Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Mol Pharmacol ; 94(2): 896-906, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29884692

RESUMO

Sodium channels are important proteins in modulating neuronal membrane excitability. Genetic studies from patients and animals have indicated neuronal sodium channels play key roles in pain sensitization. We identified WB4101 (2-(2,6-Dimethoxyphenoxyethyl)aminomethyl-1,4-benzodioxane hydrochloride), an antagonist of α1-adrenoceptor, as a Nav1.7 inhibitor from a screen. The present study characterized the effects of WB4101 on sodium channels. We demonstrated that WB4101 inhibited both Nav1.7 and Nav1.8 channels with similar levels of potency. The half-inhibition concentrations (IC50 values) of WB4101 were 11.6 ± 2.07 and 1.0 ± 0.07 µM for the resting and inactivated Nav1.7 channels, respectively, and 8.67 ± 1.31 and 0.91 ± 0.25 µM for the resting and inactivated Nav1.8 channels, respectively. WB4101 induced a hyperpolarizing shift in the voltage-dependent inactivation for both Nav1.7 (15 mV) and Nav1.8 (20 mV) channels. The IC50 values for the open-state sodium channel were 2.50 ± 1.16 µM for Nav1.7 and 1.1 ± 0.2 µM for Nav1.8, as determined by the block of persistent late currents in inactivation-deficient Nav1.7 and Nav1.8 channels, respectively. Consistent with the state-dependent block, the drug also displayed use-dependent inhibitory properties on both wild-type Nav1.7 and Nav1.8 channels, which were removed by the local anesthetic-insensitive mutations but still existed in the inactivation-deficient channels. Further, the state-dependent inhibition on sodium channels induced by WB4101 was demonstrated in dorsal root ganglion neurons. In conclusion, the present study identified WB4101 as a sodium channel blocker with an open-state-dependent property, which may contribute to WB4101's analgesic action.


Assuntos
Analgésicos/administração & dosagem , Dioxanos/administração & dosagem , Adjuvante de Freund/efeitos adversos , Dor/tratamento farmacológico , Bloqueadores do Canal de Sódio Disparado por Voltagem/administração & dosagem , Analgésicos/farmacologia , Animais , Células Cultivadas , Dioxanos/farmacologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Gânglios Espinais/citologia , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Células HEK293 , Humanos , Camundongos , Mutagênese Sítio-Dirigida , Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.8/metabolismo , Dor/induzido quimicamente , Dor/metabolismo , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia
2.
Neuropharmacology ; 117: 282-291, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28216001

RESUMO

Previous studies demonstrated that Loperamide, originally known as an anti-diarrheal drug, is a promising analgesic agent primarily targeting mu-opioid receptors. However some evidences suggested that non-opioid mechanisms may be contributing to its analgesic effect. In the present study, Loperamide was identified as a Nav1.7 blocker in a pilot screen. In HEK293 cells expressing Nav1.7 sodium channels, Loperamide blocked the resting state of Nav1.7 channels (IC50 = 1.86 ± 0.11 µM) dose-dependently and reversibly. Loperamide produced a 10.4 mV of hyperpolarizing shift for the steady-state inactivation of Nav1.7 channels without apparent effect on the voltage-dependent activation. The drug displayed a mild use- and state-dependent inhibition on Nav1.7 channels, which was removed by the local anesthetic-insensitive construct Nav1.7-F1737A. Inhibition of Nav1.7 at resting state was not altered significantly by the F1737A mutation. Compared to its effects on Nav1.7, Loperamide exhibited higher potency on recombinant Nav1.8 channels in ND7/23 cells (IC50 = 0.60 ± 0.10 µM) and weaker potency on Nav1.9 channels (3.48 ± 0.33 µM). Notably more pronounced inhibition was observed in the native Nav1.8 channels (0.11 ± 0.08 µM) in DRG neurons. Once mu-opioid receptor was antagonized by Naloxone in DRG neurons, potency of Loperamide on Nav1.8 was identical to that of recombinant Nav1.8 channels. The inhibition on Nav channels may be the main mechanism of Loperamide for pain relief beyond mu-opioid receptor. In the meanwhile, the opioid receptor pathway may also influence the blocking effect of Loperamide on sodium channels, implying a cross-talk between sodium channels and opioid receptors in pain processing.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Hiperalgesia/tratamento farmacológico , Inflamação/tratamento farmacológico , Loperamida/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Animais , Células CHO , Cricetulus , Relação Dose-Resposta a Droga , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Células HEK293 , Humanos , Hiperalgesia/etiologia , Hiperalgesia/metabolismo , Inflamação/complicações , Inflamação/metabolismo , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Técnicas de Patch-Clamp , Ratos Sprague-Dawley , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Proteínas Recombinantes/metabolismo , Canais de Sódio/genética , Canais de Sódio/metabolismo
3.
Acta Pharmacol Sin ; 37(1): 111-23, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26725739

RESUMO

AIM: hERG potassium channels display miscellaneous interactions with diverse chemical scaffolds. In this study we assessed the hERG inhibition in a large compound library of diverse chemical entities and provided data for better understanding of the mechanisms underlying promiscuity of hERG inhibition. METHODS: Approximately 300 000 compounds contained in Molecular Library Small Molecular Repository (MLSMR) library were tested. Compound profiling was conducted on hERG-CHO cells using the automated patch-clamp platform-IonWorks Quattro(™). RESULTS: The compound library was tested at 1 and 10 µmol/L. IC50 values were predicted using a modified 4-parameter logistic model. Inhibitor hits were binned into three groups based on their potency: high (IC50<1 µmol/L), intermediate (1 µmol/L< IC50<10 µmol/L), and low (IC50>10 µmol/L) with hit rates of 1.64%, 9.17% and 16.63%, respectively. Six physiochemical properties of each compound were acquired and calculated using ACD software to evaluate the correlation between hERG inhibition and the properties: hERG inhibition was positively correlative to the physiochemical properties ALogP, molecular weight and RTB, and negatively correlative to TPSA. CONCLUSION: Based on a large diverse compound collection, this study provides experimental evidence to understand the promiscuity of hERG inhibition. This study further demonstrates that hERG liability compounds tend to be more hydrophobic, high-molecular, flexible and polarizable.


Assuntos
Canal de Potássio ERG1/antagonistas & inibidores , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Células CHO , Físico-Química , Cricetulus , Células HEK293 , Ensaios de Triagem em Larga Escala , Humanos , Técnicas de Patch-Clamp , Bibliotecas de Moléculas Pequenas/química
4.
Cell ; 161(6): 1252-65, 2015 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-26046436

RESUMO

Small-molecule probes can illuminate biological processes and aid in the assessment of emerging therapeutic targets by perturbing biological systems in a manner distinct from other experimental approaches. Despite the tremendous promise of chemical tools for investigating biology and disease, small-molecule probes were unavailable for most targets and pathways as recently as a decade ago. In 2005, the NIH launched the decade-long Molecular Libraries Program with the intent of innovating in and broadening access to small-molecule science. This Perspective describes how novel small-molecule probes identified through the program are enabling the exploration of biological pathways and therapeutic hypotheses not otherwise testable. These experiences illustrate how small-molecule probes can help bridge the chasm between biological research and the development of medicines but also highlight the need to innovate the science of therapeutic discovery.


Assuntos
Descoberta de Drogas , Bibliotecas de Moléculas Pequenas , Animais , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Ensaios de Triagem em Larga Escala , Humanos , National Institutes of Health (U.S.) , Estados Unidos
5.
Br J Pharmacol ; 172(13): 3370-82, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25752739

RESUMO

BACKGROUND AND PURPOSE: A-type potassium channels (IA) are important proteins for modulating neuronal membrane excitability. The expression and activity of Kv 4.2 channels are critical for neurological functions and pharmacological inhibitors of Kv 4.2 channels may have therapeutic potential for Fragile X syndrome. While screening various compounds, we identified tyrphostin AG879, a tyrosine kinase inhibitor, as a Kv 4.2 inhibitor from. In the present study we characterized the effect of AG879 on cloned Kv 4.2/Kv channel-interacting protein 2 (KChIP2) channels. EXPERIMENTAL APPROACH: To screen the library of pharmacologically active compounds, the thallium flux assay was performed on HEK-293 cells transiently-transfected with Kv 4.2 cDNA using the Maxcyte transfection system. The effects of AG879 were further examined on CHO-K1 cells expressing Kv 4.2/KChIP2 channels using a whole-cell patch-clamp technique. KEY RESULTS: Tyrphostin AG879 selectively and dose-dependently inhibited Kv 4.2 and Kv 4.3 channels. In Kv 4.2/KChIP2 channels, AG879 induced prominent acceleration of the inactivation rate, use-dependent block and slowed the recovery from inactivation. AG879 induced a hyperpolarizing shift in the voltage-dependence of the steady-state inactivation of Kv 4.2 channels without apparent effect on the V1/2 of the voltage-dependent activation. The blocking effect of AG879 was enhanced as channel inactivation increased. Furthermore, AG879 significantly inhibited the A-type potassium currents in the cultured hippocampus neurons. CONCLUSION AND IMPLICATIONS: AG879 was identified as a selective and potent inhibitor the Kv 4.2 channel. AG879 inhibited Kv 4.2 channels by preferentially interacting with the open state and further accelerating their inactivation.


Assuntos
Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio Shal/antagonistas & inibidores , Tirfostinas/farmacologia , Animais , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Células HEK293 , Hipocampo/citologia , Humanos , Neurônios/efeitos dos fármacos , Neurônios/parasitologia , Ratos Sprague-Dawley , Canais de Potássio Shal/fisiologia
6.
PLoS One ; 10(2): e0118324, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25700001

RESUMO

Promiscuous inhibition of the human ether-à-go-go-related gene (hERG) potassium channel by drugs poses a major risk for life threatening arrhythmia and costly drug withdrawals. Current knowledge of this phenomenon is derived from a limited number of known drugs and tool compounds. However, in a diverse, naïve chemical library, it remains unclear which and to what degree chemical motifs or scaffolds might be enriched for hERG inhibition. Here we report electrophysiology measurements of hERG inhibition and computational analyses of >300,000 diverse small molecules. We identify chemical 'communities' with high hERG liability, containing both canonical scaffolds and structurally distinctive molecules. These data enable the development of more effective classifiers to computationally assess hERG risk. The resultant predictive models now accurately classify naïve compound libraries for tendency of hERG inhibition. Together these results provide a more complete reference map of characteristic chemical motifs for hERG liability and advance a systematic approach to rank chemical collections for cardiotoxicity risk.


Assuntos
Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Bloqueadores dos Canais de Potássio/química , Bibliotecas de Moléculas Pequenas/química , Animais , Células CHO , Cardiotoxicidade , Cricetinae , Cricetulus , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/metabolismo , Humanos , Bloqueadores dos Canais de Potássio/efeitos adversos , Bloqueadores dos Canais de Potássio/farmacologia , Ligação Proteica , Bibliotecas de Moléculas Pequenas/efeitos adversos , Bibliotecas de Moléculas Pequenas/farmacologia
7.
Drug Discov Today Technol ; 18: 18-23, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26723888

RESUMO

In vitro pharmacological profiling provides crucial information to eliminate drug candidates with potential toxicity early in drug discovery and reduce failure in later stages. It has become a common practice in industry to test lead compounds against a panel of ion channel targets for selectivity and safety liability at early drug discovery stages. Ion channel profiling technologies include binding assays, flux assays, fluorescent membrane potential assays, automated and conventional electrophysiology. Instead of examining compound effects on individual ion channel targets, automated current clamp, optical electrophysiology, and multi-electrode assays have evolved to investigate the integrated compound effects on cardiac myocytes. This review aims to provide an overview of ion channel profiling for cardiac safety and comparisons of various technologies.


Assuntos
Arritmias Cardíacas/induzido quimicamente , Descoberta de Drogas/métodos , Drogas em Investigação/toxicidade , Canais Iônicos/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Tecnologia Farmacêutica/métodos , Animais , Arritmias Cardíacas/metabolismo , Bioensaio , Células Cultivadas , Humanos , Potenciais da Membrana/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Técnicas de Patch-Clamp , Ligação Proteica
8.
Mol Pharmacol ; 87(2): 207-17, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25422141

RESUMO

Voltage-gated sodium (Nav) channels are essential for membrane excitability and represent therapeutic targets for treating human diseases. Recent reports suggest that these channels, e.g., Nav1.3 and Nav1.5, are inhibited by multiple structurally distinctive small molecule drugs. These studies give reason to wonder whether these drugs collectively target a single site or multiple sites in manifesting such pharmacological promiscuity. We thus investigate the pharmacological profile of Nav1.5 through systemic analysis of its sensitivity to diverse compound collections. Here, we report a dual-color fluorescent method that exploits a customized Nav1.5 [calcium permeable Nav channel, subtype 5 (SoCal5)] with engineered-enhanced calcium permeability. SoCal5 retains wild-type (WT) Nav1.5 pharmacological profiles. WT SoCal5 and SoCal5 with the local anesthetics binding site mutated (F1760A) could be expressed in separate cells, each with a different-colored genetically encoded calcium sensor, which allows a simultaneous report of compound activity and site dependence. The pharmacological profile of SoCal5 reveals a hit rate (>50% inhibition) of around 13% at 10 µM, comparable to that of hERG. The channel activity is susceptible to blockage by known drugs and structurally diverse compounds. The broad inhibition profile is highly dependent on the F1760 residue in the inner cavity, which is a residue conserved among all nine subtypes of Nav channels. Both promiscuity and dependence on F1760 seen in Nav1.5 were replicated in Nav1.4. Our evidence of a broad inhibition profile of Nav channels suggests a need to consider off-target effects on Nav channels. The site-dependent promiscuity forms a foundation to better understand Nav channels and compound interactions.


Assuntos
Cálcio/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Bloqueadores dos Canais de Sódio/farmacologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Sequência de Aminoácidos , Células HEK293 , Humanos , Dados de Sequência Molecular
9.
Bioorg Med Chem Lett ; 24(16): 3968-73, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-25017033

RESUMO

TASK-1 is a two-pore domain potassium channel that is important to modulating cell excitability, most notably in the context of neuronal pathways. In order to leverage TASK-1 for therapeutic benefit, its physiological role needs better characterization; however, designing selective inhibitors that avoid the closely related TASK-3 channel has been challenging. In this study, a series of bis-amide derived compounds were found to demonstrate improved TASK-1 selectivity over TASK-3 compared to reported inhibitors. Optimization of a marginally selective hit led to analog 35 which displays a TASK-1 IC50=16 nM with 62-fold selectivity over TASK-3 in an orthogonal electrophysiology assay.


Assuntos
Amidas/farmacologia , Proteínas do Tecido Nervoso/antagonistas & inibidores , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio de Domínios Poros em Tandem/antagonistas & inibidores , Amidas/síntese química , Amidas/química , Relação Dose-Resposta a Droga , Humanos , Estrutura Molecular , Proteínas do Tecido Nervoso/metabolismo , Bloqueadores dos Canais de Potássio/síntese química , Bloqueadores dos Canais de Potássio/química , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Relação Estrutura-Atividade
10.
Eur J Pharmacol ; 740: 603-10, 2014 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-24972239

RESUMO

Two-pore domain potassium (K2P) channels are responsible for background potassium (K+) current, which is crucial for the maintenance of resting membrane potential. K2P18.1, also called TWIK-related spinal cord K+ channel (TRESK) or KCNK18, is thought to be a major contributor to background K+ currents, particularly in sensory neurons where it is abundantly expressed. Despite its critical role and potential therapeutic implication, pharmacological tools for probing K2P18.1 activity remain unavailable. Here, we report a high-throughput screen against a collection of bioactive compounds that yielded 26 inhibitors and 8 activators of K2P18.1 channel activity with more than 10-fold selectivity over the homologous channel K2P9.1. Among these modulators, the antihistamine loratadine inhibited K2P18.1 activity with IC50 of 0.49±0.23 µM and is considerably more potent than existing K2P18.1 inhibitors. Importantly, the inhibition by loratadine remains equally efficacious upon potentiation of K2P18.1 by calcium signaling. Furthermore, the loratadine effect is dependent on transmembrane residues F145 and F352, providing orthogonal evidence that the inhibition is caused by a direct compound-channel interaction. This study reveals new pharmacological modulators of K2P18.1 activity useful in dissecting native K2P18.1 function.


Assuntos
Moduladores de Transporte de Membrana/farmacologia , Canais de Potássio/fisiologia , Células HEK293 , Humanos , Potenciais da Membrana/efeitos dos fármacos , Mutagênese Sítio-Dirigida , Canais de Potássio/agonistas , Canais de Potássio/genética
11.
Proc Natl Acad Sci U S A ; 110(21): 8732-7, 2013 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-23650380

RESUMO

Voltage-gated KCNQ1 (Kv7.1) potassium channels are expressed abundantly in heart but they are also found in multiple other tissues. Differential coassembly with single transmembrane KCNE beta subunits in different cell types gives rise to a variety of biophysical properties, hence endowing distinct physiological roles for KCNQ1-KCNEx complexes. Mutations in either KCNQ1 or KCNE1 genes result in diseases in brain, heart, and the respiratory system. In addition to complexities arising from existence of five KCNE subunits, KCNE1 to KCNE5, recent studies in heterologous systems suggest unorthodox stoichiometric dynamics in subunit assembly is dependent on KCNE expression levels. The resultant KCNQ1-KCNE channel complexes may have a range of zero to two or even up to four KCNE subunits coassembling per KCNQ1 tetramer. These findings underscore the need to assess the selectivity of small-molecule KCNQ1 modulators on these different assemblies. Here we report a unique small-molecule gating modulator, ML277, that potentiates both homomultimeric KCNQ1 channels and unsaturated heteromultimeric (KCNQ1)4(KCNE1)n (n < 4) channels. Progressive increase of KCNE1 or KCNE3 expression reduces efficacy of ML277 and eventually abolishes ML277-mediated augmentation. In cardiomyocytes, the slowly activating delayed rectifier potassium current, or IKs, is believed to be a heteromultimeric combination of KCNQ1 and KCNE1, but it is not entirely clear whether IKs is mediated by KCNE-saturated KCNQ1 channels or by channels with intermediate stoichiometries. We found ML277 effectively augments IKs current of cultured human cardiomyocytes and shortens action potential duration. These data indicate that unsaturated heteromultimeric (KCNQ1)4(KCNE1)n channels are present as components of IKs and are pharmacologically distinct from KCNE-saturated KCNQ1-KCNE1 channels.


Assuntos
Canal de Potássio KCNQ1/metabolismo , Proteínas Musculares/metabolismo , Miócitos Cardíacos/metabolismo , Piperidinas/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Multimerização Proteica/efeitos dos fármacos , Tiazóis/farmacologia , Compostos de Tosil/farmacologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/genética , Animais , Células CHO , Cricetinae , Cricetulus , Células HEK293 , Humanos , Transporte de Íons/efeitos dos fármacos , Transporte de Íons/genética , Canal de Potássio KCNQ1/genética , Proteínas Musculares/genética , Miócitos Cardíacos/citologia , Potássio/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética
12.
J Biol Chem ; 287(47): 39613-25, 2012 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-23033485

RESUMO

The human Ether-à-go-go-related gene (hERG)-encoded K(+) current, I(Kr) is essential for cardiac repolarization but is also a source of cardiotoxicity because unintended hERG inhibition by diverse pharmaceuticals can cause arrhythmias and sudden cardiac death. We hypothesized that a small molecule that diminishes I(Kr) block by a known hERG antagonist would constitute a first step toward preventing hERG-related arrhythmias and facilitating drug discovery. Using a high-throughput assay, we screened a library of compounds for agents that increase the IC(70) of dofetilide, a well characterized hERG blocker. One compound, VU0405601, with the desired activity was further characterized. In isolated, Langendorff-perfused rabbit hearts, optical mapping revealed that dofetilide-induced arrhythmias were reduced after pretreatment with VU0405601. Patch clamp analysis in stable hERG-HEK cells showed effects on current amplitude, inactivation, and deactivation. VU0405601 increased the IC(50) of dofetilide from 38.7 to 76.3 nM. VU0405601 mitigates the effects of hERG blockers from the extracellular aspect primarily by reducing inactivation, whereas most clinically relevant hERG inhibitors act at an inner pore site. Structure-activity relationships surrounding VU0405601 identified a 3-pyridiyl and a naphthyridine ring system as key structural components important for preventing hERG inhibition by multiple inhibitors. These findings indicate that small molecules can be designed to reduce the sensitivity of hERG to inhibitors.


Assuntos
Arritmias Cardíacas/induzido quimicamente , Arritmias Cardíacas/tratamento farmacológico , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Canais de Potássio Éter-A-Go-Go/metabolismo , Proteínas Musculares/antagonistas & inibidores , Proteínas Musculares/metabolismo , Naftiridinas/química , Naftiridinas/farmacologia , Fenetilaminas/efeitos adversos , Bloqueadores dos Canais de Potássio/efeitos adversos , Piridinas/química , Piridinas/farmacologia , Sulfonamidas/efeitos adversos , Animais , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/patologia , Relação Dose-Resposta a Droga , Descoberta de Drogas , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/genética , Células HEK293 , Humanos , Proteínas Musculares/genética , Miocárdio/metabolismo , Miocárdio/patologia , Fenetilaminas/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Coelhos , Relação Estrutura-Atividade , Sulfonamidas/farmacologia
13.
J Med Chem ; 55(15): 6975-9, 2012 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-22793372

RESUMO

A potent and selective inhibitor of KCNQ2, (S)-5 (ML252, IC(50) = 69 nM), was discovered after a high-throughput screen of the MLPCN library was performed. SAR studies revealed a small structural change (ethyl group to hydrogen) caused a functional shift from antagonist to agonist activity (37, EC(50) = 170 nM), suggesting an interaction at a critical site for controlling gating of KCNQ2 channels.


Assuntos
Encéfalo/metabolismo , Canal de Potássio KCNQ2/antagonistas & inibidores , Fenilbutiratos/síntese química , Bloqueadores dos Canais de Potássio/síntese química , Pirrolidinas/síntese química , Animais , Bases de Dados Factuais , Ensaios de Triagem em Larga Escala , Humanos , Canal de Potássio KCNQ2/química , Microssomos Hepáticos/metabolismo , Permeabilidade , Fenilbutiratos/química , Fenilbutiratos/farmacocinética , Bloqueadores dos Canais de Potássio/química , Bloqueadores dos Canais de Potássio/farmacocinética , Pirrolidinas/química , Pirrolidinas/farmacocinética , Ratos , Bibliotecas de Moléculas Pequenas , Estereoisomerismo , Relação Estrutura-Atividade
14.
Proc Natl Acad Sci U S A ; 109(29): 11866-71, 2012 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-22745159

RESUMO

Long QT syndrome (LQTS) is a genetic disease characterized by a prolonged QT interval in an electrocardiogram (ECG), leading to higher risk of sudden cardiac death. Among the 12 identified genes causal to heritable LQTS, ∼90% of affected individuals harbor mutations in either KCNQ1 or human ether-a-go-go related genes (hERG), which encode two repolarizing potassium currents known as I(Ks) and I(Kr). The ability to quantitatively assess contributions of different current components is therefore important for investigating disease phenotypes and testing effectiveness of pharmacological modulation. Here we report a quantitative analysis by simulating cardiac action potentials of cultured human cardiomyocytes to match the experimental waveforms of both healthy control and LQT syndrome type 1 (LQT1) action potentials. The quantitative evaluation suggests that elevation of I(Kr) by reducing voltage sensitivity of inactivation, not via slowing of deactivation, could more effectively restore normal QT duration if I(Ks) is reduced. Using a unique specific chemical activator for I(Kr) that has a primary effect of causing a right shift of V(1/2) for inactivation, we then examined the duration changes of autonomous action potentials from differentiated human cardiomyocytes. Indeed, this activator causes dose-dependent shortening of the action potential durations and is able to normalize action potentials of cells of patients with LQT1. In contrast, an I(Kr) chemical activator of primary effects in slowing channel deactivation was not effective in modulating action potential durations. Our studies provide both the theoretical basis and experimental support for compensatory normalization of action potential duration by a pharmacological agent.


Assuntos
Potenciais de Ação/fisiologia , Canais de Potássio Éter-A-Go-Go/metabolismo , Ativação do Canal Iônico/fisiologia , Canais de Potássio KCNQ/metabolismo , Síndrome do QT Longo/genética , Modelos Biológicos , Acetamidas/farmacologia , Animais , Células CHO , Células Cultivadas , Simulação por Computador , Cricetinae , Cricetulus , Primers do DNA/genética , Humanos , Canais de Potássio KCNQ/fisiologia , Síndrome do QT Longo/patologia , Miócitos Cardíacos/fisiologia , Técnicas de Patch-Clamp , Piridinas/farmacologia , Reação em Cadeia da Polimerase em Tempo Real , Bloqueadores dos Canais de Sódio/farmacologia
15.
Assay Drug Dev Technol ; 9(6): 580-8, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22149888

RESUMO

The unintended and often promiscous inhibition of the cardiac human Ether-à-go-go related gene (hERG) potassium channel is a common cause for either delay or removal of therapeutic compounds from development and withdrawal of marketed drugs. The clinical manifestion is prolongation of the duration between QRS complex and T-wave measured by surface electrocardiogram (ECG)-hence Long QT Syndrome. There are several useful online resources documenting hERG inhibition by known drugs and bioactives. However, their utilities remain somewhat limited because they are biased toward well-studied compounds and their number of data points tends to be much smaller than many commercial compound libraries. The hERGCentral ( www.hergcentral.org ) is mainly based on experimental data obtained from a primary screen by electrophysiology against more than 300,000 structurally diverse compounds. The system is aimed to display and combine three resources: primary electrophysiological data, literature, as well as online reports and chemical library collections. Currently, hERGCentral has annotated datasets of more than 300,000 compounds including structures and chemophysiological properties of compounds, raw traces, and biophysical properties. The system enables a variety of query formats, including searches for hERG effects according to either chemical structure or properties, and alternatively according to the specific biophysical properties of current changes caused by a compound. Therefore, the hERGCentral, as a unique and evolving resource, will facilitate investigation of chemically induced hERG inhibition and therefore drug development.


Assuntos
Cardiotoxinas/efeitos adversos , Bases de Dados Factuais , Descoberta de Drogas/métodos , Canais de Potássio Éter-A-Go-Go/efeitos adversos , Armazenamento e Recuperação da Informação/métodos , Estatística como Assunto/métodos , Cardiotoxinas/química , Cardiotoxinas/genética , Bases de Dados Factuais/tendências , Descoberta de Drogas/tendências , Canais de Potássio Éter-A-Go-Go/química , Canais de Potássio Éter-A-Go-Go/genética , Cardiopatias/induzido quimicamente , Cardiopatias/genética , Humanos , Armazenamento e Recuperação da Informação/tendências , Estatística como Assunto/tendências
16.
ACS Chem Neurosci ; 2(10): 572-577, 2011 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-22125664

RESUMO

Herein we report the discovery, synthesis and evaluation of a series of N-Aryl-bicyclo[2.2.1]heptane-2-carboxamides as selective KCNQ2 (K(v)7.2) and KCNQ4 (K(v)7.4) channel openers. The best compound, 1 (ML213) has an EC(50) of 230 nM (KCNQ2) and 510 nM (KCNQ4) and is selective for KCNQ2 and KCNQ4 channels versus a large battery of related potassium channels, as well as affording modest brain levels. This represents the first report of unique selectivity profile for KCNQ2 and KCNQ4 over the other channels (KCNQ1/3/5) and as such should prove to be a valuable tool compound for understanding these channels in regulating neuronal activity.

17.
Assay Drug Dev Technol ; 8(6): 743-54, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21158688

RESUMO

Compound effects on cloned human Ether-à-go-go related gene (hERG) potassium channels have been used to assess the potential cardiac safety liabilities of drug development candidate compounds. In addition to radioactive ligand displacement tests, two other common approaches are surrogate ion-based flux assays and electrophysiological recordings. The former has much higher throughput, whereas the latter measures directly the effects on ionic currents. Careful characterization in earlier reports has been performed to compare the relative effectiveness of these approaches for known hERG blockers, which often yielded good overall correlation. However, cases were reported showing significant and reproducible differences in potency and/or sensitivity by the two methods. This raises a question concerning the rationale and criteria on which an assay should be selected for evaluating unknown compounds. To provide a general basis for considering assays to profile large compound libraries for hERG activity, we have conducted parallel flux and electrophysiological analyses of 2,000 diverse compounds, representative of the 300,000 compound collection of NIH Molecular Library Small Molecular Repository (MLSMR). Our results indicate that at the conventional testing concentration 1.0 µM, the overlap between the two assays ranges from 32% to 50% depending on the hit selection criteria. There was a noticeable rate of false negatives by the thallium-based assay relative to electrophysiological recording, which may be greatly reduced under modified comparative conditions. As these statistical results identify a preferred method for cardiac safety profiling of unknown compounds, they suggest an efficient method combining flux and electrophysiological assays to rapidly profile hERG liabilities of large collection of naive compounds.


Assuntos
Descoberta de Drogas , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Bloqueadores dos Canais de Potássio/farmacologia , Bibliotecas de Moléculas Pequenas , Animais , Células CHO , Cricetinae , Cricetulus , Ensaios de Triagem em Larga Escala , Humanos , Técnicas de Patch-Clamp , Tálio/metabolismo
18.
Biochem J ; 414(1): 63-71, 2008 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-18419581

RESUMO

The MAP1 (microtubule-associated protein 1) family is a class of microtubule-binding proteins represented by mammalian MAP1A, MAP1B and the recently identified MAP1S. MAP1A and MAP1B are expressed in the nervous system and thought to mediate interactions of the microtubule-based cytoskeleton in neural development and function. The characteristic structural organization of mammalian MAP1s, which are composed of heavy- and light-chain subunits, requires proteolytic cleavage of a precursor polypeptide encoded by the corresponding map1 gene. MAP1 function in Drosophila appears to be fulfilled by a single gene, futsch. Although the futsch gene product is known to share several important functional properties with mammalian MAP1s, whether it adopts the same basic structural organization has not been addressed. Here, we report the identification of a Drosophila MAP1 light chain, LC(f), produced by proteolytic cleavage of a futsch-encoded precursor polypeptide, and confirm co-localization and co-assembly of the heavy chain and LC(f) cleavage products. Furthermore, the in vivo properties of MAP1 proteins were further defined through precise MS identification of a conserved proteolytic cleavage site within the futsch-encoded MAP1 precursor and demonstration of light-chain diversity represented by multiple LC(f) variants. Taken together, these findings establish conservation of proteolytic processing and structural organization among mammalian and Drosophila MAP1 proteins and are expected to enhance genetic analysis of conserved MAP1 functions within the neuronal cytoskeleton.


Assuntos
Sequência Conservada , Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Proteínas Associadas aos Microtúbulos/química , Fatores de Crescimento Neural/química , Fatores de Crescimento Neural/metabolismo , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Sítios de Ligação/genética , Drosophila , Proteínas de Drosophila/genética , Hidrólise , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Dados de Sequência Molecular , Fatores de Crescimento Neural/genética , Peptídeo Hidrolases/química , Peptídeo Hidrolases/genética , Peptídeo Hidrolases/metabolismo , Peptídeos/química , Peptídeos/genética , Peptídeos/metabolismo , Subunidades Proteicas/genética
19.
J Neurosci ; 24(1): 282-5, 2004 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-14715960

RESUMO

Presynaptic calcium channels play a central role in chemical synaptic transmission by providing the calcium trigger for evoked neurotransmitter release. These voltage-gated calcium channels are composed of a primary structural subunit, alpha1, as well as auxiliary beta and alpha2delta subunits. Our previous genetic, molecular, and functional analysis has shown that the cacophony (cac) gene encodes a primary presynaptic calcium channel alpha1 subunit in Drosophila. Here we report that transgenic expression of a cac-encoded alpha1 subunit fused with enhanced green fluorescent protein efficiently rescues cac lethal mutations and allows in vivo analysis of calcium channel localization at active zones. The results reported here further characterize the primary role of cac-encoded calcium channels in neurotransmitter release. In addition, these studies provide a unique genetic tool for live imaging of functional presynaptic calcium channels in vivo and define a molecular marker for immunolocalization of other presynaptic proteins relative to active zones. These findings are expected to facilitate additional analysis of synaptic development and function in this important model system.


Assuntos
Canais de Cálcio/análise , Proteínas de Drosophila/análise , Drosophila/química , Junção Neuromuscular/química , Terminações Pré-Sinápticas/química , Animais , Animais Geneticamente Modificados , Canais de Cálcio/genética , Drosophila/genética , Proteínas de Drosophila/genética , Proteínas de Fluorescência Verde , Proteínas Luminescentes/análise , Proteínas Luminescentes/genética , Microscopia de Fluorescência , Proteínas Recombinantes de Fusão/análise , Transformação Genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA