51
|
Gui SB, Zhang YZ, Sun MZ, Wang HY, He Y, Li D. Effect of bone marrow stromal cell-conditioned medium on the glutamate uptake of peroxide-injured astrocytes. J Clin Neurosci 2009; 16:1205-10. [PMID: 19589682 DOI: 10.1016/j.jocn.2008.11.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2008] [Revised: 09/24/2008] [Accepted: 11/02/2008] [Indexed: 11/28/2022]
Abstract
We aimed to investigate the effect of bone marrow stromal cell-conditioned medium (BCM) on glutamate uptake of peroxide (H(2)O(2))-injured astrocytes. Bone marrow stromal cells (BMSC) were isolated from rat bone marrow. Confluent BMSC cultures were incubated with serum-free Dulbecco's Modified Eagle's Medium to create the BCM. Astrocytes were isolated from 1-day-old rats. H(2)O(2)-injured astrocytes were cultured in BCM (experimental group) or serum-free medium (control group). The labeled glutamate ((3)H-L-glutamate) uptake by H(2)O(2)-injured astrocytes with or without BCM was compared after 1 and 3 days. We found that astrocytes cultured in BCM exhibited increased glutamate uptake compared to those cultured in serum-free medium following H(2)O(2)-induced injury (p<0.01) and concluded that BCM increased the glutamate uptake capability of H(2)O(2)-injured rat astrocytes. The therapeutic benefits associated with BMSC transplantation following brain injury might be partly due to increased glutamate uptake by astrocytes.
Collapse
Affiliation(s)
- Song-Bai Gui
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | | | | | | | | | | |
Collapse
|
52
|
Kabakov AY, Rosenberg PA. Evidence for change in current-flux coupling of GLT1 at high glutamate concentrations in rat primary forebrain neurons and GLT1a-expressing COS-7 cells. Eur J Neurosci 2009; 30:186-95. [PMID: 19614985 DOI: 10.1111/j.1460-9568.2009.06809.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Glutamate is the major excitatory neurotransmitter of the central nervous system and is toxic to neurons even at low concentrations. GLT1, the rodent analog of human EAAT2, is primarily responsible for glutamate clearance in the cerebrum. GLT1 was thought to be expressed exclusively in astrocytes in the mature brain. Recently, however, GLT1a was demonstrated in excitatory axon terminals where synaptic glutamate concentration rises above 1 mm during excitatory transmission. GLT1 function in neurons with accurate control of both intracellular and extracellular solutions mimicking synaptic concentration gradients has never been studied. Here we characterized the kinetics of coupled glutamate transporter current in whole-cell configuration and [(3)H]-l-glutamate uptake in cultured rat cerebral neurons across the entire range of synaptic glutamate concentrations. In both neurons and GLT1a-transfected COS-7 cells, the kinetics were similar and revealed two specific components: a high-affinity component with glutamate k(D) value around 15 mum and a low-affinity component with k(D) value around 0.2 mm. The specific low-affinity component was discovered as a result of significant deviation of the transporter current from Michaelis-Menten kinetics in the 100-300 mum concentration range. Activation of the specific low-affinity component led to a two-fold decrease in the current/flux ratio, implying a change in the transport coupling. Our data indicate that GLT1 endogenously expressed in cultured rat forebrain neurons displays high and low glutamate affinity uptake components that are different in current-flux coupling ratios. This property is intrinsic to the protein because it was also observed in GLT1a-transfected COS-7 cells.
Collapse
Affiliation(s)
- Anatoli Y Kabakov
- FM Kirby Neurobiology Center and Department of Neurology, Children's Hospital Boston, Boston, MA, USA
| | | |
Collapse
|
53
|
Holten AT, Talgøy HA, Danbolt NC, Christian DN, Shimamoto K, Gundersen V, Vidar G. Low-affinity excitatory amino acid uptake in hippocampal astrocytes: a possible role of Na+/dicarboxylate cotransporters. Glia 2008; 56:990-7. [PMID: 18442087 DOI: 10.1002/glia.20672] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The excitatory amino acid transporters (EAATs) underlie the so-called "high affinity" uptake of glutamate, which is well characterized. In contrast, the "low-affinity" uptake of glutamate remains poorly defined, and it has been discussed whether it may represent a mere in vitro artifact. Here we have visualized "low-affinity" excitatory amino acid uptake sites by incubating rat hippocampal slices with the glutamate analogue D-aspartate in the presence of PMB-TBOA, which blocks the EAATs. After fixation of the slices, D-aspartate taken up into the tissue was localized with the use of light microscopic immunoperoxidase and electron microscopic immunogold methods, exploiting highly specific antibodies against D-aspartate. PMB-TBOA blocked uptake of both low and high exogenous D-aspartate concentrations (0.01-1.0 mM) into nerve terminals, as well as the uptake of 0.01 mM D-aspartate into astrocytes. Interestingly, there was a residual PMB-TBOA insensitive uptake of D-aspartate in astrocytes at higher exogenous D-aspartate concentrations (0.05-1.0 mM), strongly suggesting that astrocytes have "low-affinity" uptake sites for excitatory amino acid. The PMB-TBOA insensitive D-aspartate uptake in astrocytes was sodium dependent and inhibited by succinate and to certain extent by homocysteate, but not by cystine or DIDS. We suggest that excitatory amino acid is transported into astrocytes in a "low-affinity" fashion by sodium/dicarboxylate transporters.
Collapse
|
54
|
Alekseenko AV, Waseem TV, Fedorovich SV. Ferritin, a protein containing iron nanoparticles, induces reactive oxygen species formation and inhibits glutamate uptake in rat brain synaptosomes. Brain Res 2008; 1241:193-200. [PMID: 18835382 DOI: 10.1016/j.brainres.2008.09.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2008] [Revised: 09/01/2008] [Accepted: 09/03/2008] [Indexed: 12/28/2022]
Abstract
Nanoparticles are currently used in medicine as agents for targeted drug delivery and imaging. However it has been demonstrated that nanoparticles induce neurodegeneration in vivo and kill neurons in vitro. The cellular and molecular bases of this phenomenon are still unclear. We have used the protein ferritin as a nanoparticle model. Ferritin contains iron particles (Fe(3+)) with size 7 nm and a protein shell. We investigated how ferritin influences uptake and release of [(14)C]glutamate and free radical formation as monitored by fluorescent dye DCFDA in rat brain synaptosomes. We found that even a high concentration of ferritin (800 microg/ml) did not induce spontaneous [(14)C]glutamate release. In contrast the same concentration of this protein inhibited [(14)C]glutamate uptake two fold. Furthermore ferritin induced intrasynaptosomal ROS (reactive oxygen species) formation in a dose-dependent manner. This process was insensitive to 30 microM DPI, an inhibitor of NADPH oxidase and to 10 microM CCCP, a mitochondrial uncoupler. These results indicate that iron-based nanoparticles can cause ROS and decreased glutamate uptake, potentially leading to neurodegeneration.
Collapse
Affiliation(s)
- Alexandra V Alekseenko
- Institute of Biophysics and Cell Engineering, National Academy of Sciences of Belarus, Akademicheskaya Street, 27, Minsk 220072, Belarus
| | | | | |
Collapse
|
55
|
Furness DN, Dehnes Y, Akhtar AQ, Rossi DJ, Hamann M, Grutle NJ, Gundersen V, Holmseth S, Lehre KP, Ullensvang K, Wojewodzic M, Zhou Y, Attwell D, Danbolt NC. A quantitative assessment of glutamate uptake into hippocampal synaptic terminals and astrocytes: new insights into a neuronal role for excitatory amino acid transporter 2 (EAAT2). Neuroscience 2008; 157:80-94. [PMID: 18805467 DOI: 10.1016/j.neuroscience.2008.08.043] [Citation(s) in RCA: 198] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2007] [Revised: 08/07/2008] [Accepted: 08/08/2008] [Indexed: 01/04/2023]
Abstract
The relative distribution of the excitatory amino acid transporter 2 (EAAT2) between synaptic terminals and astroglia, and the importance of EAAT2 for the uptake into terminals is still unresolved. Here we have used antibodies to glutaraldehyde-fixed d-aspartate to identify electron microscopically the sites of d-aspartate accumulation in hippocampal slices. About 3/4 of all terminals in the stratum radiatum CA1 accumulated d-aspartate-immunoreactivity by an active dihydrokainate-sensitive mechanism which was absent in EAAT2 glutamate transporter knockout mice. These terminals were responsible for more than half of all d-aspartate uptake of external substrate in the slices. This is unexpected as EAAT2-immunoreactivity observed in intact brain tissue is mainly associated with astroglia. However, when examining synaptosomes and slice preparations where the extracellular space is larger than in perfusion fixed tissue, it was confirmed that most EAAT2 is in astroglia (about 80%). Neither d-aspartate uptake nor EAAT2 protein was detected in dendritic spines. About 6% of the EAAT2-immunoreactivity was detected in the plasma membrane of synaptic terminals (both within and outside of the synaptic cleft). Most of the remaining immunoreactivity (8%) was found in axons where it was distributed in a plasma membrane surface area several times larger than that of astroglia. This explains why the densities of neuronal EAAT2 are low despite high levels of mRNA in CA3 pyramidal cell bodies, but not why EAAT2 in terminals account for more than half of the uptake of exogenous substrate by hippocampal slice preparations. This and the relative amount of terminal versus glial uptake in the intact brain remain to be discovered.
Collapse
Affiliation(s)
- D N Furness
- Institute of Science and Technology in Medicine, Keele University, Keele, Staffs, ST5 5BG, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
56
|
Crema LM, Vendite D, Horn AP, Diehl LA, Aguiar AP, Nunes E, Vinade L, Fontella FU, Salbego C, Dalmaz C. Effects of chronic restraint stress and estradiol replacement on glutamate release and uptake in the spinal cord from ovariectomized female rats. Neurochem Res 2008; 34:499-507. [PMID: 18712597 DOI: 10.1007/s11064-008-9810-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2008] [Accepted: 07/15/2008] [Indexed: 12/13/2022]
Abstract
Glutamate is an excitatory neurotransmitter involved in neuronal plasticity and neurotoxicity. Chronic stress produces several physiological changes on the spinal cord, many of them presenting sex-specific differences, which probably involve glutamatergic system alterations. The aim of the present study was to verify possible effects of exposure to chronic restraint stress and 17beta-estradiol replacement on [3H]-glutamate release and uptake in spinal cord synaptosomes of ovariectomized (OVX) rats. Female rats were subjected to OVX, and half of the animals received estradiol replacement. Animals were subdivided in controls and chronically stressed. Restraint stress or estradiol had no effect on [3H]-glutamate release. The chronic restraint stress promoted a decrease and 17beta-estradiol induced an increase on [3H]-glutamate uptake, but the uptake observed in the restraint stress +17beta-estradiol group was similar to control. Furthermore, 17beta-estradiol treatment caused a significant increase in the immunocontent of the three glutamate transporters present in spinal cord. Restraint stress had no effect on the expression of these transporters, but prevented the 17beta-estradiol effect. We suggest that changes in the glutamatergic system are likely to take part in the mechanisms involved in spinal cord plasticity following repeated stress exposure, and that 17beta-estradiol levels may affect chronic stress effects in this structure.
Collapse
Affiliation(s)
- Leonardo Machado Crema
- Programa de Pós-Graduação em Neurociências, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
57
|
Excitatory amino acid transporter expression by astrocytes is neuroprotective against microglial excitotoxicity. Brain Res 2008; 1210:11-9. [DOI: 10.1016/j.brainres.2008.03.012] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2008] [Revised: 02/25/2008] [Accepted: 03/03/2008] [Indexed: 11/20/2022]
|
58
|
Lehmann C, Eisner F, Engele J. Role of endothelins as mediators of injury-induced alterations of glial glutamate turnover. J Neurosci Res 2008; 86:660-7. [PMID: 17893916 DOI: 10.1002/jnr.21512] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Astroglia terminate glutamatergic neurotransmission and prevent excitotoxic extracellular glutamate concentration by clearing synaptically released glutamate through the high-affinity, sodium-dependent glutamate transporters GLT-1 and GLAST. Many brain injures are associated with the disturbed expression of glial glutamate transporters and a subsequent increase of extracellular glutamate to neurotoxic levels. We have now followed up initial hints pointing to endothelins, a family of injury-regulated peptides, as mediators of this injury-induced loss of glial glutamate transporter expression. We observed that, in line with such a role, endothelins not only act as potent inhibitors of basal and exogenously (dbcAMP)-induced expression of GLT-1 in cortical astrocytes as shown before, but likewise inhibit expression of GLT-1 or GLAST in astrocytes cultured from the diencephalon, mesencephalon, cerebellum, and spinal cord. We further demonstrate that endothelins equally inhibit GLT-1 expression in cortical slice cultures, a culture system closely resembling the in vivo situation. Although brain injuries are usually associated with an increase in the expression of the glutamate-converting enzyme glutamine synthetase, cultured cortical astrocytes maintained with endothelins showed an almost complete loss of glutamine synthetase. Interestingly, the inhibitory effects of endothelins on the expression of glutamine synthetase, but not of glutamate transporters, was overridden by high extracellular glutamate, indicating that the primarily inhibitory action of endothelins on the various components of glial glutamate turnover dissociates in the injured brain.
Collapse
Affiliation(s)
- Claudia Lehmann
- Institute of Anatomy, University of Leipzig, Medical Faculty, Leipzig, Germany
| | | | | |
Collapse
|
59
|
Vit JP, Ohara PT, Bhargava A, Kelley K, Jasmin L. Silencing the Kir4.1 potassium channel subunit in satellite glial cells of the rat trigeminal ganglion results in pain-like behavior in the absence of nerve injury. J Neurosci 2008; 28:4161-71. [PMID: 18417695 PMCID: PMC2533133 DOI: 10.1523/jneurosci.5053-07.2008] [Citation(s) in RCA: 140] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2007] [Revised: 02/25/2008] [Accepted: 02/26/2008] [Indexed: 12/21/2022] Open
Abstract
Growing evidence suggests that changes in the ion buffering capacity of glial cells can give rise to neuropathic pain. In the CNS, potassium ion (K+) buffering is dependent on the glia-specific inward rectifying K+ channel Kir4.1. We recently reported that the satellite glial cells that surround primary sensory neurons located in sensory ganglia of the peripheral nervous system also express Kir4.1, whereas the neurons do not. In the present study, we show that, in the rat trigeminal ganglion, the location of the primary sensory neurons for face sensation, specific silencing of Kir4.1 using RNA interference leads to spontaneous and evoked facial pain-like behavior in freely moving rats. We also show that Kir4.1 in the trigeminal ganglion is reduced after chronic constriction injury of the infraorbital nerve. These findings suggests that neuropathic pain can result from a change in expression of a single K+ channel in peripheral glial cells, raising the possibility of targeting Kir4.1 to treat pain in general and particularly neuropathic pain that occurs in the absence of nerve injury.
Collapse
Affiliation(s)
- Jean-Philippe Vit
- Department of Neurosurgery and Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, and
| | | | - Aditi Bhargava
- Surgery, University of California, San Francisco, San Francisco, California 94143
| | - Kanwar Kelley
- Department of Neurosurgery and Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, and
| | - Luc Jasmin
- Department of Neurosurgery and Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, and
| |
Collapse
|
60
|
Synthesis of enantiomerically pure HIP-A and HIP-B and investigation of their activity as inhibitors of excitatory amino acid transporters. ACTA ACUST UNITED AC 2008. [DOI: 10.1016/j.tetasy.2008.03.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
61
|
Paluzzi S, Alloisio S, Zappettini S, Milanese M, Raiteri L, Nobile M, Bonanno G. Adult astroglia is competent for Na+/Ca2+ exchanger-operated exocytotic glutamate release triggered by mild depolarization. J Neurochem 2007; 103:1196-207. [PMID: 17935604 DOI: 10.1111/j.1471-4159.2007.04826.x] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Glutamate release induced by mild depolarization was studied in astroglial preparations from the adult rat cerebral cortex, that is acutely isolated glial sub-cellular particles (gliosomes), cultured adult or neonatal astrocytes, and neuron-conditioned astrocytes. K+ (15, 35 mmol/L), 4-aminopyridine (0.1, 1 mmol/L) or veratrine (1, 10 micromol/L) increased endogenous glutamate or [3H]D-aspartate release from gliosomes. Neurotransmitter release was partly dependent on external Ca2+, suggesting the involvement of exocytotic-like processes, and partly because of the reversal of glutamate transporters. K+ increased gliosomal membrane potential, cytosolic Ca2+ concentration [Ca2+]i, and vesicle fusion rate. Ca2+ entry into gliosomes and glutamate release were independent from voltage-sensitive Ca2+ channel opening; they were instead abolished by 2-[2-[4-(4-nitrobenzyloxy)phenyl]ethyl]isothiurea (KB-R7943), suggesting a role for the Na+/Ca2+ exchanger working in reverse mode. K+ (15, 35 mmol/L) elicited increase of [Ca2+]i and Ca2+-dependent endogenous glutamate release in adult, not in neonatal, astrocytes in culture. Glutamate release was even more marked in in vitro neuron-conditioned adult astrocytes. As seen for gliosomes, K+-induced Ca2+ influx and glutamate release were abolished by KB-R7943 also in cultured adult astrocytes. To conclude, depolarization triggers in vitro glutamate exocytosis from in situ matured adult astrocytes; an aptitude grounding on Ca2+ influx driven by the Na+/Ca2+ exchanger working in the reverse mode.
Collapse
Affiliation(s)
- Silvio Paluzzi
- Department of Experimental Medicine, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy
| | | | | | | | | | | | | |
Collapse
|
62
|
Nafia I, Re DB, Masmejean F, Melon C, Kachidian P, Kerkerian-Le Goff L, Nieoullon A, Had-Aissouni L. Preferential vulnerability of mesencephalic dopamine neurons to glutamate transporter dysfunction. J Neurochem 2007; 105:484-96. [PMID: 18042178 DOI: 10.1111/j.1471-4159.2007.05146.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Nigral depletion of the main brain antioxidant GSH is the earliest biochemical event involved in Parkinson's disease pathogenesis. Its causes are completely unknown but increasing number of evidence suggests that glutamate transporters [excitatory amino acid transporters (EAATs)] are the main route by which GSH precursors may enter the cell. In this study, we report that dopamine (DA) neurons, which express the excitatory amino acid carrier 1, are preferentially affected by EAAT dysfunction when compared with non-DA neurons. In rat embryonic mesencephalic cultures, l-trans-pyrrolidine-2,4-dicarboxylate, a substrate inhibitor of EAATs, is directly and preferentially toxic for DA neurons by decreasing the availability of GSH precursors and lowering their resistance threshold to glutamate excitotoxicity through NMDA-receptors. In adult rat, acute intranigral injection of l-trans-pyrrolidine-2,4-dicarboxylate induces a large regionally selective and dose-dependent loss of DA neurons and alpha-synuclein aggregate formation. These data highlight for the first time the importance of excitatory amino acid carrier 1 function for the maintenance of antioxidant defense in DA neurons and suggest its dysfunction as a candidate mechanism for the selective death of DA neurons such as occurring in Parkinson's disease.
Collapse
Affiliation(s)
- Imane Nafia
- Equipe Interactions Cellulaires, Neurodégénérescence et Neuroplasticité, Institut de Biologie du Développement de Marseille-Luminy, UMR6216 CNRS-Université de la Méditerranée, Marseille, France
| | | | | | | | | | | | | | | |
Collapse
|
63
|
Bonanno G, Raiteri L, Milanese M, Zappettini S, Melloni E, Pedrazzi M, Passalacqua M, Tacchetti C, Usai C, Sparatore B. The high-mobility group box 1 cytokine induces transporter-mediated release of glutamate from glial subcellular particles (gliosomes) prepared from in situ-matured astrocytes. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2007; 82:73-93. [PMID: 17678956 DOI: 10.1016/s0074-7742(07)82004-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
The multifunctional protein high-mobility group box 1 (HMGB1) is expressed in restricted areas of adult brain where it can act as a proinflammatory cytokine. We report here that HMGB1 affects CNS transmission by inducing glutamatergic release from glial (gliosomes) but not neuronal (synaptosomes) resealed subcellular particles isolated from mouse cerebellum and hippocampus. Confocal microscopy showed that gliosomes are enriched with glia-specific proteins such as GFAP and S-100, but not with neuronal proteins such as PSD-95, MAP-2, and beta-tubulin III. Furthermore, gliosomes exhibit labeling neither for integrin-alphaM nor for myelin basic protein, specific for microglia and oligodendrocytes, respectively. The gliosomal fraction contains proteins of the exocytotic machinery coexisting with GFAP. Consistent with ultrastructural analysis, several approximately 30-nm nonclustered vesicles are present in the gliosome cytoplasm. Finally, gliosomes represent functional organelles that actively export glutamate when subjected to releasing stimuli, such as ionomycin or ATP, by mechanisms involving extracellular Ca(2+) and Ca(2+) release from intracellular stores. HMGB1-induced release of the stable glutamate analogue [(3)H]d-aspartate and endogenous glutamate form gliosomes, whereas nerve terminals were insensitive to the protein. The HMGB1-evoked release of glutamate was independent on modifications of cytosolic Ca(2+) concentration, but it was blocked by dl-threo-beta-benzyloxyaspartate, suggesting the involvement of transporter-mediated release mechanisms. Moreover, dihydrokainic acid, a selective inhibitor of glutamate transporter 1 does not block the HMGB1 effect, indicating a role for the glial glutamate-aspartate transporter (GLAST) subtype in this response. HMGB1 bind to gliosomes but not to synaptosomes and can physically interact with GLAST and receptor for advanced glycation end products (RAGE). Taken together, these results suggest that the HMGB1 cytokine could act as a modulator of glutamate homeostasis in adult mammalian brain.
Collapse
Affiliation(s)
- Giambattista Bonanno
- Department of Experimental Medicine, Section of Pharmacology and Toxicology University of Genoa, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Raiteri L, Zappettini S, Milanese M, Fedele E, Raiteri M, Bonanno G. Mechanisms of glutamate release elicited in rat cerebrocortical nerve endings by 'pathologically' elevated extraterminal K+ concentrations. J Neurochem 2007; 103:952-61. [PMID: 17662048 DOI: 10.1111/j.1471-4159.2007.04784.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Extracellular [K+] can increase during some pathological conditions, resulting into excessive glutamate release through multiple mechanisms. We here investigate the overflow of [3H]D-aspartate ([3H] D-ASP) and of endogenous glutamate elicited by increasing [K+] from purified rat cerebrocortical synaptosomes. Depolarization with [K+] <or= 15 mmol/L provoked [3H] D-ASP and glutamate overflows almost totally dependent on external Ca2+. Consistent with release by exocytosis, the overflow of [3H] D-ASP evoked by 12 mmol/L K+ was sensitive to clostridial toxins. The overflows evoked by 35/50 mmol/L K+ remained external Ca2+-dependent by more than 50%. The Ca2+-independent components of the [3H] D-ASP overflows evoked by [K+] > 15 mmol/L were prevented by the glutamate transporter inhibitors DL-threo-beta-benzyloxyaspartate (DL-TBOA) and dihydrokainate. Differently, the overflows of endogenous glutamate provoked by [K+] > 15 mmol/L were insensitive to both inhibitors; the external Ca2+-independent glutamate overflow caused by 50 mmol/L KCl was prevented by bafilomycin, by chelating intraterminal Ca2+, by blocking the mitochondrial Na+/Ca2+ exchanger and, for a small portion, by blocking anion channels. In contrast to purified synaptosomes, the 50 mmol/L K+-evoked release of endogenous glutamate or [3H]D-ASP was inhibited by DL-TBOA in crude synaptosomes; moreover, it was external Ca2+-insensitive and blocked by DL-TBOA in purified gliosomes, suggesting that carrier-mediated release of endogenous glutamate provoked by excessive [K+] in CNS tissues largely originates from glia.
Collapse
Affiliation(s)
- Luca Raiteri
- Department of Experimental Medicine, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy
| | | | | | | | | | | |
Collapse
|
65
|
Patti L, Raiteri L, Grilli M, Zappettini S, Bonanno G, Marchi M. Evidence that α7 nicotinic receptor modulates glutamate release from mouse neocortical gliosomes. Neurochem Int 2007; 51:1-7. [PMID: 17462791 DOI: 10.1016/j.neuint.2007.03.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2007] [Revised: 03/05/2007] [Accepted: 03/07/2007] [Indexed: 11/23/2022]
Abstract
The presence of nicotinic receptors on astrocytes in human and rat brain has been previously demonstrated however their possible functional role is still poorly understood. In this study we investigated on the presence of nicotinic receptors on gliosomes, purified from mouse cortex, and on their role in eliciting glutamate release. Epibatidine significantly increased basal release of [3H]D-aspartate and of endogenous glutamate from mouse gliosomes but not from synaptosomes. This effect was prevented by methyllycaconitine, alpha-bungarotoxin and mecamylamine but not by dihydro-beta-erythroidine. Epibatidine provoked also a significant increase of calcium concentration in gliosomes but not in synaptosomes; the increase in [Ca2+]i induced by epibatidine and KCl in gliosomes was very similar to each other. The present results indicate that alpha7 nicotinic receptors exist on mouse cortical glial particles and stimulate glutamate release.
Collapse
Affiliation(s)
- Laura Patti
- Sezione di Farmacologia e Tossicologia, Dipartimento di Medicina Sperimentale, Università degli Studi di Genova, Viale Cembrano 4, 16148 Genoa, Italy
| | | | | | | | | | | |
Collapse
|
66
|
Schroeter H, Bahia P, Spencer JPE, Sheppard O, Rattray M, Cadenas E, Rice-Evans C, Williams RJ. (-)Epicatechin stimulates ERK-dependent cyclic AMP response element activity and up-regulates GluR2 in cortical neurons. J Neurochem 2007; 101:1596-606. [PMID: 17298385 DOI: 10.1111/j.1471-4159.2006.04434.x] [Citation(s) in RCA: 137] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Emerging evidence suggests that the cellular actions of flavonoids relate not simply to their antioxidant potential but also to the modulation of protein kinase signalling pathways. We investigated in primary cortical neurons, the ability of the flavan-3-ol, (-)epicatechin, and its human metabolites at physiologically relevant concentrations, to stimulate phosphorylation of the transcription factor cAMP-response element binding protein (CREB), a regulator of neuronal viability and synaptic plasticity. (-)Epicatechin at 100-300 nmol/L stimulated a rapid, extracellular signal-regulated kinase (ERK)- and PI3K-dependent, increase in CREB phosphorylation. At micromolar concentrations, stimulation was no longer apparent and at the highest concentration tested (30 mumol/L) (-)epicatechin was inhibitory. (-)Epicatechin also stimulated ERK and Akt phosphorylation with similar bell-shaped concentration-response characteristics. The human metabolite 3'-O-methyl-(-)epicatechin was as effective as (-)epicatechin at stimulating ERK phosphorylation, but (-)epicatechin glucuronide was inactive. (-)Epicatechin and 3'-O-methyl-(-)epicatechin treatments (100 nmol/L) increased CRE-luciferase activity in cortical neurons in a partially ERK-dependent manner, suggesting the potential to increase CREB-mediated gene expression. mRNA levels of the glutamate receptor subunit GluR2 increased by 60%, measured 18 h after a 15 min exposure to (-)epicatechin and this translated into an increase in GluR2 protein. Thus, (-)epicatechin has the potential to increase CREB-regulated gene expression and increase GluR2 levels and thus modulate neurotransmission, plasticity and synaptogenesis.
Collapse
Affiliation(s)
- Hagen Schroeter
- Department of Molecular Pharmacology and Toxicology, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|
67
|
Figiel M, Allritz C, Lehmann C, Engele J. Gap junctional control of glial glutamate transporter expression. Mol Cell Neurosci 2007; 35:130-7. [PMID: 17369047 DOI: 10.1016/j.mcn.2007.02.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2006] [Revised: 02/06/2007] [Accepted: 02/08/2007] [Indexed: 11/24/2022] Open
Abstract
The uptake of glutamate into astroglia is the predominant mechanism to terminate glutamatergic neurotransmission and to prevent neurotoxic extracellular glutamate concentrations. Here, we show that uncoupling cultured cortical astrocytes with the gap junction blocker, propofol, or the Cx43 mimetic peptide, Gap27, inhibits the expression of GLT-1, the major glutamate transporter subtype in the cortex. The dependence of GLT-1 expression on gap junctions was further confirmed by the use of astrocytes in which either the expression of Cx43, the major astrocytic gap junction protein, was inhibited by RNA interference or which were derived from animals carrying an astrocyte-specific deletion of the Cx43 gene. In both cases, reduced astrocytic coupling was associated with a pronounced decline in GLT-1 expression. Finally, a luciferase reporter gene assay demonstrated that blockade of gap junctions/connexins suppressed transcriptional activity of GLT-1 promoter. These observations unravel a previously unrecognized role of gap junctions in the control of glial glutamate transport.
Collapse
Affiliation(s)
- Maciej Figiel
- Institute of Anatomy, University of Leipzig, Medical Faculty, Liebigstr. 13, 04103 Leipzig, Germany
| | | | | | | |
Collapse
|
68
|
Fernandes SP, Edwards TM, Ng KT, Robinson SR. HIV-1 protein gp120 rapidly impairs memory in chicks by interrupting the glutamate–glutamine cycle. Neurobiol Learn Mem 2007; 87:1-8. [PMID: 16714124 DOI: 10.1016/j.nlm.2006.03.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2005] [Revised: 03/27/2006] [Accepted: 03/28/2006] [Indexed: 10/24/2022]
Abstract
Learning and memory impairments are frequently observed in patients suffering from AIDS Dementia Complex (ADC). These effects have been linked to the presence of gp120, an HIV viral coat glycoprotein. The present study investigated the possibility that gp120 prevents the uptake of extracellular glutamate by astrocytes, leading to an interruption of the glutamate-glutamine cycle and a subsequent impairment of memory. Ten microliters of 10nM gp120 was bilaterally injected into the region of the intermediate medial mesopallium of day-old chicks at various times before, or after, training using a single-trial passive avoidance task. Gp120 was found to significantly impair memory retention when injected 10-40 min after training. Memory impairments were evident within 5 min of gp120 administration and remained evident 24h later. Further, the amnestic effect of gp120 could be overcome with glutamine or with precursors of glutamate synthesis, but only weakly by glutamate. These results support the conclusion that the amnestic effect of gp120 is due to an impaired uptake of glutamate by astrocytes and a subsequent interruption of glutamine supply to neurones. The data indicate that the glutamate-glutamine cycle may be a useful therapeutic target in the treatment of ADC.
Collapse
Affiliation(s)
- S P Fernandes
- School of Psychology, Psychiatry and Psychological Medicine, Monash University, 3800 Vic., Australia
| | | | | | | |
Collapse
|
69
|
van Landeghem FKH, Weiss T, Oehmichen M, von Deimling A. Decreased Expression of Glutamate Transporters in Astrocytes after Human Traumatic Brain Injury. J Neurotrauma 2006; 23:1518-28. [PMID: 17020486 DOI: 10.1089/neu.2006.23.1518] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The primary mechanism for eliminating synaptically released glutamate is uptake by astrocytes. In the present study, we examined whether traumatic brain injury (TBI) affects the cellular expression of glutamate transporters EAAT1 and EAAT2. Morphometrical immunohistochemical analysis demonstrated a predominant expression of EAAT1 and EAAT2 in astrocytes of normal human neocortex (n = 10). Following traumatic injury of human brain (n = 55), the number of EAAT2-positive cells was decreased for a prolonged survival period within the traumatized neocortex and the pericontusional region. GFAP-positive astrocytes decreased in number within the first 24 h. Thereafter, the number of GFAP-positive astrocytes increased again, indicating formation of reactive gliosis. Double immunofluorescence examinations revealed a reduction in absolute numbers of GFAP-positive astrocytes coexpressing EAAT1 or EAAT2 at survival times up to 7 days. In addition, the relative fractions of astrocytes coexpressing glutamate transporters decreased following TBI. We conclude that the posttraumatic reduction in cellular EAAT 1 and EAAT2 expression is predominantly due to degeneration of astrocytes and to downregulation in surviving astrocytes. Our results support the view that reduced glutamate uptake by astrocytes contributes to posttraumatic elevation of extracellular glutamate in humans.
Collapse
|
70
|
Pedrazzi M, Raiteri L, Bonanno G, Patrone M, Ledda S, Passalacqua M, Milanese M, Melloni E, Raiteri M, Pontremoli S, Sparatore B. Stimulation of excitatory amino acid release from adult mouse brain glia subcellular particles by high mobility group box 1 protein. J Neurochem 2006; 99:827-38. [PMID: 16911580 DOI: 10.1111/j.1471-4159.2006.04120.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The multifunctional protein high mobility group box 1 (HMGB1) is expressed in hippocampus and cerebellum of adult mouse brain. Our aim was to determine whether HMGB1 affects glutamatergic transmission by monitoring neurotransmitter release from glial (gliosomes) and neuronal (synaptosomes) re-sealed subcellular particles isolated from cerebellum and hippocampus. HMGB1 induced release of the glutamate analogue [(3)H]d-aspartate form gliosomes in a concentration-dependent manner, whereas nerve terminals were insensitive to the protein. The HMGB1-evoked release of [(3)H]d-aspartate was independent of modifications of cytosolic Ca(2+) , but it was blocked by dl-threo-beta-benzyloxyaspartate (dl-TBOA), an inhibitor of glutamate transporters. HMGB1 also stimulated the release of endogenous glutamate in a Ca(2+)-independent and dl-TBOA-sensitive manner. These findings suggest the involvement of carrier-mediated release. Moreover, dihydrokainic acid, a selective inhibitor of glutamate transporter 1 (GLT1), does not block the effect of HMGB1, indicating a role for the glial glutamate-aspartate transporter (GLAST) subtype in this response. We also demonstrate that HMGB1/glial particles association is promoted by Ca(2+). Furthermore, although HMGB1 can physically interact with GLAST and the receptor for advanced glycation end products (RAGE), only its binding with RAGE is promoted by Ca(2+). These results suggest that the HMGB1 cytokine could act as a modulator of glutamate homeostasis in adult mammal brain.
Collapse
Affiliation(s)
- Marco Pedrazzi
- Department of Experimental Medicine and Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
71
|
Rattray M, Bendotti C. Does excitotoxic cell death of motor neurons in ALS arise from glutamate transporter and glutamate receptor abnormalities? Exp Neurol 2006; 201:15-23. [PMID: 16806177 DOI: 10.1016/j.expneurol.2006.05.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2006] [Revised: 05/02/2006] [Accepted: 05/04/2006] [Indexed: 12/14/2022]
Affiliation(s)
- Marcus Rattray
- King's College London, Wolfson Centre for Age-Related Diseases, Guy's Hospital Campus, London SE1 1UL, UK
| | | |
Collapse
|
72
|
Thais ME, Carqueja CL, Santos TG, Silva RV, Stroeh E, Machado RS, Wahlheim DO, Bianchin MM, Sakamoto AC, Brentani RR, Martins VR, Walz R, Tasca CI. Synaptosomal glutamate release and uptake in mice lacking the cellular prion protein. Brain Res 2006; 1075:13-9. [PMID: 16519879 DOI: 10.1016/j.brainres.2005.12.045] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2005] [Revised: 11/21/2005] [Accepted: 12/11/2005] [Indexed: 10/24/2022]
Abstract
Glutamate plays a central role in the fast excitatory synaptic transmission and is a key neurotransmitter involved in several neurophysiological processes. Glutamate levels on the synaptic cleft are related to neural excitability, neuroplasticity, and neuronal damage associated with excitotoxicity. Mice lacking the cellular prion protein (PrP(c)) gene (Prnp) present a decreased astrocytic glutamate uptake in cultures, higher neuronal excitability in vitro and sensitivity to pro-convulsant drugs in vivo, and age-dependent memory impairment. Here, we investigate if PrP(c) might be involved in neuronal uptake and release of glutamate. For this purpose, we compared synaptosomal preparations from the cerebral cortex, entorhinal cortex, hippocampus, cerebellum, and olfactory bulb of 3- or 9-month-old PrP(c) null mice and with respective wild-type controls. Although we observed differences in synaptosomal glutamate release and uptake regarding the age of mice and the brain structure studied, these differences were similar for PrP(c) null mice and their respective wild-type controls. Therefore, despite a possible correlation between neuronal glutamate transporters, excitability, and neuronal damage, our results suggest that PrP(c) expression is not critical for neuronal glutamate transport.
Collapse
Affiliation(s)
- Maria Emília Thais
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Trindade, 88040-900 Florianópolis, SC, Brasil
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
73
|
Alele PE, Devaud LL. Differential adaptations in GABAergic and glutamatergic systems during ethanol withdrawal in male and female rats. Alcohol Clin Exp Res 2005; 29:1027-34. [PMID: 15976529 DOI: 10.1097/01.alc.0000167743.96121.40] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND There are significant and consistent sex differences in recovery from ethanol withdrawal in our animal model of ethanol dependence. We have also observed significant and varied sex differences in subunit protein levels of gamma-aminobutyric acid A (GABAA) and the N-metheyl-D-aspartate subtype of glutamate receptors occurring with ethanol dependence and withdrawal. Considering the major role of these two systems as targets of ethanol, we wanted to explore additional possible mechanisms underlying changes in GABAergic and glutamatergic responses after chronic ethanol exposure. Therefore, the objective of the present study was to examine GABAergic- and glutamatergic-associated proteins at three days of ethanol withdrawal, when female rats appear to have largely recovered but male rats still display robust signs of withdrawal. METHODS Male and female rats were fed 6% ethanol in a nutritionally complete liquid diet for 14 days according to a pair-fed design; withdrawal was initiated by replacement of the diet with chow. At three days of withdrawal, the cerebral cortex and hippocampus were dissected for use in Western blot analysis. The paired design was maintained throughout all experimental procedures. RESULTS At three days of ethanol withdrawal, we found region-specific and sex-selective alterations in levels of GAD (glutamic acid decarboxylase, GABA synthetic enzyme), GABA and glutamate transporters, and the synapse-associated proteins HSP70, PSD-95, and synaptophysin. There were also several significant differences in transporter function at this time that varied between males and females. CONCLUSIONS Taken together, these findings show differential adaptations of GABAergic and glutamatergic neurotransmission between female and male rats that are associated with withdrawal recovery. This suggests that selective withdrawal-induced neuroadaptations in regulation of these systems' activities underlie, at least in part, sex differences in withdrawal recovery between male and female rats.
Collapse
Affiliation(s)
- P E Alele
- Department of Pharmaceutical Sciences, Idaho State University, Pocatello, Idaho 83209, USA
| | | |
Collapse
|
74
|
Holmer HK, Keyghobadi M, Moore C, Meshul CK. l-dopa-induced reversal in striatal glutamate following partial depletion of nigrostriatal dopamine with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. Neuroscience 2005; 136:333-41. [PMID: 16198485 DOI: 10.1016/j.neuroscience.2005.08.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2005] [Revised: 07/21/2005] [Accepted: 08/01/2005] [Indexed: 11/17/2022]
Abstract
We have reported that 1 month following acute (20mg/kg x 4) or subchronic (30 mg/kg/day x 7d) administration of the neurotoxin, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, there is an increase or decrease, respectively, in the extracellular level of striatal glutamate as determined by in vivo microdialysis [Robinson S, Freeman P, Moore C, Touchon JC, Krentz L, Meshul CK (2003) Acute and subchronic MPTP administration differentially affects striatal glutamate synaptic function. Exp Neurol 180:73-86]. The goal of this study was to determine the effects of treatment with l-dopa (15 mg/kg) for 21 days on striatal glutamate starting on day 8 after the first dose of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine was administered to mice. Following acute administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, the increase in extracellular striatal glutamate due to lesion of the nigrostriatal pathway was completely reversed to a level below that found in the vehicle-treated group after l-dopa treatment. Subchronic 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine treatment resulted in a decrease in striatal extracellular glutamate that was reversed to the level close to that observed in the vehicle-treated group. There was no change in the density of nerve terminal glutamate immunolabeling associated with the synaptic vesicle pool, suggesting that the alterations in extracellular glutamate most likely originated from the calcium-independent pool. There was a similar decrease in the relative density of tyrosine hydroxylase immunolabeling, a marker for dopamine terminals, within the dorsolateral striatum in both the acute and subchronic 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated groups that had been administered l-dopa. There was a decrease in the relative density of immunolabeling within the dorsolateral striatum for the glutamate transporter, GLT-1, following acute 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine treatment in the groups administered either vehicle or l-dopa. There was no change in GLT-1 immunolabeling following subchronic 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. The results demonstrate that the reversal in the extracellular level of striatal glutamate following l-dopa treatment in both the acute and subchronic 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated groups is not due to changes in either striatal dopamine nerve terminals or in the density of the glutamate transporter, GLT-1.
Collapse
Affiliation(s)
- H K Holmer
- Research Services, Neurocytology Laboratory, VA Medical Center, Portland, OR 97239, USA
| | | | | | | |
Collapse
|
75
|
Raiteri L, Stigliani S, Patti L, Usai C, Bucci G, Diaspro A, Raiteri M, Bonanno G. Activation of ?-aminobutyric acid GAT-1 transporters on glutamatergic terminals of mouse spinal cord mediates glutamate release through anion channels and by transporter reversal. J Neurosci Res 2005; 80:424-33. [PMID: 15789377 DOI: 10.1002/jnr.20437] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The effects of gamma-aminobutyric acid (GABA) on the release of glutamate from mouse spinal cord nerve endings have been studied using superfused synaptosomes. GABA elicited a concentration-dependent release of [3H]D-aspartate ([3H]D-ASP; EC50= 3.76 microM). Neither muscimol nor (-)baclofen mimicked GABA, excluding receptor involvement. The GABA-evoked release was strictly Na+ dependent and was prevented by the GABA transporter inhibitor SKF89976A, suggesting involvement of GAT-1 transporters located on glutamatergic nerve terminals. GABA also potentiated the spontaneous release of endogenous glutamate; an effect sensitive to SKF89976A and low-Na+-containing medium. Confocal microscopy shows that the GABA transporter GAT-1 is coexpressed with the vesicular glutamate transporter vGLUT-1 and with the plasma membrane glutamate transporter EAAT2 in a substantial portion of synaptosomal particles. The GABA effect was external Ca2+ independent and was not decreased when cytosolic Ca2+ ions were chelated by BAPTA. The glutamate transporter blocker DL-TBOA or dihydrokainate inhibited in part (approximately 35%) the GABA (10 microM)-evoked [3H]D-ASP release; this release was strongly reduced by the anion channel blockers niflumic acid and NPPB. GABA, up to 30 microM, was unable to augment significantly the basal release of [3H]glycine from spinal cord synaptosomes, indicating selectivity for glutamatergic transmission. It is concluded that GABA GAT-1 transporters and glutamate transporters coexist on the same spinal cord glutamatergic terminals. Activation of these GABA transporters elicits release of glutamate partially by reversal of glutamate transporters present on glutamatergic terminals and largely through anion channels.
Collapse
Affiliation(s)
- Luca Raiteri
- Department of Experimental Medicine, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy
| | | | | | | | | | | | | | | |
Collapse
|
76
|
Dolman D, Drndarski S, Abbott NJ, Rattray M. Induction of aquaporin 1 but not aquaporin 4 messenger RNA in rat primary brain microvessel endothelial cells in culture. J Neurochem 2005; 93:825-33. [PMID: 15857386 DOI: 10.1111/j.1471-4159.2005.03111.x] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Aquaporins (AQPs) are a family of proteins that mediate water transport across cells, but the extent to which they are involved in water transport across endothelial cells of the blood-brain barrier is not clear. Expression of AQP1 and AQP4 in rat brain microvessel endothelial cells was investigated in order to determine whether these isoforms were present and, in particular, to examine the hypothesis that brain endothelial expression of AQPs is dynamic and regulated by astrocytic influences. Reverse-transcriptase-polymerase chain reaction (RT-PCR) and immunocytochemistry showed that AQP1 mRNA and protein are present at very low levels in primary rat brain microvessel endothelial cells, and are up-regulated in passaged cells. Upon passage, endothelial cell expression of mdr1a mRNA is decreased, indicating loss of blood-brain barrier phenotype. In passage 4 endothelial cells, AQP1 mRNA levels are reduced by coculture above rat astrocytes, demonstrating that astrocytic influences are important in maintaining the low levels of AQP1 characteristic of the blood-brain barrier endothelium. Reverse-transcriptase-PCR revealed very low levels of AQP1 mRNA present in the RBE4 rat brain microvessel endothelial cell line, with no expression detected in primary cultures of rat astrocytes or in the C6 rat glioma cell line. In contrast, AQP4 mRNA is strongly expressed in astrocytes, but no expression is found in primary or passaged brain microvessel endothelial cells, or in RBE4 or C6 cells. Our results support the concept that expression of AQP1, which is seen in many non-brain endothelia, is suppressed in the specialized endothelium of the blood-brain barrier.
Collapse
Affiliation(s)
- Diana Dolman
- Blood-Brain Barrier Research Group, Wolfson Centre for Age-Related Diseases, Guy's Kings & St Thomas' School of Biomedical Sciences, King's College London, UK
| | | | | | | |
Collapse
|
77
|
Shakil SS, Holmer HK, Moore C, Abernathy AT, Jakowec MW, Petzinger GM, Meshul CK. High and low responders to novelty show differential effects in striatal glutamate. Synapse 2005; 58:200-7. [PMID: 16138315 DOI: 10.1002/syn.20198] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The goal of this study was to determine whether there was a difference in glutamate within the dorsolateral striatum in mice exhibiting either a high (HR) or low (LR) locomotor response to a novel environment. The number of line crossings over a 30-min-period when the mice were placed in a novel environment was determined, and those mice for which the values were above the mean were in the HR group and those with the values below the mean were in the LR group. In vivo microdialysis was carried out to determine the basal extracellular level of striatal glutamate, and the contralateral striatum was taken to measure the density of glutamate immunolabeling within nerve terminals making an asymmetrical (excitatory) synaptic contact using quantitative immuno-gold electron microscopy. There was a statistically significant difference (35%) in the basal extracellular level of striatal glutamate between the HR and LR groups, with the HR group having a lower level, compared with that of the LR group. There was a 25% difference in the density of nerve terminal glutamate immuno-gold labeling associated with the synaptic vesicle pool in the HR, compared with that in the LR group, but this difference was not statistically significant. There was no change in the basal extracellular level of striatal dopamine between the two groups, but there was a statistically significant difference (73%) in the basal turnover ratio of striatal dopamine and its metabolites in the HR, compared with that in the LR group. The data suggests that the difference in extracellular striatal glutamate between the HR and LR groups is not due to an alteration in basal extracellular dopamine but could be due to an increase in dopamine turnover.
Collapse
Affiliation(s)
- Saate S Shakil
- Research Services, Neurocytology Lab, V.A. Medical Center, Portland, OR 97239
| | | | | | | | | | | | | |
Collapse
|
78
|
Holmer HK, Keyghobadi M, Moore C, Menashe RA, Meshul CK. Dietary restriction affects striatal glutamate in the MPTP-induced mouse model of nigrostriatal degeneration. Synapse 2005; 57:100-12. [PMID: 15906381 DOI: 10.1002/syn.20163] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
One month following subchronic treatment with the neurotoxin, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) (30 mg/kg/d x 7 days), there is a decrease in the extracellular level of striatal glutamate. It has been reported that following dietary restriction (DR) (fed on alternate days) of C57BL/6 mice, MPTP administration resulted in a reduction in the loss of tyrosine hydroxylase-positive neurons within the substantia nigra pars compacta (SN-PC) compared to the ad libitum (AL)-fed MPTP-treated mice. However, there have been no reports of whether the MPTP-induced alterations in brain neurochemistry or morphology can be similarly attenuated by DR if initiated after administration of the toxin. In the MPTP/AL group there is a decrease in the extracellular level of striatal glutamate compared to the Vehicle/AL group. However, 21 days of DR starting 1 day after the last subchronic dose of MPTP results in a reversal in the extracellular level of striatal glutamate compared to the MPTP/AL group. DR alone resulted in a decrease in extracellular striatal glutamate. There was no change in the relative density of the glutamate transporter, GLT-1, within the striatum or SN-PC between any of the groups, suggesting that the alterations in striatal extracellular glutamate were not due to a change in this specific transporter. There was an increase in the density of nerve terminal glutamate immunolabeling in the MPTP/AL and MPTP/DR groups compared to the Vehicle/AL group. There was a similar decrease in the relative density of tyrosine hydroxylase immunolabeling within the striatum and the SN-PC in both the MPTP/AL and MPTP/DR groups compared to the Vehicle/AL group. Since a decrease in the activity of the corticostriatal glutamate pathway has been reported in both Parkinson's disease and in animal models of nigrostriatal loss, these data suggest that DR initiated after the partial loss of striatal dopamine appears to reverse the decrease in striatal glutamate.
Collapse
Affiliation(s)
- Haley K Holmer
- Research Services, Neurocytology Lab, V.A. Medical Center, Portland, Oregon 97239, USA
| | | | | | | | | |
Collapse
|
79
|
Guebel DV, Torres NV. Dynamics of sulfur amino acids in mammalian brain: assessment of the astrocytic-neuronal cysteine interaction by a mathematical hybrid model. Biochim Biophys Acta Gen Subj 2004; 1674:12-28. [PMID: 15342110 DOI: 10.1016/j.bbagen.2004.05.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2003] [Revised: 05/07/2004] [Accepted: 05/11/2004] [Indexed: 12/29/2022]
Abstract
A mathematically hybrid model was used to analyze three mechanisms by which cysteine could be produced in the brain to be used as preferential substrate in the synthesis of neuronal glutathione. In that way, the fluxes of sulfur-compounds at the brain-blood barrier were integrated with their transport in astrocytes and neurons, and with their metabolism in astrocytes. We concluded that cysteine, in contrast with its precursor cystine, would not be taken up from the blood at the blood-brain barrier, but instead it must be lost continuously from astrocytes. Cysteine efflux is produced because the uptake of cystine in astrocytes is much greater than their cysteine demand to synthesize glutathione, hypotaurine and taurine. Once in the interstitial parenchyma, cysteine would be taken for the neurons, as backwardly by the endothelial cells. Remarkably, a close sulfur-macro balance can be maintained only if the surplus of the produced cysteine is transferred from the endothelial cells to the blood together with significant amounts of other sulfur-compounds, probably taurine and hypotaurine. In addition, the results obtained shown that alternative mechanisms of cysteine generation (i.e., nonenzymatic-thiol-disulfide exchange reaction, enzymatic cleavage of the glutathione effluxed from astrocytes) are not quantitatively significant under physiological conditions, in situ.
Collapse
Affiliation(s)
- Daniel V Guebel
- Biotechnology Counseling Services, Av San Martín 4927, (1417) Buenos Aires, Argentina
| | | |
Collapse
|
80
|
Sullivan R, Rauen T, Fischer F, Wiessner M, Grewer C, Bicho A, Pow DV. Cloning, transport properties, and differential localization of two splice variants of GLT-1 in the rat CNS: implications for CNS glutamate homeostasis. Glia 2004; 45:155-69. [PMID: 14730709 DOI: 10.1002/glia.10317] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
At least two splice variants of GLT-1 are expressed by rat brain astrocytes, albeit in different membrane domains. There is at present only limited data available as to the spatial relationship of such variants relative to the location of synapses and their functional properties. We have characterized the transport properties of GLT-1v in a heterologous expression system and conclude that its transport properties are similar to those of the originally described form of GLT-1, namely GLT-1alpha. We demonstrate that GLT-1alpha is localized to glial processes, some of which are interposed between multiple synapse types, including GABAergic synapses, whereas GLT-1v is expressed by astrocytic processes, at sites not interposed between synapses. Both splice variants can be expressed by a single astrocyte, but such expression is not uniform over the surface of the astrocytes. Neither splice variant of GLT-1 is evident in brain neurons, but both are abundantly expressed in some retinal neurons. We conclude that GLT-1v may not be involved in shaping the kinetics of synaptic signaling in the brain, but may be critical in preventing spillover of glutamate between adjacent synapses, thereby regulating intersynaptic glutamatergic and GABAergic transmission. Furthermore, GLT-1v may be crucial in ensuring that low levels of glutamate are maintained at extrasynaptic locations, especially in pathological conditions such as ischemia, motor neurone disease, and epilepsy.
Collapse
Affiliation(s)
- Robert Sullivan
- School of Biomedical Sciences, Department of Physiology and Pharmacology, University of Queensland, Brisbane 4072, Australia
| | | | | | | | | | | | | |
Collapse
|
81
|
Chen W, Mahadomrongkul V, Berger UV, Bassan M, DeSilva T, Tanaka K, Irwin N, Aoki C, Rosenberg PA. The glutamate transporter GLT1a is expressed in excitatory axon terminals of mature hippocampal neurons. J Neurosci 2004; 24:1136-48. [PMID: 14762132 PMCID: PMC2849838 DOI: 10.1523/jneurosci.1586-03.2004] [Citation(s) in RCA: 217] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
GLT1 is the major glutamate transporter of the brain and has been thought to be expressed exclusively in astrocytes. Although excitatory axon terminals take up glutamate, the transporter responsible has not been identified. GLT1 is expressed in at least two forms varying in the C termini, GLT1a and GLT1b. GLT1 mRNA has been demonstrated in neurons, without associated protein. Recently, evidence has been presented, using specific C terminus-directed antibodies, that GLT1b protein is expressed in neurons in vivo. These data suggested that the GLT1 mRNA detected in neurons encodes GLT1b and also that GLT1b might be the elusive presynaptic transporter. To test these hypotheses, we used variant-specific probes directed to the 3'-untranslated regions for GLT1a and GLT1b to perform in situ hybridization in the hippocampus. Contrary to expectation, GLT1a mRNA was the more abundant form. To investigate further the expression of GLT1 in neurons in the hippocampus, antibodies raised against the C terminus of GLT1a and against the N terminus of GLT1, found to be specific by testing in GLT1 knock-out mice, were used for light microscopic and EM-ICC. GLT1a protein was detected in neurons, in 14-29% of axons in the hippocampus, depending on the region. Many of the labeled axons formed axo-spinous, asymmetric, and, thus, excitatory synapses. Labeling also occurred in some spines and dendrites. The antibody against the N terminus of GLT1 also produced labeling of neuronal processes. Thus, the originally cloned form of GLT1, GLT1a, is expressed as protein in neurons in the mature hippocampus and may contribute significantly to glutamate uptake into excitatory terminals.
Collapse
Affiliation(s)
- Weizhi Chen
- Department of Neurology, Children's Hospital, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
82
|
King N, Lin H, McGivan JD, Suleiman MS. Aspartate transporter expression and activity in hypertrophic rat heart and ischaemia-reperfusion injury. J Physiol 2004; 556:849-58. [PMID: 14766933 PMCID: PMC1664986 DOI: 10.1113/jphysiol.2004.060616] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
This study's rationale was that the expression and activity of aspartate transporters in hypertrophied hearts might be different from normal hearts, which could affect the use of aspartate in myocardial protection of hypertrophied hearts. mRNA expression of system X(ag)(-) transporters in hearts from normal (Wistar Kyoto) and hypertrophied (spontaneously hypertensive rat) rats was investigated by RT-PCR. EAAT3 protein expression in isolated cells and vesicles from normal and hypertrophied hearts was investigated by Western blotting. The same vesicles were also used to measure aspartate uptake. The effects of 0.5 mmol l(-1) aspartate supplementation on cardiac performance during ischaemia-reperfusion were investigated in isolated and perfused hearts. Both normal and hypertrophied hearts expressed EAAT1 and EAAT3 mRNA. EAAT3 protein expression was significantly greater in cells and vesicles from hypertrophied hearts compared to normal hearts. The velocity (V(max)) of aspartate uptake was faster at 24.4 +/- 2.2 pmol mg(-1) s(-1) in vesicles from hypertrophied hearts compared to 8.2 +/- 0.8 pmol mg(-1) s(-1) (P < 0.001, t test, n= 6, means +/-s.e.m.) in normal heart vesicles. The affinity (K(m)) was similar for both preparations. When recoveries were matched, 0.5 mmol l(-1) aspartate addition reduced reperfusion injury and increased functional recovery of hypertrophied hearts but not normal hearts. This was associated with a greater preservation of ATP, glutamate and glutamine and less lactate production during ischaemia in aspartate-treated hypertrophied hearts compared to all other experimental groups. These results suggest that increased aspartate transporter expression and activity in hypertrophy helps facilitate aspartate entry into hypertrophied cardiomyocytes, which in turn leads to improved myocardial protection.
Collapse
Affiliation(s)
- Nicola King
- Bristol Heart Institute, University of Bristol, Bristol Royal Infirmary, Bristol BS2 8HW, UK.
| | | | | | | |
Collapse
|
83
|
Abstract
Activation of cannabinoid CB1 receptors reduces glutamatergic synaptic transmission in the rodent striatum and is involved in the normal control of motor function by the basal ganglia. Here we investigated CB1 receptor regulation of glutamate release and uptake and synaptic transmission in the rat striatum. We show that CB1 receptor activation reduces both the release and uptake of [3H]glutamate in striatal slices. We also demonstrate that both activation of CB1 receptors and inhibition of glutamate uptake reduce corticostriatal synaptic transmission in a mutually occlusive manner and that both forms of depression are dependent on metabotropic glutamate receptor (mGluR) activation. We propose that CB1 receptor activation in the striatum decreases glutamate transporter activity and that the resulting increase in synaptic cleft glutamate concentration causes the activation of presynaptic mGluRs, which then decrease glutamate release.
Collapse
|
84
|
Hu WH, Walters WM, Xia XM, Karmally SA, Bethea JR. Neuronal glutamate transporter EAAT4 is expressed in astrocytes. Glia 2003; 44:13-25. [PMID: 12951653 DOI: 10.1002/glia.10268] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
High-affinity excitatory amino acid transporters (EAATs) are essential to terminate glutamatergic neurotransmission and to prevent excitotoxicity. To date, five distinct EAATs have been cloned from animal and human tissues: GLAST (EAAT1), GLT-1 (EAAT2), EAAC1 (EAAT3), EAAT4, and EAAT5. EAAT1 and EAAT2 are commonly known as glial glutamate transporters, whereas EAAT3, EAAT4, and EAAT5 are neuronal. EAAT4 is largely expressed in cerebellar Purkinje cells. In this study, using immunohistochemistry and Western blotting, we found that EAAT4-like immunoreactivity (ir) is enriched in the spinal cord and forebrain. Double-labeled fluorescent immunostaining and confocal image analysis indicated that EAAT4-like ir colocalizes with an astrocytic marker, glial fibrillary acidic protein (GFAP). The astrocytic localization of EAAT4 was further confirmed in astrocyte cultures by double-labeled fluorescent immunocytochemistry and Western blotting. Reverse transcriptase-polymerase chain reaction analysis demonstrated mRNA expression of EAAT4 in astrocyte cultures. Sequencing confirmed the specificity of the amplified fragment. These results demonstrate that EAAT4 is expressed in astrocytes. This astrocytic localization of neuronal EAAT4 may reveal a new function of EAAT4 in the central nervous system.
Collapse
Affiliation(s)
- Wen-Hui Hu
- The Miami Project to Cure Paralysis, University of Miami School of Medicine, Miami, Florida 33136, USA
| | | | | | | | | |
Collapse
|
85
|
Mennini T, Fumagalli E, Gobbi M, Fattorusso C, Catalanotti B, Campiani G. Substrate inhibitors and blockers of excitatory amino acid transporters in the treatment of neurodegeneration: critical considerations. Eur J Pharmacol 2003; 479:291-6. [PMID: 14612159 DOI: 10.1016/j.ejphar.2003.08.078] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Excessive glutamate release (mediated by reversed uptake) or impaired reuptake contributes to the etiopathology of many neurodegenerative disorders. Thus great effort has been devoted to the discovery of agents that can interfere with high-affinity Na+-dependent glutamate transport, with the aim of finding new therapeutics against neurodegenerative diseases. We developed two different 3D-pharmacophore models for substrate inhibitors and blockers, which led to the rational design of novel and potent glutamate and aspartate analogues that selectively interact with excitatory amino acid transporters (EAAT). Our results indicated that all analysed EAAT ligands share the same orientation of the acidic functions and the protonatable nitrogen, even though the distance between the carboxylic carbons varies from 3.7 to 4.9 A. This distance does not discriminate between substrate inhibitors and blockers, but between glutamate and aspartate derivatives. In contrasts differences in the volume distribution of the rest of the molecule with respect to the axis connecting the two carboxylic groups are responsible for the difference in activity between transportable and nontransportable inhibitors. Thus our 3D receptor interaction model for EAAT substrates and nontransportable inhibitors could lead to the rational design of selective EAAT ligands as possible neuroprotective agents. However, some critical points, such as which glutamate transporter is present on glutamatergic nerve terminals and which glutamate transporter mediates reversed glutamate uptake, still remain to be elucidated.
Collapse
Affiliation(s)
- Tiziana Mennini
- Laboratory Receptor Pharmacology, Mario Negri Institute for Pharmacological Research, Via Eritrea 62, 20157, Milan, Italy.
| | | | | | | | | | | |
Collapse
|
86
|
Fontana ACK, Guizzo R, de Oliveira Beleboni R, Meirelles E Silva AR, Coimbra NC, Amara SG, dos Santos WF, Coutinho-Netto J. Purification of a neuroprotective component of Parawixia bistriata spider venom that enhances glutamate uptake. Br J Pharmacol 2003; 139:1297-309. [PMID: 12890709 PMCID: PMC1573954 DOI: 10.1038/sj.bjp.0705352] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
(1) In this study, we examined the effects of crude venom from the spider Parawixia bistriata on glutamate and GABA uptake into synaptosomes prepared from rat cerebral cortex. Addition of venom to cortical synaptosomes stimulated glutamate uptake and inhibited GABA uptake in a concentration-dependent manner. (2) The venom was fractionated using reverse-phase high-performance liquid chromatography on a preparative column. The fraction that retained glutamate uptake-stimulating activity was further purified on a reverse-phase analytical column followed by ion-exchange chromatography. (3) The active fraction, referred to as PbTx1.2.3, stimulated glutamate uptake in synaptosomes without changing the K(M) value, and did not affect GABA uptake. Additional experiments showed that the enhancement of glutamate uptake by PbTx1.2.3 occurs when ionotropic glutamate receptors or voltage-gated sodium and calcium channels are completely inhibited or when GABA receptors and potassium channels are activated, indicating that the compound may have a direct action on the transporters. (4) In an experimental model for glaucoma in which rat retinas are subjected to ischemia followed by reperfusion, PbTx1.2.3 protected neurons from excitotoxic death in both outer and inner nuclear layers, and ganglion cell layers. (5) This active spider venom component may serve as a basis for designing therapeutic drugs that increase glutamate clearance and limit neurodegeneration.
Collapse
Affiliation(s)
- Andréia Cristina Karklin Fontana
- Laboratory of Neurochemistry, Department of Biochemistry and Immunology, Ribeirão Preto School of Medicine, University of São Paulo, Brazil.
| | | | | | | | | | | | | | | |
Collapse
|