Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 107
1.
Exp Neurol ; 262 Pt B: 111-20, 2014 Dec.
Article En | MEDLINE | ID: mdl-24859452

For more than twenty years glial cells have been implicated in the pathogenetic cascades for genetic and sporadic forms of ALS. The biological role of glia, including the principal CNS glia, astroglia and oligodendroglia, as well as the myeloid derived microglia, has uniformly led to converging data sets that implicate these diverse cells in the degeneration of neurons in ALS. Originating as studies in postmortem human brain implicating astroglia, the research progressed to strongly implicate microglia and contributors to CNS injury in all forms of ALS. Most recently and unexpectedly, oligodendroglia have also been shown in animal model systems and human brain to play an early role in the dysfunction and death of ALS neurons. These studies have identified a number of diverse cellular cascades that could be, or have already been, the target of therapeutic interventions. Understanding the temporal and regional role of these cells and the magnitude of their contribution will be important for future interventions. Employing markers of these cell types may also allow for future important patient subgrouping and pharmacodynamic drug development tools.


Amyotrophic Lateral Sclerosis/pathology , Neuroglia/pathology , Animals , Humans
2.
J Cell Mol Med ; 13(1): 114-24, 2009 Jan.
Article En | MEDLINE | ID: mdl-18363836

Glutamate transporter associated protein 3-18 (GTRAP3-18) is an endoplasmic reticulum (ER)-localized protein belonging to the prenylated rab-acceptor-family interacting with small Rab GTPases, which regulate intracellular trafficking events. Its impact on secretory trafficking has not been investigated. We report here that GTRAP3-18 has an inhibitory effect on Rab1, which is involved in ER-to-Golg trafficking. The effects on the early secretory pathway in HEK293 cells were: reduction of the rate of ER-to-Golgi transport of the vesicular stomatitis virus glycoprotein (VSVG), slowed accumulation of a Golgi marker plasmid in pre-Golgi structures after Brefeldin A treatment and inhibition of cargo concentration of the neuronal glutamate transporter excitatory amino-acid carrier 1 (EAAC1) into transpor complexes in HEK293 cells, an effect that could be completely reversed in the presence of an excess of Rab1. In accordance with the known role of Rab1 in neurite formation, overexpression of GTRAP3-18 significantly inhibited the length of outgrowing neurites in differentiated CAD cells. The inhibitory effect of GTRAP3-18 on neurite growth was rescued by co-expression with Rab1, supporting the conclusion that GTRAP 3-18 acted by inhibiting Rab1 action. Finally, we hypothesized that expression of GTRAP3-18 in the brain shoul be lower at stages of active synaptogenesis compared to early developmental stages. This was the case as expression of GTRAP3-18 declined from E17 to P0 and adult rat brains. Thus, we propose a model where protein trafficking and neuronal differentiation are directly linked by the interaction of Rab1 and its regulator GTRAP3-18.


Carrier Proteins/metabolism , Cell Differentiation/physiology , Neurons/physiology , Protein Transport/physiology , rab1 GTP-Binding Proteins/metabolism , Animals , Biomarkers/metabolism , Brefeldin A/metabolism , Carrier Proteins/genetics , Cell Line , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Humans , Neurons/cytology , Protein Synthesis Inhibitors/metabolism , RNA Interference , Rats , Rats, Sprague-Dawley , rab1 GTP-Binding Proteins/genetics
3.
Neuroscience ; 152(4): 1086-92, 2008 Apr 09.
Article En | MEDLINE | ID: mdl-18358622

We have previously shown that the atypical methylxanthine, propentofylline, reduces mechanical allodynia after peripheral nerve transection in a rodent model of neuropathy. In the present study, we sought to determine whether propentofylline-induced glial modulation alters spinal glutamate transporters, glutamate transporter-1 (GLT-1) and glutamate-aspartate transporter (GLAST) in vivo, which may contribute to reduced behavioral hypersensitivity after nerve injury. In order to specifically examine the expression of the spinal glutamate transporters, a novel line of double transgenic GLT-1-enhanced green fluorescent protein (eGFP)/GLAST-Discosoma Red (DsRed) promoter mice was used. Adult mice received propentofylline (10 mg/kg) or saline via i.p. injection starting 1 h prior to L5-spinal nerve transection and then daily for 12 days. Mice receiving saline exhibited punctate expression of both eGFP (GLT-1 promoter activation) and DsRed (GLAST promoter activation) in the dorsal horn of the spinal cord, which was decreased ipsilateral to nerve injury on day 12. Propentofylline administration reinstated promoter activation on the injured side as evidenced by an equal number of eGFP (GLT-1) and DsRed (GLAST) puncta in both dorsal horns. As demonstrated in previous studies, propentofylline induced a concomitant reversal of L5 spinal nerve transection-induced expression of glial fibrillary acidic protein (GFAP). The ability of propentofylline to alter glial glutamate transporters highlights the importance of controlling aberrant glial activation in neuropathic pain and suggests one possible mechanism for the anti-allodynic action of this drug.


Astrocytes/drug effects , Excitatory Amino Acid Transporter 1/metabolism , Neuroprotective Agents/pharmacology , Spinal Cord Injuries/pathology , Xanthines/pharmacology , Animals , Excitatory Amino Acid Transporter 1/genetics , Excitatory Amino Acid Transporter 2/metabolism , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/metabolism , Hyperalgesia/physiopathology , Hyperalgesia/prevention & control , Indoles , Mice , Mice, Inbred C57BL , Mice, Transgenic , Time Factors
4.
Handb Exp Pharmacol ; (175): 277-303, 2006.
Article En | MEDLINE | ID: mdl-16722241

Glutamate is the primary excitatory neurotransmitter in the central nervous system. During synaptic activity, glutamate is released into the synaptic cleft and binds to glutamate receptors on the pre- and postsynaptic membrane as well as on neighboring astrocytes in order to start a number of intracellular signaling cascades. To allow for an efficient signaling to occur, glutamate levels in the synaptic cleft have to be maintained at very low levels. This process is regulated by glutamate transporters, which remove excess extracellular glutamate via a sodium-potassium coupled uptake mechanism. When extracellular glutamate levels rise to about normal, glutamate overactivates glutamate receptors, triggering a multitude of intracellular events in the postsynaptic neuron, which ultimately results in neuronal cell death. This phenomenon is known as excitotoxicity and is the underlying mechanisms of a number of neurodegenerative diseases. A dysfunction of the glutamate transporter is thought to contribute to cell death during excitotoxicity. Therefore, efforts have been made to understand the regulation of glutamate transporter function. Transporter activity can be regulated in different ways, including through gene expression, transporter protein targeting and trafficking and through posttranslational modifications of the transporter protein. The identification of these mechanisms has helped to understand the role of glutamate transporters during pathology and will aid in the development of therapeutic strategies with the transporter as a desirable target.


Excitatory Amino Acid Transporter 1/metabolism , Excitatory Amino Acid Transporter 2/metabolism , Glutamate Plasma Membrane Transport Proteins/metabolism , Protein Processing, Post-Translational , Animals , Brain/metabolism , Excitatory Amino Acid Transporter 1/genetics , Excitatory Amino Acid Transporter 2/genetics , Gene Expression Regulation , Glutamate Plasma Membrane Transport Proteins/genetics , Glutamic Acid/metabolism , Humans , Neurodegenerative Diseases/metabolism , Presynaptic Terminals/metabolism , Protein Transport , Transcription, Genetic
5.
Neuroscience ; 123(4): 1045-51, 2004.
Article En | MEDLINE | ID: mdl-14751295

The expression and distribution of the neuronal glutamate transporter, excitatory amino acid carrier-1 (EAAC1), are demonstrated in the dorsal root ganglion neurons and their central terminals. Reverse transcriptase-polymerase chain reaction shows expression of EAAC1 mRNA in the dorsal root ganglion. Immunoblotting analysis further confirms existence of EAAC1 protein in this region. Immunocytochemistry reveals that approximately 46.6% of the dorsal root ganglion neurons are EAAC1-positive. Most EAAC1-positive neurons are small and around 250-750 microm2 in surface area, and some co-label with calcitonin gene-related peptide (CGRP) or isolectin IB4. In the spinal cord, EAAC-1 immunoreactive small dot- or patch-like structures are mainly localized in the superficial dorsal horn, and some are positive for CGRP or labeled by isolectin IB4. Unilateral dorsal rhizotomy experiments further show that EAAC1 immunoreactivity is less intense in superficial dorsal horn on the side ipsilateral to the dorsal rhizotomy than on the contralateral side. The results indicate the presence of EAAC1 in the dorsal root ganglion neurons and their central terminals. Our findings suggest that EAAC1 might play an important role in transmission and modulation of nociceptive information via the regulation of pre-synaptically released glutamate.


Amino Acid Transport System X-AG/metabolism , Ganglia, Spinal/cytology , Neurons/metabolism , Symporters/metabolism , Amino Acid Transport System X-AG/genetics , Animals , Blotting, Western/methods , Calcitonin Gene-Related Peptide/metabolism , Cell Count , Excitatory Amino Acid Transporter 3 , Functional Laterality , Glutamate Plasma Membrane Transport Proteins , Immunohistochemistry/methods , Lectins/metabolism , Male , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction/methods , Rhizotomy/methods , Spinal Cord/metabolism , Symporters/genetics
6.
Neurology ; 61(4): 456-64, 2003 Aug 26.
Article En | MEDLINE | ID: mdl-12939417

OBJECTIVE: To determine if long-term topiramate therapy is safe and slows disease progression in patients with ALS. METHODS: A double-blind, placebo-controlled, multicenter randomized clinical trial was conducted. Participants with ALS (n = 296) were randomized (2:1) to receive topiramate (maximum tolerated dose up to 800 mg/day) or placebo for 12 months. The primary outcome measure was the rate of change in upper extremity motor function as measured by the maximum voluntary isometric contraction (MVIC) strength of eight arm muscle groups. Secondary endpoints included safety and the rate of decline of forced vital capacity (FVC), grip strength, ALS functional rating scale (ALSFRS), and survival. RESULTS: Patients treated with topiramate showed a faster decrease in arm strength (33.3%) during 12 months (0.0997 vs 0.0748 unit decline/month, p = 0.012). Topiramate did not significantly alter the decline in FVC and ALSFRS or affect survival. Topiramate was associated with an increased frequency of anorexia, depression, diarrhea, ecchymosis, nausea, kidney calculus, paresthesia, taste perversion, thinking abnormalities, weight loss, and abnormal blood clotting (pulmonary embolism and deep venous thrombosis). CONCLUSIONS: At the dose studied, topiramate did not have a beneficial effect for patients with ALS. High-dose topiramate treatment was associated with a faster rate of decline in muscle strength as measured by MVIC and with an increased risk for several adverse events in patients with ALS. Given the lack of efficacy and large number of adverse effects, further studies of topiramate at a dose of 800 mg or maximum tolerated dose up to 800 mg/day are not warranted.


Amyotrophic Lateral Sclerosis/drug therapy , Fructose/analogs & derivatives , Fructose/therapeutic use , Adult , Aged , Amyotrophic Lateral Sclerosis/mortality , Disease Progression , Double-Blind Method , Female , Fructose/adverse effects , Fructose/pharmacology , Hand Strength , Humans , Life Tables , Male , Middle Aged , Muscle Contraction/drug effects , Proportional Hazards Models , Safety , Survival Analysis , Thromboembolism/chemically induced , Topiramate , Treatment Failure , Vital Capacity/drug effects
7.
Neuroscience ; 116(1): 81-7, 2003.
Article En | MEDLINE | ID: mdl-12535941

GLT-1 is the predominant glutamate transporter in most brain regions and therefore plays a major role in terminating synaptic transmission and protecting neurons from glutamate neurotoxicity. In the present study we assessed (i) the regulation of GLT-1 expression in the spinal cord after peripheral nociceptive stimulation and (ii) the nociceptive behavior of rats following inhibition or transient knockdown of spinal GLT-1. Formalin injection into one hindpaw caused a rapid transient upregulation of GLT-1 protein expression in the spinal cord which did not occur when rats were pretreated with morphine (10 mg/kg, i.p.) suggesting that the nociceptive input specifically caused the increase of GLT-1 transcription. Inhibition of GLT-1 by the transportable inhibitor trans-pyrrolidine-2,4-dicarboxylic acid resulted in a significant reduction of nociceptive behavior in the rat formalin assay. Similar results were obtained with a transient reduction of GLT-1 protein expression by antisense oligonucleotides. These data suggest that inhibition of GLT-1 activity or expression reduces excitatory synaptic efficacy and thereby nociception. Mechanisms that might explain this phenomenon may include activation of inhibitory metabotropic glutamate receptors, postsynaptic desensitization or disturbance of glutamate recycling.


Excitatory Amino Acid Transporter 2/metabolism , Pain/metabolism , Spinal Cord/metabolism , Analgesics, Opioid/pharmacology , Animals , Blotting, Western , Cell Culture Techniques , Dicarboxylic Acids/pharmacology , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Formaldehyde , Male , Morphine/pharmacology , Neurotransmitter Uptake Inhibitors/pharmacology , Oligoribonucleotides, Antisense/pharmacology , Pain/chemically induced , Pyrrolidines/pharmacology , Rats , Rats, Sprague-Dawley , Up-Regulation
8.
Brain Res Dev Brain Res ; 132(2): 131-9, 2001 Dec 31.
Article En | MEDLINE | ID: mdl-11744117

Glutamate transporters are essential for maintaining the extracellular levels of glutamate at synaptic clefts and are regulated developmentally in a subtype-specific manner. We investigated chronological changes of immunoreactivities for glial glutamate transporters GLAST and GLT-1 and a neuronal glutamate transporter, EAAC1, in postnatal 7-day-old rat neocortices and hippocampi at 12, 24, 48 and 72 h after hypoxia-ischemia. Glutamate transporter subtypes are differentially expressed in the ischemic core and the boundary area of the neonatal rat brain with hypoxia-ischemia. Expressions of these glutamate transporters decreased in the ischemic core at 12 h, then immunoreactivities for GLAST and GLT-1 were recovered at the hippocampus. This was accompanied by a GFAP-positive gliosis at 72 h, whereas these immunoreactivities were reduced at the neocortex in the ischemic core. Glial glutamate transporters, especially GLAST, were noted in some astrocytes appearing as apoptosis as well as shrunken pyramidal neurons mainly in the boundary area of the neocortex. Increased perikaryal expression of EAAC1 was associated with that of MAP2 at the border of the boundary area. These temporal and regional expressions of glutamate transporters may contribute towards understanding the excitotoxic cell death mechanism in hypoxic-ischemic encephalopathy during the perinatal period.


Amino Acid Transport System X-AG/biosynthesis , Excitatory Amino Acid Transporter 2/biosynthesis , Hypoxia-Ischemia, Brain/metabolism , Symporters , Amino Acid Transport System X-AG/analysis , Animals , Animals, Newborn , Apoptosis , Brain/growth & development , Brain/metabolism , Carrier Proteins/analysis , Carrier Proteins/biosynthesis , Disease Models, Animal , Excitatory Amino Acid Transporter 1 , Excitatory Amino Acid Transporter 2/analysis , Excitatory Amino Acid Transporter 3 , Glial Fibrillary Acidic Protein/analysis , Gliosis/metabolism , Gliosis/pathology , Glutamate Plasma Membrane Transport Proteins , Glutamic Acid/metabolism , Hypoxia-Ischemia, Brain/pathology , Immunohistochemistry , Rats , Rats, Wistar
9.
Neurobiol Dis ; 8(6): 933-41, 2001 Dec.
Article En | MEDLINE | ID: mdl-11741389

The mechanisms leading to neurodegeneration in ALS (amyotrophic lateral sclerosis) are not well understood, but cytosolic protein aggregates appear to be common in sporadic and familial ALS as well as transgenic mouse models expressing mutant Cu/Zn superoxide dismutase (SOD1). In this study, we systematically evaluated the presence of these aggregates in three different mouse models (G93A, G85R, and G37R SOD1) and compared these aggregates to those seen in cases of sporadic and familial ALS. Inclusions and loss of motor neurons were observed in spinal cords of all of these three mutant transgenic lines. Since a copper-mediated toxicity hypothesis has been proposed to explain the cytotoxic gain-of-function of mutant SOD1, we sought to determine the involvement of the copper chaperone for SOD1 (CCS) in the formation of protein aggregates. Although all aggregates contained CCS, SOD1 was not uniformly found in the inclusions. Similarly, CCS-positive skein-like inclusions were rarely seen in ALS neurons. These studies do not provide strong evidence for a causal role of CCS in aggregate formation, but they do suggest that protein aggregation is a common event in all animal models of the disease. Selected proteins, such as the glutamate transporter GLT-1, were not typically observed within the inclusions. Most inclusions were positively stained with antibodies recognizing ubiquitin, proteasome, Hsc70 in transgenic lines, and some Hsc70-positive inclusions were detected in sporadic ALS cases. Overall, these observations suggest that inclusions might be sequestered into ubiquitin-proteasome pathway and some chaperone proteins such as Hsc70 may be involved in formation and/or degradation of these inclusions.


Amyotrophic Lateral Sclerosis/pathology , Central Nervous System/pathology , Inclusion Bodies/genetics , Molecular Chaperones/genetics , Motor Neurons/pathology , Nerve Tissue Proteins/metabolism , Superoxide Dismutase/genetics , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Animals , Brain Stem/metabolism , Brain Stem/pathology , Brain Stem/physiopathology , Carrier Proteins/metabolism , Central Nervous System/metabolism , Central Nervous System/physiopathology , Copper/metabolism , Cysteine Endopeptidases/metabolism , Female , Glial Fibrillary Acidic Protein/metabolism , Heat-Shock Proteins/metabolism , Humans , Immunohistochemistry , Inclusion Bodies/metabolism , Male , Mice , Mice, Neurologic Mutants , Mice, Transgenic , Molecular Chaperones/metabolism , Motor Neurons/metabolism , Multienzyme Complexes/metabolism , Nerve Tissue Proteins/genetics , Proteasome Endopeptidase Complex , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Cord/physiopathology , Superoxide Dismutase-1 , Ubiquitins/metabolism
11.
Neurosci Lett ; 311(3): 161-4, 2001 Oct 05.
Article En | MEDLINE | ID: mdl-11578819

The synaptic distribution of alpha-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid receptor subunit GluR2 and neuronal glutamate transporter subunit EAAC1 were studied using immunogold in layer II of the macaque monkey entorhinal cortex. Immunoreactivity for EAAC1 and GluR2 was frequent at asymmetric synapses and their associated membrane. The synaptic localization of EAAC1 differed considerably from that of GluR2, in that GluR2 immunolabelling was most commonly located within the postsynaptic density, but EAAC1 localization was more heterogeneous and was predominant at the edge of postsynaptic densities and perisynaptic zones. Since EAAC1 may play an important role in clearing glutamate from the synaptic cleft and intercellular spaces, the high perisynaptic expression of EAAC1 in these neurons could presumably offer a powerful mechanism through which high concentrations of glutamate could be efficiently removed from the synapses following release and interaction with glutamate receptors. The distribution of EAAC1 may also offer protection for these neurons against excessive glutamatergic stimuli that may occur under certain pathological conditions.


Amino Acid Transport System X-AG , Carrier Proteins/metabolism , Entorhinal Cortex/metabolism , Macaca fascicularis/metabolism , Neurons/metabolism , Receptors, AMPA/metabolism , Symporters , Synaptic Membranes/metabolism , Animals , Entorhinal Cortex/ultrastructure , Glutamate Plasma Membrane Transport Proteins , Glutamic Acid/metabolism , Immunohistochemistry , Macaca fascicularis/anatomy & histology , Male , Microscopy, Electron , Neurons/ultrastructure , Synaptic Membranes/ultrastructure , Synaptic Transmission/physiology
13.
Mol Psychiatry ; 6(4): 380-6, 2001 Jul.
Article En | MEDLINE | ID: mdl-11443521

We show here that clozapine, a beneficial antipsychotic, down-regulates the expression of the glutamate transporter GLT-1 in the rat cerebral cortex, thereby reducing glutamate transport and raising extracellular glutamate levels. Clozapine treatment (25--35 mg kg(-1) day(-1) orally) reduced GLT-1 immunoreactivity in several brain regions after 3 weeks; this effect was most prominent after 9 weeks and most evident in the frontal cortex. GLT-1 protein levels were reduced in the cerebral cortex of treated rats compared with controls and were more severely affected in the anterior (71.9 +/- 4.5%) than in the posterior (53.2 +/- 15.4%) cortex. L-[(3)H]-glutamate uptake in Xenopus laevis oocytes injected with mRNA extracted from the anterior cerebral cortex of rats treated for 9 weeks was remarkably reduced (to 30.6 +/- 8.6%) as compared to controls. In addition, electrophysiological recordings from oocytes following application of glutamate revealed a strong reduction in glutamate uptake currents (46.3 +/- 10.2%) as compared to controls. Finally, clozapine treatment led to increases in both the mean basal (8.1 +/- 0.7 microM) and the KCl-evoked (28.7 +/- 7.7 microM) output of glutamate that were 3.1 and 3.5, respectively, higher than in control rats. These findings indicate that clozapine may potentiate glutamatergic synaptic transmission by regulating glutamate transport.


ATP-Binding Cassette Transporters/genetics , Cerebral Cortex/metabolism , Clozapine/pharmacology , Frontal Lobe/metabolism , Gene Expression Regulation/drug effects , Glutamic Acid/metabolism , ATP-Binding Cassette Transporters/analysis , Amino Acid Transport System X-AG , Animals , Antipsychotic Agents/pharmacology , Cerebral Cortex/drug effects , Female , Frontal Lobe/drug effects , In Vitro Techniques , Microdialysis , Oocytes/drug effects , Oocytes/physiology , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Recombinant Proteins/metabolism , Xenopus laevis
15.
Nature ; 410(6824): 84-8, 2001 Mar 01.
Article En | MEDLINE | ID: mdl-11242046

Excitatory amino-acid carrier 1 (EAAC1) is a high-affinity Na+-dependent L-glutamate/D,L-aspartate cell-membrane transport protein. It is expressed in brain as well as several non-nervous tissues. In brain, EAAC1 is the primary neuronal glutamate transporter. It has a polarized distribution in cells and mainly functions perisynaptically to transport glutamate from the extracellular environment. In the kidney it is involved in renal acidic amino-acid re-absorption and amino-acid metabolism. Here we describe the identification and characterization of an EAAC1-associated protein, GTRAP3-18. Like EAAC1, GTRAP3-18 is expressed in numerous tissues. It localizes to the cell membrane and cytoplasm, and specifically interacts with carboxy-terminal intracellular domain of EAAC1. Increasing the expression of GTRAP3-18 in cells reduces EAAC1-mediated glutamate transport by lowering substrate affinity. The expression of GTRAP3-18 can be upregulated by retinoic acid, which results in a specific reduction of EAAC1-mediated glutamate transport. These studies show that glutamate transport proteins can be regulated potently and that GTRAP can modulate the transport functions ascribed to EAAC1. GTRAP3-18 may be important in regulating the metabolic function of EAAC1.


ATP-Binding Cassette Transporters/metabolism , Carrier Proteins/metabolism , Glutamic Acid/metabolism , Proteins/metabolism , Symporters , Amino Acid Transport System X-AG , Animals , Biological Transport/drug effects , Brain/metabolism , Cell Line , Cloning, Molecular , Excitatory Amino Acid Transporter 3 , Glutamate Plasma Membrane Transport Proteins , Membrane Proteins/metabolism , Molecular Sequence Data , Precipitin Tests , Protein Binding , Protein Kinase C/genetics , Protein Kinase C/metabolism , Proteins/genetics , Rats , Recombinant Fusion Proteins/metabolism , Tissue Distribution , Tretinoin/pharmacology
16.
Nature ; 410(6824): 89-93, 2001 Mar 01.
Article En | MEDLINE | ID: mdl-11242047

Glutamate is the main excitatory neurotransmitter in the mammalian central nervous system and is removed from the synaptic cleft by sodium-dependent glutamate transporters. To date, five distinct glutamate transporters have been cloned from animal and human tissue: GLAST (EAAT1), GLT-1 (EAAT2), EAAC1 (EAAT3), EAAT4, and EAAT5 (refs 1-5). GLAST and GLT-1 are localized primarily in astrocytes, whereas EAAC1 (refs 8, 9), EAAT4 (refs 9-11) and EAAT5 (ref 5) are neuronal. Studies of EAAT4 and EAAC1 indicate an extrasynaptic localization on perisynaptic membranes that are near release sites. This localization facilitates rapid glutamate binding, and may have a role in shaping the amplitude of postsynaptic responses in densely packed cerebellar terminals. We have used a yeast two-hybrid screen to identify interacting proteins that may be involved in regulating EAAT4--the glutamate transporter expressed predominately in the cerebellum--or in targeting and/or anchoring or clustering the transporter to the target site. Here we report the identification and characterization of two proteins, GTRAP41 and GTRAP48 (for glutamate transporter EAAT4 associated protein) that specifically interact with the intracellular carboxy-terminal domain of EAAT4 and modulate its glutamate transport activity.


Amino Acid Transport System X-AG , Glutamic Acid/metabolism , Neurons/metabolism , Receptors, Glutamate/metabolism , Spectrin/metabolism , Symporters , Amino Acid Sequence , Animals , Biological Transport , Blotting, Northern , Blotting, Western , Cell Line , Cloning, Molecular , Excitatory Amino Acid Transporter 1 , Excitatory Amino Acid Transporter 2 , Excitatory Amino Acid Transporter 3 , Excitatory Amino Acid Transporter 4 , Glutamate Plasma Membrane Transport Proteins , Guanine Nucleotide Exchange Factors , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Molecular Sequence Data , Precipitin Tests , Protein Binding , Rats , Receptors, Glutamate/genetics , Rho Guanine Nucleotide Exchange Factors , Sequence Homology, Amino Acid , Spectrin/genetics , Two-Hybrid System Techniques
17.
J Neurosci ; 21(6): 1876-83, 2001 Mar 15.
Article En | MEDLINE | ID: mdl-11245672

Transient focal cerebral ischemia leads to extensive neuronal damage in cerebral cortex and striatum. Normal functioning of glutamate transporters clears the synaptically released glutamate to prevent excitotoxic neuronal death. This study evaluated the functional role of the glial (GLT-1) and neuronal (EAAC1) glutamate transporters in mediating ischemic neuronal damage after transient middle cerebral artery occlusion (MCAO). Transient MCAO in rats infused with GLT-1 antisense oligodeoxynucleotides (ODNs) led to increased infarct volume (45 +/- 8%; p < 0.05), worsened neurological status, and increased mortality rate, compared with GLT-1 sense/random ODN-infused controls. Transient MCAO in rats infused with EAAC1 antisense ODNs had no significant effect on any of these parameters. This study suggests that GLT-1, but not EAAC1, knockdown exacerbates the neuronal death and thus neurological deficit after stroke.


ATP-Binding Cassette Transporters/antagonists & inhibitors , Brain/metabolism , Carrier Proteins/antagonists & inhibitors , Ischemic Attack, Transient/metabolism , Oligonucleotides, Antisense/pharmacology , Symporters , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Amino Acid Transport System X-AG , Animals , Blood Flow Velocity/drug effects , Brain/pathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cerebral Cortex/blood supply , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cerebrovascular Circulation/drug effects , Corpus Striatum/blood supply , Corpus Striatum/metabolism , Corpus Striatum/pathology , Disease Progression , Excitatory Amino Acid Transporter 3 , Glutamate Plasma Membrane Transport Proteins , Glutamic Acid/metabolism , Infarction, Middle Cerebral Artery , Ischemic Attack, Transient/pathology , Male , Neuroglia/metabolism , Neuroglia/pathology , Neurons/metabolism , Neurons/pathology , Rats , Rats, Inbred SHR , Survival Rate
18.
Int J Dev Neurosci ; 19(1): 47-52, 2001 Feb.
Article En | MEDLINE | ID: mdl-11226754

Depletion of noradrenaline in newborn rats by 6-hydroxydopamine (6-OHDA) affects the postnatal development and reduces the granular cell area in the neocerebellum (lobules V-VII). During the first postnatal month, Bergmann glial fibers guide the migration of immature granule cells to the internal granule cell layer. Microglia and Bergmann glia may play an important role in this process, but the exact mechanism behind this phenomenon is not known. We studied the effect of systemic administration of 6-OHDA on the expression and localization on microglia and Bergmann glia in the neonatal cerebellum by immunohistochemistry. In the neocerebellum, 6-OHDA treatment caused a significant increase in the number of activated microglia. The increase was observed mainly in the granule cell layer and the cerebellar medulla. Bergmann glial cells in treated brains were abnormally located, did not form intimate associations with Purkinje cells, and the glial fibers were structurally different. Our findings indicate that a noradrenergic influence may be necessary for the normal maturation and migration of granule cells, and abnormal migration may be the result of Bergmann glia destruction and the activation of microglia. Activated microglia in the granule cell layer may be used as a marker for an injured cerebellar area.


Animals, Newborn/growth & development , Cell Differentiation/physiology , Cell Movement/physiology , Cerebellum/growth & development , Microglia/metabolism , Neurons/metabolism , Norepinephrine/metabolism , Plant Lectins , Age Factors , Animals , Animals, Newborn/anatomy & histology , Animals, Newborn/metabolism , Biomarkers/analysis , Cell Differentiation/drug effects , Cell Movement/drug effects , Cerebellum/cytology , Cerebellum/metabolism , Lectins/pharmacokinetics , Microglia/cytology , Microglia/drug effects , Neurons/cytology , Neurons/drug effects , Neurotoxins/pharmacology , Oxidopamine/pharmacology , Rats , Rats, Sprague-Dawley , Sympatholytics/pharmacology
19.
Ann Neurol ; 48(5): 792-5, 2000 Nov.
Article En | MEDLINE | ID: mdl-11079544

The pathogenesis of motor neuron loss in amyotrophic lateral sclerosis (ALS) is thought to involve both glutamate-mediated excitotoxicity and oxidative damage due to the accumulation of free radicals and other toxic molecules. Cyclooxygenase-2 (COX-2) may play a key role in these processes by producing prostaglandins, which trigger astrocytic glutamate release, and by inducing free radical formation. We tested the effects of COX-2 inhibition in an organotypic spinal cord culture model of ALS. The COX-2 inhibitor (SC236) provided significant protection against loss of spinal motor neurons in this system, suggesting that it may be useful in the treatment of ALS.


Amyotrophic Lateral Sclerosis/metabolism , Isoenzymes/metabolism , Motor Neurons/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Animals , Cyclooxygenase 2 , Disease Models, Animal , Organ Culture Techniques , Pyrazoles/metabolism , Rats , Spinal Cord/metabolism , Sulfonamides/metabolism
...