Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
1.
J Gen Virol ; 98(3): 357-363, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27983470

RESUMO

Deletion or truncation of NS1, the principal IFN antagonist of influenza viruses, leads to increased IFN induction during influenza virus infection. We have studied activation of the IFN induction cascade by both wild-type and NS1-defective viruses at the single-cell level using a cell line expressing GFP under the control of the IFN-ß promoter and by examining MxA expression. The IFN-ß promoter was not activated in all infected cells even during NS1-defective virus infections. Loss of NS1 expression is therefore insufficient per se to induce IFN in an infected cell, and factors besides NS1 expression status must dictate whether the IFN response is activated. The IFN response was efficiently stimulated in these cells following infection with other viruses; the differential IFN response we observe with influenza viruses is therefore not cell specific but is likely due to differences in the nature of the infecting virus particles and their subsequent replication.


Assuntos
Vírus da Influenza A/fisiologia , Influenza Humana/imunologia , Influenza Humana/virologia , Interferon beta/genética , Regiões Promotoras Genéticas , Ativação Transcricional , Proteínas não Estruturais Virais/metabolismo , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Humanos , Vírus da Influenza A/genética , Influenza Humana/genética , Proteínas de Resistência a Myxovirus/genética , Análise de Célula Única , Proteínas não Estruturais Virais/genética , Internalização do Vírus , Replicação Viral
2.
J Virol ; 90(20): 9446-56, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27512068

RESUMO

UNLABELLED: We have previously shown that IFIT1 is primarily responsible for the antiviral action of interferon (IFN) alpha/beta against parainfluenza virus type 5 (PIV5), selectively inhibiting the translation of PIV5 mRNAs. Here we report that while PIV2, PIV5, and mumps virus (MuV) are sensitive to IFIT1, nonrubulavirus members of the paramyxoviridae such as PIV3, Sendai virus (SeV), and canine distemper virus (CDV) are resistant. The IFIT1 sensitivity of PIV5 was not rescued by coinfection with an IFIT1-resistant virus (PIV3), demonstrating that PIV3 does not specifically inhibit the antiviral activity of IFIT1 and that the inhibition of PIV5 mRNAs is regulated by cis-acting elements. We developed an in vitro translation system using purified human IFIT1 to further investigate the mechanism of action of IFIT1. While the translations of PIV2, PIV5, and MuV mRNAs were directly inhibited by IFIT1, the translations of PIV3, SeV, and CDV mRNAs were not. Using purified human mRNA-capping enzymes, we show biochemically that efficient inhibition by IFIT1 is dependent upon a 5' guanosine nucleoside cap (which need not be N7 methylated) and that this sensitivity is partly abrogated by 2'O methylation of the cap 1 ribose. Intriguingly, PIV5 M mRNA, in contrast to NP mRNA, remained sensitive to inhibition by IFIT1 following in vitro 2'O methylation, suggesting that other structural features of mRNAs may influence their sensitivity to IFIT1. Thus, surprisingly, the viral polymerases (which have 2'-O-methyltransferase activity) of rubulaviruses do not protect these viruses from inhibition by IFIT1. Possible biological consequences of this are discussed. IMPORTANCE: Paramyxoviruses cause a wide variety of diseases, and yet most of their genes encode structural proteins and proteins involved in their replication cycle. Thus, the amount of genetic information that determines the type of disease that paramyxoviruses cause is relatively small. One factor that will influence disease outcomes is how they interact with innate host cell defenses, including the interferon (IFN) system. Here we show that different paramyxoviruses interact in distinct ways with cells in a preexisting IFN-induced antiviral state. Strikingly, all the rubulaviruses tested were sensitive to the antiviral action of ISG56/IFIT1, while all the other paramyxoviruses tested were resistant. We developed novel in vitro biochemical assays to investigate the mechanism of action of IFIT1, demonstrating that the mRNAs of rubulaviruses can be directly inhibited by IFIT1 and that this is at least partially because their mRNAs are not correctly methylated.


Assuntos
Proteínas de Transporte/farmacologia , Paramyxoviridae/genética , Biossíntese de Proteínas/genética , RNA Mensageiro/genética , Rubulavirus/genética , Células A549 , Proteínas Adaptadoras de Transdução de Sinal , Linhagem Celular Tumoral , Humanos , Interferon-alfa/metabolismo , Metilação , Vírus da Caxumba/genética , Vírus da Parainfluenza 5/genética , Capuzes de RNA/genética , RNA Viral/genética , Proteínas de Ligação a RNA , Vírus Sendai/genética , Replicação Viral/genética
3.
J Virol ; 87(9): 4798-807, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23449801

RESUMO

Preparations of parainfluenza virus 5 (PIV5) that are potent activators of the interferon (IFN) induction cascade were generated by high-multiplicity passage in order to accumulate defective interfering virus genomes (DIs). Nucleocapsid RNA from these virus preparations was extracted and subjected to deep sequencing. Sequencing data were analyzed using methods designed to detect internal deletion and "copyback" DIs in order to identify and characterize the different DIs present and to approximately quantify the ratio of defective to nondefective genomes. Trailer copybacks dominated the DI populations in IFN-inducing preparations of both the PIV5 wild type (wt) and PIV5-VΔC (a recombinant virus that does not encode a functional V protein). Although the PIV5 V protein is an efficient inhibitor of the IFN induction cascade, we show that nondefective PIV5 wt is unable to prevent activation of the IFN response by coinfecting copyback DIs due to the interfering effects of copyback DIs on nondefective virus protein expression. As a result, copyback DIs are able to very rapidly activate the IFN induction cascade prior to the expression of detectable levels of V protein by coinfecting nondefective virus.


Assuntos
Vírus Defeituosos/genética , Genoma Viral , Infecções por Rubulavirus/imunologia , Infecções por Rubulavirus/virologia , Rubulavirus/genética , Animais , Linhagem Celular , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Interferons/genética , Interferons/imunologia , Infecções por Rubulavirus/genética , Proteínas Virais/genética
4.
J Gen Virol ; 94(Pt 1): 59-68, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23052390

RESUMO

Interferon (IFN) induces an antiviral state in cells that results in alterations of the patterns and levels of parainfluenza virus type 5 (PIV5) transcripts and proteins. This study reports that IFN-stimulated gene 56/IFN-induced protein with tetratricopeptide repeats 1 (ISG56/IFIT1) is primarily responsible for these effects of IFN. It was shown that treating cells with IFN after infection resulted in an increase in virus transcription but an overall decrease in virus protein synthesis. As there was no obvious decrease in the overall levels of cellular protein synthesis in infected cells treated with IFN, these results suggested that ISG56/IFIT1 selectively inhibits the translation of viral mRNAs. This conclusion was supported by in vitro translation studies. Previous work has shown that ISG56/IFIT1 can restrict the replication of viruses lacking a 2'-O-methyltransferase activity, an enzyme that methylates the 2'-hydroxyl group of ribose sugars in the 5'-cap structures of mRNA. However, the data in the current study strongly suggested that PIV5 mRNAs are methylated at the 2'-hydroxyl group and thus that ISG56/IFIT1 selectively inhibits the translation of PIV5 mRNA by some as yet unrecognized mechanism. It was also shown that ISG56/IFIT1 is primarily responsible for the IFN-induced inhibition of PIV5.


Assuntos
Proteínas de Transporte/biossíntese , Interferon-alfa/farmacologia , Infecções por Respirovirus/virologia , Respirovirus/efeitos dos fármacos , Respirovirus/genética , Proteínas Virais/biossíntese , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Chlorocebus aethiops , Replicação do DNA , Técnicas de Silenciamento de Genes , Humanos , Interferon alfa-2 , Biossíntese de Proteínas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Viral/genética , Proteínas de Ligação a RNA , Proteínas Recombinantes/farmacologia , Respirovirus/metabolismo , Infecções por Respirovirus/tratamento farmacológico , Infecções por Respirovirus/metabolismo , Transcrição Gênica , Células Vero , Proteínas Virais/genética , Proteínas Virais/metabolismo , Replicação Viral/efeitos dos fármacos , Replicação Viral/genética
5.
J Gen Virol ; 93(Pt 2): 299-307, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22049094

RESUMO

Conflicting reports exist regarding the requirement for virus replication in interferon (IFN) induction by paramyxoviruses. Our previous work has demonstrated that pathogen-associated molecular patterns capable of activating the IFN-induction cascade are not normally generated during virus replication, but are associated instead with the presence of defective interfering (DI) viruses. We demonstrate here that DIs of paramyxoviruses, including parainfluenza virus 5, mumps virus and Sendai virus, can activate the IFN-induction cascade and the IFN-ß promoter in the absence of virus protein synthesis. As virus protein synthesis is an absolute requirement for paramyxovirus genome replication, our results indicate that these DI viruses do not require replication to activate the IFN-induction cascade.


Assuntos
Interferon beta/biossíntese , Interferon beta/genética , Paramyxoviridae/imunologia , Paramyxoviridae/fisiologia , Regiões Promotoras Genéticas , Ativação Transcricional , Replicação Viral , Animais , Linhagem Celular , Vírus Defeituosos/genética , Vírus Defeituosos/imunologia , Humanos , Paramyxoviridae/genética , Rubulavirus , Proteínas Virais/biossíntese
6.
J Virol ; 83(3): 1465-73, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19019954

RESUMO

The RNA helicases encoded by melanoma differentiation-associated gene 5 (mda-5) and retinoic acid-inducible gene I (RIG-I) detect foreign cytoplasmic RNA molecules generated during the course of a virus infection, and their activation leads to induction of type I interferon synthesis. Paramyxoviruses limit the amount of interferon produced by infected cells through the action of their V protein, which binds to and inhibits mda-5. Here we show that activation of both mda-5 and RIG-I by double-stranded RNA (dsRNA) leads to the formation of homo-oligomers through self-association of the helicase domains. We identify a region within the helicase domain of mda-5 that is targeted by all paramyxovirus V proteins and demonstrate that they inhibit activation of mda-5 by blocking dsRNA binding and consequent self-association. In addition to this commonly targeted domain, some paramyxovirus V proteins target additional regions of mda-5. In contrast, V proteins cannot bind to RIG-I and consequently have no effect on the ability of RIG-I to bind dsRNA or to form oligomers.


Assuntos
RNA Helicases DEAD-box/antagonistas & inibidores , Paramyxoviridae/fisiologia , Proteínas Virais/fisiologia , Animais , Biopolímeros , Linhagem Celular , Chlorocebus aethiops , RNA Helicases DEAD-box/metabolismo , Humanos , Hidrólise , Helicase IFIH1 Induzida por Interferon , Técnicas do Sistema de Duplo-Híbrido , Células Vero
7.
J Physiol ; 587(Pt 13): 3159-73, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19403603

RESUMO

Influenza A viruses cause lung disease via an incompletely understood mechanism that involves the accumulation of liquid within the lungs. The accumulation of lung liquid is normally prevented by epithelial Na(+) absorption, a transport process regulated via several pathways including phosphoinositide-3-kinase (PI3K). Since the influenza A virus encodes a non-structural protein (NS1) that can activate this kinase, we now explore the effects of NS1 upon the biophysical properties of human airway epithelial cells. Transient expression of NS1 depolarized electrically isolated cells maintained in glucocorticoid-free medium by activating a cation conductance identical to the glucocorticoid-induced conductance seen in single cells. This response involved PI3K-independent and PI3K-dependent mechanisms. Infecting glucocorticoid-deprived cells with influenza A virus disrupted the normal electrical coupling between neighbouring cells, but also activated a conductance identical to that induced by NS1. This response to virus infection was only partially dependent upon NS1-mediated activation of PI3K. The presence of NS1 allows influenza A to modify the biophysical properties of infected cells by activating a Na(+)-permeable conductance. Whilst the activation of Na(+)-permeable channels may be expected to increase the rate of Na(+) absorption and thus reduce the volume of liquid in the lung, liquid does normally accumulate in influenza A-infected lungs. The overall effect of influenza A on lung liquid volume may therefore reflect a balance between the activation and inhibition of Na(+)-permeable channels.


Assuntos
Vírus da Influenza A/patogenicidade , Canais Iônicos/metabolismo , Sistema Respiratório/metabolismo , Sistema Respiratório/virologia , Fenômenos Biofísicos , Linhagem Celular , Dexametasona/farmacologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Humanos , Vírus da Influenza A/genética , Influenza Humana/metabolismo , Influenza Humana/virologia , Transporte de Íons/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Sistema Respiratório/citologia , Sódio/metabolismo , Transfecção , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/toxicidade
8.
J Gen Virol ; 90(Pt 11): 2731-2738, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19625458

RESUMO

Although the Enders strain of mumps virus (MuV) encodes a functional V protein that acts as an interferon (IFN) antagonist, in multi-cycle growth assays MuV Enders grew poorly in naïve ('IFN-competent' Hep2) cells but grew to high titres in 'IFN-compromised' Hep2 cells. Even so, the growth rate of MuV Enders was significantly slower in 'IFN-compromised' Hep2 cells when compared with its replication rate in Vero cells and with the replication rate of parainfluenza virus type 5 (a closely related paramyxovirus) in both naïve and 'IFN-compromised' Hep2 cells. This suggests that a consequence of slower growth is that the IFN system of naïve Hep2 cells can respond quickly enough to control the growth of MuV Enders. This is supported by the finding that rapidly growing variants of MuV Enders that were selected on 'IFN-compromised' Hep2 cells (i.e. in the absence of any selection pressure exerted by the IFN response) also grew to high titres on naïve Hep2 cells. Sequencing of the complete genome of one of these variants identified a single point mutation that resulted in a substitution of a conserved asparagine by histidine at position 498 of the haemagglutinin-neuraminidase protein, although this mutation was not present in all rapidly growing variants. These results support the concept that there is a race between the ability of a cell to detect and respond to virus infection and the ability of a virus to block the IFN response. Importantly, this emphasizes that factors other than viral IFN antagonists influence the sensitivity of viruses to IFN.


Assuntos
Interferons/antagonistas & inibidores , Interferons/imunologia , Vírus da Caxumba/imunologia , Vírus da Caxumba/fisiologia , Replicação Viral , Substituição de Aminoácidos/genética , Animais , Linhagem Celular , Chlorocebus aethiops , Análise Mutacional de DNA , Proteína HN/genética , Humanos , Mutação de Sentido Incorreto , Ensaio de Placa Viral
9.
J Cell Biol ; 109(6 Pt 2): 3273-89, 1989 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-2557352

RESUMO

The hemagglutinin-neuraminidase (HN) glycoprotein of the paramyxovirus SV5 is a type II integral membrane protein that is expressed at the infected cell surface. The intracellular assembly and transport of HN in CV1 cells was examined using conformation-specific HN mAbs and sucrose density sedimentation analysis. HN was found to oligomerize with a t1/2 of 25-30 min and these data suggest the oligomer is a tetramer consisting primarily of two noncovalently associated disulfide-linked dimers. As HN oligomers could be found that were sensitive to endoglycosidase H digestion and oligomers formed in the presence of the ER to the Golgi complex transport inhibitor, carbonylcyanide m-chlorophenylhydrazone (CCCP), these data are consistent with HN oligomerization occurring in the ER. Unfolded or immature HN molecules that could not be recognized by conformation-specific antibodies were found to specifically associate with the resident ER protein GRP78-BiP. Immunoprecipitation of BiP-HN complexes with an immunoglobulin heavy-chain binding protein (BiP) antibody indicated that newly synthesized HN associated and dissociated from GRP78-BiP (t1/2 20-25 min) in an inverse correlation with the gain in reactivity with a HN conformation-specific antibody, suggesting that the transient association of GRP78-BiP with immature HN is part of the normal HN maturation pathway. After pulse-labeling of HN in infected cells, it was found that HN is rapidly turned over in cells (t1/2 2-2.5 h). This led to the finding that the vast majority of HN expressed at the cell surface, rather than being incorporated into budding virions, is internalized and degraded after localization to endocytic vesicles and lysosomes.


Assuntos
Proteínas de Transporte/metabolismo , Retículo Endoplasmático/metabolismo , Proteína HN/metabolismo , Proteínas de Choque Térmico , Proteínas de Membrana/metabolismo , Chaperonas Moleculares , Vírus da Parainfluenza 1 Humana/metabolismo , Animais , Linhagem Celular , Eletroforese em Gel de Poliacrilamida , Chaperona BiP do Retículo Endoplasmático , Exocitose , Lisossomos/metabolismo , Camundongos , Oligopeptídeos/biossíntese , Conformação Proteica , Coelhos
10.
Gene ; 221(1): 59-68, 1998 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-9852950

RESUMO

A series of vectors is described which enables the episomal expression of proteins fused to different tag sequences in Schizosaccharomyces pombe. Proteins can be expressed with their amino termini fused to GFP/EGFP, three copies of the HA or Pk epitopes or a combined tag which contains two copies of the myc epitope and six histidine residues (MH). Fusion of the carboxyl terminus of a protein to a tag is possible with GFP/EGFP or Pk. Expression of the fusion proteins is controlled by the medium strength mutant version of the regulatable nmt1 promoter.


Assuntos
Vetores Genéticos/genética , Schizosaccharomyces/genética , Sequência de Aminoácidos , Anticorpos Monoclonais/genética , Sequência de Bases , Clonagem Molecular , Epitopos/genética , Regulação Fúngica da Expressão Gênica , Vetores Genéticos/química , Proteínas de Fluorescência Verde , Hemaglutininas/genética , Histidina/genética , Proteínas Luminescentes/genética , Dados de Sequência Molecular , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Schizosaccharomyces/química , Schizosaccharomyces/citologia
11.
J Immunol Methods ; 60(1-2): 147-65, 1983 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-6304196

RESUMO

A method is described for forming monolayers of Staphylococcus aureus Cowan strain A on plastic tissue culture plates (A plates). Bacteria remain bound to such plates even under strong protein denaturing conditions. The specific binding of antibody to A plates provides a rapid method for immune precipitation on a solid matrix. Antibody can be covalently crosslinked to staphylococcus monolayers by fixation with paraformaldehyde without significant loss of specific antigen binding capacity. Furthermore antigen-antibody complexes may be efficiently disrupted with 9 M urea and non-ionic detergent and both antibody and antigen renatured by removal of urea, antibody regaining the ability to bind fresh antigen. In the presence of 9 M urea and 1% Nonidet P-40 fixed antibody remains bound to plates while antigen is released into the urea solution, providing a method for the immunological purification of proteins. Plates with such fixed antibody may be used multiple times to bind antigen. The use of this method is illustrated by the purification of 2 adenovirus structural proteins and a herpes simplex virus glycoprotein, by means of specific monoclonal antibodies to these proteins crosslinked to A plates. A method is also described to enrich for functional antibody from immune precipitates by dissociating antibody-antigen complexes with 9 M urea and 1% Nonidet P-40 and isolating the antigen free antibody on Staphylococcus aureus Cowan strain A. A plates have also been used to separate suspension cells on the basis of their cell surface antigens (e.g., lymphocyte subpopulations or cells expressing virus antigens on their surface). Cells were either (1) reacted with specific antisera to cell surface determinants and panned over A plates, cells with antibody on their surface binding to the monolayer, or (2) panned over A plates to which specific antibody had been coupled. Separated cells may be probed directly for certain properties, such as their ability to support virus replication. The staphylococcus monolayer does not prevent the growth of tissue culture cells on the plastic surface and the selected cell population can be examined for cells giving rise to infectious centres by the subsequent addition of permissive cells.


Assuntos
Antígenos de Superfície/análise , Técnicas Imunológicas , Linfócitos/imunologia , Proteínas Virais/isolamento & purificação , Animais , Anticorpos Monoclonais/imunologia , Sítios de Ligação de Anticorpos , Separação Celular , Eletroforese em Gel de Poliacrilamida , Células HeLa/imunologia , Humanos , Linfócitos/classificação , Camundongos , Camundongos Endogâmicos BALB C , Coelhos , Simplexvirus/imunologia , Proteína Estafilocócica A/metabolismo , Staphylococcus aureus/crescimento & desenvolvimento , Staphylococcus aureus/metabolismo
12.
J Immunol Methods ; 224(1-2): 141-50, 1999 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-10357214

RESUMO

The monoclonal antibody (mAb) SV5-Pk is used widely in a variety of procedures to detect recombinant proteins tagged with the Pk tag, a 14 amino acid sequence derived from the P and V proteins of the paramyxovirus Simian Virus 5. Here we report on the isolation and characterisation of four additional SV5-Pk mAbs (termed SV5-Pk2 to 5) that bind the Pk tag. All the SV5-Pk mAbs can detect Pk tagged recombinant proteins in a variety of immunological procedures, including ELISA and immunofluorescence. Using SPOT technology, the minimal binding epitope of each SV5-Pk mAb was defined by one-sided terminal truncation analysis from either the amino- or carboxy-ends of the Pk peptide. Each mAb recognises slightly different epitopes within the Pk tag, ranging from 5 to 9 amino acids in length. The equilibrium dissociation constants (Kd) of the mAbs, as measured by surface plasmon resonance, ranged from approximately 20 to 60 pmol. Cysteine scanner mutations throughout the Pk tag revealed that some amino acids within the minimal binding epitopes were critical for mAb binding, while others could readily be substituted with little or no effect on antibody binding. The development of the Pk tag as a spacer arm for site-directed chemical coupling, and the use of the mAbs to monitor purification and coupling procedures, is discussed.


Assuntos
Anticorpos Monoclonais/imunologia , Epitopos de Linfócito B/imunologia , Fosfoproteínas/imunologia , Proteínas Virais/imunologia , Proteínas Estruturais Virais , Sequência de Aminoácidos , Animais , Afinidade de Anticorpos , Sítios de Ligação , Chlorocebus aethiops , Cricetinae , Reações Cruzadas , Cisteína , Mapeamento de Epitopos , Imunofluorescência , Células HeLa , Humanos , Hibridomas , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Proteínas de Ligação a RNA , Células Vero
13.
Am J Med ; 60(2): 293-9, 1976 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-814812

RESUMO

In two patients with terminal renal failure, manifestations of disease developed in multiple organ systems. One had a previous diagnosis of multiple myeloma with kappa light chain proteinemia and proteinuria. The other had idiopathic lobular glomerulonephritis. Hepatic and neurologic abnormalities developed in both; in the latter gastrointestinal, cardiac and endocrine disease developed as well. Clinical and pathologic correlations suggest that the retention and tissue deposition of light chains produced the organ dysfunction, inasmuch as free kappa light chain determinants were demonstrated histologically in the clinically affected organs. The deposition in these patients may be an extreme example of a common but previously unrecognized form of plasma cell dyscrasia.


Assuntos
Cadeias Leves de Imunoglobulina , Mieloma Múltiplo/imunologia , Adulto , Idoso , Membrana Basal/imunologia , Feminino , Humanos , Cadeias kappa de Imunoglobulina , Rim/imunologia , Falência Renal Crônica/imunologia , Glomérulos Renais/imunologia , Masculino
14.
AIDS Res Hum Retroviruses ; 10(6): 665-74, 1994 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8074930

RESUMO

Two commercially available expression vectors were modified to generate plasmids pGEXcPk and pQ9cPk. Proteins expressed from pGEXcPk and pQ9cPk had a short oligopeptide tag termed Pk at their carboxy termini and either glutathione S-transferase (GST) or a small histidine (His) tag, respectively, at their N termini. GST fusion proteins can be purified on immobilized glutathione and proteins coupled to the His tag selectively bind to Ni(2+)-NTA columns. The Pk tag is recognized by monoclonal antibody (MAb) SV5-P-k, previously produced in our laboratory. Thus proteins expressed from the pGEXcPk and pQ9cPk vectors can be purified in a two-step procedure, first via the N-terminal tag and second via the C-terminal tag. The combination of two affinity purification steps significantly improves the antigen purity and selects for full-size proteins. Moreover, by using the MAbSV5-P-k in the second purification step, Pk-linked antigens can be assembled directly into solid matrix-antibody-antigen (SMAA) complexes for use as vaccines. The genes for nef, endonuclease, p15, p17, p27, protease, Rev, reverse transcriptase (rt), tat, vif, vpr, and vpx of simian immunodeficiency virus (SIV mac 251) were cloned and expressed as both GST-SIV-Pk and His-SIV-Pk proteins. Multivalent SMAA complexes were made that contained His-p17-Pk, His-p27-Pk, His-rt-Pk, His-vpx-Pk, and His-vpr-Pk. Following two immunizations of mice with this mixture, antibodies could be detected to all five SIV antigens. When compared to single-protein immunizations, the immunogenicity of some of the proteins in this cocktail was either enhanced or decreased. Mice were also immunized with His-p17-Pk or His-p17-Pk-antibody complexes in the presence or absence of alum. The antibody-antigen complexes induced two- to four-fold higher antibody levels than antigen alone but did not appear to be more immunogenic in inducing lymphoproliferative responses. Sera from SIV-infected macaques were tested for the presence of antibodies reacting with the recombinant proteins by Western blot analysis. Antibodies to endonuclease, p15, p17, p27, rt, and vif were readily detected, antibodies against protease and vpx were present at much lower levels, but no antibodies were detected to nef, rev, tat, or vpr. Thus, we have developed a comprehensive range of reagents (available on request) that can be used to examine immune responses to SIV in both mice and monkeys.


Assuntos
Vírus da Imunodeficiência Símia/química , Vírus da Imunodeficiência Símia/imunologia , Proteínas Virais/imunologia , Proteínas Virais/isolamento & purificação , Animais , Complexo Antígeno-Anticorpo/imunologia , Escherichia coli/genética , Macaca , Camundongos , Plasmídeos , Proteínas Virais de Fusão , Proteínas Virais/genética
15.
J Virol Methods ; 53(1): 149-56, 1995 May.
Artigo em Inglês | MEDLINE | ID: mdl-7543487

RESUMO

A small 14 amino acid oligopeptide tag (termed SV5-Pk) was fused onto the carboxy-terminus of simian immunodeficiency virus gp160 expressed from a recombinant baculovirus. The presence of the Pk tag had no obvious effect on the expression and glycosylation of gp160 and did not interfere either with CD4 binding or with cleavage at its maturation site by the protease furin. The presence of the Pk tag did, however, facilitate the simplified purification of full-length gp160 and its incorporation into immunogenic solid matrix-antibody-antigen (SMAA) complexes.


Assuntos
Complexo Antígeno-Anticorpo/metabolismo , Antígenos CD4/metabolismo , Epitopos/metabolismo , Produtos do Gene env/metabolismo , Oligopeptídeos/metabolismo , Animais , Baculoviridae , Epitopos/genética , Produtos do Gene env/genética , Oligopeptídeos/genética , Ligação Proteica , Processamento de Proteína Pós-Traducional , Proteínas Recombinantes de Fusão/metabolismo
16.
Virology ; 415(1): 39-46, 2011 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-21511322

RESUMO

It is generally thought that pathogen-associated molecular patterns (PAMPs) responsible for triggering interferon (IFN) induction are produced during virus replication and, to limit the activation of the IFN response by these PAMPs, viruses encode antagonists of IFN induction. Here we have studied the induction of IFN by parainfluenza virus type 5 (PIV5) at the single-cell level, using a cell line expressing GFP under the control of the IFN-ß promoter. We demonstrate that a recombinant PIV5 (termed PIV5-VΔC) that lacks a functional V protein (the viral IFN antagonist) does not activate the IFN-ß promoter in the majority of infected cells. We conclude that viral PAMPs capable of activating the IFN induction cascade are not produced or exposed during the normal replication cycle of PIV5, and suggest instead that defective viruses are primarily responsible for inducing IFN during PIV5 infection in this system.


Assuntos
Interferon beta/antagonistas & inibidores , Interferon beta/genética , Regiões Promotoras Genéticas , Rubulavirus/fisiologia , Proteínas Virais/fisiologia , Animais , Linhagem Celular , Chlorocebus aethiops , Vírus Defeituosos/genética , Vírus Defeituosos/fisiologia , Imunofluorescência , Regulação da Expressão Gênica , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Humanos , Immunoblotting , Interferon beta/metabolismo , Mutação , Rubulavirus/genética , Células Vero , Proteínas Virais/genética , Replicação Viral
18.
Virology ; 407(2): 247-55, 2010 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-20833406

RESUMO

The infection of cells by RNA viruses is associated with the recognition of virus PAMPs (pathogen-associated molecular patterns) and the production of type I interferon (IFN). To counter this, most, if not all, RNA viruses encode antagonists of the IFN system. Here we present data on the dynamics of IFN production and response during developing infections by paramyxoviruses, influenza A virus and bunyamwera virus. We show that only a limited number of infected cells are responsible for the production of IFN, and that this heterocellular production is a feature of the infecting virus as opposed to an intrinsic property of the cells.


Assuntos
Vírus Bunyamwera/patogenicidade , Vírus da Influenza A/patogenicidade , Interferon Tipo I/metabolismo , Rim/virologia , Pulmão/virologia , Paramyxoviridae/patogenicidade , Animais , Vírus Bunyamwera/imunologia , Linhagem Celular Tumoral/virologia , Chlorocebus aethiops , Interações Hospedeiro-Patógeno , Humanos , Vírus da Influenza A/imunologia , Interferon Tipo I/genética , Interferon-alfa/genética , Interferon-alfa/metabolismo , Interferon beta/genética , Interferon beta/metabolismo , Rim/citologia , Rim/imunologia , Pulmão/citologia , Pulmão/imunologia , Paramyxoviridae/imunologia , Especificidade da Espécie , Células Vero/virologia , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA