Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 12(1): 4801, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34376649

RESUMEN

Hippocampal pyramidal cells encode an animal's location by single action potentials and complex spike bursts. These elementary signals are believed to play distinct roles in memory consolidation. The timing of single spikes and bursts is determined by intrinsic excitability and theta oscillations (5-10 Hz). Yet contributions of these dynamics to place fields remain elusive due to the lack of methods for specific modification of burst discharge. In mice lacking Kcnq3-containing M-type K+ channels, we find that pyramidal cell bursts are less coordinated by the theta rhythm than in controls during spatial navigation, but not alert immobility. Less modulated bursts are followed by an intact post-burst pause of single spike firing, resulting in a temporal discoordination of network oscillatory and intrinsic excitability. Place fields of single spikes in one- and two-dimensional environments are smaller in the mutant. Optogenetic manipulations of upstream signals reveal that neither medial septal GABA-ergic nor cholinergic inputs alone, but rather their joint activity, is required for entrainment of bursts. Our results suggest that altered representations by bursts and single spikes may contribute to deficits underlying cognitive disabilities associated with KCNQ3-mutations in humans.


Asunto(s)
Potenciales de Acción/fisiología , Canal de Potasio KCNQ3/fisiología , Células Piramidales/fisiología , Ritmo Teta/fisiología , Animales , Hipocampo/citología , Humanos , Canal de Potasio KCNQ3/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Optogenética/métodos
2.
J Gen Physiol ; 152(8)2020 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-32365171

RESUMEN

Retigabine is unique among anticonvulsant drugs by targeting the neuronal M-channel, which is composed of KV7.2/KV7.3 and contributes to the negative neuronal resting membrane potential. Unfortunately, retigabine causes adverse effects, which limits its clinical use. Adverse effects may be reduced by developing M-channel activators with improved KV7 subtype selectivity. The aim of this study was to evaluate the prospect of endocannabinoids as M-channel activators, either in isolation or combined with retigabine. Human KV7 channels were expressed in Xenopus laevis oocytes. The effect of extracellular application of compounds with different properties was studied using two-electrode voltage clamp electrophysiology. Site-directed mutagenesis was used to construct channels with mutated residues to aid in the mechanistic understanding of these effects. We find that arachidonoyl-L-serine (ARA-S), a weak endocannabinoid, potently activates the human M-channel expressed in Xenopus oocytes. Importantly, we show that ARA-S activates the M-channel via a different mechanism and displays a different KV7 subtype selectivity compared with retigabine. We demonstrate that coapplication of ARA-S and retigabine at low concentrations retains the effect on the M-channel while limiting effects on other KV7 subtypes. Our findings suggest that improved KV7 subtype selectivity of M-channel activators can be achieved through strategically combining compounds with different subtype selectivity.


Asunto(s)
Carbamatos/farmacología , Endocannabinoides , Canal de Potasio KCNQ2/farmacología , Canal de Potasio KCNQ3/fisiología , Fenilendiaminas/farmacología , Animales , Endocannabinoides/farmacología , Humanos , Oocitos , Xenopus laevis
3.
Commun Biol ; 3(1): 90, 2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-32111967

RESUMEN

Potassium channels form physical complexes with solute transporters in vivo, yet little is known about their range of possible signaling modalities and the underlying mechanisms. The KCNQ2/3 potassium channel, which generates neuronal M-current, is voltage-gated and its activity is also stimulated by binding of various small molecules. KCNQ2/3 forms reciprocally regulating complexes with sodium-coupled myo-inositol transporters (SMITs) in mammalian neurons. Here, we report that the neurotransmitter γ-aminobutyric acid (GABA) and other small molecules directly regulate myo-inositol transport in rat dorsal root ganglia, and by human SMIT1-KCNQ2/3 complexes in vitro, by inducing a distinct KCNQ2/3 pore conformation. Reciprocally, SMIT1 tunes KCNQ2/3 sensing of GABA and related metabolites. Ion permeation and mutagenesis studies suggest that SMIT1 and GABA similarly alter KCNQ2/3 pore conformation but via different KCNQ subunits and molecular mechanisms. KCNQ channels therefore act as chemosensors to enable co-assembled myo-inositol transporters to respond to diverse stimuli including neurotransmitters, metabolites and drugs.


Asunto(s)
Canales de Potasio KCNQ/fisiología , Proteínas de Transporte de Membrana/metabolismo , Animales , Animales Recién Nacidos , Transporte Biológico/genética , Femenino , Ganglios Espinales/metabolismo , Humanos , Inositol/metabolismo , Canales de Potasio KCNQ/genética , Canal de Potasio KCNQ2/genética , Canal de Potasio KCNQ2/metabolismo , Canal de Potasio KCNQ2/fisiología , Canal de Potasio KCNQ3/genética , Canal de Potasio KCNQ3/metabolismo , Canal de Potasio KCNQ3/fisiología , Neuronas/metabolismo , Multimerización de Proteína , Ratas , Transducción de Señal , Simportadores/fisiología , Xenopus laevis , Ácido gamma-Aminobutírico/farmacología
4.
Nat Commun ; 9(1): 1847, 2018 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-29748663

RESUMEN

Voltage-gated potassium channels KCNQ2-5 generate the M-current, which controls neuronal excitability. KCNQ2-5 subunits each harbor a high-affinity anticonvulsant drug-binding pocket containing an essential tryptophan (W265 in human KCNQ3) conserved for >500 million years, yet lacking a known physiological function. Here, phylogenetic analysis, electrostatic potential mapping, in silico docking, electrophysiology, and radioligand binding assays reveal that the anticonvulsant binding pocket evolved to accommodate endogenous neurotransmitters including γ-aminobutyric acid (GABA), which directly activates KCNQ5 and KCNQ3 via W265. GABA, and endogenous metabolites ß-hydroxybutyric acid (BHB) and γ-amino-ß-hydroxybutyric acid (GABOB), competitively and differentially shift the voltage dependence of KCNQ3 activation. Our results uncover a novel paradigm: direct neurotransmitter activation of voltage-gated ion channels, enabling chemosensing of the neurotransmitter/metabolite landscape to regulate channel activity and cellular excitability.


Asunto(s)
Anticonvulsivantes/metabolismo , Canales de Potasio KCNQ/fisiología , Canal de Potasio KCNQ3/fisiología , Neuronas/fisiología , Neurotransmisores/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Animales , Sitios de Unión/fisiología , Células CHO , Cricetulus , Ganglios Espinales/citología , Canales de Potasio KCNQ/química , Canal de Potasio KCNQ3/química , Masculino , Ratones , Simulación del Acoplamiento Molecular , Oocitos , Células PC12 , Técnicas de Placa-Clamp , Filogenia , Cultivo Primario de Células , Unión Proteica/fisiología , Ratas , Alineación de Secuencia , Triptófano/metabolismo , Xenopus laevis
5.
Elife ; 72018 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-29561262

RESUMEN

Somatosensory neurons mediate responses to diverse mechanical stimuli, from innocuous touch to noxious pain. While recent studies have identified distinct populations of A mechanonociceptors (AMs) that are required for mechanical pain, the molecular underpinnings of mechanonociception remain unknown. Here, we show that the bioactive lipid sphingosine 1-phosphate (S1P) and S1P Receptor 3 (S1PR3) are critical regulators of acute mechanonociception. Genetic or pharmacological ablation of S1PR3, or blockade of S1P production, significantly impaired the behavioral response to noxious mechanical stimuli, with no effect on responses to innocuous touch or thermal stimuli. These effects are mediated by fast-conducting A mechanonociceptors, which displayed a significant decrease in mechanosensitivity in S1PR3 mutant mice. We show that S1PR3 signaling tunes mechanonociceptor excitability via modulation of KCNQ2/3 channels. Our findings define a new role for S1PR3 in regulating neuronal excitability and establish the importance of S1P/S1PR3 signaling in the setting of mechanical pain thresholds.


Asunto(s)
Lisofosfolípidos/fisiología , Mecanorreceptores/fisiología , Dolor/fisiopatología , Transducción de Señal/fisiología , Esfingosina/análogos & derivados , Animales , Células Cultivadas , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Ganglios Espinales/fisiología , Canal de Potasio KCNQ2/metabolismo , Canal de Potasio KCNQ2/fisiología , Canal de Potasio KCNQ3/metabolismo , Canal de Potasio KCNQ3/fisiología , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Umbral del Dolor , Receptores de Lisoesfingolípidos/genética , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/fisiología , Receptores de Esfingosina-1-Fosfato
6.
Neuropharmacology ; 114: 67-76, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-27908768

RESUMEN

Acute stress stimulates corticotrophin-releasing hormone (CRH)-expressing neurons in the hypothalamic paraventricular nucleus (PVN), which is an essential component of hypothalamic-pituitary-adrenal (HPA) axis. However, the cellular and molecular mechanisms remain unclear. The M-channel is a voltage-dependent K+ channel involved in stabilizing the neuronal membrane potential and regulating neuronal excitability. In this study, we tested our hypothesis that acute stress suppresses expression of Kv7 channels to stimulate PVN-CRH neurons and the HPA axis. Rat PVN-CRH neurons were identified by expressing enhanced green fluorescent protein driven by Crh promoter. Acute restraint stress attenuated the excitatory effect of Kv7 blocker XE-991 on the firing activity of PVN-CRH neurons and blunted the increase in plasma corticosterone (CORT) levels induced by microinjection of XE-991 into the PVN. Furthermore, acute stress significantly decreased the M-currents in PVN-CRH neurons and reduced PVN expression of Kv7.3 subunit in the membrane. In addition, acute stress significantly increased phosphorylated AMP-activated protein kinase (AMPK) levels in the PVN tissue. Intracerebroventricular injection of the AMPK inhibitor dorsomorphin restored acute stress-induced elevation of CORT levels and reduction of membrane Kv7.3 protein level in the PVN. Dorsomorphin treatment increased the M-currents and reduced the firing activity of PVN-CRH neurons in acutely stressed rats. Collectively, these data suggest that acute stress diminishes Kv7 channels to stimulate PVN-CRH neurons and the HPA axis potentially via increased AMPK activity.


Asunto(s)
Sistema Hipotálamo-Hipofisario/fisiología , Canales de Potasio KCNQ/fisiología , Neuronas/fisiología , Sistema Hipófiso-Suprarrenal/fisiología , Estrés Psicológico/fisiopatología , Adenilato Quinasa/metabolismo , Animales , Corticosterona/sangre , Hormona Liberadora de Corticotropina/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Canal de Potasio KCNQ3/metabolismo , Canal de Potasio KCNQ3/fisiología , Masculino , Neuronas/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Subunidades de Proteína/metabolismo , Ratas , Ratas Sprague-Dawley , Estrés Psicológico/metabolismo
7.
J Physiol ; 595(3): 663-676, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27506413

RESUMEN

KEY POINTS: Retigabine is a KCNQ voltage-gated potassium channel opener that was recently approved as an add-on therapeutic for patients with drug-resistant epilepsy. Retigabine exhibits very little specificity between most KCNQ channel subtypes, and there is interest in generating more potent and specific KCNQ channel openers. The present study describes the marked specificity of ICA069673 for KCNQ2 vs. KCNQ3, and exploits this property to investigate determinants of KCNQ subtype specificity. ICA069673 acts on a binding site in the voltage-sensing domain that is distinct from the putative retigabine site in the channel pore. ICA069673 has two separable effects on KCNQ channel activity. We identify two channel residues required for subtype specificity of KCNQ channel openers and show that these are sufficient to generate ICA069673 sensitivity in KCNQ3. ABSTRACT: Retigabine (RTG) is the first approved anti-epileptic drug that acts via activation of voltage-gated potassium channels, targeting KCNQ channels that underlie the neuronal M-current. RTG exhibits little specificity between KCNQ2-5 as a result of conservation of a Trp residue in the pore domain that binds to the drug. The RTG analogue ICA-069673 ('ICA73') exhibits much stronger effects on KCNQ2 channels, including a large hyperpolarizing shift of the voltage-dependence of activation, an ∼2-fold enhancement of peak current and pronounced subtype specificity for KCNQ2 over KCNQ3. Based on ICA73 sensitivity of chimeric constructs of the transmembrane segments of KCNQ2 and KCNQ3, this drug appears to interact with the KCNQ2 voltage sensor (S1-S4) rather than the pore region targeted by RTG. KCNQ2 point mutants in the voltage sensor were generated based on KCNQ2/KCNQ3 sequence differences, and screened for ICA73 sensitivity. These experiments reveal that KCNQ2 residues F168 and A181 in the S3 segment are essential determinants of ICA73 subtype specificity. Mutations at either position in KCNQ2 abolish the ICA73-mediated gating shift, but preserve RTG sensitivity. Interestingly, A181P mutant channels show little ICA73-mediated gating shift but retain current potentiation by the drug. Mutations (L198F and P211A), which introduce these critical KCNQ2 residues at corresponding positions in KCNQ3, transplant partial ICA73 sensitivity. These findings demonstrate that RTG and ICA73 act via distinct mechanisms, and also reveal specific residues that underlie subtype specificity of KCNQ channel openers.


Asunto(s)
Carbamatos/farmacología , Canal de Potasio KCNQ2/fisiología , Canal de Potasio KCNQ3/fisiología , Moduladores del Transporte de Membrana/farmacología , Fenilendiaminas/farmacología , Células HEK293 , Humanos , Activación del Canal Iónico , Canal de Potasio KCNQ2/genética , Canal de Potasio KCNQ3/genética , Modelos Moleculares , Mutación
9.
Circ Res ; 115(2): 263-72, 2014 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-24838176

RESUMEN

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.


Asunto(s)
Arterias Mesentéricas/fisiología , Miocitos del Músculo Liso/fisiología , Presorreceptores/fisiología , Receptor de Angiotensina Tipo 1/fisiología , Arteria Renal/fisiología , 4-Aminopiridina/farmacología , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Antracenos/farmacología , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/fisiología , Células HEK293 , Hemorreología , Humanos , Canales de Potasio KCNQ/fisiología , Canal de Potasio KCNQ3/fisiología , Losartán/farmacología , Arterias Mesentéricas/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Presión Osmótica , Receptor de Angiotensina Tipo 1/deficiencia , Receptor de Angiotensina Tipo 1/genética , Arteria Renal/citología , Canales Catiónicos TRPC/fisiología , Canal Catiónico TRPC6 , Transcripción Genética , Resistencia Vascular/efectos de los fármacos , Resistencia Vascular/fisiología
10.
Proc Natl Acad Sci U S A ; 111(22): E2281-90, 2014 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-24843134

RESUMEN

Plasma membrane (PM) phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2] regulates the activity of many ion channels and other membrane-associated proteins. To determine precursor sources of the PM PI(4,5)P2 pool in tsA-201 cells, we monitored KCNQ2/3 channel currents and translocation of PHPLCδ1 domains as real-time indicators of PM PI(4,5)P2, and translocation of PHOSH2×2, and PHOSH1 domains as indicators of PM and Golgi phosphatidylinositol 4-phosphate [PI(4)P], respectively. We selectively depleted PI(4)P pools at the PM, Golgi, or both using the rapamycin-recruitable lipid 4-phosphatases. Depleting PI(4)P at the PM with a recruitable 4-phosphatase (Sac1) results in a decrease of PI(4,5)P2 measured by electrical or optical indicators. Depleting PI(4)P at the Golgi with the 4-phosphatase or disrupting membrane-transporting motors induces a decline in PM PI(4,5)P2. Depleting PI(4)P simultaneously at both the Golgi and the PM induces a larger decrease of PI(4,5)P2. The decline of PI(4,5)P2 following 4-phosphatase recruitment takes 1-2 min. Recruiting the endoplasmic reticulum (ER) toward the Golgi membranes mimics the effects of depleting PI(4)P at the Golgi, apparently due to the trans actions of endogenous ER Sac1. Thus, maintenance of the PM pool of PI(4,5)P2 appears to depend on precursor pools of PI(4)P both in the PM and in the Golgi. The decrease in PM PI(4,5)P2 when Sac1 is recruited to the Golgi suggests that the Golgi contribution is ongoing and that PI(4,5)P2 production may be coupled to important cell biological processes such as membrane trafficking or lipid transfer activity.


Asunto(s)
Membrana Celular/metabolismo , Aparato de Golgi/metabolismo , Canal de Potasio KCNQ2/fisiología , Canal de Potasio KCNQ3/fisiología , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatidilinositoles/metabolismo , 1-Fosfatidilinositol 4-Quinasa/metabolismo , Androstadienos/farmacología , Células Cultivadas , Humanos , Riñón/citología , Potenciales de la Membrana/fisiología , Miosina Tipo II/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Wortmanina
11.
J Neurosci ; 34(10): 3719-32, 2014 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-24599470

RESUMEN

Rapid energy-efficient signaling along vertebrate axons is achieved through intricate subcellular arrangements of voltage-gated ion channels and myelination. One recently appreciated example is the tight colocalization of K(v)7 potassium channels and voltage-gated sodium (Na(v)) channels in the axonal initial segment and nodes of Ranvier. The local biophysical properties of these K(v)7 channels and the functional impact of colocalization with Na(v) channels remain poorly understood. Here, we quantitatively examined K(v)7 channels in myelinated axons of rat neocortical pyramidal neurons using high-resolution confocal imaging and patch-clamp recording. K(v)7.2 and 7.3 immunoreactivity steeply increased within the distal two-thirds of the axon initial segment and was mirrored by the conductance density estimates, which increased from ~12 (proximal) to 150 pS µm(-2) (distal). The axonal initial segment and nodal M-currents were similar in voltage dependence and kinetics, carried by K(v)7.2/7.3 heterotetramers, 4% activated at the resting membrane potential and rapidly activated with single-exponential time constants (~15 ms at 28 mV). Experiments and computational modeling showed that while somatodendritic K(v)7 channels are strongly activated by the backpropagating action potential to attenuate the afterdepolarization and repetitive firing, axonal K(v)7 channels are minimally recruited by the forward-propagating action potential. Instead, in nodal domains K(v)7.2/7.3 channels were found to increase Na(v) channel availability and action potential amplitude by stabilizing the resting membrane potential. Thus, K(v)7 clustering near axonal Na(v) channels serves specific and context-dependent roles, both restraining initiation and enhancing conduction of the action potential.


Asunto(s)
Potenciales de Acción/fisiología , Axones/fisiología , Canal de Potasio KCNQ2/fisiología , Canal de Potasio KCNQ3/fisiología , Neocórtex/fisiología , Conducción Nerviosa/fisiología , Animales , Masculino , Fibras Nerviosas Mielínicas/fisiología , Técnicas de Cultivo de Órganos , Ratas , Ratas Wistar
12.
Pain ; 154(3): 434-448, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23352759

RESUMEN

Bone cancer pain has a strong impact on the quality of life of patients, but is difficult to treat. Better understanding of the pathogenic mechanisms underlying bone cancer pain will likely lead to the development of more effective treatments. In the present study, we investigated whether inhibition of KCNQ/M channels contributed to the hyperexcitability of primary sensory neurons and to the pathogenesis of bone cancer pain. By using a rat model of bone cancer pain based on intratibial injection of MRMT-1 tumour cells, we documented a prominent decrease in expression of KCNQ2 and KCNQ3 proteins and a reduction of M-current density in small-sized dorsal root ganglia (DRG) neurons, which were associated with enhanced excitability of these DRG neurons and the hyperalgesic behaviours in bone cancer rats. Coincidently, we found that inhibition of KCNQ/M channels with XE-991 caused a robust increase in the excitability of small-sized DRG neurons and produced an obvious mechanical allodynia in normal rats. On the contrary, activation of the KCNQ/M channels with retigabine not only inhibited the hyperexcitability of these small DRG neurons, but also alleviated mechanical allodynia and thermal hyperalgesia in bone cancer rats, and all of these effects of retigabine could be blocked by KCNQ/M-channel antagonist XE-991. These results suggest that repression of KCNQ/M channels leads to the hyperexcitability of primary sensory neurons, which in turn causes bone cancer pain. Thus, suppression of KCNQ/M channels in primary DRG neurons plays a crucial role in the development of bone cancer pain.


Asunto(s)
Neoplasias Óseas/fisiopatología , Carcinoma/fisiopatología , Ganglios Espinales/fisiopatología , Hiperalgesia/etiología , Canal de Potasio KCNQ2/fisiología , Canal de Potasio KCNQ3/fisiología , Nocicepción/fisiología , Dolor/etiología , Células Receptoras Sensoriales/fisiología , Animales , Antracenos/farmacología , Neoplasias Óseas/patología , Neoplasias Óseas/secundario , Carbamatos/farmacología , Carbamatos/uso terapéutico , Carcinoma/patología , Carcinoma/secundario , Regulación hacia Abajo , Femenino , Calor/efectos adversos , Hiperalgesia/fisiopatología , Canal de Potasio KCNQ2/antagonistas & inhibidores , Canal de Potasio KCNQ2/biosíntesis , Canal de Potasio KCNQ2/genética , Canal de Potasio KCNQ3/antagonistas & inhibidores , Canal de Potasio KCNQ3/biosíntesis , Canal de Potasio KCNQ3/genética , Neoplasias Mamarias Experimentales/patología , Trasplante de Neoplasias , Técnicas de Placa-Clamp , Fenilendiaminas/farmacología , Fenilendiaminas/uso terapéutico , Bloqueadores de los Canales de Potasio/farmacología , Ratas , Ratas Sprague-Dawley , Células Receptoras Sensoriales/metabolismo , Estrés Mecánico , Transmisión Sináptica , Tibia/patología
13.
Proc Natl Acad Sci U S A ; 109(35): E2316-23, 2012 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-22847441

RESUMEN

Phosphoinositides (PIs) are lipid components of cell membranes that regulate a wide variety of cellular functions. Here we exploited the blue light-induced dimerization between two plant proteins, cryptochrome 2 (CRY2) and the transcription factor CIBN, to control plasma membrane PI levels rapidly, locally, and reversibly. The inositol 5-phosphatase domain of OCRL (5-ptase(OCRL)), which acts on PI(4,5)P(2) and PI(3,4,5)P(3), was fused to the photolyase homology region domain of CRY2, and the CRY2-binding domain, CIBN, was fused to plasma membrane-targeting motifs. Blue-light illumination (458-488 nm) of mammalian cells expressing these constructs resulted in nearly instantaneous recruitment of 5-ptase(OCRL) to the plasma membrane, where it caused rapid (within seconds) and reversible (within minutes) dephosphorylation of its targets as revealed by diverse cellular assays: dissociation of PI(4,5)P(2) and PI(3,4,5)P(3) biosensors, disappearance of endocytic clathrin-coated pits, nearly complete inhibition of KCNQ2/3 channel currents, and loss of membrane ruffling. Focal illumination resulted in local and transient 5-ptase(OCRL) recruitment and PI(4,5)P(2) dephosphorylation, causing not only local collapse and retraction of the cell edge or process but also compensatory accumulation of the PI(4,5)P(2) biosensor and membrane ruffling at the opposite side of the cells. Using the same approach for the recruitment of PI3K, local PI(3,4,5)P(3) synthesis and membrane ruffling could be induced, with corresponding loss of ruffling distally to the illuminated region. This technique provides a powerful tool for dissecting with high spatial-temporal kinetics the cellular functions of various PIs and reversibly controlling the functions of downstream effectors of these signaling lipids.


Asunto(s)
Endocitosis/fisiología , Endocitosis/efectos de la radiación , Luz , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatidilinositoles/metabolismo , Actinas/metabolismo , Secuencias de Aminoácidos/fisiología , Animales , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Sitios de Unión/fisiología , Células COS , Membrana Celular/metabolismo , Membrana Celular/efectos de la radiación , Chlorocebus aethiops , Criptocromos/genética , Criptocromos/metabolismo , Humanos , Canal de Potasio KCNQ2/fisiología , Canal de Potasio KCNQ3/fisiología , Potenciales de la Membrana/fisiología , Potenciales de la Membrana/efectos de la radiación , Células PC12 , Fosfatidilinositol 3-Quinasas/metabolismo , Monoéster Fosfórico Hidrolasas/genética , Monoéster Fosfórico Hidrolasas/metabolismo , Fosforilación/fisiología , Fosforilación/efectos de la radiación , Ratas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal/fisiología , Transducción de Señal/efectos de la radiación
14.
J Neurosci ; 31(40): 14158-71, 2011 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-21976501

RESUMEN

Whereas neuronal M-type K(+) channels composed of KCNQ2 and KCNQ3 subunits regulate firing properties of neurons, presynaptic KCNQ2 subunits were demonstrated to regulate neurotransmitter release by directly influencing presynaptic function. Two interaction partners of M-channels, syntaxin 1A and calmodulin, are known to act presynaptically, syntaxin serving as a major protein component of the membrane fusion machinery and calmodulin serving as regulator of several processes related to neurotransmitter release. Notably, both partners specifically modulate KCNQ2 but not KCNQ3 subunits, suggesting selective presynaptic targeting to directly regulate exocytosis without interference in neuronal firing properties. Here, having first demonstrated in Xenopus oocytes, using analysis of single-channel biophysics, that both modulators downregulate the open probability of KCNQ2 but not KCNQ3 homomers, we sought to resolve the channel structural determinants that confer the isoform-specific gating downregulation and to get insights into the molecular events underlying this mechanism. We show, using optical, biochemical, electrophysiological, and molecular biology analyses, the existence of constitutive interactions between the N and C termini in homomeric KCNQ2 and KCNQ3 channels in living cells. Furthermore, rearrangement in the relative orientation of the KCNQ2 termini that accompanies reduction in single-channel open probability is induced by both regulators, strongly suggesting that closer N-C termini proximity underlies gating downregulation. Different structural determinants, identified at the N and C termini of KCNQ3, prevent the effects by syntaxin 1A and calmodulin, respectively. Moreover, we show that the syntaxin 1A and calmodulin effects can be additive or blocked at different concentration ranges of calmodulin, bearing physiological significance with regard to presynaptic exocytosis.


Asunto(s)
Calmodulina/fisiología , Activación del Canal Iónico/fisiología , Canal de Potasio KCNQ2/fisiología , Canal de Potasio KCNQ3/fisiología , Neuronas/fisiología , Sintaxina 1/fisiología , Animales , Exocitosis/fisiología , Femenino , Humanos , Canal de Potasio KCNQ2/química , Canal de Potasio KCNQ3/química , Neuronas/metabolismo , Oocitos/química , Oocitos/metabolismo , Oocitos/fisiología , Técnicas de Placa-Clamp , Isoformas de Proteínas/química , Isoformas de Proteínas/fisiología , Xenopus laevis
15.
J Neurosci Methods ; 200(1): 54-62, 2011 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-21723881

RESUMEN

KCNQ2/3 voltage-gated potassium channels conduct low-threshold, slowly activating and non-inactivating currents to repolarize the neuronal resting membrane potential. The channels negatively regulate neuronal excitability and KCNQ2/3 openers are efficacious in hyperexcited states such as epilepsy and pain. We developed and utilized thallium influx assays to profile novel KCNQ2/3 channel openers with respect to selectivity across KCNQ subtypes and on requirement for tryptophan 236 of KCNQ2, a critical residue for activity of the KCNQ opener retigabine. Using distinct chemical series of openers, a quinazolinone series showed relatively poor selectivity across multiple KCNQ channels and lacked activity at the KCNQ2(W236L) mutant channel. In contrast, several novel benzimidazole openers showed selectivity for KCNQ2/3 and KCNQ2 and retain activity at KCNQ2(W236L). Profiling of several hundred KCNQ2/3 openers across multiple diverse chemical series revealed that openers show differential degrees of selectivity across subtypes, with selectivity most difficult to achieve against KCNQ2. In addition, we report the significant finding that KCNQ openers can pharmacologically differentiate between homomeric and heteromeric channels containing subtypes in common. Moreover, most openers assayed were dependent on the W236 for activity, whereas only a small number appear to use a distinct mechanism. Collectively, we provide novel insights into the molecular pharmacology of KCNQ channels by demonstrating differential selectivity and site of action for KCNQ2/3 openers. The high-throughput thallium influx assays should prove useful for rapid characterization of KCNQ openers and in guiding efforts to identify selective compounds for advancement towards the clinic.


Asunto(s)
Activación del Canal Iónico/efectos de los fármacos , Canal de Potasio KCNQ2/metabolismo , Canal de Potasio KCNQ3/metabolismo , Talio/farmacocinética , Carbamatos/farmacología , Células HEK293 , Humanos , Activación del Canal Iónico/genética , Canales de Potasio KCNQ/genética , Canales de Potasio KCNQ/metabolismo , Canales de Potasio KCNQ/fisiología , Canal de Potasio KCNQ2/genética , Canal de Potasio KCNQ2/fisiología , Canal de Potasio KCNQ3/genética , Canal de Potasio KCNQ3/fisiología , Mutación , Fenilendiaminas/farmacología
16.
PLoS One ; 6(7): e21792, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21750731

RESUMEN

BACKGROUND: K(V)7/KCNQ channels are widely expressed in neurons and they have multiple important functions, including control of excitability, spike afterpotentials, adaptation, and theta resonance. Mutations in KCNQ genes have been demonstrated to associate with human neurological pathologies. However, little is known about whether K(V)7/KCNQ channels are expressed in oligodendrocyte lineage cells (OLCs) and what their functions in OLCs. METHODS AND FINDINGS: In this study, we characterized K(V)7/KCNQ channels expression in rat primary cultured OLCs by RT-PCR, immunostaining and electrophysiology. KCNQ2-5 mRNAs existed in all three developmental stages of rat primary cultured OLCs. K(V)7/KCNQ proteins were also detected in oligodendrocyte progenitor cells (OPCs, early developmental stages of OLCs) of rat primary cultures and cortex slices. Voltage-clamp recording revealed that the I(M) antagonist XE991 significantly reduced K(V)7/KCNQ channel current (I(K(Q))) in OPCs but not in differentiated oligodendrocytes. In addition, inhibition of K(V)7/KCNQ channels promoted OPCs motility in vitro. CONCLUSIONS: These findings showed that K(V)7/KCNQ channels were functionally expressed in rat primary cultured OLCs and might play an important role in OPCs functioning in physiological or pathological conditions.


Asunto(s)
Canales de Potasio KCNQ/metabolismo , Canal de Potasio KCNQ2/metabolismo , Canal de Potasio KCNQ3/metabolismo , Oligodendroglía/metabolismo , Células Madre/metabolismo , Animales , Animales Recién Nacidos , Antracenos/farmacología , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Relación Dosis-Respuesta a Droga , Inmunohistoquímica , Canales de Potasio KCNQ/genética , Canales de Potasio KCNQ/fisiología , Canal de Potasio KCNQ2/genética , Canal de Potasio KCNQ2/fisiología , Canal de Potasio KCNQ3/genética , Canal de Potasio KCNQ3/fisiología , Potenciales de la Membrana/efectos de los fármacos , Oligodendroglía/citología , Oligodendroglía/fisiología , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/farmacología , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/citología , Células Madre/fisiología
17.
J Physiol ; 588(Pt 22): 4519-37, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-20876197

RESUMEN

Cerebral vascular smooth muscle contractility plays a crucial role in controlling arterial diameter and, thereby, blood flow regulation in the brain. A number of K(+) channels have been suggested to contribute to the regulation of diameter by controlling smooth muscle membrane potential (E(m)) and Ca(2+) influx. Previous studies indicate that stromatoxin (ScTx1)-sensitive, Kv2-containing channels contribute to the control of cerebral arterial diameter at 80 mmHg, but their precise role and molecular composition were not determined. Here, we tested if Kv2 subunits associate with 'silent' subunits from the Kv5, Kv6, Kv8 or Kv9 subfamilies to form heterotetrameric channels that contribute to control of diameter of rat middle cerebral arteries (RMCAs) over a range of intraluminal pressure from 10 to 100 mmHg. The predominant mRNAs expressed by RMCAs encode Kv2.1 and Kv9.3 subunits. Co-localization of Kv2.1 and Kv9.3 proteins at the plasma membrane of dissociated single RMCA myocytes was detected by proximity ligation assay. ScTx1-sensitive native current of RMCA myocytes and Kv2.1/Kv9.3 currents exhibited functional identity based on the similarity of their deactivation kinetics and voltage dependence of activation that were distinct from those of homomultimeric Kv2.1 channels. ScTx1 treatment enhanced the myogenic response of pressurized RMCAs between 40 and 100 mmHg, but this toxin also caused constriction between 10 and 40 mmHg that was not previously observed following inhibition of large conductance Ca(2+)-activated K(+) (BK(Ca)) and Kv1 channels. Taken together, this study defines the molecular basis of Kv2-containing channels and contributes to our understanding of the functional significance of their expression in cerebral vasculature. Specifically, our findings provide the first evidence of heteromultimeric Kv2.1/Kv9.3 channel expression in RMCA myocytes and their distinct contribution to control of cerebral arterial diameter over a wider range of E(m) and transmural pressure than Kv1 or BK(Ca) channels owing to their negative range of voltage-dependent activation.


Asunto(s)
Arterias Cerebrales/fisiología , Canal de Potasio KCNQ3/fisiología , Péptidos/fisiología , Multimerización de Proteína/fisiología , Subunidades de Proteína/fisiología , Canales de Potasio Shab/fisiología , Vasoconstricción/fisiología , Animales , Células HEK293 , Humanos , Canal de Potasio KCNQ3/química , Masculino , Subunidades de Proteína/química , Ratas , Ratas Sprague-Dawley , Canales de Potasio Shab/antagonistas & inhibidores , Canales de Potasio Shab/química , Venenos de Araña
18.
J Gen Physiol ; 135(2): 99-114, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20100891

RESUMEN

The signaling phosphoinositide phosphatidylinositol 4,5-bisphosphate (PIP(2)) is synthesized in two steps from phosphatidylinositol by lipid kinases. It then interacts with KCNQ channels and with pleckstrin homology (PH) domains among many other physiological protein targets. We measured and developed a quantitative description of these metabolic and protein interaction steps by perturbing the PIP(2) pool with a voltage-sensitive phosphatase (VSP). VSP can remove the 5-phosphate of PIP(2) with a time constant of tau <300 ms and fully inhibits KCNQ currents in a similar time. PIP(2) was then resynthesized from phosphatidylinositol 4-phosphate (PIP) quickly, tau = 11 s. In contrast, resynthesis of PIP(2) after activation of phospholipase C by muscarinic receptors took approximately 130 s. These kinetic experiments showed that (1) PIP(2) activation of KCNQ channels obeys a cooperative square law, (2) the PIP(2) residence time on channels is <10 ms and the exchange time on PH domains is similarly fast, and (3) the step synthesizing PIP(2) by PIP 5-kinase is fast and limited primarily by a step(s) that replenishes the pool of plasma membrane PI(4)P. We extend the kinetic model for signaling from M(1) muscarinic receptors, presented in our companion paper in this issue (Falkenburger et al. 2010. J. Gen. Physiol. doi:10.1085/jgp.200910344), with this new information on PIP(2) synthesis and KCNQ interaction.


Asunto(s)
Canal de Potasio KCNQ2/metabolismo , Canal de Potasio KCNQ3/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Línea Celular , Fenómenos Electrofisiológicos , Humanos , Canal de Potasio KCNQ2/fisiología , Canal de Potasio KCNQ3/fisiología , Cinética , Monoéster Fosfórico Hidrolasas/fisiología , Fosfotransferasas (Aceptor de Grupo Alcohol)/fisiología , Unión Proteica/fisiología , Receptores Muscarínicos/metabolismo , Receptores Muscarínicos/fisiología , Transducción de Señal/fisiología
19.
Epilepsia ; 50(7): 1752-9, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19453707

RESUMEN

PURPOSE: Benign familial neonatal convulsions (BFNC) is caused by mutations in the KCNQ2 and KCNQ3 genes, which encode subunits of the M-type potassium channel. The purpose of this study was to examine the effects of orthologous BFNC-causing mutations on seizure thresholds and the acquisition of corneal kindling in mice with heterozygous expression of the mutations. METHODS: The effects of the Kcnq2 gene A306T mutation and the Kcnq3 gene G311V mutation were determined for minimal clonic, minimal tonic hindlimb extension, and partial psychomotor seizures. The rate of corneal kindling acquisition was also determined for Kcnq2 A306T and Kcnq3 G311V mice. RESULTS: Seizure thresholds were significantly altered relative to wild-type animals in the minimal clonic, minimal tonic hindlimb extension, and partial psychomotor seizure models. Differences in seizure threshold were found to be dependent on the mutation expressed, the seizure testing paradigm, the genetic background strain, and the gender of the animal. Mutations in Kcnq2 and Kcnq3 were associated with an increased rate of corneal kindling. In the Kcnq2 A306T mice, an increased incidence of death occurred during and immediately following the conclusion of the kindling acquisition period. CONCLUSIONS: These results suggest that genetic alterations in the subunits that underlie the M-current and cause BFNC alter seizure susceptibility in a sex-, mouse strain-, and seizure-test dependent manner. Although the heterozygous mice do not appear to have spontaneous seizures, the increased seizure susceptibility and incidence of death during and after kindling suggests that these mutations lead to altered excitability in these animals.


Asunto(s)
Epilepsia Benigna Neonatal/genética , Canal de Potasio KCNQ2/genética , Canal de Potasio KCNQ3/genética , Excitación Neurológica/fisiología , Mutación/genética , Convulsiones/genética , Potenciales de Acción/fisiología , Animales , Modelos Animales de Enfermedad , Estimulación Eléctrica , Electrocardiografía , Epilepsia Benigna Neonatal/fisiopatología , Femenino , Técnicas de Sustitución del Gen/métodos , Predisposición Genética a la Enfermedad , Heterocigoto , Humanos , Canal de Potasio KCNQ2/fisiología , Canal de Potasio KCNQ3/fisiología , Excitación Neurológica/genética , Masculino , Ratones , Mutación Missense/genética , Proteínas del Tejido Nervioso/genética , Convulsiones/fisiopatología , Factores Sexuales
20.
Proc Natl Acad Sci U S A ; 105(50): 19974-9, 2008 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-19060215

RESUMEN

Benign familial neonatal convulsion (BNFC) is a neurological disorder caused by mutations in the potassium channel genes KCNQ2 and KCNQ3, which are thought to contribute to the medium afterhyperpolarization (mAHP). Despite their importance in normal brain function, it is unknown whether they invariably function as heteromeric complexes. Here, we examined the contribution of KCNQ3 and KCNQ2 in mediating the apamin-insensitive mAHP current (ImAHP) in hippocampus. The ImAHP was not impaired in CA1 pyramidal neurons from mice genetically deficient for either KCNQ3 or KCNQ2 but was reduced approximately 50% in dentate granule cells. While recording from KCNQ-deficient mice, we observed that the calcium-activated slow afterhyperpolarization current (IsAHP) was also reduced in dentate granule cells, suggesting that KCNQ channels might also contribute to this potassium current whose molecular identity is unknown. Further pharmacological and molecular experiments manipulating KCNQ channels provided evidence in support of this possibility. Together our data suggest that multiple KCNQ subunit compositions can mediate the ImAHP, and that the very same subunits may also contribute to the IsAHP. We also present data suggesting that the neuronal calcium sensor protein hippocalcin may allow for these dual signaling processes.


Asunto(s)
Apamina/farmacología , Hipocampo/fisiología , Canal de Potasio KCNQ2/fisiología , Canal de Potasio KCNQ3/fisiología , Proteínas del Tejido Nervioso/fisiología , Animales , Epilepsia Benigna Neonatal/genética , Hipocalcina/fisiología , Hipocampo/citología , Hipocampo/efectos de los fármacos , Canal de Potasio KCNQ2/genética , Canal de Potasio KCNQ3/genética , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Células Piramidales/efectos de los fármacos , Células Piramidales/fisiología , Ganglio Cervical Superior/citología , Ganglio Cervical Superior/efectos de los fármacos , Ganglio Cervical Superior/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...