Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Neurosci Res ; 167: 3-10, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33872635

RESUMEN

Experience-dependent plasticity within visual cortex is controlled by postnatal maturation of inhibitory circuits, which are both morphologically diverse and precisely connected. Gene-targeted disruption of the voltage-dependent potassium channel Kv3.1 broadens action potentials and reduces net inhibitory function of parvalbumin (PV)-positive GABA subtypes within the neocortex. In mice lacking Kv3.1, the rate of input loss from an eye deprived of vision was slowed two-fold, despite otherwise normal critical period timecourse and receptive field properties. Rapid ocular dominance plasticity was restored by local or systemic enhancement of GABAergic transmission with acute benzodiazepine infusion. Diazepam instead exacerbated a global suppression of slow-wave oscillations during sleep described previously in these mutant mice, which therefore did not account for the rescued plasticity. Rapid ocular dominance shifts closely reflected Kv3.1 gene dosage that prevented prolonged spike discharge of their target pyramidal cells in vivo or the spike amplitude decrement of fast-spiking cells during bouts of high-frequency firing in vitro. Late postnatal expression of this unique channel in fast-spiking interneurons thus subtly regulates the speed of critical period plasticity with implications for mental illnesses.


Asunto(s)
Neocórtex , Canales de Potasio Shaw , Animales , Período Crítico Psicológico , Interneuronas/metabolismo , Ratones , Neocórtex/metabolismo , Plasticidad Neuronal , Parvalbúminas/metabolismo , Canales de Potasio Shaw/genética , Canales de Potasio Shaw/metabolismo
2.
J Neurosci ; 29(50): 15735-44, 2009 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-20016089

RESUMEN

The role of cerebellar Kv3.1 and Kv3.3 channels in motor coordination was examined with an emphasis on the deep cerebellar nuclei (DCN). Kv3 channel subunits encoded by Kcnc genes are distinguished by rapid activation and deactivation kinetics that support high-frequency, narrow action potential firing. Previously we reported that increased lateral deviation while ambulating and slips while traversing a narrow beam of ataxic Kcnc3-null mice were corrected by restoration of Kv3.3 channels specifically to Purkinje cells, whereas Kcnc3-mutant mice additionally lacking one Kcnc1 allele were partially rescued. Here, we report mice lacking all Kcnc1 and Kcnc3 alleles exhibit no such rescue. For Purkinje cell output to reach the rest of the brain it must be conveyed by neurons of the DCN or vestibular nuclei. As Kcnc1, but not Kcnc3, alleles are lost, mutant mice exhibit increasing gait ataxia accompanied by spike broadening and deceleration in DCN neurons, suggesting the facet of coordination rescued by Purkinje-cell-restricted Kv3.3 restoration in mice lacking just Kcnc3 is hypermetria, while gait ataxia emerges when additionally Kcnc1 alleles are lost. Thus, fast repolarization in Purkinje cells appears important for normal movement velocity, whereas DCN neurons are a prime candidate locus where fast repolarization is necessary for normal gait patterning.


Asunto(s)
Marcación de Gen , Desempeño Psicomotor/fisiología , Células de Purkinje/fisiología , Canales de Potasio Shaw/deficiencia , Canales de Potasio Shaw/genética , Canales de Potasio Shaw/fisiología , Potenciales de Acción/genética , Alelos , Animales , Núcleos Cerebelosos/metabolismo , Núcleos Cerebelosos/patología , Femenino , Ataxia de la Marcha/genética , Ataxia de la Marcha/patología , Masculino , Ratones , Ratones Noqueados , Células de Purkinje/citología , Células de Purkinje/patología , Canales de Potasio Shaw/biosíntesis
3.
J Neurosci ; 28(18): 4640-8, 2008 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-18448641

RESUMEN

The fast-activating/deactivating voltage-gated potassium channel Kv3.3 (Kcnc3) is expressed in various neuronal cell types involved in motor function, including cerebellar Purkinje cells. Spinocerebellar ataxia type 13 (SCA13) patients carrying dominant-negative mutations in Kcnc3 and Kcnc3-null mutant mice both display motor incoordination, suggested in mice by increased lateral deviation while ambulating and slips on a narrow beam. Motor skill learning, however, is spared. Mice lacking Kcnc3 also exhibit muscle twitches. In addition to broadened spikes, recordings of Kcnc3-null Purkinje cells revealed fewer spikelets in complex spikes and a lower intraburst frequency. Targeted reexpression of Kv3.3 channels exclusively in Purkinje cells in Kcnc3-null mice as well as in mice also heterozygous for Kv3.1 sufficed to restore simple spike brevity along with normal complex spikes and to rescue specifically coordination. Therefore, spike parameters requiring Kv3.3 function in Purkinje cells are involved in the ataxic null phenotype and motor coordination, but not motor learning.


Asunto(s)
Potenciales de Acción/fisiología , Destreza Motora/fisiología , Mutación/fisiología , Equilibrio Postural/fisiología , Células de Purkinje/fisiología , Canales de Potasio Shaw/fisiología , Potenciales de Acción/genética , Potenciales de Acción/efectos de la radiación , Análisis de Varianza , Animales , Conducta Animal , Cerebelo/citología , Relación Dosis-Respuesta en la Radiación , Estimulación Eléctrica/métodos , Femenino , Expresión Génica/fisiología , Proteínas Fluorescentes Verdes/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Técnicas de Placa-Clamp , Canales de Potasio Shaw/deficiencia
4.
J Neurosci ; 28(21): 5570-81, 2008 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-18495891

RESUMEN

The genes Kcnc1 and Kcnc3 encode the subunits for the fast-activating/fast-deactivating, voltage-gated potassium channels Kv3.1 and Kv3.3, which are expressed in several brain regions known to be involved in the regulation of the sleep-wake cycle. When these genes are genetically eliminated, Kv3.1/Kv3.3-deficient mice display severe sleep loss as a result of unstable slow-wave sleep. Within the thalamocortical circuitry, Kv3.1 and Kv3.3 subunits are highly expressed in the thalamic reticular nucleus (TRN), which is thought to act as a pacemaker at sleep onset and to be involved in slow oscillatory activity (spindle waves) during slow-wave sleep. We showed that in cortical electroencephalographic recordings of freely moving Kv3.1/Kv3.3-deficient mice, spectral power is reduced up to 70% at frequencies <15 Hz. In addition, the number of sleep spindles in vivo as well as rhythmic rebound firing of TRN neurons in vitro is diminished in mutant mice. Kv3.1/Kv3.3-deficient TRN neurons studied in vitro show approximately 60% increase in action potential duration and a reduction in high-frequency firing after depolarizing current injections and during rebound burst firing. The results support the hypothesis that altered electrophysiological properties of TRN neurons contribute to the reduced EEG power at slow frequencies in the thalamocortical network of Kv3-deficient mice.


Asunto(s)
Relojes Biológicos/fisiología , Corteza Cerebral/fisiopatología , Canales de Potasio Shaw/deficiencia , Núcleos Talámicos/fisiología , Acetilcolina/metabolismo , Análisis de Varianza , Animales , Monoaminas Biogénicas/metabolismo , Electroencefalografía , Electromiografía , Análisis de Fourier , Técnicas In Vitro , Ratones , Ratones Noqueados , Vías Nerviosas/fisiología , Polisomnografía , Privación de Sueño
5.
Cerebellum ; 8(3): 323-33, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19247732

RESUMEN

Different subunits of the Kv3 subfamily of voltage-gated potassium (Kv) channels (Kv3.1-Kv3.4) are expressed in distinct neuronal subpopulations in the cerebellum. Behavioral phenotypes in Kv3-null mutant mice such as ataxia with prominent hypermetria and heightened alcohol sensitivity are characteristic of cerebellar dysfunction. Here, we review how the unique biophysical properties of Kv3-type potassium channels, fast activation and fast deactivation that enable cerebellar neurons to generate brief action potentials at high frequencies, affect firing patterns and influence cerebellum-mediated behavior.


Asunto(s)
Conducta/fisiología , Cerebelo/fisiología , Canales de Potasio Shaw/metabolismo , Potenciales de Acción/genética , Potenciales de Acción/fisiología , Animales , Cerebelo/citología , Expresión Génica/fisiología , Humanos , Ratones , Ratones Noqueados , Modelos Biológicos , Trastornos del Movimiento/genética , Trastornos del Movimiento/patología , Neuronas/fisiología , Subunidades de Proteína/fisiología , Desempeño Psicomotor/fisiología , Canales de Potasio Shaw/genética
6.
J Neurosci ; 25(21): 5230-5, 2005 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-15917463

RESUMEN

Potassium (K+) channel subunits of the Kv3 subfamily (Kv3.1-Kv3.4) display a positively shifted voltage dependence of activation and fast activation/deactivation kinetics when compared with other voltage-gated K+ channels, features that confer on Kv3 channels the ability to accelerate the repolarization of the action potential (AP) efficiently and specifically. In the cortex, the Kv3.1 and Kv3.2 proteins are expressed prominently in a subset of GABAergic interneurons known as fast-spiking (FS) cells and in fact are a significant determinant of the fast-spiking discharge pattern. However, in addition to expression at FS cell somata, Kv3.1 and Kv3.2 proteins also are expressed prominently at FS cell terminals, suggesting roles for Kv3 channels in neurotransmitter release. We investigated the effect of 1.0 mM tetraethylammonium (TEA; which blocks Kv3 channels) on inhibitory synaptic currents recorded in layer II/III neocortical pyramidal cells. Spike-evoked GABA release by FS cells was enhanced nearly twofold by 1.0 mM TEA, with a decrease in the paired pulse ratio (PPR), effects not reproduced by blockade of the non-Kv3 subfamily K+ channels also blocked by low concentrations of TEA. Moreover, in Kv3.1/Kv3.2 double knock-out (DKO) mice, the large effects of TEA were absent, spike-evoked GABA release was larger, and the PPR was lower than in wild-type mice. Together, these results suggest specific roles for Kv3 channels at FS cell terminals that are distinct from those of Kv1 and large-conductance Ca2+-activated K+ channels (also present at the FS cell synapse). We propose that at FS cell terminals synaptically localized Kv3 channels keep APs brief, limiting Ca2+ influx and hence release probability, thereby influencing synaptic depression at a synapse designed for sustained high-frequency synaptic transmission.


Asunto(s)
Neocórtex/citología , Neuronas/metabolismo , Canales de Potasio/fisiología , Sinapsis/fisiología , Transmisión Sináptica/fisiología , Ácido gamma-Aminobutírico/metabolismo , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Potenciales de Acción/efectos de la radiación , Aminopiridinas/farmacología , Animales , Animales Recién Nacidos , Calcio/metabolismo , Diagnóstico por Imagen/métodos , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Esquema de Medicación , Interacciones Farmacológicas , Estimulación Eléctrica/métodos , Técnicas In Vitro , Ratones , Inhibición Neural/efectos de los fármacos , Inhibición Neural/fisiología , Inhibición Neural/efectos de la radiación , Neuronas/citología , Técnicas de Placa-Clamp/métodos , Péptidos/farmacología , Fotones , Bloqueadores de los Canales de Potasio/farmacología , Sinapsis/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Tetraetilamonio/farmacología , Ácido gamma-Aminobutírico/farmacología
7.
J Neurosci ; 23(20): 7677-84, 2003 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-12930807

RESUMEN

Micelacking both Kv3.1 and both Kv3.3 K+ channel alleles display severe motor deficits such as tremor, myoclonus, and ataxic gait. Micelacking one to three alleles at the Kv3.1 and Kv3.3 loci exhibit in an allele dose-dependent manner a modest degree of ataxia. Cerebellar granule cells coexpress Kv3.1 and Kv3.3 K+ channels and are therefore candidate neurons that might be involved in these behavioral deficits. Hence, we investigated the synaptic mechanisms of transmission in the parallel fiber-Purkinje cell system. Action potentials of parallel fibers were broader in mice lacking both Kv3.1 and both Kv3.3 alleles and in mice lacking both Kv3.1 and a single Kv3.3 allele compared with those of wild-type mice. The transmission of high-frequency trains of action potentials was only impaired at 200 Hz but not at 100 Hz in mice lacking both Kv3.1 and Kv3.3 genes. However, paired-pulse facilitation (PPF) at parallel fiber-Purkinje cell synapses was dramatically reduced in a gene dose-dependent manner in mice lacking Kv3.1 or Kv3.3 alleles. Normal PPF could be restored by reducing the extracellular Ca2+ concentration indicating that increased activity-dependent presynaptic Ca2+ influx, at least in part caused the altered PPF in mutant mice. Induction of metabotropic glutamate receptor-mediated EPSCs was facilitated, whereas longterm depression was not impaired but rather facilitated in Kv3.1/Kv3.3 double-knockout mice. These results demonstrate the importance of Kv3 potassium channels in regulating the dynamics of synaptic transmission at the parallel fiber-Purkinje cell synapse and suggest a correlation between short-term plasticity at the parallel fiber-Purkinje cell synapse and motor performance.


Asunto(s)
Corteza Cerebelosa/fisiopatología , Discinesias/etiología , Neuropéptidos/fisiología , Canales de Potasio con Entrada de Voltaje , Canales de Potasio/fisiología , Células de Purkinje/fisiología , Transmisión Sináptica , Potenciales de Acción , Animales , Ataxia/etiología , Conducta Animal , Calcio/farmacología , Células Cultivadas , Discinesias/fisiopatología , Femenino , Depresión Sináptica a Largo Plazo , Masculino , Ratones , Ratones Noqueados , Fibras Nerviosas/fisiología , Plasticidad Neuronal , Neuropéptidos/genética , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio/genética , Receptores de Glutamato Metabotrópico/metabolismo , Canales de Potasio Shaw , Sinapsis/fisiología
8.
J Neurosci ; 23(4): 1133-41, 2003 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-12598601

RESUMEN

Neurons of the medial nucleus of the trapezoid body, which transmit auditory information that is used to compute the location of sounds in space, are capable of firing at high frequencies with great temporal precision. We found that elimination of the Kv3.1 gene in mice results in the loss of a high-threshold component of potassium current and failure of the neurons to follow high-frequency stimulation. A partial decrease in Kv3.1 current can be produced in wild-type neurons of the medial nucleus of the trapezoid body by activation of protein kinase C. Paradoxically, activation of protein kinase C increases temporal fidelity and the number of action potentials that are evoked by intermediate frequencies of stimulation. Computer simulations confirm that a partial decrease in Kv3.1 current is sufficient to increase the accuracy of response at intermediate frequencies while impairing responses at high frequencies. We further establish that, of the two isoforms of the Kv3.1 potassium channel that are expressed in these neurons, Kv3.1a and Kv3.1b, the decrease in Kv3.1 current is mediated by selective phosphorylation of the Kv3.1b isoform. Using site-directed mutagenesis, we identify a specific C-terminal phosphorylation site responsible for the observed difference in response of the two isoforms to protein kinase C activation. Our results suggest that modulation of Kv3.1 by phosphorylation allows auditory neurons to tune their responses to different patterns of sensory stimulation.


Asunto(s)
Potenciales de Acción , Tronco Encefálico/fisiología , Potenciales Evocados Auditivos , Neuronas/fisiología , Neuropéptidos/metabolismo , Canales de Potasio con Entrada de Voltaje , Canales de Potasio/metabolismo , Animales , Tronco Encefálico/citología , Células CHO , Células Cultivadas , Cricetinae , Conductividad Eléctrica , Cinética , Ratones , Ratones Noqueados , Neuronas/metabolismo , Neuropéptidos/genética , Neuropéptidos/fisiología , Técnicas de Placa-Clamp , Fosforilación , Canales de Potasio/genética , Canales de Potasio/fisiología , Isoformas de Proteínas/metabolismo , Proteína Quinasa C/metabolismo , Serina/metabolismo , Canales de Potasio Shaw , Acetato de Tetradecanoilforbol/farmacología
9.
J Neurosci ; 24(33): 7335-43, 2004 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-15317859

RESUMEN

Direction-selective retinal ganglion cells show an increased activity evoked by light stimuli moving in the preferred direction. This selectivity is governed by direction-selective inhibition from starburst amacrine cells occurring during stimulus movement in the opposite or null direction. To understand the intrinsic membrane properties of starburst cells responsible for direction-selective GABA release, we performed whole-cell recordings from starburst cells in mouse retina. Voltage-clamp recordings revealed prominent voltage-dependent K(+) currents. The currents were mostly blocked by 1 mm TEA, activated rapidly at voltages more positive than -20 mV, and deactivated quickly, properties reminiscent of the currents carried by the Kv3 subfamily of K+ channels. Immunoblots confirmed the presence of Kv3.1 and Kv3.2 proteins in retina and immunohistochemistry revealed their expression in starburst cell somata and dendrites. The Kv3-like current in starburst cells was absent in Kv3.1-Kv3.2 knock-out mice. Current-clamp recordings showed that the fast activation of the Kv3 channels provides a voltage-dependent shunt that limits depolarization of the soma to potentials more positive than -20 mV. This provides a mechanism likely to contribute to the electrical isolation of individual starburst cell dendrites, a property thought essential for direction selectivity. This function of Kv3 channels differs from that in other neurons where they facilitate high-frequency repetitive firing. Moreover, we found a gradient in the intensity of Kv3.1b immunolabeling favoring proximal regions of starburst cells. We hypothesize that this Kv3 channel gradient contributes to the preference for centrifugal signal flow in dendrites underlying direction-selective GABA release from starburst amacrine cells


Asunto(s)
Células Amacrinas/fisiología , Neuropéptidos/fisiología , Canales de Potasio con Entrada de Voltaje , Canales de Potasio/fisiología , Células Amacrinas/metabolismo , Animales , Conductividad Eléctrica , Ratones , Ratones Noqueados , Neuropéptidos/análisis , Neuropéptidos/genética , Técnicas de Placa-Clamp , Canales de Potasio/análisis , Canales de Potasio/genética , Retina/química , Retina/metabolismo , Canales de Potasio Shaw , Transmisión Sináptica
10.
Eur J Neurosci ; 23(6): 1567-74, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16553620

RESUMEN

Sleep-wake behavior is tightly controlled in many animal species, suggesting genetically encoded, homeostatic control mechanisms that determine arousal-state dynamics. We reported that two voltage-gated potassium channels, Kv3.1 and Kv3.3, control sleep in wild-type and Kv3-mutant mice. Compared with wild-type (WT), homozygous double mutants (DKO) that lack these channels sleep 40% less in the light and 22% less in the dark. To understand how the lack of these channels affects sleep, we analysed arousal-state changes during the light period where the differences are greatest between WT and DKO. We determined the kinetic complexity of each arousal state from the episode durations of wakefulness, slow-wave sleep and rapid eye movement sleep (REMS). Based on the number of exponential components in episode-duration histograms, WT and DKO mice have several kinetically distinct states of wakefulness, and these states are longer in duration in DKO. For slow-wave sleep, WT mice have a single slow-wave sleep (SWS) state in contrast to DKO mice, which show two distinct SWS states, one that is 60% shorter than that in WT and a second that is similar in duration. Both WT and DKO mice have two kinetically distinct REMS states. DKO mice show an 84% reduction in the frequency of short REMS episodes (<45 s) without any change in the occurrence of long REMS episodes (>60 s). In contrast to the stochastic control of episode durations of wakefulness and SWS, the durations of both REMS states are normally distributed, indicating that the underlying control processes are fundamentally different.


Asunto(s)
Nivel de Alerta/fisiología , Periodicidad , Canales de Potasio Shaw/fisiología , Animales , Cinética , Luz , Ratones , Ratones Noqueados , Polisomnografía , Canales de Potasio Shaw/genética , Sueño/fisiología , Sueño REM/fisiología , Procesos Estocásticos , Vigilia/fisiología
11.
Mol Pharmacol ; 67(2): 480-8, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15525758

RESUMEN

Kv2.1, the prevalent delayed-rectifier K(+) channel in neuroendocrine and endocrine cells, was suggested previously by our group to be modulated in islet beta-cells by syntaxin 1A (Syx) and soluble N-ethylmaleimide-sensitive factor attachment protein-25 (SNAP-25). We also demonstrated physical interactions in neuroendocrine cells between Kv2.1, Syx, and SNAP-25, characterized their effects on Kv2.1 activation and inactivation in Xenopus laevis oocytes, and suggested that they pertain to the assembly/disassembly of the Syx/SNAP-25 (t-SNARE) complex. In the present work, we established the existence of a causal relationship between the physical and the functional interactions of Syx with the Kv2.1 channel using three different peptides that compete with the channel for binding of Syx when injected into oocytes already coexpressing Syx with Kv2.1 in the plasma membrane: one peptide corresponding to the Syx-binding region on the N-type Ca(2+) channel, and two peptides corresponding to Syx-binding regions on the Kv2.1 C terminus. All peptides reversed the effects of Syx on Kv2.1, suggesting that the hyperpolarizing shifts of the steady-state inactivation and activation of Kv2.1 caused by Syx result from cell-surface protein-protein interactions and point to participation of the C terminus in such an interaction. In line with these findings, the effects of Syx were dissipated by partial deletions of the C terminus. Furthermore, the t-SNARE complex was shown to bind to the Kv2.1 C terminus, and its effects on the inactivation of Kv2.1 were dissipated by partial deletions of the C terminus. Taken together, these findings suggest that physical interactions of both Syx and the t-SNARE complex with the C terminus of Kv2.1 are involved in channel regulation.


Asunto(s)
Antígenos de Superficie/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Fragmentos de Péptidos/fisiología , Canales de Potasio con Entrada de Voltaje/metabolismo , Proteínas de Transporte Vesicular/fisiología , Animales , Antígenos de Superficie/genética , Femenino , Proteínas del Tejido Nervioso/genética , Fragmentos de Péptidos/metabolismo , Canales de Potasio con Entrada de Voltaje/química , Canales de Potasio con Entrada de Voltaje/genética , Proteínas SNARE , Canales de Potasio Shab , Solubilidad , Sintaxina 1 , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo , Xenopus laevis
12.
J Neurophysiol ; 87(3): 1303-10, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11877504

RESUMEN

Fast spiking (FS), GABAergic neurons of the reticular thalamic nucleus (RTN) are capable of firing high-frequency trains of brief action potentials, with little adaptation. Studies in recombinant systems have shown that high-voltage-activated K(+) channels containing the Kv3.1 and/or Kv3.2 subunits display biophysical properties that may contribute to the FS phenotype. Given that RTN expresses high levels of Kv3.1, with little or no Kv3.2, we tested whether this subunit was required for the fast action potential repolarization mechanism essential to the FS phenotype. Single- and multiple-action potentials were recorded using whole-cell current clamp in RTN neurons from brain slices of wild-type and Kv3.1-deficient mice. At 23 degrees C, action potentials recorded from homozygous Kv3.1 deficient mice (Kv3.1(-/-)) compared with their wild-type (Kv3.1(+/+)) counterparts had reduced amplitudes (-6%) and fast after-hyperpolarizations (-16%). At 34 degrees C, action potentials in Kv3.1(-/-) mice had increased duration (21%) due to a reduced rate of repolarization (-30%) when compared with wild-type controls. Action potential trains in Kv3.1(-/-) were associated with a significantly greater spike decrement and broadening and a diminished firing frequency versus injected current relationship (F/I) at 34 degrees C. There was no change in either spike count or maximum instantaneous frequency during low-threshold Ca(2+) bursts in Kv3.1(-/-) RTN neurons at either temperature tested. Our findings show that Kv3.1 is not solely responsible for fast spikes or high-frequency firing in RTN neurons. This suggests genetic redundancy in the system, possibly in the form of other Kv3 members, which may suffice to maintain the FS phenotype in RTN neurons in the absence of Kv3.1.


Asunto(s)
Potenciales de Acción/fisiología , Núcleos Talámicos Intralaminares/fisiología , Neuropéptidos/genética , Canales de Potasio con Entrada de Voltaje , Canales de Potasio/genética , Animales , Conducta Animal , Núcleos Talámicos Intralaminares/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/fisiología , Neuropéptidos/metabolismo , Técnicas de Cultivo de Órganos , Técnicas de Placa-Clamp , Canales de Potasio/metabolismo , Canales de Potasio Shaw
13.
Eur J Neurosci ; 19(12): 3317-27, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15217387

RESUMEN

Double-mutant mice (DKO) lacking the two voltage-gated K(+) channels Kv3.1 and Kv3.3 display a series of phenotypic alterations that include ataxia, myoclonus, tremor and alcohol hypersensitivity. The prominent cerebellar expression of mRNAs encoding Kv3.1 and Kv3.3 subunits raised the question as to whether altered electrical activity resulting from the lack of these K(+) channels might be related to the dramatic motor changes. We used the tremorogenic agent harmaline to probe mutant mice lacking different K(+) channel alleles for altered olivocerebellar circuit properties. Harmaline induced the characteristic 13-Hz tremor in wildtype mice (WT); however, no tremor was observed in DKO suggesting that the ensemble properties of the olivocerebellar circuitry are altered in the absence of Kv3.1 and Kv3.3 subunits. Harmaline induced tremor in Kv3.1-single mutants, but it was of smaller amplitude and at a lower frequency indicating the participation of Kv3.1 subunits in normal olivocerebellar system function. In contrast, harmaline tremor was virtually absent in Kv3.3-single mutants indicating an essential role for Kv3.3 subunits in tremor induction by harmaline. Immunohistochemical staining for Kv3.3 showed clear expression in the somata and proximal dendrites of Purkinje cells and in their axonal projections to the deep cerebellar nuclei (DCN). In DCN, both Kv3.1 and Kv3.3 subunits are expressed. Action potential duration is increased by approximately 100% in Purkinje cells from Kv3.3-single mutants compared to WT or Kv3.1-single mutants. We conclude that Kv3.3 channel subunits are essential for the olivocerebellar system to generate and sustain normal harmaline tremor whereas Kv3.1 subunits influence tremor amplitude and frequency.


Asunto(s)
Alelos , Cerebelo/fisiología , Vías Nerviosas/fisiología , Núcleo Olivar/fisiología , Canales de Potasio con Entrada de Voltaje , Canales de Potasio/deficiencia , Potenciales de Acción/fisiología , Animales , Western Blotting , Estimulantes del Sistema Nervioso Central/farmacología , Cerebelo/efectos de los fármacos , Harmalina/farmacología , Inmunohistoquímica , Ratones , Ratones Mutantes , Microscopía Confocal , Actividad Motora/fisiología , Mutación , Vías Nerviosas/efectos de los fármacos , Núcleo Olivar/efectos de los fármacos , Técnicas de Cultivo de Órganos , Técnicas de Placa-Clamp , Canales de Potasio/genética , Canales de Potasio Shaw , Temblor/inducido químicamente , Temblor/genética
14.
Eur J Neurosci ; 15(1): 40-50, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11860505

RESUMEN

During the last few years a variety of genetically encodable optical probes that monitor physiological parameters such as local pH, Ca2+, Cl-, or transmembrane voltage have been developed. These sensors are based on variants of green-fluorescent protein (GFP) and can be synthesized by mammalian cells after transfection with cDNA. To use these sensor proteins in intact brain tissue, specific promoters are needed that drive protein expression at a sufficiently high expression level in distinct neuronal subpopulations. Here we investigated whether the promoter sequence of a particular potassium channel may be useful for this purpose. We produced transgenic mouse lines carrying the gene for enhanced yellow-fluorescent protein (EYFP), a yellow-green pH- and Cl- sensitive variant of GFP, under control of the Kv3.1 K+ channel promoter (pKv3.1). Transgenic mouse lines displayed high levels of EYFP expression, identified by confocal microscopy, in adult cerebellar granule cells, interneurons of the cerebral cortex, and in neurons of hippocampus and thalamus. Furthermore, using living cerebellar slices we demonstrate that expression levels of EYFP are sufficient to report intracellular pH and Cl- concentration using imaging techniques and conditions analogous to those used with conventional ion-sensitive dyes. We conclude that transgenic mice expressing GFP-derived sensors under the control of cell-type specific promoters, provide a unique opportunity for functional characterization of defined subsets of neurons.


Asunto(s)
Proteínas Bacterianas/biosíntesis , Química Encefálica/genética , Cloruros/metabolismo , Proteínas Luminiscentes/biosíntesis , Canales de Potasio con Entrada de Voltaje , Canales de Potasio/genética , Regiones Promotoras Genéticas/genética , Animales , Encéfalo/anatomía & histología , Agonistas del GABA/farmacología , Ácido Glutámico/farmacología , Concentración de Iones de Hidrógeno , Ratones , Ratones Transgénicos , Microscopía Confocal , Muscimol/farmacología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Canales de Potasio Shaw
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA