Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
J Virol ; 88(12): 6690-701, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24696472

RESUMEN

UNLABELLED: In previous work, a prototypic recombinant vesicular stomatitis virus Indiana serotype (rVSIV) vector expressing simian immunodeficiency virus (SIV) gag and human immunodeficiency virus type 1 (HIV-1) env antigens protected nonhuman primates (NHPs) from disease following challenge with an HIV-1/SIV recombinant (SHIV). However, when tested in a stringent NHP neurovirulence (NV) model, this vector was not adequately attenuated for clinical evaluation. For the work described here, the prototypic rVSIV vector was attenuated by combining specific G protein truncations with either N gene translocations or mutations (M33A and M51A) that ablate expression of subgenic M polypeptides, by incorporation of temperature-sensitive mutations in the N and L genes, and by deletion of the VSIV G gene to generate a replicon that is dependent on trans expression of G protein for in vitro propagation. When evaluated in a series of NHP NV studies, these attenuated rVSIV variants caused no clinical disease and demonstrated a very significant reduction in neuropathology compared to wild-type VSIV and the prototypic rVSIV vaccine vector. In spite of greatly increased in vivo attenuation, some of the rVSIV vectors elicited cell-mediated immune responses that were similar in magnitude to those induced by the much more virulent prototypic vector. These data demonstrate novel approaches to the rational attenuation of VSIV NV while retaining vector immunogenicity and have led to identification of an rVSIV N4CT1gag1 vaccine vector that has now successfully completed phase I clinical evaluation. IMPORTANCE: The work described in this article demonstrates a rational approach to the attenuation of vesicular stomatitis virus neurovirulence. The major attenuation strategy described here will be most likely applicable to other members of the Rhabdoviridae and possibly other families of nonsegmented negative-strand RNA viruses. These studies have also enabled the identification of an attenuated, replication-competent rVSIV vector that has successfully undergone its first clinical evaluation in humans. Therefore, these studies represent a major milestone in the development of attenuated rVSIV, and likely other vesiculoviruses, as a new vaccine platform(s) for use in humans.


Asunto(s)
Vacunas contra el SIDA/inmunología , Sistema Nervioso Central/virología , Vectores Genéticos/inmunología , Infecciones por VIH/inmunología , VIH-1/inmunología , Macaca fascicularis , Virus de la Estomatitis Vesicular Indiana/inmunología , Vacunas contra el SIDA/administración & dosificación , Vacunas contra el SIDA/genética , Animales , Anticuerpos Antivirales/inmunología , Sistema Nervioso Central/inmunología , Modelos Animales de Enfermedad , Vectores Genéticos/genética , Infecciones por VIH/prevención & control , Infecciones por VIH/virología , VIH-1/genética , Humanos , Macaca fascicularis/genética , Macaca fascicularis/inmunología , Macaca fascicularis/virología , Masculino , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Virus de la Estomatitis Vesicular Indiana/genética , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/administración & dosificación , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/inmunología
2.
J Virol ; 82(1): 207-19, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17942549

RESUMEN

Recombinant vesicular stomatitis virus (rVSV) has shown great potential as a new viral vector for vaccination. However, the prototypic rVSV vector described previously was found to be insufficiently attenuated for clinical evaluation when assessed for neurovirulence in nonhuman primates. Here, we describe the attenuation, neurovirulence, and immunogenicity of rVSV vectors expressing human immunodeficiency virus type 1 Gag. These rVSV vectors were attenuated by combinations of the following manipulations: N gene translocations (N4), G gene truncations (CT1 or CT9), noncytopathic M gene mutations (Mncp), and positioning of the gag gene into the first position of the viral genome (gag1). The resulting N4CT1-gag1, N4CT9-gag1, and MncpCT1-gag1 vectors demonstrated dramatically reduced neurovirulence in mice following direct intracranial inoculation. Surprisingly, in spite of a very high level of attenuation, the N4CT1-gag1 and N4CT9-gag1 vectors generated robust Gag-specific immune responses following intramuscular immunization that were equivalent to or greater than immune responses generated by the more virulent prototypic vectors. MncpCT1-gag1 also induced Gag-specific immune responses following intramuscular immunization that were equivalent to immune responses generated by the prototypic rVSV vector. Placement of the gag gene in the first position of the VSV genome was associated with increased in vitro expression of Gag protein, in vivo expression of Gag mRNA, and enhanced immunogenicity of the vector. These findings demonstrate that through directed manipulation of the rVSV genome, vectors that have reduced neurovirulence and enhanced immunogenicity can be made.


Asunto(s)
Vacunas contra el SIDA/inmunología , Vectores Genéticos , VIH-1/genética , Vesiculovirus/genética , Vacunas Virales/inmunología , Vacunas contra el SIDA/genética , Animales , Citocinas/biosíntesis , Anticuerpos Anti-VIH/sangre , Inyecciones Intramusculares , Dosificación Letal Mediana , Ratones , Ratones Endogámicos BALB C , Mutación Puntual , Recombinación Genética , Eliminación de Secuencia , Linfocitos T Citotóxicos/inmunología , Translocación Genética , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Vacunas Virales/genética , Virulencia , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/inmunología
3.
J Virol Methods ; 143(1): 55-64, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17382412

RESUMEN

Assessment of in vivo viral replication of live attenuated recombinant vesicular stomatitis virus (rVSV) vaccine vector candidates encoding HIV gag requires comprehensive preclinical safety studies, and development of sensitive assays to monitor the outcome of vaccination of animals is important. In this study, two 2-step quantitative real-time RT-PCR assays were developed; a singleplex assay to detect VSV genomic RNA from ferrets inoculated intra-cranially (IC) or intra-nasally (IN) with either a wild-type (wt) virus or an attenuated rVSV vector engineered to express HIV gag protein, and a duplex assay to simultaneously detect VSV-N and HIV-gag mRNAs from cynomolgus macaques inoculated intra-thalamically (IT) with the same viruses. Using synthetic oligonucleotides as standards, the lower limit of detection of VSV-N and HIV-gag was 50 copies. Results showed high levels of wt VSV(IN) genomic RNA and mRNA in ferret and macaque tissues, respectively, and significantly lower levels of VSV genomic RNA and VSV-N and HIV-gag mRNAs in tissues from animals inoculated with the attenuated rVSV vector. These assays correlated with both the course of infection for these animals, and the infectious viral load measured by a standard plaque assay, and could be used to determine the safety profile of rVSV vaccine vectors.


Asunto(s)
Vacunas contra el SIDA , Productos del Gen gag/aislamiento & purificación , VIH/genética , ARN Viral/aislamiento & purificación , Virus de la Estomatitis Vesicular Indiana/genética , Vacunas contra el SIDA/genética , Animales , Terapia Antirretroviral Altamente Activa , Hurones , Productos del Gen gag/genética , Vectores Genéticos , Macaca , ARN Viral/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sensibilidad y Especificidad , Virus de la Estomatitis Vesicular Indiana/aislamiento & purificación , Carga Viral , Replicación Viral
4.
Nat Biotechnol ; 21(8): 932-5, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12845329

RESUMEN

This report describes a transfection-independent system for packaging alphavirus replicon vectors using modified vaccinia virus Ankara (MVA) vectors to express all of the RNA components necessary for the production of Venezuelan equine encephalitis (VEE) virus replicon particles (VRP). Infection of mammalian cells with these recombinant MVA vectors resulted in robust expression of VEE structural genes, replication of the alphavirus vector and high titers of VRP. In addition, VRP packaging was achieved in a cell type (fetal rhesus lung) that has been approved for the manufacturing of vaccines destined for human use.


Asunto(s)
ADN Viral/administración & dosificación , ADN Viral/genética , Sistemas de Liberación de Medicamentos/métodos , Virus de la Encefalitis Equina Venezolana/genética , Técnicas de Transferencia de Gen , Vectores Genéticos , Replicón/genética , Transfección/métodos , Virus Vaccinia/genética , Virión/genética , Alphavirus/genética , Regulación Viral de la Expresión Génica
5.
Virus Res ; 115(1): 9-15, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16099066

RESUMEN

An intranasally delivered, live attenuated, temperature sensitive (ts) respiratory syncytial virus vaccine candidate, rA2cp248/404/1,030DeltaSH, exhibits a low level of genetic instability in clinical studies, in contrast to the relatively high stability of two similar candidates, cpts248/404 and rA2cp248/404DeltaSH. The latter strains, containing two ts mutations (248ts and 404ts), are partially growth restricted at 37 degrees C, whereas, rA2cp248/404/1,030DeltaSH contains an additional ts mutation (1,030ts) that increases attenuation and partially restricts virus growth at 35 degrees C. Since the maximum human airway temperature is 35.5 degrees C, we investigated whether growth restriction at 35 degrees C contributes to genetic instability of rA2cp248/404/1,030DeltaSH in vitro. We conducted in vitro passage studies with the three strains at 32 degrees C (a fully permissive growth temperature) and 35 degrees C (restrictive for only rA2cp248/404/1,030DeltaSH). Instability of the ts phenotype was observed only in rA2cp248/404/1,030DeltaSH passaged at 35 degrees C, and corresponded with reversion at the 248ts or 1,030ts mutation sites, as observed in clinical studies. This study indicates that ts mutations that partially restrict replication at physiologic temperatures may contribute to genetic instability of viruses in vivo. In vitro passage studies performed at appropriate temperatures can be used to assess genetic stability and to prioritize ts vaccine candidates for clinical evaluation.


Asunto(s)
Inestabilidad Genómica , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/genética , Vacunación , Vacunas Virales/genética , Administración Intranasal , Animales , Chlorocebus aethiops , Humanos , Mutación , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitiales Respiratorios/crecimiento & desarrollo , Pase Seriado , Temperatura , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Células Vero , Vacunas Virales/administración & dosificación
6.
J Virol Methods ; 135(1): 91-101, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16569439

RESUMEN

Recovery of recombinant, negative-strand, nonsegmented RNA viruses from a genomic cDNA clone requires a rescue system that promotes de novo assembly of a functional ribonucleoprotein (RNP) complex in the cell cytoplasm. This is accomplished typically by cotransfecting permissive cells with multiple plasmids that encode the positive-sense genomic RNA, the nucleocapsid protein (N or NP), and the two subunits of the viral RNA-dependent RNA polymerase (L and P). The transfected plasmids are transcribed in the cell cytoplasm by phage T7 RNA polymerase (T7 RNAP), which usually is supplied by infection with a recombinant vaccinia virus or through use of a stable cell line that expresses the polymerase. Although both methods of providing T7 RNAP are effective neither is ideal for viral vaccine development for a number of reasons. Therefore, it was necessary to modify existing technology to make it possible to routinely rescue a variety of recombinant viruses when T7 RNAP was provided by a cotransfected expression plasmid. Development of a broadly applicable procedure required optimization of the helper-virus-free methodology, which resulted in several modifications that improved rescue efficiency such as inclusion of plasmids encoding viral glycoproteins and matrix protein, heat shock treatment, and use of electroporation. The combined effect of these enhancements produced several important benefits including: (1) a helper-virus-free methodology capable of rescuing a diverse variety of paramyxoviruses and recombinant vesicular stomatitis virus (rVSV); (2) methodology that functioned effectively when using Vero cells, a suitable substrate for vaccine production; and (3) a method that enabled rescue of highly attenuated recombinant viruses, which had proven refractory to rescue using published procedures.


Asunto(s)
Paramyxovirinae/aislamiento & purificación , Vacunas Atenuadas , Virus de la Estomatitis Vesicular Indiana/aislamiento & purificación , Vacunas Virales/genética , Animales , Chlorocebus aethiops , ADN Recombinante , ADN Viral , ARN Polimerasas Dirigidas por ADN/genética , Virus Helper/genética , Mutación , Paramyxovirinae/genética , Plásmidos/genética , ARN Viral/metabolismo , Transfección , Vacunas Atenuadas/genética , Vacunas Sintéticas/genética , Células Vero , Virus de la Estomatitis Vesicular Indiana/genética , Proteínas Virales/genética , Replicación Viral/genética , Replicación Viral/fisiología
7.
AIDS Res Hum Retroviruses ; 21(7): 629-43, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16060834

RESUMEN

Of the various approaches being developed as prophylactic HIV vaccines, those based on a heterologous plasmid DNA prime, live vector boost vaccination regimen appear especially promising in the nonhuman primate/simian-human immunodeficiency virus (SHIV) challenge model. In this study, we sought to determine whether a series of intramuscular priming immunizations with a plasmid DNA vaccine expressing SIVgag p39, in combination with plasmid expressed rhesus IL-12, could effectively enhance the immunogenicity and postchallenge efficacy of two intranasal doses of recombinant vesicular stomatitis virus (rVSV)-based vectors expressing HIV-1 env 89.6P gp160 and SIVmac239 gag p55 in rhesus macaques. In macaques receiving the combination plasmid DNA prime, rVSV boost vaccination regimen we observed significantly increased SIVgag- specific cell-mediated and humoral immune responses and significantly lower viral loads postintravenous SHIV89.6P challenge relative to macaques receiving only the rVSV vectored immunizations. In addition, the plasmid DNA prime, rVSV boost vaccination regimen also tended to increase the preservation of peripheral blood CD4+ cells and reduce the morbidity and mortality associated with SHIV89.6P infection. An analysis of immune correlates of protection after SHIV89.6P challenge revealed that the prechallenge SHIV-specific IFN-gamma ELISpot response elicited by vaccination and the ability of the host to mount a virus-specific neutralizing antibody response postchallenge correlated with postchallenge clinical outcome. The correlation between vaccine-elicited cell-mediated immune responses and an improved clinical outcome after SHIV challenge provides strong justification for the continued development of a cytokine-enhanced plasmid DNA prime, rVSV vector boost immunization regimen for the prevention of HIV infection.


Asunto(s)
Vacunas contra el SIDA/inmunología , ADN/genética , Productos del Gen gag/genética , Interleucina-12/genética , Plásmidos , Virus de la Inmunodeficiencia de los Simios/genética , Virus de la Estomatitis Vesicular Indiana/genética , Animales , Secuencia de Bases , Cartilla de ADN , Ensayo de Inmunoadsorción Enzimática , Vectores Genéticos , Macaca mulatta , Pruebas de Neutralización , Recombinación Genética , Carga Viral
8.
Virus Res ; 83(1-2): 131-47, 2002 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-11864746

RESUMEN

A canine distemper virus (CDV) genomic cDNA clone and expression plasmids required to establish a CDV rescue system were generated from a laboratory-adapted strain of the Onderstepoort vaccine virus. In addition, a CDV minireplicon was prepared and used in transient expression studies performed to identify optimal virus rescue conditions. Results from the transient expression experiments indicated that minireplicon-encoded reporter gene activity was increased when transfected cell cultures were maintained at 32 rather than 37 degrees C, and when the cellular stress response was induced by heat shock. Applying these findings to rescue of recombinant CDV (rCDV) resulted in efficient recovery of virus after transfected HEp2 or A549 cells were co-cultured with Vero cell monolayers. Nucleotide sequence determination and analysis of restriction site polymorphisms confirmed that rescued virus was rCDV. A rCDV strain also was engineered that contained the luciferase gene inserted between the P and M genes; this virus directed high levels of luciferase expression in infected cells.


Asunto(s)
Virus del Moquillo Canino/genética , Expresión Génica , Vectores Genéticos/genética , Animales , Secuencia de Bases , Línea Celular , Chlorocebus aethiops , Clonación Molecular , Cricetinae , ADN Viral , ARN Polimerasas Dirigidas por ADN/genética , Virus del Moquillo Canino/aislamiento & purificación , Perros , Genes Reporteros , Humanos , Luciferasas/genética , Datos de Secuencia Molecular , Proteínas de la Nucleocápside/genética , Recombinación Genética , Replicón , Células Tumorales Cultivadas , Células Vero
9.
AIDS Res Hum Retroviruses ; 20(9): 989-1004, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15585086

RESUMEN

An experimental AIDS vaccine based on attenuated, recombinant vesicular stomatitis virus (rVSV), when administered by a combination of parenteral and mucosal routes, has proven effective at preventing AIDS in a rhesus macaque model (Rose NF, et al.: Cell 2001;106:539-549). In an effort to determine the optimal route of vaccine administration we evaluated the ability of rVSV-based vaccine vectors expressing HIV-1 Env and SIV Gag proteins, when given either intramuscularly (i.m.) or intranasally (i.n.), to elicit antigen-specific cellular and humoral immune responses, and to protect from a subsequent vaginal challenge with simian-human immunodeficiency virus (SHIV89.6P). Our results demonstrate that macaques vaccinated by the i.n. route developed significantly higher antigen-specific cellular immune responses as determined by MHC class I tetramer staining, IFN-gamma ELISPOT, and cytotoxic T cell assays. However, systemic and mucosal humoral immune responses did not vary significantly with the route of vaccine administration. Given the importance of cell-mediated immune responses in slowing AIDS progression, intranasal delivery of a VSV-based AIDS vaccine may be an optimal as well as practical route for vaccination and should be considered in design of clinical trials.


Asunto(s)
Vacunas contra el SIDA/inmunología , Productos del Gen env/inmunología , Productos del Gen gag/inmunología , Vectores Genéticos/inmunología , Vacunas Atenuadas/inmunología , Virus de la Estomatitis Vesicular Indiana/inmunología , Vacunas contra el SIDA/administración & dosificación , Administración Intranasal , Animales , Anticuerpos Antivirales/sangre , Femenino , Productos del Gen env/genética , Productos del Gen env/metabolismo , Productos del Gen gag/genética , Productos del Gen gag/metabolismo , Vectores Genéticos/administración & dosificación , Anticuerpos Anti-VIH/sangre , Infecciones por VIH/inmunología , Infecciones por VIH/prevención & control , VIH-1/inmunología , Humanos , Inyecciones Intramusculares , Interferón gamma/biosíntesis , Macaca mulatta , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Virus de la Inmunodeficiencia de los Simios/inmunología , Linfocitos T/inmunología , Vacunación , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Virus de la Estomatitis Vesicular Indiana/genética
10.
J Virol Methods ; 111(1): 29-36, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12821194

RESUMEN

A modified cDNA rescue system that improves recovery of recombinant nonsegmented, negative-strand RNA viruses from cloned DNAs is described. Rescue systems based on vaccinia virus-T7 RNA polymerase vectors have been used to derive many negative-strand viruses; however, some strains can be recalcitrant to rescue possibly because of the simultaneous replication of the vaccinia virus-T7 vector. Our goal was to engineer a system where replication of the vaccinia virus-T7 vector could be blocked, yet allow for sufficient T7 RNA polymerase expression to enable genetic rescue. To that end, a recombinant modified vaccinia virus Ankara (MVA) was engineered that contained the bacteriophage T7 gene-1 under the control of a strong early promoter that would enable T7 RNA polymerase expression in the absence of MVA DNA replication. The new T7 helper, MVAGKT7, was then utilized successfully for the genetic rescue of a measles virus minigenome and full-length cDNAs, in the presence of DNA synthesis inhibitors. In addition to blocking completely MVAGKT7 replication, AraC treatment was found to enhance minigenome-encoded gene expression and the efficiency of measles virus rescue.


Asunto(s)
Bacteriófago T7/genética , ADN Viral/biosíntesis , ARN Polimerasas Dirigidas por ADN/genética , Vectores Genéticos , Genoma Viral , Virus Vaccinia/metabolismo , Animales , Línea Celular , Replicación del ADN/efectos de los fármacos , ADN Complementario/genética , Genes Virales , Virus del Sarampión/genética , Virus del Sarampión/fisiología , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Regiones Promotoras Genéticas , Recombinación Genética , Transfección/métodos , Virus Vaccinia/genética , Proteínas Virales , Replicación Viral
11.
J Virol Methods ; 164(1-2): 43-50, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19941901

RESUMEN

Propagation-defective vesicular stomatitis virus (VSV) vectors that encode a truncated G protein (VSV-Gstem) or lack the G gene entirely (VSV-DeltaG) are attractive vaccine vectors because they are immunogenic, cannot replicate and spread after vaccination, and do not express many of the epitopes that elicit neutralizing anti-VSV immunity. To consider advancing non-propagating VSV vectors towards clinical assessment, scalable technology that is compliant with human vaccine manufacturing must be developed to produce clinical trial material. Accordingly, two propagation methods were developed for VSV-Gstem and VSV-DeltaG vectors encoding HIV gag that have the potential to support large-scale production. One method is based on transient expression of G protein after electroporating plasmid DNA into Vero cells and the second is based on a stable Vero cell line that contains a G gene controlled by a heat shock-inducible transcription unit. Both methods reproducibly supported production of 1 x 10(7) to 1 x 10(8) infectious units (I.U.s) of vaccine vector per milliliter. Results from these studies also showed that optimization of the G gene is necessary for abundant G protein expression from electroporated plasmid DNA or from DNA integrated in the genome of a stable cell line, and that the titers of VSV-Gstem vectors generally exceeded VSV-DeltaG.


Asunto(s)
Vectores Genéticos , Glicoproteínas de Membrana/deficiencia , Vesiculovirus/crecimiento & desarrollo , Vesiculovirus/genética , Proteínas del Envoltorio Viral/deficiencia , Animales , Chlorocebus aethiops , Glicoproteínas de Membrana/biosíntesis , Datos de Secuencia Molecular , ARN Viral/genética , Análisis de Secuencia de ADN , Células Vero , Proteínas del Envoltorio Viral/biosíntesis , Cultivo de Virus/métodos , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/genética
12.
Vaccine ; 27(22): 2930-9, 2009 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-19428903

RESUMEN

Recombinant vesicular stomatitis viruses (rVSVs) are being developed as potential HIV-1 vaccine candidates. To characterize the in vivo replication and dissemination of rVSV vectors in mice, high doses of a highly attenuated vector expressing HIV-1 Gag, rVSV(IN)-N4CT9-Gag1, and a prototypic reference virus, rVSV(IN)-HIVGag5, were delivered intramuscularly (IM), intranasally (IN), or intravenously (IV). We used quantitative, real-time RT-PCR (Q-PCR) and standard plaque assays to measure the temporal dissemination of these viruses to various tissues. Following IM inoculation, both viruses were detected primarily at the injection site as well as in draining lymph nodes; neither virus induced significant weight loss, pathologic signs, or evidence of neuroinvasion. In contrast, following IN inoculation, the prototypic virus was detected in all tissues tested and caused significant weight loss leading to death. IN administration of rVSV(IN)-N4CT9-Gag1 resulted in detection in numerous tissues (brain, lung, nasal turbinates, and lymph nodes) albeit in significantly reduced levels, which caused little or no weight loss nor any mortality. Following IV inoculation, both prototypic and attenuated viruses were detected by Q-PCR in all tissues tested. In contrast to the prototype, rVSV(IN)-N4CT9-Gag1 viral loads were significantly lower in all organs tested, and no infectious virus was detected in the brain following IV inoculation, despite the presence of viral RNA. These studies demonstrated significant differences in the biodistribution patterns of and the associated pathogenicity engendered by the prototypic and attenuated vectors in a highly susceptible host.


Asunto(s)
Vacunas contra el SIDA/administración & dosificación , Vacunas contra el SIDA/farmacocinética , Vectores Genéticos , Vesiculovirus/crecimiento & desarrollo , Vesiculovirus/genética , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/genética , Vacunas contra el SIDA/efectos adversos , Administración Intranasal , Animales , Femenino , Inyecciones Intramusculares , Inyecciones Intravenosas , Ratones , Ratones Endogámicos BALB C , ARN Viral/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/efectos adversos , Vacunas Atenuadas/farmacocinética , Ensayo de Placa Viral
13.
J Virol ; 81(4): 2056-64, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17151112

RESUMEN

A variety of rational approaches to attenuate growth and virulence of vesicular stomatitis virus (VSV) have been described previously. These include gene shuffling, truncation of the cytoplasmic tail of the G protein, and generation of noncytopathic M gene mutants. When separately introduced into recombinant VSV (rVSV), these mutations gave rise to viruses distinguished from their "wild-type" progenitor by diminished reproductive capacity in cell culture and/or reduced cytopathology and decreased pathogenicity in vivo. However, histopathology data from an exploratory nonhuman primate neurovirulence study indicated that some of these attenuated viruses could still cause significant levels of neurological injury. In this study, additional attenuated rVSV variants were generated by combination of the above-named three distinct classes of mutation. The resulting combination mutants were characterized by plaque size and growth kinetics in cell culture, and virulence was assessed by determination of the intracranial (IC) 50% lethal dose (LD(50)) in mice. Compared to virus having only one type of attenuating mutation, all of the mutation combinations examined gave rise to virus with smaller plaque phenotypes, delayed growth kinetics, and 10- to 500-fold-lower peak titers in cell culture. A similar pattern of attenuation was also observed following IC inoculation of mice, where differences in LD(50) of many orders of magnitude between viruses containing one and two types of attenuating mutation were sometimes seen. The results show synergistic rather than cumulative increases in attenuation and demonstrate a new approach to the attenuation of VSV and possibly other viruses.


Asunto(s)
Glicoproteínas de Membrana/genética , Infecciones por Rhabdoviridae/virología , Virus de la Estomatitis Vesicular Indiana/genética , Proteínas del Envoltorio Viral/genética , Animales , Línea Celular , Chlorocebus aethiops , Cricetinae , Femenino , Eliminación de Gen , Genes Virales/genética , Ratones , Proteínas de la Nucleocápside/genética , Células Vero , Virus de la Estomatitis Vesicular Indiana/crecimiento & desarrollo , Virus de la Estomatitis Vesicular Indiana/patogenicidad , Proteínas de la Matriz Viral/genética , Virulencia , Replicación Viral
14.
Virology ; 360(1): 36-49, 2007 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-17098273

RESUMEN

Although vesicular stomatitis virus (VSV) neurovirulence and pathogenicity in rodents have been well studied, little is known about VSV pathogenicity in non-human primates. To address this question, we measured VSV viremia, shedding, and neurovirulence in macaques. Following intranasal inoculation, macaques shed minimal recombinant VSV (rVSV) in nasal washes for 1 day post-inoculation; viremia was not detected. Following intranasal inoculation of macaques, wild type (wt) VSV, rVSV, and two rVSV-HIV vectors showed no evidence of spread to CNS tissues. However, macaques inoculated intrathalamically with wt VSV developed severe neurological disease. One of four macaques receiving rVSV developed clinical and histological signs similar to the wt group, while the remaining three macaques in this group and all of the macaques in the rVSV-HIV vector groups showed no clinical signs of disease and reduced severity of histopathology compared to the wt group. The implications of these findings for rVSV vaccine development are discussed.


Asunto(s)
Enfermedades del Sistema Nervioso Central/virología , Vectores Genéticos , Enfermedades de los Monos/virología , Infecciones por Rhabdoviridae/virología , Virus de la Estomatitis Vesicular Indiana , Vacunas contra el SIDA/administración & dosificación , Vacunas contra el SIDA/genética , Animales , Encéfalo/patología , Encéfalo/virología , Enfermedades del Sistema Nervioso Central/patología , Vectores Genéticos/administración & dosificación , Vectores Genéticos/fisiología , Inflamación/patología , Macaca mulatta , Masculino , Enfermedades de los Monos/patología , Mucosa Nasal/virología , Recombinación Genética , Infecciones por Rhabdoviridae/patología , Médula Espinal/patología , Virus de la Estomatitis Vesicular Indiana/patogenicidad , Virus de la Estomatitis Vesicular Indiana/fisiología , Viremia , Virulencia , Replicación Viral
15.
Vaccine ; 25(12): 2296-305, 2007 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-17239997

RESUMEN

The safety of a propagation-defective Venezuelan equine encephalitis virus (VEEV) replicon particle vaccine was examined in mice. After intracranial inoculation we observed approximately 5% body weight loss, modest inflammatory changes in the brain, genome replication, and foreign gene expression. These changes were transient and significantly less severe than those caused by TC-83, a live-attenuated vaccinal strain of VEEV that has been safely used to immunize military personnel and laboratory workers. Replicon particles injected intramuscularly or intravenously were detected at limited sites 3 days post-administration, and were undetectable by day 22. There was no evidence of dissemination to spinal cord or brain after systemic administration. These results demonstrate that propagation-defective VEEV replicon particles are minimally neurovirulent and lack neuroinvasive potential.


Asunto(s)
Virus de la Encefalitis Equina Venezolana/inmunología , Encefalomielitis Equina Venezolana/inmunología , Replicón/genética , Proteínas del Envoltorio Viral/genética , Animales , Peso Corporal , Encéfalo/metabolismo , Encéfalo/virología , Virus de la Encefalitis Equina Venezolana/genética , Encefalomielitis Equina Venezolana/prevención & control , Inyecciones Intramusculares , Inyecciones Intravenosas , Ratones , ARN Viral/genética , ARN Viral/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Médula Espinal/metabolismo , Médula Espinal/virología , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Replicación Viral/genética
16.
Vaccine ; 25(41): 7132-44, 2007 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-17850933

RESUMEN

Respiratory syncytial virus (RSV) is a major cause of acute respiratory tract disease in humans. Towards development of a prophylactic vaccine, we genetically engineered Venezuelan equine encephalitis virus (VEEV) replicons encoding the fusion (Fa) or attachment (Ga or Gb) proteins of the A or B subgroups of RSV. Intramuscular immunization with a formulation composed of equal amounts of each replicon particle (3vRSV replicon vaccine) generated serum neutralizing antibodies against A and B strains of RSV in BALB/c mice and rhesus macaques. When contrasted with purified natural protein or formalin-inactivated RSV formulated with alum, the 3vRSV replicon vaccine induced balanced Th1/Th2 T cell responses in mice. This was evident in the increased number of RSV-specific IFN-gamma(+) splenocytes following F or G peptide stimulation, diminished quantity of eosinophils and type 2 T cell cytokines in the lungs after challenge, and increased in vivo lysis of RSV peptide-loaded target cells. The immune responses in mice were also protective against intranasal challenge with RSV. Thus, the replicon-based platform represents a promising new strategy for vaccines against RSV.


Asunto(s)
Virus de la Encefalitis Equina Venezolana/inmunología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Vacunas Sintéticas/inmunología , Proteínas Estructurales Virales/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales/sangre , Citocinas/biosíntesis , Virus de la Encefalitis Equina Venezolana/genética , Femenino , Inyecciones Intramusculares , Pulmón/inmunología , Pulmón/virología , Macaca mulatta , Masculino , Ratones , Ratones Endogámicos BALB C , Pruebas de Neutralización , Infecciones por Virus Sincitial Respiratorio/inmunología , Vacunas contra Virus Sincitial Respiratorio/genética , Virus Sincitiales Respiratorios/genética , Bazo/inmunología , Linfocitos T/inmunología , Vacunas Sintéticas/genética , Proteínas Estructurales Virales/genética , Vacunas Virales/genética
17.
Virology ; 367(2): 275-87, 2007 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-17599381

RESUMEN

Widespread use of a live-attenuated influenza vaccine (LAIV) in the United States (licensed as FluMist) raises the possibility that vaccine viruses will contribute gene segments to the type A influenza virus gene pool. Progeny viruses possessing new genotypes might arise from genetic reassortment between circulating wild-type (wt) and vaccine strains, but it will be difficult to predict whether they will be viable or exhibit novel properties. To begin addressing these uncertainties, reverse-genetics was used to generate 34 reassortant viruses derived from wt influenza virus A/Sydney/5/97 and the corresponding live vaccine strain. The reassortants contained different combinations of vaccine and wt PB2, PB1, PA, NP, M, and NS gene segments whereas all strains encoded wt HA and NA glycoproteins. The phenotypes of the reassortant strains were compared to wt and vaccine viruses by evaluating temperature-sensitive (ts) plaque formation and replication attenuation (att) in ferrets following intranasal inoculation. The results demonstrated that the vaccine virus PB1, PB2, and NP gene segments were dominant when introduced into the wt A/Sydney/5/97 genetic background, producing recombinant viruses that expressed the ts and att phenotypes. A dominant attenuated phenotype also was evident when reassortant strains contained the vaccine M or PA gene segments, even though these polypeptides are not temperature-sensitive. Although the vaccine M and NS gene segments typically are not associated with temperature sensitivity, a number of reassortants containing these vaccine gene segments did exhibit a more restricted ts phenotype. Overall, no reassortant strains were more virulent than wt, and in fact, 33 of the 34 recombinant viruses replicated less efficiently in infected ferrets. These results suggest that genetic reassortment between wt and vaccine strains is unlikely to produce viruses having novel properties that differ substantially from either progenitor, and that the likely outcome of reassortment will be attenuated viruses.


Asunto(s)
Genes Virales , Virus de la Influenza A/genética , Vacunas contra la Influenza/biosíntesis , Virus Reordenados/genética , Recombinación Genética , Vacunas Atenuadas/genética , Proteínas Virales/genética , Animales , Hurones , Ingeniería Genética , Genotipo , Virus de la Influenza A/inmunología , Virus de la Influenza A/fisiología , Fenotipo , ARN Polimerasa Dependiente del ARN/metabolismo , Virus Reordenados/fisiología , Temperatura , Células Tumorales Cultivadas , Vacunas Atenuadas/química , Vacunas Atenuadas/inmunología , Proteínas del Núcleo Viral/química , Proteínas del Núcleo Viral/genética , Proteínas del Núcleo Viral/metabolismo , Ensayo de Placa Viral , Proteínas Virales/química , Proteínas Virales/metabolismo , Replicación Viral
18.
Virology ; 348(1): 107-19, 2006 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-16445957

RESUMEN

Measles virus V protein is a Cys-rich polypeptide that is dispensable for virus propagation in continuous cell lines, but necessary for efficient viral replication in animals. Those functions modulating virus propagation in vivo are not understood completely, although V protein is known to interfere with the host interferon response and control of viral gene expression. The ability to modulate gene expression was investigated further with a minireplicon transient expression system in which V protein was found to repress reporter activity. Two regions of the polypeptide contributed to this repressive effect including the carboxy-terminus and a region conserved in morbillivirus V proteins located between amino acids 110-131, whereas domains known to mediate the interaction between V and the nucleocapsid (N) protein were not essential. Accumulation of encapsidated minigenome in transfected cells was inhibited by V protein suggesting that it acted as a repressor of genome replication thereby limiting availability of template for reporter gene mRNA transcription.


Asunto(s)
Cloranfenicol O-Acetiltransferasa/análisis , Regulación Viral de la Expresión Génica , Virus del Sarampión/fisiología , Fosfoproteínas/fisiología , Proteínas de Unión al ARN/fisiología , Proteínas Virales/fisiología , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Fusión Artificial Génica , Línea Celular , Cloranfenicol O-Acetiltransferasa/genética , Secuencia Conservada , Análisis Mutacional de ADN , Genes Reporteros , Genoma Viral/genética , Humanos , Datos de Secuencia Molecular , Mutación Missense , Fosfoproteínas/genética , Estructura Terciaria de Proteína/genética , Estructura Terciaria de Proteína/fisiología , ARN Viral/análisis , Proteínas de Unión al ARN/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Proteínas Virales/genética , Replicación Viral
19.
Virology ; 348(1): 96-106, 2006 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-16442140

RESUMEN

Measles virus V protein represses genome replication through a poorly understood mechanism, which led us to investigate whether V protein might be an RNA-binding modulatory factor. Recombinant V protein, expressed from transfected HEp-2 cells or E. coli, formed protein-RNA complexes with poly-guanosine (poly-G) or poly-U linked to agarose beads. RNA binding was not exclusive to ribonucleotide homopolymers as complex formation between V protein and an RNA molecule equivalent to the 3' terminal 107 bases of the measles virus genome was observed with an electrophoretic mobility shift assay (EMSA). The interaction with poly-G was used to further examine the RNA binding properties of V demonstrating that protein-RNA complex formation was dependent upon the unique Cys-rich carboxy terminus, a region also required to induce maximal repression of minireplicon-encoded reporter gene expression in transient assays. Surprisingly, two mutant proteins that contained Cys-to-Ala substitutions in the C-terminus were found to retain their ability to bind poly-G binding and repress minireplicon reporter gene expression indicating that neither activity was dependent on the integrity of all 7 C-terminal Cys residues. Additional genetic analysis revealed that amino acids 238-266 were necessary for efficient RNA binding and overlapped with residues (238-278) required for maximal repression induced by the C-terminal domain. In addition, a 10 amino acid deletion was identified (residues 238-247) that blocked RNA binding and repression indicating that these two activities were related.


Asunto(s)
Virus del Sarampión/fisiología , Fosfoproteínas/fisiología , Proteínas de Unión al ARN/metabolismo , ARN/metabolismo , Proteínas Virales/fisiología , Replicación Viral , Sustitución de Aminoácidos , Fusión Artificial Génica , Línea Celular , Cisteína/genética , Cisteína/fisiología , Ensayo de Cambio de Movilidad Electroforética , Expresión Génica , Genes Reporteros , Humanos , Luciferasas/análisis , Luciferasas/genética , Mutación Missense , Fosfoproteínas/genética , Unión Proteica , Estructura Terciaria de Proteína/genética , Estructura Terciaria de Proteína/fisiología , ARN Viral/metabolismo , Proteínas de Unión al ARN/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Eliminación de Secuencia , Proteínas Virales/genética
20.
Springer Semin Immunopathol ; 28(3): 239-53, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16977404

RESUMEN

Recombinant vesicular stomatitis virus (rVSV) is currently under evaluation as a human immunodeficiency virus (HIV)-1 vaccine vector. The most compelling reasons to develop rVSV as a vaccine vector include a very low seroprevalence in humans, the ability to infect and robustly express foreign antigens in a broad range of cells, and vigorous growth in continuous cell lines used for vaccine manufacture. Numerous preclinical studies with rVSV vectors expressing antigens from a variety of human pathogens have demonstrated the versatility, flexibility, and potential efficacy of the rVSV vaccine platform. When administered to nonhuman primates (NHPs), rVSV vectors expressing HIV-1 Gag and Env elicited robust HIV-1-specific cellular and humoral immune responses, and animals immunized with rVSV vectors expressing simian immunodeficiency virus (SIV) Gag and HIV Env were protected from AIDS after challenge with a pathogenic SIV/HIV recombinant. However, results from an exploratory neurovirulence study in NHPs indicated that these prototypic rVSV vectors might not be adequately attenuated for widespread use in human populations. To address this safety concern, a variety of different attenuation strategies, designed to produce a range of further attenuated rVSV vectors, are currently under investigation. Additional modifications of further attenuated rVSV vectors to upregulate expression of HIV-1 antigens and coexpress molecular adjuvants are also being developed in an effort to balance immunogenicity and attenuation.


Asunto(s)
Vacunas contra el SIDA , Vectores Genéticos , Antígenos VIH/inmunología , Infecciones por VIH/prevención & control , VIH-1/genética , Virus de la Estomatitis Vesicular Indiana/genética , Vacunas contra el SIDA/genética , Vacunas contra el SIDA/inmunología , Vacunas contra el SIDA/uso terapéutico , Adyuvantes Inmunológicos , Animales , Terapia Antirretroviral Altamente Activa , Productos del Gen env/genética , Productos del Gen env/inmunología , Productos del Gen gag/genética , Productos del Gen gag/inmunología , Antígenos VIH/genética , VIH-1/inmunología , Humanos , Factores Inmunológicos/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Vacunas Sintéticas , Virus de la Estomatitis Vesicular Indiana/inmunología , Virus de la Estomatitis Vesicular Indiana/patogenicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA