Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
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
2.
Pharm Res ; 36(9): 137, 2019 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-31332533

RESUMEN

PURPOSE: Pitt Hopkins Syndrome (PTHS) is a rare genetic disorder caused by mutations of a specific gene, transcription factor 4 (TCF4), located on chromosome 18. PTHS results in individuals that have moderate to severe intellectual disability, with most exhibiting psychomotor delay. PTHS also exhibits features of autistic spectrum disorders, which are characterized by the impaired ability to communicate and socialize. PTHS is comorbid with a higher prevalence of epileptic seizures which can be present from birth or which commonly develop in childhood. Attenuated or absent TCF4 expression results in increased translation of peripheral ion channels Kv7.1 and Nav1.8 which triggers an increase in after-hyperpolarization and altered firing properties. METHODS: We now describe a high throughput screen (HTS) of 1280 approved drugs and machine learning models developed from this data. The ion channels were expressed in either CHO (KV7.1) or HEK293 (Nav1.8) cells and the HTS used either 86Rb+ efflux (KV7.1) or a FLIPR assay (Nav1.8). RESULTS: The HTS delivered 55 inhibitors of Kv7.1 (4.2% hit rate) and 93 inhibitors of Nav1.8 (7.2% hit rate) at a screening concentration of 10 µM. These datasets also enabled us to generate and validate Bayesian machine learning models for these ion channels. We also describe a structure activity relationship for several dihydropyridine compounds as inhibitors of Nav1.8. CONCLUSIONS: This work could lead to the potential repurposing of nicardipine or other dihydropyridine calcium channel antagonists as potential treatments for PTHS acting via Nav1.8, as there are currently no approved treatments for this rare disorder.


Asunto(s)
Dihidropiridinas/farmacología , Reposicionamiento de Medicamentos/métodos , Hiperventilación/tratamiento farmacológico , Discapacidad Intelectual/tratamiento farmacológico , Canal de Potasio KCNQ1/antagonistas & inhibidores , Canal de Sodio Activado por Voltaje NAV1.8/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Bloqueadores de los Canales de Sodio/farmacología , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacología , Animales , Teorema de Bayes , Células CHO , Cricetulus , Dihidropiridinas/química , Facies , Células HEK293 , Humanos , Canal de Potasio KCNQ1/metabolismo , Aprendizaje Automático , Bloqueadores de los Canales de Potasio/química , Bibliotecas de Moléculas Pequeñas/química , Bloqueadores de los Canales de Sodio/química , Relación Estructura-Actividad , Bloqueadores del Canal de Sodio Activado por Voltaje/química
3.
Epilepsia ; 59(4): 802-813, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29574705

RESUMEN

OBJECTIVE: Many previous studies of drug repurposing have relied on literature review followed by evaluation of a limited number of candidate compounds. Here, we demonstrate the feasibility of a more comprehensive approach using high-throughput screening to identify inhibitors of a gain-of-function mutation in the SCN8A gene associated with severe pediatric epilepsy. METHODS: We developed cellular models expressing wild-type or an R1872Q mutation in the Nav 1.6 sodium channel encoded by SCN8A. Voltage clamp experiments in HEK-293 cells expressing the SCN8A R1872Q mutation demonstrated a leftward shift in sodium channel activation as well as delayed inactivation; both changes are consistent with a gain-of-function mutation. We next developed a fluorescence-based, sodium flux assay and used it to assess an extensive library of approved drugs, including a panel of antiepileptic drugs, for inhibitory activity in the mutated cell line. Lead candidates were evaluated in follow-on studies to generate concentration-response curves for inhibiting sodium influx. Select compounds of clinical interest were evaluated by electrophysiology to further characterize drug effects on wild-type and mutant sodium channel functions. RESULTS: The screen identified 90 drugs that significantly inhibited sodium influx in the R1872Q cell line. Four drugs of potential clinical interest-amitriptyline, carvedilol, nilvadipine, and carbamazepine-were further investigated and demonstrated concentration-dependent inhibition of sodium channel currents. SIGNIFICANCE: A comprehensive drug repurposing screen identified potential new candidates for the treatment of epilepsy caused by the R1872Q mutation in the SCN8A gene.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Reposicionamiento de Medicamentos/métodos , Epilepsia/tratamiento farmacológico , Epilepsia/genética , Ensayos Analíticos de Alto Rendimiento/métodos , Canal de Sodio Activado por Voltaje NAV1.6/genética , Anticonvulsivantes/farmacología , Niño , Relación Dosis-Respuesta a Droga , Epilepsia/diagnóstico , Femenino , Células HEK293 , Humanos , Masculino , Mutación/efectos de los fármacos , Mutación/genética
4.
Proc Natl Acad Sci U S A ; 110(29): E2724-32, 2013 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-23818614

RESUMEN

Voltage-gated sodium (Nav) channels play a fundamental role in the generation and propagation of electrical impulses in excitable cells. Here we describe two unique structurally related nanomolar potent small molecule Nav channel inhibitors that exhibit up to 1,000-fold selectivity for human Nav1.3/Nav1.1 (ICA-121431, IC50, 19 nM) or Nav1.7 (PF-04856264, IC50, 28 nM) vs. other TTX-sensitive or resistant (i.e., Nav1.5) sodium channels. Using both chimeras and single point mutations, we demonstrate that this unique class of sodium channel inhibitor interacts with the S1-S4 voltage sensor segment of homologous Domain 4. Amino acid residues in the "extracellular" facing regions of the S2 and S3 transmembrane segments of Nav1.3 and Nav1.7 seem to be major determinants of Nav subtype selectivity and to confer differences in species sensitivity to these inhibitors. The unique interaction region on the Domain 4 voltage sensor segment is distinct from the structural domains forming the channel pore, as well as previously characterized interaction sites for other small molecule inhibitors, including local anesthetics and TTX. However, this interaction region does include at least one amino acid residue [E1559 (Nav1.3)/D1586 (Nav1.7)] that is important for Site 3 α-scorpion and anemone polypeptide toxin modulators of Nav channel inactivation. The present study provides a potential framework for identifying subtype selective small molecule sodium channel inhibitors targeting interaction sites away from the pore region.


Asunto(s)
Acetamidas/farmacología , Fenómenos Electrofisiológicos/fisiología , Canal de Sodio Activado por Voltaje NAV1.3/metabolismo , Tiazoles/farmacología , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacología , Secuencias de Aminoácidos/genética , Sitios de Unión/genética , Células HEK293 , Humanos , Concentración 50 Inhibidora , Datos de Secuencia Molecular , Canal de Sodio Activado por Voltaje NAV1.3/genética , Técnicas de Placa-Clamp , Alineación de Secuencia
5.
J Clin Invest ; 134(5)2024 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-38227370

RESUMEN

Two coding variants of apolipoprotein L1 (APOL1), called G1 and G2, explain much of the excess risk of kidney disease in African Americans. While various cytotoxic phenotypes have been reported in experimental models, the proximal mechanism by which G1 and G2 cause kidney disease is poorly understood. Here, we leveraged 3 experimental models and a recently reported small molecule blocker of APOL1 protein, VX-147, to identify the upstream mechanism of G1-induced cytotoxicity. In HEK293 cells, we demonstrated that G1-mediated Na+ import/K+ efflux triggered activation of GPCR/IP3-mediated calcium release from the ER, impaired mitochondrial ATP production, and impaired translation, which were all reversed by VX-147. In human urine-derived podocyte-like epithelial cells (HUPECs), we demonstrated that G1 caused cytotoxicity that was again reversible by VX-147. Finally, in podocytes isolated from APOL1 G1 transgenic mice, we showed that IFN-γ-mediated induction of G1 caused K+ efflux, activation of GPCR/IP3 signaling, and inhibition of translation, podocyte injury, and proteinuria, all reversed by VX-147. Together, these results establish APOL1-mediated Na+/K+ transport as the proximal driver of APOL1-mediated kidney disease.


Asunto(s)
Apolipoproteína L1 , Enfermedades Renales , Compuestos Organotiofosforados , Ratones , Animales , Humanos , Apolipoproteína L1/genética , Células HEK293 , Variación Genética , Enfermedades Renales/genética , Ratones Transgénicos
6.
ACS Med Chem Lett ; 12(4): 593-602, 2021 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-33859800

RESUMEN

The gene KCNT1 encodes the sodium-activated potassium channel KNa1.1 (Slack, Slo2.2). Variants in the KCNT1 gene induce a gain-of-function (GoF) phenotype in ionic currents and cause a spectrum of intractable neurological disorders in infants and children, including epilepsy of infancy with migrating focal seizures (EIMFS) and autosomal dominant nocturnal frontal lobe epilepsy (ADNFLE). Effective treatment options for KCNT1-related disease are absent, and novel therapies are urgently required. We describe the development of a novel class of oxadiazole KNa1.1 inhibitors, leading to the discovery of compound 31 that reduced seizures and interictal spikes in a mouse model of KCNT1 GoF.

7.
Bioorg Med Chem Lett ; 20(22): 6812-5, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-20855211

RESUMEN

A series of aryl-substituted nicotinamide derivatives with selective inhibitory activity against the Na(v)1.8 sodium channel is reported. Replacement of the furan nucleus and homologation of the anilide linker in subtype-selective blocker A-803467 (1) provided potent, selective derivatives with improved aqueous solubility and oral bioavailability. Representative compounds from this series displayed efficacy in rat models of inflammatory and neuropathic pain.


Asunto(s)
Niacinamida/farmacología , Bloqueadores de los Canales de Sodio/farmacología , Administración Oral , Animales , Disponibilidad Biológica , Niacinamida/química , Niacinamida/farmacocinética , Ratas , Bloqueadores de los Canales de Sodio/administración & dosificación , Bloqueadores de los Canales de Sodio/química , Bloqueadores de los Canales de Sodio/farmacocinética , Relación Estructura-Actividad
8.
Bioorg Med Chem ; 18(22): 7816-25, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-20965738

RESUMEN

Na(v)1.8 (also known as PN3) is a tetrodotoxin-resistant (TTx-r) voltage-gated sodium channel (VGSC) that is highly expressed on small diameter sensory neurons. It has been implicated in the pathophysiology of inflammatory and neuropathic pain, and we envisioned that selective blockade of Na(v)1.8 would be analgesic, while reducing adverse events typically associated with non-selective VGSC blocking therapeutic agents. Herein, we describe the preparation and characterization of a series of 6-aryl-2-pyrazinecarboxamides, which are potent blockers of the human Na(v)1.8 channel and also block TTx-r sodium currents in rat dorsal root ganglia (DRG) neurons. Selected derivatives display selectivity versus human Na(v)1.2. We further demonstrate that an example from this series is orally bioavailable and produces antinociceptive activity in vivo in a rodent model of neuropathic pain following oral administration.


Asunto(s)
Neuralgia/tratamiento farmacológico , Pirazinas/química , Bloqueadores de los Canales de Sodio/química , Canales de Sodio/química , Administración Oral , Animales , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Ganglios Espinales/citología , Humanos , Microsomas/metabolismo , Canal de Sodio Activado por Voltaje NAV1.8 , Neuronas/metabolismo , Pirazinas/farmacocinética , Pirazinas/uso terapéutico , Ratas , Bloqueadores de los Canales de Sodio/farmacocinética , Bloqueadores de los Canales de Sodio/uso terapéutico , Canales de Sodio/metabolismo , Relación Estructura-Actividad
9.
Anesth Analg ; 109(2): 632-40, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19608841

RESUMEN

BACKGROUND: A non-opioid receptor-mediated inhibition of sodium channels in dorsal root ganglia (DRGs) by kappa-opioid receptor agonists (kappa-ORAs) has been reported to contribute to the antinociceptive actions in animals and humans. In this study, we examined structurally diverse kappa-ORAs for their abilities to inhibit tetrodotoxin-resistant (TTX-r) sodium channels in adult rat DRGs. METHODS: Whole-cell recordings of TTX-r sodium currents were performed on cultured adult rat DRGs. Structurally diverse kappa-ORAs were studied for their abilities to inhibit TTX-r sodium channels. RESULTS: The racemic kappa-ORA, (+/-)U50,488, inhibited TTX-r sodium currents in a voltage-dependent manner, yielding IC(50) values of 49 and 8 muM, at prepulse potentials of -100 and -40 mV, respectively. Furthermore, we found that both the kappa-ORA U50,488 active enantiomer 1S,2S U50,488 and the inactive enantiomer 1R,2R U50,488 were equally potent inhibitors of TTX-r sodium currents. Structurally related kappa-ORAs, such as BRL 52537 and ICI 199,441 also inhibited TTX-r sodium currents. However, sodium channel inhibition and kappa-opioid receptor agonism have a distinct structure-activity relationship because another kappa-ORA (ICI 204,488) was inactive versus TTX-r sodium channels. We further investigated the sodium channel block of this class of compounds by studying (+/-)U50,488. (+/-)U50,488 was found to preferentially interact with the slow inactivated state of TTX-r sodium channels and to retard recovery from inactivation. CONCLUSION: Our results suggest that TTX-r sodium channels can be inhibited by many kappa-ORAs via an opioid receptor-independent mechanism. Although the potency for sodium channel inhibition is typically much less than apparent affinity for opioid receptors, sodium channel block may still contribute to the antinociceptive effects of this class of compounds.


Asunto(s)
Analgésicos Opioides/farmacología , Receptores Opioides kappa/agonistas , Células Receptoras Sensoriales/efectos de los fármacos , Bloqueadores de los Canales de Sodio/farmacología , Canales de Sodio/efectos de los fármacos , Tetrodotoxina/farmacología , 3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/farmacología , Analgésicos no Narcóticos/farmacología , Animales , Células Cultivadas , Ganglios Espinales/citología , Ganglios Espinales/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Dimensión del Dolor/efectos de los fármacos , Técnicas de Placa-Clamp , Piperidinas/farmacología , Pirrolidinas/farmacología , Ratas , Relación Estructura-Actividad
10.
Br J Pharmacol ; 175(12): 2272-2283, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29150838

RESUMEN

BACKGROUND AND PURPOSE: TREK two-pore-domain potassium (K2P ) channels play a critical role in regulating the excitability of somatosensory nociceptive neurons and are important mediators of pain perception. An understanding of the roles of TREK channels in pain perception and, indeed, in other pathophysiological conditions, has been severely hampered by the lack of potent and/or selective activators and inhibitors. In this study, we describe a new, selective opener of TREK channels, GI-530159. EXPERIMENTAL APPROACH: The effect of GI-530159 on TREK channels was demonstrated using 86 Rb efflux assays, whole-cell and single-channel patch-clamp recordings from recombinant TREK channels. The expression of K2P 2.1 (TREK1), K2P 10.1 (TREK2) and K2P 4.1 (TRAAK) channels was determined using transcriptome analysis from single dorsal root ganglion (DRG) cells. Current-clamp recordings from cultured rat DRG neurons were used to measure the effect of GI-530159 on neuronal excitability. KEY RESULTS: For recombinant human TREK1 channels, GI-530159 had similar low EC50 values in Rb efflux experiments and electrophysiological recordings. It activated TREK2 channels, but it had no detectable action on TRAAK channels nor any significant effect on other K channels tested. Current-clamp recordings from cultured rat DRG neurones showed that application of GI-530159 at 1 µM resulted in a significant reduction in firing frequency and a small hyperpolarization of resting membrane potential. CONCLUSIONS AND IMPLICATIONS: This study provides pharmacological evidence for the presence of mechanosensitive TREK K2P channels in sensory neurones and suggests that development of selective K2P channel openers like GI-530159 could aid in the development of novel analgesic agents. LINKED ARTICLES: This article is part of a themed section on Recent Advances in Targeting Ion Channels to Treat Chronic Pain. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.12/issuetoc.


Asunto(s)
Ganglios Espinales/efectos de los fármacos , Neuronas/efectos de los fármacos , Canales de Potasio de Dominio Poro en Tándem/agonistas , Animales , Células CHO , Línea Celular , Cricetulus , Relación Dosis-Respuesta a Droga , Ganglios Espinales/metabolismo , Células HEK293 , Humanos , Estructura Molecular , Neuronas/metabolismo , Ratas , Relación Estructura-Actividad
11.
ACS Med Chem Lett ; 9(2): 125-130, 2018 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-29456800

RESUMEN

Inhibitors of the renal outer medullary potassium channel (ROMK) show promise as novel mechanism diuretics, with potentially lower risk of diuretic-induced hypokalemia relative to current thiazide and loop diuretics. Here, we report the identification of a novel series of 3-sulfamoylbenzamide ROMK inhibitors. Starting from HTS hit 4, this series was optimized to provide ROMK inhibitors with good in vitro potencies and well-balanced ADME profiles. In contrast to previously reported small-molecule ROMK inhibitors, members of this series were demonstrated to be highly selective for inhibition of human over rat ROMK and to be insensitive to the N171D pore mutation that abolishes inhibitory activity of previously reported ROMK inhibitors.

12.
ACS Med Chem Lett ; 8(6): 666-671, 2017 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-28626530

RESUMEN

A series of TRPA1 antagonists is described which has as its core structure an indazole moiety. The physical properties and in vitro DMPK profiles are discussed. Good in vivo exposure was obtained with several analogs, allowing efficacy to be assessed in rodent models of inflammatory pain. Two compounds showed significant activity in these models when administered either systemically or topically. Protein chimeras were constructed to indicate compounds from the series bound in the S5 region of the channel, and a computational docking model was used to propose a binding mode for example compounds.

13.
Channels (Austin) ; 9(6): 376-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26556675

RESUMEN

A prerequisite for a successful target-based drug discovery program is a robust data set that increases confidence in the validation of the molecular target and the therapeutic approach. Given the significant time and resource investment required to carry a drug to market, early selection of targets that can be modulated safely and effectively forms the basis for a strong portfolio and pipeline. In this article we present some of the more useful scientific approaches that can be applied toward the validation of ion channel targets, a molecular family with a history of clinical success in therapeutic areas such as cardiovascular, respiratory, pain and neuroscience.


Asunto(s)
Descubrimiento de Drogas/métodos , Canales Iónicos/metabolismo , Moduladores del Transporte de Membrana/farmacología , Animales , Genoma Humano , Humanos , Canales Iónicos/efectos de los fármacos , Canales Iónicos/genética , Moduladores del Transporte de Membrana/química , Estudios de Validación como Asunto
14.
Br J Pharmacol ; 172(10): 2654-70, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25625641

RESUMEN

BACKGROUND AND PURPOSE: NaV 1.8 ion channels have been highlighted as important molecular targets for the design of low MW blockers for the treatment of chronic pain. Here, we describe the effects of PF-01247324, a new generation, selective, orally bioavailable Nav 1.8 channel blocker of novel chemotype. EXPERIMENTAL APPROACH: The inhibition of Nav 1.8 channels by PF-01247324 was studied using in vitro patch-clamp electrophysiology and the oral bioavailability and antinociceptive effects demonstrated using in vivo rodent models of inflammatory and neuropathic pain. KEY RESULTS: PF-01247324 inhibited native tetrodotoxin-resistant (TTX-R) currents in human dorsal root ganglion (DRG) neurons (IC50 : 331 nM) and in recombinantly expressed h Nav 1.8 channels (IC50 : 196 nM), with 50-fold selectivity over recombinantly expressed TTX-R hNav 1.5 channels (IC50 : ∼10 µM) and 65-100-fold selectivity over TTX-sensitive (TTX-S) channels (IC50 : ∼10-18 µM). Native TTX-R currents in small-diameter rodent DRG neurons were inhibited with an IC50 448 nM, and the block of both human recombinant Nav 1.8 channels and TTX-R from rat DRG neurons was both frequency and state dependent. In vitro current clamp showed that PF-01247324 reduced excitability in both rat and human DRG neurons and also altered the waveform of the action potential. In vivo experiments n rodents demonstrated efficacy in both inflammatory and neuropathic pain models. CONCLUSIONS AND IMPLICATIONS: Using PF-01247324, we have confirmed a role for Nav 1.8 channels in both inflammatory and neuropathic pain. We have also demonstrated a key role for Nav 1.8 channels in action potential upstroke and repetitive firing of rat and human DRG neurons.


Asunto(s)
Nocicepción/efectos de los fármacos , Ácidos Picolínicos/farmacología , Células Receptoras Sensoriales/efectos de los fármacos , Bloqueadores de los Canales de Sodio/farmacología , Potenciales de Acción/efectos de los fármacos , Administración Oral , Animales , Ganglios Espinales/efectos de los fármacos , Células HEK293 , Humanos , Potenciales de la Membrana/efectos de los fármacos , Canal de Sodio Activado por Voltaje NAV1.8/efectos de los fármacos , Dimensión del Dolor/efectos de los fármacos , Ácidos Picolínicos/administración & dosificación , Ácidos Picolínicos/farmacocinética , Ratas , Tetrodotoxina/antagonistas & inhibidores , Tetrodotoxina/farmacología
15.
J Biomol Screen ; 8(3): 332-9, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12857387

RESUMEN

1-Deoxy-D-xylulose 5-phosphate reductoisomerase (Dxr) is a key enzyme in a biosynthetic pathway for isoprenoids that is unique to eubacteria and plants. Dxr catalyzes the rearrangement and NADPH-dependent reduction of 1-deoxy-D-xylulose 5-phosphate to 2-C-methyl-D-erythritol 4-phosphate. The authors have purified Escherichia coli Dxr and devised a high-throughput screen (HTS) for compounds that bind to this enzyme at a functional site. Evidence is presented that the surrogate ligand directly binds or allosterically affects both the D-1-deoxyxylulose 5-phosphate (DXP) and NADPH binding sites. Compounds that bind at either or both sites that compete for binding with the surrogate ligand register as hits. The time-resolved fluorescence-based assay represents an improvement over the Dxr enzyme assay that relies on relatively insensitive measurements of NADPH oxidation. Screening 32,000 compounds from a diverse historical library, the authors obtained 89 potent inhibitors in the surrogate ligand competition assay. The results presented here suggest that peptide surrogate ligands may be useful in formatting HTS for proteins with difficult biochemical assays or targets of unknown function.


Asunto(s)
Isomerasas Aldosa-Cetosa/antagonistas & inhibidores , Fosfomicina/análogos & derivados , Complejos Multienzimáticos/antagonistas & inhibidores , Oxidorreductasas/antagonistas & inhibidores , Sitios de Unión , Unión Competitiva , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Industria Farmacéutica , Inhibidores Enzimáticos/farmacología , Escherichia coli/enzimología , Europio/química , Fosfomicina/farmacología , Concentración 50 Inhibidora , Ligandos , Modelos Químicos , NADP/química , Biblioteca de Péptidos , Péptidos/química , Espectrometría de Fluorescencia , Estreptavidina/química , Factores de Tiempo
16.
ACS Med Chem Lett ; 2(6): 481-4, 2011 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-24900334

RESUMEN

A series of N-pyridyl benzamide KCNQ2/Q3 potassium channel openers were identified and found to be active in animal models of epilepsy and pain. The best compound 12 [ICA-027243, N-(6-chloro-pyridin-3-yl)-3,4-difluoro-benzamide] has an EC50 of 0.38 µM and is selective for KCNQ2/Q3 channels. This compound was active in several rodent models of epilepsy and pain but upon repeated dosing had a number of unacceptable toxicities that prevented further development. On the basis of the structure-activity relationships developed around 12, a second compound, 51, [N-(2-chloro-pyrimidin-5-yl)-3,4-difluoro-benzamide, ICA-069673], was prepared and advanced into a phase 1 clinical study. Herein, we describe the structure-activity relationships that led to the identification of compound 12 and to the corresponding pyrimidine 51.

17.
Comb Chem High Throughput Screen ; 12(1): 107-22, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19149496

RESUMEN

Voltage dependent sodium channels are widely recognized as valuable targets for the development of therapeutic interventions for neuroexcitatory disorders such as epilepsy and pain as well as cardiac arrhythmias. An ongoing challenge for sodium channel drug discovery is the ability to readily evaluate state dependent interactions, which are known to underlie inhibition by many clinically used local anesthetic, antiepileptic and antiarrhythmic sodium channel blockers. While patch-clamp electrophysiology is still considered the most effective way of measuring ion channel function and pharmacology, it does not have the throughput to be useful in early stages of drug discovery in which there is often a need to evaluate many thousands to hundreds of thousands of compounds. Fortunately over the past five years, there has been significant progress in developing much higher throughput electrophysiology platforms like the PatchXpress and IonWorks, which are now widely used in drug discovery. This review highlights the strengths and weaknesses of these two high throughput devices for use in sodium channel inhibitor drug discovery programs. Overall, the PatchXpress and IonWorks electrophysiology platforms have individual strengths that make them complementary to each other. Both platforms are capable of measuring state dependent modulation of sodium channels. IonWorks has the throughput to allow for effective screening of libraries of tens of thousands of compounds whereas the PatchXpress has more flexibility to provide quantitative voltage clamp, which is useful in structure activity evaluations for the hit-to-lead and lead optimization stages of sodium channel drug discovery.


Asunto(s)
Descubrimiento de Drogas/métodos , Electrofisiología/instrumentación , Bloqueadores de los Canales de Sodio/farmacología , Automatización , Electrofisiología/métodos , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA