Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 264
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
FASEB J ; 34(11): 15015-15028, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32918525

RESUMO

Adult patients with dysfunction in human ether-a-go-go 2 (hERG2) protein, encoded by KCNH6, present with hypoinsulinemia and hyperglycemia. However, the mechanism of KCNH6 action in glucose disorders has not been clearly defined. Previous studies identified that sustained endoplasmic reticulum (ER) stress-mediated apoptosis of pancreatic ß-cells and directly contributed to diabetes. In the present study, we showed that Kcnh6 knockout (KO) mice had impaired glucose tolerance mediated by high ER stress levels, and showed increased apoptosis and elevated intracellular calcium levels in pancreatic ß-cells. In contrast, KCNH6 overexpression in islets isolated from C57BL/6J mice attenuated ER stress induced by thapsigargin or palmitic acid. This effect contributed to better preservation of ß-cells, as reflected in increased ß cell survival and enhanced glucose-stimulated insulin secretion. These results were further corroborated by studies evaluating KCNH6 overexpression in KO islets. Similarly, induction of Kcnh6 in KO mice by lentivirus injection improved glucose tolerance by reducing pancreatic ER stress and apoptosis. Our data provide new insights into how Kcnh6 deficiency causes ER calcium depletion and ß cell dysfunction.


Assuntos
Apoptose , Estresse do Retículo Endoplasmático , Canais de Potássio Éter-A-Go-Go/fisiologia , Células Secretoras de Insulina/citologia , Substâncias Protetoras/farmacologia , Tapsigargina/farmacologia , Animais , Cálcio/metabolismo , Inibidores Enzimáticos/farmacologia , Feminino , Glucose/farmacologia , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ácido Palmítico/farmacologia
2.
Horm Metab Res ; 52(9): 669-675, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32750722

RESUMO

Adult patients with a dysfunctional ether-a-go-go 2 (hERG2) protein, which is encoded by the KCNH6 gene, present with hyperinsulinemia and hyperglycemia. However, the mechanism of KCNH6 in glucose metabolism disorders has not been clearly defined. It has been proposed that sustained endoplasmic reticulum (ER) stress is closely concerned with hepatic insulin resistance and inflammation. Here, we demonstrate that Kcnh6 knockout (KO) mice had impaired glucose tolerance and increased levels of hepatic apoptosis, in addition to displaying an increased insulin resistance that was mediated by high ER stress levels. By contrast, overexpression of KCNH6 in primary hepatocytes led to a decrease in ER stress and apoptosis induced by thapsigargin. Similarly, induction of Kcnh6 by tail vein injection into KO mice improved glucose tolerance by reducing ER stress and apoptosis. Furthermore, we show that KCNH6 alleviated hepatic ER stress, apoptosis, and inflammation via the NFκB-IκB kinase (IKK) pathway both in vitro and in vivo. In summary, our study provides new insights into the causes of ER stress and subsequent induction of primary hepatocytes apoptosis.


Assuntos
Estresse do Retículo Endoplasmático , Canais de Potássio Éter-A-Go-Go/fisiologia , Intolerância à Glucose/patologia , Glucose/metabolismo , Resistência à Insulina , Hepatopatias/patologia , Animais , Apoptose , Intolerância à Glucose/etiologia , Intolerância à Glucose/metabolismo , Hepatopatias/etiologia , Hepatopatias/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
3.
Proc Natl Acad Sci U S A ; 113(35): 9916-21, 2016 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-27516548

RESUMO

The human human ether-à-go-go-related gene (hERG) potassium channel plays a critical role in the repolarization of the cardiac action potential. Changes in hERG channel function underlie long QT syndrome (LQTS) and are associated with cardiac arrhythmias and sudden death. A striking feature of this channel and KCNH channels in general is the presence of an N-terminal Per-Arnt-Sim (PAS) domain. In other proteins, PAS domains bind ligands and modulate effector domains. However, the PAS domains of KCNH channels are orphan receptors. We have uncovered a family of positive modulators of hERG that specifically bind to the PAS domain. We generated two single-chain variable fragments (scFvs) that recognize different epitopes on the PAS domain. Both antibodies increase the rate of deactivation but have different effects on channel activation and inactivation. Importantly, we show that both antibodies, on binding to the PAS domain, increase the total amount of current that permeates the channel during a ventricular action potential and significantly reduce the action potential duration recorded in human cardiomyocytes. Overall, these molecules constitute a previously unidentified class of positive modulators and establish that allosteric modulation of hERG channel function through ligand binding to the PAS domain can be attained.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Canais de Potássio Éter-A-Go-Go/fisiologia , Ativação do Canal Iônico/efeitos dos fármacos , Anticorpos de Cadeia Única/farmacologia , Animais , Sítios de Ligação/genética , Sítios de Ligação/imunologia , Células Cultivadas , Galinhas , Estimulação Elétrica/métodos , Epitopos/genética , Epitopos/imunologia , Canais de Potássio Éter-A-Go-Go/genética , Canais de Potássio Éter-A-Go-Go/imunologia , Células HEK293 , Humanos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Técnicas de Patch-Clamp , Anticorpos de Cadeia Única/imunologia
4.
Drug Dev Res ; 80(6): 846-856, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31301190

RESUMO

Nalbuphine (NAL) is recognized as a mixer with the κ-opioid receptor agonist and the µ-opioid receptor antagonist. However, whether this drug causes any modifications in neuronal ionic currents is unclear. The effects of NAL on ionic currents in mHippoE-14 hippocampal neurons were investigated. In the whole-cell current recordings, NAL suppressed the peak amplitude of voltage-gated Na+ current (INa ) with an IC50 value of 1.9 µM. It shifted the steady-state inactivation curve of peak INa to the hyperpolarized potential, suggesting that there is the voltage dependence of NAL-mediated inhibition of peak INa . In continued presence of NAL, subsequent application of either dynorphin A1-13 (1 µM) or naloxone (30 µM) failed to modify its suppression of peak INa . Tefluthrin (Tef; 10 µM), a pyrethroid known to activate INa , increased peak INa with slowed current inactivation; however, further application of NAL suppressed Tef-mediated suppression of peak INa followed by an additional slowing of current inactivation. In addition, NAL suppressed the amplitude of M-type K+ current [IK(M) ] with an IC50 value of 5.7 µM, while it slightly suppressed erg-mediated and delayed-rectifier K+ currents. In the inside-out current recordings, NAL failed to modify the activity of large-conductance Ca2+ -activated K+ channels. In differentiated NG108-15 neuronal cells, NAL also suppressed the peak INa , and subsequent addition of Tef reversed NAL-induced suppression of INa . Our study highlights the evidence that in addition to modulate opioid receptors, NAL has the propensity to interfere with ionic currents including INa and IK(M) , thereby influencing the functional activities of central neurons.


Assuntos
Analgésicos Opioides/farmacologia , Canais de Potássio de Retificação Tardia/antagonistas & inibidores , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Nalbufina/farmacologia , Neurônios/efeitos dos fármacos , Receptores Opioides kappa/agonistas , Receptores Opioides mu/antagonistas & inibidores , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia , Animais , Linhagem Celular , Canais de Potássio de Retificação Tardia/fisiologia , Canais de Potássio Éter-A-Go-Go/fisiologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/fisiologia , Camundongos , Neurônios/fisiologia
5.
J Physiol ; 596(10): 1813-1828, 2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29573276

RESUMO

KEY POINTS: Ion current kinetics are commonly represented by current-voltage relationships, time constant-voltage relationships and subsequently mathematical models fitted to these. These experiments take substantial time, which means they are rarely performed in the same cell. Rather than traditional square-wave voltage clamps, we fitted a model to the current evoked by a novel sum-of-sinusoids voltage clamp that was only 8 s long. Short protocols that can be performed multiple times within a single cell will offer many new opportunities to measure how ion current kinetics are affected by changing conditions. The new model predicts the current under traditional square-wave protocols well, with better predictions of underlying currents than literature models. The current under a novel physiologically relevant series of action potential clamps is predicted extremely well. The short sinusoidal protocols allow a model to be fully fitted to individual cells, allowing us to examine cell-cell variability in current kinetics for the first time. ABSTRACT: Understanding the roles of ion currents is crucial to predict the action of pharmaceuticals and mutations in different scenarios, and thereby to guide clinical interventions in the heart, brain and other electrophysiological systems. Our ability to predict how ion currents contribute to cellular electrophysiology is in turn critically dependent on our characterisation of ion channel kinetics - the voltage-dependent rates of transition between open, closed and inactivated channel states. We present a new method for rapidly exploring and characterising ion channel kinetics, applying it to the hERG potassium channel as an example, with the aim of generating a quantitatively predictive representation of the ion current. We fitted a mathematical model to currents evoked by a novel 8 second sinusoidal voltage clamp in CHO cells overexpressing hERG1a. The model was then used to predict over 5 minutes of recordings in the same cell in response to further protocols: a series of traditional square step voltage clamps, and also a novel voltage clamp comprising a collection of physiologically relevant action potentials. We demonstrate that we can make predictive cell-specific models that outperform the use of averaged data from a number of different cells, and thereby examine which changes in gating are responsible for cell-cell variability in current kinetics. Our technique allows rapid collection of consistent and high quality data, from single cells, and produces more predictive mathematical ion channel models than traditional approaches.


Assuntos
Potenciais de Ação , Capilares/fisiologia , Canais de Potássio Éter-A-Go-Go/fisiologia , Ativação do Canal Iônico , Modelos Teóricos , Animais , Células CHO , Cricetinae , Cricetulus , Cinética , Técnicas de Patch-Clamp
6.
J Biol Chem ; 292(52): 21548-21557, 2017 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-29089383

RESUMO

Voltage-activated human ether-á-go-go-related gene (hERG) potassium channels are critical for the repolarization of cardiac action potentials and tune-spike frequency adaptation in neurons. Two isoforms of mammalian ERG1 channel subunits, ERG1a and ERG1b, are the principal subunits that conduct the IKr current in the heart and are also broadly expressed in the nervous system. However, there is little direct evidence that ERG1a and ERG1b form heteromeric channels. Here, using electrophysiology, biochemistry, and fluorescence approaches, we systematically tested for direct interactions between hERG1a and hERG1b subunits. We report 1) that hERG1a dominant-negative subunits suppress hERG1b currents (and vice versa), 2) that disulfide bonds form between single cysteine residues experimentally introduced into an extracellular loop of hERG1a and hERG1b subunits and produce hERG1a-hERG1b dimers, and 3) that hERG1a and hERG1b subunits tagged with fluorescent proteins that are FRET pairs exhibit robust energy transfer at the plasma membrane. Thus, multiple lines of evidence indicated a physical interaction between hERG1a and hERG1b, consistent with them forming heteromeric channels. Moreover, co-expression of variable ratios of hERG1a and hERG1b RNA yielded channels with deactivation kinetics that reached a plateau and were different from those of hERG1b channels, consistent with a preference of hERG1b subunits for hERG1a subunits. Cross-linking studies revealed that an equal input of hERG1a and hERG1b yields more hERG1a-hERG1a or hERG1a-hERG1b dimers than hERG1b-hERG1b dimers, also suggesting that hERG1b preferentially interacts with hERG1a. We conclude that hERG1b preferentially forms heteromeric ion channels with hERG1a at the plasma membrane.


Assuntos
Canais de Potássio Éter-A-Go-Go/genética , Canais de Potássio Éter-A-Go-Go/fisiologia , Canal de Potássio ERG1/genética , Canais de Potássio Éter-A-Go-Go/metabolismo , Transferência Ressonante de Energia de Fluorescência/métodos , Coração/fisiologia , Humanos , Ativação do Canal Iônico , Síndrome do QT Longo/metabolismo , Miocárdio/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Isoformas de Proteínas , Subunidades Proteicas
7.
J Physiol ; 595(14): 4695-4723, 2017 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-28516454

RESUMO

KEY POINTS: This study represents a first step toward predicting mechanisms of sex-based arrhythmias that may lead to important developments in risk stratification and may inform future drug design and screening. We undertook simulations to reveal the conditions (i.e. pacing, drugs, sympathetic stimulation) required for triggering and sustaining reentrant arrhythmias. Using the recently solved cryo-EM structure for the Eag-family channel as a template, we revealed potential interactions of oestrogen with the pore loop hERG mutation (G604S). Molecular models suggest that oestrogen and dofetilide blockade can concur simultaneously in the hERG channel pore. ABSTRACT: Female sex is a risk factor for inherited and acquired long-QT associated torsade de pointes (TdP) arrhythmias, and sympathetic discharge is a major factor in triggering TdP in female long-QT syndrome patients. We used a combined experimental and computational approach to predict 'the perfect storm' of hormone concentration, IKr block and sympathetic stimulation that induces arrhythmia in females with inherited and acquired long-QT. More specifically, we developed mathematical models of acquired and inherited long-QT syndrome in male and female ventricular human myocytes by combining effects of a hormone and a hERG blocker, dofetilide, or hERG mutations. These 'male' and 'female' model myocytes and tissues then were used to predict how various sex-based differences underlie arrhythmia risk in the setting of acute sympathetic nervous system discharge. The model predicted increased risk for arrhythmia in females when acute sympathetic nervous system discharge was applied in the settings of both inherited and acquired long-QT syndrome. Females were predicted to have protection from arrhythmia induction when progesterone is high. Males were protected by the presence of testosterone. Structural modelling points towards two plausible and distinct mechanisms of oestrogen action enhancing torsadogenic effects: oestradiol interaction with hERG mutations in the pore loop containing G604 or with common TdP-related blockers in the intra-cavity binding site. Our study presents findings that constitute the first evidence linking structure to function mechanisms underlying female dominance of arousal-induced arrhythmias.


Assuntos
Nível de Alerta/fisiologia , Arritmias Cardíacas/fisiopatologia , Modelos Biológicos , Agonistas Adrenérgicos beta/farmacologia , Animais , Antiarrítmicos/farmacologia , Estradiol/farmacologia , Canais de Potássio Éter-A-Go-Go/fisiologia , Feminino , Cobaias , Isoproterenol/farmacologia , Masculino , Simulação de Acoplamento Molecular , Miócitos Cardíacos/fisiologia , Fenetilaminas/farmacologia , Caracteres Sexuais , Sulfonamidas/farmacologia
8.
J Physiol ; 595(16): 5465-5479, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28087896

RESUMO

KEY POINTS: The principles underlying the evolutionary selection of ion channels for expression in sensory neurons are unclear. Photoreceptor depolarization in the diurnal Drosophila melanogaster is predominantly provided by light-activated transient receptor potential (TRP) channels, whereas repolarization is mediated by sustained voltage-gated K+ channels of the Shab family. In the present study, we show that phototransduction in the nocturnal cockroach Periplaneta americana is predominantly mediated by TRP-like channels, whereas membrane repolarization is based on EAG channels. Although bright light stimulates Shab channels in Drosophila, further restricting depolarization and improving membrane bandwidth, it strongly suppresses EAG conductance in Periplaneta. This light-dependent inhibition (LDI) is caused by calcium and is abolished by chelating intracellular calcium or suppressing eag gene expression. LDI increases membrane resistance, augments gain and reduces the signalling bandwidth. This makes EAG unsuitable for light response conditioning during the day and might have resulted in the evolutionary replacement of EAG by other delayed rectifiers in diurnal insects. ABSTRACT: The principles underlying evolutionary selection of ion channels for expression in sensory neurons are unclear. Among species possessing microvillar photoreceptors, the major ionic conductances have only been identified in Drosophila melanogaster. In Drosophila, depolarization is provided by light-activated transient receptor potential (TRP) channels with a minor contribution from TRP-like (TRPL) channels, whereas repolarization is mediated by sustained voltage-gated K+ (Kv) channels of the Shab family. Bright light stimulates Shab channels, further restricting depolarization and improving membrane bandwidth. In the present study, data obtained using a combination of electrophysiological, pharmacological and molecular knockdown techniques strongly suggest that in photoreceptors of the nocturnal cockroach Periplaneta americana the major excitatory channel is TRPL, whereas the predominant delayed rectifier is EAG, a ubiquitous but enigmatic Kv channel. By contrast to the diurnal Drosophila, bright light strongly suppresses EAG conductance in Periplaneta. This light-dependent inhibition (LDI) is caused by calcium entering the cytosol and is amplified following inhibition of calcium extrusion, and it can also be abolished by chelating intracellular calcium or suppressing eag gene expression by RNA interference. LDI increases membrane resistance, augments gain and reduces the signalling bandwidth, impairing information transfer. LDI is also observed in the nocturnal cricket Gryllus integer, whereas, in the diurnal water strider Gerris lacustris, the delayed rectifier is up-regulated by light. Although LDI is not expected to reduce delayed rectifier current in the normal illumination environment of nocturnal cockroaches and crickets, it makes EAG unsuitable for light response conditioning during the day, and might have resulted in the evolutionary replacement of EAG by other delayed rectifiers in diurnal insects.


Assuntos
Canais de Potássio Éter-A-Go-Go/fisiologia , Células Fotorreceptoras de Invertebrados/fisiologia , Visão Ocular/fisiologia , Animais , Ritmo Circadiano , Canais de Potássio Éter-A-Go-Go/genética , Gryllidae/fisiologia , Heterópteros/fisiologia , Canais de Potássio KCNQ/genética , Canais de Potássio KCNQ/fisiologia , Luz , Masculino , Microvilosidades , Periplaneta/fisiologia , Células Fotorreceptoras de Invertebrados/ultraestrutura , Interferência de RNA , RNA Mensageiro/metabolismo , Canais de Potencial de Receptor Transitório/genética , Canais de Potencial de Receptor Transitório/fisiologia
9.
Proc Natl Acad Sci U S A ; 110(28): 11648-53, 2013 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-23801759

RESUMO

The human ether-à-go-go-related gene (hERG) encodes a K(+) channel crucial for repolarization of the cardiac action potential. EAG-related gene (ERG) channels contain a C-terminal cyclic nucleotide-binding homology domain coupled to the pore of the channel by a C-linker. Here, we report the structure of the C-linker/cyclic nucleotide-binding homology domain of a mosquito ERG channel at 2.5-Å resolution. The structure reveals that the region expected to form the cyclic nucleotide-binding pocket is negatively charged and is occupied by a short ß-strand, referred to as the intrinsic ligand, explaining the lack of direct regulation of ERG channels by cyclic nucleotides. In hERG channels, the intrinsic ligand harbors hereditary mutations associated with long-QT syndrome (LQTS), a potentially lethal cardiac arrhythmia. Mutations in the intrinsic ligand affected hERG channel gating and LQTS mutations abolished hERG currents and altered trafficking of hERG channels, which explains the LQT phenotype. The structure also reveals a dramatically different conformation of the C-linker compared with the structures of the related ether-à-go-go-like K(+) and hyperpolarization-activated cyclic nucleotide-modulated channels, suggesting that the C-linker region may be highly dynamic in the KCNH, hyperpolarization-activated cyclic nucleotide-modulated, and cyclic nucleotide-gated channels.


Assuntos
Canais de Potássio Éter-A-Go-Go/química , Canais de Potássio Éter-A-Go-Go/fisiologia , Potenciais de Ação , Animais , Anopheles , Canais de Potássio Éter-A-Go-Go/genética , Modelos Moleculares , Mutação , Conformação Proteica
10.
Proc Natl Acad Sci U S A ; 110(20): 8004-9, 2013 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-23630249

RESUMO

Understanding the mechanism of toxicity of nanomaterials remains a challenge with respect to both mechanisms involved and product regulation. Here we show toxicity of ultrasmall gold nanoparticles (AuNPs). Depending on the ligand chemistry, 1.4-nm-diameter AuNPs failed electrophysiology-based safety testing using human embryonic kidney cell line 293 cells expressing human ether-á-go-go-Related gene (hERG), a Food and Drug Administration-established drug safety test. In patch-clamp experiments, phosphine-stabilized AuNPs irreversibly blocked hERG channels, whereas thiol-stabilized AuNPs of similar size had no effect in vitro, and neither particle blocked the channel in vivo. We conclude that safety regulations may need to be reevaluated and adapted to reflect the fact that the binding modality of surface functional groups becomes a relevant parameter for the design of nanoscale bioactive compounds.


Assuntos
Canais de Potássio Éter-A-Go-Go/fisiologia , Ouro/química , Nanopartículas Metálicas/química , Animais , Canal de Potássio ERG1 , Eletrocardiografia/métodos , Eletrofisiologia/métodos , Canais de Potássio Éter-A-Go-Go/metabolismo , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nanotecnologia/métodos , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/química , Ligação Proteica , Temperatura
11.
Yao Xue Xue Bao ; 51(10): 1530-9, 2016 10.
Artigo em Zh | MEDLINE | ID: mdl-29932317

RESUMO

The potassium channel encoded by the human ether-a-go-go related gene(hERG) plays a very important role in the physiological and pathological processes in human. hERG potassium channel determines the outward currents which facilitate the repolarization of the myocardial cells. Some drugs were withdrawn from the market for the serious side effect of long QT interval and arrhythmia due to blockade of hERG channel. The strategies for lead compound optimization are to reduce inhibitory activity of hERG potassium channel and decrease cardiac toxicity. These methods include reduction of lipophilicity and basicity of amines, introduction of hydroxyl and acidic groups, and restricting conformation.


Assuntos
Arritmias Cardíacas/induzido quimicamente , Cardiotoxicidade/prevenção & controle , Canais de Potássio Éter-A-Go-Go/fisiologia , Síndrome do QT Longo/induzido quimicamente , Miócitos Cardíacos/fisiologia , Bloqueadores dos Canais de Potássio/efeitos adversos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Humanos
12.
J Physiol ; 593(12): 2575-85, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-25820318

RESUMO

Potassium ion channels encoded by the human ether-a-go-go related gene (hERG) form the ion-conducting subunit of the rapid delayed rectifier potassium current (IKr ). Although hERG channels exhibit a widespread tissue distribution they play a particularly important role in the heart. There has been considerable interest in hERG K(+) channels for three main reasons. First, they have very unusual gating kinetics, most notably rapid and voltage-dependent inactivation coupled to slow deactivation, which has led to the suggestion that they may play a specific role in the suppression of arrhythmias. Second, mutations in hERG are the cause of 30-40% of cases of congenital long QT syndrome (LQTS), the commonest inherited primary arrhythmia syndrome. Third, hERG is the molecular target for the vast majority of drugs that cause drug-induced LQTS, the commonest cause of drug-induced arrhythmias and cardiac death. Drug-induced LQTS has now been reported for a large range of both cardiac and non-cardiac drugs, in which this side effect is entirely undesired. In recent years there have been comprehensive reviews published on hERG K(+) channels (Vandenberg et al. 2012) and we will not re-cover this ground. Rather, we focus on more recent work on the structural basis and dynamics of hERG gating with an emphasis on how the latest developments may facilitate translational research in the area of stratifying risk of arrhythmias.


Assuntos
Canais de Potássio Éter-A-Go-Go/fisiologia , Coração/fisiologia , Humanos , Ativação do Canal Iônico
13.
J Physiol ; 593(1): 181-96, 2015 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-25556795

RESUMO

KEY POINTS: Voltage-gated KV 10.1 potassium channels are widely expressed in the mammalian brain but their function remains poorly understood. We report that KV 10.1 is enriched in the presynaptic terminals and does not take part in somatic action potentials. In parallel fibre synapses in the cerebellar cortex, we find that KV 10.1 regulates Ca(2+) influx and neurotransmitter release during repetitive high-frequency activity. Our results describe the physiological role of mammalian KV 10.1 for the first time and help understand the fine-tuning of synaptic transmission. The voltage-gated potassium channel KV 10.1 (Eag1) is widely expressed in the mammalian brain, but its physiological function is not yet understood. Previous studies revealed highest expression levels in hippocampus and cerebellum and suggested a synaptic localization of the channel. The distinct activation kinetics of KV 10.1 indicate a role during repetitive activity of the cell. Here, we confirm the synaptic localization of KV 10.1 both biochemically and functionally and that the channel is sufficiently fast at physiological temperature to take part in repolarization of the action potential (AP). We studied the role of the channel in cerebellar physiology using patch clamp and two-photon Ca(2+) imaging in KV 10.1-deficient and wild-type mice. The excitability and action potential waveform recorded at granule cell somata was unchanged, while Ca(2+) influx into axonal boutons was enhanced in mutants in response to stimulation with three APs, but not after a single AP. Furthermore, mutants exhibited a frequency-dependent increase in facilitation at the parallel fibre-Purkinje cell synapse at high firing rates. We propose that KV 10.1 acts as a modulator of local AP shape specifically during high-frequency burst firing when other potassium channels suffer cumulative inactivation.


Assuntos
Cálcio/fisiologia , Cerebelo/fisiologia , Canais de Potássio Éter-A-Go-Go/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Células de Purkinje/fisiologia , Potenciais de Ação , Animais , Cerebelo/citologia , Potenciais Pós-Sinápticos Excitadores , Células HEK293 , Humanos , Camundongos Knockout , Ratos Sprague-Dawley , Sinapses/fisiologia
14.
J Pharmacol Sci ; 127(1): 127-34, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25704028

RESUMO

The human ether-a-go-go-related gene (HERG) potassium current (IHERG) has been shown to decrease in amplitude following stimulation with Gq protein-coupled receptors (GqRs), such as α1-adrenergic and M1-muscarinic receptors (α1R and M1R, respectively), at least partly via the reduction of membrane phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2). The present study was designed to investigate the modulation of HERG channels by PI(4,5)P2 and phosphatidylinositol4-phosphate 5-kinase (PI(4)P5-K), a synthetic enzyme of PI(4,5)P2. Whole-cell patch-clamp recordings were used to examine the activity of HERG channels expressed heterologously in Chinese Hamster Ovary cells. The stimulation of α1R with phenylephrine or M1R with acetylcholine decreased the amplitude of IHERG accompanied by a significant acceleration of deactivation kinetics and the effects on IHERG were significantly attenuated in cells expressing PI(4)P5-K. The density of IHERG in cells expressing GqRs alone was significantly increased by the coexpression of PI(4)P5-K without significant differences in the voltage dependence of activation and deactivation kinetics. The kinase-deficient substitution mutant, PI(4)P5-K-K138A did not have these counteracting effects on the change in IHERG by M1R stimulation. These results suggest that the current density of IHERG is closely dependent on the membrane PI(4,5)P2 level, which is regulated by PI(4)P5-K and GqRs and that replenishing PI(4,5)P2 by PI(4)P5-K recovers IHERG.


Assuntos
Canais de Potássio Éter-A-Go-Go/efeitos dos fármacos , Fosfotransferases (Aceptor do Grupo Álcool)/fisiologia , Acetilcolina/farmacologia , Animais , Células CHO , Cricetinae , Cricetulus , Canais de Potássio Éter-A-Go-Go/fisiologia , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/efeitos dos fármacos , Humanos , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Mutação , Fenilefrina/farmacologia , Fosfatidilinositol 4,5-Difosfato/fisiologia , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Transfecção
15.
Scand J Clin Lab Invest ; 75(8): 699-709, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26403377

RESUMO

BACKGROUND: Congenital long QT syndrome (LQTS) is a hereditary cardiac channelopathy characterized by delayed ventricular repolarization, syncope, torsades de pointes and sudden cardiac death. Thirty-three members of five apparently 'unrelated' Danish families carry the KCNH2:c.87C> A; p.F29L founder mutation. METHODS AND RESULTS: Linkage disequilibrium mapping with microsatellites around KCNH2 enabled us to estimate the age of the founder mutation to be approximately 22 generations, corresponding to around 550 years. Neighbouring-Joining analysis disclosed one early and three later nodes. The median QTc time of the carriers was 490 ms (range: 415-589 ms) and no difference was seen between the different branches of the family. The mutation is malignant with a penetrance of 73%. Ten F29L carriers received implantable defibrillators (ICDs) (median age at implant 20 years), and of those four individuals experienced eight appropriate shocks. Patch-clamp analysis in HEK 293 cells, performed at 34°C disclosed a loss-of-function phenotype with fast deactivation, reduced steady-state inactivation current density and a positive voltage shift in inactivation. Western blotting of HEK 293 cells transfected with KCNH2:WT and KCNH2:c.87C> A revealed a reduced fraction of fully glycosylated hERG:p.F29L suggesting that this mutation results in defective trafficking. CONCLUSION: The altered channel gating kinetics in combination with defective trafficking of mutated channels is expected to result in reduced repolarizing current density and, thus, a LQTS phenotype.


Assuntos
Canais de Potássio Éter-A-Go-Go/genética , Síndrome do QT Longo/genética , Dinamarca , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/fisiologia , Feminino , Efeito Fundador , Estudos de Associação Genética , Células HEK293 , Haplótipos , Humanos , Ativação do Canal Iônico , Cinética , Masculino , Potenciais da Membrana , Repetições de Microssatélites , Mutação de Sentido Incorreto , Fenótipo , Polimorfismo de Nucleotídeo Único , Transporte Proteico , Análise de Sequência de DNA
16.
J Neurosci ; 33(2): 761-75, 2013 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-23303953

RESUMO

Caenorhabditis elegans regulates egg laying by alternating between an inactive phase and a serotonin-triggered active phase. We found that the conserved ERG [ether-a-go-go (EAG) related gene] potassium channel UNC-103 enables this two-state behavior by limiting excitability of the egg-laying muscles. Using both high-speed video recording and calcium imaging of egg-laying muscles in behaving animals, we found that the muscles appear to be excited at a particular phase of each locomotor body bend. During the inactive phase, this rhythmic excitation infrequently evokes calcium transients or contraction of the egg-laying muscles. During the serotonin-triggered active phase, however, these muscles are more excitable and each body bend is accompanied by a calcium transient that drives twitching or full contraction of the egg-laying muscles. We found that ERG-null mutants lay eggs too frequently, and that ERG function is necessary and sufficient in the egg-laying muscles to limit egg laying. ERG K(+) channels localize to postsynaptic sites in the egg-laying muscle, and mutants lacking ERG have more frequent calcium transients and contractions of the egg-laying muscles even during the inactive phase. Thus ERG channels set postsynaptic excitability at a threshold so that further adjustments of excitability by serotonin generate two distinct behavioral states.


Assuntos
Comportamento Animal/fisiologia , Caenorhabditis elegans/fisiologia , Canais de Potássio Éter-A-Go-Go/fisiologia , Músculos/inervação , Músculos/fisiologia , Oviposição/fisiologia , Sinapses/fisiologia , Regiões 3' não Traduzidas/genética , Animais , Sinalização do Cálcio/fisiologia , DNA/biossíntese , DNA/genética , Feminino , Microscopia Confocal , Contração Muscular/fisiologia , Domínios PDZ/genética , Reação em Cadeia da Polimerase , Serotonina/fisiologia , Sinapses/ultraestrutura , Transgenes/genética
17.
FASEB J ; 27(8): 3039-53, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23608144

RESUMO

Human ether-á-gogo-related gene (HERG) encodes a potassium channel that is highly susceptible to deleterious mutations resulting in susceptibility to fatal cardiac arrhythmias. Most mutations adversely affect HERG channel assembly and trafficking. Why the channel is so vulnerable to missense mutations is not well understood. Since nothing is known of how mRNA structural elements factor in channel processing, we synthesized a codon-modified HERG cDNA (HERG-CM) where the codons were synonymously changed to reduce GC content, secondary structure, and rare codon usage. HERG-CM produced typical IKr-like currents; however, channel synthesis and processing were markedly different. Translation efficiency was reduced for HERG-CM, as determined by heterologous expression, in vitro translation, and polysomal profiling. Trafficking efficiency to the cell surface was greatly enhanced, as assayed by immunofluorescence, subcellular fractionation, and surface labeling. Chimeras of HERG-NT/CM indicated that trafficking efficiency was largely dependent on 5' sequences, while translation efficiency involved multiple areas. These results suggest that HERG translation and trafficking rates are independently governed by noncoding information in various regions of the mRNA molecule. Noncoding information embedded within the mRNA may play a role in the pathogenesis of hereditary arrhythmia syndromes and could provide an avenue for targeted therapeutics.


Assuntos
Canais de Potássio Éter-A-Go-Go/fisiologia , Ativação do Canal Iônico/fisiologia , Biossíntese de Proteínas , RNA Mensageiro/metabolismo , Composição de Bases/genética , Membrana Celular/metabolismo , Membrana Celular/fisiologia , Códon/genética , DNA Complementar/genética , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/química , Canais de Potássio Éter-A-Go-Go/genética , Células HEK293 , Humanos , Immunoblotting , Ativação do Canal Iônico/genética , Potenciais da Membrana/genética , Potenciais da Membrana/fisiologia , Microscopia de Fluorescência , Mutação , Técnicas de Patch-Clamp , Estrutura Secundária de Proteína , Transporte Proteico/genética , Transporte Proteico/fisiologia , RNA Mensageiro/genética
18.
Exp Physiol ; 99(3): 503-9, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24121285

RESUMO

Ion channels play a key role in defining myometrial contractility. Modulation of ion channel populations is proposed to underpin gestational changes in uterine contractility associated with the transition from uterine quiescence to active labour. Of the myriad ion channels present in the uterus, this article will focus upon potassium channels encoded by the KCNQ genes and ether-à-go-go-related (ERG) genes. Voltage-gated potassium channels encoded by KCNQ and ERG (termed Kv7 and Kv11, respectively) are accepted as major determinants of neuronal excitability and the duration of the cardiac action potential. However, there is now growing appreciation that these ion channels have a major functional impact in vascular and non-vascular smooth muscle. Moreover, Kv7 channels may be potential therapeutic targets for the treatment of preterm labour.


Assuntos
Canais de Potássio Éter-A-Go-Go/fisiologia , Canais de Potássio KCNQ/fisiologia , Miométrio/fisiologia , Útero/fisiologia , Adulto , Feminino , Humanos , Músculo Liso/fisiologia , Vias Neurais/fisiologia , Canais de Potássio/fisiologia , Gravidez
19.
Biol Pharm Bull ; 37(11): 1826-30, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25366487

RESUMO

The effects of terfenadine and pentamidine on the human ether-a-go-go related gene (hERG) channel current and its intracellular trafficking were evaluated. Green fluorescent protein (GFP)-linked hERG channels were expressed in HEK293 cells, and the membrane current was measured by an automated whole cell voltage clamp system. To evaluate drug effects on channel trafficking to the cell membrane, the fraction of channel present on the cell membrane was quantified by current measurement after drug washout and confocal microscopy. Terfenadine directly blocked the hERG channel current but had no effect on trafficking of hERG channels to the cell membrane after application in culture medium for 2 d. In contrast, pentamidine had no direct effect on the hERG channel current but reduced trafficking of hERG channels. The two drugs inhibited hERG channel function through different mechanisms: terfenadine through direct channel blockade and pentamidine through inhibition of channel trafficking to the cell membrane. Combined use of automated voltage clamp and confocal microscopic analyses would provide insights into the mechanisms of drug-induced QT-prolongation and arrhythmogenesis.


Assuntos
Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Pentamidina/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Terfenadina/farmacologia , Canais de Potássio Éter-A-Go-Go/fisiologia , Células HEK293 , Humanos
20.
Biol Pharm Bull ; 37(9): 1495-504, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25177033

RESUMO

The effects of paroxetine, a selective serotonin reuptake inhibitor, on human ether-a-go-go-related gene (HERG) channels were investigated using the whole-cell patch-clamp technique. The HERG channels were stably expressed in human embryonic kidney cells. Paroxetine inhibited the peak tail currents of the HERG channel in a concentration-dependent manner, with an IC50 value of 0.45 µM and a Hill coefficient of 0.85. These effects were reversible after wash-out of the drug. The paroxetine-induced inhibition of the HERG channels was voltage-dependent. There was a steep increase in inhibition over the voltage range of the channel opening. Also, a shallow voltage-dependent inhibition was detected over the voltage range in which the channels were fully activated. The fractional electrical distance was estimated to be 0.11. Paroxetine induced a leftward shift in the voltage-dependence of the steady-state activation of the HERG channels. Before and after application of the 1 µM paroxetine, the half-maximum activation was -14.21 mV and -27.04 mV, respectively, with no shift in the slope value. The HERG channel block was not use-dependent. The characteristics of the block were dependent on open and inactivated channel states rather than closed state. Paroxetine had no effect on activation and deactivation kinetics, steady-state inactivation. These results suggest that paroxetine blocks the HERG channels by binding to these channels in the open and inactivated states.


Assuntos
Antidepressivos/farmacologia , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Paroxetina/farmacologia , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Canais de Potássio Éter-A-Go-Go/química , Canais de Potássio Éter-A-Go-Go/fisiologia , Células HEK293 , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Modelos Moleculares
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA