Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 12(2): 137-43, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21217758

RESUMEN

The 5' cap structures of higher eukaryote mRNAs have ribose 2'-O-methylation. Likewise, many viruses that replicate in the cytoplasm of eukaryotes have evolved 2'-O-methyltransferases to autonomously modify their mRNAs. However, a defined biological role for 2'-O-methylation of mRNA remains elusive. Here we show that 2'-O-methylation of viral mRNA was critically involved in subverting the induction of type I interferon. We demonstrate that human and mouse coronavirus mutants lacking 2'-O-methyltransferase activity induced higher expression of type I interferon and were highly sensitive to type I interferon. Notably, the induction of type I interferon by viruses deficient in 2'-O-methyltransferase was dependent on the cytoplasmic RNA sensor Mda5. This link between Mda5-mediated sensing of viral RNA and 2'-O-methylation of mRNA suggests that RNA modifications such as 2'-O-methylation provide a molecular signature for the discrimination of self and non-self mRNA.


Asunto(s)
Infecciones por Coronavirus/metabolismo , Coronavirus/fisiología , ARN Helicasas DEAD-box/metabolismo , Metiltransferasas/metabolismo , Proteínas Virales/metabolismo , Animales , Línea Celular , Coronavirus/patogenicidad , Infecciones por Coronavirus/genética , Infecciones por Coronavirus/inmunología , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/inmunología , Humanos , Inmunidad Innata/genética , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Helicasa Inducida por Interferón IFIH1 , Metilación , Metiltransferasas/genética , Metiltransferasas/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Viral/metabolismo , Receptor de Interferón alfa y beta/genética , Receptores de Reconocimiento de Patrones/genética , Ribosa/metabolismo , Proteínas Virales/genética , Proteínas Virales/inmunología , Virulencia/genética , Replicación Viral/genética
2.
Clin Infect Dis ; 72(4): 598-603, 2021 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-32594121

RESUMEN

BACKGROUND: Social distancing and stringent hygiene seem to be effective in reducing the number of transmitted virus particles, and therefore the infectivity, of coronavirus disease 2019 (COVID-19) and could alter the mode of transmission of the disease. However, it is not known if such practices can change the clinical course in infected individuals. METHODS: We prospectively studied an outbreak of COVID-19 in Switzerland among a population of 508 predominantly male soldiers with a median age of 21 years. We followed the number of infections in 2 spatially separated cohorts with almost identical baseline characteristics with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) before and after implementation of stringent social distancing. RESULTS: Of the 354 soldiers infected prior to the implementation of social distancing, 30% fell ill from COVID-19, while no soldier in a group of 154, in which infections appeared after implementation of social distancing, developed COVID-19 despite the detection of viral RNA in the nasal and virus-specific antibodies within this group. CONCLUSIONS: Social distancing not only can slow the spread of SARS-CoV-2 in a cohort of young, healthy adults but it can also prevent the outbreak of COVID-19 while still inducing an immune response and colonizing nasal passages. Viral inoculum during infection or mode of transmission may be a key factor determining the clinical course of COVID-19.


Asunto(s)
COVID-19 , Distanciamiento Físico , Adulto , Estudios de Cohortes , Humanos , Masculino , SARS-CoV-2 , Suiza/epidemiología , Adulto Joven
3.
Virol J ; 17(1): 136, 2020 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-32907596

RESUMEN

BACKGROUND: Coronaviruses (CoVs) were long thought to only cause mild respiratory and gastrointestinal symptoms in humans but outbreaks of Middle East Respiratory Syndrome (MERS)-CoV, Severe Acute Respiratory Syndrome (SARS)-CoV-1, and the recently identified SARS-CoV-2 have cemented their zoonotic potential and their capacity to cause serious morbidity and mortality, with case fatality rates ranging from 4 to 35%. Currently, no specific prophylaxis or treatment is available for CoV infections. Therefore we investigated the virucidal and antiviral potential of Echinacea purpurea (Echinaforce®) against human coronavirus (HCoV) 229E, highly pathogenic MERS- and SARS-CoVs, as well as the newly identified SARS-CoV-2, in vitro. METHODS: To evaluate the antiviral potential of the extract, we pre-treated virus particles and cells and evaluated remaining infectivity by limited dilution. Furthermore, we exposed cells to the extract after infection to further evaluate its potential as a prophylaxis and treatment against coronaviruses. We also determined the protective effect of Echinaforce® in re-constituted nasal epithelium. RESULTS: In the current study, we found that HCoV-229E was irreversibly inactivated when exposed to Echinaforce® at 3.2 µg/ml IC50. Pre-treatment of cell lines, however, did not inhibit infection with HCoV-229E and post-infection treatment had only a marginal effect on virus propagation at 50 µg/ml. However, we did observe a protective effect in an organotypic respiratory cell culture system by exposing pre-treated respiratory epithelium to droplets of HCoV-229E, imitating a natural infection. The observed virucidal activity of Echinaforce® was not restricted to common cold coronaviruses, as both SARS-CoV-1 and MERS-CoVs were inactivated at comparable concentrations. Finally, the causative agent of COVID-19, SARS-CoV-2 was also inactivated upon treatment with 50µg/ml Echinaforce®. CONCLUSIONS: These results show that Echinaforce® is virucidal against HCoV-229E, upon direct contact and in an organotypic cell culture model. Furthermore, MERS-CoV and both SARS-CoV-1 and SARS-CoV-2 were inactivated at similar concentrations of the extract. Therefore we hypothesize that Echinacea purpurea preparations, such as Echinaforce®, could be effective as prophylactic treatment for all CoVs due to their structural similarities.


Asunto(s)
Antivirales/farmacología , Betacoronavirus/efectos de los fármacos , Coronavirus Humano 229E/efectos de los fármacos , Infecciones por Coronavirus/tratamiento farmacológico , Coronavirus/efectos de los fármacos , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Animales , COVID-19 , Línea Celular , Chlorocebus aethiops , Resfriado Común/tratamiento farmacológico , Resfriado Común/virología , Infecciones por Coronavirus/virología , Humanos , Coronavirus del Síndrome Respiratorio de Oriente Medio/efectos de los fármacos , Pandemias , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/virología , Virus ARN/efectos de los fármacos , Ensayos Clínicos Controlados Aleatorios como Asunto , SARS-CoV-2 , Síndrome Respiratorio Agudo Grave/tratamiento farmacológico , Síndrome Respiratorio Agudo Grave/virología , Células Vero
5.
Euro Surveill ; 25(36)2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32914744

RESUMEN

In March 2020, we observed an outbreak of COVID-19 among a relatively homogenous group of 199 young (median age 21 years; 87% men) Swiss recruits. By comparing physical endurance before and in median 45 days after the outbreak, we found a significant decrease in predicted maximal aerobic capacity in COVID-19 convalescent but not in asymptomatically infected and SARS-CoV-2 naive recruits. This finding might be indicative of lung injury after apparently mild COVID-19 in young adults.


Asunto(s)
Infecciones por Coronavirus/diagnóstico , Coronavirus/aislamiento & purificación , Ejercicio Físico/fisiología , Lesión Pulmonar/etiología , Consumo de Oxígeno , Resistencia Física/fisiología , Neumonía Viral/diagnóstico , Ventilación Pulmonar/fisiología , Adulto , Infecciones Asintomáticas , Betacoronavirus , COVID-19 , Convalecencia , Coronavirus/genética , Infecciones por Coronavirus/epidemiología , Brotes de Enfermedades , Femenino , Humanos , Masculino , Personal Militar , Pandemias , Resistencia Física/inmunología , Aptitud Física , Neumonía Viral/epidemiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Riesgo , SARS-CoV-2 , Suiza/epidemiología , Adulto Joven
6.
J Virol ; 92(21)2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30111567

RESUMEN

Dengue virus (DENV) is the most prevalent mosquito-transmitted viral pathogen in humans. The recently licensed dengue vaccine has major weaknesses. Therefore, there is an urgent need to develop improved dengue vaccines. Here, we report a virion assembly-defective DENV as a vaccine platform. DENV containing an amino acid deletion (K188) in nonstructural protein 2A (NS2A) is fully competent in viral RNA replication but is completely defective in virion assembly. When trans-complemented with wild-type NS2A protein, the virion assembly defect could be rescued, generating pseudoinfectious virus (PIVNS2A) that could initiate single-round infection. The trans-complementation efficiency could be significantly improved through selection for adaptive mutations, leading to high-yield PIVNS2A production, with titers of >107 infectious-focus units (IFU)/ml. Mice immunized with a single dose of PIVNS2A elicited strong T cell immune responses and neutralization antibodies and were protected from wild-type-virus challenge. Collectively, the results proved the concept of using assembly-defective virus as a vaccine approach. The study also solved the technical bottleneck in producing high yields of PIVNS2A vaccine. The technology could be applicable to vaccine development for other viral pathogens.IMPORTANCE Many flaviviruses are significant human pathogens that pose global threats to public health. Although licensed vaccines are available for yellow fever, Japanese encephalitis, tick-borne encephalitis, and dengue viruses, new approaches are needed to develop improved vaccines. Using dengue virus as a model, we developed a vaccine platform using a virion assembly-defective virus. We show that such an assembly-defective virus could be rescued to higher titers and infect cells for a single round. Mice immunized with the assembly-defective virus were protected from wild-type-virus infection. This vaccine approach could be applicable to other viral pathogens.


Asunto(s)
Virus Defectuosos/patogenicidad , Vacunas contra el Dengue/inmunología , Virus del Dengue/patogenicidad , Dengue/virología , Proteínas no Estructurales Virales/inmunología , Ensamble de Virus , Replicación Viral , Animales , Anticuerpos Neutralizantes/inmunología , Virus Defectuosos/genética , Dengue/genética , Dengue/inmunología , Virus del Dengue/genética , Femenino , Humanos , Masculino , Ratones , Mutación , ARN Viral , Proteínas no Estructurales Virales/genética
7.
PLoS Pathog ; 13(2): e1006195, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28158275

RESUMEN

Coronaviruses are of veterinary and medical importance and include highly pathogenic zoonotic viruses, such as SARS-CoV and MERS-CoV. They are known to efficiently evade early innate immune responses, manifesting in almost negligible expression of type-I interferons (IFN-I). This evasion strategy suggests an evolutionary conserved viral function that has evolved to prevent RNA-based sensing of infection in vertebrate hosts. Here we show that the coronavirus endonuclease (EndoU) activity is key to prevent early induction of double-stranded RNA (dsRNA) host cell responses. Replication of EndoU-deficient coronaviruses is greatly attenuated in vivo and severely restricted in primary cells even during the early phase of the infection. In macrophages we found immediate induction of IFN-I expression and RNase L-mediated breakdown of ribosomal RNA. Accordingly, EndoU-deficient viruses can retain replication only in cells that are deficient in IFN-I expression or sensing, and in cells lacking both RNase L and PKR. Collectively our results demonstrate that the coronavirus EndoU efficiently prevents simultaneous activation of host cell dsRNA sensors, such as Mda5, OAS and PKR. The localization of the EndoU activity at the site of viral RNA synthesis-within the replicase complex-suggests that coronaviruses have evolved a viral RNA decay pathway to evade early innate and intrinsic antiviral host cell responses.


Asunto(s)
Coronaviridae/enzimología , Infecciones por Coronavirus/inmunología , Endonucleasas/inmunología , Evasión Inmune/fisiología , Proteínas Virales/inmunología , Animales , Coronaviridae/inmunología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata/inmunología , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena en Tiempo Real de la Polimerasa
8.
Artículo en Inglés | MEDLINE | ID: mdl-30181371

RESUMEN

The virus family Flaviviridae encompasses several viruses, including (re)emerging viruses which cause widespread morbidity and mortality throughout the world. Members of this virus family are positive-strand RNA viruses and replicate their genome in close association with reorganized intracellular host cell membrane compartments. This evolutionarily conserved strategy facilitates efficient viral genome replication and contributes to evasion from host cell cytosolic defense mechanisms. We have previously described the identification of a small-compound inhibitor, K22, which exerts a potent antiviral activity against a broad range of coronaviruses by targeting membrane-bound viral RNA replication. To analyze the antiviral spectrum of this inhibitor, we assessed the inhibitory potential of K22 against several members of the Flaviviridae family, including the reemerging Zika virus (ZIKV). We show that ZIKV is strongly affected by K22. Time-of-addition experiments revealed that K22 acts during a postentry phase of the ZIKV life cycle, and combination regimens of K22 together with ribavirin (RBV) or interferon alpha (IFN-α) further increased the extent of viral inhibition. Ultrastructural electron microscopy studies revealed severe alterations of ZIKV-induced intracellular replication compartments upon infection of K22-treated cells. Importantly, the antiviral activity of K22 was demonstrated against several other members of the Flaviviridae family. It is tempting to speculate that K22 exerts its broad antiviral activity against several positive-strand RNA viruses via a similar mechanism and thereby represents an attractive candidate for development as a panviral inhibitor.


Asunto(s)
Antivirales/farmacología , Membrana Celular/efectos de los fármacos , Infecciones por Flaviviridae/tratamiento farmacológico , Flaviviridae/efectos de los fármacos , Aedes , Animales , Línea Celular , Membrana Celular/virología , Chlorocebus aethiops , Infecciones por Flaviviridae/virología , Humanos , Interferón-alfa/farmacología , ARN Viral/genética , Ribavirina/farmacología , Células Vero , Replicación Viral/efectos de los fármacos
9.
J Virol ; 90(24): 11122-11131, 2016 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-27707930

RESUMEN

Half of the world's population is exposed to the risk of dengue virus infection. Although a vaccine for dengue virus is now available in a few countries, its reported overall efficacy of about 60% is not ideal. Protective immune correlates following natural dengue virus infection remain undefined, which makes it difficult to predict the efficacy of new vaccines. In this study, we address the protective capacity of dengue virus-specific antibodies that are produced by plasmablasts a few days after natural secondary infection. Among a panel of 18 dengue virus-reactive human monoclonal antibodies, four groups of antibodies were identified based on their binding properties. While antibodies targeting the fusion loop of the glycoprotein of dengue virus dominated the antibody response, two smaller groups of antibodies bound to previously undescribed epitopes in domain II of the E protein. The latter, largely serotype-cross-reactive antibodies, demonstrated increased stability of binding at pH 5. These antibodies possessed weak to moderate neutralization capacity in vitro but were the most efficacious in promoting the survival of infected mice. Our data suggest that the cross-reactive anamnestic antibody response has a protective capacity despite moderate neutralization in vitro and a moderate decrease of viremia in vivo IMPORTANCE: Antibodies can protect from symptomatic dengue virus infection. However, it is not easy to assess which classes of antibodies provide protection because in vitro assays are not always predictive of in vivo protection. During a repeat infection, dengue virus-specific immune memory cells are reactivated and large amounts of antibodies are produced. By studying antibodies cloned from patients with heterologous secondary infection, we tested the protective value of the serotype-cross-reactive "recall" or "anamnestic" response. We found that results from in vitro neutralization assays did not always correlate with the ability of the antibodies to reduce viremia in a mouse model. In addition, a decrease of viremia in mice did not necessarily improve survival. The most protective antibodies were stable at pH 5, suggesting that antibody binding in the endosomes, after the antibody-virus complex is internalized, might be important to block virus spread in the organism.


Asunto(s)
Anticuerpos Monoclonales/biosíntesis , Anticuerpos Neutralizantes/biosíntesis , Anticuerpos Antivirales/biosíntesis , Virus del Dengue/inmunología , Dengue/prevención & control , Proteínas del Envoltorio Viral/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/química , Anticuerpos Neutralizantes/administración & dosificación , Anticuerpos Neutralizantes/química , Anticuerpos Antivirales/administración & dosificación , Anticuerpos Antivirales/química , Reacciones Cruzadas , Dengue/inmunología , Dengue/virología , Virus del Dengue/clasificación , Virus del Dengue/genética , Modelos Animales de Enfermedad , Mapeo Epitopo , Epítopos/química , Epítopos/inmunología , Expresión Génica , Humanos , Concentración de Iones de Hidrógeno , Inmunidad Humoral/efectos de los fármacos , Memoria Inmunológica , Ratones , Pruebas de Neutralización , Unión Proteica , Estabilidad Proteica , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología
10.
Nature ; 468(7322): 452-6, 2010 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-21085181

RESUMEN

Cellular messenger RNA (mRNA) of higher eukaryotes and many viral RNAs are methylated at the N-7 and 2'-O positions of the 5' guanosine cap by specific nuclear and cytoplasmic methyltransferases (MTases), respectively. Whereas N-7 methylation is essential for RNA translation and stability, the function of 2'-O methylation has remained uncertain since its discovery 35 years ago. Here we show that a West Nile virus (WNV) mutant (E218A) that lacks 2'-O MTase activity was attenuated in wild-type primary cells and mice but was pathogenic in the absence of type I interferon (IFN) signalling. 2'-O methylation of viral RNA did not affect IFN induction in WNV-infected fibroblasts but instead modulated the antiviral effects of IFN-induced proteins with tetratricopeptide repeats (IFIT), which are interferon-stimulated genes (ISGs) implicated in regulation of protein translation. Poxvirus and coronavirus mutants that lacked 2'-O MTase activity similarly showed enhanced sensitivity to the antiviral actions of IFN and, specifically, IFIT proteins. Our results demonstrate that the 2'-O methylation of the 5' cap of viral RNA functions to subvert innate host antiviral responses through escape of IFIT-mediated suppression, and suggest an evolutionary explanation for 2'-O methylation of cellular mRNA: to distinguish self from non-self RNA. Differential methylation of cytoplasmic RNA probably serves as an example for pattern recognition and restriction of propagation of foreign viral RNA in host cells.


Asunto(s)
Proteínas Portadoras/metabolismo , Regulación de la Expresión Génica/inmunología , Inmunidad Innata/inmunología , Interferones/inmunología , Proteínas/metabolismo , Caperuzas de ARN/metabolismo , ARN Viral/metabolismo , Células 3T3 , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Reguladoras de la Apoptosis , Proteínas Portadoras/genética , Células Cultivadas , Coronavirus/enzimología , Coronavirus/genética , Coronavirus/inmunología , Coronavirus/fisiología , Fibroblastos , Regulación de la Expresión Génica/genética , Humanos , Inmunidad Innata/genética , Interferones/deficiencia , Interferones/genética , Metilación , Metiltransferasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Modelos Genéticos , Modelos Inmunológicos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Poxviridae/enzimología , Poxviridae/genética , Poxviridae/inmunología , Poxviridae/fisiología , Biosíntesis de Proteínas/inmunología , Proteínas/genética , Caperuzas de ARN/genética , Caperuzas de ARN/inmunología , ARN Viral/genética , ARN Viral/inmunología , Proteínas de Unión al ARN , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/genética , Tasa de Supervivencia , Replicación Viral , Virus del Nilo Occidental/enzimología , Virus del Nilo Occidental/genética , Virus del Nilo Occidental/inmunología , Virus del Nilo Occidental/fisiología
11.
J Virol ; 88(13): 7276-85, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24741106

RESUMEN

UNLABELLED: Dengue virus (DENV) infects an estimated 400 million people every year, causing prolonged morbidity and sometimes mortality. Development of an effective vaccine has been hampered by the lack of appropriate small animal models; mice are naturally not susceptible to DENV and only become infected if highly immunocompromised. Mouse models lacking both type I and type II interferon (IFN) receptors (AG129 mice) or the type I IFN receptor (IFNAR(-/-) mice) are susceptible to infection with mouse-adapted DENV strains but are severely impaired in mounting functional immune responses to the virus and thus are of limited use for study. Here we used conditional deletion of the type I IFN receptor (IFNAR) on individual immune cell subtypes to generate a minimally manipulated mouse model that is susceptible to DENV while retaining global immune competence. Mice lacking IFNAR expression on CD11c(+) dendritic cells and LysM(+) macrophages succumbed completely to DENV infection, while mice deficient in the receptor on either CD11c(+) or LysM(+) cells were susceptible to infection but often resolved viremia and recovered fully from infection. Conditional IFNAR mice responded with a swift and strong CD8(+) T-cell response to viral infection, compared to a weak response in IFNAR(-/-) mice. Furthermore, mice lacking IFNAR on either CD11c(+) or LysM(+) cells were also sufficiently immunocompetent to raise a protective immune response to a candidate subunit vaccine against DENV-2. These data demonstrate that mice with conditional deficiencies in expression of the IFNAR represent improved models for the study of DENV immunology and screening of vaccine candidates. IMPORTANCE: Dengue virus infects 400 million people every year worldwide, causing 100 million clinically apparent infections, which can be fatal if untreated. Despite many years of research, there are no effective vaccine and no antiviral treatment available for dengue. Development of vaccines has been hampered in particular by the lack of a suitable small animal model. Mouse models used to test dengue vaccine are deficient in interferon (IFN) type I signaling and severely immunocompromised and therefore likely not ideal for the testing of vaccines. In this study, we explored alternative models lacking the IFN receptor only on certain cell types. We show that mice lacking the IFN receptor on either CD11c- or LysM-expressing cells (conditional IFNAR mice) are susceptible to dengue virus infection. Importantly, we demonstrate that conditional IFN receptor knockout mice generate a better immune response to live virus and a candidate dengue vaccine compared to IFNAR mice and are resistant to subsequent challenge.


Asunto(s)
Células Dendríticas/inmunología , Vacunas contra el Dengue/uso terapéutico , Dengue/inmunología , Modelos Animales de Enfermedad , Interferón Tipo I/fisiología , Interferón gamma/fisiología , Macrófagos/inmunología , Animales , Citocinas/metabolismo , Células Dendríticas/virología , Dengue/prevención & control , Dengue/virología , Virus del Dengue/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Humanos , Inmunización , Macrófagos/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Viral/genética , Replicación Viral
12.
PLoS Pathog ; 9(8): e1003521, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23935499

RESUMEN

Dengue virus is transmitted by Aedes mosquitoes and infects at least 100 million people every year. Progressive urbanization in Asia and South-Central America and the geographic expansion of Aedes mosquito habitats have accelerated the global spread of dengue, resulting in a continuously increasing number of cases. A cost-effective, safe vaccine conferring protection with ideally a single injection could stop dengue transmission. Current vaccine candidates require several booster injections or do not provide protection against all four serotypes. Here we demonstrate that dengue virus mutants lacking 2'-O-methyltransferase activity are highly sensitive to type I IFN inhibition. The mutant viruses are attenuated in mice and rhesus monkeys and elicit a strong adaptive immune response. Monkeys immunized with a single dose of 2'-O-methyltransferase mutant virus showed 100% sero-conversion even when a dose as low as 1,000 plaque forming units was administrated. Animals were fully protected against a homologous challenge. Furthermore, mosquitoes feeding on blood containing the mutant virus were not infected, whereas those feeding on blood containing wild-type virus were infected and thus able to transmit it. These results show the potential of 2'-O-methyltransferase mutant virus as a safe, rationally designed dengue vaccine that restrains itself due to the increased susceptibility to the host's innate immune response.


Asunto(s)
Vacunas contra el Dengue/inmunología , Virus del Dengue/inmunología , Dengue/inmunología , Metiltransferasas/inmunología , Animales , Cricetinae , Dengue/enzimología , Dengue/genética , Dengue/prevención & control , Vacunas contra el Dengue/genética , Vacunas contra el Dengue/farmacología , Virus del Dengue/genética , Células HEK293 , Humanos , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Macaca mulatta , Metiltransferasas/genética , Ratones , Ratones Mutantes , Mutación , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Vacunas Atenuadas/farmacología
13.
J Gen Virol ; 95(Pt 4): 763-778, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24486628

RESUMEN

The 5' end of eukaryotic mRNA contains the type-1 (m7GpppNm) or type-2 (m7GpppNmNm) cap structure. Many viruses have evolved various mechanisms to develop their own capping enzymes (e.g. flavivirus and coronavirus) or to 'steal' caps from host mRNAs (e.g. influenza virus). Other viruses have developed 'cap-mimicking' mechanisms by attaching a peptide to the 5' end of viral RNA (e.g. picornavirus and calicivirus) or by having a complex 5' RNA structure (internal ribosome entry site) for translation initiation (e.g. picornavirus, pestivirus and hepacivirus). Here we review the diverse viral RNA capping mechanisms. Using flavivirus as a model, we summarize how a single methyltransferase catalyses two distinct N-7 and 2'-O methylations of viral RNA cap in a sequential manner. For antiviral development, a structural feature unique to the flavivirus methyltransferase was successfully used to design selective inhibitors that block viral methyltransferase without affecting host methyltransferases. Functionally, capping is essential for prevention of triphosphate-triggered innate immune activation; N-7 methylation is critical for enhancement of viral translation; and 2'-O methylation is important for subversion of innate immune response during viral infection. Flaviviruses defective in 2'-O methyltransferase are replicative, but their viral RNAs lack 2'-O methylation and are recognized and eliminated by the host immune response. Such mutant viruses could be rationally designed as live attenuated vaccines. This concept has recently been proved with Japanese encephalitis virus and dengue virus. The findings obtained with flavivirus should be applicable to other RNA viruses.


Asunto(s)
Flavivirus/enzimología , Flavivirus/metabolismo , Análogos de Caperuza de ARN , Procesamiento Postranscripcional del ARN , ARN Viral/metabolismo , ARNt Metiltransferasas/metabolismo , Evasión Inmune , Metilación , Biosíntesis de Proteínas
14.
Heliyon ; 10(10): e31490, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38826712

RESUMEN

Due to the discontinuation of routine smallpox vaccination after its eradication in 1980, a large part of the human population remains naïve against smallpox and other members of the orthopoxvirus genus. As a part of biosafety personnel protection programs, laboratory workers receive prophylactic vaccinations against diverse infectious agents, including smallpox. Here, we studied the levels of cross-protecting neutralizing antibodies as well as total IgG induced by either first- or third-generation smallpox vaccines against Monkeypox virus, using a clinical isolate from the 2022 outbreak. Serum neutralization tests indicated better overall neutralization capacity after vaccination with first-generation smallpox vaccines, compared to an attenuated third-generation vaccine. Results obtained from total IgG ELISA, however, did not show higher induction of orthopoxvirus-specific IgGs in first-generation vaccine recipients. Taken together, our results indicate a lower level of cross-protecting neutralizing antibodies against Monkeypox virus in recipients of third-generation smallpox vaccine compared to first-generation vaccine recipients, although total IgG levels were comparable.

15.
Eur J Immunol ; 42(9): 2311-21, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22730043

RESUMEN

Myocarditis is a potentially lethal inflammatory heart disease of children and young adults that frequently leads to dilated cardiomyopathy (DCM). Since diagnostic procedures and efficient therapies are lacking, it is important to characterize the critical immune effector pathways underlying the initial cardiac inflammation and the transition from myocarditis to DCM. We describe here a T-cell receptor (TCR) transgenic mouse model with spontaneously developing autoimmune myocarditis that progresses to lethal DCM. Cardiac magnetic resonance imaging revealed early inflammation-associated changes in the ventricle wall including transient thickening of the left ventricle wall. Furthermore, we found that IFN-γ was a major effector cytokine driving the initial inflammatory process and that the cooperation of IFN-γ and IL-17A was essential for the development of the progressive disease. This novel TCR transgenic mouse model permits the identification of the central pathophysiological and immunological processes involved in the transition from autoimmune myocarditis to DCM.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Cardiomiopatía Dilatada/inmunología , Cardiomiopatía Dilatada/patología , Miocarditis/inmunología , Miocarditis/patología , Células TH1/inmunología , Células Th17/inmunología , Animales , Autoantígenos/inmunología , Enfermedades Autoinmunes/patología , Modelos Animales de Enfermedad , Ventrículos Cardíacos/inmunología , Ventrículos Cardíacos/patología , Inflamación/inmunología , Inflamación/patología , Interferón gamma/inmunología , Interleucina-17/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/inmunología , Remodelación Ventricular/inmunología
16.
PLoS Pathog ; 7(10): e1002331, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22046132

RESUMEN

Coronaviruses (CoVs) are important human and animal pathogens that induce fatal respiratory, gastrointestinal and neurological disease. The outbreak of the severe acute respiratory syndrome (SARS) in 2002/2003 has demonstrated human vulnerability to (Coronavirus) CoV epidemics. Neither vaccines nor therapeutics are available against human and animal CoVs. Knowledge of host cell proteins that take part in pivotal virus-host interactions could define broad-spectrum antiviral targets. In this study, we used a systems biology approach employing a genome-wide yeast-two hybrid interaction screen to identify immunopilins (PPIA, PPIB, PPIH, PPIG, FKBP1A, FKBP1B) as interaction partners of the CoV non-structural protein 1 (Nsp1). These molecules modulate the Calcineurin/NFAT pathway that plays an important role in immune cell activation. Overexpression of NSP1 and infection with live SARS-CoV strongly increased signalling through the Calcineurin/NFAT pathway and enhanced the induction of interleukin 2, compatible with late-stage immunopathogenicity and long-term cytokine dysregulation as observed in severe SARS cases. Conversely, inhibition of cyclophilins by cyclosporine A (CspA) blocked the replication of CoVs of all genera, including SARS-CoV, human CoV-229E and -NL-63, feline CoV, as well as avian infectious bronchitis virus. Non-immunosuppressive derivatives of CspA might serve as broad-range CoV inhibitors applicable against emerging CoVs as well as ubiquitous pathogens of humans and livestock.


Asunto(s)
Antivirales/uso terapéutico , Ciclofilinas/metabolismo , Síndrome Respiratorio Agudo Grave/tratamiento farmacológico , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/patogenicidad , Animales , Células CACO-2 , Chlorocebus aethiops , Ciclofilinas/antagonistas & inhibidores , Ciclofilinas/efectos de los fármacos , Ciclosporina/farmacología , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Células Jurkat , Inhibidores de Proteasas/farmacología , Mapeo de Interacción de Proteínas , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/efectos de los fármacos , Técnicas del Sistema de Dos Híbridos , Células Vero , Proteínas Virales/metabolismo , Replicación Viral/efectos de los fármacos
17.
Sci Rep ; 13(1): 20556, 2023 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-37996620

RESUMEN

While the global healthcare system is slowly recovering from the COVID-19 pandemic, new multi-drug-resistant pathogens are emerging as the next threat. To tackle these challenges there is a need for safe and sustainable antiviral and antibacterial functionalized materials. Here we develop an 'easy-to-apply' procedure for the surface functionalization of textiles, rendering them antiviral and antibacterial and assessing the performance of these textiles. A metal-free quaternary ammonium-based coating was applied homogeneously and non-covalently to hospital curtains. Abrasion, durability testing, and aging resulted in little change in the performance of the treated textile. Additionally, qualitative and quantitative antibacterial assays on Staphylococcus aureus, Pseudomonas aeruginosa, and Acinetobacter baumanii revealed excellent antibacterial activity with a CFU reduction of 98-100% within only 4 h of exposure. The treated curtain was aged 6 months before testing. Similarly, the antiviral activity tested according to ISO-18184 with murine hepatitis virus (MHV) showed > 99% viral reduction with the functionalized curtain. Also, the released active compounds of the coating 24 ± 5 µg mL-1 revealed no acute in vitro skin toxicity (IC50: 95 µg mL-1) and skin sensitization. This study emphasizes the potential of safe and sustainable metal-free textile coatings for the rapid antiviral and antibacterial functionalization of textiles.


Asunto(s)
Compuestos de Amonio , Virus , Ratones , Animales , Humanos , Pandemias , Textiles/microbiología , Bacterias , Antibacterianos/farmacología , Antivirales
18.
Sci Rep ; 13(1): 15718, 2023 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-37735604

RESUMEN

Proper disinfection and inactivation of highly pathogenic viruses is an essential component of public health and prevention. Depending on environment, surfaces, and type of contaminant, various methods of disinfection must be both efficient and available. To test both established and novel chemical disinfectants against risk group 4 viruses in our maximum containment facility, we developed a standardized protocol and assessed the chemical inactivation of the two Ebola virus variants Mayinga and Makona suspended in two different biological soil loads. Standard chemical disinfectants ethanol and sodium hypochlorite completely inactivate both Ebola variants after 30 s in suspension at 70% and 0.5% v/v, respectively, concentrations recommended for disinfection by the World Health Organization. Additionally, peracetic acid is also inactivating at 0.2% v/v under the same conditions. Continued vigilance and optimization of current disinfection protocols is extremely important due to the continuous presence of Ebola virus on the African continent and increased zoonotic spillover of novel viral pathogens. Furthermore, to facilitate general pandemic preparedness, the establishment and sharing of standardized protocols is very important as it allows for rapid testing and evaluation of novel pathogens and chemical disinfectants.


Asunto(s)
Desinfectantes , Ebolavirus , Fiebre Hemorrágica Ebola , Humanos , Desinfectantes/farmacología , Fiebre Hemorrágica Ebola/prevención & control , Desinfección , Suelo
19.
Viruses ; 15(11)2023 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-38005942

RESUMEN

When infecting humans, Andes orthohantavirus (ANDV) may cause a severe disease called hantavirus cardiopulmonary syndrome (HCPS). Following non-specific symptoms, the infection may progress to a syndrome of hemorrhagic fever combined with hyper-acute cardiopulmonary failure. The case fatality rate ranges between 25-40%, depending on the outbreak. In this study, we present the follow-up of a male patient who recovered from HCPS six years ago. We demonstrate that the ANDV genome persists within the reproductive tract for at least 71 months. Genome sequence analysis early and late after infection reveals a low number of mutations (two single nucleotide variants and one deletion), suggesting limited replication activity. We can exclude the integration of the viral genome into the host genome, since the treatment of the specimen with RNAse led to a loss of signal. We demonstrate a long-lasting, strong neutralizing antibody response using pseudovirions expressing the ANDV glycoprotein. Taken together, our results show that ANDV has the potential for sexual transmission.


Asunto(s)
Infecciones por Hantavirus , Orthohantavirus , Humanos , Masculino , Orthohantavirus/genética , Semen , Anticuerpos Neutralizantes , ARN Viral/genética
20.
Clin Microbiol Infect ; 29(12): 1587-1594, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37661067

RESUMEN

OBJECTIVES: To report 5-year persistence and avidity of antibodies produced by the live-attenuated recombinant vesicular stomatitis virus (rVSV) expressing the Zaire Ebolavirus (ZEBOV) glycoprotein (GP), known as rVSV-ZEBOV (Ervebo®). METHODS: Healthy adults vaccinated with 300,000 or 10-50 million plaque-forming units of rVSV-ZEBOV in the WHO-coordinated trials of 2014-2015 were followed for up to 4 (Lambaréné, Gabon) and 5 (Geneva, Switzerland) years. We report seropositivity rates, geometric mean titres (GMTs), and population distribution of ZEBOV-GP ELISA IgG antibodies, neutralizing antibodies (pseudovirus and live-virus neutralization) and antibody avidity; the primary outcome was ZEBOV-GP ELISA IgG GMTs at 4 or 5 years compared with 1 year (Y1) after immunization. RESULTS: Among the 168 eligible vaccinees (Geneva: 97 and Lambaréné: 71) enrolled 1 year post-immunization, 146 (87%) remained enrolled at 4 years (Geneva: n = 88, Lambaréné: n = 58), and 84 (87%, Geneva) at 5 years post-vaccination. ZEBOV-GP ELISA IgG GMTs plateaued, with no declining trend from 1 year through the last time point assessed (1147.8 [95% CI 874.3-1507.0] at Y1 versus 1548.1 [95% CI 1136.6-2108.5] at Y5 in Geneva volunteers receiving ≥10 million plaque-forming units of rVSV-ZEBOV), their avidity matching that of ZEBOV convalescents. Live-virus neutralizing antibodies were detected for shorter periods and in fewer vaccinees (53/95 [56%] at Y1 versus 35/84 [42%] at Y5 in Geneva volunteers, all dose levels). DISCUSSION: Titres at Y1 emerged as a correlate of antibody persistence at Y5. The findings of persistent ZEBOV-GP ELISA IgG titres yet shorter-lasting, lower titres of live-virus neutralizing antibodies suggest the contribution of antibody-mediated protective mechanisms other than neutralization. Long-term clinical efficacy of rVSV-ZEBOV, however, requires further study.


Asunto(s)
Vacunas contra el Virus del Ébola , Ebolavirus , Fiebre Hemorrágica Ebola , Estomatitis Vesicular , Adulto , Animales , Humanos , Ebolavirus/genética , Formación de Anticuerpos , República Democrática del Congo , Anticuerpos Antivirales , Vacunación , Anticuerpos Neutralizantes , Inmunoglobulina G , Anticuerpos Bloqueadores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA