Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Nature ; 537(7618): 117-121, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27556947

RESUMEN

Ca2+ antagonist drugs are widely used in therapy of cardiovascular disorders. Three chemical classes of drugs bind to three separate, but allosterically interacting, receptor sites on CaV1.2 channels, the most prominent voltage-gated Ca2+ (CaV) channel type in myocytes in cardiac and vascular smooth muscle. The 1,4-dihydropyridines are used primarily for treatment of hypertension and angina pectoris and are thought to act as allosteric modulators of voltage-dependent Ca2+ channel activation, whereas phenylalkylamines and benzothiazepines are used primarily for treatment of cardiac arrhythmias and are thought to physically block the pore. The structural basis for the different binding, action, and therapeutic uses of these drugs remains unknown. Here we present crystallographic and functional analyses of drug binding to the bacterial homotetrameric model CaV channel CaVAb, which is inhibited by dihydropyridines and phenylalkylamines with nanomolar affinity in a state-dependent manner. The binding site for amlodipine and other dihydropyridines is located on the external, lipid-facing surface of the pore module, positioned at the interface of two subunits. Dihydropyridine binding allosterically induces an asymmetric conformation of the selectivity filter, in which partially dehydrated Ca2+ interacts directly with one subunit and blocks the pore. In contrast, the phenylalkylamine Br-verapamil binds in the central cavity of the pore on the intracellular side of the selectivity filter, physically blocking the ion-conducting pathway. Structure-based mutations of key amino-acid residues confirm drug binding at both sites. Our results define the structural basis for binding of dihydropyridines and phenylalkylamines at their distinct receptor sites on CaV channels and offer key insights into their fundamental mechanisms of action and differential therapeutic uses in cardiovascular diseases.


Asunto(s)
Aminas/química , Aminas/farmacología , Bloqueadores de los Canales de Calcio/química , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio/química , Dihidropiridinas/química , Dihidropiridinas/farmacología , Regulación Alostérica/efectos de los fármacos , Aminas/efectos adversos , Amlodipino/química , Amlodipino/farmacología , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión/efectos de los fármacos , Sitios de Unión/genética , Calcio/química , Calcio/metabolismo , Canales de Calcio/genética , Canales de Calcio/metabolismo , Línea Celular , Cristalografía por Rayos X , Dihidropiridinas/efectos adversos , Lípidos/química , Modelos Moleculares , Mariposas Nocturnas , Mutación , Niacina/análogos & derivados , Niacina/química , Niacina/farmacología , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Verapamilo/química , Verapamilo/farmacología
2.
Proc Natl Acad Sci U S A ; 116(52): 26549-26554, 2019 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-31822620

RESUMEN

Valproic acid (VPA) is an anticonvulsant drug that is also used to treat migraines and bipolar disorder. Its proposed biological targets include human voltage-gated sodium channels, among other membrane proteins. We used the prokaryotic NavMs sodium channel, which has been shown to be a good exemplar for drug binding to human sodium channels, to examine the structural and functional interactions of VPA. Thermal melt synchrotron radiation circular dichroism spectroscopic binding studies of the full-length NavMs channel (which includes both pore and voltage sensor domains), and a pore-only construct, undertaken in the presence and absence of VPA, indicated that the drug binds to and destabilizes the channel, but not the pore-only construct. This is in contrast to other antiepileptic compounds that have previously been shown to bind in the central hydrophobic core of the pore region of the channel, and that tend to increase the thermal stability of both pore-only constructs and full-length channels. Molecular docking studies also indicated that the VPA binding site is associated with the voltage sensor, rather than the hydrophobic cavity of the pore domain. Electrophysiological studies show that VPA influences the block and inactivation rates of the NavMs channel, although with lower efficacy than classical channel-blocking compounds. It thus appears that, while VPA is capable of binding to these voltage-gated sodium channels, it has a very different mode and site of action than other anticonvulsant compounds.

3.
Proc Natl Acad Sci U S A ; 115(48): 12301-12306, 2018 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-30429323

RESUMEN

TRPA1, a member of the transient receptor potential channel (TRP) family, is genetically linked to pain in humans, and small molecule inhibitors are efficacious in preclinical animal models of inflammatory pain. These findings have driven significant interest in development of selective TRPA1 inhibitors as potential analgesics. The majority of TRPA1 inhibitors characterized to date have been reported to interact with the S5 transmembrane helices forming part of the pore region of the channel. However, the development of many of these inhibitors as clinical drug candidates has been prevented by high lipophilicity, low solubility, and poor pharmacokinetic profiles. Identification of alternate compound interacting sites on TRPA1 provides the opportunity to develop structurally distinct modulators with novel structure-activity relationships and more desirable physiochemical properties. In this paper, we have identified a previously undescribed potent and selective small molecule thiadiazole structural class of TRPA1 inhibitor. Using species ortholog chimeric and mutagenesis strategies, we narrowed down the site of interaction to ankyrinR #6 within the distal N-terminal region of TRPA1. To identify the individual amino acid residues involved, we generated a computational model of the ankyrinR domain. This model was used predictively to identify three critical amino acids in human TRPA1, G238, N249, and K270, which were confirmed by mutagenesis to account for compound activity. These findings establish a small molecule interaction region on TRPA1, expanding potential avenues for developing TRPA1 inhibitor analgesics and for probing the mechanism of channel gating.


Asunto(s)
Bibliotecas de Moléculas Pequeñas/química , Canal Catiónico TRPA1/química , Canal Catiónico TRPA1/metabolismo , Secuencia de Aminoácidos , Animales , Repetición de Anquirina , Humanos , Modelos Moleculares , Unión Proteica , Ratas , Alineación de Secuencia , Bibliotecas de Moléculas Pequeñas/metabolismo , Canal Catiónico TRPA1/antagonistas & inhibidores , Canal Catiónico TRPA1/genética
4.
Proc Natl Acad Sci U S A ; 111(23): 8428-33, 2014 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-24850863

RESUMEN

Voltage-gated sodium channels are important targets for the development of pharmaceutical drugs, because mutations in different human sodium channel isoforms have causal relationships with a range of neurological and cardiovascular diseases. In this study, functional electrophysiological studies show that the prokaryotic sodium channel from Magnetococcus marinus (NavMs) binds and is inhibited by eukaryotic sodium channel blockers in a manner similar to the human Nav1.1 channel, despite millions of years of divergent evolution between the two types of channels. Crystal complexes of the NavMs pore with several brominated blocker compounds depict a common antagonist binding site in the cavity, adjacent to lipid-facing fenestrations proposed to be the portals for drug entry. In silico docking studies indicate the full extent of the blocker binding site, and electrophysiology studies of NavMs channels with mutations at adjacent residues validate the location. These results suggest that the NavMs channel can be a valuable tool for screening and rational design of human drugs.


Asunto(s)
Alphaproteobacteria/metabolismo , Proteínas Bacterianas/metabolismo , Canal de Sodio Activado por Voltaje NAV1.1/metabolismo , Canales de Sodio/metabolismo , Alphaproteobacteria/química , Alphaproteobacteria/genética , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Sitios de Unión/genética , Cristalografía por Rayos X , Células HEK293 , Humanos , Activación del Canal Iónico/efectos de los fármacos , Activación del Canal Iónico/genética , Activación del Canal Iónico/fisiología , Lamotrigina , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Canal de Sodio Activado por Voltaje NAV1.1/química , Canal de Sodio Activado por Voltaje NAV1.1/genética , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Bloqueadores de los Canales de Sodio/metabolismo , Bloqueadores de los Canales de Sodio/farmacología , Canales de Sodio/química , Canales de Sodio/genética , Triazinas/metabolismo , Triazinas/farmacología
5.
Biophys J ; 111(4): 775-784, 2016 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-27558721

RESUMEN

Potassium channels in the two-pore domain family (K2P) have various structural attributes that differ from those of other K(+) channels, including a dimeric assembly constituted of nonidentical domains and an expansive extracellular cap. Crystallization of the prototypical K2P channel, TWIK-1, finally revealed the structure of these characteristics in atomic detail, allowing computational studies to be undertaken. In this study, we performed molecular-dynamics simulations for a cumulative time of ∼1 µs to discern the mechanism of ion transport throughout TWIK-1. We observed the free passage of ions beneath the extracellular cap and identified multiple high-occupancy sites in close proximity to charged residues on the protein surface. Despite the overall topological similarity of the x-ray structure of the selectivity filter to other K(+) channels, the structure diverges significantly in molecular-dynamics simulations as a consequence of nonconserved residues in both pore domains contributing to the selectivity filter (T118 and L228). The behavior of such residues has been linked to channel inactivation and the phenomenon of dynamic selectivity, where TWIK-1 displays robust Na(+) inward flux in response to subphysiological K(+) concentrations.


Asunto(s)
Canales de Potasio de Dominio Poro en Tándem/metabolismo , Sitios de Unión , Cristalografía por Rayos X , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Simulación de Dinámica Molecular , Canales de Potasio de Dominio Poro en Tándem/química , Conformación Proteica
6.
Mol Pharm ; 13(7): 2263-73, 2016 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-27173896

RESUMEN

Potassium channels are of paramount physiological and pathological importance and therefore constitute significant drug targets. One of the keys to rationalize the way drugs modulate ion channels is to understand the ability of such small molecules to access their respective binding sites, from which they can exert an activating or inhibitory effect. Many computational studies have probed the energetics of ion permeation, and the mechanisms of voltage gating, but little is known about the role of fenestrations as possible mediators of drug entry in potassium channels. To explore the existence, structure, and conformational dynamics of transmembrane fenestrations accessible by drugs in potassium channels, molecular dynamics simulation trajectories were analyzed from three potassium channels: the open state voltage-gated channel Kv1.2, the G protein-gated inward rectifying channel GIRK2 (Kir3.2), and the human two-pore domain TWIK-1 (K2P1.1). The main results of this work were the identification of the sequence identity of four main lateral fenestrations of similar length and with bottleneck radius in the range of 0.9-2.4 Å for this set of potassium channels. It was found that the fenestrations in Kv1.2 and Kir3.2 remain closed to the passage of molecules larger than water. In contrast, in the TWIK-1 channel, both open and closed fenestrations are sampled throughout the simulation, with bottleneck radius shown to correlate with the random entry of lipid membrane molecules into the aperture of the fenestrations. Druggability scoring function analysis of the fenestration regions suggests that Kv and Kir channels studied are not druggable in practice due to steric constraining of the fenestration bottleneck. A high (>50%) fenestration sequence identity was found in each potassium channel subfamily studied, Kv1, Kir3, and K2P1. Finally, the reported fenestration sequence of TWIK-1 compared favorably with another channel, K2P channel TREK-2, reported to possess open fenestrations, suggesting that K2P channels could be druggable via fenestrations, for which we reported atomistic detail of the fenestration region, including the flexible residues M260 and L264 that interact with POPC membrane in a concerted fashion with the aperture and closure of the fenestrations.


Asunto(s)
Canales de Potasio/metabolismo , Secuencia de Aminoácidos , Sitios de Unión/fisiología , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Humanos , Canal de Potasio Kv.1.2/metabolismo , Conformación Molecular , Simulación de Dinámica Molecular
7.
Mol Pharm ; 13(11): 4001-4012, 2016 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-27704838

RESUMEN

Selective modulators of the γ-amino butyric acid (GABAA) family of receptors have the potential to treat a range of disease states related to cognition, pain, and anxiety. While the development of various α subunit-selective modulators is currently underway for the treatment of anxiety disorders, a mechanistic understanding of the correlation between their bioactivity and efficacy, based on ligand-target interactions, is currently still lacking. In order to alleviate this situation, in the current study we have analyzed, using ligand- and structure-based methods, a data set of 5440 GABAA modulators. The Spearman correlation (ρ) between binding activity and efficacy of compounds was calculated to be 0.008 and 0.31 against the α1 and α2 subunits of GABA receptor, respectively; in other words, the compounds had little diversity in structure and bioactivity, but they differed significantly in efficacy. Two compounds were selected as a case study for detailed interaction analysis due to the small difference in their structures and affinities (ΔpKi(comp1_α1 - comp2_α1) = 0.45 log units, ΔpKi(comp1_α2 - comp2_α2) = 0 log units) as compared to larger relative efficacies (ΔRE(comp1_α1 - comp2_α1) = 1.03, ΔRE(comp1_α2 - comp2_α2) = 0.21). Docking analysis suggested that His-101 is involved in a characteristic interaction of the α1 receptor with both compounds 1 and 2. Residues such as Phe-77, Thr-142, Asn-60, and Arg-144 of the γ chain of the α1γ2 complex also showed interactions with heterocyclic rings of both compounds 1 and 2, but these interactions were disturbed in the case of α2γ2 complex docking results. Binding pocket stability analysis based on molecular dynamics identified three substitutions in the loop C region of the α2 subunit, namely, G200E, I201T, and V202I, causing a reduction in the flexibility of α2 compared to α1. These amino acids in α2, as compared to α1, were also observed to decrease the vibrational and dihedral entropy and to increase the hydrogen bond content in α2 in the apo state. However, freezing of both α1 and α2 was observed in the ligand-bound state, with an increased number of internal hydrogen bonds and increased entropy. Therefore, we hypothesize that the amino acid differences in the loop C region of α2 are responsible for conformational changes in the protein structure compared to α1, as well as for the binding modes of compounds and hence their functional signaling.


Asunto(s)
Receptores de GABA/metabolismo , Secuencia de Aminoácidos , Animales , Benzodiazepinas/farmacología , Ácido Butírico/farmacología , Agonistas de Receptores de GABA-A/farmacología , Humanos , Enlace de Hidrógeno , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Análisis de Componente Principal , Estructura Secundaria de Proteína , Receptores de GABA/química
8.
Bioorg Med Chem Lett ; 26(20): 4919-4924, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27641472

RESUMEN

TRESK (Twik RElated Spinal cord K+ channel) is a member of the Twin Pore Domain potassium channel (K2P) family responsible for regulating neuronal excitability in dorsal root ganglion (DRG) and trigeminal (TG) neurons, peripheral neurons involved in pain transmission. As channel opening causes an outward K+ current responsible for cell hyperpolarisation, TRESK represents a potentially interesting target for pain treatment. However, as no crystal structure exists for this protein, the mechanisms involved in the opening action of its ligands are still poorly understood, making the development of new potent and selective openers challenging. In this work we present a structure activity relationship (SAR) of the known TRESK opener flufenamic acid (FFA) and some derivatives, investigating the functional effects of chemical modifications to build a TRESK homology model to support the biological results. A plausible binding mode is proposed, providing the first predictive hypothesis of a human TRESK opener binding site.


Asunto(s)
Ácido Flufenámico/química , Ácido Flufenámico/farmacología , Canales de Potasio/química , Animales , Sitios de Unión , Células HEK293 , Humanos , Ratones , Neuronas/efectos de los fármacos , Relación Estructura-Actividad
9.
Org Biomol Chem ; 14(28): 6676-8, 2016 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-27327397

RESUMEN

The design and synthesis of azogabazine is described, which represents a highly potent (IC50 = 23 nM) photoswitchable antagonist of the GABAA receptor. An azologization strategy is adopted, in which a benzyl phenyl ether in a high affinity gabazine analogue is replaced by an azobenzene, with resultant retention of antagonist potency. We show that cycling from blue to UV light, switching between trans and cis isomeric forms, leads to photochemically controlled antagonism of the GABA ion channel.


Asunto(s)
Antagonistas de Receptores de GABA-A/química , Antagonistas de Receptores de GABA-A/farmacología , Piridazinas/química , Piridazinas/farmacología , Compuestos Azo/síntesis química , Compuestos Azo/química , Compuestos Azo/farmacología , Diseño de Fármacos , Antagonistas de Receptores de GABA-A/síntesis química , Células HEK293 , Humanos , Piridazinas/síntesis química , Receptores de GABA-A/metabolismo
10.
Mol Pharm ; 12(4): 1299-307, 2015 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-25734225

RESUMEN

Voltage-gated potassium channels of the Kv1 family play a crucial role in the generation and transmission of electrical signals in excitable cells affecting neuronal and cardiac activities. Small-molecule blockage of these channels has been proposed to occur via a cooperative mechanism involving two main blocking sites: the inner-pore site located below the selectivity filter, and a side-pocket cavity located between the pore and the voltage sensor. Using 0.5 µs molecular dynamics simulation trajectories complemented by docking calculations, the potential binding sites of the PAP-1 (5-(4-phenoxybutoxy)psoralen) blocker to the crystal structure of Kv1.2 channel have been studied. The presence of both mentioned blocking sites at Kv1.2 is confirmed, adding evidence in favor of a cooperative channel blockage mechanism. These observations provide insight into drug modulation that will guide further developments of Kv inhibitors.


Asunto(s)
Ficusina/química , Canal de Potasio Kv.1.2/química , Secuencia de Aminoácidos , Animales , Sitios de Unión , Análisis por Conglomerados , Cristalización , Electroquímica , Humanos , Ligandos , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Neuronas/patología , Proteínas Asociadas a Pancreatitis , Ratas , Homología de Secuencia de Aminoácido , Solventes/química
11.
Angew Chem Int Ed Engl ; 53(24): 6126-30, 2014 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-24821300

RESUMEN

The benzoxazinone and dihydroquinoxalinone fragments were employed as novel acetyl lysine mimics in the development of CREBBP bromodomain ligands. While the benzoxazinone series showed low affinity for the CREBBP bromodomain, expansion of the dihydroquinoxalinone series resulted in the first potent inhibitors of a bromodomain outside the BET family. Structural and computational studies reveal that an internal hydrogen bond stabilizes the protein-bound conformation of the dihydroquinoxalinone series. The side chain of this series binds in an induced-fit pocket forming a cation-π interaction with R1173 of CREBBP. The most potent compound inhibits binding of CREBBP to chromatin in U2OS cells.


Asunto(s)
Proteína de Unión a CREB/genética , Cationes/química , Epigenómica/métodos , Ligandos , Modelos Moleculares , Unión Proteica
12.
Bioorg Med Chem Lett ; 23(3): 827-33, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23265891

RESUMEN

Several non-benzimidazole containing inhibitors of respiratory syncytial virus are described. Core template modification, analysis of antiviral activity, physicochemistry and optimisation of properties led to the thiazole-imidazole 13, that showed a good potency and pharmacokinetic profile in the rat.


Asunto(s)
Antivirales/síntesis química , Antivirales/farmacología , Virus Sincitiales Respiratorios/efectos de los fármacos , Animales , Antivirales/química , Bencimidazoles/química , Imidazoles/síntesis química , Imidazoles/farmacocinética , Imidazoles/farmacología , Concentración 50 Inhibidora , Ratas , Relación Estructura-Actividad , Tiazoles/síntesis química , Tiazoles/farmacocinética , Tiazoles/farmacología
13.
Org Biomol Chem ; 11(19): 3230-46, 2013 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-23584232

RESUMEN

Chiral N-heterocyclic carbenes (NHCs) promote the asymmetric formal [4 + 2] cycloaddition of alkylarylketenes with ß,γ-unsaturated α-ketocarboxylic esters and amides. Divergent diastereoselectivity is observed in this process, with γ-aryl-ß,γ-unsaturated α-ketocarboxylic esters and amides giving preferentially syn-dihydropyranones (up to 68 : 32 dr syn : anti, up to 98% ee), while γ-alkyl-derivatives generate anti-dihydropyranones (up to 18 : 82 dr syn : anti, up to 75% ee).


Asunto(s)
Amidas/síntesis química , Ácidos Carboxílicos/síntesis química , Ésteres/síntesis química , Etilenos/síntesis química , Compuestos Heterocíclicos/química , Cetonas/síntesis química , Metano/análogos & derivados , Amidas/química , Ácidos Carboxílicos/química , Cristalografía por Rayos X , Ciclización , Ésteres/química , Etilenos/química , Cetonas/química , Metano/química , Modelos Moleculares , Estructura Molecular , Estereoisomerismo
14.
J Antimicrob Chemother ; 67(4): 789-801, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22258929

RESUMEN

In the past, antiviral research has focused mainly on viral targets. As the search for effective and differentiated antiviral therapies continues, cellular targets are becoming more common, bringing with them a variety of challenges and concerns. Toll-like receptors (TLRs) provide a unique mechanism to induce an antiviral state in the host. In this review we introduce TLRs as targets for the pharmaceutical industry, including how they signal and thereby induce an antiviral state through the production of type I interferons. We examine how TLRs are being therapeutically targeted and discuss several clinically precedented agents for which efficacy and safety data are available. We describe some of the chemistries that have been applied to both small molecule and large molecule leads to tune agonist potency, and offer a differentiated safety profile through targeting certain compartments such as the gut or the lung, thereby limiting systemic drug exposure and affecting systemic cytokine levels. The application of low-dose agonists of TLRs as vaccine adjuvants or immunoprotective agents is also presented. Some of the challenges presented by this approach are then discussed, including viral evasion strategies and mechanism-linked inflammatory cytokine induction.


Asunto(s)
Antivirales/administración & dosificación , Factores Inmunológicos/administración & dosificación , Receptores Toll-Like/agonistas , Virosis/tratamiento farmacológico , Virosis/inmunología , Antivirales/química , Antivirales/farmacología , Humanos , Factores Inmunológicos/química , Factores Inmunológicos/farmacología , Interferones/inmunología , Interferones/metabolismo
15.
J Virol ; 85(13): 6353-68, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21507963

RESUMEN

The current standard of care for hepatitis C virus (HCV)-infected patients consists of lengthy treatment with interferon and ribavirin. To increase the effectiveness of HCV therapy, future regimens will incorporate multiple direct-acting antiviral (DAA) drugs. Recently, the HCV-encoded NS5A protein has emerged as a promising DAA target. Compounds targeting NS5A exhibit remarkable potency in vitro and demonstrate early clinical promise, suggesting that NS5A inhibitors could feature in future DAA combination therapies. Since the mechanisms through which these molecules operate are unknown, we have used NS5A inhibitors as tools to investigate their modes of action. Analysis of replicon-containing cells revealed dramatic phenotypic alterations in NS5A localization following treatment with NS5A inhibitors; NS5A was redistributed from the endoplasmic reticulum to lipid droplets. The NS5A relocalization did not occur in cells treated with other classes of HCV inhibitors, and NS5A-targeting molecules did not cause similar alterations in the localization of other HCV-encoded proteins. Time course analysis of the redistribution of NS5A revealed that the transfer of protein to lipid droplets was concomitant with the onset of inhibition, as judged by the kinetic profiles for these compounds. Furthermore, analysis of the kinetic profile of inhibition for a panel of test molecules permitted the separation of compounds into different kinetic classes based on their modes of action. Results from this approach suggested that NS5A inhibitors perturbed the function of new replication complexes, rather than acting on preformed complexes. Taken together, our data reveal novel biological consequences of NS5A inhibition, which may help enable the development of future assay platforms for the identification of new and/or different NS5A inhibitors.


Asunto(s)
Antivirales/farmacología , Retículo Endoplásmico/metabolismo , Imidazoles/farmacología , Proteínas no Estructurales Virales/antagonistas & inhibidores , Antivirales/química , Carbamatos , Línea Celular Tumoral , Retículo Endoplásmico/ultraestructura , Hepacivirus/efectos de los fármacos , Hepacivirus/genética , Hepatocitos/ultraestructura , Hepatocitos/virología , Humanos , Imidazoles/química , Concentración 50 Inhibidora , Lípidos , Microscopía Confocal , Modelos Moleculares , Pirrolidinas , Replicón , Bibliotecas de Moléculas Pequeñas , Valina/análogos & derivados , Proteínas no Estructurales Virales/metabolismo , Replicación Viral
17.
Bioorg Med Chem Lett ; 22(8): 2856-60, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22429467

RESUMEN

Aldehyde oxidase (AO) is a molybdenum-containing enzyme distributed throughout the animal kingdom and capable of metabolising a wide range of aldehydes and N-heterocyclic compounds. Although metabolism by this enzyme in man is recognised to have significant clinical impact where human AO activity was not predicted by screening in preclinical species, there is very little reported literature offering real examples where drug discoverers have successfully designed away from AO oxidation. This article reports on some strategies adopted in the Pfizer TLR7 agonist programme to successfully switch off AO metabolism that was seen principally in the rat.


Asunto(s)
Aldehído Oxidasa/metabolismo , Piridinas/síntesis química , Aldehído Oxidasa/antagonistas & inhibidores , Aldehído Oxidasa/química , Animales , Células Cultivadas , Química Farmacéutica , Citosol/enzimología , Perros , Estabilidad de Medicamentos , Humanos , Masculino , Piridinas/química , Ratas , Relación Estructura-Actividad , Receptor Toll-Like 7/agonistas
18.
Eur J Med Chem ; 229: 114087, 2022 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-34998056

RESUMEN

STING mediates innate immune responses that are triggered by the presence of cytosolic DNA. Activation of STING to boost antigen recognition is a therapeutic modality that is currently being tested in cancer patients using nucleic-acid based macrocyclic STING ligands. We describe here the discovery of 3,4-dihydroquinazolin-2(1H)-one based 6,6-bicyclic heterocyclic agonists of human STING that activate all known human variants of STING with high potency.


Asunto(s)
Antineoplásicos/síntesis química , Compuestos Bicíclicos Heterocíclicos con Puentes/síntesis química , Inmunidad Innata/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Neoplasias/tratamiento farmacológico , Bibliotecas de Moléculas Pequeñas/síntesis química , Animales , Antineoplásicos/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Citosol/química , ADN/química , Haplorrinos , Humanos , Masculino , Proteínas de la Membrana/genética , Ratones Endogámicos BALB C , Unión Proteica , Transducción de Señal , Bibliotecas de Moléculas Pequeñas/farmacología , Relación Estructura-Actividad
19.
Antimicrob Agents Chemother ; 55(7): 3105-14, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21502629

RESUMEN

We have screened 47 locked nucleic acid (LNA) antisense oligonucleotides (ASOs) targeting conserved (>95% homology) sequences in the hepatitis C virus (HCV) genome using the subgenomic HCV replicon assay and generated both antiviral (50% effective concentration [EC(50)]) and cytotoxic (50% cytotoxic concentration [CC(50)]) dose-response curves to allow measurement of the selectivity index (SI). This comprehensive approach has identified an LNA ASO with potent antiviral activity (EC(50) = 4 nM) and low cytotoxicity (CC(50) >880 nM) targeting the 25- to 40-nucleotide region (nt) of the HCV internal ribosome entry site (IRES) containing the distal and proximal miR-122 binding sites. LNA ASOs targeting previously known accessible regions of the IRES, namely, loop III and the initiation codon in loop IV, had poor SI values. We optimized the LNA ASO sequence by performing a 1-nucleotide walk through the 25- to 40-nt region and show that the boundaries for antiviral efficacy are extremely precise. Furthermore, we have optimized the format for the LNA ASO using different gapmer and mixomer patterns and show that RNase H is required for antiviral activity. We demonstrate that RNase H-refractory ASOs targeting the 25- to 40-nt region have no antiviral effect, revealing important regulatory features of the 25- to 40-nt region and suggesting that RNase H-refractory LNA ASOs can act as potential surrogates for proviral functions of miR-122. We confirm the antisense mechanism of action using mismatched LNA ASOs. Finally, we have performed pharmacokinetic experiments to demonstrate that the LNA ASOs have a very long half-life (>5 days) and attain hepatic maximum concentrations >100 times the concentration required for in vitro antiviral activity.


Asunto(s)
Antivirales/farmacología , Antivirales/farmacocinética , Hepacivirus/efectos de los fármacos , Oligonucleótidos Antisentido/farmacología , Oligonucleótidos Antisentido/farmacocinética , Ribosomas/virología , Animales , Línea Celular , Humanos , Riñón/metabolismo , Hígado/metabolismo , Ratones
20.
Bioorg Med Chem Lett ; 21(19): 5939-43, 2011 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-21885277

RESUMEN

The discovery of a series of highly potent and novel TLR7 agonist interferon inducers is described. Structure-activity relationships are presented, along with pharmacokinetic studies of a lead molecule from this series of N9-pyridylmethyl-8-oxo-3-deazapurine analogues. A rationale for the very high potency observed is offered. An investigation of the clearance mechanism of this class of compounds in rat was carried out, resulting in aldehyde oxidase mediated oxidation being identified as a key component of the high clearance observed. A possible solution to this problem is discussed.


Asunto(s)
Antivirales/síntesis química , Antivirales/farmacología , Hepacivirus/efectos de los fármacos , Hepatitis C/tratamiento farmacológico , Interferones/agonistas , Receptor Toll-Like 7/agonistas , Aldehído Oxidasa/metabolismo , Animales , Antivirales/química , Antivirales/farmacocinética , Relación Dosis-Respuesta a Droga , Descubrimiento de Drogas , Evaluación Preclínica de Medicamentos , Hepacivirus/fisiología , Hepatitis C/virología , Humanos , Inyecciones Intravenosas , Inductores de Interferón/síntesis química , Inductores de Interferón/química , Inductores de Interferón/farmacocinética , Inductores de Interferón/farmacología , Microsomas Hepáticos/metabolismo , Terapia Molecular Dirigida , Peso Molecular , Purinas/síntesis química , Purinas/metabolismo , Ratas , Solubilidad , Estereoisomerismo , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA