Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Mol Genet Metab ; 134(4): 330-336, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34802899

RESUMO

Niemann-Pick disease, type C1 is a progressive, lethal, neurodegenerative disorder due to endolysosomal storage of unesterified cholesterol. Cerebellar ataxia, as a result of progressive loss of cerebellar Purkinje neurons, is a major symptom of Nieman-Pick disease, type C1. Comparing single cell RNAseq data from control (Npc1+/+) and mutant (Npc1-/-) mice, we observed significantly decreased expression of Slc1a3 in Npc1-/- astrocytes. Slc1a3 encodes a glutamate transporter (GLAST, EAAT1) which functions to decrease glutamate concentrations in the post synaptic space after neuronal firing. Glutamate is an excitatory neurotransmitter and elevated extracellular levels of glutamate can be neurotoxic. Impaired EAAT1 function underlies type-6 episodic ataxia, a rare disorder with progressive cerebellar dysfunction, thus suggesting that impaired glutamate uptake in Niemann-Pick disease, type C1 could contribute to disease progression. We now show that decreased expression of Slc1a3 in Npc1-/- mice has functional consequences that include decreased surface protein expression and decreased glutamate uptake by Npc1-/- astrocytes. To test whether glutamate neurotoxicity plays a role in Niemann-Pick disease, type C1 progression, we treated NPC1 deficient mice with ceftriaxone and riluzole. Ceftriaxone is a ß-lactam antibiotic that is known to upregulate the expression of Slc1a2, an alternative glial glutamate transporter. Although ceftriaxone increased Slc1a2 expression, we did not observe a treatment effect in NPC1 mutant mice. Riluzole is a glutamate receptor antagonist that inhibits postsynaptic glutamate receptor signaling and reduces the release of glutamate. We found that treatment with riluzole increased median survival in Npc1-/- by 12%. Given that riluzole is an approved drug for the treatment of amyotrophic lateral sclerosis, repurposing of this drug may provide a novel therapeutic approach to decrease disease progression in Niemann-Pick disease type, C1 patients.


Assuntos
Ceftriaxona/uso terapêutico , Ácido Glutâmico/toxicidade , Doença de Niemann-Pick Tipo C/tratamento farmacológico , Riluzol/uso terapêutico , Animais , Astrócitos/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Transportador 1 de Aminoácido Excitatório/fisiologia , Feminino , Ácido Glutâmico/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Proteína C1 de Niemann-Pick/fisiologia
2.
Eur Neuropsychopharmacol ; 29(11): 1288-1294, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31582286

RESUMO

Alterations of energy metabolism and of astrocyte number/function in ventral anterior cingulate cortex (vACC) have been reported in major depressive disorder (MDD) patients and may contribute to MDD pathophysiology. We recently developed a mouse model of MDD mimicking these alterations. We knocked down the astroglial glutamate transporters GLAST and GLT-1 in infralimbic cortex (IL, rodent equivalent of vACC) using small interfering RNA (siRNA). GLAST and GLT-1 siRNA microinfusion in IL evoked a depressive-like phenotype, associated with a reduced serotonergic function and reduced forebrain BDNF expression. Neither effect occurred after siRNA application in the adjacent prelimbic cortex (PrL), thus emphasizing the critical role of vACC/IL in MDD pathogenesis. Here we examined the cellular/network basis of the changes induced in IL using intracellular recordings of layer V pyramidal neurons from mice microinjected with siRNA 24 h before. We analyzed (i) the electrophysiological characteristics of neurons; (ii) the synaptic transmission properties, by monitoring miniature, spontaneous and evoked EPSCs, and (iii) the gliotransmission, by monitoring slow inward currents (SICs), mediated by astrocytic glutamate release and activation of extra-synaptic NMDA receptors. GLT-1 and GLAST knockdown led to a more depolarized membrane potential and increased action potential firing rate of layer V pyramidal neurons, and enhanced excitatory synaptic transmission, as shown by the enhanced amplitude/frequency of spontaneous EPSCs. Gliotransmission was also increased, as indicated by the enhanced SIC amplitude/frequency. Hence, the depressive-like phenotype is associated with IL hyperactivity, likely leading to an excessive top-down inhibitory control of serotonergic activity through IL-midbrain descending pathways.


Assuntos
Astrócitos/metabolismo , Transportador 1 de Aminoácido Excitatório/fisiologia , Transportador 2 de Aminoácido Excitatório/fisiologia , Giro do Cíngulo/metabolismo , Transmissão Sináptica/fisiologia , Animais , Fator Neurotrófico Derivado do Encéfalo/biossíntese , Transtorno Depressivo Maior/metabolismo , Transtorno Depressivo Maior/fisiopatologia , Transportador 1 de Aminoácido Excitatório/metabolismo , Transportador 2 de Aminoácido Excitatório/metabolismo , Potenciais Pós-Sinápticos Excitadores/fisiologia , Técnicas de Silenciamento de Genes , Giro do Cíngulo/efeitos dos fármacos , Lobo Límbico/efeitos dos fármacos , Camundongos , Microinjeções , Prosencéfalo/metabolismo , Células Piramidais/fisiologia , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/farmacologia , Transmissão Sináptica/efeitos dos fármacos
3.
CNS Neurosci Ther ; 25(4): 509-518, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30311425

RESUMO

AIM: Deficient glutamate reuptake occurs in the cerebral cortex of Huntington's disease (HD) patients and murine models. Here, we examine the effects of partial or complete blockade of glutamate transporters on excitatory postsynaptic currents (EPSCs) of cortical pyramidal neurons (CPNs). METHODS: Whole-cell patch clamp recordings of CPNs in slices from symptomatic R6/2 mice and wild-type (WT) littermates were used to examine the effects of selective or concurrent inhibition of glutamate reuptake transporters. RESULTS: Selective inhibition of the glial glutamate transporter 1 (GLT-1) or the glutamate aspartate transporter (GLAST) produced slight decreases in decay time of evoked EPSCs in CPNs from WT and R6/2 mice with no significant differences between genotypes. In contrast, concurrent inhibition of both transporters with DL-TBOA induced a significant increase in area and decay time and this effect was significantly greater in R6/2 CPNs. Furthermore, full blockade also reduced spontaneous EPSC frequency and exacerbated epileptiform activity in CPNs from symptomatic R6/2 mice. CONCLUSIONS: R6/2 CPNs are more sensitive to glutamate accumulation during full inhibition of both glutamate transporters, and these neurons have homeostatic mechanisms to cope with inhibition of GLT-1 or GLAST by a mechanism that involves upregulation of either transporter when the other is deficient.


Assuntos
Córtex Cerebral/fisiopatologia , Modelos Animais de Doenças , Transportador 1 de Aminoácido Excitatório/antagonistas & inibidores , Transportador 2 de Aminoácido Excitatório/antagonistas & inibidores , Doença de Huntington/fisiopatologia , Animais , Ácido Aspártico/farmacologia , Benzopiranos/farmacologia , Transportador 1 de Aminoácido Excitatório/fisiologia , Transportador 2 de Aminoácido Excitatório/fisiologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Doença de Huntington/genética , Masculino , Camundongos , Camundongos Transgênicos , Técnicas de Cultura de Órgãos
4.
Proc Natl Acad Sci U S A ; 114(28): 7438-7443, 2017 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-28655840

RESUMO

Astrocytes regulate synaptic transmission through controlling neurotransmitter concentrations around synapses. Little is known, however, about their roles in neural circuit development. Here we report that Bergmann glia (BG), specialized cerebellar astrocytes that thoroughly enwrap Purkinje cells (PCs), are essential for synaptic organization in PCs through the action of the l-glutamate/l-aspartate transporter (GLAST). In GLAST-knockout mice, dendritic innervation by the main ascending climbing fiber (CF) branch was significantly weakened, whereas the transverse branch, which is thin and nonsynaptogenic in control mice, was transformed into thick and synaptogenic branches. Both types of CF branches frequently produced aberrant wiring to proximal and distal dendrites, causing multiple CF-PC innervation. Our electrophysiological analysis revealed that slow and small CF-evoked excitatory postsynaptic currents (EPSCs) were recorded from almost all PCs in GLAST-knockout mice. These atypical CF-EPSCs were far more numerous and had significantly faster 10-90% rise time than those elicited by glutamate spillover under pharmacological blockade of glial glutamate transporters. Innervation by parallel fibers (PFs) was also affected. PF synapses were robustly increased in the entire dendritic trees, leading to impaired segregation of CF and PF territories. Furthermore, lamellate BG processes were retracted from PC dendrites and synapses, leading to the exposure of these neuronal elements to the extracellular milieus. These synaptic and glial phenotypes were reproduced in wild-type mice after functional blockade of glial glutamate transporters. These findings highlight that glutamate transporter function by GLAST on BG plays important roles in development and maintenance of proper synaptic wiring and wrapping in PCs.


Assuntos
Transportador 1 de Aminoácido Excitatório/genética , Transportador 1 de Aminoácido Excitatório/fisiologia , Neuroglia/fisiologia , Células de Purkinje/fisiologia , Sinapses/fisiologia , Sistema X-AG de Transporte de Aminoácidos/genética , Sistema X-AG de Transporte de Aminoácidos/fisiologia , Animais , Astrócitos/fisiologia , Cerebelo/fisiologia , Dendritos/fisiologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Genótipo , Ácido Glutâmico , Proteínas de Fluorescência Verde/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/fisiologia , Fenótipo , Transmissão Sináptica/fisiologia
5.
Nat Neurosci ; 20(3): 393-395, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28135241

RESUMO

Contributions of glial cells to neuroenergetics have been the focus of extensive debate. Here we provide positron emission tomography evidence that activation of astrocytic glutamate transport via the excitatory amino acid transporter GLT-1 triggers widespread but graded glucose uptake in the rodent brain. Our results highlight the need for a reevaluation of the interpretation of [18F]FDG positron emission tomography data, whereby astrocytes would be recognized as contributing to the [18F]FDG signal.


Assuntos
Astrócitos/metabolismo , Transportador 1 de Aminoácido Excitatório/fisiologia , Fluordesoxiglucose F18/metabolismo , Ácido Glutâmico/metabolismo , Animais , Transporte Biológico , Encéfalo/irrigação sanguínea , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Ceftriaxona/farmacologia , Células Cultivadas , Transportador 1 de Aminoácido Excitatório/agonistas , Neuroimagem Funcional , Locomoção/efeitos dos fármacos , Masculino , Tomografia por Emissão de Pósitrons , Ratos , Vibrissas/fisiologia
6.
J Neurosci ; 36(50): 12624-12639, 2016 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-27821575

RESUMO

Continuous cerebral blood flow is essential for neuronal survival, but whether vascular tone influences resting neuronal function is not known. Using a multidisciplinary approach in both rat and mice brain slices, we determined whether flow/pressure-evoked increases or decreases in parenchymal arteriole vascular tone, which result in arteriole constriction and dilation, respectively, altered resting cortical pyramidal neuron activity. We present evidence for intercellular communication in the brain involving a flow of information from vessel to astrocyte to neuron, a direction opposite to that of classic neurovascular coupling and referred to here as vasculo-neuronal coupling (VNC). Flow/pressure increases within parenchymal arterioles increased vascular tone and simultaneously decreased resting pyramidal neuron firing activity. On the other hand, flow/pressure decreases evoke parenchymal arteriole dilation and increased resting pyramidal neuron firing activity. In GLAST-CreERT2; R26-lsl-GCaMP3 mice, we demonstrate that increased parenchymal arteriole tone significantly increased intracellular calcium in perivascular astrocyte processes, the onset of astrocyte calcium changes preceded the inhibition of cortical pyramidal neuronal firing activity. During increases in parenchymal arteriole tone, the pyramidal neuron response was unaffected by blockers of nitric oxide, GABAA, glutamate, or ecto-ATPase. However, VNC was abrogated by TRPV4 channel, GABAB, as well as an adenosine A1 receptor blocker. Differently to pyramidal neuron responses, increases in flow/pressure within parenchymal arterioles increased the firing activity of a subtype of interneuron. Together, these data suggest that VNC is a complex constitutive active process that enables neurons to efficiently adjust their resting activity according to brain perfusion levels, thus safeguarding cellular homeostasis by preventing mismatches between energy supply and demand. SIGNIFICANCE STATEMENT: We present evidence for vessel-to-neuron communication in the brain slice defined here as vasculo-neuronal coupling. We showed that, in response to increases in parenchymal arteriole tone, astrocyte intracellular Ca2+ increased and cortical neuronal activity decreased. On the other hand, decreasing parenchymal arteriole tone increased resting cortical pyramidal neuron activity. Vasculo-neuronal coupling was partly mediated by TRPV4 channels as genetic ablation, or pharmacological blockade impaired increased flow/pressure-evoked neuronal inhibition. Increased flow/pressure-evoked neuronal inhibition was blocked in the presence of adenosine A1 receptor and GABAB receptor blockade. Results provide evidence for the concept of vasculo-neuronal coupling and highlight the importance of understanding the interplay between basal CBF and resting neuronal activity.


Assuntos
Vasos Sanguíneos/inervação , Encéfalo/fisiologia , Comunicação Celular/fisiologia , Neurônios/fisiologia , Animais , Arteríolas/inervação , Arteríolas/fisiologia , Astrócitos/fisiologia , Vasos Sanguíneos/efeitos dos fármacos , Encéfalo/citologia , Cálcio/metabolismo , Comunicação Celular/efeitos dos fármacos , Circulação Cerebrovascular/efeitos dos fármacos , Circulação Cerebrovascular/fisiologia , Transportador 1 de Aminoácido Excitatório/genética , Transportador 1 de Aminoácido Excitatório/fisiologia , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/fisiologia , Neurônios/efeitos dos fármacos , Células Piramidais/fisiologia , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/fisiologia
7.
Hum Mol Genet ; 25(20): 4448-4461, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-28173092

RESUMO

Clinical phenotypes of spinocerebellar ataxia type-5 (SCA5) and spectrin-associated autosomal recessive cerebellar ataxia type-1 (SPARCA1) are mirrored in mice lacking ß-III spectrin (ß-III-/-). One function of ß-III spectrin is the stabilization of the Purkinje cell-specific glutamate transporter EAAT4 at the plasma membrane. In ß-III-/- mice EAAT4 levels are reduced from an early age. In contrast levels of the predominant cerebellar glutamate transporter GLAST, expressed in Bergmann glia, only fall progressively from 3 months onwards. Here we elucidated the roles of these two glutamate transporters in cerebellar pathogenesis mediated through loss of ß-III spectrin function by studying EAAT4 and GLAST knockout mice as well as crosses of both with ß-III-/- mice. Our data demonstrate that EAAT4 loss, but not abnormal AMPA receptor composition, in young ß-III-/- mice underlies early Purkinje cell hyper-excitability and that subsequent loss of GLAST, superimposed on the earlier deficiency of EAAT4, is responsible for Purkinje cell loss and progression of motor deficits. Yet the loss of GLAST appears to be independent of EAAT4 loss, highlighting that other aspects of Purkinje cell dysfunction underpin the pathogenic loss of GLAST. Finally, our results demonstrate that Purkinje cells in the posterior cerebellum of ß-III-/- mice are most susceptible to the combined loss of EAAT4 and GLAST, with degeneration of proximal dendrites, the site of climbing fibre innervation, most pronounced. This highlights the necessity for efficient glutamate clearance from these regions and identifies dysregulation of glutamatergic neurotransmission particularly within the posterior cerebellum as a key mechanism in SCA5 and SPARCA1 pathogenesis.


Assuntos
Ataxia Cerebelar/metabolismo , Modelos Animais de Doenças , Transportador 1 de Aminoácido Excitatório/metabolismo , Transportador 4 de Aminoácido Excitatório/metabolismo , Células de Purkinje/metabolismo , Espectrina/metabolismo , Ataxias Espinocerebelares/metabolismo , Animais , Ataxia Cerebelar/genética , Ataxia Cerebelar/patologia , Transportador 1 de Aminoácido Excitatório/fisiologia , Transportador 4 de Aminoácido Excitatório/fisiologia , Feminino , Masculino , Camundongos , Camundongos Knockout , Fenótipo , Células de Purkinje/patologia , Espectrina/fisiologia , Ataxias Espinocerebelares/genética , Ataxias Espinocerebelares/patologia
8.
Mol Pharmacol ; 86(6): 657-64, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25267718

RESUMO

Excitatory amino acid transporter 1 plays an important role in keeping the synaptic glutamate concentration below neurotoxic levels by translocating this neurotransmitter into the cell. Both reentrant hairpin loops, HP1 and -2, have been shown to take part in binding the substrate and the more deeply buried sodium ion, and might therefore be a part of the intra- or extracellular gate of the transporter. However, the shape of the motion of either loop relative to transmembrane domain (TM) 4 during the transport cycle has not yet been fully resolved. Using copper(II) (1,10-phenanthroline)3 (CuPh) for cross-linking cysteine pairs, we found strong inhibition of transport when A243C (TM4) was combined with S366C (HP1), I453C (HP2), or T456C (HP2). These findings were reinforced by the impact of cadmium on transport activity, and both approaches consistently showed that proximity was exclusively intramonomeric. Under conditions that promote the inward-facing state, inhibition by CuPh in A243C/S366C was reduced, while the opposite was seen when the outward-facing one was stabilized, suggesting that the two positions are farther apart in the former conformation than in the latter. Surprisingly, maximal cross-linking of A243C with I453C or T456C was not observed under conditions that promote the inward-facing state. Altogether, our data suggest that the transporter may undergo complex relative movement between these positions on TM4 and HP1/HP2 during the transport cycle.


Assuntos
Transportador 1 de Aminoácido Excitatório/química , Ácido Aspártico/farmacologia , Cádmio/farmacologia , Reagentes de Ligações Cruzadas/farmacologia , Transportador 1 de Aminoácido Excitatório/fisiologia , Ácido Glutâmico/farmacologia , Células HeLa , Humanos , Estrutura Terciária de Proteína
9.
Vision Res ; 103: 49-62, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25152321

RESUMO

To maintain reliable signal transmission across a synapse, free synaptic neurotransmitters must be removed from the cleft in a timely manner. In the first visual synapse, this critical task is mainly undertaken by glutamate transporters (EAATs). Here we study the differential roles of the EAAT1, EAAT2 and EAAT5 subtypes in glutamate (GLU) uptake at the photoreceptor-to-depolarizing bipolar cell synapse in intact dark-adapted retina. Various doses of EAAT blockers and/or GLU were injected into the eye before the electroretinogram (ERG) was measured. Their effectiveness and potency in inhibiting the ERG b-wave were studied to determine their relative contributions to the GLU clearing activity at the synapse. The results showed that EAAT1 and EAAT2 plays different roles. Selectively blocking glial EAAT1 alone using UCPH101 inhibited the b-wave 2-24h following injection, suggesting a dominating role of EAAT1 in the overall GLU clearing capacity in the synaptic cleft. Selectively blocking EAAT2 on photoreceptor terminals had no significant effect on the b-wave, but increased the potency of exogenous GLU in inhibiting the b-wave. These suggest that EAAT2 play a secondary yet significant role in the GLU reuptake activity at the rod and the cone output synapses. Additionally, we have verified our electrophysiological findings with double-label immunohistochemistry, and extend the literature on the spatial distribution of EAAT2 splice variants in the mouse retina.


Assuntos
Transportador 1 de Aminoácido Excitatório/fisiologia , Transportador 2 de Aminoácido Excitatório/fisiologia , Transportador 5 de Aminoácido Excitatório/fisiologia , Glutamatos/metabolismo , Retina/fisiologia , Transmissão Sináptica/efeitos dos fármacos , Animais , Transporte Biológico , Adaptação à Escuridão/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Eletrorretinografia/efeitos dos fármacos , Transportador 1 de Aminoácido Excitatório/antagonistas & inibidores , Transportador 2 de Aminoácido Excitatório/antagonistas & inibidores , Transportador 2 de Aminoácido Excitatório/metabolismo , Transportador 5 de Aminoácido Excitatório/metabolismo , Glutamatos/farmacologia , Imuno-Histoquímica , Injeções Intravítreas , Ácido Caínico/análogos & derivados , Ácido Caínico/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células Fotorreceptoras/efeitos dos fármacos , Células Fotorreceptoras/metabolismo , Retina/efeitos dos fármacos , Células Bipolares da Retina/efeitos dos fármacos , Células Bipolares da Retina/metabolismo
10.
Biol Pharm Bull ; 36(12): 1996-2004, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24292058

RESUMO

The astrocytic L-glutamate (L-Glu) transporter EAAT1 participates in the removal of L-Glu from the synaptic cleft and maintenance of non-toxic concentrations in the extracellular fluid. We have shown that niflumic acid (NFA), a non-steroidal anti-inflammatory drug (NSAIDs), alters L-Glu-induced EAAT1 currents in a voltage-dependent manner using the two-electrode voltage clamp technique in Xenopus oocytes expressing EAAT1. In this study, we characterised the effects of NFA on each type of ion-flux through EAAT1. NFA modulated currents induced by both L-Glu and L-aspartate (L-Asp) in a voltage-dependent manner. Ion-substitution experiments revealed that the activation of additional H(+) conductance was involved in the modulation of currents induced by L-Asp and L-Glu, but Cl(-) was involved only with the L-Asp currents. NFA activated additional currents of EAAT1 in a substrate-dependent manner.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Transportador 1 de Aminoácido Excitatório/fisiologia , Ácido Niflúmico/farmacologia , Animais , Ácido Aspártico/farmacologia , Ácido Glutâmico/farmacologia , Humanos , Técnicas In Vitro , Oócitos , Sódio/farmacologia , Xenopus laevis
11.
Mol Pharmacol ; 83(1): 22-32, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23012257

RESUMO

4-(2-Butyl-6,7-dichloro-2-cyclopentyl-indan-1-on-5-yl) oxobutyric acid (DCPIB) was identified as the selective blocker of volume-regulated anion channels (VRAC). VRAC are permeable to small inorganic and organic anions, including the excitatory neurotransmitter glutamate. In recent years DCPIB has been increasingly used for probing the physiologic and pathologic roles of VRAC and was found to potently suppress pathologic glutamate release in cerebral ischemia. Because ischemic glutamate release can be mediated by a plethora of mechanisms, in this study we explored the selectivity of DCPIB toward the majority of previously identified glutamate transporters and permeability pathways. l-[(3)H]glutamate, d-[(3)H]aspartate, and l-[(14)C]cystine were used to trace amino acid release and uptake. We found that in addition to its well-characterized effect on VRAC, DCPIB potently inhibited glutamate release via connexin hemichannels and glutamate uptake via the glutamate transporter GLT-1 in rat glial cells. In contrast, DCPIB had no direct effect on vesicular glutamate release from rat brain synaptosomes or the cystine/glutamate exchange in astrocytes. The compound did not affect the astrocytic glutamate transporter GLAST, nor did it block glutamate release via the P2X(7)/pannexin permeability pathway. The ability of DCPIB to directly block connexin hemichannels was confirmed using a gene-specific siRNA knockdown approach. Overall, our data demonstrate that DCPIB influences several glutamate transport pathways and that its effects on VRAC in vivo should be verified using additional pharmacological controls.


Assuntos
Sistemas de Transporte de Aminoácidos/fisiologia , Astrócitos/efeitos dos fármacos , Ciclopentanos/farmacologia , Ácido Glutâmico/metabolismo , Indanos/farmacologia , Microglia/efeitos dos fármacos , Trifosfato de Adenosina/farmacologia , Sistema y+ de Transporte de Aminoácidos/antagonistas & inibidores , Sistema y+ de Transporte de Aminoácidos/fisiologia , Sistemas de Transporte de Aminoácidos/antagonistas & inibidores , Sistemas de Transporte de Aminoácidos Acídicos , Animais , Astrócitos/metabolismo , Transporte Biológico , Células Cultivadas , Córtex Cerebral/citologia , Conexinas/antagonistas & inibidores , Conexinas/fisiologia , Transportador 1 de Aminoácido Excitatório/antagonistas & inibidores , Transportador 1 de Aminoácido Excitatório/fisiologia , Transportador 2 de Aminoácido Excitatório/antagonistas & inibidores , Transportador 2 de Aminoácido Excitatório/fisiologia , Microglia/metabolismo , Permeabilidade , Cultura Primária de Células , Ratos , Receptores Purinérgicos P2X7/fisiologia , Sinaptossomos/metabolismo
12.
Brain ; 135(Pt 11): 3416-25, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23107647

RESUMO

Episodic ataxia is a human genetic disease characterized by paroxysmal cerebellar incoordination. There are several genetically and clinically distinct forms of this disease, and one of them, episodic ataxia type 6, is caused by mutations in the gene encoding a glial glutamate transporter, the excitatory amino acid transporter-1. So far, reduced glutamate uptake by mutant excitatory amino acid transporter-1 has been thought to be the main pathophysiological process in episodic ataxia type 6. However, excitatory amino acid transporter-1 does not only mediate secondary-active glutamate transport, but also functions as an ion channel. Here, we examined the effects of a disease-associated point mutation, P290R, on glutamate transport, anion current as well as on the subcellular distribution of excitatory amino acid transporter-1 using heterologous expression in mammalian cells. P290R reduces the number of excitatory amino acid transporter-1 in the surface membrane and impairs excitatory amino acid transporter-1-mediated glutamate uptake. Cells expressing P290R excitatory amino acid transporter-1 exhibit larger anion currents than wild-type cells in the absence as well as in the presence of external l-glutamate, despite a lower number of mutant transporters in the surface membrane. Noise analysis revealed unaltered unitary current amplitudes, indicating that P290R modifies opening and closing, and not anion permeation through mutant excitatory amino acid transporter-1 anion channels. These findings identify gain-of-function of excitatory amino acid transporter anion conduction as a pathological process in episodic ataxia. Episodic ataxia type 6 represents the first human disease found to be associated with altered function of excitatory amino acid transporter anion channels and illustrates possible physiological and pathophysiological impacts of this functional mode of this class of glutamate transporters.


Assuntos
Ataxia Cerebelar/genética , Ataxia Cerebelar/fisiopatologia , Transportador 1 de Aminoácido Excitatório/fisiologia , Potenciais da Membrana/fisiologia , Mutação Puntual/genética , Mutação Puntual/fisiologia , Canais de Ânion Dependentes de Voltagem/fisiologia , Linhagem Celular , Membrana Celular/metabolismo , Membrana Celular/fisiologia , Transportador 1 de Aminoácido Excitatório/genética , Transportador 1 de Aminoácido Excitatório/metabolismo , Ácido Glutâmico/metabolismo , Humanos , Potenciais da Membrana/genética , Canais de Ânion Dependentes de Voltagem/genética
13.
Doc Ophthalmol ; 125(3): 249-65, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23054160

RESUMO

PURPOSE: To study the physiological and pathological roles of excitatory amino acid transporters in the distal retina of albino rabbits. METHODS: Albino rabbits were injected intravitreally in one eye with different doses of L- or D-isomers of glutamate or aspartate, with mixtures of L-glutamate and antagonists to glutamate receptors or with inhibitors of glutamate transporters. The other eye was injected with saline, and served as a control. The electroretinogram (ERG) was recorded 4 h and 2 weeks after injection. At the end of the ERG follow-up period, retinas were prepared for light microscopy. RESULTS: The ERG b-wave was reduced and the a-wave augmented by both isomers of EAAs when tested 4 h after injection. Long-term (2-week) follow-up indicated severe damage to the retina by both isomers of EAAs. Antagonists to glutamate-gated ionic channels failed to protect the rabbit distal retina from permanent damage. Competitive inhibitors of GLAST-1 transporter were highly effective in blocking synaptic transmission in the OPL and in inducing permanent ERG deficit. Selective inhibition of the GLT-1 transporter caused short-term augmentation of the ERG and no permanent ERG deficit. CONCLUSION: GLAST-1, the glutamate transporter of Müller cells, plays a major role in synaptic transmission within the OPL of the rabbit retina. Over-activation of GLAST-1 seems to induce permanent damage to the distal rabbit retina via yet unidentified mechanism.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/farmacologia , Ácido Aspártico/farmacologia , Eletrorretinografia/efeitos dos fármacos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Ácido Glutâmico/farmacologia , Retina/fisiologia , Animais , Transportador 1 de Aminoácido Excitatório/fisiologia , Injeções Intravítreas , Coelhos , Transmissão Sináptica/fisiologia
14.
J Physiol ; 590(10): 2317-31, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22411007

RESUMO

Stimulation of astrocytes by neuronal activity and the subsequent release of neuromodulators is thought to be an important regulator of synaptic communication. In this study we show that astrocytes juxtaposed to the glutamatergic calyx of Held synapse in the rat medial nucleus of the trapezoid body (MNTB) are stimulated by the activation of glutamate transporters and consequently release glutamine on a very rapid timescale. MNTB principal neurones express electrogenic system A glutamine transporters, and were exploited as glutamine sensors in this study. By simultaneous whole-cell voltage clamping astrocytes and neighbouring MNTB neurones in brainstem slices, we show that application of the excitatory amino acid transporter (EAAT) substrate d-aspartate stimulates astrocytes to rapidly release glutamine, which is detected by nearby MNTB neurones. This release is significantly reduced by the toxins L-methionine sulfoximine and fluoroacetate, which reduce glutamine concentrations specifically in glial cells. Similarly, glutamine release was also inhibited by localised inactivation of EAATs in individual astrocytes, using internal DL-threo-ß-benzyloxyaspartic acid (TBOA) or dissipating the driving force by modifying the patch-pipette solution. These results demonstrate that astrocytes adjacent to glutamatergic synapses can release glutamine in a temporally precise, controlled manner in response to glial glutamate transporter activation. Since glutamine can be used by neurones as a precursor for glutamate and GABA synthesis, this represents a potential feedback mechanism by which astrocytes can respond to synaptic activation and react in a way that sustains or enhances further communication. This would therefore represent an additional manifestation of the tripartite relationship between synapses and astrocytes.


Assuntos
Astrócitos/fisiologia , Ácido Glutâmico/fisiologia , Glutamina/fisiologia , Transmissão Sináptica/fisiologia , Animais , Tronco Encefálico/fisiologia , Transportador 1 de Aminoácido Excitatório/fisiologia , Transportador 2 de Aminoácido Excitatório/fisiologia , Retroalimentação Fisiológica , Neurônios/fisiologia , Ratos , Ratos Wistar
15.
J Assoc Res Otolaryngol ; 13(3): 323-33, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22350511

RESUMO

Various studies point to a crucial role of the high-affinity sodium-coupled glutamate aspartate transporter GLAST-1 for modulation of excitatory transmission as shown in the retina and the CNS. While 2-4-month-old GLAST-1 null mice did not show any functional vestibular abnormality, we observed profound circling behavior in older (7 months) animals lacking GLAST-1. An unchanged total number of otoferlin-positive vestibular hair cells (VHCs), similar ribbon numbers in VHCs, and an unchanged VGLUT3 expression in type II VHCs were detected in GLAST-1 null compared to wild-type mice. A partial loss of supporting cells and an apparent decline of a voltage-gated channel potassium subunit (KCNQ4) was observed in postsynaptic calyceal afferents contacting type I VHCs, together with a reduction of neurofilament- (NF200-) and vesicular glutamate transporter 1- (VGLUT1-) positive calyces in GLAST-1 null mice. Taken together, GLAST-1 deletion appeared to preferentially affect the maintenance of a normal postsynaptic/neuronal phenotype, evident only with increasing age.


Assuntos
Transportador 1 de Aminoácido Excitatório/fisiologia , Vestíbulo do Labirinto/fisiologia , Animais , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Células Receptoras Sensoriais/fisiologia , Vestíbulo do Labirinto/anatomia & histologia
16.
J Neurochem ; 121(4): 526-36, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22339645

RESUMO

Decreased GABAergic synaptic strength ('disinhibition') in the spinal dorsal horn is a crucial mechanism contributing to the development and maintenance of pathological pain. However, mechanisms leading to disinhibition in the spinal dorsal horn remain elusive. We investigated the role of glial glutamate transporters (GLT-1 and GLAST) and glutamine synthetase in maintaining GABAergic synaptic activity in the spinal dorsal horn. Electrically evoked GABAergic inhibitory post-synaptic currents (eIPSCs), spontaneous IPSCs (sIPSCs) and miniature IPSCs were recorded in superficial spinal dorsal horn neurons of spinal slices from young adult rats. We used (2S,3S)-3-[3-[4-(trifluoromethyl)benzoylamino]benzyloxy]aspartate (TFB-TBOA), to block both GLT-1 and GLAST and dihydrokainic acid to block only GLT-1. We found that blockade of both GLAST and GLT-1 and blockade of only GLT-1 in the spinal dorsal horn decreased the amplitude of GABAergic eIPSCs, as well as both the amplitude and frequency of GABAergic sIPSCs or miniature IPSCs. Pharmacological inhibition of glial glutamine synthetase had similar effects on both GABAergic eIPSCs and sIPSCs. We provided evidence demonstrating that the reduction in GABAergic strength induced by the inhibition of glial glutamate transporters is due to insufficient GABA synthesis through the glutamate-glutamine cycle between astrocytes and neurons. Thus, our results indicate that deficient glial glutamate transporters and glutamine synthetase significantly attenuate GABAergic synaptic strength in the spinal dorsal horn, which may be a crucial synaptic mechanism underlying glial-neuronal interactions caused by dysfunctional astrocytes in pathological pain conditions.


Assuntos
Transportador 1 de Aminoácido Excitatório/fisiologia , Glutamato-Amônia Ligase/fisiologia , Neuroglia/fisiologia , Células do Corno Posterior/fisiologia , Sinapses/fisiologia , Ácido gama-Aminobutírico/fisiologia , Animais , Ácido Aspártico/análogos & derivados , Ácido Aspártico/antagonistas & inibidores , Ácido Aspártico/farmacologia , Astrócitos/metabolismo , Estimulação Elétrica , Fenômenos Eletrofisiológicos , Transportador 1 de Aminoácido Excitatório/antagonistas & inibidores , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Glutamato-Amônia Ligase/antagonistas & inibidores , Glutamina/farmacologia , Ácido Caínico/análogos & derivados , Ácido Caínico/farmacologia , Masculino , Neuroglia/efeitos dos fármacos , Técnicas de Patch-Clamp , Células do Corno Posterior/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Sinapses/efeitos dos fármacos , Ácido gama-Aminobutírico/farmacologia
17.
Curr Eye Res ; 37(3): 170-8, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22335803

RESUMO

PURPOSE: To investigate the role of glutamate transporters (GLTs)in retinal glial cells that were treated with N-methyl-D-aspartate (NMDA), in retinal ganglion cell (RGC) survival. METHODS: Primary cultures of retinal glial cells or RGCs from 3-day-old Sprague-Dawley rats were employed in the present study. Retinal glial cells were treated with NMDA and changes in GLT mRNA and protein expression were analyzed. The effects of pretreating retinal glial cells with the GLAST-specific inhibitor, rottlerin (ROT), and the GLT-1-specific inhibitor, dihydrokainic acid (DHK), on RGC survival were investigated under exposure to NMDA. The amount of glutamate in the culture medium of retinal glial cells was measured by high-performance liquid chromatography. RESULTS: NMDA treatment increased GLAST and GLT-1 expression. GLAST and GLT-1 mRNA expression increased by 2.94-fold and 3.36-fold at 12 h after treatment with the highest concentration of NMDA (33 mM), and by 1.41-fold and 1.39-fold at 24 h, respectively. GLT-1 and GLAST protein expression also increased. MK801, an NMDA-receptor antagonist, inhibited the NMDA-induced upregulation of GLT mRNA expression. Co-culture with retinal glial cells increased the survival rate of RGCs. ROT decreased the survival rate of RGCs, whereas DHK significantly increased the survival rate of RGCs treated with 33 mM NMDA. NMDA treatment reduced the total amount of glutamate in the culture medium, particularly when 33 mM NMDA was added to the medium. ROT pretreatment increased the amount of glutamate in the culture medium, whereas DHK pretreatment decreased it. CONCLUSION: GLAST and GLT-1 may have different roles in the survival of RGCs mediated by retinal glial cells. These results suggest that the NMDA-associated induction of GLTs plays an important role in RGC survival.


Assuntos
Transportador 1 de Aminoácido Excitatório/fisiologia , Transportador 2 de Aminoácido Excitatório/fisiologia , N-Metilaspartato/farmacologia , Neuroglia/citologia , Receptores de N-Metil-D-Aspartato/metabolismo , Células Ganglionares da Retina/efeitos dos fármacos , Acetofenonas/farmacologia , Animais , Animais Recém-Nascidos , Benzopiranos/farmacologia , Western Blotting , Sobrevivência Celular/fisiologia , Células Cultivadas , Técnicas de Cocultura , Meios de Cultura , Maleato de Dizocilpina/farmacologia , Inibidores Enzimáticos/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Transportador 1 de Aminoácido Excitatório/antagonistas & inibidores , Transportador 2 de Aminoácido Excitatório/antagonistas & inibidores , Ácido Caínico/análogos & derivados , Ácido Caínico/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Células Ganglionares da Retina/citologia , Células Ganglionares da Retina/metabolismo
18.
Neurochem Int ; 61(4): 566-74, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22306776

RESUMO

GLAST is the predominant glutamate transporter in the cerebellum and contributes substantially to glutamate transport in forebrain. This astroglial glutamate transporter quickly binds and clears synaptically released glutamate and is principally responsible for ensuring that synaptic glutamate concentrations remain low. This process is associated with a significant energetic cost. Compartmentalization of GLAST with mitochondria and proteins involved in energy metabolism could provide energetic support for glutamate transport. Therefore, we performed immunoprecipitation and co-localization experiments to determine if GLAST might co-compartmentalize with proteins involved in energy metabolism. GLAST was immunoprecipitated from rat cerebellum and subunits of the Na(+)/K(+) ATPase, glycolytic enzymes, and mitochondrial proteins were detected. GLAST co-localized with mitochondria in cerebellar tissue. GLAST also co-localized with mitochondria in fine processes of astrocytes in organotypic hippocampal slice cultures. From these data, we hypothesized that mitochondria participate in a macromolecular complex with GLAST to support oxidative metabolism of transported glutamate. To determine the functional metabolic role of this complex, we measured CO(2) production from radiolabeled glutamate in cultured astrocytes and compared it to overall glutamate uptake. Within 15 min, 9% of transported glutamate was converted to CO(2). This CO(2) production was blocked by inhibitors of glutamate transport and glutamate dehydrogenase, but not by an inhibitor of glutamine synthetase. Our data support a model in which GLAST exists in a macromolecular complex that allows transported glutamate to be metabolized in mitochondria to support energy production.


Assuntos
Transportador 1 de Aminoácido Excitatório/fisiologia , Ácido Glutâmico/metabolismo , Animais , Cerebelo/metabolismo , Humanos , Masculino , Ratos , Ratos Sprague-Dawley
19.
Neuropharmacology ; 63(2): 181-9, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22342743

RESUMO

A hyperglutamatergic state has been hypothesized to drive escalation of alcohol intake. This hypothesis predicts that an impairment of glutamate clearance through inactivation of the astrocytic glutamate transporter, GLAST (EAAT1), will result in escalation of alcohol consumption. Here, we used mice with a deletion of GLAST to test this prediction. WT and GLAST KO mice were tested for alcohol consumption using two-bottle free-choice drinking. Alcohol reward was evaluated using conditioned place preference (CPP). Sensitivity to depressant alcohol effects was tested using the accelerating rotarod, alcohol-induced hypothermia, and loss of righting reflex. Extracellular glutamate was measured using microdialysis, and striatal slice electrophysiology was carried out to examine plasticity of the cortico-striatal pathway as a model system in which adaptations to the constitutive GLAST deletion can be studied. Contrary to our hypothesis, GLAST KO mice showed markedly decreased alcohol consumption, and lacked CPP for alcohol, despite a higher locomotor response to this drug. Alcohol-induced ataxia, hypothermia, and sedation were unaffected. In striatal slices from GLAST KO mice, long-term depression (LTD) induced by high frequency stimulation, or by post-synaptic depolarization combined with the l-type calcium channel activator FPL 64176 was absent. In contrast, normal synaptic depression was observed after application of the cannabinoid 1 (CB1) receptor agonist WIN55,212-2. Constitutive deletion of GLAST unexpectedly results in markedly reduced alcohol consumption and preference, associated with markedly reduced alcohol reward. Endocannabinoid signaling appears to be down-regulated upstream of the CB1 receptor as a result of the GLAST deletion, and is a candidate mechanism behind the reduction of alcohol reward observed.


Assuntos
Consumo de Bebidas Alcoólicas/metabolismo , Endocanabinoides/metabolismo , Transportador 1 de Aminoácido Excitatório/genética , Recompensa , Transdução de Sinais/fisiologia , Consumo de Bebidas Alcoólicas/genética , Animais , Aprendizagem por Associação/efeitos dos fármacos , Aprendizagem por Associação/fisiologia , Comportamento de Escolha/efeitos dos fármacos , Comportamento de Escolha/fisiologia , Condicionamento Clássico/efeitos dos fármacos , Condicionamento Clássico/fisiologia , Etanol/farmacologia , Transportador 1 de Aminoácido Excitatório/fisiologia , Camundongos , Camundongos Knockout , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Transdução de Sinais/efeitos dos fármacos
20.
Neurochem Int ; 59(6): 871-9, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21856347

RESUMO

Glutamate, the major excitatory amino acid, activates a wide variety of signal transduction cascades. Synaptic plasticity relies on activity-dependent differential protein expression. Ionotropic and metabotropic glutamate receptors have been critically involved in long-term synaptic changes, although recent findings suggest that the electrogenic Na(+)-dependent glutamate transporters, responsible of its removal from the synaptic cleft, participate in glutamate-induced signaling. Transporter proteins are expressed in neurons and glia cells albeit most of the glutamate uptake occurs in the glial compartment. Within the cerebellum, Bergmann glial cells are close to glutamatergic synapses and participate actively in the recycling of glutamate through the glutamate/glutamine shuttle. In this context, we decided to investigate a plausible role of Bergmann glia glutamate transporters as signaling entities. To this end, primary cultures of chick cerebellar Bergmann glial cells were exposed to d-aspartate (D-Asp) and other transporter ligands and the serine 2448 phosphorylation pattern of the master regulator of protein synthesis, namely the mammalian target of rapamycin (mTOR), determined. An increase in mTOR phosphorylation and activity was detected. The signaling cascade included Ca(2+) influx, activation of the phosphatidylinositol 3-kinase and protein kinase B. Furthermore, transporter signaling resulted also in an increase in activator protein-1 (AP-1) binding to DNA and the up-regulation of the transcription of an AP-1 driven gene construct. These results add a novel mediator of the glutamate effects at the translational and transcriptional levels and further strengthen the notion of the critical involvement of glia cells in synaptic function.


Assuntos
Transportador 1 de Aminoácido Excitatório/fisiologia , Neuroglia/classificação , Neuroglia/metabolismo , Transdução de Sinais/fisiologia , Animais , Ácido Aspártico/fisiologia , Cálcio/metabolismo , Cátions Bivalentes , Embrião de Galinha , Galinhas , Transportador 1 de Aminoácido Excitatório/genética , Ácido Glutâmico/metabolismo , Neuroglia/enzimologia , Fosfatidilinositol 3-Quinase/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Cultura Primária de Células , Serina-Treonina Quinases TOR/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA