Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 973
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 545: 145-149, 2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33550095

RESUMEN

In March 2013 it was reported by the World Health Organization (WHO) the first cases of human infections with avian influenza virus A (H7N9). From 2013 to December 2019, 1568 cases have been reported with 616 deaths. H7N9 infection has been associated with high morbidity and mortality rates, and vaccination is currently the most effective way to prevent infections and consequently flu-related severe illness. Developing and producing vaccines against pandemic influenza viruses is the main strategy for a response to a possible pandemic. This study aims to present the production of three industrial lots under current Good Manufacturing Practices (cGMP) of the active antigen used to produce the pandemic influenza vaccine candidate against A(H7N9). These batches were characterized and evaluated for quality standards and tested for immunogenicity in mice. The average yield was 173.50 ± 7.88 µg/mL of hemagglutinin and all the preparations met all the required specifications. The formulated H7N9 vaccine is poorly immunogenic and needs to be adjuvanted with an oil in water emulsion adjuvant (IB160) to achieve a best immune response, in a prime and in a boost scheme. These data are important for initial production planning and preparedness in the case of a H7N9 pandemic.


Asunto(s)
Subtipo H7N9 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/biosíntesis , Gripe Humana/prevención & control , Pandemias/prevención & control , Animales , Antígenos Virales/biosíntesis , Antígenos Virales/inmunología , Composición de Medicamentos/métodos , Composición de Medicamentos/estadística & datos numéricos , Industria Farmacéutica/normas , Femenino , Humanos , Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/aislamiento & purificación , Gripe Humana/inmunología , Gripe Humana/virología , Ratones , Ratones Endogámicos BALB C , Vacunas de Productos Inactivados/biosíntesis , Vacunas de Productos Inactivados/inmunología , Vacunas de Productos Inactivados/aislamiento & purificación
2.
PLoS Pathog ; 15(10): e1008068, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31648236

RESUMEN

Ebola virus (EBOV) infections are characterized by a pronounced lymphopenia that is highly correlative with fatalities. However, the mechanisms leading to T-cell depletion remain largely unknown. Here, we demonstrate that both viral mRNAs and antigens are detectable in CD4+ T cells despite the absence of productive infection. A protein phosphatase 1 inhibitor, 1E7-03, and siRNA-mediated suppression of viral antigens were used to demonstrate de novo synthesis of viral RNAs and antigens in CD4+ T cells, respectively. Cell-to-cell fusion of permissive Huh7 cells with non-permissive Jurkat T cells impaired productive EBOV infection suggesting the presence of a cellular restriction factor. We determined that viral transcription is partially impaired in the fusion T cells. Lastly, we demonstrate that exposure of T cells to EBOV resulted in autophagy through activation of ER-stress related pathways. These data indicate that exposure of T cells to EBOV results in an abortive infection, which likely contributes to the lymphopenia observed during EBOV infections.


Asunto(s)
Linfocitos T CD4-Positivos/virología , Ebolavirus/inmunología , Fiebre Hemorrágica Ebola/inmunología , Linfopenia/inmunología , Replicación Viral/fisiología , Animales , Antígenos Virales/biosíntesis , Antígenos Virales/genética , Autofagia/fisiología , Linfocitos T CD4-Positivos/inmunología , Línea Celular , Chlorocebus aethiops , Estrés del Retículo Endoplásmico/fisiología , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Indoles/farmacología , Células Jurkat , Proteína Fosfatasa 1/antagonistas & inhibidores , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Viral/biosíntesis , ARN Viral/genética , Factores de Transcripción/metabolismo , Urea/análogos & derivados , Urea/farmacología , Células Vero , Proteínas Virales/metabolismo
3.
Biotechnol Bioeng ; 118(6): 2202-2219, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33624859

RESUMEN

Serological assays are valuable tools to study SARS-CoV-2 spread and, importantly, to identify individuals that were already infected and would be potentially immune to a virus reinfection. SARS-CoV-2 Spike protein and its receptor binding domain (RBD) are the antigens with higher potential to develop SARS-CoV-2 serological assays. Moreover, structural studies of these antigens are key to understand the molecular basis for Spike interaction with angiotensin converting enzyme 2 receptor, hopefully enabling the development of COVID-19 therapeutics. Thus, it is urgent that significant amounts of this protein became available at the highest quality. In this study, we produced Spike and RBD in two human derived cell hosts: HEK293-E6 and Expi293F™. We evaluated the impact of different and scalable bioprocessing approaches on Spike and RBD production yields and, more importantly, on these antigens' quality attributes. Using negative and positive sera collected from human donors, we show an excellent performance of the produced antigens, assessed in serologic enzyme-linked immunosorbent assay (ELISA) tests, as denoted by the high specificity and sensitivity of the test. We show robust Spike productions with final yields of approx. 2 mg/L of culture that were maintained independently of the production scale or cell culture strategy. To the best of our knowledge, the final yield of 90 mg/L of culture obtained for RBD production, was the highest reported to date. An in-depth characterization of SARS-CoV-2 Spike and RBD proteins was performed, namely the antigen's oligomeric state, glycosylation profiles, and thermal stability during storage. The correlation of these quality attributes with ELISA performance show equivalent reactivity to SARS-CoV-2 positive serum, for all Spike and RBD produced, and for all storage conditions tested. Overall, we provide straightforward protocols to produce high-quality SARS-CoV-2 Spike and RBD antigens, that can be easily adapted to both academic and industrial settings; and integrate, for the first time, studies on the impact of bioprocess with an in-depth characterization of these proteins, correlating antigen's glycosylation and biophysical attributes to performance of COVID-19 serologic tests.


Asunto(s)
Antígenos Virales/biosíntesis , Glicosilación , Glicoproteína de la Espiga del Coronavirus/biosíntesis , Frío , Ensayo de Inmunoadsorción Enzimática/normas , Congelación , Células HEK293 , Humanos , Conformación Proteica , Estabilidad Proteica , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/normas , SARS-CoV-2 , Pruebas Serológicas/normas , Glicoproteína de la Espiga del Coronavirus/normas
4.
Protein Expr Purif ; 188: 105965, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34461217

RESUMEN

BACKGROUND: Egypt has a high prevalence of hepatitis C virus (HCV) infection with 92.5% of genotype-4. AIM: This study aimed to clone and express the core gene of HCV genotype-4 for using it to develop a highly sensitive, specific, and cost-effective diagnostic assay for detecting HCV infection. METHODS: Using synthetic HCV genotype-4 core gene, pET15b as E. coli expression vector, and 1 mM lactose as inducer, the HCV core protein (MW 17 kDa) was expressed in the form of inclusion bodies (IBs) that was purified and solubilized using 8 M guanidinium HCl. The recombinant core protein was in vitro refolded by a rapid dilution method for further purification using weak cation exchange liquid chromatography. The immunogenicity of the purified protein was tested by ELISA using 129 serum samples. RESULTS: The recombinant core protein was successfully expressed and purified. The results also showed that the in-house anti-HCV core assay is accurate, specific (~96.6%), and highly sensitive (~100%) in accordance with the commercial ELISA kit. CONCLUSION: The sensitivity, specificity, and reproducibility of the developed assay were high and promising to be used as a screening assay for detecting HCV infection.


Asunto(s)
Anticuerpos Antivirales/sangre , Antígenos Virales/genética , Hepacivirus/genética , Hepatitis C/diagnóstico , Proteínas del Núcleo Viral/genética , Antígenos Virales/biosíntesis , Antígenos Virales/inmunología , Antígenos Virales/aislamiento & purificación , Cromatografía por Intercambio Iónico/métodos , Clonación Molecular , Egipto/epidemiología , Ensayo de Inmunoadsorción Enzimática , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Genotipo , Guanidina/química , Hepacivirus/clasificación , Hepacivirus/inmunología , Hepatitis C/epidemiología , Hepatitis C/virología , Humanos , Sueros Inmunes/química , Cuerpos de Inclusión/química , Prevalencia , Replegamiento Proteico , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/aislamiento & purificación , Proteínas del Núcleo Viral/biosíntesis , Proteínas del Núcleo Viral/inmunología , Proteínas del Núcleo Viral/aislamiento & purificación
5.
Biochem Biophys Res Commun ; 533(4): 983-987, 2020 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-33008586

RESUMEN

JC polyomavirus (JCPyV) causes progressive multifocal leukoencephalopathy (PML), a demyelinating disease of the central nervous system, in immunocompromised patients. Although PML used to be rare, recently the incidence of PML has risen due to an increase in immunosuppressive therapy. An in vitro JCPyV infection system could be used for anti-drug screening and investigation of tropism changes, but study of JCPyV in vitro has been limited due to the difficulty of efficiently propagating the virus in cultured cells. PML-type JCPyV efficiently propagates in primary human fetal and progenitor cell-derived astrocytes, but the preparation of cells from human fetuses is associated with severe ethical problems. In this study, human iPS cell-derived astrocytes were exposed to PML-type JCPyV. Infection, replication, and VP1 and T antigens of JCPyV were detected and confirmed in this culture. The non-coding control region (NCCR) of M1-IMRb was conserved in infected cells without point mutations. In addition, PML-type JCPyV genomic DNA in infected cells was detected as a single band of approximately 5.1 kbp, with no deletions. This is the first demonstration that human iPS cell-derived astrocytes efficiently support replication of PML-type JCPyV without production of defective interfering particles. These findings indicated that a culture system using human iPS cell-derived astrocyte would be useful for studies of PML, especially for screening anti-JCPyV drugs.


Asunto(s)
Astrocitos/virología , Células Madre Pluripotentes Inducidas/virología , Virus JC/fisiología , Leucoencefalopatía Multifocal Progresiva/virología , Animales , Antígenos Virales/biosíntesis , Antígenos Virales de Tumores/biosíntesis , Astrocitos/patología , Células COS , Proteínas de la Cápside/biosíntesis , Proteínas de la Cápside/inmunología , Diferenciación Celular , Línea Celular , Chlorocebus aethiops , ADN Viral/genética , Genoma Viral , Humanos , Células Madre Pluripotentes Inducidas/patología , Virus JC/genética , Virus JC/patogenicidad , Leucoencefalopatía Multifocal Progresiva/etiología , Leucoencefalopatía Multifocal Progresiva/patología , Células-Madre Neurales/patología , Cultivo de Virus/métodos , Replicación Viral
6.
Mol Biol Rep ; 47(10): 7333-7340, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32997310

RESUMEN

Dengue virus and Zika virus are arthropod-borne flaviviruses that cause millions of infections worldwide. The co-circulation of both viruses makes serological diagnosis difficult as they share high amino acid similarities in viral proteins. Antigens are one of the key reagents in the differential diagnosis of these viruses through the detection of IgG antibodies in serological assays during the convalescent-phase of infections. Here, we report the expression of Dengue virus (DENV) and Zika virus (ZIKV) antigens containing non-conserved and immunodominant amino acid sequences using the baculovirus expression vector system in insect cells. We designed DENV and ZIKV antigens based on the domain III of the E protein (EDIII) after analyzing previously reported epitopes and by multiple alignment of the most important flaviviruses. The ZIKV and DENV multi-epitope genes were designed as tandem repeats or impaired repeats separated by tetra- or hexa-glycine linkers. The biochemical analyses revealed adequate expression of the antigens. Then, the obtained multi-epitope antigens were semi-purified in a sucrose gradient and tested using patients' sera collected during the convalescent-phase that were previously diagnosed positive for anti-DENV and -ZIKV IgG antibodies. The optimal serum dilution was 1:200, and the mean absorbance values in the preliminary tests show that multi-epitope antigens have been recognized by human sera. The production of both antigens using the multi-epitope strategy in the eukaryotic system and based on the EDIII regions provide a proof of concept for the use of antigens in the differentiation between DENV and ZIKV.


Asunto(s)
Antígenos Virales , Virus del Dengue/genética , Epítopos , Expresión Génica , Proteínas Recombinantes de Fusión , Proteínas del Envoltorio Viral , Virus Zika/genética , Animales , Antígenos Virales/biosíntesis , Antígenos Virales/genética , Baculoviridae/genética , Baculoviridae/metabolismo , Línea Celular , Epítopos/biosíntesis , Epítopos/genética , Mariposas Nocturnas , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Proteínas del Envoltorio Viral/biosíntesis , Proteínas del Envoltorio Viral/genética
7.
Metab Brain Dis ; 35(5): 819-827, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32172520

RESUMEN

As feared and deadly human diseases globally, Rabies virus contrived mechanisms to escape early immune recognition via suppression of the interferon response. This study, preliminarily investigated whether Rabies virus employs epigenetic mechanism for the suppression of the interferon using the Challenge virus standard (CVS) strain and Nigerian street Rabies virus (SRV) strain. Mice were challenged with Rabies virus (RABV) infection, and presence of RABV antigen was assessed by direct fluorescent antibody test (DFAT). A real time quantitative Polymerase chain reaction (qRT-PCR) was used to measure the expression of type II interferon gamma (IFNG) and methylation specific quantitative PCR for methylation analysis of 1FNG promoter region. Accordingly, DNA methyltransferase (DNMT) and histone acetyltransferase (HAT) enzymes activities were determined. RABV antigen was detected in all infected samples. A statistically significant increase (p < 0.05) in mRNA level of IFNG was observed at the onset of the disease and a decrease as the disease progressed. An increase in methylation in the test groups from the control group was observed, with a fluctuation in methylation as the disease progressed. DNMT and HAT activities also agree with methylation as there was an observed increase activity in test group compared with control group. Similar fluctuation pattern was observed in both CVS and SRV groups as the disease progressed with HAT, being the most active proportionally. This study suggests that epigenetic modification via DNA methylation and histone acetylation may have played a role in the expression of type II interferon gamma in Rabies virus infection. Graphical abstract.


Asunto(s)
Epigénesis Genética/genética , Interferón gamma/genética , Rabia/metabolismo , Animales , Antígenos Virales/biosíntesis , Antígenos Virales/genética , ADN (Citosina-5-)-Metiltransferasa 1/biosíntesis , ADN (Citosina-5-)-Metiltransferasa 1/genética , Metilación de ADN , Regulación de la Expresión Génica , Histona Acetiltransferasas/metabolismo , Interferón gamma/biosíntesis , Ratones , Rabia/inmunología , Virus de la Rabia
8.
Protein Expr Purif ; 153: 18-25, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30125621

RESUMEN

Saint Louis encephalitis virus (SLEV) and West Nile virus (WNV) are two of the major causes of arboviral encephalitis in the Americas. The co-circulation of related flaviviruses in the Americas and prior vaccination against flaviviruses pose problems to the diagnostic specificity of serological assays due to the development of cross-reactive antibodies. An accurate diagnosis method capable of differentiating these related viruses is needed. NS1 is a glycosylated, nonstructural protein, of about 46 kDa which has a highly conserved structure. Anti-NS1 antibodies can be detected within 4-8 days after the initial exposure and NS1 is the least cross-reactive of the flaviviral antigens. This study was aimed to generate SLEV and WNV NS1 recombinants proteins for the development of a flavivirus diagnostic test. Local Argentinian isolates were used as the source of NS1 gene cloning, expression, and purification. The protein was expressed in Escherichia coli as inclusion bodies and further purified by metal-chelating affinity chromatography (IMAC) under denaturing conditions. Human sera from SLEV and WNV positive cases showed reactivity to the recombinant NS1 proteins by western blot. The unfolded NS1 proteins were also used as immunogens. The polyclonal antibodies elicited in immunized mice recognized the two recombinant proteins with differential reactivity.


Asunto(s)
Anticuerpos Antivirales/biosíntesis , Antígenos Virales/inmunología , Virus de la Encefalitis de San Luis/inmunología , Encefalitis de San Luis/diagnóstico , Proteínas no Estructurales Virales/inmunología , Fiebre del Nilo Occidental/diagnóstico , Virus del Nilo Occidental/inmunología , Animales , Especificidad de Anticuerpos , Antígenos Virales/biosíntesis , Antígenos Virales/genética , Argentina , Western Blotting , Cromatografía de Afinidad , Clonación Molecular , Reacciones Cruzadas , Diagnóstico Diferencial , Virus de la Encefalitis de San Luis/química , Virus de la Encefalitis de San Luis/genética , Encefalitis de San Luis/inmunología , Encefalitis de San Luis/virología , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Humanos , Cuerpos de Inclusión/química , Ratones , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Solubilidad , Proteínas no Estructurales Virales/biosíntesis , Proteínas no Estructurales Virales/genética , Fiebre del Nilo Occidental/inmunología , Fiebre del Nilo Occidental/virología , Virus del Nilo Occidental/química , Virus del Nilo Occidental/genética
9.
Med Microbiol Immunol ; 208(3-4): 439-446, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31004200

RESUMEN

Roizman's definition of herpesviral latency, which applies also to cytomegaloviruses (CMVs), demands maintenance of reactivation-competent viral genomes after clearance of productive infection. It is more recent understanding that failure to complete the productive viral cycle for virus assembly and release does not imply viral gene silencing at all genetic loci and all the time. It rather appears that CMV latency is transcriptionally "noisy" in that silenced viral genes get desilenced from time to time in a stochastic manner, leading to "transcripts expressed in latency" (TELs). If a TEL happens to code for a protein that contains a CD8 T cell epitope, protein processing can lead to the presentation of the antigenic peptide and restimulation of cognate CD8 T cells during latency. This mechanism is discussed as a potential driver of epitope-selective accumulation of CD8 T cells over time, a phenomenon linked to CMV latency and known as "memory inflation" (MI). So far, expression of an epitope-encoding TEL was shown only for the major immediate-early (MIE) gene m123/ie1 of murine cytomegalovirus (mCMV), which codes for the prototypic MI-driving antigenic peptide YPHFMPTNL that is presented by the MHC class-I molecule Ld. The only known second MI-driving antigenic peptide of mCMV in the murine MHC haplotype H-2d is AGPPRYSRI presented by the MHC-I molecule Dd. This peptide is very special in that it is encoded by the early (E) phase gene m164 and by an overlapping immediate-early (IE) transcript governed by a promoter upstream of m164. If MI is driven by presentation of TEL-derived antigenic peptides, as the hypothesis says, one should find corresponding TELs. We show here that E-phase and IE-phase transcripts that code for the MI-driving antigenic peptide AGPPRYSRI are independently and stochastically expressed in latently infected lungs.


Asunto(s)
Antígenos Virales/inmunología , Linfocitos T CD8-positivos/inmunología , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/virología , Perfilación de la Expresión Génica , Muromegalovirus/inmunología , Latencia del Virus , Animales , Antígenos Virales/biosíntesis , Modelos Animales de Enfermedad , Epítopos/biosíntesis , Epítopos/inmunología , Memoria Inmunológica , Muromegalovirus/crecimiento & desarrollo
10.
Appl Microbiol Biotechnol ; 103(18): 7491-7504, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31332484

RESUMEN

Infectious bursal disease virus (IBDV) is the cause of an economically important highly contagious disease of poultry, and vaccines are regarded as the most beneficial interventions for its prevention. In this study, plants were used to produce a recombinant chimeric IBDV antigen for the formulation of an innovative subunit vaccine. The fusion protein (PD-FcY) was designed to combine the immunodominant projection domain (PD) of the viral structural protein VP2 with the constant region of avian IgY (FcY), which was selected to enhance antigen uptake by avian immune cells. The gene construct encoding the fusion protein was transiently expressed in Nicotiana benthamiana plants and an extraction/purification protocol was set up, allowing to reduce the contamination by undesired plant compounds/proteins. Mass spectrometry analysis of the purified protein revealed that the glycosylation pattern of the FcY portion was similar to that observed in native IgY, while in vitro assays demonstrated the ability of PD-FcY to bind to the avian immunoglobulin receptor CHIR-AB1. Preliminary immunization studies proved that PD-FcY was able to induce the production of protective anti-IBDV-VP2 antibodies in chickens. In conclusion, the proposed fusion strategy holds promises for the development of innovative low-cost subunit vaccines for the prevention of avian viral diseases.


Asunto(s)
Anticuerpos Antivirales/sangre , Antígenos Virales/inmunología , Inmunoglobulinas/inmunología , Enfermedades de las Aves de Corral/prevención & control , Vacunas Virales/biosíntesis , Animales , Antígenos Virales/biosíntesis , Pollos/inmunología , Inmunoglobulinas/biosíntesis , Virus de la Enfermedad Infecciosa de la Bolsa , Enfermedades de las Aves de Corral/virología , Nicotiana/genética , Vacunación , Vacunas de Subunidad/biosíntesis , Proteínas Estructurales Virales/biosíntesis , Proteínas Estructurales Virales/inmunología
11.
J Virol ; 91(19)2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28747501

RESUMEN

Latency-associated nuclear antigen (LANA) is a multifunctional protein encoded by members of the Rhadinovirus genus of gammaherpesviruses. Studies using murine gammaherpesvirus 68 (MHV68) demonstrated that LANA is important for acute replication, latency establishment, and reactivation in vivo Despite structural similarities in their DNA-binding domains (DBDs), LANA homologs from Kaposi sarcoma-associated herpesvirus (KSHV) and MHV68 exhibit considerable sequence divergence. We sought to determine if KSHV and MHV68 LANA homologs are functionally interchangeable. We generated an MHV68 virus that encodes KSHV LANA (kLANA) in place of MHV68 LANA (mLANA) and evaluated the virus's capacity to replicate, establish and maintain latency, and reactivate. kLANA knock-in (KLKI) MHV68 was replication competent in vitro and in vivo but exhibited slower growth kinetics and lower titers than wild-type (WT) MHV68. Following inoculation of mice, KLKI MHV68 established and maintained latency in splenocytes and peritoneal cells but did not reactivate efficiently ex vivo kLANA repressed the MHV68 promoter for ORF50, the gene that encodes the major lytic transactivator protein RTA, while mLANA did not, suggesting a likely mechanism for the KLKI MHV68 phenotypes. Bypassing this repression by providing MHV68 RTA in trans rescued KLKI MHV68 replication in tissue culture and enabled detection of KLKI MHV68 reactivation ex vivo These data demonstrate that kLANA and mLANA are functionally interchangeable for establishment and maintenance of latency and suggest that repression of lytic replication by kLANA, as previously shown with KSHV, is a kLANA-specific function that is transferable to MHV68.IMPORTANCE Kaposi sarcoma-associated herpesvirus (KSHV) and murine gammaherpesvirus 68 (MHV68) are members of the Rhadinovirus genus of gammaherpesviruses. These viruses establish lifelong infections that place their respective human and murine hosts at risk for cancer. Latency-associated nuclear antigen (LANA) is a conserved Rhadinovirus protein that is necessary for long-term chronic infection by these viruses. To better understand the conserved functions performed by LANA homologs, we generated a recombinant MHV68 virus that encodes the KSHV LANA protein in place of the MHV68 LANA homolog. We determined that the KSHV LANA protein is capable of supporting MHV68 latency in a mouse model of chronic infection but also functions to repress viral replication. This work describes an in vivo model system for defining evolutionarily conserved and divergent functions of LANA homologs in Rhadinovirus infection and disease.


Asunto(s)
Antígenos Virales/genética , Herpesvirus Humano 8/crecimiento & desarrollo , Proteínas Inmediatas-Precoces/genética , Proteínas Nucleares/genética , Rhadinovirus/crecimiento & desarrollo , Transactivadores/genética , Latencia del Virus/genética , Células 3T3 , Animales , Antígenos Virales/biosíntesis , Línea Celular , Femenino , Técnicas de Sustitución del Gen , Células HEK293 , Herpesvirus Humano 8/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Nucleares/biosíntesis , Regiones Promotoras Genéticas/genética , Rhadinovirus/genética , Rhadinovirus/metabolismo
12.
J Viral Hepat ; 25(6): 649-660, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29316037

RESUMEN

Hepatitis C virus (HCV) chronically infects 2%-3% of the world's population, causing liver disease and cancer with prolonged infection. The narrow host range of the virus, being restricted largely to human hepatocytes, has made the development of relevant models to evaluate the efficacy of vaccines a challenge. We have developed a novel approach to accomplish this by generating a murine hepatoma cell line stably expressing nonstructural HCV antigens which can be used in vitro or in vivo to test HCV vaccine efficacies. These HCV-recombinant hepatoma cells formed large solid-mass tumours when implanted into syngeneic mice, allowing us to test candidate HCV vaccines to demonstrate the development of an HCV-specific immune response that limited tumour growth. Using this model, we tested the therapeutic potential of recombinant anti-HCV-specific vaccines based on two fundamentally different attenuated pathogen vaccine systems-attenuated Salmonella and recombinant adenoviral vector based vaccine. While attenuated Salmonella that secreted HCV antigens limited growth of the HCV-recombinant tumours when used in a therapeutic vaccination trial, replication-competent but noninfectious adenovirus expressing nonstructural HCV antigens showed overall greater survival and reduced weight loss compared to non-replicating nondisseminating adenovirus. Our results demonstrate a model with anti-tumour responses to HCV nonstructural (NS) protein antigens and suggest that recombinant vaccine vectors should be explored as a therapeutic strategy for controlling HCV and HCV-associated cancers.


Asunto(s)
Carcinoma Hepatocelular/patología , Modelos Animales de Enfermedad , Neoplasias Hepáticas/patología , Animales , Antígenos Virales/biosíntesis , Antígenos Virales/genética , Carcinoma Hepatocelular/terapia , Carcinoma Hepatocelular/virología , Femenino , Expresión Génica , Hepacivirus/genética , Hepatocitos/virología , Neoplasias Hepáticas/terapia , Neoplasias Hepáticas/virología , Ratones Endogámicos C57BL , Modelos Biológicos , Trasplante de Neoplasias , Resultado del Tratamiento , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología , Vacunas contra Hepatitis Viral/administración & dosificación , Vacunas contra Hepatitis Viral/inmunología , Proteínas no Estructurales Virales/biosíntesis , Proteínas no Estructurales Virales/genética
13.
Protein Expr Purif ; 133: 57-65, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28274805

RESUMEN

Dengue is a mosquito-borne disease caused by four genetically and serologically related viruses that affect several millions of people. Envelope domain III (EDIII) of the viral envelope protein contains dengue virus (DENV) type-specific and DENV complex-reactive antigenic sites. Here, we describe the expression in Escherichia coli, the refolding and bio-structural analysis of envelope domain III of the four dengue serotypes as a tetravalent dengue protein (EDIIIT2), generating an attractive diagnostic candidate. In vitro refolding of denatured EDIIIT2 was performed by successive dialysis with decreasing concentrations of chaotropic reagent and in the presence of oxidized glutathione. The efficiency of refolding was demonstrated by protein mobility shifting and fluorescent visualization of labeled cysteine in non-reducing SDS-PAGE. The identity and the fully oxidized state of the protein were verified by mass spectrometry. Analysis of the structure by fluorescence, differential scanning calorimetry and circular dichroism showed a well-formed structural conformation mainly composed of ß-strands. A label-free immunoassay based on biolayer interferometry technology was subsequently used to evaluate antigenic properties of folded EDIIIT2 protein using a panel of dengue IgM positive and negative human sera. Our data collectively support the use of an oxidatively refolded EDIIIT2 recombinant chimeric protein as a promising antigen in the serological diagnosis of dengue virus infections.


Asunto(s)
Anticuerpos Antivirales , Antígenos Virales , Virus del Dengue/genética , Dengue , Epítopos , Inmunoglobulina M , Proteínas Virales , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Antígenos Virales/biosíntesis , Antígenos Virales/genética , Antígenos Virales/inmunología , Antígenos Virales/aislamiento & purificación , Dengue/sangre , Dengue/diagnóstico , Dengue/inmunología , Virus del Dengue/inmunología , Virus del Dengue/metabolismo , Epítopos/biosíntesis , Epítopos/genética , Epítopos/inmunología , Epítopos/aislamiento & purificación , Humanos , Inmunoglobulina M/sangre , Inmunoglobulina M/inmunología , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/aislamiento & purificación , Proteínas Virales/biosíntesis , Proteínas Virales/genética , Proteínas Virales/inmunología , Proteínas Virales/aislamiento & purificación
14.
Protein Expr Purif ; 130: 28-34, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27693623

RESUMEN

The Norovirus (NoV) P domain, with three surface loops for foreign antigen insertion, has been demonstrated as an excellent platform for antigen presentation and novel vaccine development. The P domain alone can self-assemble into a P dimer, 12-mer small particle or 24-mer P particle, and vaccines based on those particles may elicit different levels of immunogenicity. Currently, P particles are generally produced in soluble expression systems in Escherichia coli, mainly in the 24-mer form. However, the low yield of the soluble protein has hindered further clinical applications of P particle-based protein vaccines. In this study, we inserted the Alzheimer's disease (AD) immunogen Aß1-6 into the three loops of the P particle to generate an AD protein vaccine. To increase the yield of this chimeric protein, we tested the generation of proteins in a soluble expression system and an inclusion body expression system separately in E. coli. The result showed that the inclusion body expression system could greatly enhance the product yield of the chimeric protein compared with the soluble expression system. The refolded protein from the inclusion bodies was mainly in the 12-mer form, while the protein generated from the soluble supernatant was mainly in the 24-mer form. Moreover, the immunogenicity of soluble proteins was significantly stronger than that of the refolded proteins. Thus, comparisons between the two expression methods suggested that the soluble expression system generated chimeric P particles with better immunogenicity, while inclusion body expression system yielded more P particle proteins.


Asunto(s)
Antígenos Virales , Expresión Génica , Norovirus/genética , Proteínas Recombinantes de Fusión , Proteínas Virales , Vacunas Virales , Animales , Antígenos Virales/biosíntesis , Antígenos Virales/química , Antígenos Virales/genética , Antígenos Virales/aislamiento & purificación , Escherichia coli/genética , Escherichia coli/metabolismo , Femenino , Inmunogenicidad Vacunal , Ratones , Pichia/genética , Pichia/metabolismo , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Virales/biosíntesis , Proteínas Virales/química , Proteínas Virales/genética , Proteínas Virales/aislamiento & purificación , Vacunas Virales/biosíntesis , Vacunas Virales/química , Vacunas Virales/genética , Vacunas Virales/aislamiento & purificación
15.
Proc Natl Acad Sci U S A ; 111(1): 125-30, 2014 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-24344259

RESUMEN

The rapid dissemination of the 2009 pandemic H1N1 influenza virus emphasizes the need for universal influenza vaccines that would broadly protect against multiple mutated strains. Recent efforts have focused on the highly conserved hemagglutinin (HA) stem domain, which must undergo a significant conformational change for effective viral infection. Although the production of isolated domains of multimeric ectodomain proteins has proven difficult, we report a method to rapidly produce the properly folded HA stem domain protein from influenza virus A/California/05/2009 (H1N1) by using Escherichia coli-based cell-free protein synthesis and a simple refolding protocol. The T4 bacteriophage fibritin foldon placed at the C terminus of the HA stem domain induces trimer formation. Placing emphasis on newly exposed protein surfaces, several hydrophobic residues were mutated, two polypeptide segments were deleted, and the number of disulfide bonds in each monomer was reduced from four to two. High pH and Brij 35 detergent emerged as the most beneficial factors for improving the refolding yield. To stabilize the trimer of the HA stem-foldon fusion, new intermolecular disulfide bonds were finally introduced between foldon monomers and between stem domain monomers. The correct immunogenic conformation of the stabilized HA stem domain trimer was confirmed by using antibodies CR6261, C179, and FI6 that block influenza infection by binding to the HA stem domain trimer. These results suggest great promise for a broadly protective vaccine and also demonstrate a unique approach for producing individual domains of complex multimeric proteins.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza/biosíntesis , Subtipo H1N1 del Virus de la Influenza A/química , Vacunas contra la Influenza/biosíntesis , Anticuerpos Neutralizantes/química , Anticuerpos Antivirales/química , Antígenos Virales/biosíntesis , Bacteriófago T4/química , Sistema Libre de Células , Cristalografía por Rayos X , Disulfuros/química , Relación Dosis-Respuesta a Droga , Escherichia coli/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Gripe Humana/prevención & control , Modelos Moleculares , Desnaturalización Proteica , Pliegue de Proteína , Multimerización de Proteína , Estructura Terciaria de Proteína
16.
Vopr Virusol ; 62(1): 17-25, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29323842

RESUMEN

A task of creating a universal platform for engineering affordable recombinant producers of viral proteins conserving immunogenicity has not been solved yet. High toxicity of the viral proteins for the host cells, low yield and abnormal folding of the products often present severe obstacles to obtaining producers of the viral proteins. In this work, we report a new method of engineering and screening of deletion libraries from the viral antigen genes. This method allows selection of artificial derivatives of these genes adapted for expression in microbial producer cells. The method involves PCR amplification of the gene fragments using a system of randomized and adapter primers, which allows the spontaneous formation of duplexes from the random primers in the absence of the template DNA to be prevented. For selecting variants capable of in vivo expression, the obtained PCR products are cloned to a special vector of a direct phenotypical selection pQL30. It contains E. coli ß-galactosidase gene with an inserted polylinker producing a frame-shift mutation. Using this screening method, an artificial variant of hepatitis C (HCV) NS5a gene with optimal biotechnological properties was established. 27 clinical specimens of 1670 bp long HCV1b NS5a fragments were used as a source gene. A PCR bank of the deletion derivatives was produced. 40 LacZ-positive clones based on pQL30 vector with a 50-700 bp long insertion were selected. The LacZ activity of the cell lysates and the immunogenicity of the products were tested. As a result, a single clone encoding a soluble protein with Mr = 114 kDa was selected. Its yield reached 0.3% of the total cell protein. It was highly reactive with sera of HCV 1b infected patients but not with sera of the healthy donors.


Asunto(s)
Antígenos Virales/genética , Escherichia coli/genética , Hepacivirus/genética , Hepatitis C/diagnóstico , Ingeniería de Proteínas/métodos , Proteínas Recombinantes/genética , Proteínas no Estructurales Virales/genética , Antígenos Virales/biosíntesis , Antígenos Virales/inmunología , Antígenos Virales/aislamiento & purificación , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Cartilla de ADN/síntesis química , Cartilla de ADN/genética , Escherichia coli/metabolismo , Mutación del Sistema de Lectura , Biblioteca de Genes , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Hepacivirus/inmunología , Hepatitis C/inmunología , Hepatitis C/virología , Humanos , Sueros Inmunes/química , Operón Lac , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/aislamiento & purificación , Proteínas no Estructurales Virales/biosíntesis , Proteínas no Estructurales Virales/inmunología , Proteínas no Estructurales Virales/aislamiento & purificación , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
17.
Plant Mol Biol ; 91(4-5): 497-512, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27116001

RESUMEN

Dengue fever is a disease in many parts of the tropics and subtropics and about half the world's population is at risk of infection according to the World Health Organization. Dengue is caused by any of the four related dengue virus serotypes DEN-1, -2, -3 and -4, which are transmitted to people by Aedes aegypti mosquitoes. Currently there is only one vaccine (Dengvaxia(®)) available (limited to a few countries) on the market since 2015 after half a century's intensive efforts. Affordable and accessible vaccines against dengue are hence still urgently needed. The dengue envelop protein domain III (EDIII), which is capable of eliciting serotype-specific neutralizing antibodies, has become the focus for subunit vaccine development. To contribute to the development of an accessible and affordable dengue vaccine, in the current study we have used plant-based vaccine production systems to generate a dengue subunit vaccine candidate in tobacco. Chloroplast genome engineering was applied to express serotype-specific recombinant EDIII proteins in tobacco chloroplasts using both constitutive and ethanol-inducible expression systems. Expression of a tetravalent antigen fusion construct combining EDIII polypeptides from all four serotypes was also attempted. Transplastomic EDIII-expressing tobacco lines were obtained and homoplasmy was verified by Southern blot analysis. Northern blot analyses showed expression of EDIII antigen-encoding genes. EDIII protein accumulation levels varied for the different recombinant EDIII proteins and the different expression systems, and reached between 0.8 and 1.6 % of total cellular protein. Our study demonstrates the suitability of the chloroplast compartment as a production site for an EDIII-based vaccine candidate against dengue fever and presents a Gateway(®) plastid transformation vector for inducible transgene expression.


Asunto(s)
Antígenos Virales/biosíntesis , Cloroplastos/genética , Virus del Dengue/inmunología , Técnicas Genéticas , Nicotiana/genética , Secuencia de Aminoácidos , Antígenos Virales/química , Antígenos Virales/metabolismo , Etanol/farmacología , Vectores Genéticos/metabolismo , Plantas Modificadas Genéticamente , Dominios Proteicos , Proteínas Recombinantes/metabolismo , Regeneración , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo
18.
J Virol ; 89(17): 8922-31, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26085145

RESUMEN

The Epstein-Barr virus (EBV) capsid contains a major capsid protein, VCA; two minor capsid proteins, BDLF1 and BORF1; and a small capsid protein, BFRF3. During the lytic cycle, these capsid proteins are synthesized and imported into the host nucleus for capsid assembly. This study finds that EBV capsid proteins colocalize with promyelocytic leukemia (PML) nuclear bodies (NBs) in P3HR1 cells during the viral lytic cycle, appearing as nuclear speckles under a confocal laser scanning microscope. In a glutathione S-transferase pulldown study, we show that BORF1 interacts with PML-NBs in vitro. BORF1 also colocalizes with PML-NBs in EBV-negative Akata cells after transfection and is responsible for bringing VCA and the VCA-BFRF3 complex from the cytoplasm to PML-NBs in the nucleus. Furthermore, BDLF1 is dispersed throughout the cell when expressed alone but colocalizes with PML-NBs when BORF1 is also present in the cell. In addition, this study finds that knockdown of PML expression by short hairpin RNA does not influence the intracellular levels of capsid proteins but reduces the number of viral particles produced by P3HR1 cells. Together, these results demonstrate that BORF1 plays a critical role in bringing capsid proteins to PML-NBs, which may likely be the assembly sites of EBV capsids. The mechanisms elucidated in this study are critical to understanding the process of EBV capsid assembly. IMPORTANCE Capsid assembly is an important event during the Epstein-Barr virus (EBV) lytic cycle, as this process is required for the production of virions. In this study, confocal microscopy revealed that the EBV capsid protein BORF1 interacts with promyelocytic leukemia (PML) nuclear bodies (NBs) in the host nucleus and is responsible for transporting the other EBV capsid proteins, including VCA, BDLF1, and BFRF3, to these subnuclear locations prior to initiation of capsid assembly. This study also found that knockdown of PML expression by short hairpin RNA significantly reduces EBV capsid assembly capabilities. This enhanced understanding of capsid assembly offers potential for the development of novel antiviral strategies and therapies that can prevent the propagation and spread of EBV.


Asunto(s)
Transporte Activo de Núcleo Celular/genética , Antígenos Virales/metabolismo , Proteínas de la Cápside/metabolismo , Cápside/metabolismo , Herpesvirus Humano 4/metabolismo , Proteínas de Neoplasias/metabolismo , Antígenos Virales/biosíntesis , Antígenos Virales/genética , Proteínas de la Cápside/biosíntesis , Proteínas de la Cápside/genética , Línea Celular Tumoral , Células HEK293 , Herpesvirus Humano 4/genética , Humanos , Leucemia Promielocítica Aguda/virología , Microscopía Confocal , Proteínas Nucleares/metabolismo , Transporte de Proteínas/genética , Interferencia de ARN , ARN Interferente Pequeño
19.
J Virol ; 89(18): 9262-80, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26109721

RESUMEN

UNLABELLED: Viruses often hijack cellular pathways to facilitate infection and replication. Kaposi's sarcoma-associated herpesvirus (KSHV) is an oncogenic gammaherpesvirus etiologically associated with Kaposi's sarcoma, a vascular tumor of endothelial cells. Despite intensive studies, cellular pathways mediating KSHV infection and replication are still not well defined. Using an antibody array approach, we examined cellular proteins phosphorylated during primary KSHV infection of primary human umbilical vein endothelial cells. Enrichment analysis identified integrin/mitogen-activated protein kinase (integrin/MAPK), insulin/epidermal growth factor receptor (insulin/EGFR), and JAK/STAT as the activated networks during primary KSHV infection. The transcriptional factor CREB1 (cyclic AMP [cAMP]-responsive element-binding protein 1) had the strongest increase in phosphorylation. While knockdown of CREB1 had no effect on KSHV entry and trafficking, it drastically reduced the expression of lytic transcripts and proteins and the production of infectious virions. Chemical activation of CREB1 significantly enhanced viral lytic replication. In contrast, CREB1 neither influenced the expression of the latent gene LANA nor affected KSHV infectivity. Mechanistically, CREB1 was not activated through the classic cAMP/protein kinase A (cAMP/PKA) pathway or via the AKT, MK2, and RSK pathways. Rather, CREB1 was activated by the mitogen- and stress-activated protein kinases 1 and 2 (MSK1/2). Consequently, chemical inhibition or knockdown of MSKs significantly inhibited the KSHV lytic replication program; however, it had a minimal effect on LANA expression and KSHV infectivity. Together, these results identify the MSK1/2-CREB1 proteins as novel essential effectors of KSHV lytic replication during primary infection. The differential effect of the MSK1/2-CREB1 pathway on the expression of viral latent and lytic genes might control the robustness of viral lytic replication, and therefore the KSHV replication program, during primary infection. IMPORTANCE: Kaposi's sarcoma-associated herpesvirus (KSHV) is a human tumor virus associated with several cancers. Through genome-wide kinase screening, we found that KSHV activates the MSK1/2-CREB1 pathway during primary infection and that it depends on this pathway for viral lytic replication. Inhibition of this pathway blocks KSHV lytic replication. These results illustrate a mechanism by which KSHV hijacks a cellular pathway for its replication, and they identify a potential therapeutic target.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Infecciones por Herpesviridae/metabolismo , Herpesvirus Humano 8/fisiología , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Sistemas de Mensajero Secundario , Replicación Viral/fisiología , Antígenos Virales/biosíntesis , Antígenos Virales/genética , AMP Cíclico/genética , AMP Cíclico/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Regulación Viral de la Expresión Génica/fisiología , Técnicas de Silenciamiento del Gen , Infecciones por Herpesviridae/genética , Infecciones por Herpesviridae/patología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Fosforilación/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas S6 Ribosómicas 90-kDa/genética
20.
PLoS Pathog ; 10(6): e1004195, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24945302

RESUMEN

The devastating clinical consequences associated with human cytomegalovirus (HCMV) infection and reactivation underscores the importance of understanding triggers of HCMV reactivation in dendritic cells (DC). Here we show that ERK-mediated reactivation is dependent on the mitogen and stress activated kinase (MSK) family. Furthermore, this MSK mediated response is dependent on CREB binding to the viral major immediate early promoter (MIEP). Specifically, CREB binding to the MIEP provides the target for MSK recruitment. Importantly, MSK mediated phosphorylation of histone H3 is required to promote histone de-methylation and the subsequent exit of HCMV from latency. Taken together, these data suggest that CREB binding to the MIEP is necessary for the recruitment of the kinase activity of MSKs to initiate the chromatin remodelling at the MIEP required for reactivation. Thus the importance of CREB during HCMV reactivation is to promote chromatin modifications conducive for viral gene expression as well as acting as a classical transcription factor. Clearly, specific inhibition of this interaction between CREB and MSKs could provide a strategy for therapeutic intervention.


Asunto(s)
Antígenos Virales/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Inmediatas-Precoces/genética , MAP Quinasa Quinasa 1/metabolismo , Transactivadores/genética , Antígenos Virales/biosíntesis , Sitios de Unión , Células Cultivadas , Citomegalovirus/genética , Infecciones por Citomegalovirus/patología , Proteínas de Unión al ADN/metabolismo , Células Dendríticas/virología , Regulación Viral de la Expresión Génica/genética , Histonas/metabolismo , Humanos , Proteínas Inmediatas-Precoces/biosíntesis , Sistema de Señalización de MAP Quinasas , Ácido Ocadaico/farmacología , Fosforilación , Regiones Promotoras Genéticas/genética , Unión Proteica , Transactivadores/biosíntesis , Activación Viral/fisiología , Latencia del Virus/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA