Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
J Virol ; 93(17)2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31189709

RESUMO

Hypoxia and hypoxia inducible factors (HIFs) play important roles in the Kaposi's sarcoma-associated herpesvirus (KSHV) life cycle. KSHV is the causative agent of Kaposi's sarcoma (KS) and other AIDS-related malignancies. Kaposi's sarcoma is a highly vascular tumor, which preferentially develops in the lower extremities of the body where blood vessels are often poorly oxygenated. The main cellular responses to hypoxia are mediated mainly by two isoforms of HIF, HIF-1α and HIF-2α. HIF-1α and HIF-2α have common as well as distinct functions, although they are similar in structure and function. Previously, we showed that the KSHV ORF34 protein binds HIF-1α and facilitates its degradation through the ubiquitin-proteasome pathway causing negative regulation of HIF-1α-dependent genes (Haque and Kousoulas, J Virol 87:2164-2173, 2013, https://www.doi.org/10.1128/JVI.02460-12). Herein, we show that the ORF34 gene is involved in the regulation of KSHV lytic gene expression, since deletion of ORF34 resulted in reduced immediate early and early lytic gene expression and blocked late gene expression. Coimmunoprecipitation experiments revealed that the ORF34 protein physically interacted with HIF-2α in transfected as well as in KSHV-infected cells. Utilization of ORF34 truncations revealed that three distinct domains bind HIF-2α and that both bHLH and PAS domains of HIF-2α interacted with ORF34. Unlike HIF-1α, dose-dependent coexpression of ORF34 stabilized the HIF-2α protein, ensuring HIF-2α-dependent transcriptional activity. The ORF34 protein enhanced HIF-2α ubiquitination at the bHLH and PAS domains. The results show that the KSHV ORF34 protein is involved in the KSHV life cycle by regulating the expression of HIF-1α and HIF-2α proteins.IMPORTANCE Hypoxia inducible factor 1α (HIF-1α) and HIF-2α are transcription factors which play important roles in the Kaposi's sarcoma-associated herpesvirus (KSHV) latent and lytic gene replication. Herein, we show that the ORF34 gene is involved in the regulation of KSHV lytic gene expression, since deletion of ORF34 resulted in reduced immediate early and early lytic gene expression and blocked late gene expression. In addition, we demonstrate that the KSHV ORF34 protein binds and stabilizes HIF-2α, in contrast to its role in binding HIF-1α and causing its degradation via the proteasome pathway. Thus, the KSHV ORF34 protein plays a regulatory role in the KSHV life cycle by regulating HIF-1α and HIF-2α expression.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/química , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , Herpesvirus Humano 8/fisiologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Sítios de Ligação , Proteínas do Capsídeo/genética , Deleção de Genes , Regulação Viral da Expressão Gênica , Células HEK293 , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Estágios do Ciclo de Vida , Domínios Proteicos , Estabilidade Proteica , Transcrição Gênica , Ubiquitinação
2.
J Virol ; 92(6)2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29321326

RESUMO

Previously, we have shown that the amino terminus of glycoprotein K (gK) binds to the amino terminus of gB and that deletion of the amino-terminal 38 amino acids of gK prevents herpes simplex virus 1 (HSV-1) infection of mouse trigeminal ganglia after ocular infection and virus entry into neuronal axons. Recently, it has been shown that gB binds to Akt during virus entry and induces Akt phosphorylation and intracellular calcium release. Proximity ligation and two-way immunoprecipitation assays using monoclonal antibodies against gB and Akt-1 phosphorylated at S473 [Akt-1(S473)] confirmed that HSV-1(McKrae) gB interacted with Akt-1(S473) during virus entry into human neuroblastoma (SK-N-SH) cells and induced the release of intracellular calcium. In contrast, the gB specified by HSV-1(McKrae) gKΔ31-68, lacking the amino-terminal 38 amino acids of gK, failed to interact with Akt-1(S473) and induce intracellular calcium release. The Akt inhibitor miltefosine inhibited the entry of McKrae but not the gKΔ31-68 mutant into SK-N-SH cells. Importantly, the entry of the gKΔ31-68 mutant but not McKrae into SK-N-SH cells treated with the endocytosis inhibitors pitstop-2 and dynasore hydrate was significantly inhibited, indicating that McKrae gKΔ31-68 entered via endocytosis. These results suggest that the amino terminus of gK functions to regulate the fusion of the viral envelope with cellular plasma membranes.IMPORTANCE HSV-1 glycoprotein B (gB) functions in the fusion of the viral envelope with cellular membranes during virus entry. Herein, we show that a deletion in the amino terminus of glycoprotein K (gK) inhibits gB binding to Akt-1(S473), the release of intracellular calcium, and virus entry via fusion of the viral envelope with cellular plasma membranes.


Assuntos
Sinalização do Cálcio , Cálcio/metabolismo , Membrana Celular/metabolismo , Herpes Simples/metabolismo , Herpesvirus Humano 1/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas do Envelope Viral/metabolismo , Proteínas Virais/metabolismo , Internalização do Vírus , Animais , Linhagem Celular Tumoral , Membrana Celular/genética , Chlorocebus aethiops , Herpes Simples/genética , Herpes Simples/patologia , Herpesvirus Humano 1/genética , Humanos , Domínios Proteicos , Proteínas Proto-Oncogênicas c-akt/genética , Células Vero , Proteínas do Envelope Viral/genética , Proteínas Virais/genética
3.
Curr Eye Res ; 39(6): 596-603, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24401006

RESUMO

PURPOSE: To determine the role of the amino terminus of herpes simplex virus-1 (HSV-1) glycoprotein K (gK) in corneal infection, neuroinvasion, and establishment of virus latency in trigeminal ganglia of mice. METHODS: The recombinant virus HSV-1 (McKΔgK31-68) was constructed by engineering gK genes encoding gK lacking 38 amino acids immediately after the gK signal sequence. A rescued virus was also produced. Mouse eyes were scarified and infected with 10(5) plaque forming units (PFU) in each eye. Clinical signs of ocular disease were monitored daily. Thirty days postinfection trigeminal ganglia were collected and processed for quantitative PCR (qPCR) analysis of viral DNA and recovery of infectious virions by cell culture of ganglionic tissues. RESULTS: Deletion of the amino terminus of gK encoded by the McKΔgK31-68 mutant virus did not substantially affect its replication kinetics on African green monkey kidney cells (Vero), while it reduced cell-to-cell spread. McK viral infection of scarified mouse corneas with 10(5) PFU produced severe ocular disease. In contrast, McKΔgK31-68 viral infection with 10(5) PFU produced no significant ocular disease symptoms. All ganglia from mice infected with the McK virus produced high numbers of infectious virions upon explant culture in Vero cells, in agreement with qPCR results detecting high number of HSV-1 viral DNA in ganglionic tissues. In contrast, qPCR failed to detect any viral genomes in McKΔgK31-68 ganglia, while two of the ten ganglia revealed the presence of low numbers of infectious virions upon explant culture in Vero cells. CONCLUSIONS: The results show that the amino terminus of gK is essential for neuroinvasiveness and acute herpes keratitis in the mouse eye model. It is likely that gK is involved in efficient infection of axonal termini, since mouse eye scarification provided a direct access to the high density of neuronal axons innervating mouse corneas.


Assuntos
Córnea/inervação , Herpesvirus Humano 1/fisiologia , Ceratite Herpética/prevenção & controle , Mutação , Gânglio Trigeminal/virologia , Proteínas Virais/genética , Replicação Viral/fisiologia , Animais , Chlorocebus aethiops , DNA Viral/genética , Modelos Animais de Doenças , Feminino , Ceratite Herpética/virologia , Camundongos , Camundongos Endogâmicos BALB C , Reação em Cadeia da Polimerase , Células Vero , Ensaio de Placa Viral , Vírion/isolamento & purificação
4.
J Virol ; 84(4): 1704-14, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19955303

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) viral glycoproteins play important roles in the infectious life cycle and have been implicated in KSHV-associated endothelial cell transformation, angiogenesis, and KS-induced malignancies. KSHV-associated primary effusion lymphomas (PELs) secrete high levels of vascular endothelial growth factor (VEGF) and viral interleukin-6 (vIL-6) in vitro and VEGF, vIL-6, and basic-fibroblast growth factor (b-FGF) in mouse xenografts. KSHV-encoded glycoproteins B (gB) and K8.1 stimulate VEGF secretion, most likely mediated by direct or indirect binding to cell surface receptors, including the gB-specific alphaVbeta3 and alpha3beta1 integrins. In this study, the short interfering RNA (siRNA)-mediated inhibition of either gB or K8.1 transcription by anti-gB or -K8.1 siRNAs caused a substantial reduction in virion egress and a decrease in both vIL-6 and VEGF production. Similarly, the treatment of BCBL-1 cells with anti-gB or anti-K8.1 antibodies caused a substantial reduction in vIL-6 and VEGF production. Codon-optimized versions of either wild-type gB, mutant gB having the RGD amino acid motif changed to RAA, or K8.1 efficiently rescued virion egress and VEGF and vIL-6 production. These results suggest that the binding of gB via its RGD motif to integrin receptors was not responsible for the observed gB-associated regulation of VEGF and vIL-6 transcription. Conditioned medium collected from BCBL-1 cells transfected with anti-gB and anti-K8.1 siRNAs or treated with anti-gB and anti-K8.1 antibodies exhibited a significantly reduced ability to induce the formation of the capillary network of endothelial cells compared to the ability of medium from mock-infected BCBl-1 cells. Furthermore, medium obtained from BCBL-1 cells expressing smaller amounts of gB and K8.1 produced a substantial reduction in endothelial cell migration in a vertical migration assay compared to that of control medium containing wild-type levels of gB and K8.1. These results suggest a functional linkage between gB/K8.1 synthesis and VEGF/vIL-6 transcriptional regulation via paracrine and/or autocrine signaling pathways.


Assuntos
Glicoproteínas/fisiologia , Herpesvirus Humano 8/fisiologia , Herpesvirus Humano 8/patogenicidade , Interleucina-6/biossíntese , Fator A de Crescimento do Endotélio Vascular/biossíntese , Proteínas do Envelope Viral/fisiologia , Proteínas Virais/fisiologia , Substituição de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular Tumoral , Células Cultivadas , Primers do DNA/genética , Células Endoteliais/virologia , Genes Virais , Glicoproteínas/antagonistas & inibidores , Glicoproteínas/genética , Herpesvirus Humano 8/genética , Humanos , Interleucina-6/genética , Camundongos , Mutagênese Sítio-Dirigida , Neovascularização Patológica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfecção , Fator A de Crescimento do Endotélio Vascular/genética , Proteínas do Envelope Viral/antagonistas & inibidores , Proteínas do Envelope Viral/genética , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/genética
5.
J Virol ; 83(12): 6115-24, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19357164

RESUMO

Herpes simplex virus type 1 (HSV-1) acquires its final envelope by budding into cytoplasmic vesicles thought to be derived from trans-Golgi network membranes. This process is facilitated by interactions among the carboxyl termini of viral glycoproteins and tegument proteins. To directly investigate the relative importance of the carboxyl terminus of glycoprotein D (gD) in the presence or absence of gE, a recombinant virus (gDDeltact) was constructed to specify a truncated gD lacking the carboxy-terminal 29 amino acids. Furthermore, two additional recombinant viruses were constructed by mutating from ATG to CTG the initiation codons of gE (gEctg) or both gE and gM (gEctg+gMctg), causing lack of expression of gE or both gE and gM, respectively. A fourth mutant virus was constructed to specify the gEctg+gDDeltact mutations. The replication properties of these viruses were compared to those of a newly constructed recombinant virus unable to express UL20 due to alteration of the two initiation codons of UL20 (UL20ctgctg). All recombinant viruses were constructed by using the double-Red, site-directed mutagenesis system implemented on the HSV-1(F) genome cloned into a bacterial artificial chromosome. The gEctg, gEctg+gMctg, gDDeltact, and gEctg+gDDeltact viruses produced viral plaques on African monkey kidney cells (Vero), as well as other cells, that were on average approximately 30 to 50% smaller than those produced by the wild-type virus HSV-1(F). In contrast, the UL20ctgctg virus produced very small plaques containing three to five cells, as reported previously for the DeltaUL20 virus lacking the entire UL20 gene. Viral replication kinetics of intracellular and extracellular viruses revealed that all recombinant viruses produced viral titers similar to those produced by the wild-type HSV-1(F) virus intracellularly and extracellularly at late times postinfection, with the exception of the UL20ctgctg and DeltaUL20 viruses, which replicated more than two-and-a-half logs less efficiently than HSV-1(F). Electron microscopy confirmed that all viruses, regardless of their different gene mutations, efficiently produced enveloped virions within infected cells, with the exception of the UL20ctgctg and DeltaUL20 viruses, which accumulated high levels of unenveloped virions in the cytoplasm. These results show that the carboxyl terminus of gD and the full-length gE, either alone or in a redundant manner, are not essential in cytoplasmic virion envelopment and egress from infected cells. Similarly, gM and gE do not function alone or in a redundant manner in cytoplasmic envelopment and virion egress, confirming previous findings.


Assuntos
Herpesvirus Humano 1/fisiologia , Proteínas do Envelope Viral/genética , Montagem de Vírus , Animais , Chlorocebus aethiops , Citoplasma/virologia , DNA Viral/genética , Perfilação da Expressão Gênica , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/crescimento & desenvolvimento , Herpesvirus Humano 1/ultraestrutura , Humanos , Mutagênese Sítio-Dirigida , Mutação , Células Vero , Ensaio de Placa Viral , Proteínas Virais/genética
6.
Curr Eye Res ; 33(5): 455-67, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18568883

RESUMO

PURPOSE: To determine the role of herpes simplex virus-1 (HSV-1) glycoprotein K(gK) in corneal infection, neuroinvasion, and virus latency in trigeminal ganglia of mice. METHODS: The recombinant virus HSV-1 (McKrae) Delta gK (MKDelta gK) carrying a deletion of the gK gene was constructed by insertional/deletion mutagenesis and replaced by a gene cassette constitutively expressing the enhanced green fluorescence protein. The gK deletion of the MKDelta gK virus was rescued to produce the wild-type-like virus MKgK. Balb/c mice were infected ocularly with either virus, and the infection pattern in the eye, clinical disease progression, and establishment of viral latency was monitored. RESULTS: Mice infected with the MKDelta gK strain produced in a gK complementing cell line did not exhibit clinical signs when compared with mice infected with the MKgK virus. Direct visualization of infected eyes revealed that the MKDelta gK virus was unable to spread in mouse corneas, while the MKgK rescued virus spread efficiently. Nineteen of 20 scarified and 5/12 unscarified mice infected with the MKgK virus produced infectious virus after coculture with permissive cells, while 0/20 scarified and 0/12 unscarified mice infected with the MKDelta gK virus produced infectious virus. HSV DNA was detected in trigeminal ganglia by PCR in 19/20 scarified and 9/12 unscarified mice inoculated with MKgK, while HSV DNA was detected in the trigeminal ganglia of 3/20 scarified and 0/12 unscarified mice inoculated with MKDelta gK. CONCLUSIONS: The results show that HSV-1 gK is essential for efficient replication and spread in the corneal epithelium and trigeminal ganglia neuroinvasion in MKDelta gK inoculated mice.


Assuntos
Córnea/inervação , Herpesvirus Humano 1/patogenicidade , Ceratite Herpética/virologia , Gânglio Trigeminal/virologia , Proteínas Virais/fisiologia , Animais , Técnicas de Cocultura , Córnea/patologia , DNA Viral/análise , Feminino , Herpesvirus Humano 1/fisiologia , Ceratite Herpética/patologia , Camundongos , Camundongos Endogâmicos BALB C , Reação em Cadeia da Polimerase , Proteínas Recombinantes de Fusão/fisiologia , Latência Viral , Replicação Viral/fisiologia
7.
J Virol ; 82(14): 7144-54, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18480449

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV)-encoded glycoprotein B (gB) is an important determinant of viral infectivity and virion egress. A small interfering RNA (siRNA)-based strategy was devised to inhibit KSHV gB gene expression. Transient cotransfection of plasmids constitutively expressing gB and anti-gB siRNAs in 293 cells substantially inhibited gB mRNA levels and protein production. Similarly, transient expression of siRNAs into the primary effusion lymphoma cell line BCBL-1 caused a substantial reduction of gB transcripts and protein synthesis. TaqMan real-time PCR assays against the lytic KSHV gene ORF59 and infectivity assays on 293 cells were employed to assess the effect of inhibiting gB synthesis on virion egress from BCBL-1 cells and infectivity on 293 cells, respectively. These experiments showed that gB was essential for virion egress and infectivity. Transfection of a codon-optimized gB gene with the first 540 nucleotides altered, and therefore not recognized by anti-gB siRNAs that target the native but not the codon-optimized sequence, efficiently rescued virion egress and infectivity in BCBL-1 cells in the presence of siRNAs inhibiting wild-type gB expression. To assess the role of the cytoplasmic domain of gB in virion egress, mutant gB genes were generated specifying carboxyl terminal truncations of 25 and 58 amino acids disrupting two prominent predicted alpha-helical domains associated with virus-induced cell fusion. A third truncation removed the entire predicted cytoplasmic terminus of 84 amino acids, while a fourth truncation removed 110 amino acids, including the terminal most hydrophobic, intramembrane anchoring sequence. Virion egress experiments revealed that all truncated gBs facilitated virion egress from BCBL-1 cells, with the exception of the largest 110-amino-acid truncation, which removed the gB anchoring sequence. Importantly, the gB truncation that removed the entire predicted cytoplasmic domain increased virion egress, suggesting the presence of a egress regulation domain located proximal to the intramembrane sequence within the cytoplasmic domain of gB. All supernatant virions were infectious on 293 cells, indicating that the carboxyl terminus of gB is not essential for either virion egress or virus infectivity.


Assuntos
Herpesvirus Humano 8/fisiologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Linhagem Celular , Inativação Gênica , Humanos , Modelos Moleculares , Estrutura Terciária de Proteína , RNA Interferente Pequeno/genética , Deleção de Sequência
8.
J Virol ; 82(13): 6310-23, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18434401

RESUMO

Herpes simplex virus type 1 glycoprotein K (gK) and the UL20 protein (UL20p) are coordinately transported to the trans-Golgi network (TGN) and cell surfaces and are required for cytoplasmic virion envelopment at the TGN. In addition, cell surface expression of gK and UL20p is required for virus-induced cell fusion. Previously, confocal microscopy colocalization and intracellular transport experiments strongly suggested direct protein-protein interactions between gK and UL20p. Direct protein-protein interactions between gK and UL20p were demonstrated through reciprocal coimmunoprecipitation experiments, as well as with glutathione S-transferase (GST) pull-down experiments. A fusion protein consisting of the amino-terminal 66 amino acids of UL20p fused in-frame with GST was expressed in Escherichia coli and purified via glutathione column chromatography. Precipitation of GST-UL20p from mixtures of GST-UL20p fusion protein with cellular extracts containing gK specifically coprecipitated gK but not other viral glycoproteins. The purified UL20p-GST fusion protein reacted with all gK-associated protein species. It was concluded that the amino terminus of UL20p, most likely, interacted with gK domain III, which is predicted to lie intracellularly. UL20p-gK domain-specific interactions must serve important functions in the coordinate transport of UL20p and gK to the TGN, because retention of UL20p in the endoplasmic reticulum (ER) via the addition of an ER retention signal at the carboxyl terminus of UL20p forced the ER retention of gK and drastically inhibited intracellular virion envelopment and virus-induced cell fusion.


Assuntos
Proteínas de Membrana/metabolismo , Proteínas Virais de Fusão/metabolismo , Proteínas Virais/metabolismo , Vírion/metabolismo , Rede trans-Golgi/metabolismo , Animais , Western Blotting , Chlorocebus aethiops , Eletroforese em Gel de Poliacrilamida , Imunoprecipitação , Microscopia Eletrônica de Transmissão , Transporte Proteico/fisiologia , Células Vero , Proteínas do Envelope Viral/metabolismo , Rede trans-Golgi/ultraestrutura
9.
Virology ; 360(2): 264-74, 2007 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-17134730

RESUMO

The SARS-coronavirus (SARS-CoV) is the etiological agent of the severe acute respiratory syndrome (SARS). The SARS-CoV spike (S) glycoprotein mediates membrane fusion events during virus entry and virus-induced cell-to-cell fusion. The cytoplasmic portion of the S glycoprotein contains four cysteine-rich amino acid clusters. Individual cysteine clusters were altered via cysteine-to-alanine amino acid replacement and the modified S glycoproteins were tested for their transport to cell-surfaces and ability to cause cell fusion in transient transfection assays. Mutagenesis of the cysteine cluster I, located immediately proximal to the predicted transmembrane, domain did not appreciably reduce cell-surface expression, although S-mediated cell fusion was reduced by more than 50% in comparison to the wild-type S. Similarly, mutagenesis of the cysteine cluster II located adjacent to cluster I reduced S-mediated cell fusion by more than 60% compared to the wild-type S, while cell-surface expression was reduced by less than 20%. Mutagenesis of cysteine clusters III and IV did not appreciably affect S cell-surface expression or S-mediated cell fusion. The wild-type S was palmitoylated as evidenced by the efficient incorporation of (3)H-palmitic acid in wild-type S molecules. S glycoprotein palmitoylation was significantly reduced for mutant glycoproteins having cluster I and II cysteine changes, but was largely unaffected for cysteine cluster III and IV mutants. These results show that the S cytoplasmic domain is palmitoylated and that palmitoylation of the membrane proximal cysteine clusters I and II may be important for S-mediated cell fusion.


Assuntos
Fusão Celular , Fusão de Membrana , Glicoproteínas de Membrana/metabolismo , Ácido Palmítico/metabolismo , Processamento de Proteína Pós-Traducional , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/fisiologia , Proteínas do Envelope Viral/metabolismo , Sequência de Aminoácidos , Animais , Membrana Celular/química , Chlorocebus aethiops , Cisteína/genética , Cisteína/fisiologia , Imuno-Histoquímica , Marcação por Isótopo , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Ácido Palmítico/análise , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/química , Glicoproteína da Espícula de Coronavírus , Trítio/metabolismo , Células Vero , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética
10.
Virology ; 341(2): 215-30, 2005 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-16099010

RESUMO

The SARS-coronavirus (SARS-CoV) is the etiological agent of severe acute respiratory syndrome (SARS). The SARS-CoV spike (S) glycoprotein mediates membrane fusion events during virus entry and virus-induced cell-to-cell fusion. To delineate functional domains of the SARS-CoV S glycoprotein, single point mutations, cluster-to-lysine and cluster-to-alanine mutations, as well as carboxyl-terminal truncations were investigated in transient expression experiments. Mutagenesis of either the coiled-coil domain of the S glycoprotein amino terminal heptad repeat, the predicted fusion peptide, or an adjacent but distinct region, severely compromised S-mediated cell-to-cell fusion, while intracellular transport and cell-surface expression were not adversely affected. Surprisingly, a carboxyl-terminal truncation of 17 amino acids substantially increased S glycoprotein-mediated cell-to-cell fusion suggesting that the terminal 17 amino acids regulated the S fusogenic properties. In contrast, truncation of 26 or 39 amino acids eliminating either one or both of the two endodomain cysteine-rich motifs, respectively, inhibited cell fusion in comparison to the wild-type S. The 17 and 26 amino-acid deletions did not adversely affect S cell-surface expression, while the 39 amino-acid truncation inhibited S cell-surface expression suggesting that the membrane proximal cysteine-rich motif plays an essential role in S cell-surface expression. Mutagenesis of the acidic amino-acid cluster in the carboxyl terminus of the S glycoprotein as well as modification of a predicted phosphorylation site within the acidic cluster revealed that this amino-acid motif may play a functional role in the retention of S at cell surfaces. This genetic analysis reveals that the SARS-CoV S glycoprotein contains extracellular domains that regulate cell fusion as well as distinct endodomains that function in intracellular transport, cell-surface expression, and cell fusion.


Assuntos
Fusão Celular , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/genética , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Membrana Celular/química , Chlorocebus aethiops , Imuno-Histoquímica , Fusão de Membrana , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/fisiologia , Dados de Sequência Molecular , Mutação Puntual , Estrutura Terciária de Proteína , Transporte Proteico , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/fisiologia , Deleção de Sequência , Glicoproteína da Espícula de Coronavírus , Células Vero , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/fisiologia , Proteínas Virais de Fusão/análise
11.
J Virol ; 75(24): 12431-8, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11711633

RESUMO

To facilitate detection of glycoprotein K (gK) specified by herpes simplex virus, a 12-amino-acid epitope tag was inserted within gK domain III. Recombinant virus gKprotC-DIII, expressing the tagged gK, was isolated. This virus formed wild-type plaques and replicated as efficiently as the wild-type KOS virus in Vero cells. Anti-protein C MAb detected high-mannose and Golgi complex-dependent glycosylated gK within cells as well as on purified virions. The gK-null virus DeltagK (gK(-/-)) entered Vero cells substantially more slowly than the wild-type KOS (gK(+/+)), while DeltagK virus grown in complementing VK302 cells (gK(-/+)) entered with entry kinetics similar to those of the KOS virus.


Assuntos
Complexo de Golgi/metabolismo , Simplexvirus/fisiologia , Proteínas Virais/fisiologia , Vírion/fisiologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/imunologia , Chlorocebus aethiops , Glicosilação , Dados de Sequência Molecular , Simplexvirus/química , Células Vero , Vírion/química
12.
Virology ; 287(1): 18-29, 2001 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-11504538

RESUMO

Previous studies from our laboratory indicated that a 28-amino-acid carboxyl-terminal truncation of gB caused extensive virus-induced cell fusion (Baghian et al., 1993, J Virol 67, 2396-2401). We tested the ability of additional truncations and mutations within gB to cause cell fusion in the recently established virus-free cell fusion assay (Turner et al., 1998, J. Virol. 72, 873-875). Deletion of the carboxyl-terminal 28 amino acids of gB (gBDelta28), which removed part of the predicted alpha-helical structure H17b, caused extensive cell fusion. A gB truncation specified by gBDelta36, which removed the entire H17b domain, caused as much cell fusion as the gBDelta28 truncation. Similarly, gB(A874P) containing a substitution of an Ala with Pro within H17b caused cell fusion. Heparin, a gB-specific inhibitor of virus-induced cell fusion, inhibited both wild-type gB and gB(syn3)-mediated cell fusion. In contrast, fusion of cells transfected with gB(Delta28), gB(Delta36), or gB(A874P) was resistant to heparin inhibition of cell fusion. We concluded the following: (1) The predicted alpha-helical structure of H17b within the carboxyl terminus of gB is involved in both virus-induced and virus-free cell fusion. (2) Heparin is a specific inhibitor of gB-mediated fusion in both systems. (3) Resistance to heparin inhibition of gB-mediated cell fusion is associated with the predicted alpha-helical structure H17b within the carboxyl terminus of gB.


Assuntos
Heparina/farmacologia , Fusão de Membrana/efeitos dos fármacos , Proteínas do Envelope Viral/química , Sequência de Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Imunofluorescência , Células Gigantes/virologia , Dados de Sequência Molecular , Conformação Proteica , Estrutura Secundária de Proteína , Relação Estrutura-Atividade , Transfecção , Células Vero
13.
Blood ; 97(8): 2366-73, 2001 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-11290599

RESUMO

Human herpesvirus 8 (HHV-8) is a recently discovered gammaherpesvirus that is the etiologic agent of Kaposi sarcoma (KS). The natural history of primary HHV-8 infection, including clinical outcome and host immune responses that may be important in preventing disease related to HHV-8, has not been elucidated. The present study characterized the clinical, immunologic, and virologic parameters of primary HHV-8 infection in 5 cases detected during a 15-year longitudinal study of 108 human immunodeficiency virus type 1 seronegative men in the Multicenter AIDS Cohort Study. Primary HHV-8 infection was associated with mild, nonspecific signs and symptoms of diarrhea, fatigue, localized rash, and lymphadenopathy. There were no alterations in numbers of CD4(+) or CD8(+) T cells or CD8(+) T-cell interferon gamma (IFN-gamma) production to mitogen or nominal antigen. CD8(+) cytotoxic T-lymphocyte precursor (CTLp) and IFN-gamma reactivity were detected during primary HHV-8 infection, with broad specificity to 5 lytic cycle proteins of HHV-8 encoded by open reading frame 8 (ORF 8; glycoprotein B homolog of Epstein-Barr virus), ORF 22 (gH homolog), ORF 25 (major capsid protein homolog), ORF 26 (a minor capsid protein homolog), or ORF 57 (an early protein homolog), in association with increases in serum antibody titers and appearance of HHV-8 DNA in blood mononuclear cells. CD8(+) T-cell responses to HHV-8 decreased by 2 to 3 years after primary infection. This antiviral T-cell response may control initial HHV-8 infection and prevent development of disease.


Assuntos
Antígenos Virais/imunologia , Glicoproteínas , Infecções por Herpesviridae/imunologia , Herpesvirus Humano 8/imunologia , Proteínas Virais/imunologia , Adulto , Sequência de Aminoácidos , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Capsídeo/imunologia , DNA Viral/sangue , Exantema/etiologia , Fadiga/etiologia , Soronegatividade para HIV , Infecções por Herpesviridae/epidemiologia , Homossexualidade , Humanos , Memória Imunológica , Imunofenotipagem , Incidência , Interferon gama/biossíntese , Ionomicina/farmacologia , Estudos Longitudinais , Doenças Linfáticas/etiologia , Ativação Linfocitária , Contagem de Linfócitos , Masculino , Pessoa de Meia-Idade , Mitógenos/farmacologia , Dados de Sequência Molecular , Fosfoproteínas/imunologia , Estudos Prospectivos , Subpopulações de Linfócitos T , Acetato de Tetradecanoilforbol/farmacologia , Proteínas do Envelope Viral/imunologia , Proteínas da Matriz Viral/imunologia , Viremia/imunologia , Viremia/virologia
15.
J Med Microbiol ; 49(12): 1119-1127, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11129725

RESUMO

Numerous respiratory bovine coronaviruses (RBCV) were isolated recently from nasal swab samples and lung tissues of feedlot cattle with acute respiratory tract disease. These newly emerging RBCV isolates exhibited distinct phenotypic features that differentiated them from enteropathogenic bovine coronaviruses (EBCV). The RBCV strains had a receptor-destroying enzyme function mediated by acetylesterase (AE) activity of the haemagglutinin-esterase (HE) glycoprotein. The HE genes of wild-type EBCV strain LY138 and RBCV strains OK-0514 (OK) and LSU-94LSS-051 (LSU) were cloned, sequenced and transiently expressed in COS-7 cells. The enzymic properties of HE proteins in COS-7 cellular extracts and in purified virus preparations were assayed at room temperature, 37 degrees C and 39 degrees C by two different assays. One assay used p-nitrophenyl acetate (PNPA) as substrate and detected serine-esterase activity; the second assay monitored AE function with bovine submaxillary mucin (BSM) as substrate. The PNPA tests confirmed that HE proteins of EBCV and RBCV were functionally expressed in transfected COS-7 cells. Time-dependent determination of the AE activity of purified RBCV OK and LSU particles showed lower AE activity at 39 degrees C than at 37 degrees C, whereas the purified EBCV LY particles retained full AE activity at both 37 degrees C and 39 degrees C. Transiently expressed RBCV HE exhibited a marked reduction of AE activity after 40 min of assay time at 37 degrees C. In contrast, the AE activity of the transiently expressed EBCV HE remained stable beyond 40 min. The deduced amino-acid sequences of the HE proteins specified by the RBCV strains OK and LSU contained specific amino-acid changes in comparison with the EBCV LY strain, which may be responsible for the observed enzymic differences. These results are consistent with the hypothesis that RBCV strains have evolved to selectivelyreplicate in respiratory tissues and that HE may play a role in this tissue tropism.


Assuntos
Acetilesterase/metabolismo , Doenças dos Bovinos/microbiologia , Infecções por Coronavirus/veterinária , Coronavirus Bovino/enzimologia , Hemaglutininas Virais/metabolismo , Infecções Respiratórias/veterinária , Proteínas Virais de Fusão/metabolismo , Acetatos/metabolismo , Acetilesterase/química , Sequência de Aminoácidos , Animais , Células COS , Bovinos , Clonagem Molecular , Infecções por Coronavirus/virologia , Coronavirus Bovino/genética , Coronavirus Bovino/isolamento & purificação , Esterases/metabolismo , Hemaglutininas Virais/química , Hemaglutininas Virais/genética , Mucinas/metabolismo , Nitrofenóis/metabolismo , Infecções Respiratórias/virologia , Análise de Sequência de DNA , Glândula Submandibular/metabolismo , Temperatura , Transfecção , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/genética
16.
J Clin Microbiol ; 38(9): 3291-8, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10970373

RESUMO

Respiratory tract infections with viruses and Pasteurella spp. were determined sequentially among 26 cattle that died during two severe epizootics of shipping fever pneumonia. Nasal swab and serum samples were collected prior to onset of the epizootics, during disease progression, and after death, when necropsies were performed and lung samples were collected. Eighteen normal control cattle also were sampled at the beginning of the epizootics as well as at weekly intervals for 4 weeks. Respiratory bovine coronaviruses (RBCV) were isolated from nasal secretions of 21 and 25 cattle before and after transport. Two and 17 cattle nasally shed Pasteurella spp. before and after transport, respectively. RBCV were isolated at titers of 1 x 10(3) to 1.2 x 10(7) PFU per g of lung tissue from 18 cattle that died within 7 days of the epizootics, but not from the lungs of the remaining cattle that died on days 9 to 36. Twenty-five of the 26 lung samples were positive for Pasteurella spp., and their CFU ranged between 4.0 x 10(5) and 2.3 x 10(9) per g. Acute and subacute exudative, necrotizing lobar pneumonia characterized the lung lesions of these cattle with a majority of pneumonic lung lobes exhibiting fibronecrotic and exudative changes typical of pneumonic pasteurellosis, but other lung lobules had histological changes consisting of bronchiolitis and alveolitis typical of virus-induced changes. These cattle were immunologically naive to both infectious agents at the onset of the epizootics, but those that died after day 7 had rising antibody titers against RBCV and Pasteurella haemolytica. In contrast, the 18 clinically normal and RBCV isolation-negative cattle had high hemagglutinin inhibition antibody titers to RBCV from the beginning, while their antibody responses to P. haemolytica antigens were delayed. Evans' criteria for causation were applied to our findings because of the multifactorial nature of shipping fever pneumonia. This analysis identified RBCV as the primary inciting cause in these two epizootics. These viruses were previously not recognized as a causative agent in this complex respiratory tract disease of cattle.


Assuntos
Infecções por Coronavirus/veterinária , Coronavirus Bovino/isolamento & purificação , Pasteurella/isolamento & purificação , Pasteurelose Pneumônica/microbiologia , Pasteurelose Pneumônica/virologia , Animais , Anticorpos Antibacterianos/sangue , Anticorpos Antivirais/imunologia , Bovinos , Coronavirus Bovino/patogenicidade , Coronavirus Bovino/fisiologia , Testes de Inibição da Hemaglutinação , Pulmão/microbiologia , Pulmão/patologia , Pulmão/virologia , Mannheimia haemolytica/isolamento & purificação , Cavidade Nasal/microbiologia , Cavidade Nasal/virologia , Pasteurella/classificação , Pasteurella/patogenicidade , Pasteurella multocida/isolamento & purificação , Pasteurelose Pneumônica/fisiopatologia , Eliminação de Partículas Virais
17.
J Infect Dis ; 182(3): 928-32, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10950791

RESUMO

T cell immunity to lytic proteins of herpesviruses is important in host control of infection. We have characterized the cytotoxic T lymphocyte (CTL) response to 5 human herpesvirus 8 (HHV-8) homologues of lytic proteins in HHV-8-seropositive individuals. HLA class I-restricted, CD8(+) CTL responses to >/=1 HHV-8 lytic protein were detected in all 14 HHV-8-seropositive study subjects tested, with or without human immunodeficiency virus type 1 (HIV-1) infection, but not in any of 5 HHV-8-seronegative individuals. Seven of these study subjects with both HHV-8 and HIV-1 infection had greater anti-CTL reactivity to glycoprotein H (open-reading frame 22) than did the 7 study subjects infected only with HHV-8. Moreover, there was a strong, inverse correlation between HIV-1 load and glycoprotein H-specific CTL lysis in the study subjects infected with both viruses. CTL reactivity to HHV-8 lytic proteins may be involved in host control of HHV-8-related diseases, such as Kaposi's sarcoma.


Assuntos
Infecções Oportunistas Relacionadas com a AIDS/imunologia , Soronegatividade para HIV/imunologia , HIV-1 , Herpesvirus Humano 8 , Sarcoma de Kaposi/imunologia , Linfócitos T Citotóxicos/efeitos dos fármacos , Antígenos Virais/imunologia , Estudos de Coortes , Citotoxicidade Imunológica , Feminino , Vetores Genéticos , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Imunofenotipagem , Masculino , Fases de Leitura Aberta , Linfócitos T Citotóxicos/imunologia , Vaccinia virus , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Carga Viral
18.
Virology ; 269(1): 18-25, 2000 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-10725194

RESUMO

Human herpesvirus 8 (HHV-8) or Kaposi's sarcoma-associated herpesvirus (KSHV) is the only known human member of the Rhadinovirus genus of the gammaherpesvirus subfamily. Antibodies against peptides representing portions of the amino- and carboxyl-termini of HHV-8 gB were produced and used to detect gB expression in Vero cells transfected with the gB gene, in the HHV-8-harboring cell line, BCBL-1, and in purified virions. Expression of gB was detected in approximately 3% of uninduced BCBL-1 cells, while up to 30% of the cells expressed gB after 12-O-tetradecanoylphorbol-13-acetate (TPA) induction of virus replication. Indirect immunofluorescence assays and confocal microscopy showed that gB was distributed throughout the cytoplasm of BCBL-1 cells and transfected Vero cells. Immunoblot analyses of virion preparations revealed the presence of full-length as well as two smaller than full-length gB-derived species corresponding to the amino- and carboxy-terminal portions of gB, respectively. Biochemical analysis of the gB carbohydrate moieties using glycosylation inhibitors revealed that gB contained N-linked oligosaccharides of the high-mannose type, characteristic of precursor carbohydrate chains added in the endoplasmic reticulum.


Assuntos
Herpesvirus Humano 8/química , Fragmentos de Peptídeos/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas do Envelope Viral/metabolismo , Amidoidrolases/metabolismo , Animais , Western Blotting , Células COS , Chlorocebus aethiops , Citoplasma/virologia , Técnica Indireta de Fluorescência para Anticorpo , Glicosilação/efeitos dos fármacos , Herpesvirus Humano 8/efeitos dos fármacos , Herpesvirus Humano 8/isolamento & purificação , Herpesvirus Humano 8/metabolismo , Hexosaminidases/metabolismo , Manose/metabolismo , Peso Molecular , Fragmentos de Peptídeos/biossíntese , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Peptídeo-N4-(N-acetil-beta-glucosaminil) Asparagina Amidase , Acetato de Tetradecanoilforbol/farmacologia , Transfecção , Células Tumorais Cultivadas , Tunicamicina/farmacologia , Células Vero , Proteínas do Envelope Viral/biossíntese , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Replicação Viral/efeitos dos fármacos
19.
J Virol ; 73(10): 8457-68, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10482598

RESUMO

Herpes simplex virus type 1 (KOS)DeltagK is a mutant virus which lacks glycoprotein K (gK) and exhibits defects in virion egress (S. Jayachandra, A. Baghian, and K. G. Kousoulas, J. Virol. 69:5401-5413, 1997). To further understand the role of gK in virus egress, we constructed recombinant viruses, DeltagKhpd-1, -2, -3, and -4, that specified gK amino-terminal portions of 139, 239, 268, and 326 amino acids, respectively, corresponding to truncations immediately after each of the four putative membrane-spanning domains of gK. DeltagKhpd-1 and DeltagKhpd-2 viruses produced lower yields and smaller plaques than DeltagK. Numerous DeltagKhpd-1 capsids accumulated predominately within large double-membrane vesicles of which the inner membrane appeared to be derived from viral envelopes while the outer membrane appeared to originate from the outer nuclear membrane. The mutant virus DeltagKhpd-3 produced higher yields and larger plaques than the DeltagK virus. The mutant virus DeltagKhpd-4 produced yields and plaques similar to those of the wild-type virus strain KOS, indicating that deletion of the carboxy-terminal 12 amino acids did not adversely affect virus replication and egress. Comparisons of the gK primary sequences specified by alphaherpesviruses revealed the presence of a cysteine-rich motif (CXXCC), located within domain III in the lumen side of gK, and a tyrosine-based motif, YTKPhi (where Phi is any bulky hydrophobic amino acid), located between the second and third hydrophobic domains (domain II) in the cytoplasmic side of gK. The mutant virus gK/Y183S, which was constructed to specify gK with a single-amino-acid change (Y to S) within the YTKPhi motif, replicated less efficiently than the DeltagK virus. The mutant virus gK/C304S-C307S, which was constructed to specify two serine instead of cysteine residues within the cysteine-rich motif (CXXCC changed to SXXSC) of gK domain III, replicated more efficiently than the DeltagK virus. Our data suggests that gK contains domains in its amino-terminal portion that promote aberrant nucleocapsid envelopment and/or membrane fusion between different virion envelopes and contains domains within its domains II and III that function in virus replication and egress.


Assuntos
Herpes Simples/virologia , Herpesvirus Humano 1/fisiologia , Proteínas Virais/genética , Sequência de Aminoácidos , Animais , Chlorocebus aethiops , Regulação Viral da Expressão Gênica , Herpes Simples/genética , Herpesvirus Humano 1/ultraestrutura , Microscopia Eletrônica , Dados de Sequência Molecular , Mutação , Alinhamento de Sequência , Células Vero , Replicação Viral/genética
20.
Virology ; 258(2): 365-74, 1999 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-10366573

RESUMO

We cloned the gene specified by African monkey kidney cells (Vero) that codes for the homolog of the herpes virus entry mediator (HveA) specified by HeLa cells. The primary sequence of the monkey HveA (HveAs) differed significantly from HveA. Single amino acid differences were distributed throughout the amino and carboxyl terminal portions of the HveAs in comparison with the HveA, whereas certain regions were highly conserved. The predicted membrane spanning domains of the two receptors differed substantially due to insertions and deletions of short amino acid sequences. The ability of HveAs to mediate HSV virus entry was tested in a series of experiments using the recombinant virus KOS/EGFP, which constitutively expressed the enhanced green fluorescence protein (EGFP) and Chinese hamster ovary cells (CHO) transformed with the HveAs gene. The KOS/EGFP virus was constructed by inserting an EGFP gene cassette within the intergenic region between the UL53 (gK) and UL54 (ICP27) genes. The KOS/EGFP virus formed viral plaques and replicated as well as the wild-type KOS virus. HveAs-transformed CHO cells constitutively expressing HveAs mediated herpesvirus entry efficiently, whereas cells transformed with the HveAs gene in the noncoding orientation did not mediate virus entry. A genetically engineered protein composed of the amino-terminal portion of the HveAs protein fused to the heavy chain of mouse IgG immunoglobulin as well as mouse antibodies raised against HveAs blocked virus entry into HveAs-transformed CHO cells. Thus, HveAs is the functional homolog of HveA.


Assuntos
Herpesvirus Humano 1/metabolismo , Receptores do Fator de Necrose Tumoral , Receptores Virais/metabolismo , Sequência de Aminoácidos , Animais , Células CHO , Linhagem Celular Transformada , Chlorocebus aethiops , Clonagem Molecular , Cricetinae , Expressão Gênica , Proteínas de Fluorescência Verde , Células HeLa , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiologia , Humanos , Rim/citologia , Proteínas Luminescentes/genética , Camundongos , Dados de Sequência Molecular , Membro 14 de Receptores do Fator de Necrose Tumoral , Recombinação Genética , Células Vero , Ensaio de Placa Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA