Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 186(10): 2160-2175.e17, 2023 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-37137306

RESUMEN

The serotonin transporter (SERT) removes synaptic serotonin and is the target of anti-depressant drugs. SERT adopts three conformations: outward-open, occluded, and inward-open. All known inhibitors target the outward-open state except ibogaine, which has unusual anti-depressant and substance-withdrawal effects, and stabilizes the inward-open conformation. Unfortunately, ibogaine's promiscuity and cardiotoxicity limit the understanding of inward-open state ligands. We docked over 200 million small molecules against the inward-open state of the SERT. Thirty-six top-ranking compounds were synthesized, and thirteen inhibited; further structure-based optimization led to the selection of two potent (low nanomolar) inhibitors. These stabilized an outward-closed state of the SERT with little activity against common off-targets. A cryo-EM structure of one of these bound to the SERT confirmed the predicted geometry. In mouse behavioral assays, both compounds had anxiolytic- and anti-depressant-like activity, with potencies up to 200-fold better than fluoxetine (Prozac), and one substantially reversed morphine withdrawal effects.


Asunto(s)
Ibogaína , Inhibidores Selectivos de la Recaptación de Serotonina , Proteínas de Transporte de Serotonina en la Membrana Plasmática , Bibliotecas de Moléculas Pequeñas , Animales , Ratones , Fluoxetina/farmacología , Ibogaína/química , Ibogaína/farmacología , Conformación Molecular , Serotonina/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/química , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/ultraestructura , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología
2.
Cell ; 183(7): 1986-2002.e26, 2020 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-33333022

RESUMEN

Serotonin plays a central role in cognition and is the target of most pharmaceuticals for psychiatric disorders. Existing drugs have limited efficacy; creation of improved versions will require better understanding of serotonergic circuitry, which has been hampered by our inability to monitor serotonin release and transport with high spatial and temporal resolution. We developed and applied a binding-pocket redesign strategy, guided by machine learning, to create a high-performance, soluble, fluorescent serotonin sensor (iSeroSnFR), enabling optical detection of millisecond-scale serotonin transients. We demonstrate that iSeroSnFR can be used to detect serotonin release in freely behaving mice during fear conditioning, social interaction, and sleep/wake transitions. We also developed a robust assay of serotonin transporter function and modulation by drugs. We expect that both machine-learning-guided binding-pocket redesign and iSeroSnFR will have broad utility for the development of other sensors and in vitro and in vivo serotonin detection, respectively.


Asunto(s)
Evolución Molecular Dirigida , Aprendizaje Automático , Serotonina/metabolismo , Algoritmos , Secuencia de Aminoácidos , Amígdala del Cerebelo/fisiología , Animales , Conducta Animal , Sitios de Unión , Encéfalo/metabolismo , Células HEK293 , Humanos , Cinética , Modelos Lineales , Ratones , Ratones Endogámicos C57BL , Fotones , Unión Proteica , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Sueño/fisiología , Vigilia/fisiología
3.
Proc Natl Acad Sci U S A ; 121(18): e2319384121, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38652746

RESUMEN

Clearance of serotonin (5-hydroxytryptamine, 5-HT) from the synaptic cleft after neuronal signaling is mediated by serotonin transporter (SERT), which couples this process to the movement of a Na+ ion down its chemical gradient. After release of 5-HT and Na+ into the cytoplasm, the transporter faces a rate-limiting challenge of resetting its conformation to be primed again for 5-HT and Na+ binding. Early studies of vesicles containing native SERT revealed that K+ gradients can provide an additional driving force, via K+ antiport. Moreover, under appropriate conditions, a H+ ion can replace K+. Intracellular K+ accelerates the resetting step. Structural studies of SERT have identified two binding sites for Na+ ions, but the K+ site remains enigmatic. Here, we show that K+ antiport can drive substrate accumulation into vesicles containing SERT extracted from a heterologous expression system, allowing us to study the residues responsible for K+ binding. To identify candidate binding residues, we examine many cation binding configurations using molecular dynamics simulations, predicting that K+ binds to the so-called Na2 site. Site-directed mutagenesis of residues in this site can eliminate the ability of both K+ and H+ to drive 5-HT accumulation into vesicles and, in patch clamp recordings, prevent the acceleration of turnover rates and the formation of a channel-like state by K+ or H+. In conclusion, the Na2 site plays a pivotal role in orchestrating the sequential binding of Na+ and then K+ (or H+) ions to facilitate 5-HT uptake in SERT.


Asunto(s)
Simulación de Dinámica Molecular , Potasio , Proteínas de Transporte de Serotonina en la Membrana Plasmática , Sodio , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/química , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Potasio/metabolismo , Sitios de Unión , Humanos , Sodio/metabolismo , Serotonina/metabolismo , Unión Proteica , Animales
4.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33658361

RESUMEN

The human GlyT1 glycine transporter requires chloride for its function. However, the mechanism by which Cl- exerts its influence is unknown. To examine the role that Cl- plays in the transport cycle, we measured the effect of Cl- on both glycine binding and conformational changes. The ability of glycine to displace the high-affinity radioligand [3H]CHIBA-3007 required Na+ and was potentiated over 1,000-fold by Cl- We generated GlyT1b mutants containing reactive cysteine residues in either the extracellular or cytoplasmic permeation pathways and measured changes in the reactivity of those cysteine residues as indicators of conformational changes in response to ions and substrate. Na+ increased accessibility in the extracellular pathway and decreased it in the cytoplasmic pathway, consistent with stabilizing an outward-open conformation as observed in other members of this transporter family. In the presence of Na+, both glycine and Cl- independently shifted the conformation of GlyT1b toward an outward-closed conformation. Together, Na+, glycine, and Cl- stabilized an inward-open conformation of GlyT1b. We then examined whether Cl- acts by interacting with a conserved glutamine to allow formation of an ion pair that stabilizes the closed state of the extracellular pathway. Molecular dynamics simulations of a GlyT1 homolog indicated that this ion pair is formed more frequently as that pathway closes. Mutation of the glutamine blocked the effect of Cl-, and substituting it with glutamate or lysine resulted in outward- or inward-facing transporter conformations, respectively. These results provide an unexpected insight into the role of Cl- in this family of transporters.


Asunto(s)
Cloruros/química , Proteínas de Transporte de Glicina en la Membrana Plasmática/química , Simulación de Dinámica Molecular , Línea Celular , Cloruros/metabolismo , Proteínas de Transporte de Glicina en la Membrana Plasmática/metabolismo , Humanos , Transporte Iónico , Conformación Proteica , Sodio/química , Sodio/metabolismo
5.
Neurochem Res ; 47(1): 3-8, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33929682

RESUMEN

Baruch Kanner and this author have had parallel careers investigating neurotransmitter transporters. At multiple times during their careers, they have found themselves collaborating or competing, but always learning from each other. This commentary elaborates on the interactions between the Kanner and Rudnick laboratories, with a focus on transporters in the Neurotransmitter: Sodium Symporter (NSS) family of amino acid and amine transporters. A key focus of these interactions is the mechanism by which chloride ions activate and drive transport.


Asunto(s)
Cloruros , Simportadores , Aminoácidos/metabolismo , Historia del Siglo XX , Historia del Siglo XXI , Proteínas de Transporte de Membrana , Neurotransmisores/metabolismo , Sodio
6.
Proc Natl Acad Sci U S A ; 115(38): E8854-E8862, 2018 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-30181291

RESUMEN

The coupled transport of ions and substrates allows transporters to accumulate substrates using the energy of transmembrane ion gradients and electrical potentials. During transport, conformational changes that switch accessibility of substrate and ion binding sites from one side of the membrane to the other must be controlled so as to prevent uncoupled movement of ions or substrates. In the neurotransmitter:sodium symporter (NSS) family, Na+ stabilizes the transporter in an outward-open state, thus decreasing the likelihood of uncoupled Na+ transport. Substrate binding, in a step essential for coupled transport, must overcome the effect of Na+, allowing intracellular substrate and Na+ release from an inward-open state. However, the specific elements of the protein that mediate this conformational response to substrate binding are unknown. Previously, we showed that in the prokaryotic NSS transporter LeuT, the effect of Na+ on conformation requires the Na2 site, where it influences conformation by fostering interaction between two domains of the protein. Here, we used cysteine accessibility to measure conformational changes of LeuT in Escherichia coli membranes. We identified a conserved tyrosine residue in the substrate binding site required for substrate to convert LeuT to inward-open states by establishing an interaction between the two transporter domains. We further identify additional required interactions between the two transporter domains in the extracellular pathway. Together with our previous work on the conformational effect of Na+, these results identify mechanistic components underlying ion-substrate coupling in NSS transporters.


Asunto(s)
Proteínas de Escherichia coli/química , Escherichia coli/metabolismo , Proteínas de Transporte de Neurotransmisores en la Membrana Plasmática/química , Dominios Proteicos , Sodio/metabolismo , Sitios de Unión , Cationes Monovalentes/metabolismo , Membrana Celular/metabolismo , Cisteína/química , Cisteína/metabolismo , Citoplasma/metabolismo , Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Simulación de Dinámica Molecular , Mutación , Proteínas de Transporte de Neurotransmisores en la Membrana Plasmática/metabolismo , Unión Proteica , Transcitosis , Tirosina/química , Tirosina/metabolismo
7.
Proc Natl Acad Sci U S A ; 113(20): E2776-83, 2016 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-27140629

RESUMEN

Serotonin transporter (SERT) is responsible for reuptake and recycling of 5-hydroxytryptamine (5-HT; serotonin) after its exocytotic release during neurotransmission. Mutations in human SERT are associated with psychiatric disorders and autism. Some of these mutations affect the regulation of SERT activity by cGMP-dependent phosphorylation. Here we provide direct evidence that this phosphorylation occurs at Thr276, predicted to lie near the cytoplasmic end of transmembrane helix 5 (TM5). Using membranes from HeLa cells expressing SERT and intact rat basophilic leukemia cells, we show that agents such as Na(+) and cocaine that stabilize outward-open conformations of SERT decreased phosphorylation and agents that stabilize inward-open conformations (e.g., 5-HT, ibogaine) increased phosphorylation. The opposing effects of the inhibitors cocaine and ibogaine were each reversed by an excess of the other inhibitor. Inhibition of phosphorylation by Na(+) and stimulation by ibogaine occurred at concentrations that induced outward opening and inward opening, respectively, as measured by the accessibility of cysteine residues in the extracellular and cytoplasmic permeation pathways, respectively. The results are consistent with a mechanism of SERT regulation that is activated by the transport of 5-HT, which increases the level of inward-open SERT and may lead to unwinding of the TM5 helix to allow phosphorylation.


Asunto(s)
Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Transporte Biológico , Cocaína/farmacología , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Células HeLa , Humanos , Ibogaína/farmacología , Fosforilación , Conformación Proteica en Hélice alfa , Procesamiento Proteico-Postraduccional , Serotonina/farmacología , Transducción de Señal
8.
J Biol Chem ; 291(3): 1456-71, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26582198

RESUMEN

In LeuT, a prokaryotic homolog of neurotransmitter transporters, Na(+) stabilizes outward-open conformational states. We examined how each of the two LeuT Na(+) binding sites contributes to Na(+)-dependent closure of the cytoplasmic pathway using biochemical and biophysical assays of conformation. Mutating either of two residues that contribute to the Na2 site completely prevented cytoplasmic closure in response to Na(+), suggesting that Na2 is essential for this conformational change, whereas Na1 mutants retained Na(+) responsiveness. However, mutation of Na1 residues also influenced the Na(+)-dependent conformational change in ways that varied depending on the position mutated. Computational analyses suggest those mutants influence the ability of Na1 binding to hydrate the substrate pathway and perturb an interaction network leading to the extracellular gate. Overall, the results demonstrate that occupation of Na2 stabilizes outward-facing conformations presumably through a direct interaction between Na(+) and transmembrane helices 1 and 8, whereas Na(+) binding at Na1 influences conformational change through a network of intermediary interactions. The results also provide evidence that N-terminal release and helix motions represent distinct steps in cytoplasmic pathway opening.


Asunto(s)
Sistemas de Transporte de Aminoácidos/química , Organismos Acuáticos/metabolismo , Proteínas Bacterianas/química , Bacterias Gramnegativas/metabolismo , Modelos Moleculares , Proteínas de Transporte de Neurotransmisores en la Membrana Plasmática/química , Sodio/metabolismo , Sustitución de Aminoácidos , Sistemas de Transporte de Aminoácidos/genética , Sistemas de Transporte de Aminoácidos/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Cisteína/química , Ligandos , Liposomas , Simulación de Dinámica Molecular , Mutagénesis Sitio-Dirigida , Mutación , Proteínas de Transporte de Neurotransmisores en la Membrana Plasmática/genética , Proteínas de Transporte de Neurotransmisores en la Membrana Plasmática/metabolismo , Conformación Proteica , Pliegue de Proteína , Estabilidad Proteica , Proteolípidos , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
9.
Mol Pharmacol ; 89(1): 165-75, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26519222

RESUMEN

Determining the structural elements that define substrates and inhibitors at the monoamine transporters is critical to elucidating the mechanisms underlying these disparate functions. In this study, we addressed this question directly by generating a series of N-substituted 3,4-methylenedioxyamphetamine analogs that differ only in the number of methyl substituents on the terminal amine group. Starting with 3,4-methylenedioxy-N-methylamphetamine, 3,4-methylenedioxy-N,N-dimethylamphetamine (MDDMA) and 3,4-methylenedioxy-N,N,N-trimethylamphetamine (MDTMA) were prepared. We evaluated the functional activities of the compounds at all three monoamine transporters in native brain tissue and cells expressing the transporters. In addition, we used ligand docking to generate models of the respective protein-ligand complexes, which allowed us to relate the experimental findings to available structural information. Our results suggest that the 3,4-methylenedioxyamphetamine analogs bind at the monoamine transporter orthosteric binding site by adopting one of two mutually exclusive binding modes. 3,4-methylenedioxyamphetamine and 3,4-methylenedioxy-N-methylamphetamine adopt a high-affinity binding mode consistent with a transportable substrate, whereas MDDMA and MDTMA adopt a low-affinity binding mode consistent with an inhibitor, in which the ligand orientation is inverted. Importantly, MDDMA can alternate between both binding modes, whereas MDTMA exclusively binds to the low-affinity mode. Our experimental results are consistent with the idea that the initial orientation of bound ligands is critical for subsequent interactions that lead to transporter conformational changes and substrate translocation.


Asunto(s)
N-Metil-3,4-metilenodioxianfetamina/química , N-Metil-3,4-metilenodioxianfetamina/metabolismo , Proteínas de Transporte Vesicular de Monoaminas/química , Proteínas de Transporte Vesicular de Monoaminas/metabolismo , Animales , Sitios de Unión/fisiología , Transporte Biológico/fisiología , Células HEK293 , Células HeLa , Humanos , Masculino , Estructura Terciaria de Proteína , Ratas , Ratas Sprague-Dawley
10.
Biochemistry ; 53(33): 5444-60, 2014 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-25093911

RESUMEN

Uptake of neurotransmitters by sodium-coupled monoamine transporters of the NSS family is required for termination of synaptic transmission. Transport is tightly regulated by protein-protein interactions involving the small cytoplasmic segments at the amino- and carboxy-terminal ends of the transporter. Although structures of homologues provide information about the transmembrane regions of these transporters, the structural arrangement of the terminal domains remains largely unknown. Here, we combined molecular modeling, biochemical, and biophysical approaches in an iterative manner to investigate the structure of the 82-residue N-terminal and 30-residue C-terminal domains of human serotonin transporter (SERT). Several secondary structures were predicted in these domains, and structural models were built using the Rosetta fragment-based methodology. One-dimensional (1)H nuclear magnetic resonance and circular dichroism spectroscopy supported the presence of helical elements in the isolated SERT N-terminal domain. Moreover, introducing helix-breaking residues within those elements altered the fluorescence resonance energy transfer signal between terminal cyan fluorescent protein and yellow fluorescent protein tags attached to full-length SERT, consistent with the notion that the fold of the terminal domains is relatively well-defined. Full-length models of SERT that are consistent with these and published experimental data were generated. The resultant models predict confined loci for the terminal domains and predict that they move apart during the transport-related conformational cycle, as predicted by structures of homologues and by the "rocking bundle" hypothesis, which is consistent with spectroscopic measurements. The models also suggest the nature of binding to regulatory interaction partners. This study provides a structural context for functional and regulatory mechanisms involving SERT terminal domains.


Asunto(s)
Modelos Moleculares , Proteínas de Transporte de Serotonina en la Membrana Plasmática/química , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Secuencia de Aminoácidos , Dicroismo Circular , Citoplasma/química , Transferencia Resonante de Energía de Fluorescencia , Humanos , Espectroscopía de Resonancia Magnética , Datos de Secuencia Molecular , Conformación Proteica , Pliegue de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética
11.
Pflugers Arch ; 466(1): 25-42, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24337881

RESUMEN

The human SLC6 family is composed of approximately 20 structurally related symporters (co-transporters) that use the transmembrane electrochemical gradient to actively import their substrates into cells. Approximately half of the substrates of these transporters are amino acids, with others transporting biogenic amines and/or closely related compounds, such as nutrients and compatible osmolytes. In this short review, five leaders in the field discuss a number of currently important research themes that involve SLC6 transporters, highlighting the integrative role they play across a wide spectrum of different functions. The first essay, by Gary Rudnick, describes the molecular mechanism of their coupled transport which is being progressively better understood based on new crystal structures, functional studies, and modeling. Next, the question of multiple levels of transporter regulation is discussed by Reinhard Krämer, in the context of osmoregulation and stress response by the related bacterial betaine transporter BetP. The role of selected members of the human SLC6 family that function as nutrient amino acid transporters is then reviewed by François Verrey. He discusses how some of these transporters mediate the active uptake of (essential) amino acids into epithelial cells of the gut and the kidney tubule to support systemic amino acid requirements, whereas others are expressed in specific cells to support their specialized metabolism and/or growth. The most extensively studied members of the human SLC6 family are neurotransmitter reuptake transporters, many of which are important drug targets for the treatment of neuropsychiatric disorders. Randy Blakely discusses the role of posttranscriptional modifications of these proteins in regulating transporter subcellular localization and activity state. Finally, Dennis Murphy reviews how natural gene variants and mouse genetic models display consistent behavioral alterations that relate to altered extracellular neurotransmitter levels.


Asunto(s)
Proteínas Transportadoras de GABA en la Membrana Plasmática/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Transportadoras de GABA en la Membrana Plasmática/química , Proteínas Transportadoras de GABA en la Membrana Plasmática/genética , Humanos , Transporte Iónico , Datos de Secuencia Molecular
12.
Mol Membr Biol ; 30(7): 355-9, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24147977

RESUMEN

Abstract The availability of high-resolution atomic structures for transport proteins provides unprecedented opportunities for understanding their mechanism of action. The details of conformational change can be deduced from these structures, especially when multiple conformations are available. However, the singular ability of transporters to couple the movement of one solute to that of another requires even more information than what is supplied by a crystal structure. This short commentary discusses how recent biochemical and biophysical studies are beginning to reveal how solute coupling is achieved.


Asunto(s)
Transporte Biológico , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Transporte Iónico , Sitios de Unión , Ligandos , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/metabolismo , Modelos Moleculares , Conformación Proteica
13.
J Biol Chem ; 287(43): 36051-8, 2012 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-22942288

RESUMEN

The serotonin transporter (SERT) is responsible for reuptake of serotonin (5-hydroxytryptamine) after its exocytotic release from neurons. It is the primary target for antidepressants and stimulants, including "ecstasy" (3,4-methylenedioxymethamphetamine). SERT is regulated by several processes, including a cyclic GMP signaling pathway involving nitric oxide synthase, guanylyl cyclase, and cGMP-dependent protein kinase (PKG). Here, we show that SERT was phosphorylated in a PKG Iα-dependent manner in vitro, but that SERT was not a direct substrate of PKG. We generated an analog-sensitive gatekeeper residue mutant of PKG Iα (M438G) that efficiently used the ATP analog N(6)-benzyl-ATP. This mutant, but not the wild type (WT) kinase, used the ATP analog to phosphorylate both a model peptide substrate as well as an established protein substrate of PKG (vasodilator-stimulated phosphoprotein). PKG Iα M438G effectively substituted for the WT kinase in stimulating SERT-mediated 5-hydroxytryptamine transport in cultured cells. Addition of either WT or mutant PKG Iα M438G to membranes containing SERT in vitro led to radiolabel incorporation from [γ-(33)P]ATP but not from similarly labeled N(6)-benzyl-ATP, indicating that SERT was phosphorylated by another kinase that could not utilize the ATP analog. These results are consistent with the proposed SERT phosphorylation site, Thr-276, being highly divergent from the consensus PKG phosphorylation site sequence, which we verified through peptide library screening. Another proposed SERT kinase, the p38 mitogen-activated protein kinase, could not substitute for PKG in this assay, and p38 inhibitors did not block PKG-dependent phosphorylation of SERT. The results suggest that PKG initiates a kinase cascade that leads to phosphorylation of SERT by an as yet unidentified protein kinase.


Asunto(s)
Proteína Quinasa Dependiente de GMP Cíclico Tipo I/metabolismo , GMP Cíclico/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Serotonina/metabolismo , Sustitución de Aminoácidos , Transporte Biológico Activo/fisiología , GMP Cíclico/genética , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/genética , Células HEK293 , Humanos , Mutación Missense , Fosforilación/fisiología , Serotonina/genética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
J Biol Chem ; 287(1): 438-445, 2012 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-22072712

RESUMEN

Serotonin (5-HT) uptake by the human serotonin transporter (hSERT) is driven by ion gradients. The stoichiometry of transported 5-HT and ions is predicted to result in electroneutral charge movement. However, hSERT mediates a current when challenged with 5-HT. This discrepancy can be accounted for by an uncoupled ion flux. Here, we investigated the mechanistic basis of the uncoupled currents and its relation to the conformational cycle of hSERT. Our observations support the conclusion that the conducting state underlying the uncoupled ion flux is in equilibrium with an inward facing state of the transporter with K+ bound. We identified conditions associated with accumulation of the transporter in inward facing conformations. Manipulations that increased the abundance of inward facing states resulted in enhanced steady-state currents. We present a comprehensive kinetic model of the transport cycle, which recapitulates salient features of the recorded currents. This study provides a framework for exploring transporter-associated currents.


Asunto(s)
Conductividad Eléctrica , Fenómenos Electrofisiológicos , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Animales , Fenómenos Electrofisiológicos/efectos de los fármacos , Femenino , Células HEK293 , Humanos , Cinética , Microscopía Fluorescente , Modelos Biológicos , Potasio/metabolismo , Conformación Proteica/efectos de los fármacos , Serotonina/farmacología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/química
15.
J Biol Chem ; 287(22): 18524-34, 2012 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-22451652

RESUMEN

Ibogaine, a hallucinogenic alkaloid proposed as a treatment for opiate withdrawal, has been shown to inhibit serotonin transporter (SERT) noncompetitively, in contrast to all other known inhibitors, which are competitive with substrate. Ibogaine binding to SERT increases accessibility in the permeation pathway connecting the substrate-binding site with the cytoplasm. Because of the structural similarity between ibogaine and serotonin, it had been suggested that ibogaine binds to the substrate site of SERT. The results presented here show that ibogaine binds to a distinct site, accessible from the cell exterior, to inhibit both serotonin transport and serotonin-induced ionic currents. Ibogaine noncompetitively inhibited transport by both SERT and the homologous dopamine transporter (DAT). Ibogaine blocked substrate-induced currents also in DAT and increased accessibility of the DAT cytoplasmic permeation pathway. When present on the cell exterior, ibogaine inhibited SERT substrate-induced currents, but not when it was introduced into the cytoplasm through the patch electrode. Similar to noncompetitive transport inhibition, the current block was not reversed by increasing substrate concentration. The kinetics of inhibitor binding and dissociation, as determined by their effect on SERT currents, indicated that ibogaine does not inhibit by forming a long-lived complex with SERT, but rather binds directly to the transporter in an inward-open conformation. A kinetic model for transport describing the noncompetitive action of ibogaine and the competitive action of cocaine accounts well for the results of the present study.


Asunto(s)
Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/antagonistas & inhibidores , Antagonistas de Aminoácidos Excitadores/farmacología , Ibogaína/farmacología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/efectos de los fármacos , Animales , Sitios de Unión , Línea Celular , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Antagonistas de Aminoácidos Excitadores/metabolismo , Humanos , Ibogaína/metabolismo , Cinética , Técnicas de Placa-Clamp , Ensayo de Unión Radioligante , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Xenopus laevis
16.
Neuropharmacology ; 240: 109704, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-37703919

RESUMEN

Mephedrone (4-methylmethcathinone) is a cathinone derivative that is recreationally consumed for its energizing and empathogenic effects. The stimulating properties are believed to arise from the ability of mephedrone to interact with the high-affinity transporters for dopamine (DA) (DAT) and norepinephrine (NET), whereas the entactogenic effect presumably relies on its activity at the serotonin (5-HT) transporter (SERT). Early studies found that mephedrone acts as a releaser at NET, DAT and SERT, and thus promotes efflux of the respective monoamines. Evidence linked drug-induced reverse transport of 5-HT via SERT to prosocial effects, whereas activity at DAT is strongly correlated with abuse liability. Consequently, we sought to evaluate the pharmacology of mephedrone at human (h) DAT and SERT, heterologously expressed in human embryonic kidney 293 cells, in further detail. In line with previous studies, we report that mephedrone evokes carrier-mediated release via hDAT and hSERT. We found this effect to be sensitive to the protein kinase C inhibitor GF109203X. Electrophysiological recordings revealed that mephedrone is actively transported by hDAT and hSERT. However, mephedrone acts as a full substrate of hSERT but as a partial substrate of hDAT. Furthermore, when compared to fully efficacious releasing agents at hDAT and hSERT (i.e. S(+)-amphetamine and para-chloroamphetamine, respectively) mephedrone displays greater efficacy as a releaser at hSERT than at hDAT. In summary, this study provides additional insights into the molecular mechanism of action of mephedrone at hDAT and hSERT.

17.
J Biol Chem ; 286(4): 2461-8, 2011 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-21097501

RESUMEN

By transporting serotonin (5-HT) into neurons and other cells, serotonin transporter (SERT) modulates the action of 5-HT at cell surface receptors. SERT itself is modulated by several processes, including the cGMP signaling pathway. Activation of SERT by cGMP requires the cGMP-dependent protein kinase (PKG). Here we show that in HeLa cells lacking endogenous PKG, expression of PKGIα or PKGIß was required for 8-bromoguanosine-3',5'-cyclic monophosphate (8-Br-cGMP) to stimulate SERT phosphorylation and 5-HT influx. Catalytically inactive PKG mutants and wild-type PKGII did not support this stimulation. However, a mutant PKGII (G2A) that was not myristoylated substituted for functional PKGI, suggesting that myristoylation and subsequent membrane association blocked productive interaction with SERT. PKG also influenced SERT expression and localization. PKGI isoforms increased total and cell surface SERT levels, and PKGII decreased cell surface SERT without altering total expression. Remarkably, these changes did not require 8-Br-cGMP or functional kinase activity and were also observed with a SERT mutant resistant to activation by PKG. Both PKGIα and PKGIß formed detergent-stable complexes with SERT, and this association did not require catalytic activity. The nonmyristoylated PKGII G2A mutant stimulated SERT expression similar to PKGI isoforms. These results suggest multiple mechanisms by which PKG can modulate SERT and demonstrate that the functional difference between PKG isoforms results from myristoylation of PKGII.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Ácido Mirístico/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/biosíntesis , Membrana Celular/genética , Membrana Celular/metabolismo , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacología , Proteína Quinasa Dependiente de GMP Cíclico Tipo I , Proteínas Quinasas Dependientes de GMP Cíclico/genética , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Células HeLa , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Mutación , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética
18.
J Biol Chem ; 286(4): 2834-42, 2011 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-21115480

RESUMEN

In ion-coupled transport proteins, occupation of selective ion-binding sites is required to trigger conformational changes that lead to substrate translocation. Neurotransmitter transporters, targets of abused and therapeutic drugs, require Na(+) and Cl(-) for function. We recently proposed a chloride-binding site in these proteins not present in Cl(-)-independent prokaryotic homologues. Here we describe conversion of the Cl(-)-independent prokaryotic tryptophan transporter TnaT to a fully functional Cl(-)-dependent form by a single point mutation, D268S. Mutations in TnaT-D268S, in wild type TnaT and in serotonin transporter provide direct evidence for the involvement of each of the proposed residues in Cl(-) coordination. In both SERT and TnaT-D268S, Cl(-) and Na(+) mutually increased each other's potency, consistent with electrostatic interaction through adjacent binding sites. These studies establish the site where Cl(-) binds to trigger conformational change during neurotransmitter transport.


Asunto(s)
Proteínas Bacterianas/química , Bacilos Grampositivos Formadores de Endosporas/química , Proteínas de Transporte de Serotonina en la Membrana Plasmática/química , Sustitución de Aminoácidos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Transporte Biológico/fisiología , Cloruros/metabolismo , Bacilos Grampositivos Formadores de Endosporas/genética , Bacilos Grampositivos Formadores de Endosporas/metabolismo , Humanos , Mutación Missense , Neurotransmisores/química , Neurotransmisores/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Homología Estructural de Proteína
19.
J Biol Chem ; 286(35): 30823-30836, 2011 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-21730057

RESUMEN

Na(+)- and Cl(-)-dependent uptake of neurotransmitters via transporters of the SLC6 family, including the human serotonin transporter (SLC6A4), is critical for efficient synaptic transmission. Although residues in the human serotonin transporter involved in direct Cl(-) coordination of human serotonin transport have been identified, the role of Cl(-) in the transport mechanism remains unclear. Through a combination of mutagenesis, chemical modification, substrate and charge flux measurements, and molecular modeling studies, we reveal an unexpected role for the highly conserved transmembrane segment 1 residue Asn-101 in coupling Cl(-) binding to concentrative neurotransmitter uptake.


Asunto(s)
Asparagina/química , Cloruros/química , Neurotransmisores/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/química , Animales , Cisteína/química , Electrofisiología/métodos , Células HeLa , Humanos , Iones , Mutagénesis Sitio-Dirigida , Norepinefrina/metabolismo , Oocitos/metabolismo , Técnicas de Placa-Clamp , Plásmidos/metabolismo , Ratas , Serotonina/metabolismo , Xenopus laevis
20.
Biochemistry ; 50(35): 7462-75, 2011 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-21774491

RESUMEN

Ion-coupled solute transporters are responsible for transporting nutrients, ions, and signaling molecules across a variety of biological membranes. Recent high-resolution crystal structures of several transporters from protein families that were previously thought to be unrelated show common structural features indicating a large structural family representing transporters from all kingdoms of life. This review describes studies that led to an understanding of the conformational changes required for solute transport in this family. The first structure in this family showed the bacterial amino acid transporter LeuT, which is homologous to neurotransmitter transporters, in an extracellularly oriented conformation with a molecule of leucine occluded at the substrate site. Studies with the mammalian serotonin transporter identified positions, buried in the LeuT structure, that defined a potential pathway leading from the cytoplasm to the substrate binding site. Modeling studies utilized an inverted structural repeat within the LeuT crystal structure to predict the conformation of LeuT in which the cytoplasmic permeation pathway, consisting of positions identified in SERT, was open for diffusion of the substrate to the cytoplasm. From the difference between the model and the crystal structures, a simple "rocking bundle" mechanism was proposed, in which a four-helix bundle changed its orientation with respect to the rest of the protein to close the extracellular pathway and open the cytoplasmic one. Subsequent crystal structures from structurally related proteins provide evidence supporting this model for transport.


Asunto(s)
Permeabilidad de la Membrana Celular/fisiología , Citoplasma/metabolismo , Proteínas de Transporte de Neurotransmisores/química , Proteínas de Transporte de Neurotransmisores/metabolismo , Transducción de Señal/fisiología , Secuencia de Aminoácidos , Animales , Permeabilidad de la Membrana Celular/genética , Citoplasma/química , Citoplasma/genética , Humanos , Leucina/química , Leucina/genética , Leucina/metabolismo , Datos de Secuencia Molecular , Proteínas de Transporte de Neurotransmisores/genética , Conformación Proteica , Transducción de Señal/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA