Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Exp Mol Pathol ; 134: 104876, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37890651

RESUMEN

Molluscum contagiosum virus (MCV) is a poxvirus that causes benign, persistent skin lesions. MCV encodes a variety of immune evasion molecules to dampen host immune responses. Two of these proteins are the MC159 and MC160 proteins. Both MC159 and MC160 contain two tandem death effector domains and share homology to the cellular FLIPs, FADD, and procaspase-8. MC159 and MC160 dampen several innate immune responses such as NF-κB activation and mitochondrial antiviral signaling (MAVS)-mediated induction of type 1 interferon (IFN). The type 1 IFN response is also activated by the cytosolic DNA sensors cyclic GMP-AMP synthase (cGAS) and stimulator of interferon genes (STING). Both cGAS and STING play a vital role in sensing a poxvirus infection. In this study, we demonstrate that there are nuanced differences between both MC160 and MC159 in terms of how the viral proteins modulate the cGAS/STING and MAVS pathways. Specifically, MC160 expression, but not MC159 expression, dampens cGAS/STING-mediated induction of IFN in HEK 293 T cells. Further, MC160 expression prevented the K63-ubiquitination of both STING and TBK1, a kinase downstream of cGAS/STING. Ectopic expression of the MC160 protein, but not the MC159 protein, resulted in a measurable decrease in the TBK1 protein levels as detected via immunoblotting. Finally, using a panel of MC160 truncation mutants, we report that the MC160 protein requires both DEDs to inhibit cGAS/STING-induced activation of IFN-ß. Our model indicates MC160 likely alters the TBK1 signaling complex to decrease IFN-ß activation at the molecular intersection of the cGAS/STING and MAVS signaling pathways.


Asunto(s)
Virus del Molusco Contagioso , Humanos , Virus del Molusco Contagioso/genética , Virus del Molusco Contagioso/metabolismo , Células HEK293 , Proteínas Virales/genética , Proteínas Virales/metabolismo , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Inmunidad Innata , Interferón beta/genética , Interferón beta/metabolismo , Interferones/metabolismo
2.
JAMA Netw Open ; 6(9): e2332106, 2023 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-37669055

RESUMEN

This secondary analysis of a randomized clinical trial investigates the effects of a vegan diet on total food costs per day.


Asunto(s)
Dieta Vegana , Sobrepeso , Adulto , Humanos , Costos y Análisis de Costo , Alimentos
3.
World Neurosurg ; 141: 44-51, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32450315

RESUMEN

BACKGROUND: Spinal subdural hematomas (SDHs) have been reported secondary to direct trauma or iatrogenic causes associated with coagulopathies. Spinal SDHs found after the development of acute intracranial SDHs, without any evidence of trauma to the spine, are extremely rare. In addition to this rare presentation, there is a lack of consensus regarding whether surgical decompression is the ideal treatment strategy. Depending on the extent of SDH within the spinal canal, surgical decompression may be difficult where diffuse hematoma within the intradural space requires multilevel decompression for treatment. CASE DESCRIPTION: A 46-year-old man initially presented with an acute cranial SDH following isolated head trauma. After a period of full recovery, he developed delayed lower extremity paraparesis secondary to the formation of a thoracolumbar SDH. This hematoma coincided with resolution of the cranial SDH and likely was due to redistribution of blood from the cranial subdural space into the spinal canal. Given the diffuse multilevel nature of the spread of hematoma and lack of a focal area of compression, he was managed conservatively. He demonstrated small signs of neurologic improvement over several days and regained considerable strength over the following several weeks. CONCLUSIONS: This report demonstrates a very rare occurrence of a traumatic intracranial SDH migrating into the thoracic and lumbar spine. This case also highlights that despite acute neurologic deficits, conservative management may be a feasible strategy that can result in recovery of neurologic function.


Asunto(s)
Traumatismos Craneocerebrales/complicaciones , Hematoma Subdural Espinal/etiología , Hematoma Subdural Espinal/cirugía , Traumatismos Craneocerebrales/diagnóstico por imagen , Descompresión Quirúrgica , Hematoma Subdural Espinal/diagnóstico por imagen , Humanos , Masculino , Persona de Mediana Edad , Paraparesia/etiología , Resultado del Tratamiento
4.
Viruses ; 9(8)2017 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-28786952

RESUMEN

Cells have multiple means to induce apoptosis in response to viral infection. Poxviruses must prevent activation of cellular apoptosis to ensure successful replication. These viruses devote a substantial portion of their genome to immune evasion. Many of these immune evasion products expressed during infection antagonize cellular apoptotic pathways. Poxvirus products target multiple points in both the extrinsic and intrinsic apoptotic pathways, thereby mitigating apoptosis during infection. Interestingly, recent evidence indicates that poxviruses also hijack cellular means of eliminating apoptotic bodies as a means to spread cell to cell through a process called apoptotic mimicry. Poxviruses are the causative agent of many human and veterinary diseases. Further, there is substantial interest in developing these viruses as vectors for a variety of uses including vaccine delivery and as oncolytic viruses to treat certain human cancers. Therefore, an understanding of the molecular mechanisms through which poxviruses regulate the cellular apoptotic pathways remains a top research priority. In this review, we consider anti-apoptotic strategies of poxviruses focusing on three relevant poxvirus genera: Orthopoxvirus, Molluscipoxvirus, and Leporipoxvirus. All three genera express multiple products to inhibit both extrinsic and intrinsic apoptotic pathways with many of these products required for virulence.


Asunto(s)
Apoptosis , Interacciones Huésped-Patógeno , Evasión Inmune , Infecciones por Poxviridae/virología , Poxviridae/fisiología , Animales , Caspasas/metabolismo , Humanos , Leporipoxvirus/patogenicidad , Leporipoxvirus/fisiología , Molluscipoxvirus/patogenicidad , Molluscipoxvirus/fisiología , Orthopoxvirus/patogenicidad , Orthopoxvirus/fisiología , Poxviridae/genética , Poxviridae/patogenicidad , Infecciones por Poxviridae/inmunología , Infecciones por Poxviridae/fisiopatología , Transducción de Señal , Proteínas Virales/metabolismo , Virulencia , Replicación Viral
5.
Virus Genes ; 53(4): 522-531, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28425034

RESUMEN

The molluscum contagiosum virus (MCV) uses a variety of immune evasion strategies to antagonize host immune responses. Two MCV proteins, MC159 and MC160, contain tandem death effector domains (DEDs). They are reported to inhibit innate immune signaling events such as NF-κB and IRF3 activation, and apoptosis. The RxDL motif of MC159 is required for inhibition of both apoptosis and NF-κB activation. However, the role of the conserved RxDL motif in the MC160 DEDs remained unknown. To answer this question, we performed alanine mutations to neutralize the arginine and aspartate residues present in the MC160 RxDL in both DED1 and DED2. These mutations were further modeled against the structure of the MC159 protein. Surprisingly, the RxDL motif was not required for MC160's ability to inhibit MAVS-induced IFNß activation. Further, unlike previous results with the MC159 protein, mutations within the RxDL motif of MC160 had no effect on the ability of MC160 to dampen TNF-α-induced NF-κB activation. Molecular modeling predictions revealed no overall changes to the structure in the MC160 protein when the amino acids of both RxDL motifs were mutated to alanine (DED1 = R67A D69A; DED2 = R160A D162A). Taken together, our results demonstrate that the RxDL motifs present in the MC160 DEDs are not required for known functions of the viral protein.


Asunto(s)
Evasión Inmune , Molusco Contagioso/virología , Virus del Molusco Contagioso/inmunología , Proteínas Virales/química , Proteínas Virales/inmunología , Secuencias de Aminoácidos , Apoptosis , Humanos , Interferón beta/genética , Interferón beta/inmunología , Molusco Contagioso/genética , Molusco Contagioso/inmunología , Molusco Contagioso/fisiopatología , Virus del Molusco Contagioso/química , Virus del Molusco Contagioso/genética , Dominios Proteicos , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Proteínas Virales/genética
6.
Virology ; 505: 91-101, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28235685

RESUMEN

Apoptosis is a powerful host cell defense to prevent viruses from completing replication. Poxviruses have evolved complex means to dampen cellular apoptotic responses. The poxvirus, Molluscum Contagiosum Virus (MCV), encodes numerous host interacting molecules predicted to antagonize immune responses. However, the function of the majority of these MCV products has not been characterized. Here, we show that the MCV MC163 protein localized to the mitochondria via an N-terminal mitochondrial localization sequence and transmembrane domain. Transient expression of the MC163 protein prevented mitochondrial membrane permeabilization (MMP), an event central to cellular apoptotic responses, induced by either Tumor Necrosis Factor alpha (TNF-α) or carbonyl cyanide 3-chlorophenylhydrazone (CCCP). MC163 expression prevented the release of a mitochondrial intermembrane space reporter protein when cells were challenged with TNF-α. Inhibition of MMP was also observed in cell lines stably expressing MC163. MC163 expression may contribute to the persistence of MCV lesions by dampening cellular apoptotic responses.


Asunto(s)
Permeabilidad de la Membrana Celular/fisiología , Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo , Virus del Molusco Contagioso/metabolismo , Proteínas Virales/metabolismo , Apoptosis , Caspasa 3/metabolismo , Línea Celular Tumoral , Células HeLa , Humanos , Hidrazonas/farmacología , Molusco Contagioso/virología , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Estaurosporina/farmacología , Factor de Necrosis Tumoral alfa/farmacología
7.
Hepatology ; 65(5): 1462-1477, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28090674

RESUMEN

Hepatitis C virus (HCV) infection is a common risk factor for the development of liver cancer. The molecular mechanisms underlying this effect are only partially understood. Here, we show that the HCV protein, nonstructural protein (NS) 5B, directly binds to the tumor suppressor, NORE1A (RASSF5), and promotes its proteosomal degradation. In addition, we show that NORE1A colocalizes to sites of HCV viral replication and suppresses the replication process. Thus, NORE1A has antiviral activity, which is specifically antagonized by NS5B. Moreover, the suppression of NORE1A protein levels correlated almost perfectly with elevation of Ras activity in primary human samples. Therefore, NORE1A inactivation by NS5B may be essential for maximal HCV replication and may make a major contribution to HCV-induced liver cancer by shifting Ras signaling away from prosenescent/proapoptotic signaling pathways. CONCLUSION: HCV uses NS5B to specifically suppress NORE1A, facilitating viral replication and elevated Ras signaling. (Hepatology 2017;65:1462-1477).


Asunto(s)
Hepacivirus/fisiología , Proteínas de Unión al GTP Monoméricas/metabolismo , Proteínas no Estructurales Virales/metabolismo , Replicación Viral , Proteínas Adaptadoras Transductoras de Señales , Proteínas Reguladoras de la Apoptosis , Carcinoma Hepatocelular/virología , Regulación hacia Abajo , Células HEK293 , Humanos , Hígado/metabolismo , Hígado/virología , Neoplasias Hepáticas/virología , Complejo de la Endopetidasa Proteasomal/metabolismo
8.
Eur J Med Chem ; 112: 33-38, 2016 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-26874742

RESUMEN

The synthesis of a series of 5-carba-pterocarpens derivatives involving the cyclization of α-aryl-α-tetralones is described. Several compounds demonstrated potent activity and selectivity in vitro against HCV replicon reporter cells. The best profile in Huh7/Rep-Feo1b replicon reporter cells was observed with 2h (EC50 = 5.5 µM/SI = 20), while 2e was the most active in Huh7.5-FGR-JC1-Rluc2A replicon reporter cells (EC50 = 1.5 µM/SI = 70). Hydroxy groups at A- and D-rings are essential for anti-HCV activity, and substitutions in the A-ring at positions 3 and 4 resulted in enhanced activity of the compounds.


Asunto(s)
Antivirales/química , Antivirales/farmacología , Guanidinas/química , Guanidinas/farmacología , Hepacivirus/efectos de los fármacos , Anisoles/síntesis química , Anisoles/química , Anisoles/farmacología , Antivirales/síntesis química , Catálisis , Línea Celular , Guanidinas/síntesis química , Hepacivirus/genética , Hepatitis C/tratamiento farmacológico , Hepatitis C/virología , Humanos , Paladio/química , Replicón/efectos de los fármacos , Tetralonas/síntesis química , Tetralonas/química , Tetralonas/farmacología
9.
Bioorg Med Chem Lett ; 26(3): 1048-1051, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26750257

RESUMEN

Investigation of a series of 2,N-bisarylated 2-ethoxyacetamides resulted in the identification of four inhibitors 5, 20, 24, 29 with single-digit micromolar in vitro efficacy against two drug-resistant Plasmodium falciparum strains. These compounds are analogs of structurally-related 1,3-bisaryl-2-propen-1-ones (chalcones), the latter showing efficacy in vitro but not in a malaria-infected mouse. The 2,N-bisarylated 2-ethoxyacetamides (e.g., 2, 5, 20) were shown to possess significantly greater stability in the presence of metabolizing enzymes than the corresponding 1,3-bisaryl-2-propen-1-ones (e.g., 1, 3, 18).


Asunto(s)
Acetamidas/química , Antimaláricos/química , Acetamidas/metabolismo , Acetamidas/farmacología , Animales , Antimaláricos/metabolismo , Antimaláricos/farmacología , Chalcona/química , Cloroquina/farmacología , Resistencia a Medicamentos/efectos de los fármacos , Semivida , Ratones , Microsomas Hepáticos/metabolismo , Plasmodium falciparum/efectos de los fármacos , Relación Estructura-Actividad
10.
J Anal Toxicol ; 38(8): 524-7, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25217541

RESUMEN

Legitimate use of legal intranasal decongestants containing l-methamphetamine may complicate interpretation of urine drug tests positive for amphetamines. Our study hypotheses were that commonly used immunoassays would produce no false-positive results and a recently developed enantiomer-specific gas chromatography-mass spectrometry (GC-MS) procedure would find no d-amphetamine or d-methamphetamine in urine following controlled Vicks VapoInhaler administration at manufacturer's recommended doses. To evaluate these hypotheses, 22 healthy adults were each administered one dose (two inhalations in each nostril) of a Vicks VapoInhaler every 2 h for 10 h on Day 1 (six doses), followed by a single dose on Day 2. Every urine specimen was collected as an individual void for 32 h after the first dose and assayed for d- and l-amphetamines specific isomers with a GC-MS method with >99% purity of R-(-)-α-methoxy-α-(trifluoromethyl)phenylacetyl derivatives and 10 µg/L lower limits of quantification. No d-methamphetamine or d-amphetamine was detected in any urine specimen by GC-MS. The median l-methamphetamine maximum concentration was 62.8 µg/L (range: 11.0-1,440). Only two subjects had detectable l-amphetamine, with maximum concentrations coinciding with l-methamphetamine peak levels, and always ≤ 4% of the parent's maximum. Three commercial immunoassays for amphetamines EMIT(®) II Plus, KIMS(®) II and DRI(®) had sensitivities, specificities and efficiencies of 100, 97.8, 97.8; 100, 99.6, 99.6 and 100, 100, 100%, respectively. The immunoassays had high efficiencies, but our first hypothesis was not affirmed. The EMIT(®) II Plus assay produced 2.2% false-positive results, requiring an enantiomer-specific confirmation.


Asunto(s)
Anfetamina/orina , Metanfetamina/orina , Administración por Inhalación , Adolescente , Adulto , Anciano , Creatinina/orina , Relación Dosis-Respuesta a Droga , Reacciones Falso Positivas , Femenino , Cromatografía de Gases y Espectrometría de Masas , Voluntarios Sanos , Humanos , Inmunoensayo/métodos , Masculino , Persona de Mediana Edad , Sensibilidad y Especificidad , Manejo de Especímenes , Estereoisomerismo , Detección de Abuso de Sustancias/métodos , Adulto Joven
11.
J Virol ; 88(20): 11825-33, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25100850

RESUMEN

To combat emerging coronaviruses, developing safe and efficient platforms to evaluate viral protease activities and the efficacy of protease inhibitors is a high priority. Here, we exploit a biosafety level 2 (BSL-2) chimeric Sindbis virus system to evaluate protease activities and the efficacy of inhibitors directed against the papain-like protease (PLpro) of severe acute respiratory syndrome coronavirus (SARS-CoV), a biosafety level 3 (BSL-3) pathogen. We engineered Sindbis virus to coexpress PLpro and a substrate, murine interferon-stimulated gene 15 (ISG15), and found that PLpro mediates removal of ISG15 (deISGylation) from cellular proteins. Mutation of the catalytic cysteine residue of PLpro or addition of a PLpro inhibitor blocked deISGylation in virus-infected cells. Thus, deISGylation is a marker of PLpro activity. Infection of alpha/beta interferon receptor knockout (IFNAR(-/-)) mice with these chimeric viruses revealed that PLpro deISGylation activity removed ISG15-mediated protection during viral infection. Importantly, administration of a PLpro inhibitor protected these mice from lethal infection, demonstrating the efficacy of a coronavirus protease inhibitor in a mouse model. However, this PLpro inhibitor was not sufficient to protect the mice from lethal infection with SARS-CoV MA15, suggesting that further optimization of the delivery and stability of PLpro inhibitors is needed. We extended the chimeric-virus platform to evaluate the papain-like protease/deISGylating activity of Middle East respiratory syndrome coronavirus (MERS-CoV) to provide a small-animal model to evaluate PLpro inhibitors of this recently emerged pathogen. This platform has the potential to be universally adaptable to other viral and cellular enzymes that have deISGylating activities. Importance: Evaluating viral protease inhibitors in a small-animal model is a critical step in the path toward antiviral drug development. We modified a biosafety level 2 chimeric virus system to facilitate evaluation of inhibitors directed against highly pathogenic coronaviruses. We used this system to demonstrate the in vivo efficacy of an inhibitor of the papain-like protease of severe acute respiratory syndrome coronavirus. Furthermore, we demonstrate that the chimeric-virus system can be adapted to study the proteases of emerging human pathogens, such as Middle East respiratory syndrome coronavirus. This system provides an important tool to rapidly assess the efficacy of protease inhibitors targeting existing and emerging human pathogens, as well as other enzymes capable of removing ISG15 from cellular proteins.


Asunto(s)
Coronavirus/fisiología , Modelos Animales de Enfermedad , Papaína/metabolismo , Péptido Hidrolasas/metabolismo , Animales , Chlorocebus aethiops , Coronavirus/enzimología , Cricetinae , Ratones , Células Vero
12.
Forensic Sci Int ; 241: 87-90, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24887324

RESUMEN

Opiates are an important component for drug testing due to their high abuse potential. Proper urine opiate interpretation includes ruling out poppy seed ingestion; however, detailed elimination studies after controlled poppy seed administration with known morphine and codeine doses are not available. Therefore, we investigated urine opiate pharmacokinetics after controlled oral administration of uncooked poppy seeds with known morphine and codeine content. Participants were administered two 45 g oral poppy seed doses 8 h apart, each containing 15.7 mg morphine and 3mg codeine. Urine was collected ad libitum up to 32 h after the first dose. Specimens were analyzed with the Roche Opiates II immunoassay at 2000 and 300 µg/L cutoffs, and the ThermoFisher CEDIA(®) heroin metabolite (6-acetylmorphine, 6-AM) and Lin-Zhi 6-AM immunoassays with 10 µg/L cutoffs to determine if poppy seed ingestion could produce positive results in these heroin marker assays. In addition, all specimens were quantified for morphine and codeine by GC/MS. Participants (N=22) provided 391 urine specimens over 32 h following dosing; 26.6% and 83.4% were positive for morphine at 2000 and 300 µg/L GC/MS cutoffs, respectively. For the 19 subjects who completed the study, morphine concentrations ranged from <300 to 7522 µg/L with a median peak concentration of 5239 µg/L. The median first morphine-positive urine sample at 2000 µg/L cutoff concentration occurred at 6.6 h (1.2-12.1), with the last positive from 2.6 to 18 h after the second dose. No specimens were positive for codeine at a cutoff concentration of 2000 µg/L, but 20.2% exceeded 300 µg/L, with peak concentrations of 658 µg/L (284-1540). The Roche Opiates II immunoassay had efficiencies greater than 96% for the 2000 and 300 µg/L cutoffs. The CEDIA 6-AM immunoassay had a specificity of 91%, while the Lin-Zhi assay had no false positive results. These data provide valuable information for interpreting urine opiate results.


Asunto(s)
Analgésicos Opioides/orina , Codeína/orina , Morfina/orina , Papaver , Semillas , Femenino , Cromatografía de Gases y Espectrometría de Masas , Humanos , Inmunoensayo , Masculino
13.
Eur J Med Chem ; 69: 931-41, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24161679

RESUMEN

Hepatitis C virus (HCV) NS5B polymerase is an important and attractive target for the development of anti-HCV drugs. Here we report on the design, synthesis and evaluation of twenty-four novel allosteric inhibitors bearing the 4-thiazolidinone scaffold as inhibitors of HCV NS5B polymerase. Eleven compounds tested were found to inhibit HCV NS5B with IC50 values ranging between 19.8 and 64.9 µM. Compound 24 was the most active of this series with an IC50 of 5.6 µM. A number of these derivatives further exhibited strong inhibition against HCV 1b and 2a genotypes in cell based antiviral assays. Molecular docking analysis predicted that the thiazolidinone derivatives bind to the NS5B thumb pocket-II (TP-II). Our results suggest that further optimization of the thiazolidinone scaffold may be possible to yield new derivatives with improved enzyme- and cell-based activity.


Asunto(s)
Antivirales/farmacología , Hepacivirus/efectos de los fármacos , Hepacivirus/enzimología , Tiazolidinas/farmacología , Proteínas no Estructurales Virales/antagonistas & inhibidores , Antivirales/síntesis química , Antivirales/química , Antivirales/clasificación , Células Cultivadas , Relación Dosis-Respuesta a Droga , Genotipo , Hepacivirus/genética , Humanos , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Estructura Molecular , Relación Estructura-Actividad , Tiazolidinas/síntesis química , Tiazolidinas/química , Proteínas no Estructurales Virales/metabolismo
14.
Chem Biol Drug Des ; 81(5): 607-14, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23311976

RESUMEN

Coumarins and coumestans represent an important family of compounds with diverse pharmacological properties. We recently identified coumestans as novel inhibitors of hepatitis C virus NS5B polymerase and predicted their binding in thumb pocket-1 (TP-1) of NS5B. As the coumarins are structurally related to coumestans by virtue of their common A- and B-rings, we postulated them to also exhibit similar binding interaction with NS5B and inhibit its polymerase function. We therefore investigated 24 coumarin and neoflavone derivatives as candidate NS5B inhibitors and identified 14 compounds inhibiting NS5B polymerase activity with IC50 values between 17 and 63 µm. Of these, the newly synthesized 6,8-diallyl-5,7-dihydroxycoumarin (8a) was produced in three steps in high chemical yield from floroglucinol and found to be the most potent of this series, exhibiting activity similar to the reference coumestan LQB-34. The binding site of 8a was mapped to TP-1 of NS5B by counter screening against P495L NS5B mutant, employed as a screen for TP-1 site binders. NS5B-TP-1-8a interaction map provided insight into 8a binding and offered clues for future SAR optimization.


Asunto(s)
Antivirales , Cumarinas , Inhibidores Enzimáticos , Flavonas , Hepacivirus/enzimología , ARN Polimerasa Dependiente del ARN/antagonistas & inhibidores , Proteínas no Estructurales Virales/antagonistas & inhibidores , Anticoagulantes/síntesis química , Anticoagulantes/química , Antivirales/síntesis química , Antivirales/química , Sitios de Unión , Cumarinas/síntesis química , Cumarinas/química , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Flavonas/síntesis química , Flavonas/química , Proteínas no Estructurales Virales/química
15.
Eur J Med Chem ; 58: 258-64, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23127989

RESUMEN

Hepatitis C virus (HCV) NS5B polymerase is a key target for the development of anti-HCV drugs. Here we report on the identification of novel allosteric inhibitors of HCV NS5B through a combination of structure-based virtual screening and in vitro NS5B inhibition assays. One hundred and sixty thousand compounds from the Otava database were virtually screened against the thiazolone inhibitor binding site on NS5B (thumb pocket-2, TP-2), resulting in a sequential down-sizing of the library by 2.7 orders of magnitude to yield 59 NS5B non-nucleoside inhibitor (NNI) candidates. In vitro evaluation of the NS5B inhibitory activity of the 59 selected compounds resulted in a 14% hit rate, yielding 8 novel structural scaffolds. Of these, compound 1 bearing a 4-hydrazinoquinazoline scaffold was the most active (IC(50) = 16.0 µM). The binding site of all 8 NNIs was mapped to TP-2 of NS5B as inferred by a decrease in their inhibition potency against the M423T NS5B mutant, employed as a screen for TP-2 site binders. At 100 µM concentration, none of the eight compounds exhibited any cytotoxicity, and all except compound 8 exhibited between 40 and 60% inhibition of intracellular NS5B polymerase activity in BHK-NS5B-FRLuc reporter cells. These inhibitor scaffolds will form the basis for future optimization and development of more potent NS5B inhibitors.


Asunto(s)
Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Hidrazinas/farmacología , Quinazolinonas/farmacología , Proteínas no Estructurales Virales/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/química , Ensayos Analíticos de Alto Rendimiento , Hidrazinas/química , Modelos Moleculares , Estructura Molecular , Quinazolinonas/química , Relación Estructura-Actividad , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo
16.
PLoS One ; 7(2): e30802, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22312431

RESUMEN

Viruses have evolved elaborate mechanisms to evade or inactivate the complex system of sensors and signaling molecules that make up the host innate immune response. Here we show that human coronavirus (HCoV) NL63 and severe acute respiratory syndrome (SARS) CoV papain-like proteases (PLP) antagonize innate immune signaling mediated by STING (stimulator of interferon genes, also known as MITA/ERIS/MYPS). STING resides in the endoplasmic reticulum and upon activation, forms dimers which assemble with MAVS, TBK-1 and IKKε, leading to IRF-3 activation and subsequent induction of interferon (IFN). We found that expression of the membrane anchored PLP domain from human HCoV-NL63 (PLP2-TM) or SARS-CoV (PLpro-TM) inhibits STING-mediated activation of IRF-3 nuclear translocation and induction of IRF-3 dependent promoters. Both catalytically active and inactive forms of CoV PLPs co-immunoprecipitated with STING, and viral replicase proteins co-localize with STING in HCoV-NL63-infected cells. Ectopic expression of catalytically active PLP2-TM blocks STING dimer formation and negatively regulates assembly of STING-MAVS-TBK1/IKKε complexes required for activation of IRF-3. STING dimerization was also substantially reduced in cells infected with SARS-CoV. Furthermore, the level of ubiquitinated forms of STING, RIG-I, TBK1 and IRF-3 are reduced in cells expressing wild type or catalytic mutants of PLP2-TM, likely contributing to disruption of signaling required for IFN induction. These results describe a new mechanism used by CoVs in which CoV PLPs negatively regulate antiviral defenses by disrupting the STING-mediated IFN induction.


Asunto(s)
Coronavirus Humano NL63/enzimología , Cisteína Endopeptidasas/metabolismo , Inmunidad Innata , Proteínas de la Membrana/metabolismo , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/enzimología , Transducción de Señal/inmunología , Proteínas Virales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Biocatálisis , Membrana Celular/enzimología , Chlorocebus aethiops , Proteasas 3C de Coronavirus , Coronavirus Humano NL63/fisiología , Cisteína Endopeptidasas/química , Células HEK293 , Humanos , Quinasa I-kappa B/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Interferones/metabolismo , Proteínas de la Membrana/química , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/fisiología , Ubiquitinación/inmunología , Células Vero , Proteínas Virales/química
17.
Eur J Med Chem ; 49: 191-9, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22280819

RESUMEN

Structure-based studies led to the identification of a constrained derivative of S-trityl-l-cysteine (STLC) scaffold as a candidate inhibitor of hepatitis C virus (HCV) NS5B polymerase. A panel of STLC derivatives were synthesized and investigated for their activity against HCV NS5B. Three STLC derivatives, 9, F-3070, and F-3065, were identified as modest HCV NS5B inhibitors with IC(50) values between 22.3 and 39.7 µM. F-3070 and F-3065 displayed potent inhibition of intracellular NS5B activity in the BHK-NS5B-FRLuc reporter and also inhibited HCV RNA replication in the Huh7/Rep-Feo1b reporter system. Binding mode investigations suggested that the STLC scaffold can be used to develop new NS5B inhibitors by further chemical modification at one of the trityl phenyl group.


Asunto(s)
Antivirales/química , Antivirales/farmacología , Cisteína/análogos & derivados , Hepacivirus/enzimología , ARN Polimerasa Dependiente del ARN/antagonistas & inhibidores , Línea Celular , Cisteína/química , Cisteína/farmacología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Hepacivirus/efectos de los fármacos , Hepatitis C/tratamiento farmacológico , Hepatitis C/enzimología , Humanos , Modelos Moleculares , ARN Viral/metabolismo , ARN Polimerasa Dependiente del ARN/metabolismo
18.
Pharmacology ; 87(1-2): 96-104, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21282967

RESUMEN

4'-n-Butoxy-2,4-dimethoxy-chalcone (MBC) has been described as protecting mice from an otherwise lethal infection with Plasmodium yoelii when dosed orally at 50 mg/kg/dose, daily for 5 days. In contrast, we found that oral dosing of MBC at 640 mg/kg/dose, daily for 5 days, failed to extend the survivability of P. berghei-infected mice. The timing of compound administration and metabolic activation likely contribute to the outcome of efficacy testing in vivo. Microsomal digest of MBC yielded 4'-n-butoxy-4-hydroxy-2-methoxy-chalcone and 4'-(1-hydroxy-n-butoxy)-2,4-dimethoxy-chalcone. We propose that the latter will hydrolyze in vivo to 4'-hydroxy-2,4-dimethoxy-chalcone, which has greater efficacy than MBC in our P. berghei-infected mouse model and was detected in plasma following oral dosing of mice with MBC. Pharmacokinetic parameters suggest that poor absorption, distribution, metabolism and excretion properties contribute to the limited in vivo efficacy observed for MBC and its analogs.


Asunto(s)
Antimaláricos/farmacocinética , Chalconas/farmacocinética , Malaria/tratamiento farmacológico , Microsomas Hepáticos/metabolismo , Plasmodium berghei/efectos de los fármacos , Animales , Antimaláricos/sangre , Antimaláricos/farmacología , Antimaláricos/uso terapéutico , Biotransformación , Chalconas/sangre , Chalconas/farmacología , Chalconas/uso terapéutico , Cromatografía Líquida de Alta Presión , Relación Dosis-Respuesta a Droga , Resistencia a Medicamentos , Semivida , Humanos , Concentración 50 Inhibidora , Malaria/sangre , Malaria/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Estructura Molecular , Espectrometría de Masa por Ionización de Electrospray , Análisis de Supervivencia , Espectrometría de Masas en Tándem
19.
J Med Chem ; 53(13): 4968-79, 2010 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-20527968

RESUMEN

The design, synthesis, X-ray crystal structure, molecular modeling, and biological evaluation of a series of new generation SARS-CoV PLpro inhibitors are described. A new lead compound 3 (6577871) was identified via high-throughput screening of a diverse chemical library. Subsequently, we carried out lead optimization and structure-activity studies to provide a series of improved inhibitors that show potent PLpro inhibition and antiviral activity against SARS-CoV infected Vero E6 cells. Interestingly, the (S)-Me inhibitor 15 h (enzyme IC(50) = 0.56 microM; antiviral EC(50) = 9.1 microM) and the corresponding (R)-Me 15 g (IC(50) = 0.32 microM; antiviral EC(50) = 9.1 microM) are the most potent compounds in this series, with nearly equivalent enzymatic inhibition and antiviral activity. A protein-ligand X-ray structure of 15 g-bound SARS-CoV PLpro and a corresponding model of 15 h docked to PLpro provide intriguing molecular insight into the ligand-binding site interactions.


Asunto(s)
Antivirales/química , Inhibidores Enzimáticos/química , Piperidinas/química , Síndrome Respiratorio Agudo Grave/tratamiento farmacológico , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/enzimología , Proteínas Virales/antagonistas & inhibidores , Animales , Antivirales/síntesis química , Antivirales/metabolismo , Antivirales/farmacología , Chlorocebus aethiops , Proteasas 3C de Coronavirus , Cristalografía por Rayos X , Cisteína Endopeptidasas/química , Cisteína Endopeptidasas/metabolismo , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Concentración 50 Inhibidora , Espectroscopía de Resonancia Magnética , Espectrometría de Masas , Modelos Moleculares , Piperidinas/síntesis química , Piperidinas/metabolismo , Piperidinas/farmacología , Síndrome Respiratorio Agudo Grave/virología , Relación Estructura-Actividad , Células Vero , Proteínas Virales/química , Proteínas Virales/metabolismo , Replicación Viral/efectos de los fármacos
20.
J Med Chem ; 52(16): 5228-40, 2009 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-19645480

RESUMEN

We describe here the design, synthesis, molecular modeling, and biological evaluation of a series of small molecule, nonpeptide inhibitors of SARS-CoV PLpro. Our initial lead compound was identified via high-throughput screening of a diverse chemical library. We subsequently carried out structure-activity relationship studies and optimized the lead structure to potent inhibitors that have shown antiviral activity against SARS-CoV infected Vero E6 cells. Upon the basis of the X-ray crystal structure of inhibitor 24-bound to SARS-CoV PLpro, a drug design template was created. Our structure-based modification led to the design of a more potent inhibitor, 2 (enzyme IC(50) = 0.46 microM; antiviral EC(50) = 6 microM). Interestingly, its methylamine derivative, 49, displayed good enzyme inhibitory potency (IC(50) = 1.3 microM) and the most potent SARS antiviral activity (EC(50) = 5.2 microM) in the series. We have carried out computational docking studies and generated a predictive 3D-QSAR model for SARS-CoV PLpro inhibitors.


Asunto(s)
Antivirales/síntesis química , Benzamidas/síntesis química , Naftalenos/síntesis química , Inhibidores de Proteasas/síntesis química , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/efectos de los fármacos , Proteínas Virales/antagonistas & inhibidores , Animales , Antivirales/química , Antivirales/farmacología , Benzamidas/química , Benzamidas/farmacología , Chlorocebus aethiops , Proteasas 3C de Coronavirus , Cristalografía por Rayos X , Cisteína Endopeptidasas , Diseño de Fármacos , Modelos Moleculares , Naftalenos/química , Naftalenos/farmacología , Inhibidores de Proteasas/química , Inhibidores de Proteasas/farmacología , Relación Estructura-Actividad Cuantitativa , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/enzimología , Células Vero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA