Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 112
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Clin Microbiol Rev ; 36(1): e0004022, 2023 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-36645300

RESUMEN

Preventing and controlling influenza virus infection remains a global public health challenge, as it causes seasonal epidemics to unexpected pandemics. These infections are responsible for high morbidity, mortality, and substantial economic impact. Vaccines are the prophylaxis mainstay in the fight against influenza. However, vaccination fails to confer complete protection due to inadequate vaccination coverages, vaccine shortages, and mismatches with circulating strains. Antivirals represent an important prophylactic and therapeutic measure to reduce influenza-associated morbidity and mortality, particularly in high-risk populations. Here, we review current FDA-approved influenza antivirals with their mechanisms of action, and different viral- and host-directed influenza antiviral approaches, including immunomodulatory interventions in clinical development. Furthermore, we also illustrate the potential utility of machine learning in developing next-generation antivirals against influenza.


Asunto(s)
Vacunas contra la Influenza , Gripe Humana , Infecciones por Orthomyxoviridae , Orthomyxoviridae , Humanos , Gripe Humana/tratamiento farmacológico , Gripe Humana/prevención & control , Antivirales/farmacología , Antivirales/uso terapéutico , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Vacunas contra la Influenza/uso terapéutico
2.
Clin Infect Dis ; 76(3): e1168-e1176, 2023 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-36031405

RESUMEN

BACKGROUND: Antibody responses to non-egg-based standard-dose cell-culture influenza vaccine (containing 15 µg hemagglutinin [HA]/component) and recombinant vaccine (containing 45 µg HA/component) during consecutive seasons have not been studied in the United States. METHODS: In a randomized trial of immunogenicity of quadrivalent influenza vaccines among healthcare personnel (HCP) aged 18-64 years over 2 consecutive seasons, HCP who received recombinant-HA influenza vaccine (RIV) or cell culture-based inactivated influenza vaccine (ccIIV) during the first season (year 1) were re-randomized the second season of 2019-2020 (year 2 [Y2]) to receive ccIIV or RIV, resulting in 4 ccIIV/RIV combinations. In Y2, hemagglutination inhibition antibody titers against reference cell-grown vaccine viruses were compared in each ccIIV/RIV group with titers among HCP randomized both seasons to receive egg-based, standard-dose inactivated influenza vaccine (IIV) using geometric mean titer (GMT) ratios of Y2 post-vaccination titers. RESULTS: Y2 data from 414 HCP were analyzed per protocol. Compared with 60 IIV/IIV recipients, 74 RIV/RIV and 106 ccIIV/RIV recipients showed significantly elevated GMT ratios (Bonferroni corrected P < .007) against all components except A(H3N2). Post-vaccination GMT ratios for ccIIV/ccIIV and RIV/ccIIV were not significantly elevated compared with IIV/IIV except for RIV/ccIIV against A(H1N1)pdm09. CONCLUSIONS: In adult HCP, receipt of RIV in 2 consecutive seasons or the second season was more immunogenic than consecutive egg-based IIV for 3 of the 4 components of quadrivalent vaccine. Immunogenicity of ccIIV/ccIIV was similar to that of IIV/IIV. Differences in HA antigen content may play a role in immunogenicity of influenza vaccination in consecutive seasons. CLINICAL TRIALS REGISTRATION: NCT03722589.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A , Vacunas contra la Influenza , Gripe Humana , Vacuna contra Viruela , Adulto , Humanos , Anticuerpos Antivirales , Técnicas de Cultivo de Célula , Atención a la Salud , Pruebas de Inhibición de Hemaglutinación , Subtipo H3N2 del Virus de la Influenza A , Estados Unidos , Vacunación , Vacunas Combinadas , Vacunas de Productos Inactivados , Vacunas Sintéticas
3.
Crit Rev Immunol ; 41(3): 57-82, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35378011

RESUMEN

Influenza is a highly contagious respiratory virus that causes mild to severe respiratory illness, as well as death, and remains a serious threat to human health. Annual vaccination is the most cost-effective way to control influenza; however, the vaccine does not provide protection against emerging strains with epidemic and pandemic potential. Several antivirals have been developed to treat influenza but there is a rapid emergence of antiviral resistant strains. Therefore, there is an urgent need to understand the virus and its interactions with the host immune system so that novel strategies can be developed for prophylactic and therapeutic interventions. Innate lymphoid cells (ILCs), a family of immune cells present in the peripheral circulation and in mucosal tissues, play an important role in regulation of tissue homeostasis, inflammation, and immunity. This review examines the current understanding and therapeutic potential of ILCs during influenza virus infection in humans.


Asunto(s)
Vacunas contra la Influenza , Gripe Humana , Humanos , Inmunidad Innata , Vacunas contra la Influenza/uso terapéutico , Gripe Humana/epidemiología , Gripe Humana/prevención & control , Linfocitos , Vacunación
4.
Clin Infect Dis ; 73(11): 1973-1981, 2021 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-34245243

RESUMEN

BACKGROUND: RIV4 and cell-culture based inactivated influenza vaccine (ccIIV4) have not been compared to egg-based IIV4 in healthcare personnel, a population with frequent influenza vaccination that may blunt vaccine immune responses over time. We conducted a randomized trial among healthcare personnel (HCP) aged 18-64 years to compare humoral immune responses to ccIIV4 and RIV4 to IIV4. METHODS: During the 2018-2019 season, participants were randomized to receive ccIIV4, RIV4, or IIV4 and had serum samples collected prevaccination, 1 and 6 months postvaccination. Serum samples were tested by hemagglutination inhibition (HI) for influenza A/H1N1, B/Yamagata, and B/Victoria and microneutralization (MN) for A/H3N2 against cell-grown vaccine reference viruses. Primary outcomes at 1 month were seroconversion rate (SCR), geometric mean titers (GMT), GMT ratio, and mean fold rise (MFR) in the intention-to-treat population. RESULTS: In total, 727 participants were included (283 ccIIV4, 202 RIV4, and 242 IIV4). At 1 month, responses to ccIIV4 were similar to IIV4 by SCR, GMT, GMT ratio, and MFR. RIV4 induced higher SCRs, GMTs, and MFRs than IIV4 against A/H1N1, A/H3N2, and B/Yamagata. The GMT ratio of RIV4 to egg-based vaccines was 1.5 (95% confidence interval [CI] 1.2-1.9) for A/H1N1, 3.0 (95% CI: 2.4-3.7) for A/H3N2, 1.1 (95% CI: .9-1.4) for B/Yamagata, and 1.1 (95% CI: .9-1.3) for B/Victoria. At 6 months, ccIIV4 recipients had similar GMTs to IIV4, whereas RIV4 recipients had higher GMTs against A/H3N2 and B/Yamagata. CONCLUSIONS: RIV4 resulted in improved antibody responses by HI and MN compared to egg-based vaccines against 3 of 4 cell-grown vaccine strains 1 month postvaccination, suggesting a possible additional benefit from RIV4. CLINICAL TRIALS REGISTRATION: NCT03722589.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A , Vacunas contra la Influenza , Gripe Humana , Anticuerpos Antivirales , Técnicas de Cultivo de Célula , Atención a la Salud , Pruebas de Inhibición de Hemaglutinación , Humanos , Inmunogenicidad Vacunal , Subtipo H3N2 del Virus de la Influenza A , Virus de la Influenza B , Gripe Humana/prevención & control , Vacunas de Productos Inactivados
5.
Clin Infect Dis ; 71(7): 1704-1714, 2020 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-31828291

RESUMEN

BACKGROUND: Enhanced influenza vaccines may improve protection for older adults, but comparative immunogenicity data are limited. Our objective was to examine immune responses to enhanced influenza vaccines, compared to standard-dose vaccines, in community-dwelling older adults. METHODS: Community-dwelling older adults aged 65-82 years in Hong Kong were randomly allocated (October 2017-January 2018) to receive 2017-2018 Northern hemisphere formulations of a standard-dose quadrivalent vaccine, MF59-adjuvanted trivalent vaccine, high-dose trivalent vaccine, or recombinant-hemagglutinin (rHA) quadrivalent vaccine. Sera collected from 200 recipients of each vaccine before and at 30-days postvaccination were assessed for antibodies to egg-propagated vaccine strains by hemagglutination inhibition (HAI) and to cell-propagated A/Hong Kong/4801/2014(H3N2) virus by microneutralization (MN). Influenza-specific CD4+ and CD8+ T cell responses were assessed in 20 participants per group. RESULTS: Mean fold rises (MFR) in HAI titers to egg-propagated A(H1N1) and A(H3N2) and the MFR in MN to cell-propagated A(H3N2) were statistically significantly higher in the enhanced vaccine groups, compared to the standard-dose vaccine. The MFR in MN to cell-propagated A(H3N2) was highest among rHA recipients (4.7), followed by high-dose (3.4) and MF59-adjuvanted (2.9) recipients, compared to standard-dose recipients (2.3). Similarly, the ratio of postvaccination MN titers among rHA recipients to cell-propagated A(H3N2) recipients was 2.57-fold higher than the standard-dose vaccine, which was statistically higher than the high-dose (1.33-fold) and MF59-adjuvanted (1.43-fold) recipient ratios. Enhanced vaccines also resulted in the boosting of T-cell responses. CONCLUSIONS: In this head-to-head comparison, older adults receiving enhanced vaccines showed improved humoral and cell-mediated immune responses, compared to standard-dose vaccine recipients. CLINICAL TRIALS REGISTRATION: NCT03330132.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A , Vacunas contra la Influenza , Gripe Humana , Adyuvantes Inmunológicos , Anciano , Anciano de 80 o más Años , Anticuerpos Antivirales , Pruebas de Inhibición de Hemaglutinación , Humanos , Inmunogenicidad Vacunal , Subtipo H3N2 del Virus de la Influenza A , Gripe Humana/prevención & control , Escualeno
6.
Immunity ; 34(6): 854-65, 2011 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-21703540

RESUMEN

The nucleotide-binding domain and leucine-rich-repeat-containing (NLR) proteins regulate innate immunity. Although the positive regulatory impact of NLRs is clear, their inhibitory roles are not well defined. We showed that Nlrx1(-/-) mice exhibited increased expression of antiviral signaling molecules IFN-ß, STAT2, OAS1, and IL-6 after influenza virus infection. Consistent with increased inflammation, Nlrx1(-/-) mice exhibited marked morbidity and histopathology. Infection of these mice with an influenza strain that carries a mutated NS-1 protein, which normally prevents IFN induction by interaction with RNA and the intracellular RNA sensor RIG-I, further exacerbated IL-6 and type I IFN signaling. NLRX1 also weakened cytokine responses to the 2009 H1N1 pandemic influenza virus in human cells. Mechanistically, Nlrx1 deletion led to constitutive interaction of MAVS and RIG-I. Additionally, an inhibitory function is identified for NLRX1 during LPS activation of macrophages where the MAVS-RIG-I pathway was not involved. NLRX1 interacts with TRAF6 and inhibits NF-κB activation. Thus, NLRX1 functions as a checkpoint of overzealous inflammation.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/inmunología , Proteínas Mitocondriales/inmunología , Infecciones por Orthomyxoviridae/inmunología , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales/inmunología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Células Cultivadas , Interferón beta/biosíntesis , Interferón beta/inmunología , Interleucina-6/biosíntesis , Interleucina-6/inmunología , Macrófagos/inmunología , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Proteínas Mitocondriales/deficiencia , FN-kappa B/inmunología , FN-kappa B/metabolismo , Proteínas del Tejido Nervioso/inmunología , Proteínas del Tejido Nervioso/metabolismo , Receptores de Superficie Celular , Factor 6 Asociado a Receptor de TNF/inmunología , Factor 6 Asociado a Receptor de TNF/metabolismo
7.
J Infect Dis ; 220(5): 743-751, 2019 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-31045222

RESUMEN

BACKGROUND: Human immunodeficiency virus (HIV)-infected persons are at a higher risk of severe influenza. Although we have shown that a standard-dose intradermal influenza vaccine versus a standard-dose intramuscular influenza vaccine does not result in differences in hemagglutination-inhibition titers in this population, a comprehensive examination of cell-mediated immune responses remains lacking. METHODS: Serological, antigen-specific B-cell, and interleukin 2-, interferon γ-, and tumor necrosis factor α-secreting T-cell responses were assessed in 79 HIV-infected men and 79 HIV-uninfected men. RESULTS: The route of vaccination did not affect the immunoglobulin A and immunoglobulin G (IgG) plasmablast or memory B-cell response, although these were severely impaired in the group with a CD4+ T-cell count of <200 cells/µL. The frequencies of IgG memory B cells measured on day 28 after vaccination were highest in the HIV-uninfected group, followed by the group with a CD4+ T-cell count of ≥200 cells/µL and the group with a CD4+ T-cell count of <200 cells/µL. The route of vaccination did not affect the CD4+ or CD8+ T-cell responses measured at various times after vaccination. CONCLUSIONS: The route of vaccination had no effect on antibody responses, antibody avidity, T-cell responses, or B-cell responses in HIV-infected or HIV-uninfected subjects. With the serological and cellular immune responses to influenza vaccination being impaired in HIV-infected individuals with a CD4+ T-cell count of <200 cells/µL, passive immunization strategies need to be explored to protect this population. CLINICAL TRIALS REGISTRATION: NCT01538940.


Asunto(s)
Infecciones por VIH/inmunología , Inmunidad Celular/inmunología , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/normas , Gripe Humana/prevención & control , Adulto , Anticuerpos Antivirales/inmunología , Formación de Anticuerpos , Linfocitos B/inmunología , Recuento de Linfocito CD4 , Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos , Infecciones por VIH/complicaciones , Pruebas de Inhibición de Hemaglutinación , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Humanos , Inmunoglobulina A , Inmunoglobulina G , Subtipo H1N1 del Virus de la Influenza A/inmunología , Interferón gamma/metabolismo , Interleucina-2/metabolismo , Masculino , Persona de Mediana Edad , Tailandia , Factor de Necrosis Tumoral alfa/metabolismo , Vacunación
8.
J Virol ; 91(11)2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28331092

RESUMEN

Monkeypox virus (MPXV) is a human pathogen that is a member of the Orthopoxvirus genus, which includes Vaccinia virus and Variola virus (the causative agent of smallpox). Human monkeypox is considered an emerging zoonotic infectious disease. To identify host factors required for MPXV infection, we performed a genome-wide insertional mutagenesis screen in human haploid cells. The screen revealed several candidate genes, including those involved in Golgi trafficking, glycosaminoglycan biosynthesis, and glycosylphosphatidylinositol (GPI)-anchor biosynthesis. We validated the role of a set of vacuolar protein sorting (VPS) genes during infection, VPS51 to VPS54 (VPS51-54), which comprise the Golgi-associated retrograde protein (GARP) complex. The GARP complex is a tethering complex involved in retrograde transport of endosomes to the trans-Golgi apparatus. Our data demonstrate that VPS52 and VPS54 were dispensable for mature virion (MV) production but were required for extracellular virus (EV) formation. For comparison, a known antiviral compound, ST-246, was used in our experiments, demonstrating that EV titers in VPS52 and VPS54 knockout (KO) cells were comparable to levels exhibited by ST-246-treated wild-type cells. Confocal microscopy was used to examine actin tail formation, one of the viral egress mechanisms for cell-to-cell dissemination, and revealed an absence of actin tails in VPS52KO- or VPS54KO-infected cells. Further evaluation of these cells by electron microscopy demonstrated a decrease in levels of wrapped viruses (WVs) compared to those seen with the wild-type control. Collectively, our data demonstrate the role of GARP complex genes in double-membrane wrapping of MVs necessary for EV formation, implicating the host endosomal trafficking pathway in orthopoxvirus infection.IMPORTANCE Human monkeypox is an emerging zoonotic infectious disease caused by Monkeypox virus (MPXV). Of the two MPXV clades, the Congo Basin strain is associated with severe disease, increased mortality, and increased human-to-human transmission relative to the West African strain. Monkeypox is endemic in regions of western and central Africa but was introduced into the United States in 2003 from the importation of infected animals. The threat of MPXV and other orthopoxviruses is increasing due to the absence of routine smallpox vaccination leading to a higher proportion of naive populations. In this study, we have identified and validated candidate genes that are required for MPXV infection, specifically, those associated with the Golgi-associated retrograde protein (GARP) complex. Identifying host targets required for infection that prevents extracellular virus formation such as the GARP complex or the retrograde pathway can provide a potential target for antiviral therapy.


Asunto(s)
Endosomas/metabolismo , Interacciones Huésped-Patógeno , Proteínas de la Membrana/genética , Monkeypox virus/fisiología , Proteínas de Transporte Vesicular/metabolismo , Actinas/efectos de los fármacos , Actinas/metabolismo , Animales , Benzamidas/farmacología , Transporte Biológico , Línea Celular , Genoma Humano , Glicosaminoglicanos/biosíntesis , Glicosaminoglicanos/genética , Glicosilfosfatidilinositoles/biosíntesis , Aparato de Golgi/genética , Aparato de Golgi/metabolismo , Haploidia , Humanos , Isoindoles/farmacología , Proteínas de la Membrana/metabolismo , Mpox/virología , Mutagénesis Insercional , Proteínas de Transporte Vesicular/genética , Carga Viral , Replicación Viral
9.
J Infect Dis ; 216(suppl_4): S560-S565, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28934453

RESUMEN

Background: Protein energy malnutrition (PEM) increases susceptibility to infectious diseases, including influenza infection, but no studies have addressed the potential influences of PEM on the immunogenicity and protective efficacy of avian influenza A(H5N1) vaccine. Methods: We investigated the role of PEM on vaccine-mediated protection after a lethal challenge with recombinant A(H5N1) virus using isocaloric diets providing either adequate protein (AP; 18% protein) or very low protein (VLP; 2% protein) in an established murine model of influenza vaccination. Results: We demonstrated that mice maintained on a VLP diet succumb to lethal challenge at greater rates than mice maintained on an AP diet, despite comparable immunization regimens. Importantly, there was no virus-induced mortality in both VLP and AP groups of mice when either group was immunized with adjuvanted low-dose A(H5N1) subvirion vaccine. Conclusions: Our results suggest that adjuvanted vaccination in populations where PEM is endemic may be one strategy to boost vaccination-promoted immunity and improve outcomes associated with highly pathogenic A(H5N1).


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Formación de Anticuerpos , Vacunas contra la Influenza/administración & dosificación , Infecciones por Orthomyxoviridae/prevención & control , Desnutrición Proteico-Calórica/inmunología , Animales , Dieta con Restricción de Proteínas/efectos adversos , Proteínas en la Dieta/administración & dosificación , Proteínas en la Dieta/sangre , Modelos Animales de Enfermedad , Femenino , Pruebas de Inhibición de Hemaglutinación , Subtipo H5N1 del Virus de la Influenza A/inmunología , Ratones , Ratones Endogámicos C57BL , Desnutrición Proteico-Calórica/virología
10.
J Gen Virol ; 98(5): 1016-1026, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28530548

RESUMEN

Human noroviruses are a major cause of acute gastroenteritis worldwide, but the lack of a robust cell culture system or small animal model have hampered a better understanding of innate immunity against these viruses. Tulane virus (TV) is the prototype virus of a tentative new genus, Recovirus, in the family Caliciviridae. Its epidemiology and biological properties most closely resemble human norovirus. The host innate immune response to RNA virus infection primarily involves pathogen-sensing toll-like receptors (TLRs) TLR3 and TLR7 and retinoic acid-inducible gene I-like receptor RIG-I and melanoma differentiation associated gene 5 (MDA5). In this study, by using siRNA knockdown, we report that TV infection in LLC-MK2 cells results in an early [3 h post infection (h p.i.), P<0.05] RIG-I-dependent and type I interferon-mediated antiviral response, whereas an MDA5-mediated antiviral effect was observed at later (12 h p.i.; P<0.05) stages of TV replication. Induction of RIG-I and MDA5 was critical for inhibition of TV replication. Furthermore, pre-activation of the RIG-I/MDA5 pathway prevented TV replication (>900-fold decrease; P<0.05), suggesting that RIG-I and MDA5 ligands could be used to develop novel preventive and therapeutic measures against norovirus.


Asunto(s)
Infecciones por Caliciviridae/inmunología , Caliciviridae/inmunología , Proteína 58 DEAD Box/metabolismo , Interacciones Huésped-Patógeno , Inmunidad Innata , Helicasa Inducida por Interferón IFIH1/metabolismo , Replicación Viral , Animales , Técnicas de Silenciamiento del Gen , Macaca mulatta
11.
J Infect Dis ; 214(7): 1030-8, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27534687

RESUMEN

BACKGROUND: Emergence of antigenically drifted influenza A(H3N2) viruses resulted in reduced vaccine effectiveness in all age groups during the 2014-2015 influenza season. In children, inactivated influenza vaccine (IIV) elicited neutralizing antibodies (Abs) against drifted strains at significantly lower levels than against the vaccine strain. Little is known about the cross-reactivity of cell-mediated immunity against drifted strains in children. METHODS: Children aged 3-17 years (n = 48) received IIV during the 2014-2015 influenza season. Peripheral blood mononuclear cells, collected before (on day 0) and after (on days 7 and 21) vaccination were evaluated for induction of cross-reactive plasmablasts, memory B cells, and cytokine-secreting CD4(+) and CD8(+) T cells against the vaccine and drifted A(H3N2) viruses by an enzyme-linked immunospot assay and flow cytometry. RESULTS: IIV increased frequencies of plasmablasts and memory B cells. The overall induction of the T-cell response was not significant. Both B-cell and T-cell responses showed significant cross-reactivity against A(H3N2) viruses. Age and preexisting immunity affected virus-specific plasmablast responses and fold-change of T-cell responses, respectively. The proportion of T-helper type 1-prone (ie, interferon γ- or tumor necrosis factor α-secreting) CD4(+) T cell responses also increased with age. CONCLUSIONS: In children aged 3-17 years, B- and T-cell responses following IIV receipt showed significant cross-reactivity against A(H3N2) viruses during a vaccine mismatch season.


Asunto(s)
Linfocitos B/inmunología , Flujo Genético , Inmunidad Heteróloga , Subtipo H3N2 del Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Gripe Humana/virología , Linfocitos T/inmunología , Adolescente , Niño , Preescolar , Reacciones Cruzadas , Femenino , Humanos , Subtipo H3N2 del Virus de la Influenza A/genética , Masculino
12.
J Infect Dis ; 214(10): 1477-1486, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27571905

RESUMEN

BACKGROUND: Influenza viruses gradually accumulate point mutations, reducing the effectiveness of prior immune protection. METHODS: Children aged 9-14 years received 2010-2011 trivalent inactivated influenza vaccine (TIV). Vaccination history, hemagglutination-inhibition (HI) titers, and cell-mediated immune responses were assessed to investigate the cross-reactivity with past and future influenza virus strains. RESULTS: 2010-2011 TIV induced significant T-cell responses and HI titers of ≥160, with a fold-rise of ≥4 and titers of ≥100 maintained for >7 months in the majority of children. Pre-existing memory B cells in these children differentiated quickly to antibody-secreting cells to the new vaccine antigens. Children vaccinated in the previous year maintained high HI titers well into 2010, demonstrating elevated HI titers against A/Perth/16/2009, the future (in 2010-2011) H3N2 component. Prior vaccination enhanced CD8+ T-cell responses to A/Perth/16/2009. Children vaccinated with the prior 2009-2010 seasonal vaccine also demonstrated higher preexisting levels of interferon γ-secreting CD4+CD69+ T cells to 2009 pandemic influenza A(H1N1). Children previously vaccinated with 2009-2010 seasonal influenza vaccine also showed greater expansion of tumor necrosis factor α-secreting CD8+CD69+ T cells to 2009 pandemic influenza A(H1N1) upon vaccination in the 2010-2011 season than those who were not previously vaccinated. CONCLUSIONS: Seasonal influenza viruses continuously drift, which allows them to circumvent protective immunity, but conserved epitopes provide immunological cross-reactivity in children through either vaccination directly or through prime/boost in the prior influenza season.


Asunto(s)
Inmunidad Celular , Inmunidad Humoral , Vacunas contra la Influenza/inmunología , Gripe Humana/prevención & control , Orthomyxoviridae/inmunología , Adolescente , Anticuerpos Antivirales/sangre , Niño , Reacciones Cruzadas , Femenino , Pruebas de Inhibición de Hemaglutinación , Humanos , Vacunas contra la Influenza/administración & dosificación , Gripe Humana/inmunología , Masculino , Linfocitos T/inmunología , Factores de Tiempo , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/inmunología
13.
Eur J Immunol ; 45(3): 758-72, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25404059

RESUMEN

The NLR protein, NLRC5 is an important regulator of MHC class I gene expression, however, the role of NLRC5 in other innate immune responses is less well defined. In the present study, we report that NLRC5 binds RIG-I and that this interaction is critical for robust antiviral responses against influenza virus. Overexpression of NLRC5 in the human lung epithelial cell line, A549, and normal human bronchial epithelial cells resulted in impaired replication of influenza virus A/Puerto Rico/8/34 virus (PR8) and enhanced IFN-ß expression. Influenza virus leads to induction of IFN-ß that drives RIG-I and NLRC5 expression in host cells. Our results suggest that NLRC5 extends and stabilizes influenza virus induced RIG-I expression and delays expression of the viral inhibitor protein NS1. We show that NS1 binds to NLRC5 to suppress its function. Interaction domain mapping revealed that NLRC5 interacts with RIG-I via its N-terminal death domain and that NLRC5 enhanced antiviral activity in an leucine-rich repeat domain independent manner. Taken together, our findings identify a novel role for NLRC5 in RIG-I-mediated antiviral host responses against influenza virus infection, distinguished from the role of NLRC5 in MHC class I gene regulation.


Asunto(s)
ARN Helicasas DEAD-box/inmunología , Regulación de la Expresión Génica/inmunología , Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Péptidos y Proteínas de Señalización Intracelular/inmunología , Mucosa Respiratoria/inmunología , Proteína 58 DEAD Box , Células Epiteliales/inmunología , Células Epiteliales/patología , Células Epiteliales/virología , Células HEK293 , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Gripe Humana/patología , Estructura Terciaria de Proteína , Receptores Inmunológicos , Mucosa Respiratoria/patología , Mucosa Respiratoria/virología
14.
Cell Immunol ; 304-305: 55-8, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27106062

RESUMEN

Avian H7N9 influenza virus infection with fatal outcomes continues to pose a pandemic threat and highly immunogenic vaccines are urgently needed. In this report we show that baculovirus-derived recombinant H7 hemagglutinin protein, when delivered with RIG-I ligand, induced enhanced antibody and T cell responses and conferred protection against lethal challenge with a homologous H7N9 virus. These findings indicate the potential utility of RIG-I ligands as vaccine adjuvants to increase the immunogenicity of recombinant H7 hemagglutinin.


Asunto(s)
Proteína 58 DEAD Box/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Subtipo H7N9 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Gripe Humana/prevención & control , Infecciones por Orthomyxoviridae/prevención & control , Linfocitos T/inmunología , Adyuvantes Inmunológicos , Animales , Células Cultivadas , Femenino , Humanos , Inmunidad Humoral , Subtipo H7N9 del Virus de la Influenza A/metabolismo , Gripe Humana/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/inmunología , Receptores Inmunológicos , Receptores de Reconocimiento de Patrones/metabolismo , Linfocitos T/virología , Vacunas Sintéticas
15.
Nanomedicine ; 12(7): 1909-1917, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27112307

RESUMEN

To enhance the immunogenicity of the Influenza H5N1 vaccine, we developed an oil-in-water nanoemulsion (NE) adjuvant. NE displayed good temperature stability and maintained particle size. More importantly, it significantly enhanced IL-6 and MCP-1 production to recruit innate cells, including neutrophils, monocytes/macrophages and dendritic cells to the local environment. Furthermore, NE enhanced dendritic cell function to induce robust antigen-specific T and B cell immune responses. NE-adjuvanted H5N1 vaccine not only elicited significantly higher and long-lasting antibody responses, but also conferred enhanced protection against homologous clade 1 as well as heterologous clade 2 H5N1 virus challenge in young as well as in aged mice. The pre-existing immunity to seasonal influenza did not affect the immunogenicity of NE-adjuvanted H5N1 vaccine.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A , Vacunas contra la Influenza/administración & dosificación , Nanopartículas/química , Adyuvantes Inmunológicos , Animales , Anticuerpos Antivirales , Emulsiones , Humanos , Gripe Humana/prevención & control , Ratones
16.
J Infect Dis ; 212(12): 1923-9, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26068782

RESUMEN

We compared the innate immune response to a newly emerged swine-origin influenza A(H3N2) variant containing the M gene from 2009 pandemic influenza A(H1N1), termed "A(H3N2)vpM," to the immune responses to the 2010 swine-origin influenza A(H3N2) variant and seasonal influenza A(H3N2). Our results demonstrated that A(H3N2)vpM-induced myeloid dendritic cells secreted significantly lower levels of type I interferon (IFN) but produced significantly higher levels of proinflammatory cytokines and induced potent inflammasome activation. The reduction in antiviral immunity with increased inflammatory responses upon A(H3N2)vpM infection suggest that these viruses have the potential for increased disease severity in susceptible hosts.


Asunto(s)
Inflamasomas/metabolismo , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H3N2 del Virus de la Influenza A/inmunología , Leucocitos Mononucleares/inmunología , Animales , Línea Celular , Citocinas/metabolismo , Células Dendríticas/inmunología , Humanos , Subtipo H1N1 del Virus de la Influenza A/aislamiento & purificación , Subtipo H3N2 del Virus de la Influenza A/aislamiento & purificación , Infecciones por Orthomyxoviridae/virología , Porcinos , Enfermedades de los Porcinos/virología
17.
Trends Biochem Sci ; 36(6): 314-9, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21497095

RESUMEN

Retinoic-acid-inducible gene-I (RIG-I) is an important component of the innate immune response to many RNA viruses that limits viral replication until adaptive immunity becomes available to clear the infection. Upon binding to the nucleic acid genomes and replication intermediates of these viruses, RIG-I undergoes a complex activation process that involves post-translational modifications and structural rearrangements. Once activated, RIG-I upregulates well-studied signal transduction pathways that lead to the production of type-I interferons (IFNs) and a large variety of antiviral IFN-stimulated genes. Thus, an effective antiviral response is dependent on the interaction between pathogen-derived ligands and RIG-I. Recent work has begun to clarify the required characteristics of RIG-I activators and is setting the stage for the identification of authentic ligands used during viral infection.


Asunto(s)
ARN Helicasas DEAD-box/química , ARN Helicasas DEAD-box/metabolismo , Activación Enzimática , Inmunidad Innata/inmunología , Interferones/inmunología , Ligandos , Transducción de Señal
18.
J Virol ; 88(24): 13990-4001, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25253340

RESUMEN

UNLABELLED: Pattern recognition receptors (PRR) sense certain molecular patterns uniquely expressed by pathogens. Retinoic-acid-inducible gene I (RIG-I) is a cytosolic PRR that senses viral nucleic acids and induces innate immune activation and secretion of type I interferons (IFNs). Here, using influenza vaccine antigens, we investigated the consequences of activating the RIG-I pathway for antigen-specific adaptive immune responses. We found that mice immunized with influenza vaccine antigens coadministered with 5'ppp-double-stranded RNA (dsRNA), a RIG-I ligand, developed robust levels of hemagglutination-inhibiting antibodies, enhanced germinal center reaction, and T follicular helper cell responses. In addition, RIG-I activation enhanced antibody affinity maturation and plasma cell responses in the draining lymph nodes, spleen, and bone marrow and conferred protective immunity against virus challenge. Importantly, activation of the RIG-I pathway was able to reduce the antigen requirement by 10- to 100-fold in inducing optimal influenza-specific cellular and humoral responses, including protective immunity. The effects induced by 5'ppp-dsRNA were significantly dependent on type I IFN and IPS-1 (an adapter protein downstream of the RIG-I pathway) signaling but were independent of the MyD88- and TLR3-mediated pathways. Our results show that activation of the RIG-I-like receptor pathway programs the innate immunity to achieve qualitatively and quantitatively enhanced protective cellular adaptive immune responses even at low antigen doses, and this indicates the potential utility of RIG-I ligands as molecular adjuvants for viral vaccines. IMPORTANCE: The recently discovered RNA helicase family of RIG-I-like receptors (RLRs) is a critical component of host defense mechanisms responsible for detecting viruses and triggering innate antiviral cytokines that help control viral replication and dissemination. In this study, we show that the RLR pathway can be effectively exploited to enhance adaptive immunity and protective immune memory against viral infection. Our results show that activation of the RIG-I pathway along with influenza vaccination programs the innate immunity to induce qualitatively and quantitatively superior protective adaptive immunity against pandemic influenza viruses. More importantly, RIG-I activation at the time of vaccination allows induction of robust adaptive responses even at low vaccine antigen doses. These results highlight the potential utility of exploiting the RIG-I pathway to enhance viral-vaccine-specific immunity and have broader implications for designing better vaccines in general.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , ARN Helicasas DEAD-box/metabolismo , Centro Germinal/inmunología , Vacunas contra la Influenza/inmunología , ARN Bicatenario/administración & dosificación , Transducción de Señal , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Anticuerpos Antivirales/sangre , Proliferación Celular , Proteína 58 DEAD Box , Modelos Animales de Enfermedad , Pruebas de Inhibición de Hemaglutinación , Vacunas contra la Influenza/administración & dosificación , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Linfocitos T Colaboradores-Inductores/fisiología , Vacunación/métodos
19.
Proc Natl Acad Sci U S A ; 109(34): 13751-6, 2012 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-22869731

RESUMEN

Original antigenic sin is a phenomenon wherein sequential exposure to closely related influenza virus variants reduces antibody (Ab) response to novel antigenic determinants in the second strain and, consequently, impairs the development of immune memory. This could pose a risk to the development of immune memory in persons previously infected with or vaccinated against influenza. Here, we explored strategies to overcome original antigenic sin responses in mice sequentially exposed to two closely related hemagglutinin 1 neuraminidase 1 (H1N1) influenza strains A/PR/8/34 and A/FM/1/47. We found that dendritic cell-activating adjuvants [Bordetella pertussis toxin (PT) or CpG ODN or a squalene-based oil-in-water nanoemulsion (NE)], upon administration during the second viral exposure, completely protected mice from a lethal challenge and enhanced neutralizing-Ab titers against the second virus. Interestingly, PT and NE adjuvants when administered during the first immunization even prevented original antigenic sin in subsequent immunization without any adjuvants. As an alternative to using adjuvants, we also found that repeated immunization with the second viral strain relieved the effects of original antigenic sin. Taken together, our studies provide at least three ways of overcoming original antigenic sin.


Asunto(s)
Formación de Anticuerpos , Inmunización/métodos , Memoria Inmunológica , Orthomyxoviridae/genética , Animales , Presentación de Antígeno , Bordetella/metabolismo , Línea Celular , Islas de CpG , Perros , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Humanos , Sistema Inmunológico , Subtipo H1N1 del Virus de la Influenza A/inmunología , Ratones , Ratones Endogámicos BALB C , Oligonucleótidos , Toxina del Pertussis/metabolismo
20.
Am J Pathol ; 183(4): 1258-1268, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23938324

RESUMEN

Pathological studies on fatal cases caused by 2009 pandemic influenza H1N1 virus (2009 pH1N1) reported extensive diffuse alveolar damage and virus infection predominantly in the lung parenchyma. However, the host immune response after severe 2009 pH1N1 infection is poorly understood. Herein, we investigated viral load, the immune response, and apoptosis in lung tissues from 50 fatal cases with 2009 pH1N1 virus infection. The results suggested that 7 of the 27 cytokines/chemokines showed remarkably high expression, including IL-1 receptor antagonist protein, IL-6, tumor necrosis factor-α, IL-8, monocyte chemoattractant protein-1, macrophage inflammatory protein 1-ß, and interferon-inducible protein-10 in lung tissues of 2009 pH1N1 fatal cases. Viral load, which showed the highest level on day 7 of illness onset and persisted until day 17 of illness, was positively correlated with mRNA levels of IL-1 receptor antagonist protein, monocyte chemoattractant protein-1, macrophage inflammatory protein 1-ß, interferon-inducible protein-10, and regulated on activation normal T-cell expressed and secreted. Apoptosis was evident in lung tissues stained by the TUNEL assay. Decreased Fas and elevated FasL mRNA levels were present in lung tissues, and cleaved caspase-3 was frequently seen in pneumocytes, submucosal glands, and lymphoid tissues. The pathogenesis of the 2009 pH1N1 virus infection is associated with viral replication and production of proinflammatory mediators. FasL and caspase-3 are involved in the pathway of 2009 pH1N1 virus-induced apoptosis in lung tissues, and the disequilibrium between the Fas and FasL level in lung tissues could contribute to delayed clearance of the virus and subsequent pathological damages.


Asunto(s)
Quimiocinas/metabolismo , Interacciones Huésped-Patógeno/inmunología , Inmunidad/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Pulmón/inmunología , Pandemias , Adolescente , Adulto , Anciano , Apoptosis/genética , Caspasa 3/metabolismo , Quimiocinas/genética , Niño , Preescolar , Demografía , Femenino , Regulación de la Expresión Génica , Humanos , Lactante , Gripe Humana/epidemiología , Gripe Humana/patología , Gripe Humana/virología , Pulmón/enzimología , Pulmón/patología , Pulmón/virología , Masculino , Persona de Mediana Edad , ARN Mensajero/genética , ARN Mensajero/metabolismo , Carga Viral , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA