Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Immunol ; 202(12): 3370-3380, 2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-31092636

RESUMEN

The importance of antiviral CD8+ T cell recognition of alternative reading frame (ARF)-derived peptides is uncertain. In this study, we describe an epitope (NS1-ARF21-8) present in a predicted 14-residue peptide encoded by the +1 register of NS1 mRNA in the influenza A virus (IAV). NS1-ARF21-8 elicits a robust, highly functional CD8+ T cell response in IAV-infected BALB/c mice. NS1-ARF21-8 is presented from unspliced NS mRNA, likely from downstream initiation on a Met residue that comprises the P1 position of NS1-ARF21-8 Derived from a 14-residue peptide with no apparent biological function and negligible impacts on IAV infection, infectivity, and pathogenicity, NS1-ARF21-8 provides a clear demonstration of how immunosurveillance exploits natural errors in protein translation to provide antiviral immunity. We further show that IAV infection enhances a model cellular ARF translation, which potentially has important implications for virus-induced autoimmunity.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/metabolismo , Virus de la Influenza A/fisiología , Gripe Humana/inmunología , Infecciones por Orthomyxoviridae/inmunología , Proteínas no Estructurales Virales/metabolismo , Empalme Alternativo , Animales , Modelos Animales de Enfermedad , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/inmunología , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Vigilancia Inmunológica , Ratones , Ratones Endogámicos BALB C , Sistemas de Lectura Abierta/genética , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/inmunología
2.
J Virol ; 92(4)2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29187546

RESUMEN

Small-animal models have been used to obtain many insights regarding the pathogenesis and immune responses induced following infection with human respiratory syncytial virus (hRSV). Among those described to date, infections in cotton rats, mice, guinea pigs, chinchillas, and Syrian hamsters with hRSV strains Long and/or A2 have been well characterized, although clinical isolates have also been examined. Ferrets are also susceptible to hRSV infection, but the pathogenesis and immune responses elicited following infection have not been well characterized. Here, we describe the infection of adult ferrets with hRSV Long or A2 via the intranasal route and characterized virus replication, as well as cytokine induction, in the upper and lower airways. Virus replication and cytokine induction during the acute phase of infection (days 0 to 15 postinfection) were similar between the two strains, and both elicited high levels of F glycoprotein-specific binding and neutralizing antibodies following virus clearance (days 16 to 22 postinfection). Importantly, we demonstrate transmission from experimentally infected donor ferrets to cohoused naive recipients and have characterized virus replication and cytokine induction in the upper airways of infected contact animals. Together, these studies provide a direct comparison of the pathogenesis of hRSV Long and A2 in ferrets and highlight the potential of this animal model to study serological responses and examine interventions that limit transmission of hRSV.IMPORTANCE Ferrets have been widely used to study pathogenesis, immunity, and transmission following human influenza virus infections; however, far less is known regarding the utility of the ferret model to study hRSV infections. Following intranasal infection of adult ferrets with the well-characterized Long or A2 strain of hRSV, we report virus replication and cytokine induction in the upper and lower airways, as well as the development of virus-specific humoral responses. Importantly, we demonstrate transmission of hRSV from experimentally infected donor ferrets to cohoused naive recipients. Together, these findings significantly enhance our understanding of the utility of the ferret as a small-animal model to investigate aspects of hRSV pathogenesis and immunity.


Asunto(s)
Modelos Animales de Enfermedad , Inmunidad Humoral/inmunología , Infecciones por Virus Sincitial Respiratorio/transmisión , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/patogenicidad , Infecciones del Sistema Respiratorio/virología , Animales , Hurones , Células HeLa , Humanos , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/patología , Virus Sincitial Respiratorio Humano/inmunología , Infecciones del Sistema Respiratorio/inmunología , Carga Viral , Replicación Viral
3.
J Infect Dis ; 218(3): 406-417, 2018 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-29746640

RESUMEN

Epidemiological studies have observed that the seasonal peak incidence of influenza virus infection is sometimes separate from the peak incidence of human respiratory syncytial virus (hRSV) infection, with the peak incidence of hRSV infection delayed. This is proposed to be due to viral interference, whereby infection with one virus prevents or delays infection with a different virus. We investigated viral interference between hRSV and 2009 pandemic influenza A(H1N1) virus (A[H1N1]pdm09) in the ferret model. Infection with A(H1N1)pdm09 prevented subsequent infection with hRSV. Infection with hRSV reduced morbidity attributed to infection with A(H1N1)pdm09 but not infection, even when an increased inoculum dose of hRSV was used. Notably, infection with A(H1N1)pdm09 induced higher levels of proinflammatory cytokines, chemokines, and immune mediators in the ferret than hRSV. Minimal cross-reactive serological responses or interferon γ-expressing cells were induced by either virus ≥14 days after infection. These data indicate that antigen-independent mechanisms may drive viral interference between unrelated respiratory viruses that can limit subsequent infection or disease.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/crecimiento & desarrollo , Infecciones por Orthomyxoviridae/virología , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/crecimiento & desarrollo , Interferencia Viral , Animales , Anticuerpos Antivirales , Modelos Animales de Enfermedad , Hurones , Inmunidad Celular , Inmunidad Humoral , Interferón gamma/análisis , Leucocitos Mononucleares/inmunología , Infecciones por Orthomyxoviridae/patología , Infecciones por Virus Sincitial Respiratorio/patología , Análisis de Supervivencia
4.
J Infect Dis ; 217(4): 548-559, 2018 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-29325138

RESUMEN

Background: Two influenza B virus lineages, B/Victoria and B/Yamagata, cocirculate in the human population. While the lineages are serologically distinct, cross-reactive responses to both lineages have been detected. Viral interference describes the situation whereby infection with one virus limits infection and replication of a second virus. We investigated the potential for viral interference between the influenza B virus lineages. Methods: Ferrets were infected and then challenged 3, 10, or 28 days later with pairs of influenza B/Victoria and B/Yamagata viruses. Results: Viral interference occurred at challenge intervals of 3 and 10 days and occasionally at 28 days. At the longer interval, shedding of challenge virus was reduced, and this correlated with cross-reactive interferon γ responses from lymph nodes from virus-infected animals. Viruses from both lineages could prevent or significantly limit subsequent infection with a virus from the other lineage. Coinfections were rare, indicating the potential for reassortment between lineages is limited. Conclusions: These data suggest that innate and cross-reactive immunity mediate viral interference and that this may contribute to the dominance of a specific influenza B virus lineage in any given influenza season. Furthermore, infection with one influenza B virus lineage may be beneficial in protecting against subsequent infection with either influenza B virus lineage.


Asunto(s)
Protección Cruzada , Virus de la Influenza B/inmunología , Virus de la Influenza B/fisiología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Interferencia Viral , Animales , Reacciones Cruzadas , Modelos Animales de Enfermedad , Hurones , Inmunidad Innata
5.
J Virol ; 90(12): 5724-5734, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27053553

RESUMEN

UNLABELLED: This study seeks to assess the ability of seasonal trivalent inactivated influenza vaccine (TIV) to induce nonneutralizing antibodies (Abs) with Fc-mediated functions in HIV-uninfected and HIV-infected subjects. Functional influenza-specific Ab responses were studied in 30 HIV-negative and 27 HIV-positive subjects immunized against seasonal influenza. All 57 subjects received the 2015 TIV. Fc-mediated antihemagglutinin (anti-HA) Ab activity was measured in plasma before and 4 weeks after vaccination using Fc-receptor-binding assays, NK cell activation assays, and phagocytosis assays. At baseline, the HIV-positive group had detectable but reduced functional Ab responses to both vaccine and nonvaccine influenza antigens. TIV enhanced Fc-mediated Ab responses in both HIV-positive and HIV-negative groups. A larger rise was generally observed in the HIV-positive group, such that there was no difference in functional Ab responses between the two groups after vaccination. The 2015 TIV enhanced functional influenza-specific Ab responses in both HIV-negative and HIV-positive subjects to a range of influenza HA proteins. The increase in functional Ab responses in the HIV-positive group supports recommendations to immunize this at-risk group. IMPORTANCE: Infection with HIV is associated with increasing disease severity following influenza infections, and annual influenza vaccinations are recommended for this target group. However, HIV-infected individuals respond relatively poorly to vaccination compared to healthy individuals, particularly if immunodeficient. There is therefore a need to increase our understanding of immunity to influenza in the context of underlying HIV infection. While antibodies can mediate direct virus neutralization, interactions with cellular Fc receptors may be important for anti-influenza immunity in vivo by facilitating antibody-dependent cellular cytotoxicity (ADCC) and/or antibody-dependent phagocytosis (ADP). The ability of seasonal influenza vaccines to induce antibody responses with potent Fc-mediated antiviral activity is currently unclear. Probing the ADCC and ADP responses to influenza vaccination has provided important new information in the quest to improve immunity to influenza.


Asunto(s)
Anticuerpos Antivirales/sangre , Infecciones por VIH/inmunología , Vacunas contra la Influenza/inmunología , Receptores Fc/inmunología , Adulto , Anticuerpos Antivirales/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Hemaglutininas/inmunología , Humanos , Subtipo H1N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/administración & dosificación , Masculino , Persona de Mediana Edad , Fagocitosis , Vacunación , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/inmunología , Adulto Joven
6.
J Theor Biol ; 413: 34-49, 2017 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-27856216

RESUMEN

The cellular adaptive immune response plays a key role in resolving influenza infection. Experiments where individuals are successively infected with different strains within a short timeframe provide insight into the underlying viral dynamics and the role of a cross-reactive immune response in resolving an acute infection. We construct a mathematical model of within-host influenza viral dynamics including three possible factors which determine the strength of the cross-reactive cellular adaptive immune response: the initial naive T cell number, the avidity of the interaction between T cells and the epitopes presented by infected cells, and the epitope abundance per infected cell. Our model explains the experimentally observed shortening of a second infection when cross-reactivity is present, and shows that memory in the cellular adaptive immune response is necessary to protect against a second infection.


Asunto(s)
Inmunidad Adaptativa , Reacciones Cruzadas/inmunología , Interacciones Huésped-Patógeno/inmunología , Inmunidad Celular , Memoria Inmunológica , Gripe Humana/inmunología , Modelos Inmunológicos , Linfocitos T CD8-positivos/inmunología , Epítopos/inmunología , Humanos , Carga Viral/inmunología
7.
J Virol ; 90(6): 2838-48, 2015 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-26719259

RESUMEN

UNLABELLED: The burden of infection with seasonal influenza viruses is significant. Each year is typically characterized by the dominance of one (sub)type or lineage of influenza A or B virus, respectively. The incidence of disease varies annually, and while this may be attributed to a particular virus strain or subtype, the impacts of prior immunity, population differences, and variations in clinical assessment are also important. To improve our understanding of the impacts of seasonal influenza viruses, we directly compared clinical symptoms, virus shedding, and expression of cytokines, chemokines, and immune mediators in the upper respiratory tract (URT) of ferrets infected with contemporary A(H1N1)pdm09, A(H3N2), or influenza B virus. Gene expression in the lower respiratory tract (LRT) was also assessed. Clinical symptoms were minimal. Overall cytokine/chemokine profiles in the URT were consistent in pattern and magnitude between animals infected with influenza A and B viruses, and peak expression levels of interleukin-1α (IL-1α), IL-1ß, IL-6, IL-12p40, alpha interferon (IFN-α), IFN-ß, and tumor necrosis factor alpha (TNF-α) mRNAs correlated with peak levels of viral shedding. MCP1 and IFN-γ were expressed after the virus peak. Granzymes A and B and IL-10 reached peak expression as the virus was cleared and seroconversion was detected. Cytokine/chemokine gene expression in the LRT following A(H1N1)pdm09 virus infection reflected the observations seen for the URT but was delayed 2 or 3 days, as was virus replication. These data indicate that disease severities and localized immune responses following infection with seasonal influenza A and B viruses are similar, suggesting that other factors are likely to modulate the incidence and impact of seasonal influenza. IMPORTANCE: Both influenza A and B viruses cocirculate in the human population, and annual influenza seasons are typically dominated by an influenza A virus subtype or an influenza B virus lineage. Surveillance data indicate that the burden of disease is higher in some seasons, yet it is unclear whether this is due to specific virus strains or to other factors, such as cross-reactive immunity or clinical definitions of influenza. We directly compared disease severities and localized inflammatory responses to different seasonal influenza virus strains, including the 2009 pandemic strain, in healthy naive ferrets. We found that the disease severities and the cytokine and chemokine responses were similar irrespective of the seasonal strain or the location of the infection in the respiratory tract. This suggests that factors other than the immune response to a particular virus (sub)type contribute to the variable impact of influenza virus infection in a population.


Asunto(s)
Virus de la Influenza A/inmunología , Virus de la Influenza B/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/patología , Sistema Respiratorio/inmunología , Sistema Respiratorio/patología , Animales , Temperatura Corporal , Peso Corporal , Citocinas/análisis , Modelos Animales de Enfermedad , Femenino , Hurones , Perfilación de la Expresión Génica , Humanos , Masculino , Infecciones por Orthomyxoviridae/virología , Sistema Respiratorio/virología , Índice de Severidad de la Enfermedad , Carga Viral , Esparcimiento de Virus
8.
PLoS Comput Biol ; 11(8): e1004334, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26284917

RESUMEN

Influenza is an infectious disease that primarily attacks the respiratory system. Innate immunity provides both a very early defense to influenza virus invasion and an effective control of viral growth. Previous modelling studies of virus-innate immune response interactions have focused on infection with a single virus and, while improving our understanding of viral and immune dynamics, have been unable to effectively evaluate the relative feasibility of different hypothesised mechanisms of antiviral immunity. In recent experiments, we have applied consecutive exposures to different virus strains in a ferret model, and demonstrated that viruses differed in their ability to induce a state of temporary immunity or viral interference capable of modifying the infection kinetics of the subsequent exposure. These results imply that virus-induced early immune responses may be responsible for the observed viral hierarchy. Here we introduce and analyse a family of within-host models of re-infection viral kinetics which allow for different viruses to stimulate the innate immune response to different degrees. The proposed models differ in their hypothesised mechanisms of action of the non-specific innate immune response. We compare these alternative models in terms of their abilities to reproduce the re-exposure data. Our results show that 1) a model with viral control mediated solely by a virus-resistant state, as commonly considered in the literature, is not able to reproduce the observed viral hierarchy; 2) the synchronised and desynchronised behaviour of consecutive virus infections is highly dependent upon the interval between primary virus and challenge virus exposures and is consistent with virus-dependent stimulation of the innate immune response. Our study provides the first mechanistic explanation for the recently observed influenza viral hierarchies and demonstrates the importance of understanding the host response to multi-strain viral infections. Re-exposure experiments provide a new paradigm in which to study the immune response to influenza and its role in viral control.


Asunto(s)
Inmunidad Innata/inmunología , Gripe Humana , Infecciones por Orthomyxoviridae , Orthomyxoviridae , Animales , Biología Computacional , Modelos Animales de Enfermedad , Hurones , Interacciones Huésped-Patógeno/inmunología , Humanos , Gripe Humana/inmunología , Gripe Humana/virología , Modelos Inmunológicos , Orthomyxoviridae/inmunología , Orthomyxoviridae/patogenicidad , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Carga Viral
9.
J Infect Dis ; 212(11): 1701-10, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25943206

RESUMEN

BACKGROUND: Epidemiological studies suggest that, following infection with influenza virus, there is a short period during which a host experiences a lower susceptibility to infection with other influenza viruses. This viral interference appears to be independent of any antigenic similarities between the viruses. We used the ferret model of human influenza to systematically investigate viral interference. METHODS: Ferrets were first infected then challenged 1-14 days later with pairs of influenza A(H1N1)pdm09, influenza A(H3N2), and influenza B viruses circulating in 2009 and 2010. RESULTS: Viral interference was observed when the interval between initiation of primary infection and subsequent challenge was <1 week. This effect was virus specific and occurred between antigenically related and unrelated viruses. Coinfections occurred when 1 or 3 days separated infections. Ongoing shedding from the primary virus infection was associated with viral interference after the secondary challenge. CONCLUSIONS: The interval between infections and the sequential combination of viruses were important determinants of viral interference. The influenza viruses in this study appear to have an ordered hierarchy according to their ability to block or delay infection, which may contribute to the dominance of different viruses often seen in an influenza season.


Asunto(s)
Modelos Animales de Enfermedad , Gripe Humana/inmunología , Gripe Humana/virología , Orthomyxoviridae/inmunología , Interferencia Viral/inmunología , Animales , Coinfección , Hurones , Humanos , Esparcimiento de Virus
10.
PLoS Pathog ; 9(5): e1003354, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23671418

RESUMEN

Surveillance data indicate that most circulating A(H1N1)pdm09 influenza viruses have remained antigenically similar since they emerged in humans in 2009. However, antigenic drift is likely to occur in the future in response to increasing population immunity induced by infection or vaccination. In this study, sequential passaging of A(H1N1)pdm09 virus by contact transmission through two independent series of suboptimally vaccinated ferrets resulted in selection of variant viruses with an amino acid substitution (N156K, H1 numbering without signal peptide; N159K, H3 numbering without signal peptide; N173K, H1 numbering from first methionine) in a known antigenic site of the viral HA. The N156K HA variant replicated and transmitted efficiently between naïve ferrets and outgrew wildtype virus in vivo in ferrets in the presence and absence of immune pressure. In vitro, in a range of cell culture systems, the N156K variant rapidly adapted, acquiring additional mutations in the viral HA that also potentially affected antigenic properties. The N156K escape mutant was antigenically distinct from wildtype virus as shown by binding of HA-specific antibodies. Glycan binding assays demonstrated the N156K escape mutant had altered receptor binding preferences compared to wildtype virus, which was supported by computational modeling predictions. The N156K substitution, and culture adaptations, have been detected in human A(H1N1)pdm09 viruses with N156K preferentially reported in sequences from original clinical samples rather than cultured isolates. This study demonstrates the ability of the A(H1N1)pdm09 virus to undergo rapid antigenic change to evade a low level vaccine response, while remaining fit in a ferret transmission model of immunization and infection. Furthermore, the potential changes in receptor binding properties that accompany antigenic changes highlight the importance of routine characterization of clinical samples in human A(H1N1)pdm09 influenza surveillance.


Asunto(s)
Antígenos Virales , Flujo Genético , Subtipo H1N1 del Virus de la Influenza A , Gripe Humana , Mutación Missense/inmunología , Pandemias , Sustitución de Aminoácidos , Animales , Antígenos Virales/genética , Antígenos Virales/inmunología , Embrión de Pollo , Modelos Animales de Enfermedad , Perros , Femenino , Hurones , Humanos , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H1N1 del Virus de la Influenza A/inmunología , Gripe Humana/epidemiología , Gripe Humana/genética , Gripe Humana/inmunología , Células de Riñón Canino Madin Darby , Masculino
11.
J Infect Dis ; 210(11): 1811-22, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24916185

RESUMEN

BACKGROUND: Intravenous immunoglobulin (IVIG) is a purified pool of human antibodies from thousands of donors that is used to prevent or treat primary immune deficiency, several infectious diseases, and autoimmune diseases. The antibodies that mediate antibody-dependent cellular cytotoxicity (ADCC) against heterologous influenza strains may be present in IVIG preparations. METHODS: We tested 8 IVIG preparations prior to the 2009 H1N1 swine-origin influenza pandemic and 10 IVIG preparations made after 2010 for their ability to mediate influenza-specific ADCC. RESULTS: ADCC mediating antibodies to A(H1N1)pdm09 hemagglutinin (HA) and neuraminidase (NA) were detected in IVIG preparations prior to the 2009-H1N1 pandemic. The HA-specific ADCC targeted both the HA1 and HA2 regions of A(H1N1)pdm09 HA and was capable of recognizing a broad range of HA proteins including those from recent avian influenza strains A(H5N1) and A(H7N9). The low but detectable ADCC recognition of A(H7N9) was likely due to rare individuals in the population contributing cross-reactive antibodies to IVIG. CONCLUSIONS: IVIG preparations contain broadly cross-reactive ADCC mediating antibodies. IVIG may provide at least some level of protection for individuals at high risk of severe influenza disease, especially during influenza pandemics prior to the development of effective vaccines.


Asunto(s)
Anticuerpos Antivirales/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Reacciones Cruzadas/inmunología , Inmunoglobulinas Intravenosas/uso terapéutico , Subtipo H1N1 del Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Gripe Humana/terapia , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Antivirales/sangre , Femenino , Pruebas de Hemaglutinación , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Humanos , Subtipo H7N9 del Virus de la Influenza A/inmunología , Células Asesinas Naturales/inmunología , Masculino , Persona de Mediana Edad , Adulto Joven
12.
J Virol ; 87(24): 13706-18, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24109221

RESUMEN

Yearly vaccination with the trivalent inactivated influenza vaccine (TIV) is recommended, since current vaccines induce little cross neutralization to divergent influenza strains. Whether the TIV can induce antibody-dependent cellular cytotoxicity (ADCC) responses that can cross-recognize divergent influenza virus strains is unknown. We immunized 6 influenza-naive pigtail macaques twice with the 2011-2012 season TIV and then challenged the macaques, along with 12 control macaques, serially with H1N1 and H3N2 viruses. We measured ADCC responses in plasma to a panel of H1 and H3 hemagglutinin (HA) proteins and influenza virus-specific CD8 T cell (CTL) responses using a sensitive major histocompatibility complex (MHC) tetramer reagent. The TIV was weakly immunogenic and, although binding antibodies were detected by enzyme-linked immunosorbent assay (ELISA), did not induce detectable influenza virus-specific ADCC or CTL responses. The H1N1 challenge elicited robust ADCC to both homologous and heterologous H1 HA proteins, but not influenza virus HA proteins from different subtypes (H2 to H7). There was no anamnestic influenza virus-specific ADCC or CTL response in vaccinated animals. The subsequent H3N2 challenge did not induce or boost ADCC either to H1 HA proteins or to divergent H3 proteins but did boost CTL responses. ADCC or CTL responses were not induced by TIV vaccination in influenza-naive macaques. There was a marked difference in the ability of infection compared to that of vaccination to induce cross-reactive ADCC and CTL responses. Improved vaccination strategies are needed to induce broad-based ADCC immunity to influenza.


Asunto(s)
Anticuerpos Antivirales/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Vacunas contra la Influenza/inmunología , Gripe Humana/inmunología , Animales , Glicoproteínas Hemaglutininas del Virus de la Influenza/administración & dosificación , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Humanos , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H3N2 del Virus de la Influenza A/genética , Subtipo H3N2 del Virus de la Influenza A/inmunología , Virus de la Influenza B/genética , Virus de la Influenza B/inmunología , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/genética , Gripe Humana/prevención & control , Gripe Humana/virología , Macaca , Masculino , Vacunación
13.
J Infect Dis ; 208(7): 1051-61, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23812238

RESUMEN

BACKGROUND: During the 2009 pandemic of influenza A virus subtype H1N1 (A[H1N1]pdm09) infection, older individuals were partially protected from severe disease. It is not known whether preexisting antibodies with effector functions such as antibody-dependent cellular cytotoxicity (ADCC) contributed to the immunity observed. METHODS: We tested serum specimens obtained from 182 individuals aged 1-72 years that were collected either immediately before or after the A(H1N1)pdm09 pandemic for ADCC antibodies to the A(H1N1)pdm09 hemagglutinin (HA) protein. RESULTS: A(H1N1)pdm09 HA-specific ADCC antibodies were detected in almost all individuals aged >45 years (28/31 subjects) before the 2009 A(H1N1) pandemic. Conversely, only approximately half of the individuals aged 1-14 years (11/31) and 15-45 years (17/31) had cross-reactive ADCC antibodies before the 2009 A(H1N1) pandemic. The A(H1N1)pdm09-specific ADCC antibodies were able to efficiently mediate the killing of influenza virus-infected respiratory epithelial cells. Further, subjects >45 years of age had higher ADCC titers to a range of seasonal H1N1 HA proteins, including from the 1918 virus, compared with younger individuals. CONCLUSIONS: ADCC antibodies may have contributed to the protection exhibited in older individuals during the 2009 A(H1N1) pandemic. This work has significant implications for improved vaccination strategies for future influenza pandemics.


Asunto(s)
Anticuerpos Antivirales/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Adolescente , Adulto , Factores de Edad , Niño , Preescolar , Femenino , Humanos , Lactante , Gripe Humana/virología , Masculino , Persona de Mediana Edad , Adulto Joven
14.
Emerg Infect Dis ; 19(1): 92-101, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23260059

RESUMEN

To estimate population attack rates of influenza A(H1N1)pdm2009 in the Southern Hemisphere during June-August 2009, we conducted several serologic studies. We pooled individual-level data from studies using hemagglutination inhibition assays performed in Australia, New Zealand, and Singapore. We determined seropositive proportions (titer ≥40) for each study region by age-group and sex in pre- and postpandemic phases, as defined by jurisdictional notification data. After exclusions, the pooled database consisted of, 4,414 prepandemic assays and 7,715 postpandemic assays. In the prepandemic phase, older age groups showed greater seropositive proportions, with age-standardized, community-based proportions ranging from 3.5% in Singapore to 11.9% in New Zealand. In the postpandemic phase, seropositive proportions ranged from 17.5% in Singapore to 30.8% in New Zealand, with highest proportions seen in school-aged children. Pregnancy and residential care were associated with lower postpandemic seropositivity, whereas Aboriginal and Torres Strait Islander Australians and Pacific Peoples of New Zealand had greater postpandemic seropositivity.


Asunto(s)
Anticuerpos Antivirales/sangre , Subtipo H1N1 del Virus de la Influenza A/fisiología , Gripe Humana/epidemiología , Pandemias , Adolescente , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Anticuerpos Antivirales/inmunología , Australia/epidemiología , Niño , Preescolar , Femenino , Pruebas de Inhibición de Hemaglutinación , Humanos , Incidencia , Lactante , Gripe Humana/etnología , Gripe Humana/inmunología , Gripe Humana/virología , Masculino , Persona de Mediana Edad , Nativos de Hawái y Otras Islas del Pacífico , Nueva Zelanda/epidemiología , Embarazo , Estudios Seroepidemiológicos , Singapur/epidemiología
16.
J Infect Dis ; 202(7): 1011-20, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20715930

RESUMEN

BACKGROUND: An age bias toward children and young adults has been reported for infection and hospitalizations with pandemic H1N1 influenza (A[H1N1]pdm) in the 2009 and 2010 influenza seasons in the Southern and Northern Hemispheres. Serological analysis of prepandemic samples has shown a higher incidence of cross-reactive antibodies to A(H1N1)pdm virus in older populations; conserved T cell epitopes between viruses have been identified. The contribution of preexisting immunity to seasonal influenza to protection against A(H1N1)pdm infection was analyzed in a ferret model. METHODS: Ferrets were pre-infected with influenza A viruses and/or vaccinated with inactivated influenza viruses with adjuvant. Infection after challenge was assessed by measuring shedding virus, transmission to naive animals, and seroconversion. RESULTS: Homologous vaccination reduced the incidence of infection and delayed transmission. Pre-infection with virus induced sterilizing immunity to homologous challenge. One prior infection with seasonal influenza A virus improved clearance of A(H1N1)pdm virus. Prior infection with A(H1N1)pdm virus reduced shedding after seasonal influenza A challenge. Two infections with seasonal influenza A viruses reduced the incidence of infection, the amount and duration of virus shedding, and the frequency of transmission following A(H1N1)pdm challenge. CONCLUSION: These data suggest the reduced incidence and severity of infection with A(H1N1)pdm virus in the adult population during the 2009-2010 influenza season may be a result of previous exposure to seasonal influenza A viruses.


Asunto(s)
Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Protección Cruzada , Subtipo H1N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Animales , Hurones , Inmunización Secundaria/métodos , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Vacunación/métodos
17.
Influenza Other Respir Viruses ; 15(5): 573-576, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33955176

RESUMEN

The world has experienced five pandemics in just over one hundred years, four due to influenza and one due to coronavirus (SARS-CoV-2). In each case of pandemic influenza, the pandemic influenza strain has replaced the previous seasonal influenza virus. Notably, throughout the SARS-CoV-2 pandemic, there has been a 99% reduction in influenza isolation globally. It is anticipated that influenza will re-emerge following the SARS-CoV-2 pandemic and circulate again. The potential for which influenza viruses will emerge is examined.


Asunto(s)
COVID-19 , Gripe Humana , Orthomyxoviridae , Humanos , Gripe Humana/epidemiología , Gripe Humana/virología , Orthomyxoviridae/clasificación , Pandemias
18.
NPJ Vaccines ; 6(1): 149, 2021 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-34887440

RESUMEN

Influenza vaccines are utilised to combat seasonal and pandemic influenza. The key to influenza vaccination currently is the availability of candidate vaccine viruses (CVVs). Ideally, CVVs reflect the antigenic characteristics of the circulating virus, which may vary depending upon the isolation method. For traditional inactivated egg-based vaccines, CVVs are isolated in embryonated chicken eggs, while for cell-culture production, CVV's are isolated in either embryonated eggs or qualified cell lines. We compared isolation rates, growth characteristics, genetic stability and antigenicity of cell and egg CVV's derived from the same influenza-positive human clinical respiratory samples collected from 2008-2020. Influenza virus isolation rates in MDCK33016PF cells were twice that of eggs and mutations in the HA protein were common in egg CVVs but rare in cell CVVs. These results indicate that fully cell-based influenza vaccines will improve the choice, match and potentially the effectiveness, of seasonal influenza vaccines compared to egg-based vaccines.

19.
Vaccine ; 39(24): 3270-3278, 2021 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-33985853

RESUMEN

BACKGROUND: Epidemiological studies suggest that influenza vaccine effectiveness decreases with repeated administration. We examined antibody responses to influenza vaccination among healthcare workers (HCWs) by prior vaccination history and determined the incidence of influenza infection. METHODS: HCWs were vaccinated with the 2016 Southern Hemisphere quadrivalent influenza vaccine. Serum samples were collected pre-vaccination, 21-28 days and 7 months post-vaccination. Influenza antibody titres were measured at each time-point using the haemagglutination inhibition (HI) assay. Immunogenicity was compared by prior vaccination history. RESULTS: A total of 157 HCWs completed the study. The majority were frequently vaccinated, with only 5 reporting no prior vaccinations since 2011. Rises in titres for all vaccine strains among vaccine-naïve HCWs were significantly greater than rises observed for HCWs who received between 1 and 5 prior vaccinations (p < 0.001, respectively). Post-vaccination GMTs against influenza A but not B strains decreased as the number of prior vaccinations increased from 1 to 5. There was a significant decline in GMTs post-season for both B lineages. Sixty five (41%) HCWs reported at least one influenza-like illness episode, with 6 (4%) identified as influenza positive. CONCLUSIONS: Varying serological responses to influenza vaccination were observed among HCWs by prior vaccination history, with vaccine-naïve HCWs demonstrating greater post-vaccination responses against A(H3N2).


Asunto(s)
Vacunas contra la Influenza , Gripe Humana , Anticuerpos Antivirales , Formación de Anticuerpos , Australia/epidemiología , Personal de Salud , Humanos , Subtipo H3N2 del Virus de la Influenza A , Gripe Humana/prevención & control , Vacunación
20.
Nat Commun ; 12(1): 2691, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33976217

RESUMEN

How innate and adaptive immune responses work in concert to resolve influenza disease is yet to be fully investigated in one single study. Here, we utilize longitudinal samples from patients hospitalized with acute influenza to understand these immune responses. We report the dynamics of 18 important immune parameters, related to clinical, genetic and virological factors, in influenza patients across different severity levels. Influenza disease correlates with increases in IL-6/IL-8/MIP-1α/ß cytokines and lower antibody responses. Robust activation of circulating T follicular helper cells correlates with peak antibody-secreting cells and influenza heamaglutinin-specific memory B-cell numbers, which phenotypically differs from vaccination-induced B-cell responses. Numbers of influenza-specific CD8+ or CD4+ T cells increase early in disease and retain an activated phenotype during patient recovery. We report the characterisation of immune cellular networks underlying recovery from influenza infection which are highly relevant to other infectious diseases.


Asunto(s)
Formación de Anticuerpos/inmunología , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Citocinas/inmunología , Gripe Humana/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Estudios de Cohortes , Citocinas/metabolismo , Hospitalización/estadística & datos numéricos , Humanos , Virus de la Influenza A/clasificación , Virus de la Influenza A/genética , Virus de la Influenza A/fisiología , Vacunas contra la Influenza/inmunología , Gripe Humana/virología , Persona de Mediana Edad , Filogenia , Vacunación/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA