Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Neuroscience ; 323: 20-8, 2016 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-26546469

RESUMEN

A few decades ago it was discovered that two regions of the adult brain retain the ability to generate new neurons. These regions include the subgranular zone of the hippocampal dentate gyrus and the ventricular-subventricular zone (V-SVZ) located at the border of the lateral ventricle. In the V-SVZ, it was discovered that neural progenitor cells (NPCs) share many features of mature astrocytes and are often referred as V-SVZ astrocytes. We will first describe the markers, the morphology, and the neurophysiological characteristics of the mouse V-SVZ astrocytes. We will then discuss the fact that V-SVZ astrocytes constitute a mixed population with respect to their neurogenic properties, e.g., quiescent versus activated state, neurogenic fate, and transcription factors expression. Finally, we will describe two functions of V-SVZ astrocytes, their metabolic coupling to blood vessels and their neurogenic-supportive role consisting of providing guidance and survival cues to migrating newborn neurons.


Asunto(s)
Astrocitos/fisiología , Encéfalo/fisiología , Células-Madre Neurales/fisiología , Neurogénesis/fisiología , Nicho de Células Madre/fisiología , Animales , Astrocitos/citología , Encéfalo/citología , Ácido Glutámico/metabolismo , Humanos , Células-Madre Neurales/citología , Neuronas/fisiología
2.
Cell Death Differ ; 22(12): 2078-86, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25976303

RESUMEN

A synthetic peptide (sPIF) analogous to the mammalian embryo-derived PreImplantation Factor (PIF) enables neuroprotection in rodent models of experimental autoimmune encephalomyelitis and perinatal brain injury. The protective effects have been attributed, in part, to sPIF's ability to inhibit the biogenesis of microRNA let-7, which is released from injured cells during central nervous system (CNS) damage and induces neuronal death. Here, we uncover another novel mechanism of sPIF-mediated neuroprotection. Using a clinically relevant rat newborn brain injury model, we demonstrate that sPIF, when subcutaneously administrated, is able to reduce cell death, reverse neuronal loss and restore proper cortical architecture. We show, both in vivo and in vitro, that sPIF activates cyclic AMP dependent protein kinase (PKA) and calcium-dependent protein kinase (PKC) signaling, leading to increased phosphorylation of major neuroprotective substrates GAP-43, BAD and CREB. Phosphorylated CREB in turn facilitates expression of Gap43, Bdnf and Bcl2 known to have important roles in regulating neuronal growth, survival and remodeling. As is the case in sPIF-mediated let-7 repression, we provide evidence that sPIF-mediated PKA/PKC activation is dependent on TLR4 expression. Thus, we propose that sPIF imparts neuroprotection via multiple mechanisms at multiple levels downstream of TLR4. Given the recent FDA fast-track approval of sPIF for clinical trials, its potential clinical application for treating other CNS diseases can be envisioned.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Fármacos Neuroprotectores/farmacología , Péptidos/farmacología , Proteína Quinasa C/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , AMP Cíclico/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Proteína GAP-43/genética , Proteína GAP-43/metabolismo , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Fármacos Neuroprotectores/síntesis química , Péptidos/síntesis química , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Interferencia de ARN , Ratas , Receptor Toll-Like 4/antagonistas & inhibidores , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Proteína Letal Asociada a bcl/genética , Proteína Letal Asociada a bcl/metabolismo
3.
Nat Commun ; 5: 4563, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-25081057

RESUMEN

Tuberous sclerosis complex (TSC), caused by dominant mutations in either TSC1 or TSC2 tumour suppressor genes is characterized by the presence of brain malformations, the cortical tubers that are thought to contribute to the generation of pharmacoresistant epilepsy. Here we report that tuberless heterozygote Tsc1(+/-) mice show functional upregulation of cortical GluN2C-containing N-methyl-D-aspartate receptors (NMDARs) in an mTOR-dependent manner and exhibit recurrent, unprovoked seizures during early postnatal life (

Asunto(s)
Anticonvulsivantes/farmacología , Epilepsia/tratamiento farmacológico , Pirazoles/farmacología , Quinolonas/farmacología , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/genética , Esclerosis Tuberosa/tratamiento farmacológico , Proteínas Supresoras de Tumor/genética , Potenciales de Acción/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Electroencefalografía , Epilepsia/genética , Epilepsia/metabolismo , Epilepsia/patología , Regulación de la Expresión Génica , Heterocigoto , Humanos , Masculino , Ratones , Ratones Transgénicos , Microtomía , Neocórtex/efectos de los fármacos , Neocórtex/metabolismo , Neocórtex/patología , Técnicas de Placa-Clamp , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Técnicas de Cultivo de Tejidos , Esclerosis Tuberosa/genética , Esclerosis Tuberosa/metabolismo , Esclerosis Tuberosa/patología , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/deficiencia
4.
Neuroscience ; 219: 271-9, 2012 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-22677205

RESUMEN

The generation of the most abundant neurons of the cerebellum, the granule cells, relies on a balance between clonal expansion and apoptosis during the first 10 days after birth in the external germinal layer (EGL). The amino acid glutamate controls such critical phases of cell development in other systems through specific receptors such as metabotropic glutamate receptor 5 (mGlu(5)R). However, the function of mGlu(5)Rs on the proliferation and survival of granule cell precursors (GCPs) remains elusive. We found mGlu(5)R mRNA transcripts in EGL using RT-PCR and observed mGlu(5)R-mediated Ca(2+) responses in GCPs in acute slices as early as postnatal day (P) 2-3. Using in vivo injections of the selective non-competitive mGlu(5)R antagonist 2-methyl-6-(phenylethynyl)pyridine (MPEP) in P7-P9 mice, we found a 20% increase in the number of proliferative GCPs labeled at P7 with the S-phase marker bromodeoxyuridine (BrdU), but no increase in cell proliferation examined 2h following a BrdU injection. Furthermore, similar treatments led to a significant 70% decrease in the number of apoptotic GCPs in the EGL as determined by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining. In contrast, in vivo treatment with the mGlu(5)R agonist (RS)-2-chloro-5-hydroxyphenylglycine (CHPG) resulted in a ∼60% increase in the number of TUNEL-labeled GCPs compared to control. These findings identify a unique role for glutamate acting at mGlu(5)Rs as a functional switch regulating GCP survival in the EGL, thus controlling the total number of cerebellar granule cells produced.


Asunto(s)
Cerebelo/crecimiento & desarrollo , Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Receptores de Glutamato Metabotrópico/biosíntesis , Animales , Animales Recién Nacidos , Apoptosis/fisiología , Supervivencia Celular , Cerebelo/citología , Cerebelo/metabolismo , Femenino , Etiquetado Corte-Fin in Situ , Masculino , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor del Glutamato Metabotropico 5 , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
Epilepsy Res ; 59(2-3): 107-22, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15246115

RESUMEN

Previous studies have characterized the electrophysiological properties of astrocytes in the CA1 region of hippocampi resected from patients with intractable temporal lobe epilepsy (TLE). However, the properties of hilar astrocytes from such patients have not been studied although astrocytes display regional heterogeneity and a non-uniform response to injury. Thus, we performed patch-clamp recordings of putative astrocytes in hilar and CA1 regions of surgically removed epileptic hippocampi with and without sclerosis (mesial TLE, MTLE patients, and paradoxical TLE, PTLE patients, respectively), and non-epileptic, non-sclerotic hippocampi (tumor patients). Our data show that the current profile of hilar astrocytes undergoes significant changes in MTLE but not in PTLE or tumor hippocampi. In particular, inwardly rectifying K(+) (K(IR)) and outwardly rectifying K(+) currents were reduced, inward Na(+) currents and membrane resistances were increased in putative astrocytes from MLTE cases compared to PTLE and tumor cases. Because the conductance of K(IR) channels in cell-attached patches (approximately 34pS) from MTLE tissue was not altered, a reduction in the number of K(IR) channels likely accounts for the decrease in whole-cell K(IR) conductance. Presumed astrocytes in the CA1 region from each patient group displayed intercellular coupling and a passive current profile; these characteristics were never observed in hilar glial cells. No apparent changes in the current profile of coupled CA1 glial cells could be detected between MTLE, PTLE and tumor tissues. Additionally, CA1 glial cells expressed a high density of 34pS K(IR) channels. These data suggest that K(+) buffering via K(IR) channels may be functionally compromised in hilar astrocytes of epileptic and sclerotic (MTLE) human hippocampi. By contrast, CA1 astrocytes retained their intercellular coupling and K(IR) channel expression necessary for K(+) buffering.


Asunto(s)
Giro Dentado/fisiología , Epilepsia del Lóbulo Temporal/patología , Epilepsia del Lóbulo Temporal/fisiopatología , Neuroglía/fisiología , Giro Dentado/citología , Electrofisiología , Humanos , Neuroglía/citología , Esclerosis/patología , Esclerosis/fisiopatología
6.
Eur J Neurosci ; 18(10): 2893-8, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14656339

RESUMEN

During acute pathological processes, microglia transform into an activated state characterized by a defined morphology and current profile, and are recruited to injury sites by chemokines. No information is available on the ion channels and the mode of action of chemokines in microglia in brain slices from humans with a chronic pathology. Thus, patch-clamp recordings of microglia were performed in hippocampal slices from seven patients who underwent surgery for pharmaco-resistant epilepsy. Cells were identified as microglia by positive labelling with fluorescein-conjugated tomato lectin before recording. All the recorded cells had an ameboid morphology characteristic of activated microglia. However, they had a high input resistance (3.6 G omega), a zero-current resting potential of -16 mV, and lacked Na+ currents, inwardly rectifying and delayed rectifying K+ currents such as non-activated microglia. Importantly, recorded cells expressed Ca2+-sensitive outward currents that activated at 0 mV with non-buffered intracellular Ca2+ and were sensitive to 1 mm tetraethylammonium (TEA). The estimated single-channel conductances were 187 pS in cell-attached and 149 pS in outside-out patches, similar to those of high-conductance Ca2+-dependent K+ channels. The chemokine MIP1-alpha increased whole-cell outward current amplitudes measured at +60 mV by a factor of 3.3. Thus, microglia in hippocampi from epileptic patients express high-conductance Ca2+-dependent K+ channels that are modulated by the chemokine MIP1-alpha. This modulation may contribute to the migratory effect of MIP1-alpha on microglia.


Asunto(s)
Calcio/fisiología , Quimiocinas/farmacología , Hipocampo/patología , Proteínas Inflamatorias de Macrófagos/farmacología , Microglía/efectos de los fármacos , Canales de Potasio/fisiología , Quimiocina CCL4 , Conductividad Eléctrica , Epilepsia del Lóbulo Temporal/fisiopatología , Hipocampo/efectos de los fármacos , Humanos , Técnicas In Vitro , Potenciales de la Membrana/efectos de los fármacos , Microglía/clasificación , Microglía/fisiología , Técnicas de Placa-Clamp , Lectinas de Plantas/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Tetraetilamonio/farmacología
7.
J Physiol ; 550(Pt 3): 785-800, 2003 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-12807990

RESUMEN

Previous studies have reported the presence of migrating and dividing neuronal progenitors in the subventricular zone (SVZ) and rostral migratory stream (RMS) of the postnatal mammalian brain. Although the behaviour of these progenitors is thought to be influenced by local signals, the nature and mode of action of the local signals are largely unknown. One of the signalling molecules known to affect the behaviour of embryonic neurons is the neurotransmitter GABA. In order to determine whether GABA affects neuronal progenitors via the activation of specific receptors, we performed cell-attached, whole-cell and gramicidin perforated patch-clamp recordings of progenitors in postnatal mouse brain slices containing either the SVZ or the RMS. Recorded cells displayed a morphology typical of migrating neuronal progenitors had depolarized zero-current resting potentials, and lacked action potentials. A subset of progenitors contained GABA and stained positive for glutamic acid decarboxylase 67 (GAD-67) as shown by immunohistochemistry. In addition, every neuronal progenitor responded to GABA via picrotoxin-sensitive GABAA receptor (GABAAR) activation. GABAARs displayed an ATP-dependent rundown and a low sensitivity to Zn2+. GABA responses were sensitive to benzodiazepine agonists, an inverse agonist, as well as a barbiturate agonist. While GABA was hyperpolarizing at the zero-current resting potentials, it was depolarizing at the cell resting potentials estimated from the reversal potential of K+ currents through a cell-attached patch. Thus, our study demonstrates that neuronal progenitors of the SVZ/RMS contain GABA and are depolarized by GABA, which may constitute the basis for a paracrine signal among neuronal progenitors to dynamically regulate their proliferation and/or migration.


Asunto(s)
Ventrículos Cerebrales/fisiología , Neuronas/efectos de los fármacos , Receptores de GABA-A/efectos de los fármacos , Células Madre/efectos de los fármacos , Ácido gamma-Aminobutírico/farmacología , Adenosina Trifosfato/fisiología , Animales , Barbitúricos/farmacología , Benzodiazepinas/farmacología , Ventrículos Cerebrales/citología , Ventrículos Cerebrales/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Electrofisiología , Antagonistas del GABA/farmacología , Glutamato Descarboxilasa/metabolismo , Isoenzimas/metabolismo , Potenciales de la Membrana/fisiología , Ratones , Neostriado/citología , Neostriado/fisiología , Técnicas de Placa-Clamp , Picrotoxina/farmacología , Canales de Potasio/efectos de los fármacos , Zinc/farmacología
8.
J Neurophysiol ; 90(4): 2291-302, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12801891

RESUMEN

Previous studies have reported the presence of neuronal progenitors in the subventricular zone (SVZ) and rostral migratory stream (RMS) of the postnatal mammalian brain. Although many studies have examined the survival and migration of progenitors after transplantation and the factors influencing their proliferation or differentiation, no information is available on the electrophysiological properties of these progenitors in a near-intact environment. Thus we performed whole cell and cell-attached patch-clamp recordings of progenitors in brain slices containing either the SVZ or the RMS from postnatal day 15 to day 25 mice. Both regions displayed strong immunoreactivity for nestin and neuron-specific class III beta-tubulin, and recorded cells displayed a morphology typical of the neuronal progenitors known to migrate throughout the SVZ and RMS to the olfactory bulb. Recorded progenitors had depolarized zero-current resting potentials (mean more depolarized than -28 mV), very high input resistances (about 4 GOmega), and lacked action potentials. Using the reversal potential of K+ currents through a cell-attached patch a mean resting potential of -59 mV was estimated. Recorded progenitors displayed Ca2+-dependent K+ currents and TEA-sensitive-delayed rectifying K+ (KDR) currents, but lacked inward K+ currents and transient outward K+ currents. KDR currents displayed classical kinetics and were also sensitive to 4-aminopyridine and alpha-dendrotoxin, a blocker of Kv1 channels. Na+ currents were found in about 60% of the SVZ neuronal progenitors. No developmental changes were observed in the passive membrane properties and current profile of neuronal progenitors. Together these data suggest that SVZ neuronal progenitors display passive membrane properties and an ionic signature distinct from that of cultured SVZ neuronal progenitors and mature neurons.


Asunto(s)
Cuerpo Calloso/crecimiento & desarrollo , Cuerpo Estriado/crecimiento & desarrollo , Ventrículos Laterales/crecimiento & desarrollo , Neuronas/fisiología , Células Madre/fisiología , Animales , Animales Recién Nacidos , Fenómenos Biofísicos , Biofisica , Calcio/farmacología , Calcio/fisiología , Cuerpo Calloso/efectos de los fármacos , Cuerpo Estriado/efectos de los fármacos , Técnicas In Vitro , Canales Iónicos/fisiología , Ventrículos Laterales/efectos de los fármacos , Ratones , Neuronas/efectos de los fármacos , Células Madre/efectos de los fármacos
9.
J Membr Biol ; 189(3): 201-12, 2002 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-12395285

RESUMEN

Previous studies have reported that mature oligodendrocytes (OLGs) in vitro display various voltage-dependent K+ currents while in situ OLGs show only voltage-independent K+ currents. Given this discrepancy and the lack of information on myelinating OLG ion channel expression in situ, we characterized mature OLG currents in myelinating corpus callosum slices from 17 to 36-day old rats. OLGs were recorded in cell-attached and whole-cell patch-clamp configurations, displayed morphology typical of mature OLGs, and stained positive for myelin basic protein. OLGs displayed large voltage-independent currents that decayed during the voltage pulse and small voltage-activated outward currents. The latter were blocked by TEA, activated between -40 and -50 mV, and decayed slowly. The former were composed of large voltage-independent, time-dependent Ba2+ (1 mM)-sensitive currents, and voltage-dependent Cs+ (5 mM) and Ba2+ (100 mM)-sensitive currents that reversed near the K+ equilibrium potential and inactivated at hyperpolarized potentials, identifying them as inwardly rectifying K+ currents. Inwardly rectifying single-channel K+currents could be recorded in the cell-attached configuration. The estimated single-channel slope conductance was 30 pS. The steady-state open probability was voltage-dependent and declined from 0.9 to 0.5 between -80 and -150 mV. Overall, mature OLGs in situ possess time- and also voltage-dependent K+ currents, which may facilitate clearance of K+ released during axonal firing.


Asunto(s)
Cuerpo Calloso/fisiología , Oligodendroglía/fisiología , Canales de Potasio/fisiología , Animales , Bario/metabolismo , Bario/farmacología , Cesio/metabolismo , Cesio/farmacología , Cuerpo Calloso/efectos de los fármacos , Conductividad Eléctrica , Técnicas In Vitro , Transporte Iónico/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Oligodendroglía/citología , Oligodendroglía/efectos de los fármacos , Técnicas de Placa-Clamp , Canales de Potasio/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Sensibilidad y Especificidad , Tetraetilamonio/metabolismo , Tetraetilamonio/farmacología
10.
J Neurophysiol ; 88(3): 1407-19, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12205162

RESUMEN

Although glial GABA uptake and release have been studied in vitro, GABA transporters (GATs) have not been characterized in glia in slices. Whole cell patch-clamp recordings were obtained from Bergmann glia in rat cerebellar slices to characterize carrier-mediated GABA influx and efflux. GABA induced inward currents at -70 mV that could be pharmacologically separated into GABA(A) receptor and GAT currents. In the presence of GABA(A/B/C) receptor blockers, mean GABA-induced currents measured -48 pA at -70 mV, were inwardly rectifying between -70 and +50 mV, were inhibited by external Na(+) removal, and were diminished by reduction of external Cl(-). Nontransportable blockers of GAT-1 (SKF89976-A and NNC-711) and a transportable blocker of all the GAT subtypes (nipecotic acid) reversibly reduced GABA-induced transport currents by 68 and 100%, respectively. A blocker of BGT-1 (betaine) had no effect. SKF89976-A and NNC-711 also suppressed baseline inward currents that likely result from tonic GAT activation by background GABA. The substrate agonists, nipecotic acid and beta-alanine but not betaine, induced voltage- and Na(+)-dependent currents. With Na(+) and GABA inside the patch pipette or intracellular GABA perfusion during the recording, SKF89976-A blocked baseline outward currents that activated at -60 mV and increased with more depolarized potentials. This carrier-mediated GABA efflux induced a local accumulation of extracellular GABA detected by GABA(A) receptor activation on the recorded cell. Overall, these results indicate that Bergmann glia express GAT-1 that are activated by ambient GABA. In addition, GAT-1 in glia can work in reverse and release sufficient GABA to activate nearby GABA receptors.


Asunto(s)
Proteínas Portadoras/metabolismo , Cerebelo/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Transporte de Membrana , Neuroglía/metabolismo , Transportadores de Anión Orgánico , Ácido gamma-Aminobutírico/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Transporte Biológico/fisiología , Proteínas Portadoras/fisiología , Cerebelo/citología , Electrofisiología , Proteínas Transportadoras de GABA en la Membrana Plasmática , Técnicas In Vitro , Proteínas de la Membrana/fisiología , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Receptores de GABA-A/fisiología , Ácido gamma-Aminobutírico/farmacología
11.
J Physiol ; 541(Pt 3): 753-67, 2002 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-12068038

RESUMEN

Taurine uptake is essential for the maintenance of millimolar intracellular concentrations of taurine, which is released during ischaemia and is thought to be neuroprotective. To determine whether Bergmann glia express functional transporters that can mediate both taurine uptake and efflux, whole-cell patch-clamp recordings were obtained from these cells in rat cerebellar slices. Taurine-induced inward currents can be pharmacologically separated into GABA(A) receptor and taurine transporter currents. In the presence of GABA receptor blockers, residual taurine currents averaged -28 pA at -70 mV and were strictly inwardly rectifying between -70 and +50 mV. These residual currents were also abolished by external Na+ removal and diminished by reduction of external Cl-, consistent with transport currents. Taurine transport currents were reduced by a taurine transporter inhibitor, guanidinoethyl sulphonate (GES). Other classical inhibitors reduced taurine transport currents with an order of potency (hypotaurine > beta-alanine > GES > GABA) similar to that reported for cloned rat taurine transporters. Following intracellular taurine perfusion during the recording, a progressively developing outward current could be observed at -50 mV but not at -70 mV. Intracellular perfusion of taurine also decreased taurine-induced inward currents at both holding potentials. Outward currents induced by intracellular taurine increased in amplitude with depolarization, activated near -50 mV, and were affected by GES. For the first time, these results demonstrate that taurine activates both GABA(A) receptors and Na+/Cl--dependent taurine transporters in Bergmann glia in slices. In addition, our data show that taurine transporters can work in reverse and can probably mediate taurine efflux under ischaemic conditions.


Asunto(s)
Proteínas Portadoras/metabolismo , Cerebelo/metabolismo , Neuroglía/metabolismo , Taurina/metabolismo , Animales , Cerebelo/citología , Cerebelo/efectos de los fármacos , Cloruros/metabolismo , Antagonistas del GABA/farmacología , Técnicas In Vitro , Potenciales de la Membrana/fisiología , Neuroglía/efectos de los fármacos , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Receptores de GABA-A/efectos de los fármacos , Receptores de GABA-A/metabolismo , Sodio/metabolismo
12.
J Neurophysiol ; 85(4): 1719-31, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11287494

RESUMEN

Neocortical freeze lesions have been widely used to study neuronal mechanisms underlying hyperexcitability in dysplastic cortex. Comparatively little attention has been given to biophysical changes in the surrounding astrocytes that show profound morphological and biochemical alterations, often referred to as reactive gliosis. Astrocytes are thought to aid normal neuronal function by buffering extracellular K(+). Compromised astrocytic K(+) buffering has been proposed to contribute to neuronal dysfunction. Astrocytic K(+) buffering is mediated, partially, by the activity of inwardly rectifying K(+) channels (K(IR)) and may involve intracellular redistribution of K(+) through gap-junctions. We characterized K(+) channel expression and gap-junction coupling between astrocytes in freeze-lesion-induced dysplastic neocortex. Whole cell patch-clamp recordings were obtained from astrocytes in slices from postnatal day (P) 16--P24 rats that had received a freeze-lesion on P1. A marked increase in glial fibrillary acidic protein immunoreactivity was observed along the entire length of the freeze lesion. Clusters of proliferative (bromo-deoxyuridine nuclear staining, BrdU+) astrocytes were seen near the depth of the microsulcus. Astrocytes in cortical layer I surrounding the lesion were characterized by a significant reduction in K(IR). BrdU-positive astrocytes near the depth of the microsulcus showed essentially no expression of K(IR) channels but markedly enhanced expression of delayed rectifier K(+) (K(DR)) channels. These proliferative cells showed virtually no dye coupling, whereas astrocytes in the hyperexcitable zone adjacent to the microsulcus displayed prominent dye-coupling as well as large K(IR) and outward K(+) currents. These findings suggest that reactive gliosis is accompanied by a loss of K(IR) currents and reduced gap junction coupling, which in turn suggests a compromised K(+) buffering capacity.


Asunto(s)
Astrocitos/fisiología , Encefalopatías/fisiopatología , Neocórtex/fisiopatología , Animales , Astrocitos/patología , Encefalopatías/etiología , Encefalopatías/patología , División Celular , Electrofisiología , Colorantes Fluorescentes , Congelación , Técnicas In Vitro , Isoquinolinas , Neocórtex/patología , Canales de Potasio/fisiología , Ratas , Ratas Sprague-Dawley , Valores de Referencia
13.
Neuroreport ; 11(14): 3151-5, 2000 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-11043540

RESUMEN

Injury and diseases of the nervous system can induce astrocytes to form tenacious glial scars. We induced focal cortical freeze-lesions in neonatal rats and examined scars histologically and electrophysiologically in tissue slices isolated 2-3 weeks after lesioning. Lesions displayed marked gliosis, characterized by upregulation of GFAP labeling. Reactive astrocytes surrounding the scar showed marked hypertrophy, enlarged cell bodies and extended processes frequently terminating with endfeet-like structures on blood vessels. These reactive astrocytes showed enhanced expression of inwardly rectifying K+ (K(IR)) channels, widely believed to be an important pathway for astrocytic K+ buffering. These results suggest that a subpopulation of reactive astrocytes along a glial scar might be instrumental in buffering K+ away from the lesion.


Asunto(s)
Astrocitos/citología , Lesiones Encefálicas/patología , Cicatriz/patología , Gliosis/patología , Canales de Potasio/metabolismo , Animales , Astrocitos/metabolismo , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/ultraestructura , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/fisiopatología , Corteza Cerebral/lesiones , Corteza Cerebral/patología , Corteza Cerebral/fisiopatología , Cicatriz/metabolismo , Cicatriz/fisiopatología , Frío/efectos adversos , Desnervación/efectos adversos , Proteína Ácida Fibrilar de la Glía/metabolismo , Gliosis/etiología , Gliosis/fisiopatología , Potenciales de la Membrana/fisiología , Ratas , Ratas Sprague-Dawley
14.
J Membr Biol ; 176(1): 31-40, 2000 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-10882426

RESUMEN

Patients with cerebral tumors often present with elevated levels of acetylcholine (ACh) in their cerebrospinal fluid. This motivated us to investigate physiological effects of ACh on cultured human astrocytoma cells (U373) using a combination of videomicroscopy, calcium microspectrofluorimetry and perforated patch-clamp recording. Astrocytoma cells exhibited the typical morphological changes associated with cell migration; polarized cells displayed prominent lamellipodia and associated membrane ruffling at the anterior of the cell, and a long tail region that periodically contracted into the cell body as the cell moved forward. Bath application of the ACh receptor agonist, muscarine, reversibly inhibited cell migration. In conjunction with this inhibition, ACh induced a dose-dependent, biphasic increase in resting intracellular free calcium concentration ([Ca2+]i) associated with periodic Ca2+ oscillations during prolonged ACh applications. The early transient rise in [Ca2+]i was abolished by ionomycin and thapsigargin but was insensitive to caffeine and ryanodine while the plateau phase was strictly dependent on external calcium. The Ca2+ response to ACh was mimicked by muscarine and abolished by the muscarinic antagonists, atropine or 4-DAMP, but not by pirenzepine. Using perforated patch-clamp recordings combined with fluorescent imaging, we demonstrated that ACh-induced [Ca2+]i oscillations triggered membrane voltage oscillations that were due to the activation of voltage-dependent, Ca2+-sensitive K+ currents. These K+ currents were blocked by intracellular injection of EGTA, or by extracellular application of TEA, quinine, or charybdotoxin, but not by apamin. These studies suggest that activation of muscarinic receptors on glioma cells induce the release of Ca2+ from intracellular stores which in turn activate Ca2+-dependent (BK-type) K+ channels. Furthermore, this effect was associated with inhibition of cell migration, suggesting an interaction of this pathway with glioma cell migration.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Muscarina/metabolismo , Agonistas Muscarínicos/metabolismo , Canales de Potasio Calcio-Activados , Canales de Potasio/metabolismo , Acetilcolina/farmacología , Astrocitoma , Calcio/metabolismo , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Movimiento Celular/fisiología , Humanos , Canales de Potasio de Gran Conductancia Activados por el Calcio , Muscarina/farmacología , Agonistas Muscarínicos/farmacología , Células Tumorales Cultivadas
15.
Glia ; 30(1): 27-38, 2000 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10696142

RESUMEN

Patch-clamp recordings were obtained in brain slices from 283 rat astrocytes. The expression of voltage-activated whole-cell currents was compared in four different CNS regions (hippocampus, cerebral cortex, spinal cord, and cerebellum). Our data show that CNS astrocytes do not show significant regional differences in their ion channel complement. With the exception of cerebellar Bergmann glial cells, essentially all astrocytes express a combination of delayed rectifying outward K(+) currents, transient A-type K(+) currents, and small Na(+) currents. Developmentally, an increasing percentage of astrocytes and Bergmann glial cells express inwardly rectifying K(+) currents. We did not observe cells that were passive, i.e., lacking voltage-activated currents. A few cells that appeared "passive" in initial recordings showed voltage-activated K(+) currents after off-line leak subtraction. The heterogeneity observed in the ion channel complement was found to be identical when cell-to-cell variations observed within a given CNS region and between various CNS regions were compared, suggesting a common and fairly stereotypical complement of ion channels in CNS astrocytes. Ion channel expression in Bergmann glial cells differed from that of all other CNS regions studied. These cells typically showed very low input resistances attributable to a significant time- and voltage-independent resting K(+) conductance. However, as with electrophysiologically "passive"-appearing astrocytes, Bergmann glial cells showed expression of delayed rectifying K(+) currents after off-line leak subtraction. Inwardly rectifying K(+) currents were observed in Bergmann glial cells after postnatal day 17. Collectively, our data suggest that all astrocytes contain voltage-gated ion channels that display a common pattern of expression during development.


Asunto(s)
Astrocitos/metabolismo , Sistema Nervioso Central/metabolismo , Canales de Potasio/metabolismo , Canales de Sodio/metabolismo , Envejecimiento/metabolismo , Envejecimiento/fisiología , Animales , Animales Recién Nacidos/crecimiento & desarrollo , Animales Recién Nacidos/metabolismo , Animales Recién Nacidos/fisiología , Astrocitos/fisiología , Sistema Nervioso Central/citología , Sistema Nervioso Central/fisiología , Conductividad Eléctrica , Técnicas In Vitro , Neuroglía/metabolismo , Neuroglía/fisiología , Técnicas de Placa-Clamp , Canales de Potasio/fisiología , Ratas , Ratas Sprague-Dawley , Canales de Sodio/fisiología , Distribución Tisular
16.
J Neurophysiol ; 82(6): 3476-87, 1999 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-10601476

RESUMEN

Spinal cord astrocytes express four biophysically and pharmacologically distinct voltage-activated potassium (K(+)) channel types. The K(+) channel blocker 4-aminopyridine (4-AP) exhibited differential and concentration-dependent block of all of these currents. Specifically, 100 microM 4-AP selectively inhibited a slowly inactivating outward current (K(SI)) that was insensitive to dendrototoxin (< or = 10 microM) and that activated at -50 mV. At 2 mM, 4-AP inhibited fast-inactivating, low-threshold (-70 mV) A-type currents (K(A)) and sustained, TEA-sensitive noninactivating delayed-rectifier-type currents (K(DR)). At an even higher concentration (8 mM), 4-AP additionally blocked inwardly rectifying, Cs(+)- and Ba(2+)-sensitive K(+) currents (K(IR)). Current injection into current-clamped astrocytes in culture or in acute spinal cord slices induced an overshooting voltage response reminiscent of slow neuronal action potentials. Increasing concentrations of 4-AP selectively modulated different phases in the repolarization of these glial spikes, suggesting that all four K(+) currents serve different roles in stabilization and repolarization of the astrocytic membrane potential. Our data suggest that 4-AP is an useful, dose-dependent inhibitor of all four astrocytic K(+) channels. We show that the slowly inactivating astrocytic K(+) currents, which had not been described as separate current entities in astrocytes, contribute to the resting K(+) conductance and may thus be involved in K(+) homeostatic functions of astrocytes. The high sensitivity of these currents to micromolar 4-AP suggests that application of 4-AP to inhibit neuronal A-currents or to induce epileptiform discharges in brain slices also may influence astrocytic K(+) buffering.


Asunto(s)
4-Aminopiridina/farmacología , Neuroglía/metabolismo , Bloqueadores de los Canales de Potasio , Canales de Potasio Calcio-Activados , Canales de Potasio de Rectificación Interna , Canales de Potasio con Entrada de Voltaje , Canales de Potasio , Potenciales de Acción/efectos de los fármacos , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Células Cultivadas , Canales de Potasio de Tipo Rectificador Tardío , Electrofisiología , Canales de Potasio de Gran Conductancia Activados por el Calcio , Neuroglía/efectos de los fármacos , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Médula Espinal/citología , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Tetraetilamonio/farmacología
17.
J Membr Biol ; 166(3): 213-22, 1998 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-9843595

RESUMEN

Astrocytes that are recorded in acute tissue slices of rat hippocampus using whole-cell patch-clamp, commonly exhibit voltage-activated Na+ and K+ currents. Some reports have described astrocytes that appear to lack voltage-activated currents and proposed that these cells constitute a subpopulation of electrophysiologically passive astrocytes. We show here that these cells can spontaneously change during a recording unmasking expression of previously suppressed voltage-activated currents, suggesting that such cells do not represent a subpopulation of passive astrocytes. Superfusion of a low Ca2+/EGTA solution was able to reversibly suppress voltage-activated K+ currents in cultured astrocytes. Currents were restored upon addition of normal bath Ca2+. These effects of Ca2+ on both outward and inward K+ currents were dose- and time-dependent, with increasing concentrations of Ca2+ (from 0 to 800 micrometers) leading to a gradual unmasking of voltage-dependent outward and inward K+ currents. The transition from an apparently passive cell to one exhibiting prominent voltage-activated currents was not associated with any changes in membrane capacitance or access resistance. By contrast, in cells in which low access resistance or poor seal accounted for the absence of voltage-activated currents, improvement of cell access was always accompanied by changes in series resistance and membrane capacitance. We propose that spillage of pipette solution containing low Ca2+/EGTA during cell approach in slice recordings and/or poor cell access, lead to a transient masking of voltage-activated currents even in astrocytes that express prominent voltage-activated currents. These cells, however, do not constitute a subpopulation of electrophysiologically passive astrocytes.


Asunto(s)
Artefactos , Astrocitos/metabolismo , Calcio/farmacología , Activación del Canal Iónico , Transporte Iónico , Canales de Potasio/metabolismo , Animales , Astrocitos/clasificación , Células Cultivadas , Ácido Egtácico/farmacología , Líquido Intracelular/metabolismo , Potenciales de la Membrana , Técnicas de Placa-Clamp , Potasio/metabolismo , Canales de Potasio/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Médula Espinal/citología
18.
Epilepsy Res ; 32(1-2): 286-303, 1998 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9761328

RESUMEN

We studied physiological properties of glial cells from acute slices of biopsies from patients operated for intractable mesio-temporal lobe epilepsy using whole-cell patch-clamp recordings. Cells were filled with Lucifer Yellow (LY) during recordings to allow morphological reconstruction and immunohistochemical cell identification. Seizure-associated astrocytes had complex, arborized, highly branched processes giving them a stellate appearance, and cells stained intensely for the intermediate filament GFAP as previously reported for 'reactive' astrocytes. GFAP-positive astrocytes from epilepsy biopsies consistently expressed voltage-activated, TTX-sensitive Na+ channels that showed fast activation and inactivation kinetics. Unlike comparison astrocytes, derived from tissues that were not associated with seizure foci, these astrocytes expressed Na+ channels at densities sufficient to generate slow action potentials (spikes) in current clamp studies. In these cells, the ratio of Na+ to K+ conductance was consistently 3-4-fold higher than in comparison human or control rat astrocytes. Four of 17 astrocytes from epilepsy patients versus 14/14 from control rat hippocampus and four of five in comparison human tissue showed a lack of inwardly rectifying K+ currents, which in normal astrocytes are implicated in the control of extracellular K+ levels. These results suggest that astrocytes surrounding seizure foci differ in morphological and physiological properties, and that glial K+ buffering could be impaired at the seizure focus, thus contributing to the pathophysiology of seizures.


Asunto(s)
Epilepsias Parciales/fisiopatología , Epilepsia del Lóbulo Temporal/fisiopatología , Hipocampo/fisiopatología , Neuroglía/fisiología , Lóbulo Temporal/fisiopatología , Adulto , Animales , Astrocitos/patología , Astrocitos/fisiología , Epilepsias Parciales/patología , Epilepsias Parciales/cirugía , Epilepsia del Lóbulo Temporal/patología , Epilepsia del Lóbulo Temporal/cirugía , Femenino , Proteína Ácida Fibrilar de la Glía/análisis , Hipocampo/patología , Hipocampo/cirugía , Humanos , Técnicas In Vitro , Potenciales de la Membrana , Persona de Mediana Edad , Neuroglía/patología , Canales de Potasio/fisiología , Ratas , Valores de Referencia , Canales de Sodio/fisiología , Lóbulo Temporal/patología , Lóbulo Temporal/cirugía
19.
J Neurophysiol ; 79(5): 2782-93, 1998 May.
Artículo en Inglés | MEDLINE | ID: mdl-9582244

RESUMEN

To better understand physiological changes that accompany the neoplastic transition of astrocytes to become astrocytoma cells, we studied biopsies of low-grade, pilocytic astrocytomas. This group of tumors is most prevalent in children and the tumor cells maintain most antigenic features typical of astrocytes. Astrocytoma cells were studied with the use of whole cell patch-clamp recordings in acute biopsy slices from 4-mo- to 14-yr-old pediatric patients. Recordings from 53 cells in six cases of low-grade astrocytomas were compared to either noncancerous peritumoral astrocytes or astrocytes obtained from other surgeries. Astrocytoma cells almost exclusively displayed slowly activating, sustained, tetraethylammonium (TEA)-sensitive outward potassium currents (delayed rectifying potassium currents; IDR) and transient, tetrodotoxin (TTX)-sensitive sodium currents (INa). By contrast, comparison glial cells from peritumoral regions or other surgeries showed IDR and INa, but in addition these cells also expressed transient "A"-type K+ currents and inwardly rectifying K+ currents (IIR), both of which were absent in astrocytoma cells. IIR constituted the predominant conductance in comparison astrocytes and was responsible for a high-resting K+ conductance in these cells. Voltage-activated Na+ currents were observed in 37 of 53 astrocytoma cells. Na+ current densities in astrocytoma cells, on average, were three- to fivefold larger than in comparison astrocytes. Astrocytoma cells expressing INa could be induced to generate slow action potential-like responses (spikes) by current injections. The threshold for generating such spikes was -34 mV (from a holding potential of -70 mV). The spike amplitude and time width were 52.5 mV and 12 ms, respectively. No spikes could be elicited in comparison astrocytes, although some of them expressed Na+ currents of similar size. Comparison of astrocytes to astrocytoma cells suggests that the apparent lack of IIR, which leads to high-input resistance (>500 MOmega), allows glioma cells to be sufficiently depolarized to generate Na+ spikes, whereas the high resting K+ conductance in astrocytes prevents their depolarization and thus generation of spikes. Consistent with this notion, Na+ spikes could be induced in spinal cord astrocytes in culture when IIR was experimentally blocked by 10 microM Ba2+, suggesting that the absence of IIR in astrocytoma cells is primarily responsible for the unusual spiking behavior seen in these glial tumor cells. It is unlikely that such glial spikes ever occur in vivo.


Asunto(s)
Astrocitos/fisiología , Astrocitoma/patología , Neoplasias Encefálicas/patología , Proteínas de Neoplasias/fisiología , Células Madre Neoplásicas/fisiología , Proteínas del Tejido Nervioso/fisiología , Canales de Potasio de Rectificación Interna , Canales de Potasio con Entrada de Voltaje , Canales de Potasio/fisiología , Canales de Sodio/fisiología , Potenciales de Acción/efectos de los fármacos , Carcinoma/patología , Diferenciación Celular , Niño , Neoplasias del Plexo Coroideo/patología , Canales de Potasio de Tipo Rectificador Tardío , Humanos , Transporte Iónico , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Técnicas de Placa-Clamp , Potasio/metabolismo , Bloqueadores de los Canales de Potasio , Canales de Potasio/análisis , Bloqueadores de los Canales de Sodio , Médula Espinal/citología , Tetraetilamonio/farmacología , Tetrodotoxina/farmacología
20.
Neuroscience ; 83(4): 1161-73, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9502255

RESUMEN

Using whole-cell patch-clamp recordings, we identified a novel voltage-activated chloride current that was selectively expressed in glioma cells from 23 patient biopsies. Chloride currents were identified in 64% of glioma cells studied in acute slices of nine patient biopsies. These derived from gliomas of various pathological grades. In addition, 98% of cells acutely isolated or in short-term culture from 23 patients diagnosed with gliomas showed chloride current expression. These currents, which we termed glioma chloride currents activated at potentials >45 mV, showed pronounced outward rectification, and were sensitive to bath application of the presumed Cl- channel specific peptide chlorotoxin (approximately 600 nM) derived from Leiurus scorpion venom. Interestingly, low grade tumours (e.g., pilocytic astrocytomas), containing more differentiated, astrocyte-like cells showed expression of glioma chloride currents in concert with voltage-activated sodium and potassium currents also seen in normal astrocytes. By contrast, high grade tumours (e.g., glioblastoma multiforme) expressed almost exclusively chloride currents, suggesting a gradual loss of Na+ currents and gain of Cl- currents with increasing pathological tumour grade. To expand on the observation that these chloride currents are glioma-specific, we introduced experimental tumours in scid mice by intracranial injection of D54MG glioma cells and subsequently recorded from tumour cells and adjacent normal glial cells in acute slices. We consistently observed expression of chlorotoxin-sensitive chloride channels in implanted glioma cells, but without evidence for expression of chloride channels in surrounding "normal" host glial cells, suggesting that these chloride channels are probably a glioma-specific feature. Finding of this novel glioma specific Cl- channel in gliomas in situ and it's selective binding of chlorotoxin may provide a way to identify or target glioma cells in the future.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/fisiopatología , Canales de Cloruro/biosíntesis , Glioma/metabolismo , Glioma/fisiopatología , Adolescente , Adulto , Anciano , Animales , Astrocitoma/metabolismo , Astrocitoma/fisiopatología , Biopsia , Niño , Preescolar , Canales de Cloruro/efectos de los fármacos , Canales de Cloruro/fisiología , Femenino , Glioblastoma/metabolismo , Glioblastoma/fisiopatología , Humanos , Técnicas In Vitro , Lactante , Masculino , Ratones , Ratones SCID , Persona de Mediana Edad , Neurotoxinas/farmacología , Técnicas de Placa-Clamp , Venenos de Escorpión/farmacología , Trasplante Heterólogo , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA