Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Naunyn Schmiedebergs Arch Pharmacol ; 390(1): 15-24, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27650729

ABSTRACT

3,4-Methylenedioxy-N-methylamphetamine (MDMA) has been shown to be effective in the treatment of post-traumatic stress disorder (PTSD) in numerous clinical trials. In the present study, we have characterized the neurochemical binding profiles of three MDMA-benzofuran analogues (1-(benzofuran-5-yl)-propan-2-amine, 5-APB; 1-(benzofuran-6-yl)-N-methylpropan-2-amine, 6-MAPB; 1-(benzofuran-5-yl)-N-methylpropan-2-amine, 5-MAPB) and one MDMA-indole analogue (1-(1H-indol-5-yl)-2-methylamino-propan-1-ol, 5-IT). These compounds were screened as potential second-generation anti-PTSD drugs, against a battery of human and non-human receptors, transporters, and enzymes, and their potencies as 5-HT2 receptor agonist and monoamine uptake inhibitors determined. All MDMA analogues displayed high binding affinities for 5-HT2a,b,c and NEα2 receptors, as well as significant 5-HT, DA, and NE uptake inhibition. 5-APB revealed significant agonist activity at the 5-HT2a,b,c receptors, while 6-MAPB, 5-MAPB, and 5-IT exhibited significant agonist activity at the 5-HT2c receptor. There was a lack of correlation between the results of functional uptake and the monoamine transporter binding assay. MDMA analogues emerged as potent and selective monoamine oxidase A inhibitors. Based on 6-MAPB favorable pharmacological profile, it was further subjected to IC50 determination for monoamine transporters. Overall, all MDMA analogues displayed higher monoamine receptor/transporter binding affinities and agonist activity at the 5-HT2a,c receptors as compared to MDMA.


Subject(s)
Benzofurans/metabolism , Indoles/metabolism , N-Methyl-3,4-methylenedioxyamphetamine/metabolism , Neurotransmitter Uptake Inhibitors/metabolism , Receptors, Serotonin, 5-HT2/metabolism , Serotonin 5-HT2 Receptor Agonists/metabolism , Stress Disorders, Post-Traumatic/drug therapy , Vesicular Monoamine Transport Proteins/metabolism , Benzofurans/chemistry , Benzofurans/pharmacology , Binding Sites , Catechol O-Methyltransferase/chemistry , Catechol O-Methyltransferase/metabolism , Humans , Indoles/chemistry , Indoles/pharmacology , Monoamine Oxidase/chemistry , Monoamine Oxidase/metabolism , Monoamine Oxidase Inhibitors/chemistry , Monoamine Oxidase Inhibitors/metabolism , Monoamine Oxidase Inhibitors/pharmacology , N-Methyl-3,4-methylenedioxyamphetamine/chemistry , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Neurotransmitter Uptake Inhibitors/chemistry , Neurotransmitter Uptake Inhibitors/pharmacology , Protein Binding , Protein Conformation , Radioligand Assay , Receptors, Serotonin, 5-HT2/chemistry , Receptors, Serotonin, 5-HT2/drug effects , Serotonin 5-HT2 Receptor Agonists/chemistry , Serotonin 5-HT2 Receptor Agonists/pharmacology , Stress Disorders, Post-Traumatic/metabolism , Structure-Activity Relationship , Tyrosine 3-Monooxygenase/chemistry , Tyrosine 3-Monooxygenase/metabolism , Vesicular Monoamine Transport Proteins/antagonists & inhibitors , Vesicular Monoamine Transport Proteins/chemistry
2.
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
3.
ChemMedChem ; 10(6): 1027-39, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25899387

ABSTRACT

We herein present label-free, mass-spectrometry-based binding assays (MS Binding Assays) for the human dopamine, norepinephrine, and serotonin transporters (hDAT, hNET, and hSERT). Using this approach both enantiomers of the triple reuptake inhibitor indatraline as well as its cis-configured diastereomer were investigated toward hDAT, hNET, and hSERT in saturation experiments. The dissociation rate constants for (1R,3S)-indatraline binding at hDAT, hNET, and hSERT were determined in kinetic studies. These experiments revealed an allosteric effect of clomipramine on the dissociation of (1R,3S)-indatraline from hSERT. Finally, a comprehensive set of known monoamine transport inhibitors and substrates was studied in competition experiments at hDAT, hNET, and hSERT, using (1R,3S)-indatraline as nonlabeled marker. The results are in excellent agreement with those reported for radioligand binding assays. Therefore, the established MS Binding Assays are a promising alternative to the latter for the characterization of new monoamine reuptake inhibitors at DAT, NET, and SERT.


Subject(s)
Indans/metabolism , Methylamines/metabolism , Neurotransmitter Uptake Inhibitors/metabolism , Tandem Mass Spectrometry/methods , Vesicular Monoamine Transport Proteins/metabolism , Binding, Competitive , Half-Life , Humans , Radioligand Assay
4.
PLoS One ; 8(9): e74891, 2013.
Article in English | MEDLINE | ID: mdl-24098676

ABSTRACT

Multimodal analgesia is designed to optimize pain relief by coadministering drugs with distinct mechanisms of action or by combining multiple pharmacologies within a single molecule. In clinical settings, combinations of monoamine reuptake inhibitors and opioid receptor agonists have been explored and one currently available analgesic, tapentadol, functions as both a µ-opioid receptor agonist and a norepinephrine transporter inhibitor. However, it is unclear whether the combination of selective norepinephrine reuptake inhibition and µ-receptor agonism achieves an optimal antinociceptive synergy. In this study, we assessed the pharmacodynamic interactions between morphine and monoamine reuptake inhibitors that possess different affinities and selectivities for norepinephrine and serotonin transporters. Using the rat formalin model, in conjunction with measurements of ex vivo transporter occupancy, we show that neither the norepinephrine-selective inhibitor, esreboxetine, nor the serotonin-selective reuptake inhibitor, fluoxetine, produce antinociceptive synergy with morphine. Atomoxetine, a monoamine reuptake inhibitor that achieves higher levels of norepinephrine than serotonin transporter occupancy, exhibited robust antinociceptive synergy with morphine. Similarly, a fixed-dose combination of esreboxetine and fluoxetine which achieves comparable levels of transporter occupancy potentiated the antinociceptive response to morphine. By contrast, duloxetine, a monoamine reuptake inhibitor that achieves higher serotonin than norepinephrine transporter occupancy, failed to potentiate the antinociceptive response to morphine. However, when duloxetine was coadministered with the 5-HT3 receptor antagonist, ondansetron, potentiation of the antinociceptive response to morphine was revealed. These results support the notion that inhibition of both serotonin and norepinephrine transporters is required for monoamine reuptake inhibitor and opioid-mediated antinociceptive synergy; yet, excess serotonin, acting via 5-HT3 receptors, may reduce the potential for synergistic interactions. Thus, in the rat formalin model, the balance between norepinephrine and serotonin transporter inhibition influences the degree of antinociceptive synergy observed between monoamine reuptake inhibitors and morphine.


Subject(s)
Analgesia/methods , Morphine/metabolism , Neurotransmitter Uptake Inhibitors/metabolism , Nociceptive Pain/drug therapy , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Animals , Atomoxetine Hydrochloride , Biogenic Monoamines/metabolism , Chromatography, Liquid , Drug Synergism , Duloxetine Hydrochloride , Fluoxetine , Formaldehyde , Morpholines , Neurotransmitter Uptake Inhibitors/pharmacokinetics , Ondansetron , Propylamines , Rats , Rotarod Performance Test , Tandem Mass Spectrometry , Thiophenes
5.
Curr Med Chem ; 20(38): 4853-8, 2013.
Article in English | MEDLINE | ID: mdl-24083608

ABSTRACT

We summarize the alterations of classical neurotransmitters and neuropeptides and the corresponding subreceptors involved in major depression. Neuronal circuits in the brainstem, hippocampus and hypothalamus are developed, since they can be used to derive a multimodal pharmacotherapy. In this sense, serotonin hypoactivity could occur through a strong presynaptic inhibition of glutaminergic neurons via the subtype 5 of metabotropic glutaminergic receptors, and noradrenaline hypoactivity could be due to an enhanced presynaptic inhibition of GABAergic neurons via GABAB receptors. In the hippocampus, dopamine hypoactivity leads to a decreased positive effect. In clinical trials, the antidepressant effect of drugs interfering with the mentioned subreceptors, for example the triple reuptake inhibitor amitifadine, is being investigated. Moreover, the alterations of neuropeptides, such as corticotropin-releasing hormone, neuropeptide Y and galanin are pointed out. The additional antidepressant effect of analogs, agonists and antagonists of the mentioned neuropeptides should be examined.


Subject(s)
Antidepressive Agents/therapeutic use , Depression/drug therapy , Neuropeptides/metabolism , Depression/metabolism , Depression/pathology , GABAergic Neurons/metabolism , Hippocampus/metabolism , Humans , Neurotransmitter Agents/chemistry , Neurotransmitter Agents/metabolism , Neurotransmitter Agents/therapeutic use , Neurotransmitter Uptake Inhibitors/chemistry , Neurotransmitter Uptake Inhibitors/metabolism , Neurotransmitter Uptake Inhibitors/therapeutic use , Receptors, GABA/metabolism
6.
Drug Metab Lett ; 7(1): 23-33, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23826879

ABSTRACT

Amitifadine (EB-1010, formerly DOV 21,947) is a serotonin-preferring triple reuptake inhibitor that is a drug candidate for major depressive disorder. We investigated several relevant biopharmaceutic and drug-like characteristics of amitifadine using in vitro methodology and additionally determined the in vivo brain to plasma ratio of the drug in rats. Amitifadine was highly plasma protein bound with over 99% of drug bound to human plasma proteins. Using Caco-2 cell lines, amitifadine was bidirectionally highly permeable and showed no evidence of active secretion. Amitifadine was metabolized slowly by human hepatocytes and the major metabolite was the lactam EB-10101. In vitro studies using human liver microsomes demonstrated that EB-10101 was formed by monoamine oxidase A (MAO-A) and a NADPHdependent enzyme, possibly a cytochrome P450 (CYP) isoform. Amitifadine was a moderate inhibitor of the human isoforms of the major drug metabolizing enzymes CYP2D6, CYP3A4, CYP2C9, and CYP2C19 (IC50 = 9 - 100 µM), but was a potent inhibitor of human CYP2B6 (IC50 = 1.8 µM). The brain to plasma ratio for amitifadine varied from 3.7 - 6.5 at various time points, indicating preferential partitioning into rat brain versus plasma. The low affinity for the major drug metabolizing CYP enzymes and metabolism by multiple pathways may reduce pharmacokinetic drug-drug interactions and effects of enzyme polymorphisms. Overall, these studies suggest that amitifadine has drug-like characteristics favorable for drug development.


Subject(s)
Antidepressive Agents/pharmacokinetics , Aza Compounds/pharmacokinetics , Brain/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Neurotransmitter Uptake Inhibitors/pharmacokinetics , Animals , Antidepressive Agents/blood , Antidepressive Agents/metabolism , Aryl Hydrocarbon Hydroxylases , Aza Compounds/blood , Aza Compounds/metabolism , Biopharmaceutics , Blood Proteins/metabolism , Bridged Bicyclo Compounds, Heterocyclic/blood , Bridged Bicyclo Compounds, Heterocyclic/metabolism , Caco-2 Cells , Cell Membrane Permeability , Cytochrome P-450 CYP2C19 , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/metabolism , Enzyme Inhibitors/pharmacology , FMN Reductase/metabolism , Hepatocytes/metabolism , Humans , Lactams/metabolism , Male , Microsomes, Liver/metabolism , Monoamine Oxidase/metabolism , NADP/physiology , Neurotransmitter Uptake Inhibitors/blood , Neurotransmitter Uptake Inhibitors/metabolism , Protein Binding , Rats , Rats, Sprague-Dawley
7.
Curr Top Microbiol Immunol ; 364: 139-57, 2013.
Article in English | MEDLINE | ID: mdl-23239352

ABSTRACT

Seven serologically distinct botulinum neurotoxins and tetanus neurotoxin which cause the diseases botulism and tetanus constitute the clostridial neurotoxin family. Like many other bacterial protein toxins they exhibit a modular structure. One domain mediates highly specific binding to target cells and endocytosis, while the second translocates the third, a catalytic domain across the endosomal membrane to the target cell cytosol. In case of Clostridial neurotoxins (CNT), the latter acts as extremely specific Zn(2+)-dependent metalloproteinase. The various serotypes proteolyze each one particular peptide bond in one of the three SNARE proteins, which are the core of the membrane fusion apparatus for synaptic vesicles. SNARE cleavage causes the blockade of neurotransmitter release. This chapter details the molecular basis for the highly selective substrate recognition and cleavage mechanism of CNT.


Subject(s)
Neurotransmitter Uptake Inhibitors/metabolism , Proteolysis , SNARE Proteins/metabolism , Synaptic Transmission , Tetanus Toxin/metabolism , Amino Acid Sequence , Animals , Catalytic Domain , Clostridium/metabolism , Endocytosis , Enzyme Activation , Exocytosis , Humans , Hydrolysis , Molecular Sequence Data , Neurons/metabolism , Protein Binding , Protein Transport , Sequence Homology, Amino Acid , Substrate Specificity , Synaptic Membranes/metabolism , Synaptic Vesicles/metabolism , Zinc/metabolism
8.
Eur J Med Chem ; 54: 123-36, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22608762

ABSTRACT

A series of arylalkanol-piperidine derivatives was synthesized, and their triple reuptake inhibition and in vivo activities have been evaluated. Among them, compounds 2a, 2j, 2k, 2m and 2n exhibited high potency for 5-HT, NA and DA transporters. Optimized compounds 2j and 2m showed significant reduction of immobility time compared to that of vehicle in the mouse tail suspension test (TST) test at doses ranging from 10 to 50 mg/kg po, and were not generally motor stimulants at 50 mg/kg dose. In addition, compounds 2j and 2m displayed desirable pharmacokinetic properties in SD rats.


Subject(s)
Antidepressive Agents/chemistry , Antidepressive Agents/pharmacology , Neurotransmitter Uptake Inhibitors/chemistry , Neurotransmitter Uptake Inhibitors/pharmacology , Piperidines/chemistry , Piperidines/pharmacology , Animals , Antidepressive Agents/metabolism , Antidepressive Agents/pharmacokinetics , Chemistry Techniques, Synthetic , Male , Mice , Neurotransmitter Uptake Inhibitors/metabolism , Neurotransmitter Uptake Inhibitors/pharmacokinetics , Piperidines/metabolism , Piperidines/pharmacokinetics , Rats , Rats, Sprague-Dawley , Receptors, Biogenic Amine/metabolism
9.
J Nucl Med ; 52(7): 1150-5, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21680689

ABSTRACT

UNLABELLED: SEP-225289 is a novel compound that, based on in vitro potencies for transporter function, potentially inhibits reuptake at dopamine, norepinephrine, and serotonin transporters. An open-label PET study was conducted during the development of SEP-225289 to investigate its dopamine and serotonin transporter occupancy. METHODS: Different single doses of SEP-225289 were administered to healthy volunteers in 3 cohorts: 8 mg (n = 7), 12 mg (n = 5), and 16 mg (n = 7). PET was performed before and approximately 24 h after oral administration of SEP-225289, to assess occupancy at trough levels. Dopamine and serotonin transporter occupancies were estimated from PET using (11)C-N-(3-iodoprop-2E-enyl)-2ß-carbomethoxy-3ß-(4-methylphenyl)nortropane ((11)C-PE2I) and (11)C-N,N-dimethyl-2-(2-amino-4-cyanophenylthio)benzylamine ((11)C-DASB), respectively. Plasma concentration of SEP-225289 was assessed before ligand injection, and subjects were monitored for adverse events. RESULTS: Average dopamine and serotonin transporter occupancies increased with increasing doses of SEP-225289. Mean dopamine and serotonin transporter occupancies were 33% ± 11% and 2% ± 13%, respectively, for 8 mg; 44% ± 4% and 9% ± 10%, respectively, for 12 mg; and 49% ± 7% and 14% ± 15%, respectively, for 16 mg. On the basis of the relationship between occupancy and plasma concentration, dopamine transporter IC(50) (the plasma concentration of drug at 50% occupancy) was determined (4.5 ng/mL) and maximum dopamine transporter occupancy was extrapolated (85%); however, low serotonin transporter occupancy prevented similar serotonin transporter calculations. No serious adverse events were reported. CONCLUSION: At the doses evaluated, occupancy of the dopamine transporter was significantly higher than that of the serotonin transporter, despite similar in vitro potencies, confirming that, in addition to in vitro assays, PET occupancy studies can be instrumental to the drug development process by informing early decisions about indication, dose, and therapeutic potential.


Subject(s)
Amines/metabolism , Cyclobutanes/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Neurotransmitter Uptake Inhibitors/metabolism , Positron-Emission Tomography , Serotonin Plasma Membrane Transport Proteins/metabolism , Adolescent , Adult , Amines/adverse effects , Amines/pharmacology , Cohort Studies , Cyclobutanes/adverse effects , Cyclobutanes/pharmacology , Female , Humans , Male , Middle Aged , Neurotransmitter Uptake Inhibitors/adverse effects , Neurotransmitter Uptake Inhibitors/pharmacology , Protein Transport/drug effects , Young Adult
10.
Br J Nutr ; 105(8): 1150-63, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21205415

ABSTRACT

A healthy, balanced diet is essential for both physical and mental well-being. Such a diet must include an adequate intake of micronutrients, essential fatty acids, amino acids and antioxidants. The monoamine neurotransmitters, serotonin, dopamine and noradrenaline, are derived from dietary amino acids and are involved in the modulation of mood, anxiety, cognition, sleep regulation and appetite. The capacity of nutritional interventions to elevate brain monoamine concentrations and, as a consequence, with the potential for mood enhancement, has not been extensively evaluated. The present study investigated an extract from oregano leaves, with a specified range of active constituents, identified via an unbiased, high-throughput screening programme. The oregano extract was demonstrated to inhibit the reuptake and degradation of the monoamine neurotransmitters in a dose-dependent manner, and microdialysis experiments in rats revealed an elevation of extracellular serotonin levels in the brain. Furthermore, following administration of oregano extract, behavioural responses were observed in mice that parallel the beneficial effects exhibited by monoamine-enhancing compounds when used in human subjects. In conclusion, these data show that an extract prepared from leaves of oregano, a major constituent of the Mediterranean diet, is brain-active, with moderate triple reuptake inhibitory activity, and exhibits positive behavioural effects in animal models. We postulate that such an extract may be effective in enhancing mental well-being in humans.


Subject(s)
Anti-Anxiety Agents/therapeutic use , Antidepressive Agents/therapeutic use , Biogenic Monoamines/physiology , Dietary Supplements , Neurotransmitter Uptake Inhibitors/therapeutic use , Origanum/chemistry , Plant Extracts/therapeutic use , Animals , Anti-Anxiety Agents/chemistry , Anti-Anxiety Agents/metabolism , Antidepressive Agents/chemistry , Antidepressive Agents/metabolism , Anxiety/prevention & control , Behavior, Animal , Benzoquinones/analysis , Benzoquinones/pharmacology , Brain/metabolism , Cymenes , Depression/prevention & control , Dietary Supplements/analysis , Drug Discovery/methods , HEK293 Cells , Humans , Male , Mice , Monoamine Oxidase Inhibitors/chemistry , Monoamine Oxidase Inhibitors/metabolism , Monoamine Oxidase Inhibitors/therapeutic use , Monoterpenes/analysis , Monoterpenes/blood , Monoterpenes/pharmacology , Neurotransmitter Uptake Inhibitors/chemistry , Neurotransmitter Uptake Inhibitors/metabolism , Neurotransmitter Uptake Inhibitors/pharmacology , Plant Extracts/chemistry , Plant Extracts/metabolism , Plant Leaves/chemistry , Random Allocation , Rats , Serotonin Plasma Membrane Transport Proteins/genetics , Serotonin Plasma Membrane Transport Proteins/metabolism
12.
Mol Cell Neurosci ; 40(4): 463-73, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19340933

ABSTRACT

Evidence have accumulated that reverse glutamate uptake plays a key role in the pathophysiology of cerebral ischemia. Here, we investigated the effects of glial glutamate transporter dysfunction on neuronal survival using the substrate inhibitor of glutamate transporters, L-trans-pyrrolidine,2-4,dicarboxylate (PDC), that partly mimics reverse glutamate uptake. On mice primary cortical co-cultures of neurons and astrocytes, PDC treatment triggered an elevation of extracellular glutamate concentration, induced neuronal calcium influx and a massive NMDA receptor (NMDAR) mediated-neuronal death without having any direct agonist activity on NMDARs. We investigated the NMDAR subpopulation activated by PDC-induced glutamate release. PDC application led to the activation of both subtypes of NMDARs but the presence of astrocytes was required to activate NMDARs located extra-synaptically. Extrasynaptic NMDAR activation was also confirmed by the loss of neuronal mitochondrial membrane potential and the inhibition of pro-survival p-ERK signalling pathway. These data suggest that reverse glial glutamate uptake may trigger neuronal death through preferential activation of extrasynaptic NMDAR-related pathways.


Subject(s)
Cell Death/physiology , Glutamic Acid/metabolism , Neuroglia/metabolism , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Amino Acid Transport System X-AG/metabolism , Animals , Cells, Cultured , Coculture Techniques , Dicarboxylic Acids/metabolism , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Glial Fibrillary Acidic Protein/metabolism , Humans , Mice , Microtubule-Associated Proteins/metabolism , Mitochondria/metabolism , Neuroglia/cytology , Neurons/cytology , Neurotransmitter Uptake Inhibitors/metabolism , Pyrrolidines/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
13.
Science ; 317(5843): 1390-3, 2007 Sep 07.
Article in English | MEDLINE | ID: mdl-17690258

ABSTRACT

Tricyclic antidepressants exert their pharmacological effect-inhibiting the reuptake of serotonin, norepinephrine, and dopamine-by directly blocking neurotransmitter transporters (SERT, NET, and DAT, respectively) in the presynaptic membrane. The drug-binding site and the mechanism of this inhibition are poorly understood. We determined the crystal structure at 2.9 angstroms of the bacterial leucine transporter (LeuT), a homolog of SERT, NET, and DAT, in complex with leucine and the antidepressant desipramine. Desipramine binds at the inner end of the extracellular cavity of the transporter and is held in place by a hairpin loop and by a salt bridge. This binding site is separated from the leucine-binding site by the extracellular gate of the transporter. By directly locking the gate, desipramine prevents conformational changes and blocks substrate transport. Mutagenesis experiments on human SERT and DAT indicate that both the desipramine-binding site and its inhibition mechanism are probably conserved in the human neurotransmitter transporters.


Subject(s)
Antidepressive Agents, Tricyclic/metabolism , Bacterial Proteins/metabolism , Desipramine/metabolism , Neurotransmitter Uptake Inhibitors/metabolism , Plasma Membrane Neurotransmitter Transport Proteins/metabolism , Amino Acid Sequence , Animals , Antidepressive Agents, Tricyclic/chemistry , Bacterial Proteins/chemistry , Binding Sites , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/metabolism , Cell Line , Conserved Sequence , Crystallography, X-Ray , Desipramine/chemistry , Dopamine/chemistry , Dopamine/metabolism , Dopamine Uptake Inhibitors/chemistry , Dopamine Uptake Inhibitors/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Humans , Leucine/chemistry , Leucine/metabolism , Models, Molecular , Molecular Sequence Data , Neurotransmitter Uptake Inhibitors/chemistry , Norepinephrine/chemistry , Norepinephrine/metabolism , Norepinephrine Plasma Membrane Transport Proteins/antagonists & inhibitors , Norepinephrine Plasma Membrane Transport Proteins/chemistry , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Plasma Membrane Neurotransmitter Transport Proteins/chemistry , Protein Binding , Protein Conformation , Sequence Homology, Amino Acid , Serotonin/chemistry , Serotonin/metabolism , Selective Serotonin Reuptake Inhibitors/chemistry , Selective Serotonin Reuptake Inhibitors/metabolism
14.
Phytochemistry ; 67(20): 2201-7, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16973193

ABSTRACT

Hyperforin is a polyprenylated acylphloroglucinol derivative from Hypericum perforatum (St. John's wort). It exhibits antidepressant activity by a novel mechanism of action, antibiotic activity against gram-positive bacteria, and antitumoral activity in vivo. However, it also produces drug-drug interactions by activation of the pregnan X receptor. No total synthesis has been described. Some natural and semisynthetic analogues are available to study structure-activity relationships. Enzymatically, the skeleton of hyperforin is formed by isobutyrophenone synthase from isobutyryl-CoA and three molecules of malonyl-CoA. The first prenylation step is catalyzed by a soluble and ion-dependent dimethylallyltransferase. Hyperforin mainly accumulates in pistils and fruits where it probably serves as defensive compound.


Subject(s)
Anti-Bacterial Agents/chemistry , Antineoplastic Agents, Phytogenic/chemistry , Hypericum/chemistry , Neurotransmitter Uptake Inhibitors/chemistry , Phloroglucinol/analogs & derivatives , Terpenes/chemistry , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Antineoplastic Agents, Phytogenic/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Bridged Bicyclo Compounds/chemistry , Bridged Bicyclo Compounds/metabolism , Bridged Bicyclo Compounds/pharmacology , Hypericum/anatomy & histology , Hypericum/metabolism , Neurotransmitter Uptake Inhibitors/metabolism , Neurotransmitter Uptake Inhibitors/pharmacology , Nuclear Magnetic Resonance, Biomolecular , Phloroglucinol/chemistry , Phloroglucinol/metabolism , Phloroglucinol/pharmacology , Terpenes/metabolism , Terpenes/pharmacology
15.
Neuropsychopharmacology ; 31(1): 12-35, 2006 Jan.
Article in English | MEDLINE | ID: mdl-15920497

ABSTRACT

Glucocorticoids administered to prevent respiratory distress in preterm infants are associated with neurodevelopmental disorders. To evaluate the long-term effects on forebrain development, we treated developing rats with dexamethasone (Dex) at 0.05, 0.2, or 0.8 mg/kg, doses below or spanning the range in clinical use, testing the effects of administration during three different stages: gestational days 17-19, postnatal days 1-3, or postnatal days 7-9. In adulthood, we assessed biomarkers of neural cell number and size, cholinergic presynaptic activity, neurotransmitter receptor expression, and synaptic signaling mediated through adenylyl cyclase (AC), in the cerebral cortex, hippocampus, and striatum. Even at doses that were devoid of lasting effects on somatic growth, Dex elicited deficits in the number and size of neural cells, with the largest effect in the cerebral cortex. Indices of cholinergic synaptic function (choline acetyltransferase, hemicholinium-3 binding) indicated substantial hyperactivity in males, especially in the hippocampus, effectively eliminating the normal sex differences for these parameters. However, the largest effects were seen for cerebrocortical cell signaling mediated by AC, where Dex treatment markedly elevated overall activity while obtunding the function of G-protein-coupled catecholaminergic or cholinergic receptors that stimulate or inhibit AC; uncoupling was noted despite receptor upregulation. Again, the effects on signaling were larger in males and offset the normal sex differences in AC. These results indicate that, during critical developmental periods, Dex administration evokes lasting alterations in neural cell numbers and synaptic function in forebrain regions, even at doses below those used in preterm infants.


Subject(s)
Dexamethasone/pharmacology , Neurons/drug effects , Signal Transduction/drug effects , Synapses/drug effects , Adenylyl Cyclases/metabolism , Animals , Animals, Newborn , Brain Chemistry/drug effects , Catecholamines/metabolism , Cell Count , Cell Size/drug effects , Choline O-Acetyltransferase/metabolism , Data Interpretation, Statistical , Dose-Response Relationship, Drug , Female , Hemicholinium 3/metabolism , Male , Neurotransmitter Uptake Inhibitors/metabolism , Pregnancy , Prosencephalon/cytology , Prosencephalon/drug effects , Prosencephalon/growth & development , Rats , Receptors, G-Protein-Coupled/drug effects , Receptors, Neurotransmitter/drug effects , Receptors, Neurotransmitter/metabolism , Sex Characteristics , Up-Regulation/drug effects
16.
Biol Trace Elem Res ; 108(1-3): 205-14, 2005.
Article in English | MEDLINE | ID: mdl-16327073

ABSTRACT

In these experiments we have tested the effect of bis(acetato)tetrakis (imidazole) copper(II) on the release and uptake of 14C-GABA and 3H-glutamate from brain slices and brain cortical synaptosomes. Cu(OAc)2(Im)4 in concentrations ranging from 1 to 100 microM has increased the release of GABA and glutamate from brain slices and synaptosomal preparations in a dose-related manner when the effect on GABA release is two-fold greater than glutamate and 10-fold greater than alanine. Pretreatment with a GABA uptake inhibitor such as 1-2 mM nipecotic acid has no effect on 14C-GABA release, whereas hydroxy aspartate, the glutamate uptake inhibitor, has elevated the stimulated release of glutamate. Copper(II) chloride, the inorganic form of copper, had no significant effect either on GABA release or on glutamate release. The stimulated release of exogenous GABA and glutamate was Ca2+-dependent, because it was inhibited by EGTA, and neuronal, because it was blocked by tetrodotoxin. The recent results can explain the anticonvulsant activity of Cu(OAc)2(Im)4 against strychnine-induced seizures by increasing the net release of GABA from cortical neurons.


Subject(s)
Brain/metabolism , Cerebral Cortex/metabolism , Glutamates/metabolism , Organometallic Compounds/pharmacology , Synaptosomes/microbiology , gamma-Aminobutyric Acid/metabolism , Animals , Anticonvulsants/metabolism , Anticonvulsants/pharmacology , Brain/drug effects , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Dose-Response Relationship, Drug , Egtazic Acid/antagonists & inhibitors , Egtazic Acid/metabolism , Female , Neurotransmitter Uptake Inhibitors/antagonists & inhibitors , Neurotransmitter Uptake Inhibitors/metabolism , Rats , Synaptosomes/drug effects , Synaptosomes/enzymology , Tetrodotoxin/antagonists & inhibitors , Tetrodotoxin/metabolism
17.
J Neurosci Res ; 81(2): 199-207, 2005 Jul 15.
Article in English | MEDLINE | ID: mdl-15931685

ABSTRACT

Extracellular glutamate is kept below a toxic level by glial and neuronal glutamate transporters. Here we show that the transportable glutamate uptake inhibitor L-trans-pyrrolidine-2,4-dicarboxylate (t-PDC) induced cell death in mature, but not in immature, hippocampal neuron-enriched cultures. The cell death produced by a 24-hr treatment with t-PDC was dose-dependent and reached 85% of the cell population at a 250 microM concentration at 23 days in vitro (DIV). Immunocytochemistry experiments showed that, under these experimental conditions, t-PDC killed not only neurons as expected but also glial cells. The N-methyl-D-aspartate (NMDA) antagonist D-2-aminophosphonovalerate (D-APV; 250 microM) only partially reversed this toxicity, completely protecting the neuronal cell population but not the glial population. The antioxidant compounds alpha-tocopherol or Trolox, used at concentrations that reverse the oxidative stress-induced toxicity, did not block the gliotoxicity specifically produced by t-PDC in the presence of D-APV. The nontransportable glutamate uptake inhibitor DL-threo-beta-benzyloxyaspartate (TBOA) elicited cell death only in mature, but not in immature, hippocampal cultures. The TBOA toxic effect was dose dependent and reached a plateau at 100 microM in 23-DIV cultures. About 50% of the cell population died. TBOA affected essentially the neuronal population. D-APV (250 microM) completely reversed this toxicity. It is concluded that nontransportable glutamate uptake inhibitors are neurotoxic via overactivation of NMDA receptors, whereas transportable glutamate uptake inhibitors induce both an NMDA-dependent neurotoxicity and an NMDA- and oxidative stress-independent gliotoxicity, but only in mature hippocampal cultures.


Subject(s)
Dicarboxylic Acids/toxicity , Glutamic Acid/toxicity , Hippocampus/pathology , Neuroglia/drug effects , Neurotoxins/pharmacology , Neurotransmitter Uptake Inhibitors/toxicity , Pyrrolidines/toxicity , Amino Acid Transport System X-AG/drug effects , Amino Acid Transport System X-AG/metabolism , Animals , Aspartic Acid/toxicity , Cell Death/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Interactions , Excitatory Amino Acid Agonists/pharmacology , Hippocampus/metabolism , Neuroglia/cytology , Neuroglia/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Neurotransmitter Uptake Inhibitors/metabolism , Rats , Receptors, N-Methyl-D-Aspartate/drug effects , Valine/analogs & derivatives , Valine/pharmacology
18.
Anat Rec A Discov Mol Cell Evol Biol ; 280(1): 874-83, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15382023

ABSTRACT

This chapter reviews recent evidence that the sympathetic nervous system (SNS) regulates liver repair by modulating the phenotypes of hepatic stellate cells (HSCs), the liver's principal fibrogenic cells, and hepatic epithelial progenitors, i.e., oval cells. SNS nerve fibers touch HSCs and these cells express adrenoceptors, suggesting that HSCs may be targets for SNS neurotransmitters. HSCs also contain catecholamine biosynthetic enzymes, release norepinephrine (NE), and are growth-inhibited by adrenoceptor antagonists. In addition, HSCs from mice with reduced levels of NE grow poorly in culture and exhibit inhibited activation during liver injury. Finally, growth and injury-related fibrogenic responses are rescued by adrenoceptor agonists. Thus, certain SNS inhibitors (SNSIs) protect experimental animals from cirrhosis. Conversely, SNSIs enhance the hepatic accumulation of oval cells (OCs) in injured livers. This response is associated with improved liver injury. Because SNSIs do not affect the expression of cytokines, growth factors, or growth factor receptors that are known to regulate OCs, and OCs express adrenoceptors, it is conceivable that catecholamines influence OCs by direct interaction with OC adrenoceptors. Given evidence that the SNS regulates the viability and activation of HSCs and OCs differentially, SNSIs may be novel therapies to improve the repair of damaged livers.


Subject(s)
Epithelial Cells/metabolism , Liver Cirrhosis/physiopathology , Liver/cytology , Liver/innervation , Sympathetic Nervous System/metabolism , Acetylcholine/metabolism , Animals , Humans , Leptin/metabolism , Liver Cirrhosis/prevention & control , Mice , Neuropeptide Y/metabolism , Neurotransmitter Uptake Inhibitors/metabolism , Norepinephrine/metabolism , Sympathetic Nervous System/anatomy & histology
19.
Synapse ; 53(3): 176-83, 2004 Sep 01.
Article in English | MEDLINE | ID: mdl-15236350

ABSTRACT

In the present study we describe a novel agent, SoRI-6238 (ethyl 5-amino-3-(3,4-dichlorophenyl)-1,2-dihydropyrido[3,4-b]pyrazin-7-ylcarbamate) that partially inhibits 5-HT transporter (SERT) binding and allosterically modulates SERT function. Membranes were prepared from rat brain. SoRI-6238 partially inhibited SERT binding to brain membranes with a plateau at about 40% of control. SoRI-6238 fully inhibited norepinephrine transporter (NET) and dopamine transporter (DAT) binding with IC(50) values of 12.1 microM and 5.8 microM, respectively. The apparent K(d) of [(125)I]RTI-55 binding to SERT increased, then reached a plateau with increasing concentrations of SoRI-6238. SoRI-6238 fully inhibited [(3)H]5-HT uptake, acting to decrease the V(max) (noncompetitive inhibition). In kinetic experiments, SoRI-6238 slowed the dissociation of [(125)I]RTI-55 from SERT and slowed the initial association rate. We conclude that SoRI-6238 partially inhibits SERT binding and function, most likely via an allosteric mechanism.


Subject(s)
Brain Chemistry/drug effects , Brain/drug effects , Carbamates/metabolism , Carrier Proteins/antagonists & inhibitors , Cell Membrane/drug effects , Membrane Glycoproteins/antagonists & inhibitors , Nerve Tissue Proteins/antagonists & inhibitors , Neurons/drug effects , Neurotransmitter Uptake Inhibitors/metabolism , Pyrazines/metabolism , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Animals , Binding Sites/drug effects , Binding Sites/physiology , Binding, Competitive/drug effects , Binding, Competitive/physiology , Brain/metabolism , Brain Chemistry/physiology , Carbamates/pharmacokinetics , Carrier Proteins/metabolism , Cell Membrane/metabolism , Dopamine Plasma Membrane Transport Proteins , Membrane Glycoproteins/drug effects , Membrane Glycoproteins/metabolism , Membrane Transport Proteins/drug effects , Membrane Transport Proteins/metabolism , Molecular Structure , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neurotransmitter Uptake Inhibitors/pharmacokinetics , Norepinephrine Plasma Membrane Transport Proteins , Pyrazines/pharmacokinetics , Radioligand Assay , Rats , Serotonin Plasma Membrane Transport Proteins , Subcellular Fractions , Symporters/drug effects , Symporters/metabolism
20.
J Neurophysiol ; 90(3): 1363-74, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12966170

ABSTRACT

Many electrophysiologists view neurotransmitter transporters as tiny vacuum cleaners, operating continuously to lower extracellular neurotransmitter concentration to zero. However, this is not consistent with their known behavior, instead only reducing extracellular neurotransmitter concentration to a finite, nonzero value at which an equilibrium is reached. In addition, transporters are equally able to go in either the forward or reverse direction, and when they reverse, they release their substrate in a calcium-independent manner. Transporter reversal has long been recognized to occur in response to pathological stimuli, but new data demonstrate that some transporters can also reverse in response to physiologically relevant stimuli. This is consistent with theoretical calculations that indicate that the reversal potentials of GABA and glycine transporters are close to the resting potential of neurons under normal conditions and that the extracellular concentration of GABA is sufficiently high when the GABA transporter is at equilibrium to tonically activate high-affinity extrasynaptic GABAA receptors. The equilibrium for the GABA transporter is not static but instead varies continuously as the driving force for the transporter changes. We propose that the GABA transporter plays a dynamic role in control of brain excitability by modulating the level of tonic inhibition in response to neuronal activity.


Subject(s)
Membrane Transport Proteins/metabolism , Neurotransmitter Agents/metabolism , Neurotransmitter Uptake Inhibitors/metabolism , Animals , Humans , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Transport Modulators , Membrane Transport Proteins/antagonists & inhibitors , Neurotransmitter Agents/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...