Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
J Neurosci ; 41(39): 8126-8133, 2021 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-34400517

RESUMEN

Neurotransmitter spillover is a form of communication not readily predicted by anatomic structure. In the cerebellum, glutamate spillover from climbing fibers recruits molecular layer interneurons in the absence of conventional synaptic connections. Spillover-mediated signaling is typically limited by transporters that bind and reuptake glutamate. Here, we show that patterned expression of the excitatory amino acid transporter 4 (EAAT4) in Purkinje cells regulates glutamate spillover to molecular layer interneurons. Using male and female Aldolase C-Venus knock-in mice to visualize zebrin microzones, we find larger climbing fiber-evoked spillover EPSCs in regions with low levels of EAAT4 compared with regions with high EAAT4. This difference is not explained by presynaptic glutamate release properties or postsynaptic receptor density but rather by differences in the glutamate concentration reaching receptors on interneurons. Inhibiting glutamate transport normalizes the differences between microzones, suggesting that heterogeneity in EAAT4 expression is a primary determinant of differential spillover. These results show that neuronal glutamate transporters limit extrasynaptic transmission in a non-cell-autonomous manner and provide new insight into the functional specialization of cerebellar microzones.SIGNIFICANCE STATEMENT Excitatory amino acid transporters (EAATs) help maintain the fidelity and independence of point-to-point synaptic transmission. Whereas glial transporters are critical to maintain low ambient levels of extracellular glutamate to prevent excitotoxicity, neuronal transporters have more subtle roles in shaping excitatory synaptic transmission. Here we show that the patterned expression of neuronal EAAT4 in cerebellar microzones controls glutamate spillover from cerebellar climbing fibers to nearby interneurons. These results contribute to fundamental understanding of neuronal transporter functions and specialization of cerebellar microzones.


Asunto(s)
Cerebelo/metabolismo , Transportador 4 de Aminoácidos Excitadores/metabolismo , Potenciales Postsinápticos Excitadores/fisiología , Ácido Glutámico/metabolismo , Interneuronas/metabolismo , Transmisión Sináptica/fisiología , Animales , Transportador 4 de Aminoácidos Excitadores/genética , Ratones , Células de Purkinje/metabolismo , Sinapsis/metabolismo
2.
Hum Mol Genet ; 27(15): 2614-2627, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29741614

RESUMEN

Loss of excitatory amino acid transporters (EAATs) has been implicated in a number of human diseases including spinocerebellar ataxias, Alzhiemer's disease and motor neuron disease. EAAT4 and GLAST/EAAT1 are the two predominant EAATs responsible for maintaining low extracellular glutamate levels and preventing neurotoxicity in the cerebellum, the brain region essential for motor control. Here using genetically modified mice we identify new critical roles for EAAT4 and GLAST/EAAT1 as modulators of Purkinje cell (PC) spontaneous firing patterns. We show high EAAT4 levels, by limiting mGluR1 signalling, are essential in constraining inherently heterogeneous firing of zebrin-positive PCs. Moreover mGluR1 antagonists were found to restore regular spontaneous PC activity and motor behaviour in EAAT4 knockout mice. In contrast, GLAST/EAAT1 expression is required to sustain normal spontaneous simple spike activity in low EAAT4 expressing (zebrin-negative) PCs by restricting NMDA receptor activation. Blockade of NMDA receptor activity restores spontaneous activity in zebrin-negative PCs of GLAST knockout mice and furthermore alleviates motor deficits. In addition both transporters have differential effects on PC survival, with zebrin-negative PCs more vulnerable to loss of GLAST/EAAT1 and zebrin-positive PCs more vulnerable to loss of EAAT4. These findings reveal that glutamate transporter dysfunction through elevated extracellular glutamate and the aberrant activation of extrasynaptic receptors can disrupt cerebellar output by altering spontaneous PC firing. This expands our understanding of disease mechanisms in cerebellar ataxias and establishes EAATs as targets for restoring homeostasis in a variety of neurological diseases where altered cerebellar output is now thought to play a key role in pathogenesis.


Asunto(s)
Cerebelo/metabolismo , Transportador 1 de Aminoácidos Excitadores/genética , Transportador 4 de Aminoácidos Excitadores/genética , Células de Purkinje/fisiología , Animales , Ataxia/genética , Supervivencia Celular/genética , Transportador 1 de Aminoácidos Excitadores/metabolismo , Transportador 4 de Aminoácidos Excitadores/metabolismo , Femenino , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Células de Purkinje/citología , Receptores de Glutamato Metabotrópico/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo
3.
Sci Rep ; 8(1): 3318, 2018 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-29463856

RESUMEN

Niemann-Pick disease Type C1 (NPC1) is a rare hereditary neurodegenerative disease. NPC1-patients suffer, amongst others, from ataxia, based on a loss of cerebellar Purkinje cells (PCs). Impaired expression/function of excitatory amino acid transporters (EAATs) are suspected of contributing to PC-degeneration in hereditary spinocerebellar ataxias (SCAs). Thus, we studied EAAT-expression and its impact to PC-activity in NPC1-/-mice. Western blot revealed reduced EAAT1, EAAT2, EAAT4, and ßIII-spectrin levels in NPC1-/-mice. EAATs play a crucial role in synaptic transmission, thus we were interested in the impact of the reduced EAAT-expression on the function of PCs. Patch-clamp recordings of PCs showed no differences in the firing patterns of NPC1+/+and NPC1-/-mice using a low internal chloride concentration. Because EAAT4 also comprises a chloride permeable ion pore, we perturbed the chloride homeostasis using a high internal chloride concentration. We observed differences in the firing patterns of NPC1+/+and NPC1-/-mice, suggesting an impact of the altered EAAT4-expression. Additionally, the EAAT-antagonist DL-TBOA acts differently in NPC1+/+and NPC1-/-mice. Our data support the line of evidence that an altered EAAT-expression/function is involved in neurodegeneration of PCs observed in SCAs. Thus, we suggest that similar pathogenic mechanisms contribute the loss of PCs in NPC1.


Asunto(s)
Sistema de Transporte de Aminoácidos X-AG/metabolismo , Cerebelo/metabolismo , Transportador 2 de Aminoácidos Excitadores/metabolismo , Transportador 3 de Aminoácidos Excitadores/metabolismo , Transportador 4 de Aminoácidos Excitadores/metabolismo , Proteínas/fisiología , Células de Purkinje/fisiología , Sistema de Transporte de Aminoácidos X-AG/genética , Animales , Células Cultivadas , Cerebelo/citología , Cloruros/metabolismo , Transportador 2 de Aminoácidos Excitadores/genética , Transportador 3 de Aminoácidos Excitadores/genética , Transportador 4 de Aminoácidos Excitadores/genética , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Endogámicos BALB C , Proteína Niemann-Pick C1 , Células de Purkinje/citología , Transmisión Sináptica
4.
Brain Pathol ; 28(2): 240-263, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28268246

RESUMEN

In prion diseases, the brain lesion profile is influenced by the prion "strain" properties, the invasion route to the brain, and still unknown host cell-specific parameters. To gain insight into those endogenous factors, we analyzed the histopathological alterations induced by distinct prion strains in the mouse cerebellum. We show that 22L and ME7 scrapie prion proteins (PrP22L , PrPME7 ), but not bovine spongiform encephalopathy PrP6PB1 , accumulate in a reproducible parasagittal banding pattern in the cerebellar cortex of infected mice. Such banding pattern of PrP22L aggregation did not depend on the neuroinvasion route, but coincided with the parasagittal compartmentation of the cerebellum mostly defined by the expression of zebrins, such as aldolase C and the excitatory amino acid transporter 4, in Purkinje cells. We provide evidence that Purkinje cells display a differential, subtype-specific vulnerability to 22L prions with zebrin-expressing Purkinje cells being more resistant to prion toxicity, while in stripes where PrP22L accumulated most zebrin-deficient Purkinje cells are lost and spongiosis accentuated. In addition, in PrP22L stripes, enhanced reactive astrocyte processes associated with microglia activation support interdependent events between the topographic pattern of Purkinje cell death, reactive gliosis and PrP22L accumulation. Finally, we find that in preclinically-ill mice prion infection promotes at the membrane of astrocytes enveloping Purkinje cell excitatory synapses, upregulation of tumor necrosis factor-α receptor type 1 (TNFR1), a key mediator of the neuroinflammation process. These overall data show that Purkinje cell sensitivity to prion insult is locally restricted by the parasagittal compartmentation of the cerebellum, and that perisynaptic astrocytes may contribute to prion pathogenesis through prion-induced TNFR1 upregulation.


Asunto(s)
Cerebelo/metabolismo , Cerebelo/patología , Proteínas Priónicas/metabolismo , Animales , Astrocitos/metabolismo , Astrocitos/patología , Bovinos , Encefalopatía Espongiforme Bovina/metabolismo , Encefalopatía Espongiforme Bovina/patología , Transportador 4 de Aminoácidos Excitadores/genética , Transportador 4 de Aminoácidos Excitadores/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Inflamación/metabolismo , Inflamación/patología , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/metabolismo , Microglía/patología , Neuronas/metabolismo , Neuronas/patología , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Scrapie/metabolismo , Scrapie/patología , Sinapsis/metabolismo , Sinapsis/patología
5.
Pain ; 158(4): 705-716, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28030475

RESUMEN

Metabotropic glutamate receptor 5 (mGluR5) is an excitatory G-protein-coupled receptor (GPCR) present in the spinal cord dorsal horn (SCDH) where it has a well-established role in pain. In addition to its traditional location on the cytoplasmic membrane, recent evidence shows that these receptors are present intracellularly on the nuclear membrane in the spinal cord dorsal horn and are implicated in neuropathic pain. Nuclear mGluR5 is a functional receptor that binds glutamate entering the cell through the neuronal glutamate transporter (GT) EAAT3 and activates transcription factor c-fos, whereas plasma membrane mGluR5 is responsible for c-jun activation. Here, we extend these findings to a model of inflammatory pain using complete Freund's adjuvant (CFA) and show that nuclear mGluR5 is also upregulated in the spinal cord dorsal horn following inflammation. We also show that pretreatment with an excitatory amino acid transporter (EAAT) inhibitor attenuates pain and decreases Fos, but not Jun, expression in complete Freund's adjuvant rats. In contrast, selective glial glutamate transporter inhibitors are pronociceptive and increase spinal glutamate concentrations. Additionally, we found that permeable mGluR5 antagonists are more effective at attenuating pain and Fos expression than nonpermeable group I mGluR antagonists. Taken together, these results suggest that under inflammatory conditions, intracellular mGluR5 is actively involved in the relay of nociceptive information in the spinal cord.


Asunto(s)
Espacio Intracelular/metabolismo , Dolor/patología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptor del Glutamato Metabotropico 5/metabolismo , Médula Espinal/metabolismo , Animales , Cadherinas/metabolismo , Condicionamiento Operante/efectos de los fármacos , Ciclodextrinas/farmacología , Modelos Animales de Enfermedad , Transportador 4 de Aminoácidos Excitadores/metabolismo , Adyuvante de Freund/toxicidad , Ácido Glutámico/toxicidad , Histona Desacetilasa 1/metabolismo , Inflamación/inducido químicamente , Inflamación/complicaciones , Masculino , Microdiálisis , Dolor/etiología , Proteínas Proto-Oncogénicas c-jun/metabolismo , Ratas , Ratas Long-Evans
6.
Cell Physiol Biochem ; 40(5): 1252-1260, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27978527

RESUMEN

BACKGROUND: Cellular uptake of glutamate by the excitatory amino-acid transporters (EAATs) decreases excitation and thus participates in the regulation of neuroexcitability. Kinases impacting on neuronal function include Lithium-sensitive glycogen synthase kinase GSK3ß. The present study thus explored whether the activities of EAAT3 and/or EAAT4 isoforms are sensitive to GSK3ß. METHODS: cRNA encoding wild type EAAT3 (SLC1A1) or EAAT4 (SLC1A6) was injected into Xenopus oocytes without or with additional injection of cRNA encoding wild type GSK3ß or the inactive mutant K85AGSK3ß. Dual electrode voltage clamp was performed in order to determine glutamate-induced current (IEAAT). RESULTS: Appreciable IEAAT was observed in EAAT3 or EAAT4 expressing but not in water injected oocytes. IEAAT was significantly increased by coexpression of GSK3ß but not by coexpression of K85AGSK3ß. Coexpression of GSK3ß increased significantly the maximal IEAAT in EAAT3 or EAAT4 expressing oocytes, without significantly modifying apparent affinity of the carriers. Lithium (1 mM) exposure for 24 hours decreased IEAAT in EAAT3 and GSK3ß expressing oocytes to values similar to IEAAT in oocytes expressing EAAT3 alone. Lithium did not significantly modify IEAAT in oocytes expressing EAAT3 without GSK3ß. CONCLUSIONS: Lithium-sensitive GSK3ß is a powerful regulator of excitatory amino acid transporters EAAT3 and EAAT4.


Asunto(s)
Transportador 3 de Aminoácidos Excitadores/metabolismo , Transportador 4 de Aminoácidos Excitadores/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Litio/farmacología , Regulación hacia Arriba/efectos de los fármacos , Animales , Transporte Biológico/efectos de los fármacos , Ácido Glutámico/metabolismo , Humanos , Proteínas Mutantes/metabolismo , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Xenopus laevis
7.
J Membr Biol ; 249(3): 239-49, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26690923

RESUMEN

Excitatory amino acid transporters EAAT1 (SLC1A3), EAAT2 (SLC1A2), EAAT3 (SLC1A1), and EAAT4 (SLC1A6) serve to clear L-glutamate from the synaptic cleft and are thus important for the limitation of neuronal excitation. EAAT3 has previously been shown to form complexes with caveolin-1, a major component of caveolae, which participate in the regulation of transport proteins. The present study explored the impact of caveolin-1 on electrogenic transport by excitatory amino acid transporter isoforms EAAT1-4. To this end cRNA encoding EAAT1, EAAT2, EAAT3, or EAAT4 was injected into Xenopus oocytes without or with additional injection of cRNA encoding caveolin-1. The L-glutamate (2 mM)-induced inward current (I Glu) was taken as a measure of glutamate transport. As a result, I Glu was observed in EAAT1-, EAAT2-, EAAT3-, or EAAT4-expressing oocytes but not in water-injected oocytes, and was significantly decreased by coexpression of caveolin-1. Caveolin-1 decreased significantly the maximal transport rate. Treatment of EAATs-expressing oocytes with brefeldin A (5 µM) was followed by a decrease in conductance, which was similar in oocytes expressing EAAT together with caveolin-1 as in oocytes expressing EAAT1-4 alone. Thus, caveolin-1 apparently does not accelerate transporter protein retrieval from the cell membrane. In conclusion, caveolin-1 is a powerful negative regulator of the excitatory glutamate transporters EAAT1, EAAT2, EAAT3, and EAAT4.


Asunto(s)
Caveolina 1/metabolismo , Transportador 1 de Aminoácidos Excitadores/metabolismo , Transportador 2 de Aminoácidos Excitadores/metabolismo , Transportador 3 de Aminoácidos Excitadores/metabolismo , Transportador 4 de Aminoácidos Excitadores/metabolismo , Animales , Transporte Biológico , Caveolina 1/genética , Transportador 1 de Aminoácidos Excitadores/genética , Transportador 2 de Aminoácidos Excitadores/genética , Transportador 3 de Aminoácidos Excitadores/genética , Transportador 4 de Aminoácidos Excitadores/genética , Expresión Génica , Ácido Glutámico/metabolismo , Humanos , Oocitos/metabolismo , Xenopus laevis
8.
Hum Mol Genet ; 25(20): 4448-4461, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28173092

RESUMEN

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.


Asunto(s)
Ataxia Cerebelosa/metabolismo , Modelos Animales de Enfermedad , Transportador 1 de Aminoácidos Excitadores/metabolismo , Transportador 4 de Aminoácidos Excitadores/metabolismo , Células de Purkinje/metabolismo , Espectrina/metabolismo , Ataxias Espinocerebelosas/metabolismo , Animales , Ataxia Cerebelosa/genética , Ataxia Cerebelosa/patología , Transportador 1 de Aminoácidos Excitadores/fisiología , Transportador 4 de Aminoácidos Excitadores/fisiología , Femenino , Masculino , Ratones , Ratones Noqueados , Fenotipo , Células de Purkinje/patología , Espectrina/fisiología , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/patología
9.
Cerebellum ; 15(3): 314-21, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26194056

RESUMEN

Excitatory amino acid transporter 4 (EAAT4) is believed to be critical to the synaptic activity of cerebellar Purkinje cells by limiting extracellular glutamate concentrations and facilitating the induction of long-term depression. However, the modulation of EAAT4 expression has not been elucidated. It has been shown that Ras homolog enriched in brain (Rheb)/mammalian target of rapamycin (mTOR) signaling plays essential roles in the regulation of protein translation, cell size, and cell growth. In addition, we previously found that a cascade including mTOR suppression and Akt activation induces increased expression of EAAT2 in astrocytes. In the present work, we explored whether Rheb/mTOR signaling is involved in the regulation of EAAT4 expression using conditional Rheb1 knockout mice. Our results demonstrated that Rheb1 deficiency resulted in the downregulation of EAAT4 expression, as well as decreased activity of mTOR and increased activity of Akt. The downregulation of EAAT4 was also confirmed by reduced EAAT4 currents and slowed kinetics of α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptor-mediated currents. On the other hand, conditional knockout of Rheb1 did not alter the morphology of Purkinje cell layer and the number of Purkinje cells. Overall, our findings suggest that small GTPase Rheb1 is a modulator in the expression of EAAT4 in Purkinje cells.


Asunto(s)
Transportador 4 de Aminoácidos Excitadores/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Neuropéptidos/metabolismo , Células de Purkinje/metabolismo , Animales , Western Blotting , Femenino , Inmunohistoquímica , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Potenciales de la Membrana/fisiología , Ratones Noqueados , Proteínas de Unión al GTP Monoméricas/genética , Complejos Multiproteicos/metabolismo , Neuropéptidos/genética , Técnicas de Placa-Clamp , Células de Purkinje/citología , Proteína Homóloga de Ras Enriquecida en el Cerebro , Receptores AMPA , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de la Célula Individual , Serina-Treonina Quinasas TOR/metabolismo
10.
PLoS One ; 8(7): e70988, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23923038

RESUMEN

Klotho, a transmembrane protein, which can be cleaved off as ß-glucuronidase and hormone, is released in both, kidney and choroid plexus and encountered in blood and cerebrospinal fluid. Klotho deficiency leads to early appearance of age-related disorders and premature death. Klotho may modify transport by inhibiting 1,25(OH)2D3 formation or by directly affecting channel and carrier proteins. The present study explored whether Klotho influences the activity of the Na(+)-coupled excitatory amino acid transporters EAAT3 and EAAT4, which are expressed in kidney (EAAT3), intestine (EAAT3) and brain (EAAT3 and EAAT4). To this end, cRNA encoding EAAT3 or EAAT4 was injected into Xenopus oocytes with and without additional injection of cRNA encoding Klotho. EAAT expressing Xenopus oocytes were further treated with recombinant human ß-Klotho protein with or without ß-glucuronidase inhibitor D-saccharic acid 1,4-lactone monohydrate (DSAL). Electrogenic excitatory amino acid transport was determined as L-glutamate-induced current (Iglu) in two electrode voltage clamp experiments. EAAT3 and EAAT4 protein abundance in the Xenopus oocyte cell membrane was visualized by confocal microscopy and quantified utilizing chemiluminescence. As a result, coexpression of Klotho cRNA significantly increased Iglu in both, EAAT3 or EAAT4-expressing Xenopus oocytes. Klotho cRNA coexpression significantly increased the maximal current and cell membrane protein abundance of both EAAT3 and EAAT4. The effect of Klotho coexpression on EAAT3 and EAAT4 activity was mimicked by treating EAAT3 or EAAT4-expressing Xenopus oocytes with recombinant human ß-Klotho protein. The effects of Klotho coexpression and of treatment with recombinant human ß-Klotho protein were both abrogated in the presence of DSAL (10 µM). In conclusion, Klotho is a novel, powerful regulator of the excitatory amino acid transporters EAAT3 and EAAT4.


Asunto(s)
Transportador 3 de Aminoácidos Excitadores/metabolismo , Transportador 4 de Aminoácidos Excitadores/metabolismo , Glucuronidasa/metabolismo , Neuronas/metabolismo , Potenciales de Acción , Animales , Transporte Biológico , Transportador 3 de Aminoácidos Excitadores/genética , Transportador 4 de Aminoácidos Excitadores/genética , Expresión Génica , Glucuronidasa/genética , Ácido Glutámico/metabolismo , Proteínas Klotho , Oocitos/metabolismo , Xenopus laevis
11.
J Neurosci ; 33(3): 1068-87, 2013 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-23325245

RESUMEN

In the present study, the mechanism of action and molecular basis for the activity of the first class of selective inhibitors of the human excitatory amino acid transporter subtype 1 (EAAT1) and its rodent ortholog GLAST are elucidated. The previously reported specificity of UCPH-101 and UCPH-102 for EAAT1 over EAAT2 and EAAT3 is demonstrated to extend to the EAAT4 and EAAT5 subtypes as well. Interestingly, brief exposure to UCPH-101 induces a long-lasting inactive state of EAAT1, whereas the inhibition exerted by closely related analogs is substantially more reversible in nature. In agreement with this, the kinetic properties of UCPH-101 unblocking of the transporter are considerably slower than those of UCPH-102. UCPH-101 exhibits noncompetitive inhibition of EAAT1, and its binding site in GLAST has been delineated in an elaborate mutagenesis study. Substitutions of several residues in TM3, TM4c, and TM7a of GLAST have detrimental effects on the inhibitory potency and/or efficacy of UCPH-101 while not affecting the pharmacological properties of (S)-glutamate or the competitive EAAT inhibitor TBOA significantly. Hence, UCPH-101 is proposed to target a predominantly hydrophobic crevice in the "trimerization domain" of the GLAST monomer, and the inhibitor is demonstrated to inhibit the uptake through the monomer that it binds to exclusively and not to affect substrate translocation through the other monomers in the GLAST trimer. The allosteric mode of UCPH-101 inhibition underlines the functional importance of the trimerization domain of the EAAT and demonstrates the feasibility of modulating transporter function through ligand binding to regions distant from its "transport domain."


Asunto(s)
Regulación Alostérica/efectos de los fármacos , Benzopiranos/farmacología , Transportador 1 de Aminoácidos Excitadores/antagonistas & inhibidores , Animales , Transporte Biológico/fisiología , Células Cultivadas , Transportador 1 de Aminoácidos Excitadores/genética , Transportador 1 de Aminoácidos Excitadores/metabolismo , Transportador 2 de Aminoácidos Excitadores/genética , Transportador 2 de Aminoácidos Excitadores/metabolismo , Transportador 3 de Aminoácidos Excitadores/genética , Transportador 3 de Aminoácidos Excitadores/metabolismo , Transportador 4 de Aminoácidos Excitadores/genética , Transportador 4 de Aminoácidos Excitadores/metabolismo , Humanos , Ratas
12.
PLoS One ; 7(9): e46261, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23049999

RESUMEN

Glutamate is the neurotransmitter released from hair cells. Its clearance from the synaptic cleft can shape neurotransmission and prevent excitotoxicity. This may be particularly important in the inner ear and in other sensory organs where there is a continually high rate of neurotransmitter release. In the case of most cochlear and type II vestibular hair cells, clearance involves the diffusion of glutamate to supporting cells, where it is taken up by EAAT1 (GLAST), a glutamate transporter. A similar mechanism cannot work in vestibular type I hair cells as the presence of calyx endings separates supporting cells from hair-cell synapses. Because of this arrangement, it has been conjectured that a glutamate transporter must be present in the type I hair cell, the calyx ending, or both. Using whole-cell patch-clamp recordings, we demonstrate that a glutamate-activated anion current, attributable to a high-affinity glutamate transporter and blocked by DL-TBOA, is expressed in type I, but not in type II hair cells. Molecular investigations reveal that EAAT4 and EAAT5, two glutamate transporters that could underlie the anion current, are expressed in both type I and type II hair cells and in calyx endings. EAAT4 has been thought to be expressed almost exclusively in the cerebellum and EAAT5 in the retina. Our results show that these two transporters have a wider distribution in mice. This is the first demonstration of the presence of transporters in hair cells and provides one of the few examples of EAATs in presynaptic elements.


Asunto(s)
Transportador 4 de Aminoácidos Excitadores/metabolismo , Transportador 5 de Aminoácidos Excitadores/metabolismo , Células Ciliadas Vestibulares/metabolismo , Terminaciones Nerviosas/metabolismo , Animales , Western Blotting , Electrofisiología , Transportador 4 de Aminoácidos Excitadores/genética , Transportador 5 de Aminoácidos Excitadores/genética , Femenino , Inmunohistoquímica , Hibridación in Situ , Masculino , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Cell Physiol Biochem ; 28(4): 693-702, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22178881

RESUMEN

The Janus-activated kinase-2 JAK2 is involved in the signaling of leptin and erythropoietin receptors and mediates neuroprotective effects of the hormones. In theory, JAK2 could be effective through modulation of the glutamate transporters, carriers accounting for the clearance of glutamate released during neurotransmission. The present study thus elucidated the effect of JAK2 on the glutamate transporters EAAT1, EAAT2, EAAT3 and EAAT4. To this end, cRNA encoding the carriers was injected into Xenopus oocytes with or without cRNA encoding JAK2 and glutamate transport was estimated from glutamate induced current (I(glu)). I(glu) was observed in Xenopus oocytes expressing EAAT1 or EAAT2 or EAAT3 or EAAT4, but not in water injected oocytes. Coexpression of JAK2 resulted in an increase of I(glu) by 83% (EAAT1), 67% (EAAT2), 42% (EAAT3) and 126% (EAAT4). As shown for EAAT4 expressing Xenopus oocytes, the effect of JAK2 was mimicked by gain of function mutation (V617F)JAK2 but not by the inactive mutant (K882E)JAK2. Incubation with JAK2 inhibitor AG490 (40 µM) resulted in a gradual decrease of I(glu) by 53%, 79% and 92% within 3, 6 and 24 hours. Confocal microscopy and chemiluminescence analysis revealed that JAK2 coexpression increased EAAT4 protein abundance in the cell membrane. Disruption of transcription did not appreciably modify the up-regulation of I(glu) in EAAT4 expressing oocytes. The decay of I(glu) following inhibition of carrier insertion with brefeldin A was similar in oocytes expressing EAAT4 + JAK2 and oocytes expressing EAAT4 alone, indicating that JAK2 did not appreciably affect carrier retrieval from the membrane. In conclusion, JAK2 is a novel powerful regulator of glutamate transporters and thus participates in the protection against excitotoxicity.


Asunto(s)
Proteínas de Transporte de Glutamato en la Membrana Plasmática/metabolismo , Janus Quinasa 2/metabolismo , Sustitución de Aminoácidos , Animales , Transportador 1 de Aminoácidos Excitadores/metabolismo , Transportador 2 de Aminoácidos Excitadores/metabolismo , Transportador 3 de Aminoácidos Excitadores/metabolismo , Transportador 4 de Aminoácidos Excitadores/metabolismo , Ácido Glutámico/metabolismo , Humanos , Janus Quinasa 2/genética , Oocitos/metabolismo , Técnicas de Placa-Clamp , Regulación hacia Arriba , Xenopus laevis/genética
14.
J Neurosci ; 31(46): 16581-90, 2011 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-22090485

RESUMEN

Mutations in the gene encoding ß-III spectrin give rise to spinocerebellar ataxia type 5, a neurodegenerative disease characterized by progressive thinning of the molecular layer, loss of Purkinje cells and increasing motor deficits. A mouse lacking full-length ß-III spectrin (ß-III⁻/⁻) displays a similar phenotype. In vitro and in vivo analyses of Purkinje cells lacking ß-III spectrin, reveal a critical role for ß-III spectrin in Purkinje cell morphological development. Disruption of the normally well ordered dendritic arborization occurs in Purkinje cells from ß-III⁻/⁻ mice, specifically showing a loss of monoplanar organization, smaller average dendritic diameter and reduced densities of Purkinje cell spines and synapses. Early morphological defects appear to affect distribution of dendritic, but not axonal, proteins. This study confirms that thinning of the molecular layer associated with disease pathogenesis is a consequence of Purkinje cell dendritic degeneration, as Purkinje cells from 8-month-old ß-III⁻/⁻ mice have drastically reduced dendritic volumes, surface areas and total dendritic lengths compared with 5- to 6-week-old ß-III⁻/⁻ mice. These findings highlight a critical role of ß-III spectrin in dendritic biology and are consistent with an early developmental defect in ß-III⁻/⁻ mice, with abnormal Purkinje cell dendritic morphology potentially underlying disease pathogenesis.


Asunto(s)
Corteza Cerebral/citología , Corteza Cerebral/crecimiento & desarrollo , Dendritas/ultraestructura , Espinas Dendríticas/metabolismo , Células de Purkinje/citología , Espectrina/metabolismo , Factores de Edad , Animales , Animales Recién Nacidos , Calbindinas , Transportador 4 de Aminoácidos Excitadores/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Transportador de Glucosa de Tipo 2/metabolismo , Técnicas In Vitro , Depresión Sináptica a Largo Plazo/efectos de los fármacos , Depresión Sináptica a Largo Plazo/genética , Ratones , Ratones Noqueados , Microscopía Electrónica de Transmisión , Canal de Sodio Activado por Voltaje NAV1.6 , Proteínas del Tejido Nervioso/metabolismo , Técnicas de Placa-Clamp , Proteínas de Transporte de Fosfato/metabolismo , Proteína G de Unión al Calcio S100/metabolismo , Tinción con Nitrato de Plata/métodos , Canales de Sodio/metabolismo , Espectrina/deficiencia , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo
15.
Channels (Austin) ; 5(6): 468-74, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21849820

RESUMEN

Excitatory amino acid transporters (EAATs) do not only mediate secondary-active glutamate uptake but also function as anion channels. We recently used macroscopic current recordings and noise analysis to determine unitary current amplitudes of anion channels associated with a neuronal EAAT isoform, EAAT4. We found that, at symmetrical NO(3)(-), EAAT4 anion channels exhibit a single channel conductance of ~1 pS in the absence as well as in the presence of glutamate. These results indicate that glutamate increases EAAT4 anion currents by modifying exclusively open probabilities, however, leaves unitary current amplitudes unaffected. Noise analysis has been developed for ion channels with a single conductance state and limitations might ensue when using this approach for transporter-associated ion channels. We here performed stochastic simulations of EAAT transporter-associated anion channels and noise analysis of simulated currents to assess the reliability and possible limitations of this technique in studying this special class of ion channels.


Asunto(s)
Transportador 4 de Aminoácidos Excitadores/metabolismo , Modelos Biológicos , Nitratos/metabolismo , Transporte Iónico/fisiología , Isoformas de Proteínas/metabolismo
16.
J Biol Chem ; 286(27): 23780-8, 2011 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-21572047

RESUMEN

EAAT glutamate transporters do not only function as secondary-active glutamate transporters but also as anion channels. EAAT anion channel activity depends on transport substrates. For most isoforms, it is negligible without external Na(+) and increased by external glutamate. We here investigated gating of EAAT4 anion channels with various cations and amino acid substrates using patch clamp experiments on a mammalian cell line. We demonstrate that Li(+) can substitute for Na(+) in supporting substrate-activated anion currents, albeit with changed voltage dependence. Anion currents were recorded in glutamate, aspartate, and cysteine, and distinct time and voltage dependences were observed. For each substrate, gating was different in external Na(+) or Li(+). All features of voltage-dependent and substrate-specific anion channel gating can be described by a simplified nine-state model of the transport cycle in which only amino acid substrate-bound states assume high anion channel open probabilities. The kinetic scheme suggests that the substrate dependence of channel gating is exclusively caused by differences in substrate association and translocation. Moreover, the voltage dependence of anion channel gating arises predominantly from electrogenic cation binding and membrane translocation of the transporter. We conclude that all voltage- and substrate-dependent conformational changes of the EAAT4 anion channel are linked to transitions within the transport cycle.


Asunto(s)
Transportador 4 de Aminoácidos Excitadores/metabolismo , Activación del Canal Iónico/fisiología , Litio/metabolismo , Sodio/metabolismo , Animales , Transportador 4 de Aminoácidos Excitadores/genética , Células HEK293 , Humanos , Transporte Iónico/fisiología , Conformación Proteica , Ratas
17.
Asian J Androl ; 13(2): 254-65, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21170079

RESUMEN

Glutamate is a regulated molecule in the mammalian testis. Extracellular regulation of glutamate in the body is determined largely by the expression of plasmalemmal glutamate transporters. We have examined by PCR, western blotting and immunocytochemistry the expression of a panel of sodium-dependent plasmalemmal glutamate transporters in the rat testis. Proteins examined included: glutamate aspartate transporter (GLAST), glutamate transporter 1 (GLT1), excitatory amino acid carrier 1 (EAAC1), excitatory amino acid transporter 4 (EAAT4) and EAAT5. We demonstrate that many of the glutamate transporters in the testis are alternately spliced. GLAST is present as exon-3- and exon-9-skipping forms. GLT1 was similarly present as the alternately spliced forms GLT1b and GLT1c, whereas the abundant brain form (GLT1a) was detectable only at the mRNA level. EAAT5 was also strongly expressed, whereas EAAC1 and EAAT4 were absent. These patterns of expression were compared with the patterns of endogenous glutamate localization and with patterns of d-aspartate accumulation, as assessed by immunocytochemistry. The presence of multiple glutamate transporters in the testis, including unusually spliced forms, suggests that glutamate homeostasis may be critical in this organ. The apparent presence of many of these transporters in the testis and sperm may indicate a need for glutamate transport by such cells.


Asunto(s)
Empalme Alternativo , Sistema de Transporte de Aminoácidos X-AG/genética , Testículo/metabolismo , Secuencia de Aminoácidos , Sistema de Transporte de Aminoácidos X-AG/metabolismo , Animales , Ácido Aspártico/metabolismo , Secuencia de Bases , Encéfalo/metabolismo , Cartilla de ADN/genética , Transportador 2 de Aminoácidos Excitadores/genética , Transportador 2 de Aminoácidos Excitadores/metabolismo , Transportador 3 de Aminoácidos Excitadores/genética , Transportador 3 de Aminoácidos Excitadores/metabolismo , Transportador 4 de Aminoácidos Excitadores/genética , Transportador 4 de Aminoácidos Excitadores/metabolismo , Transportador 5 de Aminoácidos Excitadores/genética , Transportador 5 de Aminoácidos Excitadores/metabolismo , Expresión Génica , Ácido Glutámico/metabolismo , Homeostasis , Inmunohistoquímica , Masculino , Datos de Secuencia Molecular , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Retina/metabolismo
18.
J Biol Chem ; 286(5): 3935-43, 2011 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-21127051

RESUMEN

Excitatory amino acid transporters (EAATs) mediate the uptake of glutamate into neuronal and glial cells of the mammalian central nervous system. Two transporters expressed primarily in glia, EAAT1 and EAAT2, are crucial for glutamate homeostasis in the adult mammalian brain. Three neuronal transporters (EAAT3, EAAT4, and EAAT5) appear to have additional functions in regulating and processing cellular excitability. EAATs are assembled as trimers, and the existence of multiple isoforms raises the question of whether certain isoforms can form hetero-oligomers. Co-expression and pulldown experiments of various glutamate transporters showed that EAAT3 and EAAT4, but neither EAAT1 and EAAT2, nor EAAT2 and EAAT3 are capable of co-assembling into heterotrimers. To study the functional consequences of hetero-oligomerization, we co-expressed EAAT3 and the serine-dependent mutant R501C EAAT4 in HEK293 cells and Xenopus laevis oocytes and studied glutamate/serine transport and anion conduction using electrophysiological methods. Individual subunits transport glutamate independently of each other. Apparent substrate affinities are not affected by hetero-oligomerization. However, polarized localization in Madin-Darby canine kidney cells was different for homo- and hetero-oligomers. EAAT3 inserts exclusively into apical membranes of Madin-Darby canine kidney cells when expressed alone. Co-expression with EAAT4 results in additional appearance of basolateral EAAT3. Our results demonstrate the existence of heterotrimeric glutamate transporters and provide novel information about the physiological impact of EAAT oligomerization.


Asunto(s)
Transportador 3 de Aminoácidos Excitadores/metabolismo , Transportador 4 de Aminoácidos Excitadores/metabolismo , Proteínas de Transporte de Glutamato en la Membrana Plasmática/metabolismo , Neuronas/metabolismo , Multimerización de Proteína , Animales , Transporte Biológico , Línea Celular , Fenómenos Electrofisiológicos , Transportador 1 de Aminoácidos Excitadores/metabolismo , Transportador 2 de Aminoácidos Excitadores/metabolismo , Transportador 3 de Aminoácidos Excitadores/genética , Transportador 4 de Aminoácidos Excitadores/genética , Transportador 5 de Aminoácidos Excitadores/metabolismo , Ácido Glutámico/metabolismo , Humanos , Mutación Missense , Neuroglía/metabolismo , Isoformas de Proteínas , Ratas , Especificidad por Sustrato , Transfección
19.
Eur J Neurosci ; 32(11): 1843-53, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21070388

RESUMEN

Neurotransmitters diffuse out of the synaptic cleft and act on adjacent synapses to exert concerted control of the synaptic strength within neural pathways that converge on single target neurons. The excitatory transmitter released from climbing fibers (CFs), presumably glutamate, is shown to inhibit γ-aminobutyric acid (GABA) release at basket cell (BC)-Purkinje cell (PC) synapses in the rat cerebellar cortex through its extrasynaptic diffusion and activation of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors on BC axon terminals. This study aimed at examining how the CF transmitter-diffusion-mediated presynaptic inhibition is controlled by glutamate transporters. Pharmacological blockade of the PC-selective neuronal transporter EAAT4 markedly enhanced CF-induced inhibition of GABAergic transmission. Tetanic CF-stimulation elicited long-term potentiation of glutamate transporters in PCs, and thereby attenuated the CF-induced inhibition. Combined use of electrophysiology and immunohistochemistry revealed a significant inverse relationship between the level of EAAT4 expression and the inhibitory action of CF-stimulation on the GABA release at different cerebellar lobules - the CF-induced inhibition was profound in lobule III, where the EAAT4 expression level was low, whereas it was minimal in lobule X, where EAAT4 was abundant. The findings clearly demonstrate that the neuronal glutamate transporter EAAT4 in PCs plays a critical role in the extrasynaptic diffusion of CF transmitter - it appears not only to retrogradely determine the degree of CF-mediated inhibition of GABAergic inputs to the PC by controlling the glutamate concentration for intersynaptic diffusion, but also regulate synaptic information processing in the cerebellar cortex depending on its differential regional distribution as well as use-dependent plasticity of uptake efficacy.


Asunto(s)
Transportador 4 de Aminoácidos Excitadores/metabolismo , Interneuronas/metabolismo , Neurotransmisores/metabolismo , Células de Purkinje/metabolismo , Sinapsis/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Animales , Cerebelo/citología , Difusión , Transportador 4 de Aminoácidos Excitadores/antagonistas & inhibidores , Potenciales Postsinápticos Inhibidores/fisiología , Interneuronas/citología , Técnicas de Placa-Clamp , Células de Purkinje/citología , Ratas , Ratas Wistar , Transmisión Sináptica/fisiología
20.
Proc Natl Acad Sci U S A ; 107(13): 6022-7, 2010 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-20231455

RESUMEN

The spectrin membrane skeleton controls the disposition of selected membrane channels, receptors, and transporters. In the brain betaIII spectrin binds directly to the excitatory amino acid transporter (EAAT4), the glutamate receptor delta, and other proteins. Mutations in betaIII spectrin link strongly to human spinocerebellar ataxia type 5 (SCA5), correlating with alterations in EAAT4. We have explored the mechanistic basis of this phenotype by targeted gene disruption of Spnb3. Mice lacking intact betaIII spectrin develop normally. By 6 months they display a mild nonprogressive ataxia. By 1 year most Spnb3(-/-) animals develop a myoclonic seizure disorder with significant reductions of EAAT4, EAAT1, GluRdelta, IP3R, and NCAM140. Other synaptic proteins are normal. The cerebellum displays increased dark Purkinje cells (PC), a thin molecular layer, fewer synapses, a loss of dendritic spines, and a 2-fold expansion of the PC dendrite diameter. Membrane and expanded Golgi profiles fill the PC dendrite and soma, and both regions accumulate EAAT4. Correlating with the seizure disorder are enhanced hippocampal levels of neuropeptide Y and EAAT3 and increased calpain proteolysis of alphaII spectrin. It appears that betaIII spectrin disruption impairs synaptogenesis by disturbing the intracellular pathways selectively regulating protein trafficking to the synapse. The mislocalization of these proteins secondarily disrupts glutamate transport dynamics, leading to seizures, neuronal damage, and compensatory changes in EAAT3 and neuropeptide Y.


Asunto(s)
Ataxia/etiología , Convulsiones/etiología , Espectrina/deficiencia , Animales , Ataxia/genética , Ataxia/fisiopatología , Secuencia de Bases , Encéfalo/metabolismo , Encéfalo/fisiopatología , Encéfalo/ultraestructura , Cartilla de ADN/genética , Modelos Animales de Enfermedad , Transportador 4 de Aminoácidos Excitadores/metabolismo , Femenino , Marcación de Gen , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Transmisión , Degeneración Nerviosa/genética , Degeneración Nerviosa/fisiopatología , Fenotipo , Convulsiones/genética , Convulsiones/fisiopatología , Espectrina/genética , Espectrina/fisiología , Ataxias Espinocerebelosas/etiología , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/fisiopatología , Sinapsis/fisiología , Sinapsis/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...