Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 66
1.
Pharmacology ; 106(11-12): 597-605, 2021.
Article En | MEDLINE | ID: mdl-34515205

BACKGROUND: Amphetamine (AMPH) and other psychostimulants act on the norepinephrine (NE) transporter (NET) and the dopamine (DA) transporter (DAT) and enhance NE and DA signaling. Both NET and DAT share anatomical and functional characteristics and are regulated similarly by psychostimulants and receptor-linked signaling pathways. We and others have demonstrated that NET and DAT are downregulated by AMPH and substance P/neurokinin-1 receptor (NK1R)-mediated protein kinase C pathway. OBJECTIVES: Since both NET and DAT are downregulated by AMPH and NK1R activation and share high sequence homology, the objective of the study was to determine the catecholamine transporter specificity in NK1R modulation of AMPH-induced behaviors. METHODS: The effect of NK1R antagonism on AMPH-induced conditioned place preference (CPP) as well as AMPH-induced NET and DAT downregulation was examined using NET and DAT knockout mice (NET-KO and DAT-KO) along with their wild-type littermates. RESULTS: Aprepitant (5 mg/kg i.p.) significantly attenuated AMPH (2 mg/kg i.p.)-induced CPP in the wild-type and DAT-KO but not in the NET-KO. Locomotor activity measured during the post-conditioning test (in the absence of AMPH) showed higher locomotor activity in DAT-KO compared to wild-type or NET-KO. However, the locomotor activity of all 3 genotypes remained unchanged following aprepitant. Additionally, in the ventral striatum of wild-type, the AMPH-induced downregulation of NET function and surface expression but not that of DAT was attenuated by aprepitant. CONCLUSIONS: The results from the current study demonstrate that aprepitant attenuates the expression of AMPH-induced CPP in DAT-KO mice but not in NET-KO mice suggesting a role for NK1R-mediated NET regulation in AMPH-induced behaviors.


Amphetamine/pharmacology , Aprepitant/pharmacology , Central Nervous System Stimulants/pharmacology , Dopamine Plasma Membrane Transport Proteins/drug effects , Neurokinin-1 Receptor Antagonists/pharmacology , Norepinephrine Plasma Membrane Transport Proteins/drug effects , Animals , Behavior, Animal/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
2.
Pharmacol Res Perspect ; 9(5): e00855, 2021 10.
Article En | MEDLINE | ID: mdl-34423920

Several therapeutic options are currently available to treat excessive daytime sleepiness (EDS) in patients suffering from narcolepsy or obstructive sleep apnea. However, there are no comparisons between the various wake-promoting agents in terms of mechanism of action, efficacy, or safety. The goal of this study was to compare amphetamine, modafinil, solriamfetol, and pitolisant at their known primary pharmacological targets, histamine H3 receptors (H3R), dopamine, norepinephrine, and serotonin transporters, and in various in vivo preclinical models in relation to neurochemistry, locomotion, behavioral sensitization, and food intake. Results confirmed that the primary pharmacological effect of amphetamine, modafinil, and solriamfetol was to increase central dopamine neurotransmission, in part by inhibiting its transporter. Furthermore, solriamfetol increased levels of extracellular dopamine in the nucleus accumbens, and decreased the 3,4-dihydroxyphenyl acetic acid (DOPAC)/DA ratio in the striatum, as reported for modafinil and amphetamine. All these compounds produced hyperlocomotion, behavioral sensitization, and hypophagia, which are common features of psychostimulants and of compounds with abuse potential. In contrast, pitolisant, a selective and potent H3R antagonist/inverse agonist that promotes wakefulness, had no effect on striatal dopamine, locomotion, or food intake. In addition, pitolisant, devoid of behavioral sensitization by itself, attenuated the hyperlocomotion induced by either modafinil or solriamfetol. Therefore, pitolisant presents biochemical, neurochemical, and behavioral profiles different from those of amphetamine and other psychostimulants such as modafinil or solriamfetol. In conclusion, pitolisant is a differentiated therapeutic option, when compared with psychostimulants, for the treatment of EDS, as this agent does not show any amphetamine-like properties within in vivo preclinical models.


Amphetamine/pharmacology , Carbamates/pharmacology , Corpus Striatum/drug effects , Disorders of Excessive Somnolence/drug therapy , Feeding Behavior/drug effects , Locomotion/drug effects , Modafinil/pharmacology , Phenylalanine/analogs & derivatives , Piperidines/pharmacology , Wakefulness-Promoting Agents/pharmacology , 3,4-Dihydroxyphenylacetic Acid/metabolism , Adrenergic Uptake Inhibitors/pharmacology , Animals , Corpus Striatum/metabolism , Disorders of Excessive Somnolence/etiology , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins/drug effects , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopamine Uptake Inhibitors/pharmacology , Drug Evaluation, Preclinical , Drug Inverse Agonism , Histamine Antagonists/pharmacology , Mice , Narcolepsy/drug therapy , Neostriatum/drug effects , Neostriatum/metabolism , Norepinephrine Plasma Membrane Transport Proteins/drug effects , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Phenylalanine/pharmacology , Receptors, Histamine H3 , Sleep Apnea, Obstructive/complications
3.
J Integr Neurosci ; 20(2): 307-320, 2021 Jun 30.
Article En | MEDLINE | ID: mdl-34258929

We evaluated the practicability of using the rarely utilized C57BL/6N mouse as a Parkinson's disease model established via the acute MPTP/probenecid (MPTP/p) protocol. We confirmed dopaminergic degeneration in terms of decreased expression levels of tyrosine hydroxylase in the substantia nigra and striatum of MPTP/p-lesioned mice. In addition, acute MPTP/p-lesioned mice demonstrated initial motor dysfunctions followed by spontaneous recovery. Interestingly, these MPTP/p-lesioned mice exhibited anxiolytic and antidepressive behaviors upon recovery from these motor deficits. Additionally, increased expression of norepinephrine transporters in several brain regions, including the hippocampus, medial prefrontal cortex, and striatum, and an elevated rate of adult neurogenesis (in terms of increased numbers of doublecortin-positive neuroblasts) in the hippocampus were observed after recovery from motor dysfunctions. We suggest that the emotional alterations observed under these experimental conditions may be associated with enhanced adult neurogenesis, increased levels of norepinephrine transporters, and/or a possible interplay between these two factors. Consequently, this acute MPTP/p model adequately satisfies the criteria for the validity of a Parkinson's disease model regarding dopaminergic loss and motor impairment. However, the non-motor findings may offer novel evidence against the practicability of utilizing the acute MPTP/p-lesioned mice for modeling the emotional aberrations found in Parkinson's disease patients.


Behavior, Animal/drug effects , Dopamine Agents/pharmacology , Neurogenesis/drug effects , Norepinephrine Plasma Membrane Transport Proteins/drug effects , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/physiopathology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Disease Models, Animal , Male , Mice, Inbred C57BL
4.
Neuropharmacology ; 190: 108570, 2021 06 01.
Article En | MEDLINE | ID: mdl-33864800

While classical cathinones, such as methcathinone, have been shown to be monoamine releasing agents at human monoamine transporters, the subgroup of α-pyrrolidinophenones has thus far solely been characterized as monoamine transporter reuptake inhibitors. Herein, we report data from previously undescribed α-pyrrolidinopropiophenone (α-PPP) derivatives and compare them with the pharmacologically well-researched α-PVP (α-pyrrolidinovalerophenone). Radiotracer-based in vitro uptake inhibition assays in HEK293 cells show that the investigated α-PPP derivatives inhibit the human high-affinity transporters of dopamine (hDAT) and norepinephrine (hNET) in the low micromolar range, with α-PVP being ten times more potent. Similar to α-PVP, no relevant pharmacological activity was found at the human serotonin transporter (hSERT). Unexpectedly, radiotracer-based in vitro release assays reveal α-PPP, MDPPP and 3Br-PPP, but not α-PVP, to be partial releasing agents at hNET (EC50 values in the low micromolar range). Furthermore, uptake inhibition assays at low-affinity monoamine transporters, i.e., the human organic cation transporters (hOCT) 1-3 and human plasma membrane monoamine transporter (hPMAT), bring to light that all compounds inhibit hOCT1 and 2 (IC50 values in the low micromolar range) while less potently interacting with hPMAT and hOCT3. In conclusion, this study describes (i) three new hybrid compounds that efficaciously block hDAT while being partial releasers at hNET, and (ii) highlights the interactions of α-PPP-derivatives with low-affinity monoamine transporters, giving impetus to further studies investigating the interaction of drugs of abuse with OCT1-3 and PMAT.


Norepinephrine Plasma Membrane Transport Proteins/drug effects , Norepinephrine/metabolism , Propiophenones/pharmacology , Pyrroles/pharmacology , Pyrrolidines/pharmacology , Dopamine Plasma Membrane Transport Proteins/drug effects , Dopamine Plasma Membrane Transport Proteins/metabolism , Equilibrative Nucleoside Transport Proteins/drug effects , Equilibrative Nucleoside Transport Proteins/metabolism , HEK293 Cells , Humans , In Vitro Techniques , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Organic Cation Transport Proteins/drug effects , Organic Cation Transport Proteins/metabolism , Organic Cation Transporter 1/drug effects , Organic Cation Transporter 1/metabolism , Organic Cation Transporter 2/drug effects , Organic Cation Transporter 2/metabolism , Serotonin Plasma Membrane Transport Proteins/drug effects , Serotonin Plasma Membrane Transport Proteins/metabolism
5.
Behav Brain Res ; 383: 112508, 2020 04 06.
Article En | MEDLINE | ID: mdl-32017978

Two decades ago, the observation of a rapid and sustained antidepressant response after ketamine administration provided an exciting new avenue in the search for more effective therapeutics for the treatment of clinical depression. Research elucidating the mechanism(s) underlying ketamine's antidepressant properties has led to the development of several hypotheses, including that of disinhibition of excitatory glutamate neurons via blockade of N-methyl-d-aspartate (NMDA) receptors. Although the prominent understanding has been that ketamine's mode of action is mediated solely via the NMDA receptor, this view has been challenged by reports implicating other glutamate receptors such as AMPA, and other neurotransmitter systems such as serotonin and opioids in the antidepressant response. The recent approval of esketamine (Spravato™) for the treatment of depression has sparked a resurgence of interest for a deeper understanding of the mechanism(s) underlying ketamine's actions and safe therapeutic use. This review aims to present our current knowledge on both NMDA and non-NMDA mechanisms implicated in ketamine's response, and addresses the controversy surrounding the antidepressant role and potency of its stereoisomers and metabolites. There is much that remains to be known about our understanding of ketamine's antidepressant properties; and although the arrival of esketamine has been received with great enthusiasm, it is now more important than ever that its mechanisms of action be fully delineated, and both the short- and long-term neurobiological/functional consequences of its treatment be thoroughly characterized.


Antidepressive Agents/therapeutic use , Depressive Disorder, Major/drug therapy , Depressive Disorder, Treatment-Resistant/drug therapy , Ketamine/therapeutic use , Antidepressive Agents/pharmacology , Dopamine Plasma Membrane Transport Proteins/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Amino Acid Antagonists/therapeutic use , Humans , Ketamine/pharmacology , Norepinephrine Plasma Membrane Transport Proteins/drug effects , Receptor, Muscarinic M1/drug effects , Receptors, AMPA/drug effects , Receptors, Dopamine D2/drug effects , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, Opioid, delta/drug effects , Receptors, Opioid, kappa/drug effects , Receptors, Opioid, mu/drug effects , Receptors, Serotonin, 5-HT3/drug effects , Receptors, sigma/drug effects , Serotonin Plasma Membrane Transport Proteins/drug effects
6.
Int J Neuropsychopharmacol ; 22(4): 278-285, 2019 04 01.
Article En | MEDLINE | ID: mdl-30649319

BACKGROUND: The in vivo binding of clinical dose of venlafaxine on norepinephrine transporter has been questioned because venlafaxine has higher in vitro affinity to serotonin transporter than that to norepinephrine transporter. Although serotonin transporter occupancy of clinically relevant doses of venlafaxine has been reported, there has been no report of norepinephrine transporter occupancy in the human brain. METHODS: This was an open-label, single center, exploratory positron emission tomography study. Twelve major depressive disorder patients who had responded to venlafaxine extended-release and 9 control subjects were recruited. Each subject participated in one positron emission tomography measurement with [18F]FMeNER-D2. Binding potential in brain was quantified by the area under the curve ratio method with thalamus as target and white matter as reference regions. The difference of binding potential values between control and patient groups divided to 2 dose ranges were evaluated. Norepinephrine transporter occupancy (%) for all the major depressive disorder patients was calculated using mean binding potential of control subjects as baseline. The relationships between dose or plasma concentration of total active moiety and occupancies of norepinephrine transporter were also estimated. RESULTS: The binding potential of the patient group with 150 to 300 mg/d was significantly lower than that in the control subjects group (P = .0004 < .05/2). The norepinephrine transporter occupancy (8-61%) increased in a dose-dependent manner although a clear difference beyond 150 mg/d was not observed. CONCLUSIONS: This study demonstrates that clinically relevant doses of venlafaxine extended-release block the norepinephrine transporter of the major depressive disorder patient's brain. The data support the notion that the antidepressant effect of venlafaxine involves a combination of serotonin transporter and norepinephrine transporter blockades.


Brain/drug effects , Depressive Disorder, Major/drug therapy , Norepinephrine Plasma Membrane Transport Proteins/drug effects , Serotonin and Noradrenaline Reuptake Inhibitors/pharmacology , Venlafaxine Hydrochloride/pharmacology , Adult , Aged , Brain/diagnostic imaging , Delayed-Action Preparations , Depressive Disorder, Major/diagnostic imaging , Female , Humans , Male , Middle Aged , Morpholines , Positron-Emission Tomography , Serotonin and Noradrenaline Reuptake Inhibitors/administration & dosage , Venlafaxine Hydrochloride/administration & dosage , Young Adult
7.
ACS Chem Neurosci ; 10(1): 740-745, 2019 01 16.
Article En | MEDLINE | ID: mdl-30354055

Methcathinone analogs are appearing on the clandestine market at a rate nearly out-pacing the ability of investigators to examine them on an individual basis. To formulate structure-activity relationship (SAR) generalities, we examined the releasing ability of several simple methcathinone analogs at the three monoamine transporters (i.e., the dopamine, norepinephrine, and serotonin transporters, DAT, NET, and SERT, respectively) using in vitro assay methods. The analogs included methcathinone and 14 other compounds monosubstituted at the 2-, 3-, or 4-position. In general, (a) the 2-substituted analogs were less potent than either the 3- or 4-substituted analogs, (b) the 3- and 4-substituted analogs were relatively similar in potency, (c) methcathinone was the most selective as a DAT-releasing agent, and (d) the 3- and 4-CF3 analogs were the least DAT-selective. For the 15 compounds, there was a significant correlation ( r > 0.9) between DAT and NET potency, suggesting relatively similar structure-activity relationships (at least for the compounds examined here). Several of the compounds have appeared on the clandestine market since our studies were initiated, and the present results provide new information on how they might act.


Dopamine Plasma Membrane Transport Proteins/drug effects , Propiophenones/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology , Serotonin Plasma Membrane Transport Proteins/drug effects , Animals , Dopamine Plasma Membrane Transport Proteins/metabolism , Male , Norepinephrine/metabolism , Norepinephrine Plasma Membrane Transport Proteins/drug effects , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Propiophenones/chemistry , Rats, Sprague-Dawley , Serotonin Plasma Membrane Transport Proteins/metabolism , Structure-Activity Relationship , Synaptosomes/drug effects , Synaptosomes/metabolism
8.
Am J Respir Crit Care Med ; 199(11): 1421-1431, 2019 06 01.
Article En | MEDLINE | ID: mdl-30521757

Rationale: Primary treatment of obstructive sleep apnea can be accompanied by a persistence of excessive sleepiness despite adherence. Furthermore, effectiveness of sleep apnea treatment is limited by poor adherence. Currently available pharmacologic options for the treatment of sleepiness in this population are limited. Objectives: To evaluate the efficacy and safety of solriamfetol (JZP-110), a selective dopamine and norepinephrine reuptake inhibitor with robust wake-promoting effects, for the treatment of excessive sleepiness in participants with obstructive sleep apnea with current or prior sleep apnea treatment. Methods: This was a double-blind, randomized, placebo-controlled, parallel-group, 12-week trial comparing solriamfetol, 37.5, 75, 150, and 300 mg, with placebo. Measurements and Main Results: Of 476 randomized participants, 459 were included in the prespecified efficacy analyses. Coprimary endpoints (Maintenance of Wakefulness Test sleep latency and Epworth Sleepiness Scale score) were met at all solriamfetol doses (P < 0.05), with dose-dependent effects observed at Week 1 maintained over the study duration. All doses except 37.5 mg resulted in higher percentages of participants reporting improvement on Patient Global Impression of Change (key secondary endpoint; P < 0.05). Adverse events were reported in 47.9% of placebo- and 67.9% of solriamfetol-treated participants; five participants experienced serious adverse events (two [1.7%] placebo, three [0.8%] solriamfetol); none were deemed related to study drug. The most common adverse events with solriamfetol were headache (10.1%), nausea (7.9%), decreased appetite (7.6%), anxiety (7.0%), and nasopharyngitis (5.1%). Conclusions: Solriamfetol significantly increased wakefulness and reduced sleepiness in participants with obstructive sleep apnea and excessive sleepiness; most adverse events were mild or moderate in severity. Clinical trial registered with www.clinicaltrials.gov (NCT02348606) and www.eudract.ema.europa.eu (EudraCT 2014-005514-31).


Carbamates/therapeutic use , Disorders of Excessive Somnolence/drug therapy , Disorders of Excessive Somnolence/etiology , Dopamine Uptake Inhibitors/therapeutic use , Norepinephrine Plasma Membrane Transport Proteins/drug effects , Phenylalanine/analogs & derivatives , Sleep Apnea, Obstructive/complications , Adult , Aged , Double-Blind Method , Female , Humans , Male , Middle Aged , Phenylalanine/therapeutic use
9.
Molecules ; 23(10)2018 Oct 04.
Article En | MEDLINE | ID: mdl-30287800

In traditional Asian medicinal systems, preparations of the root and stem bark of Magnolia species are widely used to treat anxiety and other nervous disturbances. The biphenyl-type neolignans honokiol and magnolol are the main constituents of Magnolia bark extracts. In the central nervous system, Magnolia bark preparations that contain honokiol are thought to primarily interact with γ-aminobutyric acid A (GABAA) receptors. However, stress responses inherently involve the noradrenergic system, which has not been investigated in the pharmacological mechanism of honokiol. We present here interactions of honokiol and other synthesized biphenyl-type neolignans and diphenylmethane analogs with the norepinephrine transporter (NET), which is responsible for the synaptic clearance of norepinephrine and the target of many anxiolytics. Of the synthesized compounds, 16 are new chemical entities, which are fully characterized. The 52 compounds tested show mild, non-potent interactions with NET (IC50 > 100 µM). It is thus likely that the observed anxiolytic effects of, e.g., Magnolia preparations, are not due to direct interaction with the noradrenergic system.


Adrenergic Neurons/drug effects , Anti-Anxiety Agents/pharmacology , Central Nervous System/drug effects , Norepinephrine Plasma Membrane Transport Proteins/drug effects , Plant Extracts/pharmacology , Anti-Anxiety Agents/chemistry , Benzhydryl Compounds/chemistry , Benzhydryl Compounds/pharmacology , Biphenyl Compounds/chemistry , Biphenyl Compounds/pharmacology , Central Nervous System/metabolism , HEK293 Cells , Humans , Lignans/chemistry , Lignans/pharmacology , Magnolia/chemistry , Norepinephrine/metabolism , Plant Bark/chemistry , Plant Extracts/chemistry , Receptors, GABA-A/drug effects
10.
ACS Chem Neurosci ; 9(5): 1128-1140, 2018 05 16.
Article En | MEDLINE | ID: mdl-29300091

Dual inhibition of serotonin and norepinephrine transporters (hSERT and hNET) gives greatly improved efficacy and tolerability for treating major depressive disorder (MDD) compared with selective reuptake inhibitors. Pioneer studies provided valuable information on structure, function, and pharmacology of drugs targeting both hSERT and hNET (serotonin-norepinephrine reuptake inhibitors, SNRIs), and the differential binding mechanism between SNRIs and selective inhibitors of 5-HT (SSRIs) or NE (sNRIs) to their corresponding targets was expected to be able to facilitate the discovery of a privileged drug-like scaffold with improved efficacy. However, the dual-target mechanism of SNRIs was still elusive, and the binding mode distinguishing SNRIs from SSRIs and sNRIs was also unclear. Herein, an integrated computational strategy was adopted to discover the binding mode shared by all FDA approved SNRIs. The comparative analysis of binding free energy at the per-residue level discovered that residues Phe335, Leu337, Gly338, and Val343 located at the transmembrane domain 6 (TM6) of hSERT (the corresponding residues Phe317, Leu319, Gly320, and Val325 in hNET) were the determinants accounting for SNRIs' dual-acting inhibition, while residues lining TM3 and 8 (Ile172, Ser438, Thr439, and Leu443 in hSERT; Val148, Ser419, Ser420, and Met424 in hNET) contributed less to the binding of SNRIs than that of SSRIs and sNRIs. Based on these results, the distances between an SNRI's centroid and the centroids of its two aromatic rings (measuring the depth of rings stretching into hydrophobic pockets) were discovered as the key to the SNRIs' dual-targeting mechanism. This finding revealed SNRIs' binding mechanism at an atomistic level, which could be further utilized as structural blueprints for the rational design of privileged drug-like scaffolds treating MDD.


Depressive Disorder, Major/drug therapy , Norepinephrine Plasma Membrane Transport Proteins/drug effects , Norepinephrine/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology , Binding Sites/drug effects , Humans , Molecular Dynamics Simulation , Norepinephrine Plasma Membrane Transport Proteins/metabolism
11.
Int J Neuropsychopharmacol ; 21(2): 108-113, 2018 02 01.
Article En | MEDLINE | ID: mdl-29016993

Background: Quetiapine is effective in treating depressive symptoms in major depressive disorder and bipolar disorder, but the mechanisms underlying its antidepressants effects are unknown. Norquetiapine, a metabolite of quetiapine, has high affinity for norepinephrine transporter, which might account for its therapeutic efficacy. Methods: In this study, we used positron emission tomography with (S,S)-[11C]O-methyl reboxetine to estimate norepinephrine transporter density and assess the relationship between norepinephrine transporter occupancy by quetiapine XR and improvement in depression in patients with major depressive disorder (n=5) and bipolar disorder (n=5). After the baseline positron emission tomography scan, patients were treated with quetiapine XR with a target dose of 150 mg in major depressive disorder and 300 mg in bipolar disorder. Patients had a second positron emission tomography scan at the end of week 2 and a final scan at week 7. Results: Norepinephrine transporter density was significantly lower in locus ceruleus in patients compared with healthy subjects. Further, there was a significant positive correlation between quetiapine XR dose and norepinephrine transporter occupancy in locus ceruleus at week 2. The norepinephrine transporter occupancy at week 2 in hypothalamus but not in other regions predicted improvement in depression as reflected by reduction in MADRS scores from baseline to week 7. The estimated dose of quetiapine XR associated with 50% norepinephrine transporter occupancy in hypothalamus at week 2 was 256 mg and the estimated plasma levels of norquetiapine to achieve 50% norepinephrine transporter occupancy was 36.8 µg/L. Conclusion: These data provide preliminary support for the hypothesis that norepinephrine transporter occupancy by norquetiapine may be a contributor to the antidepressant effects of quetiapine.


Adrenergic Uptake Inhibitors , Antidepressive Agents/pharmacokinetics , Bipolar Disorder/drug therapy , Depressive Disorder, Major/drug therapy , Dibenzothiazepines/blood , Hypothalamus/drug effects , Locus Coeruleus/drug effects , Norepinephrine Plasma Membrane Transport Proteins/drug effects , Positron-Emission Tomography/methods , Quetiapine Fumarate/pharmacokinetics , Reboxetine , Adult , Antidepressive Agents/administration & dosage , Bipolar Disorder/diagnostic imaging , Delayed-Action Preparations , Depressive Disorder, Major/diagnostic imaging , Female , Humans , Hypothalamus/diagnostic imaging , Locus Coeruleus/diagnostic imaging , Male , Middle Aged , Quetiapine Fumarate/administration & dosage , Young Adult
12.
ACS Chem Neurosci ; 8(6): 1397-1403, 2017 06 21.
Article En | MEDLINE | ID: mdl-28220701

Bupropion (1), an α-aminophenone uptake inhibitor at plasma membrane transporters for dopamine (DAT) and norepinephrine (NET), is a widely prescribed antidepressant and smoking cessation aid. Cathinone (2), a structurally simpler α-aminophenone, is a substrate-type releasing agent at the same transporters and a recognized drug of abuse. Our goal was to identify the structural features of α-aminophenones that govern the mechanistic transition from uptake inhibition to substrate-induced release. Deconstructed analogues of 1 were synthesized and compared for their ability to interact with DAT, NET, and the serotonin transporter (SERT) using in vitro assay methods. Bulky amine substituents resulted in compounds that function as DAT uptake inhibitors but not release agents, whereas smaller amine substituents result in relatively nonselective releasing agents at DAT and NET. Our findings add to empirical evidence supporting distinct molecular determinants for α-aminophenone- (i.e., cathinone-) related agents acting as transporter inhibitors versus those acting as releasers.


Bupropion/pharmacology , Dopamine Plasma Membrane Transport Proteins/drug effects , Dopamine Uptake Inhibitors/pharmacology , Norepinephrine Plasma Membrane Transport Proteins/drug effects , Alkaloids/chemistry , Alkaloids/pharmacology , Animals , Brain/drug effects , Bupropion/chemistry , Central Nervous System Stimulants/chemistry , Central Nervous System Stimulants/pharmacology , Dopamine Uptake Inhibitors/chemistry , Male , Neurotransmitter Agents/metabolism , Rats , Rats, Sprague-Dawley
13.
Curr Top Behav Neurosci ; 32: 49-72, 2017.
Article En | MEDLINE | ID: mdl-27272068

Pharmacological assays carried out in transfected cells have been very useful for describing the mechanism of action of cathinone new psychoactive substances (NPS). These in vitro characterizations provide fast and reliable information on psychoactive substances soon after they emerge for recreational use. Well-investigated comparator compounds, such as methamphetamine, 3,4-methylenedioxymethamphetamine, cocaine, and lysergic acid diethylamide, should always be included in the characterization to enhance the translation of the in vitro data into clinically useful information. We classified cathinone NPS according to their pharmacology at monoamine transporters and receptors. Cathinone NPS are monoamine uptake inhibitors and most induce transporter-mediated monoamine efflux with weak to no activity at pre- or postsynaptic receptors. Cathinones with a nitrogen-containing pyrrolidine ring emerged as NPS that are extremely potent transporter inhibitors but not monoamine releasers. Cathinones exhibit clinically relevant differences in relative potencies at serotonin vs. dopamine transporters. Additionally, cathinone NPS have more dopaminergic vs. serotonergic properties compared with their non-ß-keto amphetamine analogs, suggesting more stimulant and reinforcing properties. In conclusion, in vitro pharmacological assays in heterologous expression systems help to predict the psychoactive and toxicological effects of NPS.


Alkaloids/pharmacology , Central Nervous System Stimulants/pharmacology , Dopamine Plasma Membrane Transport Proteins/drug effects , Norepinephrine Plasma Membrane Transport Proteins/drug effects , Serotonin Plasma Membrane Transport Proteins/drug effects , Adrenergic Uptake Inhibitors/pharmacology , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins/antagonists & inhibitors , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopamine Uptake Inhibitors/pharmacology , Humans , In Vitro Techniques , Norepinephrine/metabolism , Norepinephrine Plasma Membrane Transport Proteins/antagonists & inhibitors , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology
14.
Curr Top Behav Neurosci ; 32: 119-131, 2017.
Article En | MEDLINE | ID: mdl-27696217

Many cathinone analogs act as substrates or inhibitors at dopamine, norepinephrine, and serotonin transporters (DAT, NET, SERT, respectively). Drug selectivity at DAT vs. SERT is a key determinant of abuse potential for monoamine transporter substrates and inhibitors, such that potency at DAT > SERT is associated with high abuse potential, whereas potency at DAT < SERT is associated with low abuse potential. Quantitative structure-activity relationship (QSAR) studies with a series of 4-substituted methcathinone analogs identified volume of the 4-position substituent on the methcathinone phenyl ring as one structural determinant of both DAT vs. SERT selectivity and abuse-related behavioral effects in an intracranial self-stimulation procedure in rats. Subsequent modeling studies implicated specific amino acids in DAT and SERT that might interact with 4-substituent volume to determine effects produced by this series of cathinone analogs. These studies illustrate use of QSAR analysis to investigate pharmacology of cathinones and function of monoamine transporters.


Behavior, Animal/drug effects , Dopamine Plasma Membrane Transport Proteins/drug effects , Propiophenones/pharmacology , Psychotropic Drugs/pharmacology , Serotonin Plasma Membrane Transport Proteins/drug effects , Substance-Related Disorders , Amphetamine/pharmacology , Animals , Fenfluramine/pharmacology , Humans , Methamphetamine/analogs & derivatives , Methamphetamine/chemistry , Methamphetamine/pharmacology , Methylamines/chemistry , Methylamines/pharmacology , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Norepinephrine Plasma Membrane Transport Proteins/drug effects , Propiophenones/chemistry , Psychotropic Drugs/chemistry , Quantitative Structure-Activity Relationship , Rats , Self Administration , Structure-Activity Relationship
15.
Curr Top Behav Neurosci ; 32: 73-92, 2017.
Article En | MEDLINE | ID: mdl-27677783

Products containing psychoactive synthetic cathinones, such as mephedrone and 3,4-methylenedioxypyrovalerone (MDPV) are prevalent in our society. Synthetic cathinones are structurally similar to methamphetamine, and numerous synthetics have biological activity at dopamine, serotonin, and norepinephrine transporters. Importantly, monoamine transporters co-transport sodium ions along with their substrate, and movement of substrates and ions through the transporter can generate measurable ionic currents. Here we review how electrophysiological information has enabled us to determine how synthetic cathinones affect transporter-mediated currents in cells that express these transporters. Specifically, drugs that act as transporter substrates induce inward depolarizing currents when cells are held near their resting membrane potential, whereas drugs that act as transporter blockers induce apparent outward currents by blocking an inherent inward leak current. We have employed the two-electrode voltage-clamp technique in Xenopus laevis oocytes overexpressing monoamine transporters to determine whether synthetic cathinones found in the so-called bath salts products behave as blockers or substrates. We also examined the structure-activity relationships for synthetic cathinone analogs related to the widely abused compound MDPV, a common constituent in "bath salts" possessing potent actions at the dopamine transporter.


Alkaloids/pharmacology , Benzodioxoles/pharmacology , Dopamine Plasma Membrane Transport Proteins/drug effects , Norepinephrine Plasma Membrane Transport Proteins/drug effects , Oocytes/drug effects , Psychotropic Drugs/pharmacology , Pyrrolidines/pharmacology , Serotonin Plasma Membrane Transport Proteins/drug effects , Alkaloids/chemistry , Animals , Benzodioxoles/chemistry , Dopamine Plasma Membrane Transport Proteins/metabolism , Humans , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Patch-Clamp Techniques , Psychotropic Drugs/chemistry , Pyrrolidines/chemistry , Serotonin Plasma Membrane Transport Proteins/metabolism , Structure-Activity Relationship , Xenopus laevis , Synthetic Cathinone
16.
Curr Top Behav Neurosci ; 32: 93-117, 2017.
Article En | MEDLINE | ID: mdl-27830575

3,4-Methylenedioxypyrovalerone (MDPV) is a psychoactive component of so-called bath salts products that has caused serious medical consequences in humans. In this chapter, we review the neuropharmacology of MDPV and related analogs, and supplement the discussion with new results from our preclinical experiments. MDPV acts as a potent uptake inhibitor at plasma membrane transporters for dopamine (DAT) and norepinephrine (NET) in nervous tissue. The MDPV formulation in bath salts is a racemic mixture, and the S isomer is much more potent than the R isomer at blocking DAT and producing abuse-related effects. Elevations in brain extracellular dopamine produced by MDPV are likely to underlie its locomotor stimulant and addictive properties. MDPV displays rapid pharmacokinetics when injected into rats (0.5-2.0 mg/kg), with peak plasma concentrations achieved by 10-20 min and declining quickly thereafter. MDPV is metabolized to 3,4-dihydroxypyrovalerone (3,4-catechol-PV) and 4-hydroxy-3-methoxypyrovalerone (4-OH-3-MeO-PV) in vivo, but motor activation produced by the drug is positively correlated with plasma concentrations of parent drug and not its metabolites. 3,4-Catechol-PV is a potent uptake blocker at DAT in vitro but has little activity after administration in vivo. 4-OH-3-MeO-PV is the main MDPV metabolite but is weak at DAT and NET. MDPV analogs, such as α-pyrrolidinovalerophenone (α-PVP), display similar ability to inhibit DAT and increase extracellular dopamine concentrations. Taken together, these findings demonstrate that MDPV and its analogs represent a unique class of transporter inhibitors with a high propensity for abuse and addiction.


Adrenergic Uptake Inhibitors/pharmacology , Benzodioxoles/pharmacology , Dopamine Plasma Membrane Transport Proteins/drug effects , Dopamine Uptake Inhibitors/pharmacology , Norepinephrine Plasma Membrane Transport Proteins/drug effects , Psychotropic Drugs/pharmacology , Pyrrolidines/pharmacology , Animals , Behavior, Animal/drug effects , Brain/drug effects , Humans , Neuropharmacology , Substance-Related Disorders , Synthetic Cathinone
17.
Curr Top Behav Neurosci ; 32: 333-350, 2017.
Article En | MEDLINE | ID: mdl-28025810

Psychoactive compounds can cause acute and long-term health problems and lead to addiction. In addition to well-studied and legally controlled compounds like cocaine, new psychoactive substances (NPS) are appearing in street drug markets as replacement strategies and legal alternatives. NPS are effectively marketed as "designer drugs" or "research chemicals" without any knowledge of their underlying pharmacological mode of action and their potential toxicological effects and obviously devoid of any registration process. As of 2016, the knowledge of structure-activity relationships for most NPS is scarce, and predicting detailed pharmacological activity of newly emerging drugs is a challenging task. Therefore, it is important to combine different approaches and employ biological test systems that are superior to mere chemical analysis in recognizing novel and potentially harmful street drugs. In this chapter, we provide a detailed description of techniques to decipher the molecular mechanism of action of NPS that target the high-affinity transporters for dopamine, norepinephrine, and serotonin. In addition, this chapter provides insights into a combined approach to identify and characterize new psychoactive street drugs of unknown content in a collaboration with the Austrian prevention project "checkit!."


Dopamine Plasma Membrane Transport Proteins/drug effects , Norepinephrine Plasma Membrane Transport Proteins/drug effects , Psychotropic Drugs/pharmacology , Serotonin Plasma Membrane Transport Proteins/drug effects , HEK293 Cells , Humans , Illicit Drugs/pharmacology
18.
J Neurochem ; 139(2): 197-207, 2016 10.
Article En | MEDLINE | ID: mdl-27501468

MicroRNAs are short non-coding RNAs that provide global regulation of gene expression at the post-transcriptional level. Such regulation has been found to play a role in stress-induced epigenetic responses in the brain. The norepinephrine transporter (NET) and glucocorticoid receptors are closely related to the homeostatic integration and regulation after stress. Our previous studies demonstrated that NET mRNA and protein levels in rats are regulated by chronic stress and by administration of corticosterone, which is mediated through glucocorticoid receptors. Whether miRNAs are intermediaries in the regulation of these proteins remains to be elucidated. This study was undertaken to determine possible regulatory effects of miRNAs on the expression of NET and glucocorticoid receptors in the noradrenergic neuronal cell line. Using computational target prediction, we identified several candidate miRNAs potentially targeting NET and glucocorticoid receptors. Western blot results showed that over-expression of miR-181a and miR-29b significantly repressed protein levels of NET, which is accompanied by a reduced [3 H] norepinephrine uptake, and glucocorticoid receptors in PC12 cells. Luciferase reporter assays verified that both miR-181a and miR-29b bind the 3'UTR of mRNA of NET and glucocorticoid receptors. Furthermore, exposure of PC12 cells to corticosterone markedly reduced the endogenous levels of miR-29b, which was not reversed by the application of glucocorticoid receptor antagonist mifepristone. These observations indicate that miR-181a and miR-29b can function as the negative regulators of NET and glucocorticoid receptor translation in vitro. This regulatory effect may be related to stress-induced up-regulation of the noradrenergic phenotype, a phenomenon observed in stress models and depressive patients. This study demonstrated that miR-29b and miR-181a, two short non-coding RNAs that provide global regulation of gene expression, markedly repressed protein levels of norepinephrine (NE) transporter and glucocorticoid receptor (GR), as well as NE uptake by binding the 3'UTR of their mRNAs in PC12 cells. Also, exposure of cells to corticosterone significantly reduced miR-29b levels through a GR-independent way.


MicroRNAs/genetics , Norepinephrine Plasma Membrane Transport Proteins/biosynthesis , Receptors, Glucocorticoid/biosynthesis , 3' Untranslated Regions , Animals , Computer Simulation , Corticosterone/pharmacology , Down-Regulation/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , MicroRNAs/biosynthesis , Mifepristone/pharmacology , Norepinephrine/metabolism , Norepinephrine Plasma Membrane Transport Proteins/drug effects , Norepinephrine Plasma Membrane Transport Proteins/genetics , PC12 Cells , Rats , Receptors, Glucocorticoid/drug effects , Receptors, Glucocorticoid/genetics
19.
Bioorg Med Chem ; 24(9): 2137-45, 2016 May 01.
Article En | MEDLINE | ID: mdl-27041397

The limitations of established serotonin (5-hydroxytryptamine, 5-HT) and norepinephrine (NE) reuptake inhibitors necessitate the development of safer and more effective therapeutic agents. Based on the structures of 4-benzylpiperidine carboxamides and trazodone, arylpiperazine-benzylpiperidines with chemical scaffolds different from those of marketed drugs were designed, synthesized, and evaluated for their neurotransmitter reuptake inhibitory activities. The majority of the synthesized compounds showed greater NE than 5-HT reuptake inhibition. The activities were even greater than those of the standard drug, venlafaxine hydrochloride were. The derivatives with a three-carbon linker showed better activities than the derivatives with a two-carbon linker. Among the newly synthesized compounds, 2d exhibited the strongest reuptake inhibition of the neurotransmitters (IC50=0.38 µM for NE and 1.18 µM for 5-HT). The biological activity data demonstrate that arylpiperazine-benzylpiperidines have the potential to be developed as a new class of therapeutic agents to treat neuropsychiatric and neurodegenerative disorders.


Norepinephrine Plasma Membrane Transport Proteins/drug effects , Piperazines/chemical synthesis , Piperazines/pharmacology , Piperidines/chemical synthesis , Piperidines/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology , HEK293 Cells , Humans
20.
J Pharmacol Exp Ther ; 357(1): 24-35, 2016 Apr.
Article En | MEDLINE | ID: mdl-26813929

Thirty-two congeneric rigid adenine nucleoside derivatives containing a North (N)-methanocarba ribose substitution and a 2-arylethynyl group either enhanced (up to 760% of control) or inhibited [(125)I] methyl (1R,2S,3S)-3-(4-iodophenyl)-8-methyl-8-azabicyclo[3.2.1]octane-2-carboxylate (RTI-55) binding at the human dopamine (DA) transporter (DAT) and inhibited DA uptake. Several nucleosides also enhanced [(3)H]mazindol [(±)-5-(4-chlorophenyl)-3,5-dihydro-2H-imidazo[2,1-a]isoindol-5-ol] binding to the DAT. The combination of binding enhancement and functional inhibition suggests possible allosteric interaction with the tropanes. The structure-activity relationship of this novel class of DAT ligands was explored: small N(6)-substition (methyl or ethyl) was favored, while the N1 of the adenine ring was essential. Effective terminal aryl groups include thien-2-yl (compounds 9 and 16), with EC50 values of 35.1 and 9.1 nM, respectively, in [(125)I]RTI-55 binding enhancement, and 3,4-difluorophenyl as in the most potent DA uptake inhibitor (compound 6) with an IC50 value of 92 nM (3-fold more potent than cocaine), but not nitrogen heterocycles. Several compounds inhibited or enhanced binding at the norepinephrine transporter (NET) and serotonin transporter (SERT) and inhibited function in the micromolar range; truncation at the 4'-position in compound 23 allowed for weak inhibition of the SERT. We have not yet eliminated adenosine receptor affinity from this class of DAT modulators, but we identified modifications that remove DAT inhibition as an off-target effect of potent adenosine receptor agonists. Thus, we have identified a new class of allosteric DAT ligands, rigidified adenosine derivatives, and explored their initial structural requirements. They display a very atypical pharmacological profile, i.e., either enhancement by increasing affinity or inhibition of radioligand binding at the DAT, and in some cases the NET and SERT, and inhibition of neurotransmitter uptake.


Adenine/analogs & derivatives , Adenine/pharmacology , Dopamine/metabolism , Norepinephrine/metabolism , Nucleosides/chemistry , Nucleosides/pharmacology , Symporters/drug effects , Symporters/metabolism , Adenine/chemistry , Cocaine/analogs & derivatives , Cocaine/antagonists & inhibitors , Cocaine/metabolism , Cocaine/pharmacology , Dopamine Plasma Membrane Transport Proteins/drug effects , Dopamine Plasma Membrane Transport Proteins/metabolism , HEK293 Cells , Humans , Norepinephrine Plasma Membrane Transport Proteins/drug effects , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Protein Binding/drug effects , Serotonin Plasma Membrane Transport Proteins/drug effects , Serotonin Plasma Membrane Transport Proteins/metabolism , Sodium/metabolism , Structure-Activity Relationship , Vesicular Monoamine Transport Proteins/metabolism
...