Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Nat Struct Mol Biol ; 30(7): 1023-1032, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37400654

RESUMO

The inhibitory neurotransmitter γ-aminobutyric acid (GABA) is cleared from the synaptic cleft by the sodium- and chloride-coupled GABA transporter GAT1. Inhibition of GAT1 prolongs the GABAergic signaling at the synapse and is a strategy to treat certain forms of epilepsy. In this study, we present the cryo-electron microscopy structure of Rattus norvegicus GABA transporter 1 (rGAT1) at a resolution of 3.1 Å. The structure elucidation was facilitated by epitope transfer of a fragment-antigen binding (Fab) interaction site from the Drosophila dopamine transporter (dDAT) to rGAT1. The structure reveals rGAT1 in a cytosol-facing conformation, with a linear density in the primary binding site that accommodates a molecule of GABA, a displaced ion density proximal to Na site 1 and a bound chloride ion. A unique insertion in TM10 aids the formation of a compact, closed extracellular gate. Besides yielding mechanistic insights into ion and substrate recognition, our study will enable the rational design of specific antiepileptics.


Assuntos
Cloretos , Ácido gama-Aminobutírico , Ratos , Animais , Proteínas da Membrana Plasmática de Transporte de GABA/química , Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Microscopia Crioeletrônica , Cloretos/metabolismo , Cloretos/farmacologia , Ácido gama-Aminobutírico/metabolismo , Sítios de Ligação
2.
Trends Pharmacol Sci ; 44(1): 4-6, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-35985875

RESUMO

The γ-aminobutyric acid transporter 1 (GAT1) is a transporter which clears the inhibitory neurotransmitter γ-aminobutyric acid (GABA) from the synaptic cleft. The paper by Motiwala et al. documents a structure of GAT1 in complex with the antiepileptic drug tiagabine. This study will enable structure-based docking of large chemical libraries for the discovery of novel antiepileptics.


Assuntos
Anticonvulsivantes , Ácido gama-Aminobutírico , Humanos , Proteínas da Membrana Plasmática de Transporte de GABA/química , Anticonvulsivantes/farmacologia , Tiagabina
3.
Neuropharmacology ; 161: 107534, 2019 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-30790582

RESUMO

GAT1 is a member of the neurotransmitter:sodium: symporter family and mediates transport of GABA together with sodium and chloride in an electrogenic process enabling efficient synaptic transmission. Biochemical and modelling studies based on the structure of the bacterial homologue LeuT are consistent with a transport mechanism whereby the binding pocket is alternately accessible to either side of the membrane. This is achieved by the sequential opening and closing of extracellular and intracellular gates. The amino acid residues participating in the formation of these gates are highly conserved within the neurotransmitter:sodium: symporter family. Net flux requires that the gating mechanism is operative regardless if the binding pocket is loaded with substrate or empty. On the other hand, exchange of labelled for non-labelled substrate across the membrane only requires gating in the presence of substrate. To address the question if the gating requirements of the substrate-bound and empty transporters are similar or different, we analyzed the impact of mutation of intra- and extra-cellular gate residues on net GABA influx and on exchange by liposomes inlaid with the mutant transporters. Whereas net flux by all four internal gate mutants tested was severely abrogated, each exhibited significant levels of exchange. In contrast, two external gate mutants were impaired in both processes. Our results indicate that perturbation of the internal gate of GAT1 selectively impairs the gating mechanism of the empty transporter. This article is part of the issue entitled 'Special Issue on Neurotransmitter Transporters'.


Assuntos
Proteínas da Membrana Plasmática de Transporte de GABA/genética , Mutação/fisiologia , Aminoácidos/metabolismo , Animais , Sítios de Ligação , Biotinilação , Espaço Extracelular/genética , Espaço Extracelular/metabolismo , Proteínas da Membrana Plasmática de Transporte de GABA/química , Células HeLa , Humanos , Espaço Intracelular/genética , Espaço Intracelular/metabolismo , Ativação do Canal Iônico/genética , Cinética , Lipossomos/metabolismo , Modelos Moleculares , Mutagênese Sítio-Dirigida , Ratos , Membranas Sinápticas/metabolismo , Ácido gama-Aminobutírico/metabolismo
4.
Epilepsia ; 59(9): e135-e141, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30132828

RESUMO

Previous reports have identified SLC6A1 variants in patients with generalized epilepsies, such as myoclonic-atonic epilepsy and childhood absence epilepsy. However, to date, none of the identified SLC6A1 variants has been functionally tested for an effect on GAT-1 transporter activity. The purpose of this study was to determine the incidence of SLC6A1 variants in 460 unselected epilepsy patients and to evaluate the impact of the identified variants on γ-aminobutyric acid (GABA)transport. Targeted resequencing was used to screen 460 unselected epilepsy patients for variants in SLC6A1. Five missense variants, one in-frame deletion, one nonsense variant, and one intronic splice-site variant were identified, representing a 1.7% diagnostic yield. Using a [3 H]-GABA transport assay, the seven identified exonic variants were found to reduce GABA transport activity. A minigene splicing assay revealed that the splice-site variant disrupted canonical splicing of exon 9 in the mRNA transcript, leading to premature protein truncation. These findings demonstrate that SLC6A1 is an important contributor to childhood epilepsy and that reduced GAT-1 function is a common consequence of epilepsy-causing SLC6A1 variants.


Assuntos
Epilepsia/genética , Epilepsia/metabolismo , Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Regulação da Expressão Gênica/genética , Mutação/genética , Estudos de Coortes , Análise Mutacional de DNA , Feminino , Proteínas da Membrana Plasmática de Transporte de GABA/genética , Predisposição Genética para Doença/genética , Células HEK293 , Células HeLa , Humanos , Masculino , RNA Mensageiro/metabolismo , Transfecção , Trítio/farmacocinética , Ácido gama-Aminobutírico/metabolismo
5.
J Biol Chem ; 293(37): 14200-14209, 2018 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-30026234

RESUMO

In the brain, glutamate transporters terminate excitatory neurotransmission by removing this neurotransmitter from the synapse via cotransport with three sodium ions into the surrounding cells. Structural studies have identified the binding sites of the three sodium ions in glutamate transporters. The residue side-chains directly interact with the sodium ions at the Na1 and Na3 sites and are fully conserved from archaeal to eukaryotic glutamate transporters. The Na2 site is formed by three main-chain oxygens on the extracellular reentrant hairpin loop HP2 and one on transmembrane helix 7. A glycine residue on HP2 is located closely to the three main-chain oxygens in all glutamate transporters, except for the astroglial transporter GLT-1, which has a serine residue at that position. Unlike for WT GLT-1, substitution of the serine residue to glycine enables sustained glutamate transport also when sodium is replaced by lithium. Here, using functional and simulation studies, we studied the role of this serine/glycine switch on cation selectivity of substrate transport. Our results indicate that the side-chain oxygen of the serine residues can form a hydrogen bond with a main-chain oxygen on transmembrane helix 7. This leads to an expansion of the Na2 site such that water can participate in sodium coordination at Na2. Furthermore, we found other molecular determinants of cation selectivity on the nearby HP1 loop. We conclude that subtle changes in the composition of the two reentrant hairpin loops determine the cation specificity of acidic amino acid transport by glutamate transporters.


Assuntos
Transportador 2 de Aminoácido Excitatório/metabolismo , Sódio/metabolismo , Sítios de Ligação , Cátions/metabolismo , Transportador 2 de Aminoácido Excitatório/química , Transportador 2 de Aminoácido Excitatório/genética , Transportador 3 de Aminoácido Excitatório/química , Transportador 3 de Aminoácido Excitatório/genética , Transportador 3 de Aminoácido Excitatório/metabolismo , Glicina/metabolismo , Células HeLa , Humanos , Ligação de Hidrogênio , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , Serina/metabolismo
6.
J Biol Chem ; 292(13): 5418-5428, 2017 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-28213519

RESUMO

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.


Assuntos
Proteínas da Membrana Plasmática de Transporte de GABA/química , Glicina/genética , Transporte de Íons , Mutagênese Sítio-Dirigida , Aminoácidos , Sequência Conservada/genética , Proteínas da Membrana Plasmática de Transporte de GABA/genética , Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Células HeLa , Humanos , Conformação Proteica , Domínios Proteicos , Relação Estrutura-Atividade
7.
J Biol Chem ; 290(48): 28988-96, 2015 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-26475859

RESUMO

Crystal structures of the archaeal homologue GltPh have provided important insights into the molecular mechanism of transport of the excitatory neurotransmitter glutamate. Whereas mammalian glutamate transporters can translocate both glutamate and aspartate, GltPh is only one capable of aspartate transport. Most of the amino acid residues that surround the aspartate substrate in the binding pocket of GltPh are highly conserved. However, in the brain transporters, Thr-352 and Met-362 of the reentrant hairpin loop 2 are replaced by the smaller Ala and Thr, respectively. Therefore, we have studied the effects of T352A and M362T on binding and transport of aspartate and glutamate by GltPh. Substrate-dependent intrinsic fluorescence changes were monitored in transporter constructs containing the L130W mutation. GltPh-L130W/T352A exhibited an ~15-fold higher apparent affinity for l-glutamate than the wild type transporter, and the M362T mutation resulted in an increased affinity of ~40-fold. An even larger increase of the apparent affinity for l-glutamate, around 130-fold higher than that of wild type, was observed with the T352A/M362T double mutant. Radioactive uptake experiments show that GltPh-T352A not only transports aspartate but also l-glutamate. Remarkably, GltPh-M362T exhibited l-aspartate but not l-glutamate transport. The double mutant retained the ability to transport l-glutamate, but its kinetic parameters were very similar to those of GltPh-T352A alone. The differential impact of mutation on binding and transport of glutamate suggests that hairpin loop 2 not only plays a role in the selection of the substrate but also in its translocation.


Assuntos
Ácido Aspártico/química , Proteínas de Transporte de Glutamato da Membrana Plasmática/química , Ácido Glutâmico/química , Mutação de Sentido Incorreto , Proteínas do Tecido Nervoso/química , Substituição de Aminoácidos , Ácido Aspártico/genética , Ácido Aspártico/metabolismo , Encéfalo/metabolismo , Proteínas de Transporte de Glutamato da Membrana Plasmática/genética , Proteínas de Transporte de Glutamato da Membrana Plasmática/metabolismo , Ácido Glutâmico/genética , Ácido Glutâmico/metabolismo , Humanos , Transporte de Íons/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Estrutura Secundária de Proteína , Especificidade por Substrato/genética
8.
J Biol Chem ; 289(49): 34258-66, 2014 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-25339171

RESUMO

The sodium- and chloride-coupled GABA transporter GAT-1 is a member of the neurotransmitter:sodium:symporters, which are crucial for synaptic transmission. Structural work on the bacterial homologue LeuT suggests that extracellular loop 4 closes the extracellular solvent pathway when the transporter becomes inward-facing. To test whether this model can be extrapolated to GAT-1, cysteine residues were introduced at positions 359 and 448 of extracellular loop 4 and transmembrane helix 10, respectively. Treatment of HeLa cells, expressing the double cysteine mutant S359C/K448C with the oxidizing reagent copper(II)(1,10-phenantroline)3, resulted in a significant inhibition of [(3)H]GABA transport. However, transport by the single cysteine mutant S359C was also inhibited by the oxidant, whereas its activity was almost 4-fold stimulated by dithiothreitol. Both effects were attenuated when the conserved cysteine residues, Cys-164 and/or Cys-173, were replaced by serine. These cysteines are located in extracellular loop 2, the role of which in the structure and function of the eukaryotic neurotransmitter:sodium:symporters remains unknown. The inhibition of transport of S359C by the oxidant was markedly reduced under conditions expected to increase the proportion of inward-facing transporters, whereas the reactivity of the mutants to a membrane-impermeant sulfhydryl reagent was not conformationally sensitive. Our data suggest that extracellular loops 2 and 4 come into close proximity to each other in the outward-facing conformation of GAT-1.


Assuntos
Cisteína/química , Proteínas da Membrana Plasmática de Transporte de GABA/química , Oócitos/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Colina/metabolismo , Colina/farmacologia , Cisteína/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Feminino , Proteínas da Membrana Plasmática de Transporte de GABA/genética , Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Expressão Gênica , Gluconatos/metabolismo , Gluconatos/farmacologia , Células HeLa , Humanos , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Modelos Moleculares , Mutação , Oócitos/citologia , Oócitos/efeitos dos fármacos , Técnicas de Patch-Clamp , Fenantrolinas/química , Domínios e Motivos de Interação entre Proteínas , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Xenopus laevis , Ácido gama-Aminobutírico/farmacologia
9.
J Biol Chem ; 289(41): 28172-8, 2014 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-25143384

RESUMO

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.


Assuntos
Ácido Aspártico/química , Proteínas da Membrana Plasmática de Transporte de GABA/química , Ácido Glutâmico/química , Mutação , Fenilalanina/química , Ácido gama-Aminobutírico/química , Animais , Ácido Aspártico/metabolismo , Transporte Biológico , Proteínas da Membrana Plasmática de Transporte de GABA/genética , Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Expressão Gênica , Ácido Glutâmico/metabolismo , Células HeLa , Humanos , Cinética , Potenciais da Membrana , Modelos Moleculares , Oócitos/citologia , Oócitos/fisiologia , Técnicas de Patch-Clamp , Fenilalanina/metabolismo , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sódio/química , Sódio/metabolismo , Relação Estrutura-Atividade , Xenopus laevis , Ácido gama-Aminobutírico/metabolismo
10.
J Biol Chem ; 289(16): 11175-11182, 2014 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-24584931

RESUMO

Excitatory amino acid transporters remove synaptically released glutamate and maintain its concentrations below neurotoxic levels. EAATs also mediate a thermodynamically uncoupled substrate-gated anion conductance that may modulate cell excitability. A structure of an archeal homologue, which reflects an early intermediate on the proposed substrate translocation path, has been suggested to be similar to an anion conducting conformation. To probe this idea by functional studies, we have introduced two cysteine residues in the neuronal glutamate transporter EAAC1 at positions predicted to be close enough to form a disulfide bond only in outward-facing and early intermediate conformations of the homologue. Upon treatment of Xenopus laevis oocytes expressing the W441C/K269C double mutant with dithiothreitol, radioactive transport was stimulated >2-fold but potently inhibited by low micromolar concentrations of the oxidizing reagent copper(II)(1,10-phenanthroline)3. The substrate-induced currents by the untreated double mutant, reversed at approximately -20 mV, close to the reversal potential of chloride, but treatment with dithiothreitol resulted in transport currents with the same voltage dependence as the wild type. It appears therefore that in the oocyte expression system the introduced cysteine residues in many of the mutant transporters are already cross-linked and are only capable of mediating the substrate-gated anion conductance. Reduction of the disulfide bond now allows these transporters to execute the full transport cycle. Our functional data support the idea that the anion conducting conformation of the neuronal glutamate transporter is associated with an early step of the transport cycle.


Assuntos
Transportador 3 de Aminoácido Excitatório/metabolismo , Ativação do Canal Iônico/fisiologia , Multimerização Proteica/fisiologia , Substituição de Aminoácidos , Animais , Ditiotreitol/farmacologia , Transportador 3 de Aminoácido Excitatório/genética , Células HeLa , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Transporte de Íons/efeitos dos fármacos , Transporte de Íons/fisiologia , Mutação de Sentido Incorreto , Compostos Organometálicos/farmacologia , Multimerização Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína , Coelhos , Xenopus laevis
11.
Nat Struct Mol Biol ; 20(10): 1142-4, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24096399

RESUMO

Sodium-coupled glutamate transporters regulate excitatory signaling in the brain. A new crystal structure shows how the substrate induces changes in the binding pocket of an archaeal transporter homolog, providing new insights into the mechanism of transport.


Assuntos
Ácido Aspártico/metabolismo , Proteínas de Transporte/metabolismo
12.
J Biol Chem ; 288(7): 4549-56, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23288838

RESUMO

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.


Assuntos
Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Mutação , Animais , Transporte Biológico , Biotinilação , Relação Dose-Resposta a Droga , Etilmaleimida/química , Células HeLa , Humanos , Cinética , Modelos Moleculares , Conformação Molecular , Neurotransmissores/metabolismo , Oócitos/metabolismo , Transporte Proteico , Sódio/química , Relação Estrutura-Atividade , Xenopus laevis
13.
J Biol Chem ; 288(2): 964-73, 2013 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-23188832

RESUMO

Glutamate transporters in the brain remove the neurotransmitter from the synapse by cotransport with three sodium ions into the surrounding cells. Recent structural work on an archaeal homolog suggests that, during substrate translocation, the transport domain, including the peripheral transmembrane helix 3 (TM3), moves relative to the trimerization domain in an elevator-like process. Moreover, two TM3 residues have been proposed to form part of a transient Na3' site, and another, Tyr-124, appears close to both Na3' and Na1. To obtain independent evidence for the role of TM3 in glutamate transport, each of its 31 amino acid residues from the glial GLT-1 transporter was individually mutated to cysteine. Except for six mutants, substantial transport activity was detected. Aqueous accessibility of the introduced cysteines was probed with membrane-permeant and membrane-impermeant sulfhydryl reagents under a variety of conditions. Transport of six single cysteine mutants, all located on the intracellular side of TM3, was affected by membrane-permeant sulfhydryl reagents. However, only at two positions could ligands modulate the reactivity. A120C reactivity was diminished under conditions expected to favor the outward-facing conformation of the transporter. Sulfhydryl modification of Y124C by 2-aminoethyl methanethiosulfonate, but not by N-ethylmaleimide, was fully protected in the presence of sodium. Our data are consistent with the idea that TM3 moves during transport. Moreover, computational modeling indicated that electrostatic repulsion between the positive charge introduced at position 124 and the sodium ions bound at Na3' and Na1 underlies the protection by sodium.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/metabolismo , Encéfalo/metabolismo , Cisteína/genética , Proteínas de Membrana/metabolismo , Mutagênese , Sistema X-AG de Transporte de Aminoácidos/química , Sistema X-AG de Transporte de Aminoácidos/genética , Proteínas de Membrana/química , Proteínas de Membrana/genética , Modelos Moleculares , Conformação Proteica , Reagentes de Sulfidrila/química
14.
J Biol Chem ; 287(21): 17198-17205, 2012 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-22493292

RESUMO

Transporters of the major excitatory neurotransmitter glutamate play a crucial role in glutamatergic neurotransmission by removing their substrate from the synaptic cleft. The transport mechanism involves co-transport of glutamic acid with three Na(+) ions followed by countertransport of one K(+) ion. Structural work on the archeal homologue Glt(Ph) indicates a role of a conserved asparagine in substrate binding. According to a recent proposal, this residue may also participate in a novel Na(+) binding site. In this study, we characterize mutants of this residue from the neuronal transporter EAAC1, Asn-451. None of the mutants, except for N451S, were able to exhibit transport. However, the K(m) of this mutant for l-aspartate was increased ∼30-fold. Remarkably, the increase for d-aspartate and l-glutamate was 250- and 400-fold, respectively. Moreover, the cation specificity of N451S was altered because sodium but not lithium could support transport. A similar change in cation specificity was observed with a mutant of a conserved threonine residue, T370S, also implicated to participate in the novel Na(+) site together with the bound substrate. In further contrast to the wild type transporter, only l-aspartate was able to activate the uncoupled anion conductance by N451S, but with an almost 1000-fold reduction in apparent affinity. Our results not only provide experimental support for the Na(+) site but also suggest a distinct orientation of the substrate in the binding pocket during the activation of the anion conductance.


Assuntos
Asparagina/química , Asparagina/metabolismo , Transportador 3 de Aminoácido Excitatório/química , Transportador 3 de Aminoácido Excitatório/metabolismo , Substituição de Aminoácidos , Animais , Asparagina/genética , Sítios de Ligação , Transportador 3 de Aminoácido Excitatório/genética , Transporte de Íons/fisiologia , Mutação de Sentido Incorreto , Coelhos , Especificidade por Substrato , Xenopus laevis
15.
J Biol Chem ; 287(10): 7159-68, 2012 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-22235131

RESUMO

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.


Assuntos
Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Ativação do Canal Iônico/fisiologia , Neurotransmissores/metabolismo , Ácido gama-Aminobutírico/metabolismo , Substituição de Aminoácidos , Animais , Proteínas da Membrana Plasmática de Transporte de GABA/química , Proteínas da Membrana Plasmática de Transporte de GABA/genética , Células HeLa , Humanos , Transporte de Íons/fisiologia , Mutação de Sentido Incorreto , Neurotransmissores/genética , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Sódio/metabolismo , Xenopus laevis , Ácido gama-Aminobutírico/genética
16.
J Biol Chem ; 286(48): 41381-41390, 2011 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-21984827

RESUMO

In the brain, transporters of the major excitatory neurotransmitter glutamate remove their substrate from the synaptic cleft to allow optimal glutamatergic neurotransmission. Their transport cycle consists of two sequential translocation steps, namely cotransport of glutamic acid with three Na(+) ions, followed by countertransport of K(+). Recent studies, based on several crystal structures of the archeal homologue Glt(Ph), indicate that glutamate translocation occurs by an elevator-like mechanism. The resolution of these structures was not sufficiently high to unambiguously identify the sites of Na(+) binding, but functional and computational studies suggest some candidate sites. In the Glt(Ph) structure, a conserved aspartate residue (Asp-390) is located adjacent to a conserved tyrosine residue, previously shown to be a molecular determinant of ion selectivity in the brain glutamate transporter GLT-1. In this study, we characterize mutants of Asp-440 of the neuronal transporter EAAC1, which is the counterpart of Asp-390 of Glt(Ph). Except for substitution by glutamate, this residue is functionally irreplaceable. Using biochemical and electrophysiological approaches, we conclude that although D440E is intrinsically capable of net flux, this mutant behaves as an exchanger under physiological conditions, due to increased and decreased apparent affinities for Na(+) and K(+), respectively. Our present and previous data are compatible with the idea that the conserved tyrosine and aspartate residues, located at the external end of the binding pocket, may serve as a transient or stable cation binding site in the glutamate transporters.


Assuntos
Ácido Aspártico/metabolismo , Encéfalo/metabolismo , Transportador 3 de Aminoácido Excitatório/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Substituição de Aminoácidos , Animais , Ácido Aspártico/genética , Sítios de Ligação , Transportador 3 de Aminoácido Excitatório/genética , Células HeLa , Humanos , Mutação de Sentido Incorreto , Proteínas do Tecido Nervoso/genética , Coelhos , Ratos , Homologia Estrutural de Proteína , Xenopus laevis
17.
J Biol Chem ; 286(4): 2826-33, 2011 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-21098479

RESUMO

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.


Assuntos
Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Ativação do Canal Iônico/fisiologia , Substituição de Aminoácidos , Animais , Proteínas da Membrana Plasmática de Transporte de GABA/genética , Glutamina/genética , Glutamina/metabolismo , Células HeLa , Humanos , Transporte de Íons/fisiologia , Mutação de Sentido Incorreto , Neurotransmissores/genética , Neurotransmissores/metabolismo , Oócitos , Sódio/metabolismo , Xenopus laevis
18.
Protein Expr Purif ; 73(2): 152-60, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20566324

RESUMO

Neurotransmitter transporters play essential roles in the process of neurotransmission. Vesicular neurotransmitter transporters mediate storage inside secretory vesicles in a process that involves the exchange of lumenal H(+) for cytoplasmic transmitter. Retrieval of the neurotransmitter from the synaptic cleft catalyzed by sodium-coupled transporters is critical for the termination of the synaptic actions of the released neurotransmitter. Our current understanding of the mechanism of these transporters is based on functional and biochemical characterization but is lacking high-resolution structural information. Very few structures of membrane transport systems from mammalian origin have been solved to atomic resolution, mainly because of the difficulty in obtaining large amounts of purified protein. Development of high yield heterologous expression systems suitable for mammalian neurotransmitter transporters is essential to enable the production of purified protein for structural studies. Such a system makes possible also the production of mutants that can be used in biochemical and biophysical studies. We describe here a screen for the expression of the vesicular monoamine transporter 2 (VMAT2) in cell-free and baculovirus expression systems and discuss the expression of VMAT2 in other systems as well (bacterial, yeast and mammalian cell lines). After screening and optimization, we achieved high yield (2-2.5 mg/l) expression of functional VMAT2 in insect cells. The system was also used for the expression of three additional plasma membrane neurotransmitter transporters. All were functional and expressed to high levels. Our results demonstrate the advantages of the baculovirus expression system for the expression of mammalian neurotransmitter transporters in a functional state.


Assuntos
Proteínas de Transporte de Neurotransmissores/metabolismo , Animais , Baculoviridae/genética , Transporte Biológico , Sistema Livre de Células , Células Cultivadas , Técnica Direta de Fluorescência para Anticorpo , Vetores Genéticos , Imuno-Histoquímica , Proteínas de Membrana Transportadoras/metabolismo , Neurotransmissores/metabolismo , Proteínas de Transporte de Neurotransmissores/química , Spodoptera/citologia , Spodoptera/metabolismo , Transmissão Sináptica/fisiologia , Proteínas Vesiculares de Transporte de Monoamina/isolamento & purificação , Proteínas Vesiculares de Transporte de Monoamina/metabolismo
19.
J Biol Chem ; 285(23): 17725-33, 2010 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-20378543

RESUMO

The glutamate transporter excitatory amino acid carrier 1 (EAAC1) catalyzes the co-transport of three Na(+) ions, one H(+) ion, and one glutamate molecule into the cell, in exchange for one K(+) ion. Na(+) binding to the glutamate-free form of the transporter generates a high affinity binding site for glutamate and is thus required for transport. Moreover, sodium binding to the transporters induces a basal anion conductance, which is further activated by glutamate. Here, we used the [Na(+)] dependence of this conductance as a read-out of Na(+) binding to the substrate-free transporter to study the impact of a highly conserved amino acid residue, Thr(101), in transmembrane domain 3. The apparent affinity of substrate-free EAAC1 for Na(+) was dramatically decreased by the T101A but not by the T101S mutation. Interestingly, in further contrast to EAAC1(WT), in the T101A mutant this [Na(+)] dependence was biphasic. This behavior can be explained by assuming that the binding of two Na(+) ions prior to glutamate binding is required to generate a high affinity substrate binding site. In contrast to the dramatic effect of the T101A mutation on Na(+) binding, other properties of the transporter, such as its ability to transport glutamate, were impaired but not eliminated. Our results are consistent with the existence of a cation binding site deeply buried in the membrane and involving interactions with the side chain oxygens of Thr(101) and Asp(367). A theoretical valence screening approach confirms that the predicted site of cation interaction has the potential to be a novel, so far undetected sodium binding site.


Assuntos
Cátions/química , Transportador 3 de Aminoácido Excitatório/química , Treonina/química , Sequência de Aminoácidos , Animais , Sítios de Ligação , Eletrofisiologia/métodos , Transportador 3 de Aminoácido Excitatório/metabolismo , Células HeLa , Humanos , Dados de Sequência Molecular , Mutação , Ligação Proteica , Transporte Proteico , Ratos , Retina/metabolismo , Homologia de Sequência de Aminoácidos
20.
J Biol Chem ; 285(28): 21241-8, 2010 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-20424168

RESUMO

Glutamate transporters located in the brain maintain low synaptic concentrations of the neurotransmitter by coupling its flux to that of sodium and other cations. In the binding pocket of the archeal homologue Glt(Ph), a conserved methionine residue has been implicated in the binding of the benzyl moiety of the nontransportable substrate analogue threo-beta-benzyloxyaspartate. To determine whether the corresponding methionine residue of the neuronal glutamate transporter EAAC1, Met-367, fulfills a similar role, M367L, M367C, and M367S mutants were expressed in HeLa cells and Xenopus laevis oocytes to monitor radioactive transport and transport currents, respectively. The apparent affinity of the Met-367 mutants for D-aspartate and L-glutamate, but not for L-aspartate, was 10-20-fold reduced as compared with wild type. Unlike wild type, the magnitude of I(max) was different for each of the three substrates. D-glutamate, which is also a transportable substrate of EAAC1, did not elicit any detectable response with M367C and M367S but acted as a nontransportable substrate analogue in M367L. In the mutants, substrates inhibited the anion conductance as opposed to the stimulation observed with wild type. Remarkably, the apparent affinity of the blocker D,L-threo-beta-benzyloxyaspartate in the mutants was similar to that of wild type EAAC1. Our results are consistent with the idea that the side chain of Met-367 fulfills a steric role in the positioning of the substrate in the binding pocket in a step subsequent to its initial binding.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/metabolismo , Metionina/química , Sistema X-AG de Transporte de Aminoácidos/química , Animais , Transporte Biológico , Clonagem Molecular , Eletrofisiologia/métodos , Transportador 3 de Aminoácido Excitatório/química , Vetores Genéticos , Células HeLa , Humanos , Mutação , Oócitos/metabolismo , Coelhos , Especificidade por Substrato , Xenopus laevis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...