Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
J Virol ; 93(6)2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30626682

RESUMEN

Protection from influenza virus infection is canonically associated with antibodies that neutralize the virus by blocking the interaction between the viral hemagglutinin and host cell receptors. However, protection can also be conferred by other mechanisms, including antibody-mediated effector functions. Here, we report the characterization of 22 broadly cross-reactive, nonneutralizing antibodies specific for influenza B virus hemagglutinin. The majority of these antibodies recognized influenza B viruses isolated over the period of 73 years and bind the conserved stalk domain of the hemagglutinin. A proportion of the characterized antibodies protected mice from both morbidity and mortality after challenge with a lethal dose of influenza B virus. Activity in an antibody-dependent cell-mediated cytotoxicity reporter assay correlated strongly with protection, suggesting that Fc-dependent effector function determines protective efficacy. The information regarding mechanism of action and epitope location stemming from our characterization of these antibodies will inform the design of urgently needed vaccines that could induce broad protection against influenza B viruses.IMPORTANCE While broadly protective antibodies against the influenza A virus hemagglutinin have been well studied, very limited information is available for antibodies that broadly recognize influenza B viruses. Similarly, the development of a universal or broadly protective influenza B virus vaccine lags behind the development of such a vaccine for influenza A virus. More information about epitope location and mechanism of action of broadly protective influenza B virus antibodies is required to inform vaccine development. In addition, protective antibodies could be a useful tool to treat or prevent influenza B virus infection in pediatric cohorts or in a therapeutic setting in immunocompromised individuals in conjugation with existing treatment avenues.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Reacciones Cruzadas/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Virus de la Influenza B/inmunología , Infecciones por Orthomyxoviridae/inmunología , Animales , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Línea Celular , Línea Celular Tumoral , Perros , Epítopos/inmunología , Femenino , Humanos , Vacunas contra la Influenza/inmunología , Células de Riñón Canino Madin Darby , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/virología , Células Sf9
2.
J Virol ; 92(7)2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29321309

RESUMEN

Recently, two new influenza A-like viruses have been discovered in bats, A/little yellow-shouldered bat/Guatemala/060/2010 (HL17NL10) and A/flat-faced bat/Peru/033/2010 (HL18NL11). The hemagglutinin (HA)-like (HL) and neuraminidase (NA)-like (NL) proteins of these viruses lack hemagglutination and neuraminidase activities, despite their sequence and structural homologies with the HA and NA proteins of conventional influenza A viruses. We have now investigated whether the NS1 proteins of the HL17NL10 and HL18NL11 viruses can functionally replace the NS1 protein of a conventional influenza A virus. For this purpose, we generated recombinant influenza A/Puerto Rico/8/1934 (PR8) H1N1 viruses containing the NS1 protein of the PR8 wild-type, HL17NL10, and HL18NL11 viruses. These viruses (r/NS1PR8, r/NS1HL17, and r/NS1HL18, respectively) were tested for replication in bat and nonbat mammalian cells and in mice. Our results demonstrate that the r/NS1HL17 and r/NS1HL18 viruses are attenuated in vitro and in vivo However, the bat NS1 recombinant viruses showed a phenotype similar to that of the r/NS1PR8 virus in STAT1-/- human A549 cells and mice, both in vitro and in vivo systems being unable to respond to interferon (IFN). Interestingly, multiple mouse passages of the r/NS1HL17 and r/NS1HL18 viruses resulted in selection of mutant viruses containing single amino acid mutations in the viral PB2 protein. In contrast to the parental viruses, virulence and IFN antagonism were restored in the selected PB2 mutants. Our results indicate that the NS1 protein of bat influenza A-like viruses is less efficient than the NS1 protein of its conventional influenza A virus NS1 counterpart in antagonizing the IFN response and that this deficiency can be overcome by the influenza virus PB2 protein.IMPORTANCE Significant gaps in our understanding of the basic features of the recently discovered bat influenza A-like viruses HL17NL10 and HL18NL11 remain. The basic biology of these unique viruses displays both similarities to and differences from the basic biology of conventional influenza A viruses. Here, we show that recombinant influenza A viruses containing the NS1 protein from HL17NL10 and HL18NL11 are attenuated. This attenuation was mediated by their inability to antagonize the type I IFN response. However, this deficiency could be compensated for by single amino acid replacements in the PB2 gene. Our results unravel a functional divergence between the NS1 proteins of bat influenza A-like and conventional influenza A viruses and demonstrate an interplay between the viral PB2 and NS1 proteins to antagonize IFN.


Asunto(s)
Virus de la Influenza A , Interferones , Mutación , Proteínas no Estructurales Virales , Proteínas Virales , Células A549 , Células HEK293 , Humanos , Virus de la Influenza A/genética , Virus de la Influenza A/metabolismo , Interferones/genética , Interferones/metabolismo , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo
3.
J Virol ; 89(20): 10696-701, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26246567

RESUMEN

We demonstrate that novel bat HL17NL10 and HL18NL11 influenza virus NS1 proteins are effective interferon antagonists but do not block general host gene expression. Solving the RNA-binding domain structures revealed the canonical NS1 symmetrical homodimer, and RNA binding required conserved basic residues in this domain. Interferon antagonism was strictly dependent on RNA binding, and chimeric bat influenza viruses expressing NS1s defective in this activity were highly attenuated in interferon-competent cells but not in cells unable to establish antiviral immunity.


Asunto(s)
Quirópteros/virología , Virus de la Influenza A/genética , Infecciones por Orthomyxoviridae/veterinaria , ARN Bicatenario/química , Proteínas de Unión al ARN/química , Proteínas no Estructurales Virales/química , Animales , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Inmunidad Innata , Virus de la Influenza A/inmunología , Interferón beta/genética , Interferón beta/inmunología , Modelos Moleculares , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Unión Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , ARN Bicatenario/inmunología , ARN Bicatenario/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/inmunología , Transducción de Señal , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/inmunología , Replicación Viral
4.
Curr Top Microbiol Immunol ; 386: 73-107, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25007846

RESUMEN

The non-structural protein 1 of influenza virus (NS1) is a relatively small polypeptide with an outstanding number of ascribed functions. NS1 is the main viral antagonist of the innate immune response during influenza virus infection, chiefly by inhibiting the type I interferon system at multiple steps. As such, its role is critical to overcome the first barrier the host presents to halt the viral infection. However, the pro-viral activities of this well-studied protein go far beyond and include regulation of viral RNA and protein synthesis, and disruption of the host cell homeostasis by dramatically affecting general gene expression while tweaking the PI3K signaling network. Because of all of this, NS1 is a key virulence factor that impacts influenza pathogenesis, and adaptation to new hosts, making it an attractive target for control strategies. Here, we will overview the many roles that have been ascribed to the NS1 protein, and give insights into the sequence features and structural properties that make them possible, highlighting the need to understand how NS1 can actually perform all of these functions during viral infection.


Asunto(s)
Proteínas no Estructurales Virales/fisiología , Factores de Virulencia/fisiología , Animales , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/fisiología , Humanos , Inmunidad Innata , Gripe Humana/etiología , Gripe Humana/inmunología , Receptores Inmunológicos , Proteínas no Estructurales Virales/química
5.
J Virol ; 88(20): 12146-51, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25078692

RESUMEN

Although an effective interferon antagonist in human and avian cells, the novel H7N9 influenza virus NS1 protein is defective at inhibiting CPSF30. An I106M substitution in H7N9 NS1 can restore CPSF30 binding together with the ability to block host gene expression. Furthermore, a recombinant virus expressing H7N9 NS1-I106M replicates to higher titers in vivo, and is subtly more virulent, than the parental virus. Natural polymorphisms in H7N9 NS1 that enhance CPSF30 binding may be cause for concern.


Asunto(s)
Aminoácidos/genética , Subtipo H7N9 del Virus de la Influenza A/patogenicidad , Proteínas no Estructurales Virales/metabolismo , Proteínas Virales/metabolismo , Sustitución de Aminoácidos , Animales , Pollos , Expresión Génica , Humanos , Subtipo H7N9 del Virus de la Influenza A/metabolismo , Subtipo H7N9 del Virus de la Influenza A/fisiología , Proteínas no Estructurales Virales/genética , Virulencia
6.
J Virol ; 88(11): 6213-23, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24648457

RESUMEN

UNLABELLED: Human endogenous retroviruses (HERVs) are viruses that have colonized the germ line and spread through vertical passage. Only the more recently acquired HERVs, such as the HERV-K (HML-2) group, maintain coding open reading frames. Expression of HERV-Ks has been linked to different pathological conditions, including HIV infection, but our knowledge on which specific HERV-Ks are expressed in primary lymphocytes currently is very limited. To identify the most expressed HERV-Ks in an unbiased manner, we analyzed their expression patterns in peripheral blood lymphocytes using Pacific Biosciences (PacBio) single-molecule real-time (SMRT) sequencing. We observe that three HERV-Ks (KII, K102, and K18) constitute over 90% of the total HERV-K expression in primary human lymphocytes of five different donors. We also show experimentally that two of these HERV-K env sequences (K18 and K102) retain their ability to produce full-length and posttranslationally processed envelope proteins in cell culture. We show that HERV-K18 Env can be incorporated into HIV-1 but not simian immunodeficiency virus (SIV) particles. Moreover, HERV-K18 Env incorporation into HIV-1 virions is dependent on HIV-1 matrix. Taken together, we generated high-resolution HERV-K expression profiles specific for activated human lymphocytes. We found that one of the most abundantly expressed HERV-K envelopes not only makes a full-length protein but also specifically interacts with HIV-1. Our findings raise the possibility that these endogenous retroviral Env proteins could directly influence HIV-1 replication. IMPORTANCE: Here, we report the HERV-K expression profile of primary lymphocytes from 5 different healthy donors. We used a novel deep-sequencing technology (PacBio SMRT) that produces the long reads necessary to discriminate the complexity of HERV-K expression. We find that primary lymphocytes express up to 32 different HERV-K envelopes, and that at least two of the most expressed Env proteins retain their ability to make a protein. Importantly, one of them, the envelope glycoprotein of HERV-K18, is incorporated into HIV-1 in an HIV matrix-specific fashion. The ramifications of such interactions are discussed, as the possibility of HIV-1 target tissue broadening and immune evasion are considered.


Asunto(s)
Retrovirus Endógenos/metabolismo , Variación Genética , VIH-1/metabolismo , Linfocitos/virología , Proteínas del Envoltorio Viral/metabolismo , Secuencia de Bases , Western Blotting , Biología Computacional , Cartilla de ADN/genética , ADN Complementario/biosíntesis , Retrovirus Endógenos/genética , Técnica del Anticuerpo Fluorescente , Células HEK293 , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Datos de Secuencia Molecular , Plásmidos/genética , Análisis de Secuencia de ADN , Virus de la Inmunodeficiencia de los Simios/metabolismo , Transcriptoma , Virión/metabolismo
7.
PLoS Pathog ; 8(4): e1002668, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22577360

RESUMEN

Influenza viruses continue to pose a major public health threat worldwide and options for antiviral therapy are limited by the emergence of drug-resistant virus strains. The antiviral cytokine, interferon (IFN) is an essential mediator of the innate immune response and influenza viruses, like many viruses, have evolved strategies to evade this response, resulting in increased replication and enhanced pathogenicity. A cell-based assay that monitors IFN production was developed and applied in a high-throughput compound screen to identify molecules that restore the IFN response to influenza virus infected cells. We report the identification of compound ASN2, which induces IFN only in the presence of influenza virus infection. ASN2 preferentially inhibits the growth of influenza A viruses, including the 1918 H1N1, 1968 H3N2 and 2009 H1N1 pandemic strains and avian H5N1 virus. In vivo, ASN2 partially protects mice challenged with a lethal dose of influenza A virus. Surprisingly, we found that the antiviral activity of ASN2 is not dependent on IFN production and signaling. Rather, its IFN-inducing property appears to be an indirect effect resulting from ASN2-mediated inhibition of viral polymerase function, and subsequent loss of the expression of the viral IFN antagonist, NS1. Moreover, we identified a single amino acid mutation at position 499 of the influenza virus PB1 protein that confers resistance to ASN2, suggesting that PB1 is the direct target. This two-pronged antiviral mechanism, consisting of direct inhibition of virus replication and simultaneous activation of the host innate immune response, is a unique property not previously described for any single antiviral molecule.


Asunto(s)
Antivirales/farmacología , ARN Polimerasas Dirigidas por ADN/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Indoles/farmacología , Virus de la Influenza A/efectos de los fármacos , Interferones/biosíntesis , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Animales , Antivirales/química , Células Cultivadas , Perros , Inhibidores Enzimáticos/química , Haplorrinos , Humanos , Indoles/química , Virus de la Influenza A/fisiología , Ratones , Ratones Endogámicos BALB C , Replicación Viral/efectos de los fármacos
8.
J Virol ; 86(9): 5366-70, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22345452

RESUMEN

We generated influenza A viruses expressing mutant NS1 proteins unable to activate phosphoinositide 3-kinase (PI3K) in two mouse-lethal strains. The recombinant A/Puerto Rico/8/34 (rPR8) mutant virus strain was attenuated and caused reduced morbidity/mortality. For the recombinant A/WSN/33 (rWSN) virus strain, the inability to stimulate PI3K had minimal impact on replication or morbidity/mortality. Cell-based assays revealed subtly distinct intracellular sites of NS1 localization and PI3K activation between the strains. We hypothesize that specific spatially regulated NS1-activated PI3K signaling, rather than simply the total level of active PI3K, is important for virus replication and virulence.


Asunto(s)
Virus de la Influenza A/fisiología , Virus de la Influenza A/patogenicidad , Infecciones por Orthomyxoviridae/virología , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal , Proteínas no Estructurales Virales/metabolismo , Replicación Viral , Animales , Línea Celular , Perros , Humanos , Ratones , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/mortalidad , Análisis de Supervivencia , Virulencia
9.
J Virol ; 86(23): 13095-8, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22993153

RESUMEN

Conserved tryptophan-187 facilitates homodimerization of the influenza A virus NS1 protein effector domain. We generated a mutant influenza virus strain expressing NS1-W187R to destabilize this self-interaction. NS1-W187R protein exhibited lower double-stranded RNA (dsRNA)-binding activity, showed a temporal redistribution during infection, and was minimally compromised for interferon antagonism. The mutant virus replicated similarly to the wild type in vitro, but it was slightly attenuated for replication in mice, causing notably reduced morbidity and mortality. These data suggest biological relevance for the W187-mediated homotypic interaction of NS1.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Modelos Moleculares , Proteínas no Estructurales Virales/química , Replicación Viral/fisiología , Animales , Western Blotting , Dimerización , Perros , Técnica del Anticuerpo Fluorescente Indirecta , Subtipo H1N1 del Virus de la Influenza A/fisiología , Luciferasas , Células de Riñón Canino Madin Darby , Ratones , Ratones Endogámicos C57BL , Mutación Missense/genética , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo
10.
NPJ Vaccines ; 8(1): 12, 2023 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-36765053

RESUMEN

Influenza B virus (IBV) strains are one of the components of seasonal influenza vaccines in both trivalent and quadrivalent formulations. The vast majority of these vaccines are produced in embryonated chickens' eggs. While optimized backbones for vaccine production in eggs exist and are in use for influenza A viruses, no such backbones exist for IBVs, resulting in unpredictable production yields. To generate an optimal vaccine seed virus backbone, we have compiled a panel of 71 IBV strains from 1940 to present day, representing the known temporal and genetic variability of IBV circulating in humans. This panel contains strains from the B/Victoria/2/87-like lineage, B/Yamagata/16/88-like lineage and the ancestral lineage that preceded their split to provide a diverse set that would help to identify a suitable backbone which can be used in combination with hemagglutinin (HA) and neuraminidase (NA) glycoproteins from any IBV strain to be incorporated into the seasonal vaccine. We have characterized and ranked the growth profiles of the 71 IBV strains and the best performing strains were used for co-infection of eggs, followed by serial passaging to select for high-growth reassortant viruses. After serial passaging, we selected 10 clonal isolates based on their growth profiles assessed by hemagglutination and plaque-forming units. We then generated reverse genetics systems for the three clones that performed best in growth curves. The selected backbones were then used to generate different reassortant viruses with HA/NA combinations from high and low titer yielding wild type IBV. When the growth profiles of the recombinant reassortant viruses were tested, the low titer yielding HA/NA viruses with the selected backbones yielded higher titers similar to those from high titer yielding HA/NA combinations. The use of these IBV backbones with improved replication in eggs might increase yields for the influenza B virus components of seasonal influenza virus vaccines.

11.
Infect Dis Rep ; 14(3): 453-469, 2022 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-35735759

RESUMEN

Using new and original nineteenth-century sources, we analysed the epidemiology, clinical features and virology of the 1889 pandemic, which was referred to at the time as 'Russian flu' or 'Asiatic flu'. However, we rejected this identification of the disease as an 'influenza', which we believe to have been based on insufficient knowledge of the causative agent and instead posit that the pandemic was caused by a coronavirus. We provide a new account of the 1889-1893 pandemic, with a more detailed chronology that included at least four epidemiological waves. At the end of 1889, a new virus appeared in Europe, which could be identified as the coronavirus HCoV-OC43, causing crude death rates of 1.3 per 1000 population in St Petersburg; 2.1 per 1000 in Paris; 2.8 per 1000 in Bilbao and on the French-Spanish border; between 2.9 and 5.2 per 1000 in small towns in the Basque Country; and 5.8 deaths per 1000 in Madrid, which had the highest death rate. The clinical features of the disease differed from classical influenza pandemics in terms of the latency phase, duration, symptomatology, convalescence, immunity, age and death rates. Another factor to be considered was the neurotropic capacity of the disease. The most frequent form of the 1889 pandemic was the 'nervous form', with specific symptoms such as 'heavy headache' (céphalalgie gravative), tiredness, fever and delirium. There are strong parallels between the 1889-1894 pandemic and the COVID-19 pandemic, and a better understanding of the former may therefore help us to better manage the latter.

12.
J Virol ; 84(2): 1066-75, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19906934

RESUMEN

The entry of enveloped viruses into host cells is preceded by membrane fusion, which in paramyxoviruses is triggered by the fusion (F) protein. Refolding of the F protein from a metastable conformation to a highly stable postfusion form is critical for the promotion of fusion, although the mechanism is still not well understood. Here we examined the effects of mutations of individual residues of the F protein of Newcastle disease virus, located at critical regions of the protein, such as the C terminus of the N-terminal heptad repeat (HRA) and the N terminus of the C-terminal heptad repeat (HRB). Seven of the mutants were expressed at the cell surface, showing differences in antibody reactivity in comparison with the F wild type. The N211A, L461A, I463A, and I463F mutants showed a hyperfusogenic phenotype both in syncytium and in dye transfer assays. The four mutants promoted fusion more efficiently at lower temperatures than the wild type did, meaning they probably had lower energy requirements for activation. Moreover, the N211A, I463A, and I463F mutants exhibited hemagglutinin-neuraminidase (HN)-independent activity when influenza virus hemagglutinin (HA) was coexpressed as an attachment protein. The data are discussed in terms of alterations of the refolding pathway and/or the stability of the prefusion and fusion conformations.


Asunto(s)
Proteína HN/metabolismo , Mutación , Virus de la Enfermedad de Newcastle/metabolismo , Proteínas Virales de Fusión/genética , Animales , Línea Celular , Cricetinae , Células Gigantes/metabolismo , Proteína HN/genética , Células HeLa , Humanos , Fusión de Membrana , Mutagénesis Sitio-Dirigida , Virus de la Enfermedad de Newcastle/genética , Fenotipo , Conformación Proteica , Transfección , Proteínas Virales de Fusión/metabolismo
13.
J Virol ; 84(19): 9760-74, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20660196

RESUMEN

Dengue virus (DENV) is the most prevalent arthropod-borne human virus, able to infect and replicate in human dendritic cells (DCs), inducing their activation and the production of proinflammatory cytokines. However, DENV can successfully evade the immune response in order to produce disease in humans. Several mechanisms of immune evasion have been suggested for DENV, most of them involving interference with type I interferon (IFN) signaling. We recently reported that DENV infection of human DCs does not induce type I IFN production by those infected DCs, impairing their ability to prime naive T cells toward Th1 immunity. In this article, we report that DENV also reduces the ability of DCs to produce type I IFN in response to several inducers, such as infection with other viruses or exposure to Toll-like receptor (TLR) ligands, indicating that DENV antagonizes the type I IFN production pathway in human DCs. DENV-infected human DCs showed a reduced type I IFN response to Newcastle disease virus (NDV), Sendai virus (SeV), and Semliki Forest virus (SFV) infection and to the TLR3 agonist poly(I:C). This inhibitory effect is DENV dose dependent, requires DENV replication, and takes place in DENV-infected DCs as early as 2 h after infection. Expressing individual proteins of DENV in the presence of an IFN-alpha/beta production inducer reveals that a catalytically active viral protease complex is required to reduce type I IFN production significantly. These results provide a new mechanism by which DENV evades the immune system in humans.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/virología , Virus del Dengue/inmunología , Virus del Dengue/patogenicidad , Evasión Inmune , Interferón Tipo I/biosíntesis , Proteínas no Estructurales Virales/fisiología , Animales , Secuencia de Bases , Línea Celular , Cartilla de ADN/genética , Virus del Dengue/genética , Virus del Dengue/fisiología , Perros , Humanos , Técnicas In Vitro , Virus de la Enfermedad de Newcastle/inmunología , Virus de la Enfermedad de Newcastle/fisiología , Virus de los Bosques Semliki/inmunología , Virus Sendai/inmunología , Transducción de Señal/inmunología , Linfocitos T/inmunología , Receptores Toll-Like/metabolismo , Proteínas no Estructurales Virales/genética , Replicación Viral
14.
Vaccine ; 38(8): 1953-1961, 2020 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-31959425

RESUMEN

The stalk of the influenza virus hemagglutinin (HA) is an attractive target for antibody-based universal influenza virus vaccine development. While antibodies that target this part of the virus can be neutralizing, it has been shown in recent years that Fc receptor-mediated effector functions are of significant importance for the protective effect of anti-stalk antibodies. Several assays to measure Fc-Fc receptor interaction-based effector functions like antibody-dependent cellular cytotoxicity and antibody-dependent cellular phagocytosis exist, but they suffer from limitations such as low throughput and high run-to-run variability. Reporter assays for antibody-dependent cellular cytotoxicity based on reporter cells that express luciferase upon engagement of human FcγRIIIa with the Fc of antigen-bound antibodies have been developed as well. These reporter assays can be used in a higher throughput setting with limited run-to-run assay variability but since they express only one Fc receptor, their biological relevance is unclear. Here we optimized an antibody-dependent cellular cytotoxicity reporter assay to measure the activity of antibodies to the conserved stalk domain of H1 hemagglutinin. The assay was then correlated to a CD107a-based degranulation assay, and a strong and significant correlation could be observed. This data suggests that the FcγRIIIa-based reporter assay is a good substitute for functional assays, especially in settings where larger sample numbers need to be analyzed.


Asunto(s)
Anticuerpos Antivirales/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Gripe Humana/inmunología , Adolescente , Adulto , Anciano , Anticuerpos Neutralizantes/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos , Degranulación de la Célula , Preescolar , Humanos , Lactante , Proteína 1 de la Membrana Asociada a los Lisosomas/inmunología , Persona de Mediana Edad , Adulto Joven
15.
Cell Rep Med ; 1(8): 100130, 2020 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-33294855

RESUMEN

Hemagglutination-inhibitory antibodies are usually highly strain specific with little effect on infection with drifted or shifted strains. The significance of broadly cross-reactive non-HAI anti-influenza antibodies against conserved domains of virus glycoproteins, such as the hemagglutinin (HA) stalk, is of great interest. We characterize a cohort of 40 H1N1pmd09 influenza-infected patients and identify lower respiratory symptoms (LRSs) as a predictor for development of pneumonia. A binomial logistic regression of log10 pre-existing antibody values shows that the probability of LRS occurrence decreased with increased anti-HA full-length and stalk antibody ELISA titers. However, a multilevel logistic regression model adjusted by other potential serocorrelates demonstrates that only antibodies directed against the stalk of HA correlate with protection from lower respiratory infection, limiting disease progression. Our predictive model indicates that a threshold of protective immunity based on broadly cross-reactive HA stalk antibodies could be feasible.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Hemaglutininas/inmunología , Gripe Humana/inmunología , Síntomas del Sistema Urinario Inferior/inmunología , Adulto , Anciano , Animales , Línea Celular , Protección Cruzada/inmunología , Reacciones Cruzadas/inmunología , Perros , Femenino , Pruebas de Inhibición de Hemaglutinación/métodos , Humanos , Subtipo H1N1 del Virus de la Influenza A/inmunología , Síntomas del Sistema Urinario Inferior/virología , Células de Riñón Canino Madin Darby , Masculino , Persona de Mediana Edad , Pruebas de Neutralización/métodos , Adulto Joven
16.
J Virol ; 82(21): 10580-90, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18768976

RESUMEN

Type B influenza viruses can cause substantial morbidity and mortality in the population, and vaccination remains by far the best means of protection against infections with these viruses. Here, we report the construction of mutant influenza B viruses for potential use as improved live-virus vaccine candidates. Employing reverse genetics, we altered the NS1 gene, which encodes a type I interferon (IFN) antagonist. The resulting NS1 mutant viruses induced IFN and, as a consequence, were found to be attenuated in vitro and in vivo. The absence of pathogenicity of the NS1 mutants in both BALB/c and C57BL/6 PKR(-/-) mice was confirmed. We also provide evidence that influenza B virus NS1 mutants induce a self-adjuvanted immune response and confer effective protection against challenge with both homologous and heterologous B virus strains in mice.


Asunto(s)
Virus de la Influenza B/genética , Virus de la Influenza B/inmunología , Vacunas contra la Influenza/genética , Vacunas contra la Influenza/inmunología , Proteínas no Estructurales Virales/genética , Animales , Anticuerpos Antivirales/sangre , Peso Corporal , Línea Celular , Chlorocebus aethiops , Perros , Femenino , Inmunoglobulina A/análisis , Inmunoglobulina G/sangre , Interferón gamma/biosíntesis , Pulmón/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Mucosa Nasal/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Análisis de Supervivencia , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Ensayo de Placa Viral
17.
Repert. med. cir ; 33(1): 61-66, 2024. tab, graf
Artículo en Español | LILACS, COLNAL | ID: biblio-1552533

RESUMEN

Introducción: el ataque cerebrovascular (ACV) es común a nivel mundial, una de cada cuatro personas puede presentarlo a lo largo de la vida. Constituye la segunda causa de muerte y la tercera principal de discapacidad. Es importante la atención integral para lograr un impacto en la calidad de vida. Objetivo: determinar la calidad de atención en el manejo del ACV isquémico agudo de los pacientes que consultaron al servicio de neurología en los Hospitales de San José e Infantil Universitario de San José, Bogotá DC, entre enero 1/2019 y enero 1/2020. Metodología: estudio descriptivo de corte transversal. El criterio de inclusión fue pacientes mayores de 18 años con diagnóstico de ACV isquémico. La información se recolectó de las historias clínicas, se empleó estadística descriptiva para analizar los datos. Resultados: se incluyeron 411 pacientes, 88,8% sin alteración del estado de conciencia, 26,4% ingresaron antes de las 4,5 horas de ventana para trombólisis, se realizaron procedimientos de recanalización endovenosa a 11,4%. El tiempo puerta aguja tuvo una mediana de 37,2 minutos comparado con la mediana nacional de tiempo que fue 56,5 min según lo registrado en la plataforma ResQ. El 72% recibió terapia antiagregante y estatina 88.8%. Discusión y conclusiones: al identificar los síntomas es importante ser estrictos en el tiempo de atención y la implementación de plataformas para óptimos planes de atención. Se requieren campañas masivas de educación, así como que planes de mejora institucionales.


Introduction: cerebrovascular attack (CVA) is common worldwide. One in four people may have a stroke during their lifetime. It is the second leading cause of death and the third leading cause of disability. Thus, it is important to provide integrated care to achieve an impact on quality of life. Objective: to determine ischemic CVA management quality of care among patients seen at the neurology service of the San José and Infantil Universitario de San José hospitals in Bogotá DC, between January 1/2019 and January 1/2020. Methodology: a descriptive, cross-sectional study. The inclusion criteria included patients over 18 years of age diagnosed with ischemic CVA. Information was collected from clinical records. Data was analyzed using descriptive statistics. Results: 411 patients were included; 88.8% had an altered state of consciousness, 26.4% were admitted within the 4.5-hour window for thrombolysis; 11.4% underwent intravenous reperfusion procedures. Door-to-needle time: median was 37.2 minutes versus the national media of 56.5 min according to the ResQ records platform; 72% received anti-platelet therapy and 88.8% received statins. Discussion and conclusions: the establishment of a strict time to care and the implementation of platforms to improve care plans, is essential. Massive education campaigns are required, as well as, institutional improvement plans.


Asunto(s)
Humanos
18.
Cell Rep ; 29(12): 3997-4009.e5, 2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31851929

RESUMEN

Influenza A viruses (IAVs) have a remarkable tropism in their ability to circulate in both mammalian and avian species. The IAV NS1 protein is a multifunctional virulence factor that inhibits the type I interferon host response through a myriad of mechanisms. How NS1 has evolved to enable this remarkable property across species and its specific impact in the overall replication, pathogenicity, and host preference remain unknown. Here we analyze the NS1 evolutionary landscape and host tropism using a barcoded library of recombinant IAVs. Results show a surprisingly great variety of NS1 phenotypes according to their ability to replicate in different hosts. The IAV NS1 genes appear to have taken diverse and random evolutionary pathways within their multiple phylogenetic lineages. In summary, the high evolutionary plasticity of this viral protein underscores the ability of IAVs to adapt to multiple hosts and aids in our understanding of its global prevalence.


Asunto(s)
Especificidad del Huésped/genética , Interacciones Huésped-Patógeno/genética , Virus de la Influenza A/patogenicidad , Mutación , Infecciones por Orthomyxoviridae/virología , Proteínas no Estructurales Virales/metabolismo , Replicación Viral , Animales , Perros , Femenino , Inmunidad Innata , Virus de la Influenza A/genética , Células de Riñón Canino Madin Darby , Ratones , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/patología , Filogenia , Proteínas no Estructurales Virales/genética
19.
Vaccine ; 36(9): 1146-1153, 2018 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-29395518

RESUMEN

Vaccination remains the best available prophylaxis to prevent influenza virus infections, yet current inadequacies in influenza virus vaccine manufacturing often lead to vaccine shortages at times when the vaccine is most needed, as it was the case during the last influenza virus pandemic. Novel influenza virus vaccine production systems will be crucial to improve public health and safety. Here we report the optimization of influenza B virus growth in the proprietary EB66® cell line, currently in use for human vaccine production. To this end, we collected, curated and sequenced 71 influenza B viruses selected for high diversity in date of isolation and lineage. This viral collection was tested for ability to enter and replicate within EB66® cells in a single cycle assay and appears to readily infect these cells. When the collection was tested for viral progeny production in a multi-cycle assay, we found a large variation from strain to strain. The strains with the top growth characteristics from the B/Victoria and B/Yamagata lineages were selected for vaccine backbone generation using a reverse genetics system. We then showed that these backbones maintain their desirable growth within EB66® cells when the HA and NA from poorly growing strains were substituted for the parental segments, indicating that the selected backbones are viable options for vaccine production in EB66®. Finally, we show that compounds previously reported to enhance influenza virus growth in cell culture also increase virus production in the EB66® cell line.


Asunto(s)
Virus de la Influenza B/crecimiento & desarrollo , Virus de la Influenza B/genética , Genética Inversa , Animales , Línea Celular , Patos , Hemaglutinación por Virus , Vacunas contra la Influenza
20.
Nat Struct Mol Biol ; 25(9): 885-893, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30177761

RESUMEN

Viral infection perturbs host cells and can be used to uncover regulatory mechanisms controlling cellular responses and susceptibility to infections. Using cell biological, biochemical, and genetic tools, we reveal that influenza A virus (IAV) infection induces global transcriptional defects at the 3' ends of active host genes and RNA polymerase II (RNAPII) run-through into extragenic regions. Deregulated RNAPII leads to expression of aberrant RNAs (3' extensions and host-gene fusions) that ultimately cause global transcriptional downregulation of physiological transcripts, an effect influencing antiviral response and virulence. This phenomenon occurs with multiple strains of IAV, is dependent on influenza NS1 protein, and can be modulated by SUMOylation of an intrinsically disordered region (IDR) of NS1 expressed by the 1918 pandemic IAV strain. Our data identify a strategy used by IAV to suppress host gene expression and indicate that polymorphisms in IDRs of viral proteins can affect the outcome of an infection.


Asunto(s)
Gripe Humana/genética , ARN Polimerasa II/genética , Regiones Terminadoras Genéticas/genética , Humanos , Virus de la Influenza A/patogenicidad , Virus de la Influenza A/fisiología , Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA