Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
J Biol Chem ; 289(33): 23004-23019, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24973209

ABSTRACT

The serotonin transporter (SERT) plays a critical role in regulating serotonin signaling by mediating reuptake of serotonin from the extracellular space. The molecular and cellular mechanisms controlling SERT levels in the membrane remain poorly understood. To study trafficking of the surface resident SERT, two functional epitope-tagged variants were generated. Fusion of a FLAG-tagged one-transmembrane segment protein Tac to the SERT N terminus generated a transporter with an extracellular epitope suited for trafficking studies (TacSERT). Likewise, a construct with an extracellular antibody epitope was generated by introducing an HA (hemagglutinin) tag in the extracellular loop 2 of SERT (HA-SERT). By using TacSERT and HA-SERT in antibody-based internalization assays, we show that SERT undergoes constitutive internalization in a dynamin-dependent manner. Confocal images of constitutively internalized SERT demonstrated that SERT primarily co-localized with the late endosomal/lysosomal marker Rab7, whereas little co-localization was observed with the Rab11, a marker of the "long loop" recycling pathway. This sorting pattern was distinct from that of a prototypical recycling membrane protein, the ß2-adrenergic receptor. Furthermore, internalized SERT co-localized with the lysosomal marker LysoTracker and not with transferrin. The sorting pattern was further confirmed by visualizing internalization of SERT using the fluorescent cocaine analog JHC1-64 and by reversible and pulse-chase biotinylation assays showing evidence for lysosomal degradation of the internalized transporter. Finally, we found that SERT internalized in response to stimulation with 12-myristate 13-acetate co-localized primarily with Rab7- and LysoTracker-positive compartments. We conclude that SERT is constitutively internalized and that the internalized transporter is sorted mainly to degradation.


Subject(s)
Endosomes/metabolism , Lysosomes/metabolism , Proteolysis , Serotonin Plasma Membrane Transport Proteins/metabolism , Carcinogens/pharmacology , Endosomes/genetics , HEK293 Cells , Humans , Lysosomes/genetics , Protein Transport/drug effects , Protein Transport/physiology , Serotonin Plasma Membrane Transport Proteins/genetics , Tetradecanoylphorbol Acetate/pharmacology , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins
2.
J Biol Chem ; 288(38): 27534-27544, 2013 Sep 20.
Article in English | MEDLINE | ID: mdl-23884410

ABSTRACT

The dopamine transporter (DAT) is responsible for sequestration of extracellular dopamine (DA). The psychostimulant amphetamine (AMPH) is a DAT substrate, which is actively transported into the nerve terminal, eliciting vesicular depletion and reversal of DA transport via DAT. Here, we investigate the role of the DAT C terminus in AMPH-evoked DA efflux using cell-permeant dominant-negative peptides. A peptide, which corresponded to the last 24 C-terminal residues of DAT (TAT-C24 DAT) and thereby contained the Ca(2+)-calmodulin-dependent protein kinase IIα (CaMKIIα) binding domain and the PSD-95/Discs-large/ZO-1 (PDZ)-binding sequence of DAT, was made membrane-permeable by fusing it to the cell membrane transduction domain of the HIV-1 Tat protein (TAT-C24WT). The ability of TAT-C24WT but not a scrambled peptide (TAT-C24Scr) to block the CaMKIIα-DAT interaction was supported by co-immunoprecipitation experiments in heterologous cells. In heterologous cells, we also found that TAT-C24WT, but not TAT-C24Scr, decreased AMPH-evoked 1-methyl-4-phenylpyridinium efflux. Moreover, chronoamperometric recordings in striatum revealed diminished AMPH-evoked DA efflux in mice preinjected with TAT-C24WT. Both in heterologous cells and in striatum, the peptide did not further inhibit efflux upon KN-93-mediated inhibition of CaMKIIα activity, consistent with a dominant-negative action preventing binding of CaMKIIα to the DAT C terminus. This was further supported by the ability of a peptide with perturbed PDZ-binding sequence, but preserved CaMKIIα binding (TAT-C24AAA), to diminish AMPH-evoked DA efflux in vivo to the same extent as TAT-C24WT. Finally, AMPH-induced locomotor hyperactivity was attenuated following systemic administration of TAT-C24WT but not TAT-C24Scr. Summarized, our findings substantiate that DAT C-terminal protein-protein interactions are critical for AMPH-evoked DA efflux and suggest that it may be possible to target protein-protein interactions to modulate transporter function and interfere with psychostimulant effects.


Subject(s)
Amphetamine/pharmacology , Cell-Penetrating Peptides/pharmacology , Central Nervous System Stimulants/pharmacology , Corpus Striatum/metabolism , Dopamine Plasma Membrane Transport Proteins/pharmacology , Dopamine/metabolism , Amphetamine/adverse effects , Animals , Benzylamines/pharmacology , Cell-Penetrating Peptides/metabolism , Central Nervous System Stimulants/adverse effects , Dopamine Plasma Membrane Transport Proteins/pharmacokinetics , Humans , Male , Mice , Motor Activity/drug effects , PDZ Domains , Protein Kinase C-alpha/antagonists & inhibitors , Protein Kinase C-alpha/metabolism , Protein Kinase Inhibitors/pharmacology , Sulfonamides/pharmacology
3.
Bioorg Med Chem Lett ; 23(1): 323-6, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23168018

ABSTRACT

A novel fluorescent ligand was synthesized as a high-affinity, high specificity probe for visualizing the serotonin transporter (SERT). The rhodamine fluorophore was extended from an aniline substitution on the 5-position of the dihydroisobenzofuran ring of citalopram (2, 1-(3-(dimethylamino)propyl)-1-(4-fluorophenyl)-1,3-dihydroisobenzofuran-5-carbonitrile), using an ethylamino linker. The resulting rhodamine-labeled ligand 8 inhibited [(3)H]5-HT uptake in COS-7 cells (K(i)=225 nM) with similar potency to the tropane-based JHC 1-064 (1), but with higher specificity towards the SERT relative to the transporters for dopamine and norepinephrine. Visualization of the SERT with compound 8 was demonstrated by confocal microscopy in HEK293 cells stably expressing EGFP-SERT.


Subject(s)
Citalopram/analogs & derivatives , Fluorescent Dyes/chemistry , Rhodamines/chemistry , Serotonin Plasma Membrane Transport Proteins/metabolism , Animals , COS Cells , Chlorocebus aethiops , Citalopram/chemical synthesis , Citalopram/chemistry , Citalopram/pharmacology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Kinetics , Microscopy, Confocal , Protein Biosynthesis/drug effects , Rhodamines/chemical synthesis , Rhodamines/pharmacology , Serotonin Plasma Membrane Transport Proteins/genetics
4.
medRxiv ; 2023 Aug 29.
Article in English | MEDLINE | ID: mdl-37693601

ABSTRACT

Affective or mood disorders are a leading cause of disability worldwide. The serotonergic system has been heavily implicated in the complex etiology and serves as a therapeutic target. The serotonin transporter (SERT) is a major regulator of serotonin neurotransmission, yet the disease-relevance of impaired SERT function remains unknown. Here, we present the first identification and functional characterization of disruptive coding SERT variants found in patients with psychiatric diseases. In a unique cohort of 144 patients characterized by treatment-resistant chronic affective disorders with a lifetime history of electroconvulsive therapy, we identified two previously uncharacterized coding SERT variants: SERT-N217S and SERT-A500T. Both variants were significantly enriched in the patient cohort compared to GnomAD (SERT-N217S: OR = 151, P = 0.0001 and SERT-A500T: OR = 1348, P = 0.0022) and ethnicity-matched healthy controls (SERT-N217S: OR ≥ 17.7, P ≤ 0.013 and SERT-A500T: OR = ∞, P = 0.029). Functional investigations revealed that the mutations exert distinct perturbations to SERT function, but their overall effects converge on a partial loss-of-function molecular phenotype. Thus, the SERT-A500T variant compromises the catalytic activity, while SERT-N217S disrupts proper glycosylation of SERT with a resulting dominant-negative trafficking deficiency. Moreover, we demonstrate that the trafficking deficiency of SERT-N217S is amenable to pharmacochaperoning by noribogaine. Collectively, our findings describe the first disease-associated loss-of-function SERT variants and implicate serotonergic disturbances arising from SERT dysfunction as a risk factor for chronic affective disorders.

5.
J Biol Chem ; 285(35): 27289-27301, 2010 Aug 27.
Article in English | MEDLINE | ID: mdl-20551317

ABSTRACT

The dopamine transporter (DAT) mediates reuptake of released dopamine and is the target for psychostimulants, such as cocaine and amphetamine. DAT undergoes marked constitutive endocytosis, but little is known about the fate and sorting of the endocytosed transporter. To study DAT sorting in cells lines, we fused the one-transmembrane segment protein Tac to DAT, thereby generating a transporter (TacDAT) with an extracellular antibody epitope suited for trafficking studies. TacDAT was functional and endocytosed constitutively in HEK293 cells. According to an ELISA-based assay, TacDAT intracellular accumulation was increased by the lysosomal protease inhibitor leupeptin and by monensin, an inhibitor of lysosomal degradation and recycling. Monensin also reduced TacDAT surface expression consistent with partial recycling. In both HEK293 cells and in the dopaminergic cell line 1Rb3An27, constitutively internalized TacDAT displayed primary co-localization with the late endosomal marker Rab7, less co-localization with the "short loop" recycling marker Rab4, and little co-localization with the marker of "long loop" recycling endosomes, Rab11. Removal by mutation of N-terminal ubiquitination sites did not affect this sorting pattern. The sorting pattern was distinct from a bona fide recycling membrane protein, the beta(2)-adrenergic receptor, that co-localized primarily with Rab11 and Rab4. Constitutively internalized wild type DAT probed with the fluorescently tagged cocaine analogue JHC 1-64, exhibited the same co-localization pattern as TacDAT in 1Rb3An27 cells and in cultured midbrain dopaminergic neurons. We conclude that DAT is constitutively internalized and sorted in a ubiquitination-independent manner to late endosomes/lysosomes and in part to a Rab4 positive short loop recycling pathway.


Subject(s)
Dopamine Plasma Membrane Transport Proteins/metabolism , Endocytosis/physiology , Endosomes/metabolism , Mesencephalon/metabolism , Neurons/metabolism , Cell Line , Dopamine Plasma Membrane Transport Proteins/genetics , Endosomes/genetics , Humans , Lysosomes/genetics , Lysosomes/metabolism , Mesencephalon/cytology , Neurons/cytology , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , Protein Structure, Tertiary , Protein Transport/physiology , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-2/metabolism , Ubiquitination/genetics , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , rab4 GTP-Binding Proteins/genetics , rab4 GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins
6.
J Neurochem ; 113(1): 27-41, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20085610

ABSTRACT

The dopamine transporter (DAT) plays a key role in regulating dopaminergic signalling in the brain by mediating rapid clearance of dopamine from the synaptic clefts. The psychostimulatory actions of cocaine and amphetamine are primarily the result of a direct interaction of these compounds with DAT leading to attenuated dopamine clearance and for amphetamine even increased dopamine release. In the last decade, intensive efforts have been directed towards understanding the molecular and cellular mechanisms governing the activity and availability of DAT in the plasma membrane of the pre-synaptic neurons. This has led to the identification of a plethora of different kinases, receptors and scaffolding proteins that interact with DAT and hereby either modulate the catalytic activity of the transporter or regulate its trafficking and degradation. Several new tools for studying DAT regulation in live cells have also recently become available such as fluorescently tagged cocaine analogues and fluorescent substrates. Here we review the current knowledge about the role of protein-protein interactions in DAT regulation as well as we describe the most recent methodological developments that have been established to overcome the challenges associated with the study of DAT in endogenous systems.


Subject(s)
Dopamine Plasma Membrane Transport Proteins/physiology , Dopamine/metabolism , Fluorescent Antibody Technique/methods , Fluorescent Dyes/metabolism , Protein Processing, Post-Translational , Animals , Brain/cytology , Dopamine Agents/pharmacology , Dopamine Plasma Membrane Transport Proteins/chemistry , Electric Conductivity , Ion Transport/drug effects , Ion Transport/physiology , Models, Biological , Neurons/drug effects , Neurons/metabolism , Protein Conformation , Protein Processing, Post-Translational/drug effects , Protein Transport/drug effects , Protein Transport/physiology , Receptors, G-Protein-Coupled/metabolism , Ubiquitination/physiology , alpha-Synuclein/metabolism
7.
Biol Psychiatry ; 85(1): 13-24, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30144930

ABSTRACT

The diagnosis of schizophrenia rests on clinical criteria that cannot be assessed in animal models. Together with absence of a clear underlying pathology and understanding of what causes schizophrenia, this has hindered development of informative animal models. However, recent large-scale genomic studies have identified copy number variants (CNVs) that confer high risk of schizophrenia and have opened a new avenue for generation of relevant animal models. Eight recurrent CNVs have reproducibly been shown to increase the risk of schizophrenia by severalfold: 22q11.2(del), 15q13.3(del), 1q21(del), 1q21(dup), NRXN1(del), 3q29(del), 7q11.23(dup), and 16p11.2(dup). Five of these CNVs have been modeled in animals, mainly mice, but also rats, flies, and zebrafish, and have been shown to recapitulate behavioral and electrophysiological aspects of schizophrenia. Here, we provide an overview of the schizophrenia-related phenotypes found in animal models of schizophrenia high-risk CNVs. We also discuss strengths and limitations of the CNV models, and how they can advance our biological understanding of mechanisms that can lead to schizophrenia and can be used to develop new and better treatments for schizophrenia.


Subject(s)
Psychotic Disorders , Schizophrenia , Animals , Calcium-Binding Proteins , DNA Copy Number Variations , Dopamine , Mice , Models, Animal , Neural Cell Adhesion Molecules , Positron-Emission Tomography , Rats
8.
Neurochem Int ; 73: 107-12, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24462583

ABSTRACT

The serotonin transporter (SERT) terminates serotonergic signaling and enables refilling of synaptic vesicles by mediating reuptake of serotonin (5-HT) released into the synaptic cleft. The molecular and cellular mechanisms controlling SERT activity and surface expression are not fully understood. Here we demonstrate that the substrate 5-HT itself causes acute down-regulation of SERT cell surface expression. To assess surface SERT expression by ELISA, we used a SERT variant (TacSERT) where the N-terminus of SERT was fused to the intracellular tail of the extracellularly FLAG-tagged single-membrane spanning protein Tac. In stably transfected HEK293 cells, 5-HT caused a dose-dependent reduction in TacSERT surface signal with an EC50 value equivalent to the Km value observed for 5-HT uptake. The 5-HT-induced reduction in surface signal reached maximum within 40-60min and was blocked by the selective SERT inhibitor S-citalopram. 5-HT-induced reduction in SERT expression was further supported by surface biotinylation experiments showing 5-HT-induced reduction in wild type SERT plasma membrane levels. Moreover, preincubation with 5-HT lowered the Vmax for 5-HT uptake in cultured raphe serotonergic neurons, indicting that endogenous cell-surface resident SERT likewise is down-regulated in the presence of substrate.


Subject(s)
Membrane Proteins/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Serotonin/pharmacology , Biotinylation , Down-Regulation/drug effects , HEK293 Cells , Humans , Kinetics , Neurons/drug effects , Neurons/metabolism , Primary Cell Culture , Raphe Nuclei/cytology , Raphe Nuclei/drug effects , Raphe Nuclei/metabolism , Transfection
9.
ACS Med Chem Lett ; 5(6): 696-9, 2014 Jun 12.
Article in English | MEDLINE | ID: mdl-24944746

ABSTRACT

Novel rhodamine-labeled ligands, based on (S)-citalopram, were synthesized and evaluated for uptake inhibition at the human serotonin, dopamine, and norepinephrine transporters (hSERT, hDAT, and hNET, respectively) and for binding at SERT, in transiently transfected COS7 cells. Compound 14 demonstrated high affinity binding and selectivity for SERT (K i = 3 nM). Visualization of SERT, using confocal laser scanning microscopy, validated compound 14 as a novel tool for studying SERT expression and distribution in living cells.

SELECTION OF CITATIONS
SEARCH DETAIL