Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
Mol Psychiatry ; 27(10): 4201-4217, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35821415

RESUMEN

The mechanisms underlying the dichotomic cortical/basal ganglia dopaminergic abnormalities in schizophrenia are unclear. Astrocytes are important non-neuronal modulators of brain circuits, but their role in dopaminergic system remains poorly explored. Microarray analyses, immunohistochemistry, and two-photon laser scanning microscopy revealed that Dys1 hypofunction increases the reactivity of astrocytes, which express only the Dys1A isoform. Notably, behavioral and electrochemical assessments in mice selectively lacking the Dys1A isoform unraveled a more prominent impact of Dys1A in behavioral and dopaminergic/D2 alterations related to basal ganglia, but not cortical functioning. Ex vivo electron microscopy and protein expression analyses indicated that selective Dys1A disruption might alter intracellular trafficking in astrocytes, but not in neurons. In agreement, Dys1A disruption only in astrocytes resulted in decreased motivation and sensorimotor gating deficits, increased astrocytic dopamine D2 receptors and decreased dopaminergic tone within basal ganglia. These processes might have clinical relevance because the caudate, but not the cortex, of patients with schizophrenia shows a reduction of the Dys1A isoform. Therefore, we started to show a hitherto unknown role for the Dys1A isoform in astrocytic-related modulation of basal ganglia behavioral and dopaminergic phenotypes, with relevance to schizophrenia.


Asunto(s)
Dopamina , Disbindina , Esquizofrenia , Animales , Ratones , Astrocitos/metabolismo , Ganglios Basales/metabolismo , Dopamina/metabolismo , Disbindina/metabolismo , Esquizofrenia/genética
2.
Aging Clin Exp Res ; 33(6): 1713-1716, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31732960

RESUMEN

New onset epilepsy and seizures are common neurological disorders in aged people, second only to stroke and dementia. They are frequently related to other pathological conditions including stroke, trauma, tumors and neurological diseases whereas in about one-third of cases the origin is unknown. Besides the origin, the cellular and molecular events that suddenly trigger seizures are poorly defined. Using an acute model of seizure generation that better resembles new onset seizures, we studied GABAergic interneurons and astrocytes during seizure generation. We found that seizures are preceded by a GABAergic rhythmic hyperactivity that synchronizes pyramidal neurons by inducing a rebound spiking that favors seizures' onset. Furthermore, the intense activity in GABAergic interneurons evokes Ca2+ elevations in astrocytes that, by releasing glutamate, further excite neuronal network. Elucidating the cellular and molecular events that generate seizures may reveal new targets for treatment of new onset seizures and epilepsy.


Asunto(s)
Epilepsia , Convulsiones , Anciano , Humanos , Interneuronas , Neuronas
3.
J Neurosci ; 37(43): 10398-10407, 2017 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-28947576

RESUMEN

The onset of focal seizures in humans and in different animal models of focal epilepsy correlates with reduction of neuronal firing and enhanced interneuronal network activity. Whether this phenomenon contributes to seizure generation is still unclear. We used the in vitro entorhinal cortex slices bathed in 4-aminopirydine (4-AP) as an experimental paradigm model to evaluate the correlation between interneuronal GABAergic network activity and seizure-like events. Epileptiform discharges were recorded in layer V-VI pyramidal neurons and fast-spiking interneurons in slices from male and female mice and in the isolated female guinea pig brain preparation during perfusion with 4-AP. We observed that 90% of seizure-like events recorded in principal cells were preceded by outward currents coupled with extracellular potassium shifts, abolished by pharmacological blockade of GABAA receptors. Potassium elevations associated to GABAA receptor-mediated population events were confirmed in the entorhinal cortex of the in vitro isolated whole guinea pig brain. Fast-rising and sustained extracellular potassium increases associated to interneuronal network activity consistently preceded the initiation of seizure-like events. We conclude that in the 4-AP seizure model, interneuronal network activity occurs before 4-AP-induced seizures and therefore supports a role of interneuron activity in focal seizure generation.SIGNIFICANCE STATEMENT The paper focuses on the mechanisms of ictogenesis, a topic that requires a step beyond the simplistic view that seizures, and epilepsy, are due to an increase of excitatory network activity. Focal temporal lobe seizures in humans and in several experimental epilepsies likely correlate with a prevalent activation of interneurons. The potassium channel blocker 4-aminopyridine reliably induces seizure-like events in temporal lobe structures. Herein, we show that a majority of seizures in the entorhinal cortex starts with interneuronal network activity accompanied by a fast and sustained increase in extracellular potassium. Our new findings reinforce and add a new piece of evidence to the proposal that limbic seizures can be supported by GABAergic hyperactivity.


Asunto(s)
Potenciales de Acción/fisiología , Corteza Entorrinal/fisiología , Interneuronas/fisiología , Red Nerviosa/fisiología , Convulsiones/fisiopatología , Animales , Femenino , Cobayas , Potenciales Postsinápticos Inhibidores/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Técnicas de Cultivo de Órganos
4.
Glia ; 66(10): 2188-2199, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30144319

RESUMEN

The gliotransmitter glutamate in different brain regions modulates neuronal excitability and synaptic transmission through a variety of mechanisms. Among the hallmarks of astrocytic glutamate release are the slow depolarizing inward currents (SICs) in neurons mediated by N-methyl-d-aspartate receptor activation. Different stimuli that evoke Ca2+ elevations in astrocytes induce neuronal SICs suggesting a Ca2+ -dependent exocytotic glutamate release mechanism of SIC generation. To gain new insights into this mechanism, we investigated the relationship between astrocytic Ca2+ elevations and neuronal SICs in mouse hippocampal slice preparations. Here we provide evidence that SICs, occurring either spontaneously or following a hypotonic challenge, are unchanged in the virtual absence of Ca2+ signal changes at astrocytic soma and processes, including spatially restricted Ca2+ microdomains. SICs are also unchanged in the presence of Bafilomycin A1 that after prolonged slice incubation depletes glutamate from astrocytic vesicles. We also found that hemichannels and TREK family channels-that recent studies proposed to mediate astrocytic glutamate release - are not involved in SIC generation. SICs are reduced by the volume-sensitive anion channel antagonists diisothiocyanatostilbene-2,2'-disulfonic acid (DIDS), quinine and fluoxetine, suggesting a possible contribution of these channels in SIC generation. Direct measurements of astrocytic glutamate release further confirm that hypotonicity-evoked gliotransmission is impaired following DIDS, quinine and fluoxetine while the exocytotic release of glutamate-that is proposed to mediate synaptic transmission modulation by astrocytes-remains unaffected. In conclusion, our data provide evidence that the release of glutamate generating SICs occurs independently on exocytotic Ca2+ -dependent glutamate release mechanism.


Asunto(s)
Astrocitos/metabolismo , Ácido Glutámico/metabolismo , Potenciales de la Membrana/fisiología , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transmisión Sináptica/fisiología , Animales , Astrocitos/efectos de los fármacos , Calcio/metabolismo , Cationes Bivalentes/metabolismo , Fármacos del Sistema Nervioso Central/farmacología , Exocitosis/fisiología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/efectos de los fármacos , Canales de Potasio de Dominio Poro en Tándem/genética , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Transmisión Sináptica/efectos de los fármacos , Técnicas de Cultivo de Tejidos
5.
Cereb Cortex ; 26(4): 1778-94, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26819275

RESUMEN

Severe myoclonic epilepsy of infancy (SMEI) is associated with loss of function of the SCN1A gene encoding the NaV1.1 sodium channel isoform. Previous studies in Scn1a(-/+) mice during the pre-epileptic period reported selective reduction in interneuron excitability and proposed this as the main pathological mechanism underlying SMEI. Yet, the functional consequences of this interneuronal dysfunction at the circuit level in vivo are unknown. Here, we investigated whether Scn1a(-/+) mice showed alterations in cortical network function. We found that various forms of spontaneous network activity were similar in Scn1a(-/+) during the pre-epileptic period compared with wild-type (WT) in vivo. Importantly, in brain slices from Scn1a(-/+) mice, the excitability of parvalbumin (PV) and somatostatin (SST) interneurons was reduced, epileptiform activity propagated more rapidly, and complex synaptic changes were observed. However, in vivo, optogenetic reduction of firing in PV or SST cells in WT mice modified ongoing network activities, and juxtasomal recordings from identified PV and SST interneurons showed unaffected interneuronal firing during spontaneous cortical dynamics in Scn1a(-/+) compared with WT. These results demonstrate that interneuronal hypoexcitability is not observed in Scn1a(-/+) mice during spontaneous activities in vivo and suggest that additional mechanisms may contribute to homeostatic rearrangements and the pathogenesis of SMEI.


Asunto(s)
Corteza Cerebral/fisiopatología , Interneuronas/fisiología , Síndrome de Opsoclonía-Mioclonía/fisiopatología , Potenciales de Acción , Animales , Ondas Encefálicas , Modelos Animales de Enfermedad , Femenino , Interneuronas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Canal de Sodio Activado por Voltaje NAV1.1/genética , Vías Nerviosas/fisiopatología , Síndrome de Opsoclonía-Mioclonía/genética , Parvalbúminas/metabolismo , Somatostatina/metabolismo , Potenciales Sinápticos
6.
J Neurosci ; 35(26): 9544-57, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-26134638

RESUMEN

Parvalbumin (Pv)-positive inhibitory interneurons effectively control network excitability, and their optogenetic activation has been reported to block epileptic seizures. An intense activity in GABAergic interneurons, including Pv interneurons, before seizures has been described in different experimental models of epilepsy, raising the hypothesis that an increased GABAergic inhibitory signal may, under certain conditions, initiate seizures. It is therefore unclear whether the activity of Pv interneurons enhances or opposes epileptiform activities. Here we use a mouse cortical slice model of focal epilepsy in which the epileptogenic focus can be identified and the role of Pv interneurons in the generation and propagation of seizure-like ictal events is accurately analyzed by a combination of optogenetic, electrophysiological, and imaging techniques. We found that a selective activation of Pv interneurons at the focus failed to block ictal generation and induced postinhibitory rebound spiking in pyramidal neurons, enhancing neuronal synchrony and promoting ictal generation. In contrast, a selective activation of Pv interneurons distant from the focus blocked ictal propagation and shortened ictal duration at the focus. We revealed that the reduced ictal duration was a direct consequence of the ictal propagation block, probably by preventing newly generated afterdischarges to travel backwards to the original focus of ictal initiation. Similar results were obtained upon individual Pv interneuron activation by intracellular depolarizing current pulses. The functional dichotomy of Pv interneurons here described opens new perspectives to our understanding of how local inhibitory circuits govern generation and spread of focal epileptiform activities.


Asunto(s)
Potenciales de Acción/fisiología , Encéfalo/citología , Interneuronas/metabolismo , Inhibición Neural/fisiología , Parvalbúminas/metabolismo , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/genética , Animales , Animales Recién Nacidos , Calcio/metabolismo , Channelrhodopsins , Antagonistas de Aminoácidos Excitadores/farmacología , Femenino , Técnicas In Vitro , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Ratones , Ratones Transgénicos , N-Metilaspartato/farmacología , Inhibición Neural/efectos de los fármacos , Parvalbúminas/genética , Estimulación Luminosa , Células Piramidales/efectos de los fármacos , Células Piramidales/fisiología , Transducción Genética , Ácido gamma-Aminobutírico/metabolismo
7.
Glia ; 64(3): 363-73, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26496414

RESUMEN

Studies over the last decade provided evidence that in a dynamic interaction with neurons glial cell astrocytes contribut to fundamental phenomena in the brain. Most of the knowledge on this derives, however, from studies monitoring the astrocyte Ca(2+) response to glutamate. Whether astrocytes can similarly respond to other neurotransmitters, including the inhibitory neurotransmitter GABA, is relatively unexplored. By using confocal and two photon laser-scanning microscopy the astrocyte response to GABA in the mouse somatosensory and temporal cortex was studied. In slices from developing (P15-20) and adult (P30-60) mice, it was found that in a subpopulation of astrocytes GABA evoked somatic Ca(2+) oscillations. This response was mediated by GABAB receptors and involved both Gi/o protein and inositol 1,4,5-trisphosphate (IP3 ) signalling pathways. In vivo experiments from young adult mice, revealed that also cortical astrocytes in the living brain exibit GABAB receptor-mediated Ca(2+) elevations. At all astrocytic processes tested, local GABA or Baclofen brief applications induced long-lasting Ca(2+) oscillations, suggesting that all astrocytes have the potential to respond to GABA. Finally, in patch-clamp recordings it was found that Ca(2+) oscillations induced by Baclofen evoked astrocytic glutamate release and slow inward currents (SICs) in pyramidal cells from wild type but not IP3 R2(-/-) mice, in which astrocytic GABAB receptor-mediated Ca(2+) elevations are impaired. These data suggest that cortical astrocytes in the mouse brain can sense the activity of GABAergic interneurons and through their specific recruitment contribut to the distinct role played on the cortical network by the different subsets of GABAergic interneurons.


Asunto(s)
Astrocitos/efectos de los fármacos , Señalización del Calcio/efectos de los fármacos , Calcio/metabolismo , Corteza Cerebral/citología , Inhibición Neural/efectos de los fármacos , Ácido gamma-Aminobutírico/farmacología , Factores de Edad , Animales , Animales Recién Nacidos , Astrocitos/metabolismo , Relojes Biológicos/efectos de los fármacos , Relojes Biológicos/genética , Señalización del Calcio/fisiología , Fármacos actuantes sobre Aminoácidos Excitadores/farmacología , Transportador 1 de Aminoácidos Excitadores/genética , Transportador 1 de Aminoácidos Excitadores/metabolismo , Femenino , GABAérgicos/farmacología , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Inhibición Neural/genética , Neuroimagen , Neuronas/efectos de los fármacos , Neuronas/fisiología , Rodaminas/farmacocinética , Bloqueadores de los Canales de Sodio/farmacología , Tetrodotoxina/farmacología
8.
J Physiol ; 591(4): 775-85, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23230232

RESUMEN

Our current knowledge of the role of astrocytes in health and disease states supports the view that many physiological brain functions and neurological diseases are finely tuned, and in certain cases fully determined, by the continuous cross-talk between astrocytes and neurons. This novel way of interpreting brain activity as a dynamic and reciprocal interplay between astrocytic and neuronal networks has also influenced our understanding of epilepsy, not only forcing a reinterpretation of old findings, but also being a catalyst for novel experimentation. In this review, we summarize some of the recent studies that highlight these novel distinct contributions of astrocytes to the expression of convulsive and non-convulsive epileptiform discharges and seizures. The emerging picture suggests a general framework based on bilateral signalling between astrocytes and neurons for a fuller understanding of epileptogenic and epileptic mechanisms in the brain network. Astrocytes potentially represent targets for the development of those novel chemical entities with improved efficacy for the treatment of convulsive and non-convulsive epilepsy that expert groups have recognized as one of the key priorities for the management of epilepsy.


Asunto(s)
Astrocitos/fisiología , Epilepsia/fisiopatología , Animales , Humanos
9.
J Physiol ; 591(4): 807-22, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23207591

RESUMEN

In different animal models of focal epilepsy, seizure-like ictal discharge propagation is transiently opposed by feedforward inhibition. The specific cellular source of this signal and the mechanism by which inhibition ultimately becomes ineffective are, however, undefined. We used a brain slice model to study how focal ictal discharges that were repetitively evoked from the same site, and at precise times, propagate across the cortex. We used Ca(2+) imaging and simultaneous single/dual cell recordings from pyramidal neurons (PyNs) and different classes of interneurons in rodents, including G42 and GIN transgenic mice expressing the green fluorescence protein in parvalbumin (Pv)-fast spiking (FS) and somatostatin (Som) interneurons, respectively. We found that these two classes of interneurons fired intensively shortly after ictal discharge generation at the focus. The inhibitory barrages that were recorded in PyNs occurred in coincidence with Pv-FS, but not with Som interneuron burst discharges. Furthermore, the strength of inhibitory barrages increased or decreased in parallel with increased or decreased firing in Pv-FS interneurons but not in Som interneurons. A firing impairment of Pv-FS interneurons caused by a membrane depolarization was found to precede ictal discharge onset in neighbouring pyramidal neurons. This event may account for the collapse of local inhibition that allows spatially defined clusters of PyNs to be recruited into propagating ictal discharges. Our study demonstrates that Pv-FS interneurons are a major source of the inhibitory barrages that oppose ictal discharge propagation and raises the possibility that targeting Pv-FS interneurons represents a new therapeutic strategy to prevent the generalization of human focal seizures.


Asunto(s)
Corteza Cerebral/fisiopatología , Epilepsias Parciales/fisiopatología , Interneuronas/fisiología , Convulsiones/fisiopatología , Animales , Calcio/fisiología , Modelos Animales de Enfermedad , Neuronas GABAérgicas/fisiología , Técnicas In Vitro , Ratones , Ratones Transgénicos , Técnicas de Placa-Clamp , Células Piramidales/fisiología , Ratas , Ratas Wistar
10.
PLoS Biol ; 8(4): e1000352, 2010 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-20405049

RESUMEN

Seizures in focal epilepsies are sustained by a highly synchronous neuronal discharge that arises at restricted brain sites and subsequently spreads to large portions of the brain. Despite intense experimental research in this field, the earlier cellular events that initiate and sustain a focal seizure are still not well defined. Their identification is central to understand the pathophysiology of focal epilepsies and to develop new pharmacological therapies for drug-resistant forms of epilepsy. The prominent involvement of astrocytes in ictogenesis was recently proposed. We test here whether a cooperation between astrocytes and neurons is a prerequisite to support ictal (seizure-like) and interictal epileptiform events. Simultaneous patch-clamp recording and Ca2+ imaging techniques were performed in a new in vitro model of focal seizures induced by local applications of N-methyl-D-aspartic acid (NMDA) in rat entorhinal cortex slices. We found that a Ca2+ elevation in astrocytes correlates with both the initial development and the maintenance of a focal, seizure-like discharge. A delayed astrocyte activation during ictal discharges was also observed in other models (including the whole in vitro isolated guinea pig brain) in which the site of generation of seizure activity cannot be precisely monitored. In contrast, interictal discharges were not associated with Ca2+ changes in astrocytes. Selective inhibition or stimulation of astrocyte Ca2+ signalling blocked or enhanced, respectively, ictal discharges, but did not affect interictal discharge generation. Our data reveal that neurons engage astrocytes in a recurrent excitatory loop (possibly involving gliotransmission) that promotes seizure ignition and sustains the ictal discharge. This neuron-astrocyte interaction may represent a novel target to develop effective therapeutic strategies to control seizures.


Asunto(s)
Potenciales de Acción/fisiología , Astrocitos/fisiología , Convulsiones/fisiopatología , 4-Aminopiridina/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Calcio/metabolismo , Quelantes/metabolismo , Ácido Egtácico/análogos & derivados , Ácido Egtácico/metabolismo , Corteza Entorrinal/citología , Corteza Entorrinal/fisiopatología , Agonistas de Aminoácidos Excitadores/metabolismo , Ácido Glutámico/metabolismo , Cobayas , Ratones , Ratones Transgénicos , N-Metilaspartato/metabolismo , Neuronas/fisiología , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/metabolismo , Ratas , Ratas Wistar
11.
Proc Natl Acad Sci U S A ; 107(32): 14140-5, 2010 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-20660736

RESUMEN

Myotoxins play a major role in the pathogenesis of the envenomations caused by snake bites in large parts of the world where this is a very relevant public health problem. We show here that two myotoxins that are major constituents of the venom of Bothrops asper, a deadly snake present in Latin America, induce the release of large amounts of K(+) and ATP from skeletal muscle. We also show that the released ATP amplifies the effect of the myotoxins, acting as a "danger signal," which spreads and causes further damage by acting on purinergic receptors. In addition, the release of ATP and K(+) well accounts for the pain reaction characteristic of these envenomations. As Bothrops asper myotoxins are representative of a large family of snake myotoxins with phospholipase A(2) structure, these findings are expected to be of general significance for snake bite envenomation. Moreover, they suggest potential therapeutic approaches for limiting the extent of muscle tissue damage based on antipurinergic drugs.


Asunto(s)
Adenosina Trifosfato/metabolismo , Bothrops , Venenos de Crotálidos/metabolismo , Músculo Esquelético/metabolismo , Potasio/metabolismo , Animales , Dolor/etiología , Fosfolipasas A2 , Receptores Purinérgicos
12.
Neuron ; 111(7): 920-921, 2023 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-37023713

RESUMEN

In this issue of Neuron, Yang et al.1 highlight a hitherto unknown action of cocaine in VTA circuitry. They found that chronic cocaine use increased tonic inhibition selectively onto GABA neurons through Swell1 channel-dependent GABA release from astrocytes, leading to disinhibition-mediated hyperactivity in DA neurons and addictive behavior.


Asunto(s)
Trastornos Relacionados con Cocaína , Cocaína , Humanos , Astrocitos , Área Tegmental Ventral/fisiología , Cocaína/farmacología , Neuronas GABAérgicas , Neuronas Dopaminérgicas/fisiología
13.
Nat Commun ; 14(1): 1590, 2023 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-36949142

RESUMEN

Calcium dynamics in astrocytes represent a fundamental signal that through gliotransmitter release regulates synaptic plasticity and behaviour. Here we present a longitudinal study in the PS2APP mouse model of Alzheimer's disease (AD) linking astrocyte Ca2+ hypoactivity to memory loss. At the onset of plaque deposition, somatosensory cortical astrocytes of AD female mice exhibit a drastic reduction of Ca2+ signaling, closely associated with decreased endoplasmic reticulum Ca2+ concentration and reduced expression of the Ca2+ sensor STIM1. In parallel, astrocyte-dependent long-term synaptic plasticity declines in the somatosensory circuitry, anticipating specific tactile memory loss. Notably, we show that both astrocyte Ca2+ signaling and long-term synaptic plasticity are fully recovered by selective STIM1 overexpression in astrocytes. Our data unveil astrocyte Ca2+ hypoactivity in neocortical astrocytes as a functional hallmark of early AD stages and indicate astrocytic STIM1 as a target to rescue memory deficits.


Asunto(s)
Enfermedad de Alzheimer , Ratones , Femenino , Animales , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Calcio/metabolismo , Astrocitos/metabolismo , Estudios Longitudinales , Plasticidad Neuronal/fisiología , Trastornos de la Memoria/metabolismo , Señalización del Calcio/fisiología , Molécula de Interacción Estromal 1/genética , Molécula de Interacción Estromal 1/metabolismo
14.
Glia ; 60(8): 1227-33, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22389222

RESUMEN

Studies performed over the last decade, in both animal models and human epilepsy, support the view that a defective K(+) buffering due to an altered expression of K(+) and aquaporin channels in astrocytes represents a possible causative factor of the pathological neuronal hyperexcitability in the epileptic brain. More recent studies, however, reappraised the role of neurons in epileptogenesis and suggested that Ca(2+)-dependent gliotransmission directly contributes to the excessive neuronal synchronization that predisposes the brain network to seizures. Significant support for this view comes from the finding that astrocytes from hyperexcitable networks respond to neuronal signals with massive Ca(2+) elevations and generate a recurrent excitatory loop with neurons that has the potential to promote a focal seizure. The specific aim of this review is on the one hand, to provide an overview of the experimental findings that hinted at a direct role of Ca(2+)-dependent gliotransmission in the generation of seizure-like discharges in models of focal epilepsy; and on the other hand, to emphasize the importance of developing new experimental tools that could help us to understand the amazing complexity of neuron-astrocyte partnership in brain disorders.


Asunto(s)
Astrocitos/fisiología , Señalización del Calcio/fisiología , Calcio/metabolismo , Epilepsia/patología , Animales , Humanos
15.
Cells ; 11(9)2022 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-35563706

RESUMEN

At glutamatergic synapses, astrocytes respond to the neurotransmitter glutamate with intracellular Ca2+ elevations and the release of gliotransmitters that modulate synaptic transmission. While the functional interactions between neurons and astrocytes have been intensively studied at glutamatergic synapses, the role of astrocytes at GABAergic synapses has been less investigated. In the present study, we combine optogenetics with 2-photon Ca2+ imaging experiments and patch-clamp recording techniques to investigate the signaling between Somatostatin (SST)-releasing GABAergic interneurons and astrocytes in brain slice preparations from the visual cortex (VCx). We found that an intense stimulation of SST interneurons evokes Ca2+ elevations in astrocytes that fundamentally depend on GABAB receptor (GABABR) activation, and that this astrocyte response is modulated by the neuropeptide somatostatin. After episodes of SST interneuron hyperactivity, we also observed a long-lasting reduction of the inhibitory postsynaptic current (IPSC) amplitude onto pyramidal neurons (PNs). This reduction of inhibitory tone (i.e., disinhibition) is counterbalanced by the activation of astrocytes that upregulate SST interneuron-evoked IPSC amplitude by releasing ATP that, after conversion to adenosine, activates A1Rs. Our results describe a hitherto unidentified modulatory mechanism of inhibitory transmission to VCx layer II/III PNs that involves the functional recruitment of astrocytes by SST interneuron signaling.


Asunto(s)
Astrocitos , Corteza Visual , Astrocitos/metabolismo , Interneuronas/metabolismo , Somatostatina/metabolismo , Ácido gamma-Aminobutírico
16.
Front Cell Neurosci ; 16: 919493, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35936501

RESUMEN

Dravet Syndrome (DS) is a rare autosomic encephalopathy with epilepsy linked to Nav1.1 channel mutations and defective GABAergic signaling. Effective therapies for this syndrome are lacking, urging a better comprehension of the mechanisms involved. In a recognized mouse model of DS, we studied GABA tonic current, a form of inhibition largely neglected in DS, in brain slices from developing mice before spontaneous seizures are reported. In neurons from the temporal cortex (TeCx) and CA1 region, GABA tonic current was reduced in DS mice compared to controls, while in the entorhinal cortex (ECx) it was not affected. In this region however allopregnanonole potentiation of GABA tonic current was reduced in DS mice, suggesting altered extrasynaptic GABAA subunits. Using THIP as a selective agonist, we found reduced δ subunit mediated tonic currents in ECx of DS mice. Unexpectedly in the dentate gyrus (DG), a region with high δ subunit expression, THIP-evoked currents in DS mice were larger than in controls. An immunofluorescence study confirmed that δ subunit expression was reduced in ECx and increased in DG of DS mice. Finally, considering the importance of neuroinflammation in epilepsy and neurodevelopmental disorders, we evaluated classical markers of glia activation. Our results show that DS mice have increased Iba1 reactivity and GFAP expression in both ECx and DG, compared to controls. Altogether we report that before spontaneous seizures, DS mice develop significant alterations of GABA tonic currents and glial cell activation. Understanding all the mechanisms involved in these alterations during disease maturation and progression may unveil new therapeutic targets.

18.
Nat Neurosci ; 25(12): 1639-1650, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36396976

RESUMEN

The plasticity of glutamatergic transmission in the ventral tegmental area (VTA) represents a fundamental mechanism in the modulation of dopamine neuron burst firing and phasic dopamine release at target regions. These processes encode basic behavioral responses, including locomotor activity, learning and motivated behaviors. Here we describe a hitherto unidentified mechanism of long-term synaptic plasticity in mouse VTA. We found that the burst firing in individual dopamine neurons induces a long-lasting potentiation of excitatory synapses on adjacent dopamine neurons that crucially depends on Ca2+ elevations in astrocytes, mediated by endocannabinoid CB1 and dopamine D2 receptors co-localized at the same astrocytic process, and activation of pre-synaptic metabotropic glutamate receptors. Consistent with these findings, selective in vivo activation of astrocytes increases the burst firing of dopamine neurons in the VTA and induces locomotor hyperactivity. Astrocytes play, therefore, a key role in the modulation of VTA dopamine neuron functional activity.


Asunto(s)
Neuronas Dopaminérgicas , Área Tegmental Ventral , Animales , Ratones , Astrocitos , Dopamina , Receptores de Dopamina D2
19.
Front Cell Neurosci ; 15: 673433, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34163329

RESUMEN

The glial cells astrocytes have long been recognized as important neuron-supporting elements in brain development, homeostasis, and metabolism. After the discovery that the reciprocal communication between astrocytes and neurons is a fundamental mechanism in the modulation of neuronal synaptic communication, over the last two decades astrocytes became a hot topic in neuroscience research. Crucial to their functional interactions with neurons are the cytosolic Ca2+ elevations that mediate gliotransmission. Large attention has been posed to the so-called Ca2+microdomains, dynamic Ca2+ changes spatially restricted to fine astrocytic processes including perisynaptic astrocytic processes (PAPs). With presynaptic terminals and postsynaptic neuronal membranes, PAPs compose the tripartite synapse. The distinct spatial-temporal features and functional roles of astrocyte microdomain Ca2+ activity remain poorly defined. However, thanks to the development of genetically encoded Ca2+ indicators (GECIs), advanced microscopy techniques, and innovative analytical approaches, Ca2+ transients in astrocyte microdomains were recently studied in unprecedented detail. These events have been observed to occur much more frequently (∼50-100-fold) and dynamically than somatic Ca2+ elevations with mechanisms that likely involve both IP3-dependent and -independent pathways. Further progress aimed to clarify the complex, dynamic machinery responsible for astrocytic Ca2+ activity at microdomains is a crucial step in our understanding of the astrocyte role in brain function and may also reveal astrocytes as novel therapeutic targets for different brain diseases. Here, we review the most recent studies that improve our mechanistic understanding of the essential features of astrocyte Ca2+ microdomains.

20.
Nat Neurosci ; 24(3): 312-325, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33589835

RESUMEN

Reactive astrocytes are astrocytes undergoing morphological, molecular, and functional remodeling in response to injury, disease, or infection of the CNS. Although this remodeling was first described over a century ago, uncertainties and controversies remain regarding the contribution of reactive astrocytes to CNS diseases, repair, and aging. It is also unclear whether fixed categories of reactive astrocytes exist and, if so, how to identify them. We point out the shortcomings of binary divisions of reactive astrocytes into good-vs-bad, neurotoxic-vs-neuroprotective or A1-vs-A2. We advocate, instead, that research on reactive astrocytes include assessment of multiple molecular and functional parameters-preferably in vivo-plus multivariate statistics and determination of impact on pathological hallmarks in relevant models. These guidelines may spur the discovery of astrocyte-based biomarkers as well as astrocyte-targeting therapies that abrogate detrimental actions of reactive astrocytes, potentiate their neuro- and glioprotective actions, and restore or augment their homeostatic, modulatory, and defensive functions.


Asunto(s)
Envejecimiento/patología , Astrocitos/patología , Encéfalo/patología , Médula Espinal/patología , Animales , Encefalopatías/patología , Lesiones Encefálicas/patología , Humanos , Traumatismos de la Médula Espinal/patología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda