Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 120(42): e2220029120, 2023 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-37812700

RESUMO

Voltage-gated potassium channels (Kv) are tetrameric membrane proteins that provide a highly selective pathway for potassium ions (K+) to diffuse across a hydrophobic cell membrane. These unique voltage-gated cation channels detect changes in membrane potential and, upon activation, help to return the depolarized cell to a resting state during the repolarization stage of each action potential. The Kv3 family of potassium channels is characterized by a high activation potential and rapid kinetics, which play a crucial role for the fast-spiking neuronal phenotype. Mutations in the Kv3.1 channel have been shown to have implications in various neurological diseases like epilepsy and Alzheimer's disease. Moreover, disruptions in neuronal circuitry involving Kv3.1 have been correlated with negative symptoms of schizophrenia. Here, we report the discovery of a novel positive modulator of Kv3.1, investigate its biophysical properties, and determine the cryo-EM structure of the compound in complex with Kv3.1. Structural analysis reveals the molecular determinants of positive modulation in Kv3.1 channels by this class of compounds and provides additional opportunities for rational drug design for the treatment of associated neurological disorders.


Assuntos
Neurônios , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Humanos , Neurônios/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Canais de Potássio/metabolismo , Potenciais de Ação/fisiologia , Proteínas de Membrana/metabolismo
2.
Hypertension ; 70(2): 347-356, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28630209

RESUMO

Ca2+ drives aldosterone synthesis in the cytosolic and mitochondrial compartments of the adrenal zona glomerulosa cell. Membrane potential across each of these compartments regulates the amplitude of the Ca2+ signal; yet, only plasma membrane ion channels and their role in regulating cell membrane potential have garnered investigative attention as pathological causes of human hyperaldosteronism. Previously, we reported that genetic deletion of TASK-3 channels (tandem pore domain acid-sensitive K+ channels) from mice produces aldosterone excess in the absence of a change in the cell membrane potential of zona glomerulosa cells. Here, we report using yeast 2-hybrid, immunoprecipitation, and electron microscopic analyses that TASK-3 channels are resident in mitochondria, where they regulate mitochondrial morphology, mitochondrial membrane potential, and aldosterone production. This study provides proof of principle that mitochondrial K+ channels, by modulating inner mitochondrial membrane morphology and mitochondrial membrane potential, have the ability to play a pathological role in aldosterone dysregulation in steroidogenic cells.


Assuntos
Aldosterona/biossíntese , Hiperaldosteronismo/metabolismo , Mitocôndrias/fisiologia , Canais de Potássio de Domínios Poros em Tandem/fisiologia , Zona Glomerulosa/fisiologia , Células Cultivadas , Humanos , Potencial da Membrana Mitocondrial/fisiologia
3.
ChemMedChem ; 7(1): 123-33, 2012 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-21916012

RESUMO

TWIK-related acid-sensitive K(+) (K(2P) 9.1, TASK-3) ion channels have the capacity to regulate the activity of neuronal pathways by influencing the resting membrane potential of neurons on which they are expressed. The central nervous system (CNS) expression of these channels suggests potential roles in neurologic disorders, and it is believed that the development of TASK-3 antagonists could lead to the therapeutic treatment of a number of neurological conditions. While a therapeutic potential for TASK-3 channel modulation exists, there are only a few documented examples of potent and selective small-molecule channel blockers. Herein, we describe the discovery and lead optimization efforts for a novel series of TASK-3 channel antagonists based on a 5,6,7,8-tetrahydropyrido[4,3-d]pyrimidine high-throughput screening lead from which a subseries of potent and selective inhibitors were identified. One compound was profiled in detail with respect to its physical properties and demonstrated pharmacological target engagement as indicated by its ability to modulate sleep architecture in rodent electroencephalogram (EEG) telemetry models.


Assuntos
Bloqueadores dos Canais de Potássio/química , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio de Domínios Poros em Tandem/antagonistas & inibidores , Pirimidinas/química , Pirimidinas/farmacologia , Animais , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Ratos Sprague-Dawley , Sono/efeitos dos fármacos , Relação Estrutura-Atividade
4.
Brain Res ; 1416: 69-79, 2011 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-21885038

RESUMO

Modulation of TASK-3 (Kcnk9) potassium channels affect neurotransmitter release in thalamocortical centers and other sleep-related nuclei having the capacity to regulate arousal cycles and REM sleep changes associated with mood disorders and antidepressant action. Circumstantial evidence from this and previous studies suggest the potential for TASK-3 to be a novel antidepressant therapeutic target; TASK-3 knock-out mice display augmented circadian amplitude and exhibit sleep architecture characterized by suppressed REM activity. Detailed analysis of locomotor activity indicates that the amplitudes of activity bout duration and bout number are augmented in TASK-3 mutants well beyond that seen in wildtypes, findings substantiated by amplitude increases in body temperature and EEG recordings of sleep stage bouts. Polysomnographic analysis of TASK-3 mutants reveals increases in nocturnal active wake and suppressed REM sleep time while increased slow wave sleep typifies the inactive phase, findings that have implications for the cognitive impact of reduced TASK-3 activity. In direct measures of their resistance to despair behavior, TASK-3 knock-outs displayed significant decreases in immobility relative to wildtype controls in both tail suspension and forced swim tests. Treatment of wildtype animals with the antidepressant Fluoxetine markedly reduced REM sleep, while leaving active wake and slow wave sleep relatively intact. Remarkably, these effects were absent in TASK-3 mutants indicating that TASK-3 is either directly involved in the mechanism of this drug's action, or participates in parallel pathways that achieve the same effect. Together, these results support the TASK-3 channel to act as a therapeutic target for antidepressant action.


Assuntos
Antidepressivos de Segunda Geração/farmacologia , Nível de Alerta/fisiologia , Ritmo Circadiano/fisiologia , Fluoxetina/farmacologia , Canais de Potássio/metabolismo , Sono REM/fisiologia , Animais , Comportamento Animal/fisiologia , Depressão/tratamento farmacológico , Depressão/metabolismo , Comportamento Exploratório/efeitos dos fármacos , Comportamento Exploratório/fisiologia , Masculino , Análise por Pareamento , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes Neurológicos , Fenótipo , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/genética
5.
Cell Biochem Biophys ; 55(2): 81-93, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19582593

RESUMO

Low-voltage-activated (T-type) calcium channels play a role in diverse physiological responses including neuronal burst firing, hormone secretion, and cell growth. To better understand the biological role and therapeutic potential of the target, a number of structurally diverse antagonists have been identified. Multiple drug interaction sites have been identified for L-type calcium channels, suggesting a similar possibility exists for the structurally related T-type channels. Here, we radiolabel a novel amide T-type calcium channel antagonist (TTA-A1) and show that several known antagonists, including mibefradil, flunarizine, and pimozide, displace binding in a concentration-dependent manner. Further, we identify a novel quinazolinone T-type antagonist (TTA-Q4) that enhanced amide radioligand binding, increased affinity in a saturable manner and slowed dissociation. Functional evaluation showed these compounds to be state-dependent antagonists which show a positive allosteric interaction. Consistent with slowing dissociation, the duration of efficacy was prolonged when compounds were co-administered to WAG/Rij rats, a genetic model of absence epilepsy. The development of a T-type calcium channel radioligand has been used to demonstrate structurally distinct TTAs interact at allosteric sites and to confirm the potential for synergistic inhibition of T-type calcium channels with structurally diverse antagonists.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo T/metabolismo , Regulação Alostérica/efeitos dos fármacos , Sítio Alostérico/efeitos dos fármacos , Animais , Bloqueadores dos Canais de Cálcio/química , Células Cultivadas , Humanos , Masculino , Estrutura Molecular , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Estereoisomerismo , Relação Estrutura-Atividade
6.
Neuroreport ; 20(3): 257-62, 2009 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-19212242

RESUMO

T-type voltage-dependent calcium channels may play an important role in synaptic plasticity, but lack of specific antagonists has hampered investigation into this possible function. We investigated the role of the T-type channel in a canonical model of in-vivo cortical plasticity triggered by monocular deprivation. We identified a compound (TTA-I1) with subnanomolar potency in standard voltage clamp assays and high selectivity for the T-type channel. When infused intracortically, TTA-I1 reduced cortical plasticity triggered by monocular deprivation while preserving normal visual response properties. These results show that the T-type calcium channel plays a central role in cortical plasticity.


Assuntos
Canais de Cálcio Tipo T/metabolismo , Dominância Ocular/fisiologia , Plasticidade Neuronal/fisiologia , Visão Monocular/fisiologia , Córtex Visual/metabolismo , Percepção Visual/fisiologia , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo T/efeitos dos fármacos , Gatos , Linhagem Celular , Dominância Ocular/efeitos dos fármacos , Humanos , Indóis/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Plasticidade Neuronal/efeitos dos fármacos , Técnicas de Patch-Clamp , Privação Sensorial/fisiologia , Triazóis/farmacologia , Córtex Visual/efeitos dos fármacos , Vias Visuais/efeitos dos fármacos , Vias Visuais/metabolismo , Percepção Visual/efeitos dos fármacos
7.
Biophys J ; 93(7): 2341-9, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17545248

RESUMO

Extracellular Ca(2+) ions cause a rapid block of voltage-gated sodium channels, manifest as an apparent reduction of the amplitude of single-channel currents. We examined the influence of residue Tyr-401 in the isoform rNa(V)1.4 on both single-channel conductance and Ca(2+) block. An aromatic residue at this position in the outer mouth of the pore plays a critical role in high-affinity block by the guanidinium toxin tetrodotoxin, primarily due to an electrostatic attraction between the cationic blocker and the system of pi electrons on the aromatic face. We tested whether a similar attraction between small metal cations (Na(+) and Ca(2+)) and this residue would enhance single-channel conductance or pore block, using a series of fluorinated derivatives of phenylalanine at this position. Our results show a monotonic decrease in Ca(2+) block as the aromatic ring is increasingly fluorinated, a result in accord with a cation-pi interaction between Ca(2+) and the aromatic ring. This occurred without a change of single-channel conductance, consistent with a greater electrostatic effect of the pi system on divalent than on monovalent cations. High-level quantum mechanical calculations show that Ca(2+) ions likely do not bind directly to the aromatic ring because of the substantial energetic penalty of dehydrating a Ca(2+) ion. However, the complex of a Ca(2+) ion with its inner hydration shell, Ca(2+)(H(2)O)(6), interacts electrostatically with the aromatic ring in a way that affects the local concentration of Ca(2+) ions in the extracellular vestibule.


Assuntos
Biofísica/métodos , Cálcio/química , Bloqueadores dos Canais de Sódio/química , Animais , Cátions , Eletrofisiologia/métodos , Flúor/química , Guanidina/química , Humanos , Íons , Conformação Molecular , Proteínas Musculares/química , Isoformas de Proteínas , Canais de Sódio/química , Eletricidade Estática , Tetrodotoxina/química
8.
J Biol Chem ; 282(11): 8044-51, 2007 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-17237232

RESUMO

Voltage-gated sodium channels control the upstroke of the action potential in excitable cells of nerve and muscle tissue, making them ideal targets for exogenous toxins that aim to squelch electrical excitability. One such toxin, tetrodotoxin (TTX), blocks sodium channels with nanomolar affinity only when an aromatic Phe or Tyr residue is present at a specific location in the external vestibule of the ion-conducting pore. To test whether TTX is attracted to Tyr401 of NaV1.4 through a cation-pi interaction, this aromatic residue was replaced with fluorinated derivatives of Phe using in vivo nonsense suppression. Consistent with a cation-pi interaction, increased fluorination of Phe401, which reduces the negative electrostatic potential on the aromatic face, caused a monotonic increase in the inhibitory constant for block. Trifluorination of the aromatic ring decreased TTX affinity by approximately 50-fold, a reduction similar to that caused by replacement with the comparably hydrophobic residue Leu. Furthermore, we show that an energetically equivalent cation-pi interaction underlies both use-dependent and tonic block by TTX. Our results are supported by high level ab initio quantum mechanical calculations applied to a model of TTX binding to benzene. Our analysis suggests that the aromatic side chain faces the permeation pathway where it orients TTX optimally and interacts with permeant ions. These results are the first of their kind to show the incorporation of unnatural amino acids into a voltage-gated sodium channel and demonstrate that a cation-pi interaction is responsible for the obligate nature of an aromatic at this position in TTX-sensitive sodium channels.


Assuntos
Cátions , Proteínas Musculares/química , Canais de Sódio/química , Tetrodotoxina/farmacologia , Anestésicos Locais/farmacologia , Animais , Relação Dose-Resposta a Droga , Eletrofisiologia , Flúor/química , Conformação Molecular , Oócitos/metabolismo , Fenilalanina/química , Ligação Proteica , Ratos , Tetrahymena thermophila , Tirosina/química , Xenopus
9.
Assay Drug Dev Technol ; 1(5): 637-45, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15090236

RESUMO

Establishment of stable cell lines that constitutively express Ca(2+) channels at high density and that are useful for in vitro studies may be complicated by problems with seal quality and duration during whole-cell patch-clamp electrophysiology. The current studies describe the generation and characterization of cells that express the human alpha1H T-type Ca(2+) channel under the control of a tetracycline-inducible expression system. Western blot and immunostaining studies revealed that expression of the alpha1H protein occurred only in the presence of tetracycline. Using the whole-cell patch-clamp method, the cells displayed peak inward currents of 1.15 +/- 0.14 nA in response to voltage-clamp steps. The T-type Ca(2+) current was inhibited by the T-type Ca(2+) channel antagonist, mibefradil, with an IC(50) of 160 nM. This cell line, with inducible channel expression, sealed with longer duration during whole-cell patch-clamp recording when compared with a cell line that constitutively expresses the alpha1H Ca(2+) channel. Ca(2+) influx through this channel could also be detected after the addition of extracellular Ca(2+). The amount of Ca(2+) influx was dependent on the [Ca](o) with an EC(50) of 4 mM. The Ca(2+) influx was also inhibited by mibefradil with a potency (IC(50) = 183 nM) similar to that observed in the voltage-clamp studies. Overall, this inducible alpha1H Ca(2+) channel-expressing cell line is useful for the study of human T-type Ca(2+) channel function, and offers advantages over a similar cell line that constitutively expresses the channel.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo T/fisiologia , Cálcio/metabolismo , Rim/fisiologia , Potenciais da Membrana/fisiologia , Engenharia de Proteínas/métodos , Tetraciclina/farmacologia , Canais de Cálcio Tipo T/efeitos dos fármacos , Técnicas de Cultura de Células/métodos , Linhagem Celular , Células Cultivadas , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Rim/efeitos dos fármacos , Rim/embriologia , Potenciais da Membrana/efeitos dos fármacos , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...