Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 529(3): 805-811, 2020 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-32736711

RESUMEN

Zika virus (ZIKV) is a mosquito-borne flavivirus associated with Congenital Zika Syndrome (CZS), reflecting a wide range of congenital abnormalities in fetuses and infants infected with ZIKV before birth. ZIKV infections have also been associated with the neurological autoimmune disorder known as Guillian-Barré syndrome (GBS). To date, no vaccines or antiviral strategies are licensed for ZIKV. We used rational design to develop a novel ZIKV vaccine candidate using a Woodchuck Hepatitis core Antigen (WHcAg) Virus-Like Particle (VLP) scaffold for displaying selected antigens from the ZIKV Envelope (E) protein. A Zika-VLP vaccine candidate containing the CD Loop sub-structural domain from ZIKV E protein Domain III (WHcAg CD Loop) elicited a strong immune response in a murine model. Analysis of serum immunoglobulins demonstrated induction of both Th1- and Th2- mediated immune response. No cross-reacting antibodies were detected between Zika, dengue and yellow fever virus, demonstrating a high level of specificity for the ZIKV CD Loop antigen. Immunization with the WHcAg CD Loop vaccine candidate demonstrated immunoprotection in a murine model of ZIKV infection, stimulating protective antibodies associated with antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) activities. The WHcAg CD Loop candidate may represent a safer vaccine for preventing antibody dependent enhancement (ADE).


Asunto(s)
Vacunas de Partículas Similares a Virus/uso terapéutico , Proteínas del Envoltorio Viral/uso terapéutico , Infección por el Virus Zika/prevención & control , Virus Zika/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Inmunidad , Ratones , Ratones Endogámicos C57BL , Vacunas de Partículas Similares a Virus/inmunología , Proteínas del Envoltorio Viral/inmunología , Infección por el Virus Zika/inmunología
2.
Cell Mol Life Sci ; 77(21): 4315-4324, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32367191

RESUMEN

Epstein-Barr virus (EBV) was the first human tumor virus being discovered and remains to date the only human pathogen that can transform cells in vitro. 55 years of EBV research have now brought us to the brink of an EBV vaccine. For this purpose, recombinant viral vectors and their heterologous prime-boost vaccinations, EBV-derived virus-like particles and viral envelope glycoprotein formulations are explored and are discussed in this review. Even so, cell-mediated immune control by cytotoxic lymphocytes protects healthy virus carriers from EBV-associated malignancies, antibodies might be able to prevent symptomatic primary infection, the most likely EBV-associated pathology against which EBV vaccines will be initially tested. Thus, the variety of EBV vaccines reflects the sophisticated life cycle of this human tumor virus and only vaccination in humans will finally be able to reveal the efficacy of these candidates. Nevertheless, the recently renewed efforts to develop an EBV vaccine and the long history of safe adoptive T cell transfer to treat EBV-associated malignancies suggest that this oncogenic γ-herpesvirus can be targeted by immunotherapies. Such vaccination should ideally implement the very same immune control that protects healthy EBV carriers.


Asunto(s)
Infecciones por Virus de Epstein-Barr/prevención & control , Herpesvirus Humano 4/inmunología , Vacunas contra Herpesvirus/uso terapéutico , Animales , Anticuerpos Neutralizantes/inmunología , Infecciones por Virus de Epstein-Barr/inmunología , Vacunas contra Herpesvirus/inmunología , Humanos , Vacunación , Vacunas de Partículas Similares a Virus/inmunología , Vacunas de Partículas Similares a Virus/uso terapéutico , Proteínas del Envoltorio Viral/inmunología , Proteínas del Envoltorio Viral/uso terapéutico
3.
Rev Argent Microbiol ; 51(2): 119-129, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30385072

RESUMEN

Equid alphaherpesvirus 1 (EHV-1) infection causes abortion, respiratory disease, perinatal deaths and neurological disorders in horses. The natural infection and available vaccines provide only partial and short-lived protection against reinfections. In the present study, we analyzed the ability of purified baculovirus-expressed glycoprotein D (gD) administered by different routes to induce protective immunity in BALB/c mice after challenge with the EHV-1 AR8 strain. Clinical signs varied among the different groups of mice immunized by parenteral routes, and, although gD induced a specific serum IgG response, it did not prevent the virus from reaching the lungs. Intranasally immunized mice showed no clinical signs, and virus isolation from lungs, histological lesions and antigen detection by immunohistochemistry were negative. In addition, by this route, gD did not stimulate the production of serum IgG and IgA. However, a specific IgA response in the respiratory tract was confirmed in intranasally immunized mice. Thus, we conclude that the mucosal immune response could reduce the initial viral attachment and prevent the virus from reaching the lungs. Our findings provide additional data to further study new immunization strategies in the natural host.


Asunto(s)
Infecciones por Herpesviridae/prevención & control , Herpesvirus Équido 1 , Proteínas del Envoltorio Viral/uso terapéutico , Animales , Modelos Animales de Enfermedad , Inmunización , Ratones , Ratones Endogámicos BALB C , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/uso terapéutico , Proteínas del Envoltorio Viral/inmunología
4.
Sci Rep ; 8(1): 10990, 2018 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-30030466

RESUMEN

Despite recent improvements in the engineering of viral envelope proteins, it remains a significant challenge to create lentiviral vectors that allow targeted transduction to specific cell populations of interest. In this study, we developed a simple 'plug and play' strategy to retarget lentiviral vectors to any desired cell types through in vitro covalent modification of the virions with specific cell-targeting proteins (CTPs). This strategy exploits a disulfide bond-forming protein-peptide pair PDZ1 and its pentapeptide ligand (ThrGluPheCysAla, TEFCA). PDZ1 was incorporated into an engineered Sindbis virus envelope protein (Sind-PDZ1) and displayed on lentiviral particles while the TEFCA pentapeptide ligand was genetically linked to the CTP. Her2/neu-binding designed ankyrin repeat proteins (DARPin) were used as our model CTPs. DARPin-functionalized unconcentrated lentiviral vectors harboring Sind-PDZ1 envelope protein (Sind-PDZ1-pp) exhibited >800-fold higher infectious titer in HER2+ cells than the unfunctionalized virions (8.5 × 106 vs. <104 IU/mL). Moreover, by virtue of the covalent disulfide bond interaction between PDZ1 and TEFCA, the association of the CTP with the virions is nonreversible under non-reducing conditions (e.g. serum), making these functionalized virions potentially stable in an in vivo setting.


Asunto(s)
Vectores Genéticos , Lentivirus/genética , Transducción Genética/métodos , Línea Celular Tumoral , Disulfuros/química , Técnicas de Transferencia de Gen , Humanos , Péptidos/química , Proteínas/química , Receptor ErbB-2 , Virus Sindbis/química , Proteínas del Envoltorio Viral/uso terapéutico , Virión
5.
Clin Immunol ; 191: 37-43, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29567431

RESUMEN

Retroviruses including Human Endogenous Retroviruses (HERVs), contain a conserved region with highly immunomodulatory functions in the transmembrane proteins in envelope gene (env) named immunosuppressive domain (ISU). In this report, we demonstrate that Env59-GP3 peptide holds therapeutic potential in a mouse model of myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE). The results show that this specific HERV-H derived ISU peptide, but not peptide derived from another env gene HERV-K, decreased the development of EAE in C57BL/6 mice, accompanied by reduced demyelination and inhibition of inflammatory cells. Moreover, here we tested the effect of peptides on macrophages differentiation. The treatment with Env59-GPS peptide modulate the pro-inflammatory M1 profile and anti-inflammatory M2 macrophages, being shown by inhibiting inflammatory M1 hallmark genes/cytokines expression and enhancing expression of M2 associated markers. These results demonstrate that Env59-GP3 ISU peptide has therapeutic potential in EAE possibly through inducing the polarization of M2 macrophages and inhibiting inflammatory responses.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Retrovirus Endógenos/fisiología , Inmunosupresores/uso terapéutico , Esclerosis Múltiple/tratamiento farmacológico , Animales , Polaridad Celular , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/fisiología , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/etiología , Esclerosis Múltiple/inmunología , Péptidos/uso terapéutico , Médula Espinal/patología , Proteínas del Envoltorio Viral/uso terapéutico
6.
Sci Rep ; 7: 45886, 2017 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-28397835

RESUMEN

Recently, progress has been made in the development of vaccines and monoclonal antibody cocktails that target the Ebola coat glycoprotein (GP). Based on the mutation rates for Ebola virus given its natural sequence evolution, these treatment strategies are likely to impose additional selection pressure to drive acquisition of mutations in GP that escape neutralization. Given the high degree of sequence conservation among GP of Ebola viruses, it would be challenging to determine the propensity of acquiring mutations in response to vaccine or treatment with one or a cocktail of monoclonal antibodies. In this study, we analyzed the mutability of each residue using an approach that captures the structural constraints on mutability based on the extent of its inter-residue interaction network within the three-dimensional structure of the trimeric GP. This analysis showed two distinct clusters of highly networked residues along the GP1-GP2 interface, part of which overlapped with epitope surfaces of known neutralizing antibodies. This network approach also permitted us to identify additional residues in the network of the known hotspot residues of different anti-Ebola antibodies that would impact antibody-epitope interactions.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Glicoproteínas/inmunología , Fiebre Hemorrágica Ebola/prevención & control , Proteínas del Envoltorio Viral/genética , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/uso terapéutico , Vacunas contra el Virus del Ébola/genética , Vacunas contra el Virus del Ébola/inmunología , Ebolavirus/inmunología , Ebolavirus/patogenicidad , Epítopos/genética , Epítopos/inmunología , Glicoproteínas/genética , Glicoproteínas/uso terapéutico , Fiebre Hemorrágica Ebola/inmunología , Fiebre Hemorrágica Ebola/virología , Humanos , Mutación , Pruebas de Neutralización , Proteínas del Envoltorio Viral/inmunología , Proteínas del Envoltorio Viral/uso terapéutico
7.
FASEB J ; 31(3): 1179-1192, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27998907

RESUMEN

Increasing evidence shows that CC-chemokines promote inflammatory-driven angiogenesis, with little to no effect on hypoxia-mediated angiogenesis. Inhibition of the CC-chemokine class may therefore affect angiogenesis differently depending on the pathophysiological context. We compared the effect of CC-chemokine inhibition in inflammatory and physiological conditions. In vitro, the broad-spectrum CC-chemokine inhibitor "35K" inhibited inflammatory-induced endothelial cell proliferation, migration, and tubulogenesis, with more modest effects in hypoxia. In vivo, adenoviruses were used to overexpress 35K (Ad35K) and GFP (AdGFP, control virus). Plasma chemokine activity was suppressed by Ad35K in both models. In the periarterial femoral cuff model of inflammatory-driven angiogenesis, overexpression of 35K inhibited adventitial neovessel formation compared with control AdGFP-infused mice. In contrast, 35K preserved neovascularization in the hindlimb ischemia model and had no effect on physiological neovascularization in the chick chorioallantoic membrane assay. Mechanistically, 2 key angiogenic proteins (VEGF and hypoxia-inducible factor-1α) were conditionally regulated by 35K, such that expression was inhibited in inflammation but was unchanged in hypoxia. In conclusion, CC-chemokine inhibition by 35K suppresses inflammatory-driven angiogenesis while preserving physiological ischemia-mediated angiogenesis via conditional regulation of VEGF and hypoxia-inducible factor-1α. CC-chemokine inhibition may be an alternative therapeutic strategy for suppressing diseases associated with inflammatory angiogenesis without inducing the side effects caused by global inhibition.- Ridiandries, A., Tan, J. T. M., Ravindran, D., Williams, H., Medbury, H. J., Lindsay, L., Hawkins, C., Prosser, H. C. G., Bursill, C. A. CC-chemokine class inhibition attenuates pathological angiogenesis while preserving physiological angiogenesis.


Asunto(s)
Quimiocinas CC/antagonistas & inhibidores , Endotelio Vascular/efectos de los fármacos , Neovascularización Patológica/metabolismo , Proteínas del Envoltorio Viral/farmacología , Animales , Movimiento Celular , Proliferación Celular , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/tratamiento farmacológico , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas del Envoltorio Viral/uso terapéutico
8.
Vet Res ; 47(1): 64, 2016 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-27301745

RESUMEN

Classical swine fever (CSF) is a highly contagious swine disease caused by classical swine fever virus (CSFV). Previously, we demonstrated that rAdV-SFV-E2, an adenovirus-delivered, Semliki Forest virus replicon-vectored marker vaccine against CSF, is able to protect pigs against lethal CSFV challenge. From an economical point of view, it will be beneficial to reduce the minimum effective dose of the vaccine. This study was designed to test the adjuvant effects of Salmonella enteritidis-derived bacterial ghosts (BG) to enhance the protective immunity of rAdV-SFV-E2 in pigs. Groups of 5-week-old pigs (n = 4) were immunized intramuscularly twice with 10(5) median tissue culture infective doses (TCID50) rAdV-SFV-E2 combined with 10(10) colony forming units (CFU) BG, 10(6) or 10(5) TCID50 rAdV-SFV-E2 alone or 10(10) CFU BG alone at an interval of 3 weeks, and challenged with the highly virulent CSFV Shimen strain at 1 week post-booster immunization. The results show that the pigs inoculated with 10(5) TCID50 rAdV-SFV-E2 plus BG or 10(6) TCID50 rAdV-SFV-E2 alone were completely protected from lethal CSFV challenge, in contrast with the pigs vaccinated with 10(5) TCID50 rAdV-SFV-E2 or BG alone, which displayed partial or no protection following virulent challenge. The data indicate that BG are a promising adjuvant to enhance the efficacy of rAdV-SFV-E2 and possibly other vaccines.


Asunto(s)
Virus de la Fiebre Porcina Clásica/inmunología , Peste Porcina Clásica/prevención & control , Salmonella enteritidis , Vacunas Virales/uso terapéutico , Adyuvantes Inmunológicos/uso terapéutico , Animales , Formación de Anticuerpos/inmunología , Peste Porcina Clásica/inmunología , Virus de los Bosques Semliki , Porcinos , Proteínas del Envoltorio Viral/inmunología , Proteínas del Envoltorio Viral/uso terapéutico , Vacunas Virales/inmunología
9.
EBioMedicine ; 5: 59-67, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27077112

RESUMEN

Although counteracting innate defenses allows oncolytic viruses (OVs) to better replicate and spread within tumors, CD8(+) T-cells restrict their capacity to trigger systemic anti-tumor immune responses. Herpes simplex virus-1 (HSV-1) evades CD8(+) T-cells by producing ICP47, which limits immune recognition of infected cells by inhibiting the transporter associated with antigen processing (TAP). Surprisingly, removing ICP47 was assumed to benefit OV immuno-therapy, but the impact of inhibiting TAP remains unknown because human HSV-1 ICP47 is not effective in rodents. Here, we engineer an HSV-1 OV to produce bovine herpesvirus UL49.5, which unlike ICP47, antagonizes rodent and human TAP. Significantly, UL49.5-expressing OVs showed superior efficacy treating bladder and breast cancer in murine models that was dependent upon CD8(+) T-cells. Besides injected subcutaneous tumors, UL49.5-OV reduced untreated, contralateral tumor size and metastases. These findings establish TAP inhibitor-armed OVs that evade CD8(+) T-cells as an immunotherapy strategy to elicit potent local and systemic anti-tumor responses.


Asunto(s)
Neoplasias de la Mama/inmunología , Viroterapia Oncolítica , Virus Oncolíticos/inmunología , Neoplasias de la Vejiga Urinaria/inmunología , Proteínas del Envoltorio Viral/inmunología , Animales , Neoplasias de la Mama/terapia , Linfocitos T CD8-positivos/inmunología , Bovinos , Línea Celular Tumoral , Modelos Animales de Enfermedad , Herpesvirus Humano 1/inmunología , Humanos , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/inmunología , Evasión Inmune/genética , Ratones , Virus Oncolíticos/genética , Neoplasias de la Vejiga Urinaria/terapia , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/uso terapéutico
10.
PLoS Pathog ; 11(5): e1004907, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25996983

RESUMEN

Herpes simplex virus (HSV) enters cells by means of four essential glycoproteins - gD, gH/gL, gB, activated in a cascade fashion by gD binding to one of its receptors, nectin1 and HVEM. We report that the engineering in gH of a heterologous ligand - a single-chain antibody (scFv) to the cancer-specific HER2 receptor - expands the HSV tropism to cells which express HER2 as the sole receptor. The significance of this finding is twofold. It impacts on our understanding of HSV entry mechanism and the design of retargeted oncolytic-HSVs. Specifically, entry of the recombinant viruses carrying the scFv-HER2-gH chimera into HER2+ cells occurred in the absence of gD receptors, or upon deletion of key residues in gD that constitute the nectin1/HVEM binding sites. In essence, the scFv in gH substituted for gD-mediated activation and rendered a functional gD non-essential for entry via HER2. The activation of the gH moiety in the chimera was carried out by the scFv in cis, not in trans as it occurs with wt-gD. With respect to the design of oncolytic-HSVs, previous retargeting strategies were based exclusively on insertion in gD of ligands to cancer-specific receptors. The current findings show that (i) gH accepts a heterologous ligand. The viruses retargeted via gH (ii) do not require the gD-dependent activation, and (iii) replicate and kill cells at high efficiency. Thus, gH represents an additional tool for the design of fully-virulent oncolytic-HSVs retargeted to cancer receptors and detargeted from gD receptors.


Asunto(s)
ADN Recombinante/metabolismo , Receptor ErbB-2/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Simplexvirus/fisiología , Anticuerpos de Cadena Única/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Tropismo Viral , Internalización del Virus , Animales , Sitios de Unión , Línea Celular , Supervivencia Celular , ADN Recombinante/química , ADN Recombinante/uso terapéutico , Terapia Genética , Humanos , Ligandos , Mutación , Neoplasias/metabolismo , Neoplasias/terapia , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/uso terapéutico , Ingeniería de Proteínas , Receptor ErbB-2/química , Receptor ErbB-2/genética , Receptor ErbB-2/uso terapéutico , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/uso terapéutico , Transducción de Señal , Anticuerpos de Cadena Única/química , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/uso terapéutico , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/uso terapéutico
11.
Arch Virol ; 160(6): 1543-7, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25877819

RESUMEN

Porcine reproductive and respiratory syndrome (PRRS) is an economically devastating viral disease of pigs. Safer and more effective vaccines are urgently needed. In this study, a synthetic ORF5 gene of porcine reproductive and respiratory syndrome virus (PRRSV) was adsorbed onto poly(D, L-lactide-co-glycolide)/polyethylenimine (PLGA/PEI) nanoparticles. We prepared a PLGA-nanoparticle-adsorbed PRRSV DNA vaccine and a PEI-DNA complex. The results showed that these model vaccines could significantly enhance humoral and cellular immune responses when compared with the responses induced by pcDNA3.1-SynORF5, a plasmid construct for expression of PRRSV ORF5. PLGA-branched PEI nanoparticles induced the most efficient immune response. The delivery system and adjuvant provide new models for the development of vaccines against PRRSV.


Asunto(s)
Nanopartículas/uso terapéutico , Síndrome Respiratorio y de la Reproducción Porcina/prevención & control , Virus del Síndrome Respiratorio y Reproductivo Porcino/inmunología , Vacunas de ADN/uso terapéutico , Animales , Inmunidad Celular/inmunología , Inmunidad Humoral/inmunología , Ratones , Ratones Endogámicos BALB C , Síndrome Respiratorio y de la Reproducción Porcina/inmunología , Porcinos , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/uso terapéutico
12.
Arch Virol ; 158(6): 1275-85, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23392631

RESUMEN

Porcine reproductive and respiratory syndrome virus (PRRSV) induces reproductive failure in sows and respiratory problems in pigs of all ages. Live attenuated and inactivated vaccines are used on swine farms to control PRRSV. However, their protective efficacy against field strains of PRRSV remains questionable. New vaccines have been developed to improve the efficacy of these traditional vaccines. In this study, virus-like particles (VLPs) composed of the GP5 and M proteins of PRRSV were developed, and the capacity of the VLPs to elicit antigen-specific immunity was evaluated. Serum antibody titers and production of cytokines were measured in BALB/C mice immunized intramuscularly three times with different doses (0.5, 1.0, 2.0, and 4.0 µg) of the VLP vaccine. A commercial vaccine consisting of inactivated PRRSV and phosphate-buffered saline (PBS) were used as positive and negative controls, respectively. IgG titers to GP5 were significantly higher in all groups of mice vaccinated with the VLPs than in control mice. Neutralizing antibodies were only detected in mice vaccinated with 2.0 and 4.0 µg of the VLPs. Cytokine levels were determined in cell culture supernatants after in vitro stimulation of splenocytes with the VLPs for 3 days. Mice immunized with 4.0 µg of the VLPs produced a significantly higher amount of interferon-gamma (IFN-γ) than mice immunized with the commercial inactivated PRRSV vaccine and PBS. In contrast, immunization with the commercial vaccine induced higher production of IL-4 and IL-10 in mice than mice vaccinated with VLPs. These data together demonstrate the capacity of VLPs to induce both neutralizing antibodies and IFN-γ in immunized mice. The VLP vaccine developed in this study could serve as a platform for the generation of improved VLP vaccines to control PRRSV.


Asunto(s)
Virus del Síndrome Respiratorio y Reproductivo Porcino/inmunología , Vacunas de Partículas Similares a Virus/uso terapéutico , Proteínas del Envoltorio Viral/uso terapéutico , Proteínas de la Matriz Viral/uso terapéutico , Vacunas Virales/uso terapéutico , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Western Blotting , Relación Dosis-Respuesta Inmunológica , Femenino , Inmunidad Humoral/inmunología , Interferón gamma/sangre , Interleucina-10/sangre , Interleucina-4/sangre , Ratones , Ratones Endogámicos BALB C , Pruebas de Neutralización , Vacunas de Partículas Similares a Virus/inmunología , Proteínas del Envoltorio Viral/inmunología , Proteínas de la Matriz Viral/inmunología , Vacunas Virales/inmunología
13.
Int J Pharm ; 440(2): 229-37, 2013 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-22743007

RESUMEN

The purpose of this study was to investigate the potential of intranasal immunization with non-ionic surfactant vesicles (NISV) containing either the secretory recombinant form of glycoprotein B (gBs) of herpes simplex virus type 1 or a related polylysine reach peptides (DTK) for induction of protective immunity against genital herpes infection in mice. NISV were prepared by lipid film hydration method. The mean diameter of vesicles was around 390 nm for DTK-containing NISV (DTK-NISV) and 320 nm for gB1s-containing NISV (gB1s-NISV). The encapsulation efficiency of the molecules was comprised between 57% and 70%. After 7-14 day NISV maintained stable dimensions and a drug encapsulation higher than 48%. We showed that intranasal immunization with gB1s-NISV induces gB-specific IgG antibody and lymphoproliferative responses, whereas vaccination with DTK-NISV was not able to generate a gB-specific immune response. Our results indicate that vaccination of BALB/c mice with gB1s-NISV induced Th1 responses, as characterized by increased titre of IG2a in plasma and IFN-production in CD4+ splenic cells. Intranasal immunization with gB1s-NISV could elicit 90% (almost complete) protection against a heterologous lethal vaginal challenge with herpes simplex virus type 2. These data may have implications for the development of a mucosal vaccine against genital herpes.


Asunto(s)
Herpes Genital/prevención & control , Vacunas contra el Virus del Herpes Simple/uso terapéutico , Inmunización/métodos , Liposomas/uso terapéutico , Tensoactivos/uso terapéutico , Administración Intranasal , Animales , Anticuerpos Antivirales/sangre , Chlorocebus aethiops , Citocinas/metabolismo , Proteínas de Drosophila/administración & dosificación , Proteínas de Drosophila/inmunología , Herpes Genital/sangre , Herpes Genital/inmunología , Vacunas contra el Virus del Herpes Simple/administración & dosificación , Vacunas contra el Virus del Herpes Simple/inmunología , Herpesvirus Humano 2/inmunología , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulina G/metabolismo , Liposomas/administración & dosificación , Liposomas/síntesis química , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Tamaño de la Partícula , Precursores de Proteínas/administración & dosificación , Precursores de Proteínas/inmunología , Bazo/inmunología , Bazo/metabolismo , Tensoactivos/administración & dosificación , Tensoactivos/química , Taquicininas/administración & dosificación , Taquicininas/inmunología , Células Vero , Proteínas del Envoltorio Viral/administración & dosificación , Proteínas del Envoltorio Viral/inmunología , Proteínas del Envoltorio Viral/uso terapéutico
14.
Acta Pharm ; 60(4): 407-13, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21169133

RESUMEN

Future cancer therapies will be molecular cures. They will correct, block or destroy cancer cells by targeting molecular changes that lead to carcinogenesis. Destroying cancer cells can be done using oncolytic viruses. By blocking antibody mediated neutralization of oncolytic viruses, Herpes simplex virus type 1 glycoproteins E and I could be used in the adjuvant treatment of cancer for improving the chances of oncolytic viruses to kill cancer cells in vivo.


Asunto(s)
Anticuerpos Bloqueadores/inmunología , Anticuerpos Neutralizantes/inmunología , Neoplasias/terapia , Viroterapia Oncolítica , Virus Oncolíticos/fisiología , Proteínas del Envoltorio Viral/uso terapéutico , Animales , Anticuerpos Antivirales/inmunología , Ensayos Clínicos como Asunto , Herpesvirus Humano 1 , Humanos , Neoplasias/tratamiento farmacológico , Virus Oncolíticos/inmunología , Proteínas del Envoltorio Viral/inmunología , Proteínas del Envoltorio Viral/metabolismo
15.
Protein Expr Purif ; 74(1): 129-37, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20600950

RESUMEN

Arthropod-borne flaviviruses such as dengue virus (DENV) and West Nile virus (WNV) pose significant health threats to the global community. Due to escalating numbers of DENV and WNV infections worldwide, development of an effective vaccine remains a global health priority. As flavivirus envelope Domain III (DIII) protein is highly immunogenic and capable of inducing neutralizing antibodies against wild-type virus, it is both a potential protein subunit vaccine candidate and a suitable diagnostic reagent. Here, we describe the use of metal affinity membrane chromatography as a rapid and improved alternative for the purification of recombinant DIII (rDIII) antigens from DENV serotypes 1-4 and WNV - New York, Sarafend, Wengler and Kunjin strains. Optimum conditions for the expression, solubilization, renaturation and purification of these proteins were established. The purified proteins were confirmed by MALDI-TOF mass spectrometry and ELISA using antibodies raised against the respective viruses. Biological function of the purified rDIII proteins was confirmed by their ability to generate DIII-specific antibodies in mice that could neutralize the virus.


Asunto(s)
Antígenos Virales/aislamiento & purificación , Cromatografía de Afinidad/métodos , Virus del Dengue/genética , Dengue/prevención & control , Proteínas del Envoltorio Viral/aislamiento & purificación , Fiebre del Nilo Occidental/prevención & control , Virus del Nilo Occidental/genética , Animales , Antígenos Virales/genética , Antígenos Virales/uso terapéutico , Línea Celular , Cromatografía de Afinidad/economía , Culicidae/genética , Escherichia coli/genética , Femenino , Inmunización , Ratones , Ratones Endogámicos BALB C , Plásmidos/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/uso terapéutico , Factores de Tiempo , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/uso terapéutico
16.
Vet Res Commun ; 34(4): 371-80, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20432066

RESUMEN

To develop a new type vaccine for porcine reproductive and respiratory syndrome (PRRS) prevention by using canine adenovirus 2(CAV-2) as vector, the Glycoprotein 5(GP5) gene from PRRSV strain JL was amplified by RT-PCR, and the expression cassette of GP5 was constructed using the human cytomegalovirus (HCMV) promoter and the simian virus 40 (SV40) early mRNA polyadenylation signal. The expression cassette of Glycoprotein 5 was cloned into the CAV-2 genome in which E3 region had been partly deleted, and the recombinant virus (CAV-2-GP5) was obtained by transfecting the recombinant CAV-2-GP5 genome into MDCK cells together with Lipofectamine 2000. Immunization trial in pigs with the recombinant virus CAV-2-GP5 showed that CAV-2-GP5 could stimulate a specific immune response to PRRSV. Immune response to the GP5 and PRRSV was confirmed by ELISA, neutralization test and lymphocyte proliferative responses, and western blotting confirmed expression of GP5 by the vector in cells. These results indicated that CAV-2 may serve as a vector for development of PRRSV vaccine in pigs, and the CAV-2-GP5 might be a candidate vaccine to be tested for preventing PRRSV infection.


Asunto(s)
Adenovirus Caninos/genética , Síndrome Respiratorio y de la Reproducción Porcina/inmunología , Proteínas Recombinantes/inmunología , Porcinos/inmunología , Proteínas del Envoltorio Viral/inmunología , Animales , Clonación Molecular , Citomegalovirus/genética , Perros , Ensayo de Inmunoadsorción Enzimática , Vectores Genéticos/genética , Genoma Viral , Humanos , Virus del Síndrome Respiratorio y Reproductivo Porcino , Regiones Promotoras Genéticas , ARN Mensajero/genética , Proteínas Recombinantes/uso terapéutico , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Virus 40 de los Simios/genética , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/uso terapéutico
17.
FEBS J ; 275(22): 5714-24, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18959756

RESUMEN

We previously developed the bio-nanocapsule, which consists of hepatitis B virus envelope L proteins. The bio-nanocapsule can be used to deliver genes and drugs specifically to the human liver-derived tissues in xenograft models, presumably by utilizing the human liver-specific mechanism of hepatitis B virus infection. The hepatitis B virus tropism is highly restricted to humans and higher primates. Thus, to evaluate the in vivo therapeutic effects of forthcoming bio-nanocapsule-based medicines, it will be crucial to develop an animal model whose liver is susceptible to both bio-nanocapsule and hepatitis B virus. In the present study, we aimed to establish a bio-nanocapsule-susceptible animal model using transgenic rats expressing squamous cell carcinoma antigen-1 (SCCA1), which has been proposed to be a receptor for hepatitis B virus, interacting with the hepatitis B virus envelope protein and enhancing the cellular uptake of hepatitis B virus. We show that the recombinant SCCA1 protein interacts directly with bio-nanocapsule and inhibits its attachment to the cultured human liver-derived cells. Furthermore, we have established a transgenic rat that specifically expresses SCCA1 in the liver and also demonstrate that the amount of bio-nanocapsule accumulated in the liver is significantly increased by the SCCA1 expression. Histological analysis suggests that bio-nanocapsule is preferentially incorporated into the SCCA1-expressing hepatocytes but not into macrophages, such as Küppfer cells, nor into endothelial cells. Therefore, this animal model is expected to be useful for the development of bio-nanocapsule-based medicines.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Hígado/metabolismo , Nanocápsulas/uso terapéutico , Serpinas/metabolismo , Proteínas del Envoltorio Viral/farmacocinética , Animales , Virus de la Hepatitis B/química , Humanos , Nanocápsulas/virología , Ratas , Ratas Transgénicas , Distribución Tisular , Proteínas del Envoltorio Viral/uso terapéutico
19.
J Virol ; 80(12): 5757-67, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16731915

RESUMEN

The spike (S) glycoprotein of severe acute respiratory syndrome coronavirus (SARS-CoV) mediates the receptor interaction and immune recognition and is considered a major target for vaccine design. However, its antigenic and immunogenic properties remain to be elucidated. In this study, we immunized mice with full-length S protein (FL-S) or its extracellular domain (EC-S) expressed by recombinant baculoviruses in insect cells. We found that the immunized mice developed high titers of anti-S antibodies with potent neutralizing activities against SARS pseudoviruses constructed with the S proteins of Tor2, GD03T13, and SZ3, the representative strains of 2002 to 2003 and 2003 to 2004 human SARS-CoV and palm civet SARS-CoV, respectively. These data suggest that the recombinant baculovirus-expressed S protein vaccines possess excellent immunogenicity, thereby inducing highly potent neutralizing responses against human and animal SARS-CoV variants. The antigenic structure of the S protein was characterized by a panel of 38 monoclonal antibodies (MAbs) isolated from the immunized mice. The epitopes of most anti-S MAbs (32 of 38) were localized within the S1 domain, and those of the remaining 6 MAbs were mapped to the S2 domain. Among the anti-S1 MAbs, 17 MAbs targeted the N-terminal region (amino acids [aa] 12 to 327), 9 MAbs recognized the receptor-binding domain (RBD; aa 318 to 510), and 6 MAbs reacted with the C-terminal region of S1 domain that contains the major immunodominant site (aa 528 to 635). Strikingly, all of the RBD-specific MAbs had potent neutralizing activity, 6 of which efficiently blocked the receptor binding, confirming that the RBD contains the main neutralizing epitopes and that blockage of the receptor association is the major mechanism of SARS-CoV neutralization. Five MAbs specific for the S1 N-terminal region exhibited moderate neutralizing activity, but none of the MAbs reacting with the S2 domain and the major immunodominant site in S1 showed neutralizing activity. All of the neutralizing MAbs recognize conformational epitopes. These data provide important information for understanding the antigenicity and immunogenicity of S protein and for designing SARS vaccines. This panel of anti-S MAbs can be used as tools for studying the structure and function of the SARS-CoV S protein.


Asunto(s)
Glicoproteínas de Membrana/inmunología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/inmunología , Proteínas del Envoltorio Viral/inmunología , Vacunas Virales , Animales , Anticuerpos Monoclonales , Anticuerpos Antivirales/biosíntesis , Formación de Anticuerpos/efectos de los fármacos , Antígenos Virales , Baculoviridae/genética , Clonación Molecular , Mapeo Epitopo , Humanos , Inmunización , Glicoproteínas de Membrana/administración & dosificación , Glicoproteínas de Membrana/uso terapéutico , Ratones , Proteínas Recombinantes , Glicoproteína de la Espiga del Coronavirus , Proteínas del Envoltorio Viral/administración & dosificación , Proteínas del Envoltorio Viral/uso terapéutico
20.
Mol Ther ; 12(5): 813-23, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16109508

RESUMEN

Previous studies conducted in our laboratory with Sindbis viral vectors in animal models demonstrated excellent in vivo targeting of tumor cells and significant reduction of metastatic implant size. To explore the influence of Sindbis strain on these factors, we constructed new plasmids from the wild-type Ar-339 Sindbis virus strain and compared their sequences. We found differences in the replicase and envelope proteins between JT, HRSP, and Ar-339 sequences. We made chimeras combining both strains and studied their efficiency in SCID mice bearing tumor xenograft using IVIS in vivo imaging techniques. We found that JT envelope proteins targeted tumors more efficiently than those of Ar-339, while the Ar-339 replicase showed increased efficacy in tumor reduction. To determine which residues are responsible for tumor targeting, we made mutants of Ar-339 E2 envelope protein and tested them by IVIS imaging in ES-2 tumor-bearing and tumor-free mice. The change of only one amino acid from E70 to K70 in Ar-339 E2 suppressed the ability to target metastatic tumor implants in mice. A K70 and V251 double E2 mutant did not reverse the loss of targeting capability. Only the mutant with JT E2 and Ar-339 helper targeted tumor, though with less intensity.


Asunto(s)
Vectores Genéticos , Metástasis de la Neoplasia/terapia , Plásmidos/genética , Virus Sindbis/genética , Proteínas del Envoltorio Viral/química , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Ratones , Ratones SCID , Mutación , Metástasis de la Neoplasia/patología , Transporte de Proteínas , Análisis de Secuencia de ADN , Transfección , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...