Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Neurosci ; 26(5): 840-849, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37055628

RESUMO

In any given situation, the environment can be parsed in different ways to yield decision variables (DVs) defining strategies useful for different tasks. It is generally presumed that the brain only computes a single DV defining the current behavioral strategy. Here to test this assumption, we recorded neural ensembles in the frontal cortex of mice performing a foraging task admitting multiple DVs. Methods developed to uncover the currently employed DV revealed the use of multiple strategies and occasional switches in strategy within sessions. Optogenetic manipulations showed that the secondary motor cortex (M2) is needed for mice to use the different DVs in the task. Surprisingly, we found that regardless of which DV best explained the current behavior, M2 activity concurrently encoded a full basis set of computations defining a reservoir of DVs appropriate for alternative tasks. This form of neural multiplexing may confer considerable advantages for learning and adaptive behavior.


Assuntos
Córtex Motor , Camundongos , Animais , Aprendizagem , Adaptação Psicológica
2.
Neurobiol Dis ; 157: 105441, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34224862

RESUMO

Extracellular ATP is a danger signal to the brain and contributes to neurodegeneration in animal models of Alzheimer's disease through its extracellular catabolism by CD73 to generate adenosine, bolstering the activation of adenosine A2A receptors (A2AR). Convulsive activity leads to increased ATP release, with the resulting morphological alterations being eliminated by A2AR blockade. However, it is not known if upon convulsions there is a CD73-mediated coupling between ATP release and A2AR overactivation, causing neurodegeneration. We now show that kainate-induced convulsions trigger a parallel increase of ATP release and of CD73 and A2AR densities in synapses and astrocytes of the mouse hippocampus. Notably, the genetic deletion of CD73 attenuates neuronal degeneration but has no impact on astrocytic modifications in the hippocampus upon kainate-induced convulsions. Furthermore, kainate-induced convulsions cause a parallel deterioration of hippocampal long-term potentiation (LTP) and hippocampal-dependent memory performance, which is eliminated by knocking out CD73. This demonstrates the key role of the ATP release/CD73/A2AR pathway to selectively control synaptic dysfunction and neurodegeneration following an acute brain insult, paving the way to consider CD73 as a new therapeutic target to prevent neuronal damage upon acute brain damage.


Assuntos
5'-Nucleotidase/metabolismo , Trifosfato de Adenosina/metabolismo , Astrócitos/metabolismo , Hipocampo/metabolismo , Neurônios/metabolismo , Receptor A2A de Adenosina/metabolismo , Convulsões/metabolismo , Sinapses/metabolismo , 5'-Nucleotidase/genética , Animais , Astrócitos/efeitos dos fármacos , Agonistas de Aminoácidos Excitatórios/toxicidade , Hipocampo/efeitos dos fármacos , Ácido Caínico/toxicidade , Potenciação de Longa Duração/efeitos dos fármacos , Potenciação de Longa Duração/fisiologia , Memória/efeitos dos fármacos , Memória/fisiologia , Camundongos , Camundongos Knockout , Doenças Neurodegenerativas/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/patologia , Convulsões/induzido quimicamente , Sinapses/efeitos dos fármacos
3.
Elife ; 92020 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-33252331

RESUMO

Survival depends on the ability of animals to select the appropriate behavior in response to threat and safety sensory cues. However, the synaptic and circuit mechanisms by which the brain learns to encode accurate predictors of threat and safety remain largely unexplored. Here, we show that frontal association cortex (FrA) pyramidal neurons of mice integrate auditory cues and basolateral amygdala (BLA) inputs non-linearly in a NMDAR-dependent manner. We found that the response of FrA pyramidal neurons was more pronounced to Gaussian noise than to pure frequency tones, and that the activation of BLA-to-FrA axons was the strongest in between conditioning pairings. Blocking BLA-to-FrA signaling specifically at the time of presentation of Gaussian noise (but not 8 kHz tone) between conditioning trials impaired the formation of auditory fear memories. Taken together, our data reveal a circuit mechanism that facilitates the formation of fear traces in the FrA, thus providing a new framework for probing discriminative learning and related disorders.


Assuntos
Estimulação Acústica/efeitos adversos , Tonsila do Cerebelo/fisiologia , Medo/fisiologia , Lobo Frontal/fisiologia , Aprendizagem/fisiologia , Animais , Cálcio/metabolismo , Condicionamento Clássico/fisiologia , Masculino , Camundongos , Microscopia Confocal , Plasticidade Neuronal/fisiologia , Optogenética , Técnicas de Patch-Clamp
4.
Cell Rep ; 32(9): 108097, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32877679

RESUMO

Cortical plasticity improves behaviors and helps recover lost functions after injury. However, the underlying synaptic mechanisms remain unclear. In mice, we show that trimming all but one whisker enhances sensory responses from the spared whisker in the barrel cortex and occludes whisker-mediated synaptic potentiation (w-Pot) in vivo. In addition, whisker-dependent behaviors that are initially impaired by single-whisker experience (SWE) rapidly recover when associated cortical regions remap. Cross-linking the surface GluA2 subunit of AMPA receptors (AMPARs) suppresses the expression of w-Pot, presumably by blocking AMPAR surface diffusion, in mice with all whiskers intact, indicating that synaptic potentiation in vivo requires AMPAR trafficking. We use this approach to demonstrate that w-Pot is required for SWE-mediated strengthening of synaptic inputs and initiates the recovery of previously learned skills during the early phases of SWE. Taken together, our data reveal that w-Pot mediates cortical remapping and behavioral improvement upon partial sensory deafferentation.


Assuntos
Plasticidade Neuronal/genética , Receptores de AMPA/metabolismo , Animais , Humanos , Camundongos , Privação Sensorial/fisiologia
5.
Front Mol Neurosci ; 12: 238, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31611774

RESUMO

Intelligence is the ability to learn appropriate responses to stimuli and the capacity to master new skills. Synaptic integration at the dendritic level is thought to be essential for this ability through linear and non-linear processing, by allowing neurons to be tuned to relevant information and to maximize adaptive behavior. Showing that dendrites are able to generate local computations that influence how animals perceive the world, form a new memory or learn a new skill was a break-through in neuroscience, since in the past they were seen as passive elements of the neurons, just funneling information to the soma. Here, we provide an overview of the role of dendritic integration in improving the neuronal network and behavioral performance. We focus on how NMDA spikes are generated and their role in neuronal computation for optimal behavioral output based on recent in vivo studies on rodents.

6.
eNeuro ; 5(6)2018.
Artigo em Inglês | MEDLINE | ID: mdl-30627646

RESUMO

Neurodegeneration is a process transversal to neuropsychiatric diseases and the understanding of its mechanisms should allow devising strategies to prevent this irreversible step in brain diseases. Neurodegeneration caused by seizures is a critical step in the aggravation of temporal lobe epilepsy, but its mechanisms remain undetermined. Convulsions trigger an elevation of extracellular adenosine and upregulate adenosine A2A receptors (A2AR), which have been associated with the control of neurodegenerative diseases. Using the rat and mouse kainate model of temporal lobe epilepsy, we now tested whether A2AR control convulsions-induced hippocampal neurodegeneration. The pharmacological or genetic blockade of A2AR did not affect kainate-induced convulsions but dampened the subsequent neurotoxicity. This neurotoxicity began with a rapid A2AR upregulation within glutamatergic synapses (within 2 h), through local translation of synaptic A2AR mRNA. This bolstered A2AR-mediated facilitation of glutamate release and of long-term potentiation (LTP) in CA1 synapses (4 h), triggered a subsequent synaptotoxicity, heralded by decreased synaptic plasticity and loss of synaptic markers coupled to calpain activation (12 h), that predated overt neuronal loss (24 h). All modifications were prevented by the deletion of A2AR selectively in forebrain neurons. This shows that synaptic A2AR critically control synaptic excitotoxicity, which underlies the development of convulsions-induced neurodegeneration.


Assuntos
Convulsivantes/toxicidade , Ácido Caínico/toxicidade , Degeneração Neural/etiologia , Degeneração Neural/metabolismo , Neurônios/metabolismo , Receptor A2A de Adenosina/metabolismo , Antagonistas do Receptor A2 de Adenosina/uso terapêutico , Tonsila do Cerebelo/fisiologia , Animais , Células Cultivadas , Epilepsia/complicações , Epilepsia/tratamento farmacológico , Epilepsia/etiologia , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Excitação Neurológica/efeitos dos fármacos , Excitação Neurológica/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Degeneração Neural/prevenção & controle , Neurônios/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Pirimidinas/uso terapêutico , Ratos , Ratos Wistar , Receptor A2A de Adenosina/genética , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/genética , Triazóis/uso terapêutico
7.
Biol Psychiatry ; 78(11): 763-74, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25869810

RESUMO

BACKGROUND: Adenosine A2A receptors (A2AR) modulate dopamine and glutamate signaling and thereby may influence some of the psychomotor and cognitive processes associated with schizophrenia. Because astroglial A2AR regulate the availability of glutamate, we hypothesized that they might play an unprecedented role in some of the processes leading to the development of schizophrenia, which we investigated using a mouse line with a selective deletion of A2AR in astrocytes (Gfa2-A2AR knockout [KO] mice]. METHODS: We examined Gfa2-A2AR KO mice for behaviors thought to recapitulate some features of schizophrenia, namely enhanced MK-801 psychomotor response (positive symptoms) and decreased working memory (cognitive symptoms). In addition, we probed for neurochemical alterations in the glutamatergic circuitry, evaluating glutamate uptake and release and the levels of key proteins defining glutamatergic signaling (glutamate transporter-I [GLT-I], N-methyl-D-aspartate receptors [NMDA-R] and α-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors [AMPA-R]) to provide a mechanistic understanding of the phenotype encountered. RESULTS: We show that Gfa2-A2AR KO mice exhibited enhanced MK-801 psychomotor response and decreased working memory; this was accompanied by a disruption of glutamate homeostasis characterized by aberrant GLT-I activity, increased presynaptic glutamate release, NMDA-R 2B subunit upregulation, and increased internalization of AMPA-R. Accordingly, selective GLT-I inhibition or blockade of GluR1/2 endocytosis prevented the psychomotor and cognitive phenotypes in Gfa2-A2AR KO mice, namely in the nucleus accumbens. CONCLUSIONS: These results show that the dysfunction of astrocytic A2AR, by controlling GLT-I activity, triggers an astrocyte-to-neuron wave of communication resulting in disrupted glutamate homeostasis, thought to underlie several endophenotypes relevant to schizophrenia.


Assuntos
Astrócitos/metabolismo , Transtornos Cognitivos/patologia , Ácido Glutâmico/metabolismo , Homeostase/genética , Transtornos Psicomotores/patologia , Receptor A2A de Adenosina/deficiência , Animais , Transtornos Cognitivos/genética , Modelos Animais de Doenças , Maleato de Dizocilpina/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Transportador 2 de Aminoácido Excitatório/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Homeostase/efeitos dos fármacos , Ácido Caínico/análogos & derivados , Ácido Caínico/farmacologia , Locomoção/efeitos dos fármacos , Locomoção/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transtornos Psicomotores/genética , Pirimidinas/farmacologia , Receptor A2A de Adenosina/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Sinaptossomos/efeitos dos fármacos , Sinaptossomos/metabolismo , Fatores de Tempo , Triazóis/farmacologia
8.
Biol Psychiatry ; 75(11): 855-63, 2014 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23820821

RESUMO

BACKGROUND: Adenosine A2A receptors (A2ARs) are enriched in the striatum but are also present at lower levels in the extrastriatal forebrain (i.e., hippocampus, cortex), integrating dopamine, glutamate, and brain-derived neurotrophic factor (BDNF) signaling, and are thus essential for striatal neuroplasticity and fear and anxiety behavior. METHODS: We tested two brain region-specific A2AR knockout lines with A2ARs selectively deleted either in the striatum (st-A2AR KO) or the entire forebrain (striatum, hippocampus, and cortex [fb-A2AR KO]) on fear and anxiety-related responses. We also examined the effect of hippocampus-specific A2AR deletion by local injection of adeno-associated virus type 5 (AAV5)-Cre into floxed-A2AR knockout mice. RESULTS: Selectively deleting A2ARs in the striatum increased Pavlovian fear conditioning (both context and tone) in st-A2AR KO mice, but extending the deletion to the rest of the forebrain apparently spared context fear conditioning and attenuated tone fear conditioning in fb-A2AR KO mice. Moreover, focal deletion of hippocampal A2ARs by AAV5-Cre injection selectively attenuated context (but not tone) fear conditioning. Deletion of A2ARs in the entire forebrain in fb-A2AR KO mice also produced an anxiolytic phenotype in both the elevated plus maze and open field tests, and increased the startle response. These extrastriatal forebrain A2AR behavioral effects were associated with reduced BDNF levels in the fb-A2AR KO hippocampus. CONCLUSIONS: This study provides evidence that inactivation of striatal A2ARs facilitates Pavlovian fear conditioning, while inactivation of extrastriatal A2ARs in the forebrain inhibits fear conditioning and also affects anxiety-related behavior.


Assuntos
Corpo Estriado/metabolismo , Medo/fisiologia , Prosencéfalo/metabolismo , Receptor A2A de Adenosina/metabolismo , Animais , Ansiedade/metabolismo , Condicionamento Clássico , Hipocampo/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor A2A de Adenosina/genética , Reflexo de Sobressalto
9.
J Neurosci ; 33(47): 18492-502, 2013 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-24259572

RESUMO

Astrocytic glutamate transporter-1 (GLT-I) is critical to control the bulk of glutamate uptake and, thus, to regulate synaptic plasticity and excitotoxicity. GLT-I glutamate uptake is driven by the sodium gradient implemented by Na(+)/K(+)-ATPases (NKAs) and the α2 subunit of NKA (NKA-α2) is actually linked to GLT-I to regulate astrocytic glutamate transport. We recently found that adenosine A2A receptors (A2ARs), which control synaptic plasticity and neurodegeneration, regulate glutamate uptake through unknown mechanisms. Here we report that A2AR activation decreases NKA activity selectively in astrocytes to inhibit glutamate uptake. Furthermore, we found a physical association of A2ARs with NKA-α2s in astrocytes, as gauged by coimmunoprecipitation and in situ proximity ligation assays, in the cerebral cortex and striatum, two brain regions where A2ARs inhibit the astrocytic glutamate uptake. Moreover, the selective deletion of A2ARs in astrocytes (using Gfa2-A2AR-KO mice) leads to a concurrent increase of both astrocytic glutamate uptake and NKA-α2 levels and activity in the striatum and cortex. This coupling of astrocytic A2ARs to the regulation of glutamate transport through modulation of NKA-α2 activity provides a novel mechanism linking neuronal activity to ion homeostasis controlling glutamatergic activity, all of which are processes intricately associated with the etiology of several brain diseases.


Assuntos
Astrócitos/efeitos dos fármacos , Ácido Glutâmico/metabolismo , Receptor A2A de Adenosina/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Adenosina/análogos & derivados , Adenosina/farmacologia , Agonistas do Receptor A2 de Adenosina/farmacologia , Antagonistas do Receptor A2 de Adenosina/farmacologia , Análise de Variância , Animais , Ácido Aspártico/metabolismo , Astrócitos/ultraestrutura , Proteína Glial Fibrilar Ácida/genética , Imunoprecipitação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ouabaína/farmacologia , Fenetilaminas/farmacologia , Pirimidinas/farmacologia , Receptor A2A de Adenosina/deficiência , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/metabolismo , Triazóis/farmacologia , Trítio/metabolismo
10.
J Neurosci ; 33(28): 11390-9, 2013 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-23843511

RESUMO

Adenosine is a neuromodulator acting through inhibitory A1 receptors (A1Rs) and facilitatory A2ARs, which have similar affinities for adenosine. It has been shown that the activity of intracellular adenosine kinase preferentially controls the activation of A1Rs, but the source of the adenosine activating A2ARs is unknown. We now show that ecto-5'-nucleotidase (CD73), the major enzyme able to convert extracellular AMP into adenosine, colocalizes with A2ARs in the basal ganglia. In addition to astrocytes, striatal CD73 is prominently localized to postsynaptic sites. Notably, CD73 coimmunoprecipitated with A2ARs and proximity ligation assays confirmed the close proximity of CD73 and A2ARs in the striatum. Accordingly, the cAMP formation in synaptosomes as well as the hypolocomotion induced by a novel A2AR prodrug that requires CD73 metabolization to activate A2ARs were observed in wild-type mice, but not in CD73 knock-out (KO) mice or A2AR KO mice. Moreover, CD73 KO mice displayed increased working memory performance and a blunted amphetamine-induced sensitization, mimicking the phenotype of global or forebrain-A2AR KO mice, as well as upon pharmacological A2AR blockade. These results show that CD73-mediated formation of extracellular adenosine is responsible for the activation of striatal A2AR function. This study points to CD73 as a new target that can fine-tune A2AR activity, and a novel therapeutic target to manipulate A2AR-mediated control of striatal function and neurodegeneration.


Assuntos
5'-Nucleotidase/fisiologia , Adenosina/metabolismo , Corpo Estriado/fisiologia , Receptor A2A de Adenosina/fisiologia , Antagonistas do Receptor A2 de Adenosina/farmacologia , Animais , Corpo Estriado/efeitos dos fármacos , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pirimidinas/farmacologia , Triazóis/farmacologia
11.
J Alzheimers Dis ; 31(3): 555-67, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22647260

RESUMO

Alzheimer's disease (AD) is characterized by a progressive cognitive impairment tightly correlated with the accumulation of amyloid-ß (Aß) peptides (mainly Aß(1-42)). There is a precocious disruption of glutamatergic synapses in AD, in line with an ability of Aß to decrease astrocytic glutamate uptake. Accumulating evidence indicates that caffeine prevents the burden of AD, likely through the antagonism of A(2A) receptors (A(2A)R) which attenuates Aß-induced memory impairment and synaptotoxicity. Since A(2A)R also modulate astrocytic glutamate uptake, we now tested if A(2A)R blockade could prevent the decrease of astrocytic glutamate uptake caused by Aß. In cultured astrocytes, Aß(1-42). (1 µM for 24 hours) triggered an astrogliosis typified by an increased density of GFAP, which was mimicked by the A(2A)R agonist, CGS 26180 (30 nM), and prevented by the A(2A)R antagonist, SCH 58261 (100 nM). Aß1-42 also decreased D-aspartate uptake by 28 ± 4%, an effect abrogated upon genetic inactivation or pharmacological blockade of A(2A)R. In accordance with the long term control of glutamate transporter expression by A(2A)R, Aß(1-42). enhanced the expression and density of astrocytic A(2A)R and decreased GLAST and GLT-I expression in astrocytes from wild type, but not from A(2A)R knockout mice. This impact of Aß(1-42). on glutamate transporters and uptake, dependent on A(2A)R function, was also confirmed in an ex vivo astrocyte preparation (gliosomes) from rats intracerebroventricularly (icv) injected with Aß(1-42). . These results provide the first demonstration for a direct key role of astrocytic A(2A)R in the ability of Aß-induced impairment of glutamate uptake, which may underlie glutamatergic synaptic dysfunction and excitotoxicity in AD.


Assuntos
Antagonistas do Receptor A2 de Adenosina/farmacologia , Peptídeos beta-Amiloides/fisiologia , Astrócitos/metabolismo , Ácido Glutâmico/metabolismo , Fragmentos de Peptídeos/fisiologia , Receptor A2A de Adenosina/fisiologia , Animais , Astrócitos/efeitos dos fármacos , Células Cultivadas , Regulação para Baixo/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pirimidinas/farmacologia , Ratos , Ratos Wistar , Triazóis/farmacologia
12.
J Neurochem ; 123(1): 100-12, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22639925

RESUMO

Studies with multiple sclerosis patients and animal models of experimental autoimmune encephalomyelitis (EAE) implicate adenosine and adenosine receptors in modulation of neuroinflammation and brain injury. Although the involvement of the A(1) receptor has been recently demonstrated, the role of the adenosine A(2A) receptor (A(2A)R) in development of EAE pathology is largely unknown. Using mice with genetic inactivation of the A(2A) receptor, we provide direct evidence that loss of the A(2A)R exacerbates EAE pathology in mice. Compared with wild-type mice, A(2A)R knockout mice injected with myelin oligodendroglia glycoprotein peptide had a higher incidence of EAE and exhibited higher neurological deficit scores and greater decrease in body weight. A(2A)R knockout mice displayed increased inflammatory cell infiltration and enhanced microglial cell activation in cortex, brainstem, and spinal cord. In addition, demyelination and axonal damage in brainstem were exacerbated, levels of Th1 cytokines increased, and Th2 cytokines decreased. Collectively, these findings suggest that extracellular adenosine acting at A(2A)Rs triggers an important neuroprotective mechanism. Thus, the A(2A) receptor is a potential target for therapeutic approaches to multiple sclerosis.


Assuntos
Lesões Encefálicas/etiologia , Lesões Encefálicas/patologia , Encefalomielite Autoimune Experimental/complicações , Regulação da Expressão Gênica/genética , Microglia/patologia , Receptor A2A de Adenosina/genética , Antagonistas do Receptor A1 de Adenosina/farmacocinética , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/patologia , Axônios/patologia , Lesões Encefálicas/complicações , Proliferação de Células , Células Cultivadas , Córtex Cerebral/patologia , Citocinas/metabolismo , Doenças Desmielinizantes/etiologia , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/induzido quimicamente , Encefalomielite Autoimune Experimental/imunologia , Ensaio de Imunoadsorção Enzimática , Feminino , Filtração , Citometria de Fluxo , Adjuvante de Freund/toxicidade , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Glicoproteína Mielina-Oligodendrócito/toxicidade , Fragmentos de Peptídeos/toxicidade , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , RNA Mensageiro/metabolismo , Receptor A2A de Adenosina/deficiência , Medula Espinal/patologia , Baço/citologia , Estatísticas não Paramétricas , Trítio/farmacocinética , Xantinas/farmacocinética
13.
Glia ; 60(5): 702-16, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22298379

RESUMO

Glutamate is the primary excitatory neurotransmitter in the central nervous system, where its toxic build-up leads to synaptic dysfunction and excitotoxic cell death that underlies many neurodegenerative diseases. Therefore, efforts have been made to understand the regulation of glutamate transporters, which are responsible for the clearance of extracellular glutamate. We now report that adenosine A(2A) receptors (A(2A) R) control the uptake of D-aspartate in primary cultured astrocytes as well as in an ex vivo preparation enriched in glial plasmalemmal vesicles (gliosomes) from adult rats, whereas A(1) R and A(3) R were devoid of effects. Thus, the acute exposure to the A(2A) R agonist, CGS 21680, inhibited glutamate uptake, an effect prevented by the A(2A) R antagonist, SCH 58261, and abbrogated in cultured astrocytes from A(2A) R knockout mice. Furthermore, the prolonged activation of A(2A) R lead to a cAMP/protein kinase A-dependent reduction of GLT-I and GLAST mRNA and protein levels, which leads to a sustained decrease of glutamate uptake. This dual mechanism of inhibition of glutamate transporters by astrocytic A(2A) R provides a novel candidate mechanism to understand the ability of A(2) (A) R to control synaptic plasticity and neurodegeneration, two conditions tightly associated with the control of extracellular glutamate levels by glutamate transporters.


Assuntos
Astrócitos/metabolismo , Ácido Glutâmico/metabolismo , Neuroglia/metabolismo , Receptor A2A de Adenosina/fisiologia , Animais , Animais Recém-Nascidos , Ácido Aspártico/metabolismo , Células Cultivadas , Masculino , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...