Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters

Publication year range
1.
Neurochem Res ; 40(12): 2461-72, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25972039

ABSTRACT

Synaptically released L-glutamate, the most important excitatory neurotransmitter in the CNS, is removed from extracellular space by fast and efficient transport mediated by several transporters; the most abundant ones are EAAT1/GLAST and EAAT2/GLT1. The review first summarizes their location, functions and basic characteristics. We then look at genetics and epigenetics of EAAT1/GLAST and EAAT2/GLT1 and perform in silico analyses of their promoter regions. There is one CpG island in SLC1A2 (EAAT2/GLT1) gene and none in SLC1A3 (EAAT1/GLAST) suggesting that DNA methylation is not the most important epigenetic mechanism regulating EAAT1/GLAST levels in brain. There are targets for specific miRNA in SLC1A2 (EAAT2/GLT1) gene. We also note that while defects in EAAT2/GLT1 have been associated with various pathological states including chronic neurodegenerative diseases, very little is known on possible contributions of defective or dysfunctional EAAT1/GLAST to any specific brain disease. Finally, we review evidence of EAAT1/GLAST involvement in mechanisms of brain response to alcoholism and present some preliminary data showing that ethanol, at concentrations which may be reached following heavy drinking, can have an effect on the distribution of EAAT1/GLAST in cultured astrocytes; the effect is blocked by baclofen, a GABA-B receptor agonist and a drug potentially useful in the treatment of alcoholism. We argue that more research effort should be focused on EAAT1/GLAST, particularly in relation to alcoholism and drug addiction.


Subject(s)
Brain Chemistry/genetics , Epigenesis, Genetic/genetics , Epigenesis, Genetic/physiology , Excitatory Amino Acid Transporter 1/genetics , Excitatory Amino Acid Transporter 1/metabolism , Glutamic Acid/metabolism , Alcoholism/genetics , Alcoholism/metabolism , Animals , Biological Transport, Active , Glutamate Plasma Membrane Transport Proteins/metabolism , Humans
2.
Neurochem Res ; 37(11): 2364-78, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22528834

ABSTRACT

Glial fibrillary acidic protein (GFAP) is an intermediate filament protein expressed in the astrocyte cytoskeleton that plays an important role in the structure and function of the cell. GFAP can be phosphorylated at six serine (Ser) or threonine (Thr) residues but little is known about the role of GFAP phosphorylation in physiological and pathophysiological states. We have generated antibodies against two phosphorylated GFAP (pGFAP) proteins: p8GFAP, where GFAP is phosphorylated at Ser-8 and p13GFAP, where GFAP is phosphorylated at Ser-13. We examined p8GFAP and p13GFAP expression in the control neonatal pig brain and at 24 and 72 h after an hypoxic-ischemic (HI) insult. Immunohistochemistry demonstrated pGFAP expression in astrocytes with an atypical cytoskeletal morphology, even in control brains. Semi-quantitative western blotting revealed that p8GFAP expression was significantly increased at 24 h post-insult in HI animals with seizures in frontal, parietal, temporal and occipital cortices. At 72 h post-insult, p8GFAP and p13GFAP expression were significantly increased in HI animals with seizures in brain regions that are vulnerable to cellular damage (cortex and basal ganglia), but no changes were observed in brain regions that are relatively spared following an HI insult (brain stem and cerebellum). Increased pGFAP expression was associated with poor neurological outcomes such as abnormal encephalography and neurobehaviour, and increased histological brain damage. Phosphorylation of GFAP may play an important role in astrocyte remodelling during development and disease and could potentially contribute to the plasticity of the central nervous system.


Subject(s)
Animals, Newborn , Glial Fibrillary Acidic Protein/metabolism , Hypoxia-Ischemia, Brain/metabolism , Animals , Blotting, Western , Electroencephalography , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/physiopathology , Immunohistochemistry , Phosphorylation , Swine
3.
Curr Mol Pharmacol ; 14(2): 138-149, 2021.
Article in English | MEDLINE | ID: mdl-32329706

ABSTRACT

We present an overview of genetic, metabolomic, proteomic and neurochemical studies done mainly in our laboratories that could improve prediction, mechanistic understanding and possibly extend to diagnostics and treatment of alcoholism and alcohol addiction. Specific polymorphisms in genes encoding for interleukins 2 and 6, catechol-O-methyl transferase (COMT), monaminooxidase B (MAO B) and several other enzymes were identified as associated with altered risks of alcoholism in humans. A polymorphism in the gene for BDNF has been linked to the risk of developing deficiences in colour vision sometimes observed in alcoholics. Metabolomic studies of acute ethanol effects on guinea pig brain cortex in vitro, lead to the identification of specific subtypes of GABA(A) receptors involved in the actions of alcohol at various doses. Acute alcohol affected energy metabolism, oxidation and the production of actaldehyde and acetate; this could have specific consequences not only for the brain energy production/utilization but could influence the cytotoxicity of alcohol and impact the epigenetics (histone acetylation). It is unlikely that brain metabolism of ethanol occurs to any significant degree; the reduction in glucose metabolism following alcohol consumption is due to ethanol effects on receptors, such as α4ß3δ GABA(A) receptors. Metabolomics using post-mortem human brain indicated that the catecholaminergic signalling may be preferentially affected by chronic excessive drinking. Changes in the levels of glutathione were consistent with the presence of severe oxidative stress. Proteomics of the post-mortem alcoholic brains identified a large number of proteins, the expression of which was altered by chronic alcohol, with those associated with brain energy metabolism among the most numerous. Neurochemical studies found the increased expression of glutamate transporter GLAST/EAAT1 in brain as one of the largest changes caused by alcoholism. Given that GLAST/EAAT1 is one of the most abundant proteins in the nervous tissue and is intimately associated with the function of the excitatory (glutamatergic) synapses, this may be among the most important effects of chronic alcohol on brain function. It has so far been observed mainly in the prefrontal cortex. We show several experiments suggesting that acute alcohol can translocate GLAST/EAAT1 in astrocytes towards the plasma membrane (and this effect is inhibited by the GABA(B) agonist baclofen) but neither the mechanism nor the specificity (to alcohol) of this phenomenon have been established. Furthermore, as GLAST/EAAT1 is also expressed in testes and sperm (and could also be affected there by chronic alcohol), the levels of GLAST/EAAT1 in sperm could be used as a diagnostic tool in testing the severity of alcoholism in human males. We conclude that the reviewed studies present a unique set of data which could help to predict the risk of developing alcohol dependence (genetics), to improve the understanding of the intoxicating actions of alcohol (metabolomics), to aid in assessing the extent of damage to brain cells caused by chronic excessive drinking (metabolomics and proteomics) and to point to molecular targets that could be used in the treatment and diagnosis of alcoholism and alcohol addiction.


Subject(s)
Alcoholism/genetics , Alcoholism/metabolism , Ethanol/metabolism , Acetylation , Amino Acid Transport System X-AG/metabolism , Animals , Brain , Epigenesis, Genetic , Glutamate Plasma Membrane Transport Proteins/metabolism , Histones/metabolism , Humans , Metabolomics , Proteomics , Receptors, GABA/metabolism , Signal Transduction
4.
Glia ; 58(2): 181-94, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19606499

ABSTRACT

Astrocytes play a vital role in the brain; their structural integrity and sustained function are essential for neuronal viability, especially after injury or insult. In this study, we have examined the response of astrocytes to hypoxia/ischemia (H/I), employing multiple methods (immunohistochemistry, iontophoretic cell injection, Golgi-Kopsch staining, and D-aspartate uptake) in a neonatal pig model of H/I. We have identified morphological changes in cortical gray matter astrocytes in response to H/I. Initial astrocytic changes were evident as early as 8 h post-insult, before histological evidence for neuronal damage. By 72 h post-insult, astrocytes exhibited significantly fewer processes that were shorter, thicker, and had abnormal terminal swellings, compared with astrocytes from control brains that exhibited a complex structure with multiple fine branching processes. Quantification and image analysis of astrocytes at 72 h post-insult revealed significant decreases in the average astrocyte size, from 686 microm(2) in controls to 401 microm(2) in H/I brains. Sholl analysis revealed a significant decrease (>60%) in the complexity of astrocyte branching between 5 and 20 microm from the cell body. D-Aspartate uptake studies revealed that the H/I insult resulted in impaired astrocyte function, with significantly reduced clearance of the glutamate analog, D-aspartate. These results suggest that astrocytes may be involved in the pathophysiological events of H/I brain damage at a far earlier time point than first thought. Developing therapies that prevent or reverse these astrocytic changes may potentially improve neuronal survival and thus might be a useful strategy to minimize brain damage after an H/I insult.


Subject(s)
Astrocytes/pathology , Brain/pathology , Hypoxia-Ischemia, Brain/pathology , Nerve Fibers, Unmyelinated , Animals , Animals, Newborn , Astrocytes/physiology , Brain/physiopathology , Cell Size , D-Aspartic Acid/metabolism , Female , Glial Fibrillary Acidic Protein/metabolism , Glutamic Acid/metabolism , Hypoxia-Ischemia, Brain/physiopathology , Immunohistochemistry , Isoquinolines , Male , Swine , Time Factors
5.
Brain Behav Immun ; 24(7): 1058-68, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20153418

ABSTRACT

The current study, in parallel experiments, evaluated the impact of chronic psychological stress on physiological and behavioural measures, and on the activation status of microglia in 15 stress-responsive brain regions. Rats were subjected, for 14 days, to two 30 min sessions of restraint per day, applied at random times each day. In one experiment the effects of stress on sucrose preference, weight gain, core body temperature, and struggling behaviour during restraint, were determined. In the second experiment we used immunohistochemistry to investigate stress-induced changes in ionized calcium-binding adaptor molecule-1 (Iba1), a marker constitutively expressed by microglia, and major histocompatibility complex-II (MHC-II), a marker often expressed on activated microglia, in a total of 15 stress-responsive nuclei. We also investigated cellular proliferation in these regions using Ki67 immunolabelling, to check for the possibility of microglial proliferation. Collectively, the results we obtained showed that chronic stress induced a significant increase in anhedonia, a decrease in weight gain across the entire observation period, a significant elevation in core body temperature during restraint, and a progressive decrease in struggling behaviour within and over sessions. With regard to microglial activation, chronic stress induced a significant increase in the density of Iba1 immunolabelling (nine of 15 regions) and the number of Iba1-positive cells (eight of 15 regions). Within the regions that exhibited an increased number of Iba1-positive cells after chronic stress, we found no evidence of a between group difference in the number of MHC-II or Ki67 positive cells. In summary, these results clearly demonstrate that chronic stress selectively increases the number of microglia in certain stress-sensitive brain regions, and also causes a marked transition of microglia from a ramified-resting state to a non-resting state. These findings are consistent with the view that microglial activation could play an important role in controlling and/or adapting to stress.


Subject(s)
Behavior, Animal , Brain/pathology , Microglia/pathology , Stress, Psychological/pathology , Animals , Body Temperature , Brain/metabolism , Calcium-Binding Proteins/metabolism , Cell Proliferation , Chronic Disease , Food Preferences/psychology , Genes, MHC Class II , Immunohistochemistry , Ki-67 Antigen/metabolism , Male , Microfilament Proteins , Microglia/metabolism , Rats , Rats, Sprague-Dawley , Stress, Psychological/metabolism , Time Factors , Weight Gain
6.
Neurochem Res ; 35(12): 2062-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20890657

ABSTRACT

Glutamate transport (GluT) in brain is mediated chiefly by two transporters GLT and GLAST, both driven by ionic gradients generated by (Na(+), K(+))-dependent ATPase (Na(+)/K(+)-ATPase). GLAST is located in astrocytes and its function is regulated by translocations from cytoplasm to plasma membrane in the presence of GluT substrates. The phenomenon is blocked by a naturally occurring toxin rottlerin. We have recently suggested that rottlerin acts by inhibiting Na(+)/K(+)-ATPase. We now report that Na(+)/K(+)-ATPase inhibitors digoxin and ouabain also blocked the redistribution of GLAST in cultured astrocytes, however, neither of the compounds caused detectable inhibition of ATPase activity in cell-free astrocyte homogenates (rottlerin inhibited app. 80% of Pi production from ATP in the astrocyte homogenates, IC50 = 25 µM). Therefore, while we may not have established a direct link between GLAST regulation and Na(+)/K(+)-ATPase activity we have shown that both ouabain and digoxin can interfere with GluT transport and therefore should be considered potentially neurotoxic.


Subject(s)
Astrocytes/drug effects , Brain/drug effects , Digoxin/pharmacology , Excitatory Amino Acid Transporter 1/metabolism , Ouabain/pharmacology , Animals , Animals, Newborn , Astrocytes/enzymology , Astrocytes/metabolism , Brain/cytology , Brain/enzymology , Brain/metabolism , Immunohistochemistry , Rats , Rats, Sprague-Dawley , Sodium-Potassium-Exchanging ATPase/metabolism
7.
J Comp Neurol ; 528(17): 3134-3142, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32173860

ABSTRACT

Excitatory amino acid transporter 5 (EAAT5) is a protein that is known to be alternately spliced and to be abundantly expressed in the retina by populations of neurons including photoreceptors and bipolar cells. EAAT5 acts as a slow glutamate transporter and also as glutamate-gated chloride channel, the chloride conductance being large enough for EAAT5 to serve functionally as an "inhibitory" glutamate receptor. However, there has been a long-standing view that the classically spliced form of EAAT5 is not abundant or widespread in the brain and so it has not been extensively investigated in the literature. We recently identified a human-specific splicing form of EAAT5 that was not expressed by rodents but was shown to be a functional glutamate transporter. We have examined the expression of this form of EAAT5, hEAAT5v at the mRNA, and protein level in human brain, and show that populations of human cortical pyramidal neurons and cerebellar Purkinje cells show significant expression of hEAAT5v. Accordingly, we infer that EAAT5 may well be a player in modulating neuronal function in the human brain and propose that its localization in both glutamatergic and GABAergic neurons could be compatible with a role in influencing intracellular chloride and thereby neuronal parameters such as membrane potential rather than acting as a presynaptic glutamate transporter.


Subject(s)
Brain/cytology , Brain/metabolism , Excitatory Amino Acid Transporter 5/biosynthesis , Excitatory Amino Acid Transporter 5/genetics , Neurons/metabolism , Animals , Gene Expression , Humans , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Rats
8.
Neurochem Res ; 34(10): 1748-57, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19319679

ABSTRACT

Anomalies in glutamate homeostasis may contribute to the pathological processes involved in Alzheimer's disease (AD). Glutamate released from neurons or glial cells is normally rapidly cleared by glutamate transporters, most of which are expressed at the protein level by glial cells. However, in some patho-physiological situations, expression of glutamate transporters that are normally considered to be glial types, appears to be evoked in populations of distressed neurons. This study analysed the expression of exon-skipping forms of the three predominant excitatory amino acid (glutamate) transporters (EAATs1-3) in brains afflicted with AD. We demonstrate by immunocytochemistry in temporal cortex, the expression of these proteins particularly in limited subsets of neurons, some of which appeared to be dys-morphic. Whilst the neuronal expression of the "glial" glutamate transporters EAAT1 and EAAT2 is frequently considered to represent the abnormal and ectopic expression of such transporters, we suggest this may be a misinterpretation, since neurons such as cortical pyramidal cells normally express abundant mRNA for these EAATs (but little if any EAAT protein expression). We hypothesize instead that distressed neurons in the AD brain can turn on the translation of pre-existent mRNA pools, or suppress the degradation of alternately spliced glutamate transporter protein, leading to the "unmasking" of, rather than evoked expression of "glial" glutamate transporters in stressed neurons.


Subject(s)
Alzheimer Disease/metabolism , Amino Acid Transport System X-AG/biosynthesis , Brain/metabolism , Gene Expression Regulation/physiology , Neuroglia/metabolism , Neurons/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Amino Acid Transport System X-AG/genetics , Brain/pathology , Female , Humans , Male , Middle Aged , Neuroglia/pathology , Neurons/pathology , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Protein Isoforms/physiology
9.
Neurochem Res ; 34(10): 1758-66, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19440835

ABSTRACT

Neurotransmitter L-glutamate released at central synapses is taken up and "recycled" by astrocytes using glutamate transporter molecules such as GLAST and GLT. Glutamate transport is essential for prevention of glutamate neurotoxicity, it is a key regulator of neurotransmitter metabolism and may contribute to mechanisms through which neurons and glia communicate with each other. Using immunocytochemistry and image analysis we have found that extracellular D-aspartate (a typical substrate for glutamate transport) can cause redistribution of GLAST from cytoplasm to the cell membrane. The process appears to involve phosphorylation/dephosphorylation and requires intact cytoskeleton. Glutamate transport ligands L-trans-pyrrolidine-2,4-dicarboxylate and DL-threo-3-benzyloxyaspartate but not anti,endo-3,4-methanopyrrolidine dicarboxylate have produced similar redistribution of GLAST. Several representative ligands for glutamate receptors whether of ionotropic or metabotropic type, were found to have no effect. In addition, extracellular ATP induced formation of GLAST clusters in the cell membranes by a process apparently mediated by P2 receptors. The present data suggest that GLAST can rapidly and specifically respond to changes in the cellular environment thus potentially helping to fine-tune the functions of astrocytes.


Subject(s)
Adenosine Triphosphate/metabolism , Amino Acid Transport System X-AG/metabolism , Astrocytes/metabolism , Cell Membrane/metabolism , Excitatory Amino Acid Transporter 1/metabolism , Glutamic Acid/metabolism , Adenosine Triphosphate/analysis , Amino Acid Transport System X-AG/analysis , Animals , Animals, Newborn , Astrocytes/chemistry , Astrocytes/physiology , Cell Membrane/chemistry , Cells, Cultured , Excitatory Amino Acid Transporter 1/analysis , Extracellular Fluid/chemistry , Extracellular Fluid/metabolism , Glutamic Acid/analogs & derivatives , Glutamic Acid/analysis , Protein Transport/physiology , Rats , Rats, Sprague-Dawley , Substrate Specificity/physiology
10.
Neurochem Res ; 34(10): 1767-74, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19495968

ABSTRACT

The naturally occurring toxin rottlerin has been used by other laboratories as a specific inhibitor of protein kinase C-delta (PKC-delta) to obtain evidence that the activity-dependent distribution of glutamate transporter GLAST is regulated by PKC-delta mediated phosphorylation. Using immunofluorescence labelling for GLAST and deconvolution microscopy we have observed that D-aspartate-induced redistribution of GLAST towards the plasma membranes of cultured astrocytes was abolished by rottlerin. In brain tissue in vitro, rottlerin reduced apparent activity of (Na+, K+)-dependent ATPase (Na+, K+-ATPase) and increased oxygen consumption in accordance with its known activity as an uncoupler of oxidative phosphorylation ("metabolic poison"). Rottlerin also inhibited Na+, K+-ATPase in cultured astrocytes. As the glutamate transport critically depends on energy metabolism and on the activity of Na+, K+-ATPase in particular, we suggest that the metabolic toxicity of rottlerin and/or the decreased activity of the Na+, K+-ATPase could explain both the glutamate transport inhibition and altered GLAST distribution caused by rottlerin even without any involvement of PKC-delta-catalysed phosphorylation in the process.


Subject(s)
Acetophenones/pharmacology , Astrocytes/metabolism , Benzopyrans/pharmacology , Brain/metabolism , D-Aspartic Acid/metabolism , Excitatory Amino Acid Transporter 1/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Amino Acid Transport System X-AG/analysis , Amino Acid Transport System X-AG/antagonists & inhibitors , Amino Acid Transport System X-AG/metabolism , Animals , Animals, Newborn , Astrocytes/chemistry , Astrocytes/drug effects , Brain/drug effects , Brain/enzymology , Brain Chemistry/drug effects , Brain Chemistry/physiology , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cells, Cultured , D-Aspartic Acid/analysis , Enzyme Activation/drug effects , Enzyme Activation/physiology , Excitatory Amino Acid Transporter 1/analysis , Excitatory Amino Acid Transporter 1/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors
11.
Neurochem Int ; 125: 111-116, 2019 05.
Article in English | MEDLINE | ID: mdl-30817938

ABSTRACT

We have analysed post-mortem samples of prefrontal cortex from control and alcoholic human brains by the technique of Western blotting to estimate and compare the expressions of glutamate transporter GLAST (Excitatory Amino Acid Transporter One; EAAT1). Furthermore, using the non-alcoholic prefrontal cortex and custom-made GLAST (EAAT1) antibody we determined GLAST (EAAT1) "interactome" i.e. the set of proteins selectively bound by GLAST (EAAT1). We found that GLAST (EAAT1) was significantly more abundant (about 1.6-fold) in the cortical tissue from alcoholic brains compared to that from non-alcoholic controls. The greatest increase in the level of GLAST (EAAT1) was found in plasma membrane fraction (2.2-fold). Additionally, using the prefrontal cortical tissue from control brains, we identified 38 proteins specifically interacting with GLAST (EAAT1). These can be classified as contributing to the cell structure (6 proteins; 16%), energy and general metabolism (18 proteins; 47%), neurotransmitter metabolism (three proteins; 8%), signalling (6 proteins: 16%), neurotransmitter storage/release at synapses (three proteins; 8%) and calcium buffering (two proteins; 5%). We discuss possible consequences of the increased expression of GLAST (EAAT1) in alcoholic brain tissue and whether or how this could disturb the function of the proteins potentially interacting with GLAST (EAAT1) in vivo. The data represent an extension of our previous proteomic and metabolomic studies of human alcoholism revealing another aspect of the complexity of changes imposed on brain by chronic long-term consumption of ethanol.


Subject(s)
Alcoholism/metabolism , Excitatory Amino Acid Transporter 1/biosynthesis , Metabolomics/methods , Prefrontal Cortex/metabolism , Proteomics/methods , Adult , Aged , Alcoholics , Alcoholism/genetics , Alcoholism/pathology , Excitatory Amino Acid Transporter 1/genetics , Gene Expression , Humans , Male , Middle Aged , Prefrontal Cortex/pathology
12.
J Neurosci Methods ; 171(1): 140-6, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18400305

ABSTRACT

Reduction in microtubule-associated-protein-2 (MAP2) immunoreactivity is a sensitive and quantifiable early marker of neural injury in rats. This study assessed the reliability of MAP2 as an early marker of neural injury following hypoxia/ischaemia in neonatal piglets, and compared the effects of perfusion and immersion fixation on MAP2 immunoreactivity. Hypoxia was induced in newborn piglets (n=23) by reducing the FiO2 to 4% for 0, 25, 35 or 50 min. Six hours after the end of hypoxia piglets were killed, and the brain removed and immunolabelled for MAP2. Significant reductions in MAP2 immunoreactivity were seen in cortex, hippocampus, basal ganglia and thalamus. Reductions correlated with duration of hypoxia, pH at the end of hypoxia, cerebral function monitor amplitude and cerebral impedance 6h after hypoxia, and with early histological evidence of ischaemic changes. Regions with reduced immunoreactivity correlated with areas where damage is present in later histological examination in this model. Immersion fixation with postmortem delays up to 30 min did not affect MAP2 immunoreactivity compared to perfusion-fixed tissue. Results indicate that MAP2 immunoreactivity 6h after hypoxia/ischaemia is a reliable marker of neural injury in the neonatal piglet.


Subject(s)
Asphyxia Neonatorum/complications , Brain Injuries/etiology , Brain Injuries/pathology , Gene Expression Regulation/physiology , Microtubule-Associated Proteins/metabolism , Analysis of Variance , Animals , Animals, Newborn , Asphyxia Neonatorum/pathology , Disease Models, Animal , Humans , Hydrogen-Ion Concentration , Infant, Newborn , Swine , Time Factors
13.
Invest Ophthalmol Vis Sci ; 48(6): 2782-91, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17525213

ABSTRACT

PURPOSE: To determine whether structural plasticity is evident in human retinal tissues in response to age-related macular degeneration (AMD). Remodeling events such as sprouting of neuronal processes and the reconnection of synapses are essential elements in repairing any damage to adult nervous tissues such as might occur in response to insults such as strokes or in AMD. METHODS: The anatomic architecture of normal and AMD-affected human retinas was examined in the central, midperipheral, and far-peripheral regions. The retina, by virtue of its well-organized laminar structure, allows the identification and analysis of abnormal projections or connections of neuronal elements. RESULTS: In AMD-afflicted retinas, but not in normal aged human retinas, a large number of photoreceptor synapses across the entire retina retract into the outer nuclear layer. This event evokes the subsequent outgrowth of dendrites from the postsynaptic bipolar cells, again across the entire retina, and the subsequent reformation of synaptic contacts between photoreceptor and bipolar cells. CONCLUSIONS: These findings illustrate that there are anatomic changes in the AMD retina at all eccentricities, not just in the macular region. Aged human retinal neurons have the capacity to form new synapses, and this finding may be important when investigating possible means of repairing the damaged human retina.


Subject(s)
Dendrites/physiology , Macular Degeneration/metabolism , Neuronal Plasticity/physiology , Photoreceptor Cells, Vertebrate/physiology , Retinal Bipolar Cells/physiology , Synapses/physiology , Aged , Aged, 80 and over , Female , Humans , Immunoenzyme Techniques , Isoquinolines , Macular Degeneration/pathology , Male , Middle Aged , Nerve Regeneration/physiology , Photoreceptor Cells, Vertebrate/ultrastructure , Retinal Bipolar Cells/ultrastructure , Synaptophysin/metabolism , Vesicular Glutamate Transport Protein 1/metabolism
14.
Neuroreport ; 18(8): 741-5, 2007 May 28.
Article in English | MEDLINE | ID: mdl-17471058

ABSTRACT

We have raised antibodies that selectively recognize an exon 9 skipping form of GLAST. We demonstrate expression of this protein in brains of rats, cats, monkeys and humans. Immunolabelling was present in scattered populations of neurons, particularly in cerebral cortex and colliculi. Neurons were often present in small clusters and exhibited a range of morphologies, from apparently normal to highly degenerate. GLAST1b was also expressed by some glial cells. Cortical neurons expressing the exon 9 skipping form of GLAST also labelled with antibodies against the C- or N-terminal regions of GLAST. We suggest that alternate splicing of GLAST by subpopulations of neurons may indicate some dysfunction in these cells, and may be an indicator of inappropriate local excitation.


Subject(s)
Central Nervous System/metabolism , Excitatory Amino Acid Transporter 1/metabolism , Gene Expression/physiology , Animals , Blotting, Western/methods , Callithrix , Cats , Central Nervous System/cytology , Immunoblotting/methods , Immunohistochemistry/methods , Mice , Neurons/metabolism , Protein Structure, Tertiary , Rats
15.
Med Hypotheses ; 98: 57-59, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28012606

ABSTRACT

It has been known that a preconception paternal alcoholism impacts adversely on the offspring but the mechanism of the effect is uncertain. Several findings suggest that there are signalling systems in testis that are analogous to those known to be altered by alcoholism in brain. We propose that chronic alcohol affects these systems in a manner similar to that in brain. Specifically, we hypothesise that excessive alcohol may disturb glutamatergic-like signalling in testis by increasing expression of the glutamate transporter GLAST (EAAT1). We discuss ways how to test the hypothesis as well as potential significance of some of the tests as tools in the diagnostics of chronic alcoholism.


Subject(s)
Alcohol Drinking , Brain/pathology , Ethanol/chemistry , Excitatory Amino Acid Transporter 1/metabolism , Glutamic Acid/metabolism , Testis/metabolism , Alcoholism/physiopathology , Amino Acid Transport System X-AG/metabolism , Animals , Biological Transport , Congenital Abnormalities/etiology , Fathers , Female , Glutamine/metabolism , Humans , Male , Mice , Models, Biological , Paternal Exposure , Risk , Signal Transduction
16.
Invest Ophthalmol Vis Sci ; 47(9): 3853-9, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16936097

ABSTRACT

PURPOSE: Glaucoma is a common disease of the eye, a key characteristic consequence of which is the death of retinal ganglion cells. The cause of this loss is unknown, though glutamate-mediated toxicity has been implicated. Glutamate transporters are key regulators of glutamate; therefore, the purpose of the study was to determine whether unusual excitation is associated with unusual expression of one or more transporters. METHODS: The expression of a splice variant of the glutamate transporter GLT-1 (EAAT2) was examined in normal and glaucomatous retinas from humans and rats. RESULTS: In normal eyes of humans and rats, GLT-1c was expressed only in photoreceptors. In glaucoma, there was additional robust expression of GLT-1c in retinal ganglion cells, including occasional displaced ganglion cells. Conversely, cells such as displaced amacrine cells and amacrine cells were unlabeled. CONCLUSIONS: The induction of GLT-1c expression by retinal ganglion cells supports the notion that an anomaly or anomalies in glutamate homeostasis may be evident in glaucoma and that such anomalies selectively influence retinal ganglion cells. By analogy to in vitro experiments in which elevated glutamate levels induce expression of glutamate transporters, the authors hypothesize that expression of GLT-1c may represent an attempt by retinal ganglion cells to protect themselves against elevated levels of glutamate. Such anomalies in glutamate levels cannot be restricted to the ganglion cell layer, as this would not have affected displaced ganglion cells. GLT-1c may be a useful indicator of the extent of stress of the retinal ganglion cells and thus a tool for examining outcomes of potential therapeutic and experimental interventions.


Subject(s)
Excitatory Amino Acid Transporter 2/metabolism , Glaucoma/metabolism , Retinal Ganglion Cells/metabolism , Aged , Aged, 80 and over , Animals , Disease Models, Animal , Female , Glutamic Acid/metabolism , Humans , Immunoenzyme Techniques , Intraocular Pressure , Male , Photoreceptor Cells, Vertebrate/metabolism , Rats , Rats, Inbred BN
17.
Neurochem Int ; 48(6-7): 604-10, 2006.
Article in English | MEDLINE | ID: mdl-16530295

ABSTRACT

Excitatory amino acid transporters (EAATs) are responsible for homeostasis of extracellular L-glutamate, and the glial transporters are functionally dominant. EAAT expression or function is altered in acute and chronic neurological conditions, but little is known about the regulation of EAATs in reactive astroglia found in such neuropathologies. These studies examined the effects of the bacterial endotoxin lipopolysaccharide (LPS) on glial EAATs in vitro. The effects of LPS (1 microg/ml, 24-72 h) on EAAT activity and expression were examined in primary cultures of mouse astrocytes. [(3)H]D-aspartate uptake increased to 129% of control by 72 h treatment with LPS. Saturation analysis revealed that apparent K(m) was unchanged whilst V(max) was significantly increased to 172% of control by 72 h LPS treatment. Biotinylation and Western blotting indicated that cell-surface expression of GLT-1 was significantly elevated (146% control) by LPS treatment whereas GLAST expression was unchanged. Confocal analyses revealed that LPS treatment resulted in cytoskeletal changes and stellation of astrocytes, with rearrangement of F-actin (as shown by phalloidin labelling). Immunocytochemistry revealed clustering of GLAST, and increased expression and redistribution of GLT-1 to the cell-surface following treatment with LPS. Similar experiments were conducted in microglia, where LPS (50 ng/ml) was found to up-regulate expression of GLT-1 at 24 and 72 h in concert with cytoskeletal changes accompanying activation. These findings suggest an association of cytoskeletal changes in glia with EAAT activity, with the predominant adaptation involving up-regulation and redistribution of GLT-1.


Subject(s)
Excitatory Amino Acid Transporter 2/metabolism , Lipopolysaccharides/pharmacology , Neuroglia/drug effects , Actins/metabolism , Animals , Animals, Newborn , Aspartic Acid/metabolism , Astrocytes/drug effects , Astrocytes/ultrastructure , Biotinylation , Blotting, Western , Cells, Cultured , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Excitatory Amino Acid Transporter 1/biosynthesis , Excitatory Amino Acid Transporter 2/biosynthesis , Immunohistochemistry , Mice , Mice, Inbred C57BL , Microglia/metabolism , Microscopy, Confocal , Neuroglia/cytology , Neuroglia/metabolism , Phenotype , Protein Transport , Up-Regulation
18.
Neuroreport ; 17(18): 1867-70, 2006 Dec 18.
Article in English | MEDLINE | ID: mdl-17179860

ABSTRACT

GLAST is a glial glutamate transporter; mRNA for a splice variant, GLAST1a, which lacks exon 3, has previously been identified. To detect GLAST1a protein, we generated antibodies against a peptide sequence encompassing the splice site. We demonstrate by Western blotting and immunocytochemistry the expression of GLAST1a in brains and retinae. Robust immunolabelling was present in the cerebellar Bergmann glia, and weaker labelling was evident in the retinal Müller cells. GLAST1a is differentially targeted to some cellular compartments such as the end feet of the Müller cells. As GLAST1a protein may interfere with the transport of glutamate by normally spliced GLAST, differentially targeted expression of GLAST1a may represent a mechanism for selectively regulating GLAST function in the mammalian nervous system.


Subject(s)
Brain/metabolism , Excitatory Amino Acid Transporter 1/metabolism , Gene Expression/physiology , Retina/metabolism , Animals , Blotting, Western/methods , Brain/cytology , Excitatory Amino Acid Transporter 1/genetics , Immunoblotting/methods , Immunohistochemistry/methods , Neuroglia/cytology , Neuroglia/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Rats , Retina/cytology
19.
Behav Brain Res ; 303: 176-81, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26821291

ABSTRACT

The consolidation of short-term memory into long-term memory involves changing protein level and activity for the synaptic plasticity required for long-term potentiation (LTP). AMPA receptor trafficking is a key determinant of LTP and recently ubiquitination by Nedd4 has been shown to play an important role via direct action on the GluA1 subunit, although the physiological relevance of these findings are yet to be determined. We therefore investigated learning and memory in Nedd4(+/-) mice that have a 50% reduction in levels of Nedd4. These mice showed decreased long-term spatial memory as evidenced by significant increases in the time taken to learn the location of and subsequently find a platform in the Morris water maze. In contrast, there were no significant differences between Nedd4(+/+) and Nedd4(+/-) mice in terms of short-term spatial memory in a Y-maze test. Nedd4(+/-) mice also displayed a significant reduction in post-synaptic LTP measured in hippocampal brain slices. Immunofluorescence of Nedd4 in the hippocampus confirmed its expression in hippocampal neurons of the CA1 region. These findings indicate that reducing Nedd4 protein by 50% significantly impairs LTP and long-term memory thereby demonstrating an important role for Nedd4 in these processes.


Subject(s)
Endosomal Sorting Complexes Required for Transport/physiology , Hippocampus/physiology , Learning/physiology , Long-Term Potentiation , Spatial Memory/physiology , Ubiquitin-Protein Ligases/physiology , Animals , Endosomal Sorting Complexes Required for Transport/genetics , Heterozygote , Hippocampus/metabolism , Memory, Long-Term/physiology , Memory, Short-Term/physiology , Mice , Mice, Transgenic , Nedd4 Ubiquitin Protein Ligases , Neurons/metabolism , Receptors, AMPA/metabolism , Ubiquitin-Protein Ligases/genetics
20.
J Neurosci ; 22(3): RC206, 2002 Feb 01.
Article in English | MEDLINE | ID: mdl-11826152

ABSTRACT

Glutamate-mediated toxicity has been implicated in the neurodegeneration observed in Alzheimer's disease. In particular, glutamate transport dysfunction may increase susceptibility to glutamate toxicity, thereby contributing to neuronal cell injury and death. In this study, we examined the cellular localization of the glial glutamate transporter excitatory amino acid transporter 1 (EAAT1) in the cerebral cortex of control, Alzheimer's disease, and non-Alzheimer dementia cases. We found that EAAT1 was strongly expressed in a subset of cortical pyramidal neurons in dementia cases showing Alzheimer-type pathology. In addition, tau (which is a marker of neurofibrillary pathology) colocalized to those same pyramidal cells that expressed EAAT1. These findings suggest that EAAT1 changes are related to tau expression (and hence neurofibrillary tangle formation) in dementia cases showing Alzheimer-type pathology. This study implicates aberrant glutamate transporter expression as a mechanism involved in neurodegeneration in Alzheimer's disease.


Subject(s)
Alzheimer Disease/metabolism , Cerebral Cortex/metabolism , Excitatory Amino Acid Transporter 1/biosynthesis , Aged , Aged, 80 and over , Alzheimer Disease/etiology , Alzheimer Disease/pathology , Biomarkers/analysis , Cerebral Cortex/pathology , Dementia, Multi-Infarct/metabolism , Dementia, Multi-Infarct/pathology , Excitatory Amino Acid Transporter 1/analysis , Female , Humans , Immunohistochemistry , Lewy Body Disease/metabolism , Lewy Body Disease/pathology , Male , Middle Aged , Neurofibrillary Tangles/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , tau Proteins/analysis , tau Proteins/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL