Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Expert Rev Vaccines ; 21(11): 1637-1646, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36222056

RESUMEN

INTRODUCTION: Immunization is the best strategy to protect individuals from invasive meningococcal disease (IMD). To support decision-making around immunization, this paper considers what has led four countries and regions of two more to introduce the quadrivalent MenACWY vaccine in toddlers (ages 12-24 months). AREAS COVERED: A narrative literature review was conducted to identify countries that have introduced a MenACWY vaccination program for toddlers. Information from peer-reviewed publications, reports, and policy documents for each identified country was extracted. Australia, Chile, the Netherlands, Switzerland, and regions of Italy and Spain have introduced the MenACWY vaccine in their toddler programs, driven by the rising incidence of MenW and MenY and the vaccine's ability to provide protection against other serogroups. Australia and the Netherlands considered the economic impacts of implementing a MenACWY toddler vaccination program. Vaccination uptake and effects are reported for three countries; however, in two, isolating the vaccine's effect from the collateral effect of COVID-related measures is difficult. EXPERT OPINION: Increased convergence of vaccination policies and programs is needed internationally, as IMD recognizes no borders.PL AIN LANGUAGE SUMMARYVaccination is the best defense against meningitis, a deadly disease. While someone of any age can contract it, children 0-24 months of age are disproportionately affected. The increasing number of cases of meningitis has led four countries plus regions of two more to introduce into their vaccination schedules for toddlers (ages 12-24 months) a vaccine that protects against four different serogroups rather than one serogroup alone. This paper considers what has driven that shift.


Asunto(s)
COVID-19 , Infecciones Meningocócicas , Vacunas Meningococicas , Preescolar , Humanos , Lactante , Infecciones Meningocócicas/epidemiología , Infecciones Meningocócicas/prevención & control , Políticas , Vacunas Conjugadas
2.
J Virol ; 85(9): 4309-17, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21325424

RESUMEN

Marburgviruses are zoonotic pathogens that cause lethal hemorrhagic fever in humans and nonhuman primates. However, they do not cause lethal disease in immunocompetent mice unless they are adapted to this species. The adaptation process can therefore provide insight into the specific virus-host interactions that determine virulence. In primate cells, the Lake Victoria marburgvirus Musoke strain (MARV) VP40 matrix protein antagonizes alpha/beta interferon (IFN-α/ß) and IFN-γ signaling by inhibiting the activation of the cellular tyrosine kinase Jak1. Here, VP40 from the Ravn strain (RAVV VP40)-from a distinct Marburg virus clade-is demonstrated to also inhibit IFN signaling in human cells. However, neither MARV nor RAVV VP40 effectively inhibited IFN-signaling in mouse cells, as assessed by assays of the antiviral effects of IFN-α/ß and the IFN-α/ß-induced phosphorylation of Jak1, STAT1, and STAT2. In contrast, the VP40 from a mouse-adapted RAVV (maRAVV) did inhibit IFN signaling. Effective Jak1 inhibition correlated with the species from which the cells were derived and did not depend upon whether Jak1 was of human or mouse origin. Of the seven amino acid changes that accumulated in VP40 during mouse adaptation, two (V57A and T165A) are sufficient to allow efficient IFN signaling antagonism by RAVV VP40 in mouse cells. The same two changes also confer efficient IFN antagonist function upon MARV VP40 in mouse cells. The mouse-adaptive changes did not affect the budding of RAVV VP40 in mouse cells, suggesting that this second major function of VP40 did not undergo adaptation. These data identify an apparent determinant of RAVV host range and virulence and define specific genetic determinants of this function.


Asunto(s)
Interferón-alfa/antagonistas & inhibidores , Interferón beta/antagonistas & inhibidores , Interferón gamma/antagonistas & inhibidores , Marburgvirus/patogenicidad , Transducción de Señal , Proteínas de la Matriz Viral/metabolismo , Animales , Línea Celular , Humanos , Janus Quinasa 1/metabolismo , Ratones , Fosforilación , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT2/metabolismo , Proteínas de la Matriz Viral/inmunología
3.
Immunol Cell Biol ; 89(7): 792-802, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21263462

RESUMEN

Ebolaviruses naturally infect a wide variety of cells including macrophages and dendritic cells (DCs), and the resulting cytokine and interferon-α/ß (IFN) responses of infected cells are thought to influence viral pathogenesis. The VP35 protein impairs RIG-I-like receptor-dependent signaling to inhibit IFN production, and this function has been suggested to promote the ineffective host immune response characteristic of ebolavirus infection. To assess the impact of VP35 on innate immunity in biologically relevant primary cells, we used a recombinant Newcastle disease virus encoding VP35 (NDV/VP35) to infect macrophages and conventional DCs, which primarily respond to RNA virus infection via RIG-I-like pathways. VP35 suppressed not only IFN but also tumor necrosis factor (TNF)-α secretion, which are normally produced from these cells upon NDV infection. Additionally, in cells susceptible to the activity of VP35, IRF7 activation is impaired. In contrast, NDV/VP35 infection of plasmacytoid DCs, which activate IRF7 and produce IFN through TLR-dependent signaling, leads to robust IFN production. When plasmacytoid DCs deficient for TLR signaling were infected, NDV/VP35 was able to inhibit IFN production. Consistent with this, VP35 was less able to inhibit TLR-dependent versus RIG-I-dependent signaling in vitro. These data demonstrate that ebolavirus VP35 suppresses both IFN and cytokine production in multiple primary human cell types. However, cells that utilize the TLR pathway can circumvent this inhibition, suggesting that the presence of multiple viral sensors enables the host to overcome viral immune evasion mechanisms.


Asunto(s)
Células Dendríticas/inmunología , Ebolavirus/patogenicidad , Fiebre Hemorrágica Ebola/inmunología , Interferones/biosíntesis , Proteínas Reguladoras y Accesorias Virales/metabolismo , Animales , Línea Celular , Chlorocebus aethiops , Proteína 58 DEAD Box , ARN Helicasas DEAD-box , Células Dendríticas/metabolismo , Células Dendríticas/virología , Ebolavirus/genética , Ebolavirus/inmunología , Fiebre Hemorrágica Ebola/metabolismo , Humanos , Inmunidad Innata , Factor 7 Regulador del Interferón/antagonistas & inhibidores , Factor 7 Regulador del Interferón/inmunología , Factor 7 Regulador del Interferón/metabolismo , Macrófagos/virología , Ratones , Virus de la Enfermedad de Newcastle/genética , Receptores Inmunológicos , Transducción de Señal , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis , Células Vero , Proteínas Reguladoras y Accesorias Virales/genética , Proteínas Reguladoras y Accesorias Virales/inmunología
4.
Bioorg Chem ; 38(6): 247-51, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20926112

RESUMEN

Spiro[piperidine-2,2'-adamantane] 4 is one of the most potent synthetic anti-influenza A aminoadamantanes or other cage structure amines tested so far. Based on previous results Tataridis et al. (2007) [5h] which demonstrate the boost of in vitro potency by the presence of an additional amino group, we examined whether the incorporation of a second amino group into this heterocycle would increase the anti-influenza A virus activity. The new synthetic molecules 5-7 are capable of forming two hydrogen bonds within the receptor. We identified the diamino derivatives 5 and 6, which are active against influenza A H3N2 virus although less potent than amantadine and its equipotent spiropiperidine 4.


Asunto(s)
Adamantano/análogos & derivados , Adamantano/farmacología , Antivirales/química , Antivirales/farmacología , Subtipo H3N2 del Virus de la Influenza A/efectos de los fármacos , Piperazinas/química , Piperazinas/farmacología , Adamantano/síntesis química , Animales , Antivirales/síntesis química , Línea Celular , Humanos , Gripe Humana/tratamiento farmacológico , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Piperazinas/síntesis química , Compuestos de Espiro/síntesis química , Compuestos de Espiro/química , Compuestos de Espiro/farmacología
5.
PLoS Pathog ; 6(1): e1000721, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20084112

RESUMEN

Previous studies have demonstrated that Marburg viruses (MARV) and Ebola viruses (EBOV) inhibit interferon (IFN)-alpha/beta signaling but utilize different mechanisms. EBOV inhibits IFN signaling via its VP24 protein which blocks the nuclear accumulation of tyrosine phosphorylated STAT1. In contrast, MARV infection inhibits IFNalpha/beta induced tyrosine phosphorylation of STAT1 and STAT2. MARV infection is now demonstrated to inhibit not only IFNalpha/beta but also IFNgamma-induced STAT phosphorylation and to inhibit the IFNalpha/beta and IFNgamma-induced tyrosine phosphorylation of upstream Janus (Jak) family kinases. Surprisingly, the MARV matrix protein VP40, not the MARV VP24 protein, has been identified to antagonize Jak and STAT tyrosine phosphorylation, to inhibit IFNalpha/beta or IFNgamma-induced gene expression and to inhibit the induction of an antiviral state by IFNalpha/beta. Global loss of STAT and Jak tyrosine phosphorylation in response to both IFNalpha/beta and IFNgamma is reminiscent of the phenotype seen in Jak1-null cells. Consistent with this model, MARV infection and MARV VP40 expression also inhibit the Jak1-dependent, IL-6-induced tyrosine phosphorylation of STAT1 and STAT3. Finally, expression of MARV VP40 is able to prevent the tyrosine phosphorylation of Jak1, STAT1, STAT2 or STAT3 which occurs following over-expression of the Jak1 kinase. In contrast, MARV VP40 does not detectably inhibit the tyrosine phosphorylation of STAT2 or Tyk2 when Tyk2 is over-expressed. Mutation of the VP40 late domain, essential for efficient VP40 budding, has no detectable impact on inhibition of IFN signaling. This study shows that MARV inhibits IFN signaling by a mechanism different from that employed by the related EBOV. It identifies a novel function for the MARV VP40 protein and suggests that MARV may globally inhibit Jak1-dependent cytokine signaling.


Asunto(s)
Evasión Inmune/inmunología , Interferones/inmunología , Marburgvirus/inmunología , Transducción de Señal/inmunología , Proteínas de la Matriz Viral/inmunología , Animales , Western Blotting , Línea Celular , Citocinas/inmunología , Ebolavirus/inmunología , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Fiebre Hemorrágica Ebola/inmunología , Humanos , Janus Quinasa 1/inmunología , Janus Quinasa 1/metabolismo , Enfermedad del Virus de Marburg/inmunología , Enfermedad del Virus de Marburg/metabolismo , Marburgvirus/metabolismo , Reacción en Cadena de la Polimerasa , Transfección
6.
J Exp Bot ; 58(13): 3513-24, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18057034

RESUMEN

Stochastic processes are intrinsic phenomena that perturb developmental processes. However, the canalization process restricts the magnitude of perturbation and hence the magnitude of morphological variation during development. Heat-shock protein 90 (Hsp90) chaperones are a class of proteins stabilizing a network of 'client' proteins that are involved in diverse signal transduction pathways affecting development. Here it is reported that a reduction of Hsp90 gene dose creates canalization perturbations that affect many aspects of Arabidopsis development and results in a plethora of morphological alterations. Hence, Hsp90 restricts stochastic phenomena by minimizing perturbations, thereby canalizing development. It is also shown that morphogenesis is determined by three mutually inter-related parameters: genotype, environment, and time. Hsp90 is involved in the interaction of these three parameters which ultimately affect developmental processes. The amount of phenotypic variation upon the reduction of Hsp90 function could be perceived as adaptive and could have an impact on the evolutionary process.


Asunto(s)
Arabidopsis/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Arabidopsis/embriología , Arabidopsis/genética , Regulación de la Expresión Génica de las Plantas , Proteínas HSP90 de Choque Térmico/genética , Calor , Mutación , Factor de Transcripción STAT1 , Procesos Estocásticos
7.
J Virol ; 81(24): 13469-77, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17928350

RESUMEN

The Zaire ebolavirus protein VP24 was previously demonstrated to inhibit alpha/beta interferon (IFN-alpha/beta)- and IFN-gamma-induced nuclear accumulation of tyrosine-phosphorylated STAT1 (PY-STAT1) and to inhibit IFN-alpha/beta- and IFN-gamma-induced gene expression. These properties correlated with the ability of VP24 to interact with the nuclear localization signal receptor for PY-STAT1, karyopherin alpha1. Here, VP24 is demonstrated to interact not only with overexpressed but also with endogenous karyopherin alpha1. Mutational analysis demonstrated that VP24 binds within the PY-STAT1 binding region located in the C terminus of karyopherin alpha1. In addition, VP24 was found to inhibit PY-STAT1 binding to both overexpressed and endogenous karyopherin alpha1. We assessed the binding of both PY-STAT1 and the VP24 proteins from Zaire, mouse-adapted Zaire, and Reston Ebola viruses for interaction with all six members of the human karyopherin alpha family. We found, in contrast to previous studies, that PY-STAT1 can interact not only with karyopherin alpha1 but also with karyopherins alpha5 and alpha6, which together comprise the NPI-1 subfamily of karyopherin alphaS. Similarly, all three VP24s bound and inhibited PY-STAT1 interaction with karyopherins alpha1, alpha5, and alpha6. Consistent with their ability to inhibit the karyopherin-PY-STAT1 interaction, Zaire, mouse-adapted Zaire, and Reston Ebola virus VP24s displayed similar capacities to inhibit IFN-beta-induced gene expression in human and mouse cells. These findings suggest that VP24 inhibits interaction of PY-STAT1 with karyopherins alpha1, alpha5, or alpha6 by binding within the PY-STAT1 binding region of the karyopherins and that this function is conserved among the VP24 proteins of different Ebola virus species.


Asunto(s)
Ebolavirus/metabolismo , Factor de Transcripción STAT1/efectos de los fármacos , Factor de Transcripción STAT1/metabolismo , Proteínas Virales/metabolismo , alfa Carioferinas/efectos de los fármacos , alfa Carioferinas/metabolismo , Animales , Línea Celular , Ebolavirus/genética , Genes Reporteros , Humanos , Ratones , Células 3T3 NIH , Factor de Transcripción STAT1/genética , Transfección , Proteínas Virales/genética , Proteínas Virales/farmacología , alfa Carioferinas/genética
8.
Virology ; 364(2): 342-54, 2007 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-17434557

RESUMEN

Dendritic cells (DCs), important early targets of Ebola virus (EBOV) infection in vivo, are activated by Ebola virus-like particles (VLPs). To better understand this phenomenon, we have systematically assessed the response of DCs to VLPs of different compositions. VLPs containing the viral matrix protein (VP40) and the viral glycoprotein (GP), were found to induce a proinflammatory response highly similar to a prototypical DC activator, LPS. This response included the production of several proinflammatory cytokines, activation of numerous transcription factors including NF-kappaB, the functional importance of which was demonstrated by employing inhibitors of NF-kappaB activation, and activation of ERK1/2 MAP kinase. In contrast, VLPs constituted with a mutant GP lacking the heavily glycosylated mucin domain showed impaired NF-kappaB and Erk activation and induced less DC cytokine production. We conclude that the GP mucin domain is required for VLPs to stimulate human dendritic cells through NF-kappaB and MAPK signaling pathways.


Asunto(s)
Células Dendríticas/metabolismo , Células Dendríticas/virología , Ebolavirus/patogenicidad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , FN-kappa B/metabolismo , Proteínas Virales/fisiología , Línea Celular , Células Cultivadas , Citocinas/biosíntesis , Células Dendríticas/inmunología , Ebolavirus/genética , Ebolavirus/fisiología , Ebolavirus/ultraestructura , Humanos , Mediadores de Inflamación/metabolismo , Interleucina-6/biosíntesis , Sistema de Señalización de MAP Quinasas , Microscopía Electrónica , Mucinas/química , Mucinas/genética , Estructura Terciaria de Proteína , Factores de Transcripción/metabolismo , Transfección , Proteínas Virales/química , Proteínas Virales/genética , Virión/genética , Virión/patogenicidad , Virión/fisiología , Virión/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA