Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Molecules ; 28(1)2022 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-36615354

RESUMEN

The Coronavirus Disease 2019 (COVID-19) and dengue fever (DF) pandemics both remain to be significant public health concerns in the foreseeable future. Anti-SARS-CoV-2 drugs and vaccines are both indispensable to eliminate the epidemic situation. Here, two piperazine-based polyphenol derivatives DF-47 and DF-51 were identified as potential inhibitors directly blocking the active site of SARS-CoV-2 and DENV RdRp. Data through RdRp inhibition screening of an in-house library and in vitro antiviral study selected DF-47 and DF-51 as effective inhibitors of SARS-CoV-2/DENV polymerase. Moreover, in silico simulation revealed stable binding modes between the DF-47/DF-51 and SARS-CoV-2/DENV RdRp, respectively, including chelating with Mg2+ near polymerase active site. This work discovered the inhibitory effect of two polyphenols on distinct viral RdRp, which are expected to be developed into broad-spectrum, non-nucleoside RdRp inhibitors with new scaffold.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/metabolismo , Polifenoles/farmacología , ARN Polimerasa Dependiente del ARN/metabolismo , Antivirales/química , Simulación del Acoplamiento Molecular
2.
J Virol ; 94(2)2020 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-31666384

RESUMEN

To counteract the serious health threat posed by known and novel viral pathogens, drugs that target a variety of viruses through a common mechanism have attracted recent attention due to their potential in treating (re)emerging infections, for which direct-acting antivirals are not available. We found that labyrinthopeptins A1 and A2, the prototype congeners of carbacyclic lanthipeptides, inhibit the proliferation of diverse enveloped viruses, including dengue virus, Zika virus, West Nile virus, hepatitis C virus, chikungunya virus, Kaposi's sarcoma-associated herpesvirus, cytomegalovirus, and herpes simplex virus, in the low micromolar to nanomolar range. Mechanistic studies on viral particles revealed that labyrinthopeptins induce a virolytic effect through binding to the viral membrane lipid phosphatidylethanolamine (PE). These effects are enhanced by a combined equimolar application of both labyrinthopeptins, and a clear synergism was observed across a concentration range corresponding to 10% to 90% inhibitory concentrations of the compounds. Time-resolved experiments with large unilamellar vesicles (LUVs) reveal that membrane lipid raft compositions (phosphatidylcholine [PC]/PE/cholesterol/sphingomyelin at 17:10:33:40) are particularly sensitive to labyrinthopeptins in comparison to PC/PE (90:10) LUVs, even though the overall PE amount remains constant. Labyrinthopeptins exhibited low cytotoxicity and had favorable pharmacokinetic properties in mice (half-life [t1/2] = 10.0 h), which designates them promising antiviral compounds acting by an unusual viral lipid targeting mechanism.IMPORTANCE For many viral infections, current treatment options are insufficient. Because the development of each antiviral drug is time-consuming and expensive, the prospect of finding broad-spectrum antivirals that can fight multiple, diverse viruses-well-known viruses as well as (re)emerging species-has gained attention, especially for the treatment of viral coinfections. While most known broad-spectrum agents address processes in the host cell, we found that targeting lipids of the free virus outside the host cell with the natural products labyrinthopeptin A1 and A2 is a viable strategy to inhibit the proliferation of a broad range of viruses from different families, including chikungunya virus, dengue virus, Zika virus, Kaposi's sarcoma-associated herpesvirus, and cytomegalovirus. Labyrinthopeptins bind to viral phosphatidylethanolamine and induce virolysis without exerting cytotoxicity on host cells. This represents a novel and unusual mechanism to tackle medically relevant viral infections.


Asunto(s)
Bacteriocinas/farmacología , Microdominios de Membrana/metabolismo , Virosis/metabolismo , Virus/metabolismo , Aedes , Animales , Línea Celular , Microdominios de Membrana/virología , Fosfatidiletanolaminas/metabolismo , Virosis/tratamiento farmacológico
3.
J Vis Exp ; (193)2023 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-37036247

RESUMEN

Cell-based electrical impedance (CEI) technology measures changes in impedance caused by a growing or manipulated adherent cell monolayer on culture plate wells embedded with electrodes. The technology can be used to monitor the consequences of Zika virus (ZIKV) infection and adherent cell replication in real time, as this virus is highly cytopathogenic. It is a straightforward assay that does not require the use of labels or invasive methods and has the benefit of providing real-time data. The kinetics of ZIKV infection are highly dependent on the employed cell line, virus strain, and multiplicity of infection (MOI), which cannot be easily studied with conventional endpoint assays. Furthermore, the CEI assay can also be used for the evaluation and characterization of antiviral compounds, which can also have dynamic inhibitory properties over the course of infection. This methods article gives a detailed explanation of the practical execution of the CEI assay and its potential applications in ZIKV research and antiviral research in general.


Asunto(s)
Infección por el Virus Zika , Virus Zika , Chlorocebus aethiops , Animales , Virus Zika/metabolismo , Impedancia Eléctrica , Replicación Viral , Antivirales/farmacología , Células Vero
4.
Microbiol Spectr ; 10(4): e0049122, 2022 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-35862960

RESUMEN

In this study, we use electric cell-substrate impedance sensing (ECIS), an established cell-based electrical impedance (CEI) technology, to decipher the kinetic cytopathic effect (CPE) induced by Zika virus (ZIKV) in susceptible human A549 lung epithelial cells and to evaluate several classes of compounds with reported antiviral activity (two entry inhibitors and two replication inhibitors). To validate the assay, we compare the results with those obtained with more traditional in vitro methods based on cell viability and viral yield readouts. We demonstrate that CEI can detect viral infection in a sensitive manner and can be used to determine antiviral potency. Moreover, CEI has multiple benefits compared to conventional assays: the technique is less laborious and better at visualizing the dynamic antiviral activity profile of the compounds, while also it has the ability to determine interesting time points that can be selected as endpoints in assays without continuous readout. We describe several parameters to characterize the compounds' cytotoxicity and their antiviral activity profile. In addition, the CEI patterns provide valuable additional information about the presumed mechanism of action of these compounds. Finally, as a proof of concept, we used CEI to evaluate the antiviral activity of a small series of compounds, for which we demonstrate that the sulfonated polymer PRO2000 inhibits ZIKV with a response profile representative for a viral entry inhibitor. Overall, we demonstrate for the first time that CEI is a powerful technology to evaluate and characterize compounds against ZIKV replication in a real-time, label-free, and noninvasive manner. IMPORTANCE Zika virus can cause serious disease in humans. Unfortunately, no antiviral drugs are available to treat infection. Here, we use an impedance-based method to continuously monitor virus infection in-and damage to-human cells. We can determine the Zika viral dose with this technique and also evaluate whether antiviral compounds protect the cells from damage caused by virus replication. We also show that this technique can be used to further unravel the characteristics of these compounds, such as their toxicity to the cells, and that it might even give further insight in their mechanism of antiviral action. Finally, we also find a novel Zika virus inhibitor, PRO2000. Overall, in this study, we use the impedance technology to-for the first time-evaluate compounds with anti-Zika virus properties, and therefore it can add valuable information in the further search for antiviral drugs.


Asunto(s)
Infección por el Virus Zika , Virus Zika , Animales , Antivirales/farmacología , Antivirales/uso terapéutico , Chlorocebus aethiops , Impedancia Eléctrica , Humanos , Células Vero , Replicación Viral , Infección por el Virus Zika/tratamiento farmacológico
5.
Virology ; 562: 74-86, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34274562

RESUMEN

To date, there are no broad-spectrum antivirals available to treat infections with flaviviruses such as dengue (DENV) and Zika virus (ZIKV). In this study, we determine the broad antiviral activity of the lantibiotic Labyrinthopeptin A1. We show that Laby A1 inhibits all DENV serotypes and various ZIKV strains with IC50 around 1 µM. The structurally related Laby A2 also displayed a consistent, but about tenfold lower, antiviral activity. Furthermore, Laby A1 inhibits many viruses from divergent families such as HIV, YFV, RSV and Punta Torovirus. Of interest, Laby A1 does not show activity against non-enveloped viruses. Its antiviral activity is independent of the cell line or the used evaluation method, and can also be observed in MDDC, a physiologically relevant primary cell type. Furthermore, Laby A1 demonstrates low cellular toxicity and has a more favorable SI compared to duramycin, a well-described lantibiotic with broad-spectrum antiviral activity. Time-of-drug addition experiments demonstrate that Laby A1 inhibits infection and entry processes of ZIKV and DENV. We reveal that Laby A1 performs its broad antiviral activity by interacting with a viral factor rather than a cellular factor, and that it has virucidal properties. Finally, using SPR interaction studies we demonstrate that Laby A1 interacts with several phospholipids (i.e. PE and PS) present in the viral envelope. Together with other recent Labyrinthopeptin antiviral publications, this work validates the activity of Laby A1 as broad antiviral entry inhibitor with a unique mechanism of action and demonstrates its potential value as antiviral agent against emerging flaviviruses.


Asunto(s)
Antivirales/farmacología , Bacteriocinas/farmacología , Virus del Dengue/efectos de los fármacos , Fosfolípidos/metabolismo , Envoltura Viral/efectos de los fármacos , Virus Zika/efectos de los fármacos , Animales , Antivirales/metabolismo , Bacteriocinas/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Péptidos/farmacología , Envoltura Viral/metabolismo , Internalización del Virus/efectos de los fármacos , Virus/clasificación , Virus/efectos de los fármacos
6.
Virus Res ; 274: 197760, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31618614

RESUMEN

In Gordts et al. (2015), we have shown that lignosulfonic acid, a commercially available lignin derivative, possesses broad antiviral activity against human immunodeficiency virus (HIV) and Herpes simplex virus (HSV) by preventing viral entry into susceptible target cells. Because of the interesting safety profile as potential microbicide, we now determined the antiviral activity of a series of lignosulfonates in order to understand better which molecular features can contribute to their antiviral activity. Here, 24 structurally different lignosulfonates were evaluated for their capacity to inhibit HIV and HSV transmission and replication in various cellular assays. These derivatives differ in origin (hardwood or softwood), counter-ion used during sulphite processing (Na+, Ca2+, or NH4+), sulphur content, carboxylic acid percentage, and molecular weight fraction, which allowed to determine structure-activity relationships. We demonstrate that the broad antiviral activity of lignosulfonates is mainly dependent on their molecular weight and that their mechanism of action is based on interactions with the viral envelope glycoproteins. This makes the lignosulfonates a potential low-cost microbicide that protects women from sexual HIV and HSV transmission and thus prevents life-long infection.


Asunto(s)
Antivirales/farmacología , Infecciones por VIH/prevención & control , VIH/efectos de los fármacos , Lignina/análogos & derivados , Animales , Antivirales/química , Línea Celular , Infecciones por VIH/transmisión , Herpes Simple/prevención & control , Herpes Simple/transmisión , Herpesvirus Humano 2/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Lignina/química , Lignina/farmacología , Fusión de Membrana/efectos de los fármacos , Estructura Molecular , Peso Molecular , Proteínas del Envoltorio Viral/química , Internalización del Virus/efectos de los fármacos
7.
Parasitol Int ; 66(2): 119-125, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28027967

RESUMEN

We performed a comparative phylogeographic study on the monogenean flatworm Gyrodactylus gondae Huyse, Malmberg & Volckaert 2005 (Gyrodactylidae) and its sand goby host Pomatoschistus minutus (Pallas, 1770) (Gobiidae). G. gondae is a host-specific parasite with a direct life cycle and a very short generation time. These properties are expected to increase the chance to track the genealogical history of the host with genetic data of the parasite ('magnifying glass principle'). To investigate this hypothesis we screened nine sand goby populations (n=326) along the Atlantic coasts of Europe for Gyrodactylus specimens. Low parasite prevalence resulted in partially overlapping host and parasite datasets. Ninety-two G. gondae collected on five sand goby populations were subsequently sequenced for a 460bp cytochrome c oxidase subunit II (coxII) fragment, which, in combination with previously published haplotype data for the hosts, allowed for partially overlapping host and parasite datasets. Haplotype diversity was lowest in the Irish Sea while nucleotide diversity was highest in the Southern North Sea. The host population also showed the lowest diversity in the Irish Sea but the highest nucleotide diversity, based on cytochrome b sequences of 850bp, was found in Skagerrak. Phylogeographic networks suggest postglacial expansion in both the host and the parasite. Pair-wise population differentiation was however not consistently higher in the parasite than in the host, rejecting the magnifying glass hypothesis for this host-parasite system. The parasite network offered limited resolution and was characterized by many extinctions and/or missing haplotypes, which could be attributed to 1) sampling bias, 2) size fluctuations in the parasite populations resulting in frequent extinctions and genetic drift and 3) the relatively young age of the host-parasite association. A more exhaustive study including a broader geographical and genomic coverage is needed to discriminate among these competing hypotheses.


Asunto(s)
Evolución Biológica , Enfermedades de los Peces/parasitología , Interacciones Huésped-Parásitos , Perciformes/parasitología , Trematodos/fisiología , Infecciones por Trematodos/veterinaria , Animales , Océano Atlántico , Complejo IV de Transporte de Electrones/genética , Variación Genética , Haplotipos , Filogenia , Filogeografía , Análisis de Secuencia de ADN , Trematodos/genética , Infecciones por Trematodos/parasitología
8.
PLoS One ; 12(4): e0176057, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28410420

RESUMEN

The chemokine receptor CXCR4 is activated by its unique chemokine ligand CXCL12 and regulates many physiological and developmental processes such as hematopoietic cell trafficking. CXCR4 is also one of the main co-receptors for human immunodeficiency virus (HIV) entry. Dysfunction of the CXCL12/CXCR4 axis contributes to several human pathologies, including cancer and inflammatory diseases. Consequently, inhibition of CXCR4 activation is recognized as an attractive target for therapeutic intervention. In this regard, numerous agents modifying CXCR4 activity have been evaluated in in vitro experimental studies and pre-clinical models. Here, we evaluated a CXCL12 competition binding assay for its potential as a valuable initial screen for functional and competitive CXCR4 inhibitors. In total, 11 structurally diverse compounds were included in a side-by-side comparison of in vitro CXCR4 cell-based assays, such as CXCL12 competition binding, CXCL12-induced calcium signaling, CXCR4 internalization, CXCL12-guided cell migration and CXCR4-specific HIV-1 replication experiments. Our data indicated that agents that inhibit CXCL12 binding, i.e. the anti-CXCR4 peptide analogs T22, T140 and TC14012 and the small molecule antagonists AMD3100, AMD3465, AMD11070 and IT1t showed inhibitory activity with consistent relative potencies in all further applied CXCR4-related assays. Accordingly, agents exerting no or very weak receptor binding (i.e., CTCE-9908, WZ811, Me6TREN and gambogic acid) showed no or very poor anti-CXCR4 inhibitory activity. Thus, CXCL12 competition binding studies were proven to be highly valuable as an initial screening assay and indicative for the pharmacological and functional profile of competitive CXCR4 antagonists, which will help the design of new potent CXCR4 inhibitors.


Asunto(s)
Fármacos Anti-VIH/metabolismo , Receptores CXCR4/antagonistas & inhibidores , Aminopiridinas/química , Aminopiridinas/metabolismo , Aminopiridinas/toxicidad , Fármacos Anti-VIH/química , Fármacos Anti-VIH/toxicidad , Bencilaminas/química , Bencilaminas/metabolismo , Bencilaminas/toxicidad , Unión Competitiva , Señalización del Calcio/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Quimiocina CXCL12/química , Quimiocina CXCL12/metabolismo , Ciclamas , Compuestos Heterocíclicos/química , Compuestos Heterocíclicos/metabolismo , Compuestos Heterocíclicos/toxicidad , Humanos , Células Jurkat , Unión Proteica , Piridinas/química , Piridinas/metabolismo , Piridinas/toxicidad , Receptores CXCR4/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA