Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 43(5): 114158, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38722742

RESUMEN

Throughout the brain, astrocytes form networks mediated by gap junction channels that promote the activity of neuronal ensembles. Although their inputs on neuronal information processing are well established, how molecular gap junction channels shape neuronal network patterns remains unclear. Here, using astroglial connexin-deficient mice, in which astrocytes are disconnected and neuronal bursting patterns are abnormal, we show that astrocyte networks strengthen bursting activity via dynamic regulation of extracellular potassium levels, independently of glutamate homeostasis or metabolic support. Using a facilitation-depression model, we identify neuronal afterhyperpolarization as the key parameter underlying bursting pattern regulation by extracellular potassium in mice with disconnected astrocytes. We confirm this prediction experimentally and reveal that astroglial network control of extracellular potassium sustains neuronal afterhyperpolarization via KCNQ voltage-gated K+ channels. Altogether, these data delineate how astroglial gap junctions mechanistically strengthen neuronal population bursts and point to approaches for controlling aberrant activity in neurological diseases.


Asunto(s)
Astrocitos , Uniones Comunicantes , Hipocampo , Canales de Potasio KCNQ , Potasio , Animales , Ratones , Potenciales de Acción/fisiología , Astrocitos/metabolismo , Conexinas/metabolismo , Conexinas/genética , Uniones Comunicantes/metabolismo , Hipocampo/metabolismo , Canales de Potasio KCNQ/metabolismo , Canales de Potasio KCNQ/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Red Nerviosa/metabolismo , Neuronas/metabolismo , Potasio/metabolismo , Masculino , Femenino
2.
Nat Commun ; 15(1): 3583, 2024 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-38678030

RESUMEN

Fragile X syndrome (FXS) is an inherited form of intellectual disability caused by the loss of the mRNA-binding fragile X mental retardation protein (FMRP). FXS is characterized by neuronal hyperexcitability and behavioral defects, however the mechanisms underlying these critical dysfunctions remain unclear. Here, using male Fmr1 knockout mouse model of FXS, we identify abnormal extracellular potassium homeostasis, along with impaired potassium channel Kir4.1 expression and function in astrocytes. Further, we reveal that Kir4.1 mRNA is a binding target of FMRP. Finally, we show that the deficit in astroglial Kir4.1 underlies neuronal hyperexcitability and several behavioral defects in Fmr1 knockout mice. Viral delivery of Kir4.1 channels specifically to hippocampal astrocytes from Fmr1 knockout mice indeed rescues normal astrocyte potassium uptake, neuronal excitability, and cognitive and social performance. Our findings uncover an important role for astrocyte dysfunction in the pathophysiology of FXS, and identify Kir4.1 channel as a potential therapeutic target for FXS.


Asunto(s)
Astrocitos , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil , Síndrome del Cromosoma X Frágil , Neuronas , Canales de Potasio de Rectificación Interna , Animales , Masculino , Ratones , Astrocitos/metabolismo , Conducta Animal , Modelos Animales de Enfermedad , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/metabolismo , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/fisiopatología , Hipocampo/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Neuronas/fisiología , Potasio/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Canales de Potasio de Rectificación Interna/genética , ARN Mensajero/metabolismo , ARN Mensajero/genética
3.
Nat Commun ; 13(1): 2844, 2022 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-35606347

RESUMEN

The cerebral cortex develops from dorsal forebrain neuroepithelial progenitor cells. Following the initial expansion of the progenitor cell pool, these cells generate neurons of all the cortical layers and then astrocytes and oligodendrocytes. Yet, the regulatory pathways that control the expansion and maintenance of the progenitor cell pool are currently unknown. Here we define six basic pathway components that regulate proliferation of cortically specified human neuroepithelial stem cells (cNESCs) in vitro without the loss of cerebral cortex developmental potential. We show that activation of FGF and inhibition of BMP and ACTIVIN A signalling are required for long-term cNESC proliferation. We also demonstrate that cNESCs preserve dorsal telencephalon-specific potential when GSK3, AKT and nuclear CATENIN-ß1 activity are low. Remarkably, regulation of these six pathway components supports the clonal expansion of cNESCs. Moreover, cNESCs differentiate into lower- and upper-layer cortical neurons in vitro and in vivo. The identification of mechanisms that drive the neuroepithelial stem cell self-renewal and differentiation and preserve this potential in vitro is key to developing regenerative and cell-based therapeutic approaches to treat neurological conditions.


Asunto(s)
Glucógeno Sintasa Quinasa 3 , Células Neuroepiteliales , Diferenciación Celular/fisiología , Corteza Cerebral , Humanos , Células Madre , Telencéfalo
4.
Neuroscientist ; 28(2): 144-162, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-33567971

RESUMEN

Oligodendrocyte precursor cells (OPCs) give rise to oligodendrocytes which myelinate axons in the central nervous system. Although classically thought to be a homogeneous population, OPCs are reported to have different developmental origins and display regional and temporal diversity in their transcriptome, response to growth factors, and physiological properties. Similarly, evidence is accumulating that myelinating oligodendrocytes display transcriptional heterogeneity. Analyzing this reported heterogeneity suggests that OPCs, and perhaps also myelinating oligodendrocytes, may exist in different functional cell states. Here, we review the evidence indicating that OPCs and oligodendrocytes are diverse, and we discuss the implications of functional OPC states for myelination in the adult brain and for myelin repair.


Asunto(s)
Vaina de Mielina , Oligodendroglía , Axones/fisiología , Diferenciación Celular/fisiología , Linaje de la Célula , Sistema Nervioso Central , Humanos , Vaina de Mielina/fisiología , Oligodendroglía/metabolismo
5.
Nat Neurosci ; 24(11): 1508-1521, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34711959

RESUMEN

Myelin, a lipid membrane that wraps axons, enabling fast neurotransmission and metabolic support to axons, is conventionally thought of as a static structure that is set early in development. However, recent evidence indicates that in the central nervous system (CNS), myelination is a protracted and plastic process, ongoing throughout adulthood. Importantly, myelin is emerging as a potential modulator of neuronal networks, and evidence from human studies has highlighted myelin as a major player in shaping human behavior and learning. Here we review how myelin changes throughout life and with learning. We discuss potential mechanisms of myelination at different life stages, explore whether myelin plasticity provides the regenerative potential of the CNS white matter, and question whether changes in myelin may underlie neurological disorders.


Asunto(s)
Encéfalo/fisiología , Vaina de Mielina/fisiología , Plasticidad Neuronal/fisiología , Neuronas/fisiología , Sustancia Blanca/fisiología , Animales , Encéfalo/citología , Humanos , Oligodendroglía/fisiología , Sustancia Blanca/citología
6.
Cell Rep ; 36(2): 109362, 2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34260928

RESUMEN

The mediobasal hypothalamus (MBH; arcuate nucleus of the hypothalamus [ARH] and median eminence [ME]) is a key nutrient sensing site for the production of the complex homeostatic feedback responses required for the maintenance of energy balance. Here, we show that refeeding after an overnight fast rapidly triggers proliferation and differentiation of oligodendrocyte progenitors, leading to the production of new oligodendrocytes in the ME specifically. During this nutritional paradigm, ME perineuronal nets (PNNs), emerging regulators of ARH metabolic functions, are rapidly remodeled, and this process requires myelin regulatory factor (Myrf) in oligodendrocyte progenitors. In genetically obese ob/ob mice, nutritional regulations of ME oligodendrocyte differentiation and PNN remodeling are blunted, and enzymatic digestion of local PNN increases food intake and weight gain. We conclude that MBH PNNs are required for the maintenance of energy balance in lean mice and are remodeled in the adult ME by the nutritional control of oligodendrocyte differentiation.


Asunto(s)
Diferenciación Celular , Eminencia Media/citología , Red Nerviosa/fisiología , Fenómenos Fisiológicos de la Nutrición , Oligodendroglía/citología , Adulto , Animales , Linaje de la Célula , Proliferación Celular , Humanos , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones Endogámicos C57BL , Oligodendroglía/ultraestructura , Análisis de la Célula Individual , Transcriptoma/genética
7.
Neurosci Lett ; 703: 139-144, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-30904575

RESUMEN

Brain tissue undergoes substantial activity-dependent reorganisation after stroke due to neuronal plasticity, leading to partial functional recovery in patients. Concurrent myelin repair is crucial for proper neuronal network function and reorganisation. Myelin repair after stroke might occur as myelin plasticity or as remyelination through the recruitment and differentiation of oligodendrocyte precursor cells (OPCs), which become myelin-forming oligodendrocytes (OLs). These two processes might share a similar guiding mechanism, which is postulated to depend on neuronal activity and glutamate signaling to OPCs. However, with ageing, the ability of OPCs to differentiate into myelinating OLs decreases due to changes in their ion channel and neurotransmitter receptor expression profile, rendering them less sensitive to neuronal activity. Because of their unique ability to replace damaged OLs, OPCs represent a potential therapeutic target for myelin repair in the context of stroke.


Asunto(s)
Vaina de Mielina/fisiología , Neuronas/fisiología , Accidente Cerebrovascular/patología , Animales , Diferenciación Celular , Proliferación Celular , Humanos , Células-Madre Neurales/patología , Plasticidad Neuronal , Oligodendroglía/patología , Accidente Cerebrovascular/fisiopatología
8.
Eur J Neurosci ; 50(1): 1685-1699, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30633415

RESUMEN

The proper function of the nervous system is dependent on the balance of ions and water between the intracellular and extracellular space (ECS). It has been suggested that the interaction of aquaporin-4 (AQP4) and the transient receptor potential vaniloid isoform 4 (TRPV4) channels play a role in water balance and cell volume regulation, and indirectly, of the ECS volume. Using the real-time iontophoretic method, we studied the changes of the ECS diffusion parameters: ECS volume fraction α (α = ECS volume fraction/total tissue volume) and tortuosity λ (λ2  = free/apparent diffusion coefficient) in mice with a genetic deficiency of AQP4 or TRPV4 channels, and in control animals. The used models of cytotoxic edema included: mild and severe hypotonic stress or oxygen-glucose deprivation (OGD) in situ and terminal ischemia/anoxia in vivo. This study shows that an AQP4 or TRPV4 deficit slows down the ECS volume shrinkage during severe ischemia in vivo. We further demonstrate that a TRPV4 deficit slows down the velocity and attenuates an extent of the ECS volume decrease during OGD treatment in situ. However, in any of the cytotoxic edema models in situ (OGD, mild or severe hypotonic stress), we did not detect any alterations in the cell swelling or volume regulation caused by AQP4 deficiency. Overall, our results indicate that the AQP4 and TRPV4 channels may play a crucial role in severe pathological states associated with their overexpression and enhanced cell swelling. However, detailed interplay between AQP4 and TRPV4 channels requires further studies and additional research.


Asunto(s)
Acuaporina 4/metabolismo , Edema Encefálico/metabolismo , Espacio Extracelular/metabolismo , Corteza Somatosensorial/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Acuaporina 4/deficiencia , Modelos Animales de Enfermedad , Electrocardiografía , Femenino , Paro Cardíaco/metabolismo , Hipoglucemia/metabolismo , Hipoxia-Isquemia Encefálica/metabolismo , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Potasio/metabolismo , Canales Catiónicos TRPV/deficiencia
9.
Neuroscience ; 394: 127-143, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30367945

RESUMEN

Transient receptor potential vanilloid type 4 (TRPV4) channels are involved in astrocyte volume regulation; however, only limited data exist about its mechanism in astrocytes in situ. We performed middle cerebral artery occlusion in adult mice, where we found twice larger edema 1 day after the insult in trpv4-/- mice compared to the controls, which was quantified using magnetic resonance imaging. This result suggests disrupted volume regulation in the brain cells in trpv4-/- mice leading to increased edema formation. The aim of our study was to elucidate whether TRPV4 channel-based volume regulation occurs in astrocytes in situ and whether the disrupted volume regulation in trpv4-/- mice might lead to higher edema formation after brain ischemia. For our experiments, we used trpv4-/- mice crossed with transgenic mice expressing enhanced green fluorescent protein (EGFP) under the control of the glial fibrillary acidic protein promoter, which leads to astrocyte visualization by EGFP expression. For quantification of astrocyte volume changes, we used two-dimensional (2D) and three-dimensional (3D) morphometrical approaches and a quantification algorithm based on fluorescence intensity changes during volume alterations induced by hypotonicity or by oxygen-glucose deprivation. In contrast to in vitro experiments, we found little evidence of the contribution of TRPV4 channels to volume regulation in astrocytes in situ in adult mice. Moreover, we only found a rare expression of TRPV4 channels in adult mouse astrocytes. Our data suggest that TRPV4 channels are not involved in astrocyte volume regulation in situ; however, they play a protective role during the ischemia-induced brain edema formation.


Asunto(s)
Astrocitos/metabolismo , Astrocitos/patología , Edema Encefálico/metabolismo , Edema Encefálico/patología , Canales Catiónicos TRPV/metabolismo , Animales , Edema Encefálico/etiología , Isquemia Encefálica/complicaciones , Femenino , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Cultivo Primario de Células , Canales Catiónicos TRPV/genética
10.
J Neurosci Methods ; 306: 38-44, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29885815

RESUMEN

BACKGROUND: Several techniques for cell volume measurement using fluorescence microscopy have been established to date. In this study, we compare the performance of three different approaches which allow for estimations of the cell volume changes in biological samples containing individual fluorescently labeled cells either in culture or in the tissue context. The specific requirements, limitations and advantages of individual approaches are discussed. NEW METHOD: Global morphometric data are quantitatively compared with local information about the overall cell volume, represented by the concentration of a mobile fluorophore accumulated within the monitored cell. RESULTS: Volume changes induced by variations in the extracellular osmolarity in murine fibroblasts and astrocytes either in the culture or in the acute brain slices were registered by the three- and two-dimensional morphometries and by local fluorescence intensity measurements. The performance of the latter approach was verified using FRAP assessment of the fluorophore mobility. Significantly lower amplitudes of the cortical astrocytes swelling were detected by three-dimensional morphometry, when compared to the other two approaches. Consequently, it failed to detect temperature-induced cell volume changes. COMPARISON WITH EXISTING METHOD(S): The three most popular methods of cell volume measurement are compared to each other in this study. CONCLUSIONS: We show that the effectivity of global morphometry-based volumetric approaches drops with the increasing cell shape complexity or in the tissue context. In contrast to this, the performance of local fluorescence intensity monitoring, which is also fully capable of reflecting the instant cell volume variations remains stable, independent of the system used and application.


Asunto(s)
Astrocitos/citología , Astrocitos/fisiología , Tamaño de la Célula , Fibroblastos/citología , Fibroblastos/fisiología , Imagenología Tridimensional/métodos , Microscopía Fluorescente/métodos , Células 3T3 , Animales , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/fisiología , Colorantes Fluorescentes , Soluciones Hipertónicas , Soluciones Hipotónicas , Soluciones Isotónicas , Ratones , Microscopía Confocal/métodos
11.
Curr Pharm Des ; 23(33): 5056-5074, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28699523

RESUMEN

Brain ischemic injury represents one of the greatest medical challenges for the aging population in developed countries, yet despite strong efforts, possibilities to treat ischemic injury still remain poor. Stroke, the most common type of brain ischemic injury in humans, is caused by brain artery occlusion, and represents a focal form of ischemia, which leads to neuronal loss in the ischemic core, and glial scar formation in the penumbral region around the core. Such glial scar mainly comprises reactive astrocytes, reactive NG2 glia and activated microglia. Reactive astrocytes display distinct features when compared to healthy astroglia, including changes in their morphology, metabolism, gene expression profiles, production of extracellular matrix proteins or proliferation rate. Similarly to astrocytes in the healthy nervous tissue, reactive astrocytes surrounding the glial scar strongly influence the activity of surviving neurons around the ischemic lesion. Bringing insight into pathophysiological functions of reactive astrocytes within the glial scar might thus open new possibilities for stroke treatment. Here, we summarize the properties of reactive astrocytes, with emphasis on the expression and function of ion channels, transporters and neurotransmitter receptors; all of which possess the ability to change the functional state of astrocytes, such as the membrane equilibrium potentials for different ions. This may have major effects on the functioning of surviving neurons, consequently leading to changes in neuronal excitability and progression of secondary pathologies, such as epilepsy. Moreover, we provide possible clues for therapy, based on functional modulation of astrocytic ion transporting mechanisms.


Asunto(s)
Astrocitos/metabolismo , Isquemia Encefálica/metabolismo , Homeostasis/fisiología , Animales , Astrocitos/efectos de los fármacos , Isquemia Encefálica/tratamiento farmacológico , Gliosis/tratamiento farmacológico , Gliosis/metabolismo , Homeostasis/efectos de los fármacos , Humanos , Canales Iónicos/antagonistas & inhibidores , Canales Iónicos/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo
12.
J Neurochem ; 132(3): 286-300, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25279950

RESUMEN

Accumulating evidence indicates that increased intracellular Na(+) concentration ([Na(+) ]i ) in astroglial cells is associated with the development of brain edema under ischemic conditions, but the underlying mechanisms are still elusive. Here, we report that in primary cultured rat cortical astrocytes, elevations of [Na(+) ]i reflecting those achieved during ischemia cause a marked decrease in hypotonicity-evoked current mediated by volume-regulated anion channel (VRAC). Pharmacological manipulations revealed that VRAC inhibition was not due to the reverse mode of the plasma membrane sodium/calcium exchanger. The negative modulation of VRAC was also observed in an astrocytic cell line lacking the predominant astrocyte water channel aquaporin 4, indicating that [Na(+) ]i effect was not mediated by the regulation of aquaporin 4 activity. The inward rectifier Cl(-) current, which is also expressed by cultured astrocytes, was not affected by [Na(+) ]i increase. VRAC depression by high [Na(+) ]i was confirmed in adult astrocytes, suggesting that it was not developmentally regulated. Altogether, these results provide the first evidence that intracellular Na(+) dynamics can modulate astrocytic membrane conductance that controls functional processes linked to cell volume regulation and add further support to the concept that limiting astrocyte intracellular Na(+) accumulation might be a favorable strategy to counteract the development of brain edema.


Asunto(s)
Astrocitos/metabolismo , Corteza Cerebral/metabolismo , Canales Iónicos/efectos de los fármacos , Sodio/farmacología , Animales , Acuaporina 4/metabolismo , Astrocitos/efectos de los fármacos , Corteza Cerebral/citología , Corteza Cerebral/efectos de los fármacos , Canales de Cloruro/metabolismo , Femenino , Masculino , Técnicas de Placa-Clamp , Cultivo Primario de Células , Ratas , Ratas Sprague-Dawley , Sodio/metabolismo , Intercambiador de Sodio-Calcio/metabolismo
13.
Glia ; 62(12): 2004-21, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25042871

RESUMEN

Astrocytes respond to ischemic brain injury by proliferation, the increased expression of intermediate filaments and hypertrophy, which results in glial scar formation. In addition, they alter the expression of ion channels, receptors and transporters that maintain ionic/neurotransmitter homeostasis. Here, we aimed to demonstrate the expression of Hcn1-4 genes encoding hyperpolarization-activated cyclic nucleotide-gated (HCN) channels in reactive astrocytes following focal cerebral ischemia (FCI) or global cerebral ischemia (GCI) and to characterize their functional properties. A permanent occlusion of the middle cerebral artery (MCAo) was employed to induce FCI in adult GFAP/EGFP mice, while GCI was induced by transient bilateral common carotid artery occlusion combined with hypoxia in adult rats. Using FACS, we isolated astrocytes from non-injured or ischemic brains and performed gene expression profiling using single-cell RT-qPCR. We showed that 2 weeks after ischemia reactive astrocytes express high levels of Hcn1-4 transcripts, while immunohistochemical analyses confirmed the presence of HCN1-3 channels in reactive astrocytes 5 weeks after ischemia. Electrophysiological recordings revealed that post-ischemic astrocytes are significantly depolarized, and compared with astrocytes from non-injured brains, they display large hyperpolarization-activated inward currents, the density of which increased 2-3-fold in response to ischemia. Their activation was facilitated by cAMP and their amplitudes were decreased by ZD7288 or low extracellular Na(+) concentration, suggesting that they may belong to the family of HCN channels. Collectively, our results demonstrate that regardless of the type of ischemic injury, reactive astrocytes express HCN channels, which could therefore be an important therapeutic target in poststroke therapy.


Asunto(s)
Astrocitos/metabolismo , Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Regulación de la Expresión Génica/fisiología , Isquemia/patología , Animales , Astrocitos/efectos de los fármacos , Encéfalo/citología , AMP Cíclico/farmacología , Canales Catiónicos Regulados por Nucleótidos Cíclicos/genética , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Técnicas In Vitro , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Pirimidinas/farmacología , Ratas , Ratas Wistar , Sodio/metabolismo
14.
PLoS One ; 7(5): e36816, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22590616

RESUMEN

Polydendrocytes (also known as NG2 glial cells) constitute a fourth major glial cell type in the adult mammalian central nervous system (CNS) that is distinct from other cell types. Although much evidence suggests that these cells are multipotent in vitro, their differentiation potential in vivo under physiological or pathophysiological conditions is still controversial.To follow the fate of polydendrocytes after CNS pathology, permanent middle cerebral artery occlusion (MCAo), a commonly used model of focal cerebral ischemia, was carried out on adult NG2creBAC:ZEG double transgenic mice, in which enhanced green fluorescent protein (EGFP) is expressed in polydendrocytes and their progeny. The phenotype of the EGFP(+) cells was analyzed using immunohistochemistry and the patch-clamp technique 3, 7 and 14 days after MCAo. In sham-operated mice (control), EGFP(+) cells in the cortex expressed protein markers and displayed electrophysiological properties of polydendrocytes and oligodendrocytes. We did not detect any co-labeling of EGFP with neuronal, microglial or astroglial markers in this region, thus proving polydendrocyte unipotent differentiation potential under physiological conditions. Three days after MCAo the number of EGFP(+) cells in the gliotic tissue dramatically increased when compared to control animals, and these cells displayed properties of proliferating cells. However, in later phases after MCAo a large subpopulation of EGFP(+) cells expressed protein markers and electrophysiological properties of astrocytes that contribute to the formation of glial scar. Importantly, some EGFP(+) cells displayed membrane properties typical for neural precursor cells, and moreover these cells expressed doublecortin (DCX)--a marker of newly-derived neuronal cells. Taken together, our data indicate that polydendrocytes in the dorsal cortex display multipotent differentiation potential after focal ischemia.


Asunto(s)
Antígenos de Diferenciación/biosíntesis , Isquemia Encefálica/metabolismo , Diferenciación Celular , Proliferación Celular , Proteínas del Tejido Nervioso/biosíntesis , Neuroglía/metabolismo , Animales , Isquemia Encefálica/patología , Modelos Animales de Enfermedad , Proteína Doblecortina , Ratones , Ratones Transgénicos , Neuroglía/patología
15.
PLoS One ; 7(1): e29725, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22253765

RESUMEN

Recently, we have identified two astrocytic subpopulations in the cortex of GFAP-EGFP mice, in which the astrocytes are visualized by the enhanced green-fluorescent protein (EGFP) under the control of the human glial fibrillary acidic protein (GFAP) promotor. These astrocytic subpopulations, termed high response- (HR-) and low response- (LR-) astrocytes, differed in the extent of their swelling during oxygen-glucose deprivation (OGD). In the present study we focused on identifying the ion channels or transporters that might underlie the different capabilities of these two astrocytic subpopulations to regulate their volume during OGD. Using three-dimensional confocal morphometry, which enables quantification of the total astrocytic volume, the effects of selected inhibitors of K⁺ and Cl⁻ channels/transporters or glutamate transporters on astrocyte volume changes were determined during 20 minute-OGD in situ. The inhibition of volume regulated anion channels (VRACs) and two-pore domain potassium channels (K(2P)) highlighted their distinct contributions to volume regulation in HR-/LR-astrocytes. While the inhibition of VRACs or K(2P) channels revealed their contribution to the swelling of HR-astrocytes, in LR-astrocytes they were both involved in anion/K⁺ effluxes. Additionally, the inhibition of Na⁺-K⁺-Cl⁻ co-transporters in HR-astrocytes led to a reduction of cell swelling, but it had no effect on LR-astrocyte volume. Moreover, employing real-time single-cell quantitative polymerase chain reaction (PCR), we characterized the expression profiles of EGFP-positive astrocytes with a focus on those ion channels and transporters participating in astrocyte swelling and volume regulation. The PCR data revealed the existence of two astrocytic subpopulations markedly differing in their gene expression levels for inwardly rectifying K⁺ channels (Kir4.1), K(2P) channels (TREK-1 and TWIK-1) and Cl⁻ channels (ClC2). Thus, we propose that the diverse volume changes displayed by cortical astrocytes during OGD mainly result from their distinct expression patterns of ClC2 and K(2P) channels.


Asunto(s)
Astrocitos/citología , Astrocitos/metabolismo , Corteza Cerebral/citología , Canales de Cloruro/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Canales de Potasio/metabolismo , Animales , Astrocitos/efectos de los fármacos , Tamaño de la Célula/efectos de los fármacos , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/deficiencia , Humanos , Técnicas In Vitro , Masculino , Moduladores del Transporte de Membrana/farmacología , Ratones , Ratones Transgénicos , Modelos Biológicos , Oxígeno , Caracteres Sexuales , Simportadores de Cloruro de Sodio-Potasio/metabolismo , Simportadores/metabolismo , Proteínas de Transporte Vesicular de Glutamato/metabolismo , Cotransportadores de K Cl
16.
Cell Mol Neurobiol ; 31(7): 1113-22, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21630007

RESUMEN

Adipose-derived stromal cells (ASCs) are an alternative source of stem cells for cell-based therapies of neurological disorders such as spinal cord injury (SCI). In the present study, we predifferentiated ASCs (pASCs) and compared their behavior with naïve ASCs in vitro and after transplantation into rats with a balloon-induced compression lesion. ASCs were predifferentiated into spheres before transplantation, then pASCs or ASCs were injected intraspinally 1 week after SCI. The cells' fate and the rats' functional outcome were assessed using behavioral, histological, and electrophysiological methods. Immunohistological analysis of pASCs in vitro revealed the expression of NCAM, NG2, S100, and p75. Quantitative RT-PCR at different intervals after neural induction showed the up-regulated expression of the glial markers NG2 and p75 and the neural precursor markers NCAM and Nestin. Patch clamp analysis of pASCs revealed three different types of membrane currents; however, none were fast activating Na(+) currents indicating a mature neuronal phenotype. Significant improvement in both the pASC and ASC transplanted groups was observed in the BBB motor test. In vivo, pASCs survived better than ASCs did and interacted closely with the host tissue, wrapping host axons and oligodendrocytes. Some transplanted cells were NG2- or CD31-positive, but no neuronal markers were detected. The predifferentiation of ASCs plays a beneficial role in SCI repair by promoting the protection of denuded axons; however, functional improvements were comparable in both the groups, indicating that repair was induced mainly through paracrine mechanisms.


Asunto(s)
Tejido Adiposo/fisiología , Diferenciación Celular/fisiología , Células Madre Multipotentes/fisiología , Traumatismos de la Médula Espinal/cirugía , Trasplante de Células Madre/métodos , Células del Estroma/trasplante , Tejido Adiposo/citología , Animales , Conducta Animal/fisiología , Células Cultivadas , Masculino , Actividad Motora/fisiología , Células Madre Multipotentes/citología , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Ratas Wistar , Traumatismos de la Médula Espinal/patología
17.
J Cereb Blood Flow Metab ; 31(3): 894-907, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20877389

RESUMEN

To understand the structural alterations that underlie early and late changes in hippocampal diffusivity after hypoxia/ischemia (H/I), the changes in apparent diffusion coefficient of water (ADC(W)) were studied in 8-week-old rats after H/I using diffusion-weighted magnetic resonance imaging (DW-MRI). In the hippocampal CA1 region, ADC(W) analyses were performed during 6 months of reperfusion and compared with alterations in cell number/cell-type composition, glial morphology, and extracellular space (ECS) diffusion parameters obtained by the real-time iontophoretic method. In the early phases of reperfusion (1 to 3 days) neuronal cell death, glial proliferation, and developing gliosis were accompanied by an ADC(W) decrease and tortuosity increase. Interestingly, ECS volume fraction was decreased only first day after H/I. In the late phases of reperfusion (starting 1 month after H/I), when the CA1 region consisted mainly of microglia, astrocytes, and NG2-glia with markedly altered morphology, ADC(W), ECS volume fraction and tortuosity were increased. Three-dimensional confocal morphometry revealed enlarged astrocytes and shrunken NG2-glia, and in both the contribution of cell soma/processes to total cell volume was markedly increased/decreased. In summary, the ADC(W) increase in the CA1 region underlain by altered cellular composition and glial morphology suggests that considerable changes in extracellular signal transmission might occur in the late phases of reperfusion after H/I.


Asunto(s)
Agua Corporal/metabolismo , Isquemia Encefálica/patología , Isquemia Encefálica/fisiopatología , Región CA1 Hipocampal/patología , Proliferación Celular , Hipoxia/patología , Neuroglía/patología , Animales , Astrocitos/patología , Isquemia Encefálica/complicaciones , Región CA1 Hipocampal/fisiopatología , Recuento de Células , Muerte Celular , Difusión , Imagen de Difusión por Resonancia Magnética , Espacio Extracelular/metabolismo , Gliosis/etiología , Gliosis/patología , Hipoxia/complicaciones , Hipoxia/fisiopatología , Imagenología Tridimensional , Inmunohistoquímica , Masculino , Microscopía Confocal , Ratas , Ratas Wistar , Reperfusión , Factores de Tiempo
18.
Neurochem Int ; 57(7): 783-94, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20833221

RESUMEN

Astrocytes and NG2 glia respond to CNS injury by the formation of a glial scar. Since the changes in K(+) currents in astrocytes and NG2 glia that accompany glial scar formation might influence tissue outcome by altering K(+) ion homeostasis, we aimed to characterize the changes in K(+) currents in hippocampal astrocytes and NG2 glia during an extended time window of reperfusion after ischemic injury. Global cerebral ischemia was induced in adult rats by bilateral, 15-min common carotid artery occlusion combined with low-pressure oxygen ventilation. Using the patch-clamp technique, we investigated the membrane properties of hippocampal astrocytes and NG2 glia in situ 2 hours, 6 hours, 1 day, 3 days, 7 days or 5 weeks after ischemia. Astrocytes in the CA1 region of the hippocampus progressively depolarized starting 3 days after ischemia, which coincided with decreased Kir4.1 protein expression in the gliotic tissue. Other K(+) channels described previously in astrocytes, such as Kir2.1, Kir5.1 and TREK1, did not show any changes in their protein content in the hippocampus after ischemia; however, their expression switched from neurons to reactive astrocytes, as visualized by immunohistochemistry. NG2 glia displayed increased input resistance, decreased membrane capacitance, increased delayed outwardly rectifying and A-type K(+) currents and decreased inward K(+) currents 3 days after ischemia, accompanied by their proliferation. Our results show that the membrane properties of astrocytes after ischemia undergo complex alterations, which might profoundly influence the maintenance of K(+) homeostasis in the damaged tissue, while NG2 glia display membrane currents typical of proliferating cells.


Asunto(s)
Isquemia Encefálica/metabolismo , Región CA1 Hipocampal/metabolismo , Membrana Celular/metabolismo , Polaridad Celular/fisiología , Gliosis/metabolismo , Potenciales de la Membrana/fisiología , Neuroglía/metabolismo , Canales de Potasio de Rectificación Interna/antagonistas & inhibidores , Animales , Isquemia Encefálica/patología , Isquemia Encefálica/fisiopatología , Región CA1 Hipocampal/patología , Región CA1 Hipocampal/fisiopatología , Membrana Celular/patología , Regulación hacia Abajo/genética , Regulación hacia Abajo/fisiología , Gliosis/genética , Gliosis/patología , Masculino , Neuroglía/patología , Canales de Potasio de Rectificación Interna/biosíntesis , Canales de Potasio de Rectificación Interna/genética , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...