Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Biol Chem ; 293(10): 3510-3523, 2018 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-29352106

RESUMEN

The serotonin transporter (SERT) is important for reuptake of the neurotransmitter serotonin from the synaptic cleft and is also the target of most antidepressants. It has previously been shown that cholesterol in the membrane bilayer affects the conformation of SERT. Although recent crystal structures have identified several potential cholesterol-binding sites, it is unclear whether any of these potential cholesterol sites are occupied by cholesterol and functionally relevant. In the present study, we focus on the conserved cholesterol site 1 (CHOL1) located in a hydrophobic groove between TM1a, TM5, and TM7. By molecular dynamics simulations, we demonstrate a strong binding of cholesterol to CHOL1 in a membrane bilayer environment. In biochemical experiments, we find that cholesterol depletion induces a more inward-facing conformation favoring substrate analog binding. Consistent with this, we find that mutations in CHOL1 with a negative impact on cholesterol binding induce a more inward-facing conformation, and, vice versa, mutations with a positive impact on cholesterol binding induce a more outward-facing conformation. This shift in transporter conformation dictated by the ability to bind cholesterol in CHOL1 affects the apparent substrate affinity, maximum transport velocity, and turnover rates. Taken together, we show that occupation of CHOL1 by cholesterol is of major importance in the transporter conformational equilibrium, which in turn dictates ligand potency and serotonin transport activity. Based on our findings, we propose a mechanistic model that incorporates the role of cholesterol binding to CHOL1 in the function of SERT.


Asunto(s)
Colesterol/metabolismo , Membrana Dobles de Lípidos/metabolismo , Modelos Moleculares , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Sustitución de Aminoácidos , Sitios de Unión , Unión Competitiva , Transporte Biológico/efectos de los fármacos , Colesterol/química , Secuencia Conservada , Células HEK293 , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Cinética , Ligandos , Membrana Dobles de Lípidos/química , Simulación de Dinámica Molecular , Mutagénesis Sitio-Dirigida , Mutación , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Estabilidad Proteica , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/química , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , beta-Ciclodextrinas/química , beta-Ciclodextrinas/metabolismo
2.
J Biol Chem ; 290(12): 7747-55, 2015 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-25614630

RESUMEN

Serotonergic neurotransmission is modulated by the membrane-embedded serotonin transporter (SERT). SERT mediates the reuptake of serotonin into the presynaptic neurons. Conformational changes in SERT occur upon binding of ions and substrate and are crucial for translocation of serotonin across the membrane. Our understanding of these conformational changes is mainly based on crystal structures of a bacterial homolog in various conformations, derived homology models of eukaryotic neurotransmitter transporters, and substituted cysteine accessibility method of SERT. However, the dynamic changes that occur in the human SERT upon binding of ions, the translocation of substrate, and the role of cholesterol in this interplay are not fully elucidated. Here we show that serotonin induces a dualistic conformational response in SERT. We exploited the substituted cysteine scanning method under conditions that were sensitized to detect a more outward-facing conformation of SERT. We found a novel high affinity outward-facing conformational state of the human SERT induced by serotonin. The ionic requirements for this new conformational response to serotonin mirror the ionic requirements for translocation. Furthermore, we found that membrane cholesterol plays a role in the dualistic conformational response in SERT induced by serotonin. Our results indicate the existence of a subpopulation of SERT responding differently to serotonin binding than hitherto believed and that membrane cholesterol plays a role in this subpopulation of SERT.


Asunto(s)
Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Serotonina/metabolismo , Colesterol/metabolismo , Células HEK293 , Humanos , Lípidos de la Membrana/metabolismo , Unión Proteica , Conformación Proteica , Proteínas de Transporte de Serotonina en la Membrana Plasmática/química
3.
Mol Pharmacol ; 85(2): 208-17, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24214825

RESUMEN

Mazindol has been explored as a possible agent in cocaine addiction pharmacotherapy. The tetracyclic compound inhibits both the dopamine transporter and the serotonin transporter, and simple chemical modifications considerably alter target selectivity. Mazindol, therefore, is an attractive scaffold for both understanding the molecular determinants of serotonin/dopamine transporter selectivity and for the development of novel drug abuse treatments. Using molecular modeling and pharmacologic profiling of rationally chosen serotonin and dopamine transporter mutants with respect to a series of mazindol analogs has allowed us to determine the orientation of mazindol within the central binding site. We find that mazindol binds in the central substrate binding site, and that the transporter selectivity can be modulated through mutations of a few residues in the binding pocket. Mazindol is most likely to bind as the R-enantiomer. Tyrosines 95 and 175 in the human serotonin transporter and the corresponding phenylalanines 75 and 155 in the human dopamine transporter are the primary determinants of mazindol selectivity. Manipulating the interaction of substituents on the 7-position with the human serotonin transporter Tyr175 versus dopamine transporter Phe155 is found to be a strong tool in tuning the selectivity of mazindol analogs and may be used in future drug design of cocaine abuse pharmacotherapies.


Asunto(s)
Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Mazindol/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Sitios de Unión , Células Cultivadas , Trastornos Relacionados con Cocaína/tratamiento farmacológico , Humanos , Mazindol/química , Mazindol/uso terapéutico , Modelos Moleculares , Relación Estructura-Actividad
4.
J Biol Chem ; 285(11): 8363-74, 2010 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-19948720

RESUMEN

Tricyclic antidepressants (TCAs) have been used for decades, but their orientation within and molecular interactions with their primary target is yet unsettled. The recent finding of a TCA binding site in the extracellular vestibule of the bacterial leucine transporter 11 A above the central site has prompted debate about whether this vestibular site in the bacterial transporter is applicable to binding of antidepressants to their relevant physiological target, the human serotonin transporter (hSERT). We present an experimentally validated structural model of imipramine and analogous TCAs in the central substrate binding site of hSERT. Two possible binding modes were observed from induced fit docking calculations. We experimentally validated a single binding mode by combining mutagenesis of hSERT with uptake inhibition studies of different TCA analogs according to the paired mutation ligand analog complementation paradigm. Using this experimental method, we identify a salt bridge between the tertiary aliphatic amine and Asp(98). Furthermore, the 7-position of the imipramine ring is found vicinal to Phe(335), and the pocket lined by Ala(173) and Thr(439) is utilized by 3-substituents. These protein-ligand contact points unambiguously orient the TCA within the central binding site and reveal differences between substrate binding and inhibitor binding, giving important clues to the inhibition mechanism. Consonant with the well established competitive inhibition of uptake by TCAs, the resulting binding site for TCAs in hSERT is fully overlapping with the serotonin binding site in hSERT and dissimilar to the low affinity noncompetitive TCA site reported in the leucine transporter (LeuT).


Asunto(s)
Antidepresivos Tricíclicos/química , Antidepresivos Tricíclicos/metabolismo , Imipramina/química , Imipramina/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Alanina/genética , Ácido Aspártico/genética , Sitios de Unión , Unión Competitiva , Células Cultivadas , Humanos , Imipramina/análogos & derivados , Riñón/citología , Leucina/genética , Modelos Químicos , Mutagénesis Sitio-Dirigida , Fenilalanina/genética , Serotonina/farmacocinética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/química , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Relación Estructura-Actividad , Transfección , Tritio
5.
J Am Chem Soc ; 132(4): 1311-22, 2010 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-20055463

RESUMEN

The two enantiomers of the antidepressant citalopram inhibit the human serotonin transporter substantially differently. Previous studies revealed Tyr95 and Ile172 as important for citalopram binding, however, the overall orientation of the ligands in the binding site and the protein-ligand interaction points remain unknown. The binding of S- and R-citalopram to a human serotonin transporter homology model are herein examined via docking simulations including induced fit effects. For a better description of the formal charges of the ligand when bound inside the protein, polarization effects of the protein were included by additional quantum-polarized ligand docking calculations, where ligand charges are evaluated using QM/MM calculations. By this approach a much clearer picture emerged of the positions of the functional groups of citalopram. The two enantiomers are predicted to bind in the substrate binding pocket with opposite orientations of their aromatic groups. The predicted binding modes are experimentally validated using human wild type and 15 serotonin transporter mutants and 13 optically pure citalopram analogues. Important protein-ligand interaction points were identified validating one binding model for each enantiomer. In the validated model of the high affinity enantiomer, S-citalopram, the fluorine atom is located near Ala173 and Thr439 and the cyano group is in close proximity of Phe341; these contacts are found to be reversed for the R-enantiomer.


Asunto(s)
Antidepresivos de Segunda Generación/química , Antidepresivos de Segunda Generación/farmacología , Citalopram/química , Citalopram/farmacología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Sitios de Unión , Simulación por Computador , Humanos , Modelos Moleculares , Unión Proteica , Proteínas de Transporte de Serotonina en la Membrana Plasmática/química , Estereoisomerismo
6.
J Am Chem Soc ; 130(12): 3853-65, 2008 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-18314975

RESUMEN

Molecular modeling and structure-activity relationship studies were performed to propose a model for binding of the neurotransmitter serotonin (5-HT) to the human serotonin transporter (hSERT). Homology models were constructed using the crystal structure of a bacterial homologue, the leucine transporter from Aquifex aeolicus, as the template and three slightly different sequence alignments. Induced fit docking of 5-HT into hSERT homology models resulted in two different binding modes. Both show a salt bridge between Asp98 and the charged primary amine of 5-HT, and both have the 5-HT C6 position of the indole ring pointing toward Ala173. The difference between the two orientations of 5-HT is an enantiofacial discrimination of the indole ring, resulting in the 5-hydroxyl group of 5-HT being vicinal to either Ser438/Thr439 or Ala169/Ile172/Ala173. To assess the binding experimentally, binding affinities for 5-HT and 17 analogues toward wild type and 13 single point mutants of hSERT were measured using an approach termed paired mutant-ligand analogue complementation (PaMLAC). The proposed ligand-protein interaction was systematically examined by disrupting it through site-directed mutagenesis and re-establishing another interaction via a ligand analogue matching the mutated residue, thereby minimizing the risk of identifying indirect effects. The interactions between Asp98 and the primary amine of 5-HT and the interaction between the C6-position of 5-HT and hSERT position 173 was confirmed using PaMLAC. The measured binding affinities of various mutants and 5-HT analogues allowed for a distinction between the two proposed binding modes of 5-HT and biochemically support the model for 5-HT binding in hSERT where the 5-hydroxyl group is in close proximity to Thr439.


Asunto(s)
Proteínas de Transporte de Serotonina en la Membrana Plasmática/química , Serotonina/química , Sitios de Unión , Línea Celular , Células Cultivadas , Simulación por Computador , Humanos , Ligandos , Modelos Moleculares , Estructura Molecular , Unión Proteica , Serotonina/análogos & derivados , Serotonina/farmacocinética , Estereoisomerismo , Relación Estructura-Actividad , Distribución Tisular
7.
J Neurochem ; 105(5): 1794-805, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18266934

RESUMEN

The selective serotonin reuptake inhibitors and tricyclic antidepressants act by inhibiting pre-synaptic reuptake of serotonin (5-HT) leading to elevated synaptic 5-HT concentrations. However, despite extensive efforts little is known about the protein-ligand interactions of serotonin transporter (SERT) and inhibitors. To identify domains and individual amino acids important for ligand binding, we cloned the serotonin transporter from zebrafish, Danio rerio, (drSERT) and compared its pharmacological profile to that of the human serotonin transporter (hSERT) with respect to inhibition of [3H]5-HT uptake and [3H]-escitalopram binding in transiently transfected human embryonic kidney cells; HEK293-MSR. Residues responsible for altered affinities inhibitors were pinpointed by generating cross-species chimeras and subsequent point mutations by site directed mutagenesis. drSERT has a higher affinity towards compounds of the imipramine class, desipramine in particular, exhibiting a 35-fold increased affinity compared to hSERT. drSERT has a 15-30-fold lower affinity towards cocaine and cocaine analogues. The differences in ligand recognition are shown to be primarily caused by interspecies differences in TM10 and were tracked down to three residues (Ala(505), Leu(506) and Ile(507)).


Asunto(s)
Proteínas de Transporte de Serotonina en la Membrana Plasmática/química , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Proteínas de Pez Cebra/química , Proteínas de Pez Cebra/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión/genética , Transporte Biológico/genética , Línea Celular , Membrana Celular/química , Membrana Celular/genética , Membrana Celular/metabolismo , Humanos , Ligandos , Datos de Secuencia Molecular , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/fisiología , Especificidad de la Especie , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/fisiología
8.
Nat Commun ; 7: 11673, 2016 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-27221344

RESUMEN

Bacterial members of the neurotransmitter:sodium symporter (NSS) family perform Na(+)-dependent amino-acid uptake and extrude H(+) in return. Previous NSS structures represent intermediates of Na(+)/substrate binding or intracellular release, but not the inward-to-outward return transition. Here we report crystal structures of Aquifex aeolicus LeuT in an outward-oriented, Na(+)- and substrate-free state likely to be H(+)-occluded. We find a remarkable rotation of the conserved Leu25 into the empty substrate-binding pocket and rearrangements of the empty Na(+) sites. Mutational studies of the equivalent Leu99 in the human serotonin transporter show a critical role of this residue on the transport rate. Molecular dynamics simulations show that extracellular Na(+) is blocked unless Leu25 is rotated out of the substrate-binding pocket. We propose that Leu25 facilitates the inward-to-outward transition by compensating a Na(+)- and substrate-free state and acts as the gatekeeper for Na(+) binding that prevents leak in inward-outward return transitions.


Asunto(s)
Sistemas de Transporte de Aminoácidos/metabolismo , Proteínas de Transporte de Neurotransmisores en la Membrana Plasmática/metabolismo , Secuencia de Aminoácidos , Sistemas de Transporte de Aminoácidos/química , Sistemas de Transporte de Aminoácidos/aislamiento & purificación , Escherichia coli , Células HEK293 , Humanos , Leucina/metabolismo , Simulación de Dinámica Molecular , Conformación Proteica , Sodio/metabolismo
9.
Nat Commun ; 7: 10680, 2016 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-26893169

RESUMEN

Stimulator of interferon genes (STING) is known be involved in control of DNA viruses but has an unexplored role in control of RNA viruses. During infection with DNA viruses STING is activated downstream of cGAMP synthase (cGAS) to induce type I interferon. Here we identify a STING-dependent, cGAS-independent pathway important for full interferon production and antiviral control of enveloped RNA viruses, including influenza A virus (IAV). Further, IAV interacts with STING through its conserved hemagglutinin fusion peptide (FP). Interestingly, FP antagonizes interferon production induced by membrane fusion or IAV but not by cGAMP or DNA. Similar to the enveloped RNA viruses, membrane fusion stimulates interferon production in a STING-dependent but cGAS-independent manner. Abolishment of this pathway led to reduced interferon production and impaired control of enveloped RNA viruses. Thus, enveloped RNA viruses stimulate a cGAS-independent STING pathway, which is targeted by IAV.


Asunto(s)
Virus de la Influenza A/fisiología , Gripe Humana/enzimología , Proteínas de la Membrana/metabolismo , Nucleotidiltransferasas/metabolismo , Infecciones por Virus ARN/enzimología , Animales , Humanos , Virus de la Influenza A/genética , Gripe Humana/genética , Gripe Humana/metabolismo , Gripe Humana/virología , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Proteínas de la Membrana/genética , Ratones , Nucleotidiltransferasas/genética , Infecciones por Virus ARN/genética , Infecciones por Virus ARN/metabolismo , Infecciones por Virus ARN/virología , Virus ARN/genética , Virus ARN/fisiología
10.
ACS Chem Neurosci ; 3(9): 693-705, 2012 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-23019496

RESUMEN

The human serotonin transporter (hSERT), the human dopamine transporter (hDAT), and the human norepinephrine transporter (hNET) facilitate the active uptake of the neurotransmitters serotonin, dopamine, and norepinephrine from the synaptic cleft. Drugs of abuse such as MDMA (streetname "ecstasy") and certain 1-phenyl-piperazine (PP) analogs such as 1-(3-chlorophenyl)-piperazine (mCPP) elicit their stimulatory effect by elevating the synaptic concentration of serotonin by blocking or reversing the normal transport activity of hSERT. Recent data suggest that certain analogs of PP may be able to counteract the addictive effect of cocaine. Little is still known about the precise mechanism by which MDMA and PP analogs function at hSERT, hDAT, and hNET and even less is known about the specific protein-ligand interactions. In this study, we provide a comprehensive biochemical examination of a repertoire of PP analogs in hSERT, hDAT, and hNET. Combined with induced fit docking models and molecular dynamics simulations of PP and 1-(3-hydroxyphenyl)-piperazine (3-OH-PP) bound to hSERT and hDAT, we present detailed molecular insight into the promiscuous binding of PP analogs in the monoamine transporters. We find that PP analogs inhibit uptake as well as induce release in all three monoamine transporters. We also find that the selectivity of the PP analogs can be adjusted by carefully selecting substituents on the PP skeleton.


Asunto(s)
Anfetaminas/metabolismo , Piperazinas/metabolismo , Proteínas de Transporte Vesicular de Monoaminas/metabolismo , Anfetaminas/química , Animales , Análisis por Conglomerados , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Células HEK293 , Humanos , Indicadores y Reactivos , Ligandos , Masculino , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/metabolismo , Piperazinas/química , Conformación Proteica , Ratas , Ratas Sprague-Dawley , Serotonina/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Especificidad por Sustrato , Proteínas de Transporte Vesicular de Monoaminas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA