Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 164
Filtrar
1.
Eur J Neurosci ; 59(1): 3-16, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38018635

RESUMEN

The expression of IKCa (SK4) channel subunits overlaps with that of SK channel subunits, and it has been proposed that the two related subunits prefer to co-assemble to form heteromeric hSK1:hIKCa channels. This implicates hSK1:hIKCa heteromers in physiological roles that might have been attributed to activation of SK channels. We have used a mutation approach to confirm formation of heterometric hSK1:hIKCa channels. Introduction of residues within hSK1 that were predicted to impart sensitivity to the hIKCa current blocker TRAM-34 changed the pharmacology of functional heteromers. Heteromeric channels formed between wildtype hIKCa and mutant hSK1 subunits displayed a significantly higher sensitivity and maximum block to addition of TRAM-34 than heteromers formed between wildtype subunits. Heteromer formation was disrupted by a single point mutation within one COOH-terminal coiled-coil domain of the hIKCa channel subunit. This mutation only disrupted the formation of hSK1:hIKCa heteromeric channels, without affecting the formation of homomeric hIKCa channels. Finally, the Ca2+ gating sensitivity of heteromeric hSK1:hIKCa channels was found to be significantly lower than the Ca2+ gating sensitivity of homomeric hIKCa channels. These data confirmed the preferred formation of heteromeric channels that results from COOH-terminal interactions between subunits. The distinct sensitivity of the heteromer to activation by Ca2+ suggests that heteromeric channels fulfil a distinct function within those neurons that express both subunits.


Asunto(s)
Canales de Potasio de Conductancia Intermedia Activados por el Calcio , Neuronas , Canales de Potasio de Pequeña Conductancia Activados por el Calcio , Mutación , Humanos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/genética , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/fisiología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/genética , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/fisiología
2.
PLoS Biol ; 18(1): e3000596, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31905212

RESUMEN

Neurons store information by changing synaptic input weights. In addition, they can adjust their membrane excitability to alter spike output. Here, we demonstrate a role of such "intrinsic plasticity" in behavioral learning in a mouse model that allows us to detect specific consequences of absent excitability modulation. Mice with a Purkinje-cell-specific knockout (KO) of the calcium-activated K+ channel SK2 (L7-SK2) show intact vestibulo-ocular reflex (VOR) gain adaptation but impaired eyeblink conditioning (EBC), which relies on the ability to establish associations between stimuli, with the eyelid closure itself depending on a transient suppression of spike firing. In these mice, the intrinsic plasticity of Purkinje cells is prevented without affecting long-term depression or potentiation at their parallel fiber (PF) input. In contrast to the typical spike pattern of EBC-supporting zebrin-negative Purkinje cells, L7-SK2 neurons show reduced background spiking but enhanced excitability. Thus, SK2 plasticity and excitability modulation are essential for specific forms of motor learning.


Asunto(s)
Potenciales de Acción/genética , Aprendizaje/fisiología , Memoria/fisiología , Actividad Motora/fisiología , Células de Purkinje/metabolismo , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/fisiología , Animales , Cerebelo/citología , Cerebelo/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Plasticidad Neuronal/fisiología , Reflejo Vestibuloocular , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo
3.
Pflugers Arch ; 473(3): 491-506, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33411079

RESUMEN

Apamin-sensitive small-conductance calcium-activated potassium (SK) current (IKAS) plays an important role in cardiac repolarization under a variety of physiological and pathological conditions. The regulation of cardiac IKAS relies on SK channel expression, intracellular Ca2+, and interaction between SK channel and intracellular Ca2+. IKAS activation participates in multiple types of arrhythmias, including atrial fibrillation, ventricular tachyarrhythmias, and automaticity and conduction abnormality. Recently, sex dimorphisms in autonomic control have been noticed in IKAS activation, resulting in sex-differentiated action potential morphology and arrhythmogenesis. This review provides an update on the Ca2+-dependent regulation of cardiac IKAS and the role of IKAS on arrhythmias, with a special focus on sex differences in IKAS activation. We propose that sex dimorphism in autonomic control of IKAS may play a role in J wave syndrome.


Asunto(s)
Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatología , Caracteres Sexuales , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/fisiología , Animales , Femenino , Humanos , Masculino
4.
J Neurosci ; 39(40): 7826-7839, 2019 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-31420457

RESUMEN

Dendritic excitability regulates how neurons integrate synaptic inputs and thereby influences neuronal output. As active dendritic events are associated with significant calcium influx they are likely to be modulated by calcium-dependent processes, such as calcium-activated potassium channels. Here we investigate the impact of small conductance calcium-activated potassium channels (SK channels) on dendritic excitability in male and female rat cortical pyramidal neurons in vitro and in vivo Using local applications of the SK channel antagonist apamin in vitro, we show that blocking somatic SK channels enhances action potential output, whereas blocking dendritic SK channels paradoxically reduces the generation of dendritic calcium spikes and associated somatic burst firing. Opposite effects were observed using the SK channel enhancer NS309. The effect of apamin on dendritic SK channels was occluded when R-type calcium channels were blocked, indicating that the inhibitory impact of apamin on dendritic calcium spikes involved R-type calcium channels. Comparable effects were observed in vivo Intracellular application of apamin via the somatic whole-cell recording pipette reduced the medium afterhyperpolarization and increased action potential output during UP states. In contrast, extracellular application of apamin to the cortical surface to block dendritic SK channels shifted the distribution of action potentials within UP states from an initial burst to a more distributed firing pattern, while having no impact on overall action potential firing frequency or UP and DOWN states. These data indicate that somatic and dendritic SK channels have opposite effects on neuronal excitability, with dendritic SK channels counter-intuitively promoting rather than suppressing neuronal output.SIGNIFICANCE STATEMENT Neurons typically receive input from other neurons onto processes called dendrites, and use electrical events such as action potentials for signaling. As electrical events in neurons are usually associated with calcium influx they can be regulated by calcium-dependent processes. One such process is through the activation of calcium-dependent potassium channels, which usually act to reduce action potential signaling. Although this is the case for calcium-dependent potassium channels found at the cell body, we show here that calcium-dependent potassium channels in dendrites of cortical pyramidal neurons counter-intuitively promote rather than suppress action potential output.


Asunto(s)
Dendritas/fisiología , Células Piramidales/fisiología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/fisiología , Animales , Apamina/farmacología , Canales de Calcio Tipo R/efectos de los fármacos , Señalización del Calcio/efectos de los fármacos , Dendritas/efectos de los fármacos , Fenómenos Electrofisiológicos/efectos de los fármacos , Femenino , Indoles/farmacología , Masculino , Oximas/farmacología , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/farmacología , Células Piramidales/efectos de los fármacos , Ratas , Ratas Wistar
5.
Clin Nephrol ; 93(4): 195-202, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32049626

RESUMEN

AIM: By observing the expression and distribution of platelet-derived growth factor receptor α-positive (PDGFRα+) cells in ureteropelvic junction obstruction (UPJO), to explore their role in the pathogenesis of children with congenital hydronephrosis. MATERIALS AND METHODS: The control group involved specimens of the normal ureter (nephrectomy for tumor; n = 10), and the UPJO group contained specimens of ureteropelvic junction (UPJ) segment excised during pyeloplasty (n = 30). The specimens were investigated using immunofluorescence for the expression and distribution of PDGFRα+ cells in each group by light microscopy with computerized image analysis. Real-time PCR (RT-PCR) was used to study PDGFRα gene expression levels. In addition, small conductance calcium-activated potassium channel 3 (SK3) and closely associated cells consisting of smooth muscle cells (SMCs), interstitial cells of Cajal (ICCs), and nerve fibers were investigated. RESULTS: PDGFRα+ cells were in close proximity to SMCs, ICCs, and nerve fibers. PDGFRα+ cells expressed SK3 channels, which are found to regulate purinergic inhibitory neurotransmission in SMCs. Regarding the expression of PDGFRα+ cells no significant difference was seen between the two groups, while the expression of SK3 channels in PDGFRα+ cells was significantly decreased in the UPJO group versus the control group. CONCLUSION: This study identified the expression of PDGFRα+ cells in the human UPJ. Our results demonstrate the expression of SK3 channels in PDGFRα+ cells was decreased in UPJO, and SK3 channels may be involved in the pathogenesis of UPJO by perturbing the UPJ peristalsis.


Asunto(s)
Pelvis Renal/patología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/análisis , Uréter/patología , Obstrucción Ureteral/etiología , Niño , Preescolar , Constricción Patológica , Humanos , Lactante , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/fisiología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/fisiología
6.
J Neurosci ; 38(43): 9252-9262, 2018 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-30242046

RESUMEN

Hebbian synaptic plasticity at hippocampal Schaffer collateral synapses is tightly regulated by postsynaptic small conductance (SK) channels that restrict NMDA receptor activity. SK channels are themselves modulated by G-protein-coupled signaling pathways, but it is not clear under what conditions these are activated to enable synaptic plasticity. Here, we show that muscarinic M1 receptor (M1R) and type 1 metabotropic glutamate receptor (mGluR1) signaling pathways, which are known to inhibit SK channels and thereby disinhibit NMDA receptors, converge to facilitate spine calcium transients during the induction of long-term potentiation (LTP) at hippocampal Schaffer collateral synapses onto CA1 pyramidal neurons of male rats. Furthermore, mGluR1 activation is required for LTP induced by reactivated place-cell firing patterns that occur in sharp-wave ripple events during rest or sleep. In contrast, M1R activation is required for LTP induced by place-cell firing patterns during exploration. Thus, we describe a common mechanism that enables synaptic plasticity during both encoding and consolidation of memories within hippocampal circuits.SIGNIFICANCE STATEMENT Memory ensembles in the hippocampus are formed during active exploration and consolidated during rest or sleep. These two distinct phases each require strengthening of synaptic connections by long-term potentiation (LTP). The neuronal activity patterns in each phase are very different, which makes it hard to map generalized rules for LTP induction onto both formation and consolidation phases. In this study, we show that inhibition of postsynaptic SK channels is a common necessary feature of LTP induction and that SK channel inhibition is achieved by separate but convergent metabotropic signaling pathways. Thus, we reveal a common mechanism for enabling LTP under distinct behavioral conditions.


Asunto(s)
Hipocampo/fisiología , Plasticidad Neuronal/fisiología , Receptor Muscarínico M1/fisiología , Transducción de Señal/fisiología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/fisiología , Animales , Apamina/farmacología , Agonistas de Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Hipocampo/química , Hipocampo/efectos de los fármacos , Masculino , Microscopía de Fluorescencia por Excitación Multifotónica/métodos , Plasticidad Neuronal/efectos de los fármacos , Técnicas de Cultivo de Órganos , Ratas , Ratas Wistar , Receptor Muscarínico M1/agonistas , Transducción de Señal/efectos de los fármacos , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/antagonistas & inhibidores , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/química
7.
J Neurosci ; 37(44): 10738-10747, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28982705

RESUMEN

Reliable and precise signal transmission is essential in circuits of the auditory brainstem to encode timing with submillisecond accuracy. Globular bushy cells reliably and faithfully transfer spike signals to the principal neurons of the medial nucleus of the trapezoid body (MNTB) through the giant glutamatergic synapse, the calyx of Held. Thus, the MNTB works as a relay nucleus that preserves the temporal pattern of firing at high frequency. Using whole-cell patch-clamp recordings, we observed a K+ conductance mediated by small-conductance calcium-activated potassium (SK) channels in the MNTB neurons from rats of either sex. SK channels were activated by intracellular Ca2+ sparks and mediated spontaneous transient outward currents in developing MNTB neurons. SK channels were also activated by Ca2+ influx through voltage-gated Ca2+ channels and synaptically activated NMDA receptors. Blocking SK channels with apamin depolarized the resting membrane potential, reduced resting conductance, and affected the responsiveness of MNTB neurons to signal inputs. Moreover, SK channels were activated by action potentials and affected the spike afterhyperpolarization. Blocking SK channels disrupted the one-to-one signal transmission from presynaptic calyces to postsynaptic MNTB neurons and induced extra postsynaptic action potentials in response to presynaptic firing. These data reveal that SK channels play crucial roles in regulating the resting properties and maintaining reliable signal transmission of MNTB neurons.SIGNIFICANCE STATEMENT Reliable and precise signal transmission is required in auditory brainstem circuits to localize the sound source. The calyx of Held synapse in the mammalian medial nucleus of the trapezoid body (MNTB) plays an important role in sound localization. We investigated the potassium channels that shape the reliability of signal transfer across the calyceal synapse and observed a potassium conductance mediated by small-conductance calcium-activated potassium (SK) channels in rat MNTB principal neurons. We found that SK channels are tonically activated and contribute to the resting membrane properties of MNTB neurons. Interestingly, SK channels are transiently activated by calcium sparks and calcium influx during action potentials and control the one-to-one signal transmission from presynaptic calyces to postsynaptic MNTB neurons.


Asunto(s)
Potenciales de la Membrana/fisiología , Neuronas/fisiología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/fisiología , Cuerpo Trapezoide/fisiología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Apamina/farmacología , Tronco Encefálico/fisiología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Masculino , Potenciales de la Membrana/efectos de los fármacos , Neuronas/efectos de los fármacos , Técnicas de Cultivo de Órganos , Ratas , Ratas Wistar , Canales de Potasio de la Superfamilia Shaker/antagonistas & inhibidores , Canales de Potasio de la Superfamilia Shaker/farmacología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/antagonistas & inhibidores , Factores de Tiempo , Cuerpo Trapezoide/efectos de los fármacos
8.
Circ Res ; 118(7): 1078-90, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26838791

RESUMEN

RATIONALE: Recent data from mesenteric and cerebral beds have revealed spatially restricted Ca(2+) transients occurring along the vascular intima that control effector recruitment and vasodilation. Although Ca(2+) is pivotal for coronary artery endothelial function, spatial and temporal regulation of functional Ca(2+) signals in the coronary endothelium is poorly understood. OBJECTIVE: We aimed to determine whether a discrete spatial and temporal profile of Ca(2+) dynamics underlies endothelium-dependent relaxation of swine coronary arteries. METHODS AND RESULTS: Using confocal imaging, custom automated image analysis, and myography, we show that the swine coronary artery endothelium generates discrete basal Ca(2+) dynamics, including isolated transients and whole-cell propagating waves. These events are suppressed by depletion of internal stores or inhibition of inositol 1,4,5-trisphosphate receptors but not by inhibition of ryanodine receptors or removal of extracellular Ca(2+). In vessel rings, inhibition of specific Ca(2+)-dependent endothelial effectors, namely, small and intermediate conductance K(+) channels (K(Ca)3.1 and K(Ca)2.3) and endothelial nitric oxide synthase, produces additive tone, which is blunted by internal store depletion or inositol 1,4,5-trisphosphate receptor blockade. Stimulation of endothelial inositol 1,4,5-trisphosphate-dependent signaling with substance P causes idiosyncratic changes in dynamic Ca(2+) signal parameters (active sites, event frequency, amplitude, duration, and spatial spread). Overall, substance P-induced vasorelaxation corresponded poorly with whole-field endothelial Ca(2+) measurements but corresponded precisely with the concentration-dependent change in Ca(2+) dynamics (linearly translated composite of dynamic parameters). CONCLUSIONS: Our findings show that endothelium-dependent control of swine coronary artery tone is determined by spatial and temporal titration of inherent endothelial Ca(2+) dynamics that are not represented by tissue-level averaged Ca(2+) changes.


Asunto(s)
Señalización del Calcio , Circulación Coronaria/fisiología , Vasos Coronarios/metabolismo , Endotelio Vascular/metabolismo , Animales , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Circulación Coronaria/efectos de los fármacos , Femenino , Procesamiento de Imagen Asistido por Computador , Receptores de Inositol 1,4,5-Trifosfato/efectos de los fármacos , Receptores de Inositol 1,4,5-Trifosfato/fisiología , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/fisiología , Contracción Isométrica , Masculino , Microscopía Confocal , Modelos Cardiovasculares , Miografía , Óxido Nítrico Sintasa de Tipo III/fisiología , Péptidos , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/fisiología , Sustancia P/farmacología , Sus scrofa , Porcinos , Túnica Íntima/fisiología , Resistencia Vascular/efectos de los fármacos , Resistencia Vascular/fisiología
9.
Biochim Biophys Acta Bioenerg ; 1858(6): 442-458, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28342809

RESUMEN

We provide evidence for location and function of a small conductance, Ca2+-activated K+ (SKCa) channel isoform 3 (SK3) in mitochondria (m) of guinea pig, rat and human ventricular myocytes. SKCa agonists protected isolated hearts and mitochondria against ischemia/reperfusion (IR) injury; SKCa antagonists worsened IR injury. Intravenous infusion of a SKCa channel agonist/antagonist, respectively, in intact rats was effective in reducing/enhancing regional infarct size induced by coronary artery occlusion. Localization of SK3 in mitochondria was evidenced by Western blot of inner mitochondrial membrane, immunocytochemical staining of cardiomyocytes, and immunogold labeling of isolated mitochondria. We identified a SK3 splice variant in guinea pig (SK3.1, aka SK3a) and human ventricular cells (SK3.2) by amplifying mRNA, and show mitochondrial expression in mouse atrial tumor cells (HL-1) by transfection with full length and truncated SK3.1 protein. We found that the N-terminus is not required for mitochondrial trafficking but the C-terminus beyond the Ca2+ calmodulin binding domain is required for Ca2+ sensing to induce mK+ influx and/or promote mitochondrial localization. In isolated guinea pig mitochondria and in SK3 overexpressed HL-1 cells, mK+ influx was driven by adding CaCl2. Moreover, there was a greater fall in membrane potential (ΔΨm), and enhanced cell death with simulated cell injury after silencing SK3.1 with siRNA. Although SKCa channel opening protects the heart and mitochondria against IR injury, the mechanism for favorable bioenergetics effects resulting from SKCa channel opening remains unclear. SKCa channels could play an essential role in restraining cardiac mitochondria from inducing oxidative stress-induced injury resulting from mCa2+ overload.


Asunto(s)
Mitocondrias Cardíacas/metabolismo , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/fisiología , 1-Naftilamina/análogos & derivados , 1-Naftilamina/farmacología , Secuencia de Aminoácidos , Animales , Bencimidazoles/farmacología , Bencimidazoles/uso terapéutico , Cloruro de Calcio/farmacología , Hipoxia de la Célula , Línea Celular , Cobayas , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potencial de la Membrana Mitocondrial/fisiología , Ratones , Mitocondrias Cardíacas/química , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Potasio/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Isoformas de Proteínas/fisiología , Interferencia de ARN , ARN Mensajero/biosíntesis , Ratas , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/agonistas , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/antagonistas & inhibidores , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/aislamiento & purificación
10.
J Physiol ; 595(20): 6429-6442, 2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28714070

RESUMEN

KEY POINTS: Small conductance Ca2+ -activated K+ (SK) channels play an important role in regulating the excitability of magnocellular neurosecretory cells (MNCs). Although an increased SK channel function contributes to adaptive physiological responses, it remains unknown whether changes in SK channel function/expression contribute to exacerbated MNC activity under disease conditions. We show that the input-output function of MNCs in heart failure (HF) rats is enhanced. Moreover, the SK channel blocker apamin enhanced the input-output function in sham, although not in HF rats. We found that both the after-hyperpolarizing potential magnitude and the underlying apamin-sensitive IAHP are blunted in MNCs from HF rats. The magnitude of spike-induced increases in intracellular Ca2+ levels was not affected in MNCs of HF rats. We found a diminished expression of SK2/SK3 channel subunit mRNA expression in the supraoptic nucleus of HF rats. Our studies suggest that a reduction in SK channel expression, but not changes in Ca2+ -mediated activation of SK channels, contributes to exacerbated MNC activity in HF rats. ABSTRACT: Small conductance Ca2+ -activated K+ channels (SK) play an important role in regulating the activity of magnocellular neurosecretory cells (MNCs) and hormone release from the posterior pituitary. Moreover, enhanced SK activity contributes to the adaptive responses of MNCs to physiological challenge, such as lactation. Nevertheless, whether changes in SK function/expression contribute to exacerbated MNC activity during diseases such as heart failure (HF) remains unknown. In the present study, we used a combination of patch clamp electrophysiology, confocal Ca2+ imaging and molecular biology in a rat model of ischaemic HF. We found that the input-output function of MNCs was enhanced in HF compared to sham rats. Moreover, although the SK blocker apamin (200 nm) strengthened the input-output function in sham rats, it failed to have an effect in HF rats. The magnitude of the after-hyperpolarizing potential (AHP) following a train of spikes and the underlying apamin-sensitive IAHP were blunted in MNCs from HF rats. However, spike-induced increases in intracellular Ca2+ were not affected in the MNCs of HF rats. Real-time PCR measurements of SK channel subunits mRNA in supraoptic nucleus punches revealed a diminished expression of SK2/SK3 subunits in HF compared to sham rats. Together, our studies demonstrate that MNCs from HF rats exhibit increased membrane excitability and an enhanced input-output function, and also that a reduction in SK channel-mediated, apamin-sensitive AHP is a critical contributing mechanism. Moreover, our results suggest that the reduced AHP is related to a down-regulation of SK2/SK3 channel subunit expression but not the result of a blunted activity-dependent intracellular Ca2+ increase following a burst of action potentials.


Asunto(s)
Insuficiencia Cardíaca/fisiopatología , Hipotálamo/fisiología , Neuronas/fisiología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/fisiología , Animales , Masculino , Ratas Wistar , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/genética
11.
J Physiol ; 595(7): 2271-2284, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-27779751

RESUMEN

KEY POINTS: Ion channels are transmembrane proteins that are synthesized within the cells but need to be trafficked to the cell membrane for the channels to function. Small-conductance, Ca2+ -activated K+ channels (SK, KCa 2) are unique subclasses of K+ channels that are regulated by Ca2+ inside the cells; they are expressed in human atrial myocytes and responsible for shaping atrial action potentials. We have previously shown that interacting proteins of SK2 channels are important for channel trafficking to the membrane. Using total internal reflection fluorescence (TIRF) and confocal microscopy, we studied the mechanisms by which the surface membrane localization of SK2 (KCa 2.2) channels is regulated by their interacting proteins. Understanding the mechanisms of SK channel trafficking may provide new insights into the regulation controlling the repolarization of atrial myocytes. ABSTRACT: The normal function of ion channels depends critically on the precise subcellular localization and the number of channel proteins on the cell surface membrane. Small-conductance, Ca2+ -activated K+ channels (SK, KCa 2) are expressed in human atrial myocytes and are responsible for shaping atrial action potentials. Understanding the mechanisms of SK channel trafficking may provide new insights into the regulation controlling the repolarization of atrial myocytes. We have previously demonstrated that the C- and N-termini of SK2 channels interact with the actin-binding proteins α-actinin2 and filamin A, respectively. However, the roles of the interacting proteins on SK2 channel trafficking remain incompletely understood. Using total internal reflection fluorescence (TIRF) microscopy, we studied the mechanisms of surface membrane localization of SK2 (KCa 2.2) channels. When SK2 channels were co-expressed with filamin A or α-actinin2, the membrane fluorescence intensity of SK2 channels increased significantly. We next tested the effects of primaquine and dynasore on SK2 channels expression. Treatment with primaquine significantly reduced the membrane expression of SK2 channels. In contrast, treatment with dynasore failed to alter the surface membrane expression of SK2 channels. Further investigations using constitutively active or dominant-negative forms of Rab GTPases provided additional insights into the distinct roles of the two cytoskeletal proteins on the recycling processes of SK2 channels from endosomes. α-Actinin2 facilitated recycling of SK2 channels from both early and recycling endosomes while filamin A probably aids the recycling of SK2 channels from recycling endosomes.


Asunto(s)
Actinina/fisiología , Filaminas/fisiología , Miocitos Cardíacos/fisiología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/fisiología , Animales , Membrana Celular/efectos de los fármacos , Membrana Celular/fisiología , Endosomas/metabolismo , Células HEK293 , Atrios Cardíacos/citología , Humanos , Hidrazonas/farmacología , Masculino , Ratones Endogámicos C57BL , Miocitos Cardíacos/efectos de los fármacos , Primaquina/farmacología
12.
Physiol Rev ; 90(4): 1437-59, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20959620

RESUMEN

Molecular research on ion channels has demonstrated that many of these integral membrane proteins associate with partner proteins, often versatile in their function, or even assemble into stable macromolecular complexes that ensure specificity and proper rate of the channel-mediated signal transduction. Calcium-activated potassium (K(Ca)) channels that link excitability and intracellular calcium concentration are responsible for a wide variety of cellular processes ranging from regulation of smooth muscle tone to modulation of neurotransmission and control of neuronal firing pattern. Most of these functions are brought about by interaction of the channels' pore-forming subunits with distinct partner proteins. In this review we summarize recent insights into protein complexes associated with K(Ca) channels as revealed by proteomic research and discuss the results available on structure and function of these complexes and on the underlying protein-protein interactions. Finally, the results are related to their significance for the function of K(Ca) channels under cellular conditions.


Asunto(s)
Complejos Multiproteicos/fisiología , Canales de Potasio Calcio-Activados/fisiología , Animales , Regulación de la Expresión Génica/fisiología , Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Modelos Moleculares , Conformación Proteica , Transporte de Proteínas , Proteómica , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/fisiología
13.
Neural Plast ; 2017: 7282834, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29362678

RESUMEN

Evidence indicates that high salt (HS) intake activates presympathetic paraventricular nucleus (PVN) neurons, which contributes to sympathoexcitation of salt-sensitive hypertension. The present study determined whether 5 weeks of HS (2% NaCl) intake alters the small conductance Ca2+-activated potassium channel (SK) current in presympathetic PVN neurons and whether this change affects the neuronal excitability. In whole-cell voltage-clamp recordings, HS-treated rats had significantly decreased SK currents compared to rats with normal salt (NS, 0.4% NaCl) intake in PVN neurons. The sensitivity of PVN neuronal excitability in response to current injections was greater in HS group compared to NS controls. The SK channel blocker apamin augmented the neuronal excitability in both groups but had less effect on the sensitivity of the neuronal excitability in HS group compared to NS controls. In the HS group, the interspike interval (ISI) was significantly shorter than that in NS controls. Apamin significantly shortened the ISI in NS controls but had less effect in the HS group. This data suggests that HS intake reduces SK currents, which contributes to increased PVN neuronal excitability at least in part through a decrease in spike frequency adaptation and may be a precursor to the development of salt-sensitive hypertension.


Asunto(s)
Bulbo Raquídeo/fisiología , Potenciales de la Membrana , Neuronas/fisiología , Núcleo Hipotalámico Paraventricular/fisiología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/fisiología , Cloruro de Sodio/administración & dosificación , Animales , Apamina/administración & dosificación , Masculino , Bulbo Raquídeo/efectos de los fármacos , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/fisiología , Neuronas/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Ratas Sprague-Dawley , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/antagonistas & inhibidores
14.
J Neurosci ; 35(37): 12779-91, 2015 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-26377466

RESUMEN

Attention deficits in Alzheimer's disease can exacerbate its other cognitive symptoms, yet relevant disruptions of key prefrontal circuitry are not well understood. Here, in the TgCRND8 mouse model of this neurological disorder, we demonstrate and characterize a disruption of cholinergic excitation in the major corticothalamic layer of the prefrontal cortex, in which modulation by acetylcholine is essential for optimal attentional function. Using electrophysiology with concurrent multiphoton imaging, we show that layer 6 pyramidal cells are unable to sustain cholinergic excitation to the same extent as their nontransgenic littermate controls, as a result of the excessive activation of calcium-activated hyperpolarizing conductances. We report that cholinergic excitation can be improved in TgCRND8 cortex by pharmacological blockade of SK channels, suggesting a novel target for the treatment of cognitive dysfunction in Alzheimer's disease. SIGNIFICANCE STATEMENT: Alzheimer's disease is accompanied by attention deficits that exacerbate its other cognitive symptoms. In brain slices of a mouse model of this neurological disorder, we demonstrate, characterize, and rescue impaired cholinergic excitation of neurons essential for optimal attentional performance. In particular, we show that the excessive activation of a calcium-activated potassium conductance disrupts the acetylcholine excitation of prefrontal layer 6 pyramidal neurons and that its blockade normalizes responses. These findings point to a novel potential target for the treatment of cognitive dysfunction in Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Atención/fisiología , Señalización del Calcio/fisiología , Neuronas Colinérgicas/fisiología , Proteínas del Tejido Nervioso/fisiología , Corteza Prefrontal/fisiopatología , Células Piramidales/fisiología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/fisiología , Transmisión Sináptica/fisiología , Acetilcolina/farmacología , Acetilcolina/fisiología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animales , Apamina/farmacología , Atropina/farmacología , Atención/efectos de los fármacos , Señalización del Calcio/efectos de los fármacos , Neuronas Colinérgicas/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Genotipo , Activación del Canal Iónico/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Proteínas del Tejido Nervioso/efectos de los fármacos , Técnicas de Placa-Clamp , Células Piramidales/efectos de los fármacos , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos
15.
Annu Rev Physiol ; 74: 245-69, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-21942705

RESUMEN

Small-conductance Ca(2+)-activated K(+) channels (SK channels) are widely expressed throughout the central nervous system. These channels are activated solely by increases in intracellular Ca(2+). SK channels are stable macromolecular complexes of the ion pore-forming subunits with calmodulin, which serves as the intrinsic Ca(2+) gating subunit, as well as with protein kinase CK2 and protein phosphatase 2A, which modulate Ca(2+) sensitivity. Well-known for their roles in regulating somatic excitability in central neurons, SK channels are also expressed in the postsynaptic membrane of glutamatergic synapses, where their activation and regulated trafficking modulate synaptic transmission and the induction and expression of synaptic plasticity, thereby affecting learning and memory. In this review we discuss the molecular and functional properties of SK channels and their physiological roles in central neurons.


Asunto(s)
Canales de Potasio de Pequeña Conductancia Activados por el Calcio/fisiología , Animales , Calcio/metabolismo , Calcio/fisiología , Humanos , Activación del Canal Iónico/fisiología , Aprendizaje/fisiología , Memoria/fisiología , Plasticidad Neuronal/fisiología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/efectos de los fármacos , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/genética , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/ultraestructura , Transmisión Sináptica/fisiología
16.
Mol Pharmacol ; 87(2): 338-48, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25468883

RESUMEN

Small/intermediate conductance KCa channels (KCa2/3) are Ca(2+)/calmodulin regulated K(+) channels that produce membrane hyperpolarization and shape neurologic, epithelial, cardiovascular, and immunologic functions. Moreover, they emerged as therapeutic targets to treat cardiovascular disease, chronic inflammation, and some cancers. Here, we aimed to generate a new pharmacophore for negative-gating modulation of KCa2/3 channels. We synthesized a series of mono- and dibenzoates and identified three dibenzoates [1,3-phenylenebis(methylene) bis(3-fluoro-4-hydroxybenzoate) (RA-2), 1,2-phenylenebis(methylene) bis(3-fluoro-4-hydroxybenzoate), and 1,4-phenylenebis(methylene) bis(3-fluoro-4-hydroxybenzoate)] with inhibitory efficacy as determined by patch clamp. Among them, RA-2 was the most drug-like and inhibited human KCa3.1 with an IC50 of 17 nM and all three human KCa2 subtypes with similar potencies. RA-2 at 100 nM right-shifted the KCa3.1 concentration-response curve for Ca(2+) activation. The positive-gating modulator naphtho[1,2-d]thiazol-2-ylamine (SKA-31) reversed channel inhibition at nanomolar RA-2 concentrations. RA-2 had no considerable blocking effects on distantly related large-conductance KCa1.1, Kv1.2/1.3, Kv7.4, hERG, or inwardly rectifying K(+) channels. In isometric myography on porcine coronary arteries, RA-2 inhibited bradykinin-induced endothelium-derived hyperpolarization (EDH)-type relaxation in U46619-precontracted rings. Blood pressure telemetry in mice showed that intraperitoneal application of RA-2 (≤100 mg/kg) did not increase blood pressure or cause gross behavioral deficits. However, RA-2 decreased heart rate by ≈145 beats per minute, which was not seen in KCa3.1(-/-) mice. In conclusion, we identified the KCa2/3-negative-gating modulator, RA-2, as a new pharmacophore with nanomolar potency. RA-2 may be of use to generate structurally new types of negative-gating modulators that could help to define the physiologic and pathomechanistic roles of KCa2/3 in the vasculature, central nervous system, and during inflammation in vivo.


Asunto(s)
Bradicardia/inducido químicamente , Vasos Coronarios/efectos de los fármacos , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/antagonistas & inhibidores , Vasodilatación/efectos de los fármacos , Animales , Benzoatos/química , Benzoatos/farmacología , Bradicardia/fisiopatología , Vasos Coronarios/fisiología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiología , Femenino , Células HEK293 , Frecuencia Cardíaca/efectos de los fármacos , Frecuencia Cardíaca/fisiología , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/antagonistas & inhibidores , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/fisiología , Activación del Canal Iónico , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Técnicas de Cultivo de Órganos , Bloqueadores de los Canales de Potasio/química , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/fisiología , Vasodilatación/fisiología
17.
Circulation ; 129(4): 430-40, 2014 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-24190961

RESUMEN

BACKGROUND: Recent evidence points to functional Ca²âº-dependent K⁺ (SK) channels in the heart that may govern atrial fibrillation (AF) risk, but the underlying mechanisms are unclear. This study addressed the role of SK channels in atrial repolarization and AF persistence in a canine AF model. METHODS AND RESULTS: Electrophysiological variables were assessed in dogs subjected to atrial remodeling by 7-day atrial tachypacing (AT-P), as well as controls. Ionic currents and single-channel properties were measured in isolated canine atrial cardiomyocytes by patch clamp. NS8593, a putative selective SK blocker, suppressed SK current with an IC50 of ≈5 µmol/L, without affecting Na⁺, Ca²âº, or other K⁺ currents. Whole-cell SK current sensitive to NS8593 was significantly larger in pulmonary vein (PV) versus left atrial (LA) cells, without a difference in SK single-channel open probability (P(o)), whereas AT-P enhanced both whole-cell SK currents and single-channel P(o). SK-current block increased action potential duration in both PV and LA cells after AT-P; but only in PV cells in absence of AT-P. SK2 expression was more abundant at both mRNA and protein levels for PV versus LA in control dogs, in both control and AT-P; AT-P upregulated only SK1 at the protein level. Intravenous administration of NS8593 (5 mg/kg) significantly prolonged atrial refractoriness and reduced AF duration without affecting the Wenckebach cycle length, left ventricular refractoriness, or blood pressure. CONCLUSIONS: SK currents play a role in canine atrial repolarization, are larger in PVs than LA, are enhanced by atrial-tachycardia remodeling, and appear to participate in promoting AF maintenance. These results are relevant to the potential mechanisms underlying the association between SK single-nucleotide polymorphisms and AF and suggest SK blockers as potentially interesting anti-AF drugs.


Asunto(s)
Fibrilación Atrial/patología , Fibrilación Atrial/fisiopatología , Fenómenos Electrofisiológicos/fisiología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/fisiología , 1-Naftilamina/análogos & derivados , 1-Naftilamina/farmacología , Animales , Modelos Animales de Enfermedad , Perros , Técnicas Electrofisiológicas Cardíacas , Atrios Cardíacos/efectos de los fármacos , Atrios Cardíacos/patología , Miocitos Cardíacos/patología , Miocitos Cardíacos/fisiología , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/farmacología , Venas Pulmonares/efectos de los fármacos , Venas Pulmonares/patología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/efectos de los fármacos
18.
J Neurophysiol ; 114(1): 1-5, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25185803

RESUMEN

Voltage-dependent calcium channels (VDCCs) couple neuronal activity to diverse intracellular signals with exquisite spatiotemporal specificity. Using calcium imaging and electrophysiology, Jones and Stuart (J Neurosci 33: 19396-19405, 2013) examined the intimate relationship between distinct types of VDCCs and small-conductance calcium-activated potassium (SK) channels that contribute to the compartmentalized control of excitability in the soma and dendrites of cortical pyramidal neurons. Here we discuss the importance of calcium domains for signal specificity, explore the possible functions and mechanisms for local control of SK channels, and highlight technical considerations for the optical detection of calcium signals.


Asunto(s)
Potenciales de Acción/fisiología , Calcio/metabolismo , Dendritas/fisiología , Células Piramidales/fisiología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/fisiología , Animales
19.
IUBMB Life ; 67(5): 331-7, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25988524

RESUMEN

The family of p90 ribosomal S6 kinases (RSKs) are pleiotropic effectors for extracellular signal-regulated kinase signaling pathways. Recently, RSK3 was shown to be important for pathological remodeling of the heart. Although cardiac myocyte hypertrophy can be compensatory for increased wall stress, in chronic heart diseases, this nonmitotic cell growth is usually associated with interstitial fibrosis, increased cell death, and decreased cardiac function. Although RSK3 is less abundant in the cardiac myocyte than other RSK family members, RSK3 appears to serve a unique role in cardiac myocyte stress responses. A potential mechanism conferring the unique function of RSK3 in the heart is anchoring by the scaffold protein muscle A-kinase anchoring protein ß (mAKAPß). Recent findings suggest that RSK3 should be considered as a therapeutic target for the prevention of heart failure, a clinical syndrome of major public health significance.


Asunto(s)
Miocitos Cardíacos/patología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/fisiología , Animales , Humanos , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Estrés Fisiológico
20.
J Cardiovasc Pharmacol ; 66(1): 118-27, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25815673

RESUMEN

Endothelial KCa2.3 and KCa3.1 channels contribute to the regulation of myogenic tone in resistance arteries by Ca(2+)-mobilizing vasodilatory hormones. To define further the functional role of these channels in distinct vascular beds, we have examined the vasodilatory actions of the KCa channel activator SKA-31 in myogenically active rat cremaster and middle cerebral arteries. Vessels pressurized to 70 mm Hg constricted by 80-100 µm (ie, 25%-45% of maximal diameter). SKA-31 (10 µM) inhibited myogenic tone by 80% in cremaster and ∼65% in middle cerebral arteries, with IC50 values of ∼2 µM in both vessels. These vasodilatory effects were largely prevented by the KCa2.3 blocker UCL1684 and the KCa3.1 blocker TRAM-34 and abolished by endothelial denudation. Preincubation with N(G) nitro L-arginine methyl ester (L-NAME, 0.1 mM) did not affect the inhibitory response to SKA-31, but attenuated the ACh-evoked dilation by ∼45%. Penitrem-A, a blocker of BK(Ca) channels, did not alter SKA-31 evoked vasodilation but did reduce the inhibition of myogenic tone by ACh, the BKCa channel activator NS1619, and sodium nitroprusside. Collectively, these data demonstrate that SKA-31 produces robust inhibition of myogenic tone in resistance arteries isolated from distinct vascular beds in an endothelium-dependent manner.


Asunto(s)
Benzotiazoles/farmacología , Arterias Cerebrales/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/agonistas , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/agonistas , Vasodilatación/efectos de los fármacos , Animales , Arterias Cerebrales/fisiología , Endotelio Vascular/fisiología , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/fisiología , Masculino , Ratas , Ratas Sprague-Dawley , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/fisiología , Vasodilatación/fisiología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda