Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 258
Filter
1.
Acta Pharm ; 71(2): 279-291, 2021 Jun 01.
Article in English | MEDLINE | ID: mdl-33151165

ABSTRACT

The current study investigates the anorectic interaction and safety of the mazindol-metformin combination in rats. Isobologram and interaction index were used to determine anorectic interaction between mazindol and metformin in the sweetened milk model. The safety profile of the mazindol-metformin combination was determined by measuring anxiety, blood pressure, hematic biometry and blood chemistry. An acute dose of mazindol and metformin administered per os, individually or as a mixture, has reduced the milk consumption in rats in a dose-dependent manner. Theoretical effective dose 40 (ED40t) did not differ from the experimental effective dose 40 (ED40e) obtained with the mazindol-metformin mixture in the anorexia experiments, by Student's t-test. In addition, the interaction index confirmed the additive anorectic effect between both drugs. A single oral dose of ED40e mazindol-metformin mixture induced anxiolysis in the elevated plus-maze test. Moreover, oral administration of mazindol-metformin combination for 3 months significantly decreased glycemia, but not blood pressure nor other parameters of hematic biometry and blood chemistry. Results suggest that mazindol-metformin combination exerts an additive anorectic effect, as well as anxiolytic and hypoglycemic properties. Mazindol-metformin combination might be useful in obese patients with anxiety disorders or diabetes risk factors.


Subject(s)
Appetite Depressants/pharmacology , Mazindol/pharmacology , Metformin/pharmacology , Administration, Oral , Animals , Appetite Depressants/administration & dosage , Appetite Depressants/toxicity , Dose-Response Relationship, Drug , Drug Therapy, Combination , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/toxicity , Male , Maze Learning/drug effects , Mazindol/administration & dosage , Mazindol/toxicity , Metformin/administration & dosage , Metformin/toxicity , Rats , Rats, Wistar
2.
Psychopharmacology (Berl) ; 234(3): 323-328, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27766370

ABSTRACT

RATIONALE: The beneficial effects of psychostimulant drugs in the treatment of psychiatric disorders occur because they increase the extracellular dopamine concentration by inhibiting re-uptake of extracellular dopamine at dopamine transporters. However, the psychological effects at low dopamine transporter occupancy have not been well demonstrated. OBJECTIVES: The purpose of the study was to evaluate the psychological effects, dopamine transporter occupancy, and dopamine release induced by a single oral administration of a clinical dose of mazindol. METHODS: Ten healthy male volunteers were orally administered a placebo and a clinical dose of mazindol (1.5 mg) on separate days. The psychological effects of mazindol were assessed using a visual analogue scale to detect alterations in the state of consciousness. The amount of blockade of dopamine transporters was assessed using positron emission tomography with [18F]FE-PE2I and extracellular dopamine release was measured as the amount of change in [11C]raclopride binding. RESULTS: Following administration of a clinical dose of mazindol, the dopamine transporters were blocked by 24-25 %, and the binding potential of [11C]raclopride was reduced by 2.8-4.6 %. The differences of a score measuring derealisation and depersonalization associated with a positive basic mood were significantly correlated with the change in the [11C]raclopride binding in the limbic striatum. CONCLUSIONS: A subtle alteration in the state of consciousness was detected with a correlation to the changes in the [11C]raclopride binding, which implies that a subtle alteration in extracellular dopamine concentration in the limbic striatum by a small amount of dopamine transporter occupancy can affect the state of consciousness. TRIAL REGISTRATION HTTPS://UPLOAD.UMIN.AC.JP/CGI-OPEN-BIN/CTR_E/CTR_VIEW.CGI?RECPTNO=R000009703 : UMIN000008232.


Subject(s)
Brain/metabolism , Consciousness/drug effects , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopamine Uptake Inhibitors/pharmacology , Mazindol/pharmacology , Brain/diagnostic imaging , Carbon Radioisotopes , Corpus Striatum/metabolism , Depersonalization/chemically induced , Dopamine/metabolism , Dopamine Antagonists , Humans , Male , Positron-Emission Tomography , Raclopride , Young Adult
3.
Neurosci Lett ; 633: 141-145, 2016 10 28.
Article in English | MEDLINE | ID: mdl-27658895

ABSTRACT

In this study, we evaluated the preventive effect of mazindol on the development of obesity and sought to elucidate the drug's effects on the reward system. In mice, body weight gain and hyperphagia induced by high-fat diet (HFD) were decreased by 38.6% and 13.9%, respectively, by subcutaneous infusion of mazindol (1.5mg/kg/day) for 28days. A single intraperitoneal administration of mazindol (1.5mg/kg) significantly reduced lipid preference, as assessed using the two-bottle preference paradigm (vehicle, 89.98±1.66%; mazindol, 75.65±5.47%; p<0.05). In addition, the conditioned place preference (CPP) test demonstrated that mazindol (1.5mg/kg) significantly decreased CPP score for HFD as compared with vehicle (vehicle, 330.44±58.61s; mazindol, 144.72±43.02s; p<0.05). Moreover, at the dose required for these effects, mazindol did not elicit abuse potential or induce psychostimulant-like behavior. These results confirm that mazindol prevents diet-induced obesity without addictive behavior and demonstrate that its action is mediated at least in part via the reward system, advancing our understanding of mazindol in clinical practice.


Subject(s)
Anti-Obesity Agents/pharmacology , Mazindol/pharmacology , Reward , Animals , Conditioning, Psychological/drug effects , Dietary Fats/administration & dosage , Drug Tolerance , Food Preferences/drug effects , Infusions, Subcutaneous , Male , Mice, Inbred C57BL , Motor Activity/drug effects
4.
Acta Neuropsychiatr ; 26(3): 146-54, 2014 Jun.
Article in English | MEDLINE | ID: mdl-25142190

ABSTRACT

OBJECTIVES: Mazindol is a sympathomimetic amine, widely used as an anorectic agent in the treatment of obesity. This drug causes psychostimulant effects because of its pharmacological profile similar to amphetamine, acting like a monoamine reuptake inhibitor. However, the mechanisms underlying the action of mazindol are still not clearly understood. METHODS: Swiss mice received a single acute administration of mazindol (0.25, 1.25 and 2.5 mg/kg, ip) or saline. After 2 h, the animals were killed by decapitation; the brain was removed and used for the evaluation of activities of mitochondrial respiratory chain complexes, Krebs cycle enzymes and creatine kinase. RESULTS: Acute administration of mazindol decreased complex I activity only in the hippocampus. Complex IV activity was increased in the cerebellum (2.5 mg/kg) and cerebral cortex (0.25 mg/kg). Citrate synthase activity was increased in the cerebellum (1.25 mg/kg) and cerebral cortex (1.25 mg/kg), and creatine kinase activity was increased in the cerebellum (1.25 mg/kg). CONCLUSION: We suggest that the inhibition of complex I in the hippocampus only and activation of complex IV, citrate synthase and creatine kinase occurs because of a stimulus effect of mazindol in the central nervous system, which causes a direct impairment on energy metabolism.


Subject(s)
Brain/drug effects , Brain/metabolism , Central Nervous System Stimulants/pharmacology , Energy Metabolism/drug effects , Mazindol/pharmacology , Animals , Central Nervous System Stimulants/administration & dosage , Central Nervous System Stimulants/therapeutic use , Male , Mazindol/administration & dosage , Mazindol/therapeutic use , Mice , Obesity/drug therapy , Obesity/metabolism
5.
Behav Brain Res ; 270: 206-12, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24859175

ABSTRACT

Recent research has shown that pharmacological enhancement of dopaminergic function increases an optimism bias in humans. The present study investigated whether acute dopaminergic system stimulation through the administration of two dopamine-mimetic drugs, cocaine and mazindol, have similar effects in rats. To accomplish this goal, after initial behavioural training, two groups of rats received single injections of either cocaine or mazindol and were subsequently tested with the ambiguous-cue interpretation (ACI) paradigm. Both drugs were administered in three doses using the fully randomised Latin square designs. Cocaine (1, 2 and 5mg/kg) had no significant effect on the interpretation of the ambiguous cue. Mazindol at all three doses (0.5, 1 and 2mg/kg) significantly biased animals towards negative interpretation of the ambiguous cue. The results are discussed in relation to pharmacological and behaviourally evoked actions of tested compounds.


Subject(s)
Cocaine/pharmacology , Cognition/drug effects , Discrimination Learning/drug effects , Dopamine Uptake Inhibitors/pharmacology , Judgment/drug effects , Mazindol/pharmacology , Animals , Cocaine/administration & dosage , Cues , Dopamine Uptake Inhibitors/administration & dosage , Dose-Response Relationship, Drug , Male , Mazindol/administration & dosage , Neuropsychological Tests , Rats , Rats, Sprague-Dawley
6.
Psychopharmacology (Berl) ; 231(11): 2321-5, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24398821

ABSTRACT

RATIONALE: Mazindol, an appetite suppressant, inhibits the reuptake of dopamine in the synaptic cleft. It has been considered that mazindol might enhance dopamine transmission in the human brain. However, there has been no study that investigated the extracellular dopamine concentration in vivo. OBJECTIVE: Using positron emission tomography (PET), we aimed to measure the effect of mazindol on the extracellular dopamine concentration and to evaluate how mazindol affects the dopamine system in the healthy human brain. METHODS: Eleven healthy individuals (six males, five females, age 30.9 ± 4.9 years) were enrolled in this study. Each participant was scanned with [(11)C]raclopride on 1 day without any medicine as baseline condition, and on another day with mazindol as drug condition. In the drug condition, participants took mazindol 0.5 mg (N = 5) or 1.5 mg (N = 6) 2 h before the PET scan. Plasma concentrations of mazindol were measured before the injection of [(11)C]raclopride, and urine concentrations of mazindol were measured after the scan. RESULTS: After taking mazindol, the calculated decrease in binding potential (ΔBP) in the striatum was 1.74 % for 0.5 mg and 8.14 for 1.5 mg, and the correlation with the blood concentration of mazindol was significant (P = 0.0016, R (2) = 0.69). ΔBP was not significantly correlated with the urine concentration of mazindol (P = 0.84, R (2) = 0.005). CONCLUSIONS: Mazindol increased the extracellular concentration of dopamine in the human brain, and its effect was dose dependent. A single administration of mazindol, even at usual dosage, elevated dopamine concentration similarly to other addictive drugs, suggesting that the risk of dependence may increase with the mazindol dose.


Subject(s)
Brain/diagnostic imaging , Brain/metabolism , Dopamine Uptake Inhibitors/pharmacology , Dopamine/metabolism , Extracellular Space/metabolism , Mazindol/pharmacology , Adult , Appetite Depressants/analysis , Appetite Depressants/pharmacology , Blood Chemical Analysis , Carbon Radioisotopes , Corpus Striatum/diagnostic imaging , Corpus Striatum/metabolism , Dopamine Uptake Inhibitors/blood , Dopamine Uptake Inhibitors/urine , Dose-Response Relationship, Drug , Female , Humans , Male , Mazindol/blood , Positron-Emission Tomography , Raclopride , Young Adult
7.
Nature ; 503(7474): 141-5, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24121440

ABSTRACT

The biogenic amine transporters (BATs) regulate endogenous neurotransmitter concentrations and are targets for a broad range of therapeutic agents including selective serotonin reuptake inhibitors (SSRIs), serotonin-noradrenaline reuptake inhibitors (SNRIs) and tricyclic antidepressants (TCAs). Because eukaryotic BATs are recalcitrant to crystallographic analysis, our understanding of the mechanism of these inhibitors and antidepressants is limited. LeuT is a bacterial homologue of BATs and has proven to be a valuable paradigm for understanding relationships between their structure and function. However, because only approximately 25% of the amino acid sequence of LeuT is in common with that of BATs, and as LeuT is a promiscuous amino acid transporter, it does not recapitulate the pharmacological properties of BATs. Indeed, SSRIs and TCAs bind in the extracellular vestibule of LeuT and act as non-competitive inhibitors of transport. By contrast, multiple studies demonstrate that both TCAs and SSRIs are competitive inhibitors for eukaryotic BATs and bind to the primary binding pocket. Here we engineered LeuT to harbour human BAT-like pharmacology by mutating key residues around the primary binding pocket. The final LeuBAT mutant binds the SSRI sertraline with a binding constant of 18 nM and displays high-affinity binding to a range of SSRIs, SNRIs and a TCA. We determined 12 crystal structures of LeuBAT in complex with four classes of antidepressants. The chemically diverse inhibitors have a remarkably similar mode of binding in which they straddle transmembrane helix (TM) 3, wedge between TM3/TM8 and TM1/TM6, and lock the transporter in a sodium- and chloride-bound outward-facing open conformation. Together, these studies define common and simple principles for the action of SSRIs, SNRIs and TCAs on BATs.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacology , Antidepressive Agents, Tricyclic/pharmacology , Biogenic Amines/metabolism , Plasma Membrane Neurotransmitter Transport Proteins , Recombinant Fusion Proteins/chemistry , Selective Serotonin Reuptake Inhibitors/pharmacology , Serotonin Plasma Membrane Transport Proteins/chemistry , Serotonin Plasma Membrane Transport Proteins/metabolism , Antidepressive Agents, Second-Generation/metabolism , Antidepressive Agents, Tricyclic/metabolism , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding, Competitive/drug effects , Chlorides/metabolism , Crystallography, X-Ray , Humans , Mazindol/metabolism , Mazindol/pharmacology , Models, Molecular , Mutation , Norepinephrine/metabolism , Plasma Membrane Neurotransmitter Transport Proteins/antagonists & inhibitors , Plasma Membrane Neurotransmitter Transport Proteins/chemistry , Plasma Membrane Neurotransmitter Transport Proteins/genetics , Plasma Membrane Neurotransmitter Transport Proteins/metabolism , Protein Conformation/drug effects , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Reproducibility of Results , Serotonin Plasma Membrane Transport Proteins/genetics , Selective Serotonin Reuptake Inhibitors/metabolism , Sertraline/metabolism , Sertraline/pharmacology , Sodium/metabolism , Structure-Activity Relationship
8.
Eur J Med Chem ; 47(1): 24-37, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22071255

ABSTRACT

The two main groups of antidepressant drugs, the tricyclic antidepressants (TCAs) and the selective serotonin reuptake inhibitors (SSRIs), as well as several other compounds, act by inhibiting the serotonin transporter (SERT). However, the binding mode and molecular mechanism of inhibition in SERT are not fully understood. In this study, five classes of SERT inhibitors were docked into an outward-facing SERT homology model using a new 4D ensemble docking protocol. Unlike other docking protocols, where protein flexibility is not considered or is highly dependent on the ligand structure, flexibility was here obtained by side chain sampling of the amino acids of the binding pocket using biased probability Monte Carlo (BPMC) prior to docking. This resulted in the generation of multiple binding pocket conformations that the ligands were docked into. The docking results showed that the inhibitors were stacked between the aromatic amino acids of the extracellular gate (Y176, F335) presumably preventing its closure. The inhibitors interacted with amino acids in both the putative substrate binding site and more extracellular regions of the protein. A general structure-docking-based pharmacophore model was generated to explain binding of all studied classes of SERT inhibitors. Docking of a test set of actives and decoys furthermore showed that the outward-facing ensemble SERT homology model consistently and selectively scored the majority of active compounds above decoys, which indicates its usefulness in virtual screening.


Subject(s)
Models, Molecular , Selective Serotonin Reuptake Inhibitors/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Serotonin Plasma Membrane Transport Proteins/metabolism , Binding Sites , Drug Evaluation, Preclinical , Ligands , Mazindol/chemistry , Mazindol/metabolism , Mazindol/pharmacology , Monte Carlo Method , Protein Conformation , Sequence Homology, Amino Acid , Serotonin Plasma Membrane Transport Proteins/chemistry , Selective Serotonin Reuptake Inhibitors/chemistry , Tropanes/chemistry , Tropanes/metabolism , Tropanes/pharmacology
9.
Eur Neuropsychopharmacol ; 20(1): 37-48, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19729284

ABSTRACT

Cognitive impairments associated with schizophrenia await an effective treatment. In order to model schizophrenia-like cognitive deficits in rats, we evaluated the effects of ketamine, a dissociative anesthetic NMDA/glutamate receptor channel blocker in the attentional set-shifting task (ASST). Acute administration of ketamine (10 but not 3mg/kg) selectively impaired solving of the extradimensional (ED) set-shifting component. Next, we investigated whether the co-administration of mazindol, a dopamine and norepinephrine reuptake inhibitor would protect rats from ketamine-induced deficits. Mazindol dose-dependently and selectively alleviated ketamine-induced ED deficit with a minimal effective dose of 0.5mg/kg. The ED component improvement was noted primarily in ketamine - but not in vehicle co-treated rats, in which the drug facilitated ED shift solving at the dose as high as 5mg/kg. A "positive control", sertindole (2.5mg/kg) also ameliorated ketamine-induced ED deficit. Microdialysis of the prefrontal cortex in a separate group of animals revealed that 2-3h after the administration of 5mg/kg of mazindol and ketamine (i.e., at the time of ED component solving), the extracellular concentrations of dopamine were enhanced by ~300% as compared to the baseline and were intermediate between the mazindol- and ketamine-treated reference groups. However, at that time the levels of norepinephrine, serotonin and glutamate appeared unaffected. We conclude that ketamine may be useful in mimicking deficits specifically related to cognitive inflexibility observed in schizophrenia, and suggest that these anomalies could be ameliorated by mazindol. The beneficial effects of mazindol on ASST performance may have therapeutic implications for the treatment of schizophrenia.


Subject(s)
Attention/drug effects , Central Nervous System Stimulants/pharmacology , Cognition Disorders/chemically induced , Cognition Disorders/physiopathology , Ketamine , Mazindol/pharmacology , Analysis of Variance , Animals , Antipsychotic Agents/pharmacology , Behavior, Animal/drug effects , Brain Chemistry/drug effects , Brain Chemistry/physiology , Discrimination, Psychological/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Glutamic Acid/metabolism , Imidazoles/pharmacology , Indoles/pharmacology , Male , Microdialysis/methods , Neuropsychological Tests , Neurotransmitter Agents/metabolism , Rats , Rats, Sprague-Dawley
10.
Neuropharmacology ; 55(5): 737-42, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18634808

ABSTRACT

The mechanism by which the dopamine neurons of the substantia nigra pars compacta degenerate in Parkinson's disease, is partly unknown. Dopamine could be implicated in this phenomenon, and in order to explain its toxicity several hypotheses have been suggested. The similarity between apomorphine and dopamine as regards their chemical, pharmacological and toxicological properties provided a basis for investigating the nature of the toxicity of the former agent. In this study we describe some effects of apomorphine on mouse mesencephalic cell cultures at relatively low concentrations (from 0.5 to 2.5microM), apomorphine produced a neurotrophic effect, consisting of a 60% increase in dopaminergic neuron survival as measured by [(3)H] dopamine uptake. At high concentrations (over 20microM), however, apomorphine induced an increasing cytotoxic effect, as measured by the marked decrease in [(3)H] dopamine uptake, and by the direct observation of the dopaminergic neurons after TH immunostaining. This study may offer a new strategy for investigating the mechanisms underlying DA neuron vulnerability.


Subject(s)
Apomorphine/pharmacology , Dopamine Agonists/pharmacology , Dopamine/metabolism , Mesencephalon/cytology , Neurons/drug effects , Animals , Cell Survival/drug effects , Cells, Cultured , Choline/metabolism , Dopamine Uptake Inhibitors/pharmacology , Dose-Response Relationship, Drug , Embryo, Mammalian , Female , L-Lactate Dehydrogenase/metabolism , Mazindol/pharmacology , Mice , Monoamine Oxidase Inhibitors/pharmacology , Pregnancy , Selegiline/pharmacology , Tyrosine 3-Monooxygenase/metabolism
11.
J Psychopharmacol ; 22(5): 532-5, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18208906

ABSTRACT

The aim of the present study was to determine the impact of the appetite suppressant mazindol on meal pattern in rats. Meal patterns were monitored in adult male rats after mazindol dosing during the first three hours of the dark cycle using automated feeding chambers (BioDAQ). Mazindol (0, 0.25, 1.25 and 2.5 mg/kg, IP) produced a dose-dependent hypophagia and hypodipsia. Meal size and meal number were significantly suppressed by mazindol. The meal pattern findings indicate that mazindol inhibits eating in the rat via a suppression of both meal size and meal number.


Subject(s)
Dopamine Uptake Inhibitors/pharmacology , Eating/drug effects , Feeding Behavior/drug effects , Mazindol/pharmacology , Animals , Dopamine Uptake Inhibitors/administration & dosage , Dose-Response Relationship, Drug , Male , Mazindol/administration & dosage , Rats , Rats, Sprague-Dawley
12.
Brain Res ; 1131(1): 68-76, 2007 Feb 02.
Article in English | MEDLINE | ID: mdl-17169338

ABSTRACT

Cocaine, amphetamines and other psychostimulants inhibit synaptic dopamine uptake by interfering with dopamine transporter (DAT) function. The resultant potentiation of dopaminergic neurotransmission is associated with psychostimulant addiction. Fluctuations in dopamine uptake inhibition potency (DUIP) were observed for classical DAT blockers including cocaine, mazindol, methylphenidate (Ritalintrade mark) and benztropine in CHO cells expressing wild type DAT; cocaine potency also decreased in DAT-expressing non-neuronal COS-7 cells and neuronal N2A neuroblastoma cells. In contrast, the DAT substrate (+)-amphetamine did not display this DUIP fluctuation. In parallel experiments, no fluctuation was observed for the apparent binding affinities of these 5 drugs. The DUIP decrease appeared to correlate with an increase in cell surface DAT expression level, as measured by B(max) values and confocal microscopy. The fact that the DUIP profile of amphetamine diverged from that of the classical DAT blockers is consistent with the idea of fundamental differences between the mechanisms of abused psychostimulant DAT substrates and inhibitors. Identification of the cellular factors that underlie the DAT inhibitor DUIP fluctuation phenomenon may be relevant to anti-psychostimulant drug discovery efforts.


Subject(s)
Amphetamine/pharmacology , Brain Chemistry/drug effects , Cocaine/pharmacology , Dopamine Plasma Membrane Transport Proteins/drug effects , Dopamine Uptake Inhibitors/pharmacology , Dopamine/metabolism , Amphetamine-Related Disorders/metabolism , Amphetamine-Related Disorders/physiopathology , Animals , Benztropine/pharmacology , Binding, Competitive/drug effects , Binding, Competitive/physiology , Brain/drug effects , Brain/metabolism , Brain Chemistry/physiology , CHO Cells , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Cocaine-Related Disorders/metabolism , Cocaine-Related Disorders/physiopathology , Cricetinae , Cricetulus , Dopamine Plasma Membrane Transport Proteins/metabolism , Mazindol/pharmacology , Methylphenidate/pharmacology , Neurons/drug effects , Neurons/metabolism , Radioligand Assay
13.
J Biol Chem ; 281(4): 2012-23, 2006 Jan 27.
Article in English | MEDLINE | ID: mdl-16272152

ABSTRACT

In previous studies examining the structural determinants of antidepressant and substrate recognition by serotonin transporters (SERTs), we identified Tyr-95 in transmembrane segment 1 (TM1) of human SERT as a major determinant of binding for several antagonists, including racemic citalopram ((RS)-CIT). Here we described a separate site in hSERT TM3 (Ile-172) that impacts (RS)-CIT recognition when switched to the corresponding Drosophila SERT residue (I172M). The hSERT I172M mutant displays a marked loss of inhibitor potency for multiple inhibitors such as (RS)-CIT, clomipramine, RTI-55, fluoxetine, cocaine, nisoxetine, mazindol, and nomifensine, whereas recognition of substrates, including serotonin and 3,4-methylenedioxymethamphetamine, is unaffected. Selectivity for antagonist interactions is evident with this substitution because the potencies of the antidepressants tianeptine and paroxetine are unchanged. Reduced cocaine analog recognition was verified in photoaffinity labeling studies using [(125)I]MFZ 2-24. In contrast to the I172M substitution, other substitutions at this position significantly affected substrate recognition and/or transport activity. Additionally, the mouse mutation (mSERT I172M) exhibits similar selective changes in inhibitor potency. Unlike hSERT or mSERT, analogous substitutions in mouse dopamine transporter (V152M) or human norepinephrine transporter (V148M) result in transporters that bind substrate but are deficient in the subsequent translocation of the substrate. A double mutant hSERT Y95F/I172M had a synergistic impact on (RS)-CIT recognition ( approximately 10,000-fold decrease in (RS)-CIT potency) in the context of normal serotonin recognition. The less active enantiomer (R)-CIT responded to the I172M substitution like (S)-CIT but was relatively insensitive to the Y95F substitution and did not display a synergistic loss at Y95F/I172M. An hSERT mutant with single cysteine substitutions in TM1 and TM3 resulted in formation of a high affinity cadmium metal coordination site, suggesting proximity of these domains in the tertiary structure of SERT. These studies provided evidence for distinct binding sites coordinating SERT antagonists and revealed a close interaction between TM1 and TM3 differentially targeted by stereoisomers of CIT.


Subject(s)
Antidepressive Agents/pharmacology , Isoleucine/chemistry , Receptors, Serotonin/chemistry , Tyrosine/chemistry , Adrenergic Uptake Inhibitors/pharmacology , Amino Acid Sequence , Animals , Binding Sites , Binding, Competitive , Blotting, Western , Cadmium/chemistry , Cell Line , Cell Membrane/metabolism , Citalopram/pharmacology , Clomipramine/pharmacology , Cocaine/analogs & derivatives , Cocaine/pharmacology , Cysteine/chemistry , Dopamine Uptake Inhibitors/pharmacology , Fluoxetine/analogs & derivatives , Fluoxetine/pharmacology , HeLa Cells , Humans , Immunoprecipitation , Kinetics , LLC-PK1 Cells , Mazindol/pharmacology , Methionine/chemistry , Mice , Models, Chemical , Molecular Sequence Data , Mutation , N-Methyl-3,4-methylenedioxyamphetamine/chemistry , Nomifensine/pharmacology , Protein Binding , Protein Structure, Tertiary , Protein Transport , Radiopharmaceuticals/pharmacology , Serotonin/chemistry , Selective Serotonin Reuptake Inhibitors/pharmacology , Species Specificity , Stereoisomerism , Substrate Specificity
14.
Neurochem Res ; 30(5): 633-9, 2005 May.
Article in English | MEDLINE | ID: mdl-16176067

ABSTRACT

The molecular mechanism of 1-methyl-4-phenylpyridinium (MPP+), a Parkinsonism-inducing neurotoxin, has been studied in PC12 cells. The cells treated with MPP+ (100 microM) induced a rapid increase in phosphorylation of tyrosine residues of several proteins, including synaptophysin, a major 38 kDa synaptic vesicle protein implicated in exocytosis. An accelerated release of dopamine by MPP+ correlated with phosphorylation of synaptophysin. Exposing the cells to MPP+ triggered reactive oxygen species (ROS) generation within 60 min of treatment and the said effect was blocked by mazindol, a dopamine uptake blocker. In addition, pretreatment with 50-100 microM of selegiline, a selective MAO-B inhibitor, significantly suppressed MPP+-mediated ROS generation. These effects of MPP+ result in the generation of ROS, which may be involved in neuronal degeneration seen in Parkinson's disease.


Subject(s)
1-Methyl-4-phenylpyridinium/pharmacology , Dopamine/metabolism , Adrenal Medulla/drug effects , Adrenal Medulla/metabolism , Animals , Calcium/metabolism , Dopamine Uptake Inhibitors/pharmacology , Exocytosis , Mazindol/pharmacology , Monoamine Oxidase Inhibitors/pharmacology , PC12 Cells , Phosphorylation , Rats , Reactive Oxygen Species , Selegiline/pharmacology , Synaptophysin/metabolism , Tyrosine/metabolism
15.
J Pharmacol Exp Ther ; 314(1): 346-54, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15831439

ABSTRACT

Ecstasy samples often contain byproducts of the illegal, uncontrolled synthesis of N-methyl-3,4-methylenedioxy-amphetamine or 3,4-methylenedioxymethamphetamine (MDMA). MDMA and eight chemically defined byproducts of MDMA synthesis were investigated for their interaction with the primary sites of action of MDMA, namely the human plasmalemmal monamine transporters for norepinephrine, serotonin, and dopamine [(norepinephrine transporter (NET), serotonin transporter (SERT), and dopamine transporter (DAT)]. SK-N-MC neuroblastoma and human embryonic kidney cells stably transfected with the transporter cDNA were used for uptake and release experiments. Two of the eight compounds, 1,3-bis (3,4-methylenedioxyphenyl)-2-propanamine (12) and N-formyl-1,3-bis (3,4-methylenedioxyphenyl)-prop-2-yl-amine (13) had uptake inhibitory potencies with IC50 values in the low micromolar range similar to MDMA. Compounds with nitro instead of amino groups and a phenylethenyl instead of a phenylethyl structure or a formamide or acetamide modification had IC50 values beyond 100 microM. MDMA, 12, and 13 were examined for induction of carrier-mediated release by superfusion of transporter expressing cells preloaded with the metabolically inert transporter substrate [3H]1-methyl-4-phenylpyridinium. MDMA induced release mediated by NET, SERT, or DAT with EC50 values of 0.64, 1.12, and 3.24 microM, respectively. 12 weakly released from NET- and SERT-expressing cells with maximum effects less than one-tenth of that of MDMA and did not release from DAT cells. 13 had no releasing activity. 12 and 13 inhibited release induced by MDMA, and the concentration dependence of this effect correlated with their uptake inhibitory potency at the various transporters. These results do not support a neurotoxic potential of the examined ecstasy synthesis byproducts and provide interesting structure-activity relationships on the transporters.


Subject(s)
Biogenic Monoamines/metabolism , Carrier Proteins/drug effects , Hallucinogens/pharmacology , N-Methyl-3,4-methylenedioxyamphetamine/analogs & derivatives , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Cell Line , Clomipramine/pharmacology , Dopamine Plasma Membrane Transport Proteins , Dopamine Uptake Inhibitors/pharmacology , Hallucinogens/chemistry , Hallucinogens/pharmacokinetics , Humans , Indicators and Reagents , Kinetics , Magnetic Resonance Spectroscopy , Mazindol/pharmacology , Membrane Glycoproteins/metabolism , Membrane Transport Proteins/metabolism , N-Methyl-3,4-methylenedioxyamphetamine/pharmacokinetics , Nerve Tissue Proteins/metabolism , Norepinephrine Plasma Membrane Transport Proteins , Recombinant Proteins/drug effects , Serotonin Plasma Membrane Transport Proteins , Selective Serotonin Reuptake Inhibitors/pharmacology , Structure-Activity Relationship , Symporters/metabolism
16.
Naunyn Schmiedebergs Arch Pharmacol ; 370(1): 9-16, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15300361

ABSTRACT

The norepinephrine transporter (NET) is the carrier that drives the neuronal norepinephrine uptake mechanism (uptake1) in mammalian hearts. The radioligand [3H]mazindol binds with high affinity to NET. In this study, the kinetics of [3H]mazindol binding to NET were measured using a rat heart membrane preparation. Results from these studies were used to set up saturation binding assays designed to measure cardiac NET densities (Bmax) and competitive inhibition assays designed to measure inhibitor binding affinities (KI) for NET. Saturation binding assays measured NET densities in rat, rabbit, and canine hearts. Assay reproducibility was assessed and the effect of NaCl concentration on [3H]mazindol binding to NET was studied using membranes from rat and canine hearts. Specificity of [3H]mazindol binding to NET was determined in experiments in which the neurotoxin 6-hydroxydopamine (6-OHDA) was used to selectively destroy cardiac sympathetic nerve terminals in rats. Competitive inhibition studies measured KI values for several NET inhibitors and substrates. In kinetic studies using rat heart membranes, [3H]mazindol exhibited a dissociation rate constant koff=0.0123+/-0.0007 min(-1) and an association rate constant kon=0.0249+/-0.0019 nM(-1)min(-1). In saturation binding assays, [3H]mazindol binding was monophasic and saturable in all cases. Increasing the concentration of NaCl in the assay buffer increased binding affinity significantly, while only modestly increasing Bmax. Injections of 6-OHDA in rats decreased measured cardiac NET Bmax values in a dose-dependent manner, verifying that [3H]mazindol binds specifically to NET from sympathetic nerve terminals. Competitive inhibition studies provided NET inhibitor and substrate KI values consistent with previously reported values. These studies demonstrate the high selectivity of [3H]mazindol binding for the norepinephrine transporter in membrane preparations from mammalian hearts.


Subject(s)
Kinetics , Mazindol/metabolism , Mazindol/pharmacology , Symporters/drug effects , Symporters/metabolism , Animals , Binding, Competitive/drug effects , Dogs , Dose-Response Relationship, Drug , Heart Rate/drug effects , Hydroxydopamines/adverse effects , Hydroxydopamines/metabolism , Methods , Myocardium/cytology , Myocardium/metabolism , Norepinephrine Plasma Membrane Transport Proteins , Rabbits , Radioligand Assay/methods , Rats , Rats, Sprague-Dawley , Sodium Chloride/pharmacology , Species Specificity , Tritium/metabolism
17.
Neurosci Lett ; 355(1-2): 13-6, 2004 Jan 23.
Article in English | MEDLINE | ID: mdl-14729223

ABSTRACT

The present study examined the interaction between mazindol (MZ), an anorectic drug extensively used in Brazil and opioid/non-opioid endogenous analgesic systems activated by swim-stress. Further, the role of opioid, dopamine and N-methyl-D-aspartate (NMDA) receptors in mediating the analgesic effect was evaluated. The stress-induced analgesia of a 3-min swimming at 32 degrees C (opioid/non-opioid) and 20 degrees C (non-opioid) were assessed using the formalin test. Male Swiss mice were intraperitoneally injected with naloxone (1.0 mg/kg), sulpiride (3.0 mg/kg), MK-801 (0.075 mg/kg) or saline/vehicle 15 min prior, and with MZ (0.5 mg/kg) or saline/vehicle 5 min prior to swimming. The dose of MZ (0.5 mg/kg) did not cause analgesic effect, however, the association of MZ and swim-stress at both temperatures displayed synergistic interaction on analgesia that was blocked by sulpiride and MK-801 but not by naloxone. The present results suggest that MZ and swim-stress acted synergistically on analgesic responses, involving mainly the non-opioid component and possibly mediated by dopamine D2 receptors and NMDA receptors.


Subject(s)
Brain/metabolism , Dopamine/metabolism , Mazindol/pharmacology , Opioid Peptides/metabolism , Pain/metabolism , Stress, Physiological/metabolism , Animals , Brain/drug effects , Dopamine Antagonists/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Dose-Response Relationship, Drug , Drug Interactions/physiology , Excitatory Amino Acid Antagonists/pharmacology , Male , Mice , Narcotic Antagonists/pharmacology , Neural Pathways/drug effects , Neural Pathways/metabolism , Pain/drug therapy , Pain/physiopathology , Pain Measurement/drug effects , Receptors, Dopamine/drug effects , Receptors, Dopamine/metabolism , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Opioid/drug effects , Receptors, Opioid/metabolism , Stress, Physiological/physiopathology
18.
Cell Mol Neurobiol ; 23(4-5): 839-50, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14514035

ABSTRACT

1. The ability to target specific neurons can be used to produce selective neural lesions and potentially to deliver therapeutically useful moieties for treatment of disease. In the present study, we sought to determine if a monoclonal antibody to the dopamine transporter (anti-DAT) could be used to target midbrain dopaminergic neurons. 2. The monoclonal antibody recognizes the second, large extracellular loop of DAT. The antibody was conjugated to the "ribosome-inactivating protein"; saporin, and stereotactically pressure microinjected into either the center of the striatum or the left lateral ventricle of adult, male Sprague-Dawley rats. 3. Local intrastriatal injections produced destruction of dopaminergic neurons in the ipsilateral substantia nigra consistent with suicide transport of the immunotoxin. Intraventricular injections (i.c.v.) produced significant loss of dopaminergic neurons in the substantia nigra and ventral tegmental area bilaterally without evident damage to any other aminergic structures such as the locus coeruleus and raphe nuclei. To confirm the anatomic findings, binding of [3-H]mazindol to DAT in the striatum and midbrain was assessed using densitometric analysis of autoradiograms. Anti-DAT-saporin injected i.c.v. at a dose of 21 microg, but not 8 microg, produced highly significant decreases in mazindol binding consistent with loss of the dopaminergic neurons. 4. These results show that anti-DAT can be used to target midbrain dopaminergic neurons and that anti-DAT-saporin may be useful for producing a lesion very similar to the naturally occurring neural degeneration seen in Parkinson's disease. Anti-DAT-saporin joins the growing list of neural lesioning agents based on targeted cytotoxins.


Subject(s)
Disease Models, Animal , Dopamine/metabolism , Immunotoxins/pharmacology , Membrane Glycoproteins , Membrane Transport Modulators , Membrane Transport Proteins/antagonists & inhibitors , Nerve Degeneration/chemically induced , Nerve Tissue Proteins , Substantia Nigra/drug effects , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Binding, Competitive/drug effects , Binding, Competitive/physiology , Cell Death/drug effects , Cell Death/physiology , Denervation/methods , Dopamine Plasma Membrane Transport Proteins , Dose-Response Relationship, Drug , Immunotoxins/toxicity , Male , Mazindol/metabolism , Mazindol/pharmacology , Membrane Transport Proteins/immunology , N-Glycosyl Hydrolases/toxicity , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Parkinson Disease/physiopathology , Plant Proteins/toxicity , Rats , Rats, Sprague-Dawley , Ribosome Inactivating Proteins, Type 1 , Saporins , Substantia Nigra/pathology , Substantia Nigra/physiopathology
19.
Drug Alcohol Depend ; 70(3): 255-64, 2003 Jun 05.
Article in English | MEDLINE | ID: mdl-12757963

ABSTRACT

A drug discrimination procedure was used to examine the neuropharmacology of (-)-ephedrine (5 mg/kg), a sympathomimetic amine found in a variety of dietary supplements. (-)-Ephedrine has caused concern because of its use as a precursor in the manufacture of street drugs (e.g. methamphetamine) and its potential for abuse and toxicity. In the present study, the catecholamine reuptake inhibitors mazindol and nomifensine, the norepinephrine (NE) reuptake inhibitor desipramine, and the dopamine D(2)-like (e.g. D(2), D(3) and D(4)) agonist quinpirole substituted for (-)-ephedrine (>/=80% (-)-ephedrine-lever responding). The NE reuptake inhibitor nisoxetine, the D(1)-like (e.g. D(1) and D(5)) agonists (+/-)-SKF 38393 and SKF 82958, and the mixed D(1)-/D(2)-like agonist apomorphine occasioned intermediate levels of responding (50-79% (-)-ephedrine-lever responding). The (-)-ephedrine cue was antagonized by the D(1)-like antagonist SCH 23390 and the alpha(1)-adrenoceptor antagonist prazosin as well as the D(2)-like antagonists (-)-eticlopride and haloperidol, although only at doses that disrupted responding in some rats. The discriminative stimulus effects of a small dose of (-)-ephedrine (1.25 mg/kg) were enhanced by the alpha(2)-adrenoceptor antagonist idazoxan and to a lesser extent by the beta-adrenoceptor antagonist (-)-propranolol. However, the alpha(2)-adrenoceptor agonist clonidine (0.04 mg/kg) did not attenuate the (-)-ephedrine stimulus. These results suggest that D(1)-, D(2)-like, and alpha(1)-adrenergic receptors mediate the discriminative stimulus effects of (-)-ephedrine. Substitution of desipramine for (-)-ephedrine and not for some other stimulants suggests that NE transmission is a prominent feature of the (-)-ephedrine discriminative stimulus, and that NE underlies therapeutic and abuse-related effects of (-)-ephedrine that diverge from those of other stimulants.


Subject(s)
Catecholamines/physiology , Central Nervous System Stimulants/pharmacology , Ephedrine/pharmacology , Adrenergic Uptake Inhibitors/pharmacology , Animals , Benzazepines/pharmacology , Discrimination, Psychological/drug effects , Dopamine Antagonists/pharmacology , Ephedrine/antagonists & inhibitors , Ligands , Male , Mazindol/pharmacology , Membrane Transport Proteins/analysis , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic/drug effects
20.
Eur J Pharmacol ; 458(3): 263-73, 2003 Jan 05.
Article in English | MEDLINE | ID: mdl-12504782

ABSTRACT

The current studies compared mazindane (5-(4-chlorophenyl)-2,3-dihydro-5H-imidazo [2,1a] isoindole) hydrogen sulfate, a water soluble pro-drug of mazindol (5-(4-chlorophenyl-2,3-dihydro-5H-imidazo [2,1-a] isoindol-5-ol), with mazindol in assays used to define cocaine treatment agents. Both compounds enhanced motor activity (LMA) in Swiss Webster mice with ED(50) values of 2.5 mg/kg i.p. for mazindane and 3.9 mg/kg i.p. for mazindol. At 25 mg/kg mazindane displayed toxic effects and death while mazindol was effect/death free at 50 mg/kg. In Sprague-Dawley rats trained to discriminate cocaine from saline both compounds fully substituted for cocaine with mazindane being fourfold more potent in the total session (0.33 vs. 1.3 mg/kg i.p.) and first reinforcer (0.29 vs. 1.2 mg/kg i.p). Complete substitution was observed in rhesus monkeys trained to discriminate cocaine from saline with ED(50) values for mazindane (0.134 mg/kg i.m.) and mazindol (0.119 mg/kg i.m.). Mazindol exhibited little or no activity at 10(-5) M in inhibiting radioligand binding at 14 neurotransmitter sites while mazindane gave weak activity at the histamine H(1) and 5-hydroxytryptamine 5-HT(3) sites. These results demonstrate that mazindane could be a useful alternative to mazindol as a pharmacological tool because of its similar profile of activity and enhanced water solubility.


Subject(s)
Cocaine/analogs & derivatives , Cocaine/pharmacology , Discrimination, Psychological/drug effects , Dopamine Uptake Inhibitors/pharmacology , Mazindol/analogs & derivatives , Mazindol/pharmacology , Analysis of Variance , Animals , Binding, Competitive/drug effects , Cell Membrane/metabolism , Cocaine/metabolism , Discrimination Learning/drug effects , Dose-Response Relationship, Drug , Isoindoles , Macaca mulatta , Male , Mazindol/metabolism , Mice , Motor Activity/drug effects , Neurotransmitter Agents/metabolism , Proadifen/metabolism , Proadifen/pharmacology , Rats , Rats, Sprague-Dawley , Tritium
SELECTION OF CITATIONS
SEARCH DETAIL