Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 292(13): 5418-5428, 2017 03 31.
Article in English | MEDLINE | ID: mdl-28213519

ABSTRACT

The GABA transporter GAT-1 mediates electrogenic transport of its substrate together with sodium and chloride. It is a member of the neurotransmitter:sodium:symporters, which are crucial for synaptic transmission. Compared with all other neurotransmitter:sodium:symporters, GAT-1 and other members of the GABA transporter subfamily all contain an extra amino acid residue at or near a conserved glycine in transmembrane segment 10. Therefore, we studied the functional impact of deletion and replacement mutants of Gly-457 and its two adjacent residues in GAT-1. The glycine replacement mutants were devoid of transport activity, but remarkably the deletion mutant was active, as were mutants obtained by deleting positions on either side of Gly-457. However, the inward rectification of GABA-induced transport currents by all three deletion mutants was diminished, and the charge-to-flux ratio was increased by more than 2.5-fold, both of which indicate substantial uncoupled transport. These observations suggest that the deletions render the transporters less tightly packed. Consistent with this interpretation, the inactive G457A mutant was partially rescued by removing the adjacent serine residue. Moreover, the activity of several gating mutants was also partially rescued upon deletion of Gly-457. Structural modeling showed that the stretch surrounding Gly-457 is likely to form a π-helix. Our data indicate that the "extra" residue in transmembrane domain 10 of the GABA transporter GAT-1 provides extra bulk, probably in the form of a π-helix, which is required for stringent gating and tight coupling of ion and substrate fluxes in the GABA transporter family.


Subject(s)
GABA Plasma Membrane Transport Proteins/chemistry , Glycine/genetics , Ion Transport , Mutagenesis, Site-Directed , Amino Acids , Conserved Sequence/genetics , GABA Plasma Membrane Transport Proteins/genetics , GABA Plasma Membrane Transport Proteins/metabolism , HeLa Cells , Humans , Protein Conformation , Protein Domains , Structure-Activity Relationship
2.
J Biol Chem ; 289(41): 28172-8, 2014 Oct 10.
Article in English | MEDLINE | ID: mdl-25143384

ABSTRACT

GAT-1 is a sodium- and chloride-coupled GABA transporter and a member of the neurotransmitter:sodium:symporters, which are crucial for synaptic transmission. The structure of bacterial homologue LeuT shows a thin extracellular gate consisting of a charge and an aromatic pair. Here we addressed the question of whether mutation of the aromatic and charge pair residues of GAT-1 has similar consequences. In contrast to charge pair mutants, significant radioactive GABA transport was retained by mutants of the aromatic pair residue Phe-294. Moreover, the magnitude of maximal transport currents induced by GABA by these mutants was comparable with those by wild type GAT-1. However, the apparent affinity of the nonconserved mutants for GABA was reduced up to 20-fold relative to wild type. The voltage dependence of the sodium-dependent transient currents of the Phe-294 mutants was similar to that of the wild type. On the other hand, the conserved charge pair mutant D451E exhibited a right-shifted voltage dependence, indicating an increased apparent affinity for sodium. In further contrast to D451E, whereas the extracellular aqueous accessibility of an endogenous cysteine residue to a membrane-impermeant sulfhydryl reagent was increased relative to wild type, this was not the case for the aromatic pair mutants. Our data indicate that, in contrast to the charge pair, the aromatic pair is not essential for gating. Instead they are compatible with the idea that they serve to diminish dissociation of the substrate from the binding pocket.


Subject(s)
Aspartic Acid/chemistry , GABA Plasma Membrane Transport Proteins/chemistry , Glutamic Acid/chemistry , Mutation , Phenylalanine/chemistry , gamma-Aminobutyric Acid/chemistry , Animals , Aspartic Acid/metabolism , Biological Transport , GABA Plasma Membrane Transport Proteins/genetics , GABA Plasma Membrane Transport Proteins/metabolism , Gene Expression , Glutamic Acid/metabolism , HeLa Cells , Humans , Kinetics , Membrane Potentials , Models, Molecular , Oocytes/cytology , Oocytes/physiology , Patch-Clamp Techniques , Phenylalanine/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sodium/chemistry , Sodium/metabolism , Structure-Activity Relationship , Xenopus laevis , gamma-Aminobutyric Acid/metabolism
3.
J Biol Chem ; 288(7): 4549-56, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23288838

ABSTRACT

The GABA transporter GAT-1 belongs to the neurotransmitter:sodium:symporters which are crucial for synaptic transmission. GAT-1 mediates electrogenic transport of GABA together with sodium and chloride. Structure-function studies indicate that the bacterial homologue LeuT, which possess extra- and intracellular thin gates, is an excellent model for this class of neurotransmitter transporters. We recently showed that a conserved aspartate residue of GAT-1, Asp-451, whose LeuT equivalent participates in its thin extracellular gate, is functionally irreplaceable in GAT-1. Only the D451E mutant exhibited residual transport activity but with an elevated apparent sodium affinity as a consequence of an increased proportion of outward-facing transporters. Because during transport the opening and closing of external and internal gates should be tightly coupled, we have addressed the question of whether mutations of the intracellular thin gate residues Arg-44 and Asp-410 can compensate for the effects of their extracellular counterparts. Mutation of Asp-410 to glutamate resulted in impaired transport activity and a reduced apparent affinity for sodium. However, the transport activity of the double mutant D410E/D451E was increased by approximately 10-fold of that of each of the single mutants. Similar compensatory effects were also seen when other combinations of intra- and extracellular thin gate mutants were analyzed. Moreover, the introduction of D410E into the D451E background resulted in lower apparent sodium affinity than that of D451E alone. Our results indicate that a functional interaction of the external and internal gates of GAT-1 is essential for transport.


Subject(s)
GABA Plasma Membrane Transport Proteins/metabolism , Mutation , Animals , Biological Transport , Biotinylation , Dose-Response Relationship, Drug , Ethylmaleimide/chemistry , HeLa Cells , Humans , Kinetics , Models, Molecular , Molecular Conformation , Neurotransmitter Agents/metabolism , Oocytes/metabolism , Protein Transport , Sodium/chemistry , Structure-Activity Relationship , Xenopus laevis
4.
J Biol Chem ; 287(10): 7159-68, 2012 Mar 02.
Article in English | MEDLINE | ID: mdl-22235131

ABSTRACT

GAT-1 mediates transport of GABA together with sodium and chloride in an electrogenic process enabling efficient GABAergic transmission. Biochemical and modeling studies based on the structure of the bacterial homologue LeuT are consistent with a mechanism whereby the binding pocket is alternately accessible to either side of the membrane and which predicts that the extracellular part of transmembrane domain 10 (TM10) exhibits aqueous accessibility in the outward-facing conformation only. In this study we have engineered cysteine residues in the extracellular half of TM10 of GAT-1 and probed their state-dependent accessibility to sulfhydryl reagents. In three out of four of the accessible cysteine mutants, the inhibition of transport by a membrane impermeant sulfhydryl reagent was diminished under conditions expected to increase the proportion of inward-facing transporters, such as the presence of GABA together with the cotransported ions. A conserved TM10 aspartate residue, whose LeuT counterpart participates in a "thin" extracellular gate, was found to be essential for transport and only the D451E mutant exhibited residual transport activity. D451E exhibited robust sodium-dependent transient currents with a voltage-dependence indicative of an increased apparent affinity for sodium. Moreover the accessibility of an endogenous cysteine to a membrane impermeant sulfhydryl reagent was enhanced by the D451E mutation, suggesting that sodium binding promotes an outward-facing conformation of the transporter. Our results support the idea that TM10 of GAT-1 lines an accessibility pathway from the extracellular space into the binding pocket and plays a role in the opening and closing of the extracellular transporter gate.


Subject(s)
GABA Plasma Membrane Transport Proteins/metabolism , Ion Channel Gating/physiology , Neurotransmitter Agents/metabolism , gamma-Aminobutyric Acid/metabolism , Amino Acid Substitution , Animals , GABA Plasma Membrane Transport Proteins/chemistry , GABA Plasma Membrane Transport Proteins/genetics , HeLa Cells , Humans , Ion Transport/physiology , Mutation, Missense , Neurotransmitter Agents/genetics , Protein Structure, Secondary , Protein Structure, Tertiary , Sodium/metabolism , Xenopus laevis , gamma-Aminobutyric Acid/genetics
5.
J Biol Chem ; 286(4): 2826-33, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21098479

ABSTRACT

Neurotransmitter:sodium symporters are crucial for efficient synaptic transmission. The transporter GAT-1 mediates electrogenic cotransport of GABA, sodium, and chloride. The presence of chloride enables the transporter to couple the transport of the neurotransmitter to multiple sodium ions, thereby enabling its accumulation against steep concentration gradients. Here we study the functional impact of mutations of the putative chloride-binding residues on transport by GAT-1, with the emphasis on a conserved glutamine residue. In contrast to another putative chloride coordinating residue, Ser-331, where mutation to glutamate led to chloride-independent GABA transport, the Q291E mutant was devoid of any transport activity, despite substantial expression at the plasma membrane. Low but significant transport activity was observed with substitution mutants with small side chains such as Q291S/A/G. Remarkably, when these mutations were combined with the S331E mutation, transport was increased significantly, even though the activity of the S331E single mutant was only ∼25% of that of wild type GAT-1. Transport by these double mutants was largely chloride-independent. Like mutants of other putative chloride coordinating residues, the apparent affinity of the active Gln-291 single mutants for chloride was markedly reduced along with a change their anion selectivity. In addition to the interaction of the transporter with chloride, Gln-291 is also required at an additional step during transport. Electrophysiological analysis of the Q291N and Q291S mutants, expressed in Xenopus laevis oocytes, is consistent with the idea that this additional step is associated with the gating of the transporter.


Subject(s)
GABA Plasma Membrane Transport Proteins/metabolism , Ion Channel Gating/physiology , Amino Acid Substitution , Animals , GABA Plasma Membrane Transport Proteins/genetics , Glutamine/genetics , Glutamine/metabolism , HeLa Cells , Humans , Ion Transport/physiology , Mutation, Missense , Neurotransmitter Agents/genetics , Neurotransmitter Agents/metabolism , Oocytes , Sodium/metabolism , Xenopus laevis
6.
J Biol Chem ; 284(15): 9727-32, 2009 Apr 10.
Article in English | MEDLINE | ID: mdl-19201752

ABSTRACT

GAT-1 is a sodium- and chloride-coupled gamma-aminobutyric acid (GABA) transporter, which fulfills an essential role in the synaptic transmission by this neurotransmitter. Cysteine-399 is the major site of inhibition of GAT-1 by membrane-permeant sulfhydryl reagents. This cysteine residue was previously thought to reside on a cytoplasmic loop connecting transmembrane domains (TMs) 8 and 9. However, the crystal structure of LeuT, a bacterial homologue of the mammalian neurotransmitter:sodium symporters, revealed that the residue corresponding to Cys-399 is in fact located in the middle of TM 8. This residue is located to the cytoplasmic side of Asp-395 and Ser-396, whose side chains are thought to ligand one of the two cotransported sodium ions. To determine how the sulfhydryl reagents approach cysteine-399, a cysteine scan of all 35 residues of TM 8 was performed. Sulfhydryl reagents inhibited transport when a cysteine residue was present at either of the positions 399, 402, 406, and 410. SKF-89976A and other non-transportable analogues, which are expected to lock the transporter in a conformation facing the extracellular medium, protected against the sulfhydryl modification at positions 399, 402, and 406. Such a protection was not seen by GABA itself, which actually modestly potentiated the modification at positions 399 and 402. Our results point to an alpha-helical stripe on TM8 lining an aqueous access pathway from the cytoplasm into the binding pocket, which gets occluded in the conformation of the transporter where the binding pocket is exposed to the extracellular medium.


Subject(s)
GABA Plasma Membrane Transport Proteins/chemistry , GABA Plasma Membrane Transport Proteins/metabolism , Aspartic Acid/chemistry , Cysteine/chemistry , Cytoplasm/metabolism , HeLa Cells , Humans , Models, Biological , Mutation , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Protein Transport , Serine/chemistry , gamma-Aminobutyric Acid/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...