Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
Sci Rep ; 11(1): 18641, 2021 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-34545126

RESUMEN

Human respiratory syncytial virus (hRSV) infection is a major pediatric health concern worldwide. Despite more than half a century of efforts, there is still no commercially available vaccine. In this study, we constructed and purified the recombinant protein CTA1-DD-RBF composed of a CTA1-DD mucosal adjuvant and prefusion F protein (RBF) using Escherichia coli BL21 cells. We studied the immunogenicity of CTA1-DD-RBF in mice. Intranasal immunization with CTA1-DD-RBF stimulated hRSV F-specific IgG1, IgG2a, sIgA, and neutralizing antibodies as well as T cell immunity without inducing lung immunopathology upon hRSV challenge. Moreover, the protective immunity of CTA1-DD-RBF was superior to that of the RBF protein, as confirmed by the assessment of serum-neutralizing activity and viral clearance after challenge. Compared to formalin-inactivated hRSV (FI-RSV), intranasal immunization with CTA1-DD-RBF induced a Th1 immune response. In summary, intranasal immunization with CTA1-DD-RBF is safe and effective in mice. Therefore, CTA1-DD-RBF represents a potential mucosal vaccine candidate for the prevention of human infection with hRSV.


Asunto(s)
Toxina del Cólera/administración & dosificación , Toxina del Cólera/inmunología , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/inmunología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Virus Sincitial Respiratorio Humano/inmunología , Proteínas Virales de Fusión/administración & dosificación , Proteínas Virales de Fusión/inmunología , Adyuvantes de Vacunas/administración & dosificación , Administración Intranasal , Animales , Anticuerpos Neutralizantes/biosíntesis , Anticuerpos Antivirales/biosíntesis , Toxina del Cólera/genética , Femenino , Humanos , Inmunidad Mucosa , Inmunización , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Proteínas Recombinantes de Fusión/genética , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/genética , Células TH1/inmunología , Proteínas Virales de Fusión/genética , Replicación Viral
2.
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
3.
Arch Virol ; 165(9): 1959-1968, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32519007

RESUMEN

Newcastle disease (ND), caused by virulent Newcastle disease virus (NDV) strains, has been one of the most problematic diseases affecting the poultry industry worldwide. Conventional vaccines provide effective protection for birds to survive ND outbreaks, but they may not completely suppress NDV shedding. NDV strains circulate on farms for a long time after the initial infection and cause potential risks. A new vaccine with fast clearance ability and low viral shedding is needed. In this study, we used interleukin-12 (IL-12) as an adjuvant and electroporation (EP) as an advanced delivery system to improve a DNA vaccine candidate. The fusion (F) protein gene from an NDV strain of the prevalent genotype VII.1.1 was cloned to prepare the vaccine. Chickens immunized with the F gene DNA vaccine co-delivered with an IL-12-expressing plasmid DNA showed higher neutralizing antibody levels and stronger concanavalin-A-induced lymphocyte proliferation than those treated with the F gene DNA vaccine alone. The co-delivered vaccine provided 100% protection, and less viral shedding and a shorter release time were observed in challenged chickens than when the F gene DNA vaccine was administered alone. The use of F gene DNA combined with IL-12 delivered by electroporation is a promising approach for vaccination against ND.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Interleucina-12/inmunología , Enfermedad de Newcastle/prevención & control , Virus de la Enfermedad de Newcastle/inmunología , Enfermedades de las Aves de Corral/prevención & control , Vacunas de ADN/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales/inmunología , Pollos , Electroporación , Interleucina-12/administración & dosificación , Enfermedad de Newcastle/inmunología , Enfermedad de Newcastle/virología , Virus de la Enfermedad de Newcastle/genética , Virus de la Enfermedad de Newcastle/fisiología , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/virología , Vacunación , Vacunas de ADN/administración & dosificación , Vacunas de ADN/genética , Proteínas Virales de Fusión/administración & dosificación , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/genética , Esparcimiento de Virus
4.
Virol J ; 17(1): 58, 2020 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-32321526

RESUMEN

BACKGROUND: Prophylactic vaccines are critical in preventing hand, foot, and mouth disease (HFMD) primarily caused by human enterovirus 71 (EV71) infection. Children aged less than 5 years are especially susceptible to EV71 infections. In addition to the development of vaccines containing the inactivated virus, those containing virus-like particles (VLPs) with repeated antigens also constitute an effective preventive strategy for EV71 infections, with safety and productivity advantages. We previously developed a fusion protein composed with truncated peptides of the EV71 capsid protein, which assembled into spherical particles. This study aimed to assess the immunoprotective effects of this fusion protein as a vaccine candidate in a mouse model of EV71 infection. METHODS: To evaluate the protective effect of fusion protein vaccine candidate, neonatal mice born by immunized female mice, as well as normal neonatal mice immunized twice were infected with EV71 virus. Whereafter, the survival rates, clinical scores and viral loads were measured. RESULTS: The high dosage and booster immunization helped induce specific serum antibodies with high neutralization titers, which were transferred to neonatal mice, thereby facilitating effective resistance towards EV71 infection. An active immune response was also observed in neonatal mice which generated following immunization. CONCLUSIONS: The present results suggest that this fusion protein is a suitable vaccine candidate in treating EV71 infections.


Asunto(s)
Enterovirus Humano A/genética , Infecciones por Enterovirus/prevención & control , Péptidos/inmunología , Proteínas Virales de Fusión/inmunología , Vacunas Virales/inmunología , Animales , Animales Recién Nacidos , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Línea Celular Tumoral , Enterovirus Humano A/inmunología , Infecciones por Enterovirus/inmunología , Femenino , Humanos , Inmunización Secundaria , Ratones , Ratones Endogámicos BALB C , Péptidos/genética , Proteínas Virales de Fusión/administración & dosificación , Carga Viral , Vacunas Virales/genética
5.
Vaccine ; 38(9): 2122-2127, 2020 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-32007293

RESUMEN

Human metapneumovirus (hMPV) is an important respiratory pathogen especially in young children and elderly subjects. Our objective was to assess the immunogenicity and protection conferred by predominant pre- and post-fusion (F) hMPV-F constructs in Balb/C mice. Immunizations without adjuvant were not immunogenic whereas alum-adjuvanted hMPV-F proteins, regardless of their conformations, generated comparable neutralizing antibody titers with undetectable pulmonary viral titers following viral challenge. In conclusion, we found no apparent advantage for mixtures of predominant pre-fusion F proteins over post-fusion conformations for hMPV vaccination in opposite to recent data obtained with the human respiratory syncytial virus.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Metapneumovirus , Infecciones por Paramyxoviridae , Proteínas Virales de Fusión/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Metapneumovirus/inmunología , Ratones , Ratones Endogámicos BALB C , Infecciones por Paramyxoviridae/prevención & control , Vacunas de Subunidad/inmunología , Proteínas Virales de Fusión/administración & dosificación
6.
Vaccine ; 38(5): 1258-1270, 2020 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-31761502

RESUMEN

Globally, human respiratory syncytial virus (RSV) is a major cause of severe lower respiratory infection in infants and young children. There are no licensed vaccines despite the high worldwide disease burden. RSV fusion (F) glycoprotein vaccine is the most advanced candidate for maternal immunization. In this report, a baboon maternal immunization model was used to assess the immunogenicity and protection of infants against pulmonary challenge with human RSV/A. Vaccination in the third trimester produced high anti-RSV F IgG titers and virus-neutralizing antibodies. Infants born to immunized females had high levels of serum RSV antibodies that were comparable to maternal levels at birth and persisted for over 50 days with a half-life of 14-24 days. Furthermore, infants from immunized females and challenged with RSV/A were healthy, developed less severe disease, and had only mild pulmonary inflammatory changes whereas infants born to non-vaccinated females developed more severe disease with marked to moderate interstitial pneumonia, pulmonary edema, and bronchiolar obstruction. These results support the further development of the RSV F vaccine for maternal immunization.


Asunto(s)
Glicoproteínas/inmunología , Infecciones por Virus Sincitial Respiratorio , Vacunas contra Virus Sincitial Respiratorio/administración & dosificación , Proteínas Virales de Fusión/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Femenino , Glicoproteínas/administración & dosificación , Madres , Papio , Infecciones por Virus Sincitial Respiratorio/prevención & control , Virus Sincitial Respiratorio Humano/inmunología , Vacunación , Proteínas Virales de Fusión/administración & dosificación
7.
Viruses ; 11(7)2019 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-31337019

RESUMEN

Andes virus (ANDV) and Sin Nombre virus (SNV) are the main causative agents responsible for hantavirus cardiopulmonary syndrome (HCPS) in the Americas. HCPS is a severe respiratory disease with a high fatality rate for which there are no approved therapeutics or vaccines available. Some vaccine approaches for HCPS have been tested in preclinical models, but none have been tested in infectious models in regard to their ability to protect against multiple species of HCPS-causing viruses. Here, we utilize recombinant vesicular stomatitis virus-based (VSV) vaccines for Andes virus (ANDV) and Sin Nombre virus (SNV) and assess their ability to provide cross-protection in infectious challenge models. We show that, while both rVSVΔG/ANDVGPC and rVSVΔG/SNVGPC display attenuated growth as compared to wild type VSV, each vaccine is able to induce a cross-reactive antibody response. Both vaccines protected against both homologous and heterologous challenge with ANDV and SNV and prevented HCPS in a lethal ANDV challenge model. This study provides evidence that the development of a single vaccine against HCPS-causing hantaviruses could provide protection against multiple agents.


Asunto(s)
Anticuerpos Antivirales/sangre , Protección Cruzada , Síndrome Pulmonar por Hantavirus/prevención & control , Orthohantavirus/inmunología , Virus Sin Nombre/inmunología , Vesiculovirus/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Cricetinae , Femenino , Mesocricetus , Vacunación , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología , Vesiculovirus/genética , Proteínas Virales de Fusión/administración & dosificación , Proteínas Virales de Fusión/inmunología , Vacunas Virales/genética
8.
Virology ; 534: 1-13, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31163351

RESUMEN

Alum adjuvanted formalin-inactivated respiratory syncytial virus (RSV) vaccination resulted in enhanced respiratory disease in young children upon natural infection. Here, we investigated the adjuvant effects of monophosphoryl lipid A (MPL) and oligodeoxynucleotide CpG (CpG) on vaccine-enhanced respiratory disease after fusion (F) protein prime vaccination and RSV challenge in infant and adult mouse models. Combination CpG + MPL adjuvant in RSV F protein single dose priming of infant and adult age mice was found to promote the induction of IgG2a isotype antibodies and neutralizing activity, and lung viral clearance after challenge. CpG + MPL adjuvanted F protein (Fp) priming of infant and adult age mice was effective in avoiding lung histopathology, in reducing interleukin-4+ CD4 T cells and cellular infiltration of monocytes and neutrophils after RSV challenge. This study suggests that combination CpG and MPL adjuvant in RSV subunit vaccination might contribute to priming protective immune responses and preventing inflammatory RSV disease after infection.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Pulmón/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Vacunas contra Virus Sincitial Respiratorio/administración & dosificación , Virus Sincitial Respiratorio Humano/inmunología , Proteínas Virales de Fusión/administración & dosificación , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Linfocitos T CD4-Positivos/inmunología , Femenino , Humanos , Pulmón/patología , Pulmón/virología , Masculino , Ratones , Ratones Endogámicos BALB C , Neutrófilos/inmunología , Oligodesoxirribonucleótidos/administración & dosificación , Oligodesoxirribonucleótidos/inmunología , Infecciones por Virus Sincitial Respiratorio/patología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Infecciones por Virus Sincitial Respiratorio/virología , Vacunas contra Virus Sincitial Respiratorio/genética , Vacunas contra Virus Sincitial Respiratorio/inmunología , Virus Sincitial Respiratorio Humano/genética , Virus Sincitial Respiratorio Humano/fisiología , Vacunación , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/inmunología
9.
PLoS Biol ; 17(2): e3000164, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30789898

RESUMEN

Throughout the last several decades, vaccination has been key to prevent and eradicate infectious diseases. However, many pathogens (e.g., respiratory syncytial virus [RSV], influenza, dengue, and others) have resisted vaccine development efforts, largely because of the failure to induce potent antibody responses targeting conserved epitopes. Deep profiling of human B cells often reveals potent neutralizing antibodies that emerge from natural infection, but these specificities are generally subdominant (i.e., are present in low titers). A major challenge for next-generation vaccines is to overcome established immunodominance hierarchies and focus antibody responses on crucial neutralization epitopes. Here, we show that a computationally designed epitope-focused immunogen presenting a single RSV neutralization epitope elicits superior epitope-specific responses compared to the viral fusion protein. In addition, the epitope-focused immunogen efficiently boosts antibodies targeting the palivizumab epitope, resulting in enhanced neutralization. Overall, we show that epitope-focused immunogens can boost subdominant neutralizing antibody responses in vivo and reshape established antibody hierarchies.


Asunto(s)
Anticuerpos Neutralizantes/biosíntesis , Anticuerpos Antivirales/biosíntesis , Epítopos/química , Receptores de Antígenos de Linfocitos B/inmunología , Proteínas Recombinantes de Fusión/química , Virus Sincitiales Respiratorios/inmunología , Proteínas Virales de Fusión/química , Animales , Anticuerpos Monoclonales Humanizados/química , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Neutralizantes/genética , Anticuerpos Antivirales/genética , Clonación Molecular , Diseño Asistido por Computadora , Epítopos/inmunología , Escherichia coli/genética , Escherichia coli/metabolismo , Femenino , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Inmunización/métodos , Inmunogenicidad Vacunal , Ratones , Ratones Endogámicos BALB C , Nanopartículas/administración & dosificación , Nanopartículas/química , Palivizumab/química , Palivizumab/inmunología , Receptores de Antígenos de Linfocitos B/química , Receptores de Antígenos de Linfocitos B/genética , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Vacunas contra Virus Sincitial Respiratorio/administración & dosificación , Vacunas contra Virus Sincitial Respiratorio/biosíntesis , Vacunas contra Virus Sincitial Respiratorio/genética , Homología Estructural de Proteína , Proteínas Virales de Fusión/administración & dosificación , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/inmunología
10.
Braz J Microbiol ; 50(1): 279-286, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30637649

RESUMEN

Porcine rotavirus (PoRV) and porcine epidemic diarrhea virus (PEDV) usually co-infect pigs in modern large-scale piggery, which both can cause severe diarrhea in newborn piglets and lead to significant economic losses to the pig industry. The VP7 protein is the main coat protein of PoRV, and the S protein is the main structural protein of PEDV, which are capable of inducing neutralizing antibodies in vivo. In this study, a DNA vaccine pPI-2.EGFP.VP7.S co-expressing VP7 protein of PoRV and S protein of PEDV was constructed. Six 8-week-old mice were immunized with the recombinant plasmid pPI-2.EGFP.VP7.S. The high humoral immune responses (virus specific antibody) and cellular immune responses (IFN-γ, IL-4, and spleen lymphocyte proliferation) were evaluated. The immune effect through intramuscular injection increased with plasmid dose when compared with subcutaneous injection. The immune-enhancing effect of IFN-α adjuvant was excellent compared with pig spleen transfer factor and IL-12 adjuvant. These results demonstrated that pPI-2.EGFP.VP7.S possess the immunological functions of the VP7 proteins of PoRV and S proteins of PEDV, indicating that pPI-2.EGFP.VP7.S is a candidate vaccine for porcine rotaviral infection (PoR) and porcine epidemic diarrhea (PED).


Asunto(s)
Antígenos Virales/inmunología , Proteínas de la Cápside/inmunología , Infecciones por Coronavirus/veterinaria , Plásmidos/inmunología , Infecciones por Rotavirus/veterinaria , Rotavirus/inmunología , Enfermedades de los Porcinos/prevención & control , Proteínas Virales de Fusión/inmunología , Vacunas Virales/inmunología , Animales , Antígenos Virales/administración & dosificación , Antígenos Virales/genética , Proteínas de la Cápside/administración & dosificación , Proteínas de la Cápside/genética , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/prevención & control , Infecciones por Coronavirus/virología , ADN Recombinante/administración & dosificación , ADN Recombinante/genética , ADN Recombinante/inmunología , Evaluación Preclínica de Medicamentos , Ratones , Plásmidos/administración & dosificación , Plásmidos/genética , Virus de la Diarrea Epidémica Porcina/genética , Virus de la Diarrea Epidémica Porcina/inmunología , Rotavirus/genética , Infecciones por Rotavirus/inmunología , Infecciones por Rotavirus/prevención & control , Infecciones por Rotavirus/virología , Porcinos , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/virología , Proteínas Virales de Fusión/administración & dosificación , Proteínas Virales de Fusión/genética , Vacunas Virales/administración & dosificación , Vacunas Virales/genética
11.
Vaccine ; 37(6): 817-826, 2019 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-30638800

RESUMEN

Conventional influenza vaccines are hampered by slow and limited production capabilities, whereas DNA vaccines can be rapidly produced for global coverage in the event of an emerging pandemic. However, a drawback of DNA vaccines is their generally low immunogenicity in non-human primates and humans. We have previously demonstrated that targeting of influenza hemagglutinin to human HLA class II molecules can increase antibody responses in larger animals such as ferrets and pigs. Here, we extend these observations by immunizing non-human primates (rhesus macaques) with a DNA vaccine encoding a bivalent fusion protein that targets influenza virus hemagglutinin (HA) to Mamu class II molecules. Such immunization induced neutralizing antibodies and antigen-specific T cells. The DNA was delivered by pain- and needle-free jet injections intradermally. No adverse effects were observed. Most importantly, the immunized rhesus macaques were protected against a challenge with influenza virus.


Asunto(s)
Anticuerpos Antivirales/sangre , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Vacunas de ADN/administración & dosificación , Proteínas Virales de Fusión/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Citocinas/inmunología , Subtipo H1N1 del Virus de la Influenza A , Inyecciones Intradérmicas/métodos , Inyecciones a Chorro , Macaca mulatta , Masculino , Vacunación/métodos , Vacunas de ADN/inmunología , Proteínas Virales de Fusión/administración & dosificación
12.
Fish Shellfish Immunol ; 85: 99-105, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29969707

RESUMEN

The highly effective DNA vaccines against diseases caused by fish rhabdoviruses in farmed fish consist of a DNA plasmid vector encoding the viral glycoprotein under the control of a constitutive cytomegalovirus promoter (CMV). Among others, attempts to improve efficacy and safety of these DNA vaccines have focused on regulatory elements of plasmid vectors, which play a major role in controlling expression levels of vaccine antigens. Depending on the context, use of a fish-derived promoter with minimal activity in mammalian cells could be preferable. Another aspect related to the CMV promoter is that constitutive expression of the vaccine antigen may lead to rapid elimination of antigen expressing cells in the fish and thereby potentially reduce the long-term effects of the vaccine. In this study, we compared DNA vaccines with the interferon-inducible Mx promoter from rainbow trout and the CMV promoter, respectively. Plasmid constructs encoding the enhanced green fluorescent protein (EGFP) were used for the in vitro analysis, whereas DNA vaccines encoding the glycoprotein (G) of the viral haemorrhagic septicaemia virus (VHSV) were applied for the in vivo examination. The in vitro analysis showed that while the DNA vaccine with the CMV promoter constitutively drove the expression of EGFP in both fish and human cell lines, the DNA vaccine with the Mx promoter inducibly enhanced the expression of EGFP in the fish cell line. To address the impact on protection, a time-course model was followed as suggested by Kurath et al. (2006), where vaccinated fish were challenged with VHSV at 2, 8 and 78 weeks post-vaccination (wpv). The DNA vaccine with the CMV promoter protected at all times, while vaccination with the DNA vaccine containing the Mx promoter only protected the fish at 8 wpv. However, following induction with Poly (I:C) one week before the challenge, high protection was also evident at 2 wpv. In conclusion, the results revealed a more fish host dependent activity of the trout Mx promoter compared to the traditionally used cross species-active CMV promoter, but improvements will be needed for its application in DNA vaccines to ensure long term protection.


Asunto(s)
Enfermedades de los Peces/prevención & control , Septicemia Hemorrágica Viral/prevención & control , Novirhabdovirus/inmunología , Oncorhynchus mykiss , Vacunas de ADN/farmacología , Vacunas Virales/farmacología , Animales , Línea Celular , Cyprinidae , Femenino , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/virología , Células HeLa , Septicemia Hemorrágica Viral/inmunología , Septicemia Hemorrágica Viral/virología , Humanos , Interferones/inmunología , Perciformes , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacología , Vacunas de ADN/administración & dosificación , Proteínas Virales de Fusión/administración & dosificación , Proteínas Virales de Fusión/farmacología , Vacunas Virales/administración & dosificación
13.
Antiviral Res ; 162: 54-60, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30550799

RESUMEN

Human parainfluenza virus 3 (PIV3) and respiratory syncytial virus (RSV) are major causative agents of serious respiratory tract illness in newborns and infants. Maternal vaccination could be a promising approach to provide immediate protection against severe PIV3 and RSV infection in young infants. Previously, we demonstrated that maternal immunization with a subunit vaccine consisting of the RSV fusion (F) protein formulated with TriAdj, an adjuvant consisting of poly(I:C), immune defense regulatory peptide and polyphosphazene, protects newborn lambs from RSV. In the present study we evaluated the protective efficacy of a novel bivalent RSV-PIV3 vaccine candidate, FRipScHN/TriAdj, as a maternal vaccine against PIV3 infection in a neonatal lamb model. This vaccine consists of the pre-fusion form of the RSV F protein linked to the haemagglutinin-neuraminidase (HN) of PIV3, formulated with TriAdj. First, we successfully established PIV3 infection in neonatal lambs. Lambs infected with human PIV3 showed gross pathology, bronchointerstitial pneumonia and viral replication in the lungs. Subsequently, ewes were immunized with FRipScHN/TriAdj. RSV FRipSc- and PIV3 HN-specific antibodies with virus-neutralizing activity were detected in both the serum and the colostrum of the vaccinated ewes. The newborn lambs had RSV- and PIV3- neutralizing antibodies in their serum, which demonstrates that maternal antibodies were transferred to the neonates. At three days of age, the newborn lambs received an intrapulmonary challenge with PIV3. The lung pathology and virus production were significantly reduced in lambs that had received PIV3-specific maternal antibodies compared to lambs born to non-vaccinated ewes. These results suggest that maternal vaccination with a bivalent FRipScHN/TriAdj vaccine might be an effective method to provide protection against both PIV3 and RSV in neonates.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Anticuerpos Antivirales/sangre , Inmunidad Materno-Adquirida , Vacunas contra la Parainfluenza/administración & dosificación , Infecciones por Respirovirus/veterinaria , Animales , Animales Recién Nacidos , Anticuerpos Neutralizantes/sangre , Femenino , Glicoproteínas/genética , Glicoproteínas/inmunología , Humanos , Vacunas contra la Parainfluenza/inmunología , Virus de la Parainfluenza 3 Humana/inmunología , Embarazo , Virus Sincitiales Respiratorios/genética , Infecciones por Respirovirus/inmunología , Infecciones por Respirovirus/prevención & control , Ovinos , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/inmunología , Proteínas Virales de Fusión/administración & dosificación , Proteínas Virales de Fusión/inmunología
14.
Proc Natl Acad Sci U S A ; 115(48): 12265-12270, 2018 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-30420505

RESUMEN

Parainfluenza virus types 1-4 (PIV1-4) are highly infectious human pathogens, of which PIV3 is most commonly responsible for severe respiratory illness in newborns, elderly, and immunocompromised individuals. To obtain a vaccine effective against all four PIV types, we engineered mutations in each of the four PIV fusion (F) glycoproteins to stabilize their metastable prefusion states, as such stabilization had previously enabled the elicitation of high-titer neutralizing antibodies against the related respiratory syncytial virus. A cryoelectron microscopy structure of an engineered PIV3 F prefusion-stabilized trimer, bound to the prefusion-specific antibody PIA174, revealed atomic-level details for how introduced mutations improved stability as well as how a single PIA174 antibody recognized the trimeric apex of prefusion PIV3 F. Nine combinations of six newly identified disulfides and two cavity-filling mutations stabilized the prefusion PIV3 F immunogens and induced 200- to 500-fold higher neutralizing titers in mice than were elicited by PIV3 F in the postfusion conformation. For PIV1, PIV2, and PIV4, we also obtained stabilized prefusion Fs, for which prefusion versus postfusion titers were 2- to 20-fold higher. Elicited murine responses were PIV type-specific, with little cross-neutralization of other PIVs. In nonhuman primates (NHPs), quadrivalent immunization with prefusion-stabilized Fs from PIV1-4 consistently induced potent neutralizing responses against all four PIVs. For PIV3, the average elicited NHP titer from the quadrivalent immunization was more than fivefold higher than any titer observed in a cohort of over 100 human adults, highlighting the ability of a prefusion-stabilized immunogen to elicit especially potent neutralization.


Asunto(s)
Virus de la Parainfluenza 1 Humana/inmunología , Virus de la Parainfluenza 2 Humana/inmunología , Virus de la Parainfluenza 3 Humana/inmunología , Virus de la Parainfluenza 4 Humana/inmunología , Infecciones por Respirovirus/inmunología , Proteínas Virales de Fusión/química , Vacunas Virales/química , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Microscopía por Crioelectrón , Femenino , Humanos , Macaca mulatta , Masculino , Ratones , Virus de la Parainfluenza 1 Humana/química , Virus de la Parainfluenza 1 Humana/genética , Virus de la Parainfluenza 2 Humana/química , Virus de la Parainfluenza 2 Humana/genética , Virus de la Parainfluenza 3 Humana/química , Virus de la Parainfluenza 3 Humana/genética , Virus de la Parainfluenza 4 Humana/química , Virus de la Parainfluenza 4 Humana/genética , Infecciones por Virus Sincitial Respiratorio , Infecciones por Respirovirus/prevención & control , Infecciones por Respirovirus/virología , Proteínas Virales de Fusión/administración & dosificación , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/genética , Vacunas Virales/inmunología
15.
Viruses ; 10(8)2018 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-30110893

RESUMEN

Prevention of severe lower respiratory tract infections in infants caused by the human respiratory syncytial virus (hRSV) remains a major public health priority. Currently, the major focus of vaccine development relies on the RSV fusion (F) protein since it is the main target protein for neutralizing antibodies induced by natural infection. The protein conserves 5 N-glycosylation sites, two of which are located in the F2 subunit (N27 and N70), one in the F1 subunit (N500) and two in the p27 peptide (N116 and N126). To study the influence of the loss of one or more N-glycosylation sites on RSV F immunogenicity, BALB/c mice were immunized with plasmids encoding RSV F glycomutants. In comparison with F WT DNA immunized mice, higher neutralizing titres were observed following immunization with F N116Q. Moreover, RSV A2-K-line19F challenge of mice that had been immunized with mutant F N116Q DNA was associated with lower RSV RNA levels compared with those in challenged WT F DNA immunized animals. Since p27 is assumed to be post-translationally released after cleavage and thus not present on the mature RSV F protein, it remains to be elucidated how deletion of this glycan can contribute to enhanced antibody responses and protection upon challenge. These findings provide new insights to improve the immunogenicity of RSV F in potential vaccine candidates.


Asunto(s)
Anticuerpos Neutralizantes/biosíntesis , Anticuerpos Antivirales/biosíntesis , Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio/administración & dosificación , Vacunas de ADN/administración & dosificación , Proteínas Virales de Fusión/genética , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Femenino , Glicosilación , Humanos , Hidrólisis , Inmunización , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Mutación , Plásmidos/administración & dosificación , Plásmidos/genética , Plásmidos/inmunología , Ingeniería de Proteínas , Subunidades de Proteína/administración & dosificación , Subunidades de Proteína/genética , Subunidades de Proteína/inmunología , 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 Sincitial Respiratorio Humano/efectos de los fármacos , Virus Sincitial Respiratorio Humano/genética , Virus Sincitial Respiratorio Humano/inmunología , Vacunas de ADN/genética , Vacunas de ADN/inmunología , Proteínas Virales de Fusión/administración & dosificación , Proteínas Virales de Fusión/inmunología , Carga Viral/efectos de los fármacos
16.
Nat Commun ; 9(1): 1904, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29765035

RESUMEN

Respiratory syncytial virus (RSV) is the most common cause of bronchiolitis in infants. Maternal immunization is an option to increase maternal antibody levels and protect infants from infection. Here we assess the efficacy of virus-like particle (VLP) vaccine candidates containing stabilized pre-fusion (pre-F) or post-fusion (post-F) conformations of the RSV F protein and the attachment RSV G protein in a maternal immunization model using cotton rats. VLP vaccines containing RSV F and G proteins strongly boost pre-existing RSV immunity in dams preventing their perinatal drop in immunity. Boosting is stronger for the pre-F VLP than for the post-F VLP or purified subunit F protein vaccines, giving an advantage on mothers' protection. VLP immunization of dams provides significant protection to pups from RSV challenge and reduced pulmonary inflammation. Collectively, our results show that a VLP vaccine with RSV F and G proteins is safe and effective for maternal and adult vaccination.


Asunto(s)
Inmunidad Materno-Adquirida , Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio/inmunología , Virus Sincitial Respiratorio Humano/inmunología , Animales , Anticuerpos Antivirales/inmunología , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunización , Pulmón/inmunología , Pulmón/virología , Masculino , Ratas , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/virología , Vacunas contra Virus Sincitial Respiratorio/administración & dosificación , Vacunas contra Virus Sincitial Respiratorio/genética , Virus Sincitial Respiratorio Humano/genética , Sigmodontinae , Proteínas del Envoltorio Viral/administración & dosificación , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología , Proteínas Virales de Fusión/administración & dosificación , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/inmunología
17.
Vet Microbiol ; 216: 99-106, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29519533

RESUMEN

Administration of vaccines combined with the good management and strict biosecurity is an effective way for Newcastle disease (ND) control. However, vaccine failure is continuously reported in some countries mainly because the antigenic difference between the used vaccine and field strains even they are of one serotype. Therefore, development of antigen-matched ND vaccines is needed to improve the vaccine efficacy in birds. In this study, we introduced four site mutations, K1756A, D1881A, K1917A and E1954Q, respectively, into the large protein gene of the virulent genotype VII Newcastle disease virus (NDV) G7 strain using reverse genetics technology. Four rescued NDVs were sharply attenuated for the pathogenicity in chickens. One of these mutants, E1954Q, was further manipulated by replacing the F cleavage site sequence of typical velogenic strains with that of the LaSota vaccine, resulting in a new mutant, G7M. Biological characterization showed that G7M was safe and genetically stable after serial passages in embryos and chickens. Vaccination of chickens with G7M induced a progressive elevation of the homologous antibodies and markedly higher CD8+ T cell percentage, T cell proliferation and IFN-γ than LaSota. G7M conferred full protection against genotype VII NDV challenge, and more importantly, it effectively reduced the challenge virus replication and shedding in chickens. Together, our data suggest that G7M is a promising genotype VII vaccine candidate, and the novel attenuation approach designed in this study could be used to develop new antigen-matched NDV vaccines.


Asunto(s)
Genes Virales/genética , Genotipo , Enfermedad de Newcastle/prevención & control , Virus de la Enfermedad de Newcastle/genética , Enfermedades de las Aves de Corral/prevención & control , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Pollos/inmunología , Pollos/virología , Mutación , Enfermedad de Newcastle/virología , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/virología , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Proteínas Virales de Fusión/administración & dosificación , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/genética , Virulencia , Replicación Viral/inmunología , Esparcimiento de Virus/inmunología
18.
Virology ; 514: 211-215, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29197721

RESUMEN

Sheep and goat pox (SGP) with peste des petits ruminants (PPR) are transboundary viral diseases of small ruminants that cause huge economic losses. Recombinant vaccines that can protect from both infections have been reported as a promising solution for the future. SGP was used as a vector to express two structural proteins hemagglutinin or the fusion protein of PPRV. We compared immunity conferred by recombinant capripoxvirus vaccines expressing H or F or both HF. Safety and efficacy were evaluated in goats and sheep. Two vaccine doses were tested in sheep, 104.5TCDI50 in 1ml dose was retained for the further experiment. Results showed that the recombinant HF confers an earlier and stronger immunity against both SGP and PPR. This recombinant vaccine protect also against the disease in exposed and unexposed sheep. The potential Differentiating Infected from Vaccinated Animals of recombinant vaccines is of great advantage in any eradication program.


Asunto(s)
Capripoxvirus/inmunología , Enfermedades de las Cabras/prevención & control , Peste de los Pequeños Rumiantes/virología , Virus de la Peste de los Pequeños Rumiantes/inmunología , Infecciones por Poxviridae/veterinaria , Enfermedades de las Ovejas/prevención & control , Proteínas Virales/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales/inmunología , Capripoxvirus/genética , Capripoxvirus/aislamiento & purificación , Capripoxvirus/fisiología , Enfermedades de las Cabras/inmunología , Enfermedades de las Cabras/virología , Cabras , Hemaglutininas/administración & dosificación , Hemaglutininas/genética , Hemaglutininas/inmunología , Virus de la Peste de los Pequeños Rumiantes/genética , Virus de la Peste de los Pequeños Rumiantes/aislamiento & purificación , Virus de la Peste de los Pequeños Rumiantes/fisiología , Infecciones por Poxviridae/inmunología , Infecciones por Poxviridae/prevención & control , Infecciones por Poxviridae/virología , Ovinos , Enfermedades de las Ovejas/inmunología , Enfermedades de las Ovejas/virología , Proteínas Virales de Fusión/administración & dosificación , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/inmunología , Proteínas Virales/administración & dosificación , Proteínas Virales/genética , Vacunas Virales/administración & dosificación , Vacunas Virales/genética
19.
Vaccine ; 35(51): 7139-7146, 2017 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-29153777

RESUMEN

Human parainfluenza virus type 3 (PIV3) is a major cause of lower respiratory disease i.e. bronchitis, bronchiolitis or pneumonia, in infants and young children. Presently there is no licensed vaccine against PIV3. To produce an effective subunit vaccine, a chimeric FHN glycoprotein consisting of the N-terminal ectodomain of the fusion (F) protein linked to the haemagglutinin-neuraminidase (HN) protein without transmembrane domain, and secreted forms of the individual F and HN glycoproteins, were expressed in mammalian cells and purified. Mice and cotton rats were immunized intramuscularly (IM) with FHN or both F and HN proteins (F + HN), formulated with poly(I:C) and an innate defense regulator peptide in polyphosphazene (TriAdj). Significantly higher levels of systemic virus-neutralizing antibodies were observed in mice and cotton rats immunized with FHN/TriAdj when compared to animals immunized with the combination of F and HN proteins (F + HN/TriAdj). As PIV3 is a pneumotropic virus, another goal is to produce an effective mucosal subunit vaccine. Intranasal (IN) administration with FHN/TriAdj resulted in mucosal IgA production in the lung and virus neutralizing antibodies in the sera. After PIV3 challenge no virus was detected in cotton rats immunized with FHN/TriAdj regardless of the route of delivery. Protective immunity against PIV3 was also induced by FHN/TriAdj in hamsters. In conclusion, the FHN protein formulated with TriAdj has potential for development of a safe and effective vaccine against PIV3.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Proteína HN/inmunología , Virus de la Parainfluenza 3 Humana/inmunología , Proteínas Virales de Fusión/inmunología , Vacunas Virales/inmunología , Adyuvantes Inmunológicos/química , Animales , Anticuerpos Neutralizantes/biosíntesis , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/sangre , Cricetinae , Proteína HN/administración & dosificación , Proteína HN/genética , Humanos , Inmunización , Ratones , Poli I-C/administración & dosificación , Poli I-C/inmunología , Polilisina/administración & dosificación , Polilisina/inmunología , Sigmodontinae , Vacunación , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/efectos adversos , Vacunas de Subunidad/genética , Vacunas de Subunidad/inmunología , Proteínas Virales de Fusión/administración & dosificación , Proteínas Virales de Fusión/genética , Vacunas Virales/administración & dosificación , Vacunas Virales/efectos adversos
20.
J Infect Dis ; 216(11): 1362-1370, 2017 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-29029260

RESUMEN

Background: Respiratory syncytial virus (RSV) is an important cause of illness in older adults. This study assessed efficacy of a vaccine for prevention of RSV-associated acute respiratory illness (ARI), defined by specified symptoms with virologic confirmation. Methods: This phase 2b study evaluated RSV postfusion F protein (120 µg) with glucopyranosyl lipid adjuvant (5 µg) in 2% stable emulsion. Subjects aged ≥60 years were randomly assigned at a ratio of 1:1 to receive vaccine or placebo (all received inactivated influenza vaccine). Ill subjects recorded symptoms and provided blood and nasal swab samples. Results: In the per-protocol population (n = 1894), the incidence of RSV-associated ARI occurring ≥14 days after dosing was 1.7% and 1.6% in the vaccine and placebo groups, respectively, for a vaccine efficacy (VE) of -7.1% (90% confidence interval [CI], -106.9%-44.3%). Efficacy was not observed in secondary analyses that included seroresponse to nonvaccine RSV antigens (VE, 8.9%; 90% CI, -28.5%-35.4%) or symptoms combined with seroresponse (VE, 10.0%; 90% CI, -45.4%-44.4%). On day 29, 92.9% of vaccinees had an anti-F immunoglobulin G antibody seroresponse. Overall, 48.5% and 30.9% of RSV vaccine recipients reported local and systemic solicited symptoms, respectively. Conclusion: The RSV vaccine was immunogenic but did not protect older adults from RSV illness. Clinical Trials Registration: NCT02508194.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio/inmunología , Vacunas contra Virus Sincitial Respiratorio/uso terapéutico , Virus Sincitial Respiratorio Humano/inmunología , Proteínas Virales de Fusión/inmunología , Adyuvantes Farmacéuticos , Anciano , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Formación de Anticuerpos , Antígenos Bacterianos , Antígenos Virales/inmunología , Método Doble Ciego , Femenino , Glucósidos/farmacología , Humanos , Inmunoglobulina G/sangre , Vacunas contra la Influenza/inmunología , Lípido A/farmacología , Masculino , Persona de Mediana Edad , Proteínas Virales de Fusión/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...