Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 234
Filter
Add more filters

Publication year range
1.
Cell ; 186(10): 2160-2175.e17, 2023 05 11.
Article in English | MEDLINE | ID: mdl-37137306

ABSTRACT

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.


Subject(s)
Ibogaine , Selective Serotonin Reuptake Inhibitors , Serotonin Plasma Membrane Transport Proteins , Small Molecule Libraries , Animals , Mice , Fluoxetine/pharmacology , Ibogaine/chemistry , Ibogaine/pharmacology , Molecular Conformation , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/chemistry , Serotonin Plasma Membrane Transport Proteins/metabolism , Serotonin Plasma Membrane Transport Proteins/ultrastructure , Selective Serotonin Reuptake Inhibitors/pharmacology , Small Molecule Libraries/pharmacology
2.
Proc Natl Acad Sci U S A ; 121(18): e2319384121, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38652746

ABSTRACT

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.


Subject(s)
Molecular Dynamics Simulation , Potassium , Serotonin Plasma Membrane Transport Proteins , Sodium , Serotonin Plasma Membrane Transport Proteins/metabolism , Serotonin Plasma Membrane Transport Proteins/chemistry , Serotonin Plasma Membrane Transport Proteins/genetics , Potassium/metabolism , Binding Sites , Humans , Sodium/metabolism , Serotonin/metabolism , Protein Binding , Animals
3.
Nature ; 569(7754): 141-145, 2019 05.
Article in English | MEDLINE | ID: mdl-31019304

ABSTRACT

The serotonin transporter (SERT) regulates neurotransmitter homeostasis through the sodium- and chloride-dependent recycling of serotonin into presynaptic neurons1-3. Major depression and anxiety disorders are treated using selective serotonin reuptake inhibitors-small molecules that competitively block substrate binding and thereby prolong neurotransmitter action2,4. The dopamine and noradrenaline transporters, together with SERT, are members of the neurotransmitter sodium symporter (NSS) family. The transport activities of NSSs can be inhibited or modulated by cocaine and amphetamines2,3, and genetic variants of NSSs are associated with several neuropsychiatric disorders including attention deficit hyperactivity disorder, autism and bipolar disorder2,5. Studies of bacterial NSS homologues-including LeuT-have shown how their transmembrane helices (TMs) undergo conformational changes during the transport cycle, exposing a central binding site to either side of the membrane1,6-12. However, the conformational changes associated with transport in NSSs remain unknown. To elucidate structure-based mechanisms for transport in SERT we investigated its complexes with ibogaine, a hallucinogenic natural product with psychoactive and anti-addictive properties13,14. Notably, ibogaine is a non-competitive inhibitor of transport but displays competitive binding towards selective serotonin reuptake inhibitors15,16. Here we report cryo-electron microscopy structures of SERT-ibogaine complexes captured in outward-open, occluded and inward-open conformations. Ibogaine binds to the central binding site, and closure of the extracellular gate largely involves movements of TMs 1b and 6a. Opening of the intracellular gate involves a hinge-like movement of TM1a and the partial unwinding of TM5, which together create a permeation pathway that enables substrate and ion diffusion to the cytoplasm. These structures define the structural rearrangements that occur from the outward-open to inward-open conformations, and provide insight into the mechanism of neurotransmitter transport and ibogaine inhibition.


Subject(s)
Cryoelectron Microscopy , Ibogaine/chemistry , Ibogaine/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology , Serotonin Plasma Membrane Transport Proteins/metabolism , Serotonin Plasma Membrane Transport Proteins/ultrastructure , Serotonin/metabolism , Binding Sites/drug effects , Binding, Competitive , Biological Transport/drug effects , Hallucinogens/chemistry , Hallucinogens/pharmacology , Humans , Models, Molecular , Protein Conformation/drug effects , Serotonin Plasma Membrane Transport Proteins/chemistry , Serotonin Plasma Membrane Transport Proteins/genetics , Selective Serotonin Reuptake Inhibitors/chemistry , Structure-Activity Relationship
4.
Subcell Biochem ; 104: 119-137, 2024.
Article in English | MEDLINE | ID: mdl-38963486

ABSTRACT

Transporters of the monoamine transporter (MAT) family regulate the uptake of important neurotransmitters like dopamine, serotonin, and norepinephrine. The MAT family functions using the electrochemical gradient of ions across the membrane and comprises three transporters, dopamine transporter (DAT), serotonin transporter (SERT), and norepinephrine transporter (NET). MAT transporters have been observed to exist in monomeric states to higher-order oligomeric states. Structural features, allosteric modulation, and lipid environment regulate the oligomerization of MAT transporters. NET and SERT oligomerization are regulated by levels of PIP2 present in the membrane. The kink present in TM12 in the MAT family is crucial for dimer interface formation. Allosteric modulation in the dimer interface hinders dimer formation. Oligomerization also influences the transporters' function, trafficking, and regulation. This chapter will focus on recent studies on monoamine transporters and discuss the factors affecting their oligomerization and its impact on their function.


Subject(s)
Protein Multimerization , Humans , Animals , Serotonin Plasma Membrane Transport Proteins/metabolism , Serotonin Plasma Membrane Transport Proteins/chemistry , Serotonin Plasma Membrane Transport Proteins/genetics , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Norepinephrine Plasma Membrane Transport Proteins/genetics , Norepinephrine Plasma Membrane Transport Proteins/chemistry , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopamine Plasma Membrane Transport Proteins/chemistry , Dopamine Plasma Membrane Transport Proteins/genetics , Allosteric Regulation
5.
J Chem Inf Model ; 64(10): 4059-4070, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38739718

ABSTRACT

Central nervous system (CNS) drugs have had a significant impact on treating a wide range of neurodegenerative and psychiatric disorders. In recent years, deep learning-based generative models have shown great potential for accelerating drug discovery and improving efficacy. However, specific applications of these techniques in CNS drug discovery have not been widely reported. In this study, we developed the CNSMolGen model, which uses a framework of bidirectional recurrent neural networks (Bi-RNNs) for de novo molecular design of CNS drugs. Results showed that the pretrained model was able to generate more than 90% of completely new molecular structures, which possessed the properties of CNS drug molecules and were synthesizable. In addition, transfer learning was performed on small data sets with specific biological activities to evaluate the potential application of the model for CNS drug optimization. Here, we used drugs against the classical CNS disease target serotonin transporter (SERT) as a fine-tuned data set and generated a focused database against the target protein. The potential biological activities of the generated molecules were verified by using the physics-based induced-fit docking study. The success of this model demonstrates its potential in CNS drug design and optimization, which provides a new impetus for future CNS drug development.


Subject(s)
Central Nervous System Agents , Drug Design , Neural Networks, Computer , Central Nervous System Agents/pharmacology , Central Nervous System Agents/chemistry , Molecular Docking Simulation , Humans , Serotonin Plasma Membrane Transport Proteins/metabolism , Serotonin Plasma Membrane Transport Proteins/chemistry
6.
J Biol Chem ; 298(3): 101613, 2022 03.
Article in English | MEDLINE | ID: mdl-35065961

ABSTRACT

The human serotonin transporter (hSERT) terminates neurotransmission by removing serotonin (5HT) from the synaptic cleft, an essential process for proper functioning of serotonergic neurons. Structures of the hSERT have revealed its molecular architecture in four conformations, including the outward-open and occluded states, and show the transporter's engagement with co-transported ions and the binding mode of inhibitors. In this study, we investigated the molecular mechanism by which the hSERT occludes and sequesters the substrate 5HT. This first step of substrate uptake into cells is a structural change consisting of the transition from the outward-open to the occluded state. Inhibitors such as the antidepressants citalopram, fluoxetine, and sertraline inhibit this step of the transport cycle. Using molecular dynamics simulations, we reached a fully occluded state, in which the transporter-bound 5HT becomes fully shielded from both sides of the membrane by two closed hydrophobic gates. Analysis of 5HT-triggered occlusion showed that bound 5HT serves as an essential trigger for transporter occlusion. Moreover, simulations revealed a complex sequence of steps and showed that movements of bundle domain helices are only partially correlated. 5HT-triggered occlusion is initially dominated by movements of transmembrane helix 1b, while in the final step, only transmembrane helix 6a moves and relaxes an intermediate change in its secondary structure.


Subject(s)
Serotonin Plasma Membrane Transport Proteins , Serotonin , Citalopram/chemistry , Citalopram/pharmacology , Humans , Molecular Dynamics Simulation , Protein Domains , Protein Structure, Secondary , Serotonin/chemistry , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/chemistry , Serotonin Plasma Membrane Transport Proteins/metabolism , Selective Serotonin Reuptake Inhibitors/chemistry , Selective Serotonin Reuptake Inhibitors/pharmacology , Structure-Activity Relationship
7.
Biophys J ; 121(5): 715-730, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35114149

ABSTRACT

The serotonin transporter (SERT) initiates the reuptake of extracellular serotonin in the synapse to terminate neurotransmission. The cryogenic electron microscopy structures of SERT bound to ibogaine and the physiological substrate serotonin resolved in different states have provided a glimpse of the functional conformations at atomistic resolution. However, the conformational dynamics and structural transitions to intermediate states are not fully understood. Furthermore, the molecular basis of how serotonin is recognized and transported remains unclear. In this study, we performed unbiased microsecond-long simulations of the human SERT to investigate the structural dynamics to various intermediate states and elucidated the complete substrate import pathway. Using Markov state models, we characterized a sequential order of conformational-driven ion-coupled substrate binding and transport events and calculated the free energy barriers of conformation transitions associated with the import mechanism. We find that the transition from the occluded to inward-facing state is the rate-limiting step for substrate import and that the substrate decreases the free energy barriers to achieve the inward-facing state. Our study provides insights on the molecular basis of dynamics-driven ion-substrate recognition and transport of SERT that can serve as a model for other closely related neurotransmitter transporters.


Subject(s)
Serotonin Plasma Membrane Transport Proteins , Serotonin , Biological Transport , Humans , Molecular Dynamics Simulation , Protein Conformation , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/chemistry , Serotonin Plasma Membrane Transport Proteins/metabolism
8.
Mol Pharmacol ; 101(2): 95-105, 2022 02.
Article in English | MEDLINE | ID: mdl-34866045

ABSTRACT

Folding-deficient mutants of solute carrier 6 (SLC6) family members have been linked to human diseases. The serotonin transporter [(SERT)/SLC6A4] is an important drug target in the treatment of depression, anxiety, and obsessive-compulsive disorders and-with structural information in several conformational states-one of the best understood transporters. Here, we surmised that thermal unfolding offered a glimpse on the folding energy landscape of SLC6 transporters. We carried out molecular dynamic (MD) simulations to understand the mechanistic basis for enhanced and reduced stability, respectively, of the thermostabilized variant SERT-Y110A/I291A/T439S, which had previously been used for crystallization of human SERT in the outward-facing state, and of the folding-deficient SERT-P601A/G602A. We also examined the hydrophobic mismatch caused by the absence of cholesterol to explore the contribution of cholesterol to protein stability. When compared with wild type SERT, the thermodynamic and kinetic stability of SERT-Y110A/I291A/T439S was enhanced. In the other instances, changes in these two components were not correlated: the mutations in SERT-P601A/G602A led to a drop in thermodynamic but an increase in kinetic stability. The divergence was even more pronounced after cholesterol depletion, which reduced thermodynamic stability but increased the kinetic stability of wild type SERT to a level comparable to that of SERT-Y110A/I291A/T439S. We conclude that the low cholesterol content of the endoplasmic reticulum facilitates progression of the folding trajectory by reducing the energy difference between folding intermediates and the native state. SIGNIFICANCE STATEMENT: Point mutations in solute carrier 6 (SLC6) family members cause folding diseases. The serotonin transporter [(SERT)/SLC6A4] is a target for antidepressants and the best understood SLC6. This study produced molecular dynamics simulations and examined thermal unfolding of wild type and mutant SERT variants to understand their folding energy landscape. In the folding-deficient SERT-P012A/G602A, changes in kinetic and thermodynamic stability were not correlated. Similarly, cholesterol depletion lowered thermodynamic but enhanced kinetic stability. These observations allow for rationalizing the action of pharmacochaperones.


Subject(s)
Cholesterol/metabolism , Mutation/genetics , Protein Unfolding/drug effects , Serotonin Plasma Membrane Transport Proteins/genetics , Serotonin Plasma Membrane Transport Proteins/metabolism , Thermodynamics , Antidepressive Agents/metabolism , Antidepressive Agents/pharmacology , Genetic Variation/genetics , HEK293 Cells , Humans , Kinetics , Molecular Dynamics Simulation , Protein Binding/physiology , Protein Stability/drug effects , Protein Structure, Secondary , Serotonin Plasma Membrane Transport Proteins/chemistry
9.
J Pharmacol Sci ; 148(3): 295-299, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35177208

ABSTRACT

Serotonin transporter (SERT) is a membrane transporter which terminates neurotransmission of serotonin through its reuptake. This transporter as well as its substrate have long drawn attention as a key mediator and drug target in a variety of diseases including mental disorders. Accordingly, its structural basis has been studied by X-ray crystallography to gain insights into a design of ligand with high affinity and high specificity over closely related transporters. Recent progress in structural biology including single particle cryo-EM have made big strides also in determination of the structures of human SERT in complex with its ligands. Moreover, rapid progress in machine learning such as deep learning accelerates computer-assisted drug design. Here, we would like to summarize recent progresses in our understanding of SERT using these two rapidly growing technologies, limitations, and future perspectives.


Subject(s)
Drug Design , Serotonin Plasma Membrane Transport Proteins , Computer Simulation , Crystallography, X-Ray , Deep Learning , Depressive Disorder, Major , Drug Design/methods , Drug Design/trends , Humans , Ligands , Serotonin Plasma Membrane Transport Proteins/chemistry
10.
Nature ; 532(7599): 334-9, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-27049939

ABSTRACT

The serotonin transporter (SERT) terminates serotonergic signalling through the sodium- and chloride-dependent reuptake of neurotransmitter into presynaptic neurons. SERT is a target for antidepressant and psychostimulant drugs, which block reuptake and prolong neurotransmitter signalling. Here we report X-ray crystallographic structures of human SERT at 3.15 Å resolution bound to the antidepressants (S)-citalopram or paroxetine. Antidepressants lock SERT in an outward-open conformation by lodging in the central binding site, located between transmembrane helices 1, 3, 6, 8 and 10, directly blocking serotonin binding. We further identify the location of an allosteric site in the complex as residing at the periphery of the extracellular vestibule, interposed between extracellular loops 4 and 6 and transmembrane helices 1, 6, 10 and 11. Occupancy of the allosteric site sterically hinders ligand unbinding from the central site, providing an explanation for the action of (S)-citalopram as an allosteric ligand. These structures define the mechanism of antidepressant action in SERT, and provide blueprints for future drug design.


Subject(s)
Serotonin Plasma Membrane Transport Proteins/chemistry , Serotonin Plasma Membrane Transport Proteins/metabolism , Allosteric Regulation/drug effects , Allosteric Site/drug effects , Antidepressive Agents/chemistry , Antidepressive Agents/metabolism , Antidepressive Agents/pharmacology , Citalopram/chemistry , Citalopram/metabolism , Citalopram/pharmacology , Crystallography, X-Ray , Dopamine Plasma Membrane Transport Proteins/chemistry , Drug Design , Extracellular Space/metabolism , Humans , Immunoglobulin Fab Fragments/immunology , Intracellular Space/metabolism , Ions/chemistry , Ions/metabolism , Ligands , Models, Molecular , Paroxetine/chemistry , Paroxetine/metabolism , Paroxetine/pharmacology , Protein Binding/drug effects , Protein Conformation/drug effects , Protein Stability , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/immunology , Structure-Activity Relationship
11.
Molecules ; 27(9)2022 May 06.
Article in English | MEDLINE | ID: mdl-35566327

ABSTRACT

Despite the recent promising results of MDMA (3,4-methylenedioxy-methamphetamine) as a psychotherapeutic agent and its history of misuse, little is known about its molecular mode of action. MDMA enhances monoaminergic neurotransmission in the brain and its valuable psychoactive effects are associated to a dual action on the 5-HT transporter (SERT). This drug inhibits the reuptake of 5-HT (serotonin) and reverses its flow, acting as a substrate for the SERT, which possesses a central binding site (S1) for antidepressants as well as an allosteric (S2) one. Previously, we characterized the spatial binding requirements for MDMA at S1. Here, we propose a structure-based mechanistic model of MDMA occupation and translocation across both binding sites, applying ensemble binding space analyses, electrostatic complementarity, and Monte Carlo energy perturbation theory. Computed results were correlated with experimental data (r = 0.93 and 0.86 for S1 and S2, respectively). Simulations on all hSERT available structures with Gibbs free energy estimations (ΔG) revealed a favourable and pervasive dual binding mode for MDMA at S2, i.e., adopting either a 5-HT or an escitalopram-like orientation. Intermediate ligand conformations were identified within the allosteric site and between the two sites, outlining an internalization pathway for MDMA. Among the strongest and more frequent interactions were salt bridges with Glu494 and Asp328, a H-bond with Thr497, a π-π with Phe556, and a cation-π with Arg104. Similitudes and differences with the allosteric binding of 5-HT and antidepressants suggest that MDMA may have a distinctive chemotype. Thus, our models may provide a framework for future virtual screening studies and pharmaceutical design and to develop hSERT allosteric compounds with a unique psychoactive MDMA-like profile.


Subject(s)
N-Methyl-3,4-methylenedioxyamphetamine , Serotonin Plasma Membrane Transport Proteins , Antidepressive Agents/chemistry , Citalopram/chemistry , Humans , Monte Carlo Method , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/chemistry , Selective Serotonin Reuptake Inhibitors/chemistry
12.
Biochem Biophys Res Commun ; 534: 583-589, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33243462

ABSTRACT

To elucidate the regulation of serotonin transporter (SERT) function via its membrane trafficking, we investigated the involvement of the ubiquitin E3 ligase HRD1 (HMG-CoA reductase degradation protein), which participates in endoplasmic reticulum (ER)-associated degradation (ERAD), in the functional regulation of SERT. Cells transiently expressing wild-type SERT or a SERT C-terminal deletion mutant (SERTΔCT), a SERT protein predicted to be misfolded, were used for experiments. Studies using HRD1-overexpressing or HRD1-knockdown cells demonstrated that HRD1 is involved in SERT proteolysis. Overexpression of HRD1 promoted SERT ubiquitination, the effect of which was augmented by treatment with the proteasome inhibitor MG132. Immunoprecipitation studies revealed that HRD1 interacts with SERT in the presence of MG132. In addition, HRD1 was intracellularly colocalized with SERT, especially with aggregates of SERTΔCT in the ER. HRD1 also affected SERT uptake activity in accordance with the expression levels of the SERT protein. These results suggest that HRD1 contributes to the membrane trafficking and functional regulation of SERT through its involvement in ERAD-mediated SERT degradation.


Subject(s)
Serotonin Plasma Membrane Transport Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , COS Cells , Chlorocebus aethiops , Endoplasmic Reticulum-Associated Degradation , Gene Knockdown Techniques , HEK293 Cells , Humans , Leupeptins/pharmacology , Proteasome Inhibitors/pharmacology , Protein Folding , Protein Interaction Domains and Motifs , Proteolysis , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Deletion , Serotonin Plasma Membrane Transport Proteins/chemistry , Serotonin Plasma Membrane Transport Proteins/genetics , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics , Ubiquitination/drug effects
13.
Chembiochem ; 22(2): 349-353, 2021 01 15.
Article in English | MEDLINE | ID: mdl-32840058

ABSTRACT

Neuropsychiatric disorders such as major depressive disorder (MDD) arise from a complex set of genetic and environmental factors. The serotonin transporter (SERT) is a key regulator of synaptic serotonin (5-HT), and its inhibition is an important pharmacological target for treating MDD. The SERT-linked polymorphic region (5-HTTLPR) contains two major variants (short and long) that have been implicated in modulating susceptibility to MDD by altering the level of expression of SERT. Both variants contain C-rich repeats that conform to consensus i-motif folding sequences. i-Motifs are quadruplex DNA structures that have been proposed to have a role in transcription regulation. With spectroscopic techniques, we demonstrate that both alleles are able to form i-motifs at acidic pH, and at neutral pH under conditions of molecular crowding. This highlights the potential for i-motif formation to contribute to transcriptional regulation of the serotonin transporter, with a potential role in the pathophysiology of neuropsychiatric disorders.


Subject(s)
Serotonin Plasma Membrane Transport Proteins/chemistry , G-Quadruplexes , Hydrogen-Ion Concentration , Oligonucleotides/chemistry , Oligonucleotides/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism
14.
Bioorg Med Chem Lett ; 30(4): 126882, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31889666

ABSTRACT

Development of highly effective, safe, and fast-acting anti-depressants is urgently required for the treatment of major depressive disorder. It has been suggested that targeting 5-HT2A and 5-HT2C in addition to inhibition of serotonin reuptake may be beneficial in generating anti-depressant agents with better pharmacology and less adverse effects. We have developed phthalazinone-based compounds that potently bind to 5-HT2A, 5-HT2C, and the serotonin transporter. The representative compounds 11j and 11l displayed strong binding affinities against these targets, and showed favorable toxicity profiles as determined by hERG binding and CYP inhibition assays. Furthermore, these compounds presented promising anti-depressant effects comparable to fluoxetine and also synergistic effects with fluoxetine in forced swimming test, which implicates these compounds can be developed to help the treatment of major depressive disorder.


Subject(s)
Antidepressive Agents/chemistry , Azoles/chemistry , Receptor, Serotonin, 5-HT2A/chemistry , Receptor, Serotonin, 5-HT2C/chemistry , Serotonin Plasma Membrane Transport Proteins/chemistry , Animals , Antidepressive Agents/metabolism , Antidepressive Agents/pharmacology , Behavior, Animal/drug effects , Drug Design , Fluoxetine/chemistry , Fluoxetine/pharmacology , Humans , Inhibitory Concentration 50 , Mice , Receptor, Serotonin, 5-HT2A/metabolism , Receptor, Serotonin, 5-HT2C/metabolism , Serotonin 5-HT2 Receptor Antagonists/chemistry , Serotonin 5-HT2 Receptor Antagonists/metabolism , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Serotonin Plasma Membrane Transport Proteins/metabolism , Selective Serotonin Reuptake Inhibitors/chemistry , Selective Serotonin Reuptake Inhibitors/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Structure-Activity Relationship
15.
Phys Chem Chem Phys ; 22(9): 5132-5144, 2020 Mar 07.
Article in English | MEDLINE | ID: mdl-32073004

ABSTRACT

Vilazodone is a novel antidepressant used for the treatment of major depressive disorder (MDD) with a primary action mechanism of inhibiting the human serotonin reuptake transporter (hSERT) and acting as a 5-HT1A receptor partial agonist. The interaction between vilazodone and the 5-HT1A receptor has been reported, however, the binding mode of vilazodone in the hSERT remains elusive. In the current study, to elucidate the molecular mechanism of vilazodone binding in the hSERT, the drug and its five analogs were docked into the hSERT crystal structure as initial conformations and were sampled by 400 ns molecular dynamics (MD) simulations. Through the analysis of the profiles of protein-ligand binding free energies, interaction fingerprints, and conformational rearrangements, the binding mode of vilazodone in the hSERT was revealed. As a result, unlike the classical antidepressants located in the S1 site of the hSERT, vilazodone adopted a linear pose in the binding pocket. Its arylpiperazine fragment occupies the central site (S1) and interacts with Y95, D98, I172, Y176, F335, F341, S438, and T439, while the indole fragment extends to the allosteric site (S2) via interacting with the ionic switch (R104/E403) between the two sites. The new insights obtained are not only helpful in understanding the binding mode of vilazodone in the hSERT, but also provide valuable guidance to the discovery of novel antidepressant drugs.


Subject(s)
Molecular Docking Simulation , Molecular Dynamics Simulation , Serotonin Plasma Membrane Transport Proteins/chemistry , Vilazodone Hydrochloride/chemistry , Allosteric Site , Antidepressive Agents/chemistry , Antidepressive Agents/metabolism , Binding Sites , Humans , Ligands , Protein Binding , Serotonin Plasma Membrane Transport Proteins/metabolism , Thermodynamics , Vilazodone Hydrochloride/metabolism
16.
Molecules ; 25(22)2020 Nov 13.
Article in English | MEDLINE | ID: mdl-33203055

ABSTRACT

Amphetamine derivatives have been used in a wide variety of pathologies because of their pharmacological properties as psychostimulants, entactogens, anorectics, and antidepressants. However, adverse cardiovascular effects (sympathomimetics) and substance abuse problems (psychotropic and hallucinogenic effects) have limited their use. 4-Methylthioamphetamine (MTA) is an amphetamine derivative that has shown to inhibit monoamine uptake and monoamine oxidase. However, the pharmacological characterization (neurochemical, behavioral, and safety) of its derivatives 4-ethylthioamphetamine (ETA) and 4-methylthio-phenil-2-butanamine (MT-But) have not been studied. In the current experiments, we show that ETA and MT-But do not increase locomotor activity and conditioned place preference with respect to MTA. At the neurochemical level, ETA and MT-But do not increase in vivo DA release in striatum, but ETA and MT-But affect the nucleus accumbens bioaccumulation of DA and DOPAC. Regarding cardiovascular effects, the administration of MTA and ETA increased the mean arterial pressure and only ETA significantly increases the heart rate. Our results show that the pharmacological and safety profiles of MTA are modulated by changing the methyl-thio group or the methyl group of the aminoethyl chain.


Subject(s)
3,4-Dihydroxyphenylacetic Acid/pharmacology , Amphetamine/pharmacology , Amphetamines/pharmacology , Corpus Striatum/drug effects , Dopamine/pharmacology , Nucleus Accumbens/drug effects , Amphetamines/chemistry , Animals , Behavior, Animal , Body Temperature , Ligands , Locomotion/drug effects , Male , Molecular Docking Simulation , Oxygen/chemistry , Rats , Rats, Sprague-Dawley , Serotonin Plasma Membrane Transport Proteins/chemistry
17.
J Biol Chem ; 293(10): 3510-3523, 2018 03 09.
Article in English | MEDLINE | ID: mdl-29352106

ABSTRACT

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.


Subject(s)
Cholesterol/metabolism , Lipid Bilayers/metabolism , Models, Molecular , Serotonin Plasma Membrane Transport Proteins/metabolism , Amino Acid Substitution , Binding Sites , Binding, Competitive , Biological Transport/drug effects , Cholesterol/chemistry , Conserved Sequence , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Kinetics , Ligands , Lipid Bilayers/chemistry , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Mutation , Protein Conformation , Protein Interaction Domains and Motifs , Protein Stability , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Serotonin Plasma Membrane Transport Proteins/chemistry , Serotonin Plasma Membrane Transport Proteins/genetics , beta-Cyclodextrins/chemistry , beta-Cyclodextrins/metabolism
18.
Anal Chem ; 91(17): 10970-10978, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31408320

ABSTRACT

Insight into the structure-function relationship of membrane proteins is important to understand basic cell function and inform drug development, as these are common targets for drugs. Hydrogen/deuterium exchange mass spectrometry (HDX-MS) is an established technique for the study of protein conformational dynamics and has shown compatibility with membrane proteins. However, the digestion and mass analysis of peptides from membrane proteins can be challenging, severely limiting the HDX-MS experiment. Here we compare the digestion of four integral membrane proteins-Cl-/H+ exchange transporter (ClC-ec1), leucine transporter (LeuT), dopamine transporter (DAT), and serotonin transporter (SERT)-by the use of porcine pepsin and three alternative aspartic proteases either in-solution or immobilized on-column in an optimized HDX-MS-compatible workflow. Pepsin was the most favorable for the digestion of ClC-ec1 and LeuT, providing coverage of 82.2 and 33.2% of the respective protein sequence; however, the alternative proteases surpassed pepsin for the digestion of DAT and SERT. By also screening quench solution additives, we observe that the denaturant urea was beneficial, resulting in improved sequence coverage of all membrane proteins, in contrast to guanidine hydrochloride. Furthermore, significant improvements in sequence coverage were achieved by tailoring the chromatography to handle hydrophobic peptides. Overall, we demonstrate that the susceptibility of membrane proteins to proteolytic digestion during HDX-MS is highly protein-specific. Our results highlight the importance of having multiple proteases and different quench buffer additives in the HDX-MS toolbox and the need to carefully screen a range of digestion conditions to successfully optimize the HDX-MS analysis of integral membrane proteins.


Subject(s)
Antiporters/analysis , Dopamine Plasma Membrane Transport Proteins/analysis , Drosophila Proteins/analysis , Escherichia coli Proteins/analysis , Hydrogen Deuterium Exchange-Mass Spectrometry/methods , Peptide Fragments/analysis , Serotonin Plasma Membrane Transport Proteins/analysis , Amino Acid Sequence , Animals , Antiporters/chemistry , Aquifex , Aspartic Acid Proteases/chemistry , Bacteria , Dopamine Plasma Membrane Transport Proteins/chemistry , Drosophila Proteins/chemistry , Drosophila melanogaster , Escherichia coli , Escherichia coli Proteins/chemistry , Humans , Models, Molecular , Pepsin A/chemistry , Proteolysis , Serotonin Plasma Membrane Transport Proteins/chemistry , Structure-Activity Relationship , Swine , Urea/chemistry
19.
Biochem Soc Trans ; 47(3): 861-874, 2019 06 28.
Article in English | MEDLINE | ID: mdl-31064865

ABSTRACT

Point mutations in the coding sequence for solute carrier 6 (SLC6) family members result in clinically relevant disorders, which are often accounted for by a loss-of-function phenotype. In many instances, the mutated transporter is not delivered to the cell surface because it is retained in the endoplasmic reticulum (ER). The underlying defect is improper folding of the transporter and is the case for many of the known dopamine transporter mutants. The monoamine transporters, i.e. the transporters for norepinephrine (NET/SLC6A2), dopamine (DAT/SLC6A3) and serotonin (SERT/SLC6A4), have a rich pharmacology; hence, their folding-deficient mutants lend themselves to explore the concept of pharmacological chaperoning. Pharmacochaperones are small molecules, which bind to folding intermediates with exquisite specificity and scaffold them to a folded state, which is exported from the ER and delivered to the cell surface. Pharmacochaperoning of mutant monoamine transporters, however, is not straightforward: ionic conditions within the ER are not conducive to binding of most typical monoamine transporter ligands. A collection of compounds exists, which are classified as atypical ligands because they trap monoamine transporters in unique conformational states. The atypical binding mode of some DAT inhibitors has been linked to their anti-addictive action. Here, we propose that atypical ligands and also compounds recently classified as partial releasers can serve as pharmacochaperones.


Subject(s)
Dopamine Plasma Membrane Transport Proteins/metabolism , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Animals , Dopamine Plasma Membrane Transport Proteins/chemistry , Endoplasmic Reticulum/metabolism , Humans , Molecular Chaperones/metabolism , Norepinephrine Plasma Membrane Transport Proteins/chemistry , Protein Conformation , Protein Folding , Serotonin Plasma Membrane Transport Proteins/chemistry
20.
Protein Expr Purif ; 164: 105479, 2019 12.
Article in English | MEDLINE | ID: mdl-31442583

ABSTRACT

The serotonin transporter belongs to the family of sodium-chloride coupled neurotransmitter transporter and is related to depression in humans. It is therefore an important drug target to support treatment of depression. Recently, structures of human serotonin transporter in complex with inhibitor molecules have been published. However, the production of large protein amounts for crystallization experiments remains a bottleneck. Here we present the possibility to obtain purified serotonin transporter from E. coli. Fos-choline 12 solubilized target protein was obtained with a purity of >95% and a yield of 1.2 mg L-1 culture in autoinduction medium. CD spectroscopic analysis of protein stability allowed identifying CHS and POPX as stabilizing components, which increased hSERT thermostability by 7 °C. The kinetic dissociation constant KD of 2.8 µM (±0.05) for of the inhibitor Desipramine was determined with a ka of 10,848 M - 1 s-1 (±220) and a kd of 0.03 s-1 (±4.7 × 10-5).


Subject(s)
Escherichia coli/genetics , Serotonin Plasma Membrane Transport Proteins/chemistry , Serotonin Plasma Membrane Transport Proteins/genetics , Amino Acid Sequence , Binding Sites , Cholesterol/chemistry , Gene Expression , Humans , Phospholipids/chemistry , Protein Stability , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Solubility
SELECTION OF CITATIONS
SEARCH DETAIL