Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
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
2.
Nature ; 630(8015): 247-254, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38750358

ABSTRACT

The noradrenaline transporter has a pivotal role in regulating neurotransmitter balance and is crucial for normal physiology and neurobiology1. Dysfunction of noradrenaline transporter has been implicated in numerous neuropsychiatric diseases, including depression and attention deficit hyperactivity disorder2. Here we report cryo-electron microscopy structures of noradrenaline transporter in apo and substrate-bound forms, and as complexes with six antidepressants. The structures reveal a noradrenaline transporter dimer interface that is mediated predominantly by cholesterol and lipid molecules. The substrate noradrenaline binds deep in the central binding pocket, and its amine group interacts with a conserved aspartate residue. Our structures also provide insight into antidepressant recognition and monoamine transporter selectivity. Together, these findings advance our understanding of noradrenaline transporter regulation and inhibition, and provide templates for designing improved antidepressants to treat neuropsychiatric disorders.


Subject(s)
Antidepressive Agents , Cryoelectron Microscopy , Norepinephrine Plasma Membrane Transport Proteins , Norepinephrine , Protein Multimerization , Humans , Antidepressive Agents/chemistry , Antidepressive Agents/metabolism , Antidepressive Agents/pharmacology , Apoproteins/chemistry , Apoproteins/metabolism , Apoproteins/ultrastructure , Aspartic Acid/chemistry , Aspartic Acid/metabolism , Binding Sites , Cholesterol/metabolism , Cholesterol/chemistry , Models, Molecular , Norepinephrine/metabolism , Norepinephrine/chemistry , Norepinephrine Plasma Membrane Transport Proteins/antagonists & inhibitors , Norepinephrine Plasma Membrane Transport Proteins/chemistry , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Norepinephrine Plasma Membrane Transport Proteins/ultrastructure , Protein Binding , Substrate Specificity
3.
Int J Mol Sci ; 22(16)2021 Aug 07.
Article in English | MEDLINE | ID: mdl-34445205

ABSTRACT

The neurobiology of schizophrenia is multifactorial, comprising the dysregulation of several biochemical pathways and molecules. This research proposes a peripheral biomarker for schizophrenia that involves the second extracellular loop of norepinephrine transporter (NEText), the tropomyosin receptor kinase C (TrkC), and the neurotrophin-3 (NT-3) in T cells. The study of NEText, NT-3, and TrkC was performed in T cells and plasma extracted from peripheral blood of 54 patients with schizophrenia and 54 healthy controls. Levels of NT-3, TrkC, and NET were significantly lower in plasma and T cells of patients compared to healthy controls. Co-immunoprecipitation (co-IPs) showed protein interactions with Co-IP NEText-NT-3 and Co-IP NEText-TrkC. Computational modelling of protein-peptide docking by CABS-dock provided a medium-high accuracy model for NT-3-NEText (4.6935 Å) and TrkC-NEText (2.1365 Å). In summary, immunocomplexes reached statistical relevance in the T cells of the control group contrary to the results obtained with schizophrenia. The reduced expression of NT-3, TrkC, and NET, and the lack of molecular complexes in T cells of patients with schizophrenia may lead to a peripheral dysregulation of intracellular signaling pathways and an abnormal reuptake of norepinephrine (NE) by NET. This peripheral molecular biomarker underlying schizophrenia reinforces the role of neurotrophins, and noradrenergic and immune systems in the pathophysiology of schizophrenia.


Subject(s)
Molecular Docking Simulation , Neurotrophin 3/chemistry , Norepinephrine Plasma Membrane Transport Proteins/chemistry , Receptor, trkC/chemistry , Schizophrenia/etiology , Adult , Biomarkers/metabolism , Female , Humans , Male , Middle Aged , Neurotrophin 3/genetics , Neurotrophin 3/metabolism , Norepinephrine Plasma Membrane Transport Proteins/genetics , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Protein Structure, Secondary , Receptor, trkC/genetics , Receptor, trkC/metabolism , Schizophrenia/genetics , Schizophrenia/metabolism
4.
Bioorg Med Chem ; 43: 116278, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34157571

ABSTRACT

Polymer-drug conjugates are growing in interest as novel anticancer agents for targeted cancer therapy. The aim of this study was to synthesize a poly(ethylene glycol) (PEG) conjugated anticancer drug for neuroblastoma, which is the most common extracranial solid tumor of childhood and the deadliest tumor of infancy. In our previous studies, we designed and synthesized a dual targeting agent using benzylguanidine (BG) conjugated with the high affinity thyrointegrin αvß3 antagonist TriAzole Tetraiodothyroacetic acid (TAT) via non-cleavable bonding to PEG400 to make BG-P400-TAT and its derivatives as agents against neuroblastoma. Here, we improved the pharmacodynamic properties and increased the solubility by changing the polymer length to 1600 molecular weight. The TAT group, which acts as an integrin αvß3 antagonist, and the BG group, which can be taken up by neuroblastoma cells through the norepinephrine transporter (NET) system, are conjugated to PEG1600 to make BG-PEG1600-TAT. The binding affinity of BG-PEG1600-TAT was 40-fold higher to integrin αvß3 versus BG-P400-TAT and was associated with greater anticancer activities against neuroblastoma cells (SK-N-F1 and SKNAS) implanted in SCID mice along with broad spectrum anti-angiogenesis activities versus the FDA approved anti-Vascular Endothelial Growth Factor (VEGF) monoclonal antibody Avastin (bevacizumab). In conclusion, our novel dual targeting of NET and αvß3 receptor antagonist, BG-P1600-TAT demonstrated broad spectrum anti-angiogenesis and anti-cancer activities in suppressing neuroblastoma tumor progression and metastasis. Thus, BG-PEG1600-TAT represents a potential clinical candidate for targeted therapy in neuroblastoma management.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery , Integrin alphaVbeta3/metabolism , Neuroblastoma/drug therapy , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Polyethylene Glycols/pharmacology , Antineoplastic Agents/chemistry , Dose-Response Relationship, Drug , Humans , Integrin alphaVbeta3/chemistry , Molecular Structure , Neuroblastoma/metabolism , Norepinephrine Plasma Membrane Transport Proteins/chemistry , Polyethylene Glycols/chemistry , Structure-Activity Relationship
5.
J Nanobiotechnology ; 18(1): 95, 2020 Jul 13.
Article in English | MEDLINE | ID: mdl-32660596

ABSTRACT

BACKGROUND: Currently, the diagnosis and treatment of neuroblastomas-the most frequent solid tumors in children-exploit the norepinephrine transporter (hNET) via radiolabeled norepinephrine analogs. We aim to develop a nanomedicine-based strategy towards precision therapy by targeting hNET cell-surface protein with hNET-derived homing peptides. RESULTS: The peptides (seq. GASNGINAYL and SLWERLAYGI) were shown to bind high-resolution homology models of hNET in silico. In particular, one unique binding site has marked the sequence and structural similarities of both peptides, while most of the contribution to the interaction was attributed to the electrostatic energy of Asn and Arg (< - 228 kJ/mol). The peptides were comprehensively characterized by computational and spectroscopic methods showing ~ 21% ß-sheets/aggregation for GASNGINAYL and ~ 27% α-helix for SLWERLAYGI. After decorating 12-nm ferritin-based nanovehicles with cysteinated peptides, both peptides exhibited high potential for use in actively targeted neuroblastoma nanotherapy with exceptional in vitro biocompatibility and stability, showing minor yet distinct influences of the peptides on the global expression profiles. Upon binding to hNET with fast binding kinetics, GASNGINAYLC peptides enabled rapid endocytosis of ferritins into neuroblastoma cells, leading to apoptosis due to increased selective cytotoxicity of transported payload ellipticine. Peptide-coated nanovehicles significantly showed higher levels of early apoptosis after 6 h than non-coated nanovehicles (11% and 7.3%, respectively). Furthermore, targeting with the GASNGINAYLC peptide led to significantly higher degree of late apoptosis compared to the SLWERLAYGIC peptide (9.3% and 4.4%, respectively). These findings were supported by increased formation of reactive oxygen species, down-regulation of survivin and Bcl-2 and up-regulated p53. CONCLUSION: This novel homing nanovehicle employing GASNGINAYLC peptide was shown to induce rapid endocytosis of ellipticine-loaded ferritins into neuroblastoma cells in selective fashion and with successful payload. Future homing peptide development via lead optimization and functional analysis can pave the way towards efficient peptide-based active delivery of nanomedicines to neuroblastoma cells.


Subject(s)
Drug Delivery Systems/methods , Endocytosis/genetics , Nanostructures/chemistry , Neuroblastoma/metabolism , Norepinephrine Plasma Membrane Transport Proteins , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Ferritins/chemistry , Humans , Nanomedicine , Norepinephrine Plasma Membrane Transport Proteins/chemistry , Norepinephrine Plasma Membrane Transport Proteins/genetics , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Peptides/chemistry , Peptides/genetics , Peptides/metabolism
6.
Nat Struct Mol Biol ; 26(7): 545-556, 2019 07.
Article in English | MEDLINE | ID: mdl-31270469

ABSTRACT

Monoamine transporters (MATs) regulate neurotransmission via the reuptake of dopamine, serotonin and norepinephrine from extra-neuronal regions and thus maintain neurotransmitter homeostasis. As targets of a wide range of compounds, including antidepressants, substances of abuse and drugs for neuropsychiatric and neurodegenerative disorders, their mechanism of action and their modulation by small molecules have long been of broad interest. Recent advances in the structural characterization of dopamine and serotonin transporters have opened the way for structure-based modeling and simulations, which, together with experimental data, now provide mechanistic understanding of their transport function and interactions. Here we review recent progress in the elucidation of the structural dynamics of MATs and their conformational landscape and transitions, as well as allosteric regulation mechanisms.


Subject(s)
Dopamine Plasma Membrane Transport Proteins/metabolism , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Allosteric Regulation/drug effects , Animals , Binding Sites/drug effects , Dopamine Plasma Membrane Transport Proteins/chemistry , Drug Discovery , Humans , Models, Molecular , Norepinephrine Plasma Membrane Transport Proteins/chemistry , Protein Conformation/drug effects , Serotonin Plasma Membrane Transport Proteins/chemistry
7.
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
8.
PLoS One ; 12(3): e0172070, 2017.
Article in English | MEDLINE | ID: mdl-28282374

ABSTRACT

In heart failure (HF), a disturbed cardiac norepinephrine (NE) homeostasis is characterized by depleted cardiac NE stores, impairment of the cardiac NE re-uptake by the neuronal norepinephrine transporter (NET) and enhanced cardiac NE net release. Reduced cardiac NE content appears to be caused by enhanced cardiac NE net release from sympathetic neurons in HF, triggered by neurohumoral activation. However, it remains unclear whether reduced NE itself has an impact on cardiac NE re-uptake, independent of neurohumoral activation. Here, we evaluated whether depletion of cardiac NE stores alone can regulate cardiac NE re-uptake. Treatment of Wistar rats with reserpine (5 mg/kg/d) for one (1d) or five days (5d) resulted in markedly reduced cardiac NE content, comparable to NE stores in experimental HF due to pressure overload. In order to assess cardiac NE re-uptake, the specific cardiac [3H]-NE uptake via the NET in a Langendorff preparation was measured. Reserpine treatment led to decreased NE re-uptake at 1d and 5d compared to saline treatment. Expression of tyrosine hydroxylase (TH), the rate-limiting enzyme of the NE synthesis, was elevated in left stellate ganglia after reserpine. Mechanistically, measurement of NET mRNA expression in left stellate ganglia and myocardial NET density revealed a post-transcriptional downregulation of the NET by reserpine. In summary, present data demonstrate that depletion of cardiac NE stores alone is sufficient to impair cardiac NE re-uptake via downregulation of the NET, independent of systemic neurohumoral activation. Knowledge about the regulation of the cardiac NE homeostasis may offer novel therapeutic strategies in HF.


Subject(s)
Catecholamines/metabolism , Myocardium/metabolism , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Animals , Down-Regulation/drug effects , Echocardiography , Heart/diagnostic imaging , Heart Failure/metabolism , Heart Failure/pathology , Heart Failure/therapy , Heart Rate , Male , Norepinephrine Plasma Membrane Transport Proteins/chemistry , Norepinephrine Plasma Membrane Transport Proteins/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Reserpine/pharmacology , Stellate Ganglion/metabolism , Stellate Ganglion/pathology , Tritium/chemistry , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
9.
Sci Rep ; 6: 26883, 2016 05 27.
Article in English | MEDLINE | ID: mdl-27230580

ABSTRACT

Selective norepinephrine reuptake inhibitors (sNRIs) provide an effective class of approved antipsychotics, whose inhibitory mechanism could facilitate the discovery of privileged scaffolds with enhanced drug efficacy. However, the crystal structure of human norepinephrine transporter (hNET) has not been determined yet and the inhibitory mechanism of sNRIs remains elusive. In this work, multiple computational methods were integrated to explore the inhibitory mechanism of approved sNRIs (atomoxetine, maprotiline, reboxetine and viloxazine), and 3 lines of evidences were provided to verify the calculation results. Consequently, a binding mode defined by interactions between three chemical moieties in sNRIs and eleven residues in hNET was identified as shared by approved sNRIs. In the meantime, binding modes of reboxetine's enantiomers with hNET were compared. 6 key residues favoring the binding of (S, S)-reboxetine over that of (R, R)-reboxetine were discovered. This is the first study reporting that those 11 residues are the common determinants for the binding of approved sNRIs. The identified binding mode shed light on the inhibitory mechanism of approved sNRIs, which could help identify novel scaffolds with improved drug efficacy.


Subject(s)
Antipsychotic Agents/chemistry , Molecular Dynamics Simulation , Morpholines/chemistry , Neurotransmitter Uptake Inhibitors/chemistry , Norepinephrine Plasma Membrane Transport Proteins/chemistry , Norepinephrine/chemistry , Amino Acid Motifs , Antipsychotic Agents/metabolism , Atomoxetine Hydrochloride/chemistry , Atomoxetine Hydrochloride/metabolism , Binding Sites , Humans , Maprotiline/chemistry , Maprotiline/metabolism , Molecular Docking Simulation , Morpholines/metabolism , Neurotransmitter Uptake Inhibitors/metabolism , Norepinephrine/metabolism , Norepinephrine Plasma Membrane Transport Proteins/antagonists & inhibitors , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Reboxetine , Stereoisomerism , Structural Homology, Protein , Thermodynamics , Viloxazine/chemistry , Viloxazine/metabolism
10.
PLoS One ; 11(2): e0148608, 2016.
Article in English | MEDLINE | ID: mdl-26863431

ABSTRACT

The aim of this study was to design, synthesize and validate a multifunctional antidepressant probe that is modified at two distinct positions. The purpose of these modifications was to allow covalent linkage of the probe to interaction partners, and decoration of probe-target complexes with fluorescent reporter molecules. The strategy for the design of such a probe (i.e., azidobupramine) was guided by the need for the introduction of additional functional groups, conveying the required properties while keeping the additional moieties as small as possible. This should minimize the risk of changing antidepressant-like properties of the new probe azidobupramine. To control for this, we evaluated the binding parameters of azidobupramine to known target sites such as the transporters for serotonin (SERT), norepinephrine (NET), and dopamine (DAT). The binding affinities of azidobupramine to SERT, NET, and DAT were in the range of structurally related and clinically active antidepressants. Furthermore, we successfully visualized azidobupramine-SERT complexes not only in SERT-enriched protein material but also in living cells stably overexpressing SERT. To our knowledge, azidobupramine is the first structural analogue of a tricyclic antidepressant that can be covalently linked to target structures and further attached to reporter molecules while preserving antidepressant-like properties and avoiding radioactive isotopes.


Subject(s)
Antidepressive Agents, Tricyclic/chemistry , Azepines/chemistry , Dopamine Plasma Membrane Transport Proteins/metabolism , Fluorescent Dyes/chemistry , Molecular Probes/chemistry , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Amines/chemistry , Antidepressive Agents, Tricyclic/chemical synthesis , Azepines/chemical synthesis , Binding Sites , Cell Line , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins/chemistry , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fluorescent Dyes/chemical synthesis , Gene Expression , Humans , Kinetics , Ligands , Molecular Probes/chemical synthesis , Norepinephrine/metabolism , Norepinephrine Plasma Membrane Transport Proteins/chemistry , Protein Binding , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/chemistry
11.
Bioorg Med Chem ; 23(19): 6418-26, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26337019

ABSTRACT

A series of 4-benzylpiperidine carboxamides were designed and synthesized, and tested for their dual (serotonin and norepinephrine) reuptake inhibition. The synthesis of 4-benzylpiperidine carboxamides involved two main steps: amidation and substitution. Derivatives with 3 carbon linker displayed better activity than with 2 carbon linker. 4-Biphenyl- and 2-naphthyl-substituted derivatives 7e and 7j showed greater dual reuptake inhibition than standard drug venlafaxine HCl.


Subject(s)
Amides/chemistry , Drug Design , Selective Serotonin Reuptake Inhibitors/chemical synthesis , Serotonin and Noradrenaline Reuptake Inhibitors/chemical synthesis , Amides/chemical synthesis , Amides/metabolism , HEK293 Cells , Humans , Norepinephrine Plasma Membrane Transport Proteins/chemistry , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Piperidines/chemistry , Protein Binding , Serotonin Plasma Membrane Transport Proteins/chemistry , Serotonin Plasma Membrane Transport Proteins/metabolism , Selective Serotonin Reuptake Inhibitors/chemistry , Selective Serotonin Reuptake Inhibitors/metabolism , Serotonin and Noradrenaline Reuptake Inhibitors/chemistry , Serotonin and Noradrenaline Reuptake Inhibitors/metabolism , Structure-Activity Relationship
12.
Bioorg Med Chem ; 23(15): 5000-5014, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26051602

ABSTRACT

Centrally acting noradrenaline reuptake inhibitor (NRI) is reportedly effective for patients with stress urinary incontinence (SUI) by increasing urethral closure in the clinical Phase IIa study with esreboxetine. Noradrenaline transporters are expressed in both central and peripheral nervous systems and the contribution of each site to efficacy has not been clarified. This report describes the development of a series of peripheral-selective 7-phenyl-1,4-oxazepane NRIs to investigate the contribution of the peripheral site to increasing urethral resistance in rats. (6S,7R)-1,4-Oxazepane derivative 7 exhibited noradrenaline transporter inhibition with high selectivity against inhibitions of serotonin and dopamine transporters. A replacement of hydroxyl with acetamide group contributed to enhancement of peripheral selectivity by increasing molecular polarity. Compound 12, N-{[(6S,7R)-7-(3,4-dichlorophenyl)-1,4-oxazepan-6-yl]methyl}acetamide 0.5 fumarate, which showed effectively no brain penetration in rats, increased urethral resistance in a dose-dependent manner and exhibited a maximal effect on par with esreboxetine. These results demonstrate that the urethral resistance-increasing effects of NRI in rats are mainly caused by the inhibition of noradrenaline transporters in the peripheral sites.


Subject(s)
Drug Design , Heterocyclic Compounds/chemistry , Serotonin and Noradrenaline Reuptake Inhibitors/chemical synthesis , Animals , Cerebral Cortex/metabolism , Dopamine Plasma Membrane Transport Proteins/chemistry , Dopamine Plasma Membrane Transport Proteins/metabolism , Female , Heterocyclic Compounds/chemical synthesis , Heterocyclic Compounds/therapeutic use , Humans , Molecular Conformation , Morpholines/therapeutic use , Norepinephrine Plasma Membrane Transport Proteins/chemistry , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Rats , Rats, Sprague-Dawley , Serotonin Plasma Membrane Transport Proteins/chemistry , Serotonin Plasma Membrane Transport Proteins/metabolism , Serotonin and Noradrenaline Reuptake Inhibitors/chemistry , Serotonin and Noradrenaline Reuptake Inhibitors/therapeutic use , Stereoisomerism , Structure-Activity Relationship , Urinary Incontinence, Stress/drug therapy
13.
J Biol Chem ; 290(17): 10814-27, 2015 Apr 24.
Article in English | MEDLINE | ID: mdl-25724654

ABSTRACT

The noradrenergic and p38 mitogen-activated protein kinase (p38 MAPK) systems are implicated in cocaine-elicited behaviors. Previously, we demonstrated a role for p38 MAPK-mediated norepinephrine transporter (NET) Thr(30) phosphorylation in cocaine-induced NET up-regulation (Mannangatti, P., Arapulisamy, O., Shippenberg, T. S., Ramamoorthy, S., and Jayanthi, L. D. (2011) J. Biol. Chem. 286, 20239-20250). The present study explored the functional interaction between p38 MAPK-mediated NET regulation and cocaine-induced behaviors. In vitro cocaine treatment of mouse prefrontal cortex synaptosomes resulted in enhanced NET function, surface expression, and phosphorylation. Pretreatment with PD169316, a p38 MAPK inhibitor, completely blocked cocaine-mediated NET up-regulation and phosphorylation. In mice, in vivo administration of p38 MAPK inhibitor SB203580 completely blocked cocaine-induced NET up-regulation and p38 MAPK activation in the prefrontal cortex and nucleus accumbens. When tested for cocaine-induced locomotor sensitization and conditioned place preference (CPP), mice receiving SB203580 on cocaine challenge day or on postconditioning test day exhibited significantly reduced cocaine sensitization and CPP. A transactivator of transcription (TAT) peptide strategy was utilized to test the involvement of the NET-Thr(30) motif. In vitro treatment of synaptosomes with TAT-NET-Thr(30) (wild-type peptide) completely blocked cocaine-mediated NET up-regulation and phosphorylation. In vivo administration of TAT-NET-Thr(30) peptide but not TAT-NET-T30A (mutant peptide) completely blocked cocaine-mediated NET up-regulation and phosphorylation. In the cocaine CPP paradigm, mice receiving TAT-NET-Thr(30) but not TAT-NET-T30A on postconditioning test day exhibited significantly reduced cocaine CPP. Following extinction, TAT-NET-Thr(30) when given prior to cocaine challenge significantly reduced reinstatement of cocaine CPP. These results demonstrate that the direct inhibition of p38 MAPK or the manipulation of NET-Thr(30) motif/phosphorylation via a TAT peptide strategy prevents cocaine-induced NET up-regulation, locomotor sensitization, and CPP, suggesting a role for Thr(30)-linked NET regulation in cocaine-elicited behaviors.


Subject(s)
Cocaine-Related Disorders/physiopathology , Conditioning, Psychological/physiology , Norepinephrine Plasma Membrane Transport Proteins/chemistry , Norepinephrine Plasma Membrane Transport Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cocaine/pharmacology , Cocaine-Related Disorders/psychology , Conditioning, Psychological/drug effects , Gene Products, tat/metabolism , Imidazoles/pharmacology , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Phosphorylation , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Synaptosomes/drug effects , Synaptosomes/metabolism , Threonine/chemistry , Up-Regulation/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
14.
Molecules ; 20(1): 1712-30, 2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25608857

ABSTRACT

Since the norepinephrine transporter (NET) is involved in a variety of diseases, the investigation of underlying dysregulation-mechanisms of the norepinephrine (NE) system is of major interest. Based on the previously described highly potent and selective NET ligand 1-(3-(methylamino)-1-phenylpropyl)-3-phenyl-1,3-dihydro-2H-benzimidaz- ol-2-one (Me@APPI), this paper aims at the development of several fluorinated methylamine-based analogs of this compound. The newly synthesized compounds were computationally evaluated for their interactions with the monoamine transporters and represent reference compounds for PET-based investigation of the NET.


Subject(s)
Computer Simulation , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Positron-Emission Tomography , Radiopharmaceuticals/chemical synthesis , Humans , Ligands , Norepinephrine Plasma Membrane Transport Proteins/chemistry , Radiopharmaceuticals/chemistry , Reference Standards , Sequence Alignment
15.
Biochem Biophys Res Commun ; 454(1): 25-9, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25305483

ABSTRACT

A ubiquitous feature of neurotransmitter transporters is the presence of short C-terminal PDZ binding motifs acting as important trafficking elements. Depending on their very C-terminal sequences, PDZ binding motifs are usually divided into at least three groups; however this classification has recently been questioned. To introduce a 3D aspect into transporter's PDZ motif similarities, we compared their interactions with the natural collection of all 13 PDZ domains of the largest PDZ binding protein MUPP1. The GABA, glycine and serotonin transporters showed unique binding preferences scattered over one or several MUPP1 domains. On the contrary, the dopamine and norepinephrine transporter PDZ motifs did not show any significant affinity to MUPP1 domains. Interestingly, despite their terminal sequence diversity all three GABA transporter PDZ motifs interacted with MUPP1 domain 7. These results indicate that similarities in binding schemes of individual transporter groups might exist. Results also suggest the existence of variable PDZ binding modes, allowing several transporters to interact with identical PDZ domains and potentially share interaction partners in vivo.


Subject(s)
Carrier Proteins/chemistry , Neurotransmitter Transport Proteins/chemistry , PDZ Domains , Amino Acid Sequence , Animals , Brain/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Dopamine Plasma Membrane Transport Proteins/chemistry , Dopamine Plasma Membrane Transport Proteins/genetics , Dopamine Plasma Membrane Transport Proteins/metabolism , GABA Plasma Membrane Transport Proteins/chemistry , GABA Plasma Membrane Transport Proteins/genetics , GABA Plasma Membrane Transport Proteins/metabolism , Glycine Plasma Membrane Transport Proteins/chemistry , Glycine Plasma Membrane Transport Proteins/genetics , Glycine Plasma Membrane Transport Proteins/metabolism , Membrane Proteins , Mice , Molecular Sequence Data , Neurotransmitter Transport Proteins/genetics , Neurotransmitter Transport Proteins/metabolism , Norepinephrine Plasma Membrane Transport Proteins/chemistry , Norepinephrine Plasma Membrane Transport Proteins/genetics , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Serotonin Plasma Membrane Transport Proteins/chemistry , Serotonin Plasma Membrane Transport Proteins/genetics , Serotonin Plasma Membrane Transport Proteins/metabolism
16.
ACS Chem Neurosci ; 5(9): 784-92, 2014 Sep 17.
Article in English | MEDLINE | ID: mdl-25003748

ABSTRACT

Discovery of new inhibitors of the plasmalemmal monoamine transporters (MATs) continues to provide pharmacotherapeutic options for depression, addiction, attention deficit disorders, psychosis, narcolepsy, and Parkinson's disease. The windfall of high-resolution MAT structural information afforded by X-ray crystallography has enabled the construction of credible computational models. Elucidation of lead compounds, creation of compound structure-activity series, and pharmacologic testing are staggering expenses that could be reduced by using a MAT computational model for virtual screening (VS) of structural libraries containing millions of compounds. Here, VS of the PubChem small molecule structural database using the S1 (primary substrate) ligand pocket of a serotonin transporter homology model yielded 19 prominent "hit" compounds. In vitro pharmacology of these VS hits revealed four structurally unique MAT substrate uptake inhibitors with high nanomolar affinity at one or more of the three MATs. In vivo characterization of three of these hits revealed significant activity in a mouse model of acute depression at doses that did not elicit untoward locomotor effects. This constitutes the first report of MAT inhibitor discovery using exclusively the primary substrate pocket as a VS tool. Novel-scaffold MAT inhibitors offer hope of new medications that lack the many classic adverse effects of existing antidepressant drugs.


Subject(s)
Computer Simulation , Models, Molecular , Serotonin Plasma Membrane Transport Proteins/chemistry , Serotonin Plasma Membrane Transport Proteins/metabolism , Animals , Antidepressive Agents/pharmacology , Dose-Response Relationship, Drug , Humans , Ligands , Mice , Norepinephrine Plasma Membrane Transport Proteins/chemistry , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Serotonin Agents/pharmacology
17.
J Med Chem ; 57(3): 1000-13, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-24494745

ABSTRACT

2-[(Diphenylmethyl)sulfinyl]acetamide (modafinil, (±)-1) is a unique dopamine uptake inhibitor that binds the dopamine transporter (DAT) differently than cocaine and may have potential for the treatment of psychostimulant abuse. To further investigate structural requirements for this divergent binding mode, novel thio- and sulfinylacetamide and ethanamine analogues of (±)-1 were synthesized wherein (1) the diphenyl rings were substituted with methyl, trifluoromethyl, and halogen substituents and (2) substituents were added to the terminal amide/amine nitrogen. Halogen substitution of the diphenyl rings of (±)-1 gave several amide analogues with improved binding affinity for DAT and robust selectivity over the serotonin transporter (SERT), whereas affinity improved at SERT over DAT for the p-halo-substituted amine analogues. Molecular docking studies, using a subset of analogues with DAT and SERT homology models, and functional data obtained with DAT (A480T) and SERT (T497A) mutants defined a role for TM10 in the substrate/inhibitor S1 binding sites of DAT and SERT.


Subject(s)
Benzhydryl Compounds/chemical synthesis , Dopamine Plasma Membrane Transport Proteins/metabolism , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Animals , Benzhydryl Compounds/chemistry , Benzhydryl Compounds/pharmacology , Binding Sites , Brain/metabolism , COS Cells , Chlorocebus aethiops , Dopamine Plasma Membrane Transport Proteins/chemistry , Dopamine Plasma Membrane Transport Proteins/genetics , Male , Modafinil , Molecular Docking Simulation , Mutation , Norepinephrine Plasma Membrane Transport Proteins/chemistry , Norepinephrine Plasma Membrane Transport Proteins/genetics , Protein Binding , Radioligand Assay , Rats , Rats, Sprague-Dawley , Serotonin Plasma Membrane Transport Proteins/chemistry , Serotonin Plasma Membrane Transport Proteins/genetics , Stereoisomerism , Structure-Activity Relationship , Thioacetamide/analogs & derivatives , Thioacetamide/chemical synthesis , Thioacetamide/chemistry , Thioacetamide/pharmacology
18.
Bioorg Med Chem Lett ; 23(24): 6920-6922, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24211020

ABSTRACT

Selective σ2 ligands continue to be an active target for medications to attenuate the effects of psychostimulants. In the course of our studies to determine the optimal substituents in the σ2-selective phenyl piperazines analogues with reduced activity at other neurotransmitter systems, we discovered that 1-(3-chlorophenyl)-4-phenethylpiperazine actually had preferentially increased affinity for dopamine transporters (DAT), yielding a highly selective DAT ligand.


Subject(s)
Dopamine Plasma Membrane Transport Proteins/chemistry , Ligands , Piperazines/chemistry , Dopamine Plasma Membrane Transport Proteins/metabolism , Norepinephrine Plasma Membrane Transport Proteins/chemistry , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Piperazines/metabolism , Protein Binding , Receptors, sigma/chemistry , Receptors, sigma/metabolism , Serotonin Plasma Membrane Transport Proteins/chemistry , Serotonin Plasma Membrane Transport Proteins/metabolism , Structure-Activity Relationship
19.
Neurochem Res ; 38(7): 1303-14, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23525969

ABSTRACT

The thiol reagent N-ethylmaleimide (NEM) is known to inhibit irreversibly ligand binding by the norepinephrine transporter (NET), while the simultaneous presence of NET substrates or ligands protects from this inhibition. Therefore, cysteine residues located within the substrate binding pocket of the NET were assumed to play an important role in ligand binding. To examine which (if any) of the 10 cysteines (Cys) of the human (h) NET might be involved in transport and/or binding function, we mutated all hNET cysteines to alanine. Using transfected HEK293 cells we studied NEM effects on the hNET with respect to [(3)H]nisoxetine binding. Two cysteines (Cys176 and Cys185) within the extracellular loop of the NET have been proposed to form a disulfide bond. We could demonstrate that this is of crucial importance as corresponding hNET mutants, in which these cysteines have been replaced, showed a lack of plasma membrane expression. However, due to their oxidized state in the native NET protein, Cys176 and Cys185 may not be targets for NEM. All other Cys-to-Ala hNET mutants were fully active and showed no change in inhibition of [(3)H]nisoxetine binding by NEM. These observations clearly exclude cysteines as being involved in hNET ligand binding. Since NEM also interacts with histidin (His), we mutated all 13 histidins of the hNET to alanine and examined the NET mutants in functional and binding assays. His222 within the large extracellular loop of the transporter was identified as an interaction partner of NEM since in the corresponding hNET mutant NEM exhibited a significantly reduced inhibitory potency. Furthermore, we could show that histidins in position 296, 370 and 372 are important for nisoxetine binding, while His220, 441, 598 and 599 are crucial for plasma membrane expression of the hNET.


Subject(s)
Cysteine/metabolism , Histidine/metabolism , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Ethylmaleimide/metabolism , Fluoxetine/analogs & derivatives , Fluoxetine/metabolism , HEK293 Cells , Humans , Mutagenesis, Site-Directed , Norepinephrine Plasma Membrane Transport Proteins/chemistry , Norepinephrine Plasma Membrane Transport Proteins/genetics , Radioligand Assay , Subcellular Fractions/metabolism
20.
ACS Chem Neurosci ; 4(2): 295-309, 2013 Feb 20.
Article in English | MEDLINE | ID: mdl-23421681

ABSTRACT

The amino acid compositions of the substrate binding pockets of the three human monoamine transporters are compared as is the orientation of the endogenous substrates, serotonin, dopamine, and norepinephrine, bound in these. Through a combination of homology modeling, induced fit dockings, molecular dynamics simulations, and uptake experiments in mutant transporters, we propose a common binding mode for the three substrates. The longitudinal axis of the substrates is similarly oriented with these, forming an ionic interaction between the ammonium group and a highly conserved aspartate, Asp98 (serotonin transporter, hSERT), Asp79 (dopamine transporter, hDAT), and Asp75 (norepinephrine transporter, hNET). The 6-position of serotonin and the para-hydroxyl groups of dopamine and norepinephrine were found to face Ala173 in hSERT, Gly153 in hDAT, and Gly149 in hNET. Three rotations of the substrates around the longitudinal axis were identified. In each mode, an aromatic hydroxyl group of the substrates occupied equivalent volumes of the three binding pockets, where small changes in amino acid composition explains the differences in selectivity. Uptake experiments support that the 5-hydroxyl group of serotonin and the meta-hydroxyl group norepinephrine and dopamine are placed in the hydrophilic pocket around Ala173, Ser438, and Thr439 in hSERT corresponding to Gly149, Ser419, Ser420 in hNET and Gly153 Ser422 and Ala423 in hDAT. Furthermore, hDAT was found to possess an additional hydrophilic pocket around Ser149 to accommodate the para-hydroxyl group. Understanding these subtle differences between the binding site compositions of the three transporters is imperative for understanding the substrate selectivity, which could eventually aid in developing future selective medicines.


Subject(s)
Dopamine Plasma Membrane Transport Proteins/chemistry , Dopamine/metabolism , Norepinephrine Plasma Membrane Transport Proteins/chemistry , Norepinephrine/metabolism , Serotonin Plasma Membrane Transport Proteins/chemistry , Serotonin/metabolism , Binding Sites , Cell Line , Dopamine Plasma Membrane Transport Proteins/metabolism , Humans , Models, Molecular , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Plasma Membrane Neurotransmitter Transport Proteins/chemistry , Plasma Membrane Neurotransmitter Transport Proteins/metabolism , Protein Binding , Serotonin Plasma Membrane Transport Proteins/metabolism , Structural Homology, Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...