Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros











Intervalo de año de publicación
1.
J Virol ; 78(18): 9998-10008, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15331733

RESUMEN

A chimeric yellow fever-dengue 1 (ChimeriVax-DEN1) virus was produced by the transfection of Vero cells with chimeric in vitro RNA transcripts. The cell culture supernatant was subjected to plaque purification for the identification of a vaccine candidate without mutations. Of 10 plaque-purified clones, 1 containing no mutation (clone J) was selected for production of the vaccine virus. During subsequent cell culture passaging of this clone for vaccine production, a single amino acid substitution (K to R) occurred in the envelope (E) protein at residue 204 (E204) (F. Guirakhoo, K. Pugachev, Z. Zhang, G. Myers, I. Levenbook, K. Draper, J. Lang, S. Ocran, F. Mitchell, M. Parsons, N. Brown, S. Brandler, C. Fournier, B. Barrere, F. Rizvi, A. Travassos, R. Nichols, D. Trent, and T. Monath, J. Virol. 78:4761-4775, 2004). The same mutation was observed in another clone (clone E). This mutation attenuated the virus in 4-day-old suckling mice inoculated by the intracerebral (i.c.) route and led to reduced viremia in monkeys inoculated by the subcutaneous or i.c. route. The histopathology scores of lesions in the brain tissue of monkeys inoculated with either the E204K or E204R virus were reduced compared to those for monkeys inoculated with the reference virus, a commercial yellow fever 17D vaccine (YF-VAX). Both viruses grew to significantly lower titers than YF-VAX in HepG2, a human hepatoma cell line. After intrathoracic inoculation into mosquitoes, both viruses grew to a similar level as YF-VAX, which was significantly lower than that of their wild-type DEN1 parent virus. A comparison of the E-protein structures of nonmutant and mutant viruses suggested the appearance of new intramolecular bonds between residues 204R, 261H, and 257E in the mutant virus. These changes may be responsible for virus attenuation through a change in the pH threshold for virus envelope fusion with the host cell membrane.


Asunto(s)
Virus del Dengue/genética , Proteínas del Envoltorio Viral/genética , Virus de la Fiebre Amarilla/genética , Aedes , Sustitución de Aminoácidos , Animales , Animales Lactantes , Anticuerpos Antivirales/sangre , Línea Celular , Quimera/genética , Chlorocebus aethiops , Dengue/etiología , Dengue/patología , Dengue/prevención & control , Virus del Dengue/patogenicidad , Femenino , Humanos , Macaca fascicularis , Macaca mulatta , Masculino , Fusión de Membrana , Ratones , Ratones Endogámicos ICR , Modelos Moleculares , Mutación Puntual , Vacunas Atenuadas/genética , Células Vero , Proteínas del Envoltorio Viral/química , Vacunas Virales/genética , Viremia/etiología , Virulencia/genética , Fiebre Amarilla/etiología , Vacuna contra la Fiebre Amarilla/genética , Virus de la Fiebre Amarilla/patogenicidad
2.
J Virol ; 75(2): 934-42, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11134306

RESUMEN

A yellow fever virus (YFV)/Japanese encephalitis virus (JEV) chimera in which the structural proteins prM and E of YFV 17D are replaced with those of the JEV SA14-14-2 vaccine strain is under evaluation as a candidate vaccine against Japanese encephalitis. The chimera (YFV/JEV SA14-14-2, or ChimeriVax-JE) is less neurovirulent than is YFV 17D vaccine in mouse and nonhuman primate models (F. Guirakhoo et al., Virology 257:363-372, 1999; T. P. Monath et al., Vaccine 17:1869-1882, 1999). Attenuation depends on the presence of the JEV SA14-14-2 E protein, as shown by the high neurovirulence of an analogous YFV/JEV Nakayama chimera derived from the wild JEV Nakayama strain (T. J. Chambers, A. Nestorowicz, P. W. Mason, and C. M. Rice, J. Virol. 73:3095-3101, 1999). Ten amino acid differences exist between the E proteins of ChimeriVax-JE and the YFV/JEV Nakayama virus, four of which are predicted to be neurovirulence determinants based on various sequence comparisons. To identify residues that are involved in attenuation, a series of intratypic YFV/JEV chimeras containing either single or multiple amino acid substitutions were engineered and tested for mouse neurovirulence. Reversions in at least three distinct clusters were required to restore the neurovirulence typical of the YFV/JEV Nakayama virus. Different combinations of cluster-specific reversions could confer neurovirulence; however, residue 138 of the E protein (E(138)) exhibited a dominant effect. No single amino acid reversion produced a phenotype significantly different from that of the ChimeriVax-JE parent. Together with the known genetic stability of the virus during prolonged cell culture and mouse brain passage, these findings support the candidacy of this experimental vaccine as a novel live-attenuated viral vaccine against Japanese encephalitis.


Asunto(s)
Virus de la Encefalitis Japonesa (Especie)/genética , Virus de la Encefalitis Japonesa (Especie)/patogenicidad , Vacunas contra la Encefalitis Japonesa , Glicoproteínas de Membrana/genética , Vacunas Sintéticas , Proteínas del Envoltorio Viral/genética , Virus de la Fiebre Amarilla/genética , Secuencia de Aminoácidos , Animales , Virus de la Encefalitis Japonesa (Especie)/inmunología , Virus de la Encefalitis Japonesa (Especie)/metabolismo , Encefalitis Japonesa/virología , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos ICR , Datos de Secuencia Molecular , Sistema Nervioso/virología , Plásmidos , Proteínas Recombinantes de Fusión , Análisis de Secuencia de ADN , Transcripción Genética , Transfección , Vacunas Atenuadas , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo , Virulencia/genética , Virus de la Fiebre Amarilla/inmunología , Virus de la Fiebre Amarilla/metabolismo
3.
J Virol ; 74(12): 5477-85, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10823852

RESUMEN

A chimeric yellow fever (YF)-dengue type 2 (dengue-2) virus (ChimeriVax-D2) was constructed using a recombinant cDNA infectious clone of a YF vaccine strain (YF 17D) as a backbone into which we inserted the premembrane (prM) and envelope (E) genes of dengue-2 virus (strain PUO-218 from a case of dengue fever in Bangkok, Thailand). The chimeric virus was recovered from the supernatant of Vero cells transfected with RNA transcripts and amplified once in these cells to yield a titer of 6.3 log(10) PFU/ml. The ChimeriVax-D2 was not neurovirulent for 4-week-old outbred mice inoculated intracerebrally. This virus was evaluated in rhesus monkeys for its safety (induction of viremia) and protective efficacy (induction of anti-dengue-2 neutralizing antibodies and protection against challenge). In one experiment, groups of non-YF-immune monkeys received graded doses of ChimeriVax-D2; a control group received only the vaccine diluents. All monkeys (except the control group) developed a brief viremia and showed no signs of illness. Sixty-two days postimmunization, animals were challenged with 5.0 log(10) focus forming units (FFU) of a wild-type dengue-2 virus. No viremia (<1.7 log(10) FFU/ml) was detected in any vaccinated group, whereas all animals in the placebo control group developed viremia. All vaccinated monkeys developed neutralizing antibodies in a dose-dependent response. In another experiment, viremia and production of neutralizing antibodies were determined in YF-immune monkeys that received either ChimeriVax-D2 or a wild-type dengue-2 virus. Low viremia was detected in ChimeriVax-D2-inoculated monkeys, whereas all dengue-2-immunized animals became viremic. All of these animals were protected against challenge with a wild-type dengue-2 virus, whereas all YF-immune monkeys and nonimmune controls became viremic upon challenge. Genetic stability of ChimeriVax-D2 was assessed by continuous in vitro passage in VeroPM cells. The titer of ChimeriVax-D2, the attenuated phenotype for 4-week-old mice, and the sequence of the inserted prME genes were unchanged after 18 passages in Vero cells. The high replication efficiency, attenuation phenotype in mice and monkeys, immunogenicity and protective efficacy, and genomic stability of ChimeriVax-D2 justify it as a novel vaccine candidate to be evaluated in humans.


Asunto(s)
Anticuerpos Antivirales/biosíntesis , Virus del Dengue/inmunología , Dengue/inmunología , Dengue/prevención & control , Vacunas Virales/inmunología , Virus de la Fiebre Amarilla/inmunología , Sustitución de Aminoácidos , Animales , Animales Recién Nacidos , Anticuerpos Antivirales/inmunología , Línea Celular , Dengue/virología , Virus del Dengue/genética , Virus del Dengue/patogenicidad , Virus del Dengue/fisiología , Relación Dosis-Respuesta Inmunológica , Genes Virales/genética , Macaca mulatta , Ratones , Pruebas de Neutralización , Pase Seriado , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/genética , Viremia/inmunología , Viremia/prevención & control , Viremia/virología , Virulencia , Replicación Viral , Virus de la Fiebre Amarilla/genética , Virus de la Fiebre Amarilla/patogenicidad , Virus de la Fiebre Amarilla/fisiología
4.
J Virol ; 74(4): 1742-51, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10644345

RESUMEN

ChimeriVax-JE is a live, attenuated recombinant virus prepared by replacing the genes encoding two structural proteins (prM and E) of yellow fever 17D virus with the corresponding genes of an attenuated strain of Japanese encephalitis virus (JE), SA14-14-2 (T. J. Chambers et al., J. Virol. 73:3095-3101, 1999). Since the prM and E proteins contain antigens conferring protective humoral and cellular immunity, the immune response to vaccination is directed principally at JE. The prM-E genome sequence of the ChimeriVax-JE in diploid fetal rhesus lung cells (FRhL, a substrate acceptable for human vaccines) was identical to that of JE SA14-14-2 vaccine and differed from sequences of virulent wild-type strains (SA14 and Nakayama) at six amino acid residues in the envelope gene (E107, E138, E176, E279, E315, and E439). ChimeriVax-JE was fully attenuated for weaned mice inoculated by the intracerebral (i.c.) route, whereas commercial yellow fever 17D vaccine (YF-Vax) caused lethal encephalitis with a 50% lethal dose of 1.67 log(10) PFU. Groups of four rhesus monkeys were inoculated by the subcutaneous route with 2.0, 3.0, 4.0, and 5. 0 log(10) PFU of ChimeriVax-JE. All 16 monkeys developed low viremias (mean peak viremia, 1.7 to 2.1 log(10) PFU/ml; mean duration, 1.8 to 2.3 days). Neutralizing antibodies appeared between days 6 and 10; by day 30, neutralizing antibody responses were similar across dose groups. Neutralizing antibody titers to the homologous (vaccine) strain were higher than to the heterologous wild-type JE strains. All immunized monkeys and sham-immunized controls were challenged i.c. on day 54 with 5.2 log(10) PFU of wild-type JE. None of the immunized monkeys developed viremia or illness and had mild residual brain lesions, whereas controls developed viremia, clinical encephalitis, and severe histopathologic lesions. Immunized monkeys developed significant (>/=4-fold) increases in serum and cerebrospinal fluid neutralizing antibodies after i.c. challenge. In a standardized test for neurovirulence, ChimeriVax-JE and YF-Vax were compared in groups of 10 monkeys inoculated i.c. and analyzed histopathologically on day 30. Lesion scores in brains and spinal cord were significantly higher for monkeys inoculated with YF-Vax. ChimeriVax-JE meets preclinical safety and efficacy requirements for a human vaccine; it appears safer than yellow fever 17D vaccine but has a similar profile of immunogenicity and protective efficacy.


Asunto(s)
Virus de la Encefalitis Japonesa (Especie)/inmunología , Vectores Genéticos , Glicoproteínas de Membrana/inmunología , Proteínas del Envoltorio Viral/inmunología , Vacunas Virales/inmunología , Virus de la Fiebre Amarilla , Animales , Seguridad de Productos para el Consumidor , Virus de la Encefalitis Japonesa (Especie)/genética , Encefalitis Japonesa/prevención & control , Femenino , Humanos , Inyecciones Subcutáneas , Macaca mulatta , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos ICR , Pruebas de Neutralización , Recombinación Genética , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Proteínas del Envoltorio Viral/genética , Vacunas Virales/genética , Viremia , Virulencia , Virus de la Fiebre Amarilla/genética , Virus de la Fiebre Amarilla/patogenicidad
5.
Virology ; 257(2): 363-72, 1999 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-10329547

RESUMEN

Yellow fever (YF) 17D vaccine virus, having a 60-year history of safe and effective use, is an ideal vector to deliver heterologous genes from other medically important flaviviruses. A chimeric YF/Japanese encephalitis (JE) virus (ChimeriVax-JE virus) was constructed by insertion of the premembrane and envelope (prME) genes of an attenuated human vaccine strain (SA14-14-2) of Japanese encephalitis (JE) virus between core and nonstructural (NS) genes of a YF 17D infectious clone. The virus grew to high titers in cell cultures and was not neurovirulent for 3- to 4-week-old mice at doses /=10(3) pfu of ChimeriVax-JE virus were solidly protected against intraperitoneal challenge with a virulent JE virus. Genetic stability of the chimera was assessed by sequential passages in cell cultures or in mouse brain. All attenuating residues and the avirulent phenotype were preserved after 18 passages in cell cultures or 6 passages in mouse brains.


Asunto(s)
Virus de la Encefalitis Japonesa (Especie)/inmunología , Encefalitis Japonesa/prevención & control , Vectores Genéticos , Glicoproteínas de Membrana/inmunología , Vacunas de ADN/inmunología , Proteínas del Envoltorio Viral/inmunología , Vacunas Virales/inmunología , Virus de la Fiebre Amarilla , Animales , Encéfalo/virología , Línea Celular , Chlorocebus aethiops , Modelos Animales de Enfermedad , Virus de la Encefalitis Japonesa (Especie)/genética , Genes Virales , Vectores Genéticos/genética , Vectores Genéticos/fisiología , Humanos , Macaca mulatta , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Recombinación Genética , Análisis de Secuencia de ADN , Vacunas Atenuadas/inmunología , Vacunas de ADN/genética , Células Vero , Proteínas del Envoltorio Viral/genética , Vacunas Virales/genética , Replicación Viral , Virus de la Fiebre Amarilla/genética , Virus de la Fiebre Amarilla/crecimiento & desarrollo , Virus de la Fiebre Amarilla/fisiología
6.
Vaccine ; 17(15-16): 1869-82, 1999 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-10217584

RESUMEN

Yellow fever 17D virus, a safe and effective live, attenuated vaccine, was used as a vector for genes encoding the protective antigenic determinants of a heterologous member of the genus Flavivirus, Japanese encephalitis (JE) virus, the leading cause of acute viral central nervous system infection and death throughout Asia. The viral envelope (prM and E) genes of a full-length cDNA clone of YF 17D virus were replaced with the corresponding genes of JE SA14-14-2, a strain licensed as a live, attenuated vaccine in China. Full-length RNA transcripts of the YF/JE chimaera were used to transfect Vero cells. The progeny virus (named 'ChimeriVax-JE'), was used to define safety after intracerebral (i.c.) inoculation of rhesus monkeys. Monkeys (N = 3) inoculated with a high dose (6.6 log10 pfu) developed a brief viremia, showed no signs of illness, developed high titers of anti-JE neutralizing antibody, and had minimal brain and spinal cord lesion scores according to criteria specified in the WHO monkey neurovirulence test. A control group of 3 monkeys that received a lower dose (4.2 log10 pfu) of commercial YF 17D vaccine had slightly higher lesion scores. To develop a lethal monkey model of JE for vaccine protection tests, we inoculated groups of monkeys i.c. or intranasally (i.n.) with a JE virus strain found to be highly neurovirulent and neuroinvasive for mice. Monkeys inoculated i.c., but not i.n., developed severe encephalitis after an incubation period of 8-13 days. The ChimeriVax-JE virus was passed in a cell line acceptable for human use (diploid fetal rhesus lung) and 4.3 or 5.3 log10 pfu were inoculated into groups of 3 monkeys by the subcutaneous route. All 6 animals developed brief viremias (peak titer < 2.0 log10 pfu/ml) and subsequently had anti-JE but no yellow fever neutralizing antibodies. On day 64, the monkeys were challenged i.c. with 5.5 log10 pfu of virulent JE virus. The immunized animals had no detectable viremia post-challenge, whereas 4 unimmunized controls became viremic. Only 1 of 6 (17%) vaccinated monkeys but 4 of 4 (100%) unvaccinated controls developed encephalitis. Histopathological examination 30 days after challenge confirmed that the protected, immunized animals had no or minimal evidence of encephalitis. These data demonstrated the ability of the ChimeriVax-JE to induce a rapid humoral immune response and to protect against a very severe, direct intracerebral virus challenge. Target areas of neuronal damage and inflammation in monkeys infected IC with wild-type JE, the chimaeric virus and YF 17D were similar, indicating that the histopathological scoring system used for the WHO yellow fever monkey neurovirulence test will be applicable to control testing of chimaeric seed viruses and vaccines.


Asunto(s)
Virus de la Encefalitis Japonesa (Especie)/inmunología , Encefalitis Japonesa/prevención & control , Vacunas Sintéticas/inmunología , Proteínas no Estructurales Virales/inmunología , Proteínas Virales/inmunología , Vacunas Virales/inmunología , Virus de la Fiebre Amarilla/inmunología , Animales , Cápside/genética , Cápside/inmunología , Línea Celular , Sistema Nervioso Central/patología , Sistema Nervioso Central/virología , Relación Dosis-Respuesta Inmunológica , Evaluación Preclínica de Medicamentos , Virus de la Encefalitis Japonesa (Especie)/genética , Virus de la Encefalitis Japonesa (Especie)/crecimiento & desarrollo , Encefalitis Japonesa/patología , Encefalitis Japonesa/virología , Macaca mulatta , Pruebas de Neutralización , Análisis de Secuencia de ADN , Vacunación , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología , Proteínas no Estructurales Virales/genética , Proteínas Virales/genética , Vacunas Virales/administración & dosificación , Vacunas Virales/genética , Viremia/virología , Virus de la Fiebre Amarilla/genética , Virus de la Fiebre Amarilla/crecimiento & desarrollo
7.
Immunotechnology ; 2(3): 219-28, 1996 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9373314

RESUMEN

BACKGROUND: HNK20 is a murine IgA which is currently being investigated in clinical trials against respiratory syncytial virus (RSV) infections in infants and young children. OBJECTIVE: To produce a single chain antibody fragment (scFv) from HNK20 hybridoma cells and assess its functional activities in vitro and in vivo (mouse model). STUDY DESIGN: The V regions of heavy and light chains were cloned and linked by a sequence encoding for (Gly4 Ser)3 and expressed in Escherichia coli. RESULTS: Over 100 mg/l of the HNK20-scFv was produced in shake flasks after induction with isopropyl (beta-D-thiogalactopyranoside (IPTG). ScFv was purified under native conditions on a Ni2+ affinity column and migrated as a single band of 34 kDa on sodium dodecyl sulfate (SDS)-gels. ScFv demonstrated similar affinity as its parent IgA molecule, neutralized RSV in vitro and significantly reduced RSV titers in lungs of mice when administered intranasally shortly before or a day after RSV challenge. CONCLUSION: It is possible that this scFv or its derivatives, when applied by intranasal or pulmonary route, will be useful for treatment of RSV infections in infants and young children.


Asunto(s)
Proteína HN , Inmunoglobulina A/fisiología , Fragmentos de Inmunoglobulinas/fisiología , Región Variable de Inmunoglobulina/fisiología , Proteínas Virales de Fusión/inmunología , Proteínas Virales/inmunología , Animales , Clonación Molecular , Femenino , Expresión Génica , Inmunoglobulina A/biosíntesis , Inmunoglobulina A/genética , Fragmentos de Inmunoglobulinas/biosíntesis , Fragmentos de Inmunoglobulinas/genética , Región Variable de Inmunoglobulina/biosíntesis , Región Variable de Inmunoglobulina/genética , Cinética , Ratones , Ratones Endogámicos BALB C , Pruebas de Neutralización , Plásmidos/genética , Plásmidos/metabolismo , Reacción en Cadena de la Polimerasa , Virus Sincitiales Respiratorios/genética , Virus Sincitiales Respiratorios/inmunología , Temperatura , Transformación Genética , Proteínas del Envoltorio Viral , Proteínas Virales de Fusión/genética , Proteínas Virales/genética
8.
Virology ; 203(2): 241-9, 1994 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-8053148

RESUMEN

We have previously isolated and characterized two dengue (DEN) 2 viruses mutant in their fusion-from-within (FFWI) phenotype in the insect cell line C6/36. Both viruses lost a potential glycosylation site (Asn-153) in the envelope (E) glycoprotein. To determine whether the change in FFWI phenotype was due to a change in E-glycoprotein glycosylation, we characterized the patterns of glycosylation on the E-glycoprotein of wild-type DEN 1 and DEN 2 viruses. The E-glycoproteins were isolated from purified virus grown in Aedes albopictus C6/36 cells, by use of high-performance size-exclusion chromatography. The tryptic maps of wild-type glycosylated and enzymatically (PNGase F) deglycosylated E-glycoproteins were compared by reverse-phase high-performance liquid chromatography. The DEN 1 virus E-glycoprotein was found to have two peaks in the tryptic map that exhibited shifts after deglycosylation, whereas the DEN 2 virus E-glycoprotein had only one. Besides the potential glycosylation site at Asn-153, both DEN 1 and DEN 2 virus E-glycoproteins have another potential site located at Asn-67. Amino-terminal sequencing of the shifted peaks revealed that DEN 2 virus E-glycoprotein is glycosylated only at Asn-67; however, DEN 1 virus E-glycoprotein is glycosylated at both Asn-67 and Asn-153. These DEN virus serotypes are thus heterogeneous in their use of glycosylation sites. We also determined by a lectin-binding assay that the attached carbohydrates for both viruses were likely to be of the high-mannose type.


Asunto(s)
Virus del Dengue/química , Glicoproteínas de Membrana/análisis , Proteínas del Envoltorio Viral/análisis , Aedes , Secuencia de Aminoácidos , Animales , Línea Celular , Cromatografía en Gel , Glicosilación , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/aislamiento & purificación , Datos de Secuencia Molecular , Peso Molecular , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/aislamiento & purificación
9.
Arch Virol Suppl ; 9: 339-48, 1994.
Artículo en Inglés | MEDLINE | ID: mdl-7913359

RESUMEN

Viral membrane proteins play an important role in the assembly and disassembly of enveloped viruses. Oligomerization and proteolytic cleavage events are involved in controlling the functions of these proteins during virus entry and release. Using tick-borne encephalitis virus as a model we have studied the role of the flavivirus envelope proteins E and prM/M in these processes. Experiments with acidotropic agents provide evidence that the virus is taken up by receptor-mediated endocytosis and that the acidic pH in endosomes plays an important role for virus entry. The envelope glycoprotein E undergoes irreversible conformational changes at acidic pH, as indicated by the loss of several monoclonal antibody-defined epitopes, which coincide with the viral fusion activity in vitro. Sedimentation analysis reveals that these conformational changes lead to aggregation of virus particles, apparently by the exposure of hydrophobic sequence elements. None of these features are exhibited by immature virions containing E and prM rather than E and M. Detergent solubilization, sedimentation, and crosslinking experiments provide evidence that prM forms a complex with protein E which prevents the conformational changes necessary for fusion activity. The functional role of prM before its endoproteolytic cleavage by a cellular protease thus seems to be the protection of protein E from acid-inactivation during its passage through acidic trans Golgi vesicles in the course of virus release.


Asunto(s)
Virus de la Encefalitis Transmitidos por Garrapatas/crecimiento & desarrollo , Macrólidos , Proteínas del Envoltorio Viral/metabolismo , Ácidos , Cloruro de Amonio/farmacología , Animales , Antibacterianos/farmacología , Compartimento Celular , Células Cultivadas , Culicidae/citología , Endocitosis , Modelos Biológicos , Modelos Moleculares , Modelos Estructurales
10.
Virology ; 194(1): 219-23, 1993 May.
Artículo en Inglés | MEDLINE | ID: mdl-8480420

RESUMEN

Two types of dengue (DEN) 2 virus mutants were selected either by repeated exposure to acidic pH (acid mutant, AM), or by the addition of ammonium chloride to Aedes albopictus C6/36 cells prior to and during viral infection (fusion mutant, FM). Both mutants grew more slowly than the parent strain and induced smaller plaques in Vero cells. The 50% fusion from within index for both mutants occurred at least 0.65 pH units higher than with the wild-type DEN virus. A single amino acid substitution (Asn-153 to Asp) was found in the envelope (E)-glycoprotein of the AM virus. Three amino acid substitutions were detected on the E-glycoprotein of the FM virus: Ile-6 to Met, Asn-134 to Ser, and Asn-153 to Tyr. No mutations were found in the precursor to the membrane protein, prM. The DEN virus E-glycoprotein has two potential glycosylation sites: Asn-67 and Asn-153. The loss of the potential glycosylation site at Asn-153 or the change in the chemical characteristics resultant from the amino acid substitutions in both mutants implicates these regions of the E-glycoprotein in virus-mediated membrane fusion.


Asunto(s)
Virus del Dengue/crecimiento & desarrollo , Virus del Dengue/genética , Proteínas del Envoltorio Viral/genética , Proteínas Virales de Fusión/genética , Ácidos/farmacología , Aedes , Cloruro de Amonio/farmacología , Animales , Fusión Celular , Células Cultivadas , Chlorocebus aethiops , Concentración de Iones de Hidrógeno , Riñón/citología , Mutagénesis , Replicación Viral
11.
Virology ; 191(2): 921-31, 1992 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-1280384

RESUMEN

To study the role of the precursor to the membrane protein (prM) in flavivirus maturation, we inhibited the proteolytic processing of the Murray Valley encephalitis (MVE) virus prM to membrane protein in infected cells by adding the acidotropic agent ammonium chloride late in the virus replication cycle. Viruses purified from supernatants of ammonium chloride-treated cells contained prM protein and were unable to fuse C6/36 mosquito cells from without. When ammonium chloride was removed from the cells, both the processing of prM and the fusion activity of the purified viruses were partially restored. By using monoclonal antibodies (MAbs) specific for the envelope (E) glycoprotein of MVE virus, we found that at least three epitopes were less accessible to their corresponding antibodies in the prM-containing MVE virus particles. Amino-terminal sequencing of proteolytic fragments of the E protein which were reactive with sequence-specific peptide antisera or MAb enabled us to estimate the site of the E protein interacting with the prM to be within amino acids 200 to 327. Since prM-containing viruses were up to 400-fold more resistant to a low pH environment, we conclude that the E-prM interaction might be necessary to protect the E protein from irreversible conformational changes caused by maturation into the acidic vesicles of the exocytic pathway.


Asunto(s)
Ácidos/farmacología , Epítopos/biosíntesis , Flavivirus/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Proteínas de la Matriz Viral/metabolismo , Cloruro de Amonio/farmacología , Anticuerpos Monoclonales , Fusión Celular/efectos de los fármacos , Farmacorresistencia Microbiana , Flavivirus/efectos de los fármacos , Concentración de Iones de Hidrógeno , Modelos Moleculares , Precursores de Proteínas/efectos de los fármacos , Precursores de Proteínas/inmunología , Precursores de Proteínas/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Virales de Fusión/efectos de los fármacos , Proteínas Virales de Fusión/metabolismo , Proteínas de la Matriz Viral/efectos de los fármacos , Proteínas de la Matriz Viral/inmunología
12.
J Gen Virol ; 72 ( Pt 6): 1323-9, 1991 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-1710648

RESUMEN

The fusion activity of flaviviruses [tick-borne encephalitis (TBE) virus and Japanese encephalitis virus] was assessed by inducing fusion from without of C6/36 mosquito cells with purified virus preparations. Membrane fusion and polykaryocyte formation was observed only after incubating the viruses at acidic pH. Two groups of monoclonal antibodies reacting with distinct non-overlapping antigenic domains on the TBE virus protein E inhibited fusion from without. One of these domains contains the most highly conserved and putative fusion-active sequence of the flavivirus protein E. Of five TBE virus monoclonal antibody escape mutants, each defined by a single amino acid substitution in the envelope protein E, one revealed a reduced fusion activity and another one a lower pH threshold. TBE virus grown in the presence of ammonium chloride as well as Langat virus purified from the supernatant of infected chick embryo cells contained the precursor of protein M (prM) rather than M itself. These 'immature' virions did not cause fusion from without, suggesting that the proteolytic processing of prM may be necessary for the generation of fusion-competent virions.


Asunto(s)
Virus de la Encefalitis Japonesa (Especie)/fisiología , Virus de la Encefalitis Transmitidos por Garrapatas/fisiología , Fusión de Membrana , Proteínas del Envoltorio Viral/análisis , Aedes , Animales , Anticuerpos Monoclonales , Línea Celular , Embrión de Pollo , Epítopos/análisis , Concentración de Iones de Hidrógeno , Cinética , Células Vero , Proteínas del Envoltorio Viral/inmunología
13.
J Virol ; 64(10): 5156-9, 1990 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-2398538

RESUMEN

We have determined the virulence characteristics of seven monoclonal antibody escape mutants of tick-borne encephalitis virus in the mouse model. One of the mutants with an amino acid substitution from tyrosine to histidine at residue 384 revealed strongly reduced pathogenicity after peripheral inoculation of adult mice but retained its capacity to replicate in the mice and to induce a high-titered antibody response. Infection with the attenuated mutant resulted in resistance to challenge with virulent virus. Assessment of nonconservative amino acid substitutions in other attenuated flaviviruses suggests that a structural element including residue 384 may represent an important determinant of flavivirus virulence in general.


Asunto(s)
Virus de la Encefalitis Transmitidos por Garrapatas/genética , Encefalitis Transmitida por Garrapatas/inmunología , Mutación , Vacunas Atenuadas , Proteínas del Envoltorio Viral/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales , Formación de Anticuerpos , Embrión de Pollo , Modelos Animales de Enfermedad , Virus de la Encefalitis Transmitidos por Garrapatas/inmunología , Virus de la Encefalitis Transmitidos por Garrapatas/patogenicidad , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Proteínas del Envoltorio Viral/genética , Virulencia
14.
Zentralbl Bakteriol ; 272(4): 477-84, 1990 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-2360967

RESUMEN

Human sera obtained after tick-borne encephalitis (TBE) without prior vaccination were compared with sera from patients after a vaccination breakdown. Most sera previously shown to have high titers of IgG and IgM against TBE virus as detected in the ELISA and hemagglutination inhibition (HI) tests also reacted in Western blot with TBE virus E protein which is involved in virus neutralization. The serum of a patient with a vaccination breakdown, however, reacted only very weakly with the E protein in the Western blot in spite of a high amount of antibodies detectable in ELISA. Using SDS-denaturated virus as an antigen in ELISA (imitating the blotting condition), this serum revealed a significant reduction in its reactivity with denatured virus compared to the control sera. This indicates that the patient had an insufficient immune response against certain denaturation resistant epitopes which might contribute to development of disease despite vaccination. The analysis of the immune response of human sera at the epitope level revealed a characteristic "fingerprint" for each serum reflecting the genetic control of the production of antibody populations against different antigenic determinants.


Asunto(s)
Anticuerpos Antivirales/biosíntesis , Virus de la Encefalitis Transmitidos por Garrapatas/inmunología , Encefalitis Transmitida por Garrapatas/inmunología , Vacunación , Proteínas del Envoltorio Viral/inmunología , Unión Competitiva , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Pruebas de Inhibición de Hemaglutinación , Humanos , Inmunoglobulina G/biosíntesis , Inmunoglobulina M/biosíntesis
15.
Virology ; 169(1): 90-9, 1989 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-2466373

RESUMEN

A panel of monoclonal antibodies (MAbs) was prepared to analyze the antigenic structure of the tick-borne encephalitis (TBE) virus glycoprotein E. Nineteen different epitopes were identified and characterized with respect to serological specificity, functional activity, structural properties, and topological relationships. Except for 3 isolated epitopes (i1, i2, and i3), these cluster to form three non-overlapping domains termed A, B, and C. The structural properties of epitopes were assessed by analyzing the effect of different treatments (SDS denaturation, reduction and carboxymethylation, performic acid oxidation, exposure to pH 5.0, CNBr, and trypsin cleavage) on the antigenic reactivities of each epitope. Only 3 epitopes of domain A as well as i2 were sensitive to SDS alone, whereas all others were SDS resistant. Reduction and carboxymethylation, however, destroyed the antigenic reactivity of all epitopes of domain B and also that of two SDS-resistant epitopes of domain A, indicating the role of disulfide bridges in stabilizing the conformation of these epitopes. Deglycosylation by N-Glycanase abolished the SDS resistance of domain C, providing evidence of the role of the carbohydrate side chain in stabilizing these epitopes. A conformational change induced by acid pH was revealed by differences in protease (proteinase K) cleavage maps before and after acid pH treatment. The conformational change involved the epitopes of domain A and occurred between pH 6.0 and 5.5 with the the threshold at pH 7.0.


Asunto(s)
Antígenos Virales/análisis , Virus de la Encefalitis Transmitidos por Garrapatas/inmunología , Glicoproteínas/inmunología , Proteínas del Envoltorio Viral/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Unión Competitiva , Western Blotting , Carbohidratos/inmunología , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Epítopos/análisis , Pruebas de Hemaglutinación , Concentración de Iones de Hidrógeno , Immunoblotting , Pruebas de Neutralización , Conformación Proteica
16.
J Virol ; 63(2): 564-71, 1989 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-2463377

RESUMEN

A model of the tick-borne encephalitis virus envelope protein E is presented that contains information on the structural organization of this flavivirus protein and correlates epitopes and antigenic domains to defined sequence elements. It thus reveals details of the structural and functional characteristics of the corresponding protein domains. The localization of three antigenic domains (composed of 16 distinct epitopes) within the primary structure was performed by (i) amino-terminal sequencing of three immunoreactive fragments of protein E and (ii) sequencing the protein E-coding regions of seven antigenic variants of tick-borne encephalitis virus that had been selected in the presence of neutralizing monoclonal antibodies directed against the E protein. Further information about variable and conserved regions was obtained by a comparative computer analysis of flavivirus E protein amino acid sequences. The search for potential T-cell determinants revealed at least one sequence compatible with an amphipathic alpha-helix which is conserved in all flaviviruses sequenced so far. By combining these data with those on the location of disulfide bridges (T. Nowak and G. Wengler, Virology 156:127-137, 1987) and the structural characteristics of epitopes, such as dependency on conformation or on intact disulfide bridges or both, a model was established that goes beyond the location of epitopes in the primary sequence and reveals features of the folding of the polypeptide chain, including the generation of discontinuous protein domains.


Asunto(s)
Antígenos Virales/inmunología , Virus de la Encefalitis Transmitidos por Garrapatas/inmunología , Proteínas del Envoltorio Viral/inmunología , Secuencia de Aminoácidos , Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/inmunología , Epítopos/inmunología , Estructura Molecular , Conformación Proteica
17.
J Gen Virol ; 68 ( Pt 3): 859-64, 1987 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-3029309

RESUMEN

Strains of tick-borne encephalitis (TBE) virus isolated from ticks in natural foci in Austria were compared to strains isolated from the same foci 14 years previously. Comparative peptide mapping of the envelope (E) glycoproteins as well as analysis of the antigenic structure of the E proteins by the use of 14 monoclonal antibodies defining different epitopes did not provide evidence for antigenic variation. The same also holds true for isolates from a probably newly established natural focus in Western Austria. These results confirm previous data by showing that under natural ecological conditions TBE virus is quite stable and does not undergo major antigenic changes.


Asunto(s)
Antígenos Virales/análisis , Virus de la Encefalitis Transmitidos por Garrapatas/inmunología , Proteínas del Envoltorio Viral/análisis , Animales , Anticuerpos Monoclonales , Austria , Virus de la Encefalitis Transmitidos por Garrapatas/aislamiento & purificación , Variación Genética , Mapeo Peptídico , Garrapatas/microbiología
18.
J Biol Stand ; 14(2): 133-41, 1986 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-2428820

RESUMEN

On the basis of an epitope model, capture enzyme immunoassay systems using monoclonal antibodies have been devised for the detection and quantification of Tick-borne encephalitis virus and compared with a reference system employing polyclonal sera. Monoclonal antibodies were used both as capture and detector antibodies, their suitability depending primarily on their avidity and intrinsic background activity. A considerable increase in sensitivity was achieved by combining antibodies to different non-overlapping epitopes. Biotinylation of the detector antibodies allowed the construction of multiple site simultaneous binding assays. Furthermore the use of monoclonal antibodies of defined serological specificity made virus type identification possible. This assay can therefore be used as a rapid 'test of identity' as required during the manufacture of viral vaccines.


Asunto(s)
Anticuerpos Monoclonales , Antígenos Virales/análisis , Cisteína Endopeptidasas/sangre , Virus de la Encefalitis Transmitidos por Garrapatas/aislamiento & purificación , Epítopos , Técnicas para Inmunoenzimas , Complejos Multienzimáticos , Animales , Afinidad de Anticuerpos , Sitios de Unión de Anticuerpos , Virus de la Encefalitis Transmitidos por Garrapatas/inmunología , Cobayas , Inmunización , Ratones , Complejo de la Endopetidasa Proteasomal , Conejos , Vacunas Virales/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA