Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 223
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 659: 54-61, 2023 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-37037066

RESUMEN

Previously, we demonstrated the utility of a recombinant chimeric flavivirus (DV2ChimV), which carries the premembrane (prM) and envelope (E) genes of a type 2 DENV clinical (Thai) isolate on a backbone of Japanese encephalitis virus, for evaluating the protective efficacy of antidengue envelope antibodies both in vitro and in vivo. Here, to assess the potential use of this model for pathological studies, we aimed to characterize interferon-α/ß-γ-receptor double-knockout mice (IFN-α/ß/γR dKO mice) infected with DV2ChimV. Vascular leakage and bone marrow suppression are unique features of severe dengue. In the current model, DV2ChimV caused vascular leakage in the liver and intestine at the moribund stage. High levels of virus were detected in the bone marrow, and strong bone marrow suppression (i.e., disappearance of megakaryocytes and erythroblastic islets) was observed. These observations suggest that the DV2ChimV-infected mouse model mimics the vascular leakage and bone marrow suppression observed in human cases.


Asunto(s)
Virus del Dengue , Dengue , Flavivirus , Ratones , Humanos , Animales , Médula Ósea/patología , Ratones Noqueados , Anticuerpos Antivirales
2.
Biochem Biophys Res Commun ; 495(1): 1221-1226, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29175328

RESUMEN

Producing virus at high yield is critically important for development of whole virion inactivated vaccines or live attenuated vaccines. Most dengue virus (DENV) clinical isolates, however, replicate at low levels in cultured cells, which limits their use for vaccine development. The present study examined differences between low-replicating DENV clinical isolates and high-replicating laboratory strains with the aim of engineering high-yield DENV clinical isolates. Construction of a series of recombinant chimeric viruses derived from a high-replicating laboratory DENV type 4 (DENV-4) H241 strain and a clinical isolate revealed that the NS3-NS4B region of H241 conferred a replication advantage in cultured cells. Furthermore, northern blot analysis revealed that this advantage was due to more efficient synthesis of viral RNA. Importantly, replacement of the NS3-NS4B region of H241 did not increase virulence in mice, suggesting that viral production can be increased safely. This study provided information that will facilitate engineering of safe and high-yield viruses that can be used for vaccine development.


Asunto(s)
Técnicas de Cultivo Celular por Lotes/métodos , Virus del Dengue/crecimiento & desarrollo , Virus del Dengue/genética , Mejoramiento Genético/métodos , Carga Viral/genética , Proteínas no Estructurales Virales/metabolismo , Virulencia/fisiología , Recombinación Genética/genética , Carga Viral/fisiología , Proteínas no Estructurales Virales/genética
3.
PLoS Pathog ; 12(4): e1005583, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27097026

RESUMEN

A major determinant in the change of the avian influenza virus host range to humans is the E627K substitution in the PB2 polymerase protein. However, the polymerase activity of avian influenza viruses with a single PB2-E627K mutation is still lower than that of seasonal human influenza viruses, implying that avian viruses require polymerase mutations in addition to PB2-627K for human adaptation. Here, we used a database search of H5N1 clade 2.2.1 virus sequences with the PB2-627K mutation to identify other polymerase adaptation mutations that have been selected in infected patients. Several of the mutations identified acted cooperatively with PB2-627K to increase viral growth in human airway epithelial cells and mouse lungs. These mutations were in multiple domains of the polymerase complex other than the PB2-627 domain, highlighting a complicated avian-to-human adaptation pathway of avian influenza viruses. Thus, H5N1 viruses could rapidly acquire multiple polymerase mutations that function cooperatively with PB2-627K in infected patients for optimal human adaptation.


Asunto(s)
Adaptación Fisiológica/genética , ARN Polimerasas Dirigidas por ADN/genética , Gripe Humana/genética , Proteínas Virales/genética , Animales , Western Blotting , Modelos Animales de Enfermedad , Femenino , Humanos , Subtipo H5N1 del Virus de la Influenza A , Ratones , Ratones Endogámicos BALB C , Mutagénesis Sitio-Dirigida , Mutación , Reacción en Cadena en Tiempo Real de la Polimerasa , Transfección
4.
Microbiol Immunol ; 61(9): 371-379, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28752940

RESUMEN

Bordetella pertussis causes whooping cough, a severe and prolonged respiratory disease that results inhas high morbidity and mortality rates, particularly in developing countries. The number incidence of whooping cough cases is increasing in many countries despite high vaccine coverage. Causes for the re-emergence of the disease include the limited duration of protection conferred by the acellular pertussis vaccines (aP)s and pathogenic adaptations that involve antigenic divergence from vaccine strains. Therefore, current vaccines therefore need to be improved. In the present study, we focused on five autotransporters: namely SphB1, BatB, SphB2, Phg, and Vag8, which were previously found to be expressed by B. bronchiseptica during the course of infection in rats and examined their protective efficiencies as vaccine antigens. The passenger domains of these proteins were produced in recombinant forms and used as antigens. An intranasal murine challenge assay showed that immunization with a mixture of SphB1 and Vag8 (SV) significantly reduced bacterial load in the lower respiratory tract and a combination of aP and SV acts synergistically in effects of conferring protection against B. pertussis infection, implying that these antigens have potential as components to for improvinge th the currently available acellular pertussis vaccine.


Asunto(s)
Antígenos Bacterianos/inmunología , Bordetella pertussis/inmunología , Vacuna contra la Tos Ferina/inmunología , Sistemas de Secreción Tipo V/inmunología , Tos Ferina/prevención & control , Animales , Anticuerpos Antibacterianos/inmunología , Variación Antigénica/inmunología , Carga Bacteriana/inmunología , Proteínas Bacterianas/inmunología , Femenino , Ratones , Ratones Endogámicos BALB C , Sistema Respiratorio/inmunología , Sistema Respiratorio/microbiología , Serina Endopeptidasas/inmunología , Vacunación , Tos Ferina/inmunología , Tos Ferina/microbiología
5.
J Biol Chem ; 290(17): 10627-42, 2015 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-25673693

RESUMEN

The highly pathogenic avian influenza (AI) virus, H5N1, is a serious threat to public health worldwide. Both the currently circulating H5N1 and previously circulating AI viruses recognize avian-type receptors; however, only the H5N1 is highly infectious and virulent in humans. The mechanism(s) underlying this difference in infectivity remains unclear. The aim of this study was to clarify the mechanisms responsible for the difference in infectivity between the current and previously circulating strains. Primary human small airway epithelial cells (SAECs) were transformed with the SV40 large T-antigen to establish a series of clones (SAEC-Ts). These clones were then used to test the infectivity of AI strains. Human SAEC-Ts could be broadly categorized into two different types based on their susceptibility (high or low) to the viruses. SAEC-T clones were poorly susceptible to previously circulating AI but were completely susceptible to the currently circulating H5N1. The hemagglutinin (HA) of the current H5N1 virus showed greater membrane fusion activity at higher pH levels than that of previous AI viruses, resulting in broader cell tropism. Moreover, the endosomal pH was lower in high susceptibility SAEC-T clones than that in low susceptibility SAEC-T clones. Taken together, the results of this study suggest that the infectivity of AI viruses, including H5N1, depends upon a delicate balance between the acid sensitivity of the viral HA and the pH within the endosomes of the target cell. Thus, one of the mechanisms underlying H5N1 pathogenesis in humans relies on its ability to fuse efficiently with the endosomes in human airway epithelial cells.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Subtipo H5N1 del Virus de la Influenza A/fisiología , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Mucosa Respiratoria/virología , Animales , Aves , Línea Celular , Células Clonales , Perros , Endosomas/metabolismo , Endosomas/virología , Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Humanos , Concentración de Iones de Hidrógeno , Gripe Aviar/metabolismo , Gripe Aviar/transmisión , Gripe Aviar/virología , Gripe Humana/metabolismo , Gripe Humana/transmisión , Gripe Humana/virología , Estabilidad Proteica , Receptores de Superficie Celular/metabolismo , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Virulencia/fisiología , Internalización del Virus
6.
Virus Genes ; 52(6): 867-871, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27369428

RESUMEN

Avian influenza viruses impose serious public health burdens with significant mortality and morbidity not only in poultry but also in humans. While poultry susceptibility to avian influenza virus infection is well characterized, pigeons have been thought to have low susceptibility to these viruses. However, recent studies reported natural pigeon infections with highly pathogenic avian influenza H5N1 viruses. In Egypt, which is one of the H5N1 endemic areas for birds, pigeons are raised in towers built on farms in backyards and on house roofs, providing a potential risk for virus transmission from pigeons to humans. In this study, we performed genetic analysis of two H5N1 virus strains that were isolated from naturally infected pigeons in Egypt. Genetic and phylogenetic analyses showed that these viruses originated from Egyptian H5N1 viruses that were circulating in chickens or ducks. Several unique mutations, not reported before in any Egyptian isolates, were detected in the internal genes (i.e., polymerase residues PB1-V3D, PB1-K363R, PA-A369V, and PA-V602I; nucleoprotein residue NP-R38K; and nonstructural protein residues NS1-D120N and NS2-F55C). Our findings suggested that pigeons are naturally infected with H5N1 virus and can be a potential reservoir for transmission to humans, and showed the importance of genetic analysis of H5N1 internal genes.


Asunto(s)
Columbidae/virología , Subtipo H5N1 del Virus de la Influenza A/genética , Gripe Aviar/epidemiología , Gripe Aviar/virología , Animales , Egipto/epidemiología , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Mutación , Filogenia , ARN Viral , Análisis de Secuencia de ADN , Factores de Virulencia
7.
Nature ; 463(7277): 84-7, 2010 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-20054395

RESUMEN

Retroviruses are the only group of viruses known to have left a fossil record, in the form of endogenous proviruses, and approximately 8% of the human genome is made up of these elements. Although many other viruses, including non-retroviral RNA viruses, are known to generate DNA forms of their own genomes during replication, none has been found as DNA in the germline of animals. Bornaviruses, a genus of non-segmented, negative-sense RNA virus, are unique among RNA viruses in that they establish persistent infection in the cell nucleus. Here we show that elements homologous to the nucleoprotein (N) gene of bornavirus exist in the genomes of several mammalian species, including humans, non-human primates, rodents and elephants. These sequences have been designated endogenous Borna-like N (EBLN) elements. Some of the primate EBLNs contain an intact open reading frame (ORF) and are expressed as mRNA. Phylogenetic analyses showed that EBLNs seem to have been generated by different insertional events in each specific animal family. Furthermore, the EBLN of a ground squirrel was formed by a recent integration event, whereas those in primates must have been formed more than 40 million years ago. We also show that the N mRNA of a current mammalian bornavirus, Borna disease virus (BDV), can form EBLN-like elements in the genomes of persistently infected cultured cells. Our results provide the first evidence for endogenization of non-retroviral virus-derived elements in mammalian genomes and give novel insights not only into generation of endogenous elements, but also into a role of bornavirus as a source of genetic novelty in its host.


Asunto(s)
Bornaviridae/genética , Genes Virales/genética , Genoma/genética , Mamíferos/genética , Mamíferos/virología , Integración Viral/genética , Secuencia de Aminoácidos , Animales , Virus de la Enfermedad de Borna/genética , Virus de la Enfermedad de Borna/fisiología , Bornaviridae/fisiología , Línea Celular , Secuencia Conservada/genética , Evolución Molecular , Interacciones Huésped-Patógeno/genética , Humanos , Modelos Genéticos , Datos de Secuencia Molecular , Sistemas de Lectura Abierta/genética , Filogenia , Transcripción Reversa , Factores de Tiempo
8.
Biologicals ; 44(5): 403-11, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27461242

RESUMEN

Hepatitis E virus (HEV) causes viral hepatitis, and is considered a risk factor for blood products. Although some HEV inactivation/removal studies have been reported, detailed investigations of different manufacturing steps as heat treatment, partitioning during cold ethanol fractionation, low pH treatment, and virus filtration have yet to be reported for plasma-derived medicinal products. In this study, human serum- and swine faeces-derived HEVs, with and without detergent treatment, were used. The kinetic patterns of inactivation, log reduction value, or partitioning during the process were evaluated. In addition, the mouse encephalomyocarditis virus (EMCV) and canine and porcine parvoviruses (CPV/PPV) were also evaluated as model viruses for HEV. Small pore size (19 or 15 nm) virus filtration demonstrated effective removal of HEV. Middle pore size (35 nm) virus filtration and 60 °C liquid heating demonstrated moderate inactivation/removal. Ethanol fractionation steps demonstrated limited removal of HEV. Unpurified HEV exhibited different properties than the detergent-treated HEV, and both forms displayed differences when compared with EMCV, CPV, and PPV. Limited or no inactivation of HEV was observed during low pH treatment. Untreated plasma-derived HEV from humans showed different properties compared to that of HEV treated with detergent or derived from swine faeces. Therefore, HEV spike preparation requires more attention.


Asunto(s)
Desinfección/métodos , Virus de la Hepatitis E/química , Virus de la Hepatitis E/aislamiento & purificación , Plasma/virología , Inactivación de Virus , Animales , Perros , Femenino , Hepatitis E , Humanos , Concentración de Iones de Hidrógeno , Masculino , Ratones , Porcinos
9.
Biologicals ; 44(5): 394-402, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27464991

RESUMEN

In previous studies, hybridomas producing human immunoglobulin G, the antibodies 5E4 and 5A7 against influenza A and B virus were established using a novel human lymphocyte fusion partner, SPYMEG. In the present study, we succeeded in achieving the recombinant production and secretion of 5E4 and 5A7 in Chinese hamster ovary cells. Our N-glycan analysis by intact-mass detection and liquid chromatography mass spectrometry showed that recombinant 5E4 and 5A7 have one N-glycan and the typical mammalian-type N-glycan structures similar to those in hybridomas. However, the glycan distribution was slightly different among these antibodies. The amount of high-mannose-type structures was under 10% of the total N-glycans of recombinant 5E4 and 5A7, compared to 20% of the 5E4 and 5A7 produced in hybridomas. The amount of galactosylated N-glycans was increased in recombinants. Approximately 80% of the N-glycans of all antibodies was fucosylated, and no sialylated N-glycan was found. Recombinant 5E4 and 5A7 neutralized pandemic influenza A virus specifically, and influenza B virus broadly, quite similar to the 5E4 and 5A7 produced in hybridomas, respectively. Here we demonstrated that recombinants of antibodies identified from hybridomas fused with SPYMEG have normal N-glycans and that their neutralizing activities bear comparison with those of the original antibodies.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Inmunoglobulina G/inmunología , Virus de la Influenza A/inmunología , Virus de la Influenza B/inmunología , Animales , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Neutralizantes/biosíntesis , Anticuerpos Antivirales/sangre , Fusión Celular/métodos , Cricetinae , Humanos , Hibridomas/inmunología , Hibridomas/metabolismo , Inmunoglobulina G/biosíntesis , Linfocitos/inmunología , Linfocitos/metabolismo
10.
J Clin Microbiol ; 53(2): 382-8, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25411170

RESUMEN

Chikungunya fever is a mosquito-borne disease of key public health importance in tropical and subtropical countries. Although severe joint pain is the most distinguishing feature of chikungunya fever, diagnosis remains difficult because the symptoms of chikungunya fever are shared by many pathogens, including dengue fever. The present study aimed to develop a new immunochromatographic diagnosis test for the detection of chikungunya virus antigen in serum. Mice were immunized with isolates from patients with Thai chikungunya fever, East/Central/South African genotype, to produce mouse monoclonal antibodies against chikungunya virus. Using these monoclonal antibodies, a new diagnostic test was developed and evaluated for the detection of chikungunya virus. The newly developed diagnostic test reacted with not only the East/Central/South African genotype but also with the Asian and West African genotypes of chikungunya virus. Testing of sera from patients suspected to have chikungunya fever in Thailand (n = 50), Laos (n = 54), Indonesia (n = 2), and Senegal (n = 6) revealed sensitivity, specificity, and real-time PCR (RT-PCR) agreement values of 89.4%, 94.4%, and 91.1%, respectively. In our study using serial samples, a new diagnostic test showed high agreement with the RT-PCR within the first 5 days after onset. A rapid diagnostic test was developed using mouse monoclonal antibodies that react with chikungunya virus envelope proteins. The diagnostic accuracy of our test is clinically acceptable for chikungunya fever in the acute phase.


Asunto(s)
Antígenos Virales/sangre , Fiebre Chikungunya/diagnóstico , Virus Chikungunya/aislamiento & purificación , Cromatografía de Afinidad/métodos , Pruebas Diagnósticas de Rutina/métodos , Suero/virología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/inmunología , Fiebre Chikungunya/virología , Humanos , Indonesia , Ratones Endogámicos BALB C , Senegal , Sensibilidad y Especificidad , Tailandia , Factores de Tiempo
11.
PLoS Pathog ; 9(2): e1003150, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23408886

RESUMEN

Influenza virus has the ability to evade host immune surveillance through rapid viral genetic drift and reassortment; therefore, it remains a continuous public health threat. The development of vaccines producing broadly reactive antibodies, as well as therapeutic strategies using human neutralizing monoclonal antibodies (HuMAbs) with global reactivity, has been gathering great interest recently. Here, three hybridoma clones producing HuMAbs against influenza B virus, designated 5A7, 3A2 and 10C4, were prepared using peripheral lymphocytes from vaccinated volunteers, and were investigated for broad cross-reactive neutralizing activity. Of these HuMAbs, 3A2 and 10C4, which recognize the readily mutable 190-helix region near the receptor binding site in the hemagglutinin (HA) protein, react only with the Yamagata lineage of influenza B virus. By contrast, HuMAb 5A7 broadly neutralizes influenza B strains that were isolated from 1985 to 2006, belonging to both Yamagata and Victoria lineages. Epitope mapping revealed that 5A7 recognizes 316G, 318C and 321W near the C terminal of HA1, a highly conserved region in influenza B virus. Indeed, no mutations in the amino acid residues of the epitope region were induced, even after the virus was passaged ten times in the presence of HuMAb 5A7. Moreover, 5A7 showed significant therapeutic efficacy in mice, even when it was administered 72 hours post-infection. These results indicate that 5A7 is a promising candidate for developing therapeutics, and provide insight for the development of a universal vaccine against influenza B virus.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Neutralizantes/uso terapéutico , Anticuerpos Antivirales/uso terapéutico , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Virus de la Influenza B/inmunología , Gripe Humana/prevención & control , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Secuencia de Bases , Mapeo Epitopo , Femenino , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Humanos , Hibridomas , Virus de la Influenza B/genética , Gripe Humana/tratamiento farmacológico , Gripe Humana/inmunología , Inyecciones Intraperitoneales , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Pruebas de Neutralización , Alineación de Secuencia , Análisis de Secuencia de ADN , Resultado del Tratamiento
12.
Bioorg Med Chem Lett ; 25(9): 1876-9, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25857943

RESUMEN

Antibody-integrated magnetic beads have been functionalized for influenza A virus capture. First, ammonia plasma produced by a radio frequency power source was reacted with the surface of graphite-encapsulated magnetic beads to introduce amino groups. Anti-influenza A virus hemagglutinin antibody was then anchored by its surface sulfide groups to the amino groups on the beads via N-succinimidyl 3-(2-pyridyldithio) propionate. After incubation with influenza A virus, adsorption of the virus to the beads was confirmed by immunochromatography, polymerase chain reaction (PCR), enzyme-linked immunosorbent assay (ELISA), and inoculation of chicken embryonated eggs, indicating that virus infectivity is maintained and that the proposed method is useful for the enhanced detection and isolation of influenza A virus.


Asunto(s)
Amoníaco/química , Anticuerpos Inmovilizados/química , Grafito/química , Separación Inmunomagnética , Virus de la Influenza A/aislamiento & purificación , Gases em Plasma/química , Propiedades de Superficie
13.
J Biol Chem ; 288(7): 4981-90, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23264630

RESUMEN

The development of vaccination methods that can overcome the emergence of new types of influenza strains caused by escape mutations is desirable to avoid future pandemics. Here, a novel type of immunogen was designed that targeted the conformation of a highly conserved region of influenza A virus hemagglutinin (HA) composed of two separate sequences that associate to form an anti-parallel ß-sheet structure. Our previous study identified this ß-sheet region as the structural core in the epitope of a characteristic antibody (B-1) that strongly neutralizes a wide variety of strains within the H3N2 serotype, and therefore this ß-sheet region was considered a good target to induce broadly reactive immunity against the influenza A virus. To design the immunogen, residues derived from the B-1 epitope were introduced directly onto a part of enhanced green fluorescent protein (EGFP), whose surface is mostly composed of ß-sheets. Through site-directed mutagenesis, several modified EGFPs with an epitope-mimicking structure embedded in their surface were prepared. Two EGFP variants, differing from wild-type (parental) EGFP by only five and nine residues, induced mice to produce antibodies that specifically bind to H3-type HA and neutralize H3N2 virus. Moreover, three of five mice immunized with each of these EGFP variants followed by a booster with equivalent mCherry variants acquired anti-viral immunity against challenge with H3N2 virus at a lethal dosage. In contrast to conventional methods, such as split HA vaccine, preparation of this type of immunogen requires less time and is therefore expected to be quickly responsive to newly emerged influenza viral strains.


Asunto(s)
Epítopos/química , Proteínas Fluorescentes Verdes/metabolismo , Hemaglutininas/química , Animales , Dicroismo Circular , Escherichia coli/metabolismo , Femenino , Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Hibridomas/metabolismo , Inmunización , Vacunas contra la Influenza/inmunología , Ratones , Ratones Endogámicos BALB C , Mutagénesis , Pruebas de Neutralización , Orthomyxoviridae/inmunología , Ingeniería de Proteínas/métodos , Estructura Secundaria de Proteína , Proteínas Virales/química
14.
Retrovirology ; 11: 32, 2014 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-24758333

RESUMEN

BACKGROUND: The CD4 binding site (CD4bs) of envelope glycoprotein (Env) gp120 is a functionally conserved, important target of anti-human immunodeficiency virus type 1 (HIV-1) neutralizing antibodies. Two neutralizing human monoclonal antibodies, IgG1 b12 (b12) and VRC01, are broadly reactive neutralizing antibodies which recognize conformational epitopes that overlap the CD4bs of Env gp120; however, many CRF01_AE viruses are resistant to neutralization mediated by these antibodies. We examined the mechanism underlying the b12 resistance of the viruses using CRF01_AE Env (AE-Env)-recombinant viruses in this study. RESULTS: Our results showed that an amino acid substitution at position 185 in the V2 region of gp120 played a crucial role in regulating the b12 susceptibility of AE-Env-recombinant viruses by cooperating with 2 previously reported potential N-linked glycosylation (PNLG) sites at positions 186 (N186) and 197 (N197) in the V2 and C2 regions of Env gp120. The amino acid residue at position 185 and 2 PNLG sites were responsible for the b12 resistance of 21 of 23 (>91%) AE-Env clones tested. Namely, the introduction of aspartic acid at position 185 (D185) conferred b12 susceptibility of 12 resistant AE-Env clones in the absence of N186 and/or N197, while the introduction of glycine at position 185 (G185) reduced the b12 susceptibility of 9 susceptible AE-Env clones in the absence of N186 and/or N197. In addition, these amino acid mutations altered the VRC01 susceptibility of many AE-Env clones. CONCLUSIONS: We propose that the V2 and C2 regions of AE-Env gp120 contain the major determinants of viral resistance to CD4bs antibodies. CRF01_AE is a major circulating recombinant form of HIV-1 prevalent in Southeast Asia. Our data may provide important information to understand the molecular mechanism regulating the neutralization susceptibility of CRF01_AE viruses to CD4bs antibodies.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Linfocitos T CD4-Positivos/inmunología , Anticuerpos Anti-VIH/inmunología , Proteína gp120 de Envoltorio del VIH/inmunología , VIH-1/inmunología , Mutación Missense , Sustitución de Aminoácidos , Asia Sudoriental , Sitios de Unión , Genotipo , Proteína gp120 de Envoltorio del VIH/genética , Proteína gp120 de Envoltorio del VIH/fisiología , Infecciones por VIH/virología , VIH-1/clasificación , VIH-1/genética , VIH-1/aislamiento & purificación , Humanos , Proteínas Mutantes/genética , Proteínas Mutantes/inmunología , Acoplamiento Viral
15.
Biochem Biophys Res Commun ; 450(1): 42-8, 2014 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-24858683

RESUMEN

Influenza viruses are a continuous threat to human public health because of their ability to evolve rapidly through genetic drift and reassortment. Three human monoclonal antibodies (HuMAbs) were generated in this study, 1H11, 2H5 and 5G2, and they cross-neutralize a diverse range of group 1 influenza A viruses, including seasonal H1N1, 2009 pandemic H1N1 (H1N1pdm) and avian H5N1 and H9N2. The three HuMAbs were prepared by fusing peripheral blood lymphocytes from an H1N1pdm-infected patient with a newly developed fusion partner cell line, SPYMEG. All the HuMAbs had little hemagglutination inhibition activity but had strong membrane-fusion inhibition activity against influenza viruses. A protease digestion assay showed the HuMAbs targeted commonly a short α-helix region in the stalk of the hemagglutinin. Furthermore, Ile45Phe and Glu47Gly double substitutions in the α-helix region made the HA unrecognizable by the HuMAbs. These two amino acid residues are highly conserved in the HAs of H1N1, H5N1 and H9N2 viruses. The HuMAbs reported here may be potential candidates for the development of therapeutic antibodies against group 1 influenza viruses.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Hemaglutininas/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Gripe Humana/virología , Animales , Anticuerpos Antivirales/inmunología , Línea Celular , Haplorrinos , Humanos , Riñón/inmunología , Riñón/virología , Pruebas de Neutralización
16.
Biochem Biophys Res Commun ; 452(3): 865-70, 2014 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-25204499

RESUMEN

Most neutralizing antibodies elicited during influenza virus infection or by vaccination have a narrow spectrum because they usually target variable epitopes in the globular head region of hemagglutinin (HA). In this study, we describe a human monoclonal antibody (HuMAb), 5D7, that was prepared from the peripheral blood lymphocytes of a vaccinated volunteer using the fusion method. The HuMAb heterosubtypically neutralizes group 1 influenza A viruses, including seasonal H1N1, 2009 pandemic H1N1 (H1N1pdm) and avian H9N2, with a strong hemagglutinin inhibition activity. Selection of an escape mutant showed that the HuMAb targets a novel conformational epitope that is located in the HA head region but is distinct from the receptor binding site. Furthermore, Phe114Ile substitution in the epitope made the HA unrecognizable by the HuMAb. Amino acid residues in the predicted epitope region are also highly conserved in the HAs of H1N1 and H9N2. The HuMAb reported here may be a potential candidate for the development of therapeutic/prophylactic antibodies against H1 and H9 influenza viruses.


Asunto(s)
Anticuerpos Monoclonales/biosíntesis , Anticuerpos Neutralizantes/biosíntesis , Anticuerpos Antivirales/biosíntesis , Protección Cruzada , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H9N2 del Virus de la Influenza A/inmunología , Gripe Humana/prevención & control , Secuencia de Aminoácidos , Animales , Antígenos Virales/química , Antígenos Virales/inmunología , Mapeo Epitopo , Epítopos/química , Epítopos/inmunología , Pruebas de Inhibición de Hemaglutinación , Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Humanos , Hibridomas/inmunología , Vacunas contra la Influenza/administración & dosificación , Gripe Humana/inmunología , Gripe Humana/virología , Masculino , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Vacunación , Adulto Joven
17.
Virus Genes ; 49(3): 485-9, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25113745

RESUMEN

Chikungunya fever (CHIKF) is an acute febrile illness caused by a mosquito-borne alphavirus, chikungunya virus (CHIKV). This disease re-emerged in Kenya in 2004, and spread to the countries in and around the Indian Ocean. The re-emerging epidemics rapidly spread to regions like India and Southeast Asia, and it was subsequently identified in Europe in 2007, probably as a result of importation of chikungunya cases. On the one hand, chikungunya is one of the neglected diseases and has only attracted strong attention during large outbreaks. In 2008-2009, there was a major outbreak of chikungunya fever in Thailand, resulting in the highest number of infections in any country in the region. However, no update of CHIKV circulating in Thailand has been published since 2009. In this study, we examined the viral growth kinetics and sequences of the structural genes derived from CHIKV clinical isolates obtained from the serum specimens of CHIKF-suspected patients in Central Thailand in 2010. We identified the CHIKV harboring two mutations E1-A226V and E2-I211T, indicating that the East, Central, and South African lineage of CHIKV was continuously circulating as an indigenous population in Thailand.


Asunto(s)
Fiebre Chikungunya/epidemiología , Fiebre Chikungunya/virología , Virus Chikungunya/aislamiento & purificación , Virus Chikungunya/clasificación , Virus Chikungunya/genética , Análisis por Conglomerados , Variación Genética , Humanos , Modelos Moleculares , Epidemiología Molecular , Datos de Secuencia Molecular , Filogenia , Conformación Proteica , ARN Viral/genética , Análisis de Secuencia de ADN , Homología de Secuencia , Suero/virología , Tailandia/epidemiología , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/genética
18.
Appl Microbiol Biotechnol ; 98(5): 2113-20, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23880875

RESUMEN

The abnormal prion protein (scrapie-associated prion protein, PrP(Sc)) is considered to be included in the group of infectious agents of transmissible spongiform encephalopathies. Since PrP(Sc) is highly resistant to normal sterilization procedures, the decontamination of PrP(Sc) is a significant public health issue. In the present study, a hyperthermostable protease, Tk-subtilisin, was used to degrade PrP(Sc). Although PrP(Sc) is known to be resistant toward proteolytic enzymes, Tk-subtilisin was able to degrade PrP(Sc) under extreme conditions. The level of PrP(Sc) in brain homogenates was found to decrease significantly in vitro following Tk-subtilisin treatment at 100 °C, whereas some protease-resistant fractions remain after proteinase K treatment. Rather small amounts of Tk-subtilisin (0.3 U) were required to degrade PrP(Sc) at 100 °C and pH 8.0. In addition, Tk-subtilisin was observed to degrade PrP(Sc) in the presence of sodium dodecyl sulfate or other industrial surfactants. Although several proteases degrading PrP(Sc) have been reported, practical decontamination procedures using enzymes are not available. This report aims to provide basic information for the practical use of a proteolytic enzyme for PrP(Sc) degradation.


Asunto(s)
Proteínas PrPSc/metabolismo , Subtilisina/aislamiento & purificación , Subtilisina/metabolismo , Thermococcus/enzimología , Detergentes/metabolismo , Inhibidores Enzimáticos/metabolismo , Estabilidad de Enzimas , Calor , Concentración de Iones de Hidrógeno , Proteolisis , Dodecil Sulfato de Sodio/metabolismo , Subtilisina/química
19.
Biochem Biophys Res Commun ; 440(3): 393-8, 2013 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-24070610

RESUMEN

The non-structural protein NS2B/NS3 serine-protease complex of the dengue virus (DENV) is required for the maturation of the viral polyprotein. Dissociation of the NS2B cofactor from NS3 diminishes the enzymatic activity of the complex. In this study, we identified a small molecule inhibitor that interferes with the interaction between NS2B and NS3 using structure-based screening and a cell-based viral replication assay. A library containing 661,417 small compounds derived from the Molecular Operating Environment lead-like database was docked to the NS2B/NS3 structural model. Thirty-nine compounds with high scores were tested in a secondary screening using a cell-based viral replication assay. SK-12 was found to inhibit replication of all DENV serotypes (EC50=0.74-4.92 µM). In silico studies predicted that SK-12 pre-occupies the NS2B-binding site of NS3. Steady-state kinetics using a fluorogenic short peptide substrate demonstrated that SK-12 is a noncompetitive inhibitor against the NS2B/NS3 protease. Inhibition to Japanese encephalitis virus by SK-12 was relatively weak (EC50=29.81 µM), and this lower sensitivity was due to difference in amino acid at position 27 of NS3. SK-12 is the promising small-molecule inhibitor that targets the interaction between NS2B and NS3.


Asunto(s)
Antivirales/farmacología , Dengue/tratamiento farmacológico , Naftoles/farmacología , Serina Proteasas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Sulfonamidas/farmacología , Proteínas no Estructurales Virales/química , Replicación Viral/efectos de los fármacos , Simulación por Computador , Dengue/enzimología , Humanos , Modelos Químicos , Conformación Proteica
20.
BMC Biotechnol ; 13: 19, 2013 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-23448268

RESUMEN

BACKGROUND: Tk-SP is a member of subtilisin-like serine proteases from a hyperthermophilic archaeon Thermococcus kodakarensis. It has been known that the hyper-stable protease, Tk-SP, could exhibit enzymatic activity even at high temperature and in the presence of chemical denaturants. In this work, the enzymatic activity of Tk-SP was measured in the presence of detergents and EDTA. In addition, we focused to demonstrate that Tk-SP could degrade the abnormal prion protein (PrPSc), a protease-resistant isoform of normal prion protein (PrPC). RESULTS: Tk-SP was observed to maintain its proteolytic activity with nonionic surfactants and EDTA at 80°C. We optimized the condition in which Tk-SP functions efficiently, and demonstrated that the enzyme is highly stable in the presence of 0.05% (w/v) nonionic surfactants and 0.01% (w/v) EDTA, retaining up to 80% of its activity. Additionally, we also found that Tk-SP can degrade PrPSc to a level undetectable by western-blot analysis. CONCLUSIONS: Our results indicate that Tk-SP has a great potential for technological applications, such as thermo-stable detergent additives. In addition, it is also suggested that Tk-SP-containing detergents can be developed to decrease the secondary infection risks of transmissible spongiform encephalopathies (TSE).


Asunto(s)
Proteínas Arqueales/metabolismo , Detergentes/química , Priones/metabolismo , Subtilisina/metabolismo , Thermococcus/enzimología , Proteínas Arqueales/química , Proteínas Arqueales/genética , Ácido Edético/química , Escherichia coli/metabolismo , Calor , Concentración de Iones de Hidrógeno , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Subtilisina/química , Subtilisina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA