Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Int J Med Microbiol ; 308(1): 148-154, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29089241

ABSTRACT

Human red blood cells infected with the malaria parasite Plasmodium falciparum show an increased permeability to a number of solutes. We have previously demonstrated that such infected cells take up glutamate via a member of the excitatory amino acid transporter protein family (EAAT), namely EAAT3. Babesia divergens is a parasite that also infects human erythrocytes, and also induces increased solute permeability, including for glutamate. Here we have investigated whether glutamate uptake in B. divergens infected human red blood cells is also dependent on EAAT3 activity. We find that, although B. divergens infected cells do take up glutamate, this uptake is independent on EAAT3. Thus, though infecting the same host cell, two related parasites have developed distinct pathways to obtain access to nutrients from the extracellular milieu.


Subject(s)
Babesia/physiology , Erythrocytes/parasitology , Excitatory Amino Acid Transporter 3/metabolism , Glutamic Acid/metabolism , Cell Membrane Permeability/drug effects , Choline/pharmacology , Erythrocyte Membrane/drug effects , Erythrocyte Membrane/physiology , Erythrocytes/drug effects , Erythrocytes/physiology , Excitatory Amino Acid Transporter 3/antagonists & inhibitors , Glutamates/pharmacology , Nitrobenzoates/pharmacology
2.
BMC Anesthesiol ; 15: 116, 2015 Aug 08.
Article in English | MEDLINE | ID: mdl-26253075

ABSTRACT

BACKGROUND: Many researchers have suggested that the glutamatergic system may be involved in the effects of antidepressant therapies. We investigated the effects of doxepin, imipramine, and fluoxetine on the excitatory amino acid transporter type 3 (EAAT3). METHODS: EAAT3 was expressed in Xenopus oocytes by injection of EAAT3 mRNA. Membrane currents were recorded after application of L-glutamate (30 µM) in the presence or absence of various concentrations of doxepin, imipramine, and fluoxetine. To study the effects of protein kinase C (PKC) activation on EAAT3 activity, oocytes were pre-incubated with phorbol 12-myristate-13-acetate (PMA) before application of imipramine and doxepin. RESULTS: Doxepin at 0.063-1.58 µM significantly decreased EAAT3 activity. Imipramine reduced EAAT3 activity in a concentration-dependent manner at 0.16-0.95 µM. However, fluoxetine did not affect EAAT3 activity, and PMA increased EAAT3 activity. At 0.32 µM, imipramine caused an equivalent decrease in EAAT3 activity in the presence or absence of PMA. However, 0.79 µM doxepin did not abolish the enhancement of EAAT3 activity by PMA. CONCLUSIONS: We showed that doxepin and imipramine, but not fluoxetine, inhibited EAAT3 activity at clinically relevant concentrations. This reveals a novel mechanism of action for doxepin and imipramine; that they increase glutamatergic neurotransmission. PKC may be involved in the effects of doxepin on EAAT3, but is not involved in the effects of imipramine at the concentrations studied.


Subject(s)
Doxepin/pharmacology , Excitatory Amino Acid Transporter 3/antagonists & inhibitors , Fluoxetine/pharmacology , Imipramine/pharmacology , Animals , Antidepressive Agents/administration & dosage , Antidepressive Agents/pharmacology , Dose-Response Relationship, Drug , Doxepin/administration & dosage , Female , Fluoxetine/administration & dosage , Glutamic Acid/metabolism , Imipramine/administration & dosage , Oocytes/drug effects , Oocytes/metabolism , Protein Kinase C/metabolism , RNA, Messenger/metabolism , Rats , Tetradecanoylphorbol Acetate/pharmacology , Xenopus laevis
3.
Mol Pharmacol ; 85(5): 747-57, 2014 May.
Article in English | MEDLINE | ID: mdl-24569088

ABSTRACT

Canonically, opioids influence cells by binding to a G protein-coupled opioid receptor, initiating intracellular signaling cascades, such as protein kinase, phosphatidylinositol 3-kinase, and extracellular receptor kinase pathways. This results in several downstream effects, including decreased levels of the reduced form of glutathione (GSH) and elevated oxidative stress, as well as epigenetic changes, especially in retrotransposons and heterochromatin, although the mechanism and consequences of these actions are unclear. We characterized the acute and long-term influence of morphine on redox and methylation status (including DNA methylation levels) in cultured neuronal SH-SY5Y cells. Acting via µ-opioid receptors, morphine inhibits excitatory amino acid transporter type 3-mediated cysteine uptake via multiple signaling pathways, involving different G proteins and protein kinases in a temporal manner. Decreased cysteine uptake was associated with decreases in both the redox and methylation status of neuronal cells, as defined by the ratios of GSH to oxidized forms of glutathione and S-adenosylmethionine to S-adenosylhomocysteine levels, respectively. Further, morphine induced global DNA methylation changes, including CpG sites in long interspersed nuclear elements (LINE-1) retrotransposons, resulting in increased LINE-1 mRNA. Together, these findings illuminate the mechanism by which morphine, and potentially other opioids, can influence neuronal-cell redox and methylation status including DNA methylation. Since epigenetic changes are implicated in drug addiction and tolerance phenomenon, this study could potentially extrapolate to elucidate a novel mechanism of action for other drugs of abuse.


Subject(s)
Cysteine/antagonists & inhibitors , DNA Methylation/drug effects , Excitatory Amino Acid Transporter 3/antagonists & inhibitors , Morphine/pharmacology , Retroelements/drug effects , Transcription, Genetic/drug effects , Cell Line, Tumor , Cysteine/metabolism , DNA Methylation/physiology , Excitatory Amino Acid Transporter 3/physiology , Humans , Oxidation-Reduction/drug effects , Retroelements/physiology , Transcription, Genetic/physiology
4.
Biochemistry ; 51(27): 5486-95, 2012 Jul 10.
Article in English | MEDLINE | ID: mdl-22703277

ABSTRACT

Glutamate transporters play an important role in the regulation of extracellular glutamate concentrations in the mammalian brain and are, thus, promising targets for therapeutics. Despite this importance, the development of pharmacological tools has mainly focused on the synthesis of competitive inhibitors, which are amino acid analogues that bind to the substrate binding site. In this report, we describe the characterization of the mechanism of glutamate transporter inhibition by a constrained, cyclic glutamate analogue, (+)-3-hydroxy-4,5,6,6a-tetrahydro-3aH-pyrrolo[3,4-d]isoxazole-6-carboxylic acid [(+)-(3aS,6S,6aS)-HIP-B]. Our results show that (+)-HIP-B is a nontransportable amino acid that inhibits glutamate transporter function in a mixed mechanism. Although (+)-HIP-B inhibits the glutamate-associated anion conductance, it has no effect on the leak anion conductance, in contrast to competitive inhibitors. Furthermore, (+)-HIP-B is unable to alleviate the effect of the competitive inhibitor dl-threo-ß-benzyloxyaspartic acid (TBOA), which binds to the substrate binding site. (+)-HIP-B is more potent in inhibiting forward transport compared to reverse transport. In a mutant transporter, which is activated by glutamine, but not glutamate, (+)-HIP-B still acts as an inhibitor, although this mutant transporter is insensitive to TBOA. Finally, we analyzed the effect of (+)-HIP-B on the pre-steady-state kinetics of the glutamate transporter. The results can be explained with a mixed mechanism at a site that may be distinct from the substrate binding site, with a preference for the inward-facing configuration of the transporter and slow inhibitor binding. (+)-HIP-B may represent a new paradigm of glutamate transporter inhibition that is based on targeting of a regulatory site.


Subject(s)
Carboxylic Acids/chemistry , Carboxylic Acids/pharmacology , Excitatory Amino Acid Transporter 3/antagonists & inhibitors , Glutamates/chemistry , Molecular Conformation , Oxazoles/chemistry , Oxazoles/pharmacology , Binding Sites , Biological Transport/drug effects , Carboxylic Acids/metabolism , Excitatory Amino Acid Transporter 3/genetics , Excitatory Amino Acid Transporter 3/metabolism , Glutamates/metabolism , HEK293 Cells , Humans , Kinetics , Mutation , Oxazoles/metabolism
5.
Exp Neurol ; 336: 113538, 2021 02.
Article in English | MEDLINE | ID: mdl-33253705

ABSTRACT

Accumulating evidence indicates time-of-day variations in ischemic neuronal injury. Under ischemic conditions, Zn2+ is massively released from hippocampal glutamatergic neurons, and intracellular Zn2+ accumulation results in neuron death. Notably, excitatory amino acid carrier 1 (EAAC1), known as a cysteine transporter, is involved in Zn2+ homeostasis, and its expressions exhibit a diurnal fluctuation. This study aimed to investigate whether time of day of an ischemic insult affects Zn2+ accumulation and neuronal injury and determine whether altered Zn2+ accumulation is modulated by EAAC1 diurnal fluctuation in the hippocampus in a mouse model of ischemic stroke. Mice subjected to transient global ischemia for 40 min at Zeitgeber time 18 (ZT18) (23:00) exhibited reduced Zn2+ accumulation and neuronal death in the hilar region of the hippocampus compared to those at ZT4 (09:00). The EAAC1 protein expression in the hippocampus was increased at ZT18 relative to ZT4. Intracerebroventricular injection of a non-selective excitatory amino acid transporter inhibitor, DL-threo-ß-benzyloxyaspartate, or a selective EAAC1 inhibitor, L-aspartic acid ß-hydroxamate, increased ischemia-induced Zn2+ accumulation and neuronal death in the hilus at ZT18. These findings suggest that ischemia-induced Zn2+ accumulation displays circadian fluctuations through diurnal variations in EAAC1 expressions and affects susceptibility to ischemic neuronal injury in the hippocampal hilar region.


Subject(s)
Brain Ischemia/metabolism , Circadian Rhythm/physiology , Excitatory Amino Acid Transporter 3/metabolism , Hippocampus/metabolism , Zinc/metabolism , Animals , Cell Death , Excitatory Amino Acid Transporter 3/antagonists & inhibitors , Glutathione/metabolism , Injections, Intraventricular , Male , Mice , Mice, Inbred C57BL , Nerve Degeneration/pathology , Neurons/pathology
6.
J Neurosci ; 29(24): 7898-908, 2009 Jun 17.
Article in English | MEDLINE | ID: mdl-19535601

ABSTRACT

Glutamate released from synaptic vesicles mediates excitatory neurotransmission by stimulating glutamate receptors. Glutamate transporters maintain low synaptic glutamate levels critical for this process, a role primarily attributed to astrocytes. Recently, vesicular release of glutamate from unmyelinated axons in the rat corpus callosum has been shown to elicit AMPA receptor-mediated currents in glial progenitor cells. Glutamate transporters are the only mechanism of glutamate clearance, yet very little is known about the role of glutamate transporters in normal development of oligodendrocytes (OLs) or in excitotoxic injury to OLs. We found that OLs in culture are capable of sodium-dependent glutamate uptake with a K(m) of 10 +/- 2 microm and a V(max) of 2.6, 5.0, and 3.8 nmol x min(-1) x mg(-1) for preoligodendrocytes, immature, and mature OLs, respectively. Surprisingly, EAAC1, thought to be exclusively a neuronal transporter, contributes more to [(3)H]l-glutamate uptake in OLs than GLT1 or GLAST. These data suggest that glutamate transporters on oligodendrocytes may serve a critical role in maintaining glutamate homeostasis at a time when unmyelinated callosal axons are engaging in glutamatergic signaling with glial progenitors. Furthermore, GLT1 was significantly increased in cultured mature OLs contrary to in vivo data in which we have shown that, although GLT1 is present on developing OLs when unmyelinated axons are prevalent in the developing rat corpus callosum, after myelination, GLT1 is not expressed on mature OLs. The absence of GLT1 in mature OLs in the rat corpus callosum and its presence in mature rat cultured OLs may indicate that a signaling process in vivo is not activated in vitro.


Subject(s)
Brain/cytology , Brain/growth & development , Excitatory Amino Acid Transporter 2/physiology , Excitatory Amino Acid Transporter 3/physiology , Gene Expression Regulation, Developmental/physiology , Glutamic Acid/metabolism , Oligodendroglia/metabolism , Animals , Animals, Newborn , Aspartic Acid/pharmacology , Benzodiazepines/pharmacology , Bicuculline/pharmacology , Cells, Cultured , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Excitatory Amino Acid Transporter 3/antagonists & inhibitors , Female , Fibroblast Growth Factors/metabolism , GABA Antagonists/pharmacology , Gangliosides/metabolism , Gene Expression Regulation, Developmental/drug effects , Kainic Acid/analogs & derivatives , Kainic Acid/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neurons/metabolism , O Antigens/metabolism , Patch-Clamp Techniques/methods , Platelet-Derived Growth Factor/metabolism , Pregnancy , Quinoxalines/pharmacology , Rats , Rats, Long-Evans , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology , Tritium/metabolism
7.
J Neurophysiol ; 103(5): 2581-6, 2010 May.
Article in English | MEDLINE | ID: mdl-20220082

ABSTRACT

In the cochlea, afferent transmission between inner hair cells and auditory neurons is mediated by glutamate receptors. Glutamate transporters located near the synapse and in spiral ganglion neurons are thought to maintain low synaptic levels of glutamate. We analyzed three glutamate transporter blockers for their ability to alter the effects of glutamate, exogenously applied to the synapse via perfusion of the scala tympani of the mouse, and compared that action to their ability to alter the effects of intense acoustic stimulation. Threo-beta-benzyloxyaspartate (TBOA) is a broad-spectrum glutamate transporter antagonist, affecting all three transporters [glutamate/aspartate transporter (GLAST), glutamate transporter-1 (GLT1), and excitatory amino acid carrier 1 (EAAC1)]. l-serine-O-sulfate (SOS) blocks both GLAST and EAAC1 without effect on GLT1. Dihydrokainate (DHK) is selective for GLT1. Infusion of glutamate (10 microM for 220 min), TBOA (200 microM for 220 min), or SOS (100 microM for 180 min) alone did not alter auditory neural thresholds. When infused together with glutamate, TBOA and SOS produced significant neural threshold shifts, leaving otoacoustic emissions intact. In addition, both TBOA and SOS exacerbated noise-induced hearing loss by producing larger neural threshold shifts and delaying recovery. DHK did not alter glutamate- or noise-induced hearing loss. The evidence points to a major role for GLAST, both in protecting the synapse from exposure to excess extracellular glutamate and in attenuating hearing loss due to acoustic overstimulation.


Subject(s)
Cochlea/physiology , Excitatory Amino Acid Transporter 1/metabolism , Excitatory Amino Acid Transporter 3/metabolism , Glucose Transporter Type 1/metabolism , Hearing/physiology , Synaptic Transmission/physiology , Acoustic Stimulation , Animals , Auditory Threshold/drug effects , Auditory Threshold/physiology , Cochlea/drug effects , Cochlear Nerve/drug effects , Cochlear Nerve/physiology , Excitatory Amino Acid Transporter 1/antagonists & inhibitors , Excitatory Amino Acid Transporter 3/antagonists & inhibitors , Extracellular Space/metabolism , Glucose Transporter Type 1/antagonists & inhibitors , Glutamic Acid/metabolism , Hair Cells, Auditory, Inner/drug effects , Hair Cells, Auditory, Inner/physiology , Hearing/drug effects , Hearing Loss, Noise-Induced/physiopathology , Mice , Mice, Inbred CBA , Neurons, Afferent/drug effects , Neurons, Afferent/physiology , Scala Tympani/drug effects , Scala Tympani/physiology , Spiral Ganglion/drug effects , Spiral Ganglion/physiology , Synaptic Transmission/drug effects
8.
Planta Med ; 76(6): 595-8, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19937551

ABSTRACT

The major excitatory neurotransmitter transporter EAAC1 in the mammalian central nervous system is considered a possible target for Chinese herbal medicine. Extracts of Acorus tatarinowii (Schott) were tested for their effects on EAAC1 activity. XENOPUS oocytes with heterologously expressed EAAC1 were used as the model system. Rate of glutamate uptake was determined by means of the isotopic tracer technique. Glutamate-induced current was recorded under a two-electrode voltage clamp. As a highly effective component, alpha-asarone was identified. The rate of glutamate uptake was stimulated by 200 microM of alpha-asarone by about 15 %. In contrast, the same concentration reduced the EAAC1-mediated current by about 35 % at a holding potential of - 60 mV; half maximum inhibition was obtained at about 60 microM. Our experimental data suggest that both stimulation of glutamate uptake and inhibition of EAAC1-mediated current by alpha-asarone could contribute to reduced excitatory activity.


Subject(s)
Anisoles/pharmacology , Excitatory Amino Acid Transporter 3/antagonists & inhibitors , Allylbenzene Derivatives , Animals , Electrophysiological Phenomena , Ion Channel Gating , Oocytes , Xenopus
9.
J Pharm Pharmacol ; 61(5): 577-81, 2009 May.
Article in English | MEDLINE | ID: mdl-19405995

ABSTRACT

OBJECTIVES: Evidence suggests that glutamatergic systems may be involved in the pathophysiology of major depression and the mechanism of action of antidepressants. We have investigated the effects of amitriptyline, a tricyclic antidepressant, on the activity of the excitatory amino acid transporter type 3 (EAAT3), a protein that can regulate extracellular glutamate concentrations in the brain. METHODS: EAAT3 was expressed in Xenopus oocytes. Using a two-electrode voltage clamp, membrane currents were recorded after application of 30 microM L-glutamate in the presence or absence of various concentrations of amitriptyline or after application of various concentrations of L-glutamate in the presence or absence of 0.64 microM amitriptyline. KEY FINDINGS: Amitriptyline concentration-dependently reduced EAAT3 activity. This inhibition reached statistical significance at 0.38-1.27 microM amitriptyline. Amitriptyline 0.64 microM reduced the pharmacokinetic parameter Vmax, but did not affect the pharmacokinetic parameter Km, of EAAT3 for L-glutamate. The amitriptyline inhibition disappeared after a 4-min washout. Phorbol-12-myristate-13-acetate, a protein kinase C activator, increased EAAT3 activity. However, 0.64 microM amitriptyline induced a similar degree of decrease in EAAT3 activity in the presence or absence of phorbol-12-myristate-13-acetate. CONCLUSIONS: Our results suggested that amitriptyline at clinically relevant concentrations reversibly reduced EAAT3 activity via decreasing its maximal velocity of glutamate transporting function. The effects of amitriptyline on EAAT3 activity may have represented a novel site of action for amitriptyline to increase glutamatergic neurotransmission. Protein kinase C may not have been involved in the effects of amitriptyline on EAAT3.


Subject(s)
Amitriptyline/pharmacology , Antidepressive Agents, Tricyclic/pharmacology , Excitatory Amino Acid Transporter 3/antagonists & inhibitors , Oocytes/drug effects , Animals , Dose-Response Relationship, Drug , Enzyme Activators/pharmacology , Excitatory Amino Acid Transporter 3/metabolism , Female , Glutamic Acid/pharmacology , Oocytes/physiology , Patch-Clamp Techniques , Protein Kinase C/physiology , Rats , Tetradecanoylphorbol Acetate/pharmacology , Xenopus laevis
10.
ACS Chem Neurosci ; 10(10): 4414-4429, 2019 10 16.
Article in English | MEDLINE | ID: mdl-31573179

ABSTRACT

In the present study, screening of a library of 49,087 compounds at the excitatory amino acid transporter subtype 3 (EAAT3) led to the identification of 2-(furan-2-yl)-8-methyl-N-(o-tolyl)imidazo[1,2-a]pyridin-3-amine (3a) which showed a >20-fold preference for inhibition of EAAT3 (IC50 = 13 µM) over EAAT1,2,4 (EAAT1: IC50 ∼ 250 µM; EAAT2,4: IC50 > 250 µM). It was shown that a small lipophilic substituent (methyl or bromine) at the 7- and/or 8-position was essential for activity. Furthermore, the substitution pattern of the o-tolyl group (compound 5b) and the chemical nature of the substituent in the 2-position (compound 7b) were shown to be essential for the selectivity toward EAAT3 over EAAT1,2. The most prominent analogues to come out of this study are 3a and 3e that display ∼35-fold selectivity for EAAT3 (IC50 = 7.2 µM) over EAAT1,2,4 (IC50 ∼ 250 µM).


Subject(s)
Excitatory Amino Acid Transporter 3/antagonists & inhibitors , Pyridines/chemistry , Pyridines/pharmacology , Biogenic Amines/chemistry , Biogenic Amines/pharmacology , Chromatography, Thin Layer/methods , Dose-Response Relationship, Drug , Excitatory Amino Acid Transporter 3/metabolism , HEK293 Cells , Humans , Structure-Activity Relationship
11.
ACS Sens ; 4(9): 2358-2366, 2019 09 27.
Article in English | MEDLINE | ID: mdl-31393114

ABSTRACT

Glutamate is the main excitatory neurotransmitter in the mammalian central nervous system. Excitatory amino acid transporters (EAATs) are a family of transmembrane transporters responsible for glutamate uptake into cells, and their malfunction is related to a variety of diseases, including neurodegenerative diseases and stroke. Screening for and developing inhibitors of EAATs as well as related transporters is a significant field of study for biomedical and pharmaceutical applications. Rapid, high-throughput methods are critical for the study of glutamate transporters, and fluorescent methods are appealing for this purpose as compared to more traditional electrophysiological methods. In this study, we present a method for studying glutamate transporters and inhibitors by utilizing a mutated version of a yellow fluorescent protein (YFP) highly sensitive to quenching by anions (mClY). We applied this YFP variant to fluorescent imaging of anion flux in HEK293 cells caused by transiently expressed excitatory amino acid carrier 1 (EAAC1) and excitatory amino acid transporter 2 (EAAT2) and its inhibition by competitive blockers. This method enables rapid identification of inhibitors and, potentially, activators of EAAT function, which is critical for glutamate transport research.


Subject(s)
Glutamic Acid/metabolism , Halogens/metabolism , Optical Imaging/methods , Bacterial Proteins/genetics , Biological Transport , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Excitatory Amino Acid Transporter 2/genetics , Excitatory Amino Acid Transporter 2/metabolism , Excitatory Amino Acid Transporter 3/antagonists & inhibitors , Excitatory Amino Acid Transporter 3/genetics , Excitatory Amino Acid Transporter 3/metabolism , HEK293 Cells , Humans , Luminescent Proteins/genetics , Time Factors
12.
Neuroscience ; 151(4): 1042-52, 2008 Feb 19.
Article in English | MEDLINE | ID: mdl-18207650

ABSTRACT

The transport of excitatory amino acids (EAA) in CNS is performed by a family of high affinity, sodium dependent carriers. One of these transporters, excitatory amino acid carrier 1 (EAAC1), is known to be regulated by several mechanisms that modify carrier abundance on the plasma membrane. Much less is known on EAAC1 regulation at the level of gene expression. Here we report that, in C6 rat glioma cells, a line recently described to contain neural stem-like cells, EAAC1 is markedly induced by all trans-retinoic acid (ATRA), a well known differentiating agent. Consistently, ATRA stimulates EAA transport, with the maximal effect observed at concentrations>or=1 microM. After 4 days of treatment with 10 microM ATRA, the transport Vmax is fivefold enhanced, Slc1a1 mRNA is increased by 400% compared with control, EAAC1 carrier is sixfold overexpressed and the C6 culture is greatly enriched of cells with bipolar morphology strongly positive for EAAC1 immunoreactivity. Compared with untreated cells, ATRA-treated C6 cells express less Slc1a3 mRNA, for the transporter GLAST, but significantly higher levels of Slc1a2 mRNA, for the transporter GLT-1, although no expression of either protein is detected with Western blot in both untreated and ATRA-treated cells. Consistently, the inhibition pattern of aspartate transport and its stimulation by phorbol esters are indicative of a transport process due to EAAC1 operation. Under the conditions adopted, ATRA treatment causes the induction of proteolipid protein, an oligodendrocytic marker. These results indicate that, in C6 cells, ATRA stimulates the expression of EAAC1, possibly as a step toward oligodendrocytic differentiation, and constitute the first demonstration of the induction of this transporter by a differentiating agent.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Differentiation/drug effects , Excitatory Amino Acid Transporter 3/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Tretinoin/pharmacology , Animals , Aspartic Acid/metabolism , Aspartic Acid/pharmacology , Cell Line, Tumor , Dose-Response Relationship, Drug , Excitatory Amino Acid Transporter 3/antagonists & inhibitors , Excitatory Amino Acids/pharmacology , Glioma/pathology , Nerve Tissue Proteins/metabolism , Rats
13.
Synapse ; 62(11): 842-50, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18720515

ABSTRACT

Group II metabotropic glutamate receptors (mGluR2 and mGluR3, also called mGlu2 and mGlu3, encoded by GRM2 and GRM3, respectively) are therapeutic targets for several psychiatric disorders. GRM3 may also be a schizophrenia susceptibility gene. mGluR2-/- and mGluR3-/- mice provide the only unequivocal means to differentiate between these receptors, yet interpretation of in vivo findings may be complicated by secondary effects on expression of other genes. To address this issue, we examined the expression of NMDA receptor subunits (NR1, NR2A, NR2B) and glutamate transporters (EAAT1-3), as well as the remaining group II mGluR, in the hippocampus of mGluR2-/- and mGluR3-/- mice, compared with wild-type controls. mGluR2 mRNA was increased in mGluR3-/- mice, and vice versa. NR2A mRNA was increased in both knockout mice. EAAT1 (GLAST) mRNA and protein, and EAAT2 (GLT-1) protein, were reduced in mGluR3-/- mice, whereas EAAT3 (EAAC1) mRNA was decreased in mGluR2-/- mice. Transcripts for NR1 and NR2B were unchanged. The findings show a compensatory upregulation of the remaining group II metabotropic glutamate receptor in the knockout mice. Upregulation of NR2A expression suggests modified NMDA receptor signaling in mGluR2-/- and mGluR3-/- mice, and downregulation of glutamate transporter expression suggests a response to altered synaptic glutamate levels. The results show a mutual interplay between mGluR2 and mGluR3, and also provide a context in which to interpret behavioral and electrophysiological results in these mice.


Subject(s)
Gene Expression Regulation/genetics , Glutamate Plasma Membrane Transport Proteins/deficiency , Glutamate Plasma Membrane Transport Proteins/genetics , Hippocampus/metabolism , Receptors, Metabotropic Glutamate/deficiency , Receptors, Metabotropic Glutamate/genetics , Amino Acid Transport System X-AG/biosynthesis , Amino Acid Transport System X-AG/deficiency , Amino Acid Transport System X-AG/genetics , Animals , Down-Regulation/genetics , Excitatory Amino Acid Transporter 1/antagonists & inhibitors , Excitatory Amino Acid Transporter 1/biosynthesis , Excitatory Amino Acid Transporter 1/genetics , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Excitatory Amino Acid Transporter 2/biosynthesis , Excitatory Amino Acid Transporter 2/genetics , Excitatory Amino Acid Transporter 3/antagonists & inhibitors , Excitatory Amino Acid Transporter 3/biosynthesis , Excitatory Amino Acid Transporter 3/genetics , Glutamate Plasma Membrane Transport Proteins/biosynthesis , Hippocampus/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/biosynthesis , Receptors, Glutamate/biosynthesis , Receptors, Glutamate/genetics , Receptors, Metabotropic Glutamate/biosynthesis , Up-Regulation/genetics
14.
Bioorg Med Chem ; 16(16): 7740-8, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18650095

ABSTRACT

A series of beta-benzylaspartate derivatives were prepared from N-trityl-L-aspartate dimethyl ester and evaluated as inhibitors of neuronal glutamate transporter EAAT3. The result of the structure-activity studies suggests that the position occupied by the aromatic ring of beta-benzylaspartate within the binding site of EAAT3 may be different from that occupied by comparable groups in previously identified inhibitors, such as L-threo-benzyloxy aspartate (TBOA). Further, halogen substitutions at the 3-position of the aromatic ring of beta-benzylaspartate can increase the potency with which the analogues inhibit EAAT3.


Subject(s)
Aspartic Acid/analogs & derivatives , Excitatory Amino Acid Transporter 3/antagonists & inhibitors , Animals , Aspartic Acid/chemical synthesis , Aspartic Acid/chemistry , Aspartic Acid/pharmacology , Cell Line , Excitatory Amino Acid Transporter 3/chemistry , Magnetic Resonance Spectroscopy , Mass Spectrometry , Models, Molecular , Neurons/drug effects , Structure-Activity Relationship
15.
J Med Chem ; 61(17): 7741-7753, 2018 09 13.
Article in English | MEDLINE | ID: mdl-30011368

ABSTRACT

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


Subject(s)
Ammonia-Lyases/metabolism , Aspartic Acid/analogs & derivatives , Aspartic Acid/pharmacology , Excitatory Amino Acid Transporter 1/antagonists & inhibitors , Excitatory Amino Acid Transporter 3/antagonists & inhibitors , Excitatory Amino Acid Transporter 4/antagonists & inhibitors , Glutamate Plasma Membrane Transport Proteins/antagonists & inhibitors , Aspartic Acid/chemical synthesis , Excitatory Amino Acid Transporter 2 , HEK293 Cells , Humans , Molecular Structure , Structure-Activity Relationship
16.
ACS Chem Neurosci ; 8(8): 1668-1672, 2017 08 16.
Article in English | MEDLINE | ID: mdl-28414419

ABSTRACT

Excitatory amino acid transporters clear glutamate from the synaptic cleft and play a critical role in glutamatergic neurotransmission. Their differential roles in astrocytes, microglia, and neurons are poorly understood due in part to a lack of pharmacological tools that can be targeted to specific cells and tissues. We now describe a photoswitchable inhibitor, termed ATT, that interacts with the major mammalian forebrain transporters EAAT1-3 in a manner that can be reversibly switched between trans (high-affinity) and cis (low-affinity) configurations using light of different colors. In the dark, ATT competitively inhibited the predominant glial transporter EAAT2 with ∼200-fold selectivity over the neuronal transporter EAAT3. Brief exposure to 350 nm light reduced the steady-state blocker affinity by more than an order of magnitude. Illumination of EAAT2 complexed with ATT induced a corresponding increase in the blocker off-rate monitored in the presence of glutamate. ATT can be used to reversibly manipulate glutamate transporter activity with light and may be useful to gain insights into the dynamic physiological roles of glutamate transporters in the brain, as well as to study the molecular interactions of transporters with ligands.


Subject(s)
Aspartic Acid/analogs & derivatives , Excitatory Amino Acid Transporter 1/antagonists & inhibitors , Excitatory Amino Acid Transporter 3/antagonists & inhibitors , Glutamate Plasma Membrane Transport Proteins/antagonists & inhibitors , Membrane Transport Modulators/pharmacology , Animals , Aspartic Acid/chemical synthesis , Aspartic Acid/chemistry , Aspartic Acid/pharmacology , Dose-Response Relationship, Drug , Excitatory Amino Acid Transporter 1/metabolism , Excitatory Amino Acid Transporter 2 , Excitatory Amino Acid Transporter 3/metabolism , Glutamate Plasma Membrane Transport Proteins/metabolism , Humans , Isomerism , Light , Membrane Potentials/drug effects , Membrane Transport Modulators/chemical synthesis , Membrane Transport Modulators/chemistry , Molecular Structure , Oocytes , Patch-Clamp Techniques , Photochemical Processes , Xenopus laevis
17.
J Med Chem ; 49(1): 172-8, 2006 Jan 12.
Article in English | MEDLINE | ID: mdl-16392801

ABSTRACT

The natural product kainic acid is used as template for the rational design of a novel conformationally restricted (S)-glutamic acid (Glu) analogue, (1R,4S,5R,6S)-3-azabicyclo[3.3.0]octane-4,6-dicarboxylic acid (1a). The target structure 1a was synthesized from commercially available (S)-pyroglutaminol, in an enantioselective fashion, in 14 steps. Pharmacological characterization of 1a at human glutamate transporter subtypes 1, 2, and 3 yielded K(i) values of 127, 52, and 46 microM, respectively. Furthermore, binding studies at native ionotropic Glu (iGlu) receptors revealed low affinity for alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionate (AMPA)-preferring iGlu receptors (IC(50) > 100 microM), whereas affinities for the KAIN-preferring iGlu receptors and the N-methyl-d-aspartate (NMDA)-preferring group of iGlu receptors were in the low micromolar range (IC(50) = 14 and 2.9 microM, respectively). At metabotropic Glu receptors (mGluR), EC(50) values for 1a were >1000 microM for mGluR1 and 4, representing group I and III, respectively, and approximately 1000 microM (agonist) for mGluR2, representing group II.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/chemical synthesis , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Dicarboxylic Acids/chemical synthesis , Dicarboxylic Acids/pharmacology , Excitatory Amino Acid Transporter 1/antagonists & inhibitors , Excitatory Amino Acid Transporter 3/antagonists & inhibitors , Glutamate Plasma Membrane Transport Proteins/antagonists & inhibitors , Animals , Binding Sites , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Cell Line , Dicarboxylic Acids/chemistry , Drug Design , Excitatory Amino Acid Transporter 2 , Humans , Models, Molecular , Molecular Conformation , Rats , Receptors, Glutamate/drug effects , Stereoisomerism , Structure-Activity Relationship
18.
ACS Chem Neurosci ; 7(5): 534-9, 2016 05 18.
Article in English | MEDLINE | ID: mdl-26918289

ABSTRACT

Glutamate is the major excitatory neurotransmitter in the mammalian brain. Its rapid clearance after the release into the synaptic cleft is vital in order to avoid toxic effects and is ensured by several transmembrane transport proteins, so-called excitatory amino acid transporters (EAATs). Impairment of glutamate removal has been linked to several neurodegenerative diseases and EAATs have therefore received increased attention as therapeutic targets. O-Benzylated l-threo-ß-hydroxyaspartate derivatives have been developed previously as highly potent inhibitors of EAATs with TFB-TBOA ((2S,3S)-2-amino-3-((3-(4-(trifluoromethyl)benzamido)benzyl)oxy)succinic acid) standing out as low-nanomolar inhibitor. We report the stereoselective synthesis of all four stereoisomers of TFB-TBOA in less than a fifth of synthetic steps than the published route. For the first time, the inhibitory activity and isoform selectivity of these TFB-TBOA enantio- and diastereomers were assessed on human glutamate transporters EAAT1-3. Furthermore, we synthesized potent photoaffinity probes based on TFB-TBOA using our novel synthetic strategy.


Subject(s)
Aspartic Acid/analogs & derivatives , Excitatory Amino Acid Transporter 1/antagonists & inhibitors , Excitatory Amino Acid Transporter 3/antagonists & inhibitors , Glutamate Plasma Membrane Transport Proteins/antagonists & inhibitors , Photoaffinity Labels/chemical synthesis , Aspartic Acid/chemical synthesis , Aspartic Acid/pharmacology , Chemistry, Pharmaceutical , Dose-Response Relationship, Drug , Excitatory Amino Acid Transporter 1/metabolism , Excitatory Amino Acid Transporter 2 , Excitatory Amino Acid Transporter 3/metabolism , Glutamate Plasma Membrane Transport Proteins/metabolism , HEK293 Cells , Humans , Photoaffinity Labels/pharmacology , Stereoisomerism
19.
J Med Chem ; 59(19): 8771-8786, 2016 10 13.
Article in English | MEDLINE | ID: mdl-27636002

ABSTRACT

In this study inspired by previous work on 3-substituted Asp analogues, we designed and synthesized a total of 32 ß-sulfonamide Asp analogues and characterized their pharmacological properties at the excitatory amino acid transporter subtypes EAAT1, EAAT2, and EAAT3. In addition to several potent EAAT inhibitors displaying IC50 values ∼1 µM at all three subtypes, this elaborate structure-activity relationship also identified analogues exhibiting distinct preferences or selectivities for specific transporter subtypes. Introduction of two fluorine atoms on the phenyl ring yielded analogue 4y that displayed an IC50 of 0.8 µM at EAAT1 with a 14- and 9-fold preference over EAAT2 and EAAT3, respectively. Conversely, the m-CF3-phenyl analogue 4r was a potent selective EAAT2-inhibitor (IC50 = 2.8 µM) exhibiting 30- and 50-fold selectivity over EAAT1 and EAAT3, respectively. In conclusion, even small structural differences in these ß-sulfonamide Asp analogues provide analogues with diverse EAAT subtype selectivity profiles.


Subject(s)
Aspartic Acid/analogs & derivatives , Aspartic Acid/pharmacology , Excitatory Amino Acid Transporter 1/antagonists & inhibitors , Excitatory Amino Acid Transporter 3/antagonists & inhibitors , Glutamate Plasma Membrane Transport Proteins/antagonists & inhibitors , Sulfonamides/chemistry , Sulfonamides/pharmacology , Biological Transport/drug effects , Excitatory Amino Acid Transporter 1/metabolism , Excitatory Amino Acid Transporter 2 , Excitatory Amino Acid Transporter 3/metabolism , Glutamate Plasma Membrane Transport Proteins/metabolism , HEK293 Cells , Humans
20.
Nat Commun ; 7: 10604, 2016 Feb 03.
Article in English | MEDLINE | ID: mdl-26837579

ABSTRACT

Spinal mGluR5 is a key mediator of neuroplasticity underlying persistent pain. Although brain mGluR5 is localized on cell surface and intracellular membranes, neither the presence nor physiological role of spinal intracellular mGluR5 is established. Here we show that in spinal dorsal horn neurons >80% of mGluR5 is intracellular, of which ∼60% is located on nuclear membranes, where activation leads to sustained Ca(2+) responses. Nerve injury inducing nociceptive hypersensitivity also increases the expression of nuclear mGluR5 and receptor-mediated phosphorylated-ERK1/2, Arc/Arg3.1 and c-fos. Spinal blockade of intracellular mGluR5 reduces neuropathic pain behaviours and signalling molecules, whereas blockade of cell-surface mGluR5 has little effect. Decreasing intracellular glutamate via blocking EAAT-3, mimics the effects of intracellular mGluR5 antagonism. These findings show a direct link between an intracellular GPCR and behavioural expression in vivo. Blockade of intracellular mGluR5 represents a new strategy for the development of effective therapies for persistent pain.


Subject(s)
Behavior, Animal , Calcium/metabolism , Glutamic Acid/metabolism , Hyperalgesia/metabolism , Neuralgia/metabolism , Posterior Horn Cells/metabolism , Receptor, Metabotropic Glutamate 5/metabolism , Sciatic Neuropathy/metabolism , Analgesics, Opioid/pharmacology , Animals , Blotting, Western , Cells, Cultured , Cytoskeletal Proteins/metabolism , Excitatory Amino Acid Transporter 3/antagonists & inhibitors , Glutamic Acid/pharmacology , Hyperalgesia/pathology , Immunohistochemistry , Injections, Spinal , Male , Microdialysis , Microscopy, Confocal , Microscopy, Electron , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Morphine/pharmacology , Nerve Tissue Proteins/metabolism , Posterior Horn Cells/pathology , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Rats , Rats, Long-Evans , Sciatic Nerve/injuries , Sciatic Neuropathy/pathology
SELECTION OF CITATIONS
SEARCH DETAIL