Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
PLoS One ; 19(4): e0301773, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38593167

RESUMEN

Respiratory syncytial virus (RSV) is the leading viral cause of bronchiolitis and pneumonia in infants and toddlers, but there currently is no licensed pediatric vaccine. A leading vaccine candidate that has been evaluated for intranasal immunization in a recently completed phase 1/2 clinical trial is an attenuated version of RSV strain A2 called RSV/ΔNS2/Δ1313/I1314L (hereafter called ΔNS2). ΔNS2 is attenuated by deletion of the interferon antagonist NS2 gene and introduction into the L polymerase protein gene of a codon deletion (Δ1313) that confers temperature-sensitivity and is stabilized by a missense mutation (I1314L). Previously, introduction of four amino acid changes derived from a second RSV strain "line 19" (I79M, K191R, T357K, N371Y) into the F protein of strain A2 increased the stability of infectivity and the proportion of F protein in the highly immunogenic pre-fusion (pre-F) conformation. In the present study, these four "line 19" assignments were introduced into the ΔNS2 candidate, creating ΔNS2-L19F-4M. During in vitro growth in Vero cells, ΔNS2-L19F-4M had growth kinetics and peak titer similar to the ΔNS2 parent. ΔNS2-L19F-4M exhibited an enhanced proportion of pre-F protein, with a ratio of pre-F/total F that was 4.5- to 5.0-fold higher than that of the ΔNS2 parent. The stability of infectivity during incubation at 4°C, 25°C, 32°C and 37°C was greater for ΔNS2-L19F-4M; for example, after 28 days at 32°C, its titer was 100-fold greater than ΔNS2. ΔNS2-L19F-4M exhibited similar levels of replication in human airway epithelial (HAE) cells as ΔNS2. The four "line 19" F mutations were genetically stable during 10 rounds of serial passage in Vero cells. In African green monkeys, ΔNS2-L19F-4M and ΔNS2 had similar growth kinetics, peak titer, and immunogenicity. These results suggest that ΔNS2-L19F-4M is an improved live attenuated vaccine candidate whose enhanced stability may simplify its manufacture, storage and distribution, which merits further evaluation in a clinical trial in humans.


Asunto(s)
Infecciones por Virus Sincitial Respiratorio , Vacunas contra Virus Sincitial Respiratorio , Virus Sincitial Respiratorio Humano , Animales , Humanos , Chlorocebus aethiops , Niño , Vacunas contra Virus Sincitial Respiratorio/genética , Células Vero , Anticuerpos Antivirales , Proteínas Virales de Fusión/genética , Virus Sincitial Respiratorio Humano/genética , Anticuerpos Neutralizantes , Mutación Missense
2.
Cell Rep ; 40(12): 111399, 2022 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-36130517

RESUMEN

Human metapneumovirus (hMPV) is a major cause of acute respiratory infections in infants and older adults, for which no vaccines or therapeutics are available. The viral fusion (F) glycoprotein is required for entry and is the primary target of neutralizing antibodies; however, little is known about the humoral immune response generated from natural infection. Here, using prefusion-stabilized F proteins to interrogate memory B cells from two older adults, we obtain over 700 paired non-IgM antibody sequences representing 563 clonotypes, indicative of a highly polyclonal response. Characterization of 136 monoclonal antibodies reveals broad recognition of the protein surface, with potently neutralizing antibodies targeting each antigenic site. Cryo-EM studies further reveal two non-canonical sites and the molecular basis for recognition of the apex of hMPV F by two prefusion-specific neutralizing antibodies. Collectively, these results provide insight into the humoral response to hMPV infection in older adults and will help guide vaccine development.


Asunto(s)
Metapneumovirus , Anciano , Anticuerpos Monoclonales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Humanos , Metapneumovirus/fisiología , Proteínas Virales de Fusión
3.
NPJ Vaccines ; 7(1): 74, 2022 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-35773301

RESUMEN

Respiratory syncytial virus (RSV) G glycoprotein has recently reemerged as a vaccine antigen due to its ability to elicit potent neutralizing antibodies and ameliorate disease in animal models. Here we designed three constructs to display the G central conserved domain (Gcc) focused on inducing broad and potent neutralizing antibodies. One construct displaying Gcc from both RSV subgroups trimerized via a C-terminal foldon (Gcc-Foldon) was highly immunogenic in mice and in MIMIC, a pre-immune human in vitro model. To explore an optimal RSV vaccine, we combined the Gcc-Foldon antigen with a stabilized pre-fusion-F nanoparticle (pre-F-NP) as a bivalent vaccine and detected no antigenic interference between the two antigens in the MIMIC model. In RSV-primed macaques, the bivalent vaccine elicited potent humoral responses. Furthermore, both Gcc-Foldon and the bivalent vaccine conferred effective protection against RSV challenge in mice. This two-component vaccine could potentially provide effective protection against RSV infection in humans and warrants further clinical evaluation.

4.
Hum Vaccin Immunother ; 17(7): 2336-2348, 2021 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-33427044

RESUMEN

Adjuvants are central to the efficacy of subunit vaccines. Although several new adjuvants have been approved in human vaccines over the last decade, the panel of adjuvants in licensed human vaccines remains small. There is still a need for novel adjuvants that can be safely used in humans, easy to source and to formulate with a wide range of antigens and would be broadly applicable to a wide range of vaccines. In this article, using the Respiratory Syncytial Virus (RSV) nanoparticulate prefusion F model antigen developed by Sanofi, we demonstrate in the macaque model that the polyacrylate (PAA)-based adjuvant SPA09 is well tolerated and increases vaccine antigen-specific humoral immunity (sustained neutralizing antibodies, memory B cells and mucosal immunity) and elicits strong TH1-type responses (based on IFNγ and IL-2 ELISpots) in a dose-dependent manner. These data warrant further development of the SPA09 adjuvant for evaluation in clinical trials.


Asunto(s)
Infecciones por Virus Sincitial Respiratorio , Vacunas contra Virus Sincitial Respiratorio , Adyuvantes Inmunológicos , Animales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Humanos , Inmunidad Celular , Inmunidad Humoral , Macaca fascicularis
5.
Virology ; 550: 21-26, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32866728

RESUMEN

Human respiratory syncytial virus (RSV) is a major cause of serious respiratory tract infections in infants and the elderly. Recently it was shown that the RSV G glycoprotein mediates attachment to cells using CX3CR1 as a receptor, and that G-specific neutralizing antibodies can be detected using human airway epithelial (HAE) cell cultures. To investigate the contributions of G-specific antibodies to RSV neutralization, we performed HAE neutralization assays on sera from RSV G-immunized mice or RSV-infected infants. We confirmed that G-specific neutralization using serum from mice or humans could only be detected on HAE cultures. We also found that RSV G-specific antibodies in infants were either subgroup specific or cross-neutralizing. Altogether, our results suggest that G is an important target for generating neutralizing antibodies and would be beneficial to include in an RSV vaccine. Further, inclusion of G antigens from both RSV subgroups may enhance the vaccine cross protection potency.


Asunto(s)
Anticuerpos Neutralizantes/biosíntesis , Anticuerpos Antivirales/biosíntesis , Antígenos Virales/inmunología , Receptor 1 de Quimiocinas CX3C/inmunología , Virus Sincitial Respiratorio Humano/inmunología , Proteínas Virales de Fusión/inmunología , Animales , Antígenos Virales/administración & dosificación , Antígenos Virales/genética , Receptor 1 de Quimiocinas CX3C/genética , Chlorocebus aethiops , Células Epiteliales/inmunología , Células Epiteliales/virología , Femenino , Expresión Génica , Humanos , Sueros Inmunes/química , Inmunización , Ratones , Ratones Endogámicos BALB C , Modelos Biológicos , Pruebas de Neutralización , Unión Proteica , Receptores Virales/genética , Receptores Virales/inmunología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/virología , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/genética , Virus Sincitial Respiratorio Humano/patogenicidad , Células Vero , Proteínas Virales de Fusión/administración & dosificación , Proteínas Virales de Fusión/genética
6.
J Infect Dis ; 222(Suppl 7): S658-S665, 2020 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-32794560

RESUMEN

Respiratory syncytial virus (RSV) is the leading viral pathogen associated with acute lower respiratory tract infection and hospitalization in children < 5 years of age worldwide. While there are known clinical risk factors for severe RSV infection, the majority of those hospitalized are previously healthy infants. There is consequently an unmet need to identify biomarkers that predict host response, disease severity, and sequelae. The primary objective is to identify biomarkers of severe RSV acute respiratory tract infection (ARTI) in infants. Secondary objectives include establishing biomarkers associated with respiratory sequelae following RSV infection and characterizing the viral load, RSV whole-genome sequencing, host immune response, and transcriptomic, proteomic, metabolomic and epigenetic signatures associated with RSV disease severity. Six hundred thirty infants will be recruited across 3 European countries: the Netherlands, Spain, and the United Kingdom. Participants will be recruited into 2 groups: (1) infants with confirmed RSV ARTI (includes upper and lower respiratory tract infections), 500 without and 50 with comorbidities; and (2) 80 healthy controls. At baseline, participants will have nasopharyngeal, blood, buccal, stool, and urine samples collected, plus complete a questionnaire and 14-day symptom diary. At convalescence (7 weeks ± 1 week post-ARTI), specimen collection will be repeated. Laboratory measures will be correlated with symptom severity scores to identify corresponding biomarkers of disease severity. CLINICAL TRIALS REGISTRATION: NCT03756766.


Asunto(s)
Progresión de la Enfermedad , Infecciones por Virus Sincitial Respiratorio/diagnóstico , Infecciones por Virus Sincitial Respiratorio/virología , Índice de Severidad de la Enfermedad , Biomarcadores , Estudios de Casos y Controles , Epigenómica , Europa (Continente) , Femenino , Humanos , Lactante , Masculino , Metabolómica , Nasofaringe/virología , Países Bajos , Proteómica , Virus Sincitial Respiratorio Humano/aislamiento & purificación , Infecciones del Sistema Respiratorio/diagnóstico , Infecciones del Sistema Respiratorio/virología , Factores de Riesgo , España , Encuestas y Cuestionarios , Transcriptoma , Reino Unido , Carga Viral
7.
Sci Immunol ; 5(47)2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32358170

RESUMEN

A stabilized form of the respiratory syncytial virus (RSV) fusion (F) protein has been explored as a vaccine to prevent viral infection because it presents several potent neutralizing epitopes. Here, we used a structure-based rational design to optimize antigen presentation and focus antibody (Ab) responses to key epitopes on the pre-fusion (pre-F) protein. This protein was fused to ferritin nanoparticles (pre-F-NP) and modified with glycans to mask nonneutralizing or poorly neutralizing epitopes to further focus the Ab response. The multimeric pre-F-NP elicited durable pre-F-specific Abs in nonhuman primates (NHPs) after >150 days and elicited potent neutralizing Ab (NAb) responses in mice and NHPs in vivo, as well as in human cells evaluated in the in vitro MIMIC system. This optimized pre-F-NP stimulated a more potent Ab response than a representative pre-F trimer, DS-Cav1. Collectively, this pre-F vaccine increased the generation of NAbs targeting the desired pre-F conformation, an attribute that facilitates the development of an effective RSV vaccine.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Nanopartículas/química , Vacunas contra Virus Sincitial Respiratorio/inmunología , Virus Sincitial Respiratorio Humano/química , Proteínas Virales de Fusión/inmunología , Animales , Formación de Anticuerpos , Antígenos Virales/inmunología , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Vacunas contra Virus Sincitial Respiratorio/química , Virus Sincitial Respiratorio Humano/inmunología , Proteínas Virales de Fusión/química
8.
PLoS One ; 13(6): e0199452, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29920563

RESUMEN

A safe and effective vaccine against RSV remains an important unmet public health need. Intranasally (IN) delivered live-attenuated vaccines represent the most extensively studied approach for immunization of RSV-naïve infants and children, however, achieving an effective balance of attenuation and immunogenicity has proven challenging. Here we report pre-clinical immunogenicity and efficacy data utilizing a live-attenuated vaccine candidate, RGΔM2-2, which was obtained by deleting the M2-2 open reading frame from the genome of the MSA1 clinical isolate. Intramuscular (IM) administration of RGΔM2-2 in cotton rats induced immunity and protective efficacy that was comparable to that induced by intranasal (IN) immunization. In contrast, the protective efficacy of RGΔM2-2 delivered by the IM route to African green monkeys was substantially reduced as compared to the efficacy following IN administration, despite comparable levels of serum neutralizing antibodies. This result suggests that mucosal immunity may play an important role in RSV protection. The RGΔM2-2 vaccine also demonstrated different attenuation profiles when tested in cotton rats, non-human primates, and a human airway epithelial (HAE) cell model. The data suggest RGΔM2-2 is less attenuated than a similarly designed vaccine candidate constructed on the A2 genetic background. These findings have important implications with regard to both the design and the preclinical safety testing of live-attenuated vaccines.


Asunto(s)
Inmunización , Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio/administración & dosificación , Vacunas Atenuadas/administración & dosificación , Administración Intranasal , Animales , Chlorocebus aethiops/inmunología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Genoma Viral/genética , Humanos , Inyecciones Intramusculares , Sistemas de Lectura Abierta , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/virología , Vacunas contra Virus Sincitial Respiratorio/genética , Vacunas contra Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Virus Sincitiales Respiratorios/patogenicidad , Sigmodontinae/inmunología , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología
9.
Hum Vaccin Immunother ; 13(12): 2982-2986, 2017 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-28925795

RESUMEN

The RepliVax vaccine platform(RV) is based on flavivirus genomes that are rationally attenuated by deletion. The self-limiting infection provided by RV has been demonstrated to be safe, highly immunogenic and efficacious for several vaccine candidates against flaviviruses. Here respiratory syncytial virus (RSV) F, influenza virus HA, and simian immunodeficiency virus (SIV) Env proteins were expressed in place of either prM-E or C-prM-E gene deletions of the West Nile (WN) virus genome. The resulting RV-RSV, -influenza and -SIV vaccine prototypes replicated efficiently in complementing helper cells expressing the WN structural proteins in trans. Expressed antigens exhibited correct post-translational processing and the RV recombinants were shown to be highly attenuated and immunogenic in mice, eliciting strong antigen-specific antibodies as well as detectable T-cell responses. These data support the utility of RV vectors for development of vaccines against non-flavivirus targets including rabies and HIV.


Asunto(s)
Virus Defectuosos/genética , Portadores de Fármacos , Vectores Genéticos , Vacunas Virales/inmunología , Virus del Nilo Occidental/genética , Animales , Anticuerpos Antivirales/sangre , Citomegalovirus/genética , Citomegalovirus/inmunología , Expresión Génica , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Ratones Endogámicos BALB C , Orthomyxoviridae/genética , Orthomyxoviridae/inmunología , Virus de la Inmunodeficiencia de los Simios/genética , Virus de la Inmunodeficiencia de los Simios/inmunología , Linfocitos T/inmunología , Linfocitos T Colaboradores-Inductores , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/genética , Replicación Viral
10.
Vaccine ; 34(32): 3690-6, 2016 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-27238375

RESUMEN

Respiratory syncytial virus (RSV) is an important human pathogen, and is the most frequent viral cause of severe respiratory disease in infants. In addition, it is increasingly being recognized as an important cause of respiratory disease in the elderly and immunocompromised. Although a passive prophylactic treatment does exist for high-risk neonates and children, the overall disease burden warrants the development of a safe and effective prophylactic vaccine for use in otherwise healthy newborns and children. RSV is known to be an extremely labile virus, prone to aggregation and loss of infectious titer during virus handling and preparation procedures. To date infective RSV virions have been prepared by methods which are not readily scalable, such as density gradient ultracentrifugation. In this study we describe a scalable, chromatography-based purification procedure for preparation of highly pure, infectious RSV. The purification scheme is based on core bead technology and hollow fiber tangential flow filtration (TFF) and results in a ∼60% recovery of infectious virus titer. This method can be used to prepare highly purified wild type or live-attenuated vaccine strain viruses with titers as high as 1×10(8) plaque forming units per mL. A live-attenuated RSV vaccine prepared by this method was found to be immunogenic and protective in vivo, and its purity was 50-200-fold greater with respect to host cell dsDNA and Vero host cell proteins, than the raw feed stream. The results presented here can be considered a starting point for downstream process development of a live-attenuated vaccine approach for prevention of disease by RSV.


Asunto(s)
Cromatografía , Virus Sincitiales Respiratorios/aislamiento & purificación , Cultivo de Virus , Animales , Chlorocebus aethiops , Femenino , Inmunogenicidad Vacunal , Ratas , Vacunas contra Virus Sincitial Respiratorio/inmunología , Vacunas Atenuadas/inmunología , Células Vero , Virión
11.
PLoS One ; 10(6): e0130517, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26107373

RESUMEN

Respiratory syncytial virus (RSV) is the principal cause of bronchiolitis in infants and a significant healthcare problem. The RSV Glycoprotein (G) mediates attachment of the virus to the cell membrane, which facilitates interaction of the RSV Fusion (F) protein with nucleolin, thereby triggering fusion of the viral and cellular membranes. However, a host protein ligand for G has not yet been identified. Here we show that CX3CR1 is expressed in the motile cilia of differentiated human airway epithelial (HAE) cells, and that CX3CR1 co-localizes with RSV particles. Upon infection, the distribution of CX3CR1 in these cells is significantly altered. Complete or partial deletion of RSV G results in viruses binding at least 72-fold less efficiently to cells, and reduces virus replication. Moreover, an antibody targeting an epitope near the G protein's CX3CR1-binding motif significantly inhibits binding of the virus to airway cells. Given previously published evidence of the interaction of G with CX3CR1 in human lymphocytes, these findings suggest a role for G in the interaction of RSV with ciliated lung cells. This interpretation is consistent with past studies showing a protective benefit in immunizing against G in animal models of RSV infection, and would support targeting the CX3CR1-G protein interaction for prophylaxis or therapy. CX3CR1 expression in lung epithelial cells may also have implications for other respiratory diseases such as asthma.


Asunto(s)
Células Epiteliales/metabolismo , Receptores de Quimiocina/genética , Mucosa Respiratoria/metabolismo , Virus Sincitial Respiratorio Humano/genética , Proteínas del Envoltorio Viral/genética , Proteínas Virales de Fusión/genética , Anticuerpos/farmacología , Secuencia de Bases , Sitios de Unión , Receptor 1 de Quimiocinas CX3C , Diferenciación Celular , Niño , Cilios/metabolismo , Cilios/patología , Cilios/virología , Células Epiteliales/patología , Células Epiteliales/virología , Epítopos/química , Epítopos/inmunología , Expresión Génica , Humanos , Datos de Secuencia Molecular , Cultivo Primario de Células , Unión Proteica , Receptores de Quimiocina/antagonistas & inhibidores , Receptores de Quimiocina/química , Receptores de Quimiocina/metabolismo , Mucosa Respiratoria/patología , Mucosa Respiratoria/virología , Virus Sincitial Respiratorio Humano/metabolismo , Eliminación de Secuencia , Proteínas del Envoltorio Viral/antagonistas & inhibidores , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/metabolismo
12.
J Immunother ; 33(8): 743-58, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20842062

RESUMEN

New therapies are urgently required for the treatment of patients with melanoma. Here we describe the generation and preclinical evaluation of 3 new recombinant ALVAC(2) poxviruses vCP2264, vCP2291, and vCP2292 for their ability to induce the desired cellular immune responses against the encoded melanoma-associated antigens. This was done either in HLA-A2/K transgenic mice or using in vitro antigen-presentation studies. These studies demonstrated that the vaccine was able to induce HLA-A*0201-restricted T-cell responses against gp100 and NY-ESO-1, detectable directly ex vivo, in HLA-A2/K-transgenic mice. The in vitro antigen presentation studies, in the absence of appropriate animal models, demonstrated that target cells infected with the vaccine construct were lysed by MAGE-1, MAGE-3 or MART-1 peptide-specific T cells. These data indicate that ALVAC(2)-encoded melanoma-associated antigens can be properly processed and presented to induce antigen-specific cytotoxic T-cell responses. To enhance the immunogenicity of the melanoma antigens, a TRIad of COstimulatory Molecules (TRICOM) were also cloned into all 3 vectors. Increased in vitro proliferation and IFN-γ production was observed with all ALVAC(2) poxviruses encoding TRICOM, confirming the immune-enhancing effect of the ALVAC-encoded TRICOM. These studies demonstrated that all components of the vaccine were functionally active and provide a rationale for moving this candidate vaccine to the clinic.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Vacunas contra el Cáncer , Melanoma/inmunología , Infecciones por Poxviridae/inmunología , Poxviridae/inmunología , Linfocitos T Citotóxicos/metabolismo , Vacunas Virales , Animales , Antígenos CD/genética , Antígenos CD/inmunología , Antígenos CD/metabolismo , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Células Cultivadas , Clonación Molecular , Citotoxicidad Inmunológica , Evaluación Preclínica de Medicamentos , Antígeno HLA-A2/genética , Humanos , Activación de Linfocitos , Melanoma/patología , Melanoma/terapia , Ratones , Ratones Transgénicos , Metástasis de la Neoplasia , Poxviridae/genética , Poxviridae/patogenicidad , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/patología
13.
Virus Res ; 110(1-2): 169-76, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15845268

RESUMEN

In order to optimize foreign gene expression in the E3 region of BAdV-3, we constructed full-length BAdV-3 genomic DNA clones containing a reporter gene (truncated glycoprotein gD of bovine herpesvirus 1, gDt), under the control of exogenous promoters inserted in either direction in the E3 region. Irrespective of exogenous transcriptional elements, viable recombinant BAdV-3 viruses could only be isolated when the gDt expression cassettes were inserted in the E3 region parallel to the direction of E3 transcription. Introduction of exogenous promoters altered the kinetics and amount of gDt expression in recombinant BAdV-3 infected cells. Interestingly, recombinant BAdV-3 containing gDt under the control of the mouse cytomegalovirus (MCMV) immediate early (IE) promoter expressed gDt more efficiently with noticeable differences in the amount and kinetics of expression. Moreover, animals immunized with recombinant BAdV-3 expressing gDt under the control of the MCMV IE promoter induced strong immune responses with reduced pathological lesions. These results suggest that BAdV vectors with the MCMV IE promoter may be useful for transgene expression and the development of vaccines.


Asunto(s)
Proteínas E3 de Adenovirus/genética , Regulación Viral de la Expresión Génica , Mastadenovirus/genética , Regiones Promotoras Genéticas , Proteínas Recombinantes/biosíntesis , Proteínas Virales/biosíntesis , Animales , Anticuerpos Antivirales/sangre , Bovinos/virología , Línea Celular , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática , Genes Reporteros , Vectores Genéticos , Inmunoprecipitación , Muromegalovirus/genética , Ratas , Recombinación Genética , Proteínas Virales/análisis , Proteínas Virales/genética , Proteínas Virales/inmunología
14.
J Gen Virol ; 84(Pt 11): 2947-2956, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14573799

RESUMEN

The assembly of adenovirus particles is a multistep process, in which viral genomic DNA is selected and subsequently inserted into preformed empty capsids. The selective encapsidation of the adenovirus genome is directed by cis-acting packaging motifs, termed A repeats due to their AT-rich character in DNA sequence. A repeats are usually located at the left end of the viral genome. In this report, the construction and analysis of bovine adenovirus type 3 (BAdV-3) mutants containing deletion mutations introduced into the AT-rich regions are described. The main cis-acting packaging domains of BAdV-3 were localized between nt 224 and 540 relative to the left end of the viral genome. They displayed a functional redundancy and followed a hierarchy of importance. In addition, the results demonstrated that not all of the AT-rich units functioned as cis-acting packaging motifs.


Asunto(s)
ADN Viral/fisiología , Mastadenovirus/genética , Ensamble de Virus , Secuencia de Bases , Genoma Viral , Mastadenovirus/fisiología , Datos de Secuencia Molecular , Mutación , Secuencias Repetitivas de Ácidos Nucleicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...