Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Cell ; 146(5): 785-98, 2011 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-21855979

RESUMEN

Basal synaptic transmission involves the release of neurotransmitters at individual synapses in response to a single action potential. Recent discoveries show that astrocytes modulate the activity of neuronal networks upon sustained and intense synaptic activity. However, their ability to regulate basal synaptic transmission remains ill defined and controversial. Here, we show that astrocytes in the hippocampal CA1 region detect synaptic activity induced by single-synaptic stimulation. Astrocyte activation occurs at functional compartments found along astrocytic processes and involves metabotropic glutamate subtype 5 receptors. In response, astrocytes increase basal synaptic transmission, as revealed by the blockade of their activity with a Ca(2+) chelator. Astrocytic modulation of basal synaptic transmission is mediated by the release of purines and the activation of presynaptic A(2A) receptors by adenosine. Our work uncovers an essential role for astrocytes in the regulation of elementary synaptic communication and provides insight into fundamental aspects of brain function.


Asunto(s)
Astrocitos/metabolismo , Hipocampo/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Sinapsis/metabolismo , Animales , Astrocitos/citología , Encéfalo/metabolismo , Ratones , Neuroglía/citología , Neuroglía/metabolismo , Neuronas/metabolismo , Ratas , Receptor del Glutamato Metabotropico 5
2.
Nature ; 583(7817): 603-608, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32641832

RESUMEN

Astrocytes take up glucose from the bloodstream to provide energy to the brain, thereby allowing neuronal activity and behavioural responses1-5. By contrast, astrocytes are under neuronal control through specific neurotransmitter receptors5-7. However, whether the activation of astroglial receptors can directly regulate cellular glucose metabolism to eventually modulate behavioural responses is unclear. Here we show that activation of mouse astroglial type-1 cannabinoid receptors associated with mitochondrial membranes (mtCB1) hampers the metabolism of glucose and the production of lactate in the brain, resulting in altered neuronal functions and, in turn, impaired behavioural responses in social interaction assays. Specifically, activation of astroglial mtCB1 receptors reduces the phosphorylation of the mitochondrial complex I subunit NDUFS4, which decreases the stability and activity of complex I. This leads to a reduction in the generation of reactive oxygen species by astrocytes and affects the glycolytic production of lactate through the hypoxia-inducible factor 1 pathway, eventually resulting in neuronal redox stress and impairment of behavioural responses in social interaction assays. Genetic and pharmacological correction of each of these effects abolishes the effect of cannabinoid treatment on the observed behaviour. These findings suggest that mtCB1 receptor signalling can directly regulate astroglial glucose metabolism to fine-tune neuronal activity and behaviour in mice.


Asunto(s)
Astrocitos/metabolismo , Metabolismo Energético , Glucosa/metabolismo , Mitocondrias/metabolismo , Receptor Cannabinoide CB1/metabolismo , Animales , Astrocitos/citología , Astrocitos/efectos de los fármacos , Agonistas de Receptores de Cannabinoides/farmacología , Células Cultivadas , Dronabinol/farmacología , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/metabolismo , Metabolismo Energético/efectos de los fármacos , Glucólisis/efectos de los fármacos , Humanos , Factor 1 Inducible por Hipoxia/metabolismo , Ácido Láctico/metabolismo , Masculino , Ratones , Mitocondrias/efectos de los fármacos , Membranas Mitocondriales/metabolismo , Oxidación-Reducción , Fosforilación , Especies Reactivas de Oxígeno/metabolismo , Receptor Cannabinoide CB1/agonistas , Conducta Social
3.
Neurobiol Dis ; 160: 105533, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34673149

RESUMEN

Memory impairment is one of the disabling manifestations of multiple sclerosis (MS) possibly present from the early stages of the disease and for which there is no specific treatment. Hippocampal synaptic dysfunction and dendritic loss, associated with microglial activation, can underlie memory deficits, yet the molecular mechanisms driving such hippocampal neurodegeneration need to be elucidated. In early-stage experimental autoimmune encephalomyelitis (EAE) female mice, we assessed the expression level of molecules involved in microglia-neuron interactions within the dentate gyrus and found overexpression of genes of the complement pathway. Compared to sham immunized mice, the central element of the complement cascade, C3, showed the strongest and 10-fold upregulation, while there was no increase of downstream factors such as the terminal component C5. The combination of in situ hybridization with immunofluorescence showed that C3 transcripts were essentially produced by activated microglia. Pharmacological inhibition of C3 activity, by daily administration of rosmarinic acid, was sufficient to prevent early dendritic loss, microglia-mediated phagocytosis of synapses in the dentate gyrus, and memory impairment in EAE mice, while morphological markers of microglial activation were still observed. In line, when EAE was induced in C3 deficient mice (C3KO), dendrites and spines of the dentate gyrus as well as memory abilities were preserved. Altogether, these data highlight the central role of microglial C3 in early hippocampal neurodegeneration and memory impairment in EAE and, therefore, pave the way toward new neuroprotective strategies in MS to prevent cognitive deficit using complement inhibitors.


Asunto(s)
Complemento C3/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Hipocampo/metabolismo , Trastornos de la Memoria/metabolismo , Degeneración Nerviosa/metabolismo , Animales , Cinamatos/farmacología , Complemento C3/antagonistas & inhibidores , Complemento C3/genética , Convertasas de Complemento C3-C5/farmacología , Dendritas/efectos de los fármacos , Dendritas/metabolismo , Depsidos/farmacología , Encefalomielitis Autoinmune Experimental/patología , Hipocampo/efectos de los fármacos , Hipocampo/patología , Trastornos de la Memoria/patología , Ratones , Ratones Noqueados , Microglía/efectos de los fármacos , Microglía/metabolismo , Molibdoferredoxina , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Degeneración Nerviosa/patología , Fagocitosis/efectos de los fármacos , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Ácido Rosmarínico
4.
Int J Mol Sci ; 22(14)2021 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-34298875

RESUMEN

Synaptic plasticity is an extensively studied cellular correlate of learning and memory in which NMDARs play a starring role. One of the most interesting features of NMDARs is their ability to act as a co-incident detector. It is unique amongst neurotransmitter receptors in this respect. Co-incident detection is possible because the opening of NMDARs requires membrane depolarisation and the binding of glutamate. Opening of NMDARs also requires a co-agonist. Although the dynamic regulation of glutamate and membrane depolarization have been well studied in coincident detection, the role of the co-agonist site is unexplored. It turns out that non-neuronal glial cells, astrocytes, regulate co-agonist availability, giving them the ability to influence synaptic plasticity. The unique morphology and spatial arrangement of astrocytes at the synaptic level affords them the capacity to sample and integrate information originating from unrelated synapses, regardless of any pre-synaptic and post-synaptic commonality. As astrocytes are classically considered slow responders, their influence at the synapse is widely recognized as modulatory. The aim herein is to reconsider the potential of astrocytes to participate directly in ongoing synaptic NMDAR activity and co-incident detection.


Asunto(s)
Astrocitos/metabolismo , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Ácido Glutámico/metabolismo , Humanos , Plasticidad Neuronal/fisiología , Sinapsis/metabolismo
5.
Neuroimage ; 172: 357-368, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29409838

RESUMEN

The hippocampus contains distinct populations of neurons organized into separate anatomical subfields and layers with differential vulnerability to pathological mechanisms. The ability of in vivo neuroimaging to pinpoint regional vulnerability is especially important for better understanding of hippocampal pathology at the early stage of neurodegenerative disorders and for monitoring future therapeutic strategies. This is the case for instance in multiple sclerosis whose neurodegenerative component can affect the hippocampus from the early stage. We challenged the capacity of two models, i.e. the classical diffusion tensor imaging (DTI) model and the neurite orientation dispersion and density imaging (NODDI) model, to compute quantitative diffusion MRI that could capture microstructural alterations in the individual hippocampal layers of experimental-autoimmune encephalomyelitis (EAE) mice, the animal model of multiple sclerosis. To achieve this, the hippocampal anatomy of a healthy mouse brain was first explored ex vivo with high resolution DTI and NODDI. Then, 18 EAE mice and 18 control mice were explored 20 days after immunization with in vivo diffusion MRI prior to sacrifice for the histological quantification of neurites and glial markers in each hippocampal layer. Fractional anisotropy (FA), axial diffusivity (AD), radial diffusivity (RD) and mean diffusivity (MD) maps were computed from the DTI model while the orientation dispersion index (ODI), the neurite density index (NDI) and the volume fraction of isotropic diffusivity (isoVF) maps were computed from the NODDI model. We first showed in control mice that color-coded FA and ODI maps can delineate three main hippocampal layers. The quantification of FA, AD, RD, MD, ODI, NDI and isoVF presented differences within these 3 layers, especially within the molecular layer of the dentate gyrus which displayed a specific signature based on a combination of AD (or MD), ODI and NDI. Then, the comparison between EAE and control mice showed a decrease of AD (p = 0.036) and of MD (p = 0.033) selectively within the molecular layer of EAE mice while NODDI indices did not present any difference between EAE and control mice in any layer. Histological analyses confirmed the differential vulnerability of the molecular layer of EAE mice that exhibited decreased dendritic length and decreased dendritic complexity together with activated microglia. Dendritic length and intersections within the molecular layer were independent contributors to the observed decrease of AD (R2 = 0.37 and R2 = 0.40, p < 0.0001) and MD (R2 = 0.41 and R2 = 0.42, p < 0.0001). We therefore identified that NODDI maps can help to highlight the internal microanatomy of the hippocampus but NODDI still presents limitations in grey matter as it failed to capture selective dendritic alterations occurring at early stages of a neurodegenerative disease such as multiple sclerosis, whereas DTI maps were significantly altered.


Asunto(s)
Encefalomielitis Autoinmune Experimental/patología , Hipocampo/patología , Neuroimagen/métodos , Animales , Imagen de Difusión Tensora/métodos , Femenino , Ratones , Ratones Endogámicos C57BL
6.
Cereb Cortex ; 27(12): 5635-5651, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-28968740

RESUMEN

Planar cell polarity (PCP) signaling is well known to play a critical role during prenatal brain development; whether it plays specific roles at postnatal stages remains rather unknown. Here, we investigated the role of a key PCP-associated gene scrib in CA1 hippocampal structure and function at postnatal stages. We found that Scrib is required for learning and memory consolidation in the Morris water maze as well as synaptic maturation and NMDAR-dependent bidirectional plasticity. Furthermore, we unveiled a direct molecular interaction between Scrib and PP1/PP2A phosphatases whose levels were decreased in postsynaptic density of conditional knock-out mice. Remarkably, exposure to enriched environment (EE) preserved memory formation in CaMK-Scrib-/- mice by recovering synaptic plasticity and maturation. Thus, Scrib is required for synaptic function involved in memory formation and EE has beneficiary therapeutic effects. Our results demonstrate a distinct new role for a PCP-associated protein, beyond embryonic development, in cognitive functions during adulthood.


Asunto(s)
Disfunción Cognitiva/fisiopatología , Disfunción Cognitiva/terapia , Ambiente , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Plasticidad Neuronal/fisiología , Animales , Células COS , Chlorocebus aethiops , Disfunción Cognitiva/patología , Hipocampo/crecimiento & desarrollo , Hipocampo/metabolismo , Hipocampo/ultraestructura , Vivienda para Animales , Péptidos y Proteínas de Señalización Intracelular/genética , Discapacidades para el Aprendizaje/patología , Discapacidades para el Aprendizaje/fisiopatología , Discapacidades para el Aprendizaje/terapia , Masculino , Trastornos de la Memoria/patología , Trastornos de la Memoria/fisiopatología , Trastornos de la Memoria/terapia , Ratones Noqueados , Modelos Moleculares , Densidad Postsináptica/metabolismo , Densidad Postsináptica/ultraestructura , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis/metabolismo , Sinapsis/ultraestructura
7.
Glia ; 65(3): 502-513, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28063222

RESUMEN

Astrocytes regulate hippocampal synaptic plasticity by the Ca2+ dependent release of the N-methyl d-aspartate receptor (NMDAR) co-agonist d-serine. Previous evidence indicated that d-serine release would be regulated by the intracellular Ca2+ release channel IP3 receptor (IP3 R), however, genetic deletion of IP3 R2, the putative astrocytic IP3 R subtype, had no impact on synaptic plasticity or transmission. Although IP3 R2 is widely believed to be the only functional IP3 R in astrocytes, three IP3 R subtypes (1, 2, and 3) have been identified in vertebrates. Therefore, to better understand gliotransmission, we investigated the functionality of IP3 R and the contribution of the three IP3 R subtypes to Ca2+ signalling. As a proxy for gliotransmission, we found that long-term potentiation (LTP) was impaired by dialyzing astrocytes with the broad IP3 R blocker heparin, and rescued by exogenous d-serine, indicating that astrocytic IP3 Rs regulate d-serine release. To explore which IP3 R subtypes are functional in astrocytes, we used pharmacology and two-photon Ca2+ imaging of hippocampal slices from transgenic mice (IP3 R2-/- and IP3 R2-/- ;3-/- ). This approach revealed that underneath IP3 R2-mediated global Ca2+ events are an overlooked class of IP3 R-mediated local events, occurring in astroglial processes. Notably, multiple IP3 Rs were recruited by high frequency stimulation of the Schaffer collaterals, a classical LTP induction protocol. Together, these findings show the dependence of LTP and gliotransmission on Ca2+ release by astrocytic IP3 Rs. GLIA 2017;65:502-513.


Asunto(s)
Astrocitos/metabolismo , Señalización del Calcio/fisiología , Hipocampo/citología , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Potenciación a Largo Plazo/fisiología , Factores de Edad , Animales , Animales Recién Nacidos , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Estimulación Eléctrica , Técnicas In Vitro , Receptores de Inositol 1,4,5-Trifosfato/genética , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Metoxihidroxifenilglicol/análogos & derivados , Metoxihidroxifenilglicol/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Técnicas de Cultivo de Órganos , Técnicas de Placa-Clamp , Transfección
8.
Brain Behav Immun ; 60: 240-254, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27847283

RESUMEN

Memory impairment is an early and disabling manifestation of multiple sclerosis whose anatomical and biological substrates are still poorly understood. We thus investigated whether memory impairment encountered at the early stage of the disease could be explained by a differential vulnerability of particular hippocampal subfields. By using experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis, we identified that early memory impairment was associated with selective alteration of the dentate gyrus as pinpointed in vivo with diffusion-tensor-imaging (DTI). Neuromorphometric analyses and electrophysiological recordings confirmed dendritic degeneration, alteration in glutamatergic synaptic transmission and impaired long-term synaptic potentiation selectively in the dentate gyrus, but not in CA1, together with a more severe pattern of microglial activation in this subfield. Systemic injections of the microglial inhibitor minocycline prevented DTI, morphological, electrophysiological and behavioral impairments in EAE-mice. Furthermore, daily infusions of minocycline specifically within the dentate gyrus were sufficient to prevent memory impairment in EAE-mice while infusions of minocycline within CA1 were inefficient. We conclude that early memory impairment in EAE is due to a selective disruption of the dentate gyrus associated with microglia activation. These results open new pathophysiological, imaging, and therapeutic perspectives for memory impairment in multiple sclerosis.


Asunto(s)
Encefalomielitis Autoinmune Experimental/metabolismo , Potenciación a Largo Plazo/fisiología , Trastornos de la Memoria/metabolismo , Esclerosis Múltiple/complicaciones , Animales , Giro Dentado/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/fisiopatología , Femenino , Ratones Endogámicos C57BL , Microglía/metabolismo , Sinapsis/fisiología , Transmisión Sináptica/fisiología
9.
Nat Commun ; 15(1): 6842, 2024 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-39122700

RESUMEN

Astrocytes control brain activity via both metabolic processes and gliotransmission, but the physiological links between these functions are scantly known. Here we show that endogenous activation of astrocyte type-1 cannabinoid (CB1) receptors determines a shift of glycolysis towards the lactate-dependent production of D-serine, thereby gating synaptic and cognitive functions in male mice. Mutant mice lacking the CB1 receptor gene in astrocytes (GFAP-CB1-KO) are impaired in novel object recognition (NOR) memory. This phenotype is rescued by the gliotransmitter D-serine, by its precursor L-serine, and also by lactate and 3,5-DHBA, an agonist of the lactate receptor HCAR1. Such lactate-dependent effect is abolished when the astrocyte-specific phosphorylated-pathway (PP), which diverts glycolysis towards L-serine synthesis, is blocked. Consistently, lactate and 3,5-DHBA promoted the co-agonist binding site occupancy of CA1 post-synaptic NMDA receptors in hippocampal slices in a PP-dependent manner. Thus, a tight cross-talk between astrocytic energy metabolism and gliotransmission determines synaptic and cognitive processes.


Asunto(s)
Astrocitos , Cognición , Glucólisis , Ácido Láctico , Ratones Noqueados , Serina , Animales , Masculino , Astrocitos/metabolismo , Cognición/fisiología , Ratones , Ácido Láctico/metabolismo , Serina/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Hipocampo/metabolismo , Sinapsis/metabolismo , Ratones Endogámicos C57BL , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética
10.
Cell Metab ; 34(5): 654-655, 2022 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-35508106

RESUMEN

Recent work from Bonvento and colleagues indicated that synaptic and memory deficits in early Alzheimer's disease (AD) are related to a shortage in L-serine production in astrocytes. Here, the authors, responding to correspondence from Chen and colleagues, discuss how this deficiency does not necessarily require a decrease in PHGDH expression and conclude that the primary event leading to lower serine production is more likely related to altered glycolytic flux in early AD than to PHGDH expression.


Asunto(s)
Enfermedad de Alzheimer , Serina , Enfermedad de Alzheimer/metabolismo , Astrocitos/metabolismo , Glucólisis , Humanos , Fosfoglicerato-Deshidrogenasa/metabolismo , Serina/metabolismo
11.
Front Cell Neurosci ; 15: 695817, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34393726

RESUMEN

Astrocytes are sensitive to ongoing neuronal/network activities and, accordingly, regulate neuronal functions (synaptic transmission, synaptic plasticity, behavior, etc.) by the context-dependent release of several gliotransmitters (e.g., glutamate, glycine, D-serine, ATP). To sense diverse input, astrocytes express a plethora of G-protein coupled receptors, which couple, via Gi/o and Gq, to the intracellular Ca2+ release channel IP3-receptor (IP3R). Indeed, manipulating astrocytic IP3R-Ca2+ signaling is highly consequential at the network and behavioral level: Depleting IP3R subtype 2 (IP3R2) results in reduced GPCR-Ca2+ signaling and impaired synaptic plasticity; enhancing IP3R-Ca2+ signaling affects cognitive functions such as learning and memory, sleep, and mood. However, as a result of discrepancies in the literature, the role of GPCR-IP3R-Ca2+ signaling, especially under physiological conditions, remains inconclusive. One primary reason for this could be that IP3R2 has been used to represent all astrocytic IP3Rs, including IP3R1 and IP3R3. Indeed, IP3R1 and IP3R3 are unique Ca2+ channels in their own right; they have unique biophysical properties, often display distinct distribution, and are differentially regulated. As a result, they mediate different physiological roles to IP3R2. Thus, these additional channels promise to enrich the diversity of spatiotemporal Ca2+ dynamics and provide unique opportunities for integrating neuronal input and modulating astrocyte-neuron communication. The current review weighs evidence supporting the existence of multiple astrocytic-IP3R isoforms, summarizes distinct sub-type specific properties that shape spatiotemporal Ca2+ dynamics. We also discuss existing experimental tools and future refinements to better recapitulate the endogenous activities of each IP3R isoform.

12.
PLoS Biol ; 5(8): e214, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17683201

RESUMEN

The role of adult hippocampal neurogenesis in spatial learning remains a matter of debate. Here, we show that spatial learning modifies neurogenesis by inducing a cascade of events that resembles the selective stabilization process characterizing development. Learning promotes survival of relatively mature neurons, apoptosis of more immature cells, and finally, proliferation of neural precursors. These are three interrelated events mediating learning. Thus, blocking apoptosis impairs memory and inhibits learning-induced cell survival and cell proliferation. In conclusion, during learning, similar to the selective stabilization process, neuronal networks are sculpted by a tightly regulated selection and suppression of different populations of newly born neurons.


Asunto(s)
Apoptosis/fisiología , Proliferación Celular , Hipocampo/citología , Aprendizaje por Laberinto/fisiología , Plasticidad Neuronal/fisiología , Neuronas/fisiología , Conducta Espacial/fisiología , Animales , Supervivencia Celular , Giro Dentado/citología , Giro Dentado/fisiología , Electrofisiología , Hipocampo/metabolismo , Masculino , Memoria/fisiología , Neuronas/citología , Ratas , Ratas Sprague-Dawley
14.
Nat Commun ; 11(1): 1906, 2020 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-32312988

RESUMEN

Astrocytic Ca2+ signals can be fast and local, supporting the idea that astrocytes have the ability to regulate single synapses. However, the anatomical basis of such specific signaling remains unclear, owing to difficulties in resolving the spongiform domain of astrocytes where most tripartite synapses are located. Using 3D-STED microscopy in living organotypic brain slices, we imaged the spongiform domain of astrocytes and observed a reticular meshwork of nodes and shafts that often formed loop-like structures. These anatomical features were also observed in acute hippocampal slices and in barrel cortex in vivo. The majority of dendritic spines were contacted by nodes and their sizes were correlated. FRAP experiments and Ca2+ imaging showed that nodes were biochemical compartments and Ca2+ microdomains. Mapping astrocytic Ca2+ signals onto STED images of nodes and dendritic spines showed they were associated with individual synapses. Here, we report on the nanoscale organization of astrocytes, identifying nodes as a functional astrocytic component of tripartite synapses that may enable synapse-specific communication between neurons and astrocytes.


Asunto(s)
Astrocitos/citología , Astrocitos/metabolismo , Señalización del Calcio/fisiología , Sinapsis/metabolismo , Animales , Encéfalo , Calcio/metabolismo , Hipocampo , Imagenología Tridimensional , Masculino , Ratones , Microscopía , Neuronas/metabolismo
15.
Cell Metab ; 31(3): 503-517.e8, 2020 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-32130882

RESUMEN

Alteration of brain aerobic glycolysis is often observed early in the course of Alzheimer's disease (AD). Whether and how such metabolic dysregulation contributes to both synaptic plasticity and behavioral deficits in AD is not known. Here, we show that the astrocytic l-serine biosynthesis pathway, which branches from glycolysis, is impaired in young AD mice and in AD patients. l-serine is the precursor of d-serine, a co-agonist of synaptic NMDA receptors (NMDARs) required for synaptic plasticity. Accordingly, AD mice display a lower occupancy of the NMDAR co-agonist site as well as synaptic and behavioral deficits. Similar deficits are observed following inactivation of the l-serine synthetic pathway in hippocampal astrocytes, supporting the key role of astrocytic l-serine. Supplementation with l-serine in the diet prevents both synaptic and behavioral deficits in AD mice. Our findings reveal that astrocytic glycolysis controls cognitive functions and suggest oral l-serine as a ready-to-use therapy for AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Astrocitos/metabolismo , Disfunción Cognitiva/metabolismo , Glucólisis , Serina/biosíntesis , Administración Oral , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/fisiopatología , Animales , Astrocitos/efectos de los fármacos , Sitios de Unión , Encéfalo/patología , Encéfalo/fisiopatología , Disfunción Cognitiva/patología , Disfunción Cognitiva/fisiopatología , Metabolismo Energético/efectos de los fármacos , Femenino , Glucosa/metabolismo , Glucólisis/efectos de los fármacos , Humanos , Masculino , Ratones Transgénicos , Persona de Mediana Edad , Plasticidad Neuronal/efectos de los fármacos , Fosfoglicerato-Deshidrogenasa/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Serina/administración & dosificación , Serina/farmacología , Serina/uso terapéutico , Memoria Espacial/efectos de los fármacos
16.
Neuron ; 108(5): 919-936.e11, 2020 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-32976770

RESUMEN

Extrasynaptic actions of glutamate are limited by high-affinity transporters expressed by perisynaptic astroglial processes (PAPs): this helps maintain point-to-point transmission in excitatory circuits. Memory formation in the brain is associated with synaptic remodeling, but how this affects PAPs and therefore extrasynaptic glutamate actions is poorly understood. Here, we used advanced imaging methods, in situ and in vivo, to find that a classical synaptic memory mechanism, long-term potentiation (LTP), triggers withdrawal of PAPs from potentiated synapses. Optical glutamate sensors combined with patch-clamp and 3D molecular localization reveal that LTP induction thus prompts spatial retreat of astroglial glutamate transporters, boosting glutamate spillover and NMDA-receptor-mediated inter-synaptic cross-talk. The LTP-triggered PAP withdrawal involves NKCC1 transporters and the actin-controlling protein cofilin but does not depend on major Ca2+-dependent cascades in astrocytes. We have therefore uncovered a mechanism by which a memory trace at one synapse could alter signal handling by multiple neighboring connections.


Asunto(s)
Astrocitos/metabolismo , Ácido Glutámico/metabolismo , Potenciación a Largo Plazo/fisiología , Sinapsis/metabolismo , Animales , Astrocitos/ultraestructura , Femenino , Imagenología Tridimensional/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Técnicas de Cultivo de Órganos , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Sinapsis/ultraestructura
17.
Nat Commun ; 9(1): 4254, 2018 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-30315174

RESUMEN

Astrocytes are important regulators of excitatory synaptic networks. However, astrocytes regulation of inhibitory synaptic systems remains ill defined. This is particularly relevant since GABAergic interneurons regulate the activity of excitatory cells and shape network function. To address this issue, we combined optogenetics and pharmacological approaches, two-photon confocal imaging and whole-cell recordings to specifically activate hippocampal somatostatin or paravalbumin-expressing interneurons (SOM-INs or PV-INs), while monitoring inhibitory synaptic currents in pyramidal cells and Ca2+ responses in astrocytes. We found that astrocytes detect SOM-IN synaptic activity via GABABR and GAT-3-dependent Ca2+ signaling mechanisms, the latter triggering the release of ATP. In turn, ATP is converted into adenosine, activating A1Rs and upregulating SOM-IN synaptic inhibition of pyramidal cells, but not PV-IN inhibition. Our findings uncover functional interactions between a specific subpopulation of interneurons, astrocytes and pyramidal cells, involved in positive feedback autoregulation of dendritic inhibition of pyramidal cells.


Asunto(s)
Astrocitos/metabolismo , Interneuronas/metabolismo , Células Piramidales/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Calcio/metabolismo , Potenciales Postsinápticos Inhibidores/fisiología , Ratones , Sinapsis/metabolismo , Transmisión Sináptica/fisiología
18.
Cell Rep ; 23(6): 1678-1690, 2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29742425

RESUMEN

The external globus pallidus (GP) is a key GABAergic hub in the basal ganglia (BG) circuitry, a neuronal network involved in motor control. In Parkinson's disease (PD), the rate and pattern of activity of GP neurons are profoundly altered and contribute to the motor symptoms of the disease. In rodent models of PD, the striato-pallidal pathway is hyperactive, and extracellular GABA concentrations are abnormally elevated in the GP, supporting the hypothesis of an alteration of neuronal and/or glial clearance of GABA. Here, we discovered the existence of persistent GABAergic tonic inhibition in GP neurons of dopamine-depleted (DD) rodent models. We showed that glial GAT-3 transporters are downregulated while neuronal GAT-1 function remains normal in DD rodents. Finally, we showed that blocking GAT-3 activity in vivo alters the motor coordination of control rodents, suggesting that GABAergic tonic inhibition in the GP contributes to the pathophysiology of PD.


Asunto(s)
Proteínas Transportadoras de GABA en la Membrana Plasmática/metabolismo , Globo Pálido/patología , Globo Pálido/fisiopatología , Inhibición Neural , Neuronas/patología , Enfermedad de Parkinson/fisiopatología , Animales , Dopamina/deficiencia , Neuronas GABAérgicas/efectos de los fármacos , Neuronas GABAérgicas/metabolismo , Globo Pálido/efectos de los fármacos , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Inhibición Neural/efectos de los fármacos , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ratas Sprague-Dawley , Receptores Dopaminérgicos/metabolismo , Receptores de GABA/metabolismo , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Ácido gamma-Aminobutírico/farmacología
19.
Acta Neuropathol Commun ; 6(1): 104, 2018 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-30322407

RESUMEN

Astrocyte reactivity and neuroinflammation are hallmarks of CNS pathological conditions such as Alzheimer's disease. However, the specific role of reactive astrocytes is still debated. This controversy may stem from the fact that most strategies used to modulate astrocyte reactivity and explore its contribution to disease outcomes have only limited specificity. Moreover, reactive astrocytes are now emerging as heterogeneous cells and all types of astrocyte reactivity may not be controlled efficiently by such strategies.Here, we used cell type-specific approaches in vivo and identified the JAK2-STAT3 pathway, as necessary and sufficient for the induction and maintenance of astrocyte reactivity. Modulation of this cascade by viral gene transfer in mouse astrocytes efficiently controlled several morphological and molecular features of reactivity. Inhibition of this pathway in mouse models of Alzheimer's disease improved three key pathological hallmarks by reducing amyloid deposition, improving spatial learning and restoring synaptic deficits.In conclusion, the JAK2-STAT3 cascade operates as a master regulator of astrocyte reactivity in vivo. Its inhibition offers new therapeutic opportunities for Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Astrocitos/patología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Apolipoproteínas E/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Astrocitos/metabolismo , Modelos Animales de Enfermedad , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Hipocampo/citología , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Transgénicos , Mutación/genética , Presenilina-1/genética , Presenilina-1/metabolismo , Factor de Transcripción STAT1/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas/genética , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo
20.
Neuron ; 98(5): 935-944.e5, 2018 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-29779943

RESUMEN

Bidirectional communication between neurons and astrocytes shapes synaptic plasticity and behavior. D-serine is a necessary co-agonist of synaptic N-methyl-D-aspartate receptors (NMDARs), but the physiological factors regulating its impact on memory processes are scantly known. We show that astroglial CB1 receptors are key determinants of object recognition memory by determining the availability of D-serine at hippocampal synapses. Mutant mice lacking CB1 receptors from astroglial cells (GFAP-CB1-KO) displayed impaired object recognition memory and decreased in vivo and in vitro long-term potentiation (LTP) at CA3-CA1 hippocampal synapses. Activation of CB1 receptors increased intracellular astroglial Ca2+ levels and extracellular levels of D-serine in hippocampal slices. Accordingly, GFAP-CB1-KO displayed lower occupancy of the co-agonist binding site of synaptic hippocampal NMDARs. Finally, elevation of D-serine levels fully rescued LTP and memory impairments of GFAP-CB1-KO mice. These data reveal a novel mechanism of in vivo astroglial control of memory and synaptic plasticity via the D-serine-dependent control of NMDARs.


Asunto(s)
Astrocitos/metabolismo , Neuronas/metabolismo , Receptor Cannabinoide CB1/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Reconocimiento en Psicología/fisiología , Serina/metabolismo , Sinapsis/metabolismo , Animales , Región CA1 Hipocampal/metabolismo , Región CA3 Hipocampal/metabolismo , Hipocampo , Técnicas In Vitro , Potenciación a Largo Plazo , Memoria , Ratones , Ratones Noqueados , Plasticidad Neuronal , Receptor Cannabinoide CB1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA