Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros











Intervalo de año de publicación
1.
J Virol ; 98(6): e0164123, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38690874

RESUMEN

Numerous viruses have been found to exploit glycoconjugates expressed on human cells as their initial attachment factor for viral entry and infection. The virus-cell glycointeractome, when characterized, may serve as a template for antiviral drug design. Heparan sulfate proteoglycans extensively decorate the human cell surface and were previously described as a primary receptor for human metapneumovirus (HMPV). After respiratory syncytial virus, HMPV is the second most prevalent respiratory pathogen causing respiratory tract infection in young children. To date, there is neither vaccine nor drug available to prevent or treat HMPV infection. Using a multidisciplinary approach, we report for the first time the glycointeractome of the HMPV fusion (F) protein, a viral surface glycoprotein that is essential for target-cell recognition, attachment, and entry. Our glycan microarray and surface plasmon resonance results suggest that Galß1-3/4GlcNAc moieties that may be sialylated or fucosylated are readily recognized by HMPV F. The bound motifs are highly similar to the N-linked and O-linked glycans primarily expressed on the human lung epithelium. We demonstrate that the identified glycans have the potential to compete with the cellular receptors used for HMPV entry and consequently block HMPV infection. We found that lacto-N-neotetraose demonstrated the strongest HMPV binding inhibition in a cell infection assay. Our current findings offer an encouraging and novel avenue for the design of anti-HMPV drug candidates using oligosaccharide templates.IMPORTANCEAll cells are decorated with a dense coat of sugars that makes a sugar code. Many respiratory viruses exploit this sugar code by binding to these sugars to cause infection. Human metapneumovirus is a leading cause for acute respiratory tract infections. Despite its medical importance, there is no vaccine or antiviral drug available to prevent or treat human metapneumovirus infection. This study investigates how human metapneumovirus binds to sugars in order to more efficiently infect the human host. We found that human metapneumovirus binds to a diverse range of sugars and demonstrated that these sugars can ultimately block viral infection. Understanding how viruses can take advantage of the sugar code on our cells could identify new intervention and treatment strategies to combat viral disease.


Asunto(s)
Metapneumovirus , Infecciones por Paramyxoviridae , Polisacáridos , Metapneumovirus/metabolismo , Metapneumovirus/fisiología , Humanos , Polisacáridos/metabolismo , Infecciones por Paramyxoviridae/virología , Infecciones por Paramyxoviridae/metabolismo , Proteínas Virales de Fusión/metabolismo , Internalización del Virus , Acoplamiento Viral , Unión Proteica , Receptores Virales/metabolismo , Línea Celular
2.
Sci Rep ; 6: 24138, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-27053240

RESUMEN

Human parainfluenza type-3 virus (hPIV-3) is one of the principal aetiological agents of acute respiratory illness in infants worldwide and also shows high disease severity in the elderly and immunocompromised, but neither therapies nor vaccines are available to treat or prevent infection, respectively. Using a multidisciplinary approach we report herein that the approved drug suramin acts as a non-competitive in vitro inhibitor of the hPIV-3 haemagglutinin-neuraminidase (HN). Furthermore, the drug inhibits viral replication in mammalian epithelial cells with an IC50 of 30 µM, when applied post-adsorption. Significantly, we show in cell-based drug-combination studies using virus infection blockade assays, that suramin acts synergistically with the anti-influenza virus drug zanamivir. Our data suggests that lower concentrations of both drugs can be used to yield high levels of inhibition. Finally, using NMR spectroscopy and in silico docking simulations we confirmed that suramin binds HN simultaneously with zanamivir. This binding event occurs most likely in the vicinity of the protein primary binding site, resulting in an enhancement of the inhibitory potential of the N-acetylneuraminic acid-based inhibitor. This study offers a potentially exciting avenue for the treatment of parainfluenza infection by a combinatorial repurposing approach of well-established approved drugs.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Virus de la Parainfluenza 3 Humana/efectos de los fármacos , Suramina/farmacología , Zanamivir/farmacología , Animales , Antivirales/metabolismo , Antivirales/farmacología , Sitios de Unión , Línea Celular , Sinergismo Farmacológico , Células Epiteliales/virología , Proteína HN/metabolismo , Interacciones Huésped-Patógeno/efectos de los fármacos , Riñón/citología , Cinética , Macaca mulatta , Espectroscopía de Resonancia Magnética , Simulación de Dinámica Molecular , Virus de la Parainfluenza 3 Humana/metabolismo , Virus de la Parainfluenza 3 Humana/fisiología , Unión Proteica , Suramina/metabolismo , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/metabolismo , Replicación Viral/efectos de los fármacos , Zanamivir/metabolismo
3.
Nat Commun ; 5: 5268, 2014 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-25327774

RESUMEN

Human parainfluenza viruses (hPIVs) cause upper and lower respiratory tract disease in children that results in a significant number of hospitalizations and impacts health systems worldwide. To date, neither antiviral drugs nor vaccines are approved for clinical use against parainfluenza virus, which reinforces the urgent need for new therapeutic discovery strategies. Here we use a multidisciplinary approach to develop potent inhibitors that target a structural feature within the hPIV type 3 haemagglutinin-neuraminidase (hPIV-3 HN). These dual-acting designer inhibitors represent the most potent designer compounds and efficiently block both hPIV cell entry and virion progeny release. We also define the binding mode of these inhibitors in the presence of whole-inactivated hPIV and recombinantly expressed hPIV-3 HN by Saturation Transfer Difference NMR spectroscopy. Collectively, our study provides an antiviral preclinical candidate and a new direction towards the discovery of potential anti-parainfluenza drugs.


Asunto(s)
Antivirales/química , Diseño de Fármacos , Hemaglutininas Virales/química , Neuraminidasa/antagonistas & inhibidores , Virus de la Parainfluenza 3 Humana , Dominio Catalítico , Línea Celular Tumoral , Simulación por Computador , Cristalografía por Rayos X , Inhibidores Enzimáticos/química , Ensayo de Inmunoadsorción Enzimática , Mapeo Epitopo , Humanos , Concentración 50 Inhibidora , Espectroscopía de Resonancia Magnética , Conformación Molecular , Simulación de Dinámica Molecular , Ácidos Neuramínicos/química , Proteínas Recombinantes/química , Solventes/química , Propiedades de Superficie
4.
Glycoconj J ; 27(1): 61-8, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19757028

RESUMEN

The secretor (Se)/nonsecretor (se) histo-blood group variation depends on the action of the FUT2 enzyme and has major implications for human susceptibility to infections. To characterize the functionality of FUT2 variants, we assessed the correlation between saliva phenotypes and sequence variation at the FUT2 gene in sixty seven individuals from northern Portugal. While most non-secretor haplotypes were found to carry the 428G > A nonsense mutation in association with a 739G > A missense substitution, we have also identified a recombinant haplotype carrying the 739*A allele together with the efficient 428*G variant in individuals with the Se phenotype. This finding suggested, in contrast to previous results, that the 739*A allele encodes an efficient Se allele. To test this hypothesis we evaluated the in vivo enzyme activity of full coding expression constructs in transient transfection of CHO-K1 cells using FACS (fluorescence-activated cell sorting) analysis and expression of type 2 and type 3 chain H structures as read out. We detected FUT2 activity for the 739*A expression construct, demonstrating that the 739G > A substitution is indeed not inactivating. In accordance with the hypothesis that FUT2 is under long standing balancing selection, we estimated that the time depth of FUT2 global genetic variation is as old as 3 million years. Age estimates of specific variants suggest that the 428G > A mutation occurred at least 1.87 million years ago while the 739G > A substitution is about 816,000 years old. The 385A > T missense mutation underlying the non-secretor phenotype in East Asians appears to be more recent and is likely to have occurred about 256,000 years ago.


Asunto(s)
Fucosiltransferasas/genética , Fucosiltransferasas/metabolismo , Variación Genética , Infecciones/enzimología , Alelos , Animales , Secuencia de Bases , Células CHO , Cricetinae , Cricetulus , Haplotipos/genética , Humanos , Antígenos del Grupo Sanguíneo de Lewis/metabolismo , Datos de Secuencia Molecular , Proteínas Mutantes/metabolismo , Polimorfismo Genético , Saliva/enzimología , Transfección , Galactósido 2-alfa-L-Fucosiltransferasa
5.
Glycobiology ; 18(12): 1085-93, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18818423

RESUMEN

Severe acute respiratory syndrome coronavirus (SARS-CoV) is a highly pathogenic emergent virus which replicates in cells that can express ABH histo-blood group antigens. The heavily glycosylated SARS-CoV spike (S) protein binds to angiotensin-converting enzyme 2 which serves as a cellular receptor. Epidemiological analysis of a hospital outbreak in Hong Kong revealed that blood group O was associated with a low risk of infection. In this study, we used a cellular model of adhesion to investigate whether natural antibodies of the ABO system could block the S protein and angiotensin-converting enzyme 2 interaction. To this aim, a C-terminally EGFP-tagged S protein was expressed in chinese hamster ovary cells cotransfected with an alpha1,2-fucosyltransferase and an A-transferase in order to coexpress the S glycoprotein ectodomain and the A antigen at the cell surface. We observed that the S protein/angiotensin-converting enzyme 2-dependent adhesion of these cells to an angiotensin-converting enzyme 2 expressing cell line was specifically inhibited by either a monoclonal or human natural anti-A antibodies, indicating that these antibodies may block the interaction between the virus and its receptor, thereby providing protection. In order to more fully appreciate the potential effect of the ABO polymorphism on the epidemiology of SARS, we built a mathematical model of the virus transmission dynamics that takes into account the protective effect of ABO natural antibodies. The model indicated that the ABO polymorphism could contribute to substantially reduce the virus transmission, affecting both the number of infected individuals and the kinetics of the epidemic.


Asunto(s)
Sistema del Grupo Sanguíneo ABO/inmunología , Autoanticuerpos/inmunología , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Animales , Antígenos Virales/inmunología , Células CHO , Adhesión Celular/inmunología , Chlorocebus aethiops , Cricetinae , Cricetulus , Citometría de Flujo , Humanos , Glicoproteínas de Membrana/inmunología , Glicoproteína de la Espiga del Coronavirus , Células Vero , Proteínas del Envoltorio Viral/antagonistas & inhibidores , Proteínas del Envoltorio Viral/inmunología
6.
Biochem J ; 393(Pt 3): 627-34, 2006 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-16266293

RESUMEN

Breast-feeding-associated protection against calicivirus diarrhoea is associated with the presence of high levels of 2-linked oligosaccharides in mother's milk, and human calicivirus strains including the NV (Norwalk virus) use gut 2-linked fucosylated glycans as receptors, suggesting the presence of decoy receptors in milk. Our aim was to analyse the ability of human milk to inhibit the attachment of rNV VLPs (recombinant NV-like particles) to their carbohydrate ligands and to characterize potential inhibitors found in milk. Milk from women with the secretor phenotype was strongly inhibitory, unlike milk from women that are non-secretors, which is devoid of 2-linked fucosylated structures. At least two fractions in human milk acted as inhibitors for the NV capsid attachment. The first fraction corresponded to BSSL (bile-salt-stimulated lipase) and the second to associated mucins MUC1 and MUC4. These proteins present tandem repeat O-glycosylated sequences that should act as decoy receptors for the NV, depending on the combined mother/child secretor status.


Asunto(s)
Antígenos/metabolismo , Proteínas de la Cápside/metabolismo , Carbohidratos , Glicoproteínas/metabolismo , Lipasa/metabolismo , Leche Humana/química , Mucinas/metabolismo , Virus Norwalk/metabolismo , Antígenos de Neoplasias , Duodeno/citología , Duodeno/metabolismo , Femenino , Humanos , Ligandos , Leche Humana/enzimología , Leche Humana/virología , Mucina-1 , Mucina 4 , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA