Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Virol ; 98(6): e0164123, 2024 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-38690874

RESUMO

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.


Assuntos
Metapneumovirus , Infecções por Paramyxoviridae , Polissacarídeos , Receptores Virais , Proteínas Virais de Fusão , Ligação Viral , Humanos , Linhagem Celular , Metapneumovirus/metabolismo , Metapneumovirus/fisiologia , Infecções por Paramyxoviridae/virologia , Infecções por Paramyxoviridae/metabolismo , Polissacarídeos/metabolismo , Ligação Proteica , Receptores Virais/química , Receptores Virais/metabolismo , Proteínas Virais de Fusão/metabolismo , Internalização do Vírus , Interações entre Hospedeiro e Microrganismos , Proteoglicanas de Heparan Sulfato/metabolismo
2.
mBio ; 15(4): e0211423, 2024 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-38470050

RESUMO

Multidrug-resistant bacteria such as the opportunistic pathogen Pseudomonas aeruginosa, which causes life-threatening infections especially in immunocompromised individuals and cystic fibrosis patients, pose an increasing threat to public health. In the search for new treatment options, P. aeruginosa uridine diphosphate-glucose pyrophosphorylase (PaUGP) has been proposed as a novel drug target because it is required for the biosynthesis of important virulence factors and linked to pathogenicity in animal models. Here, we show that UGP-deficient P. aeruginosa exhibits severely reduced virulence against human lung tissue and cells, emphasizing the enzyme's suitability as a drug target. To establish a basis for the development of selective PaUGP inhibitors, we solved the product-bound crystal structure of tetrameric PaUGP and conducted a comprehensive structure-function analysis, identifying key residues at two different molecular interfaces that are essential for tetramer integrity and catalytic activity and demonstrating that tetramerization is pivotal for PaUGP function. Importantly, we show that part of the PaUGP oligomerization interface is uniquely conserved across bacterial UGPs but does not exist in the human enzyme, therefore representing an allosteric site that may be targeted to selectively inhibit bacterial UGPs.IMPORTANCEInfections with the opportunistic bacterial pathogen Pseudomonas aeruginosa are becoming increasingly difficult to treat due to multidrug resistance. Here, we show that the enzyme uridine diphosphate-glucose pyrophosphorylase (UGP) is involved in P. aeruginosa virulence toward human lung tissue and cells, making it a potential target for the development of new antibacterial drugs. Our exploration of P. aeruginosa (Pa)UGP structure-function relationships reveals that the activity of PaUGP depends on the formation of a tetrameric enzyme complex. We found that a molecular interface involved in tetramer formation is conserved in all bacterial UGPs but not in the human enzyme, and therefore hypothesize that it provides an ideal point of attack to selectively inhibit bacterial UGPs and exploit them as drug targets.


Assuntos
Infecções por Pseudomonas , Fatores de Virulência , Animais , Humanos , Fatores de Virulência/genética , Pseudomonas aeruginosa , Antibacterianos/farmacologia , Infecções por Pseudomonas/microbiologia , Glucose , Difosfato de Uridina
3.
Sci Rep ; 6: 24138, 2016 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-27053240

RESUMO

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.


Assuntos
Células Epiteliais/efeitos dos fármacos , Vírus da Parainfluenza 3 Humana/efeitos dos fármacos , Suramina/farmacologia , Zanamivir/farmacologia , Animais , Antivirais/metabolismo , Antivirais/farmacologia , Sítios de Ligação , Linhagem Celular , Sinergismo Farmacológico , Células Epiteliais/virologia , Proteína HN/metabolismo , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Rim/citologia , Cinética , Macaca mulatta , Espectroscopia de Ressonância Magnética , Simulação de Dinâmica Molecular , Vírus da Parainfluenza 3 Humana/metabolismo , Vírus da Parainfluenza 3 Humana/fisiologia , Ligação Proteica , Suramina/metabolismo , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/metabolismo , Replicação Viral/efeitos dos fármacos , Zanamivir/metabolismo
4.
Nat Commun ; 5: 5268, 2014 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-25327774

RESUMO

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.


Assuntos
Antivirais/química , Desenho de Fármacos , Hemaglutininas Virais/química , Neuraminidase/antagonistas & inibidores , Vírus da Parainfluenza 3 Humana , Domínio Catalítico , Linhagem Celular Tumoral , Simulação por Computador , Cristalografia por Raios X , Inibidores Enzimáticos/química , Ensaio de Imunoadsorção Enzimática , Mapeamento de Epitopos , Humanos , Concentração Inibidora 50 , Espectroscopia de Ressonância Magnética , Conformação Molecular , Simulação de Dinâmica Molecular , Ácidos Neuramínicos/química , Proteínas Recombinantes/química , Solventes/química , Propriedades de Superfície
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA