Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
PLoS Pathog ; 16(3): e1008373, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32150583

RESUMEN

Lasting protection has long been a goal for malaria vaccines. The major surface antigen on Plasmodium falciparum sporozoites, the circumsporozoite protein (PfCSP), has been an attractive target for vaccine development and most protective antibodies studied to date interact with the central NANP repeat region of PfCSP. However, it remains unclear what structural and functional characteristics correlate with better protection by one antibody over another. Binding to the junctional region between the N-terminal domain and central NANP repeats has been proposed to result in superior protection: this region initiates with the only NPDP sequence followed immediately by NANP. Here, we isolated antibodies in Kymab mice immunized with full-length recombinant PfCSP and two protective antibodies were selected for further study with reactivity against the junctional region. X-ray and EM structures of two monoclonal antibodies, mAb667 and mAb668, shed light on their differential affinity and specificity for the junctional region. Importantly, these antibodies also bind to the NANP repeat region with equal or better affinity. A comparison with an NANP-only binding antibody (mAb317) revealed roughly similar but statistically distinct levels of protection against sporozoite challenge in mouse liver burden models, suggesting that junctional antibody protection might relate to the ability to also cross-react with the NANP repeat region. Our findings indicate that additional efforts are necessary to isolate a true junctional antibody with no or much reduced affinity to the NANP region to elucidate the role of the junctional epitope in protection.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/química , Anticuerpos Antiprotozoarios/química , Sitios de Unión de Anticuerpos , Epítopos/química , Plasmodium falciparum/química , Proteínas Protozoarias/química , Animales , Anticuerpos Monoclonales de Origen Murino/inmunología , Anticuerpos Antiprotozoarios/inmunología , Epítopos/inmunología , Femenino , Masculino , Ratones , Ratones Transgénicos , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología , Relación Estructura-Actividad
2.
J Clin Invest ; 127(4): 1463-1474, 2017 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-28240600

RESUMEN

The antiviral restriction factor IFN-induced transmembrane protein 3 (IFITM3) inhibits cell entry of a number of viruses, and genetic diversity within IFITM3 determines susceptibility to viral disease in humans. Here, we used the murine CMV (MCMV) model of infection to determine that IFITM3 limits herpesvirus-associated pathogenesis without directly preventing virus replication. Instead, IFITM3 promoted antiviral cellular immunity through the restriction of virus-induced lymphopenia, apoptosis-independent NK cell death, and loss of T cells. Viral disease in Ifitm3-/- mice was accompanied by elevated production of cytokines, most notably IL-6. IFITM3 inhibited IL-6 production by myeloid cells in response to replicating and nonreplicating virus as well as following stimulation with the TLR ligands Poly(I:C) and CpG. Although IL-6 promoted virus-specific T cell responses, uncontrolled IL-6 expression in Ifitm3-/- mice triggered the loss of NK cells and subsequently impaired control of MCMV replication. Thus, IFITM3 represents a checkpoint regulator of antiviral immunity that controls cytokine production to restrict viral pathogenesis. These data suggest the utility of cytokine-targeting strategies in the treatment of virus-infected individuals with impaired IFITM3 activity.


Asunto(s)
Citocinas/fisiología , Infecciones por Herpesviridae/metabolismo , Proteínas de la Membrana/fisiología , Animales , Células Cultivadas , Infecciones por Herpesviridae/inmunología , Inmunidad Celular , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Muromegalovirus/fisiología , Receptores de Interleucina-6/metabolismo , Transducción de Señal , Internalización del Virus , Replicación Viral
3.
Traffic ; 17(9): 997-1013, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27219333

RESUMEN

Interferon inducible transmembrane proteins (IFITMs) are broad-spectrum antiviral factors. In cell culture the entry of many enveloped viruses, including orthomyxo-, flavi-, and filoviruses, is inhibited by IFITMs, though the mechanism(s) involved remain unclear and may vary between viruses. We demonstrate that Sindbis and Semliki Forest virus (SFV), which both use endocytosis and acid-induced membrane fusion in early endosomes to infect cells, are restricted by the early endosomal IFITM3. The late endosomal IFITM2 is less restrictive and the plasma membrane IFITM1 does not inhibit normal infection by either virus. IFITM3 inhibits release of the SFV capsid into the cytosol, without inhibiting binding, internalization, trafficking to endosomes or low pH-induced conformational changes in the envelope glycoprotein. Infection by SFV fusion at the cell surface was inhibited by IFITM1, but was equally inhibited by IFITM3. Furthermore, an IFITM3 mutant (Y20A) that is localized to the plasma membrane inhibited infection by cell surface fusion more potently than IFITM1. Together, these results indicate that IFITMs, in particular IFITM3, can restrict alphavirus infection by inhibiting viral fusion with cellular membranes. That IFITM3 can restrict SFV infection by fusion at the cell surface equivalently to IFITM1 suggests that IFITM3 has greater antiviral potency against SFV.


Asunto(s)
Antígenos de Diferenciación/metabolismo , Endosomas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Unión al ARN/metabolismo , Virus de los Bosques Semliki/fisiología , Virus Sindbis/fisiología , Células A549 , Infecciones por Alphavirus/metabolismo , Infecciones por Alphavirus/virología , Antígenos de Diferenciación/genética , Endocitosis/fisiología , Endosomas/virología , Humanos , Proteínas de la Membrana/genética , Proteínas de Unión al ARN/genética , Internalización del Virus
4.
Nat Rev Microbiol ; 13(11): 662, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26439086

RESUMEN

This month's Genome Watch highlights insights into different selection pressures in dengue virus evolution.


Asunto(s)
Antivirales/farmacología , Virus del Dengue/efectos de los fármacos , Virus del Dengue/genética , Dengue/tratamiento farmacológico , Dengue/virología , Animales , Humanos
5.
PLoS Pathog ; 11(8): e1005056, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26252219

RESUMEN

Genetic exchange by a process of genome-segment 'reassortment' represents an important mechanism for evolutionary change in all viruses with segmented genomes, yet in many cases a detailed understanding of its frequency and biological consequences is lacking. We provide a comprehensive assessment of reassortment in bluetongue virus (BTV), a globally important insect-borne pathogen of livestock, during recent outbreaks in Europe. Full-genome sequences were generated and analysed for over 150 isolates belonging to the different BTV serotypes that have emerged in the region over the last 5 decades. Based on this novel dataset we confirm that reassortment is a frequent process that plays an important and on-going role in evolution of the virus. We found evidence for reassortment in all ten segments without a significant bias towards any particular segment. However, we observed biases in the relative frequency at which particular segments were associated with each other during reassortment. This points to selective constraints possibly caused by functional relationships between individual proteins or genome segments and genome-wide epistatic interactions. Sites under positive selection were more likely to undergo amino acid changes in newly reassorted viruses, providing additional evidence for adaptive dynamics as a consequence of reassortment. We show that the live attenuated vaccines recently used in Europe have repeatedly reassorted with field strains, contributing to their genotypic, and potentially phenotypic, variability. The high degree of plasticity seen in the BTV genome in terms of segment origin suggests that current classification schemes that are based primarily on serotype, which is determined by only a single genome segment, are inadequate. Our work highlights the need for a better understanding of the mechanisms and epidemiological consequences of reassortment in BTV, as well as other segmented RNA viruses.


Asunto(s)
Virus de la Lengua Azul/genética , Lengua Azul/epidemiología , Lengua Azul/genética , Virus Reordenados/genética , Europa (Continente) , Evolución Molecular , Datos de Secuencia Molecular , Filogenia , Reacción en Cadena en Tiempo Real de la Polimerasa
6.
J Gen Virol ; 96(Pt 5): 991-1005, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25614588

RESUMEN

IFN-induced transmembrane protein 3 (IFITM3) is a restriction factor that blocks cytosolic entry of numerous viruses that utilize acidic endosomal entry pathways. In humans and mice, IFITM3 limits influenza-induced morbidity and mortality. Although many IFITM3-sensitive viruses are zoonotic, whether IFITMs function as antiviral restriction factors in mammalian species other than humans and mice is unknown. Here, IFITM3 orthologues in the microbat (Myotis myotis) and pig (Sus scrofa domesticus) were identified using rapid amplification of cDNA ends. Amino acid residues known to be important for IFITM3 function were conserved in the pig and microbat orthologues. Ectopically expressed pig and microbat IFITM3 co-localized with transferrin (early endosomes) and CD63 (late endosomes/multivesicular bodies). Pig and microbat IFITM3 restricted cell entry mediated by multiple influenza haemagglutinin subtypes and lyssavirus glycoproteins. Expression of pig or microbat IFITM3 in A549 cells reduced influenza virus yields and nucleoprotein expression. Conversely, small interfering RNA knockdown of IFITM3 in pig NPTr cells and primary microbat cells enhanced virus replication, demonstrating that these genes are functional in their species of origin at endogenous levels. In summary, we showed that IFITMs function as potent broad-spectrum antiviral effectors in two mammals - pigs and bats - identified as major reservoirs for emerging viruses.


Asunto(s)
Interferones/inmunología , Lyssavirus/inmunología , Proteínas de la Membrana/metabolismo , Orthomyxoviridae/inmunología , Proteínas de Unión al ARN/metabolismo , Internalización del Virus , Animales , Quirópteros , Secuencia Conservada , Lyssavirus/fisiología , Proteínas de la Membrana/genética , Orthomyxoviridae/fisiología , Proteínas de Unión al ARN/genética , Homología de Secuencia de Aminoácido , Porcinos
8.
Nat Rev Microbiol ; 11(3): 150, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23411861

RESUMEN

This month's Genome Watch highlights how deep sequencing technologies have vastly reduced the time and prior knowledge needed to generate viral genomes.


Asunto(s)
Genoma Viral , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Virus/genética , Farmacorresistencia Viral , Humanos , Filogeografía , Factores de Tiempo , Virus/clasificación
9.
J Gen Virol ; 93(Pt 10): 2118-2130, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22815272

RESUMEN

Recent RNA interference (RNAi) studies have identified many host proteins that modulate virus infection, but small interfering RNA 'off-target' effects and the use of transformed cell lines limit their conclusiveness. As murine embryonic stem (mES) cells can be genetically modified and resources exist where many and eventually all known mouse genes are insertionally inactivated, it was reasoned that mES cells would provide a useful alternative to RNAi screens. Beyond allowing investigation of host-pathogen interactions in vitro, mES cells have the potential to differentiate into other primary cell types, as well as being used to generate knockout mice for in vivo studies. However, mES cells are poorly characterized for virus infection. To investigate whether ES cells can be used to explore host-virus interactions, this study characterized the responses of mES cells following infection by herpes simplex virus type 1 (HSV-1) and influenza A virus. HSV-1 replicated lytically in mES cells, although mES cells were less permissive than most other cell types tested. Influenza virus was able to enter mES cells and express some viral proteins, but the replication cycle was incomplete and no infectious virus was produced. Knockdown of the host protein AHCYL1 in mES cells reduced HSV-1 replication, showing the potential for using mES cells to study host-virus interactions. Transcriptional profiling, however, indicated the lack of an efficient innate immune response in these cells. mES cells may thus be useful to identify host proteins that play a role in virus replication, but they are not suitable to determine factors that are involved in innate host defence.


Asunto(s)
Células Madre Embrionarias/fisiología , Células Madre Embrionarias/virología , Virus de la Influenza A/fisiología , Infecciones por Orthomyxoviridae/genética , Replicación Viral/genética , Animales , Línea Celular , Cricetinae , Replicación del ADN/genética , Perros , Células Madre Embrionarias/metabolismo , Células HeLa , Herpes Simple/genética , Herpes Simple/metabolismo , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/metabolismo , Herpesvirus Humano 1/fisiología , Interacciones Huésped-Patógeno , Humanos , Virus de la Influenza A/genética , Virus de la Influenza A/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/virología , ARN Interferente Pequeño/genética , Transcripción Genética , Proteínas Virales/genética , Proteínas Virales/metabolismo
10.
Nature ; 484(7395): 519-23, 2012 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-22446628

RESUMEN

The 2009 H1N1 influenza pandemic showed the speed with which a novel respiratory virus can spread and the ability of a generally mild infection to induce severe morbidity and mortality in a subset of the population. Recent in vitro studies show that the interferon-inducible transmembrane (IFITM) protein family members potently restrict the replication of multiple pathogenic viruses. Both the magnitude and breadth of the IFITM proteins' in vitro effects suggest that they are critical for intrinsic resistance to such viruses, including influenza viruses. Using a knockout mouse model, we now test this hypothesis directly and find that IFITM3 is essential for defending the host against influenza A virus in vivo. Mice lacking Ifitm3 display fulminant viral pneumonia when challenged with a normally low-pathogenicity influenza virus, mirroring the destruction inflicted by the highly pathogenic 1918 'Spanish' influenza. Similar increased viral replication is seen in vitro, with protection rescued by the re-introduction of Ifitm3. To test the role of IFITM3 in human influenza virus infection, we assessed the IFITM3 alleles of individuals hospitalized with seasonal or pandemic influenza H1N1/09 viruses. We find that a statistically significant number of hospitalized subjects show enrichment for a minor IFITM3 allele (SNP rs12252-C) that alters a splice acceptor site, and functional assays show the minor CC genotype IFITM3 has reduced influenza virus restriction in vitro. Together these data reveal that the action of a single intrinsic immune effector, IFITM3, profoundly alters the course of influenza virus infection in mouse and humans.


Asunto(s)
Virus de la Influenza A/patogenicidad , Proteínas de la Membrana/metabolismo , Infecciones por Orthomyxoviridae/mortalidad , Proteínas de Unión al ARN/metabolismo , Alelos , Secuencia de Aminoácidos , Animales , Citocinas/inmunología , Inglaterra/epidemiología , Eliminación de Gen , Humanos , Subtipo H1N1 del Virus de la Influenza A/clasificación , Subtipo H1N1 del Virus de la Influenza A/crecimiento & desarrollo , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Subtipo H3N2 del Virus de la Influenza A/clasificación , Subtipo H3N2 del Virus de la Influenza A/crecimiento & desarrollo , Subtipo H3N2 del Virus de la Influenza A/patogenicidad , Virus de la Influenza A/clasificación , Virus de la Influenza A/crecimiento & desarrollo , Virus de la Influenza B/clasificación , Virus de la Influenza B/crecimiento & desarrollo , Virus de la Influenza B/patogenicidad , Gripe Humana/complicaciones , Gripe Humana/epidemiología , Gripe Humana/mortalidad , Gripe Humana/virología , Leucocitos/inmunología , Pulmón/patología , Pulmón/virología , Proteínas de la Membrana/química , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Infecciones por Orthomyxoviridae/complicaciones , Infecciones por Orthomyxoviridae/patología , Neumonía Viral/etiología , Neumonía Viral/patología , Neumonía Viral/prevención & control , Polimorfismo de Nucleótido Simple/genética , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Escocia/epidemiología , Replicación Viral
11.
J Virol ; 85(21): 11479-89, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21865388

RESUMEN

Bluetongue is a major infectious disease of ruminants that is caused by bluetongue virus (BTV). In this study, we analyzed virulence and genetic differences of (i) three BTV field strains from Italy maintained at either a low (L strains) or high (H strains) passage number in cell culture and (ii) three South African "reference" wild-type strains and their corresponding live attenuated vaccine strains. The Italian BTV L strains, in general, were lethal for both newborn NIH-Swiss mice inoculated intracerebrally and adult type I interferon receptor-deficient (IFNAR(-/-)) mice, while the virulence of the H strains was attenuated significantly in both experimental models. Similarly, the South African vaccine strains were not pathogenic for IFNAR(-/-) mice, while the corresponding wild-type strains were virulent. Thus, attenuation of the virulence of the BTV strains used in this study is not mediated by the presence of an intact interferon system. No clear distinction in virulence was observed for the South African BTV strains in newborn NIH-Swiss mice. Full genomic sequencing revealed relatively few amino acid substitutions, scattered in several different viral proteins, for the strains found to be attenuated in mice compared to the pathogenic related strains. However, only the genome segments encoding VP1, VP2, and NS2 consistently showed nonsynonymous changes between all virulent and attenuated strain pairs. This study established an experimental platform for investigating the determinants of BTV virulence. Future studies using reverse genetics will allow researchers to precisely map and "weight" the relative influences of the various genome segments and viral proteins on BTV virulence.


Asunto(s)
Virus de la Lengua Azul/patogenicidad , Lengua Azul/patología , Lengua Azul/virología , Factores de Virulencia/genética , Sustitución de Aminoácidos/genética , Animales , Animales Recién Nacidos , Virus de la Lengua Azul/aislamiento & purificación , Modelos Animales de Enfermedad , Genoma Viral , Italia , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Receptor de Interferón alfa y beta/deficiencia , Enfermedades de los Roedores/patología , Enfermedades de los Roedores/virología , Análisis de Secuencia de ADN , Pase Seriado , Análisis de Supervivencia , Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...