encoded by reticuloendotheliosis virus T strain (Rev-T) is a member of the rel/NF-κB family of transcription factors capable of transformation of primary chicken spleen and bone marrow cells. Rapid transformation of avian haematopoietic cells by v-rel occurs through a process of deregulation of multiple protein-encoding genes through its direct effect on their promoters. More recently, upregulation of oncogenic miR-155 and its precursor pre-miR-155 was demonstrated in both Rev-T-infected chicken embryo fibroblast cultures and Rev-T-induced B-cell lymphomas. Through electrophoresis mobility shift assay and reporter analysis on the gga-miR-155 promoter, we showed that the v-rel-induced miR-155 overexpression occurred by the direct binding to one of the putative NF-κB binding sites. Using the v-rel-induced transformation model on chicken embryonic splenocyte cultures, we could demonstrate a dynamic increase in miR-155 levels during the transformation. Transcriptome profiles of lymphoid cells transformed by v-rel showed upregulation of miR-155 accompanied by downregulation of a number of putative miR-155 targets such as Pu.1 and CEBPß. We also showed that v-rel could rescue the suppression of miR-155 expression observed in Marek's disease virus (MDV)-transformed cell lines, where its functional viral homologue MDV-miR-M4 is overexpressed. Demonstration of gene expression changes affecting major molecular pathways, including organismal injury and cancer in avian macrophages transfected with synthetic mature miR-155, underlines its potential direct role in transformation. Our study suggests that v-rel-induced transformation involves a complex set of events mediated by the direct activation of NF-κB targets, together with inhibitory effects on microRNA targets.


Asunto(s)
Transformación Celular Viral , Interacciones Huésped-Patógeno , Proteínas Oncogénicas v-rel/metabolismo , ARN Mensajero/biosíntesis , Virus de la Reticuloendoteliosis/patogenicidad , Animales , Células Cultivadas , Pollos , Perfilación de la Expresión Génica , Leucocitos Mononucleares/virología , Regiones Promotoras Genéticas , Unión Proteica
20.
Vet Res ; 48(1): 48, 2017 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-28903753

RESUMEN

Avian leukosis virus (ALV) is a retrovirus that causes tumors in avian species, and its vertical and horizontal transmission in poultry flocks results in enormous economic losses. Despite the discovery of specific host receptors, there have been few reports on the modulation of viral susceptibility via genetic modification. We therefore engineered acquired resistance to ALV subgroup B using CRISPR/Cas9-mediated genome editing technology in DF-1 chicken fibroblasts. Using this method, we efficiently modified the tumor virus locus B (tvb) gene, encoding the TVB receptor, which is essential for ALV subgroup B entry into host cells. By expanding individual DF-1 clones, we established that artificially generated premature stop codons in the cysteine-rich domain (CRD) of TVB receptor confer resistance to ALV subgroup B. Furthermore, we found that a cysteine residue (C80) of CRD2 plays a crucial role in ALV subgroup B entry. These results suggest that CRISPR/Cas9-mediated genome editing can be used to efficiently modify avian cells and establish novel chicken cell lines with resistance to viral infection.


Asunto(s)
Virus de la Leucosis Aviar/inmunología , Leucosis Aviar/inmunología , Resistencia a la Enfermedad/genética , Mutación/genética , Animales , Leucosis Aviar/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Cisteína , Resistencia a la Enfermedad/inmunología , Fibroblastos/virología , Edición Génica/veterinaria , Mutación/inmunología , Análisis de Secuencia de ADN/veterinaria
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA