Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 103
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.
Cell Physiol Biochem ; 51(5): 2275-2289, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30537735

RESUMEN

BACKGROUND/AIMS: Parkinson's disease (PD) is a frequently occurring condition that resulted from the loss of midbrain neurons, which synthesize the neurotransmitter dopamine. In this study, we established mouse models of PD to investigate the expression of microRNA-128 (miR-128) and mechanism through which it affects apoptosis of dopamine (DA) neurons and the expression of excitatory amino acid transporter 4 (EAAT4) via binding to axis inhibition protein 1 (AXIN1). METHODS: Gene expression microarray analysis was performed to screen differentially expressed miRNAs that are associated with PD. The targeting relationship between miR-128 and AXIN1 was verified via a bioinformatics prediction and dual-luciferase reporter gene assay. After separation, DA neurons were subjected to a series of inhibitors, activators and shRNAs to validate the mechanisms of miR-128 in controlling of AXIN1 in PD. Positive protein expression of AXIN1 and EAAT4 in DA neurons was determined using immunocytochemistry. miR-128 expression and the mRNA and protein levels of AXIN1 and EAAT4 were evaluated via RT-qPCR and Western blot analysis, respectively. DA neuron apoptosis was evaluated using TUNEL staining. RESULTS: We identified AXIN1 as an upregulated gene in PD based on the microarray data of GSE7621. AXIN1 was targeted and negatively mediated by miR-128. In the DA neurons, upregulated miR-128 expression or sh-AXIN1 increased the positive expression rate of EAAT4 together with mRNA and protein levels, but decreased the mRNA and protein levels of AXIN1, apoptosis rate along with the positive expression rate of AXIN1; however, the opposite trend was found in response to transfection with miR-128 inhibitors. CONCLUSION: Evidence from experimental models revealed that miR-128 might reduce apoptosis of DA neurons while increasing the expression of EAAT4 which might be related to the downregulation of AXIN1. Thus, miR-128 may serve as a potential target for the treatment of PD.


Asunto(s)
Proteína Axina/genética , Neuronas Dopaminérgicas/patología , Transportador 4 de Aminoácidos Excitadores/genética , Regulación de la Expresión Génica , MicroARNs/genética , Enfermedad de Parkinson/genética , Animales , Apoptosis , Neuronas Dopaminérgicas/metabolismo , Redes Reguladoras de Genes , Humanos , Masculino , Ratones Endogámicos C57BL , Enfermedad de Parkinson/patología , Regulación hacia Arriba
3.
J Med Chem ; 61(17): 7741-7753, 2018 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-30011368

RESUMEN

Aspartate (Asp) derivatives are privileged compounds for investigating the roles governed by excitatory amino acid transporters (EAATs) in glutamatergic neurotransmission. Here, we report the synthesis of various Asp derivatives with (cyclo)alkyloxy and (hetero)aryloxy substituents at C-3. Their pharmacological properties were characterized at the EAAT1-4 subtypes. The l- threo-3-substituted Asp derivatives 13a-e and 13g-k were nonsubstrate inhibitors, exhibiting pan activity at EAAT1-4 with IC50 values ranging from 0.49 to 15 µM. Comparisons between (dl- threo)-19a-c and (dl- erythro)-19a-c Asp analogues confirmed that the threo configuration is crucial for the EAAT1-4 inhibitory activities. Analogues (3b-e) of l-TFB-TBOA (3a) were shown to be potent EAAT1-4 inhibitors, with IC50 values ranging from 5 to 530 nM. Hybridization of the nonselective EAAT inhibitor l-TBOA with EAAT2-selective inhibitor WAY-213613 or EAAT3-preferring inhibitor NBI-59159 yielded compounds 8 and 9, respectively, which were nonselective EAAT inhibitors displaying considerably lower IC50 values at EAAT1-4 (11-140 nM) than those displayed by the respective parent molecules.


Asunto(s)
Amoníaco-Liasas/metabolismo , Ácido Aspártico/análogos & derivados , Ácido Aspártico/farmacología , Transportador 1 de Aminoácidos Excitadores/antagonistas & inhibidores , Transportador 3 de Aminoácidos Excitadores/antagonistas & inhibidores , Transportador 4 de Aminoácidos Excitadores/antagonistas & inhibidores , Proteínas de Transporte de Glutamato en la Membrana Plasmática/antagonistas & inhibidores , Ácido Aspártico/síntesis química , Transportador 2 de Aminoácidos Excitadores , Células HEK293 , Humanos , Estructura Molecular , Relación Estructura-Actividad
4.
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
5.
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
6.
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
8.
Hum Pathol ; 60: 174-179, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27816723

RESUMEN

Ossifying fibromyxoid tumors (OFMT) of soft parts are rare, slow-growing tumors that have potential for local recurrence and may metastasize. While OFMT originally was considered benign, several cases of malignant OFMT have been documented. There is no universally accepted risk stratification, although this study emphasizes the importance of utilizing histology, immunohistochemistry and FISH in establishing the diagnosis. Herein, we describe six cases of atypical and malignant OFMT with differences in morphologic features, 5 of which display the proposed morphological criteria for malignancy. The patients were mostly male (M=5, F=1) with an age range of 33-69 years. The tumors arose from the extremities (3 cases), the shoulder (1 case), the head and neck area (1 case), and the paraspinal area (1 case). One tumor had high grade and overtly sarcomatous changes, while another invaded the underlying clavicle. Two cases showed cytological atypia and necrosis. Fluorescence in situ hybridization (FISH) detected rearrangement of the PHF1 gene in 5 cases. All cases were positive for EAAT4 and actin by immunohistochemistry, while negative for desmin. Three tumors were immunoreactive for S100 protein. INI-1 immunohistochemical staining was conserved in all but 2 cases in which a mosaic loss of expression was noted. All but two patients are currently alive and free of disease.


Asunto(s)
Neoplasias Óseas/patología , Fibroma Osificante/patología , Actinas/análisis , Adulto , Anciano , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/genética , Biopsia , Neoplasias Óseas/química , Neoplasias Óseas/genética , Neoplasias Óseas/terapia , Proteínas de Unión al ADN/genética , Supervivencia sin Enfermedad , Transportador 4 de Aminoácidos Excitadores/análisis , Femenino , Fibroma Osificante/química , Fibroma Osificante/genética , Fibroma Osificante/terapia , Reordenamiento Génico , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Masculino , Persona de Mediana Edad , Necrosis , Proteínas del Grupo Polycomb/genética , Proteínas S100/análisis , Proteína SMARCB1/análisis , Factores de Tiempo , Resultado del Tratamiento
9.
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
10.
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
11.
Epilepsia ; 57(6): 984-93, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27153812

RESUMEN

OBJECTIVE: Posttraumatic seizures (PTS) commonly occur following severe traumatic brain injury (sTBI). Risk factors for PTS have been identified, but variability in who develops PTS remains. Excitotoxicity may influence epileptogenesis following sTBI. Glutamate transporters manage glutamate levels and excitatory neurotransmission, and they have been associated with both epilepsy and TBI. Therefore, we aimed to determine if genetic variation in neuronal glutamate transporter genes is associated with accelerated epileptogenesis and increased PTS risk after sTBI. METHODS: Individuals (N = 253) 18-75 years of age with sTBI were assessed for genetic relationships with PTS. Single nucleotide polymorphisms (SNPs) within SLC1A1 and SLC1A6 were assayed. Kaplan-Meier estimates and log-rank statistics were used to compare seizure rates from injury to 3 years postinjury for SNPs by genotype. Hazard ratios (HRs) were estimated using Cox proportional hazards regression for SNPs significant in Kaplan-Meier analyses adjusting for known PTS risk factors. RESULTS: Thirty-two tagging SNPs were examined (SLC1A1: n = 28, SLC1A6: n = 4). Forty-nine subjects (19.37%) had PTS. Of these, 18 (36.7%) seized within 7 days, and 31 (63.3%) seized between 8 days and 3 years post-TBI. With correction for multiple comparisons, genotypes at SNP rs10974620 (SLC1A1) were significantly associated with time to first seizure across the full 3-year follow-up (seizure rates: 77.1% minor allele homozygotes, 24.8% heterozygotes, 16.6% major allele homozygotes; p = 0.001). When seizure follow-up began day 2 postinjury, genotypes at SNP rs7858819 (SLC1A1) were significantly associated with PTS risk (seizure rates: 52.7% minor allele homozygotes, 11.8% heterozygotes, 21.1% major allele homozygotes; p = 0.002). After adjusting for covariates, we found that rs10974620 remained significant (p = 0.017, minor allele versus major allele homozygotes HR 3.4, 95% confidence interval [CI] 1.3-9.3). rs7858819 also remained significant in adjusted models (p = 0.023, minor allele versus major allele homozygotes HR 3.4, 95%CI 1.1-10.5). SIGNIFICANCE: Variations within SLC1A1 are associated with risk of epileptogenesis following sTBI. Future studies need to confirm findings, but variation within neuronal glutamate transporter genes may represent a possible pharmaceutical target for PTS prevention and treatment.


Asunto(s)
Epilepsia Postraumática/genética , Transportador 3 de Aminoácidos Excitadores/genética , Transportador 4 de Aminoácidos Excitadores/genética , Predisposición Genética a la Enfermedad/genética , Polimorfismo de Nucleótido Simple/genética , Adolescente , Adulto , Anciano , Femenino , Estudios de Seguimiento , Estudios de Asociación Genética , Genotipo , Escala de Coma de Glasgow , Humanos , Masculino , Persona de Mediana Edad , Evaluación de Resultado en la Atención de Salud , Modelos de Riesgos Proporcionales , Estadísticas no Paramétricas , Adulto Joven
12.
Handb Clin Neurol ; 133: 377-403, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27112688

RESUMEN

Neuromyelitis optica (NMO) spectrum disorders (SD) represent an evolving group of central nervous system (CNS)-inflammatory autoimmune demyelinating diseases unified by a pathogenic autoantibody specific for the aquaporin-4 (AQP4) water channel. It was historically misdiagnosed as multiple sclerosis (MS), which lacks a distinguishing biomarker. The discovery of AQP4-IgG moved the focus of CNS demyelinating disease research from emphasis on the oligodendrocyte and myelin to the astrocyte. NMO is recognized today as a relapsing disease, extending beyond the optic nerves and spinal cord to include brain (especially in children) and skeletal muscle. Brain magnetic resonance imaging abnormalities, identifiable in 60% of patients at the second attack, are consistent with MS in 10% of cases. NMOSD-typical lesions (another 10%) occur in AQP4-enriched regions: circumventricular organs (causing intractable nausea and vomiting) and the diencephalon (causing sleep disorders, endocrinopathies, and syndrome of inappropriate antidiuresis). Advances in understanding the immunobiology of AQP4 autoimmunity have necessitated continuing revision of NMOSD clinical diagnostic criteria. Assays that selectively detect pathogenic AQP4-IgG targeting extracellular epitopes of AQP4 are promising prognostically. When referring to AQP4 autoimmunity, we suggest substituting the term "autoimmune aquaporin-4 channelopathy" for the term "NMO spectrum disorders." Randomized clinical trials are currently assessing the efficacy and safety of newer immunotherapies. Increasing therapeutic options based on understanding the molecular pathogenesis is anticipated to improve the outcome for patients with AQP4 channelopathy.


Asunto(s)
Acuaporina 4/inmunología , Autoanticuerpos/metabolismo , Canalopatías/complicaciones , Neuromielitis Óptica/complicaciones , Canalopatías/metabolismo , Transportador 4 de Aminoácidos Excitadores/inmunología , Humanos , Neuromielitis Óptica/metabolismo , PubMed/estadística & datos numéricos
13.
Mol Cell Endocrinol ; 424: 34-41, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26774511

RESUMEN

Genomic profiling of immortalized human mammary epithelial (hTERT-HME1) cells identified several metabolic genes, including the membrane glutamate transporter, SLC1A1, as 1,25-dihydroxyvitamin D3 (1,25D) regulated. In these studies we have surveyed the effects of 1,25D on known glutamate transporters and evaluated its impact on cellular glutamate handling. We confirm that expression of SLC1A1 and all of its known transcript variants are significantly upregulated in hTERT-HME1 cells following 1,25D treatment. Expression of the full-length cognate protein, EAAT3, is correspondingly increased in 1,25D treated hTERT-HME1 cells. Under the same conditions, the expression of two other glutamate transporters--SLC1A6 (EAAT4) and SLC1A2 (EAAT2 or GLT-1)--is enhanced by 1,25D while that of SLC1A3 (EAAT1 or GLAST) and SLC7A11 (xCT) is decreased. Glutamate is not essential for growth of hTERT-HME1 cells, and supplemental glutamate (up to 0.5 mM) does not abrogate the growth inhibitory effects of 1,25D. These data suggest that extracellular glutamate is not a major contributor to cellular energy metabolism in hTERT-HME1 cells under basal conditions and that the growth inhibitory effects of 1,25D are not secondary to its effects on glutamate handling. Instead, the effects of 1,25D on glutamate transporters translated to a decrease in cellular glutamate concentration and an increase in media glutamate concentration, suggesting that one or more of these transporters functions to export glutamate in response to 1,25D exposure. The reduced cellular glutamate concentration may also reflect its incorporation into the cellular glutathione (GSH) pool, which is increased upon 1,25D treatment. In support of this concept, the expression of GCLC (which codes for the rate-limiting enzyme in GSH synthesis) and genes which generate reducing equivalents in the form of NADPH (ie, G6PD, PGD, IDH2) are elevated in 1,25D-treated cells. Taken together, these data identify 1,25D as a physiological regulator of multiple membrane glutamate transporters that impacts on overall cellular glutamate handling.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Transportador 3 de Aminoácidos Excitadores/genética , Transportador 3 de Aminoácidos Excitadores/metabolismo , Ácido Glutámico/farmacología , Glándulas Mamarias Humanas/citología , Vitamina D/análogos & derivados , Sistema de Transporte de Aminoácidos y+/genética , Línea Celular , Medios de Cultivo/farmacología , Células Epiteliales/citología , Células Epiteliales/metabolismo , Transportador 1 de Aminoácidos Excitadores/genética , Transportador 2 de Aminoácidos Excitadores , Transportador 4 de Aminoácidos Excitadores/genética , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas de Transporte de Glutamato en la Membrana Plasmática/genética , Humanos , Glándulas Mamarias Humanas/efectos de los fármacos , Glándulas Mamarias Humanas/metabolismo , Isoformas de Proteínas/metabolismo , Vitamina D/farmacología
14.
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
15.
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
16.
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
17.
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
18.
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
19.
World J Biol Psychiatry ; 14(7): 490-9, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22424243

RESUMEN

OBJECTIVES: The glutamatergic hypothesis of schizophrenia proposes alterations of excitatory amino acid transporters (solute carrier family, SLCs) expression and cerebellar dysfunctions. The influence of the neuregulin-1 (NRG1) risk genotype or effects of antipsychotics on expression of EAATs are unknown. METHODS: We compared post-mortem samples from the cerebellar hemispheres and vermis of 10 schizophrenia patients with nine normal subjects by investigating gene expression of SLC1A3, SLC1A1 and SLC1A6 by in-situ hybridization. We further assessed the allelic composition regarding the polymorphism rs35753505 (SNP8NRG221533) near the NRG1 gene. To control for effects due to antipsychotic treatment, we chronically treated rats with the antipsychotics haloperidol or clozapine and assessed gene expression of SLCs. RESULTS: Schizophrenia patients showed increased expression of SLC1A3 in the molecular layer of the vermis. Individuals carrying at least one C allele of rs35753505 (SNP8NRG221533) showed decreased expression of SLC1A6 in the molecular layer of both hemispheres, compared to individuals homozygous for the T allele. The animal model revealed suppression of SLC1A6 by clozapine. CONCLUSIONS: Increased SLC1A3 expression indicates facilitated transport and may result in reduced glutamate neurotransmission. Decreased SLC1A6 expression in NRG1 risk variant may be an adaptive effect to restore glutamate signalling, but treatment effects cannot be excluded.


Asunto(s)
Cerebelo/metabolismo , Transportador 1 de Aminoácidos Excitadores/genética , Transportador 3 de Aminoácidos Excitadores/genética , Transportador 4 de Aminoácidos Excitadores/genética , Regulación de la Expresión Génica/genética , Esquizofrenia/metabolismo , Anciano , Alelos , Animales , Antipsicóticos/administración & dosificación , Antipsicóticos/uso terapéutico , Cerebelo/efectos de los fármacos , Cerebelo/patología , Clozapina/uso terapéutico , Modelos Animales de Enfermedad , Transportador 1 de Aminoácidos Excitadores/biosíntesis , Transportador 3 de Aminoácidos Excitadores/biosíntesis , Transportador 4 de Aminoácidos Excitadores/biosíntesis , Femenino , Haloperidol/uso terapéutico , Humanos , Masculino , Persona de Mediana Edad , Neurregulina-1/genética , Polimorfismo de Nucleótido Simple/efectos de los fármacos , Polimorfismo de Nucleótido Simple/genética , Ratas , Ratas Sprague-Dawley , Factores de Riesgo , Esquizofrenia/tratamiento farmacológico , Esquizofrenia/genética
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA