Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 139
Filtrar
1.
Protein Expr Purif ; 190: 106010, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34737040

RESUMEN

Recombinantly expressed VP1 of norovirus self-assembled and formed norovirus-like particles (NoV-LPs). This native VP1 was expressed using the Bombyx mori nucleopolyhedrovirus (BmNPV) expression system in silkworm larva. NoV-LPs were collected from silkworm fat body lysate by density gradient centrifugation. To improve the purity of the NoV-LP, the proteins were further purified using immobilized metal affinity chromatography based on the surface exposed side chain of histidine residues. The additional purification led to a highly purified virus-like particle (VLP). The morphology and size of the purified VLPs were examined using a transmission electron microscope, and dynamic light scattering revealed a monodispersed spherical morphology with a diameter of 34 nm. The purified product had a purity of >90% with a recovery yield of 48.7% (equivalent to 930 µg) from crude lysate, obtained from seven silkworm larvae. In addition, the purified VLP could be recognized by antibodies against GII norovirus in sandwich enzyme-linked immunosorbent assay, which indicated that the silkworm-derived VLP is biologically functional as a NoV-LP in its native state, is structurally correct, and exerts its biological function. Our results suggest that the silkworm-derived NoV-LP may be useful for subsequent applications, such as in a vaccine platform. Moreover, the silkworm-based expression system is known for its robustness, facile up-scalability, and relatively low expense compared to insect cell systems.


Asunto(s)
Partículas Similares a Virus Artificiales/química , Bombyx/genética , Norovirus , Proteínas Virales de Fusión , Animales , Bombyx/metabolismo , Norovirus/química , Norovirus/genética , Norovirus/aislamiento & purificación , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Virales de Fusión/biosíntesis , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/aislamiento & purificación
2.
Sci Rep ; 10(1): 4746, 2020 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-32179788

RESUMEN

Ginkgolic acids (GA) are alkylphenol constituents of the leaves and fruits of Ginkgo biloba. GA has shown pleiotropic effects in vitro, including: antitumor effects through inhibition of lipogenesis; decreased expression of invasion associated proteins through AMPK activation; and potential rescue of amyloid-ß (Aß) induced synaptic impairment. GA was also reported to have activity against Escherichia coli and Staphylococcus aureus. Several mechanisms for this activity have been suggested including: SUMOylation inhibition; blocking formation of the E1-SUMO intermediate; inhibition of fatty acid synthase; non-specific SIRT inhibition; and activation of protein phosphatase type-2C. Here we report that GA inhibits Herpes simplex virus type 1 (HSV-1) by inhibition of both fusion and viral protein synthesis. Additionally, we report that GA inhibits human cytomegalovirus (HCMV) genome replication and Zika virus (ZIKV) infection of normal human astrocytes (NHA). We show a broad spectrum of fusion inhibition by GA of all three classes of fusion proteins including HIV, Ebola virus (EBOV), influenza A virus (IAV) and Epstein Barr virus (EBV). In addition, we show inhibition of a non-enveloped adenovirus. Our experiments suggest that GA inhibits virion entry by blocking the initial fusion event. Data showing inhibition of HSV-1 and CMV replication, when GA is administered post-infection, suggest a possible secondary mechanism targeting protein and DNA synthesis. Thus, in light of the strong effect of GA on viral infection, even after the infection begins, it may potentially be used to treat acute infections (e.g. Coronavirus, EBOV, ZIKV, IAV and measles), and also topically for the successful treatment of active lesions (e.g. HSV-1, HSV-2 and varicella-zoster virus (VZV)).


Asunto(s)
Antivirales/farmacología , Infecciones por Virus ADN/metabolismo , Virus ADN/efectos de los fármacos , Infecciones por Virus ARN/metabolismo , Virus ARN/efectos de los fármacos , Salicilatos/farmacología , Proteínas del Envoltorio Viral/antagonistas & inhibidores , Proteínas Virales de Fusión/antagonistas & inhibidores , Animales , Astrocitos/metabolismo , Chlorocebus aethiops , Replicación del ADN/efectos de los fármacos , Infecciones por Virus ADN/virología , Virus ADN/genética , ADN Viral/genética , Células HEK293 , Humanos , Infecciones por Virus ARN/virología , Virus ARN/genética , Células Vero , Proteínas del Envoltorio Viral/biosíntesis , Proteínas Virales de Fusión/biosíntesis , Virión/efectos de los fármacos , Internalización del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos
3.
Mater Sci Eng C Mater Biol Appl ; 108: 110426, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31923928

RESUMEN

A DNA fragment amplification/expression technology for the production of new generation biomaterials for scientific, industrial and biomedical applications is described. The technology enables the formation of artificial Open Reading Frames (ORFs) encoding concatemeric RNAs and proteins. It recruits the Type IIS SapI restriction endonuclease (REase) for an assembling of DNA fragments in an ordered head-to-tail-orientation. The technology employs a vector-enzymatic system, dedicated to the expression of newly formed, concatemeric ORFs from strong promoters. Four vector series were constructed to suit specialised needs. As a proof of concept, a model amplification of a 7-amino acid (aa) epitope from the S protein of HBV virus was performed, resulting in 500 copies of the epitope-coding DNA segment, consecutively linked and expressed in Escherichia coli (E. coli). Furthermore, a peptide with potential pro-regenerative properties (derived from an angiopoietin-related growth factor) was designed. Its aa sequence was back-translated, codon usage optimized and synthesized as a continuous ORF 10-mer. The 10-mer was cloned into the amplification vector, enabling the N-terminal fusion and multiplication of the encoded protein with MalE signal sequence. The obtained genes were expressed, and the proteins were purified. Conclusively, we show that the proteins are neither cytotoxic nor immunogenic and they have a very low allergic potential.


Asunto(s)
Materiales Biocompatibles , ADN Concatenado , Escherichia coli , Expresión Génica , Técnicas de Amplificación de Ácido Nucleico , Sistemas de Lectura Abierta , ADN Concatenado/genética , ADN Concatenado/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Virus de la Hepatitis B/genética , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Virales de Fusión/biosíntesis , Proteínas Virales de Fusión/genética
4.
Viruses ; 11(9)2019 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-31450681

RESUMEN

Newcastle disease (ND) is responsible for significant economic losses in the poultry industry. The disease is caused by virulent strains of Avian avulavirus 1 (AAvV-1), a species within the family Paramyxoviridae. We developed a recombinant construct based on the herpesvirus of turkeys (HVT) as a vector expressing two genes: F and HN (HVT-NDV-F-HN) derived from the AAvV-1 genotype VI ("pigeon variant" of AAvV-1). This recombinant viral vaccine candidate was used to subcutaneously immunize one group of specific pathogen-free (SPF) chickens and two groups of broiler chickens (20 one-day-old birds/group). Humoral immune response was evaluated by hemagglutination-inhibition test and enzyme-linked immunosorbent assay (ELISA). The efficacy of the immunization was assessed in two separate challenge studies performed at 6 weeks of age with the use of virulent AAvV-1 strains representing heterologous genotypes IV and VII. The developed vaccine candidate elicited complete protection in SPF chickens since none of the birds became sick or died during the 2-week observation period. In the broiler groups, 90% and 100% clinical protection were achieved after challenges with AAvV-1 of IV and VII genotypes, respectively. We found no obvious relationship between antibody levels and protection assessed in broilers in the challenge study. The developed recombinant HVT-NDV-F-HN construct containing genes from a genotype VI AAvV-1 offers promising results as a potential vaccine candidate against ND in chickens.


Asunto(s)
Proteína HN/inmunología , Inmunización/veterinaria , Virus de la Enfermedad de Newcastle , Vacunas Sintéticas/inmunología , Proteínas Virales de Fusión/inmunología , Animales , Antígenos Virales/biosíntesis , Antígenos Virales/genética , Pollos/virología , Protección Cruzada , Genes Virales , Proteína HN/biosíntesis , Proteína HN/genética , Pruebas de Inhibición de Hemaglutinación , Herpesvirus Meleágrido 1/genética , Herpesvirus Meleágrido 1/inmunología , Herpesvirus Meleágrido 1/metabolismo , Inmunidad Heteróloga , Enfermedad de Newcastle/inmunología , Enfermedad de Newcastle/prevención & control , Virus de la Enfermedad de Newcastle/genética , Virus de la Enfermedad de Newcastle/inmunología , Enfermedades de las Aves de Corral/virología , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Vacunas Sintéticas/virología , Proteínas Virales de Fusión/biosíntesis , Proteínas Virales de Fusión/genética , Vacunas Virales/biosíntesis , Vacunas Virales/inmunología
5.
Drug Discov Ther ; 11(5): 246-252, 2017 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-29070744

RESUMEN

The effect of glycyrrhizin on the replication of human parainfluenza virus type 2 (hPIV-2) was examined. Cell fusion induced by hPIV-2 was inhibited by glycyrrhizin, and glycyrrhizin reduced the number of viruses released from the cells. Glycyrrhizin did not change cell morphology at 1 day of culture, but caused some damage at 4 days, as determined by the effect on actin microfilaments. However, it affected the cell viability at 1 day: about 20% of the cells were not alive by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay at 1 day of culture. Real-time polymerase chain reaction (PCR) and PCR showed that virus genome synthesis was largely inhibited. mRNA synthesis was also inhibited by glycyrrhizin. Viral protein synthesis was largely inhibited as observed by an indirect immunofluorescence study. Multinucleated giant cell formation was studied using a recombinant green fluorescence protein (GFP)-expressing hPIV-2 without matrix protein (rhPIV-2ΔMGFP). A few single cells with fluorescence were observed, but the formation of giant cells was completely blocked. Taken together, it was shown that viral genome, mRNA and protein syntheses, including F and HN proteins, were inhibited by glycyrrhizin, and consequently multinucleated giant cell formation was not observed and the infectious virus was not detected in the culture medium.


Asunto(s)
Ácido Glicirrínico/farmacología , Virus de la Parainfluenza 2 Humana/efectos de los fármacos , ARN Mensajero/efectos de los fármacos , ARN Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Citoesqueleto de Actina/efectos de los fármacos , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Genoma Viral/efectos de los fármacos , Células Gigantes/efectos de los fármacos , Proteína HN/biosíntesis , Proteína HN/efectos de los fármacos , Macaca mulatta , Virus de la Parainfluenza 2 Humana/genética , Virus de la Parainfluenza 2 Humana/metabolismo , Inhibidores de la Síntesis de la Proteína/farmacología , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Virales de Fusión/biosíntesis , Proteínas Virales de Fusión/efectos de los fármacos , Proteínas Virales/biosíntesis , Proteínas Virales/efectos de los fármacos , Replicación Viral/genética
6.
Protein Expr Purif ; 128: 73-80, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27546452

RESUMEN

Newcastle disease (ND), a highly contagious, acute, and potent infectious disease caused by Newcastle disease virus (NDV), has a considerable impact on the global poultry industry. Although both live attenuated and inactivated vaccines are used to prevent and control the spread of ND among chickens, the increasing number of ND outbreaks in commercial poultry flocks worldwide indicates that routine vaccinations are insufficient to control ND. Hence, efforts are being invested into developing alternative and more effective vaccination strategies. In this study, we focus on F protein, the neutralizing and protective antigen of NDV, and flagellin (FliC), a toll-like receptor 5 (TLR5) agonist that is an effective inducer of innate immune responses. We amplified F gene from velogenic NDV strain F48E8. The recombinant histidine (His)-tagged F protein was efficiently expressed in a Pichia pastoris (P. pastoris) eukaryotic system and verified by sodium dodecyl sulfate polyacrylamide gel electrophoresis and western blotting. The conditions for F protein expression in P. pastoris were optimal. The immunogenicity of F protein with FliC as the adjuvant was evaluated in a C3H/HeJ mouse model. FliC was found to enhance both F-specific and NDV-specific IgG responses and F-specific cellular immune responses following intraperitoneal co-administration with F protein. Thus, the recombinant F protein expressed by P. pastoris when used with flagellin as the adjuvant has potential as a subunit vaccine candidate.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Proteínas de Escherichia coli , Flagelina , Expresión Génica , Virus de la Enfermedad de Newcastle/genética , Pichia/metabolismo , Proteínas Virales de Fusión , Vacunas Virales , Animales , Proteínas de Escherichia coli/inmunología , Proteínas de Escherichia coli/farmacología , Flagelina/inmunología , Flagelina/farmacología , Ratones , Virus de la Enfermedad de Newcastle/inmunología , Pichia/genética , Pichia/inmunología , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/farmacocinética , Proteínas Virales de Fusión/biosíntesis , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/inmunología , Proteínas Virales de Fusión/farmacocinética , Vacunas Virales/biosíntesis , Vacunas Virales/genética , Vacunas Virales/inmunología , Vacunas Virales/farmacología
7.
J Virol ; 89(18): 9499-510, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26157122

RESUMEN

UNLABELLED: Respiratory syncytial virus (RSV) and human parainfluenza virus type 3 (HPIV3) are the first and second leading viral agents of severe respiratory tract disease in infants and young children worldwide. Vaccines are not available, and an RSV vaccine is particularly needed. A live attenuated chimeric recombinant bovine/human PIV3 (rB/HPIV3) vector expressing the RSV fusion (F) glycoprotein from an added gene has been under development as a bivalent vaccine against RSV and HPIV3. Previous clinical evaluation of this vaccine candidate suggested that increased genetic stability and immunogenicity of the RSV F insert were needed. This was investigated in the present study. RSV F expression was enhanced 5-fold by codon optimization and by modifying the amino acid sequence to be identical to that of an early passage of the original clinical isolate. This conferred a hypofusogenic phenotype that presumably reflects the original isolate. We then compared vectors expressing stabilized prefusion and postfusion versions of RSV F. In a hamster model, prefusion F induced increased quantity and quality of RSV-neutralizing serum antibodies and increased protection against wild-type (wt) RSV challenge. In contrast, a vector expressing the postfusion F was less immunogenic and protective. The genetic stability of the RSV F insert was high and was not affected by enhanced expression or the prefusion or postfusion conformation of RSV F. These studies provide an improved version of the previously well-tolerated rB/HPIV3-RSV F vaccine candidate that induces a superior RSV-neutralizing serum antibody response. IMPORTANCE: Respiratory syncytial virus (RSV) and human parainfluenza virus type 3 (HPIV3) are two major causes of pediatric pneumonia and bronchiolitis. The rB/HPIV3 vector expressing RSV F protein is a candidate bivalent live vaccine against HPIV3 and RSV. Previous clinical evaluation indicated the need to increase the immunogenicity and genetic stability of the RSV F insert. Here, we increased RSV F expression by codon optimization and by modifying the RSV F amino acid sequence to conform to that of an early passage of the original isolate. This resulted in a hypofusogenic phenotype, which likely represents the original phenotype before adaptation to cell culture. We also included stabilized versions of prefusion and postfusion RSV F protein. Prefusion RSV F induced a larger quantity and higher quality of RSV-neutralizing serum antibodies and was highly protective. This provides an improved candidate for further clinical evaluation.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Expresión Génica , Virus de la Parainfluenza 3 Humana/inmunología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Virus Sincitial Respiratorio Humano/inmunología , Proteínas Virales de Fusión/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Neutralizantes/metabolismo , Anticuerpos Antivirales/metabolismo , Chlorocebus aethiops , Cricetinae , Humanos , Virus de la Parainfluenza 3 Humana/genética , Proteínas Recombinantes , Infecciones por Virus Sincitial Respiratorio/genética , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/metabolismo , Virus Sincitial Respiratorio Humano/genética , Virus Sincitial Respiratorio Humano/metabolismo , Células Vero , Proteínas Virales de Fusión/biosíntesis , Proteínas Virales de Fusión/genética , Vacunas Virales/genética , Vacunas Virales/metabolismo
8.
J Virol ; 89(17): 8713-26, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26063431

RESUMEN

UNLABELLED: Human metapneumovirus (HMPV) is a major cause of respiratory disease in infants, the elderly, and immunocompromised individuals worldwide. There is currently no licensed HMPV vaccine. Virus-like particles (VLPs) are an attractive vaccine candidate because they are noninfectious and elicit a neutralizing antibody response. However, studies show that serum neutralizing antibodies are insufficient for complete protection against reinfection and that adaptive T cell immunity is important for viral clearance. HMPV and other respiratory viruses induce lung CD8(+) T cell (TCD8) impairment, mediated by programmed death 1 (PD-1). In this study, we generated HMPV VLPs by expressing the fusion and matrix proteins in mammalian cells and tested whether VLP immunization induces functional HMPV-specific TCD8 responses in mice. C57BL/6 mice vaccinated twice with VLPs and subsequently challenged with HMPV were protected from lung viral replication for at least 20 weeks postimmunization. A single VLP dose elicited F- and M-specific lung TCD8s with higher function and lower expression of PD-1 and other inhibitory receptors than TCD8s from HMPV-infected mice. However, after HMPV challenge, lung TCD8s from VLP-vaccinated mice exhibited inhibitory receptor expression and functional impairment similar to those of mice experiencing secondary infection. HMPV challenge of VLP-immunized µMT mice also elicited a large percentage of impaired lung TCD8s, similar to mice experiencing secondary infection. Together, these results indicate that VLPs are a promising vaccine candidate but do not prevent lung TCD8 impairment upon HMPV challenge. IMPORTANCE: Human metapneumovirus (HMPV) is a leading cause of acute respiratory disease for which there is no licensed vaccine. Virus-like particles (VLPs) are an attractive vaccine candidate and induce antibodies, but T cell responses are less defined. Moreover, HMPV and other respiratory viruses induce lung CD8(+) T cell (TCD8) impairment mediated by programmed death 1 (PD-1). In this study, HMPV VLPs containing viral fusion and matrix proteins elicited epitope-specific TCD8s that were functional with low PD-1 expression. Two VLP doses conferred sterilizing immunity in C57BL/6 mice and facilitated HMPV clearance in antibody-deficient µMT mice without enhancing lung pathology. However, regardless of whether responding lung TCD8s had previously encountered HMPV antigens in the context of VLPs or virus, similar proportions were impaired and expressed comparable levels of PD-1 upon viral challenge. These results suggest that VLPs are a promising vaccine candidate but do not prevent lung TCD8 impairment upon HMPV challenge.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Pulmón/inmunología , Metapneumovirus/inmunología , Infecciones por Paramyxoviridae/inmunología , Vacunas de Partículas Similares a Virus/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Células HEK293 , Humanos , Pulmón/citología , Depleción Linfocítica , Macaca mulatta , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Infecciones por Paramyxoviridae/prevención & control , Infecciones por Paramyxoviridae/virología , Receptor de Muerte Celular Programada 1/inmunología , Infecciones del Sistema Respiratorio/inmunología , Infecciones del Sistema Respiratorio/prevención & control , Infecciones del Sistema Respiratorio/virología , Vacunación , Proteínas Virales de Fusión/biosíntesis , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/inmunología , Proteínas de la Matriz Viral/biosíntesis , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/inmunología , Replicación Viral/inmunología
9.
J Virol ; 89(17): 8982-98, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26085152

RESUMEN

To clarify the function(s) of the herpes simplex virus 1 (HSV-1) major virion structural protein UL47 (also designated VP13/14), we screened cells overexpressing UL47 for UL47-binding cellular proteins. Tandem affinity purification of transiently expressed UL47 coupled with mass spectrometry-based proteomics technology and subsequent analyses showed that UL47 interacted with cell protein p32 in HSV-1-infected cells. Unlike in mock-infected cells, p32 accumulated at the nuclear rim in HSV-1-infected cells, and this p32 recruitment to the nuclear rim required UL47. p32 formed a complex(es) with HSV-1 proteins UL31, UL34, Us3, UL47, and/or ICP22 in HSV-1-infected cells. All these HSV-1 proteins were previously reported to be important for HSV-1 nuclear egress, in which nucleocapsids bud through the inner nuclear membrane (primary envelopment) and the enveloped nucleocapsids then fuse with the outer nuclear membrane (de-envelopment). Like viral proteins UL31, UL34, Us3, and UL47, p32 was detected in primary enveloped virions. p32 knockdown reduced viral replication and induced membranous invaginations adjacent to the nuclear rim containing primary enveloped virions and aberrant localization of UL31 and UL34 in punctate structures at the nuclear rim. These effects of p32 knockdown were reduced in the absence of UL47. Therefore, the effects of p32 knockdown in HSV-1 nuclear egress were similar to those of the previously reported mutation(s) in HSV-1 regulatory proteins for HSV-1 de-envelopment during viral nuclear egress. Collectively, these results suggested that p32 regulated HSV-1 de-envelopment and replication in a UL47-dependent manner. IMPORTANCE In this study, we have obtained data suggesting that (i) the HSV-1 major virion structural protein UL47 interacted with host cell protein p32 and mediated the recruitment of p32 to the nuclear rim in HSV-1-infected cells; (ii) p32 was a component of the HSV-1 nuclear egress complex (NEC), whose core components were UL31 and UL34; and (iii) p32 regulated HSV-1 de-envelopment during viral nuclear egress. It has been reported that p32 was a component of human cytomegalovirus NEC and was required for efficient disintegration of nuclear lamina, which has been thought to facilitate HSV-1 primary envelopment during viral nuclear egress. Thus, p32 appeared to be a core component of herpesvirus NECs, like UL31 and UL34 homologs in other herpesviruses, and to play multiple roles in herpesvirus nuclear egress.


Asunto(s)
Proteínas Portadoras/metabolismo , Herpesvirus Humano 1/metabolismo , Proteínas Mitocondriales/metabolismo , Membrana Nuclear/virología , Proteínas Virales de Fusión/metabolismo , Replicación Viral/genética , Transporte Activo de Núcleo Celular/fisiología , Animales , Proteínas Portadoras/genética , Línea Celular , Chlorocebus aethiops , Citomegalovirus/metabolismo , Células HEK293 , Humanos , Proteínas Mitocondriales/genética , Proteínas Nucleares/metabolismo , Nucleocápside/metabolismo , Unión Proteica , Transporte de Proteínas/fisiología , Proteómica/métodos , Células Vero , Proteínas Virales de Fusión/biosíntesis , Proteínas Virales/metabolismo , Ensamble de Virus/genética , Liberación del Virus
10.
J Gen Virol ; 95(Pt 9): 1886-1891, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24914066

RESUMEN

Respiratory syncytial virus (RSV) is the most common cause of respiratory infection in infants and the elderly, and no vaccine against this virus has yet been licensed. Here, we report a recombinant PR8 influenza virus with the RSV fusion (F) protein epitopes of the subgroup A gene inserted into the influenza virus non-structural (NS) gene (rFlu/RSV/F) that was generated as an RSV vaccine candidate. The rescued viruses were assessed by microscopy and Western blotting. The proper expression of NS1, the NS gene product, and the nuclear export protein (NEP) of rFlu/RSV/F was also investigated using an immunofluorescent assay. The rescued virus replicated well in the MDCK kidney cell line, A549 lung adenocarcinoma cell line and CNE-2Z nasopharyngeal carcinoma cell line. BALB/c mice immunized intranasally with rFlu/RSV/F had specific haemagglutination inhibition antibody responses against the PR8 influenza virus and RSV neutralization test proteins. Furthermore, intranasal immunization with rFlu/RSV/F elicited T helper type 1-dominant cytokine profiles against the RSV strain A2 virus. Taken together, our findings suggested that rFlu/RSV/F was immunogenic in vivo and warrants further development as a promising candidate vaccine.


Asunto(s)
Virus de la Influenza A/genética , Virus Sincitiales Respiratorios/genética , Virus Sincitiales Respiratorios/inmunología , Proteínas Virales de Fusión/inmunología , Administración Intranasal , Animales , Anticuerpos Antivirales/inmunología , Células COS , Línea Celular Tumoral , Embrión de Pollo , Chlorocebus aethiops , Perros , Femenino , Técnicas de Transferencia de Gen , Vectores Genéticos , Pruebas de Inhibición de Hemaglutinación , Humanos , Inmunización , Virus de la Influenza A/inmunología , Células de Riñón Canino Madin Darby , Ratones , Ratones Endogámicos BALB C , Pruebas de Neutralización , Infecciones por Virus Sincitial Respiratorio/inmunología , Proteínas Virales de Fusión/biosíntesis , Proteínas Virales de Fusión/genética , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/inmunología
11.
Protein Expr Purif ; 88(1): 7-12, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23220378

RESUMEN

Chimeric peptide MVF-EGFR(237-267), comprising a B-cell epitope from the dimerization interface of human epidermal growth factor receptor (EGFR) and a promiscuous T-cell epitope from measles virus fusion protein (MVF), is a promising candidate antigen peptide for therapeutic vaccine. To establish a high-efficiency preparation process of this small peptide, the coding sequence was cloned into pET-21b and pET-32a respectively, to be expressed alone or in the form of fusion protein with thioredoxin (Trx) and His(6)-tag in Escherichia coli BL21 (DE3). The chimeric peptide failed to be expressed alone, but over-expressed in the fusion form, which presented as soluble protein and took up more than 30% of total proteins of host cells. The fusion protein was seriously degraded during the cell disruption, in which endogenous metalloproteinase played a key role. Degradation of target peptide was inhibited by combined application of EDTA in the cell disruption buffer and a step of Source 30Q anion exchange chromatography (AEC) before metal-chelating chromatography (MCAC) for purifying His(6)-tagged fusion protein. The chimeric peptide was recovered from the purified fusion protein by enterokinase digestion at a yield of 3.0 mg/L bacteria culture with a purity of more than 95%. Immunogenicity analysis showed that the recombinant chimeric peptide was able to arouse more than 1×10(4) titers of specific antibody in BALB/c mice. Present work laid a solid foundation for the development of therapeutic peptide vaccine targeting EGFR dimerization and provided a convenient and low-cost preparation method for small peptides.


Asunto(s)
Epítopos de Linfocito B/genética , Epítopos de Linfocito T/aislamiento & purificación , Receptores ErbB/aislamiento & purificación , Proteínas Recombinantes de Fusión/aislamiento & purificación , Animales , Epítopos de Linfocito B/biosíntesis , Epítopos de Linfocito B/aislamiento & purificación , Epítopos de Linfocito T/biosíntesis , Epítopos de Linfocito T/genética , Receptores ErbB/biosíntesis , Receptores ErbB/genética , Escherichia coli , Expresión Génica , Humanos , Masculino , Virus del Sarampión/química , Virus del Sarampión/genética , Ratones , Péptidos/genética , Péptidos/aislamiento & purificación , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Proteínas Virales de Fusión/biosíntesis , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/aislamiento & purificación
12.
Biosci Rep ; 32(3): 333-43, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22385277

RESUMEN

Protein interactions play key roles throughout all subcellular compartments. In the present paper, we report the visualization of protein interactions throughout living mammalian cells using two oligomerizing MV (measles virus) transmembrane glycoproteins, the H (haemagglutinin) and the F (fusion) glycoproteins, which mediate MV entry into permissive cells. BiFC (bimolecular fluorescence complementation) has been used to examine the dimerization of these viral glycoproteins. The H glycoprotein is a type II membrane-receptor-binding homodimeric glycoprotein and the F glycoprotein is a type I disulfide-linked membrane glycoprotein which homotrimerizes. Together they co-operate to allow the enveloped virus to enter a cell by fusing the viral and cellular membranes. We generated a pair of chimaeric H glycoproteins linked to complementary fragments of EGFP (enhanced green fluorescent protein)--haptoEGFPs--which, on association, generate fluorescence. Homodimerization of H glycoproteins specifically drives this association, leading to the generation of a fluorescent signal in the ER (endoplasmic reticulum), the Golgi and at the plasma membrane. Similarly, the generation of a pair of corresponding F glycoprotein-haptoEGFP chimaeras also produced a comparable fluorescent signal. Co-expression of H and F glycoprotein chimaeras linked to complementary haptoEGFPs led to the formation of fluorescent fusion complexes at the cell surface which retained their biological activity as evidenced by cell-to-cell fusion.


Asunto(s)
Proteínas Fluorescentes Verdes/biosíntesis , Fusión de Membrana , Proteínas de la Membrana/metabolismo , Mapeo de Interacción de Proteínas/métodos , Multimerización de Proteína , Animales , Chlorocebus aethiops , Proteínas Fluorescentes Verdes/genética , Microscopía Confocal , Fragmentos de Péptidos/biosíntesis , Fragmentos de Péptidos/genética , Unión Proteica , Proteínas Recombinantes de Fusión/biosíntesis , Análisis de la Célula Individual , Células Vero , Proteínas Virales de Fusión/biosíntesis
13.
J Immunol ; 188(4): 1647-55, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22266281

RESUMEN

CD4 Th differentiation is influenced by costimulatory molecules expressed on conventional dendritic cells (DCs) in regional lymph nodes and results in specific patterns of cytokine production. However, the function of costimulatory molecules on inflammatory (CD11b(+)) DCs in the lung during recall responses is not fully understood, but it is important for development of novel interventions to limit immunopathological responses to infection. Using a mouse model in which vaccination with vaccinia virus vectors expressing the respiratory syncytial virus (RSV) fusion protein (rVVF) or attachment protein (rVVG) leads to type 1- or type 2-biased cytokine responses, respectively, upon RSV challenge, we found expression of CD40 and OX40 ligand (OX40L) on lung inflammatory DCs was higher in rVVF-primed mice than in rVVG-primed mice early after RSV challenge, whereas the reverse was observed later in the response. Conversely, programmed cell death 1 ligand 2 (PD-L2) was higher in rVVG-primed mice throughout. Inflammatory DCs isolated at the resolution of inflammation revealed that OX40L on type 1-biased DCs promoted IL-5, whereas OX40L on type 2-biased DCs enhanced IFN-γ production by Ag-reactive Th cells. In contrast, PD-L2 promoted IFN-γ production, irrespective of conditions, suppressing IL-5 only if expressed on type 1-biased DCs. Thus, OX40L and PD-L2 expressed on DCs differentially regulate cytokine production during recall responses in the lung. Manipulation of these costimulatory pathways may provide a novel approach to controlling pulmonary inflammatory responses.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Citocinas/biosíntesis , Células Dendríticas/inmunología , Pulmón/inmunología , Glicoproteínas de Membrana/biosíntesis , Proteína 2 Ligando de Muerte Celular Programada 1/biosíntesis , Infecciones por Virus Sincitial Respiratorio/inmunología , Factores de Necrosis Tumoral/biosíntesis , Animales , Linfocitos T CD4-Positivos/metabolismo , Citocinas/inmunología , Células Dendríticas/metabolismo , Femenino , Memoria Inmunológica , Interferón gamma/biosíntesis , Interleucina-5/biosíntesis , Pulmón/metabolismo , Pulmón/patología , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ligando OX40 , Proteína 2 Ligando de Muerte Celular Programada 1/metabolismo , Infecciones por Virus Sincitial Respiratorio/patología , Vacunas contra Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/genética , Virus Sincitiales Respiratorios/inmunología , Factores de Necrosis Tumoral/metabolismo , Virus Vaccinia/genética , Proteínas Virales de Fusión/biosíntesis
14.
Sheng Wu Gong Cheng Xue Bao ; 27(10): 1499-506, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22260067

RESUMEN

Glycoprotein D (gD) of Herpes simplex virus type 2 (HSV-2) is a key factor mediating the entry of HSV-2 into host cells. In order to explain the mechanism underlying the gD-mediated receptor-binding and viral entry, we performed a structural study on HSV-2 gD. The ectodomain of the gD protein encompassing residues 1 to 285 was expressed by baculovirus-infected insect cells as a secreted soluble protein with a C-terminal hexa-his tag. The protein was then purified by affinity and size-exclusion chromatography. The purified protein was successfully crystallized using the hanging-drop vapor-diffusion at 18 degrees C in a condition consisting of 0.1 mol/L Hepes pH 7.2, 5% (V/V) 2-methyl-2,4-pentanediol (MPD) and 10% PEG 10 000. The crystals diffracted to 1.8 angstroms resolution and belonged to space group P21, with unit-cell parameters alpha = 63.6, b = 55.4, c = 65.3 angstroms, beta = 96.3 degrees.


Asunto(s)
Cristalografía por Rayos X , Herpesvirus Humano 2/química , Proteínas Virales de Fusión/química , Animales , Baculoviridae , Cristalización , Insectos/genética , Insectos/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Virales de Fusión/biosíntesis , Proteínas Virales de Fusión/genética
15.
Mol Biotechnol ; 48(3): 201-9, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21153716

RESUMEN

Transgenic plants have been used as a safe and economic expression system for the production of edible vaccines. A synthetic cholera toxin B subunit gene (CTB) was fused with a synthetic neutralizing epitope gene of the porcine epidemic diarrhea virus (sCTB-sCOE), and the sCTB-sCOE fusion gene was introduced into a plant expression vector under the control of the ubiquitin promoter. This plant expression vector was transformed into lettuce (Lactuca sativa L.) using the Agrobacterium-mediated transformation method. Stable integration and transcriptional expression of the sCTB-sCOE fusion gene was confirmed using genomic DNA PCR analysis and northern blot analysis, respectively. The results of western blot analysis with anti-cholera toxin and anti-COE antibody showed the synthesis and assembly of CTB-COE fusion protein into oligomeric structures with pentameric sizing. The biological activity of CTB-COE fusion protein to its receptor, G(M1)-ganglioside, in transgenic plants was confirmed via G(M1)-ELISA with anti-cholera toxin and anti-COE antibody. Based on G(M1)-ELISA, the expression level of CTB-COE fusion proteins reached 0.0065% of the total soluble protein in transgenic lettuce leaf tissues. Transgenic lettuce successfully expressing CTB-COE fusion protein will be tested to induce efficient immune responses against porcine epidemic diarrhea virus infection by administration with raw material.


Asunto(s)
Toxina del Cólera/biosíntesis , Lactuca/genética , Plantas Modificadas Genéticamente/genética , Virus de la Diarrea Epidémica Porcina/genética , Vacunas Comestibles/biosíntesis , Proteínas Virales de Fusión/biosíntesis , Northern Blotting , Western Blotting , Toxina del Cólera/genética , Clonación Molecular , Ensayo de Inmunoadsorción Enzimática , Epítopos/genética , Gangliósido G(M1)/metabolismo , Immunoblotting , Lactuca/metabolismo , Plantas Modificadas Genéticamente/metabolismo , Reacción en Cadena de la Polimerasa , Virus de la Diarrea Epidémica Porcina/inmunología , Ingeniería de Proteínas , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Vacunas Comestibles/genética , Proteínas Virales de Fusión/genética
16.
Vet Microbiol ; 145(1-2): 23-33, 2010 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-20303682

RESUMEN

The central nervous system (CNS) represents an important target organ of the phocine distemper virus (PDV). The aim of the present study was to characterize pathological changes in the CNS of harbor seals suffering from natural PDV-infection. The distribution of virus protein and mRNA was investigated by immunohistochemistry (IHC) and in situ hybridization, respectively. In addition, inflammatory and glial cells were characterized by IHC. Polioencephalitis with glial activation, neuronal death and perivascular mononuclear infiltrations in the cerebral cortex was the main histopathological finding. Inflammatory responses, dominated by CD3(+) T-cells and activated microglia/macrophages were associated with a prominent MHC-II upregulation within the CNS. Viral protein was found predominantly in neurofilament-expressing neurons within inflamed areas as demonstrated by immunohistochemical double-labeling. Morbillivirus nucleo-, phospho-, matrix-, fusion- and hemagglutinin-proteins were found in CNS-lesions. The expressions of viral matrix- and fusion-proteins were reduced in severely inflamed plaques. Comparison of viral protein and mRNA expression revealed a diminished amount of viral phosphoprotein preferentially associated with perivascular inflammation. In summary, CNS-lesions in PDV-infected seals are similar to canine distemper virus-induced acute polioencephalitis in dogs and measles virus inclusion body polioencephalitis in men, respectively.


Asunto(s)
Sistema Nervioso Central/virología , Virus del Moquillo Focino/genética , Moquillo/virología , Phoca/virología , Animales , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/fisiopatología , Moquillo/inmunología , Moquillo/fisiopatología , Virus del Moquillo Focino/fisiología , Encefalitis/veterinaria , Encefalitis/virología , Femenino , Regulación Viral de la Expresión Génica/fisiología , Inmunidad Celular/inmunología , Hibridación in Situ/veterinaria , Masculino , Fenotipo , Phoca/inmunología , ARN Mensajero/genética , Proteínas Virales de Fusión/biosíntesis , Proteínas de la Matriz Viral/biosíntesis , Proteínas Virales/biosíntesis
17.
Virus Genes ; 40(2): 212-21, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20111897

RESUMEN

Studies of the fusion activity of respiratory syncytial virus (RSV) F protein are significantly hindered by low recombinant expression levels. While infection produces F protein levels detectable by western blot, recombinant expression produces undetectable to low levels of F protein. Identifying the obstacles that hinder recombinant F protein expression may lead to improved expression and facilitate the study of F protein function. We hypothesized that nuclear localization and/or inefficient RNA polymerase II-mediated transcription contribute to poor recombinant F protein expression. This study shows a combination of stalled nuclear export, premature polyadenylation, and low mRNA abundance all contribute to low recombinant F protein expression levels. In addition, this study provides an expression optimization strategy that results in greater F protein expression levels than observed by codon-optimization of the F protein gene, which will be useful for future studies of F protein function.


Asunto(s)
Transporte Activo de Núcleo Celular , Expresión Génica , Procesamiento Postranscripcional del ARN , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Proteínas Virales de Fusión/biosíntesis , Animales , Línea Celular , Humanos , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Virales de Fusión/genética
18.
J Hepatol ; 51(3): 520-7, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19596480

RESUMEN

BACKGROUND/AIMS: Vaccination strategies able to induce strong T-cell responses might contribute to eradicate hepatitis C virus (HCV) infection. We previously demonstrated that fusion of an antigen to the extra domain A from fibronectin (EDA) targets the antigen to TLR4-expressing dendritic cells (DC) and improves its immunogenicity. Here, we studied if fusion of EDA with the non-structural HCV protein NS3 might constitute an effective immunogen against HCV. METHODS: Recombinant NS3 and the fusion protein EDA-NS3 were produced and purified from E. coli, and tested in vitro for their capacity to activate maturation of DC and to favour antigen presentation. HHD transgenic mice expressing the human HLA-A2 molecule were immunized with recombinant proteins in the absence or presence of poly(I:C) and anti-CD40 agonistic antibodies and responses elicited by vaccination were tested in vitro, and in vivo, by their capacity to downregulate intrahepatic expression of HCV-NS3 RNA. RESULTS: EDA-NS3, but not NS3 alone, upregulated the expression of maturation markers, as well as Delta-like 1 and Delta-like 4 Notch ligands in DC and induced the production of IL-12. Mice immunized with EDA-NS3 had strong and long lasting NS3-specific CD4+ and CD8+ T-cell responses and, in combination with poly(I:C) and anti-CD40, downregulated intrahepatic expression of HCV-NS3 RNA. CONCLUSIONS: Recombinant EDA-NS3 may be considered for the development of vaccines against HCV infection.


Asunto(s)
Fibronectinas/uso terapéutico , Hepacivirus/inmunología , Hepatitis C/prevención & control , Proteínas Recombinantes/uso terapéutico , Proteínas Virales de Fusión/uso terapéutico , Proteínas no Estructurales Virales/uso terapéutico , Vacunas Virales/uso terapéutico , Animales , Antivirales/uso terapéutico , Línea Celular , Células Cultivadas , Células Dendríticas/citología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Quimioterapia Combinada , Escherichia coli/metabolismo , Femenino , Fibronectinas/biosíntesis , Antígeno HLA-A2/genética , Antígeno HLA-A2/inmunología , Antígeno HLA-A2/metabolismo , Hepacivirus/genética , Hepacivirus/metabolismo , Hepatitis C/inmunología , Hepatitis C/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Poli I-C/uso terapéutico , Estructura Terciaria de Proteína , ARN Viral/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Virales de Fusión/biosíntesis , Proteínas no Estructurales Virales/biosíntesis , Proteínas no Estructurales Virales/genética , Vacunas Virales/biosíntesis
19.
Protein Expr Purif ; 62(2): 206-15, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18799134

RESUMEN

This study presents the heterologous production and purification of a soluble and functional form of the hemagglutinin esterase (HE) of the infectious salmon anemia virus (ISAV) isolate 4 (Glesvaer/2/90). The HE possesses receptor binding and receptor destroying enzyme (RDE) activity and is probably involved in the infection process. The recombinant HE protein (recHE 4) was expressed in insect cells (Sf9) using the baculovirus expression vector system. Both the transmembrane region and the cytoplasmic tail were deleted, and a C-terminal His(6)-tag was attached to facilitate identification and purification of the recHE 4 protein. As determined by Western analysis the recHE 4 was secreted at 20 degrees C and not at 28 degrees C. By testing three HE constructs differing in their promoter and secretion signal sequences it was clear that the HE's own secretion signal sequence is more important than the promoter with respect to the amount of secreted recHE 4 obtained under the conditions used. A one-step purification by nickel-affinity chromatography resulted in a highly purified recHE 4, identified by matrix assisted laser desorption/ionization time of flight mass spectrometry (MALDI-TOF MS) analysis. Also, the recHE 4 is glycosylated and contains disulfide bridges within the molecule. Functional studies including the verification of the receptor destroying enzyme (RDE) activity as well as the binding to Atlantic salmon erythrocytes (hemagglutination) indicate that the recHE 4 has similar functions as its native counterpart. In conclusion, insect cells secrete a functional form of the ISAV 4 HE. This is suitable for further analyses on its function and immunogenicity.


Asunto(s)
Baculoviridae/metabolismo , Hemaglutininas Virales/biosíntesis , Hemaglutininas Virales/aislamiento & purificación , Isavirus/enzimología , Proteínas Virales de Fusión/biosíntesis , Proteínas Virales de Fusión/aislamiento & purificación , Acetilesterasa/metabolismo , Secuencia de Aminoácidos , Animales , Western Blotting , Línea Celular , Estabilidad de Enzimas , Eritrocitos/citología , Glicosilación , Hemaglutinación , Hemaglutininas Virales/química , Insectos , Datos de Secuencia Molecular , Desnaturalización Proteica , Salmo salar/virología , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/metabolismo
20.
J Virol ; 82(21): 10510-8, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18753197

RESUMEN

Morbilliviruses, including measles and canine distemper virus (CDV), are nonsegmented, negative-stranded RNA viruses that cause severe diseases in humans and animals. The transcriptional units in their genomes are separated by untranslated regions (UTRs), which contain essential transcription and translation signals. Due to its increased length, the region between the matrix (M) protein and fusion (F) protein open reading frames is of particular interest. In measles virus, the entire F 5' region is untranslated, while several start codons are found in most other morbilliviruses, resulting in a long F protein signal peptide (Fsp). To characterize the role of this region in morbillivirus pathogenesis, we constructed recombinant CDVs, in which either the M-F UTR was replaced with that between the nucleocapsid (N) and phosphoprotein (P) genes, or 106 Fsp residues were deleted. The Fsp deletion alone had no effect in vitro and in vivo. In contrast, substitution of the UTR was associated with a slight increase in F gene and protein expression. Animals infected with this virus either recovered completely or experienced prolonged disease and death due to neuroinvasion. The combination of both changes resulted in a virus with strongly increased F gene and protein expression and complete attenuation. Taken together, our results provide evidence that the region between the morbillivirus M and F genes modulates virulence through transcriptional control of the F gene expression.


Asunto(s)
ADN Intergénico , Virus del Moquillo Canino/patogenicidad , Proteínas Virales de Fusión/biosíntesis , Regiones no Traducidas 5' , Animales , Línea Celular , Moquillo/virología , Virus del Moquillo Canino/genética , Hurones , Humanos , Masculino , Recombinación Genética , Eliminación de Secuencia , Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...