Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 591(7849): 327-331, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33597752

RESUMEN

Glutamate is the most abundant excitatory neurotransmitter in the central nervous system, and its precise control is vital to maintain normal brain function and to prevent excitotoxicity1. The removal of extracellular glutamate is achieved by plasma-membrane-bound transporters, which couple glutamate transport to sodium, potassium and pH gradients using an elevator mechanism2-5. Glutamate transporters also conduct chloride ions by means of a channel-like process that is thermodynamically uncoupled from transport6-8. However, the molecular mechanisms that enable these dual-function transporters to carry out two seemingly contradictory roles are unknown. Here we report the cryo-electron microscopy structure of a glutamate transporter homologue in an open-channel state, which reveals an aqueous cavity that is formed during the glutamate transport cycle. The functional properties of this cavity, combined with molecular dynamics simulations, reveal it to be an aqueous-accessible chloride permeation pathway that is gated by two hydrophobic regions and is conserved across mammalian and archaeal glutamate transporters. Our findings provide insight into the mechanism by which glutamate transporters support their dual function, and add information that will assist in mapping the complete transport cycle shared by the solute carrier 1A transporter family.


Asunto(s)
Sistema de Transporte de Aminoácidos X-AG/química , Sistema de Transporte de Aminoácidos X-AG/metabolismo , Canales de Cloruro/química , Canales de Cloruro/metabolismo , Interacciones Hidrofóbicas e Hidrofílicas , Sistema de Transporte de Aminoácidos X-AG/genética , Sistema de Transporte de Aminoácidos X-AG/ultraestructura , Animales , Encéfalo/metabolismo , Canales de Cloruro/genética , Canales de Cloruro/ultraestructura , Cloruros/metabolismo , Microscopía por Crioelectrón , Cristalografía por Rayos X , Transportador 1 de Aminoácidos Excitadores/química , Transportador 1 de Aminoácidos Excitadores/genética , Transportador 1 de Aminoácidos Excitadores/metabolismo , Transportador 1 de Aminoácidos Excitadores/ultraestructura , Femenino , Ácido Glutámico/metabolismo , Humanos , Modelos Moleculares , Mutación , Oocitos , Conformación Proteica , Xenopus laevis
2.
J Biol Chem ; 298(8): 102178, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35752361

RESUMEN

The solute carrier 1A family comprises a group of membrane proteins that act as dual-function amino acid transporters and chloride (Cl-) channels and includes the alanine serine cysteine transporters (ASCTs) as well as the excitatory amino acid transporters. ASCT2 is regarded as a promising target for cancer therapy, as it can transport glutamine and other neutral amino acids into cells and is upregulated in a range of solid tumors. The compound L-γ-glutamyl-p-nitroanilide (GPNA) is widely used in studies probing the role of ASCT2 in cancer biology; however, the mechanism by which GPNA inhibits ASCT2 is not entirely clear. Here, we used electrophysiology and radiolabelled flux assays to demonstrate that GPNA activates the Cl- conductance of ASCT2 to the same extent as a transported substrate, whilst not undergoing the full transport cycle. This is a previously unreported phenomenon for inhibitors of the solute carrier 1A family but corroborates a body of literature suggesting that the structural requirements for transport are distinct from those for Cl- channel formation. We also show that in addition to its currently known targets, GPNA inhibits several of the excitatory amino acid transporters. Together, these findings raise questions about the true mechanisms of its anticancer effects.


Asunto(s)
Aminoácidos Neutros , Neoplasias , Sistema de Transporte de Aminoácidos ASC/genética , Sistema de Transporte de Aminoácidos ASC/metabolismo , Sistemas de Transporte de Aminoácidos , Glutamina/metabolismo , Humanos , Antígenos de Histocompatibilidad Menor/genética , Antígenos de Histocompatibilidad Menor/metabolismo , Neoplasias/metabolismo
3.
Physiol Rev ; 93(4): 1621-57, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24137018

RESUMEN

L-Glutamate is the predominant excitatory neurotransmitter in the mammalian central nervous system and plays important roles in a wide variety of brain functions, but it is also a key player in the pathogenesis of many neurological disorders. The control of glutamate concentrations is critical to the normal functioning of the central nervous system, and in this review we discuss how glutamate transporters regulate glutamate concentrations to maintain dynamic signaling mechanisms between neurons. In 2004, the crystal structure of a prokaryotic homolog of the mammalian glutamate transporter family of proteins was crystallized and its structure determined. This has paved the way for a better understanding of the structural basis for glutamate transporter function. In this review we provide a broad perspective of this field of research, but focus primarily on the more recent studies with a particular emphasis on how our understanding of the structure of glutamate transporters has generated new insights.


Asunto(s)
Proteínas de Transporte de Glutamato en la Membrana Plasmática/fisiología , Glutamatos/metabolismo , Proteínas de Transporte Vesicular de Glutamato/fisiología , Secuencia de Aminoácidos , Animales , Transporte Biológico/fisiología , Sistema Nervioso Central/fisiología , Proteínas de Transporte de Glutamato en la Membrana Plasmática/análisis , Proteínas de Transporte de Glutamato en la Membrana Plasmática/química , Humanos , Datos de Secuencia Molecular , Transducción de Señal/fisiología , Proteínas de Transporte Vesicular de Glutamato/análisis , Proteínas de Transporte Vesicular de Glutamato/química
4.
Bioorg Med Chem ; 26(22): 5852-5869, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30401502

RESUMEN

Dual-specificity tyrosine phosphorylation-related kinase 1A (DYRK1A) is a dual-specificity protein kinase that catalyses phosphorylation and autophosphorylation. Higher DYRK1A expression correlates with cancer, in particular glioblastoma present within the brain. We report here the synthesis and biological evaluation of new heterocyclic diphenolic derivatives designed as novel DYRK1A inhibitors. The generation of these heterocycles such as benzimidazole, imidazole, naphthyridine, pyrazole-pyridines, bipyridine, and triazolopyrazines was made based on the structural modification of the lead DANDY and tested for their ability to inhibit DYRK1A. None of these derivatives showed significant DYRK1A inhibition but provide valuable knowledge around the importance of the 7-azaindole moiety. These data will be of use for developing further structure-activity relationship studies to improve the selective inhibition of DYRK1A.


Asunto(s)
Compuestos Heterocíclicos/farmacología , Fenoles/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Compuestos Heterocíclicos/síntesis química , Compuestos Heterocíclicos/química , Humanos , Estructura Molecular , Fenoles/síntesis química , Fenoles/química , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Proteínas Serina-Treonina Quinasas/aislamiento & purificación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/aislamiento & purificación , Proteínas Tirosina Quinasas/metabolismo , Relación Estructura-Actividad , Quinasas DyrK
5.
Biochemistry ; 55(49): 6801-6810, 2016 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-27951659

RESUMEN

Crystal structures of the prokaryotic aspartate transporter, GltPh, have provided important insights into the mechanism of amino acid transport by GltPh and related eukaryotic members of the glutamate transporter family (SLC1A family). Identification of inhibitors of GltPh can provide valuable tools for understanding the molecular basis for substrate and inhibitor specificity and selectivity of SLC1A members, but at present, few inhibitors of GltPh have been identified. We have screened a collection of commercially available aspartate analogues and identified new transportable and nontransportable GltPh inhibitors. We have explored the inhibition profile of GltPh by utilizing a thiol modification assay that isolates sided populations of the transporters reconstituted in liposomes to determine if any aspartate analogues display a preference for either the inwardly or outwardly directed binding sites. Here, we have characterized several new inhibitors of GltPh and identified three ß-carbon-substituted molecules that display a strong preference for the outwardly directed binding site of GltPh.


Asunto(s)
Sistema de Transporte de Aminoácidos X-AG/metabolismo , Ácido Aspártico/metabolismo , Sistema de Transporte de Aminoácidos X-AG/química , Sitios de Unión
6.
J Biol Chem ; 290(15): 9780-8, 2015 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-25713135

RESUMEN

The aspartate transporter from Pyrococcus horikoshii (GltPh) is a model for the structure of the SLC1 family of amino acid transporters. Crystal structures of GltPh provide insight into mechanisms of ion coupling and substrate transport; however, structures have been solved in the absence of a lipid bilayer so they provide limited information regarding interactions that occur between the protein and lipids of the membrane. Here, we investigated the effect of the lipid environment on aspartate transport by reconstituting GltPh into liposomes of defined lipid composition where the primary lipid is phosphatidylethanolamine (PE) or its methyl derivatives. We showed that the rate of aspartate transport and the transmembrane orientation of GltPh were influenced by the primary lipid in the liposomes. In PE liposomes, we observed the highest transport rate and showed that 85% of the transporters were orientated right-side out, whereas in trimethyl PE liposomes, 50% of transporters were right-side out, and we observed a 4-fold reduction in transport rate. Differences in orientation can only partially explain the lipid composition effect on transport rate. Crystal structures of GltPh revealed a tyrosine residue (Tyr-33) that we propose interacts with lipid headgroups during the transport cycle. Based on site-directed mutagenesis, we propose that a cation-π interaction between Tyr-33 and the lipid headgroups can influence conformational flexibility of the trimerization domain and thus the rate of transport. These results provide a specific example of how interactions between membrane lipids and membrane-bound proteins can influence function and highlight the importance of the role of the membrane in transporter function.


Asunto(s)
Sistema de Transporte de Aminoácidos X-AG/genética , Proteínas Arqueales/metabolismo , Membrana Dobles de Lípidos/metabolismo , Pyrococcus horikoshii/metabolismo , Sistema de Transporte de Aminoácidos X-AG/química , Sistema de Transporte de Aminoácidos X-AG/metabolismo , Proteínas Arqueales/química , Proteínas Arqueales/genética , Ácido Aspártico/metabolismo , Transporte Biológico , Cristalografía por Rayos X , Cinética , Membrana Dobles de Lípidos/química , Liposomas/química , Liposomas/metabolismo , Modelos Moleculares , Mutación , Fosfatidiletanolaminas/química , Fosfatidiletanolaminas/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Pyrococcus horikoshii/genética , Tirosina/química , Tirosina/genética , Tirosina/metabolismo
7.
Neurochem Res ; 41(3): 593-9, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26303507

RESUMEN

Transporters and ion channels are conventionally categorised into distinct classes of membrane proteins. However, some membrane proteins have a split personality and can function as both transporters and ion channels. The excitatory amino acid transporters (EAATs) in particular, function as both glutamate transporters and chloride (Cl(-)) channels. The EAATs couple the transport of glutamate to the co-transport of three Na(+) ions and one H(+) ion into the cell, and the counter-transport of one K(+) ion out of the cell. The EAAT Cl(-) channel is activated by the binding of glutamate and Na(+), but is thermodynamically uncoupled from glutamate transport and involves molecular determinants distinct from those responsible for glutamate transport. Several crystal structures of an EAAT archaeal homologue, GltPh, at different stages of the transport cycle, alongside numerous functional studies and molecular dynamics simulations, have provided extensive insights into the mechanism of substrate transport via these transporters. However, the molecular determinants involved in Cl(-) permeation, and the mechanism by which this channel is activated are not entirely understood. Here we will discuss what is currently known about the molecular determinants involved in EAAT-mediated Cl(-) permeation and the mechanisms that underlie their split personality.


Asunto(s)
Sistema de Transporte de Aminoácidos X-AG/metabolismo , Canales de Cloruro/metabolismo , Sistema de Transporte de Aminoácidos X-AG/química , Animales , Proteínas Arqueales/química , Proteínas Arqueales/metabolismo , Canales de Cloruro/química , Simulación de Dinámica Molecular , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína
8.
J Biol Chem ; 289(25): 17468-79, 2014 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-24808181

RESUMEN

The alanine, serine, cysteine transporters (ASCTs) belong to the solute carrier family 1A (SLC1A), which also includes the excitatory amino acid transporters (EAATs) and the prokaryotic aspartate transporter GltPh. Acidic amino acid transport by the EAATs is coupled to the co-transport of three Na(+) ions and one proton, and the counter-transport of one K(+) ion. In contrast, neutral amino acid exchange by the ASCTs does not require protons or the counter-transport of K(+) ions and the number of Na(+) ions required is not well established. One property common to SLC1A family members is a substrate-activated anion conductance. We have investigated the number and location of Na(+) ions required by ASCT1 by mutating residues in ASCT1 that correspond to residues in the EAATs and GltPh that are involved in Na(+) binding. Mutations to all three proposed Na(+) sites influence the binding of substrate and/or Na(+), or the rate of substrate exchange. A G422S mutation near the Na2 site reduced Na(+) affinity, without affecting the rate of exchange. D467T and D467A mutations in the Na1 site reduce Na(+) and substrate affinity and also the rate of substrate exchange. T124A and D380A mutations in the Na3 site selectively reduce the affinity for Na(+) and the rate of substrate exchange without affecting substrate affinity. In many of the mutants that reduce the rate of substrate transport the amplitudes of the substrate-activated anion conductances are not substantially affected indicating altered ion dependence for channel activation compared with substrate exchange.


Asunto(s)
Sistema de Transporte de Aminoácidos ASC/química , Sodio/química , Sustitución de Aminoácidos , Sistema de Transporte de Aminoácidos ASC/genética , Sistema de Transporte de Aminoácidos ASC/metabolismo , Sitios de Unión , Cationes Monovalentes/química , Cationes Monovalentes/metabolismo , Humanos , Transporte Iónico/fisiología , Mutación Missense , Sodio/metabolismo
9.
Biophys J ; 107(3): 621-629, 2014 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-25099801

RESUMEN

The concentration of glutamate within the glutamatergic synapse is tightly regulated by the excitatory amino-acid transporters (EAATs). In addition to their primary role of clearing extracellular glutamate, the EAATs also possess a thermodynamically uncoupled Cl(-) conductance. Several crystal structures of an archaeal EAAT homolog, GltPh, at different stages of the transport cycle have been solved. In a recent structure, an aqueous cavity located at the interface of the transport and trimerization domains has been identified. This cavity is lined by polar residues, several of which have been implicated in Cl(-) permeation. We hypothesize that this cavity opens during the transport cycle to form the Cl(-) channel. Residues lining this cavity in EAAT1, including Ser-366, Leu-369, Phe-373, Arg-388, Pro-392, and Thr-396, were mutated to small hydrophobic residues. Wild-type and mutant transporters were expressed in Xenopus laevis oocytes and two-electrode voltage-clamp electrophysiology, and radiolabeled substrate uptake was used to investigate function. Significant alterations in substrate-activated Cl(-) conductance were observed for several mutant transporters. These alterations support the hypothesis that this aqueous cavity at the interface of the transport and trimerization domains is a partially formed Cl(-) channel, which opens to form a pore through which Cl(-) ions pass. This study enhances our understanding as to how glutamate transporters function as both amino-acid transporters and Cl(-) channels.


Asunto(s)
Cloruros/metabolismo , Transportador 1 de Aminoácidos Excitadores/química , Secuencia de Aminoácidos , Animales , Transportador 1 de Aminoácidos Excitadores/metabolismo , Humanos , Transporte Iónico , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Xenopus
10.
J Biol Chem ; 288(12): 8250-8257, 2013 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-23393130

RESUMEN

The ASCTs (alanine, serine, and cysteine transporters) belong to the solute carrier family 1 (SLC1), which also includes the human glutamate transporters (excitatory amino acid transporters, EAATs) and the prokaryotic aspartate transporter GltPh. Despite the high degree of amino acid sequence identity between family members, ASCTs function quite differently from the EAATs and GltPh. The aim of this study was to mutate ASCT1 to generate a transporter with functional properties of the EAATs and GltPh, to further our understanding of the structural basis for the different transport mechanisms of the SLC1 family. We have identified three key residues involved in determining differences between ASCT1, the EAATs and GltPh. ASCT1 transporters containing the mutations A382T, T459R, and Q386E were expressed in Xenopus laevis oocytes, and their transport and anion channel functions were investigated. A382T and T459R altered the substrate selectivity of ASCT1 to allow the transport of acidic amino acids, particularly l-aspartate. The combination of A382T and T459R within ASCT1 generates a transporter with a similar profile to that of GltPh, with preference for l-aspartate over l-glutamate. Interestingly, the amplitude of the anion conductance activated by the acidic amino acids does not correlate with rates of transport, highlighting the distinction between these two processes. Q386E impaired the ability of ASCT1 to bind acidic amino acids at pH 5.5; however, this was reversed by the additional mutation A382T. We propose that these residues differences in TM7 and TM8 combine to determine differences in substrate selectivity between members of the SLC1 family.


Asunto(s)
Sistema de Transporte de Aminoácidos ASC/metabolismo , Proteínas Arqueales/metabolismo , Proteínas de Transporte de Glutamato en la Membrana Plasmática/metabolismo , Sustitución de Aminoácidos , Sistema de Transporte de Aminoácidos ASC/genética , Sistema de Transporte de Aminoácidos ASC/fisiología , Animales , Proteínas Arqueales/fisiología , Ácido Aspártico/metabolismo , Ácido Aspártico/fisiología , Sitios de Unión , Transporte Biológico , Células Cultivadas , Proteínas de Transporte de Glutamato en la Membrana Plasmática/fisiología , Ácido Glutámico/metabolismo , Ácido Glutámico/fisiología , Humanos , Concentración de Iones de Hidrógeno , Cinética , Potenciales de la Membrana , Mutagénesis Sitio-Dirigida , Serina/metabolismo , Serina/fisiología , Especificidad por Sustrato , Xenopus laevis
11.
Int J Cancer ; 135(5): 1060-71, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24531984

RESUMEN

Amino acids, especially leucine and glutamine, are important for tumor cell growth, survival and metabolism. A range of different transporters deliver each specific amino acid into cells, some of which are increased in cancer. These amino acids consequently activate the mTORC1 pathway and drive cell cycle progression. The leucine transporter LAT1/4F2hc heterodimer assembles as part of a large complex with the glutamine transporter ASCT2 to transport amino acids. In this study, we show that the expression of LAT1 and ASCT2 is significantly increased in human melanoma samples and is present in both BRAF(WT) (C8161 and WM852) and BRAF(V600E) mutant (1205Lu and 451Lu) melanoma cell lines. While inhibition of LAT1 by BCH did not suppress melanoma cell growth, the ASCT2 inhibitor BenSer significantly reduced both leucine and glutamine transport in melanoma cells, leading to inhibition of mTORC1 signaling. Cell proliferation and cell cycle progression were significantly reduced in the presence of BenSer in melanoma cells in 2D and 3D cell culture. This included reduced expression of the cell cycle regulators CDK1 and UBE2C. The importance of ASCT2 expression in melanoma was confirmed by shRNA knockdown, which inhibited glutamine uptake, mTORC1 signaling and cell proliferation. Taken together, our study demonstrates that ASCT2-mediated glutamine transport is a potential therapeutic target for both BRAF(WT) and BRAF(V600E) melanoma.


Asunto(s)
Sistema de Transporte de Aminoácidos ASC/biosíntesis , Glutamina/metabolismo , Transportador de Aminoácidos Neutros Grandes 1/biosíntesis , Melanoma/patología , Complejos Multiproteicos/antagonistas & inhibidores , Neoplasias Cutáneas/patología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Sistema de Transporte de Aminoácidos ASC/antagonistas & inhibidores , Sistema de Transporte de Aminoácidos ASC/genética , Aminoácidos Cíclicos/farmacología , Compuestos de Bencilo/farmacología , Transporte Biológico , Proteína Quinasa CDC2/biosíntesis , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/genética , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular , Humanos , Leucina/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Melanoma/metabolismo , Antígenos de Histocompatibilidad Menor , Complejos Multiproteicos/genética , Proteínas Proto-Oncogénicas B-raf/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Serina/análogos & derivados , Serina/farmacología , Transducción de Señal , Neoplasias Cutáneas/metabolismo , Esferoides Celulares , Serina-Treonina Quinasas TOR/genética , Células Tumorales Cultivadas , Enzimas Ubiquitina-Conjugadoras/biosíntesis
12.
Nature ; 445(7126): 387-93, 2007 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-17230192

RESUMEN

Secondary transporters are integral membrane proteins that catalyse the movement of substrate molecules across the lipid bilayer by coupling substrate transport to one or more ion gradients, thereby providing a mechanism for the concentrative uptake of substrates. Here we describe crystallographic and thermodynamic studies of Glt(Ph), a sodium (Na+)-coupled aspartate transporter, defining sites for aspartate, two sodium ions and d,l-threo-beta-benzyloxyaspartate, an inhibitor. We further show that helical hairpin 2 is the extracellular gate that controls access of substrate and ions to the internal binding sites. At least two sodium ions bind in close proximity to the substrate and these sodium-binding sites, together with the sodium-binding sites in another sodium-coupled transporter, LeuT, define an unwound alpha-helix as the central element of the ion-binding motif, a motif well suited to the binding of sodium and to participation in conformational changes that accompany ion binding and unbinding during the transport cycle.


Asunto(s)
Sistemas de Transporte de Aminoácidos/metabolismo , Ácido Aspártico/metabolismo , Pyrococcus horikoshii/química , Sodio/metabolismo , Sodio/farmacología , Secuencias de Aminoácidos , Sistemas de Transporte de Aminoácidos/antagonistas & inhibidores , Sistemas de Transporte de Aminoácidos/química , Sistemas de Transporte de Aminoácidos/genética , Ácido Aspártico/farmacología , Sitios de Unión , Cristalografía por Rayos X , Ligandos , Modelos Moleculares , Mutación/genética , Conformación Proteica , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Especificidad por Sustrato , Termodinámica
13.
Nat Commun ; 14(1): 6374, 2023 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-37821493

RESUMEN

Organic Cation Transporter 1 (OCT1) plays a crucial role in hepatic metabolism by mediating the uptake of a range of metabolites and drugs. Genetic variations can alter the efficacy and safety of compounds transported by OCT1, such as those used for cardiovascular, oncological, and psychological indications. Despite its importance in drug pharmacokinetics, the substrate selectivity and underlying structural mechanisms of OCT1 remain poorly understood. Here, we present cryo-EM structures of full-length human OCT1 in the inward-open conformation, both ligand-free and drug-bound, indicating the basis for its broad substrate recognition. Comparison of our structures with those of outward-open OCTs provides molecular insight into the alternating access mechanism of OCTs. We observe that hydrophobic gates stabilize the inward-facing conformation, whereas charge neutralization in the binding pocket facilitates the release of cationic substrates. These findings provide a framework for understanding the structural basis of the promiscuity of drug binding and substrate translocation in OCT1.


Asunto(s)
Proteínas de Transporte de Catión Orgánico , Transportador 1 de Catión Orgánico , Humanos , Transportador 1 de Catión Orgánico/genética , Transportador 1 de Catión Orgánico/química , Transportador 1 de Catión Orgánico/metabolismo , Proteínas de Transporte de Catión Orgánico/química , Transporte Biológico , Transportador 2 de Cátion Orgánico/metabolismo
14.
Nat Struct Mol Biol ; 14(5): 365-71, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17435767

RESUMEN

Glutamate transporters (EAATs) are pivotal in mammalian synaptic transmission, tightly regulating synaptic levels of this excitatory neurotransmitter. In addition to coupled glutamate transport, the EAATs also show an uncoupled Cl(-) conductance, whose physiological importance has recently been demonstrated. Little is yet known about the molecular mechanism of chloride permeation. Here we show that Glt(Ph), a bacterial EAAT homolog whose structure has been determined, displays an uncoupled Cl(-) conductance that can determine the rate of substrate uptake. A mutation analogous to one known to specifically affect Cl(-) movement in EAAT1 has similar effects on Glt(Ph), suggesting that this protein is an excellent structural model for understanding Cl(-) permeation through the EAATs. We also observed an uncoupled Cl(-) conductance in another bacterial EAAT homolog but not in a homolog of the Na(+)/Cl(-)-coupled neurotransmitter transporters.


Asunto(s)
Cloro/metabolismo , Proteínas de Transporte de Glutamato en la Membrana Plasmática/metabolismo , Ácido Aspártico/metabolismo , Proteínas Bacterianas , Evolución Molecular , Proteínas de Transporte de Glutamato en la Membrana Plasmática/genética , Transporte Iónico , Cinética , Mutación , Permeabilidad
15.
Biochem J ; 439(2): 333-40, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-21732909

RESUMEN

Glutamate transport is coupled to the co-transport of 3 Na(+) and 1 H(+) followed by the counter-transport of 1 K(+). In addition, glutamate and Na(+) binding to glutamate transporters generates an uncoupled anion conductance. The human glial glutamate transporter EAAT1 (excitatory amino acid transporter 1) also allows significant passive and active water transport, which suggests that water permeation through glutamate transporters may play an important role in glial cell homoeostasis. Urea also permeates EAAT1 and has been used to characterize the permeation properties of the transporter. We have previously identified a series of mutations that differentially affect either the glutamate transport process or the substrate-activated channel function of EAAT1. The water and urea permeation properties of wild-type EAAT1 and two mutant transporters were measured to identify which permeation pathway facilitates the movement of these molecules. We demonstrate that there is a significant rate of L-glutamate-stimulated passive and active water transport. Both the passive and active L-glutamate-stimulated water transport is most closely associated with the glutamate transport process. In contrast, L-glutamate-stimulated [(14)C]urea permeation is associated with the anion channel of the transporter. However, there is also likely to be a transporter-specific, but glutamate independent, flux of water via the anion channel.


Asunto(s)
Transportador 1 de Aminoácidos Excitadores/metabolismo , Urea/metabolismo , Agua/metabolismo , Animales , Transporte Biológico , Humanos , Permeabilidad , Xenopus laevis
16.
Curr Opin Struct Biol ; 75: 102405, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35709614

RESUMEN

Glutamate transporters facilitate the removal of this excitatory neurotransmitter from the synapse. Increasing evidence indicates that this process is linked to intrinsic chloride channel activity that is thermodynamically uncoupled from substrate transport. A recent cryo-EM structure of GltPh - an archaeal homolog of the glutamate transporters - in an open channel state has shed light on the structural basis for channel opening formed at the interface of two domains within the transporter which is gated by two clusters of hydrophobic residues. These transporters cycle through several conformational states during the transport process, including the chloride conducting state, which appears to be stabilised by protein-membrane interactions and membrane deformation. Several point mutations that perturb the chloride conductance can have detrimental effects and are linked to the pathogenesis of the neurological disorder, episodic ataxia type 6.


Asunto(s)
Sistema de Transporte de Aminoácidos X-AG , Cloruros , Sistema de Transporte de Aminoácidos X-AG/química , Sistema de Transporte de Aminoácidos X-AG/metabolismo , Transporte Biológico , Glutamatos , Transporte Iónico
17.
ACS Chem Biol ; 17(2): 426-437, 2022 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-35015506

RESUMEN

Biosynthesis of the hydroxamic acid siderophore desferrioxamine D1 (DFOD1, 6), which is the N-acetylated analogue of desferrioxamine B (DFOB, 5), has been delineated. Enzyme-independent Ac-CoA-mediated N-acetylation of 5 produced 6, in addition to three constitutional isomers containing an N-O-acetyl group installed at either one of the three hydroxamic acid groups of 5. The formation of N-Ac-DFOB (DFOD1, 6) and the composite of N-O-acetylated isomers N-O-Ac-DFOB[001] (6a), N-O-Ac-DFOB[010] (6b), and N-O-Ac-DFOB[100] (6c) (defined as the N-O-Ac motif positioned within the terminal amine, internal, or N-acetylated region of 5, respectively), was pH-dependent, with 6a-6c dominant at pH < 8.5 and 6 dominant at pH > 8.5. The trend in the pH dependence was consistent with the pKa values of the NH3+ (pKa ∼ 10) and N-OH (pKa ∼ 8.5-9) groups in 5. The N- and N-O-acetyl motifs can be conceived as a post-biosynthetic modification (PBM) of a nonproteinaceous secondary metabolite, akin to a post-translational modification (PTM) of a protein. The pH-labile N-O-acetyl group could act as a reversible switch to modulate the properties and functions of secondary metabolites, including hydroxamic acid siderophores. An alternative (most likely minor) biosynthetic pathway for 6 showed that the nonribosomal peptide synthetase-independent siderophore synthetase DesD was competent in condensing N'-acetyl-N-succinyl-N-hydroxy-1,5-diaminopentane (N'-Ac-SHDP, 7) with the dimeric hydroxamic acid precursor (AHDP-SHDP, 4) native to 5 biosynthesis to generate 6. The strategy of diversifying protein structure and function using PTMs could be paralleled in secondary metabolites with the use of PBMs.


Asunto(s)
Deferoxamina , Sideróforos , Acetilcoenzima A/metabolismo , Vías Biosintéticas , Deferoxamina/metabolismo , Concentración de Iones de Hidrógeno , Sideróforos/metabolismo
18.
J Clin Invest ; 132(7)2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35167492

RESUMEN

Glutamate is the predominant excitatory neurotransmitter in the mammalian central nervous system (CNS). Excitatory amino acid transporters (EAATs) regulate extracellular glutamate by transporting it into cells, mostly glia, to terminate neurotransmission and to avoid neurotoxicity. EAATs are also chloride (Cl-) channels, but the physiological role of Cl- conductance through EAATs is poorly understood. Mutations of human EAAT1 (hEAAT1) have been identified in patients with episodic ataxia type 6 (EA6). One mutation showed increased Cl- channel activity and decreased glutamate transport, but the relative contributions of each function of hEAAT1 to mechanisms underlying the pathology of EA6 remain unclear. Here we investigated the effects of 5 additional EA6-related mutations on hEAAT1 function in Xenopus laevis oocytes, and on CNS function in a Drosophila melanogaster model of locomotor behavior. Our results indicate that mutations resulting in decreased hEAAT1 Cl- channel activity but with functional glutamate transport can also contribute to the pathology of EA6, highlighting the importance of Cl- homeostasis in glial cells for proper CNS function. We also identified what we believe is a novel mechanism involving an ectopic sodium (Na+) leak conductance in glial cells. Together, these results strongly support the idea that EA6 is primarily an ion channelopathy of CNS glia.


Asunto(s)
Ataxia , Drosophila melanogaster , Animales , Ataxia/genética , Ataxia/metabolismo , Canales de Cloruro/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Transportador 1 de Aminoácidos Excitadores , Ácido Glutámico/genética , Ácido Glutámico/metabolismo , Humanos , Mamíferos/metabolismo , Mutación , Neuroglía/metabolismo
19.
Front Cell Neurosci ; 15: 670346, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33927596

RESUMEN

Neurotransmitter transporters limit spillover between synapses and maintain the extracellular neurotransmitter concentration at low yet physiologically meaningful levels. They also exert a key role in providing precursors for neurotransmitter biosynthesis. In many cases, neurons and astrocytes contain a large intracellular pool of transporters that can be redistributed and stabilized in the plasma membrane following activation of different signaling pathways. This means that the uptake capacity of the brain neuropil for different neurotransmitters can be dynamically regulated over the course of minutes, as an indirect consequence of changes in neuronal activity, blood flow, cell-to-cell interactions, etc. Here we discuss recent advances in the mechanisms that control the cell membrane trafficking and biophysical properties of transporters for the excitatory, inhibitory and modulatory neurotransmitters glutamate, GABA, and dopamine.

20.
J Biol Chem ; 284(52): 36424-36430, 2009 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-19875446

RESUMEN

Concentrations of extracellular glycine in the central nervous system are regulated by Na(+)/Cl(-)-dependent glycine transporters, GLYT1 and GLYT2. N-Arachidonylglycine (NAGly) is an endogenous inhibitor of GLYT2 with little or no effect on GLYT1 and is analgesic in rat models of neuropathic and inflammatory pain. Understanding the molecular basis of NAGly interactions with GLYT2 may allow for the development of novel therapeutics. In this study, chimeric transporters were used to determine the structural basis for differences in NAGly sensitivity between GLYT1 and GLYT2 and also the actions of a series of related N-arachidonyl amino acids. Extracellular loops 2 and 4 of GLYT2 are important in the selective inhibition of GLYT2 by NAGly and by the related compounds N-arachidonyl-gamma-aminobutyric acid and N-arachidonyl-d-alanine, whereas only the extracellular loop 4 of GLYT2 is required for N-arachidonyl-l-alanine inhibition of transport. These observations suggest that the structure of the head group of these compounds is important in determining how they interact with extracellular loops 2 and 4 of GLYT2. Site-directed mutagenesis of GLYT2 EL4 residues was used to identify the key residues Arg(531), Lys(532), and Ile(545) that contribute to the differences in NAGly sensitivity.


Asunto(s)
Ácidos Araquidónicos/farmacología , Proteínas de Transporte de Glicina en la Membrana Plasmática/metabolismo , Glicina/análogos & derivados , Glicina/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Transporte Biológico/genética , Glicina/genética , Glicina/farmacología , Proteínas de Transporte de Glicina en la Membrana Plasmática/genética , Mutagénesis Sitio-Dirigida , Dolor/genética , Dolor/metabolismo , Estructura Secundaria de Proteína/genética , Ratas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Xenopus laevis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA