Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Free Radic Biol Med ; 77: 168-82, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25224033

ABSTRACT

The contribution of oxidative stress to ischemic brain damage is well established. Nevertheless, for unknown reasons, several clinically tested antioxidant therapies have failed to show benefits in human stroke. Based on our previous in vitro work, we hypothesized that the neuroprotective potency of antioxidants is related to their ability to limit the release of the excitotoxic amino acids glutamate and aspartate. We explored the effects of two antioxidants, tempol and edaravone, on amino acid release in the brain cortex, in a rat model of transient occlusion of the middle cerebral artery (MCAo). Amino acid levels were quantified using a microdialysis approach, with the probe positioned in the ischemic penumbra as verified by a laser Doppler technique. Two-hour MCAo triggered a dramatic increase in the levels of glutamate, aspartate, taurine, and alanine. Microdialysate delivery of 10mM tempol reduced the amino acid release by 60-80%, whereas matching levels of edaravone had no effect. In line with these data, an intracerebroventricular injection of tempol but not edaravone (500 nmol each, 15 min before MCAo) reduced infarction volumes by ~50% and improved neurobehavioral outcomes. In vitro assays showed that tempol was superior at removing superoxide anion, whereas edaravone was more potent at scavenging hydrogen peroxide, hydroxyl radical, and peroxynitrite. Overall, our data suggest that the neuroprotective properties of tempol are probably related to its ability to reduce tissue levels of the superoxide anion and pathological glutamate release and, in such a way, limit progression of brain infarction within ischemic penumbra. These new findings may be instrumental in developing new antioxidant therapies for treatment of stroke.


Subject(s)
Cyclic N-Oxides/pharmacology , Glutamic Acid/metabolism , Infarction, Middle Cerebral Artery/drug therapy , Neuroprotective Agents/pharmacology , Alanine/metabolism , Animals , Antipyrine/analogs & derivatives , Antipyrine/chemistry , Antipyrine/pharmacology , Astrocytes/metabolism , Brain/drug effects , Brain/pathology , Cells, Cultured , Cyclic N-Oxides/chemistry , Drug Evaluation, Preclinical , Edaravone , Free Radical Scavengers/chemistry , Free Radical Scavengers/pharmacology , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Male , Molecular Mimicry , Neuroprotective Agents/chemistry , Oxidative Stress , Rats, Sprague-Dawley , Spin Labels , Superoxides/metabolism , Synaptosomes/drug effects , Taurine/metabolism
2.
J Physiol ; 592(22): 4855-62, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25172945

ABSTRACT

In mammals, cellular swelling activates release of small organic osmolytes, including the excitatory amino acids (EAA) glutamate and aspartate, via a ubiquitously expressed volume-regulated chloride/anion channel (VRAC). Pharmacological evidence suggests that VRAC plays plural physiological and pathological roles, including excitotoxic release of glutamate in stroke. However, the molecular identity of this pathway was unknown. Two recent studies discovered that LRRC8 gene family members encode heteromeric VRAC composed of LRRC8A plus LRRC8B-E, which mediate swelling-activated Cl(-) currents and taurine release in human non-neural cells (Z. Qiu et al. Cell 157: 447, 2014; F.K. Voss et al. Science 344: 634, 2014). Here, we tested the contribution of LRRC8A to the EAA release in brain glia. We detected and quantified expression levels of LRRC8A-E in primary rat astrocytes with quantitative RT-PCR and then downregulated LRRC8A with gene-specific siRNAs. In astrocytes exposed to hypo-osmotic media, LRRC8A knockdown dramatically reduced swelling-activated release of the EAA tracer D-[(3)H]aspartate. In parallel HPLC assays, LRRC8A siRNA prevented hypo-osmotic media-induced loss of the endogenous intracellular L-glutamate and taurine. Furthermore, downregulation of LRRC8A completely ablated the ATP-stimulated release of D-[(3)H]aspartate and [(14)C]taurine from non-swollen astrocytes. Overall, these data indicate that LRRC8A is an indispensable component of a permeability pathway that mediates both swelling-activated and agonist-induced amino acid release in brain glial cells.


Subject(s)
Aspartic Acid/metabolism , Astrocytes/metabolism , Glutamic Acid/metabolism , Membrane Proteins/metabolism , Adenosine Triphosphate/metabolism , Animals , Cells, Cultured , Membrane Proteins/genetics , Osmotic Pressure , Rats , Rats, Sprague-Dawley , Taurine/metabolism
3.
Biochem J ; 458(3): 575-83, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24341532

ABSTRACT

The proposed Ca2+-activated Cl- channel protein Best1 (bestrophin 1) is expressed and functionally important in the retina and in the brain. Human BEST1 has two known splice variants, Best1V1 and Best1V2, which arise from alternative splicing of two exons: exon 2 splicing results in a unique N-terminal domain, whereas alternative splicing of exon 11 produces two mutually exclusive C-termini. Prior studies were limited to Best1V1 and its clinically relevant mutations. In the present work, we cloned a novel splice variant of Best1V1 missing exon 2 (Best1V1Δex2) and differing from each of the two previously identified isoforms by one alternatively spliced domain. This finding allowed us to determine the role for alternative splicing of the Best1 N- and C-termini. We heteroexpressed Best1V1Δex2 in HEK (human embryonic kidney)-293 cells, and compared its properties with Best1V1 and Best1V2. Western blot analysis confirmed protein expression from all three splice variants. Both Best1V1 and Best1V1Δex2 successfully formed Ca2+-activated Cl- channels, demonstrating that the N-terminus encoded by exon 2 is not essential for channel function. In contrast, Best1V2-expressing cells had no detectable Ca2+-activated Cl- currents, pointing to a critical role for splicing of the C-terminus. Surface protein biotinylation demonstrated that Best1V1 and Best1V1Δex2 are trafficked to the plasma membrane, whereas Best1V2 is not. These results define the impact of alternative splicing on Best1 function, and should be taken into consideration in future modelling of the Best1 protein structure.


Subject(s)
Alternative Splicing , Chloride Channels/genetics , Eye Proteins/genetics , Astrocytes/metabolism , Bestrophins , Cell Line, Tumor , Chloride Channels/metabolism , Cloning, Molecular , Exons , Eye Proteins/metabolism , Glioma/metabolism , HEK293 Cells , Humans , Neuroglia/metabolism , Protein Isoforms/metabolism
4.
Biomaterials ; 35(5): 1439-49, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24246642

ABSTRACT

Bioengineered fiber substrates are increasingly studied as a means to promote regeneration and remodeling in the injured central nervous system (CNS). Previous reports largely focused on the ability of oriented scaffolds to bridge injured regions and direct outgrowth of axonal projections. In the present work, we explored the effects of electrospun microfibers on the migration and physiological properties of brain astroglial cells. Primary rat astrocytes were cultured on either fibronectin-coated poly-L-lactic acid (PLLA) films, fibronectin-coated randomly oriented PLLA electrospun fibers, or fibronectin-coated aligned PLLA electrospun fibers. Aligned PLLA fibers strongly altered astrocytic morphology, orienting cell processes, actin microfilaments, and microtubules along the length of the fibers. On aligned fibers, astrocytes also significantly increased their migration rates in the direction of fiber orientation. We further investigated if fiber topography modifies astrocytic neuroprotective properties, namely glutamate and glutamine transport and metabolism. This was done by quantifying changes in mRNA expression (qRT-PCR) and protein levels (Western blotting) for a battery of relevant biomolecules. Interestingly, we found that cells grown on random and/or aligned fibers increased the expression levels of two glutamate transporters, GLAST and GLT-1, and an important metabolic enzyme, glutamine synthetase, as compared to the fibronectin-coated films. Functional assays revealed increases in glutamate transport rates due to GLT-1 mediated uptake, which was largely determined by the dihydrokainate-sensitive GLT-1. Overall, this study suggests that aligned PLLA fibers can promote directed astrocytic migration, and, of most importance, our in vitro results indicate for the first time that electrospun PLLA fibers can positively modify neuroprotective properties of glial cells by increasing rates of glutamate uptake.


Subject(s)
Astrocytes/metabolism , Excitatory Amino Acid Transporter 2/physiology , Fibronectins/chemistry , Glutamic Acid/metabolism , Lactic Acid/chemistry , Polymers/chemistry , Animals , Astrocytes/cytology , Cell Adhesion , Cells, Cultured , Polyesters , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Tissue Scaffolds
5.
Pflugers Arch ; 465(9): 1249-60, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23515871

ABSTRACT

The Ca(2+) sensor stromal interacting molecule 1 (STIM1) and the Ca(2+) channel Orai1 mediate the ubiquitous store-operated Ca(2+) entry (SOCE) pathway activated by depletion of internal Ca(2+) stores and mediated through the highly Ca(2+)-selective, Ca(2+) release-activated Ca(2+) (CRAC) current. Furthermore, STIM1 and Orai1, along with Orai3, encode store-independent Ca(2+) currents regulated by either arachidonate or its metabolite, leukotriene C4. Orai channels are emerging as important contributors to numerous cell functions, including proliferation, migration, differentiation, and apoptosis. Recent studies suggest critical involvement of STIM/Orai proteins in controlling the development of several cancers, including malignancies of the breast, prostate, and cervix. Here, we quantitatively compared the magnitude of SOCE and the expression levels of STIM1 and Orai1 in non-malignant human primary astrocytes (HPA) and in primary human cell lines established from surgical samples of the brain tumor glioblastoma multiforme (GBM). Using Ca(2+) imaging, patch-clamp electrophysiology, pharmacological reagents, and gene silencing, we established that in GBM cells, SOCE and CRAC are mediated by STIM1 and Orai1. We further found that GBM cells show upregulation of SOCE and increased Orai1 levels compared to HPA. The functional significance of SOCE was evaluated by studying the effects of STIM1 and Orai1 knockdown on cell proliferation and invasion. Utilizing Matrigel assays, we demonstrated that in GBM, but not in HPA, downregulation of STIM1 and Orai1 caused a dramatic decrease in cell invasion. In contrast, the effects of STIM1 and Orai1 knockdown on GBM cell proliferation were marginal. Overall, these results demonstrate that STIM1 and Orai1 encode SOCE and CRAC currents and control invasion of GBM cells. Our work further supports the potential use of channels contributed by Orai isoforms as therapeutic targets in cancer.


Subject(s)
Brain Neoplasms/metabolism , Calcium Channels/metabolism , Calcium Signaling , Glioblastoma/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Action Potentials , Astrocytes/metabolism , Brain Neoplasms/pathology , Calcium/metabolism , Calcium Channels/genetics , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Humans , Membrane Proteins/genetics , Neoplasm Invasiveness , Neoplasm Proteins/genetics , ORAI1 Protein , Stromal Interaction Molecule 1 , Transcription, Genetic , Up-Regulation
6.
Am J Physiol Cell Physiol ; 301(4): C814-22, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21734190

ABSTRACT

Here we report and validate a simple method for measuring intracellular activities of glial glutamine synthetase (GS) and glutaminase (GLNase) in intact glial cells. These enzymes are responsible for glutamate and glutamine recycling in the brain, where glutamate and glutamine transport from the blood stream is strongly limited by the blood-brain barrier. The intracellular levels of glutamate and glutamine are dependent on activities of numerous enzymatic processes, including 1) cytosolic production of glutamine from glutamate by GS, 2) production of glutamate from glutamine by GLNase that is primarily localized between mitochondrial membranes, and 3) mitochondrial conversion of glutamate to the tricarboxylic cycle intermediate α-ketoglutarate in the reactions of oxidative deamination and transamination. We measured intracellular activities of GS and GLNase by quantifying enzymatic interconversions of L-[(3)H]glutamate and L-[(3)H]glutamine in cultured rat astrocytes. The intracellular substrate and the products of enzymatic reactions were separated in one step using commercially available anion exchange columns and quantified using a scintillation counter. The involvement of GS and GLNase in the conversion of (3)H-labeled substrates was verified using irreversible pharmacological inhibitors for each of the enzymes and additionally validated by measuring intracellular amino acid levels using an HPLC. Overall, this paper describes optimized conditions and pharmacological controls for measuring GS and GLNase activities in intact glial cells.


Subject(s)
Astrocytes/metabolism , Enzyme Assays/methods , Glutamate-Ammonia Ligase/metabolism , Glutaminase/metabolism , Neuroglia/enzymology , Animals , Astrocytes/cytology , Diazooxonorleucine/pharmacology , Glutamate-Ammonia Ligase/antagonists & inhibitors , Glutamate-Ammonia Ligase/genetics , Glutaminase/antagonists & inhibitors , Glutaminase/genetics , Methionine Sulfoximine/pharmacology , Neuroglia/metabolism , Rats , Rats, Sprague-Dawley
7.
J Neurochem ; 118(1): 140-52, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21517854

ABSTRACT

In our previous work, we found that perfusion of the rat cerebral cortex with hypo-osmotic medium triggers massive release of the excitatory amino acid L-glutamate but decreases extracellular levels of L-glutamine (R. E. Haskew-Layton et al., PLoS ONE, 3: e3543). The release of glutamate was linked to activation of volume-regulated anion channels, whereas mechanism(s) responsible for alterations in extracellular glutamine remained unclear. When mannitol was added to the hypo-osmotic medium to reverse reductions in osmolarity, changes in microdialysate levels of glutamine were prevented, indicating an involvement of cellular swelling. As the main source of brain glutamine is astrocytic synthesis and export, we explored the impact of hypo-osmotic medium on glutamine synthesis and transport in rat primary astrocyte cultures. In astrocytes, a 40% reduction in medium osmolarity moderately stimulated the release of L-[(3) H]glutamine by ∼twofold and produced no changes in L-[(3) H]glutamine uptake. In comparison, hypo-osmotic medium stimulated the release of glutamate (traced with D-[(3) H]aspartate) by more than 20-fold. In whole-cell enzymatic assays, we discovered that hypo-osmotic medium caused a 20% inhibition of astrocytic conversion of L-[(3) H]glutamate into L-[(3) H]glutamine by glutamine synthetase. Using an HPLC assay, we further found a 35% reduction in intracellular levels of endogenous glutamine. Overall, our findings suggest that cellular swelling (i) inhibits astrocytic glutamine synthetase activity, and (ii) reduces substrate availability for this enzyme because of the activation of volume-regulated anion channels. These combined effects likely lead to reductions in astrocytic glutamine export in vivo and may partially explain occurrence of hyperexcitability and seizures in human hyponatremia.


Subject(s)
Astrocytes/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Glutamic Acid/metabolism , Glutamine/metabolism , Animals , Animals, Newborn , Aspartic Acid/metabolism , Cells, Cultured , Cerebral Cortex/drug effects , Chromatography, High Pressure Liquid/methods , Dose-Response Relationship, Drug , Extracellular Fluid/metabolism , Glutamate-Ammonia Ligase/metabolism , Glutaminase/metabolism , Male , Microdialysis/methods , Models, Biological , Rats , Rats, Sprague-Dawley , Saline Solution, Hypertonic/pharmacology , Tritium/metabolism
8.
Channels (Austin) ; 2(3): 191-201, 2008.
Article in English | MEDLINE | ID: mdl-18836298

ABSTRACT

Almost all lipid-exposed transmembrane domains of integral proteins contain aromatic residues flanking the hydrophobic segment of the domains. These residues generally reside close to the carbonyl region of the membrane, and several structural and functional roles have been associated to these residues. Although the roles and physicochemical reasons for aromatic preference have been extensively studied using model systems, few studies have been done in a native membrane system. To gain insight about the mechanistic implication for this aromatic preference, we selected position alpha F426 of the muscle-type nicotinic acetylcholine receptor (nAChR). alpha F426 is a lipid-exposed residue at the extracellular segment of the alpha M4 transmembrane domain and is highly conserved among different nAChR subunits and species. We used site-directed mutagenesis, alpha-Bungarotoxin-binding assay, and two-electrodes voltage clamp in Xenopus laevis oocytes to characterize mutations at position alpha F426, which impart different physicochemical properties like volume, polarity, hydrogen bonds, aromaticity and net electrical charge. All mutations except the aromatic residues resulted in a significant reduction of the nAChR cell-surface levels and the macroscopic currents to acetylcholine. These results suggest that position alpha F426 contributes to structural stability and open-close transitions of the nAChR. Finally, the present study also provides information about how intermolecular interactions at position alpha 426 modulate open-close transitions of the nAChR.


Subject(s)
Hydrocarbons/chemistry , Receptors, Nicotinic/chemistry , Water/chemistry , Amino Acids/chemistry , Animals , Bungarotoxins/chemistry , Cell Membrane/metabolism , Humans , Lipids/chemistry , Mice , Models, Biological , Models, Molecular , Molecular Conformation , Mutagenesis, Site-Directed , Receptors, Nicotinic/metabolism , Xenopus laevis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...