Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros











Intervalo de año de publicación
1.
Antiviral Res ; 210: 105506, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36565756

RESUMEN

Massive efforts on both vaccine development and antiviral research were launched to combat the new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We contributed, amongst others, by the development of a high-throughput screening (HTS) antiviral assay against SARS-CoV-2 using a fully automated, high-containment robot system. Here, we describe the development of this novel, convenient and phenotypic dual-reporter virus-cell-based high-content imaging assay using the A549+hACE2+TMPRSS2_mCherry reporter lung carcinoma cell line and an ancestral SARS-CoV-2_Wuhan_mNeonGreen reporter virus. Briefly, by means of clonal selection, a host cell subclone was selected that (i) efficiently supports replication of the reporter virus with high expression, upon infection, of the NeonGreen fluorescent reporter protein, (ii) that is not affected by virus-induced cytopathogenic effects and, (iii) that expresses a strong fluorescent mCherry signal in the nucleus. The selected clone matched these criteria with an infection rate on average of 75% with limited cell death. The average (R)Z'-factors of the assay plates were all >0.8, which indicates a robust assay suitable for HTS purposes. A selection of reference compounds that inhibits SARS-CoV-2 replication in vitro were used to validate this novel dual-reporter assay and confirms the data reported in the literature. This assay is a convenient and powerful tool for HTS of large compound libraries against SARS-CoV-2.


Asunto(s)
Antivirales , COVID-19 , Humanos , Antivirales/farmacología , Antivirales/metabolismo , Ensayos Analíticos de Alto Rendimiento/métodos , SARS-CoV-2 , Descubrimiento de Drogas , Replicación Viral
2.
Mol Ther ; 30(9): 2952-2967, 2022 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-35546782

RESUMEN

The COVID-19 pandemic continues to have devastating consequences on health and economy, even after the approval of safe and effective vaccines. Waning immunity, the emergence of variants of concern, breakthrough infections, and lack of global vaccine access and acceptance perpetuate the epidemic. Here, we demonstrate that a single injection of an adenoassociated virus (AAV)-based COVID-19 vaccine elicits at least 17-month-long neutralizing antibody responses in non-human primates at levels that were previously shown to protect from viral challenge. To improve the scalability of this durable vaccine candidate, we further optimized the vector design for greater potency at a reduced dose in mice and non-human primates. Finally, we show that the platform can be rapidly adapted to other variants of concern to robustly maintain immunogenicity and protect from challenge. In summary, we demonstrate this class of AAV can provide durable immunogenicity, provide protection at dose that is low and scalable, and be adapted readily to novel emerging vaccine antigens thus may provide a potent tool in the ongoing fight against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2).


Asunto(s)
COVID-19 , Vacunas Virales , Animales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , COVID-19/prevención & control , Vacunas contra la COVID-19 , Dependovirus/genética , Humanos , Macaca , Ratones , Pandemias/prevención & control , SARS-CoV-2/genética
3.
Nat Genet ; 53(4): 435-444, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33686287

RESUMEN

The ongoing COVID-19 pandemic has caused a global economic and health crisis. To identify host factors essential for coronavirus infection, we performed genome-wide functional genetic screens with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and human coronavirus 229E. These screens uncovered virus-specific as well as shared host factors, including TMEM41B and PI3K type 3. We discovered that SARS-CoV-2 requires the lysosomal protein TMEM106B to infect human cell lines and primary lung cells. TMEM106B overexpression enhanced SARS-CoV-2 infection as well as pseudovirus infection, suggesting a role in viral entry. Furthermore, single-cell RNA-sequencing of airway cells from patients with COVID-19 demonstrated that TMEM106B expression correlates with SARS-CoV-2 infection. The present study uncovered a collection of coronavirus host factors that may be exploited to develop drugs against SARS-CoV-2 infection or future zoonotic coronavirus outbreaks.


Asunto(s)
COVID-19/genética , Sistemas CRISPR-Cas , Genoma Humano/genética , Estudio de Asociación del Genoma Completo/métodos , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Líquido del Lavado Bronquioalveolar/citología , COVID-19/epidemiología , COVID-19/virología , Línea Celular Tumoral , Células Cultivadas , Coronavirus Humano 229E/genética , Epidemias , Células Epiteliales/virología , Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Provirus/fisiología , SARS-CoV-2/fisiología , Internalización del Virus
4.
mBio ; 10(4)2019 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-31409686

RESUMEN

Encephalomyocarditis virus (EMCV) is an animal pathogen and an important model organism, whose receptor requirements are poorly understood. Here, we employed a genome-wide haploid genetic screen to identify novel EMCV host factors. In addition to the previously described picornavirus receptors sialic acid and glycosaminoglycans, this screen unveiled important new host factors for EMCV. These factors include components of the fibroblast growth factor (FGF) signaling pathway, such as the potential receptors FGFR1 and ADAM9, a cell-surface metalloproteinase. By employing various knockout cells, we confirmed the importance of the identified host factors for EMCV infection. The largest reduction in infection efficiency was observed in cells lacking ADAM9. Pharmacological inhibition of the metalloproteinase activity of ADAM9 did not affect virus infection. Moreover, reconstitution of inactive ADAM9 in knockout cells restored susceptibility to EMCV, pointing to a proteinase-independent role of ADAM9 in mediating EMCV infection. Using neutralization assays with ADAM9-specific antiserum and soluble receptor proteins, we provided evidence for a role of ADAM9 in EMCV entry. Finally, binding assays showed that ADAM9 facilitates attachment of EMCV to the cell surface. Together, our findings reveal a role for ADAM9 as a novel receptor or cofactor for EMCV.IMPORTANCE EMCV is an animal pathogen that causes acute viral infections, usually myocarditis or encephalitis. It is thought to circulate mainly among rodents, from which it is occasionally transmitted to other animal species, including humans. EMCV causes fatal outbreaks of myocarditis and encephalitis in pig farms and zoos, making it an important veterinary pathogen. Although EMCV has been widely used as a model to study mechanisms of viral disease in mice, little is known about its entry mechanism. Here, we employ a haploid genetic screen for EMCV host factors and identify an essential role for ADAM9 in EMCV entry.


Asunto(s)
Proteínas ADAM/metabolismo , Infecciones por Cardiovirus/virología , Virus de la Encefalomiocarditis/fisiología , Proteínas de la Membrana/metabolismo , Internalización del Virus , Proteínas ADAM/antagonistas & inhibidores , Proteínas ADAM/genética , Animales , Infecciones por Cardiovirus/metabolismo , Línea Celular Tumoral , Membrana Celular/metabolismo , Virus de la Encefalomiocarditis/metabolismo , Técnicas de Inactivación de Genes , Genoma Humano/genética , Humanos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Ratones , Acoplamiento Viral , Replicación Viral
5.
Nat Commun ; 10(1): 3171, 2019 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-31320648

RESUMEN

Enteroviruses are a major cause of human disease. Adipose-specific phospholipase A2 (PLA2G16) was recently identified as a pan-enterovirus host factor and potential drug target. In this study, we identify a possible mechanism of PLA2G16 evasion by employing a dual glycan receptor-binding enterovirus D68 (EV-D68) strain. We previously showed that this strain does not strictly require the canonical EV-D68 receptor sialic acid. Here, we employ a haploid screen to identify sulfated glycosaminoglycans (sGAGs) as its second glycan receptor. Remarkably, engagement of sGAGs enables this virus to bypass PLA2G16. Using cryo-EM analysis, we reveal that, in contrast to sialic acid, sGAGs stimulate genome release from virions via structural changes that enlarge the putative openings for genome egress. Together, we describe an enterovirus that can bypass PLA2G16 and identify additional virion destabilization as a potential mechanism to circumvent PLA2G16.


Asunto(s)
Enterovirus Humano D/crecimiento & desarrollo , Glicosaminoglicanos/metabolismo , Fosfolipasas A2 Calcio-Independiente/metabolismo , Receptores Virales/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Internalización del Virus , Desencapsidación Viral/fisiología , Línea Celular Tumoral , Microscopía por Crioelectrón , Enterovirus Humano D/genética , Infecciones por Enterovirus/patología , Genoma Viral/genética , Células HEK293 , Células HeLa , Humanos , Ácido N-Acetilneuramínico/metabolismo
6.
Nature ; 541(7637): 412-416, 2017 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-28077878

RESUMEN

Picornaviruses are a leading cause of human and veterinary infections that result in various diseases, including polio and the common cold. As archetypical non-enveloped viruses, their biology has been extensively studied. Although a range of different cell-surface receptors are bound by different picornaviruses, it is unclear whether common host factors are needed for them to reach the cytoplasm. Using genome-wide haploid genetic screens, here we identify the lipid-modifying enzyme PLA2G16 (refs 8, 9, 10, 11) as a picornavirus host factor that is required for a previously unknown event in the viral life cycle. We find that PLA2G16 functions early during infection, enabling virion-mediated genome delivery into the cytoplasm, but not in any virion-assigned step, such as cell binding, endosomal trafficking or pore formation. To resolve this paradox, we screened for suppressors of the ΔPLA2G16 phenotype and identified a mechanism previously implicated in the clearance of intracellular bacteria. The sensor of this mechanism, galectin-8 (encoded by LGALS8), detects permeated endosomes and marks them for autophagic degradation, whereas PLA2G16 facilitates viral genome translocation and prevents clearance. This study uncovers two competing processes triggered by virus entry: activation of a pore-activated clearance pathway and recruitment of a phospholipase to enable genome release.


Asunto(s)
Citoplasma/virología , Genoma Viral , Factores Celulares Derivados del Huésped/metabolismo , Fosfolipasas A2 Calcio-Independiente/metabolismo , Picornaviridae/genética , Picornaviridae/fisiología , Proteínas Supresoras de Tumor/metabolismo , Internalización del Virus , Animales , Autofagia , Transporte Biológico , Línea Celular , Citoplasma/genética , Endosomas/metabolismo , Femenino , Galectinas/genética , Galectinas/metabolismo , Factores Celulares Derivados del Huésped/deficiencia , Factores Celulares Derivados del Huésped/genética , Humanos , Masculino , Ratones , Mutación , Fenotipo , Fosfolipasas A2 Calcio-Independiente/deficiencia , Fosfolipasas A2 Calcio-Independiente/genética , Supresión Genética , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética , Virión/genética , Virión/metabolismo , Replicación Viral
7.
Free Radic Biol Med ; 97: 223-235, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27288283

RESUMEN

UNLABELLED: 9-Norbornyl-6-chloropurine (NCP) is a representative of a series of antienteroviral bicycle derivatives with selective cytotoxicity towards leukemia cell lines. In this work we explored the mechanism of the antileukemic activity of NCP in T-cell lymphoblast cells (CCRF-CEM). Specifically, we searched for a potential link between its ability to induce cell death on the one hand and to modulate intracellular glutathione (GSH) that is necessary to its metabolic transformation via glutathione-S-transferase on the other hand. We have observed that GSH levels decreased rapidly in NCP-treated cells. Despite a complete regeneration following 24h of incubation with NCP, this profound drop in cellular GSH content triggered ER stress, ROS production and lipid peroxidation leading to the loss of mitochondrial membrane potential (MMP). These events induced concentration-dependent cell cycle arrest in G2/M phase and apoptosis. Both MMP loss and apoptosis were reversed by sulfhydryl-containing compounds (GSH, N-acetyl-l-cysteine). Furthermore, we have also shown that NCP-induced GSH decrease activated the Nrf2 pathway and its downstream targets NAD(P)H: quinone oxidoreductase (NQO-1) and glutamate cysteine ligase modifier subunit (GCLm), thus explaining the fast restoration of GSH pool and ROS decrease. Importantly, we confirmed that the cell death-inducing properties of the compounds were co-dependent on their ability to diminish cellular GSH level by analyzing the relationships between the GSH-depleting potency and cytotoxicity in a series of other norbornylpurine analogs. Altogether, the results demonstrated that in CCRF-CEM cells NCP triggered apoptosis through GSH depletion-associated oxidative and ER stress and mitochondrial depolarization.


Asunto(s)
Glutatión Transferasa/genética , Leucemia de Células T/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Purinas/administración & dosificación , Apoptosis/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Glutamato-Cisteína Ligasa/genética , Glutatión/metabolismo , Humanos , Leucemia de Células T/genética , Leucemia de Células T/metabolismo , Leucemia de Células T/patología , Peroxidación de Lípido/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , NAD(P)H Deshidrogenasa (Quinona)/genética , Especies Reactivas de Oxígeno/metabolismo
8.
Eur Heart J ; 36(42): 2909-19, 2015 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-26206211

RESUMEN

AIMS: Viral myocarditis (VM) is an important cause of heart failure and sudden cardiac death in young healthy adults; it is also an aetiological precursor of dilated cardiomyopathy. We explored the role of the miR-221/-222 family that is up-regulated in VM. METHODS AND RESULTS: Here, we show that microRNA-221 (miR-221) and miR-222 levels are significantly elevated during acute VM caused by Coxsackievirus B3 (CVB3). Both miRs are expressed by different cardiac cells and by infiltrating inflammatory cells, but their up-regulation upon myocarditis is mostly exclusive for the cardiomyocyte. Systemic inhibition of miR-221/-222 in mice increased cardiac viral load, prolonged the viraemic state, and strongly aggravated cardiac injury and inflammation. Similarly, in vitro, overexpression of miR-221 and miR-222 inhibited enteroviral replication, whereas knockdown of this miR-cluster augmented viral replication. We identified and confirmed a number of miR-221/-222 targets that co-orchestrate the increased viral replication and inflammation, including ETS1/2, IRF2, BCL2L11, TOX, BMF, and CXCL12. In vitro inhibition of IRF2, TOX, or CXCL12 in cardiomyocytes significantly dampened their inflammatory response to CVB3 infection, confirming the functionality of these targets in VM and highlighting the importance of miR-221/-222 as regulators of the cardiac response to VM. CONCLUSIONS: The miR-221/-222 cluster orchestrates the antiviral and inflammatory immune response to viral infection of the heart. Its inhibition increases viral load, inflammation, and overall cardiac injury upon VM.


Asunto(s)
Infecciones por Coxsackievirus/virología , MicroARNs/fisiología , Miocarditis/virología , Animales , Infecciones por Coxsackievirus/inmunología , Humanos , Inmunidad Celular/inmunología , Macrófagos/inmunología , Masculino , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , MicroARNs/antagonistas & inhibidores , MicroARNs/metabolismo , Miocarditis/inmunología , Miocitos Cardíacos/inmunología , Linfocitos T/inmunología , Regulación hacia Arriba , Carga Viral/inmunología , Replicación Viral/inmunología
9.
Antiviral Res ; 117: 110-4, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25752737

RESUMEN

Enteroviruses, e.g., polio-, coxsackie- and rhinoviruses, constitute a large genus within the Picornaviridae family of positive-strand RNA viruses and include many important pathogens linked to a variety of acute and chronic diseases. Despite their huge medical and economic impact, no approved antiviral therapy is yet available. Recently, the oxysterol-binding protein (OSBP) was implicated as a host factor for enterovirus replication. Here, we investigated the antiviral activity of the natural compound OSW-1, a ligand of OSBP that is under investigation as an anti-cancer drug. OSW-1 potently inhibited the replication of all enteroviruses tested, with IC50 values in the low nanomolar range, acted at the genome replication stage and was effective in all tested cell types of three different species. Importantly, OSBP overexpression rescued viral replication, demonstrating that the antiviral effect of OSW-1 is due to targeting OSBP. Together, we here report the anti-enterovirus activity of the natural anti-cancer compound OSW-1.


Asunto(s)
Antivirales/farmacología , Colestenonas/farmacología , Enterovirus/efectos de los fármacos , Receptores de Esteroides/metabolismo , Saponinas/farmacología , Replicación Viral/efectos de los fármacos , Animales , Línea Celular , Chlorocebus aethiops , Enterovirus/crecimiento & desarrollo , Genoma Viral/efectos de los fármacos , Células HeLa , Humanos , Ligandos , Células Vero , Carga Viral/efectos de los fármacos
10.
Cell Rep ; 10(4): 600-15, 2015 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-25640182

RESUMEN

Itraconazole (ITZ) is a well-known antifungal agent that also has anticancer activity. In this study, we identify ITZ as a broad-spectrum inhibitor of enteroviruses (e.g., poliovirus, coxsackievirus, enterovirus-71, rhinovirus). We demonstrate that ITZ inhibits viral RNA replication by targeting oxysterol-binding protein (OSBP) and OSBP-related protein 4 (ORP4). Consistently, OSW-1, a specific OSBP/ORP4 antagonist, also inhibits enterovirus replication. Knockdown of OSBP inhibits virus replication, whereas overexpression of OSBP or ORP4 counteracts the antiviral effects of ITZ and OSW-1. ITZ binds OSBP and inhibits its function, i.e., shuttling of cholesterol and phosphatidylinositol-4-phosphate between membranes, thereby likely perturbing the virus-induced membrane alterations essential for viral replication organelle formation. ITZ also inhibits hepatitis C virus replication, which also relies on OSBP. Together, these data implicate OSBP/ORP4 as molecular targets of ITZ and point to an essential role of OSBP/ORP4-mediated lipid exchange in virus replication that can be targeted by antiviral drugs.


Asunto(s)
Enterovirus/efectos de los fármacos , Enterovirus/metabolismo , Itraconazol/farmacología , Receptores de Esteroides/metabolismo , Replicación Viral/efectos de los fármacos , Antivirales/farmacología , Línea Celular Tumoral , Humanos
11.
Antiviral Res ; 110: 1-9, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25043639

RESUMEN

A novel small molecule, H1PVAT, was identified as a potent and selective inhibitor of the in vitro replication of all three poliovirus serotypes, whereas no activity was observed against other enteroviruses. Time-of-drug-addition studies revealed that the compound interfered with an early stage of virus replication. Four independently-selected H1PVAT-resistant virus variants uniformly carried the single amino acid substitution I194F in the VP1 capsid protein. Poliovirus type 1 strain Sabin, reverse-engineered to contain this substitution, proved to be completely insensitive to the antiviral effect of H1PVAT and was cross-resistant to the capsid-binding inhibitors V-073 and pirodavir. The VP1 I194F mutant had a smaller plaque phenotype than wild-type virus, and the amino acid substitution rendered the virus more susceptible to heat inactivation. Both for the wild-type and VP1 I194F mutant virus, the presence of H1PVAT increased the temperature at which the virus was inactivated, providing evidence that the compound interacts with the viral capsid, and that capsid stabilization and antiviral activity are not necessarily correlated. Molecular modeling suggested that H1PVAT binds with high affinity in the pocket underneath the floor of the canyon that is involved in receptor binding. Introduction of the I194F substitution in the model of VP1 induced a slight concerted rearrangement of the core ß-barrel in this pocket, which disfavors binding of the compound. Taken together, the compound scaffold, to which H1PVAT belongs, may represent another promising class of poliovirus capsid-binding inhibitors next to V-073 and pirodavir. Potent antivirals against poliovirus will be essential in the poliovirus eradication end-game.


Asunto(s)
Antivirales/farmacología , Proteínas de la Cápside/antagonistas & inhibidores , Poliomielitis/tratamiento farmacológico , Pirazoles/farmacología , Pirimidinas/farmacología , Replicación Viral/efectos de los fármacos , Sustitución de Aminoácidos/genética , Animales , Secuencia de Bases , Sitios de Unión , Cápside/efectos de los fármacos , Proteínas de la Cápside/genética , Línea Celular Tumoral , Chlorocebus aethiops , Farmacorresistencia Viral , Células HeLa , Humanos , Modelos Moleculares , Piperidinas/farmacología , Poliovirus/efectos de los fármacos , Poliovirus/genética , Piridazinas/farmacología , ARN Viral/genética , Análisis de Secuencia de ARN
12.
PLoS Pathog ; 10(4): e1004039, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24722756

RESUMEN

Enteroviruses (family of the Picornaviridae) cover a large group of medically important human pathogens for which no antiviral treatment is approved. Although these viruses have been extensively studied, some aspects of the viral life cycle, in particular morphogenesis, are yet poorly understood. We report the discovery of TP219 as a novel inhibitor of the replication of several enteroviruses, including coxsackievirus and poliovirus. We show that TP219 binds directly glutathione (GSH), thereby rapidly depleting intracellular GSH levels and that this interferes with virus morphogenesis without affecting viral RNA replication. The inhibitory effect on assembly was shown not to depend on an altered reducing environment. Using TP219, we show that GSH is an essential stabilizing cofactor during the transition of protomeric particles into pentameric particles. Sequential passaging of coxsackievirus B3 in the presence of low GSH-levels selected for GSH-independent mutants that harbored a surface-exposed methionine in VP1 at the interface between two protomers. In line with this observation, enteroviruses that already contained this surface-exposed methionine, such as EV71, did not rely on GSH for virus morphogenesis. Biochemical and microscopical analysis provided strong evidence for a direct interaction between GSH and wildtype VP1 and a role for this interaction in localizing assembly intermediates to replication sites. Consistently, the interaction between GSH and mutant VP1 was abolished resulting in a relocalization of the assembly intermediates to replication sites independent from GSH. This study thus reveals GSH as a novel stabilizing host factor essential for the production of infectious enterovirus progeny and provides new insights into the poorly understood process of morphogenesis.


Asunto(s)
Cápside/metabolismo , Enterovirus Humano B/fisiología , Infecciones por Enterovirus/metabolismo , Glutatión/metabolismo , ARN Viral/biosíntesis , Replicación Viral/fisiología , Animales , Chlorocebus aethiops , Infecciones por Enterovirus/genética , Glutatión/genética , Células HeLa , Humanos , Mutación , ARN Viral/genética , Células Vero
13.
Eur J Med Chem ; 49: 279-88, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22305341

RESUMEN

To further explore the anti-enteroviral activity of 9-aryl-6-chloropurines, three different series of compounds with a dialkylamino, (alkyl)amido, or oxazolidinone substituent at the aryl ring have been synthesized, in most cases with the aid of microwave-assisted synthesis. The resulting compounds efficiently inhibit Coxsackie virus type B3 (CVB3) replication with EC(50) values varying from 3 to 15 µM, and with no significant toxicity in Vero cells. The most potent compounds also selectively inhibit the replication of other enteroviruses including Coxsackie virus B4 and Echo virus 11. The cross-resistance studies performed with different 9-aryl-6-chloropurines indicate that they all belong to the same pharmacological family and differ from other CVB3 drugs such as enviroxime.


Asunto(s)
Antivirales/química , Antivirales/farmacología , Enterovirus Humano B/efectos de los fármacos , Oxazolidinonas/química , Oxazolidinonas/farmacología , Purinas/química , Purinas/farmacología , Animales , Antivirales/síntesis química , Línea Celular , Chlorocebus aethiops , Infecciones por Enterovirus/tratamiento farmacológico , Halogenación , Humanos , Pruebas de Sensibilidad Microbiana , Microondas , Modelos Moleculares , Oxazolidinonas/síntesis química , Purinas/síntesis química , Células Vero
14.
Antimicrob Agents Chemother ; 53(5): 1850-7, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19237651

RESUMEN

A novel compound, TTP-8307, was identified as a potent inhibitor of the replication of several rhino- and enteroviruses. TTP-8307 inhibits viral RNA synthesis in a dose-dependent manner, without affecting polyprotein synthesis and/or processing. Drug-resistant variants of coxsackievirus B3 were all shown to carry at least one amino acid mutation in the nonstructural protein 3A. In particular, three mutations located in a nonstructured region preceding the hydrophobic domain (V45A, I54F, and H57Y) appeared to contribute to the drug-resistant phenotype. This region has previously been identified as a hot sport for mutations that resulted in resistance to enviroxime, the sole 3A-targeting enterovirus inhibitor reported thus far. This was corroborated by the fact that TTP-8307 and enviroxime proved cross-resistant. It is hypothesized that TTP-8307 and enviroxime disrupt proper interactions of 3A(B) with other viral or cellular proteins that are required for efficient replication.


Asunto(s)
Antivirales/farmacología , Farmacorresistencia Viral/genética , Enterovirus/efectos de los fármacos , Mutación , Proteínas no Estructurales Virales/genética , Replicación Viral/efectos de los fármacos , Animales , Antivirales/química , Bencimidazoles/química , Bencimidazoles/farmacología , Chlorocebus aethiops , Enterovirus/genética , Enterovirus/metabolismo , Enterovirus/fisiología , Enterovirus Humano B/efectos de los fármacos , Enterovirus Humano B/genética , Enterovirus Humano B/metabolismo , Enterovirus Humano B/fisiología , Células HeLa/virología , Humanos , Pruebas de Sensibilidad Microbiana/métodos , Oximas , Poliovirus/efectos de los fármacos , Poliovirus/genética , Rhinovirus/efectos de los fármacos , Rhinovirus/genética , Rhinovirus/metabolismo , Rhinovirus/fisiología , Sulfonamidas , Células Vero/virología , Proteínas no Estructurales Virales/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA