Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Curr Microbiol ; 81(9): 296, 2024 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-39105989

RESUMO

Duck enteritis virus (DEV) may lead to vascular injury, gastrointestinal mucosal erosion, lymphoid organ injury, and Polyinosinic-polycytidylic acid (Poly I:C) has an antiviral effect by inducing low levels of interferon. The purpose of this study was to explore the pathogenesis of DEV-induced intestinal injury in ducks and to verify the therapeutic effects of different concentrations of Poly I:C. In this study, duck enteritis model was established by infecting healthy Pekin ducks with DEV. Duck intestinal tissues were extracted from normal control group, model group, and treatment group with different doses of Poly I:C. In vivo, HE and TUNEL staining were used to observe the morphological changes and apoptosis. In vitro, the proliferation and apoptosis of duck intestinal epithelial cells were evaluated by MTT assay, TUNEL staining, and flow cytometry. The results showed that Poly I:C protected ducks from DEV toxicity by improving intestinal morphology and inhibiting apoptosis. In addition, the antiviral effect of Poly I:C on DEV was found in a dose-dependent manner, with a more relatively obvious effect at a high dose of Poly I:C. All in all, these results demonstrated that Poly I:C played a vital role in the apoptosis induced by DEV in ducks and modest dose of Poly I:C treatment worked well and may provide important reference for the development of new antiviral drugs in the future.


Assuntos
Apoptose , Patos , Enterite , Poli I-C , Animais , Patos/virologia , Poli I-C/farmacologia , Poli I-C/administração & dosagem , Apoptose/efeitos dos fármacos , Enterite/virologia , Enterite/tratamento farmacológico , Enterite/veterinária , Doenças das Aves Domésticas/virologia , Doenças das Aves Domésticas/tratamento farmacológico , Intestinos/virologia , Intestinos/patologia , Antivirais/farmacologia , Mardivirus/efeitos dos fármacos , Mucosa Intestinal/virologia , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/patologia
2.
BMC Vet Res ; 16(1): 371, 2020 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-33008383

RESUMO

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.


Assuntos
Flavonoides/farmacologia , Mardivirus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Animais , Antivirais/farmacologia , Células Cultivadas , Embrião de Galinha , Citocinas/metabolismo , Fibroblastos , Flavonoides/toxicidade , Doença de Marek/virologia , RNA Mensageiro
3.
Sci Rep ; 10(1): 10919, 2020 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-32616820

RESUMO

Marek's disease virus (MDV) is a highly cell-associated alphaherpesvirus that causes deadly lymphomas in chickens. While vaccination protects against clinical symptoms, MDV field strains can still circulate in vaccinated flocks and continuously evolve towards greater virulence. MDV vaccines do not provide sterilizing immunity, allowing the virus to overcome vaccine protection, and has increased the need for more potent vaccines or alternative interventions. In this study, we addressed if the CRISPR/Cas9 system can protect cells from MDV replication. We first screened a number of guide RNAs (gRNAs) targeting essential MDV genes for their ability to prevent virus replication. Single gRNAs significantly inhibited virus replication, but could result in the emergence of escape mutants. Strikingly, combining two or more gRNAs completely abrogated virus replication and no escape mutants were observed upon serial passaging. Our study provides the first proof-of-concept, demonstrating that the CRISPR/Cas9 system can be efficiently used to block MDV replication. The presented findings lay the foundation for future research to completely protect chickens from this deadly pathogen.


Assuntos
Sistemas CRISPR-Cas , Mardivirus/efeitos dos fármacos , RNA Guia de Cinetoplastídeos/farmacologia , Replicação Viral/efeitos dos fármacos , Animais , Embrião de Galinha , Galinhas , Patos , Genes Virais , Células HEK293 , Humanos , Mardivirus/genética , Mardivirus/fisiologia , Doença de Marek/prevenção & controle , Vacinas contra Doença de Marek , Mutação , Estudo de Prova de Conceito , RNA Guia de Cinetoplastídeos/genética , Organismos Livres de Patógenos Específicos , Replicação Viral/genética
4.
Dev Comp Immunol ; 98: 205-213, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30986433

RESUMO

Galectin-1, as a typical animal galactose-binding protein, it is found on the cell surface and in the extracellular matrix. Cloning the full-length coding sequence of galectin-1 from the spleens of Cherry Valley ducks revealed that the coding sequence of duck galectin-1 (duGal-1) comprises 405 bp, encoding 134 amino acids. Homologic analysis revealed its amino acid sequence is most identical to that of Anas platyrhynchos (98.8%) followed by Gallus gallus. Quantitative real-time PCR analysis indicated that duGal-1 mRNA is broadly expressed in healthy Cherry Valley duck tissues, primarily in the heart and trachea but minimally in the lung and skin. Meanwhile, the duGal-1 expression is slightly upregulated in the infected liver and spleen. Furthermore, the expression levels of ISGs (Mx, PKR, OAS) and some cytokines such as IFN-α, IL-1ß, IL-2, are up-regulated to varying degrees after overexpression the duGal-1, In contrast, Knockdown of duGal-1 found that the expression levels of ISGs and some inflammatory cytokines were down-regulated. Antiviral assay showed that duGal-1 could inhibit viral replications early during infection. This is the first study of the cloning, tissue distribution, and antiviral immune responses of duGal-1, and findings imply it is involved in the early stages of antiviral innate immune responses to duck plague virus infections in ducks.


Assuntos
Antivirais/imunologia , Patos/imunologia , Galectina 1/imunologia , Perfilação da Expressão Gênica/métodos , Mardivirus/imunologia , Doenças das Aves Domésticas/imunologia , Sequência de Aminoácidos , Animais , Antivirais/metabolismo , Antivirais/farmacologia , Células Cultivadas , Clonagem Molecular , Citocinas/genética , Citocinas/imunologia , Citocinas/metabolismo , Patos/genética , Patos/virologia , Galectina 1/classificação , Galectina 1/genética , Mardivirus/efeitos dos fármacos , Mardivirus/fisiologia , Filogenia , Doenças das Aves Domésticas/prevenção & controle , Doenças das Aves Domésticas/virologia , Interferência de RNA , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos
5.
Virol J ; 14(1): 183, 2017 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-28931412

RESUMO

BACKGROUND: There is little information regarding the duck enteritis virus (DEV) US10 gene and its molecular characterization. METHODS: Duck enteritis virus US10 was amplified and cloned into the recombinant vector pET32a(+). The recombinant US10 protein was expressed in Escherichia coli BL21 cells and used to immunize rabbits for the preparation of polyclonal antibodies. The harvested rabbit antiserum against DEV US10 was detected and analyzed by agar immunodiffusion. Using this antibody, western blotting and indirect immunofluorescence analysis were used to analyze the expression level and subcellular localization of US10 in infected cells at different time points. Quantitative reverse-transcription PCR (qRT-PCR) and pharmacological inhibition tests were used to ascertain the kinetic class of the US10 gene. A mass spectrometry-based strategy was used to identify US10 in purified DEV virions and quantify its abundance. RESULTS: The recombinant pET32a(+)/US10 protein was expressed as inclusion bodies, purified by gradient urea washing, and used to prepare specific antibodies. The results of qRT-PCR, western blotting, and pharmacological inhibition tests revealed that US10 is mainly transcribed in the late stage of viral replication. However, the presence of the DNA polymerase inhibitor ganciclovir and the protein synthesis inhibitor cycloheximide blocked transcription. Therefore, US10 is a γ2 (true late) gene. Indirect immunofluorescence analysis showed that US10 proteins were initially diffusely distributed throughout the cytoplasm, but with the passage of time, they gradually relocated to a perinuclear region. The US10 protein was detected in purified DEV virions by mass spectrometry, but was not detected by western blotting, indicating that DEV US10 is a minor virion protein. CONCLUSIONS: The DEV US10 gene is a γ2 gene and the US10 protein is localized in the perinuclear region. DEV US10 is a virion component.


Assuntos
Patos/virologia , Mardivirus/classificação , Mardivirus/genética , Animais , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/isolamento & purificação , Linhagem Celular , Técnica Indireta de Fluorescência para Anticorpo , Regulação Viral da Expressão Gênica , Mardivirus/efeitos dos fármacos , Mardivirus/isolamento & purificação , Doença de Marek/virologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Virais/genética , Proteínas Virais/imunologia , Vírion
6.
Genet Mol Res ; 14(4): 13779-90, 2015 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-26535693

RESUMO

The US2 protein has been reported to contribute to duck enteritis virus (DEV) infection; however, its kinetics and localization during infection, and whether it is a component of virion, have not been previously reported. To elucidate the function of DEV US2, US2 was amplified by polymerase chain reaction (PCR) and inserted into pET-32a(+); this was expressed, the recombinant US2 protein was purified, and a polyclonal antibody generated. In addition, the kinetics and localization of the US2 gene and protein were determined by quantitative real-time fluorescent PCR, ganciclovir (GCV), and cycloheximide (CHX) treatment, western-blot, and indirect immunofluorescence assay. The packaging of US2 into DEV virions was revealed by a protease protection assay. US2 was found to be transcribed 24 h post-infection (pi) and peaked at 72 h pi; the US2 protein was detected 48 h pi, except in the presence of GCV or CHX. US2 was packed into virions and also localized to the plasma membrane and cytoplasm in infected cells. The results showed that the DEV US2 is a late gene, and that its encoding protein could be a tegument component localized mainly in the cytoplasm. This study provides useful data for the further analysis of DEV US2, including an explanation for the genetic conservation among alphaherpesviruses.


Assuntos
Mardivirus/genética , Proteínas do Envelope Viral/genética , Animais , Linhagem Celular , Patos , Fibroblastos , Expressão Gênica , Mardivirus/efeitos dos fármacos , Transporte Proteico , Proteínas Recombinantes , Proteínas do Envelope Viral/isolamento & purificação , Proteínas do Envelope Viral/metabolismo , Vírion
7.
J Virol Methods ; 199: 11-6, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24412629

RESUMO

Newcastle disease virus (NDV) of paramyxovirus and Marek's disease virus (MDV) of herpesvirus, two of the most serious threats to the poultry industry, can give rise to complex co-infections that hinder diagnosis and prevention. In the current study, two different peptides, derived from the MDV gH (gHH2L) and gB (gBH3), respectively, exhibit antiviral activity against NDV in vitro. The potent inhibitory effect of heptad repeat 2 from fusion glycoprotein of the NDV on MDV infection also has been demonstrated. Plaque formation and embryo infectivity assays confirmed these antiviral results. Furthermore, each tandem peptide consisting of two motifs from different viruses exhibits more potent antiviral activity than the constituent peptides. The current work provides a new strategy for developing novel peptides and vaccines against virus infection and co-infections.


Assuntos
Antivirais/farmacologia , Glicoproteínas/farmacologia , Mardivirus/efeitos dos fármacos , Vírus da Doença de Newcastle/efeitos dos fármacos , Peptídeos/farmacologia , Proteínas Virais de Fusão/farmacologia , Animais , Antivirais/uso terapêutico , Linhagem Celular , Embrião de Galinha , Glicoproteínas/genética , Glicoproteínas/uso terapêutico , Mardivirus/genética , Doença de Marek/prevenção & controle , Testes de Sensibilidade Microbiana , Doença de Newcastle/prevenção & controle , Vírus da Doença de Newcastle/genética , Peptídeos/genética , Peptídeos/uso terapêutico , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/uso terapêutico , Ensaio de Placa Viral
8.
Antiviral Res ; 98(2): 344-51, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23523763

RESUMO

Duck enteritis virus (DEV) of the family Herpesviridae is one of the main diseases in waterfowl. Despite the wide use of vaccines to control the disease, infection with the virus cannot be completely prevented. Therefore, antiviral agents against DEV should be developed. This study presents a novel sulfated polysaccharide from Chuanminshen violaceum (sCVPS), which exhibits significant antiviral activity against DEV with 50% inhibitory concentrations (IC50) ranging from 77.12 µg/mL to 104.81 µg/mL. sCVPS is more effective than heparan sulfate (HS, as a positive control) with IC50=132.61 µg/mL. sCVPS and HS inhibit viral activity by preventing virus adsorption with IC50 values ranging from 82.83 µg/mL to 109.28 µg/mL for sCVPS and 150.22 µg/mL for HS. Direct immunofluorescence assay and transmission electron microscopy demonstrated that the mechanism of action was the interference with virus adsorption. The amount of inhibited virus during adsorption was quantified using fluorescent quantitative polymerase chain reaction, which revealed that both sCVPS and HS can significantly reduce all viruses attached to cells. sCVPS also prevented the cell-to-cell spread of DEV. These results indicated that sCVPSs perform more effectively than does HS as antiviral agents against DEV and can be further examined for potential effects as an alternative control measure for DEV infection.


Assuntos
Antivirais/farmacologia , Apiaceae/química , Enterite/veterinária , Mardivirus/efeitos dos fármacos , Extratos Vegetais/farmacologia , Polissacarídeos/farmacologia , Doenças das Aves Domésticas/virologia , Animais , Antivirais/química , Patos , Enterite/virologia , Mardivirus/fisiologia , Extratos Vegetais/química , Polissacarídeos/química
9.
Virol J ; 8: 190, 2011 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-21518442

RESUMO

BACKGROUND: Marek's disease virus (MDV), which is widely considered to be a natural model of virus-induced lymphoma, has the potential to cause tremendous losses in the poultry industry. To investigate the structural basis of MDV membrane fusion and to identify new viral targets for inhibition, we examined the domains of the MDV glycoproteins gH and gB. RESULTS: Four peptides derived from the MDV glycoprotein gH (gHH1, gHH2, gHH3, and gHH5) and one peptide derived from gB (gBH1) could efficiently inhibit plaque formation in primary chicken embryo fibroblast cells (CEFs) with 50% inhibitory concentrations (IC50) of below 12 µM. These peptides were also significantly able to reduce lesion formation on chorioallantoic membranes (CAMs) of infected chicken embryos at a concentration of 0.5 mM in 60 µl of solution. The HR2 peptide from Newcastle disease virus (NDVHR2) exerted effects on MDV specifically at the stage of virus entry (i.e., in a cell pre-treatment assay and an embryo co-treatment assay), suggesting cross-inhibitory effects of NDV HR2 on MDV infection. None of the peptides exhibited cytotoxic effects at the concentrations tested. Structural characteristics of the five peptides were examined further. CONCLUSIONS: The five MDV-derived peptides demonstrated potent antiviral activity, not only in plaque formation assays in vitro, but also in lesion formation assays in vivo. The present study examining the antiviral activity of these MDV peptides, which are useful as small-molecule antiviral inhibitors, provides information about the MDV entry mechanism.


Assuntos
Antivirais/farmacologia , Produtos Biológicos/farmacologia , Glicoproteínas/farmacologia , Mardivirus/crescimento & desenvolvimento , Doença de Marek/prevenção & controle , Peptídeos/farmacologia , Proteínas Estruturais Virais/farmacologia , Animais , Produtos Biológicos/genética , Células Cultivadas , Embrião de Galinha , Membrana Corioalantoide/patologia , Membrana Corioalantoide/virologia , Fibroblastos/virologia , Glicoproteínas/genética , Concentração Inibidora 50 , Mardivirus/efeitos dos fármacos , Mardivirus/genética , Doença de Marek/virologia , Testes de Sensibilidade Microbiana/métodos , Vírus da Doença de Newcastle/genética , Peptídeos/genética , Ensaio de Placa Viral , Proteínas Estruturais Virais/genética
10.
Wei Sheng Wu Xue Bao ; 45(5): 697-701, 2005 Oct.
Artigo em Chinês | MEDLINE | ID: mdl-16342758

RESUMO

The recombinant transfer vector pFastBacl-ChIFN-y was constructed by plasmid pcDNA-ChIFN-gamma digested with EcoR I and Not I enzymes and cloned into pFastbacl. Then the transfer vector was transformed into E. coli competent cells DH10Bac which contained the bacmid with amini-attTn7 target site and the helper plasmid. The recombinant bacmid-ChIFN-gamma was generated by transposing themini-Tn7 element located in pFastBacl-ChIFN-gamma to themini-attTn7 attachment site on the Bacmid. Subsequently the recombinant Bacmid-ChIFN-gamma was transfected into the Sf9 insect cells mediated by lipofectin to produce recombinant baculovirus and express recombinant ChIFN-gamma (rChIFN-gamma) products. The result showed that the rChIFN-gamma was successfully expressed in Sf9 cells infected with the recombinant virus by indirect immunofluorescence assay (IFA) at 5 days post-transfection. The biological activity of rChIFN-gamma was identified by its inhibition to Vesicular stomatitis virus-induced cytotoxicity of chicken embryonic fibroblasts (CEF) in vitro. The results showed that the most efficient expression of rChIFN-gamma could be obtained at 96h post-infection with multiplicity of infection (MOI) equal to 1. It is interesting that the viruses such as Avian influenza virus H5N1 or Marek's disease virus (GA strain) could not grow in CEF pre-treated with rChIFN-gamma. Cell pathogenic efficient (CPE) in the CEF infected with H5N1 and GA strain is apparently inhibited by the rChIFN-gamma. However only difference between the HA titres of the supernatant of the pre-treated cells is observed without any obvious inhibition effect in CEF infected with Newcastle disease virus (F48E8 strain).


Assuntos
Interferon gama/biossíntese , Animais , Galinhas , Virus da Influenza A Subtipo H5N1/efeitos dos fármacos , Interferon gama/genética , Interferon gama/farmacologia , Mardivirus/efeitos dos fármacos , Vírus da Doença de Newcastle/efeitos dos fármacos , Proteínas Recombinantes , Spodoptera/citologia , Spodoptera/genética
11.
Biochem Biophys Res Commun ; 331(1): 69-73, 2005 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-15845359

RESUMO

Ovotransferrin and lactoferrin are iron-binding proteins with antiviral and antibacterial activities related to natural immunity, showing marked sequence and structural homologies. The antiviral activity of two hen ovotransferrin fragments DQKDEYELL (hOtrf(219-227)) and KDLLFK (hOtrf(269-301) and hOtrf(633-638)) towards Marek's disease virus infection of chicken embryo fibroblasts is reported here. These fragments have sequence homology with two bovine lactoferrin fragments with antiviral activity towards herpes simplex virus, suggesting that these fragments could have a role for the exploitation of the antiviral activity of the intact proteins towards herpes viruses. NMR analysis showed that these peptides, chemically synthetized, did not possess any favourite conformation in solution, indicating that both the aminoacid sequence and the conformation they display in the intact protein are essential for the antiviral activity.


Assuntos
Antivirais/química , Antivirais/farmacologia , Conalbumina/farmacologia , Fragmentos de Peptídeos/farmacologia , Animais , Células Cultivadas , Embrião de Galinha , Conalbumina/química , Lactoferrina/química , Lactoferrina/farmacologia , Mardivirus/efeitos dos fármacos , Fragmentos de Peptídeos/química
12.
Biochem Cell Biol ; 80(1): 125-30, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-11908636

RESUMO

Ovotransferrin (formerly conalbumin) is an iron-binding protein present in birds. It belongs to the transferrin family and shows about 50% sequence homology with mammalian serum transferrin and lactoferrin. This protein has been demonstrated to be capable of delivering iron to cells and of inhibiting bacterial multiplication. However, no antiviral activity has been reported for ovotransferrin, although the antiviral activity of human and bovine lactoferrins against several viruses, including human herpes simplex viruses, has been well established. In this report, the antiviral activity of ovotransferrin towards chicken embryo fibroblast infection by Marek's disease virus (MDV), an avian herpesvirus, was clearly demonstrated. Ovotransferrin was more effective than human and bovine lactoferrins in inhibiting MDV infection and no correlation between antiviral efficacy and iron saturation was found. The observations reported here are of interest from an evolutionary point of view since it is likely that the defensive properties of transferrins appeared early in evolution. In birds, the defensive properties of ovotransferrin remained joined to iron transport functions; in mammals, iron transport functions became peculiar to serum transferrin, and the defensive properties towards infections were optimised in lactoferrin.


Assuntos
Antivirais/farmacologia , Conalbumina/farmacologia , Lactoferrina/farmacologia , Animais , Antivirais/metabolismo , Antivirais/toxicidade , Bovinos , Embrião de Galinha , Chlorocebus aethiops , Conalbumina/metabolismo , Conalbumina/toxicidade , Relação Dose-Resposta a Droga , Fibroblastos , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 1/fisiologia , Humanos , Ferro/metabolismo , Ferro/farmacologia , Lactoferrina/metabolismo , Lactoferrina/toxicidade , Mardivirus/efeitos dos fármacos , Mardivirus/fisiologia , Células Vero
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA