Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Gen Virol ; 102(3)2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33729125

RESUMEN

Multiple strains of human cytomegalovirus (HCMV) can cause congenital cytomegalovirus (cCMV) by primary or secondary infection. The viral gB glycoprotein is a leading vaccine candidate, essential for infection of all cell-types, and immunodominant antibody target. Guinea pig cytomegalovirus (GPCMV) is the only small animal model for cCMV. Various gB vaccines have shown efficacy but studies have utilized truncated gB and protection against prototype strain 22122 with preferential tropism to fibroblasts despite encoding a gH-based pentamer complex for non-fibroblast infection. A highly cell-associated novel strain of GPCMV (TAMYC) with 99 % identity in gB sequence to 22122 exhibited preferred tropism to epithelial cells. An adenovirus vaccine encoding full-length gB (AdgB) was highly immunogenic and partially protected against 22122 strain challenge in vaccinated animals but not when challenged with TAMYC strain. GPCMV studies with AdgB vaccine sera on numerous cell-types demonstrated impaired neutralization (NA50) compared to fibroblasts. GPCMV-convalescent sera including pentamer complex antibodies increased virus neutralization on non-fibroblasts and anti-gB depletion from GPCMV-convalescent sera had minimal impact on epithelial cell neutralization. GPCMV(PC+) 22122-convalescent animals challenged with TAMYC exhibited higher protection compared to AdgB vaccine. Overall, results suggest that antibody response to both gB and PC are important components of a GPCMV vaccine.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Infecciones por Citomegalovirus/inmunología , Vacunas contra Citomegalovirus/inmunología , Roseolovirus/patogenicidad , Animales , Infecciones por Citomegalovirus/prevención & control , Cobayas , Pruebas de Neutralización , Reacción en Cadena en Tiempo Real de la Polimerasa , Roseolovirus/fisiología , Proteínas del Envoltorio Viral/inmunología , Carga Viral
2.
Viruses ; 11(12)2019 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-31801268

RESUMEN

Viruses of the genus Roseolovirus belong to the subfamily Betaherpesvirinae, family Herpesviridae. Roseoloviruses have been studied in humans, mice and pigs, but they are likely also present in other species. This is the first comparative analysis of roseoloviruses in humans and animals. The human roseoloviruses human herpesvirus 6A (HHV-6A), 6B (HHV-6B), and 7 (HHV-7) are relatively well characterized. In contrast, little is known about the murine roseolovirus (MRV), also known as murine thymic virus (MTV) or murine thymic lymphotrophic virus (MTLV), and the porcine roseolovirus (PRV), initially incorrectly named porcine cytomegalovirus (PCMV). Human roseoloviruses have gained attention because they can cause severe diseases including encephalitis in immunocompromised transplant and AIDS patients and febrile seizures in infants. They have been linked to a number of neurological diseases in the immunocompetent including multiple sclerosis (MS) and Alzheimer's. However, to prove the causality in the latter disease associations is challenging due to the high prevalence of these viruses in the human population. PCMV/PRV has attracted attention because it may be transmitted and pose a risk in xenotransplantation, e.g., the transplantation of pig organs into humans. Most importantly, all roseoloviruses are immunosuppressive, the humoral and cellular immune responses against these viruses are not well studied and vaccines as well as effective antivirals are not available.


Asunto(s)
Genoma Viral/genética , Infecciones por Roseolovirus/virología , Roseolovirus/fisiología , Animales , Antivirales/uso terapéutico , Humanos , Ratones , Roseolovirus/genética , Roseolovirus/inmunología , Roseolovirus/patogenicidad , Infecciones por Roseolovirus/tratamiento farmacológico , Infecciones por Roseolovirus/epidemiología , Infecciones por Roseolovirus/transmisión , Porcinos , Integración Viral , Latencia del Virus
3.
J Virol ; 90(17): 7715-27, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27307567

RESUMEN

UNLABELLED: Guinea pig cytomegalovirus (GPCMV) provides a valuable model for congenital cytomegalovirus transmission. Salivary gland (SG)-passaged stocks of GPCMV are pathogenic, while tissue culture (TC) passage in fibroblasts results in attenuation. Nonpathogenic TC-derived virus N13R10 (cloned as a bacterial artificial chromosome [BAC]) has a 4-bp deletion that disrupts GP129, which encodes a subunit of the GPCMV pentameric complex (PC) believed to govern viral entry into select cell types, and GP130, an overlapping open reading frame (ORF) of unknown function. To determine if this deletion contributes to attenuation of N13R10, markerless gene transfer in Escherichia coli was used to construct virus r129, a variant of N13R10 in which the 4-bp deletion is repaired. Virions from r129 were found to contain GP129 as well as two other PC subunit proteins, GP131 and GP133, whereas these three PC subunits were absent from N13R10 virions. Replication of r129 in fibroblasts appeared unaltered compared to that of N13R10. However, following experimental challenge of immunocompromised guinea pigs, r129 induced significant weight loss, longer duration of viremia, and dramatically higher (up to 1.5 × 10(6)-fold) viral loads in blood and end organs compared to N13R10. In pregnant guinea pigs, challenge with doses of r129 virus of ≥5 × 10(6) PFU resulted in levels of maternal viremia, congenital transmission, pup viral loads, intrauterine growth restriction, and pup mortality comparable to that induced by pathogenic SG virus, although higher doses of r129 were required. These results suggest that the GP129-GP130 mutation is a significant contributor to attenuation of N13R10, likely by abrogating expression of a functional PC. IMPORTANCE: Tissue culture adaptation of cytomegaloviruses rapidly selects for mutations, deletions, and rearrangements in the genome, particularly for viruses passaged in fibroblast cells. Some of these mutations are focused in the region of the genome encoding components of the pentameric complex (PC), in particular homologs of human cytomegalovirus (HCMV) proteins UL128, UL130, and UL131A. These mutations can attenuate the course of infection when the virus is reintroduced into animals for vaccine and pathogenesis studies. This study demonstrates that a deletion that arose during the process of tissue culture passage can be repaired, with subsequent restoration of pathogenicity, using BAC-based mutagenesis. Restoration of pathogenicity by repair of a frameshift mutation in GPCMV gene GP129 using this approach provides a valuable genetic platform for future studies using the guinea pig model of congenital CMV infection.


Asunto(s)
Infecciones por Citomegalovirus/congénito , Infecciones por Citomegalovirus/patología , Fibroblastos/virología , Mutación , Multimerización de Proteína , Roseolovirus/genética , Roseolovirus/patogenicidad , Animales , Peso Corporal , Cromosomas Artificiales Bacterianos , Infecciones por Citomegalovirus/virología , Modelos Animales de Enfermedad , Escherichia coli/genética , Glicoproteínas/genética , Cobayas , Roseolovirus/crecimiento & desarrollo , Eliminación de Secuencia , Pase Seriado , Carga Viral , Proteínas Estructurales Virales/genética , Viremia , Virulencia , Factores de Virulencia/genética
5.
J Gen Virol ; 95(Pt 6): 1376-1382, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24659103

RESUMEN

The GP129, GP131 and GP133 genes of guinea pig cytomegalovirus (GPCMV) are homologues of human cytomegalovirus UL128, UL130 and UL131A, respectively, which are essential for infection of endothelial and epithelial cells, and for viral transmission to leukocytes. Our previous study demonstrated that a GPCMV strain lacking the 1.6 kb locus that contains the GP129, GP131 and GP133 genes had a growth defect in animals. Here, we demonstrated that the WT strain, but not the 1.6 kb-deleted strain, formed capsids in macrophages prepared from the peritoneal fluid. To understand the mechanism, we prepared GPCMV strains defective in each of GP129, GP131 and GP133, and found that they were all essential for the infection of peritoneal, splenic and PBMC-derived macrophages/monocytes, and for expression of immediate-early antigens in the macrophages/monocytes, although they were dispensable for infection of fibroblasts. Monocyte/macrophage tropism could be one of the important determinants for viral dissemination in vivo.


Asunto(s)
Citomegalovirus/patogenicidad , Macrófagos Peritoneales/virología , Monocitos/virología , Roseolovirus/patogenicidad , Proteínas Virales/fisiología , Animales , Citomegalovirus/genética , Citomegalovirus/fisiología , Eliminación de Gen , Genes Inmediatos-Precoces , Genes Virales , Cobayas , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiología , Roseolovirus/genética , Roseolovirus/fisiología , Especificidad de la Especie , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/fisiología , Proteínas Virales/genética , Virulencia/genética , Virulencia/fisiología
6.
Vaccine ; 31(31): 3199-205, 2013 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-23684839

RESUMEN

BACKGROUND: Cytomegalovirus (CMV) is the most common cause of congenital virus infection. Infection of guinea pigs with guinea pig CMV (GPCMV) can provide a useful model for the analysis of its pathogenesis as well as for the evaluation of vaccines. Although glycoprotein B (gB) vaccines have been reported to reduce the incidence and mortality of congenital infection in human clinical trials and guinea pig animal models, the mechanisms of protection remain unclear. METHODS: To understand the gB vaccine protection mechanisms, we analyzed the spread of challenged viruses in the placentas and fetuses of guinea pig dams immunized with recombinant adenoviruses expressing GPCMV gB and ß-galactosidase, rAd-gB and rAd-LacZ, respectively. RESULTS: Mean body weight of the fetuses in the dams immunized with rAd-LacZ followed by GPCMV challenge 3 weeks after immunization was 78% of that observed for dams immunized with rAd-gB. Under conditions in which congenital infection occurred in 75% of fetuses in rAd-LacZ-immunized dams, only 13% of fetuses in rAd-gB-immunized dams were congenitally infected. The placentas were infected less frequently in the gB-immunized animals. In the placentas of the rAd-LacZ- and rAd-gB-immunized animals, CMV early antigens were detected mainly in the spongiotrophoblast layer. Focal localization of viral antigens in the spongiotrophoblast layer suggests cell-to-cell viral spread in the placenta. In spite of a similar level of antibodies against gB and avidity indices among fetuses in each gB-immunized dam, congenital infection was sometimes observed in a littermate fetus. In such infected fetuses, CMV spread to most organs. CONCLUSIONS: Our results suggest that antibodies against gB protected against infection mainly at the interface of the placenta rather than from the placenta to the fetus. The development of strategies to block cell-to-cell viral spread in the placenta is, therefore, required for effective protection against congenital CMV infection.


Asunto(s)
Infecciones por Citomegalovirus/prevención & control , Placenta/virología , Roseolovirus/patogenicidad , Proteínas del Envoltorio Viral/inmunología , Vacunas Virales/inmunología , Adenoviridae , Animales , Anticuerpos Antivirales/sangre , Infecciones por Citomegalovirus/congénito , Infecciones por Citomegalovirus/inmunología , ADN Viral/aislamiento & purificación , Femenino , Feto/inmunología , Cobayas , Placenta/inmunología , Embarazo , beta-Galactosidasa/inmunología
7.
Virology ; 379(1): 45-54, 2008 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-18656220

RESUMEN

Guinea pig cytomegalovirus (GPCMV) provides a useful model for studies of congenital CMV infection. During characterization of the GPCMV genome sequence, we identified two types of strains in a virus stock purchased from ATCC. One of them, GPCMV/del, lacks a 1.6 kb locus that positionally corresponds to murine CMV (MCMV) M129-M133. Growth of GPCMV/del in cell culture was marginally better than that of the other strain, GPCMV/full, which harbors the 1.6 kb locus. However, in animals infected intraperitoneally with virus stocks containing both strains, GPCMV/full disseminated more efficiently than GPCMV/del, including 200-fold greater viral load in salivary glands. Viral DNA, transcripts of the immediate-early 2 gene homolog, and viral antigens were more abundant in animals infected with GPCMV/full than in those infected with GPCMV/del. Although the observed phenomena have some similarity with the growth properties of MCMV strains defective in mck-1/mck-2(M129/131) and those defective in sgg(M132), no M129-M132 homologs were found in the 1.6 kb locus. Since one of the ORFs in the locus has a weak sequence similarity with HCMV UL130, which relates to cell tropism, further studies will be required to learn the mechanism for efficient GPCMV growth in animal.


Asunto(s)
Roseolovirus/crecimiento & desarrollo , Roseolovirus/patogenicidad , Eliminación de Secuencia , Replicación Viral , Animales , Antígenos Virales/biosíntesis , Técnicas de Cultivo de Célula , Línea Celular , ADN Viral/biosíntesis , ADN Viral/química , ADN Viral/genética , Genoma Viral , Cobayas , Hígado/patología , Datos de Secuencia Molecular , Filogenia , ARN Viral/biosíntesis , Roseolovirus/genética , Infecciones por Roseolovirus/virología , Glándulas Salivales/virología , Análisis de Secuencia de ADN , Homología de Secuencia , Bazo/patología , Ensayo de Placa Viral
8.
J Virol ; 78(18): 9872-89, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15331722

RESUMEN

We recently identified the genes encoding the guinea pig cytomegalovirus (GPCMV) homologs of the upper and lower matrix proteins of human CMV, pp71 (UL82) and pp65 (UL83), which we designated GP82 and GP83, respectively. Transient-expression studies with a GP82 plasmid demonstrated that the encoded protein targets the nucleus and that the infectivity and plaquing efficiency of cotransfected GPCMV viral DNA was enhanced by GP82. The transactivation function of GP82 was not limited to GPCMV, but was also observed for a heterologous virus, herpes simplex virus type 1 (HSV-1). This was confirmed by its ability to complement the growth of an HSV-1 VP16 transactivation-defective mutant virus in an HSV viral DNA cotransfection assay. Study of a GP82 "knockout" virus (and its attendant rescuant), generated on a GPCMV bacterial artificial chromosome construct, confirmed the essential nature of the gene. Conventional homologous recombination was used to generate a GP83 mutant to examine the role of GP83 in the viral life cycle. Comparison of the one-step growth kinetics of the GP83 mutant (vAM409) and wild-type GPCMV indicated that GP83 protein is not required for viral replication in tissue culture. The role of GP83 in vivo was examined by comparing the pathogenesis of wild-type GPCMV, vAM409, and a control virus, vAM403, in guinea pigs. The vAM409 mutant was significantly attenuated for dissemination in immunocompromised strain 2 guinea pigs, suggesting that the GP83 protein is essential for full pathogenicity in vivo.


Asunto(s)
Citomegalovirus/genética , Fosfoproteínas/genética , Roseolovirus/genética , Proteínas de la Matriz Viral/genética , Proteínas Virales/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , Cromosomas Artificiales Bacterianos/genética , Clonación Molecular , Citomegalovirus/crecimiento & desarrollo , Citomegalovirus/patogenicidad , Citomegalovirus/fisiología , ADN Viral/genética , Genes Virales , Cobayas , Humanos , Datos de Secuencia Molecular , Mutagénesis , Sistemas de Lectura Abierta , Roseolovirus/crecimiento & desarrollo , Roseolovirus/patogenicidad , Roseolovirus/fisiología , Homología de Secuencia de Aminoácido , Especificidad de la Especie , Activación Transcripcional , Virulencia/genética , Replicación Viral/genética
9.
J Virol Methods ; 108(1): 103-10, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12565160

RESUMEN

Human cytomegalovirus (HCMV) is the most common cause of congenital viral infection in the developed world, and can lead to significant morbidity. Animal models of HCMV infection are required for study of pathogenesis, because of the strict species-specificity of cytomegalovirus (CMV). Among the small animal CMV models, the guinea pig CMV (GPCMV) has unique advantages, in particular its propensity to cross the placenta, causing disease in utero. In order to develop quantitative endpoints for vaccine and antiviral therapeutic studies in the GPCMV model, a quantitative-competitive PCR (qcPCR) assay was developed, based on the GPCMV homolog of the HCMV UL83 gene, GP83. Optimal amplification of GPCMV DNA was observed using primers spanning a 248 base pair (bp) region of this gene. A 91 bp deletion of this cloned fragment was generated for use as an internal standard (IS) for PCR amplification. Standard curves based upon the fluorescent intensity of full-length external target to IS were compared with signal intensity of DNA extracted from blood and organs of experimentally infected guinea pigs in order to quantify viral load. Viral load in newborn guinea pigs infected transplacentally was determined and compared with that of pups infected with GPCMV as neonates. Viral loads were highest in pups infected as neonates. The most consistent isolation and highest quantities of viral DNA were observed in liver and spleen, although viral genome could be readily identified in brain, lung, and salivary gland. Viral load determination should be useful for monitoring outcomes following vaccine studies, as well as responses to experimental antiviral agents.


Asunto(s)
Reacción en Cadena de la Polimerasa/métodos , Roseolovirus/genética , Roseolovirus/patogenicidad , Virología/métodos , Animales , Animales Recién Nacidos , Secuencia de Bases , Infecciones por Citomegalovirus/congénito , Infecciones por Citomegalovirus/etiología , Infecciones por Citomegalovirus/virología , Cartilla de ADN/genética , ADN Viral/análisis , ADN Viral/genética , Modelos Animales de Enfermedad , Femenino , Genes Virales , Genoma Viral , Cobayas , Humanos , Especificidad de Órganos , Embarazo , Roseolovirus/aislamiento & purificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA