Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Neurosci ; 2024 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-38858080

RESUMEN

The resurgent sodium current (INaR) activates on membrane repolarization, such as during the downstroke of neuronal action potentials. Due to its unique activation properties, INaR is thought to drive high rates of repetitive neuronal firing. However, INaR is often studied in combination with the persistent or non-inactivating portion of sodium currents (INaP). We used dynamic clamp to test how INaR and INaP individually affect repetitive firing in adult cerebellar Purkinje neurons from male and female mice. We learned INaR does not scale repetitive firing rates due to its rapid decay at subthreshold voltages, and that subthreshold INaP is critical in regulating neuronal firing rate. Adjustments to the Nav conductance model used in these studies revealed INaP and INaR can be inversely scaled by adjusting occupancy in the slow inactivated kinetic state. Together with additional dynamic clamp experiments, these data suggest the regulation of sodium channel slow inactivation can fine-tune INaP and Purkinje neuron repetitive firing rates.Significance Statement Across neuronal cell types, the resurgent sodium current (INaR-) is often implicated in driving high rates of repetitive firing. Using dynamic clamp, we determined INaR is ineffective at driving subsequent action potentials, and that the subthreshold persistent sodium current (INaP) is the critical parameter for scaling repetitive firing rates. We propose INaR measured in native neurons may reflect a mechanism by which the magnitude of INaP is fine-tuned.

2.
Cell Mol Life Sci ; 75(19): 3495-3505, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29982847

RESUMEN

Purkinje neurons, the sole output of the cerebellar cortex, deliver GABA-mediated inhibition to the deep cerebellar nuclei. To subserve this critical function, Purkinje neurons fire repetitively, and at high frequencies, features that have been linked to the unique properties of the voltage-gated sodium (Nav) channels expressed. In addition to the rapidly activating and inactivating, or transient, component of the Nav current (INaT) present in many types of central and peripheral neurons, Purkinje neurons, also expresses persistent (INaP) and resurgent (INaR) Nav currents. Considerable progress has been made in detailing the biophysical properties and identifying the molecular determinants of these discrete Nav current components, as well as defining their roles in the regulation of Purkinje neuron excitability. Here, we review this important work and highlight the remaining questions about the molecular mechanisms controlling the expression and the functioning of Nav currents in Purkinje neurons. We also discuss the impact of the dynamic regulation of Nav currents on the functioning of individual Purkinje neurons and cerebellar circuits.


Asunto(s)
Potenciales de Acción/fisiología , Cerebelo/citología , Células de Purkinje/fisiología , Canales de Sodio Activados por Voltaje/fisiología , Potenciales de Acción/genética , Animales , Cerebelo/fisiología , Humanos , Activación del Canal Iónico/genética , Activación del Canal Iónico/fisiología , Neuronas/metabolismo , Neuronas/fisiología , Células de Purkinje/citología , Canales de Sodio Activados por Voltaje/clasificación , Canales de Sodio Activados por Voltaje/genética
3.
J Neurosci ; 35(17): 6752-69, 2015 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-25926453

RESUMEN

Mutations in FGF14, which encodes intracellular fibroblast growth factor 14 (iFGF14), have been linked to spinocerebellar ataxia (SCA27). In addition, mice lacking Fgf14 (Fgf14(-/-)) exhibit an ataxia phenotype resembling SCA27, accompanied by marked changes in the excitability of cerebellar granule and Purkinje neurons. It is not known, however, whether these phenotypes result from defects in neuronal development or if they reflect a physiological requirement for iFGF14 in the adult cerebellum. Here, we demonstrate that the acute and selective Fgf14-targeted short hairpin RNA (shRNA)-mediated in vivo "knock-down" of iFGF14 in adult Purkinje neurons attenuates spontaneous and evoked action potential firing without measurably affecting the expression or localization of voltage-gated Na(+) (Nav) channels at Purkinje neuron axon initial segments. The selective shRNA-mediated in vivo "knock-down" of iFGF14 in adult Purkinje neurons also impairs motor coordination and balance. Repetitive firing can be restored in Fgf14-targeted shRNA-expressing Purkinje neurons, as well as in Fgf14(-/-) Purkinje neurons, by prior membrane hyperpolarization, suggesting that the iFGF14-mediated regulation of the excitability of mature Purkinje neurons depends on membrane potential. Further experiments revealed that the loss of iFGF14 results in a marked hyperpolarizing shift in the voltage dependence of steady-state inactivation of the Nav currents in adult Purkinje neurons. We also show here that expressing iFGF14 selectively in adult Fgf14(-/-) Purkinje neurons rescues spontaneous firing and improves motor performance. Together, these results demonstrate that iFGF14 is required for spontaneous and evoked action potential firing in adult Purkinje neurons, thereby controlling the output of these cells and the regulation of motor coordination and balance.


Asunto(s)
Potenciales de Acción/genética , Cerebelo/citología , Factores de Crecimiento de Fibroblastos/metabolismo , Equilibrio Postural/genética , Desempeño Psicomotor/fisiología , Células de Purkinje/fisiología , Potenciales de Acción/fisiología , Animales , Ancirinas/metabolismo , Axones/metabolismo , Línea Celular Transformada , Cricetulus , Femenino , Factores de Crecimiento de Fibroblastos/genética , Regulación de la Expresión Génica/genética , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , MicroARNs/metabolismo , Canal de Sodio Activado por Voltaje NAV1.1/metabolismo , Células de Purkinje/citología
4.
J Neurosci ; 33(24): 9950-6, 2013 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-23761890

RESUMEN

Biological and theoretical evidence suggest that individual neurons may achieve similar outputs by differentially balancing variable underlying ionic conductances. Despite the substantial amount of data consistent with this idea, a direct biological demonstration that cells with conserved output, particularly within the same network, achieve these outputs via different solutions has been difficult to achieve. Here we demonstrate definitively that neurons from native neural networks with highly similar output achieve this conserved output by differentially tuning underlying conductance magnitudes. Multiple motor neurons of the crab (Cancer borealis) cardiac ganglion have highly conserved output within a preparation, despite showing a 2-4-fold range of conductance magnitudes. By blocking subsets of these currents, we demonstrate that the remaining conductances become unbalanced, causing disparate output as a result. Therefore, as strategies to understand neuronal excitability become increasingly sophisticated, it is important that such variability in excitability of neurons, even among those within the same individual, is taken into account.


Asunto(s)
Fenómenos Biofísicos/fisiología , Neuronas Motoras/fisiología , Red Nerviosa/fisiología , Conducción Nerviosa/fisiología , Potenciales de Acción/fisiología , Animales , Braquiuros , Estimulación Eléctrica , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Ganglios de Invertebrados/citología , Masculino , Red Nerviosa/citología , Técnicas de Placa-Clamp , Estadísticas no Paramétricas
5.
bioRxiv ; 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38187680

RESUMEN

The resurgent sodium current (INaR) activates on membrane repolarization, such as during the downstroke of neuronal action potentials. Due to its unique activation properties, INaR is thought to drive high rates of repetitive neuronal firing. However, INaR is often studied in combination with the persistent or non-inactivating portion of sodium currents (INaP). We used dynamic clamp to test how INaR and INaP individually affect repetitive firing in adult cerebellar Purkinje neurons from male and female mice. We learned INaR does not scale repetitive firing rates due to its rapid decay at subthreshold voltages, and that subthreshold INaP is critical in regulating neuronal firing rate. Adjustments to the Nav conductance model used in these studies revealed INaP and INaR can be inversely scaled by adjusting occupancy in the slow inactivated kinetic state. Together with additional dynamic clamp experiments, these data suggest the regulation of sodium channel slow inactivation can fine-tune INaP and Purkinje neuron repetitive firing rates.

6.
bioRxiv ; 2024 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-38746081

RESUMEN

Mutations in FGF14 , which encodes intracellular fibroblast growth factor 14 (iFGF14), have been linked to spinocerebellar ataxia type 27 (SCA27), a multisystem disorder associated with progressive deficits in motor coordination and cognitive function. Mice ( Fgf14 -/- ) lacking iFGF14 display similar phenotypes, and we have previously shown that the deficits in motor coordination reflect reduced excitability of cerebellar Purkinje neurons, owing to the loss of iFGF14-mediated regulation of the voltage-dependence of inactivation of the fast transient component of the voltage-gated Na + (Nav) current, I NaT . Here, we present the results of experiments designed to test the hypothesis that loss of iFGF14 also attenuates the intrinsic excitability of mature hippocampal and cortical pyramidal neurons. Current-clamp recordings from adult mouse hippocampal CA1 pyramidal neurons in acute in vitro slices, however, revealed that repetitive firing rates were higher in Fgf14 -/- , than in wild type (WT), cells. In addition, the waveforms of individual action potentials were altered in Fgf14 -/- hippocampal CA1 pyramidal neurons, and the loss of iFGF14 reduced the time delay between the initiation of axonal and somal action potentials. Voltage-clamp recordings revealed that the loss of iFGF14 altered the voltage-dependence of activation, but not inactivation, of I NaT in CA1 pyramidal neurons. Similar effects of the loss of iFGF14 on firing properties were evident in current-clamp recordings from layer 5 visual cortical pyramidal neurons. Additional experiments demonstrated that the loss of iFGF14 does not alter the distribution of anti-Nav1.6 or anti-ankyrin G immunofluorescence labeling intensity along the axon initial segments (AIS) of mature hippocampal CA1 or layer 5 visual cortical pyramidal neurons in situ . Taken together, the results demonstrate that, in contrast with results reported for neonatal (rat) hippocampal pyramidal neurons in dissociated cell culture, the loss of iFGF14 does not disrupt AIS architecture or Nav1.6 localization/distribution along the AIS of mature hippocampal (or cortical) pyramidal neurons in situ .

7.
J Neurosci ; 32(28): 9649-58, 2012 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-22787050

RESUMEN

Neurons and networks undergo a process of homeostatic plasticity that stabilizes output by integrating activity levels with network and cellular properties to counter longer-term perturbations. Here we describe a rapid compensatory interaction among a pair of potassium currents, I(A) and I(KCa), that stabilizes both intrinsic excitability and network function in the cardiac ganglion of the crab, Cancer borealis. We determined that mRNA levels in single identified neurons for the channels which encode I(A) and I(KCa) are positively correlated, yet the ionic currents themselves are negatively correlated, across a population of motor neurons. We then determined that these currents are functionally coupled; decreasing levels of either current within a neuron causes a rapid increase in the other. This functional interdependence results in homeostatic stabilization of both the individual neuronal and the network output. Furthermore, these compensatory increases are mechanistically independent, suggesting robustness in the maintenance of neural network output that is critical for survival. Together, we generate a complete model for homeostatic plasticity from mRNA to network output where rapid post-translational compensatory mechanisms acting on a reservoir of channels proteins regulated at the level of gene expression provide homeostatic stabilization of both cellular and network activity.


Asunto(s)
Homeostasis , Neuronas Motoras/fisiología , Red Nerviosa/fisiología , Plasticidad Neuronal/fisiología , 4-Aminopiridina/farmacología , Potenciales de Acción/efectos de los fármacos , Análisis de Varianza , Animales , Braquiuros , Quelantes/farmacología , Ciclosporina/farmacología , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Estimulación Eléctrica , Inhibidores Enzimáticos/farmacología , Femenino , Ganglios de Invertebrados/citología , Homeostasis/efectos de los fármacos , Masculino , Neuronas Motoras/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio/genética , Canales de Potasio/metabolismo , ARN Mensajero , Bloqueadores de los Canales de Sodio/farmacología , Tetraetilamonio/farmacología , Tetrodotoxina/farmacología
8.
J Neurophysiol ; 110(1): 42-54, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23576706

RESUMEN

Large cell motoneurons in the Cancer borealis cardiac ganglion generate rhythmic bursts of action potentials responsible for cardiac contractions. While it is well known that these burst potentials are dependent on coordinated interactions among depolarizing and hyperpolarizing conductances, the depolarizing currents present in these cells, and their biophysical characteristics, have not been thoroughly described. In this study we used a combined molecular biology and electrophysiology approach to look at channel identity, expression, localization, and biophysical properties for two distinct high-voltage-activated calcium currents present in these cells: a slow calcium current (ICaS) and a transient calcium current (ICaT). Our data indicate that CbCaV1 is a putative voltage-gated calcium channel subunit in part responsible for an L-type current, while CbCaV2 (formerly cacophony) is a subunit in part responsible for a P/Q-type current. These channels appear to be localized primarily to the somata of the motoneurons. A third calcium channel gene (CbCaV3) was identified that encodes a putative T-type calcium channel subunit and is expressed in these cells, but electrophysiological studies failed to detect this current in motoneuron somata. In addition, we identify and characterize for the first time in these cells a calcium-activated nonselective cationic current (ICAN), as well as a largely noninactivating TTX-sensitive current reminiscent of a persistent sodium current. The identification and further characterization of these currents allow both biological and modeling studies to move forward with more attention to the complexity of interactions among these distinct components underlying generation of bursting output in motoneurons.


Asunto(s)
Canales de Calcio/fisiología , Ganglios de Invertebrados/fisiología , Neuronas Motoras/fisiología , Potenciales de Acción , Animales , Braquiuros , Corazón/inervación
9.
Elife ; 112022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-35076394

RESUMEN

The resurgent component of the voltage-gated sodium current (INaR) is a depolarizing conductance, revealed on membrane hyperpolarizations following brief depolarizing voltage steps, which has been shown to contribute to regulating the firing properties of numerous neuronal cell types throughout the central and peripheral nervous systems. Although mediated by the same voltage-gated sodium (Nav) channels that underlie the transient and persistent Nav current components, the gating mechanisms that contribute to the generation of INaR remain unclear. Here, we characterized Nav currents in mouse cerebellar Purkinje neurons, and used tailored voltage-clamp protocols to define how the voltage and the duration of the initial membrane depolarization affect the amplitudes and kinetics of INaR. Using the acquired voltage-clamp data, we developed a novel Markov kinetic state model with parallel (fast and slow) inactivation pathways and, we show that this model reproduces the properties of the resurgent, as well as the transient and persistent, Nav currents recorded in (mouse) cerebellar Purkinje neurons. Based on the acquired experimental data and the simulations, we propose that resurgent Na+ influx occurs as a result of fast inactivating Nav channels transitioning into an open/conducting state on membrane hyperpolarization, and that the decay of INaR reflects the slow accumulation of recovered/opened Nav channels into a second, alternative and more slowly populated, inactivated state. Additional simulations reveal that extrinsic factors that affect the kinetics of fast or slow Nav channel inactivation and/or impact the relative distribution of Nav channels in the fast- and slow-inactivated states, such as the accessory Navß4 channel subunit, can modulate the amplitude of INaR.


Asunto(s)
Potenciales de Acción/fisiología , Activación del Canal Iónico , Células de Purkinje/metabolismo , Sodio/metabolismo , Subunidad beta-4 de Canal de Sodio Activado por Voltaje/deficiencia , Animales , Animales Recién Nacidos , Cerebelo/citología , Femenino , Cinética , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/citología , Neuronas/metabolismo , Técnicas de Placa-Clamp , Equilibrio Postural/fisiología , Subunidad beta-4 de Canal de Sodio Activado por Voltaje/metabolismo
10.
Cell Rep ; 19(3): 532-544, 2017 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-28423317

RESUMEN

The resurgent component of voltage-gated Na+ (Nav) currents, INaR, has been suggested to provide the depolarizing drive for high-frequency firing and to be generated by voltage-dependent Nav channel block (at depolarized potentials) and unblock (at hyperpolarized potentials) by the accessory Navß4 subunit. To test these hypotheses, we examined the effects of the targeted deletion of Scn4b (Navß4) on INaR and on repetitive firing in cerebellar Purkinje neurons. We show here that Scn4b-/- animals have deficits in motor coordination and balance and that firing rates in Scn4b-/- Purkinje neurons are markedly attenuated. Acute, in vivo short hairpin RNA (shRNA)-mediated "knockdown" of Navß4 in adult Purkinje neurons also reduced spontaneous and evoked firing rates. Dynamic clamp-mediated addition of INaR partially rescued firing in Scn4b-/- Purkinje neurons. Voltage-clamp experiments revealed that INaR was reduced (by ∼50%), but not eliminated, in Scn4b-/- Purkinje neurons, revealing that additional mechanisms contribute to generation of INaR.


Asunto(s)
Potenciales de Acción/fisiología , Activación del Canal Iónico , Actividad Motora/fisiología , Equilibrio Postural/fisiología , Células de Purkinje/metabolismo , Sodio/metabolismo , Subunidad beta-4 de Canal de Sodio Activado por Voltaje/deficiencia , Envejecimiento , Animales , Animales Recién Nacidos , Diferenciación Celular , Separación Celular , Femenino , Eliminación de Gen , Técnicas de Silenciamiento del Gen , Marcación de Gen , Masculino , Ratones Endogámicos C57BL , Neuronas/citología , Neuronas/metabolismo , Técnicas de Placa-Clamp , Subunidad beta-4 de Canal de Sodio Activado por Voltaje/metabolismo
11.
Elife ; 52016 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-27552052

RESUMEN

Motor neurons of the crustacean cardiac ganglion generate virtually identical, synchronized output despite the fact that each neuron uses distinct conductance magnitudes. As a result of this variability, manipulations that target ionic conductances have distinct effects on neurons within the same ganglion, disrupting synchronized motor neuron output that is necessary for proper cardiac function. We hypothesized that robustness in network output is accomplished via plasticity that counters such destabilizing influences. By blocking high-threshold K(+) conductances in motor neurons within the ongoing cardiac network, we discovered that compensation both resynchronized the network and helped restore excitability. Using model findings to guide experimentation, we determined that compensatory increases of both GA and electrical coupling restored function in the network. This is one of the first direct demonstrations of the physiological regulation of coupling conductance in a compensatory context, and of synergistic plasticity across cell- and network-level mechanisms in the restoration of output.


Asunto(s)
Potenciales de Acción , Crustáceos , Ganglios de Invertebrados/fisiología , Neuronas Motoras/fisiología , Vías Nerviosas/fisiología , Plasticidad Neuronal , Animales , Técnicas de Placa-Clamp
12.
Neuron ; 84(2): 311-23, 2014 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-25374357

RESUMEN

The promise of using reprogrammed human neurons for disease modeling and regenerative medicine relies on the ability to induce patient-derived neurons with high efficiency and subtype specificity. We have previously shown that ectopic expression of brain-enriched microRNAs (miRNAs), miR-9/9* and miR-124 (miR-9/9*-124), promoted direct conversion of human fibroblasts into neurons. Here we show that coexpression of miR-9/9*-124 with transcription factors enriched in the developing striatum, BCL11B (also known as CTIP2), DLX1, DLX2, and MYT1L, can guide the conversion of human postnatal and adult fibroblasts into an enriched population of neurons analogous to striatal medium spiny neurons (MSNs). When transplanted in the mouse brain, the reprogrammed human cells persisted in situ for over 6 months, exhibited membrane properties equivalent to native MSNs, and extended projections to the anatomical targets of MSNs. These findings highlight the potential of exploiting the synergism between miR-9/9*-124 and transcription factors to generate specific neuronal subtypes.


Asunto(s)
Diferenciación Celular/fisiología , Cuerpo Estriado/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , MicroARNs/metabolismo , Neostriado/citología , Neuronas/metabolismo , Animales , Células Cultivadas , Humanos , Ratones , Factores de Transcripción/metabolismo
14.
Front Mol Neurosci ; 3: 116, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21119779

RESUMEN

In addition to the overall complexity of transcriptional regulation, cells also must take into account the subcellular distribution of these gene products. This is particularly challenging for morphologically complex cells such as neurons. Yet the interaction between cellular morphology and gene expression is poorly understood. Here we provide some of the first evidence for a relationship between neuronal compartment size and maintenance of mRNA levels in neurons. We find that single-cell transcript levels of 18S rRNA, GAPDH, and EF1-alpha, all gene products with primary functions in the cell soma, are strongly correlated to soma size in multiple distinct neuronal types. Levels of mRNA for the K+ channel shal, which is localized exclusively to the soma, are negatively correlated with soma size, suggesting that gene expression does not simply track positively with compartment size. Conversely, levels of beta-actin and beta-tubulin mRNA, which are major cytoskeletal proteins of neuronal processes, do not correlate with soma size, but are strongly correlated with one another. Additionally, actin/tubulin expression levels correlate with voltage-gated ion channels that are uniquely localized to axons. These results suggest that steady-state transcript levels are differentially regulated based on the subcellular compartment within which a given gene product primarily acts.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA