Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
PLoS Biol ; 17(5): e3000260, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31083648

RESUMO

Members of the solute carrier 17 (SLC17) family use divergent mechanisms to concentrate organic anions. Membrane potential drives uptake of the principal excitatory neurotransmitter glutamate into synaptic vesicles, whereas closely related proteins use proton cotransport to drive efflux from the lysosome. To delineate the divergent features of ionic coupling by the SLC17 family, we determined the structure of Escherichia coli D-galactonate/H+ symporter D-galactonate transporter (DgoT) in 2 states: one open to the cytoplasmic side and the other open to the periplasmic side with substrate bound. The structures suggest a mechanism that couples H+ flux to substrate recognition. A transition in the role of H+ from flux coupling to allostery may confer regulation by trafficking to and from the plasma membrane.


Assuntos
Metabolismo Energético , Escherichia coli/metabolismo , Transportadores de Ânions Orgânicos/química , Transportadores de Ânions Orgânicos/metabolismo , Transporte Biológico , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Modelos Moleculares , Conformação Proteica , Prótons , Açúcares Ácidos/metabolismo
2.
Nat Commun ; 9(1): 4005, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30275448

RESUMO

Multidrug resistance (MDR) poses a major challenge to medicine. A principle cause of MDR is through active efflux by MDR transporters situated in the bacterial membrane. Here we present the crystal structure of the major facilitator superfamily (MFS) drug/H+ antiporter MdfA from Escherichia coli in an outward open conformation. Comparison with the inward facing (drug binding) state shows that, in addition to the expected change in relative orientations of the N- and C-terminal lobes of the antiporter, the conformation of TM5 is kinked and twisted. In vitro reconstitution experiments demonstrate the importance of selected residues for transport and molecular dynamics simulations are used to gain insights into antiporter switching. With the availability of structures of alternative conformational states, we anticipate that MdfA will serve as a model system for understanding drug efflux in MFS MDR antiporters.


Assuntos
Antiporters/química , Proteínas de Escherichia coli/química , Escherichia coli/metabolismo , Proteínas de Membrana Transportadoras/química , Modelos Moleculares , Substituição de Aminoácidos , Antiporters/genética , Antiporters/metabolismo , Membrana Celular/metabolismo , Cloranfenicol/metabolismo , Cristalografia por Raios X , Resistência a Múltiplos Medicamentos/fisiologia , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Interações Hidrofóbicas e Hidrofílicas , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Simulação de Dinâmica Molecular , Estrutura Secundária de Proteína , Transporte Proteico , Relação Estrutura-Atividade
3.
Biochim Biophys Acta Biomembr ; 1860(11): 2456-2464, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30028956

RESUMO

Human MATE1 (multidrug and toxin extrusion 1, hMATE1) is a H+/organic cation (OC) exchanger responsible for the final step of toxic organic cation excretion in the kidney and liver. To investigate the mechanism of transport, we have established an in vitro assay procedure that includes its expression in insect cells, solubilization with octyl glucoside, purification, and reconstitution into liposomes. The resultant proteoliposomes containing hMATE1 as the sole protein component took up radiolabeled tetraethylammonium (TEA) in a ∆pH-dependent and electroneutral fashion. Furthermore, lipid-detergent micelle containing hMATE1 showed ∆pH-dependent TEA binding similar to transport. Mutated hMATE1 with replacement E273Q completely lacked these TEA binding and transport. In the case of divalent substrates, transport was electrogenic. These observations indicate that the stoichiometry of OC/H+ exchange is independent of substrate charge. Purification and reconstitution of hMATE1 is considered to be suitable for understanding the detailed molecular mechanisms of hMATE1. The results suggest that Glu273 of hMATE1 plays essential roles in substrate binding and transport.


Assuntos
Proteínas de Transporte de Cátions Orgânicos/metabolismo , Tetraetilamônio/metabolismo , Cátions/química , Cátions/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Potenciais da Membrana , Mutagênese Sítio-Dirigida , Proteínas de Transporte de Cátions Orgânicos/química , Proteínas de Transporte de Cátions Orgânicos/genética , Ligação Proteica , Proteolipídeos/química , Proteolipídeos/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Especificidade por Substrato , Tetraetilamônio/química
4.
J Proteome Res ; 17(3): 1108-1119, 2018 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-29350038

RESUMO

Structural analysis of purified active membrane proteins can be performed by mass spectrometry (MS). However, no large-scale expression systems for active eukaryotic membrane proteins are available. Moreover, because membrane proteins cannot easily be digested by trypsin and ionized, they are difficult to analyze by MS. We developed a method for mass spectral analysis of eukaryotic membrane proteins combined with an overexpression system in Escherichia coli. Vesicular glutamate transporter 2 (VGLUT2/SLC17A6) with a soluble α-helical protein and histidine tag on the N- and C-terminus, respectively, was overexpressed in E. coli, solubilized with detergent, and purified by Ni-NTA affinity chromatography. Proteoliposomes containing VGLUT2 retained glutamate transport activity. For MS analysis, the detergent was removed from purified VGLUT2 by trichloroacetic acid precipitation, and VGLUT2 was then subjected to reductive alkylation and tryptic digestion. The resulting peptides were detected with 88% coverage by matrix-assisted laser desorption ionization time-of-flight (MALDI-TOF) MS with or without liquid chromatography. Vesicular excitatory amino acid transporter and vesicular acetylcholine transporter were also detected with similar coverage by the same method. Thus this methodology could be used to analyze purified eukaryotic active transporters. Structural analysis with chemical modifiers by MS could have applications in functional binding analysis for drug discovery.


Assuntos
Transportador 1 de Aminoácido Excitatório/análise , Fragmentos de Peptídeos/análise , Proteínas Vesiculares de Transporte de Acetilcolina/análise , Proteína Vesicular 2 de Transporte de Glutamato/análise , Animais , Precipitação Química , Clonagem Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Transportador 1 de Aminoácido Excitatório/genética , Transportador 1 de Aminoácido Excitatório/metabolismo , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Humanos , Camundongos , Mapeamento de Peptídeos , Proteólise , Ratos , Proteínas Recombinantes/análise , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Ácido Tricloroacético/química , Tripsina/química , Proteínas Vesiculares de Transporte de Acetilcolina/genética , Proteínas Vesiculares de Transporte de Acetilcolina/metabolismo , Proteína Vesicular 2 de Transporte de Glutamato/genética , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo
5.
Epilepsia ; 58(5): 845-857, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28294308

RESUMO

OBJECTIVE: The ketogenic diet is clinically used to treat drug-resistant epilepsy. The diet treatment markedly increases ketone bodies (acetoacetate and ß-hydroxybutyrate), which work as energy metabolites in the brain. Here, we investigated effects of acetoacetate on voltage-dependent Ca2+ channels (VDCCs) in pyramidal cells of the hippocampus. We further explored an acetoacetate analog that inhibited VDCCs in pyramidal cells, reduced excitatory postsynaptic currents (EPSCs), and suppressed seizures in vivo. METHODS: The effects of acetoacetate and its analogs on VDCCs and EPSCs were evaluated using patch-clamp recordings from CA1 pyramidal cells of mouse hippocampal slices. The in vivo effects of these reagents were also evaluated using a chronic seizure model induced by intrahippocampal injection of kainate. RESULTS: Acetoacetate inhibited VDCCs in pyramidal cells of hippocampal slices, and reduced EPSCs in slices exhibiting epileptiform activity. More potent EPSC inhibitors were then explored by modifying the chemical structure of acetoacetate, and 2-phenylbutyrate was identified as an acetoacetate analog that inhibited VDCCs and EPSCs more potently. Although acetoacetate is known to inhibit vesicular glutamate transporters (VGLUTs), 2-phenylbutyrate did not inhibit VGLUTs, showing that 2-phenylbutyrate is an acetoacetate analog that preferably inhibits VDCCs. In addition, 2-phenylbutyrate markedly reduced EPSCs in slices exhibiting epileptiform activity, and suppressed hippocampal seizures in vivo in a mouse model of epilepsy. The in vivo antiseizure effects of 2-phenylbutyrate were more potent than those of acetoacetate. Finally, intraperitoneal 2-phenylbutyrate was delivered to the brain, and its brain concentration reached the level enough to reduce EPSCs. SIGNIFICANCE: These results demonstrate that 2-phenylbutyrate is an acetoacetate analog that inhibits VDCCs and EPSCs in pyramidal cells, suppresses hippocampal seizures in vivo, and has brain penetration ability. Thus 2-phenylbutyrate provides a useful chemical structure as a lead compound to develop new antiseizure drugs originating from ketone bodies.


Assuntos
Acetoacetatos/farmacologia , Dieta Cetogênica , Hipocampo/efeitos dos fármacos , Inibição Neural/efeitos dos fármacos , Fenilbutiratos/farmacologia , Células Piramidais/efeitos dos fármacos , Animais , Canais de Cálcio/efeitos dos fármacos , Modelos Animais de Doenças , Eletroencefalografia/efeitos dos fármacos , Feminino , Técnicas In Vitro , Injeções , Ácido Caínico , Masculino , Camundongos , Camundongos Endogâmicos ICR , Técnicas de Cultura de Órgãos
6.
Annu Rev Pharmacol Toxicol ; 56: 385-402, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26514205

RESUMO

Vesicular neurotransmitter transporters are responsible for the accumulation of neurotransmitters in secretory vesicles and play essential roles in chemical transmission. The SLC17 family contributes to sequestration of anionic neurotransmitters such as glutamate, aspartate, and nucleotides. Identification and subsequent cellular and molecular biological studies of SLC17 transporters unveiled the principles underlying the actions of these transporters. Recent progress in reconstitution methods in combination with postgenomic approaches has advanced studies on neurotransmitter transporters. This review summarizes the molecular properties of SLC17-type transporters and recent findings regarding the novel SLC18 transporter.


Assuntos
Transporte Biológico/fisiologia , Interações Medicamentosas/fisiologia , Proteínas Vesiculares de Transporte de Neurotransmissores/metabolismo , Animais , Humanos
7.
Am J Physiol Cell Physiol ; 309(2): C71-80, 2015 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-25972451

RESUMO

Membrane potential (Δψ)-driven and Cl(-)-dependent organic anion transport is a primary function of the solute carrier family 17 (SLC17) transporter family. Although the transport substrates and physiological relevance of the major members are well understood, SLC17A2 protein known to be Na(+)-phosphate cotransporter 3 (NPT3) is far less well characterized. In the present study, we investigated the transport properties and expression patterns of mouse SLC17A2 protein (mNPT3). Proteoliposomes containing the purified mNPT3 protein took up radiolabeled p-aminohippuric acid (PAH) in a Δψ- and Cl(-)-dependent manner. The mNPT3-mediated PAH uptake was inhibited by 4,4'-diisothiocyanostilbene-2,2'-disulfonic acid (DIDs) and Evans blue, common inhibitors of SLC17 family members. The PAH uptake was also inhibited by various anionic compounds, such as hydrophilic nonsteroidal anti-inflammatory drugs (NSAIDs) and urate. Consistent with these observations, the proteoliposome took up radiolabeled urate in a Δψ- and Cl(-)-dependent manner. Immunohistochemistry with specific antibodies against mNPT3 combined with RT-PCR revealed that mNPT3 is present in various tissues, including the hepatic bile duct, luminal membranes of the renal urinary tubules, maternal side of syncytiotrophoblast in the placenta, apical membrane of follicle cells in the thyroid, bronchiole epithelial cells in the lungs, and astrocytes around blood vessels in the cerebrum. These results suggested that mNPT3 is a polyspecific organic anion transporter that is involved in circulation of urate throughout the body.


Assuntos
Membrana Celular/metabolismo , Cloretos/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo I/metabolismo , Ácido Úrico/metabolismo , Animais , Transporte Biológico , Membrana Celular/efeitos dos fármacos , Regulação da Expressão Gênica , Hipuratos/metabolismo , Cinética , Potenciais da Membrana , Camundongos Endogâmicos C57BL , Proteínas Cotransportadoras de Sódio-Fosfato Tipo I/antagonistas & inibidores , Proteínas Cotransportadoras de Sódio-Fosfato Tipo I/genética
8.
Proc Natl Acad Sci U S A ; 112(11): 3356-61, 2015 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-25733858

RESUMO

Extrusion of chloroquine (CQ) from digestive vacuoles through the Plasmodium falciparum CQ resistance transporter (PfCRT) is essential to establish CQ resistance of the malaria parasite. However, the physiological relevance of PfCRT and how CQ-resistant PfCRT gains the ability to transport CQ remain unknown. We prepared proteoliposomes containing purified CQ-sensitive and CQ-resistant PfCRTs and measured their transport activities. All PfCRTs tested actively took up tetraethylammonium, verapamil, CQ, basic amino acids, polypeptides, and polyamines at the expense of an electrochemical proton gradient. CQ-resistant PfCRT exhibited decreased affinity for CQ, resulting in increased CQ uptake. Furthermore, CQ competitively inhibited amino acid transport. Thus, PfCRT is a H(+)-coupled polyspecific nutrient and drug exporter.


Assuntos
Proteínas de Membrana Transportadoras/metabolismo , Prótons , Proteínas de Protozoários/metabolismo , Aminoácidos/metabolismo , Transporte Biológico/efeitos dos fármacos , Cloroquina/farmacologia , Resistência a Medicamentos/efeitos dos fármacos , Tetraetilamônio/metabolismo , Verapamil/farmacologia
9.
J Neurochem ; 127(4): 482-6, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23919636

RESUMO

Vesicular GABA transporter (VGAT) is expressed in GABAergic and glycinergic neurons, and is responsible for vesicular storage and subsequent exocytosis of these inhibitory amino acids. In this study, we show that VGAT recognizes ß-alanine as a substrate. Proteoliposomes containing purified VGAT transport ß-alanine using Δψ but not ΔpH as a driving force. The Δψ-driven ß-alanine uptake requires Cl(-). VGAT also facilitates Cl(-) uptake in the presence of ß-alanine. A previously described VGAT mutant (Glu213Ala) that disrupts GABA and glycine transport similarly abrogates ß-alanine uptake. These findings indicated that VGAT transports ß-alanine through a mechanism similar to those for GABA and glycine, and functions as a vesicular ß-alanine transporter. Vesicular GABA transporter (VGAT) is expressed in GABAergic and glycinergic neurons, and is responsible for vesicular storage and subsequent exocytosis of these inhibitory amino acids. In the present study, we showed that proteoliposomes containing purified VGAT transport ß-alanine using Δψ as a driving force. VGAT also facilitates Cl(-) uptake. Our findings indicated that VGAT functions as a vesicular ß-alanine transporter.


Assuntos
Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/química , beta-Alanina/química , Transporte Biológico , Lipossomos/química , Mutação , Proteolipídeos/química , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/genética
10.
Structure ; 20(4): 582-92, 2012 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-22483106

RESUMO

In spite of its recent achievements, the technique of single particle electron cryomicroscopy (cryoEM) has not been widely used to study proteins smaller than 100 kDa, although it is a highly desirable application of this technique. One fundamental limitation is that images of small proteins embedded in vitreous ice do not contain adequate features for accurate image alignment. We describe a general strategy to overcome this limitation by selecting a fragment antigen binding (Fab) to form a stable and rigid complex with a target protein, thus providing a defined feature for accurate image alignment. Using this approach, we determined a three-dimensional structure of an ∼65 kDa protein by single particle cryoEM. Because Fabs can be readily generated against a wide range of proteins by phage display, this approach is generally applicable to study many small proteins by single particle cryoEM.


Assuntos
Proteínas de Escherichia coli/química , Fragmentos Fab das Imunoglobulinas/química , Pró-Proteína Convertases/química , Serina Endopeptidases/química , Proteínas Vesiculares de Transporte de Glutamato/química , Microscopia Crioeletrônica/métodos , Escherichia coli , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Humanos , Processamento de Imagem Assistida por Computador , Fragmentos Fab das Imunoglobulinas/genética , Fragmentos Fab das Imunoglobulinas/metabolismo , Modelos Moleculares , Peso Molecular , Biblioteca de Peptídeos , Pró-Proteína Convertase 9 , Pró-Proteína Convertases/genética , Pró-Proteína Convertases/metabolismo , Conformação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Proteínas Vesiculares de Transporte de Glutamato/genética , Proteínas Vesiculares de Transporte de Glutamato/metabolismo
11.
Biochemistry ; 50(25): 5558-65, 2011 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-21612282

RESUMO

Glutamate plays essential roles in chemical transmission as a major excitatory neurotransmitter. The accumulation of glutamate in secretory vesicles is mediated by vesicular glutamate transporters (VGLUTs) that together with the driving electrochemical gradient of proteins influence the subsequent quantum release of glutamate and the function of higher-order neurons. The vesicular content of glutamate is well correlated with membrane potential (Δψ), which suggests that Δψ determines the vesicular glutamate concentration. The transport of glutamate into secretory vesicles is highly dependent on Cl(-). This anion stimulates glutamate transport but is inhibitory at higher concentrations. Accumulating evidence indicates that Cl(-) regulates glutamate transport through control of VGLUT activity and the H(+) electrochemical gradient. Recently, a comprehensive study demonstrated that Cl(-) regulation of VGLUT is competitively inhibited by metabolic intermediates such as ketone bodies. It also showed that ketone bodies are effective in controlling epilepsy. These results suggest a correlation between metabolic state and higher-order brain function. We propose a novel function for Cl(-) as a fundamental regulator for signal transmission.


Assuntos
Metabolismo Energético , Ácido Glutâmico/química , Ácido Glutâmico/metabolismo , Proteínas Vesiculares de Transporte de Neurotransmissores/química , Proteínas Vesiculares de Transporte de Neurotransmissores/metabolismo , Sequência de Aminoácidos , Humanos , Dados de Sequência Molecular , Transporte Proteico , Transdução de Sinais , Proteínas Vesiculares de Transporte de Glutamato/química , Proteínas Vesiculares de Transporte de Glutamato/metabolismo , Proteínas Vesiculares de Transporte de Glutamato/fisiologia , Proteínas Vesiculares de Transporte de Neurotransmissores/fisiologia
12.
Neuron ; 68(1): 99-112, 2010 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-20920794

RESUMO

Fasting has been used to control epilepsy since antiquity, but the mechanism of coupling between metabolic state and excitatory neurotransmission remains unknown. Previous work has shown that the vesicular glutamate transporters (VGLUTs) required for exocytotic release of glutamate undergo an unusual form of regulation by Cl(-). Using functional reconstitution of the purified VGLUTs into proteoliposomes, we now show that Cl(-) acts as an allosteric activator, and the ketone bodies that increase with fasting inhibit glutamate release by competing with Cl(-) at the site of allosteric regulation. Consistent with these observations, acetoacetate reduced quantal size at hippocampal synapses and suppresses glutamate release and seizures evoked with 4-aminopyridine in the brain. The results indicate an unsuspected link between metabolic state and excitatory neurotransmission through anion-dependent regulation of VGLUT activity.


Assuntos
Ácido Glutâmico/metabolismo , Neurônios/metabolismo , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo , 4-Aminopiridina/farmacologia , Acetoacetatos/farmacologia , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/fisiologia , Comportamento Animal , Células Cultivadas , Cloretos/metabolismo , Cromatografia Líquida de Alta Pressão/métodos , Modelos Animais de Doenças , Dopamina/metabolismo , Relação Dose-Resposta a Droga , Embrião de Mamíferos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Exocitose/efeitos dos fármacos , Exocitose/genética , Regulação da Expressão Gênica , Hipocampo/citologia , Humanos , Técnicas In Vitro , Corpos Cetônicos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Potencial da Membrana Mitocondrial/genética , Camundongos , Camundongos Endogâmicos C57BL , Microdiálise/métodos , Modelos Biológicos , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp/métodos , Bloqueadores dos Canais de Potássio/farmacologia , Ratos , Convulsões/induzido quimicamente , Convulsões/fisiopatologia , Vesículas Sinápticas/metabolismo , Proteína Vesicular 2 de Transporte de Glutamato/química , Proteína Vesicular 2 de Transporte de Glutamato/genética
13.
J Biol Chem ; 284(50): 35073-8, 2009 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-19843525

RESUMO

The vesicular inhibitory amino acid transporter (VIAAT) is a synaptic vesicle protein responsible for the vesicular storage of gamma-aminobutyrate (GABA) and glycine which plays an essential role in GABAergic and glycinergic neurotransmission. The transport mechanism of VIAAT remains largely unknown. Here, we show that proteoliposomes containing purified VIAAT actively took up GABA upon formation of membrane potential (Deltapsi) (positive inside) but not DeltapH. VIAAT-mediated GABA uptake had an absolute requirement for Cl(-) and actually accompanied Cl(-) movement. Kinetic analysis indicated that one GABA molecule and two Cl(-) equivalents were transported during one transport cycle. VIAAT in which Glu(213) was specifically mutated to alanine completely lost the ability to take up both GABA and Cl(-). Essentially the same results were obtained with glycine, another substrate of VIAAT. These results demonstrated that VIAAT is a vesicular Cl(-) transporter that co-transports Cl(-) with GABA or glycine in a Deltapsi dependent manner. It is concluded that Cl(-) plays an essential role in vesicular storage of GABA and glycine.


Assuntos
Cloretos/metabolismo , Simportadores/metabolismo , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Glicina/metabolismo , Ionóforos/metabolismo , Lipossomos/metabolismo , Potenciais da Membrana/fisiologia , Ratos , Simportadores/genética , Valinomicina/metabolismo , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/genética
14.
Proc Natl Acad Sci U S A ; 105(15): 5683-6, 2008 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-18375752

RESUMO

ATP is a major chemical transmitter in purinergic signal transmission. Before secretion, ATP is stored in secretory vesicles found in purinergic cells. Although the presence of active transport mechanisms for ATP has been postulated for a long time, the proteins responsible for its vesicular accumulation remains unknown. The transporter encoded by the human and mouse SLC17A9 gene, a novel member of an anion transporter family, was predominantly expressed in the brain and adrenal gland. The mouse and bovine counterparts were associated with adrenal chromaffin granules. Proteoliposomes containing purified transporter actively took up ATP, ADP, and GTP by using membrane potential as the driving force. The uptake properties of the reconstituted transporter were similar to that of the ATP uptake by synaptic vesicles and chromaffin granules. Suppression of endogenous SLC17A9 expression in PC12 cells decreased exocytosis of ATP. These findings strongly suggest that SLC17A9 protein is a vesicular nucleotide transporter and should lead to the elucidation of the molecular mechanism of ATP secretion in purinergic signal transmission.


Assuntos
Trifosfato de Adenosina/metabolismo , Proteínas de Transporte de Nucleotídeos/metabolismo , Nucleotídeos/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Difosfato de Adenosina , Animais , Exocitose , Guanosina Trifosfato , Humanos , Camundongos , Proteínas de Transporte de Nucleotídeos/isolamento & purificação , Células PC12 , Ratos , Transfecção , Proteínas de Transporte Vesicular/isolamento & purificação
16.
J Biol Chem ; 281(51): 39499-506, 2006 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-17046815

RESUMO

Vesicular glutamate transporters (VGLUTs) are responsible for the vesicular storage of l-glutamate and play an essential role in glutamatergic signal transmission in the central nervous system. The molecular mechanism of the transport remains unknown. Here, we established a novel in vitro assay procedure, which includes purification of wild and mutant VGLUT2 and their reconstitution with purified bacterial F(o)F(1)-ATPase (F-ATPase) into liposomes. Upon the addition of ATP, the proteoliposomes facilitated l-glutamate uptake in a membrane potential (DeltaPsi)-dependent fashion. The ATP-dependent l-glutamate uptake exhibited an absolute requirement for approximately 4 mm Cl(-), was sensitive to Evans blue, but was insensitive to d,l-aspartate. VGLUT2s with mutations in the transmembrane-located residues Arg(184), His(128), and Glu(191) showed a dramatic loss in l-glutamate transport activity, whereas Na(+)-dependent inorganic phosphate (P(i)) uptake remained comparable to that of the wild type. Furthermore, P(i) transport did not require Cl(-) and was not inhibited by Evans blue. Thus, VGLUT2 appears to possess two intrinsic transport machineries that are independent of each other: a DeltaPsi-dependent l-glutamate uptake and a Na(+)-dependent P(i) uptake.


Assuntos
Cloretos/química , Proteína Vesicular 2 de Transporte de Glutamato/fisiologia , Trifosfato de Adenosina/química , Sequência de Aminoácidos , Animais , Arginina/química , Transporte Biológico , Ácido Glutâmico/química , Histidina/química , Insetos , Potenciais da Membrana , Dados de Sequência Molecular , Mutação , Fosfatos/química , Sódio/química , Proteína Vesicular 2 de Transporte de Glutamato/química
17.
EMBO J ; 25(18): 4175-86, 2006 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-16957773

RESUMO

Osteoclasts are involved in the catabolism of the bone matrix and eliminate the resulting degradation products through transcytosis, but the molecular mechanism and regulation of transcytosis remain poorly understood. Upon differentiation, osteoclasts express vesicular glutamate transporter 1 (VGLUT1), which is essential for vesicular storage and subsequent exocytosis of glutamate in neurons. VGLUT1 is localized in transcytotic vesicles and accumulates L-glutamate. Osteoclasts secrete L-glutamate and the bone degradation products upon stimulation with KCl or ATP in a Ca2+-dependent manner. KCl- and ATP-dependent secretion of L-glutamate was absent in osteoclasts prepared from VGLUT1-/- knockout mice. Osteoclasts express mGluR8, a class III metabotropic glutamate receptor. Its stimulation by a specific agonist inhibits secretion of L-glutamate and bone degradation products, whereas its suppression by a specific antagonist stimulates bone resorption. Finally, it was found that VGLUT1-/- mice develop osteoporosis. Thus, in bone-resorbing osteoclasts, L-glutamate and bone degradation products are secreted through transcytosis and the released L-glutamate is involved in autoregulation of transcytosis. Glutamate signaling may play an important role in the bone homeostasis.


Assuntos
Exocitose/fisiologia , Ácido Glutâmico/metabolismo , Osteoclastos/metabolismo , Células 3T3 , Animais , Reabsorção Óssea/metabolismo , Linhagem Celular , Células Cultivadas , Homeostase , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Imunoeletrônica , Modelos Biológicos , Osteoclastos/ultraestrutura , Receptores de Glutamato Metabotrópico/agonistas , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Receptores de Glutamato Metabotrópico/metabolismo , Transdução de Sinais , Proteína Vesicular 1 de Transporte de Glutamato/deficiência , Proteína Vesicular 1 de Transporte de Glutamato/genética
18.
J Neurochem ; 96(2): 550-60, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16336630

RESUMO

Vesicular glutamate transporter (VGLUT) is responsible for the vesicular storage of l-glutamate, and plays an essential role in glutamate-mediated intercellular signal transmission in the CNS and in some neuroendocrine cells. Intestinal L cells are the glucose-responsive neuroendocrine cells responsible for the secretion of glucagon-like peptide 1 (GLP-1). We have shown that intestinal L cells express VGLUT2, a VGLUT isoform, which suggests that L cells secrete L-glutamate. In the present study, we investigated this possibility using GLUTag mouse clonal L cells. RT-PCR and northern blot analyses revealed expression of the VGLUT1 and VGLUT2 genes, but not of the VGLUT3 gene. Western blot analysis revealed immunological counterparts for VGLUT2, whereas an immunological counterpart of VGLUT1 was not detected. Indirect immunofluorescence microscopy revealed a punctate distribution of VGLUT2 immunoreactivity throughout the cells, which co-localized with GLP-1. Double-labeling immunoelectronmicroscopy confirmed the association of VGLUT2 with GLP-1-containing secretory granules. The membrane fraction exhibited ATP-dependent L-glutamate uptake, which was sensitive to bafilomycin A1 (a vacuolar proton ATPase inhibitor) and Evans blue (a VGLUT inhibitor) but insensitive to D,L-aspartate. Upon depolarization with KCl, GLUTag cells secreted appreciable amounts of L-glutamate and GLP-1. D-Glucose and methyl-alpha-D-glucopyranoside, stimulators of exocytosis of GLP-1, also triggered the secretion of L-glutamate. The L-glutamate secretion was partially dependent on Ca2+ and sensitive to bafilomycin A1. These results demonstrated that GLUTag cells stored L-glutamate in secretory granules and secreted it with GLP-1 by exocytosis. As GLUTag cells and intestinal L cells express kainate receptors and plasma membrane glutamate transporters, these results support the concept of L-glutamate-mediated intercellular signaling in the vicinity of intestinal L cells.


Assuntos
Peptídeo 1 Semelhante ao Glucagon/metabolismo , Ácido Glutâmico/metabolismo , Intestino Delgado/metabolismo , Vesículas Secretórias/metabolismo , Animais , Células Clonais , Exocitose , Ácido Glutâmico/farmacocinética , Intestino Delgado/citologia , Camundongos , Transdução de Sinais , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...