Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
3.
Proc Natl Acad Sci U S A ; 118(45)2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34670842

RESUMEN

Vaccination against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and other pathogens with pandemic potential requires safe, protective, inexpensive, and easily accessible vaccines that can be developed and manufactured rapidly at a large scale. DNA vaccines can achieve these criteria, but induction of strong immune responses has often required bulky, expensive electroporation devices. Here, we report an ultra-low-cost (<1 USD), handheld (<50 g) electroporation system utilizing a microneedle electrode array ("ePatch") for DNA vaccination against SARS-CoV-2. The low cost and small size are achieved by combining a thumb-operated piezoelectric pulser derived from a common household stove lighter that emits microsecond, bipolar, oscillatory electric pulses and a microneedle electrode array that targets delivery of high electric field strength pulses to the skin's epidermis. Antibody responses against SARS-CoV-2 induced by this electroporation system in mice were strong and enabled at least 10-fold dose sparing compared to conventional intramuscular or intradermal injection of the DNA vaccine. Vaccination was well tolerated with mild, transient effects on the skin. This ePatch system is easily portable, without any battery or other power source supply, offering an attractive, inexpensive approach for rapid and accessible DNA vaccination to combat COVID-19, as well as other epidemics.


Asunto(s)
Vacunas contra la COVID-19/administración & dosificación , COVID-19/inmunología , COVID-19/prevención & control , Electroporación/instrumentación , SARS-CoV-2 , Vacunas de ADN/administración & dosificación , Animales , Vacunas contra la COVID-19/genética , Vacunas contra la COVID-19/inmunología , Costos y Análisis de Costo , Electroporación/economía , Electroporación/métodos , Diseño de Equipo , Femenino , Genes Reporteros , Humanos , Ratones , Ratones Endogámicos BALB C , Microelectrodos , Agujas , Pandemias/prevención & control , Prueba de Estudio Conceptual , Ratas , Ratas Wistar , Piel/inmunología , Piel/metabolismo , Transfección , Vacunación/economía , Vacunación/instrumentación , Vacunación/métodos , Vacunas de ADN/genética , Vacunas de ADN/inmunología
4.
Front Microbiol ; 11: 304, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32174901

RESUMEN

Ebolavirus (EBOV) infection in humans causes severe hemorrhagic fevers with high mortality rates that range from 30 to 80% as shown in different outbreaks. Thus the development of safe and efficacious EBOV vaccines remains an important goal for biomedical research. We have shown in early studies that immunization with insect cell-produced EBOV virus-like particles (VLPs) is able to induce protect vaccinated mice against lethal EBOV challenge. In the present study, we investigated immune responses induced by Ebola VLPs via two different routes, intramuscular and intradermal immunizations, in guinea pigs. Analyses of antibody responses revealed that similar levels of total IgG antibodies against the EBOV glycoprotein (GP) were induced by the two different immunization methods. However, further characterization showed that the EBOV GP-specific antibodies induced by intramuscular immunization were mainly of the IgG2 subtype whereas both IgG1 and IgG2 antibodies against EBOV GP were induced by intradermal immunization. In contrast, antibody responses against the EBOV matrix protein VP40 induced by intramuscular or intradermal immunizations exhibited similar IgG1 and IgG2 profiles. More interestingly, we found that the sites that the IgG1 antibodies induced by intradermal immunizations bind to in GP are different from those that bind to the IgG2 antibodies induced by intramuscular immunization. Further analyses revealed that sera from all vaccinated guinea pigs exhibited neutralizing activity against Ebola GP-mediated HIV pseudovirion infection at high levels. Moreover, all EBOV VLP-vaccinated guinea pigs survived the challenge by a high dose (1000 pfu) of guinea pig-adapted EBOV, while all control guinea pigs immunized with irrelevant VLPs succumbed to the challenge. The induction of both IgG1 and IgG2 antibody responses that recognized broader sites in GP by intradermal immunization of EBOV VLPs indicates that this approach may represent a more advantageous route of vaccination against virus infection.

5.
Sci Rep ; 8(1): 11193, 2018 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-30046140

RESUMEN

Development of a safe and efficacious filovirus vaccine is of high importance to public health. In this study, we compared immune responses induced by Ebola virus (EBOV) glycoprotein (GP) subunit vaccines via intradermal immunization with microneedle (MN) patches and the conventional intramuscular (IM) injection in mice, which showed that MN delivery of GP induced higher levels and longer lasting antibody responses against GP than IM injection. Further, we found that EBOV GP in formulation with a saponin-based adjuvant, Matrix-M, can be efficiently loaded onto MN patches. Co-delivery of Matrix-M with GP significantly enhanced induction of antibody responses by MN delivery, as also observed for IM injection. Results from challenge studies showed that all mice that received the GP/adjuvant formulation by MN or IM immunizations were protected from lethal EBOV challenge. Further, 4 out of 5 mice vaccinated by MN delivery of unadjuvanted GP also survived the challenge, whereas only 1 out of 5 mice vaccinated by IM injection of unadjuvanted GP survived the challenge. These results demonstrate that MN patch delivery of EBOV GP subunit vaccines, which is expected to enable improved safety and thermal stability, can confer effective protection against EBOV infection that is superior to IM vaccination.


Asunto(s)
Anticuerpos Antivirales/inmunología , Vacunas contra el Virus del Ébola/inmunología , Glicoproteínas/administración & dosificación , Fiebre Hemorrágica Ebola/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Animales , Anticuerpos Antivirales/genética , Formación de Anticuerpos/inmunología , Ebolavirus/inmunología , Ebolavirus/patogenicidad , Glicoproteínas/inmunología , Fiebre Hemorrágica Ebola/genética , Fiebre Hemorrágica Ebola/prevención & control , Humanos , Inmunización , Inyecciones Intradérmicas , Ratones , Vacunación , Vacunas de Subunidad/genética , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/uso terapéutico
6.
J Infect Dis ; 218(suppl_5): S545-S552, 2018 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-29893888

RESUMEN

In this study, we investigated immune responses induced by purified Ebola virus (EBOV) soluble glycoprotein (sGP) subunit vaccines via intradermal immunization with microneedle (MN) patches in comparison with intramuscular (IM) injection in mice. Our results showed that MN delivery of EBOV sGP was superior to IM injection in eliciting higher levels and longer lasting antibody responses against EBOV sGP and GP antigens. Moreover, sGP-specific immune responses induced by MN or IM immunizations were effectively augmented by formulating sGP with a saponin-based adjuvant, and they were shown to confer complete protection of mice against lethal mouse-adapted EBOV (MA-EBOV) challenge. In comparison, mice that received sGP without adjuvant by MN or IM immunizations succumbed to lethal MA-EBOV challenge. These results show that immunization with EBOV sGP subunit vaccines with adjuvant by MN patches, which have been shown to provide improved safety and thermal stability, is a promising approach to protect against EBOV infection.


Asunto(s)
Vacunas contra el Virus del Ébola/inmunología , Vacunación , Adyuvantes Inmunológicos/administración & dosificación , Animales , Anticuerpos Antivirales/biosíntesis , Formación de Anticuerpos , Vacunas contra el Virus del Ébola/administración & dosificación , Femenino , Células HEK293 , Células HeLa , Fiebre Hemorrágica Ebola/prevención & control , Humanos , Inyecciones Intramusculares , Ratones , Ratones Endogámicos BALB C , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/inmunología
7.
Microbes Infect ; 19(12): 626-634, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28899815

RESUMEN

Elderly humans over 65 years old are at great risk to pathogenesis by influenza virus infection. However, although influenza vaccines provide effective protection in healthy young adults, protection of elderly adults is substantially lower even with a good match between the vaccine and the circulating influenza virus. To gain insight of the underlying mechanism for the reduced immunogenicity of influenza vaccines in the aged population, we investigated immunogenicity of influenza virus-like particle vaccines in aged mice, which represent a useful model for studying aging associated impairment in immune responses. Specifically, we investigated the effect of inhibiting regulatory T cells in aged mice on induction of protective immune responses by influenza vaccines. Our results showed that injecting anti-CD25 antibodies could down-regulate CD25 on the surface of regulatory T cells and significantly increase the levels of antibody responses induced by VLP immunization in aged mice. Further, the profiles of antibody responses were also changed towards Th1 type by regulatory T cell blockage in aged mice. Moreover, aged mice that were treated by anti-CD25 antibodies prior to vaccination were more effectively protected against lethal influenza virus challenge.


Asunto(s)
Envejecimiento/inmunología , Anticuerpos Antivirales/sangre , Subtipo H1N1 del Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Linfocitos T Reguladores/inmunología , Vacunas de Partículas Similares a Virus/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/inmunología , Formación de Anticuerpos/inmunología , Femenino , Inmunogenicidad Vacunal/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Vacunas contra la Influenza/inmunología , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/virología , Células TH1/inmunología , Vacunación
9.
Emerg Microbes Infect ; 5: e65, 2016 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-27329850

RESUMEN

After three decades of intensive research efforts, an effective vaccine against HIV-1 remains to be developed. Several broadly neutralizing antibodies to HIV-1, such as 10E8, recognize the membrane proximal external region (MPER) of the HIV-1 gp41 protein. Thus, the MPER is considered to be a very important target for vaccine design. However, the MPER segment has very weak immunogenicity and tends to insert its epitope residues into the cell membrane, thereby avoiding antibody binding. To address this complication in vaccine development, we herein designed a peptide, designated 10E8-4P, containing four copies of the 10E8 epitope as an immunogen. As predicted by structural simulation, 10E8-4P exhibits a well-arranged tandem helical conformation, with the key residues in the 10E8 epitope oriented at different angles, thus suggesting that some of these key residues may be exposed outside of the lipid membrane. Compared with a peptide containing a single 10E8 epitope (10E8-1P), 10E8-4P not only exhibited better antigenicity but also elicited neutralizing antibody response against HIV-1 pseudoviruses, whereas 10E8-1P could not induce detectable neutralizing antibody response. Importantly, antibodies elicited by 10E8-4P also possessed a strong ability to activate an antibody-dependent cell-mediated cytotoxicity (ADCC) reporter gene, thus suggesting that they may have ADCC activity. Therefore, this strategy shows promise for further optimization and application in future HIV-1 vaccine design.


Asunto(s)
Vacunas contra el SIDA/inmunología , Anticuerpos Neutralizantes/biosíntesis , Citotoxicidad Celular Dependiente de Anticuerpos/genética , Epítopos/inmunología , Anticuerpos Anti-VIH/inmunología , VIH-1/inmunología , Vacunas contra el SIDA/química , Animales , Dicroismo Circular , Ensayo de Inmunoadsorción Enzimática , Epítopos/química , Genes Reporteros , Anticuerpos Anti-VIH/biosíntesis , Humanos , Pruebas de Neutralización , Unión Proteica , Conejos
10.
Sci Rep ; 6: 24897, 2016 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-27109916

RESUMEN

Polyclonal antibodies, derived from humans or hyperimmunized animals, have been used prophylactically or therapeutically as countermeasures for a variety of infectious diseases. SAB Biotherapeutics has successfully developed a transchromosomic (Tc) bovine platform technology that can produce fully human immunoglobulins rapidly, and in substantial quantities, against a variety of disease targets. In this study, two Tc bovines expressing high levels of fully human IgG were hyperimmunized with a recombinant glycoprotein (GP) vaccine consisting of the 2014 Ebola virus (EBOV) Makona isolate. Serum collected from these hyperimmunized Tc bovines contained high titers of human IgG against EBOV GP as determined by GP specific ELISA, surface plasmon resonance (SPR), and virus neutralization assays. Fully human polyclonal antibodies against EBOV were purified and evaluated in a mouse challenge model using mouse adapted Ebola virus (maEBOV). Intraperitoneal administration of the purified anti-EBOV IgG (100 mg/kg) to BALB/c mice one day after lethal challenge with maEBOV resulted in 90% protection; whereas 100% of the control animals succumbed. The results show that hyperimmunization of Tc bovines with EBOV GP can elicit protective and potent neutralizing fully human IgG antibodies rapidly and in commercially viable quantities.


Asunto(s)
Animales Modificados Genéticamente , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/uso terapéutico , Bovinos , Ebolavirus/inmunología , Fiebre Hemorrágica Ebola/prevención & control , Animales , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Humanos , Inmunización Pasiva , Inmunoglobulina G/sangre , Inmunoglobulina G/uso terapéutico , Ratones Endogámicos BALB C , Pruebas de Neutralización , Resonancia por Plasmón de Superficie , Resultado del Tratamiento
11.
Pharm Res ; 33(4): 868-78, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26620313

RESUMEN

PURPOSE: This study tested the hypothesis that encapsulation of influenza vaccine in microneedle patches increases vaccine stability during storage at elevated temperature. METHODS: Whole inactivated influenza virus vaccine (A/Puerto Rico/8/34) was formulated into dissolving microneedle patches and vaccine stability was evaluated by in vitro and in vivo assays of antigenicity and immunogenicity after storage for up to 3 months at 4, 25, 37 and 45°C. RESULTS: While liquid vaccine completely lost potency as determined by hemagglutination (HA) activity within 1-2 weeks outside of refrigeration, vaccine in microneedle patches lost 40-50% HA activity during or shortly after fabrication, but then had no significant additional loss of activity over 3 months of storage, independent of temperature. This level of stability required reduced humidity by packaging with desiccant, but was not affected by presence of oxygen. This finding was consistent with additional stability assays, including antigenicity of the vaccine measured by ELISA, virus particle morphological structure captured by transmission electron microscopy and protective immune responses by immunization of mice in vivo. CONCLUSIONS: These data show that inactivated influenza vaccine encapsulated in dissolving microneedle patches has enhanced stability during extended storage at elevated temperatures.


Asunto(s)
Sistemas de Liberación de Medicamentos , Subtipo H1N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/administración & dosificación , Agujas , Infecciones por Orthomyxoviridae/prevención & control , Vacunas de Productos Inactivados/administración & dosificación , Animales , Almacenaje de Medicamentos , Calor , Humanos , Inmunización , Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/uso terapéutico , Gripe Humana/inmunología , Gripe Humana/prevención & control , Ratones , Infecciones por Orthomyxoviridae/inmunología , Parche Transdérmico , Vacunas de Productos Inactivados/inmunología , Vacunas de Productos Inactivados/uso terapéutico
12.
PLoS One ; 10(8): e0133910, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26263554

RESUMEN

BACKGROUND: Human infections with avian influenza viruses (AIVs) have frequently raised global concerns of emerging, interspecies-transmissible viruses with pandemic potential. Waterfowl, the predominant reservoir of influenza viruses in nature, harbor precursors of different genetic lineages that have contributed to novel pandemic influenza viruses in the past. METHODS: Two duck influenza H5N2 viruses, DV518 and DV413, isolated through virological surveillance at a live-poultry market in Taiwan, showed phylogenetic relatedness but exhibited different replication capabilities in mammalian Madin-Darby Canine Kidney (MDCK) cells. This study characterizes the replication properties of the two duck H5N2 viruses and the determinants involved. RESULTS: The DV518 virus replicated more efficiently than DV413 in both MDCK and chicken DF1 cells. Interestingly, the infection of MDCK cells by DV518 formed heterogeneous plaques with great differences in size [large (L) and small (S)], and the two viral strains (p518-L and p518-S) obtained from plaque purification exhibited distinguishable replication kinetics in MDCK cells. Nonetheless, both plaque-purified DV518 strains still maintained their growth advantages over the plaque-purified p413 strain. Moreover, three amino acid substitutions in PA (P224S), PB2 (E72D), and M1 (A128T) were identified in intra-duck variations (p518-L vs p518-S), whereas other changes in HA (N170D), NA (I56T), and NP (Y289H) were present in inter-duck variations (DV518 vs DV413). Both p518-L and p518-S strains had the N170D substitution in HA, which might be related to their greater binding to MDCK cells. Additionally, polymerase activity assays on 293T cells demonstrated the role of vRNP in modulating the replication capability of the duck p518-L viruses in mammalian cells. CONCLUSION: These results demonstrate that intra-host phenotypic variation occurs even within an individual duck. In view of recent human infections by low pathogenic AIVs, this study suggests possible determinants involved in the stepwise selection of virus variants from the duck influenza virus population which may facilitate inter-species transmission.


Asunto(s)
Patos , Genotipo , Subtipo H5N2 del Virus de la Influenza A/fisiología , Gripe Aviar/virología , Fenotipo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Evolución Biológica , Línea Celular , Células Cultivadas , Embrión de Pollo , Variación Genética , Genoma Viral , Humanos , Subtipo H5N2 del Virus de la Influenza A/aislamiento & purificación , Taiwán , Ensayo de Placa Viral , Proteínas Virales/química , Proteínas Virales/genética , Replicación Viral
13.
J Infect Dis ; 212 Suppl 2: S398-403, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25877553

RESUMEN

In addition to its surface glycoprotein (GP), Ebola virus directs the production of large quantities of a truncated glycoprotein isoform (sGP) that is secreted into the extracellular space. We recently reported that sGP actively diverts host antibody responses against the epitopes that it shares with GP and thereby allows itself to absorb anti-GP antibodies, a phenomenon we termed "antigenic subversion." To investigate the effect of antigenic subversion by sGP on protection against virus infection, we compared immune responses induced by different prime-boost immunization regimens with GP and sGP DNA vaccines in mice and their efficacy against lethal Ebola virus challenge. Similar levels of anti-GP antibodies were induced by 2 immunizations with sGP and GP DNA vaccines. However, 2 immunizations with GP but not sGP DNA vaccine fully protected mice from lethal challenge. Boosting with sGP or GP DNA vaccine in mice that had been primed by GP or sGP DNA vaccine augmented the levels of anti-GP antibody responses and further improved protective efficacy against Ebola virus infection. These results show that both the quality and the levels of anti-GP antibody responses affect the efficacy of protection against Ebola virus infection.


Asunto(s)
Vacunas contra el Virus del Ébola/inmunología , Ebolavirus/inmunología , Glicoproteínas/inmunología , Fiebre Hemorrágica Ebola/inmunología , Isoformas de Proteínas/inmunología , Vacunas de ADN/inmunología , Proteínas Virales/inmunología , Animales , Anticuerpos Antivirales/inmunología , Formación de Anticuerpos/inmunología , Femenino , Células HEK293 , Fiebre Hemorrágica Ebola/virología , Humanos , Inmunización Secundaria/métodos , Ratones , Ratones Endogámicos BALB C , Vacunación/métodos
14.
J Virol ; 89(2): 1205-17, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25392212

RESUMEN

UNLABELLED: The Ebola virus (EBOV) surface glycoprotein (GP1,2) mediates host cell attachment and fusion and is the primary target for host neutralizing antibodies. Expression of GP1,2 at high levels disrupts normal cell physiology, and EBOV uses an RNA-editing mechanism to regulate expression of the GP gene. In this study, we demonstrate that high levels of GP1,2 expression impair production and release of EBOV virus-like particles (VLPs) as well as infectivity of GP1,2-pseudotyped viruses. We further show that this effect is mediated through two mechanisms. First, high levels of GP1,2 expression reduce synthesis of other proteins needed for virus assembly. Second, viruses containing high levels of GP1,2 are intrinsically less infectious, possibly due to impaired receptor binding or endosomal processing. Importantly, proteolysis can rescue the infectivity of high-GP1,2-containing viruses. Taken together, our findings indicate that GP1,2 expression levels have a profound effect on factors that contribute to virus fitness and that RNA editing may be an important mechanism employed by EBOV to regulate GP1,2 expression in order to optimize virus production and infectivity. IMPORTANCE: The Ebola virus (EBOV), as well as other members of the Filoviridae family, causes severe hemorrhagic fever that is highly lethal, with up to 90% mortality. The EBOV surface glycoprotein (GP1,2) plays important roles in virus infection and pathogenesis, and its expression is tightly regulated by an RNA-editing mechanism during virus replication. Our study demonstrates that the level of GP1,2 expression profoundly affects virus particle production and release and uncovers a new mechanism by which Ebola virus infectivity is regulated by the level of GP1,2 expression. These findings extend our understanding of EBOV infection and replication in adaptation of host environments, which will aid the development of countermeasures against EBOV infection.


Asunto(s)
Ebolavirus/fisiología , Regulación Viral de la Expresión Génica , Glicoproteínas de Membrana/biosíntesis , Internalización del Virus , Liberación del Virus , Replicación Viral , Línea Celular , Humanos , Edición de ARN
15.
Microbes Infect ; 17(2): 98-108, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25526819

RESUMEN

Ebola virus infection causes severe hemorrhagic fevers with high fatality rates up to 90% in humans, for which no effective treatment is currently available. The ongoing Ebola outbreak in West Africa that has caused over 14,000 human infections and over 5000 deaths underscores its serious threat to the public health. While licensed vaccines against Ebola virus infection are still not available, a number of vaccine approaches have been developed and shown to protect against lethal Ebola virus infection in animal models. This review aims to summarize the advancement of different strategies for Ebola vaccine development with a focus on the discussion of their protective efficacies and possible limitations. In addition, the development of animal models for efficacy evaluation of Ebola vaccines and the mechanism of immune protection against Ebola virus infection are also discussed.


Asunto(s)
Vacunas contra el Virus del Ébola/uso terapéutico , Ebolavirus/inmunología , Fiebre Hemorrágica Ebola/prevención & control , Animales , Control de Enfermedades Transmisibles , Modelos Animales de Enfermedad , Fiebre Hemorrágica Ebola/inmunología , Humanos
16.
Cell Signal ; 26(7): 1532-8, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24686088

RESUMEN

Stathmin is an oncoprotein and is expressed at high levels in a wide variety of human malignancies, which plays important roles in maintenance of malignant phenotypes. The regulation of Stathmin gene overexpression has been wildly explored, but the exact mechanism still needs to be elucidated. It is believed that regulation of an oncogene protein abundance through post-translational modifications is essential for maintenance of malignant phenotypes. Here we identified the Rlim, a Ring H2 zinc finger protein with intrinsic ubiquitin ligase activity, as a Stathmin-interacting protein that could increase Stathmin turnover through binding with this targeted protein and then induce its degradation by proteasome in a ubiquitin-dependent manner. Inhibition of endogenous Rlim expression by siRNA could increase the level of Stathmin protein, which further led to cell proliferation and cell cycle changes in human osteosarcoma cell lines. On the other hand, forced overexpression of Rlim could decrease the level of Stathmin protein. These results demonstrate that Rlim is involved in the negative regulation of Stathmin protein level through physical interaction and ubiquitin-mediated proteolysis. Hence, Rlim is a novel regulator of Stathmin protein in a ubiquitin-dependent manner, and represents a new pathway for malignant phenotype turnover by modulating the level of Stathmin protein in human osteosarcomas.


Asunto(s)
Neoplasias Óseas/metabolismo , Osteosarcoma/metabolismo , Estatmina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Proliferación Celular/genética , Células HEK293 , Humanos , Complejo de la Endopetidasa Proteasomal , Unión Proteica , Procesamiento Proteico-Postraduccional , Proteolisis , Interferencia de ARN , ARN Interferente Pequeño , Estatmina/biosíntesis , Ubiquitina/química , Ubiquitina-Proteína Ligasas/biosíntesis , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
17.
J Virol ; 88(10): 5677-86, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24623422

RESUMEN

UNLABELLED: Sporadic activity by H5N2 influenza viruses has been observed in chickens in Taiwan from 2003 to 2012. The available information suggests that these viruses were generated by reassortment between a Mexican-like H5N2 virus and a local enzootic H6N1 virus. Yet the origin, prevalence, and pathogenicity of these H5N2 viruses have not been fully defined. Following the 2012 highly pathogenic avian influenza (HPAI) outbreaks, surveillance was conducted from December 2012 to July 2013 at a live-poultry wholesale market in Taipei. Our findings showed that H5N2 and H6N1 viruses cocirculated at low levels in chickens in Taiwan. Phylogenetic analyses revealed that all H5N2 viruses had hemagglutinin (HA) and neuraminidase (NA) genes derived from a 1994 Mexican-like virus, while their internal gene complexes were incorporated from the enzootic H6N1 virus lineage by multiple reassortment events. Pathogenicity studies demonstrated heterogeneous results even though all tested viruses had motifs (R-X-K/R-R) supportive of high pathogenicity. Serological surveys for common subtypes of avian viruses confirmed the prevalence of the H5N2 and H6N1 viruses in chickens and revealed an extraordinarily high seroconversion rate to an H9N2 virus, a subtype that is not found in Taiwan but is prevalent in mainland China. These findings suggest that reassortant H5N2 viruses, together with H6N1 viruses, have become established and enzootic in chickens throughout Taiwan and that a large-scale vaccination program might have been conducted locally that likely led to the introduction of the 1994 Mexican-like virus to Taiwan in 2003. IMPORTANCE: H5N2 avian influenza viruses first appeared in chickens in Taiwan in 2003 and caused a series of outbreaks afterwards. Phylogenetic analyses show that the chicken H5N2 viruses have H5 and N2 genes that are closely related to those of a vaccine strain originating from Mexico in 1994, while the contemporary duck H5N2 viruses in Taiwan belong to the Eurasian gene pool. The unusually high similarity of the chicken H5N2 viruses to the Mexican vaccine strain suggests that these viruses might have been introduced to Taiwan by using inadequately inactivated or attenuated vaccines. These chicken H5N2 viruses are developing varying levels of pathogenicity that could lead to significant consequences for the local poultry industry. These findings emphasize the need for strict quality control and competent oversight in the manufacture and usage of avian influenza virus vaccines and indicate that alternatives to widespread vaccination may be desirable.


Asunto(s)
Evolución Molecular , Subtipo H5N2 del Virus de la Influenza A/clasificación , Subtipo H5N2 del Virus de la Influenza A/genética , Gripe Aviar/virología , Animales , Pollos , Análisis por Conglomerados , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Subtipo H5N2 del Virus de la Influenza A/aislamiento & purificación , Datos de Secuencia Molecular , Neuraminidasa/genética , Filogenia , Virus Reordenados/clasificación , Virus Reordenados/genética , Virus Reordenados/aislamiento & purificación , Análisis de Secuencia de ADN , Taiwán , Proteínas Virales/genética
18.
PLoS Pathog ; 8(12): e1003065, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23271969

RESUMEN

In addition to its surface glycoprotein (GP(1,2)), Ebola virus (EBOV) directs the production of large quantities of a truncated glycoprotein isoform (sGP) that is secreted into the extracellular space. The generation of secreted antigens has been studied in several viruses and suggested as a mechanism of host immune evasion through absorption of antibodies and interference with antibody-mediated clearance. However such a role has not been conclusively determined for the Ebola virus sGP. In this study, we immunized mice with DNA constructs expressing GP(1,2) and/or sGP, and demonstrate that sGP can efficiently compete for anti-GP(12) antibodies, but only from mice that have been immunized by sGP. We term this phenomenon "antigenic subversion", and propose a model whereby sGP redirects the host antibody response to focus on epitopes which it shares with membrane-bound GP(1,2), thereby allowing it to absorb anti-GP(1,2) antibodies. Unexpectedly, we found that sGP can also subvert a previously immunized host's anti-GP(1,2) response resulting in strong cross-reactivity with sGP. This finding is particularly relevant to EBOV vaccinology since it underscores the importance of eliciting robust immunity that is sufficient to rapidly clear an infection before antigenic subversion can occur. Antigenic subversion represents a novel virus escape strategy that likely helps EBOV evade host immunity, and may represent an important obstacle to EBOV vaccine design.


Asunto(s)
Anticuerpos Antivirales/inmunología , Ebolavirus/inmunología , Fiebre Hemorrágica Ebola/inmunología , Evasión Inmune/inmunología , Animales , Reacciones Cruzadas/efectos de los fármacos , Reacciones Cruzadas/genética , Vacunas contra el Virus del Ébola/inmunología , Vacunas contra el Virus del Ébola/farmacología , Ebolavirus/genética , Femenino , Células HeLa , Fiebre Hemorrágica Ebola/genética , Fiebre Hemorrágica Ebola/prevención & control , Humanos , Evasión Inmune/genética , Inmunización , Ratones , Ratones Endogámicos BALB C , Vacunas de ADN/inmunología , Vacunas de ADN/farmacología , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología
19.
PLoS One ; 7(7): e41332, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22859976

RESUMEN

The evolution of the H5N1 highly pathogenic avian influenza virus (HPAIV) has resulted in high sequence variations and diverse antigenic properties in circulating viral isolates. We investigated immune responses induced by HA DNA vaccines of two contemporary H5N1 HPAIV isolates, A/bar-headed goose/Qinghai/3/2005 (QH) and A/chicken/Shanxi/2/2006 (SX) respectively, against the homologous as well as the heterologous virus isolate for comparison. Characterization of antibody responses induced by immunization with QH-HA and SX-HA DNA vaccines showed that the two isolates are antigenically distinctive. Interestingly, after immunization with the QH-HA DNA vaccine, subsequent boosting with the SX-HA DNA vaccine significantly augmented antibody responses against the QH isolate but only induced low levels of antibody responses against the SX isolate. Conversely, after immunization with the SX-HA DNA vaccine, subsequent boosting with the QH-HA DNA vaccine significantly augmented antibody responses against the SX isolate but only induced low levels of antibody responses against the QH isolate. In contrast to the antibody responses, cross-reactive T cell responses are readily detected between these two isolates at similar levels. These results indicate the existence of original antigenic sin (OAS) between concurrently circulating H5N1 HPAIV strains, which may need to be taken into consideration in vaccine development against the potential H5N1 HPAIV pandemic.


Asunto(s)
Inmunidad Adaptativa , Antígenos Virales/inmunología , Hemaglutininas Virales/inmunología , Subtipo H5N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/genética , Gripe Aviar/prevención & control , Vacunación , Secuencia de Aminoácidos , Animales , Anticuerpos Antivirales/sangre , Antígenos Virales/biosíntesis , Antígenos Virales/genética , Pollos , Epítopos de Linfocito T/química , Epítopos de Linfocito T/inmunología , Femenino , Gansos , Células HeLa , Hemaglutininas Virales/biosíntesis , Hemaglutininas Virales/genética , Humanos , Inmunización Secundaria , Subtipo H5N1 del Virus de la Influenza A/genética , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/inmunología , Gripe Aviar/inmunología , Gripe Aviar/virología , Ratones , Ratones Endogámicos BALB C , Linfocitos T/inmunología , Vacunas de ADN/administración & dosificación , Vacunas de ADN/genética , Vacunas de ADN/inmunología
20.
Virology ; 432(2): 327-35, 2012 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-22726244

RESUMEN

Nipah virus (NiV), a member of the Paramyxoviridae family, causes deadly encephalitis in humans and huge economic losses to the pig industry. Here, we generated recombinant avirulent Newcastle disease virus (NDV) LaSota strains expressing the NiV G and F proteins respectively (designated as rLa-NiVG and rLa-NiVF), and evaluated their immunogenicity in mice and pigs. Both rLa-NiVG and rLa-NiVF displayed growth properties similar to those of LaSota virus in chicken eggs. Co-infection of rLa-NiVG and rLa-NiVF caused marked syncytia formation, while intracerebral co-inoculation of these viruses in mice showed they were safe in at least one mammalian species. Animal immunization studies showed rLa-NiVG and rLa-NiVF induced NiV neutralizing antibody responses in mice and pigs, and F protein-specific CD8+ T cell responses in mice. Most importantly, rLa-NiVG and rLa-NiVF administered alone or together, induced a long-lasting neutralizing antibody response in pigs. Recombinant rLa-NiVG/F thus appear to be promising NiV vaccine candidates for pigs and potentially humans.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Encefalitis Viral/inmunología , Vectores Genéticos/genética , Virus de la Enfermedad de Newcastle/genética , Virus Nipah/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales/sangre , Linfocitos B/inmunología , Línea Celular , Encefalitis Viral/prevención & control , Encefalitis Viral/virología , Femenino , Infecciones por Henipavirus/inmunología , Infecciones por Henipavirus/prevención & control , Infecciones por Henipavirus/virología , Humanos , Ratones , Ratones Endogámicos BALB C , Virus Nipah/genética , Virus Nipah/metabolismo , Porcinos , Linfocitos T/inmunología , Factores de Tiempo , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología , Vacunas Virales/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...