Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Aging Cell ; : e14201, 2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38769809

RESUMEN

The hippocampus is a brain area linked to cognition. The mechanisms that maintain cognitive activity in humans are poorly understood. Centenarians display extreme longevity which is generally accompanied by better quality of life, lower cognitive impairment, and reduced incidence of pathologies including neurodegenerative diseases. We performed transcriptomic studies in hippocampus samples from individuals of different ages (centenarians [≥97 years], old, and young) and identified a differential gene expression pattern in centenarians compared to the other two groups. In particular, several isoforms of metallothioneins (MTs) were highly expressed in centenarians. Moreover, we identified that MTs were mainly expressed in astrocytes. Functional studies in human primary astrocytes revealed that MT1 and MT3 are necessary for their homeostasis maintenance. Overall, these results indicate that the expression of MTs specifically in astrocytes is a mechanism for protection during aging.

2.
Science ; 381(6663): 1176-1182, 2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-37708272

RESUMEN

Neuronal cell loss is a defining feature of Alzheimer's disease (AD), but the underlying mechanisms remain unclear. We xenografted human or mouse neurons into the brain of a mouse model of AD. Only human neurons displayed tangles, Gallyas silver staining, granulovacuolar neurodegeneration (GVD), phosphorylated tau blood biomarkers, and considerable neuronal cell loss. The long noncoding RNA MEG3 was strongly up-regulated in human neurons. This neuron-specific long noncoding RNA is also up-regulated in AD patients. MEG3 expression alone was sufficient to induce necroptosis in human neurons in vitro. Down-regulation of MEG3 and inhibition of necroptosis using pharmacological or genetic manipulation of receptor-interacting protein kinase 1 (RIPK1), RIPK3, or mixed lineage kinase domain-like protein (MLKL) rescued neuronal cell loss in xenografted human neurons. This model suggests potential therapeutic approaches for AD and reveals a human-specific vulnerability to AD.


Asunto(s)
Enfermedad de Alzheimer , Necroptosis , Neuronas , ARN Largo no Codificante , Animales , Humanos , Ratones , Enfermedad de Alzheimer/patología , Xenoinjertos , Necroptosis/genética , Neuronas/patología , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Proteínas Quinasas/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética
3.
Cell Rep ; 42(9): 113129, 2023 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-37708025

RESUMEN

Lee et al.1 report that loss of the Alzheimer's disease risk factor SORL1 results in neuron-specific reduction in APOE and CLU, altered lipid homeostasis, and increased Aß levels and phosphorylated Tau, both rescued by stabilizing retromer or enhancing autophagy.

5.
Ann N Y Acad Sci ; 1518(1): 120-130, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36285711

RESUMEN

The evolution of the nervous system progressed through cellular diversification and specialization of functions. Conceptually, the nervous system is composed of electrically excitable neuronal networks connected by chemical synapses and nonexcitable glial cells that provide for homeostasis and defense. The evolution of neuroglia began with the emergence of the centralized nervous system and proceeded through a continuous increase in their complexity. In the primate brain, especially in the brain of humans, the astrocyte lineage is exceedingly complex, with the emergence of new types of astroglial cells possibly involved in interlayer communication and integration.


Asunto(s)
Astrocitos , Neuroglía , Humanos , Animales , Neuroglía/fisiología , Astrocitos/fisiología , Neuronas/fisiología , Sinapsis/fisiología , Encéfalo/fisiología , Oligodendroglía/fisiología
6.
Mol Neurodegener ; 16(1): 68, 2021 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-34563212

RESUMEN

BACKGROUND: Increasing evidence for a direct contribution of astrocytes to neuroinflammatory and neurodegenerative processes causing Alzheimer's disease comes from molecular and functional studies in rodent models. However, these models may not fully recapitulate human disease as human and rodent astrocytes differ considerably in morphology, functionality, and gene expression. RESULTS: To address these challenges, we established an approach to study human astrocytes within the mouse brain by transplanting human induced pluripotent stem cell (hiPSC)-derived astrocyte progenitors into neonatal brains. Xenografted hiPSC-derived astrocyte progenitors differentiated into astrocytes that integrated functionally within the mouse host brain and matured in a cell-autonomous way retaining human-specific morphologies, unique features, and physiological properties. In Alzheimer´s chimeric brains, transplanted hiPSC-derived astrocytes responded to the presence of amyloid plaques undergoing morphological changes that seemed independent of the APOE allelic background. CONCLUSIONS: In sum, we describe here a promising approach that consist of transplanting patient-derived and genetically modified astrocytes into the mouse brain to study human astrocyte pathophysiology in the context of Alzheimer´s disease.


Asunto(s)
Enfermedad de Alzheimer , Células Madre Pluripotentes Inducidas , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Astrocitos/metabolismo , Encéfalo/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Placa Amiloide/metabolismo
7.
Cells ; 10(3)2021 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-33806259

RESUMEN

Astrocytes perform a wide variety of essential functions defining normal operation of the nervous system and are active contributors to the pathogenesis of neurodegenerative disorders such as Alzheimer's among others. Recent data provide compelling evidence that distinct astrocyte states are associated with specific stages of Alzheimer´s disease. The advent of transcriptomics technologies enables rapid progress in the characterisation of such pathological astrocyte states. In this review, we provide an overview of the origin, main functions, molecular and morphological features of astrocytes in physiological as well as pathological conditions related to Alzheimer´s disease. We will also explore the main roles of astrocytes in the pathogenesis of Alzheimer´s disease and summarize main transcriptional changes and altered molecular pathways observed in astrocytes during the course of the disease.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Astrocitos/metabolismo , Transcriptoma/genética , Humanos
8.
Lancet Neurol ; 18(4): 406-414, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30795987

RESUMEN

BACKGROUND: Astrocytes, also called astroglia, maintain homoeostasis of the brain by providing trophic and metabolic support to neurons. They recycle neurotransmitters, stimulate synaptogenesis and synaptic neurotransmission, form part of the blood-brain barrier, and regulate regional blood flow. Although astrocytes have been known to display morphological alterations in Alzheimer's disease for more than a century, research has remained neurocentric. Emerging evidence suggests that these morphological changes reflect functional alterations that affect disease. RECENT DEVELOPMENTS: Genetic studies indicate that most of the risk of developing late onset Alzheimer's disease, the most common form of the disease, affecting patients aged 65 years and older, is associated with genes (ie, APOE, APOJ, and SORL) that are mainly expressed by glial cells (ie, astrocytes, microglia, and oligodendrocytes). This insight has moved the focus of research away from neurons and towards glial cells and neuroinflammation. Molecular studies in rodent models suggest a direct contribution of astrocytes to neuroinflammatory and neurodegenerative processes causing Alzheimer's disease; however, these models might insufficiently mimic the human disease, because rodent astrocytes differ considerably in morphology, functionality, and gene expression. In-vivo studies using stem-cell derived human astrocytes are allowing exploration of the human disease and providing insights into the neurotoxic or protective contributions of these cells to the pathogenesis of disease. The first attempts to develop astrocytic biomarkers and targeted therapies are emerging. WHERE NEXT?: Single-cell transcriptomics allows the fate of individual astrocytes to be followed in situ and provides the granularity needed to describe healthy and pathological cellular states at different stages of Alzheimer's disease. Given the differences between human and rodent astroglia, study of human cells in this way will be crucial. Although refined single-cell transcriptomic analyses of human post-mortem brains are important for documentation of pathology, they only provide snapshots of a dynamic reality. Thus, functional work studying human astrocytes generated from stem cells and exposed to pathological conditions in rodent brain or cell culture are needed to understand the role of these cells in the pathogenesis of Alzheimer's disease. These studies will lead to novel biomarkers and hopefully a series of new drug targets to tackle this disease.


Asunto(s)
Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Astrocitos/patología , Anciano , Enfermedad de Alzheimer/genética , Animales , Astrocitos/fisiología , Humanos , Transcriptoma
9.
Proc Natl Acad Sci U S A ; 116(1): 277-286, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30578322

RESUMEN

The mitochondrial intramembrane rhomboid protease PARL has been implicated in diverse functions in vitro, but its physiological role in vivo remains unclear. Here we show that Parl ablation in mouse causes a necrotizing encephalomyelopathy similar to Leigh syndrome, a mitochondrial disease characterized by disrupted energy production. Mice with conditional PARL deficiency in the nervous system, but not in muscle, develop a similar phenotype as germline Parl KOs, demonstrating the vital role of PARL in neurological homeostasis. Genetic modification of two major PARL substrates, PINK1 and PGAM5, do not modify this severe neurological phenotype. Parl-/- brain mitochondria are affected by progressive ultrastructural changes and by defects in Complex III (CIII) activity, coenzyme Q (CoQ) biosynthesis, and mitochondrial calcium metabolism. PARL is necessary for the stable expression of TTC19, which is required for CIII activity, and of COQ4, which is essential in CoQ biosynthesis. Thus, PARL plays a previously overlooked constitutive role in the maintenance of the respiratory chain in the nervous system, and its deficiency causes progressive mitochondrial dysfunction and structural abnormalities leading to neuronal necrosis and Leigh-like syndrome.


Asunto(s)
Complejo III de Transporte de Electrones/metabolismo , Enfermedad de Leigh/etiología , Metaloproteasas/deficiencia , Proteínas Mitocondriales/deficiencia , Ubiquinona/metabolismo , Animales , Encéfalo/metabolismo , Calcio/metabolismo , Enfermedad de Leigh/metabolismo , Enfermedad de Leigh/fisiopatología , Hígado/metabolismo , Masculino , Potencial de la Membrana Mitocondrial , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Encefalomiopatías Mitocondriales/metabolismo , Encefalomiopatías Mitocondriales/fisiopatología , Músculo Esquelético/metabolismo , Especies Reactivas de Oxígeno/metabolismo
11.
Rev Neurosci ; 28(8): 869-892, 2017 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-28779572

RESUMEN

Mutant mice deficient in hyaluronan (HA) have an epileptic phenotype. HA is one of the major constituents of the brain extracellular matrix. HA has a remarkable hydration capacity, and a lack of HA causes reduced extracellular space (ECS) volume in the brain. Reducing ECS volume can initiate or exacerbate epileptiform activity in many in vitro models of epilepsy. There is both in vitro and in vivo evidence of a positive feedback loop between reduced ECS volume and synchronous neuronal activity. Reduced ECS volume promotes epileptiform activity primarily via enhanced ephaptic interactions and increased extracellular potassium concentration; however, the epileptiform activity in many models, including the brain slices from HA synthase-3 knockout mice, may still require glutamate-mediated synaptic activity. In brain slice epilepsy models, hyperosmotic solution can effectively shrink cells and thus increase ECS volume and block epileptiform activity. However, in vivo, the intravenous administration of hyperosmotic solution shrinks both brain cells and brain ECS volume. Instead, manipulations that increase the synthesis of high-molecular-weight HA or decrease its breakdown may be used in the future to increase brain ECS volume and prevent seizures in patients with epilepsy. The prevention of epileptogenesis is also a future target of HA manipulation. Head trauma, ischemic stroke, and other brain insults that initiate epileptogenesis are known to be associated with an early decrease in high-molecular-weight HA, and preventing that decrease in HA may prevent the epileptogenesis.


Asunto(s)
Encéfalo/metabolismo , Epilepsia/metabolismo , Espacio Extracelular/metabolismo , Hialuronano Sintasas/genética , Ácido Hialurónico/metabolismo , Animales , Encéfalo/fisiopatología , Epilepsia/tratamiento farmacológico , Epilepsia/genética , Hialuronano Sintasas/metabolismo , Ácido Hialurónico/uso terapéutico , Ratones
13.
Mol Neurodegener ; 12(1): 25, 2017 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-28279219

RESUMEN

BACKGROUND: The mechanisms behind Aß-peptide accumulation in non-familial Alzheimer's disease (AD) remain elusive. Proteins of the tetraspanin family modulate Aß production by interacting to γ-secretase. METHODS: We searched for tetraspanins with altered expression in AD brains. The function of the selected tetraspanin was studied in vitro and the physiological relevance of our findings was confirmed in vivo. RESULTS: Tetraspanin-6 (TSPAN6) is increased in AD brains and overexpression in cells exerts paradoxical effects on Amyloid Precursor Protein (APP) metabolism, increasing APP-C-terminal fragments (APP-CTF) and Aß levels at the same time. TSPAN6 affects autophagosome-lysosomal fusion slowing down the degradation of APP-CTF. TSPAN6 recruits also the cytosolic, exosome-forming adaptor syntenin which increases secretion of exosomes that contain APP-CTF. CONCLUSIONS: TSPAN6 is a key player in the bifurcation between lysosomal-dependent degradation and exosome mediated secretion of APP-CTF. This corroborates the central role of the autophagosomal/lysosomal pathway in APP metabolism and shows that TSPAN6 is a crucial player in APP-CTF turnover.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Tetraspaninas/metabolismo , Animales , Western Blotting , Exosomas/metabolismo , Exosomas/ultraestructura , Humanos , Imagenología Tridimensional , Inmunohistoquímica , Lisosomas/metabolismo , Lisosomas/ultraestructura , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Neuronas/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
14.
PLoS One ; 12(2): e0171968, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28207852

RESUMEN

Tetraspanins (Tspan) are transmembrane proteins with important scaffold and signalling functions. Deletions of Tetraspanin 6 (Tspan6) gene, a member of the tetraspanin family, have been reported in patients with Epilepsy Female-restricted with Mental Retardation (EFMR). Interestingly, mutations in Tspan7, highly homologous to Tspan6, are associated with X-linked intellectual disability, suggesting that these two proteins are important for cognition. Considering recent evidences showing that Tspan7 plays a key role in synapse development and AMPAR trafficking, we initiated the study of Tspan6 in synaptic function using a Tspan6 knock out mouse model. Here we report that hippocampal field recordings from Tspan6 knock out mice show an enhanced basal synaptic transmission and impaired long term potentiation (LTP). A normal paired-pulse facilitation response suggests that Tspan6 affects the properties of the postsynaptic rather than the presynaptic terminal. However, no changes in spine morphology or postsynaptic markers could be detected in Tspan6 KO mice compared with wild types. In addition, Tspan6 KO mice show normal locomotor behaviour and no defects in hippocampus-dependent memory tests.


Asunto(s)
Conducta Animal , Hipocampo/fisiología , Potenciación a Largo Plazo/fisiología , Transmisión Sináptica/fisiología , Tetraspaninas/fisiología , Animales , Femenino , Hipocampo/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Plasticidad Neuronal
15.
Neuron ; 93(5): 1066-1081.e8, 2017 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-28238547

RESUMEN

Human pluripotent stem cells (PSCs) provide a unique entry to study species-specific aspects of human disorders such as Alzheimer's disease (AD). However, in vitro culture of neurons deprives them of their natural environment. Here we transplanted human PSC-derived cortical neuronal precursors into the brain of a murine AD model. Human neurons differentiate and integrate into the brain, express 3R/4R Tau splice forms, show abnormal phosphorylation and conformational Tau changes, and undergo neurodegeneration. Remarkably, cell death was dissociated from tangle formation in this natural 3D model of AD. Using genome-wide expression analysis, we observed upregulation of genes involved in myelination and downregulation of genes related to memory and cognition, synaptic transmission, and neuron projection. This novel chimeric model for AD displays human-specific pathological features and allows the analysis of different genetic backgrounds and mutations during the course of the disease.


Asunto(s)
Enfermedad de Alzheimer/patología , Encéfalo , Diferenciación Celular/fisiología , Neuritas/metabolismo , Neuronas/metabolismo , Células Madre Pluripotentes/citología , Proteínas tau/metabolismo , Enfermedad de Alzheimer/diagnóstico , Animales , Encéfalo/metabolismo , Encéfalo/patología , Muerte Celular/fisiología , Humanos , Ratones , Fosforilación
17.
J Cell Sci ; 128(3): 541­52, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25501810

RESUMEN

Mutations in leucine-rich repeat kinase 2 (LRRK2) are associated with Parkinson's disease, but the precise physiological function of the protein remains ill-defined. Recently, our group proposed a model in which LRRK2 kinase activity is part of an EndoA phosphorylation cycle that facilitates efficient vesicle formation at synapses in the Drosophila melanogaster neuromuscular junctions.Flies harbor only one Lrrk gene, which might encompass the functions of both mammalian LRRK1 and LRRK2. We therefore studied the role of LRRK2 in mammalian synaptic function and provide evidence that knockout or pharmacological inhibition of LRRK2 results in defects in synaptic vesicle endocytosis, altered synaptic morphology and impairments in neurotransmission. In addition, our data indicate that mammalian endophilin A1 (EndoA1,also known as SH3GL2) is phosphorylated by LRRK2 in vitro at T73 and S75, two residues in the BAR domain. Hence, our results indicate that LRRK2 kinase activity has an important role in the regulation of clathrin-mediated endocytosis of synaptic vesicles and subsequent neurotransmission at the synapse.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Endocitosis/genética , Proteínas Serina-Treonina Quinasas/genética , Transmisión Sináptica/genética , Vesículas Sinápticas/genética , Animales , Células Cultivadas , Clatrina/metabolismo , Drosophila melanogaster , Dinamina I/antagonistas & inhibidores , Endocitosis/efectos de los fármacos , Hipocampo/citología , Hidrazonas/farmacología , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/fisiología , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Ratas , Ratas Long-Evans , Sacarosa/farmacología , Transmisión Sináptica/efectos de los fármacos
18.
J Neurosci ; 34(18): 6164-76, 2014 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-24790187

RESUMEN

Hyaluronan (HA), a large anionic polysaccharide (glycosaminoglycan), is a major constituent of the extracellular matrix of the adult brain. To address its function, we examined the neurophysiology of knock-out mice deficient in hyaluronan synthase (Has) genes. Here we report that these Has mutant mice are prone to epileptic seizures, and that in Has3(-/-) mice, this phenotype is likely derived from a reduction in the size of the brain extracellular space (ECS). Among the three Has knock-out models, namely Has3(-/-), Has1(-/-), and Has2(CKO), the seizures were most prevalent in Has3(-/-) mice, which also showed the greatest HA reduction in the hippocampus. Electrophysiology in Has3(-/-) brain slices demonstrated spontaneous epileptiform activity in CA1 pyramidal neurons, while histological analysis revealed an increase in cell packing in the CA1 stratum pyramidale. Imaging of the diffusion of a fluorescent marker revealed that the transit of molecules through the ECS of this layer was reduced. Quantitative analysis of ECS by the real-time iontophoretic method demonstrated that ECS volume was selectively reduced in the stratum pyramidale by ∼ 40% in Has3(-/-) mice. Finally, osmotic manipulation experiments in brain slices from Has3(-/-) and wild-type mice provided evidence for a causal link between ECS volume and epileptiform activity. Our results provide the first direct evidence for the physiological role of HA in the regulation of ECS volume, and suggest that HA-based preservation of ECS volume may offer a novel avenue for development of antiepileptogenic treatments.


Asunto(s)
Encéfalo/patología , Epilepsia/patología , Espacio Extracelular/metabolismo , Glucuronosiltransferasa/deficiencia , Ácido Hialurónico/deficiencia , Neuronas/fisiología , Potenciales de Acción/genética , Animales , Estimulación Eléctrica , Electroencefalografía , Epilepsia/genética , Antagonistas de Aminoácidos Excitadores/farmacología , Espacio Extracelular/genética , Glucuronosiltransferasa/genética , Hialuronano Sintasas , Técnicas In Vitro , Ratones , Ratones Noqueados , Modelos Neurológicos , Mutación/genética , Red Nerviosa/metabolismo , Red Nerviosa/patología , Neuronas/efectos de los fármacos , Fosfopiruvato Hidratasa/metabolismo , Quinoxalinas/farmacología
19.
Neurobiol Dis ; 37(1): 156-65, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19804828

RESUMEN

Transient focal cerebral ischemia leads to extensive excitotoxic glial damage in the subcortical white matter. Efficient reuptake of released glutamate is essential for preventing glutamate receptor overstimulation and neuronal and glial death. The present study evaluates the expression of the main glutamate transporters (EAAT1, EAAT2, and EAAT3) in subcortical white matter of the rat after transient middle cerebral artery occlusion. Western blot analysis and immunohistochemistry show an increase in the expression of EAAT1 and EAAT2 in subcortical white matter early after ischemia which subsequently decreases at longer reperfusion periods. However, expression of both EAAT1 and EAAT2 remains higher in astrocytes forming the gliotic scar and in microglial/macrophage cells at the border of or within the infarct area, respectively. Taken together, these results indicate that there is a transient enhanced expression of EAATs in the subcortical white matter early after ischemia. Our findings reveal an adaptive response of subcortical white matter to increased levels of glutamate during focal cerebral ischemia which may limit excitotoxic damage.


Asunto(s)
Encéfalo/metabolismo , Transportador 1 de Aminoácidos Excitadores/metabolismo , Transportador 2 de Aminoácidos Excitadores/metabolismo , Transportador 3 de Aminoácidos Excitadores/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Fibras Nerviosas Mielínicas/metabolismo , Animales , Astrocitos/metabolismo , Astrocitos/patología , Western Blotting , Encéfalo/patología , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Inmunohistoquímica , Infarto de la Arteria Cerebral Media/patología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Microglía/metabolismo , Microglía/patología , Fibras Nerviosas Mielínicas/patología , Examen Neurológico , Ratas , Ratas Wistar , Factores de Tiempo
20.
Glia ; 56(12): 1353-67, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18551624

RESUMEN

Recent findings suggest that synaptic-type glutamate signaling operates between axons and their supporting glial cells. Glutamate reuptake will be a necessary component of such a system. Evidence for glutamate-mediated damage of oligodendroglia somata and processes in white matter suggests that glutamate regulation in white matter structures is also of clinical importance. The expression of glutamate transporters was examined in postnatal Day 14-17 (P14-17) mouse and in mature mouse and rat optic nerve using immuno-histochemistry and immuno-electron microscopy. EAAC1 was the major glutamate transporter detected in oligodendroglia cell membranes in both developing and mature optic nerve, while GLT-1 was the most heavily expressed transporter in the membranes of astrocytes. Both EAAC1 and GLAST were also seen in adult astrocytes, but there was little membrane expression of either at P14-17. GLAST, EAAC1, and GLT-1 were expressed in P14-17 axons with marked GLT-1 expression in the axolemma, while in mature axons EAAC1 was abundant at the node of Ranvier. Functional glutamate transport was probed in P14-17 mouse optic nerve revealing Na+-dependent, TBOA-blockable uptake of D-aspartate in astrocytes, axons, and oligodendrocytes. The data show that in addition to oligodendroglia and astrocytes, axons represent a potential source for extracellular glutamate in white matter during ischaemic conditions, and have the capacity for Na(+)-dependent glutamate uptake. The findings support the possibility of functional synaptic-type glutamate release from central axons, an event that will require axonal glutamate reuptake.


Asunto(s)
Sistema de Transporte de Aminoácidos X-AG/fisiología , Axones/metabolismo , Ácido Glutámico/metabolismo , Neuroglía/metabolismo , Nervio Óptico/metabolismo , Sistema de Transporte de Aminoácidos X-AG/ultraestructura , Animales , Axones/ultraestructura , Femenino , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Neuroglía/ultraestructura , Nervio Óptico/ultraestructura , Transporte de Proteínas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...