Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters











Publication year range
1.
J Neurosci ; 31(7): 2584-94, 2011 Feb 16.
Article in English | MEDLINE | ID: mdl-21325526

ABSTRACT

The potential for relapse following cessation of drug use can last for years, implying the induction of stable changes in neural circuitry. In hippocampal slices from rats treated with nicotine for 1 week, withdrawal from nicotine in vivo produces an increase in CA1 pyramidal cell excitability that persists up to 9 months. Immediately upon drug cessation, the enhanced excitability depends on input from regions upstream of CA1, while the long-term excitability change (> 4 weeks) is expressed as an increase in the intrinsic excitability of CA1 neurons. Re-exposure to nicotine in vitro restores hippocampal function to control levels via activation of high-affinity nicotinic acetylcholine receptors after 1 d of withdrawal, but not at times >4 weeks. Thus, nicotine in vivo first induces homeostatic adaptations followed by other more robust neural changes. These mechanisms may contribute to hippocampal localized cue-motivated reinstatement of drug-seeking and/or cognitive deficits observed during withdrawal.


Subject(s)
Action Potentials/drug effects , Excitatory Postsynaptic Potentials/drug effects , Hippocampus/drug effects , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , 2-Amino-5-phosphonovalerate/pharmacology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Dihydro-beta-Erythroidine/pharmacology , Dose-Response Relationship, Drug , Drug Administration Schedule , Electric Stimulation/methods , Excitatory Amino Acid Antagonists/pharmacology , In Vitro Techniques , Male , Patch-Clamp Techniques/methods , Rats , Rats, Sprague-Dawley , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology , Time Factors
2.
J Gen Physiol ; 134(6): 489-521, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19948998

ABSTRACT

The mouse gamma-aminobutyric acid (GABA) transporter mGAT1 was expressed in neuroblastoma 2a cells. 19 mGAT1 designs incorporating fluorescent proteins were functionally characterized by [(3)H]GABA uptake in assays that responded to several experimental variables, including the mutations and pharmacological manipulation of the cytoskeleton. Oligomerization and subsequent trafficking of mGAT1 were studied in several subcellular regions of live cells using localized fluorescence, acceptor photobleach Förster resonance energy transfer (FRET), and pixel-by-pixel analysis of normalized FRET (NFRET) images. Nine constructs were functionally indistinguishable from wild-type mGAT1 and provided information about normal mGAT1 assembly and trafficking. The remainder had compromised [(3)H]GABA uptake due to observable oligomerization and/or trafficking deficits; the data help to determine regions of mGAT1 sequence involved in these processes. Acceptor photobleach FRET detected mGAT1 oligomerization, but richer information was obtained from analyzing the distribution of all-pixel NFRET amplitudes. We also analyzed such distributions restricted to cellular subregions. Distributions were fit to either two or three Gaussian components. Two of the components, present for all mGAT1 constructs that oligomerized, may represent dimers and high-order oligomers (probably tetramers), respectively. Only wild-type functioning constructs displayed three components; the additional component apparently had the highest mean NFRET amplitude. Near the cell periphery, wild-type functioning constructs displayed the highest NFRET. In this subregion, the highest NFRET component represented approximately 30% of all pixels, similar to the percentage of mGAT1 from the acutely recycling pool resident in the plasma membrane in the basal state. Blocking the mGAT1 C terminus postsynaptic density 95/discs large/zona occludens 1 (PDZ)-interacting domain abolished the highest amplitude component from the NFRET distributions. Disrupting the actin cytoskeleton in cells expressing wild-type functioning transporters moved the highest amplitude component from the cell periphery to perinuclear regions. Thus, pixel-by-pixel NFRET analysis resolved three distinct forms of GAT1: dimers, high-order oligomers, and transporters associated via PDZ-mediated interactions with the actin cytoskeleton and/or with the exocyst.


Subject(s)
GABA Plasma Membrane Transport Proteins/chemistry , GABA Plasma Membrane Transport Proteins/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Biological Transport , Cell Line , Cell Membrane , Fluorescence Resonance Energy Transfer , GABA Plasma Membrane Transport Proteins/genetics , Humans , Mice
3.
Neuropharmacology ; 55(5): 763-70, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18602929

ABSTRACT

Plasma membrane serotonin transporters (SERTs) regulate serotonin (5HT) levels in brain and are a site of action of antidepressants and psychostimulant drugs of abuse. Syntaxin 1A is a component of the synaptic vesicle docking and fusion apparatus and has been shown to interact with multiple plasma membrane neurotransmitter transporters including SERT. Previously, we showed that syntaxin 1A regulates the transport stoichiometry of SERT. When not bound to syntaxin 1A, SERT shows both substrate-independent Na(+) fluxes and substrate-dependent Na(+) fluxes of variable stoichiometry; these fluxes are eliminated in the presence of syntaxin 1A as Na(+) flux becomes strictly coupled to 5HT uptake. However, not known are the endogenous signaling molecules that determine the conducting states that SERT exhibits. In the present experiments, we show that inhibitors of calcium/calmodulin-dependent kinase II (CaM kinase II) modulate the stoichiometry of 5HT flux and that this effect requires syntaxin 1A. The modulation correlates with a shift in the affinity of SERT for syntaxin 1A binding. The regulation by CaM kinase II is eliminated by a mutation in the N-terminal domain of SERT. In neonatal thalomocortical neurons that endogenously express SERT and syntaxin 1A, inhibition of CaM kinase II reveals SERT-mediated currents. These data suggest that calcium-mediated signals can serve as a trigger for regulating protein-protein interactions that control SERT conducting states.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/physiology , Serotonin Plasma Membrane Transport Proteins/metabolism , Syntaxin 1/metabolism , Analysis of Variance , Animals , Brain/cytology , Cells, Cultured , Dose-Response Relationship, Drug , Drug Interactions , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Microinjections , Neurons/drug effects , Oocytes , Paroxetine/pharmacology , Patch-Clamp Techniques , Rats , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/genetics , Selective Serotonin Reuptake Inhibitors/pharmacology , Syntaxin 1/genetics , Xenopus laevis
4.
Neuropharmacology ; 54(2): 309-18, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17991494

ABSTRACT

The uptake of neurotransmitter by plasma membrane transporters is a principal method for regulating extracellular transmitter levels. Neurotransmitter-mediated signals in turn are able to regulate transporter expression and function. Thus, there is a continual interplay between transporters and the transmitters they transport. Previously we showed that extracellular gamma-aminobutyric acid (GABA) increases the expression of the GABA transporter 1 (GAT1) on a time scale of minutes by acting via the transporter to slow transporter internalization. This mechanism requires in part direct tyrosine phosphorylation of the transporter. In the present study we show that the presence of GABA on a longer time scale causes a net decrease in GAT surface expression. The decrease in expression represents the contributions of transporter-mediated up-regulation and a more substantial GABA-receptor-mediated down-regulation. This receptor-mediated down-regulation is the result of both changes in the rates of transporter trafficking and in the number of transporters available for trafficking. As with transporter-mediated regulation of GAT1, the receptor-mediated regulation is associated with changes in the direct phosphorylation of GAT1. These data suggest that multiple pathways, perhaps converging upon mechanisms involving protein phosphorylation, act to regulate GAT1 expression in neurons.


Subject(s)
Brain Chemistry/physiology , GABA Plasma Membrane Transport Proteins/biosynthesis , Animals , Bicuculline/pharmacology , Biotin/metabolism , Brain Chemistry/drug effects , Cells, Cultured , Down-Regulation/drug effects , Endocytosis/drug effects , Exocytosis/drug effects , Female , GABA Antagonists/pharmacology , Immunoprecipitation , Neurons/drug effects , Neurons/physiology , Nipecotic Acids/pharmacology , Phosphorylation , Pregnancy , Rats , Receptors, GABA-A/drug effects , Serine/metabolism , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/pharmacology
5.
Mol Biol Cell ; 17(7): 3021-30, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16641366

ABSTRACT

Sodium-dependent neurotransmitter transporters participate in the clearance and/or recycling of neurotransmitters from synaptic clefts. The snf-11 gene in Caenorhabditis elegans encodes a protein of high similarity to mammalian GABA transporters (GATs). We show here that snf-11 encodes a functional GABA transporter; SNF-11-mediated GABA transport is Na+ and Cl- dependent, has an EC50 value of 168 microM, and is blocked by the GAT1 inhibitor SKF89976A. The SNF-11 protein is expressed in seven GABAergic neurons, several additional neurons in the head and retrovesicular ganglion, and three groups of muscle cells. Therefore, all GABAergic synapses are associated with either presynaptic or postsynaptic (or both) expression of SNF-11. Although a snf-11 null mutation has no obvious effects on GABAergic behaviors, it leads to resistance to inhibitors of acetylcholinesterase. In vivo, a snf-11 null mutation blocks GABA uptake in at least a subset of GABAergic cells; in a cell culture system, all GABA uptake is abolished by the snf-11 mutation. We conclude that GABA transport activity is not essential for normal GABAergic function in C. elegans and that the localization of SNF-11 is consistent with a GABA clearance function rather than recycling.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/metabolism , GABA Plasma Membrane Transport Proteins/physiology , Genes, Helminth/physiology , Synapses/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Caenorhabditis elegans/chemistry , Caenorhabditis elegans Proteins/analysis , Caenorhabditis elegans Proteins/genetics , GABA Agents/pharmacology , GABA Plasma Membrane Transport Proteins/analysis , GABA Plasma Membrane Transport Proteins/genetics , Mutation , Nipecotic Acids/pharmacology , Phenotype , Phylogeny , Sodium/metabolism , Synaptic Transmission
6.
J Neurosci ; 25(14): 3712-23, 2005 Apr 06.
Article in English | MEDLINE | ID: mdl-15814802

ABSTRACT

Alpha7 nicotinic acetylcholine receptors (nAChRs) modulate network activity in the CNS. Thus, functional regulation of alpha7 nAChRs could influence the flow of information through various brain nuclei. It is hypothesized here that these receptors are amenable to modulation by tyrosine phosphorylation. In both Xenopus oocytes and rat hippocampal interneurons, brief exposure to a broad-spectrum protein tyrosine kinase inhibitor, genistein, specifically and reversibly potentiated alpha7 nAChR-mediated responses, whereas a protein tyrosine phosphatase inhibitor, pervanadate, caused depression. Potentiation was associated with an increased expression of surface alpha7 subunits and was not accompanied by detectable changes in receptor open probability, implying that the increased function results from an increased number of alpha7 nAChRs. Soluble N-ethylmaleimide-sensitive factor attachment protein receptor-mediated exocytosis was shown to be a plausible mechanism for the rapid delivery of additional alpha7 nAChRs to the plasma membrane. Direct phosphorylation/dephosphorylation of alpha7 subunits was unlikely because mutation of all three cytoplasmic tyrosine residues did not prevent the genistein-mediated facilitation. Overall, these data are consistent with the hypothesis that the number of functional cell surface alpha7 nAChRs is controlled indirectly via processes involving tyrosine phosphorylation.


Subject(s)
Interneurons/physiology , Receptors, Nicotinic/metabolism , Tyrosine/metabolism , Acetylcholine/pharmacology , Animals , Biotinylation/methods , Blotting, Western/methods , Bungarotoxins/pharmacokinetics , Choline/pharmacology , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Drug Interactions , Electric Stimulation/methods , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Genistein/pharmacology , Hippocampus/cytology , Insulin/pharmacology , Interneurons/drug effects , Iodine Isotopes/pharmacokinetics , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Microinjections/methods , Mutagenesis/physiology , N-Methylaspartate/pharmacology , Oocytes , Patch-Clamp Techniques/methods , Phosphoric Monoester Hydrolases/pharmacology , Phosphorylation/drug effects , Protein-Tyrosine Kinases/pharmacology , RNA, Messenger/biosynthesis , Radioligand Assay/methods , Rats , Receptors, Nicotinic/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , SNARE Proteins/metabolism , Time Factors , Up-Regulation/drug effects , Up-Regulation/physiology , Vanadates/pharmacology , Xenopus , alpha7 Nicotinic Acetylcholine Receptor
7.
J Biol Chem ; 280(19): 18703-9, 2005 May 13.
Article in English | MEDLINE | ID: mdl-15778221

ABSTRACT

Plasma membrane neurotransmitter transporters rapidly traffic to and from the cell surface in neurons. This trafficking may be important in regulating neuronal signaling. Such regulation will be subject to the number of trafficking transporters and their trafficking rates. In the present study, we define an acutely recycling pool of endogenous gamma-aminobutyric acid transporters (GAT1) in cortical neurons that comprises approximately one-third of total cellular GAT1. Kinetic analysis of this pool estimates exocytosis and endocytosis time constants of 1.6 and 0.9 min, respectively, and thus approximately one-third of the recycling pool is plasma membrane resident in the basal state. Recent evidence shows that GAT1 substrates, second messengers, and interacting proteins regulate GAT1 trafficking. These triggers could act by altering trafficking rates or by changing the recycling pool size. In the present study we examine three GAT1 modulators. Calcium depletion decreases GAT1 surface expression by diminishing the recycling pool size. Sucrose increases GAT1 surface expression by blocking clathrin- and dynamin-dependent endocytosis, but it does not change the recycling pool size. Protein kinase C decreases surface GAT1 expression by increasing the endocytosis rate, but it does not change the exocytosis rate or the recycling pool size. Based upon estimates of GAT1 molecules in cortical boutons, the present data suggest that approximately 1000 transporters comprise the acutely recycling pool, of which 300 are on the surface in the basal state, and five transporters insert into the plasma membrane every second. This insertion could represent the fusion of one transporter-containing vesicle.


Subject(s)
Cell Membrane/metabolism , Membrane Transport Proteins/metabolism , Animals , Biological Transport , Biotinylation , CHO Cells , Calcium/chemistry , Cells, Cultured , Cerebral Cortex/metabolism , Clathrin/chemistry , Cricetinae , Dynamins/chemistry , Endocytosis , Exocytosis , GABA Plasma Membrane Transport Proteins , Immunoblotting , Kinetics , Neurons/metabolism , Protein Kinase C/chemistry , Protein Kinase C/metabolism , Protein Transport , Rats , Signal Transduction , Sucrose/chemistry , Tetradecanoylphorbol Acetate , Time Factors
8.
J Neurosci ; 24(16): 4082-7, 2004 Apr 21.
Article in English | MEDLINE | ID: mdl-15102923

ABSTRACT

Plasma membrane neurotransmitter transporters determine in part the concentration, time course, and diffusion of extracellular transmitter. Much has been learned about how substrate translocation through the transporter occurs; however, the precise way in which transporter structure maps onto transporter function has not yet been fully elucidated. Here, biochemical and electrophysiological approaches were used to test the hypothesis that intracellular domains of the rat brain GABA transporter (GAT1) contribute to the transport process. Injection of a peptide corresponding to the presumed fourth intracellular loop of the transporter (IL4) into oocytes expressing GAT1 greatly reduced both forward and reverse transport and reduced the transport rate in a dose-dependent manner. Coinjection of the IL4 peptide with a peptide corresponding to the N-terminal cytoplasmic tail of GAT1 reversed the IL4-mediated inhibition; this reversal, and direct binding between these two domains, was prevented by mutagenesis of charged residues in either the IL4 or N-terminal domains. Furthermore, syntaxin 1A, a soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) protein that inhibits GAT1 transport rates via interactions with the N-terminal tail of GAT1 was unable to regulate the GAT1 IL4 mutant. Together, these data suggest a model in which the GAT1 IL4 domain serves as a barrier for transport, and this barrier can be regulated through intra-molecular and inter-molecular interactions.


Subject(s)
Brain/metabolism , Carrier Proteins/metabolism , Membrane Proteins/metabolism , Membrane Transport Proteins , Animals , Antigens, Surface/genetics , Antigens, Surface/metabolism , Biological Transport/physiology , Carrier Proteins/genetics , Cells, Cultured , GABA Plasma Membrane Transport Proteins , Membrane Proteins/genetics , Mutagenesis, Site-Directed , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Oocytes/metabolism , Patch-Clamp Techniques , Protein Structure, Tertiary/genetics , Protein Structure, Tertiary/physiology , Protein Transport/genetics , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Structure-Activity Relationship , Syntaxin 1 , Xenopus , gamma-Aminobutyric Acid/metabolism
9.
J Biol Chem ; 279(16): 15961-7, 2004 Apr 16.
Article in English | MEDLINE | ID: mdl-14761965

ABSTRACT

A feature of the rat brain gamma-aminobutyric acid transporter GAT1, and other members of the neurotransmitter transporter family, is its regulated redistribution between intracellular locations and the plasma membrane. Recent studies have focused upon defining the signaling molecules that facilitate this redistribution. Agents that promote direct tyrosine phosphorylation of GAT1 promote a relative increase in surface GAT1 levels, and this results from a slowing of the transporter internalization rate. Agents that act to increase protein kinase C (PKC) activity promote a relative decrease in surface GAT1 levels; whether this effect is caused by direct transporter phosphorylation is unknown. The opposing actions of tyrosine kinase activity and PKC activity raise the possibility that the subcellular distribution of GAT1 is associated with mutually exclusive transporter phosphorylation events. The present experiments show that GAT1 is phosphorylated on serine residues in a PKC-dependent manner, but this state is only revealed when GAT1 tyrosine phosphorylation is eliminated or greatly reduced. The relative levels of serine phosphorylation and tyrosine phosphorylation are negatively correlated. The amount of serine phosphorylation is regulated by agents that affect tyrosine phosphorylation, and vice versa. In addition, the ability of agents that affect tyrosine kinase activity to regulate GAT1 serine phosphorylation requires a change in its tyrosine phosphorylation state. These data support the ideas that GAT1 can exist in either of two mutually exclusive phosphorylation states and that the relative abundance of these states determines in part the relative subcellular distribution of the transporter.


Subject(s)
Carrier Proteins/metabolism , Hippocampus/metabolism , Membrane Proteins/metabolism , Membrane Transport Proteins , Animals , Biological Transport , Cells, Cultured , GABA Plasma Membrane Transport Proteins , Phosphorylation , Rats , Serine , Tyrosine
10.
Pflugers Arch ; 447(5): 519-31, 2004 Feb.
Article in English | MEDLINE | ID: mdl-12719981

ABSTRACT

The SLC6 family is a diverse set of transporters that mediate solute translocation across cell plasma membranes by coupling solute transport to the cotransport of sodium and chloride down their electrochemical gradients. These transporters probably have 12 transmembrane domains, with cytoplasmic N- and C-terminal tails, and at least some may function as homo-oligomers. Family members include the transporters for the inhibitory neurotransmitters GABA and glycine, the aminergic transmitters norepinephrine, serotonin, and dopamine, the osmolytes betaine and taurine, the amino acid proline, and the metabolic compound creatine. In addition, this family includes a system B(0+) cationic and neutral amino acid transporter, and two transporters for which the solutes are unknown. In general, SLC6 transporters act to regulate the level of extracellular solute concentrations. In the central and the peripheral nervous system, these transporters can regulate signaling among neurons, are the sites of action of various drugs of abuse, and naturally occurring mutations in several of these proteins are associated with a variety of neurological disorders. For example, transgenic animals lacking specific aminergic transporters show profoundly disturbed behavioral phenotypes and probably represent excellent systems for investigating psychiatric disease. SLC6 transporters are also found in many non-neural tissues, including kidney, intestine, and testis, consistent with their diverse physiological roles. Transporters in this family represent attractive therapeutic targets because they are subject to multiple forms of regulation by many different signaling cascades, and because a number of pharmacological agents have been identified that act specifically on these proteins.


Subject(s)
Chlorides/metabolism , Membrane Transport Proteins/physiology , Neurotransmitter Agents/metabolism , Sodium/metabolism , Synapses/metabolism , Biological Transport/physiology , Humans , Membrane Transport Proteins/chemistry , Multigene Family/physiology
11.
Neuron ; 40(3): 537-49, 2003 Oct 30.
Article in English | MEDLINE | ID: mdl-14642278

ABSTRACT

Serotonin transporters (SERTs), sites of psychostimulant action, display multiple conducting states in expression systems. These include a substrate-independent transient conductance, two separate substrate-independent leak conductances associated with Na(+) and H(+), and a substrate-dependent conductance of variable stoichiometry, which exceeds that predicted from electroneutral substrate transport. The present data show that the SNARE protein syntaxin 1A binds the N-terminal tail of SERT, and this interaction regulates two SERT-conducting states. First, substrate-induced currents are absent because Na(+) flux becomes strictly coupled to 5HT transport. Second, Na(+)-mediated leak currents are eliminated. These two SERT-conducting states are present endogenously in thalamocortical neurons, act to depolarize the membrane potential, and are modulated by molecules that disrupt SERT and syntaxin 1A interactions. These data show that protein interactions govern SERT activity and suggest that both cell excitability and psychostimulant-mediated effects will be dependent upon the state of association among SERT and its interacting partners.


Subject(s)
Antigens, Surface/metabolism , Carrier Proteins/physiology , Cocaine/analogs & derivatives , Electric Conductivity , Membrane Glycoproteins/physiology , Membrane Transport Proteins , Nerve Tissue Proteins/metabolism , Serotonin/metabolism , Vesicular Transport Proteins , Animals , Binding Sites , Botulinum Toxins, Type A/pharmacology , Carrier Proteins/genetics , Cells, Cultured , Choline/metabolism , Cocaine/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Embryo, Mammalian , Embryo, Nonmammalian , Fluoxetine/pharmacology , Humans , Immunoblotting , Isotope Labeling , Kidney , Membrane Glycoproteins/genetics , Membrane Potentials/drug effects , Membrane Potentials/physiology , Microinjections , Molecular Biology , Munc18 Proteins , Mutation , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Neurons/drug effects , Neurons/physiology , Oocytes , Patch-Clamp Techniques/methods , Peptide Fragments/pharmacology , Proteins/pharmacology , Radiopharmaceuticals/metabolism , Rats , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Serotonin Agents/pharmacology , Serotonin Plasma Membrane Transport Proteins , Selective Serotonin Reuptake Inhibitors/pharmacology , Sodium/metabolism , Syntaxin 1 , Thalamus/cytology , Thalamus/metabolism , Time Factors , Tritium/metabolism , Xenopus laevis
12.
Mol Pharmacol ; 64(4): 905-13, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14500747

ABSTRACT

GABA transporters control extracellular GABA levels by coupling transmitter uptake to the sodium and chloride cotransport. The rat brain GABA transporter GAT1 and other members of this family are regulated by direct interactions with syntaxin 1A, a protein involved in vesicle docking and in the regulation of several ion channels and transporters. We have shown previously that syntaxin 1A exerts its effects on GAT1 by decreasing the net uptake of GABA and its associated ions through interactions with aspartic acid residues in the N-terminal tail of GAT1. This reduction in net uptake could be caused by many steps in the transport cycle, including substrate binding, substrate flux, substrate efflux, or reorientation of the unliganded transporter. To address this question, we performed GABA flux assays, measured flux- and efflux-associated ion currents, and assessed GABA exchange in multiple experimental systems expressing syntaxin 1A and wild-type GAT1 or GAT1 mutants. Syntaxin 1A caused similar reductions in forward and reverse transport that did not involve changes in apparent transport affinities for sodium, chloride, or GABA. The syntaxin 1A-mediated reduction in GABA flux and efflux was mimicked by mutations in GAT1 at the syntaxin 1A binding site. The binding site GAT1 mutant also caused a reduction in exchange. These data suggest that syntaxin 1A exerts its effects, directly or indirectly, on GAT1 function through interactions with GAT1's N-terminal tail and that the inhibition occurs at a step in the translocation process after substrate binding but which involves both unidirectional transport and transmitter exchange.


Subject(s)
Antigens, Surface/pharmacology , Carrier Proteins/metabolism , Hippocampus/drug effects , Membrane Proteins/metabolism , Membrane Transport Proteins , Nerve Tissue Proteins/pharmacology , Organic Anion Transporters , gamma-Aminobutyric Acid/metabolism , Animals , Binding Sites , Biological Transport/drug effects , Carrier Proteins/genetics , GABA Plasma Membrane Transport Proteins , Hippocampus/metabolism , Membrane Proteins/genetics , Oocytes/drug effects , Oocytes/metabolism , Rats , Syntaxin 1 , Xenopus laevis
13.
Br J Pharmacol ; 138(5): 801-10, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12642381

ABSTRACT

(1) Atropine, a classical muscarinic antagonist, has been reported previously to inhibit neuronal nicotinic acetylcholine receptors (nAChRs). In the present study, the action of atropine has been examined on alpha3beta4 receptors expressed heterologously in Xenopus oocytes and native nAChRs in medial habenula neurons. (2) At concentrations of atropine often used to inhibit muscarinic receptors (1 micro M), responses induced by near-maximal nicotine concentrations (100 micro M) at negative holding potentials (-65 mV) are inhibited (14-30%) in a reversible manner in both alpha4 and alpha3 subunit-containing heteromeric nAChRs. Half-maximal effective concentrations (IC(50) values) for atropine inhibition are similar for the four classes of heteromeric receptors studied (4-13 micro M). (3) For alpha3beta4 nAChRs in oocytes, inhibition by atropine (10 micro M) is not overcome at higher concentrations of agonist, and is increased with membrane hyperpolarization. These results are consistent with non-competitive antagonism--possibly ion channel block. (4) At low concentrations of both nicotine (10 micro M) and atropine (<10 micro M), potentiation ( approximately 25%) of alpha3beta4 nAChR responses in oocytes is observed. The relative balance between potentiation and inhibition is dependent upon membrane potential. (5) In rat medial habenula (MHb) neurons, atropine (0.3-3.0 micro M) inhibited nicotine-induced responses in both a concentration and membrane potential-dependent manner (at -40 mV, IC(50)=4 micro M), similar to the effects on alpha3beta4-nAChRs in oocytes. However, unlike heterologously expressed receptors, potentiation was barely detectable at depolarized membrane potentials using low concentrations of nicotine (3-10 micro M). Conversely, the weak agonist, choline (1-3 mM) was observed to augment responses of MHb nAChRs.


Subject(s)
Atropine/pharmacology , Gene Expression Regulation/physiology , Receptors, Nicotinic/biosynthesis , Animals , Dose-Response Relationship, Drug , Female , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neurons/drug effects , Neurons/metabolism , Oocytes/drug effects , Oocytes/metabolism , Rats , Receptors, Nicotinic/genetics , Xenopus laevis
14.
J Neurosci ; 23(5): 1563-8, 2003 Mar 01.
Article in English | MEDLINE | ID: mdl-12629157

ABSTRACT

Plasma membrane neurotransmitter transporters affect synaptic signaling through transmitter sequestration. Transporters redistribute to and from the plasma membrane, suggesting a role for trafficking in regulating synaptic transmitter levels. One method for controlling transmitter levels would be to regulate transporter redistribution in parallel with transmitter release. Thus, how similar are these processes? We show that the trafficking of the GABA transporter GAT1 resembles the trafficking of neurotransmitter-filled synaptic vesicles: (1) transporters located on the plasma membrane are internalized and reinserted into the plasma membrane on the order of minutes; (2) the rate of recycling is depolarization and calcium dependent; (3) GAT1 internalization is associated with clathrin and dynamin; and (4) intracellular GAT1 is associated with multiple compartments and, more importantly, is found on a distinct class of vesicles. These vesicles are clear, approximately 50 nm in diameter, and contain many proteins found on neurotransmitter-containing small synaptic vesicles; however, they appear to lack several traditional small synaptic vesicle proteins, such as synaptophysin and the vesicular GABA transporter. These data provide additional support for the hypothesis that GABA transporters traffic in parallel with neurotransmitter-containing small synaptic vesicles and also raise the possibility that some fraction of vesicles found in GABAergic neurons may not be participating in transmitter release but rather in the rapid regulated redistribution of membrane proteins involved in transmitter uptake.


Subject(s)
Carrier Proteins/metabolism , Cell Membrane/metabolism , Membrane Proteins/metabolism , Membrane Transport Proteins , Neurons/metabolism , Organic Anion Transporters , Synaptic Vesicles/metabolism , Animals , Avidin/chemistry , Biotin/chemistry , Brain Chemistry , CHO Cells , Cells, Cultured , Clathrin/metabolism , Cricetinae , GABA Plasma Membrane Transport Proteins , Hippocampus/cytology , Neurons/cytology , Protein Transport/physiology , Rats , Synapses/metabolism , Synaptic Vesicles/chemistry , Synaptosomes/chemistry , Synaptosomes/metabolism
15.
J Neurosci ; 23(5): 1697-709, 2003 Mar 01.
Article in English | MEDLINE | ID: mdl-12629174

ABSTRACT

Norepinephrine (NE) transporters (NETs) terminate noradrenergic synaptic transmission and represent a major therapeutic target for antidepressant medications. NETs and related transporters are under intrinsic regulation by receptor and kinase-linked pathways, and clarification of these pathways may suggest candidates for the development of novel therapeutic approaches. Syntaxin 1A, a presynaptic soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) protein, interacts with NET and modulates NET intrinsic activity. NETs colocalize with and bind to syntaxin 1A in both native preparations and heterologous systems. Protein kinase C activation disrupts surface NET/syntaxin 1A interactions and downregulates NET activity in a syntaxin-dependent manner. Syntaxin 1A binds the NH(2) terminal domain of NET, and a deletion of this domain both eliminates NET/syntaxin 1A associations and prevents phorbol ester-triggered NET downregulation. Whereas syntaxin 1A supports the surface trafficking of NET proteins, its direct interaction with NET limits transporter catalytic function. These two contradictory roles of syntaxin 1A on NET appear to be linked and reveal a dynamic cycle of interactions that allow for the coordinated control between NE release and reuptake.


Subject(s)
Antigens, Surface/metabolism , Catecholamines/metabolism , Nerve Tissue Proteins/metabolism , Symporters/metabolism , Vesicular Transport Proteins , Animals , Antidepressive Agents/pharmacology , Antigens, Surface/genetics , Botulinum Toxins/pharmacology , Brain Chemistry , Catecholamines/pharmacokinetics , Cells, Cultured , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Activators/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Norepinephrine/metabolism , Norepinephrine/pharmacokinetics , Norepinephrine Plasma Membrane Transport Proteins , Oligonucleotides, Antisense/pharmacology , Patch-Clamp Techniques , Protein Binding/physiology , Protein Structure, Tertiary/physiology , Protein Transport/physiology , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , SNARE Proteins , Sequence Deletion , Symporters/drug effects , Symporters/genetics , Synaptosomes/chemistry , Synaptosomes/metabolism , Syntaxin 1 , Vas Deferens/chemistry , Vas Deferens/metabolism
16.
Nat Neurosci ; 6(3): 243-50, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12592409

ABSTRACT

The molecular mechanisms underlying polarized sorting of proteins in neurons are poorly understood. Here we report the identification of a 16 amino-acid, dileucine-containing motif that mediates dendritic targeting in a variety of neuronal cell types in slices of rat brain. This motif is present in the carboxy (C) termini of Shal-family K+ channels and is highly conserved from C. elegans to humans. It is necessary for dendritic targeting of potassium channel Kv4.2 and is sufficient to target the axonally localized channels Kv1.3 and Kv1.4 to the dendrites. It can also mediate dendritic targeting of a non-channel protein, CD8.


Subject(s)
Dendrites/metabolism , Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Amino Acid Motifs/genetics , Amino Acid Motifs/physiology , Amino Acid Sequence , Animals , CD8 Antigens/genetics , CD8 Antigens/metabolism , Cells, Cultured , Conserved Sequence , Endocytosis , In Vitro Techniques , Kv1.3 Potassium Channel , Kv1.4 Potassium Channel , Leucine , Molecular Sequence Data , Potassium Channels/genetics , Protein Transport/physiology , Pyramidal Cells/metabolism , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Shal Potassium Channels
17.
J Biol Chem ; 278(5): 2876-85, 2003 Jan 31.
Article in English | MEDLINE | ID: mdl-12446681

ABSTRACT

Syntaxin 1A binds to and inhibits epithelial cystic fibrosis transmembrane conductance regulator (CFTR) Cl(-) channels and synaptic Ca(2+) channels in addition to participating in SNARE complex assembly and membrane fusion. We exploited the isoform-specific nature of the interaction between syntaxin 1A and CFTR to identify residues in the H3 domain of this SNARE (SNARE motif) that influence CFTR binding and regulation. Mutating isoform-specific residues that map to the surface of syntaxin 1A in the SNARE complex led to the identification of two sets of hydrophilic residues that are important for binding to and regulating CFTR channels or for binding to the syntaxin regulatory protein Munc-18a. None of these mutations affected syntaxin 1A binding to other SNAREs or the assembly and stability of SNARE complexes in vitro. Conversely, the syntaxin 1A-CFTR interaction was unaffected by mutating hydrophobic residues in the H3 domain that influence SNARE complex stability and Ca(2+) channel regulation. Thus, CFTR channel regulation by syntaxin 1A involves hydrophilic interactions that are mechanistically distinct from the hydrophobic interactions that mediate SNARE complex formation and Ca(2+) channel regulation by this t-SNARE.


Subject(s)
Antigens, Surface/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Vesicular Transport Proteins , Amino Acid Sequence , Amino Acid Substitution , Animals , Carrier Proteins/metabolism , Cell Line , Chloride Channels/metabolism , Cricetinae , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Electrophysiology , Female , Humans , Kinetics , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Oocytes/physiology , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Structure, Secondary , Qb-SNARE Proteins , Qc-SNARE Proteins , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , SNARE Proteins , Syntaxin 1 , Transfection , Xenopus
18.
J Neurobiol ; 53(4): 457-78, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12436413

ABSTRACT

The loss of functional response upon continuous or repeated exposure to agonist, desensitization, is an intriguing phenomenon if not as yet a well-defined physiological mechanism. However, detailed evaluation of the properties of desensitization, especially for the superfamily of ligand-gated ion channels, reveals how the nervous system could make important use of this process that goes far beyond simply curtailing excessive receptor stimulation and the prevention of excitotoxicity. Here we will review the mechanistic basis of desensitization and discuss how the subunit-dependent properties and regulation of nicotinic acetylcholine receptor (nAChR) desensitization contribute to the functional diversity of these channels. These studies provide the essential framework for understanding how the physiological regulation of desensitization could be a major determinant of synaptic efficacy by controlling, in both the short and long term, the number of functional receptors. This type of mechanism can be extended to explain how the continuous occupation of desensitized receptors during chronic nicotine exposure contributes to drug addiction, and highlights the potential significance of prolonged nAChR desensitization that would also occur as a result of extended acetylcholine lifetime during treatment of Alzheimer's disease with cholinesterase inhibitors. Thus, a clearer picture of the importance of nAChR desensitization in both normal information processing and in various diseased states is beginning to emerge.


Subject(s)
Drug Tolerance/physiology , Receptors, Nicotinic/physiology , Acetylcholine/metabolism , Animals , Humans , Ion Channels , Membrane Potentials/physiology , Nicotine/pharmacology
19.
J Neurosci ; 22(23): 10251-66, 2002 Dec 01.
Article in English | MEDLINE | ID: mdl-12451126

ABSTRACT

GABA transporter subtype 1 (GAT1) molecules were counted near GABAergic synapses, to a resolution of approximately 0.5 microm. Fusions between GAT1 and green fluorescent protein (GFP) were tested in heterologous expression systems, and a construct was selected that shows function, expression level, and trafficking similar to that of wild-type (WT) GAT1. A strain of knock-in mice was constructed that expresses this mGAT1-GFP fusion in place of the WT GAT1 gene. The pattern of fluorescence in brain slices agreed with previous immunocytochemical observations. [3H]GABA uptake, synaptic electrophysiology, and subcellular localization of the mGAT1-GFP construct were also compared with WT mice. Quantitative fluorescence microscopy was used to measure the density of mGAT1-GFP at presynaptic structures in CNS preparations from the knock-in mice. Fluorescence measurements were calibrated with transparent beads and gels that have known GFP densities. Surface biotinylation defined the fraction of transporters on the surface versus those in the nearby cytoplasm. The data show that the presynaptic boutons of GABAergic interneurons in cerebellum and hippocampus have a membrane density of 800-1300 GAT1 molecules per square micrometer, and the axons that connect boutons have a linear density of 640 GAT1 molecules per micrometer. A cerebellar basket cell bouton, a pinceau surrounding a Purkinje cell axon, and a cortical chandelier cell cartridge carry 9000, 7.8 million, and 430,000 GAT1 molecules, respectively; 61-63% of these molecules are on the surface membrane. In cultures from hippocampus, the set of fluorescent cells equals the set of GABAergic interneurons. Knock-in mice carrying GFP fusions of membrane proteins provide quantitative data required for understanding the details of synaptic transmission in living neurons.


Subject(s)
Carrier Proteins/biosynthesis , Cell Membrane/metabolism , Cytoplasm/metabolism , Membrane Proteins/biosynthesis , Membrane Transport Proteins , Organic Anion Transporters , Presynaptic Terminals/metabolism , Recombinant Fusion Proteins/biosynthesis , Animals , Biotinylation , Carrier Proteins/genetics , Cell Line , Cerebellum/cytology , Cerebellum/metabolism , DNA, Complementary/genetics , DNA, Complementary/metabolism , GABA Plasma Membrane Transport Proteins , Green Fluorescent Proteins , Hippocampus/cytology , Hippocampus/metabolism , Humans , Kidney/cytology , Kidney/metabolism , Luminescent Proteins/genetics , Membrane Proteins/genetics , Mice , Mice, Transgenic , Microscopy, Fluorescence , Neurons/cytology , Neurons/metabolism , Organ Specificity , Patch-Clamp Techniques , Recombinant Fusion Proteins/genetics , Stem Cells/cytology , Synaptic Transmission/physiology , gamma-Aminobutyric Acid/metabolism
20.
Int J Dev Neurosci ; 20(3-5): 219-24, 2002.
Article in English | MEDLINE | ID: mdl-12175857

ABSTRACT

Neurotransmitter transporters are regulated through a variety of signal transduction mechanisms which may operate in order to maintain appropriate levels of transmitter in the synaptic cleft. GABA and glycine transporters both interact with components of the neurotransmitter release, such as the SNARE protein syntaxin 1A, suggesting that protein-protein interactions are a common method for regulating members of the neurotransmitter transporter family, and thus, linking the release of transmitter to its subsequent re-uptake. In the present report, the interaction of syntaxin 1A with endogenous serotonin transporters (SERT) expressed in developing thalamocortical neurons is examined. Incubation of thalamocortical cultures with botulinum toxin C1, which specifically cleaves syntaxin 1A, decreased SERT function. Serotonin (5HT) saturation analysis showed that the effect of the toxin was to decrease maximum transport capacity with little change to the affinity of the transporter for 5HT. The 5HT uptake data were consistent with biotinylation experiments showing a decrease in the surface expression of SERT following toxin treatment. In addition, co-immunoprecipitation experiments showed that SERT and syntaxin 1A form a protein complex in these neurons. These data show that components of the transmitter release machinery interact with endogenously expressed amine transporters, and suggest a mechanism for the control of transmitter levels in disorders related to aminergic signaling.


Subject(s)
Antigens, Surface/metabolism , Carrier Proteins/metabolism , Cell Differentiation/physiology , Cerebral Cortex/metabolism , Membrane Glycoproteins/metabolism , Membrane Transport Proteins , Nerve Tissue Proteins/metabolism , Neural Pathways/metabolism , Presynaptic Terminals/metabolism , Thalamus/metabolism , Animals , Botulinum Toxins/pharmacology , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Fetus , Immunohistochemistry , Nerve Tissue Proteins/antagonists & inhibitors , Neural Pathways/cytology , Neural Pathways/embryology , Protein Transport/drug effects , Protein Transport/physiology , Rats , Rats, Sprague-Dawley , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins , Synaptic Transmission/physiology , Syntaxin 1 , Thalamus/cytology , Thalamus/embryology
SELECTION OF CITATIONS
SEARCH DETAIL