Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Sci Signal ; 12(601)2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31575732

RESUMEN

The retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs) RIG-I, MDA5, and LGP2 stimulate inflammatory and antiviral responses by sensing nonself RNA molecules produced during viral replication. Here, we investigated how LGP2 regulates the RIG-I- and MDA5-dependent induction of type I interferon (IFN) signaling and showed that LGP2 interacted with different components of the RNA-silencing machinery. We identified a direct protein-protein interaction between LGP2 and the IFN-inducible, double-stranded RNA binding protein PACT. The LGP2-PACT interaction was mediated by the regulatory C-terminal domain of LGP2 and was necessary for inhibiting RIG-I-dependent responses and for amplifying MDA5-dependent responses. We described a point mutation within LGP2 that disrupted the LGP2-PACT interaction and led to the loss of LGP2-mediated regulation of RIG-I and MDA5 signaling. These results suggest a model in which the LGP2-PACT interaction regulates the inflammatory responses mediated by RIG-I and MDA5 and enables the cellular RNA-silencing machinery to coordinate with the innate immune response.


Asunto(s)
Antivirales/metabolismo , Proteína 58 DEAD Box/metabolismo , Helicasa Inducida por Interferón IFIH1/metabolismo , ARN Helicasas/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Chlorocebus aethiops , Proteína 58 DEAD Box/genética , Enterovirus Humano B/genética , Enterovirus Humano B/fisiología , Células HEK293 , Células HeLa , Humanos , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Helicasa Inducida por Interferón IFIH1/genética , Mengovirus/genética , Mengovirus/fisiología , Unión Proteica , ARN Helicasas/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Proteínas de Unión al ARN/genética , Receptores Inmunológicos , Transducción de Señal/genética , Células Vero
2.
Int J Food Microbiol ; 202: 57-65, 2015 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-25771512

RESUMEN

Noroviruses (genogroup I (NoV GI) and genogroup II (NoV GII)) and the hepatitis A virus (HAV) are frequently involved in foodborne infections worldwide. They are mainly transmitted via the fecal-oral route, direct person-to-person contact or consumption of contaminated water and foods. In food virology, detection methods are currently based on identifying viral genomes using real-time reverse transcriptase PCR (RT-qPCR). One of the general requirements for detecting these viruses in food involves the use of a process control virus to monitor the quality of the entire viral extraction procedure as described in the ISO/TS 15216-1 and 15216-2 standards published in 2013. The selected process control virus should have similar morphological and physicochemical properties as the screened pathogenic virus and thus have the potential to provide comparable extraction efficiency. The aim of this study was to determine which virus should be used for process control, murine norovirus (MNV-1) or Mengovirus, when testing for the presence of HAV, NoV GI and NoV GII in bottled water, lettuce and semi-dried tomatoes. Food samples were spiked with HAV, NoV GI or NoV GII alone or in the presence of MNV-1 or Mengovirus. Recovery rates of each pathogenic virus were compared to those of both process control viruses using a multiple comparison procedure. Neither process control virus influenced the recovery of pathogenic virus regardless of the type of food matrix. MNV-1 was the most appropriate virus for validating the detection of HAV and NoV GII in all three food matrices as well as NoV GI in lettuce. Mengovirus proved to be the most appropriate control for NoV GI detection in bottled water and semi-dried tomatoes. The process control virus is essential for validating viral detection in food and the choice of virus depends on food type and the screened pathogenic virus.


Asunto(s)
Microbiología de Alimentos/métodos , Alimentos/virología , Virus de la Hepatitis A/fisiología , Norovirus/fisiología , Microbiología del Agua , Lactuca/virología , Solanum lycopersicum/virología , Mengovirus/fisiología , Reproducibilidad de los Resultados
3.
J Virol ; 87(11): 6314-25, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23536668

RESUMEN

Virus infection can initiate a type I interferon (IFN-α/ß) response via activation of the cytosolic RNA sensors retinoic acid-inducible gene-I (RIG-I) and melanoma differentiation-associated gene 5 (MDA5). Furthermore, it can activate kinases that phosphorylate eukaryotic translation initiation factor 2α (eIF2α), which leads to inhibition of (viral) protein translation and formation of stress granules (SG). Most viruses have evolved mechanisms to suppress these cellular responses. Here, we show that a mutant mengovirus expressing an inactive leader (L) protein, which we have previously shown to be unable to suppress IFN-α/ß, triggered SG formation in a protein kinase R (PKR)-dependent manner. Furthermore, we show that infection of cells that are defective in SG formation yielded higher viral RNA levels, suggesting that SG formation acts as an antiviral defense mechanism. Since the induction of both IFN-α/ß and SG is suppressed by mengovirus L, we set out to investigate a potential link between these pathways. We observed that MDA5, the intracellular RNA sensor that recognizes picornaviruses, localized to SG. However, activation of the MDA5 signaling pathway did not trigger and was not required for SG formation. Moreover, cells that were unable to form SG-by protein kinase R (PKR) depletion, using cells expressing a nonphosphorylatable eIF2α protein, or by drug treatment that inhibits SG formation-displayed a normal IFN-α/ß response. Thus, although MDA5 localizes to SG, this localization seems to be dispensable for induction of the IFN-α/ß pathway.


Asunto(s)
Infecciones por Cardiovirus/enzimología , Gránulos Citoplasmáticos/enzimología , ARN Helicasas DEAD-box/metabolismo , Interferón-alfa/genética , Interferón beta/genética , Mengovirus/fisiología , Animales , Infecciones por Cardiovirus/genética , Infecciones por Cardiovirus/virología , Gránulos Citoplasmáticos/genética , ARN Helicasas DEAD-box/genética , Humanos , Helicasa Inducida por Interferón IFIH1 , Interferón-alfa/metabolismo , Interferón beta/metabolismo , Mengovirus/genética , Ratones , Ratones Noqueados , Transporte de Proteínas
4.
J Virol ; 86(10): 5574-83, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22438537

RESUMEN

Viruses often elicit cell injury (cytopathic effect [CPE]), a major cause of viral diseases. CPE is usually considered to be a prerequisite for and/or consequence of efficient viral growth. Recently, we proposed that viral CPE may largely be due to host defensive and viral antidefensive activities. This study aimed to check the validity of this proposal by using as a model HeLa cells infected with mengovirus (MV). As we showed previously, infection of these cells with wild-type MV resulted in necrosis, whereas a mutant with incapacitated antidefensive ("security") viral leader (L) protein induced apoptosis. Here, we showed that several major morphological and biochemical signs of CPE (e.g., alterations in cellular and nuclear shape, plasma membrane, cytoskeleton, chromatin, and metabolic activity) in cells infected with L(-) mutants in the presence of an apoptosis inhibitor were strongly suppressed or delayed for long after completion of viral reproduction. These facts demonstrate that the efficient reproduction of a lytic virus may not directly require development of at least some pathological alterations normally accompanying infection. They also imply that L protein is involved in the control of many apparently unrelated functions. The results also suggest that the virus-activated program with competing necrotic and apoptotic branches is host encoded, with the choice between apoptosis and necrosis depending on a variety of intrinsic and extrinsic conditions. Implementation of this defensive suicidal program could be uncoupled from the viral reproduction. The possibility of such uncoupling has significant implications for the pathogenesis and treatment of viral diseases.


Asunto(s)
Infecciones por Cardiovirus/virología , Efecto Citopatogénico Viral , Regulación hacia Abajo , Interacciones Huésped-Patógeno , Mengovirus/fisiología , Replicación Viral , Infecciones por Cardiovirus/inmunología , Infecciones por Cardiovirus/patología , Células HeLa , Humanos , Mengovirus/genética , Mengovirus/inmunología , Proteínas Virales/genética , Proteínas Virales/inmunología
5.
J Virol ; 83(7): 3150-61, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19144712

RESUMEN

Representatives of several picornavirus genera have been shown previously to significantly enhance non-controllable bidirectional exchange of proteins between nuclei and cytoplasm. In enteroviruses and rhinoviruses, enhanced permeabilization of the nuclear pores appears to be primarily due to proteolytic degradation of some nucleoporins (protein components of the pore), whereas this effect in cardiovirus-infected cells is triggered by the leader (L) protein, devoid of any enzymatic activities. Here, we present evidence that expression of L alone was sufficient to cause permeabilization of the nuclear envelope in HeLa cells. In contrast to poliovirus, mengovirus infection of these cells did not elicit loss of nucleoporins Nup62 and Nup153 from the nuclear pore complex. Instead, nuclear envelope permeabilization was accompanied by hyperphosphorylation of Nup62 in cells infected with wild-type mengovirus, whereas both of these alterations were suppressed in L-deficient virus mutants. Since phosphorylation of Nup62 (although less prominent) did accompany permeabilization of the nuclear envelope prior to its mitotic disassembly in uninfected cells, we hypothesize that cardiovirus L alters the nucleocytoplasmic traffic by hijacking some components of the normal cell division machinery. The variability and biological significance of picornaviral interactions with the nucleocytoplasmic transport in the infected cells are discussed.


Asunto(s)
Mengovirus/fisiología , Proteínas de Complejo Poro Nuclear/metabolismo , Proteínas Virales/metabolismo , Células HeLa , Humanos , Membrana Nuclear/virología , Permeabilidad , Fosforilación , Proteínas Virales/genética
6.
Biochem Pharmacol ; 76(7): 831-40, 2008 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-18703022

RESUMEN

Unlike polyribonucleotides, such as poly(I:C), chlorite-oxidized oxyamylose (COAM) has been poorly characterized as a polyanionic antiviral. COAM possesses a controversial interferon (IFN)-inducing capacity and its mechanism of action has not been elucidated. In this study, COAM was biochemically characterized and fractionated according to molecular mass. In comparison with a strong IFN induction and upregulation of the helicase RIG-I and MDA-5 mRNAs by poly(I:C), COAM did not enhance IFN-alpha or -beta and IFN-inducible RNA helicases in mouse fibroblastoid cells. Instead, COAM inhibited virus entry by blocking the attachment to the cells. These results suggest that COAM can alter the outcome of infection, not by IFN induction and in turn modifying the cellular antiviral state, but through inhibition of virus entry into cells.


Asunto(s)
Amilosa/análogos & derivados , Antivirales/farmacología , Mengovirus/efectos de los fármacos , Internalización del Virus/efectos de los fármacos , Amilosa/farmacología , Animales , Línea Celular , Interferón-alfa/genética , Interferón beta/genética , Mengovirus/fisiología , Ratones , Poli I-C/farmacología , ARN Mensajero/metabolismo , ARN Viral/genética , Replicación Viral/efectos de los fármacos
7.
J Gen Virol ; 88(Pt 4): 1206-1217, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17374764

RESUMEN

Previously, it was shown that pyrrolidine dithiocarbamate (PDTC) inhibits proteolytic polyprotein processing and replication of human rhinovirus by transporting metal ions into cells. Here, it is shown that PDTC also inhibits replication of two other picornaviruses: coxsackievirus B3 (CVB3), a closely related virus that belongs to the genus Enterovirus, and mengovirus, an encephalomyocarditis virus strain that belongs to the genus Cardiovirus, and that this inhibition is due to the dithiocarbamate moiety of the compound. Making use of subgenomic replicons, evidence is provided that PDTC inhibits replication of these two viruses by disturbing viral RNA synthesis. Furthermore, it is shown that PDTC transports zinc ions into cells and that these zinc ions play an important role in the antiviral activity mediated by PDTC. Finally, it is shown that PDTC interferes with proteolytic processing of the polyproteins of both CVB3 and mengovirus, but that the underlying mechanism between these two viruses differs. In CVB3-infected cells, PDTC interferes strongly with the proteolytic activity of 3CD(pro), as shown by the impaired production of the mature capsid proteins as well as the autocleavage of 3CD(pro) into 3C(pro) and 3D(pol). In mengovirus-infected cells, however, PDTC had no effect on the proteolytic production of capsid proteins or the autocleavage of 3CD(pro). Instead, PDTC caused the accumulation of a high-molecular-mass precursor protein, due to an impairment in the primary 'break' that normally occurs at the 2A-2B junction. Thus, PDTC disturbs polyprotein processing and replication of two groups of picornaviruses, enteroviruses and cardioviruses, but the underlying mechanism is different.


Asunto(s)
Antivirales/farmacología , Enterovirus Humano B/efectos de los fármacos , Mengovirus/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Pirrolidinas/farmacología , ARN Viral/biosíntesis , Tiocarbamatos/farmacología , Zinc/metabolismo , Proteasas Virales 3C , Animales , Línea Celular , Chlorocebus aethiops , Cricetinae , Cisteína Endopeptidasas , Enterovirus Humano B/fisiología , Mengovirus/fisiología , Poliproteínas/biosíntesis , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/biosíntesis , Replicación Viral/efectos de los fármacos
8.
PDA J Pharm Sci Technol ; 60(1): 72-8, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17089680

RESUMEN

In this work, we evaluate the stability of a new freeze-dried and albumin-free formulation of recombinant human IFN alpha 2b (rhIFN-alpha2b) to be used in humans. The freeze-dried, albumin-free formulation was stored at the recommended temperature of 4 degrees C, and under accelerated storage conditions (28 degrees C). The stability of this product was also compared with the stability of a liquid albumin-free formulation of this cytokine. Finally, the stability of the freeze-dried albumin-free formulation was examined after reconstitution and storage at 4 degrees C and room temperature (28 degrees C) for 30 days. Samples were periodically subjected to biological activity assay (antiviral titration), reversed phase high-performance liquid chromatography (RP-HPLC), pyrogens, sterility and enzyme-linked immunosorbent assay (ELISA) testing, abnormal toxicity screening, organoleptic evaluation, and measurement of residual moisture and pH. Accelerated storage (28 degrees C) data for the freeze-dried albumin-free formulation showed biochemical stability of the active ingredient throughout the 6-month study, showing activity between 85 and 125% of its nominal value. RP-HPLC-determined purity showed that rhIFN-alpha2b remained above 95%. Additionally, the formulation was non-pyrogenic, non-toxic, sterile, and organoleptically acceptable. The real-time storage data confirmed the good biochemical long-term (30 months) stability of the freeze-dried formulation of this cytokine. Comparison with the liquid rhIFN-alpha2b albumin-free preparation showed that the freeze-dried albumin-free formulation maintained the stability of the active ingredient better than the liquid preparation. The formulation was also stable after reconstitution and storage at 4 degrees C and 28 degrees C, for 30 days.


Asunto(s)
Antineoplásicos/química , Antivirales/química , Interferón-alfa/química , Albúminas/análisis , Antineoplásicos/farmacología , Antivirales/farmacología , Línea Celular Tumoral , Química Farmacéutica , Cromatografía Líquida de Alta Presión , Efecto Citopatogénico Viral/efectos de los fármacos , Estabilidad de Medicamentos , Almacenaje de Medicamentos , Ensayo de Inmunoadsorción Enzimática , Estudios de Evaluación como Asunto , Liofilización , Humanos , Interferón alfa-2 , Interferón-alfa/farmacología , Mengovirus/efectos de los fármacos , Mengovirus/fisiología , Proteínas Recombinantes , Temperatura , Factores de Tiempo
9.
Arch Virol ; 151(10): 1933-46, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16732496

RESUMEN

Interferons induced by viral infections can have powerful immuno- modulatory effects, and several epidemiologic studies have found an association between certain viral infections and reduced prevalence of allergy. We hypothesized that allergenic proteins could be synthesized by a replicating virus, and this construct could be useful as an immunomodulator. To test this hypothesis, we cloned an allergenic protein (ovomucoid [OVM]) into a murine picornavirus (Mengo virus) vector. This plasmid has a multicloning site surrounded by auto-catalytic sequences so that a foreign protein will be cleaved from viral proteins during replication. OVM sequences were cloned in the context of full-length viral genome cDNA, T7 RNA transcripts of this plasmid were transfected into HeLa cells, and recombinant virus plaques appeared on the second passage. Sequence analysis of recombinant viruses derived from individual plaques demonstrated that three viral isolates contained up to 2/3 of the OVM coding sequence, which was retained by the viruses after 5 additional passages in HeLa cells. The experiments verify the stable expression of immunoreactive OVM subunits by replicating viruses. These virus/allergen constructs could provide a tool to evaluate whether intracellular presentation of allergenic proteins in the context of a viral infection could prevent allergic sensitization upon re-challenge.


Asunto(s)
Alérgenos/biosíntesis , Mengovirus/fisiología , Ovomucina/biosíntesis , Virus Reordenados/fisiología , Alérgenos/genética , Animales , Infecciones por Cardiovirus/virología , Vectores Genéticos/metabolismo , Vectores Genéticos/fisiología , Genoma Viral/genética , Células HeLa/metabolismo , Humanos , Mengovirus/genética , Mengovirus/metabolismo , Ratones , Ovomucina/genética , Ovomucina/inmunología , Plásmidos/genética , Virus Reordenados/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/inmunología , Recombinación Genética , Pase Seriado , Especificidad de la Especie , Transfección , Replicación Viral
10.
Virology ; 312(2): 481-94, 2003 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-12919752

RESUMEN

Short poly(C)-tract Mengoviruses have proven vaccine efficacy in many species of animals. A novel vector for the delivery of foreign proteins was created by insertion of a second autoproteolytic primary cleavage cassette linked to a multiple cloning site (MCS) into an attenuated variant of Mengo. Nineteen cDNAs from foreign sequences that ranged from 39 to 1653 bases were cloned into the MCS. The viral reading frame was maintained and translation resulted in dual, autocatalytic excision of the foreign peptides without disruption of any Mengo proteins. All cDNAs except those with the largest insertions produced viable virus. Active proteins such as GFP, CAT, and SIV p27 were expressed within infected cells. Relative to parental Mengo, the growth kinetics and genetic stability of each vector was inversely proportional to the size of the inserted sequence. While segments up to 1000 bases could be carried, inserts greater than 500-600 bases were usually reduced in size during serial passage. The limit on carrying capacity was probably due to difficulties in virion assembly or particle stability. Yet for inserts less than 500-600 bases, the Mengo vectors provided an effective system for the delivery of foreign epitopes into cells and mice.


Asunto(s)
Ingeniería Genética , Vectores Genéticos/genética , Mengovirus/genética , Mengovirus/patogenicidad , Animales , ADN Complementario/genética , ADN Viral/genética , Expresión Génica , Genoma Viral , Células HeLa , Humanos , Mengovirus/fisiología , Ratones , Ensayo de Placa Viral , Virulencia
11.
J Virol ; 76(19): 9664-72, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12208945

RESUMEN

In our studies on the biological function of the mengovirus leader protein, we identified a casein kinase II (CK-2) phosphorylation site in the protein. Here we report that the mengovirus leader protein can be phosphorylated by CK-2 in vitro. Expression of a recombinant leader protein in which the consensus CK-2 sequence around threonine 47 was disturbed resulted in a mutant protein that could no longer be phosphorylated. The CK-2 consensus sequence was modified by site-directed mutagenesis and subsequently introduced into a mengovirus cDNA clone to investigate the effect of the phosphorylation of the leader protein on virus replication and on the host cell response. Modifications by which the CK-2 consensus sequence was disturbed resulted in mutant viruses with reduced growth kinetics. We demonstrated that the integrity of the CK-2 phosphorylation site of the mengovirus leader protein was specifically related to the suppression of NF-kappa B activation and subsequent suppression of alpha/beta interferon production in infected cells. We also found that the integrity of the CK-2 phosphorylation site of the leader protein coincided with an increase of ferritin expression in the infected cell. These data indicate that the leader protein suppresses the iron-mediated activation of NF-kappa B and thereby inhibits alpha/beta interferon expression in the infected cell.


Asunto(s)
Ferritinas/fisiología , Interferón-alfa/biosíntesis , Interferón beta/biosíntesis , Hierro/fisiología , Mengovirus/fisiología , FN-kappa B/metabolismo , Señales de Clasificación de Proteína/fisiología , Proteínas Virales/fisiología , Animales , Apoferritinas/biosíntesis , Quinasa de la Caseína II , Células HeLa , Humanos , Ratones , Óxido Nítrico Sintasa/biosíntesis , Óxido Nítrico Sintasa de Tipo II , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Especies Reactivas de Oxígeno
12.
Viral Immunol ; 15(1): 155-63, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-11952137

RESUMEN

Encephalomyocarditis virus (EMCV) and Mengo virus are highly virulent murine cardioviruses that are found in abundant quantities in the spleen and lymph nodes after infection. T lymphocytes are pivotal mediators of humoral and cellular immunity against cardioviral challenge, and are highly suspect candidates of EMCV and Mengo virus infection. We found T lymphocyte-like cell lines CTLL-2, EL-4, LY1+2/9, and LBRM33 were susceptible to productive viral infection and exhibited cytopathology after infection with virulent EMCV-R or attenuated Mengo virus strains vMC0 and vMC24. Flow cytometric analysis demonstrated progressive intracellular accumulation of viral proteins, such as the replication-dependent 3D viral polymerase, in EL-4 cells during infection. Conversely, freshly isolated and mitogen-stimulated CD4+ and CD8+ T cells were resistant to productive infection with these viruses, exhibiting no viral-induced cytopathic effects or intracellular presence of viral proteins. These data indicate that although T-lymphocyte-like tumor cell lines are highly susceptible to viral infection and cytopathic effects, primary/freshly isolated T cells are resistant to infection by EMCV-R or Mengo virus.


Asunto(s)
Virus de la Encefalomiocarditis/fisiología , Mengovirus/fisiología , Linfocitos T/virología , Animales , Línea Celular , Efecto Citopatogénico Viral , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Proteínas Virales/análisis , Replicación Viral , eIF-2 Quinasa/fisiología
13.
J Virol ; 75(7): 3111-20, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11238838

RESUMEN

An alignment of cardiovirus sequences led to the prediction of three conserved stem-loops in the 3' untranslated region (UTR) of mengovirus. Deletions of each stem were engineered in mengovirus cDNAs and also in mengovirus replicons, in which part of the viral capsid sequences were replaced with the firefly luciferase gene. The effect of deletion on RNA infectivity and plaque phenotype was evaluated after transfection of viral transcripts into HeLa cells or by luciferase assays of cellular extracts after transfection with RNA replicons. Stem I (mengovirus bases 7666 to 7687) was found to be dispensable for viral growth or exponential luciferase expression. Deletion of stem III (bases 7711 to 7721) was lethal to the virus, and the replicons were incapable of RNA synthesis. Deletion of stem II (DeltaII; bases 7692 to 7705) produced an intermediate phenotype, in that replicons had marginal RNA synthesis activity but transfection with genomic RNA usually failed to produce plaques after normal incubation times (31 h, 37 degrees C). In a few of the DeltaII transfections, however, plaques were observed after long incubation, especially if the cells received large amounts of RNA (3 microg per 3 x 10(6) cells). Viruses from two DeltaII-derived plaques were isolated and amplified. Their RNAs were converted into cDNA, sequenced, and mapped for genotype. Each maintained the DeltaII deletion and, in addition, had one or two reversion mutations, which were characterized by reverse genetics as responsible for the phenotypes. One reversion caused an amino acid change in the polymerase (3D(pol)), and the other was localized to the 3' UTR, upstream of stem I.


Asunto(s)
Regiones no Traducidas 3'/química , Mengovirus/genética , Animales , Secuencia de Bases , Femenino , Células HeLa , Humanos , Mengovirus/patogenicidad , Mengovirus/fisiología , Ratones , Ratones Endogámicos ICR , Datos de Secuencia Molecular , Fenotipo , Filogenia , ARN Viral/biosíntesis
14.
J Virol ; 74(24): 11983-7, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11090201

RESUMEN

We have previously shown that Xenopus oocytes require coinjection of both poliovirus RNA and HeLa cell extracts to support a complete cycle of viral replication yielding high levels of infectious viral particles. This novel system provides a tool for identifying host factors and for biochemically dissect individual steps that lead to virus production. Here we demonstrate that Xenopus oocytes are able to support replication of other picornaviruses such as human rhinovirus 14 and mengovirus. Unlike poliovirus, microinjection of mengovirus RNA yields high viral titers (about 10(7) PFU/oocyte) without the need for coinjection of additional cell extracts. In contrast, formation of infectious rhinovirus particles requires coinjection of human cell extracts. We found that one of these human factors is required for efficient rhinovirus translation. Our findings uncover differences in the host factor requirements among members of the picornavirus family and provide the means to identify the human protein(s) involved in rhinovirus production.


Asunto(s)
Mengovirus/fisiología , Rhinovirus/fisiología , Replicación Viral , Xenopus , Animales , Femenino , Humanos , Oocitos
15.
Viral Immunol ; 13(1): 27-35, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-10733166

RESUMEN

We have shown that genetically engineered mengoviruses containing artificially shortened 5' noncoding poly(C) tracts (e.g., C0 or C13UC10) are dramatically attenuated in adult Swiss/ICR mice when compared to wild-type virus or to a genetically engineered virus containing a wild-type length poly(C) tract (C44UC10). To explore further the relationship between poly(C) tracts and virulence, we have conducted more extensive characterizations of several engineered viruses in the murine model. Both short and long poly(C) tract viruses were highly virulent in newborn mice, underscoring the importance of age in poly(C)-mediated attenuation. Virus vMC24, with a tract sequence of C13UC10, was as attenuated in 4-week-old BALB/c, C.C3-H2k/LiMcdJ, and DBA/2 mice as in Swiss/ICR mice. But it was more pathogenic for C57BL/6 mice, and highly virulent for C3H/Hej and C3H/Hen mice, demonstrating the importance of murine genotype. As expected from its virulence in all mouse strains, vMwt, with a poly(C) of C44UC10, induced higher levels of viremia than vMC24. The vMwt also induced higher levels of circulating interferon and had reduced pathogenicity in chemically immunosuppressed Swiss/ICR mice. Similar immunosuppression did not increase the virulence of vMC24. Collectively, the data suggest that endogenous immune components and the immune competence of the host play significant roles in determining the susceptibility of mice to mengovirus infection.


Asunto(s)
Infecciones por Cardiovirus/virología , Ingeniería Genética , Mengovirus/genética , Mengovirus/patogenicidad , Animales , Animales Recién Nacidos , Encéfalo/virología , Femenino , Terapia de Inmunosupresión , Interferones/sangre , Dosificación Letal Mediana , Mengovirus/fisiología , Ratones , Ratones Endogámicos , Ratones Desnudos , Viremia/virología , Virulencia , Replicación Viral
16.
J Virol ; 74(7): 3074-81, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10708422

RESUMEN

Many virulent aphthoviruses and cardioviruses have long homopolymeric poly(C) tracts in the 5' untranslated regions of their RNA genomes. A panel of genetically engineered mengo-type cardioviruses has been described which contain a variety of different poly(C) tract lengths. Studies of these viruses have shown the poly(C) tract to be dispensable for growth in HeLa cells, although the relative murine virulence of the viruses correlates directly and positively with tract length. Compared with wild-type mengovirus strain M, mutants with shortened poly(C) tracts grow poorly in mice and protectively immunize rather than kill recipient animals. In the present study, several murine cell populations were tested to determine whether, unlike HeLa cells, they allowed a differential amplification of viruses with long or short poly(C) tracts. Replication and cytopathic studies with four hematopoietically derived cell lines (CH2B, RAW 264.7, A20.J, and P815) and two murine fibroblast cell lines [L929 and L(Y)] demonstrated that several of these cell types indeed allowed differential virus replication as a function of viral poly(C) tract length. Among the most discerning of these cells, RAW 264.7 macrophages supported vigorous lytic growth of a long-tract virus, vMwt (C(44)UC(10)), but supported only substantially diminished and virtually nonlytic growth of vMC(24) (C(13)UC(10)) and vMC(0) short-tract viruses. The viral growth differences evident in all cell lines were apparent early and continuously during every cycle of virus amplification. The data suggest that poly(C) tract-dependent attenuation of mengovirus may be due in part to a viral replication defect manifest in similar hematopoietic-type cells shortly after murine infection. The characterized cultures should provide excellent tools for molecular study of poly(C) tract-mediated virulence.


Asunto(s)
Virus de la Encefalomiocarditis/crecimiento & desarrollo , Mengovirus/crecimiento & desarrollo , Poli C/genética , Animales , Línea Celular , Efecto Citopatogénico Viral , Virus de la Encefalomiocarditis/genética , Virus de la Encefalomiocarditis/fisiología , Células HeLa , Humanos , Interferones/fisiología , Mengovirus/genética , Mengovirus/fisiología , Ratones
17.
Vaccine ; 18(20): 2132-41, 2000 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-10715528

RESUMEN

Vaccines containing minigenes - isolated antigenic epitopes encoded by short open reading frames - can, under certain circumstances, confer protective immunity upon the vaccinee. Here we evaluate the efficacy of the minigene vaccine approach using DNA immunization and find that, to be immunogenic, a minigene-encoded epitope requires a perfect "Kozak" translational initiation region. In addition, using intracellular cytokine staining, we show that immunization with a plasmid encoding a full-length protein induces epitope-specific CD8(+) T cells which are detectable directly ex vivo, and constitute approximately 2% of the vaccinee's splenic CD8(+) T cells. In contrast, such cells are undetectable directly ex vivo in recipients of a minigene vaccine. Nevertheless, the minigene plasmid does induce a low number of epitope-specific CD8(+) T cells, which can be amplified to detectable levels by in vivo stimulation. Indeed, 4 days after in vivo stimulation (by virus infection), all vaccinated mice - regardless of whether they had been vaccinated with the minigene or with the full-length gene - had similar numbers of epitope-specific CD8(+) T cells. However, despite these strong responses at 4 days post-infection, recipients of the minigene vaccine showed no enhanced ability to limit virus replication and dissemination. We therefore observe a dichotomy; minigene vaccinees are not protected, despite the presence of strong virus-specific immune responses at 4 days post-challenge. We suggest that the protective benefits of vaccination exert themselves very soon - perhaps within minutes or hours - after virus challenge. If the vaccine-induced immune response is too low to achieve this early protective effect, virus-specific T cells will expand rapidly, but ineffectually, leading to the strong but non-protective response measured at 4 days post-infection. Thus, vaccine-induced immunity should be monitored very early in infection, since the extent to which these responses may later be amplified is largely irrelevant to the protection observed.


Asunto(s)
Antígenos Virales/inmunología , Epítopos/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Mengovirus/inmunología , Virus Sincitiales Respiratorios/inmunología , Respirovirus/inmunología , Vacunas de ADN/inmunología , Virus de la Estomatitis Vesicular Indiana/inmunología , Vacunas Virales/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Antivirales/biosíntesis , Anticuerpos Antivirales/inmunología , Antígenos Virales/genética , Secuencia de Bases , Linfocitos T CD8-positivos/inmunología , Codón/genética , Citocinas/biosíntesis , Epítopos/genética , Genes Sintéticos , Inmunidad Celular , Recuento de Linfocitos , Virus de la Coriomeningitis Linfocítica/genética , Virus de la Coriomeningitis Linfocítica/fisiología , Mengovirus/genética , Mengovirus/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Sistemas de Lectura Abierta , Plásmidos/genética , Plásmidos/inmunología , Secuencias Reguladoras de Ácidos Nucleicos , Virus Sincitiales Respiratorios/genética , Respirovirus/genética , Bazo/inmunología , Factores de Tiempo , Vacunación , Virus de la Estomatitis Vesicular Indiana/genética , Replicación Viral
18.
J Virol ; 73(8): 6444-52, 1999 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10400738

RESUMEN

We describe the selective irreversible inhibition of mengovirus growth in cultured cells by a combination of two pyrrolopyrimidine nucleoside analogues, 5-bromotubercidin (BrTu) and tubercidin (Tu). At a concentration of 5 microgram/ml, BrTu reversibly blocked the synthesis of cellular mRNA and rRNA but did not inhibit either mengovirus RNA synthesis or multiplication. BrTu is a potent inhibitor of adenosine kinase, and low concentrations of BrTu (e.g., 0.5 microgram/ml), which did not by themselves inhibit cell growth, blocked phosphorylation of Tu and thus protected uninfected cells against irreversible cytotoxicity resulting from Tu incorporation into nucleic acids. In contrast, in mengovirus-infected cells, BrTu did not completely inhibit Tu incorporation into mengovirus RNA, allowing the formation of Tu-containing functionally defective polynucleotides that aborted the virus development cycle. This increased incorporation of Tu coupled to mengovirus infection could be attributed either to a reduction in the inhibitory action of BrTu and/or its nucleotide derivatives at the level of nucleoside and nucleotide kinases and/or, perhaps, to an effect upon the nucleoside transport system. The virus life cycle in nucleoside-treated cells progressed to the point of synthesis of negative strands and probably to the production of a few defective new positive strands. Irreversible virus growth arrest was achieved if the nucleoside mixture of BrTu (0.5 to 10 microgram/ml) and Tu (1 to 20 microgram/ml) was added no later than 30 min after virus infection and maintained for periods of 2 to 8 h. The cultures thus "cured" of mengovirus infection could be maintained and transferred for several weeks, during which they neither produced detectable virus nor showed a visible cytopathic effect; however, the infected and cured cells themselves, while metabolically viable, were permanently impaired in RNA synthesis and unable to divide. Although completely resistant to superinfecting picornaviruses, they retained the ability to support the growth of several other viruses (vaccinia virus, reovirus, and vesicular stomatitis virus), showing that cured cells had, in general, retained the metabolic and structural machinery needed for virus production. The resistance of cured cells to superinfection with picornaviruses seemed attributable neither to interferon action nor to destruction or blockade of virus receptors but more likely to the consumption of some host factor(s) involved in the expression of early viral functions during the original infection.


Asunto(s)
Antivirales/farmacología , Mengovirus/efectos de los fármacos , Tubercidina/análogos & derivados , Replicación Viral/efectos de los fármacos , Adenosina/análogos & derivados , Animales , Embrión de Pollo , Células HeLa , Humanos , Mengovirus/genética , Mengovirus/fisiología , Virus de la Enfermedad de Newcastle/efectos de los fármacos , Nucleósidos/farmacología , Virus Sindbis/efectos de los fármacos , Sobreinfección , Tubercidina/metabolismo , Tubercidina/farmacología , Tubercidina/toxicidad , Virus Vaccinia/efectos de los fármacos
19.
Virology ; 212(2): 438-50, 1995 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-7571413

RESUMEN

We reported previously that murine L-929 cells expressing a human interferon (IFN)-gamma cDNA lacking a signal peptide sequence synthesize but fail to secrete human IFN-gamma and support viral replication at a reduced level. These cells also had elevated levels of IFN-inducible gene products. We show here that a similar response is seen in human cells expressing a mutated murine IFN-gamma cDNA. The ability of human IFN-gamma to induce gene expression in murine cells is shown to be due to the intracellular IFN-gamma rather than to clonal variation, induction of endogenous murine IFN, or alternative mediators of antiviral activity. We have used a murine cell line, Ltk-aprt-, which is resistant to both type I and II IFNs but responsive to combined treatment with both. Ltk-aprt- cells transfected with human IFN-gamma cDNA lacking a signal sequence support virus replication at the same level as control cells. However, unlike transfectants containing only the neoR selection gene, clones expressing the mutated human IFN-gamma gene show strong protection against viral infection and elevated levels of 2,5 A synthetase mRNA and MHC class I protein after treatment with IFN-beta alone. Reverse transcriptase-PCR rules out the induction of endogenous murine IFN expression as a mediator of these effects. Thus, expression of intracellular human IFN-gamma mimics treatment with extracellular murine IFN-gamma in permitting a synergistic response to IFN-beta. Given the inability of human IFN-gamma to bind to the murine cell-surface receptor our results show that intracellular IFN-gamma can activate certain responses independent of cell-surface binding.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Interferón gamma/fisiología , Replicación Viral , Animales , Secuencia de Bases , Citoplasma/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Antígenos H-2/genética , Humanos , Interferón beta/biosíntesis , Interferón beta/farmacología , Interferón gamma/biosíntesis , Interferón gamma/genética , Interferón gamma/farmacología , Células L/metabolismo , Células L/virología , Mengovirus/fisiología , Ratones , Datos de Secuencia Molecular , Polinucleótido Ligasas/genética , Señales de Clasificación de Proteína/genética , ARN Mensajero/biosíntesis , Eliminación de Secuencia , Especificidad de la Especie , Virus de la Estomatitis Vesicular Indiana/fisiología , Replicación Viral/efectos de los fármacos , Replicación Viral/fisiología
20.
J Virol ; 69(4): 2697-9, 1995 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-7884926

RESUMEN

We have constructed three cDNA clones of encephalomyocarditis virus strain R (EMCV-R) with poly(C) tracts of C4, C9, and C20. RNA transcribed from these cDNAs was infectious to HeLa cells, and the resultant viruses grew well in this system, albeit with plaque sizes that were proportional to the poly(C) length. When injected into mice, the progeny viruses were only slightly less pathogenic than EMCV-R, and the observed degree of attenuation was not nearly as dramatic as for equivalent mengoviruses with similar short poly(C)s. Short-tract poly(C)-mediated attenuation is therefore highly dependent on viral genomic context.


Asunto(s)
Virus de la Encefalomiocarditis/patogenicidad , Mengovirus/patogenicidad , Poli C/genética , Animales , Clonación Molecular , ADN Complementario , Virus de la Encefalomiocarditis/genética , Virus de la Encefalomiocarditis/fisiología , Femenino , Células HeLa , Humanos , Mengovirus/genética , Mengovirus/fisiología , Ratones , Ensayo de Placa Viral , Virulencia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...