Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biochim Biophys Acta ; 1555(1-3): 92-5, 2002 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-12206897

RESUMO

Glutamate transporters are essential for terminating synaptic excitation and for maintaining extracellular glutamate concentrations below neurotoxic levels. These transporters also mediate a thermodynamically uncoupled chloride flux, activated by two of the molecules they transport, sodium and glutamate. Five eukaryotic glutamate transporters have been cloned and identified. They exhibit approximately 50% identity and this homology is even greater at the carboxyl terminal half, which is predicted to have an unusual topology. Determination of the topology shows that the carboxyl terminal part contains several transmembrane domains separated by two reentrant loops that are in close proximity to each other. We have identified several conserved amino acid residues in the carboxyl terminal half that play crucial roles in the interaction of the transporter with its substrates: sodium, potassium and glutamate. The conformation of the transporter gating the anion conductance is different from that during substrate translocation. However, there exists a dynamic equilibrium between these conformations.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/química , Sistema X-AG de Transporte de Aminoácidos/genética , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Arginina/química , Sítios de Ligação , Cloretos/química , Clonagem Molecular , Transportador 2 de Aminoácido Excitatório/química , Transportador 2 de Aminoácido Excitatório/metabolismo , Ácido Glutâmico/metabolismo , Conformação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Sódio/metabolismo
2.
Biochem Soc Trans ; 29(Pt 6): 707-10, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11709060

RESUMO

Glutamate transporters are essential for terminating synaptic excitation and for maintaining extracellular glutamate concentrations below neurotoxic levels. These transporters also mediate a thermodynamically uncoupled chloride flux that is activated by two of the molecules that they transport - sodium and glutamate. Five eukaryotic glutamate transporters have been cloned and identified. They exhibit approximately 50% identity and this homology is even greater in the carboxyl terminal half, which is predicted to have an unusual topology. Determination of the topology shows that the carboxyl terminal part of the molecule contains several transmembrane domains that are separated by at least two re-entrant loops. In these structural elements, we have identified several conserved amino acid residues that play crucial roles in the interaction with the transporter substrates sodium, potassium and glutamate.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/química , Animais , Arginina/química , Sítios de Ligação , Sequência Conservada , Ácido Glutâmico/química , Humanos , Modelos Biológicos , Modelos Químicos , Família Multigênica , Potássio/química , Ligação Proteica , Sódio/química , Relação Estrutura-Atividade
3.
J Biol Chem ; 276(44): 40476-85, 2001 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-11527967

RESUMO

Two high affinity Zn(2+) binding sites were engineered in the otherwise Zn(2+)-insensitive rat gamma-aminobutyric acid (GABA) transporter-1 (rGAT-1) based on structural information derived from Zn(2+) binding sites engineered previously in the homologous dopamine transporter. Introduction of a histidine (T349H) at the extracellular end of transmembrane segment (TM) 7 together with a histidine (E370H) or a cysteine (Q374C) at the extracellular end of TM 8 resulted in potent inhibition of [3H]GABA uptake by Zn(2+) (IC(50) = 35 and 44 microM, respectively). Upon expression in Xenopus laevis oocytes it was similarly observed that Zn(2+) was a potent inhibitor of the GABA-induced current (IC(50) = 21 microM for T349H/E370H and 51 microM for T349H/Q374C), albeit maximum inhibition was only approximately 40% in T349H/E370H versus approximately 90% in T349H/Q374C. In the wild type, Zn(2+) did not affect the Na(+)-dependent transient currents elicited by voltage jumps and thought to reflect capacitive charge movements associated with Na(+) binding. However, in both mutants Zn(2+) caused a reduction of the inward transient currents upon jumping to hyperpolarized potentials as reflected in rightward-shifted Q/V relationships. This suggests that Zn(2+) is inhibiting transporter function by stabilizing the outward-facing Na(+)-bound state. Translocation of lithium by the transporter does not require GABA binding and analysis of this uncoupled Li(+) conductance revealed a potent inhibition by Zn(2+) in T349H/E370H, whereas surprisingly the T349H/Q374C leak was unaffected. This differential effect supports that the leak conductance represents a unique operational mode of the transporter involving conformational changes different from those of the substrate translocation process. Altogether our results support both an evolutionary conserved structural organization of the TM 7/8 domain and a key role of this domain in GABA-dependent and -independent conformational changes of the transporter.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras , Transportadores de Ânions Orgânicos , Zinco/metabolismo , Ácido gama-Aminobutírico/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Células COS , Proteínas de Transporte/química , Proteínas de Transporte/genética , Proteínas da Membrana Plasmática de Transporte de GABA , Proteínas de Membrana/química , Proteínas de Membrana/genética , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Xenopus laevis
4.
J Biol Chem ; 276(44): 40396-401, 2001 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-11479303

RESUMO

In the central nervous system a family of related (Na(+)-K(+))-coupled glutamate transporters remove the transmitter from the cleft and prevent its neurotoxic actions. In addition to this coupled uptake, these transporters also mediate a sodium- and glutamate-dependent uncoupled anion conductance. Most models assume that the initial steps for both processes are the same, leading to the anticipation that both may exhibit a similar requirement for cations. In this study we have tested this idea in the neuronal glutamate transporter EAAC-1. We report that in this transporter lithium can replace sodium in the coupled uptake. Strikingly, the glutamate-dependent gating of the uncoupled conductance mediated by EAAC-1 has a strict requirement for sodium; lithium cannot substitute for it. Moreover, we describe two mutants, T370S and G410S, in which the cation selectivity of the two processes is affected differently. In both mutants sodium, but not lithium, can support coupled transport. On the other hand, the sodium selectivity of the gated anion conductance in oocytes expressing the mutant transporters is not affected. Our observations indicate that although both the coupled and the uncoupled fluxes are sodium-dependent, the conformation gating the anion conductance is different from that during substrate translocation.


Assuntos
Sistema X-AG de Transporte de Aminoácidos , Proteínas de Transporte/metabolismo , Lítio/farmacologia , Neurônios/efeitos dos fármacos , Simportadores , Clonagem Molecular , Proteínas de Transporte de Glutamato da Membrana Plasmática , Células HeLa , Humanos , Ativação do Canal Iônico , Transporte de Íons , Mutagênese , Neurônios/metabolismo , Sódio/metabolismo , Especificidade por Substrato
5.
J Biol Chem ; 275(48): 37436-42, 2000 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-10978338

RESUMO

Glutamate transporters from the central nervous system play a crucial role in the clearance of the transmitter from the synaptic cleft. Glutamate is cotransported with sodium ions, and the electrogenic translocation cycle is completed by countertransport of potassium. Mutants that cannot interact with potassium are only capable of catalyzing electroneutral exchange. Here we identify a residue involved in controlling substrate recognition in the neuronal transporter EAAC-1 that transports acidic amino acids as well as cysteine. When arginine 447, a residue conserved in all glutamate transporters, is replaced by cysteine, transport of glutamate or aspartate is abolished, but sodium-dependent cysteine transport is left intact. Analysis of other substitution mutants shows that the replacement of arginine rather than the introduced cysteine is responsible for the observed phenotype. In further contrast to wild type, acidic amino acids are unable to inhibit cysteine transport in R447C-EAAC-1, indicating that the selectivity change is manifested at the binding step. Electrophysiological analysis shows that in the mutant cysteine, transport has become electroneutral, and its interaction with the countertransported potassium is impaired. Thus arginine 447 plays a pivotal role in the sequential interaction of acidic amino acids and potassium with the transporter and, thereby, constitutes one of the molecular determinants of coupling their fluxes.


Assuntos
Sistema X-AG de Transporte de Aminoácidos , Arginina/metabolismo , Proteínas de Transporte/metabolismo , Glutamatos/metabolismo , Neurônios/metabolismo , Simportadores , Sequência de Aminoácidos , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/fisiologia , Proteínas de Transporte de Glutamato da Membrana Plasmática , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Ligação Proteica , Xenopus laevis
6.
J Biol Chem ; 275(44): 34106-13, 2000 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-10926932

RESUMO

The gamma-aminobutyric acid (GABA) transporter GAT-1 is a prototype of a large family of neurotransmitter transporters that includes those of dopamine and serotonin. GAT-1 maintains low synaptic concentrations of neurotransmitter by coupling GABA uptake to the fluxes of sodium and chloride. Here we identify a stretch of four amino acid residues predicted to lie in the juxtamembrane region prior to transmembrane domain 1 in the cytoplasmic amino-terminal tail of GAT-1, which is critical for its function. Two residues, arginine 44 and tryptophan 47, are fully conserved within the transporter family, and their deletion abolishes GABA transport in the HeLa cell expression system used. Tryptophan 47 can be replaced only by aromatic residues without loss of activity. Arginine 44 is essential for activity. Only when it is replaced by lysine, low activity levels (around 15% of those of the wild type) are observed. Using a reconstitution assay, we show that mutants in which this residue is replaced by lysine or histidine exhibit sodium- and chloride-dependent GABA exchange similar to the wild type. This indicates that these mutants are selectively impaired in the reorientation of the unloaded transporter, a step in the translocation cycle by which net flux and exchange differ. The high degree of conservation in the consensus sequence RXXW suggests that this region may influence the reorientation step in related transporters as well.


Assuntos
Arginina/metabolismo , Encéfalo/metabolismo , Proteínas de Transporte/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras , Transportadores de Ânions Orgânicos , Substituição de Aminoácidos , Animais , Arginina/química , Arginina/genética , Proteínas de Transporte/química , Proteínas de Transporte/genética , Proteínas da Membrana Plasmática de Transporte de GABA , Células HeLa , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/genética , Mutagênese Sítio-Dirigida , Ratos , Triptofano/química , Triptofano/genética , Triptofano/metabolismo
7.
J Biol Chem ; 275(13): 9684-9, 2000 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-10734120

RESUMO

The excitatory neurotransmitter glutamate is removed from the synaptic cleft by several related sodium- and potassium-coupled transporters. They thereby restrict the neurotoxicity of this transmitter. Based on the accessibility of single cysteines to the large sulfhydryl reagent 3-N-maleimidyl(propionyl)biocytin, we have proposed a topological model for the astroglial glutamate transporter GLT-1 (Grunewald, M., Bendahan, A. and Kanner, B. I. (1998) Neuron 21, 623-632). Because of several unexpected observations, we have investigated the topological disposition of 19 cysteine residues engineered into a loop proposed to be intracellular. We have probed the accessibility of these cysteines to small and large sulfhydryl reagents. The impermeant hydrophilic sulfhydryl reagent [(2-trimethylammonium)ethyl] methanethiosulfonate inhibits transport activity only at two of these positions, weakly at G365C and potently at A364C. Glutamate and its nontransportable analogue dihydrokainate markedly protect A364C transporters against this impermeant reagent. Using a biotinylated maleimide, we found that, among the 14 mutants tested with it, only A364C is accessible to it from the extracellular side. This, together with our previous observations, indicates that the loop-including amino acid residues 354, 359, 373, and 379-is largely intracellular, but a short region of it forms a reentrant pore-loop-like structure, the accessibility of which is dependent on the conformation of the transporter.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Ácido Glutâmico/metabolismo , Transportadores de Cassetes de Ligação de ATP/química , Alanina/metabolismo , Sequência de Aminoácidos , Sistema X-AG de Transporte de Aminoácidos , Transporte Biológico , Células HeLa , Humanos , Indicadores e Reagentes , Dados de Sequência Molecular , Especificidade por Substrato
8.
J Biol Chem ; 274(33): 23020-6, 1999 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-10438469

RESUMO

The gamma-aminobutyric acid (GABA) transporter GAT-1 is a prototype of neurotransmitter transporters that maintain low synaptic levels of the transmitter. Transport by GAT-1 is sensitive to the polar sulfhydryl reagent 2-aminoethyl methanethiosulfonate. Following replacement of endogenous cysteines to other residues by site-directed mutagenesis, we have identified cysteine 399 as the major determinant of the sensitivity of the transporter to sulfhydryl modification. Cysteine-399 is located in the intracellular loop connecting putative transmembrane domains eight and nine. Binding of both sodium and chloride leads to a reduced sensitivity to sulfhydryl reagents, whereas subsequent binding of GABA increases it. Strikingly binding of the nontransportable GABA analogue SKF100330A gives rise to a marked protection against sulfhydryl modification. These effects were not observed in C399S transporters. Under standard conditions GAT-1 is almost insensitive toward the impermeant 2-(trimethylammonium)ethyl methanethiosulfonate. However, in a chloride-free medium, addition of SKF100330A renders wild type GAT-1, but not C399S, very sensitive to this impermeant reagent. These observations indicate that the accessibility of cysteine 399 is highly dependent on the conformation of GAT-1. Consequently, topological assignments based on accessibility of endogeneous or engineered cysteines to small polar sulfhydryl reagents need to be interpreted with extreme caution.


Assuntos
Proteínas de Transporte/metabolismo , Metanossulfonato de Etila/análogos & derivados , Proteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras , Transportadores de Ânions Orgânicos , Reagentes de Sulfidrila/farmacologia , Ácido gama-Aminobutírico/metabolismo , Proteínas de Transporte/química , Proteínas de Transporte/genética , Metanossulfonato de Etila/farmacologia , Proteínas da Membrana Plasmática de Transporte de GABA , Células HeLa , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/genética , Mutagênese Sítio-Dirigida , Ácidos Nicotínicos/farmacologia , Estrutura Secundária de Proteína
9.
Proc Natl Acad Sci U S A ; 96(4): 1710-5, 1999 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-9990089

RESUMO

The neurotoxicity of glutamate in the central nervous system is restricted by several (Na+ + K+)-coupled transporters for this neurotransmitter. The astroglial transporter GLT-1 is the only subtype that exhibits high sensitivity to the nontransportable glutamate analogue dihydrokainate. A marked reduction in sensitivity to the blocker is observed when serine residues 440 and 443 are mutated to glycine and glutamine, which, respectively, occupy these positions in the other homologous glutamate transporters. They are located in the ascending limb of the recently identified pore-loop-like structure. Strikingly, mutation of serine-440 to glycine enables not only sodium but also lithium ions to drive net influx of acidic amino acids. Moreover, the efficiency of lithium as a driving ion for glutamate transport depends on the nature of the amino acid residue present at position 443. Mutant transporters containing single cysteines at the position of either serine residue become sensitive to positively as well as negatively charged methanethiosulfonate derivatives. In S440C transporters significant protection against this inhibition is provided both by transportable and nontransportable glutamate analogues, but not by sodium alone. Our observations indicate that the pore-loop-like structure plays a pivotal role in coupling ion and glutamate fluxes and suggest that it is close to the glutamate-binding site.


Assuntos
Transportadores de Cassetes de Ligação de ATP/química , Transportadores de Cassetes de Ligação de ATP/metabolismo , Ácido Glutâmico/metabolismo , Serina , Sódio/metabolismo , Substituição de Aminoácidos , Sistema X-AG de Transporte de Aminoácidos , Ácido Aspártico/metabolismo , Sítios de Ligação , Transporte Biológico/efeitos dos fármacos , Glicina , Células HeLa , Humanos , Ácido Caínico/análogos & derivados , Ácido Caínico/farmacologia , Cinética , Mutagênese Sítio-Dirigida , Neurotransmissores/metabolismo , Proteínas Recombinantes/metabolismo
10.
Neuron ; 21(3): 623-32, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9768848

RESUMO

In the central nervous system, (Na+ + K+)-coupled glutamate transporters restrict the neurotoxicity of this transmitter and limit the duration of synaptic excitation at some synapses. The various isotransporters exhibit a particularly high homology in an extended hydrophobic domain of ill-defined topology that contains several determinants involved in ion and transmitter binding. Here, we describe the determination of the membrane topology of the cloned astroglial glutamate transporter GLT-1. A series of functional transporters containing single cysteines was engineered. Their topological disposition was determined by using a biotinylated sulfhydryl reagent. The glutamate transporter has eight transmembrane domains long enough to span the membrane as et heiices. Strikingly, between the seventh and eighth domains, a structure reminiscent of a pore loop and an outward-facing hydrophobic linker are positioned.


Assuntos
Transportadores de Cassetes de Ligação de ATP/química , Transportadores de Cassetes de Ligação de ATP/metabolismo , Astrócitos/metabolismo , Encéfalo/metabolismo , Cisteína , Sequência de Aminoácidos , Substituição de Aminoácidos , Sistema X-AG de Transporte de Aminoácidos , Animais , Transporte Biológico , Biotinilação , Clonagem Molecular , Glicosilação , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fragmentos de Peptídeos/química , Mutação Puntual , Conformação Proteica , Ratos , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
11.
J Biol Chem ; 273(23): 14231-7, 1998 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-9603927

RESUMO

Glutamate transporters remove this transmitter from the extracellular space by cotransport with three sodium ions and a proton. The cycle is completed by translocation of a potassium ion in the opposite direction. Recently we have identified two adjacent amino acid residues of the glutamate transporter GLT-1 that influence potassium coupling. Using the scanning cysteine accessibility method we have now explored the highly conserved region surrounding them. Replacement of each of the five consecutive residues 396-400 by cysteine abolished transport activity but at several other positions the substitution is tolerated. One residue, tyrosine 403, was identified where cysteine substitution renders the transporter sensitive to modification by positively charged methanethiosulfonate derivates in a sodium-protectable fashion. In the presence of sodium, the nontransported glutamate analogue dihydrokainate potentiated the covalent modification, presumably by binding to the glutamate site and locking the protein in a conformation in which tyrosine 403 is accessible from the external bulk medium. In contrast, transported substrates significantly slowed the reaction, suggesting that during the transport cycle residue 403 becomes occluded. On the other hand, transportable substrates are not able to protect Y403C transporters against N-ethylmaleimide, which is highly permeant but unable to modify cysteine residues buried within membrane proteins. These results indicate that tyrosine 403 is alternately accessible from either side of the membrane, consistent with its role as structural determinant of the potassium binding site.


Assuntos
Transportadores de Cassetes de Ligação de ATP/química , Potássio/metabolismo , Conformação Proteica , Transportadores de Cassetes de Ligação de ATP/genética , Sistema X-AG de Transporte de Aminoácidos , Aminoácidos/farmacologia , Ácido Aspártico/farmacocinética , Sítios de Ligação/genética , Transporte Biológico/fisiologia , Sequência Conservada/fisiologia , Cisteína/metabolismo , Etilmaleimida/farmacologia , Células HeLa , Humanos , Ácido Caínico/análogos & derivados , Ácido Caínico/farmacologia , Mesilatos/farmacologia , Mutagênese Sítio-Dirigida , Sódio/farmacologia , Reagentes de Sulfidrila/farmacologia
12.
Proc Natl Acad Sci U S A ; 95(2): 751-5, 1998 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-9435265

RESUMO

Glutamate transporters remove this neurotransmitter from the synaptic cleft by a two-stage electrogenic process, in which glutamate is first cotransported with three sodium ions and a proton. Subsequently, the cycle is completed by translocation of a potassium ion in the opposite direction. Recently, we have identified an amino acid residue of the glutamate transporter GLT-1 (Glu-404) that influences potassium coupling. We have now analyzed the effect of seven other amino acid residues in the highly conserved region surrounding this site. One of these residues, Tyr-403, also proved important for potassium coupling, because mutation to Phe (Y403F) resulted in an electroneutral obligate exchange mode of glutamate transport. This mutation in the transporter also caused an approximately 8-fold increase in the apparent sodium affinity, with no change in the apparent affinity for L-glutamate or D-aspartate. Strikingly, although exchange catalyzed by the wild-type transporter is strictly dependent on sodium, the selectivity of Y403F mutant transporters is altered so that sodium can be replaced by other alkaline metal cations including lithium and cesium. These results indicate the presence of interacting sites in or near the transporter pore that control selectivity for sodium and potassium.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Encéfalo/metabolismo , Sistema X-AG de Transporte de Aminoácidos , Animais , Células HeLa , Humanos , Transporte de Íons , Técnicas de Patch-Clamp , Potássio/metabolismo , Ratos , Sódio/metabolismo , Transfecção
13.
J Biol Chem ; 272(26): 16096-102, 1997 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-9195904

RESUMO

The gamma-aminobutyric acid (GABA) transporter GAT-1 is located in nerve terminals and catalyzes the electrogenic reuptake of the neurotransmitter with two sodium ions and one chloride. We now identify a single tyrosine residue that is critical for GABA recognition and transport. It is completely conserved throughout the superfamily, and even substitution to the other aromatic amino acids, phenylalanine (Y140F) and tryptophan (Y140W), results in completely inactive transporters. Electrophysiological characterization reveals that both mutant transporters exhibit the sodium-dependent transient currents associated with sodium binding as well as the chloride-dependent lithium leak currents characteristic of GAT-1. On the other hand, in both mutants GABA is neither able to induce a steady-state transport current nor to block their transient currents. The nontransportable analog SKF 100330A potently inhibits the sodium-dependent transient in the wild type GAT-1 but not in the Y140W transporter. It partly blocks the transient of Y140F. Thus, although sodium and chloride binding are unimpaired in the tyrosine mutants, they have a specific defect in the binding of GABA. The total conservation of the residue throughout the family suggests that tyrosine 140 may be involved in the liganding of the amino group, the moiety common to all of the neurotransmitters.


Assuntos
Proteínas de Transporte/química , Proteínas de Membrana/química , Proteínas de Membrana Transportadoras , Proteínas do Tecido Nervoso/química , Transportadores de Ânions Orgânicos , Ácido gama-Aminobutírico/metabolismo , Transporte Biológico , Proteínas de Transporte/fisiologia , Cloretos/metabolismo , Proteínas da Membrana Plasmática de Transporte de GABA , Células HeLa , Humanos , Proteínas de Membrana/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Sódio/metabolismo , Relação Estrutura-Atividade , Tirosina
14.
J Biol Chem ; 272(3): 1703-8, 1997 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-8999849

RESUMO

Glutamate transporters maintain low synaptic concentrations of neurotransmitter by coupling uptake to flux of other ions. After cotransport of glutamic acid with Na+, the cycle is completed by countertransport of K+. We have identified an amino acid residue (glutamate 404) influencing ion coupling in a domain of the transporter implicated previously in kainate binding. Mutation of this residue to aspartate (E404D) prevents both forward and reverse transport induced by K+. Sodium-dependent transmitter exchange and a transporter-mediated chloride conductance are unaffected by the mutation, indicating that this residue selectively influences potassium flux coupling. The results support a kinetic model in which sodium and potassium are translocated in distinct steps and suggest that this highly conserved region of the transporter is intimately associated with the ion permeation pathway.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Potássio/metabolismo , Transportadores de Cassetes de Ligação de ATP/genética , Sistema X-AG de Transporte de Aminoácidos , Animais , Transporte Biológico , Células HeLa , Humanos , Mutagênese Sítio-Dirigida , Ratos
15.
J Biol Chem ; 272(2): 1203-10, 1997 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-8995422

RESUMO

The membrane topology of GAT-1, a sodium- and chloride-coupled gamma-aminobutyric acid transporter from rat brain, has been probed using N-glycosylation scanning mutagenesis. Overall, the results support the theoretical 12-transmembrane segment model. This model (based on hydropathy analysis) was originally proposed for GAT-1 and adopted for all other members of the sodium- and chloride-dependent neurotransmitter transporter superfamily. However, our data indicate that the loop connecting putative transmembrane domains 2 and 3, which was predicted to be located intracellularly, can be glycosylated in vivo. Furthermore, studies with permeant and impermeant methanesulfonate reagents suggest that cysteine 74, located in the hydrophilic loop connecting transmembrane domains 1 and 2, is intracellular rather than extracellular. We present a model in which the topology deviates from the theoretical one in the amino-terminal third of the transporter. It also contains 12 transmembrane segments, but the highly conserved domain 1 does not form a conventional transmembrane alpha-helix.


Assuntos
Química Encefálica , Proteínas de Transporte/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras , Transportadores de Ânions Orgânicos , Sequência de Aminoácidos , Animais , Sequência de Bases , Sequência Consenso , Metanossulfonato de Etila/análogos & derivados , Metanossulfonato de Etila/metabolismo , Proteínas da Membrana Plasmática de Transporte de GABA , Glicosilação , Células HeLa , Humanos , Indicadores e Reagentes , Modelos Moleculares , Dados de Sequência Molecular , Ratos , Ácido gama-Aminobutírico/metabolismo
16.
J Neurosci ; 16(17): 5405-14, 1996 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-8757253

RESUMO

This study addresses the binding of ions and the permeation of substrates during function of the GABA transporter GAT1. GAT1 was expressed in Xenopus oocytes and studied electrophysiologically as well as with [3H]GABA flux; GAT1 was also expressed in mammalian cells and studied with [3H]GABA and [3H]tiagabine binding. Voltage jumps, Na+ and Cl- concentration jumps, and exposure to high-affinity blockers (NO-05-711 and SKF-100330A) all produce capacitive charge movements. Occlusive interactions among these three types of perturbations show that they all measure the same population of charges. The concentration dependences of the charge movements reveal (1) that two Na+ ions interact with the transporter even in the absence of GABA, and (2) that Cl- facilitates the binding of Na+. Comparison between the charge movements and the transport-associated current shows that this initial Na(+)-transporter interaction limits the overall transport rate when [GABA] is saturating. However, two classes of manipulation--treatment with high-affinity uptake blockers and the W68L mutation-"lock" Na+ onto the transporter by slowing or preventing the subsequent events that release the substrates to the intracellular medium. The Na+ substitutes Li+ and Cs+ do not support charge movements, but they can permeate the transporter in an uncoupled manner. Our results (1) support the hypothesis that efficient removal of synaptic transmitter by the GABA transporter GAT1 depends on the previous binding of Na+ and Cl-, and (2) indicate the important role of the conserved putative transmembrane domain 1 in interactions with the permeant substrates.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras , Transportadores de Ânions Orgânicos , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/fisiologia , Cloretos/farmacologia , Eletrofisiologia , Proteínas da Membrana Plasmática de Transporte de GABA , Células HeLa , Homeostase , Humanos , Íons , Proteínas de Membrana/genética , Proteínas de Membrana/fisiologia , Mutação , Proteínas do Tecido Nervoso/metabolismo , Inibidores da Captação de Neurotransmissores/farmacologia , Oócitos/metabolismo , Permeabilidade , Sódio/metabolismo , Xenopus , Ácido gama-Aminobutírico/metabolismo
18.
FEBS Lett ; 371(1): 39-42, 1995 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-7664880

RESUMO

We have investigated the possible role of selected negatively-charged amino acids of the sodium and chloride-coupled GABA transporter GAT-1 on sodium binding. These residues located adjacent to putative transmembrane domains and which are conserved throughout the large superfamily of neurotransmitter transporters were changed by site-directed mutagenesis. The functional consequences were that one of the residues, glutamate-101, was critical for transport. Its replacement by aspartate left only 1% of the activity, and no activity could be detected when it was replaced by other residues. Expression levels and targeting to the plasma membrane of the mutant transporters appeared normal. Transient sodium currents were not observed in the mutants, and increased sodium concentrations did not affect the percentage of wild type transport of the E101D mutant. It is concluded that residue glutamate-101 is critical for one or more of the conformational changes of GAT-1 during its transport cycle.


Assuntos
Proteínas de Transporte/metabolismo , Ácido Glutâmico/fisiologia , Proteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras , Transportadores de Ânions Orgânicos , Ácido gama-Aminobutírico/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Transporte Biológico , Proteínas de Transporte/química , Proteínas de Transporte/genética , Membrana Celular/metabolismo , Cloretos/fisiologia , Condutividade Elétrica , Proteínas da Membrana Plasmática de Transporte de GABA , Células HeLa , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/genética , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Conformação Proteica , Sódio/fisiologia
19.
J Biol Chem ; 270(29): 17093-7, 1995 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-7615503

RESUMO

Sodium-coupled glutamate transporters, located in the plasma membrane of nerve terminals and glial processes, serve to keep its extracellular glutamate concentration below extracellular levels. Moreover, they help in conjunction with diffusion to terminate the transmitter's action in synaptic transmission. We have investigated the role of negatively charged amino acid residues of GLT-1, a cloned (Na+ + K+)-coupled glutamate transporter from rat brain. Using site-directed mutagenesis we modified these negative residues, which are located in hydrophobic surroundings and are highly conserved within the glutamate transporter family. Out of five residues meeting these criteria, three, aspartate 398, glutamate 404, and aspartate 470, are critical for heterologously expressed glutamate transport. This defective transport cannot be attributed to the mere requirement of a negative charge at these positions. After prelabeling of the proteins with [35S]methionine, immunoprecipitation of all mutant transporters indicates that their expression levels are similar to that of wild type. No cryptic activity was revealed by reconstitution experiments aimed to monitor the activity of transporter molecules not located in the plasma membrane. Significantly, whereas all of the mutants at the glutamate 404 position exhibit impaired transport of glutamate, they possess considerable transport of D- and L-aspartate, up to 80% of wild type values. Binding of glutamate is not impaired in these mutants. Our observations indicate that the glutamate 404 residue may be located in the vicinity of the glutamate-aspartate permeation pathway.


Assuntos
Transportadores de Cassetes de Ligação de ATP/química , Encéfalo/metabolismo , Ácido Glutâmico/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Sistema X-AG de Transporte de Aminoácidos , Animais , Sequência de Bases , Transporte Biológico , Células HeLa , Humanos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Ratos , Relação Estrutura-Atividade
20.
FEBS Lett ; 356(2-3): 191-4, 1994 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-7805836

RESUMO

Transporter proteins consist of multiple transmembrane domains connected by hydrophillic loops. As the importance of these loops in transport processes is poorly understood, we have studied this question using the cDNA coding for GAT-1, a Na+/Cl(-)-coupled gamma-aminobutyric acid transporter from rat brain. Deletions of randomly picked non-conserved single amino acids in the loops connecting helices 7 and 8 or 8 and 9 result in inactive transport upon expression in HeLa cells. However, transporters where these amino acids are replaced with glycine retain significant activity. The expression level of the inactive mutant transporters was similar to that of the wild-type, but one of these, delta Val-348, appears to be defectively targetted to the plasma membrane. Our data are compatible with the idea that a minimal length of the loops is required, presumably to enable the transmembrane domains to interact optimally with each other.


Assuntos
Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras , Transportadores de Ânions Orgânicos , Estrutura Secundária de Proteína , Ácido gama-Aminobutírico/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Encéfalo/metabolismo , Proteínas de Transporte/biossíntese , Clonagem Molecular , Sequência Conservada , Primers do DNA , Escherichia coli , Proteínas de Escherichia coli , Proteínas da Membrana Plasmática de Transporte de GABA , Células HeLa , Humanos , Lipossomos , Proteínas de Membrana/biossíntese , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Proteolipídeos/metabolismo , Ratos , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Deleção de Sequência , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...