Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
1.
Cell Commun Signal ; 20(1): 164, 2022 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-36280829

RESUMO

BACKGROUND: Triple negative breast cancer (TNBC) is one of the most lethal breast cancer subtypes. Due to a lack of effective therapeutic targets, chemotherapy is still the main medical treatment for TNBC patients. Thus, it is important and necessary to find new therapeutic targets for TNBC. Recent genomic studies implicated the Hippo / Yap signal is over activated in TNBC, manifesting it plays a key role in TNBC carcinogenesis and cancer progression. RBCK1 was firstly identified as an important component for linear ubiquitin assembly complex (LUBAC) and facilitates NFKB signaling in immune response. Further studies showed RBCK1 also facilitated luminal type breast cancer growth and endocrine resistance via trans-activation estrogen receptor alpha. METHODS: RBCK1 and YAP protein expression levels were measured by western blotting, while the mRNA levels of YAP target genes were measured by RT-PCR. RNA sequencing data were analyzed by Ingenuity Pathway Analysis. Identification of Hippo signaling activity was accomplished with luciferase assays, RT-PCR and western blotting. Protein stability assays and ubiquitin assays were used to detect YAP protein degradation. Ubiquitin-based immunoprecipitation assays were used to detect the specific ubiquitination modification on the YAP protein. RESULTS: In our current study, our data revealed an opposite function for RBCK1 in TNBC progression. RBCK1 over-expression inhibited TNBC cell progression in vitro and in vivo, while RBCK1 depletion promoted TNBC cell invasion. The whole genomic expression profiling showed that RBCK1 depletion activated Hippo/YAP axis. RBCK1 depletion increased YAP protein level and Hippo target gene expression in TNBC. The molecular biology studies confirmed that RBCK1 could bind to YAP protein and enhance the stability of YAP protein by promoting YAP K48-linked poly-ubiquitination at several YAP lysine sites (K76, K204 and K321). CONCLUSION: Our study revealed the multi-faced RBCK1 function in different subtypes of breast cancer patients and a promising therapeutic target for TNBC treatment. Video abstract.


Assuntos
Neoplasias de Mama Triplo Negativas , Humanos , Neoplasias de Mama Triplo Negativas/metabolismo , Receptor alfa de Estrogênio/metabolismo , Lisina , Fatores de Transcrição/metabolismo , Linhagem Celular Tumoral , Ubiquitina-Proteína Ligases , RNA Mensageiro , Ubiquitinas , Proliferação de Células
2.
Arch Virol ; 166(2): 439-449, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33389105

RESUMO

Chicken infectious anemia (CIA), caused by chicken anemia virus (CAV), is an important immunosuppressive disease that seriously threatens the global poultry industry. Here, we isolated and identified 30 new CAV strains from CAV-positive flocks. The VP1 genes of these strains were sequenced and analyzed at the nucleotide and amino acid levels and were found to have very similar nucleotide sequences (> 97% identity); however, they showed 93.9-100.0% sequence identity to the VP1 genes of 55 reference strains. Furthermore, alignment of the deduced amino acid sequences revealed some unique mutations. Phylogenetic analysis indicated the division of VP1 amino acid sequences into two groups (A and B) and four subgroups (A1, A2, A3 and A4). Interestingly, 22 of the newly isolated strains and some Asian reference strains belonged to the A1 group, whereas the remaining eight new isolates belonged to the A3 group. To evaluate the pathogenicity of the epidemic CAV strains from China, the representative strains CAV-JL16/8901 and CAV-HeN19/3001 and the reference strain Cux-1 were selected for animal experiments. Chickens infected with the isolates and reference strain all showed thymus atrophy and bone marrow yellowing. The mortality rates for CAV-JL16/8901, CAV-HeN19/3001, and the reference strain was 30%, 20%, and 0%, respectively, indicating that the epidemic strains pose a more serious threat to chickens. We not only analyzed the molecular evolution of the epidemic strains but also showed for the first time that the epidemic strains in China are more pathogenic than reference strain Cux-1. Effective measures should be established to prevent the spread of CIA in China.


Assuntos
Vírus da Anemia da Galinha/genética , Vírus da Anemia da Galinha/patogenicidade , Galinhas/virologia , Animais , China , Infecções por Circoviridae/virologia , DNA Viral/genética , Evolução Molecular , Genótipo , Epidemiologia Molecular/métodos , Filogenia , Doenças das Aves Domésticas/virologia , Análise de Sequência de DNA/métodos , Virulência/genética
3.
Biomed Chromatogr ; 34(2): e4724, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31755117

RESUMO

Lipid metabolism has a significant function in the central nervous system and Alzheimer's disease (AD) is an age-related senile disease characterized by central nerve degeneration. The pathological development of AD is closely related to lipid metabolism disorders. To reveal the influence of Kai-Xin-San (KXS) on lipid metabolism in APP/PSI transgenic mice and potential therapeutic targets for treating AD, brain tissue samples were collected and analyzed by high-throughput lipidomics based on UPLC-Q/TOF-MS. The collected raw data were processed by multivariate data analysis to discover the potential biomarkers and lipid metabolic profiles. Compared with the control wild-type mouse group, nine potential lipid biomarkers were found in the AD model group, of which seven were up-regulated and two were down-regulated. Orally administrated KXS can reverse the changes in these potential biomarkers. Compared with the model group, a total of six differential metabolites showed a recovery trend and may be potential targets for KXS to treat AD. This study showed that high-throughput lipidomics can be used to discover the perturbed pathways and lipid biomarkers as potential targets to reveal the therapeutic effects of KXS.


Assuntos
Doença de Alzheimer/metabolismo , Encéfalo/efeitos dos fármacos , Medicamentos de Ervas Chinesas/farmacologia , Lipidômica/métodos , Lipídeos/análise , Animais , Biomarcadores/análise , Encéfalo/metabolismo , Cromatografia Líquida de Alta Pressão/métodos , Modelos Animais de Doenças , Espectrometria de Massas/métodos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
4.
J Virol ; 91(5)2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-27974565

RESUMO

Autophagy functions as an intrinsic antiviral defense. However, some viruses can subvert or even enhance host autophagic machinery to increase viral replication and pathogenesis. The role of autophagy during avibirnavirus infection, especially late stage infection, remains unclear. In this study, infectious bursal disease virus (IBDV) was used to investigate the role of autophagy in avibirnavirus replication. We demonstrated IBDV induction of autophagy as a significant increase in puncta of LC3+ autophagosomes, endogenous levels of LC3-II, and ultrastructural characteristics typical of autophagosomes during the late stage of infection. Induction of autophagy enhances IBDV replication, whereas inhibition of autophagy impairs viral replication. We also demonstrated that IBDV infection induced autophagosome-lysosome fusion, but without active degradation of their contents. Moreover, inhibition of fusion or of lysosomal hydrolysis activity significantly reduced viral replication, indicating that virions utilized the low-pH environment of acidic organelles to facilitate viral maturation. Using immuno-transmission electron microscopy (TEM), we observed that a large number of intact IBDV virions were arranged in a lattice surrounded by p62 proteins, some of which lay between virions. Additionally, many virions were encapsulated within the vesicular membranes, with an obvious release stage observed by TEM. The autophagic endosomal pathway facilitates low-pH-mediated maturation of viral proteins and membrane-mediated release of progeny virions.IMPORTANCE IBDV is the most extensively studied virus in terms of molecular characteristics and pathogenesis; however, mechanisms underlying the IBDV life cycle require further exploration. The present study demonstrated that autophagy enhances viral replication at the late stage of infection, and the autophagy pathway facilitates IBDV replication complex function and virus assembly, which is critical to completion of the virus life cycle. Moreover, the virus hijacks the autophagic vacuoles to mature in an acidic environment and release progeny virions in a membrane-mediated cell-to-cell manner. This autophagic endosomal pathway is proposed as a new mechanism that facilitates IBDV maturation, release, and reinternalization. This report presents a concordance in exit strategies among some RNA and DNA viruses, which exploit autophagy pathway for their release from cells.


Assuntos
Autofagia , Infecções por Birnaviridae/veterinária , Vírus da Doença Infecciosa da Bursa/fisiologia , Doenças das Aves Domésticas/virologia , Vacúolos/virologia , Animais , Infecções por Birnaviridae/virologia , Linhagem Celular , Embrião de Galinha , Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Imunidade Inata , Transdução de Sinais , Vacúolos/fisiologia , Internalização do Vírus , Liberação de Vírus , Replicação Viral
5.
J Biol Chem ; 291(28): 14815-25, 2016 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-27226547

RESUMO

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


Assuntos
Fusão Celular , Metapneumovirus/fisiologia , Infecções por Paramyxoviridae/fisiopatologia , Receptores de Vitronectina/fisiologia , Proteínas Virais de Fusão/fisiologia , Animais , Linhagem Celular , Infecções por Paramyxoviridae/virologia , Replicação Viral
6.
J Virol ; 90(24): 11231-11246, 2016 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-27707927

RESUMO

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


Assuntos
Interações Hospedeiro-Patógeno , Metapneumovirus/genética , Infecções por Paramyxoviridae/enzimologia , Doenças das Aves Domésticas/enzimologia , Proteínas Virais de Fusão/química , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Linhagem Celular , Galinhas , Chlorocebus aethiops , Cricetulus , Células Epiteliais/enzimologia , Células Epiteliais/imunologia , Células Epiteliais/virologia , Fibroblastos/enzimologia , Fibroblastos/imunologia , Fibroblastos/virologia , Regulação da Expressão Gênica , Metapneumovirus/crescimento & desenvolvimento , Metapneumovirus/imunologia , Modelos Moleculares , Infecções por Paramyxoviridae/imunologia , Infecções por Paramyxoviridae/virologia , Doenças das Aves Domésticas/imunologia , Doenças das Aves Domésticas/virologia , Domínios Proteicos , Estrutura Secundária de Proteína , Proteólise , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Células Vero , Proteínas Virais de Fusão/antagonistas & inibidores , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/metabolismo , Internalização do Vírus , Replicação Viral
7.
Virol J ; 14(1): 38, 2017 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-28222746

RESUMO

BACKGROUND: Chicken anemia virus (CAV) causes anemia and immune suppression, which are important diseases in the poultry industry. CAV VP3, also referred as 'apoptin', has been shown to selectively kill tumor cells, raising great hopes for its utilization as an anticancer therapy. The ability of apoptin to induce apoptosis is closely related to its nuclear localization. The C-terminal region of apoptin contains a bipartite nuclear localization signals (NLS), and a nuclear export signal (NES) is located between the arms of the NLS. Most previous studies have expressed apoptin of different lengths in vitro to understand the relationship between its localization and its induction of apoptosis. METHODS: In this study, we investigated the replication of CAV and its induction of apoptosis in vitro and in vivo with VP3-truncated infectious virus. Quantitative PCR was used to detect viral replication in MDCC-MSB1 cells, and the viral localization was observed by confocal microscopy. Flow cytometry was uesed to analyze virus-induced apoptosis in MDCC-MSB1 cells. Additionally, chickens infected with the rescued viruses compared with the parental virus rM9905 to evaluate the viral replication in vivo and virulence. RESULTS: Based on the infectious clone, we rescued two viruses in which were deleted NES-NLS2 (rCAV-VP3N88) or NLS1-NES-NLS2 (rCAV-VP3N80) in the C-terminal region of apoptin. The viral load of rCAV-VP3N88 decreased significantly between 60 and 108 hpi, and was always 10-100-fold lower than that of the parental virus rM9905. The levels of rCAV-VP3N80 were also 10-100-fold lower than that of rM9905 and declined significantly at three time points. There was almost no difference in the viral loads of rCAV-VP3N88 and rCAV-VP3N80. Additionally, rM9905 induced 85.39 ± 2.18% apoptosis at 96 hpi, whereas rCAV-VP3N88 and rCAV-VP3N80 induced 63.08 ± 4.78% and 62.56 ± 7.35% apoptosis, respectively, which were significantly (about 20%) lower than that induced by the parental virus. The rescued viruses altered the nuclear localization in MDCC-MSB1 cells. Moreover, deletion of C-terminal region of apoptin impaired viral replication in vivo and reduced the virulence of CAV in chickens. CONCLUSIONS: In summary, we have demonstrated that the C-terminal deletion of apoptin in infectious CAV affected the replication of the virus. The deletion of the C-terminal region of apoptin not only significantly reduced viral replication in vitro but also reduced its induction of apoptosis, which correlated with the loss of its nuclear localization. The deletion of the C-terminal region of apoptin also impaired the replication of CAV and attenuated its virulence in chickens.


Assuntos
Apoptose , Proteínas do Capsídeo/genética , Vírus da Anemia da Galinha/fisiologia , Vírus da Anemia da Galinha/patogenicidade , Fatores de Virulência/genética , Replicação Viral , Transporte Ativo do Núcleo Celular , Animais , Proteínas do Capsídeo/metabolismo , Linhagem Celular , Galinhas , Análise Mutacional de DNA , Citometria de Fluxo , Microscopia Confocal , Reação em Cadeia da Polimerase em Tempo Real , Virulência , Fatores de Virulência/metabolismo
8.
Arch Virol ; 161(10): 2871-7, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27438076

RESUMO

Avian leukosis virus (ALV) is an avian oncogenic retrovirus that can induce various clinical tumors. The capsid protein P27 is the group-specific antigen of ALV and has many viral antigen sites that are easy to detect. In this study, we produced a monoclonal antibody (mAb), 3A9, that is specific for the P27 protein. A series of partially overlapping peptides were screened to define (181)PPSAR(185) as the minimal linear epitope recognized by mAb 3A9. The identified epitope could be recognized by chicken anti-ALV and mouse anti-ALV P27 sera. The epitope was highly conserved among a number of ALV-A, ALV-B and ALV-J strains. MAb 3A9 might be a valuable tool for the development of new immunodiagnostic approaches for ALV, and the defined linear epitope might help further our understanding of the antigenic structure of the P27 protein.


Assuntos
Vírus da Leucose Aviária/imunologia , Proteínas do Capsídeo/imunologia , Mapeamento de Epitopos , Epitopos de Linfócito B/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/isolamento & purificação , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/isolamento & purificação , Vírus da Leucose Aviária/genética , Proteínas do Capsídeo/genética , Galinhas , Sequência Conservada , Epitopos de Linfócito B/genética , Feminino , Camundongos Endogâmicos BALB C
9.
Virus Genes ; 52(1): 51-60, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26611441

RESUMO

During the course of our continuous surveillance of Gallid herpesvirus 2 (GaHV-2), 44 isolates were obtained from GaHV-2-positive chickens of different flocks in China from 2009 to 2013. The meq gene, considered as a major GaHV-2 oncogene, was sequenced and was found to contain an open reading frame of 1020 nucleotides encoding a 339 amino acid (aa) polypeptide in all isolates. Compared with the GaHV-2 GA strain, the meq genes in 15.9 % (7/44) of the isolates analyzed in this study contained an aa substitution mutation at position 88 (A to T) of which is the first report. The main characteristics of Chinese GaHV-2 isolates meq genes included the substitutions K77E, D80Y, V115A, T139A, P176R, and P217A, and the aa substitution frequency at positions 139 and 176 showed an increase. To test the pathogenicity of the isolates, a pathogenicity study and a vaccination-challenge test were performed on three selected isolates (ZY/1203, WC/1203, and WC/1110) and reference strain GA. The results showed that the three isolates induced gross Marek's disease (MD) lesions in 95.0-100 % cases, which was a higher rate than that obtained for strain GA (82.4 %). Three isolates induced mortality in 10-21.1 % of specific-pathogen-free chickens, which was similar to results with strain GA (23.5 %). The commercially available CVI988 vaccine induced lower protective indices (PIs) against ZY/1203 (82.4) and WC/1110 (83.3) as compared to those against WC/1203 (100) and GA (100). These results showed an evolving trend in the meq genes of the isolates; three isolates exhibited higher morbidity as compared to the reference strain and the vaccine induced lower PIs against two isolates as compared to that against the reference strain.


Assuntos
Galinhas/virologia , Herpesvirus Galináceo 2/patogenicidade , Animais , China/epidemiologia , Herpesvirus Galináceo 2/classificação , Herpesvirus Galináceo 2/genética , Doença de Marek/epidemiologia , Doença de Marek/virologia , Proteínas Oncogênicas Virais/genética , Filogenia , Virulência
10.
J Gen Virol ; 96(Pt 1): 150-158, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25274857

RESUMO

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


Assuntos
Vírus da Leucose Aviária/genética , Leucose Aviária/virologia , Galinhas/virologia , Replicação do DNA/genética , Doenças das Aves Domésticas/virologia , Vírus do Sarcoma de Rous/genética , Replicação Viral/genética , Animais , China
11.
Protein Expr Purif ; 114: 64-70, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26102339

RESUMO

The protease (PR) encoded by most retroviruses is deeply involved in the lifecycle and infection process of retroviruses by possessing the specificity necessary to correctly cleave the viral polyproteins and host cell proteins. However, as an important representative of avian retroviruses, the enzymatic properties of PR from reticuloendotheliosis virus (REV) have not been clearly documented. The recombinant PR, its mutant fused with a His-tag, and its substrate p18-p30 fused with a GST-tag were expressed in the Escherichia coli system as soluble enzymes. The soluble PR and p18-p30 were purified using Ni-NTA His Bind Resin and Glutathione Sepharose 4B, respectively. The enzymatic activity of PR was analyzed using the substrate of p18-p30. The expressed prokaryotic protease has enzyme activity that is dependent on such conditions as temperature, pH, and ions, and its activity can be inhibited by caspase inhibitor and the divalent metal ions Ca(2+) and Ni(2+). In addition, the key role of the residue Thr (amino acids 28) for the enzymatic activity of PR was identified. Furthermore, the caspase inhibitor Z-VAD-FMK was confirmed to inhibit the PR enzymatic activity of REV. For the first time, the PR of REV was expressed in the soluble form, and the optimal enzymatic reaction system in vitro was developed and preliminarily used. This study provides essential tools and information for further understanding the infection mechanism of REV and for the development of antiviral drugs treating retroviruses.


Assuntos
Peptídeo Hidrolases/metabolismo , Proteínas Recombinantes/metabolismo , Vírus da Reticuloendoteliose/enzimologia , Proteínas Virais/metabolismo , Escherichia coli/genética , Mutação , Peptídeo Hidrolases/química , Peptídeo Hidrolases/genética , Peptídeo Hidrolases/isolamento & purificação , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Vírus da Reticuloendoteliose/genética , Proteínas Virais/química , Proteínas Virais/genética , Proteínas Virais/isolamento & purificação
12.
Arch Virol ; 160(10): 2445-53, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26175070

RESUMO

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


Assuntos
Variação Genética , Metapneumovirus/genética , Infecções por Paramyxoviridae/veterinária , Doenças das Aves Domésticas/virologia , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Chlorocebus aethiops , Humanos , Metapneumovirus/química , Metapneumovirus/classificação , Metapneumovirus/metabolismo , Dados de Sequência Molecular , Infecções por Paramyxoviridae/virologia , Alinhamento de Sequência , Perus/virologia , Células Vero , Proteínas Virais de Fusão/genética
13.
Arch Virol ; 160(9): 2231-5, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26112763

RESUMO

Reticuloendotheliosis virus (REV) causes an immunosuppressive and oncogenic disease in chickens and other birds. In this study, based on an infectious clone of REV, named HLJR0901, a recombinant virus containing the enhanced green fluorescence protein (EGFP) gene was constructed by inserting the EGFP expression cassette downstream of the 3' terminus of the viral env gene. An EGFP-tagged REV that stably expresses EGFP was rescued. This visible recombinant REV could contribute to the further understanding of the molecular mechanism involved in the replication and pathogenicity of REV.


Assuntos
Proteínas de Fluorescência Verde/biossíntese , Vírus da Reticuloendoteliose/genética , Vírus da Reticuloendoteliose/fisiologia , Coloração e Rotulagem/métodos , Virologia/métodos , Replicação Viral , Animais , Aves , Galinhas , Proteínas de Fluorescência Verde/genética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Recombinação Genética
14.
Arch Virol ; 160(4): 995-1004, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25655260

RESUMO

Avian leukosis virus subgroup J (ALV-J) is an avian oncogenic retrovirus that has caused severe economic losses in China. Gp85 protein is the main envelope protein and the most variable structural protein of ALV-J. It is also involved in virus neutralization. In this study, a specific monoclonal antibody, 4A3, was produced against the ALV-J gp85 protein. Immunofluorescence assays showed that 4A3 could react with different strains of ALV-J, including the British prototype isolate HPRS103, the American strains, an early Chinese broiler isolate, and layer isolates. A linear epitope on the gp85 protein was identified using a series of partially overlapping fragments spanning the gp85-encoding gene and subjecting them to western blot analysis. The results indicated that (134)AEAELRDFI(142) was the minimal linear epitope that could be recognized by mAb 4A3. Enzyme-linked immunosorbent assay (ELISA) revealed that chicken anti-ALV-J sera and mouse anti-ALV-J gp85 sera could also recognize the minimal linear epitope. Alignment analysis of amino acid sequences indicated that the epitope was highly conserved among 34 ALV-J strains. Furthermore, the epitope was not conserved among subgroup A and B of avian leukosis virus (ALV). Taken together, the mAb and the identified epitope may provide valuable tools for the development of new diagnostic methods for ALV-J.


Assuntos
Vírus da Leucose Aviária/imunologia , Leucose Aviária/virologia , Epitopos de Linfócito B/química , Epitopos de Linfócito B/imunologia , Doenças das Aves Domésticas/virologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/imunologia , Leucose Aviária/imunologia , Vírus da Leucose Aviária/química , Vírus da Leucose Aviária/genética , Vírus da Leucose Aviária/isolamento & purificação , Galinhas , Mapeamento de Epitopos , Epitopos de Linfócito B/genética , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Doenças das Aves Domésticas/imunologia , Especificidade da Espécie , Proteínas do Envelope Viral/genética
15.
Virus Genes ; 50(3): 418-24, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25850423

RESUMO

Reticuloendotheliosis virus (REV), classified as a gammaretrovirus, has a variety of hosts, including chickens, ducks, geese, turkeys, and wild birds. REV causes a series of pathological syndromes, especially the immunosuppression of the host, which may lead to an increased susceptibility to other pathogens, thus greatly damaging the poultry industry. Mixed infections of REV and Marek's disease virus (MDV) have been reported in many countries, including China. Previous reports revealed that MDV vaccines were not efficacious, and even less-virulent MDV strains would cause some losses due to mixed infections with REV. Additionally, contaminants in the MDV vaccine might be the main source of REV. In this study, two clinical samples were collected from two flocks of chickens that were diagnosed with MDV. Subsequently, two REV isolates were obtained from the clinical samples. The isolates, named CY1111 and SY1209, were further confirmed through an indirect immunofluorescence assay and electron microscopy. Complete genome sequences of the two REV strains were determined to test the relationship between them and other REV strains. Phylogenetic trees showed that the two REV strains were closely related to most REV strains that were isolated from a variety of hosts. Therefore, REVs might spread freely among these hosts under natural conditions. Additionally, most REV strains in China were in the same clade. The present work offers some information regarding REV in China.


Assuntos
Coinfecção/veterinária , Coinfecção/virologia , Genoma Viral , Doenças das Aves Domésticas/virologia , Vírus da Reticuloendoteliose/genética , Vírus da Reticuloendoteliose/isolamento & purificação , Infecções por Retroviridae/veterinária , Animais , Galinhas , China , Análise por Conglomerados , Herpesvirus Galináceo 2/isolamento & purificação , Doença de Marek/complicações , Microscopia , Dados de Sequência Molecular , Filogenia , RNA Viral/genética , Infecções por Retroviridae/complicações , Infecções por Retroviridae/virologia , Análise de Sequência de DNA , Homologia de Sequência
16.
J Gen Virol ; 95(Pt 4): 888-897, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24431234

RESUMO

Infectious bursal disease virus (IBDV) causes an economically significant disease of chickens worldwide. The emergence of very virulent IBDV (vvIBDV) has brought more challenges for effective prevention of this disease. The molecular basis for the virulence of vvIBDV is not fully understood. In this study, 20 IBDV strains were analysed phylogenically and clustered in three branches based on their full-length B segments. The amino acid triplet located at positions 145/146/147 of VP1 was found highly conserved in branch I non-vvIBDVs as asparagine/glutamic acid/glycine (NEG), in branch II vvIBDVs as threonine/glutamic acid/glycine (TEG) and in branch III vvIBDVs as threonine/aspartic acid/asparagine (TDN). Further studies showed that the three amino acids play a critical role in the replication and pathogenicity of vvIBDV. Substitution of the TDN triplet with TEG or NEG reduced viral replication and pathogenicity of the vvIBDV HuB-1 strain in chickens. However, the replication of the attenuated IBDV Gt strain was reduced in chicken embryo fibroblast cells, whilst it was enhanced in the bursa by substituting NEG with TEG or TDN. The exchange of the three amino acids was also found to be capable of affecting the polymerase activity of VP1. The important role of segment B in the pathogenicity of IBDV was confirmed in this study. These results also provided new insights into the mechanism of the virulence of vvIBDVs and may offer new targets for their attenuation to develop potential vaccines using reverse genetics.


Assuntos
Aminoácidos/genética , RNA Polimerases Dirigidas por DNA/genética , Vírus da Doença Infecciosa da Bursa/genética , Vírus da Doença Infecciosa da Bursa/patogenicidade , Proteínas Estruturais Virais/genética , Fatores de Virulência/genética , Substituição de Aminoácidos , Animais , Bolsa de Fabricius/virologia , Células Cultivadas , Galinhas , Análise por Conglomerados , Análise Mutacional de DNA , Fibroblastos/virologia , Mutagênese Sítio-Dirigida , Filogenia , Virulência , Replicação Viral
17.
J Clin Microbiol ; 52(1): 37-44, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24131697

RESUMO

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


Assuntos
Vírus da Leucose Aviária/classificação , Vírus da Leucose Aviária/isolamento & purificação , Leucose Aviária/diagnóstico , Leucose Aviária/virologia , Técnicas de Diagnóstico Molecular/métodos , Reação em Cadeia da Polimerase Multiplex/métodos , Medicina Veterinária/métodos , Animais , Vírus da Leucose Aviária/genética , Epidemiologia Molecular/métodos , Aves Domésticas , Sensibilidade e Especificidade , Fatores de Tempo
18.
Arch Virol ; 159(7): 1821-6, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24488027

RESUMO

Avian leukosis virus subgroup J (ALV-J), first isolated in 1989, preferentially infects meat-type birds. Chinese layer flocks have experienced outbreaks of this virus since 2008. To analyze the status of ALV-J infection in wild birds in China, 585 wild birds collected from three provinces of Northeast China from 2010 to 2012 were tested, and six ALV-J strains were isolated for the first time. Furthermore, the gp85 genes of the six strains were amplified, cloned, and sequenced. The results indicated that two different ALV-J strains coexisted in Chinese wild birds from 2010 to 2012. These results not only expand the epidemiological data available for ALV-J and provide necessary information for the further understanding of the evolution of ALV-J, but they also highlight the potential role of wild-bird migration in the spread of ALV-J.


Assuntos
Vírus da Leucose Aviária/genética , Vírus da Leucose Aviária/metabolismo , Leucose Aviária/virologia , Variação Genética , Proteínas do Envelope Viral/genética , Animais , Animais Selvagens , Leucose Aviária/epidemiologia , Aves , China/epidemiologia , Dados de Sequência Molecular , Filogenia
19.
Arch Virol ; 159(8): 2051-7, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24643331

RESUMO

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


Assuntos
Doenças das Aves/virologia , Vírus da Reticuloendoteliose/isolamento & purificação , Infecções por Retroviridae/veterinária , Animais , Anseriformes/virologia , Feminino , Masculino , Dados de Sequência Molecular , Filogenia , Vírus da Reticuloendoteliose/classificação , Vírus da Reticuloendoteliose/genética , Infecções por Retroviridae/virologia
20.
Virus Genes ; 49(2): 250-8, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24854142

RESUMO

The J-subgroup avian leukosis virus (ALV-J) strain WB11098J was isolated from a wild Eurasian teal, and its proviral genomic sequences were determined. The complete proviral sequence of WB11098J was 7868 nt long. WB11098J was 95.3.9 % identical to the prototype strain HPRS-103, 94.2 % identical to the American strain ADOL-7501, 94.5-94.7 % identical to Chinese broiler isolates, 94.8-97.5 % identical to layer chicken isolates, and 94.4-95.0 % identical to Chinese local chicken isolates at the nucleotide level. Phylogenetic analysis showed that the WB11098J isolate shared the greatest homology with the layer strain SD09DP03 and was included in the same cluster. Interestingly, two 19-bp insertions in the U3 regions of the 5'LTR and 5'UTR that were most likely derived from other retroviruses were found in the WB11098J isolate. These insertions separately introduced one E2BP-binding site in the U3 region of the 5'LTR and a RNA polymerase II transcription factor IIB and core promoter motif of ten elements in the 5'UTR. A 5-bp deletion was identified in the U3 region of the 5'LTR. No nucleotides were deleted in the rTM or DR-1 regions in the 3'UTR. A 1-bp deletion was detected in the E element and introduced a specific and distinct binding site for c-Ets-1. Our study is the first to report the molecular characteristics of the complete genome of an ALV-J that was isolated from a wild bird and will provide necessary information for further understanding of the evolution of ALV-J.


Assuntos
Anseriformes/virologia , Vírus da Leucose Aviária/classificação , Vírus da Leucose Aviária/isolamento & purificação , DNA Viral/genética , Genoma Viral , Análise de Sequência de DNA , Animais , Vírus da Leucose Aviária/genética , China , Análise por Conglomerados , DNA Viral/química , Genótipo , Dados de Sequência Molecular , Filogenia , Provírus/classificação , Provírus/genética , Provírus/isolamento & purificação , Homologia de Sequência
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa