Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 112
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Neurophysiol ; 126(1): 1-10, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-34038189

RESUMO

The exercise pressor reflex (EPR) originates in skeletal muscle and is activated by exercise-induced signals to increase arterial blood pressure and cardiac output. Muscle ischemia can elicit the EPR, which can be inappropriately activated in patients with peripheral vascular disease or heart failure to increase the incidence of myocardial infarction. We seek to better understand the receptor/channels that control excitability of group III and group IV muscle afferent fibers that give rise to the EPR. Bradykinin (BK) is released within contracting muscle and can evoke the EPR. However, the mechanism is incompletely understood. KV7 channels strongly regulate neuronal excitability and are inhibited by BK. We have identified KV7 currents in muscle afferent neurons by their characteristic activation/deactivation kinetics, enhancement by the KV7 activator retigabine, and block by KV7 specific inhibitor XE991. The blocking of KV7 current by different XE991 concentrations suggests that the KV7 current is generated by both KV7.2/7.3 (high affinity) and KV7.5 (low affinity) channels. The KV7 current was inhibited by 300 nM BK in neurons with diameters consistent with both group III and group IV afferents. The inhibition of KV7 by BK could be a mechanism by which this metabolic mediator generates the EPR. Furthermore, our results suggest that KV7 channel activators such as retigabine, could be used to reduce cardiac stress resulting from the exacerbated EPR in patients with cardiovascular disease.NEW & NOTEWORTHY KV7 channels control neuronal excitability. We show that these channels are expressed in muscle afferents and generate currents that are blocked by XE991 and bradykinin (BK). The XE991 block suggests that KV7 current is generated by KV7.2/3 and KV7.5 channels. The BK inhibition of KV7 channels may explain how BK activates the exercise pressor reflex (EPR). Retigabine can enhance KV7 current, which could help control the inappropriately activated EPR in patients with cardiovascular disease.


Assuntos
Canais de Potássio KCNQ/fisiologia , Contração Muscular/fisiologia , Músculo Esquelético/fisiologia , Condicionamento Físico Animal/fisiologia , Reflexo/fisiologia , Animais , Antracenos/farmacologia , Anticonvulsivantes/farmacologia , Carbamatos/farmacologia , Relação Dose-Resposta a Droga , Canais de Potássio KCNQ/antagonistas & inibidores , Masculino , Contração Muscular/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Fenilenodiaminas/farmacologia , Ratos , Ratos Sprague-Dawley , Reflexo/efeitos dos fármacos
2.
Arterioscler Thromb Vasc Biol ; 38(9): 2091-2102, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30002060

RESUMO

Objective- In renal arteries, inhibitors of G protein ßγ subunits (Gßγ) reduce Kv7 activity and inhibit Kv7-dependent receptor-mediated vasorelaxations. However, the mechanisms underlying receptor-mediated relaxation are artery specific. Consequently, the aim of this study was to ascertain the role of Gßγ in Kv7-dependent vasorelaxations of the rat vasculature. Approach and Results- Isometric tension recording was performed in isolated rat renal, mesenteric, and cerebral arteries to study isoproterenol and calcitonin gene-related peptide relaxations. Kv7.4 was knocked down via morpholino transfection while inhibition of Gßγ was investigated with gallein and M119K. Proximity ligation assay was performed on isolated myocytes to study the association between Kv7.4 and G protein ß subunits or signaling intermediaries. Isoproterenol or calcitonin gene-related peptide-induced relaxations were attenuated by Kv7.4 knockdown in all arteries studied. Inhibition of Gßγ with gallein or M119K had no effect on isoproterenol-mediated relaxations in mesenteric artery but had a marked effect on calcitonin gene-related peptide-induced responses in mesenteric artery and cerebral artery and isoproterenol responses in renal artery. Isoproterenol increased association with Kv7.4 and Rap1a in mesenteric artery which were not sensitive to gallein, whereas in renal artery, isoproterenol increased Kv7.4-AKAP (A-kinase anchoring protein) associations in a gallein-sensitive manner. Conclusions- The Gßγ-Kv7 relationship differs between vessels and is an essential requirement for AKAP, but not Rap-mediated regulation of the channel.


Assuntos
Subunidades beta da Proteína de Ligação ao GTP/fisiologia , Subunidades gama da Proteína de Ligação ao GTP/fisiologia , Canais de Potássio KCNQ/fisiologia , Músculo Liso Vascular/fisiologia , Vasodilatação , Proteínas de Ancoragem à Quinase A/metabolismo , Animais , Peptídeo Relacionado com Gene de Calcitonina/farmacologia , Artérias Cerebrais/efeitos dos fármacos , Artérias Cerebrais/fisiologia , Isoproterenol/farmacologia , Masculino , Artérias Mesentéricas/efeitos dos fármacos , Artérias Mesentéricas/fisiologia , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Ratos Wistar , Artéria Renal/efeitos dos fármacos , Artéria Renal/fisiologia , Vasoconstritores/farmacologia , Vasodilatadores/farmacologia , Xantenos/farmacologia
3.
Microcirculation ; 25(1)2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28976052

RESUMO

Potassium channels are recognized as important regulators of cellular functions in most, if not all cell types. These cellular proteins assemble to form gated pores in the plasma membrane, which serve to regulate the flow of potassium ions (K+ ) from the cytosol to the extracellular space. In VSMCs, the open state of potassium channels enables the efflux of K+ and thereby establishes a negative resting voltage across the plasma membrane that inhibits the opening of VSCCs. Under these conditions, cytosolic Ca2+ concentrations are relatively low and Ca2+ -dependent contraction is inhibited. Recent research has identified Kv7 family potassium channels as important contributors to resting membrane voltage in VSMCs, with much of the research focusing on the effects of drugs that specifically activate or block these channels to produce corresponding effects on VSMC contraction and vascular tone. Increasingly, evidence is emerging that these channels are not just good drug targets-they are also essential intermediates in vascular signal transduction, mediating vasoconstrictor or vasodilator responses to a variety of physiological stimuli. This review will summarize recent research findings that support a crucial function of Kv7 channels in both positive (vasoconstrictive) and negative (vasorelaxant) regulation of microvascular tone.


Assuntos
Canais de Potássio KCNQ/fisiologia , Microcirculação/fisiologia , Músculo Liso Vascular/fisiologia , Animais , Cálcio/metabolismo , Humanos , Transdução de Sinais/fisiologia , Vasoconstrição , Vasodilatação
4.
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
5.
J Physiol ; 595(3): 713-738, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-27506492

RESUMO

KEY POINTS: Neuroinflammation associated with CNS insults leads to neuronal hyperexcitability, which may culminate in epileptiform discharges. Application of the endotoxin lipopolysaccharide (LPS) to brain tissue initiates a neuroinflammatory cascade, providing an experimental model to study the mechanisms of neuroinflammatory neuronal hyperexcitability. Here we show that LPS application to hippocampal slices markedly enhances the excitability of CA1 pyramidal cells by inhibiting a specific potassium current, the M-current, generated by KV 7/M channels, which controls the excitability of almost every neuron in the CNS. The LPS-induced M-current inhibition is triggered by sequential activation of microglia, astrocytes and pyramidal cells, mediated by metabotropic purinergic and glutamatergic transmission, leading to blockade of KV 7/M channels by calcium released from intracellular stores. The identification of the downstream molecular target of neuroinflammation, namely the KV 7/M channel, potentially has far reaching implications for the understanding and treatment of many acute and chronic brain disorders. ABSTRACT: Acute brain insults and many chronic brain diseases manifest an innate inflammatory response. The hallmark of this response is glia activation, which promotes repair of damaged tissue, but also induces structural and functional changes that may lead to an increase in neuronal excitability. We have investigated the mechanisms involved in the modulation of neuronal activity by acute inflammation. Initiating inflammatory responses in hippocampal tissue rapidly led to neuronal depolarization and repetitive firing even in the absence of active synaptic transmission. This action was mediated by a complex metabotropic purinergic and glutamatergic glia-to-neuron signalling cascade, leading to the blockade of neuronal KV 7/M channels by Ca2+ released from internal stores. These channels generate the low voltage-activating, non-inactivating M-type K+ current (M-current) that controls intrinsic neuronal excitability, and its inhibition was the predominant cause of the inflammation-induced hyperexcitability. Our discovery that the ubiquitous KV 7/M channels are the downstream target of the inflammation-induced cascade, has far reaching implications for the understanding and treatment of many acute and chronic brain disorders.


Assuntos
Canais de Potássio KCNQ/fisiologia , Lipopolissacarídeos/farmacologia , Células Piramidais/efeitos dos fármacos , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/fisiologia , Região CA1 Hipocampal/citologia , Cálcio/fisiologia , Masculino , Células Piramidais/fisiologia , Ratos Sprague-Dawley , Receptor de Glutamato Metabotrópico 5/fisiologia , Receptores de Glutamato Metabotrópico/fisiologia , Receptores Purinérgicos P2Y1/fisiologia
6.
J Neurophysiol ; 116(2): 503-21, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27121577

RESUMO

The vestibular nerve is characterized by two broad groups of neurons that differ in the timing of their interspike intervals; some fire at highly regular intervals, whereas others fire at highly irregular intervals. Heterogeneity in ion channel properties has been proposed as shaping these firing patterns (Highstein SM, Politoff AL. Brain Res 150: 182-187, 1978; Smith CE, Goldberg JM. Biol Cybern 54: 41-51, 1986). Kalluri et al. (J Neurophysiol 104: 2034-2051, 2010) proposed that regularity is controlled by the density of low-voltage-activated potassium currents (IKL). To examine the impact of IKL on spike timing regularity, we implemented a single-compartment model with three conductances known to be present in the vestibular ganglion: transient sodium (gNa), low-voltage-activated potassium (gKL), and high-voltage-activated potassium (gKH). Consistent with in vitro observations, removing gKL depolarized resting potential, increased input resistance and membrane time constant, and converted current step-evoked firing patterns from transient (1 spike at current onset) to sustained (many spikes). Modeled neurons were driven with a time-varying synaptic conductance that captured the random arrival times and amplitudes of glutamate-driven synaptic events. In the presence of gKL, spiking occurred only in response to large events with fast onsets. Models without gKL exhibited greater integration by responding to the superposition of rapidly arriving events. Three synaptic conductance were modeled, each with different kinetics to represent a variety of different synaptic processes. In response to all three types of synaptic conductance, models containing gKL produced spike trains with irregular interspike intervals. Only models lacking gKL when driven by rapidly arriving small excitatory postsynaptic currents were capable of generating regular spiking.


Assuntos
Gânglios Sensitivos/citologia , Canais de Potássio KCNQ/fisiologia , Modelos Neurológicos , Neurônios/fisiologia , Animais , Biofísica , Estimulação Elétrica , Potenciais Pós-Sinápticos Excitadores/fisiologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Neurônios/efeitos dos fármacos , Potássio/metabolismo , Ratos , Sódio/metabolismo , Sinapses/fisiologia , Nervo Vestibular/anatomia & histologia , Nervo Vestibular/fisiologia
7.
J Neurophysiol ; 116(5): 2114-2124, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27512022

RESUMO

KCNQ/Kv7 channels form a slow noninactivating K+ current, also known as the M current. They activate in the subthreshold range of membrane potentials and regulate different aspects of excitability in neurons of the central nervous system. In spinal motoneurons (MNs), KCNQ/Kv7 channels have been identified in the somata, axonal initial segment, and nodes of Ranvier, where they generate a slow, noninactivating, K+ current sensitive to both muscarinic receptor-mediated inhibition and KCNQ/Kv7 channel blockers. In this study, we thoroughly reevaluated the function of up- and downregulation of KCNQ/Kv7 channels in mouse immature spinal MNs. Using electrophysiological techniques together with specific pharmacological modulators of the activity of KCNQ/Kv7 channels, we show that enhancement of the activity of these channels decreases the excitability of spinal MNs in mouse neonates. This action on MNs results from a combination of hyperpolarization of the resting membrane potential, a decrease in the input resistance, and depolarization of the voltage threshold. On the other hand, the effect of inhibition of KCNQ/Kv7 channels suggested that these channels play a limited role in regulating basal excitability. Computer simulations confirmed that pharmacological enhancement of KCNQ/Kv7 channel activity decreases excitability and also suggested that the effects of inhibition of KCNQ/Kv7 channels on the excitability of spinal MNs do not depend on a direct effect in these neurons but likely on spinal cord synaptic partners. These results indicate that KCNQ/Kv7 channels have a fundamental role in the modulation of the excitability of spinal MNs acting both in these neurons and in their local presynaptic partners.


Assuntos
Regulação para Baixo/fisiologia , Canais de Potássio KCNQ/fisiologia , Neurônios Motores/fisiologia , Medula Espinal/fisiologia , Regulação para Cima/fisiologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Animais Recém-Nascidos , Regulação para Baixo/efeitos dos fármacos , Agonistas de Aminoácidos Excitatórios/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Canais de Potássio KCNQ/agonistas , Canais de Potássio KCNQ/antagonistas & inibidores , Camundongos , Neurônios Motores/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Medula Espinal/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
8.
Circ Res ; 115(2): 263-72, 2014 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-24838176

RESUMO

RATIONALE: Vascular wall stretch is the major stimulus for the myogenic response of small arteries to pressure. The molecular mechanisms are elusive, but recent findings suggest that G protein-coupled receptors can elicit a stretch response. OBJECTIVE: To determine whether angiotensin II type 1 receptors (AT1R) in vascular smooth muscle cells exert mechanosensitivity and identify the downstream ion channel mediators of myogenic vasoconstriction. METHODS AND RESULTS: We used mice deficient in AT1R signaling molecules and putative ion channel targets, namely AT1R, angiotensinogen, transient receptor potential channel 6 (TRPC6) channels, or several subtypes of the voltage-gated K+ (Kv7) gene family (KCNQ3, 4, or 5). We identified a mechanosensing mechanism in isolated mesenteric arteries and in the renal circulation that relies on coupling of the AT1R subtype a to a Gq/11 protein as a critical event to accomplish the myogenic response. Arterial mechanoactivation occurs after pharmacological block of AT1R and in the absence of angiotensinogen or TRPC6 channels. Activation of AT1R subtype a by osmotically induced membrane stretch suppresses an XE991-sensitive Kv channel current in patch-clamped vascular smooth muscle cells, and similar concentrations of XE991 enhance mesenteric and renal myogenic tone. Although XE991-sensitive KCNQ3, 4, and 5 channels are expressed in vascular smooth muscle cells, XE991-sensitive K+ current and myogenic contractions persist in arteries deficient in these channels. CONCLUSIONS: Our results provide definitive evidence that myogenic responses of mouse mesenteric and renal arteries rely on ligand-independent, mechanoactivation of AT1R subtype a. The AT1R subtype a signal relies on an ion channel distinct from TRPC6 or KCNQ3, 4, or 5 to enact vascular smooth muscle cell activation and elevated vascular resistance.


Assuntos
Artérias Mesentéricas/fisiologia , Miócitos de Músculo Liso/fisiologia , Pressorreceptores/fisiologia , Receptor Tipo 1 de Angiotensina/fisiologia , Artéria Renal/fisiologia , 4-Aminopiridina/farmacologia , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Animais , Antracenos/farmacologia , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/fisiologia , Células HEK293 , Hemorreologia , Humanos , Canais de Potássio KCNQ/fisiologia , Canal de Potássio KCNQ3/fisiologia , Losartan/farmacologia , Artérias Mesentéricas/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pressão Osmótica , Receptor Tipo 1 de Angiotensina/deficiência , Receptor Tipo 1 de Angiotensina/genética , Artéria Renal/citologia , Canais de Cátion TRPC/fisiologia , Canal de Cátion TRPC6 , Transcrição Gênica , Resistência Vascular/efeitos dos fármacos , Resistência Vascular/fisiologia
9.
Acta Pharmacol Sin ; 37(1): 25-33, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26687932

RESUMO

M channels, an important regulator of neural excitability, are composed of four subunits of the Kv7 (KCNQ) K(+) channel family. M channels were named as such because their activity was suppressed by stimulation of muscarinic acetylcholine receptors. These channels are of particular interest because they are activated at the subthreshold membrane potentials. Furthermore, neural KCNQ channels are drug targets for the treatments of epilepsy and a variety of neurological disorders, including chronic and neuropathic pain, deafness, and mental illness. This review will update readers on the roles of KCNQ channels in the sensory system and neural circuits as well as discuss their respective mechanisms and the implications for physiology and medicine. We will also consider future perspectives and the development of additional pharmacological models, such as seizure, stroke, pain and mental illness, which work in combination with drug-design targeting of KCNQ channels. These models will hopefully deepen our understanding of KCNQ channels and provide general therapeutic prospects of related channelopathies.


Assuntos
Canais de Potássio KCNQ/fisiologia , Sensação/fisiologia , Células Receptoras Sensoriais/fisiologia , Animais , Audição/fisiologia , Humanos , Ativação do Canal Iônico , Dor/metabolismo , Dor/fisiopatologia , Transdução de Sinais , Tato/fisiologia , Visão Ocular/fisiologia
10.
J Physiol ; 593(7): 1551-80, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25656084

RESUMO

In rodent hippocampi, the connections, gene expression and functions differ along the dorsoventral (D-V) axis. CA1 pyramidal cells show increasing excitability along the D-V axis, although the underlying mechanism is not known. In the present study, we investigated how the M-current (IM ), caused by Kv7/M (KCNQ) potassium channels, and known to often control neuronal excitability, contributes to D-V differences in intrinsic properties of CA1 pyramidal cells. Using whole-cell patch clamp recordings and the selective Kv7/M blocker 10,10-bis(4-pyridinylmethyl)-9(10H)-anthracenone dihydrochloride (XE991) in hippocampal slices from 3- to 4-week-old rats, we found that: (i) IM had a stronger impact on subthreshold electrical properties in dorsal than ventral CA1 pyramidal cells, including input resistance, temporal summation of artificial synaptic potentials, and M-resonance; (ii) IM activated at more negative potentials (left-shifted) and had larger peak amplitude in the dorsal than ventral CA1; and (iii) the initial spike threshold (during ramp depolarizations) was elevated, and the medium after-hyperpolarization and spike frequency adaptation were increased (i.e. excitability was lower) in the dorsal rather than ventral CA1. These differences were abolished or reduced by application of XE991, indicating that they were caused by IM . Thus, it appears that IM has stronger effects in dorsal than in ventral rat CA1 pyramidal cells because of a larger maximal M-conductance and left-shifted activation curve in the dorsal cells. These mechanisms may contribute to D-V differences in the rate and phase coding of position by CA1 place cells, and may also enhance epileptiform activity in ventral CA1.


Assuntos
Canais de Potássio KCNQ/fisiologia , Células Piramidais/fisiologia , Animais , Antracenos/farmacologia , Região CA1 Hipocampal/citologia , Região CA1 Hipocampal/fisiologia , Técnicas In Vitro , Canais de Potássio KCNQ/antagonistas & inibidores , Masculino , Bloqueadores dos Canais de Potássio/farmacologia , Ratos Wistar
11.
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
12.
Neural Plast ; 2015: 472676, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26113994

RESUMO

Striatal projection neurons (SPNs) process motor and cognitive information. Their activity is affected by Parkinson's disease, in which dopamine concentration is decreased and acetylcholine concentration is increased. Acetylcholine activates muscarinic receptors in SPNs. Its main source is the cholinergic interneuron that responds with a briefer latency than SPNs during a cortical command. Therefore, an important question is whether muscarinic G-protein coupled receptors and their signaling cascades are fast enough to intervene during synaptic responses to regulate synaptic integration and firing. One of the most known voltage dependent channels regulated by muscarinic receptors is the KV7/KCNQ channel. It is not known whether these channels regulate the integration of suprathreshold corticostriatal responses. Here, we study the impact of cholinergic muscarinic modulation on the synaptic response of SPNs by regulating KV7 channels. We found that KV7 channels regulate corticostriatal synaptic integration and that this modulation occurs in the dendritic/spines compartment. In contrast, it is negligible in the somatic compartment. This modulation occurs on sub- and suprathreshold responses and lasts during the whole duration of the responses, hundreds of milliseconds, greatly altering SPNs firing properties. This modulation affected the behavior of the striatal microcircuit.


Assuntos
Potenciais de Ação , Neurônios GABAérgicos/fisiologia , Canais de Potássio KCNQ/fisiologia , Neostriado/fisiologia , Sinapses/fisiologia , Potenciais de Ação/efeitos dos fármacos , Animais , Córtex Cerebral/fisiologia , Neurônios Colinérgicos/fisiologia , Estimulação Elétrica , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Neurônios GABAérgicos/citologia , Neurônios GABAérgicos/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos Transgênicos , Muscarina/farmacologia , Agonistas Muscarínicos/farmacologia , Neostriado/citologia , Neostriado/metabolismo , Peptídeos/farmacologia , Receptor Muscarínico M1/agonistas , Receptor Muscarínico M1/antagonistas & inibidores , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo
13.
Mol Pharmacol ; 86(3): 330-41, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24944189

RESUMO

Recent research suggests that smooth muscle cells express Kv7.4 and Kv7.5 voltage-activated potassium channels, which contribute to maintenance of their resting membrane voltage. New pharmacologic activators of Kv7 channels, ML213 (N-mesitybicyclo[2.2.1]heptane-2-carboxamide) and ICA-069673 N-(6-chloropyridin-3-yl)-3,4-difluorobenzamide), have been reported to discriminate among channels formed from different Kv7 subtypes. We compared the effects of ML213 and ICA-069673 on homomeric human Kv7.4, Kv7.5, and heteromeric Kv7.4/7.5 channels exogenously expressed in A7r5 vascular smooth muscle cells. We found that, despite its previous description as a selective activator of Kv7.2 and Kv7.4, ML213 significantly increased the maximum conductance of homomeric Kv7.4 and Kv7.5, as well as heteromeric Kv7.4/7.5 channels, and induced a negative shift of their activation curves. Current deactivation rates decreased in the presence of the ML213 (10 µM) for all three channel combinations. Mutants of Kv7.4 (W242L) and Kv7.5 (W235L), previously found to be insensitive to another Kv7 channel activator, retigabine, were also insensitive to ML213 (10 µM). In contrast to ML213, ICA-069673 robustly activated Kv7.4 channels but was significantly less effective on homomeric Kv7.5 channels. Heteromeric Kv7.4/7.5 channels displayed intermediate responses to ICA-069673. In each case, ICA-069673 induced a negative shift of the activation curves without significantly increasing maximal conductance. Current deactivation rates decreased in the presence of ICA-069673 in a subunit-specific manner. Kv7.4 W242L responded to ICA-069673-like wild-type Kv7.4, but a Kv7.4 F143A mutant was much less sensitive to ICA-069673. Based on these results, ML213 and ICA-069673 likely bind to different sites and are differentially selective among Kv7.4, Kv7.5, and Kv7.4/7.5 channel subtypes.


Assuntos
Anilidas/farmacologia , Benzamidas/farmacologia , Compostos Bicíclicos com Pontes/farmacologia , Canais de Potássio KCNQ/agonistas , Músculo Liso Vascular/metabolismo , Piridinas/farmacologia , Animais , Células Cultivadas , Humanos , Canais de Potássio KCNQ/genética , Canais de Potássio KCNQ/fisiologia , Músculo Liso Vascular/citologia , Mutação , Técnicas de Patch-Clamp , Ratos
14.
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
15.
J Neurosci ; 32(47): 16943-52, 2012 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-23175845

RESUMO

Chemosensitive neurons in the retrotrapezoid nucleus (RTN) regulate breathing in response to CO(2)/H(+) changes. Their activity is also sensitive to neuromodulatory inputs from multiple respiratory centers, and thus they serve as a key nexus of respiratory control. However, molecular mechanisms that control their activity and susceptibility to neuromodulation are unknown. Here, we show in vitro and in vivo that KCNQ channels are critical determinants of RTN neural activity. In particular, we find that pharmacological block of KCNQ channels (XE991, 10 µm) increased basal activity and CO(2) responsiveness of RTN neurons in rat brain slices, whereas KCNQ channel activation (retigabine, 2-40 µm) silenced these neurons. Interestingly, we also find that KCNQ and apamin-sensitive SK channels act synergistically to regulate firing rate of RTN chemoreceptors; simultaneous blockade of both channels led to a increase in CO(2) responsiveness. Furthermore, we also show that KCNQ channels but not SK channels are downstream effectors of serotonin modulation of RTN activity in vitro. In contrast, inhibition of KCNQ channel did not prevent modulation of RTN activity by Substance P or thyrotropin-releasing hormone, previously identified neuromodulators of RTN chemoreception. Importantly, we also show that KCNQ channels are critical for RTN activity in vivo. Inhibition of KCNQ channels lowered the CO(2) threshold for phrenic nerve discharge in anesthetized rats and decreased the ventilatory response to serotonin in awake and anesthetized animals. Given that serotonergic dysfunction may contribute to respiratory failure, our findings suggest KCNQ channels as a new therapeutic avenue for respiratory complications associated with multiple neurological disorders.


Assuntos
Tronco Encefálico/fisiologia , Células Quimiorreceptoras/fisiologia , Impulso (Psicologia) , Canais de Potássio KCNQ/fisiologia , Mecânica Respiratória/fisiologia , Serotonina/fisiologia , Anestesia , Animais , Animais Recém-Nascidos , Carbamatos/farmacologia , Dióxido de Carbono/fisiologia , Fenômenos Eletrofisiológicos , Técnicas In Vitro , Canais de Potássio KCNQ/agonistas , Canais de Potássio KCNQ/antagonistas & inibidores , Masculino , Fenilenodiaminas/farmacologia , Nervo Frênico/efeitos dos fármacos , Nervo Frênico/fisiologia , Ratos , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Substância P/farmacologia , Hormônio Liberador de Tireotropina/farmacologia
16.
J Neurosci ; 31(33): 11825-35, 2011 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-21849543

RESUMO

Multiple K(+) conductances are targets for many peripheral and central signals involved in the control of energy homeostasis. Potential K(+) channel targets are the KCNQ subunits that form the channels underlying the M-current, a subthreshold, non-inactivating K(+) current that is a common target for G-protein-coupled receptors. Whole-cell recordings were made from GFP (Renilla)-tagged neuropeptide Y (NPY) neurons from the arcuate nucleus of the hypothalamus using protocols to isolate and characterize the M-current in these orexigenic neurons. We recorded robust K(+) currents in the voltage range of the M-current, which were inhibited by the selective KCNQ channel blocker 10,10-bis(4-pyridinylmethyl)-9(10H)-anthracenone dihydrochloride (XE991) (40 µm), in both intact males and ovariectomized, 17ß-estradiol (E2)-treated females. Since NPY neurons are orexigenic and are active during fasting, the M-current was measured in fed and fasted male mice. Fasting attenuated the XE991-sensitive current by threefold, which correlated with decreased expression of the KCNQ2 and KCNQ3 subunits as measured with quantitative real-time PCR. Furthermore, E2 treatment augmented the XE991-sensitive M-current by threefold in ovariectomized (vs oil-treated) female mice. E2 treatment increased the expression of the KCNQ5 subunit in females but not KCNQ2 or KCNQ3 subunits. Fasting in females abrogated the effects of E2 on M-current activity, at least in part, by decreasing KCNQ2 and KCNQ3 expression. In summary, these data suggest that the M-current plays a pivotal role in the modulation of NPY neuronal excitability and may be an important cellular target for neurotransmitter and hormonal signals in the control of energy homeostasis in both males and females.


Assuntos
Potenciais de Ação/fisiologia , Estradiol/fisiologia , Jejum/fisiologia , Canais de Potássio KCNQ/fisiologia , Neurônios/fisiologia , Neuropeptídeo Y/fisiologia , Potenciais de Ação/efeitos dos fármacos , Animais , Metabolismo Energético/fisiologia , Estradiol/farmacologia , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Receptores Acoplados a Proteínas G/fisiologia
17.
J Physiol ; 590(19): 4839-58, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22733663

RESUMO

Natural stimuli are often characterized by statistics that can vary over orders of magnitude. Experiments have shown that sensory neurons continuously adapt their responses to changes in these statistics, thereby optimizing information transmission. However, such adaptation can also alter the neuronal transfer function by attenuating if not eliminating responses to the low frequency components of time varying stimuli,which can create ambiguity in the neural code. We recorded from electrosensory pyramidal neurons before and after pharmacological inactivation of either calcium-activated (I(AHP)) or KCNQ voltage-gated potassium currents (I(M)). We found that blocking each current decreased adaptation in a similar fashion but led to opposite changes in the neuronal transfer function. Indeed, blocking I(AHP) increased while blocking I(M) instead decreased the response to low temporal frequencies. To understand this surprising result, we built a mathematical model incorporating each channel type. This model predicted that these differential effects could be accounted for by differential activation properties. Our results show that the mechanisms that mediate adaptation can either increase or decrease the response to low frequency stimuli. As such, they suggest that the nervous system resolves ambiguity resulting from adaptation through independent control of adaptation and the neuronal transfer function.


Assuntos
Adaptação Fisiológica/fisiologia , Canais de Potássio KCNQ/fisiologia , Modelos Neurológicos , Canais de Potássio Cálcio-Ativados/fisiologia , Células Piramidais/fisiologia , Alcanos/farmacologia , Animais , Antracenos/farmacologia , Feminino , Peixes , Técnicas In Vitro , Masculino , Bloqueadores dos Canais de Potássio/farmacologia , Compostos de Quinolínio/farmacologia
18.
J Physiol ; 590(4): 793-807, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22155935

RESUMO

M-type (Kv7, KCNQ) K(+) channels control the resting membrane potential of many neurons, including peripheral nociceptive sensory neurons. Several M channel enhancers were suggested as prospective analgesics, and targeting M channels specifically in peripheral nociceptors is a plausible strategy for peripheral analgesia. However, receptor-induced inhibition of M channels in nociceptors is often observed in inflammation and may contribute to inflammatory pain. Such inhibition is predominantly mediated by phospholipase C. We investigated four M channel enhancers (retigabine, flupirtine, zinc pyrithione and H(2)O(2)) for their ability to overcome M channel inhibition via two phospholipase C-mediated mechanisms, namely depletion of membrane phosphatidylinositol 4,5-bisphosphate (PIP(2)) and a rise in intracellular Ca(2+) (an action mediated by calmodulin). Data from overexpressed Kv7.2/Kv7.3 heteromers and native M currents in dorsal root ganglion neurons suggest the following conclusions. (i) All enhancers had a dual effect on M channel activity, a negative shift in voltage dependence and an increase of the maximal current at saturating voltages. The enhancers differed in their efficacy to produce these effects. (ii) Both PIP(2) depletion and Ca(2+)/calmodulin strongly reduced the M current amplitude; however, at voltages near the threshold for M channel activation (-60 mV) all enhancers were able to restore M channel activity to a control level or above, while at saturating voltages the effects were more variable. (iii) Receptor-mediated inhibition of M current in nociceptive dorsal root ganglion neurons did not reduce the efficacy of retigabine or flupirtine to hyperpolarize the resting membrane potential. In conclusion, we show that all four M channel enhancers tested could overcome both PIP(2) and Ca(2+)-calmodulin-induced inhibition of Kv7.2/7.3 at voltages close to the threshold for action potential firing (-60 mV) but generally had reduced efficacy at a saturating voltage (0 mV). We suggest that the efficacy of an M channel enhancer to shift the voltage dependence of activation may be most important for rescuing M channel function in sensory neurons innervating inflamed tissue.


Assuntos
Inflamação/fisiopatologia , Canais de Potássio KCNQ/agonistas , Canais de Potássio KCNQ/fisiologia , Dor/fisiopatologia , Células Receptoras Sensoriais/fisiologia , Aminopiridinas/farmacologia , Animais , Bradicinina/fisiologia , Células CHO , Cálcio/fisiologia , Carbamatos/farmacologia , Cricetinae , Cricetulus , Gânglios Espinais/fisiologia , Peróxido de Hidrogênio/farmacologia , Compostos Organometálicos/farmacologia , Fenilenodiaminas/farmacologia , Fosfatidilinositol 4,5-Difosfato/deficiência , Piridinas/farmacologia , Ratos , Ratos Wistar , Fosfolipases Tipo C/fisiologia
20.
J Biol Chem ; 286(2): 1517-27, 2011 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-20966080

RESUMO

Despite advances in identifying deafness genes, determination of the underlying cellular and functional mechanisms for auditory diseases remains a challenge. Mutations of the human K(+) channel hKv7.4 lead to post-lingual progressive hearing loss (DFNA2), which affects world-wide population with diverse racial backgrounds. Here, we have generated the spectrum of point mutations in the hKv7.4 that have been identified as diseased mutants. We report that expression of five point mutations in the pore region, namely L274H, W276S, L281S, G285C, and G296S, as well as the C-terminal mutant G321S in the heterologous expression system, yielded non-functional channels because of endoplasmic reticulum retention of the mutant channels. We mimicked the dominant diseased conditions by co-expressing the wild-type and mutant channels. As compared with expression of wild-type channel alone, the blend of wild-type and mutant channel subunits resulted in reduced currents. Moreover, the combinatorial ratios of wild type:mutant and the ensuing current magnitude could not be explained by the predictions of a tetrameric channel and a dominant negative effect of the mutant subunits. The results can be explained by the dependence of cell surface expression of the mutant on the wild-type subunit. Surprisingly, a transmembrane mutation F182L, which has been identified in a pre-lingual progressive hearing loss patient in Taiwan, yielded cell surface expression and functional features that were similar to that of the wild type, suggesting that this mutation may represent redundant polymorphism. Collectively, these findings provide traces of the cellular mechanisms for DFNA2.


Assuntos
Perda Auditiva Neurossensorial/genética , Perda Auditiva Neurossensorial/fisiopatologia , Canais de Potássio KCNQ , Mutação Puntual , Animais , Células CHO , Cricetinae , Cricetulus , Progressão da Doença , Retículo Endoplasmático/fisiologia , Genes Dominantes , Humanos , Canais de Potássio KCNQ/química , Canais de Potássio KCNQ/genética , Canais de Potássio KCNQ/fisiologia , Potenciais da Membrana/fisiologia , Proteínas de Membrana/química , Proteínas de Membrana/genética , Proteínas de Membrana/fisiologia , Estrutura Terciária de Proteína , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA