Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 105
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Neurobiol Dis ; 181: 106107, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37001612

RESUMEN

Hyperpolarization-activation cyclic nucleotide-gated (HCN) channels were for the first time implicated in absence seizures (ASs) when an abnormal Ih (the current generated by these channels) was reported in neocortical layer 5 neurons of a mouse model. Genetic studies of large cohorts of children with Childhood Absence Epilepsy (where ASs are the only clinical symptom) have identified only 3 variants in HCN1 (one of the genes that code for the 4 HCN channel isoforms, HCN1-4), with one (R590Q) mutation leading to loss-of-function. Due to the multi-faceted effects that HCN channels exert on cellular excitability and neuronal network dynamics as well as their modulation by environmental factors, it has been difficult to identify the detailed mechanism by which different HCN isoforms modulate ASs. In this review, we systematically and critically analyze evidence from established AS models and normal non-epileptic animals with area- and time-selective ablation of HCN1, HCN2 and HCN4. Notably, whereas knockout of rat HCN1 and mouse HCN2 leads to the expression of ASs, the pharmacological block of all HCN channel isoforms abolishes genetically determined ASs. These seemingly contradictory results could be reconciled by taking into account the well-known opposite effects of Ih on cellular excitability and network function. Whereas existing evidence from mouse and rat AS models indicates that pan-HCN blockers may provide a novel approach for the treatment of human ASs, the development of HCN isoform-selective drugs would greatly contribute to current research on the role for these channels in ASs generation and maintenance as well as offer new potential clinical applications.


Asunto(s)
Epilepsia Tipo Ausencia , Animales , Niño , Humanos , Ratones , Ratas , Epilepsia Tipo Ausencia/genética , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Neuronas/metabolismo , Convulsiones/genética , Convulsiones/metabolismo
2.
Neurobiol Dis ; 178: 106025, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36731682

RESUMEN

Spike-and-wave discharges (SWDs), generated by the cortico-thalamo-cortical (CTC) network, are pathological, large amplitude oscillations and the hallmark of absence seizures (ASs). SWDs begin in a cortical initiation network in both humans and animal models, including the Genetic Absence Epilepsy Rats from Strasbourg (GAERS), where it is located in the primary somatosensory cortex (S1). The behavioral manifestation of an AS occurs when SWDs spread from the cortical initiation site to the whole brain, however, the mechanisms behind this rapid propagation remain unclear. Here we investigated these processes beyond the principal CTC network, in higher-order (HO) thalamic nuclei (lateral posterior (LP) and posterior (PO) nuclei) since their diffuse connectivity and known facilitation of intracortical communications make these nuclei key candidates to support SWD generation and maintenance. In freely moving GAERS, multi-site LFP in LP, PO and multiple cortical regions revealed a novel feature of SWDs: during SWDs there are short periods (named SWD-breaks) when cortical regions far from S1, such the primary visual cortex (V1), become transiently unsynchronized from the ongoing EEG rhythm. Inactivation of HO nuclei with local muscimol injections or optogenetic perturbation of HO nuclei activity increased the occurrence of SWD-breaks and the former intervention also increased the SWD propagation-time from S1. The neural underpinnings of these findings were explored further by silicon probe recordings from single units of PO which uncovered two previously unknown groups of excitatory neurons based on their burst firing dynamics at SWD onset. Moreover, a switch from tonic to burst firing at SWD onset was shown to be an important feature since it was much less prominent for non-generalized events, i.e. SWDs that remained local to S1. Additionally, one group of neurons showed a reverse of this switch during SWD-breaks, demonstrating the importance of this firing pattern throughout the SWD. In summary, these results support the view that multiple HO thalamic nuclei are utilized at SWD onset and contribute to cortical synchrony throughout the paroxysmal discharge.


Asunto(s)
Epilepsia Tipo Ausencia , Humanos , Ratas , Animales , Epilepsia Tipo Ausencia/genética , Electroencefalografía , Núcleos Talámicos/fisiología , Convulsiones , Neuronas/fisiología , Tálamo , Modelos Animales de Enfermedad
3.
Neurobiol Dis ; 186: 106275, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37648038

RESUMEN

Typical absence seizures (ASs) are brief periods of lack of consciousness, associated with 2.5-4 Hz spike-wave discharges (SWDs) in the EEG, which are highly prevalent in children and teenagers. The majority of probands in these young epileptic cohorts show neuropsychological comorbidities, including cognitive, memory and mood impairments, even after the seizures are pharmacologically controlled. Similar cognition and memory deficits have been reported in different, but not all, genetic animal models of ASs. However, since these impairments are subtle and highly task-specific their presence may be confounded by an anxiety-like phenotype and no study has tested anxiety and memory in the same animals. Moreover, the majority of studies used non-epileptic inbred animals as the only control strain and this may have contributed to a misinterpretation of these behavioural results. To overcome these issues, here we used a battery of behavioural tests to compare anxiety and memory in the same animals from the well-established inbred model of Genetic Absence Epilepsy Rats from Strasbourg (GAERS), their inbred strain of Non-Epileptic Control (NEC) strain (that lack ASs) and normal outbred Wistar rats. We found that GAERS do not exhibit increased anxiety-like behavior and neophobia compared to both NEC and Wistar rats. In contrast, GAERS show decreased spontaneous alternation, spatial working memory and cross-modal object recognition compared to both NEC and Wistar rats. Furthermore, GAERS preferentially used egocentric strategies to perform spatial memory tasks. In summary, these results provide solid evidence of memory deficits in GAERS rats that do not depend on an anxiety or neophobic phenotype. Moreover, the presence of differences between NEC and Wistar rats stresses the need of using both outbred and inbred control rats in behavioural studies involving genetic models of ASs.


Asunto(s)
Ansiedad , Convulsiones , Humanos , Niño , Adolescente , Ratas , Animales , Ratas Wistar , Cognición , Trastornos de la Memoria
4.
Nat Rev Neurosci ; 19(2): 107-118, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29321683

RESUMEN

During inattentive wakefulness and non-rapid eye movement (NREM) sleep, the neocortex and thalamus cooperatively engage in rhythmic activities that are exquisitely reflected in the electroencephalogram as distinctive rhythms spanning a range of frequencies from <1 Hz slow waves to 13 Hz alpha waves. In the thalamus, these diverse activities emerge through the interaction of cell-intrinsic mechanisms and local and long-range synaptic inputs. One crucial feature, however, unifies thalamic oscillations of different frequencies: repetitive burst firing driven by voltage-dependent Ca2+ spikes. Recent evidence reveals that thalamic Ca2+ spikes are inextricably linked to global somatodendritic Ca2+ transients and are essential for several forms of thalamic plasticity. Thus, we propose herein that alongside their rhythm-regulation function, thalamic oscillations of low-vigilance states have a plasticity function that, through modifications of synaptic strength and cellular excitability in local neuronal assemblies, can shape ongoing oscillations during inattention and NREM sleep and may potentially reconfigure thalamic networks for faithful information processing during attentive wakefulness.


Asunto(s)
Nivel de Alerta/fisiología , Plasticidad Neuronal/fisiología , Sueño de Onda Lenta/fisiología , Tálamo/fisiología , Animales , Humanos
5.
Epilepsia ; 62(7): 1729-1743, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34018186

RESUMEN

OBJECTIVE: Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are known to be involved in the generation of absence seizures (ASs), and there is evidence that cortical and thalamic HCN channel dysfunctions may have a proabsence role. Many HCN channel blockers are available, but their role in ASs has been investigated only by localized brain injection or in in vitro model systems due to their limited brain availability. Here, we investigated the effect on ASs of orally administered ivabradine (an HCN channel blocker approved for the treatment of heart failure in humans) following injection of the P-glycoprotein inhibitor elacridar, which is known to increase penetration into the brain of drug substrates for this efflux transporter. The action of ivabradine was also tested following in vivo microinjection into the cortical initiation network (CIN) of the somatosensory cortex and in the thalamic ventrobasal nucleus (VB) as well as on cortical and thalamocortical neurons in brain slices. METHODS: We used electroencephalographic recordings in freely moving Genetic Absence Epilepsy Rats From Strasbourg (GAERSs) to assess the action of oral administration of ivabradine, with and without elacridar, on ASs. Ivabradine was also microinjected into the CIN and VB of GAERSs in vivo and applied to Wistar CIN and GAERS VB slices while recording patch-clamped cortical Layer 5/6 and thalamocortical neurons, respectively. RESULTS: Oral administration of ivabradine markedly and dose-dependently reduced ASs. Ivabradine injection into CIN abolished ASs and elicited small-amplitude 4-7-Hz waves (without spikes), whereas in the VB it was less potent. Moreover, ivabradine applied to GAERS VB and Wistar CIN slices selectively decreased HCN channel-dependent properties of cortical Layer 5/6 pyramidal and thalamocortical neurons, respectively. SIGNIFICANCE: These results provide the first demonstration of the antiabsence action of a systemically administered HCN channel blocker, indicating the potential of this class of drugs as a novel therapeutic avenue for ASs.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Canales Catiónicos Regulados por Nucleótidos Cíclicos/antagonistas & inhibidores , Ivabradina/uso terapéutico , Convulsiones/prevención & control , Animales , Anticonvulsivantes/farmacología , Corteza Cerebral , Relación Dosis-Respuesta a Droga , Electroencefalografía/efectos de los fármacos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Ivabradina/farmacología , Masculino , Microinyecciones , Red Nerviosa , Neuronas/efectos de los fármacos , Células Piramidales/efectos de los fármacos , Ratas , Ratas Wistar , Convulsiones/genética , Corteza Somatosensorial , Núcleos Talámicos Ventrales
6.
Brain ; 143(8): 2341-2368, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32437558

RESUMEN

Absence seizures in children and teenagers are generally considered relatively benign because of their non-convulsive nature and the large incidence of remittance in early adulthood. Recent studies, however, show that 30% of children with absence seizures are pharmaco-resistant and 60% are affected by severe neuropsychiatric comorbid conditions, including impairments in attention, cognition, memory and mood. In particular, attention deficits can be detected before the epilepsy diagnosis, may persist even when seizures are pharmacologically controlled and are aggravated by valproic acid monotherapy. New functional MRI-magnetoencephalography and functional MRI-EEG studies provide conclusive evidence that changes in blood oxygenation level-dependent signal amplitude and frequency in children with absence seizures can be detected in specific cortical networks at least 1 min before the start of a seizure, spike-wave discharges are not generalized at seizure onset and abnormal cortical network states remain during interictal periods. From a neurobiological perspective, recent electrical recordings and imaging of large neuronal ensembles with single-cell resolution in non-anaesthetized models show that, in contrast to the predominant opinion, cortical mechanisms, rather than an exclusively thalamic rhythmogenesis, are key in driving seizure ictogenesis and determining spike-wave frequency. Though synchronous ictal firing characterizes cortical and thalamic activity at the population level, individual cortico-thalamic and thalamocortical neurons are sparsely recruited to successive seizures and consecutive paroxysmal cycles within a seizure. New evidence strengthens previous findings on the essential role for basal ganglia networks in absence seizures, in particular the ictal increase in firing of substantia nigra GABAergic neurons. Thus, a key feature of thalamic ictogenesis is the powerful increase in the inhibition of thalamocortical neurons that originates at least from two sources, substantia nigra and thalamic reticular nucleus. This undoubtedly provides a major contribution to the ictal decrease in total firing and the ictal increase of T-type calcium channel-mediated burst firing of thalamocortical neurons, though the latter is not essential for seizure expression. Moreover, in some children and animal models with absence seizures, the ictal increase in thalamic inhibition is enhanced by the loss-of-function of the astrocytic GABA transporter GAT-1 that does not necessarily derive from a mutation in its gene. Together, these novel clinical and experimental findings bring about paradigm-shifting views of our understanding of absence seizures and demand careful choice of initial monotherapy and continuous neuropsychiatric evaluation of affected children. These issues are discussed here to focus future clinical and experimental research and help to identify novel therapeutic targets for treating both absence seizures and their comorbidities.


Asunto(s)
Convulsiones/fisiopatología , Convulsiones/terapia , Adolescente , Animales , Niño , Comorbilidad , Humanos
7.
Int J Mol Sci ; 22(17)2021 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-34502374

RESUMEN

(1) Background: Absence seizures (ASs) are sudden, transient lapses of consciousness associated with lack of voluntary movements and generalized 2.5-4 Hz spike-wave discharges (SWDs) in the EEG. In addition to the thalamocortical system, where these pathological oscillations are generated, multiple neuronal circuits have been involved in their modulation and associated comorbidities including the serotonergic system. Neuronal activity in one of the major synaptic input structures to the brainstem dorsal raphé nucleus (DRN), the lateral hypothalamus (LH), has not been characterized. (2) Methods: We used viral tract tracing and optogenetics combined with in vitro and in vivo electrophysiology to assess the involvement of the LH in absence epilepsy in a genetic rodent model. (3) Results: We found that a substantial fraction of LH neurons project to the DRN of which a minority is GABAergic. The LH to DRN projection can lead to monosynaptic iGluR mediated excitation in DRN 5-HT neurons. Neuronal activity in the LH is coupled to SWDs. (4) Conclusions: Our results indicate that a brain area involved in the regulation of autonomic functions and heavily innervating the RN is involved in ASs. The decreased activity of LH neurons during SWDs could lead to both a decreased excitation and disinhibition in the DRN. These results support a long-range subcortical regulation of serotonergic neuromodulation during ASs and further our understanding of the state-dependence of these seizures and some of their associated comorbidities.


Asunto(s)
Área Hipotalámica Lateral/fisiología , Convulsiones/fisiopatología , Potenciales de Acción , Animales , Tronco Encefálico/fisiología , Modelos Animales de Enfermedad , Núcleo Dorsal del Rafe/metabolismo , Núcleo Dorsal del Rafe/fisiología , Electroencefalografía , Epilepsia Tipo Ausencia/genética , Epilepsia Tipo Ausencia/metabolismo , Epilepsia Tipo Ausencia/fisiopatología , Neuronas GABAérgicas/fisiología , Área Hipotalámica Lateral/metabolismo , Masculino , Optogenética/métodos , Ratas , Ratas Endogámicas , Convulsiones/genética , Convulsiones/metabolismo , Neuronas Serotoninérgicas/fisiología , Serotonina/metabolismo
8.
J Neurosci ; 38(30): 6615-6627, 2018 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-29925625

RESUMEN

Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels and the Ih current they generate contribute to the pathophysiological mechanisms of absence seizures (ASs), but their precise role in neocortical and thalamic neuronal populations, the main components of the network underlying AS generation, remains controversial. In diverse genetic AS models, Ih amplitude is smaller in neocortical neurons and either larger or unchanged in thalamocortical (TC) neurons compared with nonepileptic strains. A lower expression of neocortical HCN subtype 1 channels is present in genetic AS-prone rats, and HCN subtype 2 knock-out mice exhibit ASs. Furthermore, whereas many studies have characterized Ih contribution to "absence-like" paroxysmal activity in vitro, no data are available on the specific role of cortical and thalamic HCN channels in behavioral seizures. Here, we show that the pharmacological block of HCN channels with the antagonist ZD7288 applied via reverse microdialysis in the ventrobasal thalamus (VB) of freely moving male Genetic Absence Epilepsy Rats from Strasbourg decreases TC neuron firing and abolishes spontaneous ASs. A similar effect is observed on γ-hydroxybutyric acid-elicited ASs in normal male Wistar rats. Moreover, thalamic knockdown of HCN channels via virally delivered shRNA into the VB of male Stargazer mice, another genetic AS model, decreases spontaneous ASs and Ih-dependent electrophysiological properties of VB TC neurons. These findings provide the first evidence that block of TC neuron HCN channels prevents ASs and suggest that any potential anti-absence therapy that targets HCN channels should carefully consider the opposite role for cortical and thalamic Ih in the modulation of absence seizures.SIGNIFICANCE STATEMENT Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels play critical roles in the fine-tuning of cellular and network excitability and have been suggested to be a key element of the pathophysiological mechanism underlying absence seizures. However, the precise contribution of HCN channels in neocortical and thalamic neuronal populations to these nonconvulsive seizures is still controversial. In the present study, pharmacological block and genetic suppression of HCN channels in thalamocortical neurons in the ventrobasal thalamic nucleus leads to a marked reduction in absence seizures in one pharmacological and two genetic rodent models of absence seizures. These results provide the first evidence that block of TC neuron HCN channels prevents absence seizures.


Asunto(s)
Epilepsia Tipo Ausencia/metabolismo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/antagonistas & inhibidores , Neuronas/metabolismo , Pirimidinas/farmacología , Núcleos Talámicos Ventrales/metabolismo , Animales , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Epilepsia Tipo Ausencia/fisiopatología , Ratones , Neuronas/efectos de los fármacos , Ratas , Núcleos Talámicos Ventrales/efectos de los fármacos
9.
J Neurosci ; 37(21): 5319-5333, 2017 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-28450536

RESUMEN

Backpropagating action potentials (bAPs) are indispensable in dendritic signaling. Conflicting Ca2+-imaging data and an absence of dendritic recording data means that the extent of backpropagation in thalamocortical (TC) and thalamic reticular nucleus (TRN) neurons remains unknown. Because TRN neurons signal electrically through dendrodendritic gap junctions and possibly via chemical dendritic GABAergic synapses, as well as classical axonal GABA release, this lack of knowledge is problematic. To address this issue, we made two-photon targeted patch-clamp recordings from rat TC and TRN neuron dendrites to measure bAPs directly. These recordings reveal that "tonic"' and low-threshold-spike (LTS) "burst" APs in both cell types are always recorded first at the soma before backpropagating into the dendrites while undergoing substantial distance-dependent dendritic amplitude attenuation. In TC neurons, bAP attenuation strength varies according to firing mode. During LTS bursts, somatic AP half-width increases progressively with increasing spike number, allowing late-burst spikes to propagate more efficiently into the dendritic tree compared with spikes occurring at burst onset. Tonic spikes have similar somatic half-widths to late burst spikes and undergo similar dendritic attenuation. In contrast, in TRN neurons, AP properties are unchanged between LTS bursts and tonic firing and, as a result, distance-dependent dendritic attenuation remains consistent across different firing modes. Therefore, unlike LTS-associated global electrical and calcium signals, the spatial influence of bAP signaling in TC and TRN neurons is more restricted, with potentially important behavioral-state-dependent consequences for synaptic integration and plasticity in thalamic neurons.SIGNIFICANCE STATEMENT In most neurons, action potentials (APs) initiate in the axosomatic region and propagate into the dendritic tree to provide a retrograde signal that conveys information about the level of cellular output to the locations that receive most input: the dendrites. In thalamocortical and thalamic reticular nucleus neurons, the site of AP generation and the true extent of backpropagation remain unknown. Using patch-clamp recordings, this study measures dendritic propagation of APs directly in these neurons. In either cell type, high-frequency low-threshold spike burst or lower-frequency tonic APs undergo substantial voltage attenuation as they spread into the dendritic tree. Therefore, backpropagating spikes in these cells can only influence signaling in the proximal part of the dendritic tree.


Asunto(s)
Potenciales de Acción , Neuronas GABAérgicas/fisiología , Núcleos Talámicos/fisiología , Animales , Dendritas/fisiología , Femenino , Masculino , Ratas , Ratas Wistar , Núcleos Talámicos/citología
10.
J Neurosci ; 36(13): 3735-54, 2016 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-27030759

RESUMEN

Thalamocortical neurons have thousands of synaptic connections from layer VI corticothalamic neurons distributed across their dendritic trees. Although corticothalamic synapses provide significant excitatory input, it remains unknown how different spatial and temporal input patterns are integrated by thalamocortical neurons. Using dendritic recording, 2-photon glutamate uncaging, and computational modeling, we investigated how rat dorsal lateral geniculate nucleus thalamocortical neurons integrate excitatory corticothalamic feedback. We find that unitary corticothalamic inputs produce small somatic EPSPs whose amplitudes are passively normalized and virtually independent of the site of origin within the dendritic tree. Furthermore, uncaging of MNI glutamate reveals that thalamocortical neurons have postsynaptic voltage-dependent mechanisms that can amplify integrated corticothalamic input. These mechanisms, involving NMDA receptors and T-type Ca(2+)channels, require temporally synchronous synaptic activation but not spatially coincident input patterns. In hyperpolarized thalamocortical neurons, T-type Ca(2+)channels produce nonlinear amplification of temporally synchronous inputs, whereas asynchronous inputs are not amplified. At depolarized potentials, the input-output function for synchronous synaptic input is linear but shows enhanced gain due to activity-dependent recruitment of NMDA receptors. Computer simulations reveal that EPSP amplification by T-type Ca(2+)channels and NMDA receptors occurs when synaptic inputs are either clustered onto individual dendrites or when they are distributed throughout the dendritic tree. Consequently, postsynaptic EPSP amplification mechanisms limit the "modulatory" effects of corticothalamic synaptic inputs on thalamocortical neuron membrane potential and allow these synapses to act as synchrony-dependent "drivers" of thalamocortical neuron firing. These complex thalamocortical input-output transformations significantly increase the influence of corticothalamic feedback on sensory information transfer. SIGNIFICANCE STATEMENT: Neurons in first-order thalamic nuclei transmit sensory information from the periphery to the cortex. However, the numerically dominant synaptic input to thalamocortical neurons comes from the cortex, which provides a strong, activity-dependent modulatory feedback influence on information flow through the thalamus. Here, we reveal how individual quantal-sized corticothalamic EPSPs propagate within thalamocortical neuron dendrites and how different spatial and temporal input patterns are integrated by these cells. We find that thalamocortical neurons have voltage- and synchrony-dependent postsynaptic mechanisms, involving NMDA receptors and T-type Ca(2+)channels that allow nonlinear amplification of integrated corticothalamic EPSPs. These mechanisms significantly increase the responsiveness of thalamocortical neurons to cortical excitatory input and broaden the "modulatory" influence exerted by corticothalamic synapses.


Asunto(s)
Corteza Cerebral/citología , Dendritas/fisiología , Retroalimentación Fisiológica/fisiología , Neuronas/citología , Sinapsis/fisiología , Animales , Animales Recién Nacidos , Calcio/farmacología , Simulación por Computador , Dendritas/efectos de los fármacos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Retroalimentación Fisiológica/efectos de los fármacos , Femenino , Antagonistas del GABA/farmacología , Ácido Glutámico/farmacología , Técnicas In Vitro , Masculino , Modelos Neurológicos , Vías Nerviosas/fisiología , Neuronas/efectos de los fármacos , Ácidos Fosfínicos/farmacología , Propanolaminas/farmacología , Piridazinas/farmacología , Ratas , Ratas Wistar , Sinapsis/efectos de los fármacos , Tálamo/citología
11.
J Neurosci ; 35(47): 15505-22, 2015 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-26609149

RESUMEN

Low-threshold Ca(2+) spikes (LTS) are an indispensible signaling mechanism for neurons in areas including the cortex, cerebellum, basal ganglia, and thalamus. They have critical physiological roles and have been strongly associated with disorders including epilepsy, Parkinson's disease, and schizophrenia. However, although dendritic T-type Ca(2+) channels have been implicated in LTS generation, because the properties of low-threshold spiking neuron dendrites are unknown, the precise mechanism has remained elusive. Here, combining data from fluorescence-targeted dendritic recordings and Ca(2+) imaging from low-threshold spiking cells in rat brain slices with computational modeling, the cellular mechanism responsible for LTS generation is established. Our data demonstrate that key somatodendritic electrical conduction properties are highly conserved between glutamatergic thalamocortical neurons and GABAergic thalamic reticular nucleus neurons and that these properties are critical for LTS generation. In particular, the efficiency of soma to dendrite voltage transfer is highly asymmetric in low-threshold spiking cells, and in the somatofugal direction, these neurons are particularly electrotonically compact. Our data demonstrate that LTS have remarkably similar amplitudes and occur synchronously throughout the dendritic tree. In fact, these Ca(2+) spikes cannot occur locally in any part of the cell, and hence we reveal that LTS are generated by a unique whole-cell mechanism that means they always occur as spatially global spikes. This all-or-none, global electrical and biochemical signaling mechanism clearly distinguishes LTS from other signals, including backpropagating action potentials and dendritic Ca(2+)/NMDA spikes, and has important consequences for dendritic function in low-threshold spiking neurons. SIGNIFICANCE STATEMENT: Low-threshold Ca(2+) spikes (LTS) are critical for important physiological processes, including generation of sleep-related oscillations, and are implicated in disorders including epilepsy, Parkinson's disease, and schizophrenia. However, the mechanism underlying LTS generation in neurons, which is thought to involve dendritic T-type Ca(2+) channels, has remained elusive due to a lack of knowledge of the dendritic properties of low-threshold spiking cells. Combining dendritic recordings, two-photon Ca(2+) imaging, and computational modeling, this study reveals that dendritic properties are highly conserved between two prominent low-threshold spiking neurons and that these properties underpin a whole-cell somatodendritic spike generation mechanism that makes the LTS a unique global electrical and biochemical signal in neurons.


Asunto(s)
Potenciales de Acción/fisiología , Señalización del Calcio/fisiología , Dendritas/fisiología , Neuronas/fisiología , Animales , Calcio/fisiología , Femenino , Cuerpos Geniculados/fisiología , Masculino , Ratas , Ratas Wistar
12.
J Neurosci ; 35(14): 5442-58, 2015 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-25855163

RESUMEN

During sleep and anesthesia, neocortical neurons exhibit rhythmic UP/DOWN membrane potential states. Although UP states are maintained by synaptic activity, the mechanisms that underlie the initiation and robust rhythmicity of UP states are unknown. Using a physiologically validated model of UP/DOWN state generation in mouse neocortical slices whereby the cholinergic tone present in vivo is reinstated, we show that the regular initiation of UP states is driven by an electrophysiologically distinct subset of morphologically identified layer 5 neurons, which exhibit intrinsic rhythmic low-frequency burst firing at ~0.2-2 Hz. This low-frequency bursting is resistant to block of glutamatergic and GABAergic transmission but is absent when slices are maintained in a low Ca(2+) medium (an alternative, widely used model of cortical UP/DOWN states), thus explaining the lack of rhythmic UP states and abnormally prolonged DOWN states in this condition. We also characterized the activity of various other pyramidal and nonpyramidal neurons during UP/DOWN states and found that an electrophysiologically distinct subset of layer 5 regular spiking pyramidal neurons fires earlier during the onset of network oscillations compared with all other types of neurons recorded. This study, therefore, identifies an important role for cell-type-specific neuronal activity in driving neocortical UP states.


Asunto(s)
Potenciales de Acción/fisiología , Ondas Encefálicas/fisiología , Neocórtex/citología , Red Nerviosa/fisiología , Periodicidad , Células Piramidales/fisiología , Potenciales de Acción/efectos de los fármacos , Animales , Biotina/análogos & derivados , Biotina/metabolismo , Ondas Encefálicas/efectos de los fármacos , Calcio/metabolismo , Electroencefalografía , Antagonistas de Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Técnicas In Vitro , Lisina/análogos & derivados , Lisina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Red Nerviosa/efectos de los fármacos , Neurotransmisores/farmacología , Células Piramidales/efectos de los fármacos
13.
Cereb Cortex ; 25(10): 3420-33, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25037920

RESUMEN

The thalamus plays important roles as a relay station for sensory information in the central nervous system (CNS). Although thalamic glial cells participate in this activity, little is known about their properties. In this study, we characterized the formation of coupled networks between astrocytes and oligodendrocytes in the murine ventrobasal thalamus and compared these properties with those in the hippocampus and cortex. Biocytin filling of individual astrocytes or oligodendrocytes revealed large panglial networks in all 3 gray matter regions. Combined analyses of mice with cell type-specific deletion of connexins (Cxs), semiquantitative reverse transcription-polymerase chain reaction (RT-PCR) and western blotting showed that Cx30 is the dominant astrocytic Cx in the thalamus. Many thalamic astrocytes even lack expression of Cx43, while in the hippocampus astrocytic coupling is dominated by Cx43. Deletion of Cx30 and Cx47 led to complete loss of panglial coupling, which was restored when one allele of either Cxs was present. Immunohistochemistry revealed a unique antigen profile of thalamic glia and identified an intermediate cell type expressing both Olig2 and Cx43. Our findings further the emerging concept of glial heterogeneity across brain regions.


Asunto(s)
Astrocitos/metabolismo , Conexina 43/metabolismo , Conexinas/metabolismo , Hipocampo/metabolismo , Neocórtex/metabolismo , Oligodendroglía/metabolismo , Tálamo/metabolismo , Animales , Conexina 30 , Femenino , Hipocampo/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Neocórtex/citología , Red Nerviosa/citología , Red Nerviosa/metabolismo , Tálamo/citología
14.
J Neurosci ; 33(9): 3780-5, 2013 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-23447590

RESUMEN

Tonic inhibitory GABA(A) receptor-mediated currents are observed in numerous cell types in the CNS, including thalamocortical neurons of the ventrobasal thalamus, dentate gyrus granule cells, and cerebellar granule cells. Here we show that in rat brain slices, activation of postsynaptic GABA(B) receptors enhances the magnitude of the tonic GABA(A) current recorded in these cell types via a pathway involving G G proteins, adenylate cyclase, and cAMP-dependent protein kinase. Using a combination of pharmacology and knockout mice, we show that this pathway is independent of potassium channels or GABA transporters. Furthermore, the enhancement in tonic current is sufficient to significantly alter the excitability of thalamocortical neurons. These results demonstrate for the first time a postsynaptic crosstalk between GABA(B) and GABA(A) receptors.


Asunto(s)
Encéfalo/citología , Neuronas/fisiología , Receptores de GABA-A/metabolismo , Receptores de GABA-B/fisiología , Sinapsis/fisiología , Adenina/análogos & derivados , Adenina/farmacología , Animales , Biofisica , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/farmacología , Interacciones Farmacológicas , Estimulación Eléctrica , Inhibidores Enzimáticos/farmacología , Femenino , GABAérgicos/farmacología , Guanosina Difosfato/análogos & derivados , Guanosina Difosfato/farmacología , Técnicas In Vitro , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Inhibición Neural/efectos de los fármacos , Inhibición Neural/genética , Neuronas/efectos de los fármacos , Técnicas de Placa-Clamp , Ratas , Ratas Wistar , Receptores de GABA-A/deficiencia , Receptores de GABA-B/deficiencia , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Bloqueadores de los Canales de Sodio/farmacología , Sinapsis/efectos de los fármacos , Tetrodotoxina/farmacología , Tionucleótidos/farmacología
15.
J Neurosci ; 33(50): 19599-610, 2013 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-24336724

RESUMEN

Slow waves represent one of the prominent EEG signatures of non-rapid eye movement (non-REM) sleep and are thought to play an important role in the cellular and network plasticity that occurs during this behavioral state. These slow waves of natural sleep are currently considered to be exclusively generated by intrinsic and synaptic mechanisms within neocortical territories, although a role for the thalamus in this key physiological rhythm has been suggested but never demonstrated. Combining neuronal ensemble recordings, microdialysis, and optogenetics, here we show that the block of the thalamic output to the neocortex markedly (up to 50%) decreases the frequency of slow waves recorded during non-REM sleep in freely moving, naturally sleeping-waking rats. A smaller volume of thalamic inactivation than during sleep is required for observing similar effects on EEG slow waves recorded during anesthesia, a condition in which both bursts and single action potentials of thalamocortical neurons are almost exclusively dependent on T-type calcium channels. Thalamic inactivation more strongly reduces spindles than slow waves during both anesthesia and natural sleep. Moreover, selective excitation of thalamocortical neurons strongly entrains EEG slow waves in a narrow frequency band (0.75-1.5 Hz) only when thalamic T-type calcium channels are functionally active. These results demonstrate that the thalamus finely tunes the frequency of slow waves during non-REM sleep and anesthesia, and thus provide the first conclusive evidence that a dynamic interplay of the neocortical and thalamic oscillators of slow waves is required for the full expression of this key physiological EEG rhythm.


Asunto(s)
Potenciales de Acción/fisiología , Neuronas/fisiología , Sueño/fisiología , Tálamo/fisiología , Animales , Canales de Calcio Tipo T/metabolismo , Corteza Cerebral/fisiología , Electroencefalografía , Masculino , Ratas , Ratas Wistar
16.
Pflugers Arch ; 466(4): 735-45, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24578015

RESUMEN

Since their discovery more than 30 years ago, low-threshold T-type Ca(2+) channels (T channels) have been suggested to play a key role in many EEG waves of non-REM sleep, which has remained exclusively linked to the ability of these channels to generate low-threshold Ca(2+) potentials and associated high-frequency bursts of action potentials. Our present understanding of the biophysics and physiology of T channels, however, highlights a much more diverse and complex picture of the pivotal contributions that they make to different sleep rhythms. In particular, recent experimental evidence has conclusively demonstrated the essential contribution of thalamic T channels to the expression of slow waves of natural sleep and the key role played by Ca(2+) entry through these channels in the activation or modulation of other voltage-dependent channels that are important for the generation of both slow waves and sleep spindles. However, the precise contribution to sleep rhythms of T channels in cortical neurons and other sleep-controlling neuronal networks remains unknown, and a full understanding of the cellular and network mechanisms of sleep delta waves is still lacking.


Asunto(s)
Potenciales de Acción/fisiología , Encéfalo/fisiología , Canales de Calcio Tipo T/fisiología , Fases del Sueño/fisiología , Animales , Electroencefalografía/métodos , Humanos , Red Nerviosa/fisiología
17.
Pflugers Arch ; 466(3): 415-23, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24043572

RESUMEN

Since the discovery of low-voltage-activated T-type calcium channels in sensory neurons and the initial characterization of their physiological function mainly in inferior olive and thalamic neurons, studies on neuronal T-type currents have predominantly focused on the generation of low-threshold spike (and associated action potential burst firing) which is strictly conditioned by a preceding hyperpolarization. This T-type current mediated activity has become an archetype of the function of these channels, constraining our view of the potential physiological and pathological roles that they may play in controlling the excitability of single cells and neural networks. However, greatly helped by the recent availability of the first potent and selective antagonists for this class of calcium channels, novel T-type current functions are rapidly being uncovered, including their surprising involvement in neuronal excitability at depolarized membrane potentials and their complex control of dendritic integration and neurotransmitter release. These and other data summarized in this short review clearly indicate a much wider physiological involvement of T-type channels in neuronal activity than previously expected.


Asunto(s)
Potenciales de Acción , Canales de Calcio Tipo T/metabolismo , Animales , Bloqueadores de los Canales de Calcio/farmacología , Humanos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/fisiología , Potenciales Sinápticos
18.
Rev Neurosci ; 25(2): 195-206, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24468610

RESUMEN

In recent years, it has become evident that many neurotransmitters and endogenous ligands differentially modulate synaptic γ-aminobutyric acid type A receptors (sGABAARs) and extrasynaptic GABAAR (eGABAARs). In this mini-review, we will summarize the available evidence on the ability of the monoamines serotonin (5-HT), noradrenaline (NA), and, in particular, dopamine (DA) to alter the functional response of eGABAARs, thus either increasing or decreasing tonic GABAA inhibition. Although this field of research is still in its infancy, it has already been demonstrated that eGABAARs show a nucleus-selective and neuronal-type-selective regulation by monoamines in a way that differs from that of sGABAARs. Further work will undoubtedly advance our knowledge of the intricate talk between monoamines and eGABAAR and may ultimately provide new leads for the treatment of neurological and neuropsychiatric disorders, where alteration in GABAAR function is one of the underlying causes.


Asunto(s)
Monoaminas Biogénicas/metabolismo , Encéfalo/metabolismo , Dopamina/metabolismo , Norepinefrina/metabolismo , Receptores de GABA-A/metabolismo , Serotonina/metabolismo , Animales , Encéfalo/efectos de los fármacos , Dopamina/farmacología , Humanos , Neurotransmisores/metabolismo , Neurotransmisores/farmacología , Norepinefrina/farmacología , Serotonina/farmacología
19.
CNS Neurosci Ther ; 30(3): e14206, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-37072918

RESUMEN

AIM: Many biophysical and non-biophysical models have been able to reproduce the corticothalamic activities underlying different EEG sleep rhythms but none of them included the known ability of neocortical networks and single thalamic neurons to generate some of these waves intrinsically. METHODS: We built a large-scale corticothalamic model with a high fidelity in anatomical connectivity consisting of a single cortical column and first- and higher-order thalamic nuclei. The model is constrained by different neocortical excitatory and inhibitory neuronal populations eliciting slow (<1 Hz) oscillations and by thalamic neurons generating sleep waves when isolated from the neocortex. RESULTS: Our model faithfully reproduces all EEG sleep waves and the transition from a desynchronized EEG to spindles, slow (<1 Hz) oscillations, and delta waves by progressively increasing neuronal membrane hyperpolarization as it occurs in the intact brain. Moreover, our model shows that slow (<1 Hz) waves most often start in a small assembly of thalamocortical neurons though they can also originate in cortical layer 5. Moreover, the input of thalamocortical neurons increases the frequency of EEG slow (<1 Hz) waves compared to those generated by isolated cortical networks. CONCLUSION: Our simulations challenge current mechanistic understanding of the temporal dynamics of sleep wave generation and suggest testable predictions.


Asunto(s)
Corteza Cerebral , Neocórtex , Corteza Cerebral/fisiología , Electroencefalografía , Tálamo , Sueño/fisiología , Neuronas/fisiología
20.
CNS Neurosci Ther ; 30(3): e14204, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-37032628

RESUMEN

AIMS: Recurrent network activity in corticothalamic circuits generates physiological and pathological EEG waves. Many computer models have simulated spike-and-wave discharges (SWDs), the EEG hallmark of absence seizures (ASs). However, these models either provided detailed simulated activity only in a selected territory (i.e., cortical or thalamic) or did not test whether their corticothalamic networks could reproduce the physiological activities that are generated by these circuits. METHODS: Using a biophysical large-scale corticothalamic model that reproduces the full extent of EEG sleep waves, including sleep spindles, delta, and slow (<1 Hz) waves, here we investigated how single abnormalities in voltage- or transmitter-gated channels in the neocortex or thalamus led to SWDs. RESULTS: We found that a selective increase in the tonic γ-aminobutyric acid type A receptor (GABA-A) inhibition of first-order thalamocortical (TC) neurons or a selective decrease in cortical phasic GABA-A inhibition is sufficient to generate ~4 Hz SWDs (as in humans) that invariably start in neocortical territories. Decreasing the leak conductance of higher-order TC neurons leads to ~7 Hz SWDs (as in rodent models) while maintaining sleep spindles at 7-14 Hz. CONCLUSION: By challenging key features of current mechanistic views, this simulated ictal corticothalamic activity provides novel understanding of ASs and makes key testable predictions.


Asunto(s)
Corteza Cerebral , Epilepsia Tipo Ausencia , Humanos , Corteza Cerebral/patología , Electroencefalografía , Tálamo , Sueño/fisiología , Convulsiones/patología , Ácido gamma-Aminobutírico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA