Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Pathog ; 18(6): e1010564, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35679257

RESUMO

The development of thermostable vaccines can relieve the bottleneck of existing vaccines caused by thermal instability and subsequent poor efficacy, which is one of the predominant reasons for the millions of deaths caused by vaccine-preventable diseases. Research into the mechanism of viral thermostability may provide strategies for developing thermostable vaccines. Using Newcastle disease virus (NDV) as model, we identified the negative surface charge of attachment glycoprotein as a novel determinant of viral thermostability. It prevented the temperature-induced aggregation of glycoprotein and subsequent detachment from virion surface. Then structural stability of virion surface was improved and virus could bind to and infect cells efficiently after heat-treatment. Employing the approach of surface charge engineering, thermal stability of NDV and influenza A virus (IAV) vaccines was successfully improved. The increase in the level of vaccine thermal stability was determined by the value-added in the negative surface charge of the attachment glycoprotein. The engineered live and inactivated vaccines could be used efficiently after storage at 37°C for at least 10 and 60 days, respectively. Thus, our results revealed a novel surface-charge-mediated link between HN protein and NDV thermostability, which could be used to design thermal stable NDV and IAV vaccines rationally.


Assuntos
Doença de Newcastle , Vacinas Virais , Animais , Galinhas/metabolismo , Glicoproteínas , Proteína HN/metabolismo , Doença de Newcastle/prevenção & controle , Vírus da Doença de Newcastle/metabolismo
2.
Gene Ther ; 28(12): 697-717, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-32409746

RESUMO

The direct oncolytic effect of Newcastle disease virus (NDV) depends on the following two aspects: the susceptibility of cancer cells to virus infection and the ability of virus itself to lyse cancer cells. First, we investigate the susceptibility of cancer cells to NDV infection, HepG2, MDA-MB-231, and SH-SY5Y cells were susceptible, A549, MCF7, and LoVo cells were less susceptible. To investigate the molecular mechanism responsible for cancer cell susceptibility, transcriptome sequencing was carried out. We found that the levels of alpha-sialic acid acyltransferase were upregulated in MDA-MB-231 cells compared with MCF7 cells, and the interferon was downregulated. Second, to optimize the oncolytic capacity of the wild-type rClone30, a series of chimeric viruses rClone30-Anh(HN), rClone30-Anh(F), and rClone30-Anh(HN-F) were constructed by exchanging the HN gene, F gene or both of non-lytic rClone30 strain with lytic strain Anhinga. rClone30-Anh(F) and rClone30-Anh(HN-F) enhanced the oncolytic effect of the rClone30, and this enhancement is more obvious in the susceptible cells. The oncolytic mechanism of rClone30-Anh(F) was analyzed by transcriptome analyses, in comparison with rClone30, rClone30-Anh(F) upregulated the expression of ATG5, Beclin 1, and MAP1LC3B, thus activating autophagy and promoting the production of syncytia. In conclusion, our study provides a strategy to enhance the oncolytic effect of rClone30.


Assuntos
Neoplasias , Terapia Viral Oncolítica , Vírus Oncolíticos , Animais , Linhagem Celular Tumoral , Vírus da Doença de Newcastle/genética , Vírus Oncolíticos/genética , Replicação Viral
3.
J Med Virol ; 93(6): 3890-3898, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-32779745

RESUMO

Oncolytic virus therapy is perhaps the next major breakthrough in cancer treatment following the success in immunotherapy using immune checkpoint inhibitors. However, the potential oncolytic ability of the recombinant newcastle disease virus (NDV) Anhinga strain carried with tumor necrosis factor-related apoptosis inducing ligand (TRAIL) has not been fully explored at present. In the present study, the recombinant NDV/Anh-TRAIL that secretes soluble TRAIL was constructed and the experiment results suggested NDV/Anh-TRAIL as a promising candidate for glioma therapy. Growth kinetic and TRAIL secreted quantity of recombinant NDV/Anh-TRAIL virus were measured. Cytotoxic and cell apoptosis were analyzed for its anti-glioma therapy in vitro. Nude mice were used for the in vivo evaluation. Both tumor volume and mice behavior after injection were observed. The recombinant virus replicated with the same kinetics as the parental virus and the highest expression of TRAIL (77.8 ng/L) was found at 48 hours. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, a tetrazole and flow cytometry data revealed that the recombinant NDV/Anh-TRAIL (56.1 ± 8.2%) virus could induce a more severe apoptosis rate, when compared with the NDV wild type (37.2 ± 7.0%) and mock (7.0 ± 1.8%) groups (P < .01), in U251 cells. Furthermore, in the present animal study, the average tumor volume was smaller in the NDV/Anh-TRAIL group (97.21 mm3 ), when compared with the NDV wild type (205.03 mm3 , P < .05) and PBS (310.30 mm3 , P < .01) groups.


Assuntos
Glioma/terapia , Vírus da Doença de Newcastle/genética , Vírus da Doença de Newcastle/imunologia , Terapia Viral Oncolítica/métodos , Ligante Indutor de Apoptose Relacionado a TNF/genética , Animais , Apoptose , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Camundongos Nus , Vírus Oncolíticos , Replicação Viral
4.
Arch Virol ; 166(12): 3333-3341, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34591172

RESUMO

The 2014 Ebola outbreak in West Africa resulted in more than 11,000 deaths, highlighting the need for a vaccine. A phase I clinical trial of a human adenovirus type 5 vector-based Ebola virus (EBOV) vaccine (HAdV-5-MakGP) showed that a homologous prime-boost regimen with HAdV-5 vaccine elicited a robust humoral response but a weak cellular immune response. Due to pre-existing anti-vector immunity, boosting with the same vaccine did not increase the magnitude of the cellular immune response, which contributes significantly to protection against EBOV infection. Here, we generated a recombinant Newcastle disease virus (NDV), based on the LaSota vaccine strain, expressing the GP protein of the EBOV variant Makona (rLS/EB-GP) using reverse genetics technology. The humoral and cellular immune responses to this vaccine candidate in mice immunized using a homologous prime-boost regimen or a heterologous prime-boost regimen with the HAdV-5-vectored Ebola vaccine were assessed using ELISA and ELISPOT assays. The ELISA and ELISPOT results showed that mice primed with rLS/EB-GP and boosted with HAdV-5-MakGP (NDV+HAdV-5) or, reversed, primed with HAdV-5-MakGP and boosted with rLS/EB-GP (HAdV-5+NDV) exhibited more-potent EBOV GP-specific antibody and cellular immune responses than those receiving the same vaccine twice. The most robust EBOV GP-specific antibody and T-cell responses were detected in the HAdV-5-MakGP-primed and rLS/EB-GP-boosted (HAdV-5+NDV) mice. These results suggest that the HAdV-5 prime-NDV boost regimen is more effective in stimulating EBOV-specific immunity than homologous regimens alone, indicating the potential boosting ability of the NDV vector in human vaccine use.


Assuntos
Vacinas contra Ebola , Ebolavirus , Doença pelo Vírus Ebola , Animais , Ebolavirus/genética , Vetores Genéticos , Doença pelo Vírus Ebola/prevenção & controle , Imunidade Celular , Camundongos , Vírus da Doença de Newcastle
6.
J Pharmacol Sci ; 132(1): 24-30, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27174862

RESUMO

Newcastle disease virus (NDV) have shown oncolytic therapeutic efficacy in preclinical study and are currently approved for clinical trials. NDV Anhinga strain which is a mesogenic strain should be classified as lytic strain and has a therapeutic efficacy in hepatocellular cancer. In this study, we evaluated the capacity of NDV Anhinga strain to elicit immune reaction in vivo and the possibility for using as a vaccine vector for expressing tumor therapeutic factors. Interleukin-2 (IL-2) could boost the immune response against the tumor cells. Therefore, we use NDV Anhinga strain as backbone to construct a recombinant virus (NDV/Anh-IL-2) expressing IL-2. The virus growth curve showed that the production of recombinant NDV/Anh-IL-2 was slightly delayed compared to the wild type. The NDV/Anh-IL-2 strain could express soluble IL-2 and effectively inhibit the growth of hepatocellular carcinoma in vivo. 60 days post-treatment, mice which were completely cured by previous treatment were well protected when rechallenged with the same tumor cell. From the H&E-stained sections, intense infiltration of lymphocyte was observed in the NDV Anhinga strain treated group, especially in NDV/Anh-IL-2 group. The NDV Anhinga strain could not only kill the tumor directly, but could also elicit immune reaction and a potent immunological memory when killing tumor in vivo. In conclusion, the Anhinga strain could be an effective vector for tumor therapy; the recombinant NDV/Anh-IL-2 strain expressing soluble IL-2 is a promising candidate for hepatoma therapy.


Assuntos
Carcinoma Hepatocelular/terapia , Interleucina-2/genética , Neoplasias Hepáticas/terapia , Vírus da Doença de Newcastle/genética , Animais , Vacinas Anticâncer , Carcinoma Hepatocelular/patologia , Linhagem Celular , Linhagem Celular Tumoral , Galinhas , Cricetinae , Feminino , Humanos , Neoplasias Hepáticas/patologia , Camundongos , Carga Tumoral
7.
J Gen Virol ; 96(Pt 1): 40-45, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25274858

RESUMO

Newcastle disease virus (NDV) has been developed as a vector for vaccine and gene therapy purposes. However, the optimal insertion site for foreign gene expression remained to be determined. In the present study, we inserted the green fluorescence protein (GFP) gene into five different intergenic regions of the enterotropic NDV VG/GA vaccine strain using reverse genetics technology. The rescued recombinant viruses retained lentogenic pathotype and displayed delayed growth dynamics, particularly when the GFP gene was inserted between the NP and P genes of the virus. The GFP mRNA level was most abundant when the gene was inserted closer to the 3' end and gradually decreased as the gene was inserted closer to the 5' end. Measurement of the GFP fluorescence intensity in recombinant virus-infected cells demonstrated that the non-coding region between the P and M genes is the optimal insertion site for foreign gene expression in the VG/GA vaccine vector.


Assuntos
Expressão Gênica/genética , Genes Virais/genética , Vetores Genéticos/genética , Mutagênese Insercional/genética , Vírus da Doença de Newcastle/genética , Vacinas Virais/genética , Proteínas de Fluorescência Verde/genética , Recombinação Genética/genética
8.
J Gen Virol ; 96(8): 2028-2035, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25872740

RESUMO

In the present study, we developed a novel approach for foreign gene expression by Newcastle disease virus (NDV) from a second ORF through an internal ribosomal entry site (IRES). Six NDV LaSota strain-based recombinant viruses vectoring the IRES and a red fluorescence protein (RFP) gene behind the nucleocapsid (NP), phosphoprotein (P), matrix (M), fusion (F), haemagglutinin-neuraminidase (HN) or large polymerase (L) gene ORF were generated using reverse genetics technology. The insertion of the second ORF slightly attenuated virus pathogenicity, but did not affect ability of the virus to grow. Quantitative measurements of RFP expression in virus-infected DF-1 cells revealed that the abundance of viral mRNAs and red fluorescence intensity were positively correlated with the gene order of NDV, 3'-NP-P-M-F-HN-L-5', proving the sequential transcription mechanism for NDV. The results herein suggest that the level of foreign gene expression could be regulated by selecting the second ORF insertion site to maximize the efficacy of vaccine and gene therapy.


Assuntos
Vetores Genéticos/genética , Vírus da Doença de Newcastle/genética , Genética Reversa/métodos , Ribossomos/genética , Transcrição Gênica , Animais , Galinhas , Expressão Gênica , Regulação Viral da Expressão Gênica , Vetores Genéticos/metabolismo , Doença de Newcastle , Vírus da Doença de Newcastle/metabolismo , Doenças das Aves Domésticas , Ribossomos/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Vacinas Virais/genética , Vacinas Virais/metabolismo
9.
J Gen Virol ; 96(Pt 6): 1219-1228, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25626679

RESUMO

Thermostable Newcastle disease virus (NDV) vaccines have been used widely to control Newcastle disease for village poultry flocks, due to their independence of cold chains for delivery and storage. To explore the potential use of thermostable NDV as a vaccine vector, an infectious clone of thermostable avirulent NDV strain TS09-C was developed using reverse genetics technology. The GFP gene, along with the self-cleaving 2A gene of foot-and-mouth disease virus and ubiquitin monomer (2AUbi), were inserted immediately upstream of the NP (nucleocapsid protein), M (matrix protein) or L (large polymerase protein) gene translation start codon in the TS09-C infectious clone. Detection of GFP expression in the recombinant virus-infected cells showed that the recombinant virus, rTS-GFP/M, with the GFP gene inserted into the M gene expressed the highest level of GFP. The rTS-GFP/M virus retained the same thermostability, growth dynamics and pathogenicity as its parental rTS09-C virus. Vaccination of specific-pathogen-free chickens with the rTS-GFP/M virus conferred complete protection against virulent NDV challenge. Taken together, the data suggested that the rTS09-C virus could be used as a vaccine vector to develop bivalent thermostable vaccines against Newcastle disease and the target avian diseases for village chickens, especially in the developing and least-developed countries.


Assuntos
Portadores de Fármacos , Vetores Genéticos , Instabilidade Genômica , Vírus da Doença de Newcastle/genética , Vacinas Virais/imunologia , Animais , Galinhas , Expressão Gênica , Genes Reporter , Proteínas de Fluorescência Verde/genética , Doença de Newcastle/prevenção & controle , Genética Reversa , Análise de Sobrevida , Ubiquitina/genética , Vacinação/métodos , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas Atenuadas/isolamento & purificação , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/isolamento & purificação , Proteínas Virais/genética , Vacinas Virais/administração & dosagem , Vacinas Virais/genética , Vacinas Virais/isolamento & purificação
10.
J Virol ; 88(15): 8397-406, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24829337

RESUMO

UNLABELLED: Infectious laryngotracheitis (ILT) is a highly contagious acute respiratory disease of chickens caused by infectious laryngotracheitis virus (ILTV). The disease is controlled mainly through biosecurity and vaccination with live attenuated strains of ILTV and vectored vaccines based on turkey herpesvirus (HVT) and fowlpox virus (FPV). The current live attenuated vaccines (chicken embryo origin [CEO] and tissue culture origin [TCO]), although effective, can regain virulence, whereas HVT- and FPV-vectored ILTV vaccines are less efficacious than live attenuated vaccines. Therefore, there is a pressing need to develop safer and more efficacious ILTV vaccines. In the present study, we generated Newcastle disease virus (NDV) recombinants, based on the LaSota vaccine strain, expressing glycoproteins B (gB) and D (gD) of ILTV using reverse genetics technology. These recombinant viruses, rLS/ILTV-gB and rLS/ILTV-gD, were slightly attenuated in vivo yet retained growth dynamics, stability, and virus titers in vitro that were similar to those of the parental LaSota virus. Expression of ILTV gB and gD proteins in the recombinant virus-infected cells was detected by immunofluorescence assay. Vaccination of specific-pathogen-free chickens with these recombinant viruses conferred significant protection against virulent ILTV and velogenic NDV challenges. Immunization of commercial broilers with rLS/ILTV-gB provided a level of protection against clinical disease similar to that provided by the live attenuated commercial vaccines, with no decrease in body weight gains. The results of the study suggested that the rLS/ILTV-gB and -gD viruses are safe, stable, and effective bivalent vaccines that can be mass administered via aerosol or drinking water to large chicken populations. IMPORTANCE: This paper describes the development and evaluation of novel bivalent vaccines against chicken infectious laryngotracheitis (ILT) and Newcastle disease (ND), two of the most economically important infectious diseases of poultry. The current commercial ILT vaccines are either not safe or less effective. Therefore, there is a pressing need to develop safer and more efficacious ILT vaccines. In the present study, we generated Newcastle disease virus (NDV) recombinants expressing glycoproteins B (gB) and D (gD) of infectious laryngotracheitis virus (ILTV) using reverse genetics technology. These recombinant viruses were safe, stable, and immunogenic and replicated efficiently in birds. Vaccination of chickens with these recombinant viruses conferred complete protection against ILTV and NDV challenge. These novel bivalent vaccines can be mass administered via aerosol or drinking water to large chicken populations at low cost, which will have a direct impact on poultry health, fitness, and performance.


Assuntos
Infecções por Herpesviridae/veterinária , Doença de Newcastle/prevenção & controle , Vírus da Doença de Newcastle/imunologia , Doenças das Aves Domésticas/prevenção & controle , Proteínas do Envelope Viral/imunologia , Vacinas Virais/imunologia , Administração Oral , Animais , Anticorpos Antivirais/sangue , Galinhas , Vetores Genéticos , Infecções por Herpesviridae/patologia , Infecções por Herpesviridae/prevenção & controle , Doença de Newcastle/patologia , Vírus da Doença de Newcastle/genética , Índice de Gravidade de Doença , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Proteínas do Envelope Viral/genética , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
11.
J Virol ; 88(21): 12348-63, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25122790

RESUMO

UNLABELLED: Avian metapneumovirus (aMPV), also known as avian pneumovirus or turkey rhinotracheitis virus, is the causative agent of turkey rhinotracheitis and is associated with swollen head syndrome in chickens. Since its discovery in the 1970s, aMPV has been recognized as an economically important pathogen in the poultry industry worldwide. The conserved region VI (CR VI) of the large (L) polymerase proteins of paramyxoviruses catalyzes methyltransferase (MTase) activities that typically methylate viral mRNAs at guanine N-7 (G-N-7) and ribose 2'-O positions. In this study, we generated a panel of recombinant aMPV (raMPV) Colorado strains carrying mutations in the S-adenosyl methionine (SAM) binding site in the CR VI of L protein. These recombinant viruses were specifically defective in ribose 2'-O, but not G-N-7 methylation and were genetically stable and highly attenuated in cell culture and viral replication in the upper and lower respiratory tracts of specific-pathogen-free (SPF) young turkeys. Importantly, turkeys vaccinated with these MTase-defective raMPVs triggered a high level of neutralizing antibody and were completely protected from challenge with homologous aMPV Colorado strain and heterologous aMPV Minnesota strain. Collectively, our results indicate (i) that aMPV lacking 2'-O methylation is highly attenuated in vitro and in vivo and (ii) that inhibition of mRNA cap MTase can serve as a novel target to rationally design live attenuated vaccines for aMPV and perhaps other paramyxoviruses. IMPORTANCE: Paramyxoviruses include many economically and agriculturally important viruses such as avian metapneumovirus (aMPV), and Newcastle disease virus (NDV), human pathogens such as human respiratory syncytial virus, human metapneumovirus, human parainfluenza virus type 3, and measles virus, and highly lethal emerging pathogens such as Nipah virus and Hendra virus. For many of them, there is no effective vaccine or antiviral drug. These viruses share common strategies for viral gene expression and replication. During transcription, paramyxoviruses produce capped, methylated, and polyadenylated mRNAs. Using aMPV as a model, we found that viral ribose 2'-O methyltransferase (MTase) is a novel approach to rationally attenuate the virus for vaccine purpose. Recombinant aMPV (raMPV) lacking 2'-O MTase were not only highly attenuated in turkeys but also provided complete protection against the challenge of homologous and heterologous aMPV strains. This novel approach can be applicable to other animal and human paramyxoviruses for rationally designing live attenuated vaccines.


Assuntos
Proteção Cruzada , Metapneumovirus/enzimologia , Metapneumovirus/imunologia , Infecções por Paramyxoviridae/veterinária , Doenças das Aves Domésticas/prevenção & controle , Doenças das Aves Domésticas/virologia , Vacinas Virais/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Pulmão/virologia , Metapneumovirus/genética , Metiltransferases/deficiência , Infecções por Paramyxoviridae/prevenção & controle , Infecções por Paramyxoviridae/virologia , Traqueia/virologia , Perus , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Carga Viral , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
12.
Biologicals ; 43(2): 136-45, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25511007

RESUMO

While there is typically 100% survivability in birds challenged with vNDV under experimental conditions, either with vaccines formulated with a strain homologous or heterologous (different genotype) to the challenge virus, vaccine deficiencies are often noted in the field. We have developed an improved and more stringent protocol to experimentally evaluate live NDV vaccines, and showed for the first time under experimental conditions that a statistically significant reduction in mortality can be detected with genotype matched vaccines. Using both vaccine evaluation protocols (traditional and improved), birds were challenged with a vNDV of genotype XIII and the efficacy of live heterologous (genotype II) and homologous (genotype XIII) NDV vaccines was compared. Under traditional vaccination conditions there were no differences in survival upon challenge, but the homologous vaccine induced significantly higher levels of antibodies specific to the challenge virus. With the more stringent challenge system (multiple vaccine doses and early challenge with high titers of vNDV), the birds administered the homologous vaccine had superior humoral responses, reduced clinical signs, and reduced mortality levels than those vaccinated with the heterologous vaccine. These results provide basis for the implementation of more sensitive methods to evaluate vaccine efficacy.


Assuntos
Doença de Newcastle/prevenção & controle , Vírus da Doença de Newcastle , Vacinas Virais , Animais , Galinhas , Avaliação de Medicamentos , Vírus da Doença de Newcastle/química , Vírus da Doença de Newcastle/imunologia , Vacinação/métodos , Vacinas Virais/química , Vacinas Virais/imunologia , Vacinas Virais/farmacologia
13.
Avian Dis ; 58(1): 83-9, 2014 03.
Artigo em Inglês | MEDLINE | ID: mdl-24758118

RESUMO

We developed a recombinant Newcastle disease virus (NDV) LaSota (rLS) expressing the infectious bronchitis virus (IBV) S2 gene (rLS/IBV.S2). The recombinant virus showed somewhat-reduced pathogenicity compared to the parental lentogenic LaSota strain but effectively elicited hemagglutination inhibition antibodies against NDV and protected chickens against lethal challenge with virulent NDV/CA02. IBV heterotypic protection was assessed using a prime-boost approach with a commercially available attenuated IBV Massachusetts (Mass)-type vaccine. Specific-pathogen-free chickens primed ocularly with rLS/IBV.S2 at 4 days of age and boosted with Mass at 18 days of age were completely protected against challenge at 41 days of age with a virulent Ark-type strain. In a second experiment, we compared protection conferred by priming with rLS/IBV.S2 and boosting with Mass (rLS/IBV.S2+Mass) versus priming and boosting with Mass (Mass+Mass). We also modified the timing of vaccination to prime at 1 day of age and boost at 12 days of age. Challenge with virulent Ark was performed at 21 days of age. Based on clinical signs, both vaccinated groups appeared equally protected against challenge compared to unvaccinated challenged chickens. Viral loads in lachrymal fluids of birds receiving rLS/IBV.S2+Mass showed a clear tendency of improved protection compared to Mass+Mass; however, the difference did not achieve statistical significance. A significant difference (P < 0.05) was determined between these groups regarding incidence of detection of challenge IBV RNA in the trachea; viral RNA was detected in 50% of rLS/IBV.S2+Mass-vaccinated chickens while chickens vaccinated with Mass+Mass and unvaccinated challenged controls showed 84 and 90% incidence of IBV RNA detection in the trachea, respectively. These results demonstrate that overexposing the IBV S2 to the chicken immune system by means of a vectored vaccine, followed by boost with whole virus, protects chickens against IBV showing dissimilar S1.


Assuntos
Galinhas , Infecções por Coronavirus/veterinária , Vírus da Bronquite Infecciosa/imunologia , Vírus da Doença de Newcastle/imunologia , Doenças das Aves Domésticas/prevenção & controle , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/sangue , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/prevenção & controle , Testes de Inibição da Hemaglutinação/veterinária , Vírus da Bronquite Infecciosa/patogenicidade , Vírus da Doença de Newcastle/genética , Reação em Cadeia da Polimerase/veterinária , Doenças das Aves Domésticas/imunologia , Organismos Livres de Patógenos Específicos , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/imunologia , Vacinas Atenuadas/imunologia , Carga Viral/veterinária
14.
Yao Xue Xue Bao ; 49(7): 985-92, 2014 Jul.
Artigo em Zh | MEDLINE | ID: mdl-25233628

RESUMO

To investigate the cell-killing effect and its possible mechanism of rClone30-hDR5 in combination with TRAIL on human hepatic carcinoma (HCC) cell line, first of all, recombinant plasmid pee12.4-hDR5 was introduced into HepG2 cells by liposome transfection. After five rounds of screening by flow cytometry, HepG2 cells expressing high levels of DR5 on cell surface were isolated. The cytotoxicity of TRAIL to selected cells was higher than that of TRAIL to HepG2 cells by MTT method (P < 0.01). The result suggested that the cloned hDR5 gene had biological activity. MTT assay showed that, rClone30- hDR5 in combination with TRAIL more efficiently inhibited the tumor growth of HepG2 cells compared to rClone30-hDR5 or TRAIL in vitro. The results of Annexin V-FITC/PI staining and Quantitative Real-time PCR indicated that rClone30-hDR5 in combination with TRAIL significantly increased the mRNA levels of caspase 3 and caspase 8, and induced the apoptosis of tumor cells. HepG2 cells were infected with rClone30-hDR5 or rClone30 at MOI of 1. The expression of hDR5 on tumor surface increased significantly by rClone30-hDR5 compared to that by rClone30, which contributed to the sensitivity to TRAIL. In conclusion, rClone30-hDR5 in combination with TRAIL has potential application value in cancer treatment.


Assuntos
Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/patologia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Apoptose , Caspase 3/metabolismo , Caspase 8/metabolismo , Sinergismo Farmacológico , Células Hep G2 , Humanos , Reação em Cadeia da Polimerase em Tempo Real , Transfecção
15.
J Gen Virol ; 94(Pt 6): 1189-1194, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23426356

RESUMO

The fusion (F) and haemagglutinin-neuraminidase (HN) proteins of Newcastle disease virus (NDV) are multifunctional proteins that play critical roles during infection. Here, we assessed the ability of NDV to replicate in macrophages and investigated the contribution of the F and HN proteins to NDV infection/replication in these cells. Results of our study revealed that, while presenting similar replication kinetics in a fibroblast cell line (DF1) or in primary non-adherent splenocytes, the NDV strain CA02 replicates better in macrophages (HD11 and primary adherent splenocytes) than the NDV strain Anhinga/93. Notably, exchange of the HN or both F and HN genes of NDV Anhinga/93 by the corresponding genes from NDV CA02 markedly improved the ability of the chimeric viruses to replicate in macrophages. These results indicate that the F and HN proteins are determinants of NDV macrophage host range. This represents the first description of productive NDV infection in macrophages.


Assuntos
Proteína HN/imunologia , Especificidade de Hospedeiro , Macrófagos/virologia , Doença de Newcastle/virologia , Vírus da Doença de Newcastle/fisiologia , Doenças das Aves Domésticas/virologia , Proteínas Virais de Fusão/imunologia , Animais , Células Cultivadas , Galinhas , Proteína HN/genética , Macrófagos/imunologia , Dados de Sequência Molecular , Doença de Newcastle/imunologia , Vírus da Doença de Newcastle/genética , Vírus da Doença de Newcastle/imunologia , Proteínas Virais de Fusão/genética , Replicação Viral
16.
Plasmid ; 70(3): 314-20, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23928042

RESUMO

Newcastle disease virus (NDV) can cause serious diseases and substantial economic losses to the poultry industry. To gain a better understanding of NDV pathogenesis, several reverse genetics systems for different NDV strains have been established. However, the construction of infectious cDNA clone by conventional restriction digestion/ligation cloning methods is a time-consuming process and has many drawbacks by its nature. To address the problems, we employed a novel and robust ligation-independent cloning (LIC) method for efficient assembly of multiple DNA fragments. Using this method, we successfully generated a NDV minigenome construct within three weeks by assembling RT-PCR products of the VG/GA strain genomic termini and a cDNA coding for the green fluorescence protein (GFP), as a reporter, into a modified pBluescript vector. Co-transfection of the NDV minigenome with three supporting plasmids expressing the N, P, and L proteins into MVA-T7 infected HEp-2 cells and followed by infection with NDV VG/GA resulted in the minigenome replication, transcription, and packaging as evidenced by the reporter gene GFP expression. These results suggest that this LIC approach is a powerful tool for all sequence-independent DNA cloning and multi-DNA fragment assembly, which has a potential application for rapid development of gene therapy and recombinant vaccines.


Assuntos
Cromossomos Artificiais , Clonagem Molecular/métodos , Genoma Viral , Vírus da Doença de Newcastle/genética , RNA Viral/genética , Linhagem Celular Tumoral , DNA Complementar/química , DNA Complementar/genética , Células Epiteliais/virologia , Genes Reporter , Engenharia Genética/métodos , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Vírus da Doença de Newcastle/química , Plasmídeos , RNA Viral/química , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
Virol J ; 10: 38, 2013 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-23363433

RESUMO

BACKGROUND: Avian metapneumovirus subtype C (aMPV/C) causes severe upper respiratory disease in turkeys. Previous report revealed the presence of aMPV/C in wild birds in the southeast regions of the U.S. METHODS: In this study, aMPV/C positive oral swabs from American coots (AC) and Canada geese (CG) were passaged three times in the respiratory tract of specific pathogen free (SPF) turkeys and used as aMPV/C P3 virus isolates in subsequent studies. RESULTS: Wild bird P3 isolates showed similar growth characteristics when compared to virulent aMPV/C in chicken embryo fibroblast ( CEF) cell cultures and their glycoprotein G gene sequence was closely related to the G gene of aMPV/C Colorado reference virus. Three-day-old commercial or SPF turkeys were inoculated oculonasally with wild bird aMPV/C P3 isolates. At 5 and 7 days post-inoculation (DPI), severe clinical signs were observed in both of the AC and CG virus-exposed groups. Viral RNA was detected in tracheal swabs by reverse transcriptase polymerase chain reaction (RT-PCR). In addition, immunohistochemistry showed virus replication in the nasal turbinate and trachea. All virus-exposed turkeys developed positive antibody response by 14 DPI. CONCLUSIONS: Our data demonstrate that aMPV/C wild bird isolates induced typical aMPV/C disease in the domestic turkeys.


Assuntos
Metapneumovirus/patogenicidade , Infecções por Paramyxoviridae/veterinária , Doenças das Aves Domésticas/virologia , Perus/virologia , Animais , Aves/virologia , Gansos/virologia , Genes Virais , Metapneumovirus/genética , Metapneumovirus/isolamento & purificação , Infecções por Paramyxoviridae/virologia , Sistema Respiratório/patologia , Sistema Respiratório/virologia , Cultura de Vírus
18.
Virus Genes ; 47(3): 498-504, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24037042

RESUMO

The hemagglutinin-neuraminidase (HN) of Newcastle disease virus (NDV) is a multifunctional protein that has receptor recognition, neuraminidase, and fusion promotion activities. Sequence analysis revealed that the HN gene of many extremely low virulence NDV strains encodes a larger open-reading frame (616 amino acids, aa) with additional 45 aa at its C-terminus when compared with that (571 aa) of virulent NDV strains. Therefore, it has been suspected that the 45 aa extension at the C-terminus of the HN may affect the NDV virulence. In this study, we generated an NDV mesogenic strain Anhinga-based recombinant virus with an HN C-terminal extension of 45 aa (rAnh-HN-ex virus) using reverse genetics technology. The biological characterization of the recombinant virus showed that the rAnh-HN-ex virus had similar growth ability to its parental virus rAnh-wt both in embryonating chicken eggs and DF-1 cells. However, the pathogenicity of this recombinant virus in embryonating chicken eggs and day-old chickens decreased, as evidenced by a longer mean death time and lower intracerebral pathogenicity index when compared with the parental virus. This is consistent with our previous finding that the recombinant LaSota virus with a 45-aa extension at its HN C-terminal was attenuated in chickens and embryonating eggs. These results suggest that the HN protein C-terminal extension may contribute to the reduced virulence in some low virulence NDV strains.


Assuntos
Proteína HN/química , Proteína HN/metabolismo , Doença de Newcastle/virologia , Vírus da Doença de Newcastle/metabolismo , Vírus da Doença de Newcastle/patogenicidade , Doenças das Aves Domésticas/virologia , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Sequência de Bases , Galinhas , Proteína HN/genética , Dados de Sequência Molecular , Vírus da Doença de Newcastle/química , Vírus da Doença de Newcastle/genética , Fases de Leitura Aberta , Virulência
19.
Virus Genes ; 47(1): 27-33, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23549884

RESUMO

The hemagglutinin-neuraminidase (HN) protein of Newcastle disease virus (NDV) plays an important role in virus pathogenicity and tissue tropism. Sequence analysis revealed that the HN gene of many asymptomatic enteric NDV strains encodes a larger open reading frame (616 amino acids, aa) with additional 39 aa at its C-terminus when compared with that (577 aa) of respirotropic NDV strains. Therefore, it has been suspected that the HN C-terminal extension may contribute to the enteric tropism. In the present study, we generated a NDV respirotropic strain LaSota-based recombinant virus with a HN C-terminal extension of 39 aa derived from an enterotropic NDV strain using reverse genetics technology. The biological characterization of the recombinant virus, rLS-HN-ex, showed that the HN C-terminal extension slightly attenuated the virus pathogenicity in embryonated eggs and in day-old chicks when compared to the parental LaSota virus. However, the HN C-terminal extension did not alter virus tissue tropism. In infected chickens, the recombinant virus was detected and re-isolated from the tracheal tissue, but not from the intestinal tissue, exhibiting a similar respirotropic tissue preference as its parental LaSota strain. These results demonstrated that the HN protein C-terminal extension of NDV is not the determinant of the virus enteric tropism.


Assuntos
Proteína HN/química , Proteína HN/metabolismo , Intestinos/virologia , Doença de Newcastle/virologia , Vírus da Doença de Newcastle/fisiologia , Doenças das Aves Domésticas/virologia , Tropismo Viral , Sequência de Aminoácidos , Animais , Galinhas , Proteína HN/genética , Dados de Sequência Molecular , Vírus da Doença de Newcastle/química , Vírus da Doença de Newcastle/genética , Especificidade de Órgãos , Estrutura Terciária de Proteína , Traqueia/virologia
20.
Vaccine ; 41(40): 5884-5891, 2023 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-37598026

RESUMO

Marek's disease (MD) is a highly contagious viral neoplastic disease of chickens caused by Marek's disease virus (MDV), resulting in significant economic losses to the poultry industry worldwide. The commonly used live and/or vectored MDV vaccines are expensive to produce and difficult to handle due to the requirement of liquid nitrogen for manufacturing and delivering frozen infected cells that are viable. In this study, we aimed to develop a Newcastle disease virus (NDV) vectored MDV vaccine that can be lyophilized, stored, and transported at 4 °C. Four NDV LaSota (LS) vaccine strain-based recombinant viruses expressing MDV glycoproteins gB, gC, gE, or gI were generated using reverse genetics technology. The biological assessments showed that these recombinant viruses were slightly attenuated in vivo yet retained similar growth kinetics and virus titers in vitro compared to the parental LaSota virus. Vaccination of leghorn chickens (Lines 15I5x71 F1 cross) with these recombinant viruses via intranasal and intraocular routes conferred different levels of protection against virulent MDV challenge. The recombinant expressing the MDV gB protein, rLS/MDV-gB, protected vaccinated birds significantly against MDV-induced tumor formation when challenged at 14 days post-vaccination (DPV) but moderately at 5 DPV. Whereas the other three recombinants provided little protection against the MDV challenge. All four recombinants conferred complete protection against the velogenic NDV challenge. These results demonstrated that the rLS/MDV-gB virus is a safe and efficacious dual vaccine candidate that can be lyophilized and potentially mass-administered via aerosol or drinking water to large chicken populations at a meager cost.


Assuntos
Herpesvirus Galináceo 2 , Doença de Marek , Animais , Galinhas , Vírus da Doença de Newcastle/genética , Doença de Marek/prevenção & controle , Herpesvirus Galináceo 2/genética , Comércio
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA