Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Cell ; 175(7): 1917-1930.e13, 2018 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-30550789

RESUMEN

Ebola virus (EBOV) infection often results in fatal illness in humans, yet little is known about how EBOV usurps host pathways during infection. To address this, we used affinity tag-purification mass spectrometry (AP-MS) to generate an EBOV-host protein-protein interaction (PPI) map. We uncovered 194 high-confidence EBOV-human PPIs, including one between the viral transcription regulator VP30 and the host ubiquitin ligase RBBP6. Domain mapping identified a 23 amino acid region within RBBP6 that binds to VP30. A crystal structure of the VP30-RBBP6 peptide complex revealed that RBBP6 mimics the viral nucleoprotein (NP) binding to the same interface of VP30. Knockdown of endogenous RBBP6 stimulated viral transcription and increased EBOV replication, whereas overexpression of either RBBP6 or the peptide strongly inhibited both. These results demonstrate the therapeutic potential of biologics that target this interface and identify additional PPIs that may be leveraged for novel therapeutic strategies.


Asunto(s)
Proteínas Portadoras , Proteínas de Unión al ADN , Ebolavirus/fisiología , Fiebre Hemorrágica Ebola/metabolismo , Factores de Transcripción , Proteínas Virales , Replicación Viral/fisiología , Proteínas Portadoras/química , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Cristalografía por Rayos X , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células HEK293 , Células HeLa , Fiebre Hemorrágica Ebola/genética , Fiebre Hemorrágica Ebola/patología , Humanos , Mapeo de Interacción de Proteínas , Factores de Transcripción/química , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas , Proteínas Virales/química , Proteínas Virales/genética , Proteínas Virales/metabolismo
2.
EMBO J ; 40(18): e105658, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34260076

RESUMEN

The Ebola virus VP30 protein interacts with the viral nucleoprotein and with host protein RBBP6 via PPxPxY motifs that adopt non-canonical orientations, as compared to other proline-rich motifs. An affinity tag-purification mass spectrometry approach identified additional PPxPxY-containing host proteins hnRNP L, hnRNPUL1, and PEG10, as VP30 interactors. hnRNP L and PEG10, like RBBP6, inhibit viral RNA synthesis and EBOV infection, whereas hnRNPUL1 enhances. RBBP6 and hnRNP L modulate VP30 phosphorylation, increase viral transcription, and exert additive effects on viral RNA synthesis. PEG10 has more modest inhibitory effects on EBOV replication. hnRNPUL1 positively affects viral RNA synthesis but in a VP30-independent manner. Binding studies demonstrate variable capacity of the PPxPxY motifs from these proteins to bind VP30, define PxPPPPxY as an optimal binding motif, and identify the fifth proline and the tyrosine as most critical for interaction. Competition binding and hydrogen-deuterium exchange mass spectrometry studies demonstrate that each protein binds a similar interface on VP30. VP30 therefore presents a novel proline recognition domain that is targeted by multiple host proteins to modulate viral transcription.


Asunto(s)
Ebolavirus/fisiología , Fiebre Hemorrágica Ebola/metabolismo , Fiebre Hemorrágica Ebola/virología , Prolina/metabolismo , Tirosina/metabolismo , Proteínas Portadoras , Regulación Viral de la Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Unión Proteica , Replicación Viral
3.
Pharm Res ; 36(7): 104, 2019 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-31101988

RESUMEN

PURPOSE: Since the 2014 Ebola virus (EBOV) outbreak in West Africa there has been considerable effort towards developing drugs to treat Ebola virus disease and yet to date there is no FDA approved treatment. This is important as at the time of writing this manuscript there is an ongoing outbreak in the Democratic Republic of the Congo which has killed over 1000. METHODS: We have evaluated a small number of natural products, some of which had shown antiviral activity against other pathogens. This is exemplified with eugenol, which is found in high concentrations in multiple essential oils, and has shown antiviral activity against feline calicivirus, tomato yellow leaf curl virus, Influenza A virus, Herpes Simplex virus type 1 and 2, and four airborne phages. RESULTS: Four compounds possessed EC50 values less than or equal to 11 µM. Of these, eugenol, had an EC50 of 1.3 µM against EBOV and is present in several plants including clove, cinnamon, basil and bay. Eugenol is much smaller and structurally unlike any compound that has been previously identified as an inhibitor of EBOV, therefore it may provide new mechanistic insights. CONCLUSION: This compound is readily accessible in bulk quantities, is inexpensive, and has a long history of human consumption, which endorses the idea for further assessment as an antiviral therapeutic. This work also suggests that a more exhaustive assessment of natural product libraries against EBOV and other viruses is warranted to improve our ability to identify compounds that are so distinct from FDA approved drugs.


Asunto(s)
Antivirales/farmacología , Productos Biológicos/farmacología , Ebolavirus/efectos de los fármacos , Eugenol/farmacología , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Fiebre Hemorrágica Ebola/virología , Células HeLa , Humanos
4.
J Infect Dis ; 218(suppl_5): S612-S626, 2018 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-29860496

RESUMEN

Background: For most classes of drugs, rapid development of therapeutics to treat emerging infections is challenged by the timelines needed to identify compounds with the desired efficacy, safety, and pharmacokinetic profiles. Fully human monoclonal antibodies (mAbs) provide an attractive method to overcome many of these hurdles to rapidly produce therapeutics for emerging diseases. Methods: In this study, we deployed a platform to generate, test, and develop fully human antibodies to Zaire ebolavirus. We obtained specific anti-Ebola virus (EBOV) antibodies by immunizing VelocImmune mice that use human immunoglobulin variable regions in their humoral responses. Results: Of the antibody clones isolated, 3 were selected as best at neutralizing EBOV and triggering FcγRIIIa. Binding studies and negative-stain electron microscopy revealed that the 3 selected antibodies bind to non-overlapping epitopes, including a potentially new protective epitope not targeted by other antibody-based treatments. When combined, a single dose of a cocktail of the 3 antibodies protected nonhuman primates (NHPs) from EBOV disease even after disease symptoms were apparent. Conclusions: This antibody cocktail provides complementary mechanisms of actions, incorporates novel specificities, and demonstrates high-level postexposure protection from lethal EBOV disease in NHPs. It is now undergoing testing in normal healthy volunteers in preparation for potential use in future Ebola epidemics.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Neutralizantes/uso terapéutico , Anticuerpos Antivirales/uso terapéutico , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Animales , Anticuerpos Monoclonales/aislamiento & purificación , Glicoproteínas/inmunología , Cobayas , Células HEK293 , Humanos , Macaca mulatta , Masculino , Ratones
5.
PLoS Pathog ; 11(11): e1005263, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26562011

RESUMEN

Ebola virus outbreaks, such as the 2014 Makona epidemic in West Africa, are episodic and deadly. Filovirus antivirals are currently not clinically available. Our findings suggest interferon gamma, an FDA-approved drug, may serve as a novel and effective prophylactic or treatment option. Using mouse-adapted Ebola virus, we found that murine interferon gamma administered 24 hours before or after infection robustly protects lethally-challenged mice and reduces morbidity and serum viral titers. Furthermore, we demonstrated that interferon gamma profoundly inhibits Ebola virus infection of macrophages, an early cellular target of infection. As early as six hours following in vitro infection, Ebola virus RNA levels in interferon gamma-treated macrophages were lower than in infected, untreated cells. Addition of the protein synthesis inhibitor, cycloheximide, to interferon gamma-treated macrophages did not further reduce viral RNA levels, suggesting that interferon gamma blocks life cycle events that require protein synthesis such as virus replication. Microarray studies with interferon gamma-treated human macrophages identified more than 160 interferon-stimulated genes. Ectopic expression of a select group of these genes inhibited Ebola virus infection. These studies provide new potential avenues for antiviral targeting as these genes that have not previously appreciated to inhibit negative strand RNA viruses and specifically Ebola virus infection. As treatment of interferon gamma robustly protects mice from lethal Ebola virus infection, we propose that interferon gamma should be further evaluated for its efficacy as a prophylactic and/or therapeutic strategy against filoviruses. Use of this FDA-approved drug could rapidly be deployed during future outbreaks.


Asunto(s)
Antivirales/farmacología , Ebolavirus/efectos de los fármacos , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Interferón gamma/farmacología , Macrófagos/efectos de los fármacos , Animales , Células Cultivadas , Humanos , Macrófagos/metabolismo , Ratones Endogámicos BALB C , ARN Viral/genética , Replicación Viral/efectos de los fármacos
6.
Antimicrob Agents Chemother ; 60(8): 4471-81, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27161622

RESUMEN

Filoviruses are highly infectious, and no FDA-approved drug therapy for filovirus infection is available. Most work to find a treatment has involved only a few strains of Ebola virus and testing of relatively small drug libraries or compounds that have shown efficacy against other virus types. Here we report the findings of a high-throughput screening of 319,855 small molecules from the Molecular Libraries Small Molecule Repository library for their activities against Marburg virus and Ebola virus. Nine of the most potent, novel compounds that blocked infection by both viruses were analyzed in detail for their mechanisms of action. The compounds inhibited known key steps in the Ebola virus infection mechanism by blocking either cell surface attachment, macropinocytosis-mediated uptake, or endosomal trafficking. To date, very few specific inhibitors of macropinocytosis have been reported. The 2 novel macropinocytosis inhibitors are more potent inhibitors of Ebola virus infection and less toxic than ethylisopropylamiloride, one commonly accepted macropinocytosis inhibitor. Each compound blocked infection of primary human macrophages, indicating their potential to be developed as new antifiloviral therapies.


Asunto(s)
Antivirales/farmacología , Ebolavirus/efectos de los fármacos , Marburgvirus/efectos de los fármacos , Animales , Línea Celular , Chlorocebus aethiops , Ebolavirus/genética , Ebolavirus/metabolismo , Células HeLa , Humanos , Marburgvirus/genética , Marburgvirus/metabolismo , Células Vero
7.
Antimicrob Agents Chemother ; 60(8): 4552-62, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27185801

RESUMEN

Viral emergence and reemergence underscore the importance of developing efficacious, broad-spectrum antivirals. Here, we report the discovery of tetrahydrobenzothiazole-based compound 1, a novel, broad-spectrum antiviral lead that was optimized from a hit compound derived from a cytopathic effect (CPE)-based antiviral screen using Venezuelan equine encephalitis virus. Compound 1 showed antiviral activity against a broad range of RNA viruses, including alphaviruses, flaviviruses, influenza virus, and ebolavirus. Mechanism-of-action studies with metabolomics and molecular approaches revealed that the compound inhibits host pyrimidine synthesis and establishes an antiviral state by inducing a variety of interferon-stimulated genes (ISGs). Notably, the induction of the ISGs by compound 1 was independent of the production of type 1 interferons. The antiviral activity of compound 1 was cell type dependent with a robust effect observed in human cell lines and no observed antiviral effect in mouse cell lines. Herein, we disclose tetrahydrobenzothiazole compound 1 as a novel lead for the development of a broad-spectrum, antiviral therapeutic and as a molecular probe to study the mechanism of the induction of ISGs that are independent of type 1 interferons.


Asunto(s)
Antivirales/farmacología , Interferón Tipo I/metabolismo , Pirimidinas/biosíntesis , Línea Celular , VIH-1/efectos de los fármacos , Humanos , Reacción en Cadena en Tiempo Real de la Polimerasa , Replicación Viral/efectos de los fármacos
8.
Transfusion ; 56 Suppl 1: S6-15, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27001363

RESUMEN

BACKGROUND: Transfusion of plasma from recovered patients after Ebolavirus (EBOV) infection, typically called "convalescent plasma," is an effective treatment for active disease available in endemic areas, but carries the risk of introducing other pathogens, including other strains of EBOV. A pathogen reduction technology using ultraviolet light and riboflavin (UV+RB) is effective against multiple enveloped, negative-sense, single-stranded RNA viruses that are similar in structure to EBOV. We hypothesized that UV+RB is effective against EBOV in blood products without activating complement or reducing protective immunoglobulin titers that are important for the treatment of Ebola virus disease (EVD). STUDY DESIGN AND METHODS: Four in vitro experiments were conducted to evaluate effects of UV+RB on green fluorescent protein EBOV (EBOV-GFP), wild-type EBOV in serum, and whole blood, respectively, and on immunoglobulins and complement in plasma. Initial titers for Experiments 1 to 3 were 4.21 log GFP units/mL, 4.96 log infectious units/mL, and 4.23 log plaque-forming units/mL. Conditions tested in the first three experiments included the following: 1-EBOV-GFP plus UV+RB; 2-EBOV-GFP plus RB only; 3-EBOV-GFP plus UV only; 4-EBOV-GFP without RB or UV; 5-virus-free control plus UV only; and 6-virus-free control without RB or UV. RESULTS: UV+RB reduced EBOV titers to nondetectable levels in both nonhuman primate serum (≥2.8- to 3.2-log reduction) and human whole blood (≥3.0-log reduction) without decreasing protective antibody titers in human plasma. CONCLUSION: Our in vitro results demonstrate that the UV+RB treatment efficiently reduces EBOV titers to below limits of detection in both serum and whole blood. In vivo testing to determine whether UV+RB can improve convalescent blood product safety is indicated.


Asunto(s)
Sangre/virología , Desinfección/métodos , Ebolavirus , Fiebre Hemorrágica Ebola/prevención & control , Riboflavina/farmacología , Rayos Ultravioleta , Inactivación de Virus/efectos de la radiación , Animales , Chlorocebus aethiops , Humanos , Macaca fascicularis , Células Vero
9.
J Antimicrob Chemother ; 67(2): 444-51, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22114132

RESUMEN

BACKGROUND: Japanese encephalitis virus (JEV) is a major cause of viral encephalitis in South-East Asia and there is a pressing need to develop novel therapeutic options against it. METHODS: Gene silencing by RNA interference has therapeutic potential by way of degrading the RNA genome of JEV. Four small hairpin RNAs (shRNAs) targeting different locations in the JEV genome were evaluated for antiviral activity against JEV in different cell lines and the mouse model of disease. RESULTS: shN8010, an shRNA targeting the NS5-coding sequence of JEV, had significant antiviral activity in cultured cells. JEV titres were suppressed by 99% in human embryonic kidney cells at 24 h post-infection (p.i.) when shN8010 was delivered using a plasmid. Further, shN8010 delivered using recombinant adenovirus caused 99% and 95% suppression of JEV titres at 24 h p.i. in porcine stable kidney and Vero cells, respectively. In Neuro-2a cells, JEV titres at 24 h p.i. were suppressed by 90% when shN8010 was delivered using a recombinant adenovirus or retrovirus. Four-week-old FvB/J mice treated intracerebrally with recombinant adenovirus-delivered shN8010 1 week prior to lethal intraperitoneal JEV challenge showed no protection, although the mean survival time was prolonged. In a similar experiment, retrovirus-delivered shN8010 provided 100% protection to mice following the lethal JEV challenge. CONCLUSIONS: NS5-targeting shRNA (shN8010) had very significant antiviral activity in both cultured cells and the mouse model of JEV infection.


Asunto(s)
Productos Biológicos/administración & dosificación , Productos Biológicos/farmacología , Virus de la Encefalitis Japonesa (Especie)/efectos de los fármacos , Encefalitis Japonesa/tratamiento farmacológico , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/farmacología , Animales , Antivirales/administración & dosificación , Antivirales/farmacología , Células Cultivadas , Modelos Animales de Enfermedad , Virus de la Encefalitis Japonesa (Especie)/genética , Silenciador del Gen , Humanos , Ratones , Análisis de Supervivencia , Carga Viral
10.
J Virol ; 85(1): 86-97, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20962101

RESUMEN

Many aspects of the assembly of hepatitis C virus (HCV) remain incompletely understood. To characterize the role of NS2 in the production of infectious virus, we determined NS2 interaction partners among other HCV proteins during productive infection. Pulldown assays showed that NS2 forms complexes with both structural and nonstructural proteins, including E1, E2, p7, NS3, and NS5A. Confocal microscopy also demonstrated that NS2 colocalizes with E1, E2, and NS5A in dot-like structures near lipid droplets. However, NS5A did not coprecipitate with E2 and interacted only weakly with NS3 in pulldown assays. Also, there was no demonstrable interaction between p7 and E2 or NS3 in such assays. Therefore, NS2 is uniquely capable of interacting with both structural and nonstructural proteins. Among mutations in p7, NS2, and NS3 that prevent production of infectious virus, only p7 mutations significantly reduced NS2-mediated protein interactions. These p7 mutations altered the intracellular distribution of NS2 and E2 and appeared to modulate the membrane topology of the C-terminal domain of NS2. These results suggest that NS2 acts to coordinate virus assembly by mediating interactions between envelope proteins and NS3 and NS5A within replication complexes adjacent to lipid droplets, where virus particle assembly is thought to occur. p7 may play an accessory role by regulating NS2 membrane topology, which is important for NS2-mediated protein interactions and therefore NS2 function.


Asunto(s)
Hepacivirus/metabolismo , Proteínas no Estructurales Virales/metabolismo , Ensamble de Virus/fisiología , Línea Celular , Hepacivirus/genética , Hepacivirus/fisiología , Humanos , Mutación , Proteínas no Estructurales Virales/genética , Proteínas Virales/genética , Proteínas Virales/metabolismo , Proteínas Estructurales Virales/genética , Proteínas Estructurales Virales/metabolismo
11.
ACS Infect Dis ; 8(5): 942-957, 2022 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-35357134

RESUMEN

Ebola virus (EBOV) is an aggressive filoviral pathogen that can induce severe hemorrhagic fever in humans with up to 90% fatality rate. To date, there are no clinically effective small-molecule drugs for postexposure therapies to treat filoviral infections. EBOV cellular entry and infection involve uptake via macropinocytosis, navigation through the endocytic pathway, and pH-dependent escape into the cytoplasm. We report the inhibition of EBOV cell entry via selective inhibition of vacuolar (V)-ATPase by a new series of phenol-substituted derivatives of the natural product scaffold diphyllin. In cells challenged with Ebola virus, the diphyllin derivatives inhibit viral entry dependent upon structural variations to low nanomolar potencies. Mechanistically, the diphyllin derivatives had no effect on uptake and colocalization of viral particles with endocytic marker LAMP1 but directly modulated endosomal pH. The most potent effects were reversible exhibiting higher selectivity than bafilomycin or the parent diphyllin. Unlike general lysosomotrophic agents, the diphyllin derivatives showed no major disruptions of endocytic populations or morphology when examined with Rab5 and LAMP1 markers. The dilated vacuole phenotype induced by apilimod treatment or in constitutively active Rab5 mutant Q79L-expressing cells was both blocked and reversed by the diphyllin derivatives. The results are consistent with the action of the diphyllin scaffold as a selective pH-dependent viral entry block in late endosomes. Overall, the compounds show improved selectivity and minimal cytotoxicity relative to classical endosomal acidification blocking agents.


Asunto(s)
Ebolavirus , Fiebre Hemorrágica Ebola , Benzodioxoles/farmacología , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Humanos , Lignanos , Fenol/farmacología , Fenol/uso terapéutico , Internalización del Virus
12.
Antiviral Res ; 206: 105399, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36007601

RESUMEN

Filoviruses enter cells through macropinocytosis and trafficking into the endosomes in which they bind to the receptor Niemann-Pick C1 protein (NPC1) for membrane fusion and entry into the cytoplasm. The endosomal receptor-binding is critical step for filovirus entry. Designing inhibitors to block receptor binding will prevent viral entry. Using available binding structural information from the co-crystal structures of the viral GP with the receptor NPC1 or with monoclonal antibodies, we have conducted structure-based design of peptide inhibitors to target the receptor binding site (RBS). The designed peptides were tested for their inhibition activity against pseudo-typed or replication-competent viruses in a cell-based assay. The results indicate that these peptides exhibited strong activities against both Ebola and Marburg virus infection. It is expected that these peptides can be further developed for therapeutic use to treat filovirus infection and combat the outbreaks.


Asunto(s)
Filoviridae , Receptores Virales , Inhibidores de Proteínas Virales de Fusión , Sitios de Unión , Proteínas Portadoras/metabolismo , Línea Celular , Ebolavirus/fisiología , Endosomas/metabolismo , Filoviridae/química , Filoviridae/efectos de los fármacos , Fiebre Hemorrágica Ebola , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ligandos , Glicoproteínas de Membrana/metabolismo , Proteína Niemann-Pick C1/metabolismo , Receptores Virales/química , Receptores Virales/metabolismo , Inhibidores de Proteínas Virales de Fusión/química , Inhibidores de Proteínas Virales de Fusión/farmacología , Internalización del Virus/efectos de los fármacos
13.
bioRxiv ; 2022 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-33907750

RESUMEN

Identification of host factors contributing to replication of viruses and resulting disease progression remains a promising approach for development of new therapeutics. Here, we evaluated 6710 clinical and preclinical compounds targeting 2183 host proteins by immunocytofluorescence-based screening to identify SARS-CoV-2 infection inhibitors. Computationally integrating relationships between small molecule structure, dose-response antiviral activity, host target and cell interactome networking produced cellular networks important for infection. This analysis revealed 389 small molecules, >12 scaffold classes and 813 host targets with micromolar to low nanomolar activities. From these classes, representatives were extensively evaluated for mechanism of action in stable and primary human cell models, and additionally against Beta and Delta SARS-CoV-2 variants and MERS-CoV. One promising candidate, obatoclax, significantly reduced SARS-CoV-2 viral lung load in mice. Ultimately, this work establishes a rigorous approach for future pharmacological and computational identification of novel host factor dependencies and treatments for viral diseases.

14.
iScience ; 25(9): 104925, 2022 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-35992305

RESUMEN

Pharmacologically active compounds with known biological targets were evaluated for inhibition of SARS-CoV-2 infection in cell and tissue models to help identify potent classes of active small molecules and to better understand host-virus interactions. We evaluated 6,710 clinical and preclinical compounds targeting 2,183 host proteins by immunocytofluorescence-based screening to identify SARS-CoV-2 infection inhibitors. Computationally integrating relationships between small molecule structure, dose-response antiviral activity, host target, and cell interactome produced cellular networks important for infection. This analysis revealed 389 small molecules with micromolar to low nanomolar activities, representing >12 scaffold classes and 813 host targets. Representatives were evaluated for mechanism of action in stable and primary human cell models with SARS-CoV-2 variants and MERS-CoV. One promising candidate, obatoclax, significantly reduced SARS-CoV-2 viral lung load in mice. Ultimately, this work establishes a rigorous approach for future pharmacological and computational identification of host factor dependencies and treatments for viral diseases.

15.
Microorganisms ; 9(1)2021 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-33445537

RESUMEN

Ongoing efforts to develop effective therapies against filoviruses rely, to different extents, on quantifying the amount of viable virus in samples by plaque, TCID50, and focus assays. Unfortunately, these techniques have inherent variance, and laboratory-specific preferences make direct comparison of data difficult. Additionally, human errors such as operator errors and subjective bias can further compound the differences in outcomes. To overcome these biases, we developed a computer-based automated image-processing method for a focus assay based on the open-source CellProfiler software platform, which enables high-throughput screening of many treatment samples at one time. We compared virus titers calculated using this platform to plaque and TCID50 assays using common stocks of virus for 3 major Filovirus species, Zaire ebolavirus, Sudan ebolavirus, and Marburg marburgvirus with each assay performed by multiple operators on multiple days. We show that plaque assays give comparable findings that differ by less than 3-fold. Focus-forming unit (FFU) and TCID50 assays differ by 10-fold or less from the plaque assays due a higher (FFU) and lower (TCID50) sensitivity. However, reproducibility and accuracy of each assay differs significantly with Neutral Red Agarose Overlay plaque assays and TCID50 with the lowest reproducibility due to subjective analysis and operator error. Both crystal violet methylcellulose overlay plaque assay and focus assays perform best for accuracy and the focus assay performs best for speed and throughput.

16.
Antiviral Res ; 189: 105059, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33705865

RESUMEN

Filoviruses, mainly consisting of Ebola viruses (EBOV) and Marburg viruses (MARV), are enveloped negative-strand RNA viruses which can infect humans to cause severe hemorrhagic fevers and outbreaks with high mortality rates. The filovirus infection is mediated by the interaction of viral envelope glycoprotein (GP) and the human endosomal receptor Niemann-Pick C1 (NPC1). Blocking this interaction will prevent the infection. Therefore, we utilized an In silico screening approach to conduct virtual compound screening against the NPC1 receptor-binding site (RBS). Twenty-six top-hit compounds were purchased and evaluated by in vitro cell based inhibition assays against pseudotyped or replication-competent filoviruses. Two classes (A and U) of compounds were identified to have potent inhibitory activity against both Ebola and Marburg viruses. The IC50 values are in the lower level of micromolar concentrations. One compound (compd-A) was found to have a sub-micromolar IC50 value (0.86 µM) against pseudotyped Marburg virus. The cytotoxicity assay (MTT) indicates that compd-A has a moderate cytotoxicity level but the compd-U has much less toxicity and the CC50 value was about 100 µM. Structure-activity relationship (SAR) study has found some analogs of compd-A and -U have reduced the toxicity and enhanced the inhibitory activity. In conclusion, this work has identified several qualified lead-compounds for further drug development against filovirus infection.


Asunto(s)
Antivirales/farmacología , Ebolavirus/efectos de los fármacos , Infecciones por Filoviridae/virología , Marburgvirus/efectos de los fármacos , Proteína Niemann-Pick C1/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus/efectos de los fármacos , Antivirales/química , Sitios de Unión , Supervivencia Celular , Descubrimiento de Drogas , Ebolavirus/fisiología , Infecciones por Filoviridae/tratamiento farmacológico , Células HeLa , Interacciones Microbiota-Huesped/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Marburgvirus/fisiología , Simulación del Acoplamiento Molecular , Proteína Niemann-Pick C1/química , Unión Proteica , Receptores Virales/química , Receptores Virales/metabolismo
17.
J Antimicrob Chemother ; 65(5): 953-61, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20299495

RESUMEN

BACKGROUND: Japanese encephalitis virus (JEV) has a significant impact on public health throughout Asia, and there is a pressing need for development of new therapeutics against it. METHODS: Peptide-conjugated phosphorodiamidate morpholino oligomers (PPMOs) are antisense agents that enter cells readily and interfere with gene expression. Four PPMOs, targeting various locations in the JEV genome, were evaluated for antiviral activity against JEV in cultured cells and the mouse model of JEV infection. RESULTS: A PPMO (P10882) targeting the JEV 3' cyclization sequence (3'CSI) had significant antiviral activity in Vero (epithelial), Neuro2A (neuronal) and J774E (macrophage) cells at concentrations that were not cytotoxic. P10882 added before infection suppressed JEV replication to an undetectable level in Vero cells and produced a 93% and 66% reduction in titre in J774E and Neuro2A cells, respectively, when measured at 24 h post-infection. In uninfected cells, fluorescein-labelled PPMOs entered J774E cells most efficiently, followed by Vero and Neuro2A cells. The antiviral effect of P10882 was also demonstrated in vivo, where 60%-80% of 1-week-old mice treated intracerebrally with a 20 mg/kg dose of P10882 every 12 h for 5 days were protected from a lethal dose of JEV and showed an undetectable level of virus in brain tissue at 2 days post-infection. CONCLUSIONS: P10882, which targets sequence that is highly conserved across members of the JEV serocomplex, was previously shown to be effective in a mouse model of West Nile disease, and represents a candidate antiviral agent against members of the JEV serocomplex.


Asunto(s)
Antivirales/farmacología , Virus de la Encefalitis Japonesa (Especie)/efectos de los fármacos , Oligonucleótidos/farmacología , Péptidos/farmacología , Animales , Antivirales/uso terapéutico , Línea Celular , Chlorocebus aethiops , Encefalitis Japonesa/tratamiento farmacológico , Células Epiteliales/virología , Macrófagos/virología , Ratones , Ratones Endogámicos BALB C , Neuronas/virología , Oligonucleótidos/uso terapéutico , Péptidos/uso terapéutico , Análisis de Supervivencia
18.
ACS Infect Dis ; 6(10): 2783-2799, 2020 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-32870648

RESUMEN

Marburg virus (MARV) causes sporadic outbreaks of severe disease with high case fatality rates in humans. To date, neither therapeutics nor prophylactic approaches have been approved for MARV disease. The MARV matrix protein VP40 (mVP40) plays central roles in virus assembly and budding. mVP40 also inhibits interferon signaling by inhibiting the function of Janus kinase 1. This suppression of host antiviral defenses likely contributes to MARV virulence and therefore is a potential therapeutic target. We developed and optimized a cell-based high-throughput screening (HTS) assay in 384-well format to measure mVP40 interferon (IFN) antagonist function such that inhibitors could be identified. We performed a pilot screen of 1280 bioactive compounds and identified 3 hits, azaguanine-8, tosufloxacin hydrochloride, and linezolid, with Z scores > 3 and no significant cytotoxicity. Of these, azaguanine-8 inhibited MARV growth at noncytotoxic concentrations. These data demonstrate the suitability of the HTS mVP40 assay for drug discovery and suggest potential directions for anti-MARV therapeutic development.


Asunto(s)
Enfermedad del Virus de Marburg , Marburgvirus , Animales , Ensayos Analíticos de Alto Rendimiento , Humanos , Interferones , Ensamble de Virus
19.
Antiviral Res ; 181: 104863, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32682926

RESUMEN

The recent outbreaks of the Ebola virus (EBOV) in Africa have brought global visibility to the shortage of available therapeutic options to treat patients infected with this or closely related viruses. We have recently computationally identified three molecules which have all demonstrated statistically significant efficacy in the mouse model of infection with mouse adapted Ebola virus (ma-EBOV). One of these molecules is the antimalarial pyronaridine tetraphosphate (IC50 range of 0.82-1.30 µM against three strains of EBOV and IC50 range of 1.01-2.72 µM against two strains of Marburg virus (MARV)) which is an approved drug in the European Union and used in combination with artesunate. To date, no small molecule drugs have shown statistically significant efficacy in the guinea pig model of EBOV infection. Pharmacokinetics and range-finding studies in guinea pigs directed us to a single 300 mg/kg or 600 mg/kg oral dose of pyronaridine 1hr after infection. Pyronaridine resulted in statistically significant survival of 40% at 300 mg/kg and protected from a lethal challenge with EBOV. In comparison, oral favipiravir (300 mg/kg dosed once a day) had 43.5% survival. All animals in the vehicle treatment group succumbed to disease by study day 12 (100% mortality). The in vitro metabolism and metabolite identification of pyronaridine and another of our EBOV active molecules, tilorone, suggested significant species differences which may account for the efficacy or lack thereof, respectively in guinea pig. In summary, our studies with pyronaridine demonstrates its utility for repurposing as an antiviral against EBOV and MARV.


Asunto(s)
Antivirales/uso terapéutico , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Naftiridinas/uso terapéutico , Animales , Antivirales/farmacocinética , Modelos Animales de Enfermedad , Reposicionamiento de Medicamentos , Ebolavirus/efectos de los fármacos , Femenino , Cobayas , Humanos , Concentración 50 Inhibidora , Masculino , Marburgvirus/efectos de los fármacos , Ratones , Microsomas , Naftiridinas/farmacocinética
20.
J Med Chem ; 63(13): 7211-7225, 2020 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-32490678

RESUMEN

The recent Ebola epidemics in West Africa underscore the great need for effective and practical therapies for future Ebola virus outbreaks. We have discovered a new series of remarkably potent small molecule inhibitors of Ebola virus entry. These 4-(aminomethyl)benzamide-based inhibitors are also effective against Marburg virus. Synthetic routes to these compounds allowed for the preparation of a wide variety of structures, including a conformationally restrained subset of indolines (compounds 41-50). Compounds 20, 23, 32, 33, and 35 are superior inhibitors of Ebola (Mayinga) and Marburg (Angola) infectious viruses. Representative compounds (20, 32, and 35) have shown good metabolic stability in plasma and liver microsomes (rat and human), and 32 did not inhibit CYP3A4 nor CYP2C9. These 4-(aminomethyl)benzamides are suitable for further optimization as inhibitors of filovirus entry, with the potential to be developed as therapeutic agents for the treatment and control of Ebola virus infections.


Asunto(s)
Antivirales/farmacología , Benzamidas/farmacología , Fiebre Hemorrágica Ebola/virología , Enfermedad del Virus de Marburg/virología , Internalización del Virus/efectos de los fármacos , Células A549 , Animales , Antivirales/química , Benzamidas/química , Chlorocebus aethiops , Inhibidores del Citocromo P-450 CYP3A/química , Inhibidores del Citocromo P-450 CYP3A/farmacología , Evaluación Preclínica de Medicamentos , Humanos , Microsomas Hepáticos/efectos de los fármacos , Simulación del Acoplamiento Molecular , Relación Estructura-Actividad , Toremifeno/química , Toremifeno/metabolismo , Toremifeno/farmacología , Células Vero , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA