Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Virol ; 97(8): e0068123, 2023 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-37493545

RESUMO

Feline herpesvirus type 1 (FHV-1) is an enveloped dsDNA virus belonging to the Herpesviridae family and is considered one of the two primary viral etiological factors of feline upper respiratory tract disease. In this study, we investigated the entry of FHV-1 into host cells using two models: the AK-D cell line and primary feline skin fibroblasts (FSFs). We employed confocal microscopy, siRNA silencing, and selective inhibitors of various entry pathways. Our observations revealed that the virus enters cells via pH and dynamin-dependent endocytosis, as the infection was significantly inhibited by NH4Cl, bafilomycin A1, dynasore, and mitmab. Additionally, genistein, nystatin, and filipin treatments, siRNA knock-down of caveolin-1, as well as FHV-1 and caveolin-1 colocalization suggest the involvement of caveolin-mediated endocytosis during the entry process. siRNA knock-down of clathrin heavy chain and analysis of virus particle colocalization with clathrin indicated that clathrin-mediated endocytosis also takes part in the primary cells. This is the first study to systematically examine FHV-1 entry into host cells, and for the first time, we describe FHV-1 replication in AK-D and FSFs. IMPORTANCE Feline herpesvirus 1 (FHV-1) is one of the most prevalent viruses in cats, causing feline viral rhinotracheitis, which is responsible for over half of viral upper respiratory diseases in cats and can lead to ocular lesions resulting in loss of sight. Although the available vaccine reduces the severity of the disease, it does not prevent infection or limit virus shedding. Despite the clinical relevance, the entry mechanisms of FHV-1 have not been thoroughly studied. Considering the limitations of commonly used models based on immortalized cells, we sought to verify our findings using primary feline skin fibroblasts, the natural target for infection in cats.


Assuntos
Doenças do Gato , Endocitose , Infecções por Herpesviridae , Varicellovirus , Animais , Gatos , Doenças do Gato/virologia , Caveolina 1/metabolismo , Clatrina/metabolismo , Infecções por Herpesviridae/veterinária , RNA Interferente Pequeno/genética , Varicellovirus/metabolismo
2.
Molecules ; 24(4)2019 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-30795541

RESUMO

Guanine-rich sequences in the genomes of herpesviruses can fold into G-quadruplexes. Compared with the widely-studied G3-quadruplexes, the dynamic G2-quadruplexes are more sensitive to the cell microenvironment, but they attract less attention. Pseudorabies virus (PRV) is the model species for the study of the latency and reactivation of herpesvirus in the nervous system. A total of 1722 G2-PQSs and 205 G3-PQSs without overlap were identified in the PRV genome. Twelve G2-PQSs from the CDS region exhibited high conservation in the genomes of the Varicellovirus genus. Eleven G2-PQSs were 100% conserved in the repeated region of the annotated PRV genomes. There were 212 non-redundant G2-PQSs in the 3' UTR and 19 non-redundant G2-PQSs in the 5' UTR, which would mediate gene expression in the post-transcription and translation processes. The majority of examined G2-PQSs formed parallel structures and exhibited different sensitivities to cations and small molecules in vitro. Two G2-PQSs, respectively, from 3' UTR of UL5 (encoding helicase motif) and UL9 (encoding sequence-specific ori-binding protein) exhibited diverse regulatory activities with/without specific ligands in vivo. The G-quadruplex ligand, NMM, exhibited a potential for reducing the virulence of the PRV Ea strain. The systematic analysis of the distribution of G2-PQSs in the PRV genomes could guide further studies of the G-quadruplexes' functions in the life cycle of herpesviruses.


Assuntos
DNA Viral/química , Quadruplex G/efeitos dos fármacos , Regulação Viral da Expressão Gênica , Genoma Viral , Herpesvirus Suídeo 1/genética , Regiões 3' não Traduzidas/efeitos dos fármacos , Regiões 5' não Traduzidas/efeitos dos fármacos , Acridinas/química , Acridinas/farmacologia , Aminoquinolinas/química , Aminoquinolinas/farmacologia , Animais , Bovinos , Linhagem Celular , Biologia Computacional/métodos , DNA Helicases/genética , DNA Helicases/metabolismo , DNA Primase/genética , DNA Primase/metabolismo , DNA Viral/genética , DNA Viral/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/virologia , Células HEK293 , Herpesvirus Suídeo 1/efeitos dos fármacos , Herpesvirus Suídeo 1/metabolismo , Humanos , Ligantes , Mesoporfirinas/química , Mesoporfirinas/farmacologia , Ácidos Picolínicos/química , Ácidos Picolínicos/farmacologia , Regiões Promotoras Genéticas/efeitos dos fármacos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Suínos , Varicellovirus/efeitos dos fármacos , Varicellovirus/genética , Varicellovirus/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Ativação Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
3.
J Virol ; 92(7)2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29343566

RESUMO

Simian varicella virus (SVV), the primate counterpart of varicella-zoster virus, causes varicella (chickenpox), establishes latency in ganglia, and reactivates to produce zoster. We previously demonstrated that a recombinant SVV expressing enhanced green fluorescent protein (rSVV.eGFP) is slightly attenuated both in culture and in infected monkeys. Here, we generated two additional recombinant SVVs to visualize infected cells in vitro and in vivo One harbors eGFP fused to the N terminus of open reading frame 9 (ORF9) (rSVV.eGFP-2a-ORF9), and another harbors eGFP fused to the C terminus of ORF66 (rSVV.eGFP-ORF66). Both recombinant viruses efficiently expressed eGFP in cultured cells. Both recombinant SVV infections in culture were comparable to that of wild-type SVV (SVV.wt). Unlike SVV.wt, eGFP-tagged SVV did not replicate in rhesus cells in culture. Intratracheal (i.t.) or i.t. plus intravenous (i.v.) inoculation of rhesus macaques with these new eGFP-tagged viruses resulted in low viremia without varicella rash, although SVV DNA was abundant in bronchoalveolar lavage (BAL) fluid at 10 days postinoculation (dpi). SVV DNA was also found in trigeminal ganglia of one monkey inoculated with rSVV.eGFP-ORF66. Intriguingly, a humoral response to both SVV and eGFP was observed. In addition, monkeys inoculated with the eGFP-expressing viruses were protected from superinfection with SVV.wt, suggesting that the monkeys had mounted an efficient immune response. Together, our results show that eGFP expression could be responsible for their reduced pathogenesis.IMPORTANCE SVV infection in nonhuman primates has served as an extremely useful animal model to study varicella-zoster virus (VZV) pathogenesis. eGFP-tagged viruses are a great tool to investigate their pathogenesis. We constructed and tested two new recombinant SVVs with eGFP inserted into two different locations in the SVV genome. Both recombinant SVVs showed robust replication in culture but reduced viremia compared to that with SVV.wt during primary infection in rhesus macaques. Our results indicate that conclusions on eGFP-tagged viruses based on in vitro results should be handled with care, since eGFP expression could result in attenuation of the virus.


Assuntos
Regulação Viral da Expressão Gênica , Proteínas de Fluorescência Verde , Infecções por Herpesviridae , Doenças dos Macacos , Fases de Leitura Aberta , Varicellovirus , Animais , Linhagem Celular , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Infecções por Herpesviridae/genética , Infecções por Herpesviridae/metabolismo , Infecções por Herpesviridae/patologia , Infecções por Herpesviridae/veterinária , Macaca mulatta , Doenças dos Macacos/genética , Doenças dos Macacos/metabolismo , Doenças dos Macacos/patologia , Varicellovirus/genética , Varicellovirus/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo
4.
Antiviral Res ; 115: 17-20, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25542973

RESUMO

Feline herpes virus-1 (FHV-1) is ubiquitous in the cat population and is a major cause of blindness for which antiviral drugs, including acyclovir, are not completely effective. Recurrent infections, due to reactivation of latent FHV-1 residing in the trigeminal ganglia, can lead to epithelial keratitis and stromal keratitis and eventually loss of sight. This has prompted the medical need for an antiviral drug that will specifically inhibit FHV-1 infection. A new antiviral target is the DNA polymerase and its associated processivity factor, which forms a complex that is essential for extended DNA strand synthesis. In this study we have cloned and expressed the FHV-1 DNA polymerase (f-UL30) and processivity factor (f-UL42) and demonstrated that both proteins are required to completely synthesize the 7249 nucleotide full-length DNA from the M13 primed-DNA template in vitro. Significantly, a known inhibitor of human herpes simplex virus-1 (HSV-1) processivity complex was shown to inhibit FHV-1 processive DNA synthesis in vitro and block infection of cells. This validates using f-UL42/f-UL30 as a new antiviral drug target to treat feline ocular herpes infection.


Assuntos
Antivirais/farmacologia , DNA Viral/biossíntese , DNA Polimerase Dirigida por DNA/metabolismo , Exodesoxirribonucleases/metabolismo , Sulfonamidas/farmacologia , Tiadiazóis/farmacologia , Varicellovirus/efeitos dos fármacos , Varicellovirus/metabolismo , Proteínas Virais/metabolismo , Sequência de Aminoácidos , Animais , Gatos , Linhagem Celular , Clonagem Molecular , Replicação do DNA/efeitos dos fármacos , DNA Polimerase Dirigida por DNA/genética , Exodesoxirribonucleases/genética , Varicellovirus/genética , Proteínas Virais/genética
5.
Vet Microbiol ; 164(3-4): 222-8, 2013 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-23490557

RESUMO

Caprine herpesvirus 1 (CpHV1) is a member of ruminant alphaherpesviruses antigenically related to the prototype bovine herpesvirus 1 (BoHV1). Although cross reactivity between the two viruses involves many structural glycoproteins, the use of two competitive BoHV1 ELISAs detecting anti gB and gE antibodies has been proposed for CpHV1 infection, resulting mainly in a gB+/gE- reactivity and leading to suppose that CpHV1 gE may represent an useful target for the development of specific diagnostic test. Since CpHV1 gE gene has been only partially characterized so far, in this study the genome fragment of the short unique unit (Us) encompassing gI and gE gene was amplified and sequenced. Gene fragments encoding the ectodomain of both glycoproteins were subcloned into pSECTag2/Hygro and expressed in HEK293T cells as secreted form in serum free medium. Due to the lack of specific monoclonal antibodies (Mabs), the same recombinant glycoproteins were obtained from BoHV1 and used as positive control with a panel of specific gE and gI Mabs as well as in some ELISA assays. Results clearly indicate that the ectodomain of CpHV1 gE, immobilized on solid face in an indirect ELISA format, represents a sensitive and specific marker of infection, when compared with neutralization test, with absence of very low degree of cross-reactivity with BoHV1 gE counterpart, while the use of CpHV1 gI-ELISA or a combination of gE/gI complex did not significantly improve the sensitivity of the assay. In addition, in the rare event in which cross species barrier occurs for both viruses from their natural host to other species, the use of both BoHV1 and CpHV1 gE in a comparative assay may be proposed.


Assuntos
Ensaio de Imunoadsorção Enzimática , Varicellovirus/genética , Varicellovirus/metabolismo , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Animais , Anticorpos Monoclonais/metabolismo , Bovinos , Células HEK293 , Infecções por Herpesviridae/virologia , Herpesvirus Bovino 1/genética , Herpesvirus Bovino 1/metabolismo , Humanos , Estrutura Terciária de Proteína , Proteínas Recombinantes/genética , Sensibilidade e Especificidade
6.
Antiviral Res ; 97(3): 329-33, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23267833

RESUMO

Although vaccination is an important strategy for controlling H5N1 avian influenza virus infections, broilers (short-lived meat chickens) remain the major victims in disease-endemic countries. Inactivated vaccine usually requires 2-3weeks to establish solid protection, and recombinant vaccines, including the recombinant fowlpox virus vaccine and the recombinant Newcastle disease virus vaccine are affected by maternal antibodies against the vectors. These disadvantages compromise the protective efficacy of these vaccines in broilers. Here, we evaluated the safety and efficacy of a new recombinant duck enteritis virus that expresses the HA gene of an H5N1 virus (rDEV-re6) in specific-pathogen-free chickens and broilers. We found this new rDEV-re6 virus to be safe in chickens and to induce rapid and solid protection after a single dose. This virus may therefore serve as an ideal single-dose vaccine for broilers.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Virus da Influenza A Subtipo H5N1/imunologia , Vacinas contra Influenza/imunologia , Influenza Aviária/prevenção & controle , Doenças das Aves Domésticas/prevenção & controle , Varicellovirus/genética , Animais , Anticorpos Antivirais/imunologia , Galinhas , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Glicoproteínas de Hemaglutininação de Vírus da Influenza/administração & dosagem , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Virus da Influenza A Subtipo H5N1/genética , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/genética , Influenza Aviária/imunologia , Influenza Aviária/virologia , Doenças das Aves Domésticas/imunologia , Doenças das Aves Domésticas/virologia , Varicellovirus/metabolismo
7.
J Virol ; 87(5): 2979-82, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23269790

RESUMO

Ganglia of monkeys with reactivated simian varicella virus (SVV) contained more CD8 than CD4 T cells around neurons. The abundance of CD8 T cells was greater less than 2 months after reactivation than that at later times and correlated with that of CXCL10 RNA but not with those of SVV protein or open reading frame 61 (ORF61) antisense RNA. CXCL10 RNA colocalized with T-cell clusters. After SVV reactivation, transient T-cell infiltration, possibly mediated by CXCL10, parallels varicella zoster virus (VZV) reactivation in humans.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Quimiocina CXCL10/metabolismo , Gânglios/imunologia , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/virologia , Neurônios/imunologia , Varicellovirus/imunologia , Varicellovirus/metabolismo , Varicellovirus/fisiologia , Animais , Linfócitos T CD8-Positivos/metabolismo , DNA Viral/genética , Gânglios/metabolismo , Gânglios/virologia , Infecções por Herpesviridae/genética , Macaca fascicularis , Neurônios/virologia , Fases de Leitura Aberta , RNA Antissenso/biossíntese , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Varicellovirus/genética , Carga Viral , Ativação Viral , Latência Viral/genética
8.
Mol Immunol ; 48(15-16): 2038-51, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21764135

RESUMO

Viral infections are counteracted by virus-specific cytotoxic T cells that recognize the infected cell via MHC class I (MHC I) molecules presenting virus-derived peptides. The loading of the peptides onto MHC I molecules occurs in the endoplasmic reticulum (ER) and is facilitated by the peptide loading complex. A key player in this complex is the transporter associated with antigen processing (TAP), which translocates the viral peptides from the cytosol into the ER. Herpesviruses have developed many strategies to evade cytotoxic T cells. Several members of the genus Varicellovirus encode a UL49.5 protein that prevents peptide transport through TAP. These include bovine herpesvirus (BoHV) 1, BoHV-5, bubaline herpesvirus 1, cervid herpesvirus 1, pseudorabies virus, felid herpesvirus 1, and equine herpesvirus 1 and 4. BoHV-1 UL49.5 inhibits TAP by preventing conformational changes essential for peptide transport and by inducing degradation of the TAP complex. UL49.5 consists of an ER luminal N-terminal domain, a transmembrane domain and a cytosolic C-terminal tail domain. In this study, the following features of UL49.5 were deciphered: (1) chimeric constructs of BoHV-1 and VZV UL49.5 attribute the lack of TAP inhibition by VZV UL49.5 to its ER-luminal domain, (2) the ER-luminal and TM domains of UL49.5 are required for efficient interaction with and inhibition of TAP, (3) the C-terminal RXRX sequence is essential for TAP degradation by BoHV-1 UL49.5, and (4) in addition to the RXRX sequence, the cytoplasmic tail of BoHV-1 UL49.5 carries a motif that is required for efficient TAP inhibition by the protein. A model is presented depicting how the different domains of UL49.5 may block the translocation of peptides by TAP and target TAP for proteasomal degradation.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Varicellovirus/química , Varicellovirus/metabolismo , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/imunologia , Transportadores de Cassetes de Ligação de ATP/imunologia , Linhagem Celular , Separação Celular , Citometria de Fluxo , Humanos , Immunoblotting , Imunoprecipitação , Reação em Cadeia da Polimerase , Estrutura Terciária de Proteína , Transporte Proteico/fisiologia , Varicellovirus/imunologia , Proteínas do Envelope Viral/metabolismo
9.
J Virol ; 81(15): 8149-56, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17507490

RESUMO

Simian varicella virus (SVV) and varicella-zoster virus (VZV) are closely related alphaherpesviruses that cause varicella (chickenpox) in nonhuman primates and humans, respectively. After resolution of the primary disease, SVV and VZV establish latent infection of neural ganglia and may later reactivate to cause a secondary disease (herpes zoster). This study investigated SVV gene expression in neural ganglia derived from latently infected vervet monkeys. SVV transcripts were detected in neural ganglia, but not in liver or lung tissues, of latently infected animals. A transcript mapping to open reading frame (ORF) 61 (herpes simplex virus type 1 [HSV-1] ICP0 homolog) was consistently detected in latently infected trigeminal, cervical, and lumbar ganglia by reverse transcriptase PCR. Further analysis confirmed that this SVV latency-associated transcript (LAT) was oriented antisense to the gene 61 mRNA. SVV ORF 21 transcripts were also detected in 42% of neural ganglia during latency. In contrast, SVV ORF 28, 29, 31, 62, and 63 transcripts were not detected in ganglia, liver, or lung tissues of latently infected animals. The results demonstrate that viral gene expression is limited during SVV latency and that a LAT antisense to an ICP0 homolog is expressed. In this regard, SVV gene expression during latency is similar to that of HSV-1 and other neurotropic animal alphaherpesviruses but differs from that reported for VZV.


Assuntos
Varicela , Gânglios/virologia , Herpesvirus Humano 3 , Fases de Leitura Aberta , Varicellovirus , Latência Viral , Animais , Chlorocebus aethiops , Herpesvirus Humano 3/genética , Herpesvirus Humano 3/metabolismo , Humanos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Varicellovirus/genética , Varicellovirus/metabolismo
10.
Microbes Infect ; 8(11): 2657-67, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16962359

RESUMO

Glycoprotein G (gG) orthologues have been described in several alphaherpesviruses. gG is expressed both as a membrane-anchored form on infected cells and as a secreted form. Recently, we reported that both forms of gG encoded by alphaherpesviruses infecting large herbivores and by Felid herpesvirus 1 (FeHV-1) bind with high affinity to a broad range of CXC, CC and C-chemokines. Based on the viral species, gG has been reported either as a structural or a non-structural protein. To date, the incorporation of FeHV-1 gG into virions has never been tested, nor the property of alphaherpesvirus structural gG to bind chemokines on the virion surface. In the present study, to address these questions, various FeHV-1 gG recombinant strains were produced using an original technique based on an infectious FeHV-1 BAC clone and restriction endonuclease mediated recombination. Using the recombinants produced, we were able to determine that FeHV-1 gG is a structural protein that acts as a chemokine-binding protein on the virion surface. In the light of these results, putative roles of gG in alphaherpesvirus infections are discussed, and an evolutionary scenario is proposed to explain the structural versus non-structural property of gG amongst alphaherpesviruses.


Assuntos
Quimiocinas/metabolismo , Glicoproteínas/metabolismo , Varicellovirus/metabolismo , Proteínas do Envelope Viral/metabolismo , Proteínas Estruturais Virais/metabolismo , Animais , Gatos , Linhagem Celular , Cromossomos Artificiais Bacterianos/genética , Genoma Viral , Microscopia Imunoeletrônica , Ligação Proteica , Recombinação Genética , Varicellovirus/genética , Vírion/ultraestrutura
11.
J Gen Virol ; 87(Pt 6): 1501-1508, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16690914

RESUMO

Simian varicella virus (SVV) is a neurotropic alphaherpesvirus that causes a natural, varicella-like disease in non-human primates. After resolution of the primary disease, SVV, like its human counterpart, varicella-zoster virus (VZV), establishes latent infection in the neural ganglia of the host. In this study, gene expression of SVV open reading frames (ORFs) 28 and 29, which encode the viral DNA polymerase and DNA-binding protein, respectively, was characterized during lytic infection of Vero cells. The results indicate that the intergenic region controlling gene 28 and 29 expression includes overlapping, divergent promoters. The ORF 28 and 29 promoters are active in SVV-infected Vero cells, but not in uninfected cells. The SVV immediate-early gene 62 (IE62) product transactivates ORF 28 and 29 expression, and a cellular upstream stimulatory factor-binding site is important for efficient IE62 induction of genes 28 and 29. DNA sequence analysis of the 185 bp intergenic region identified putative cellular transcription factor-binding sites. Transcriptional analysis mapped ORF 28 and 29 RNA start sites. A recombinant SVV was employed to demonstrate that the ORF 29 promoter can express a heterologous gene (green fluorescent protein) when inserted into a novel site (the ORF 12/13 intergenic region) within the SVV genome. The findings demonstrate similarities between SVV and VZV ORF 28/29 expression and indicate that the simian varicella model may be useful to investigate the differential regulation of viral genes during lytic and latent infection.


Assuntos
Chlorocebus aethiops/virologia , Proteínas de Ligação a DNA/metabolismo , DNA Polimerase Dirigida por DNA/metabolismo , Fases de Leitura Aberta/genética , Regiões Promotoras Genéticas/genética , Varicellovirus/patogenicidade , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , DNA Viral , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , DNA Polimerase Dirigida por DNA/química , DNA Polimerase Dirigida por DNA/genética , Modelos Animais de Doenças , Regulação Viral da Expressão Gênica , Humanos , Proteínas Imediatamente Precoces/metabolismo , Dados de Sequência Molecular , Transativadores/metabolismo , Ativação Transcricional , Fatores Estimuladores Upstream/metabolismo , Varicellovirus/genética , Varicellovirus/metabolismo , Proteínas do Envelope Viral/metabolismo , Proteínas Virais/química , Proteínas Virais/genética , Proteínas Virais/metabolismo
13.
Virus Res ; 70(1-2): 13-23, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11074121

RESUMO

We constructed two recombinant feline herpesvirus type 1 (FHV-1) expressing the envelope (Env) protein of feline immunodeficiency virus (FIV). One recombinant, designated dlTK-env, has the whole FIV env gene inserted at a thymidine kinase (TK) deletion site. The second recombinant, designated dlTK(gCp)-env, has a cassette containing a partial FIV env gene fused with the signal sequence of the gC protein of FHV-1 (under the control of the gC promoter) inserted at the same site. Growth kinetics of both the recombinants in Crandell feline kidney (CRFK) cells were similar to that of the parent strain of FHV-1. By indirect immunofluorescence assays and immunoblot analyses, we confirmed the expression of the FIV Env protein in CRFK cells infected with both recombinants. Enzyme-linked immunosorbent assays showed that the maximum Env expression level achieved by dlTK(gCp)-env was more than four times higher than that observed for dlTK-env. Flow cytometric analyses revealed that the Env protein produced by both recombinants was efficiently expressed on the cell surface. The dlTK(gCp)-env reported here may thus be a promising candidate for a live recombinant vaccine to protect against FIV infection.


Assuntos
Genes Virais/genética , Vírus da Imunodeficiência Felina/genética , Regiões Promotoras Genéticas , Varicellovirus/genética , Proteínas do Envelope Viral/genética , Animais , Antígenos Virais/genética , Antígenos Virais/metabolismo , Southern Blotting , Gatos , Células Cultivadas , Vetores Genéticos , Immunoblotting , Vírus da Imunodeficiência Felina/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Varicellovirus/metabolismo , Proteínas do Envelope Viral/metabolismo
14.
Arch Virol ; 145(7): 1489-96, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10963353

RESUMO

In order to identify the products of the equine herpesvirus type 4 (EHV-4) gI and gE genes, we have constructed recombinant vaccinia viruses containing the putative gI or gE genes. These recombinant viruses synthesized EHV-4 gI and gE with apparent molecular masses of 75 and 80kDa, respectively. Antibodies raised against both recombinant viruses detected a 75 kDa gI and a 95 kDa gE in EHV-4-infected cells. The results also suggest that the EHV-4 gI and gE would form a complex like in other herpesviruses.


Assuntos
Genes Virais , Varicellovirus/genética , Proteínas do Envelope Viral/genética , Animais , Imunofluorescência , Regulação Viral da Expressão Gênica , Varicellovirus/metabolismo , Proteínas do Envelope Viral/biossíntese
15.
J Gen Virol ; 75 ( Pt 11): 3219-27, 1994 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7964632

RESUMO

The varicella-zoster virus (VZV) glycoprotein B (gB) is a major viral antigen which elicits immunity and neutralizing antibodies. In this study, the genomic map position and DNA sequence of a simian varicella virus (SVV) homologue of the VZV gB gene was identified and the transcript analysed. A 32P-labelled VZV gB DNA probe hybridized to a subclone of the SVV BamHI B restriction endonuclease fragment indicating the fine map position of SVV DNA sequences homologous to the VZV gB gene. The SVV gB DNA sequence was determined and analysis revealed a 2751 base pair open reading frame (ORF) with 71.1% identity to the VZV gB gene and 53.8% identity to the herpes simplex type 1 gB gene. The SVV gB ORF encodes a 916 amino acid polypeptide with a predicted molecular mass of 104K. The deduced SVV and VZV gB polypeptides share 78.9% amino acid identity and predicted N-linked glycosylation sites, cleavage sites and transmembrane regions. 32P-labelled SVV gB DNA and RNA probes hybridized to a 3.5 kilobase SVV polyadenylated transcript. Primer extension experiments identified transcript start sites for the SVV and VZV gB genes and permitted a comparison of the sequences upstream of the SVV and VZV gB ORFs. The SVV and VZV gB promoter elements are similar in content and align closely. The VZV gB transcript start site suggests a gB polypeptide initiation site which is inconsistent with the previously reported ATG start codon.


Assuntos
DNA Viral/química , DNA Viral/metabolismo , Genes Virais , Regiões Promotoras Genéticas , Transcrição Gênica , Varicellovirus/genética , Proteínas do Envelope Viral/biossíntese , Sequência de Aminoácidos , Sequência de Bases , Northern Blotting , Primers do DNA , Sondas de DNA , Herpesvirus Humano 3/metabolismo , Dados de Sequência Molecular , RNA Mensageiro/biossíntese , Mapeamento por Restrição , Homologia de Sequência de Aminoácidos , Homologia de Sequência do Ácido Nucleico , Varicellovirus/metabolismo , Proteínas do Envelope Viral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...