Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Molecules ; 27(2)2022 Jan 14.
Article in English | MEDLINE | ID: mdl-35056845

ABSTRACT

Antidepressants target a variety of proteins in the central nervous system (CNS), the most important belonging to the family of G-protein coupled receptors and the family of neurotransmitter transporters. The increasing number of crystallographic structures of these proteins have significantly contributed to the knowledge of their mechanism of action, as well as to the design of new drugs. Several computational approaches such as molecular docking, molecular dynamics, and virtual screening are useful for elucidating the mechanism of drug action and are important for drug design. This review is a survey of molecular targets for antidepressants in the CNS and computer based strategies to discover novel compounds with antidepressant activity.


Subject(s)
Antidepressive Agents/pharmacology , Central Nervous System/drug effects , Neurotransmitter Transport Proteins/antagonists & inhibitors , Receptors, G-Protein-Coupled/antagonists & inhibitors , Central Nervous System/metabolism , Drug Design , Humans , Molecular Docking Simulation
2.
Pharmacol Res ; 176: 106078, 2022 02.
Article in English | MEDLINE | ID: mdl-35026403

ABSTRACT

Antipsychotics represent the mainstay of schizophrenia pharmacological therapy, and their role has been expanded in the last years to mood disorders treatment. Although introduced in 1952, many years of research were required before an accurate picture of how antipsychotics work began to emerge. Despite the well-recognized characterization of antipsychotics in typical and atypical based on their liability to induce motor adverse events, their main action at dopamine D2R to elicit the "anti-psychotic" effect, as well as the multimodal action at other classes of receptors, their effects on intracellular mechanisms starting with receptor occupancy is still not completely understood. Significant lines of evidence converge on the impact of these compounds on multiple molecular signaling pathways implicated in the regulation of early genes and growth factors, dendritic spine shape, brain inflammation, and immune response, tuning overall the function and architecture of the synapse. Here we present, based on PRISMA approach, a comprehensive and systematic review of the above mechanisms under a translational perspective to disentangle those intracellular actions and signaling that may underline clinically relevant effects and represent potential targets for further innovative strategies in antipsychotic therapy.


Subject(s)
Antipsychotic Agents/pharmacology , Animals , Antipsychotic Agents/chemistry , Antipsychotic Agents/therapeutic use , Brain/drug effects , Chromatin Assembly and Disassembly/drug effects , Epigenesis, Genetic , Gene Expression Regulation/drug effects , Genes, Immediate-Early , Humans , Neuronal Plasticity/drug effects , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Neurotransmitter Transport Proteins/antagonists & inhibitors , Neurotransmitter Transport Proteins/metabolism
3.
Neuropharmacology ; 134(Pt A): 133-140, 2018 05 15.
Article in English | MEDLINE | ID: mdl-28823611

ABSTRACT

BACKGROUND: Methylphenidate-based designer drugs are new psychoactive substances (NPS) that are used outside medical settings and their pharmacology is largely unexplored. The aim of the present study was to characterize the pharmacology of methylphenidate-based substances in vitro. METHODS: We determined the potencies of the methylphenidate-based NPS N-benzylethylphenidate, 3,4-dichloroethylphenidate, 3,4-dichloromethylphenidate, ethylnaphthidate, ethylphenidate, 4-fluoromethylphenidate, isopropylphenidate, 4-methylmethylphenidate, methylmorphenate, and propylphenidate and the potencies of the related compounds cocaine and modafinil with respect to norepinephrine, dopamine, and serotonin transporter inhibition in transporter-transfected human embryonic kidney 293 cells. We also investigated monoamine efflux and monoamine receptor and transporter binding affinities. Furthermore, we assessed the cell integrity under assay conditions. RESULTS: All methylphenidate-based substances inhibited the norepinephrine and dopamine transporters 4 to >1000-fold more potently than the serotonin transporter. Similar to methylphenidate and cocaine, methylphenidate-based NPS did not elicit transporter-mediated efflux of monoamines. Besides binding to monoamine transporters, several test drugs had affinity for adrenergic, serotonergic, and rat trace amine-associated receptors but not for dopaminergic or mouse trace amine-associated receptors. No cytotoxicity was observed after drug treatment at assay concentrations. CONCLUSION: Methylphenidate-based substances had pharmacological profiles similar to methylphenidate and cocaine. The predominant actions on dopamine transporters vs. serotonin transporters may be relevant when considering abuse liability. This article is part of the Special Issue entitled 'Designer Drugs and Legal Highs.'


Subject(s)
Central Nervous System Stimulants/pharmacology , Designer Drugs/pharmacology , Methylphenidate/pharmacology , Animals , Biogenic Amines/metabolism , Central Nervous System Stimulants/chemistry , Designer Drugs/chemistry , HEK293 Cells , Humans , Methylphenidate/chemistry , Neurotransmitter Transport Proteins/antagonists & inhibitors , Neurotransmitter Transport Proteins/metabolism , Protein Binding/drug effects , Receptors, Biogenic Amine/metabolism , Transfection
4.
Toxicol In Vitro ; 45(Pt 1): 60-71, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28506818

ABSTRACT

The prevalence and use of new psychoactive substances (NPS) is increasing and currently over 600 NPS exist. Many illicit drugs and NPS increase brain monoamine levels by inhibition and/or reversal of monoamine reuptake transporters (DAT, NET and SERT). This is often investigated using labor-intensive, radiometric endpoint measurements. We investigated the applicability of a novel and innovative assay that is based on a fluorescent monoamine mimicking substrate. DAT, NET or SERT-expressing human embryonic kidney (HEK293) cells were exposed to common drugs (cocaine, dl-amphetamine or MDMA), NPS (4-fluoroamphetamine, PMMA, α-PVP, 5-APB, 2C-B, 25B-NBOMe, 25I-NBOMe or methoxetamine) or the antidepressant fluoxetine. We demonstrate that this fluorescent microplate reader-based assay detects inhibition of different transporters by various drugs and discriminates between drugs. Most IC50 values were in line with previous results from radiometric assays and within estimated human brain concentrations. However, phenethylamines showed higher IC50 values on hSERT, possibly due to experimental differences. Compared to radiometric assays, this high-throughput fluorescent assay is uncomplicated, can measure at physiological conditions, requires no specific facilities and allows for kinetic measurements, enabling detection of transient effects. This assay is therefore a good alternative for radiometric assays to investigate effects of illicit drugs and NPS on monoamine reuptake transporters.


Subject(s)
Amphetamines/pharmacology , Neurotransmitter Transport Proteins/antagonists & inhibitors , Psychotropic Drugs/pharmacology , Cocaine/pharmacology , HEK293 Cells , Humans , Molecular Structure , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Psychotropic Drugs/chemistry
5.
Drug Discov Today ; 20(1): 105-13, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25263697

ABSTRACT

Recent advances in the understanding of molecular recognition and protein-ligand interactions have facilitated rapid development of potent and selective ligands for therapeutically relevant targets. Over the past two decades, a variety of useful approaches and emerging techniques have been developed to promote the identification and optimization of leads that have high potential for generating new therapeutic agents. Intriguingly, the innovation of a fragment-based drug design (FBDD) approach has enabled rapid and efficient progress in drug discovery. In this critical review, we focus on the construction of fragment libraries and the advantages and disadvantages of various fragment-based screening (FBS) for constructing such libraries. We also highlight the deconstruction-reconstruction strategy by utilizing privileged fragments of reported ligands.


Subject(s)
Drug Design , Small Molecule Libraries , Glucokinase/metabolism , Ligands , Neurotransmitter Transport Proteins/antagonists & inhibitors , Receptors, AMPA/metabolism , Receptors, sigma/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , Sigma-1 Receptor
6.
J Neurosci ; 33(25): 10534-43, 2013 Jun 19.
Article in English | MEDLINE | ID: mdl-23785165

ABSTRACT

Mood disorders cause much suffering and lost productivity worldwide, compounded by the fact that many patients are not effectively treated by currently available medications. The most commonly prescribed antidepressant drugs are the selective serotonin (5-HT) reuptake inhibitors (SSRIs), which act by blocking the high-affinity 5-HT transporter (SERT). The increase in extracellular 5-HT produced by SSRIs is thought to be critical to initiate downstream events needed for therapeutic effects. A potential explanation for their limited therapeutic efficacy is the recently characterized presence of low-affinity, high-capacity transporters for 5-HT in brain [i.e., organic cation transporters (OCTs) and plasma membrane monoamine transporter], which may limit the ability of SSRIs to increase extracellular 5-HT. Decynium-22 (D-22) is a blocker of these transporters, and using this compound we uncovered a significant role for OCTs in 5-HT uptake in mice genetically modified to have reduced or no SERT expression (Baganz et al., 2008). This raised the possibility that pharmacological inactivation of D-22-sensitive transporters might enhance the neurochemical and behavioral effects of SSRIs. Here we show that in wild-type mice D-22 enhances the effects of the SSRI fluvoxamine to inhibit 5-HT clearance and to produce antidepressant-like activity. This antidepressant-like activity of D-22 was attenuated in OCT3 KO mice, whereas the effect of D-22 to inhibit 5-HT clearance in the CA3 region of hippocampus persisted. Our findings point to OCT3, as well as other D-22-sensitive transporters, as novel targets for new antidepressant drugs with improved therapeutic potential.


Subject(s)
Antidepressive Agents/pharmacology , Depression/drug therapy , Quinolines/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology , Animals , Blood-Brain Barrier , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Drug Synergism , Electrophysiological Phenomena , Fluvoxamine/pharmacology , Hindlimb Suspension , Hippocampus , Mice , Mice, Inbred C57BL , Mice, Knockout , Microinjections , Neurotransmitter Transport Proteins/antagonists & inhibitors , Neurotransmitter Transport Proteins/metabolism , Octamer Transcription Factor-3/genetics , Quinolines/pharmacokinetics , Serotonin/metabolism , Serotonin Syndrome/psychology , Spectrophotometry, Ultraviolet
7.
Neuropharmacology ; 70: 338-47, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23499664

ABSTRACT

Levomilnacipran (LVM; F2695) is the more active enantiomer of the serotonin/norepinephrine (5-HT/NE) reuptake inhibitor (SNRI) milnacipran and is currently under development for the treatment of major depressive disorder. LVM was benchmarked against two other SNRIs, duloxetine and venlafaxine, in biochemical, neurochemical and pharmacological assays. LVM exhibited high affinity for human NE (Ki = 92.2 nM) and 5-HT (11.2 nM) transporters, and potently inhibited NE (IC50 = 10.5 nM) and 5-HT (19.0 nM) reuptake (human transporter) in vitro. LVM had 2-fold greater potency for norepinephrine relative to serotonin reuptake inhibition (i.e. NE/5-HT potency ratio: 0.6) and 17 and 27 times higher selectivity for NE reuptake inhibition compared with venlafaxine and duloxetine, respectively. LVM did not exhibit affinity for 23 off-target receptors. LVM (i.p.) increased cortical extracellular levels of 5-HT, and NE (minimal effective doses: MEDs = 20 and 10 mg/kg, respectively). In anti-depressive/anti-stress models, i.p. LVM diminished immobility time in the mouse forced swim (MED = 20 mg/kg) and tail suspension (MED = 2.5 mg/kg) tests, and reduced shock-induced ultrasonic vocalizations in rats (MED = 5 mg/kg). Duloxetine and venlafaxine were less potent (MEDs ≥ 10 mg/kg). At doses active in these three therapeutically-relevant models, LVM (i.p.) did not significantly affect spontaneous locomotor activity. In summary, LVM is a potent, selective inhibitor of NE and 5-HT transporters with preferential activity at the former. It is efficacious in models of anti-depressive/anti-stress activity, with minimal potential for locomotor side effects.


Subject(s)
Anxiety/drug therapy , Behavior, Animal/drug effects , Cyclopropanes/pharmacology , Depression/drug therapy , Neurotransmitter Transport Proteins/antagonists & inhibitors , Adrenergic Uptake Inhibitors , Animals , Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cyclohexanols/pharmacology , Cyclopropanes/therapeutic use , Dopamine/metabolism , Duloxetine Hydrochloride , Humans , Male , Mice , Milnacipran , Motor Activity/drug effects , Norepinephrine/metabolism , Rats , Serotonin/metabolism , Synaptosomes/drug effects , Thiophenes/pharmacology , Venlafaxine Hydrochloride
8.
Psychopharmacology (Berl) ; 217(2): 289-95, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21484238

ABSTRACT

RATIONALE: The antidepressant trimipramine shows an atypical pharmacological profile and its mechanism of action is still obscure. OBJECTIVES: The present study investigated whether trimipramine and three of its metabolites interact with targets of other antidepressants, namely, the human monoamine transporters for noradrenaline (hNAT), serotonin (hSERT), and dopamine (hDAT), and with the human organic cation transporters (hOCT1, hOCT2, and hOCT3) which are expressed in the brain and are known to be involved in the uptake of monoamines. METHODS: HEK293 cells heterologously expressing the abovementioned transporters were used to determine the inhibition of [(3)H]MPP(+) uptake by trimipramine and its main metabolites. RESULTS: At concentrations up to 30 µM, all transporters, except hOCT3, were inhibited by all examined substances. With IC(50) values between 2 and 10 µM, trimipramine inhibited hSERT, hNAT, hOCT1, and hOCT2, whereas clearly higher concentrations were needed for half-maximal inhibition of hDAT. Desmethyl-trimipramine showed about the same potencies as trimipramine, whereas 2-hydroxy-trimipramine was less potent at hNAT, hSERT, and hOCT1. Trimipramine-N-oxide preferentially inhibited hSERT. CONCLUSIONS: Neither trimipramine nor its metabolites are highly potent inhibitors of the examined monoamine transporters. However, since at a steady state the sum of the concentrations of the parent compound and its active metabolites is almost two times higher than the plasma concentration of trimipramine and since it is known that tricyclic antidepressants accumulate in the brain (up to tenfold), at least partial inhibition by trimipramine and its metabolites of hSERT and hNAT (but not of hOCT3) may contribute to the antidepressant action of trimipramine.


Subject(s)
Antidepressive Agents , Biogenic Monoamines/metabolism , Neurotransmitter Transport Proteins/antagonists & inhibitors , Organic Cation Transport Proteins/antagonists & inhibitors , Trimipramine , Antidepressive Agents/metabolism , Antidepressive Agents/pharmacology , Cell Culture Techniques , Data Interpretation, Statistical , HEK293 Cells , Humans , Molecular Structure , Neurotransmitter Transport Proteins/genetics , Organic Cation Transport Proteins/genetics , Regression Analysis , Transfection , Trimipramine/metabolism , Trimipramine/pharmacology
9.
J Neurosci ; 31(4): 1284-91, 2011 Jan 26.
Article in English | MEDLINE | ID: mdl-21273413

ABSTRACT

Because insulin acutely enhances the function of dopamine transporters, the tyrosine kinase receptors activated by this hormone may modulate transporter-dependent neurochemical and behavioral effects of psychoactive drugs. In this respect, we examined the effects of insulin on exocytotic monoamine release and the efficacy of the monoamine transporter blocker cocaine in rat nucleus accumbens. Whereas insulin reduced electrically evoked exocytotic [(3)H]dopamine release in nucleus accumbens slices, the hormone potentiated the release-enhancing effect of cocaine thereon. The phosphatidylinositol 3-kinase inhibitor LY294002 abolished these effects, indicating the involvement of insulin receptors. Similar insulin effects were observed on the release of [(3)H]norepinephrine in nucleus accumbens slices, but not on that of [(3)H]serotonin, and were also apparent in medial prefrontal cortex slices. As might then be expected, insulin also potentiated the dopamine and norepinephrine release-enhancing effects of the selective monoamine uptake inhibitors GBR12909 and desmethylimipramine, respectively. In subsequent behavioral experiments, we investigated the role of insulin in motor impulsivity that depends on monoamine neurotransmission in the nucleus accumbens. Intracranial administration of insulin in the nucleus accumbens alone reduced premature responses in the five-choice serial reaction time task and enhanced the stimulatory effect of peripheral cocaine administration on impulsivity, resembling the observed neurochemical effects of the hormone. In contrast, cocaine-induced locomotor activity remained unchanged by intra-accumbal insulin application. These data reveal that insulin presynaptically regulates cocaine-sensitive monoamine transporter function in the nucleus accumbens and, as a consequence, impulsivity. Therefore, insulin signaling proteins may represent targets for the treatment of inhibitory control deficits such as addictive behaviors.


Subject(s)
Biogenic Monoamines/metabolism , Cocaine/pharmacology , Impulsive Behavior/psychology , Insulin/physiology , Neurotransmitter Transport Proteins/antagonists & inhibitors , Animals , Dopamine/metabolism , Impulsive Behavior/physiopathology , In Vitro Techniques , Insulin/pharmacology , Male , Motor Activity/drug effects , Neural Inhibition/drug effects , Neurotransmitter Transport Proteins/physiology , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Rats , Rats, Wistar
10.
Brain Res ; 1309: 172-8, 2010 Jan 14.
Article in English | MEDLINE | ID: mdl-19912995

ABSTRACT

A wide range of data support a role for ambient glutamate (Glu) in epilepsy, although temporal patterns associated with the cellular uptake of Glu have not been addressed in detail. We report on the effects of Glu uptake inhibitors on recurrent seizure-like events (SLEs) evoked by low-[Mg(2+)] condition in juvenile rat hippocampal slices. Effects were compared for inhibitors such as L-trans-pyrrolidine-2,4-dicarboxylate (tPDC), DL-threo-beta-benzyloxyaspartate (DL-TBOA) and dihydrokainic acid (DHK), representing different transporter specificity and transportability profiles. Latency to the first SLE after drug application was shortened by the inhibitors (in % of control: 500 microM tPDC: 54+/-7, 15 microM DL-TBOA: 74+/-5, 50 microM dl-TBOA: 70+/-6, 100 microM DHK: 69+/-4, 300 microM DHK: 71+/-7). Further SLEs were frequently aborted by higher inhibitor concentrations applied (500 microM tPDC: 2/6, 50 microM TBOA: 5/5, 100 microM DHK: 6/8, 300 microM DHK: 3/3). Simultaneous field potential and whole-cell voltage recordings showed depolarization-induced inactivation of CA3 pyramidal neurons during inhibitor application. In the presence of inhibitors, the amplitude of forthcoming SLE was also decreased (in % of control: 500 microM tPDC: 66+/-9, 15 microM dl-TBOA: 88+/-5, 50 microM dl-TBOA: 59+/-6, 100 microM DHK: 67+/-4, 300 microM DHK: 68+/-1). Dependent on type and concentration of the inhibitor, the duration of the first SLE of drug application either increased (100 microM DHK: 375+/-90 %; 100 microM tPDC: 137+/-13 %) or decreased (50 microM TBOA: 62+/-13 %; 300 microM DHK: 60+/-15 %) reflecting differences in subtype-specificity or mechanism of action of the inhibitors. Our findings suggest a role for ambient Glu in the genesis and maintenance of recurrent epileptiform discharges.


Subject(s)
Epilepsy/metabolism , Glutamic Acid/metabolism , Hippocampus/metabolism , Magnesium Deficiency/metabolism , Magnesium/metabolism , Aging/physiology , Animals , Aspartic Acid/pharmacology , CA3 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/metabolism , CA3 Region, Hippocampal/physiopathology , Dicarboxylic Acids/pharmacology , Dose-Response Relationship, Drug , Drug Synergism , Enzyme Inhibitors/pharmacology , Epilepsy/etiology , Epilepsy/physiopathology , Hippocampus/drug effects , Hippocampus/physiopathology , Kainic Acid/analogs & derivatives , Kainic Acid/pharmacology , Magnesium Deficiency/complications , Magnesium Deficiency/physiopathology , Male , Neurons/drug effects , Neurons/metabolism , Neurotransmitter Transport Proteins/antagonists & inhibitors , Neurotransmitter Transport Proteins/metabolism , Organ Culture Techniques , Patch-Clamp Techniques , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Pyrrolidines/pharmacology , Rats , Rats, Wistar , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
12.
Biochem Pharmacol ; 75(9): 1835-47, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18329002

ABSTRACT

Monoamine transporters play key roles in controlling monoamine levels and modulating monoamine reuptake. The objective of the present study was to identify monoamine transporter inhibitors from herbal sources. We discovered that bakuchiol analogs isolated from Fructus Psoraleae inhibited monoamine transporter uptake to differing degrees. The bakuchiol analog, Delta3,2-hydroxybakuchiol was the most potent and efficacious reuptake blocker and was thus selected as the candidate target. Monoamine transporter inhibition by Delta3,2-hydroxybakuchiol was more selective for the dopamine transporter (DAT) (IC50=0.58+/-0.1 microM) and norepinephrine transporter (NET) (IC50=0.69+/-0.12 microM) than for the serotonin transporter (SERT) (IC50=312.02+/-56.69 microM). Delta3,2-Hydroxybakuchiol exhibited greater potency (pEC50 for DAT and NET) than bupropion and exhibited similar efficacy (E(max) for DAT and/or NET) to bupropion and GBR12,935. Pharmacokinetically, Delta3,2-hydroxybakuchiol competitively inhibited DAT and NET with partial reversibility and occupied cocaine binding sites. Moreover, Delta3,2-hydroxybakuchiol counteracted 1-methyl-4-phenylpyridinium-induced toxicity in cells expressing DAT with similar efficacy to GBR12,935. In vivo studies showed that Delta3,2-hydroxybakuchiol increased the activity of intact mice and improved the decreased activity of reserpinized mice. In the conditioned place preference test, preference scores in intact mice were unaffected by Delta3,2-hydroxybakuchiol treatment. Bakuchiol analogs, especially Delta3,2-hydroxybakuchiol, are monoamine transporter inhibitors involved in regulating dopaminergic and noradrenergic neurotransmission and may have represented potential pharmacotherapies for disorders such as Parkinson's disease, depression, and cocaine addiction.


Subject(s)
Biogenic Monoamines/metabolism , Neurotransmitter Transport Proteins/antagonists & inhibitors , Phenols/pharmacology , Psoralea/chemistry , Animals , CHO Cells , Cell Survival/drug effects , Conditioning, Classical/drug effects , Cricetinae , Cricetulus , Fruit/chemistry , Humans , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Neurotransmitter Transport Proteins/genetics , Phenols/chemistry , Phenols/isolation & purification , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Rats , Rats, Sprague-Dawley , Synaptosomes/drug effects , Synaptosomes/metabolism , Transfection
14.
J Pharmacol Exp Ther ; 321(3): 1208-25, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17325229

ABSTRACT

Bicifadine (1-p-tolyl-3-azabicyclo[3.1.0]hexane) inhibits monoamine neurotransmitter uptake by recombinant human transporters in vitro with a relative potency of norepinephrine > serotonin > dopamine (approximately 1:2:17). This in vitro profile is supported by microdialysis studies in freely moving rats, where bicifadine (20 mg/kg i.p.) increased extrasynaptic norepinephrine and serotonin levels in the prefrontal cortex, norepinephrine levels in the locus coeruleus, and dopamine levels in the striatum. Orally administered bicifadine is an effective antinociceptive in several models of acute, persistent, and chronic pain. Bicifadine potently suppressed pain responses in both the Randall-Selitto and kaolin models of acute inflammatory pain and in the phenyl-p-quinone-induced and colonic distension models of persistent visceral pain. Unlike many transport inhibitors, bicifadine was potent and completely efficacious in both phases of the formalin test in both rats and mice. Bicifadine also normalized the nociceptive threshold in the complete Freund's adjuvant model of persistent inflammatory pain and suppressed mechanical and thermal hyperalgesia and mechanical allodynia in the spinal nerve ligation model of chronic neuropathic pain. Mechanical hyperalgesia was also reduced by bicifadine in the streptozotocin model of neuropathic pain. Administration of the D(2) receptor antagonist (-)-sulpiride reduced the effects of bicifadine in the mechanical hyperalgesia assessment in rats with spinal nerve ligations. These results indicate that bicifadine is a functional triple reuptake inhibitor with antinociceptive and antiallodynic activity in acute, persistent, and chronic pain models, with activation of dopaminergic pathways contributing to its antihyperalgesic actions.


Subject(s)
Analgesics/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Pain/drug therapy , Acute Disease , Analgesics/metabolism , Animals , Brain/drug effects , Brain/metabolism , Bridged Bicyclo Compounds, Heterocyclic/metabolism , Chronic Disease , Desipramine/pharmacology , Disease Models, Animal , Dopamine/metabolism , Dose-Response Relationship, Drug , Humans , Male , Mice , Microdialysis , Motor Activity/drug effects , Neurotransmitter Transport Proteins/antagonists & inhibitors , Neurotransmitter Transport Proteins/metabolism , Norepinephrine/metabolism , Pain/metabolism , Pain/physiopathology , Pain Threshold/drug effects , Rats , Rats, Sprague-Dawley , Rats, Wistar , Reaction Time/drug effects , Receptors, Neurotransmitter/antagonists & inhibitors , Receptors, Neurotransmitter/metabolism , Serotonin/metabolism
15.
Mol Interv ; 7(6): 306-9, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18199851

ABSTRACT

The neurotransmitter transporters belonging to the solute carrier 6 (SLC6) family, including the gamma-aminobutyric acid (GAT), norepinephrine (NET), serotonin (SERT) and dopamine (DAT) transporters are extremely important drug targets of great clinical relevance. These Na+, Cl(-)-dependent transporters primarily function following neurotransmission to reset neuronal signaling by transporting neurotransmitter out of the synapse and back into the pre-synaptic neuron. Recent studies have tracked down an elusive binding site for Cl(-) that facilitates neurotransmitter transport using structural differences evident with bacterial family members (e.g., the Aquifex aeolicus leucine transporter LeuT Aa) that lack Cl(-) dependence. Additionally, the crystal structures of antidepressant-bound LeuT Aa reveals a surprising mode of drug interaction that may have relevance for medication development. The study of sequence and structural divergence between LeuT Aa and human SLC6 family transporters can thus inform us as to how and why neurotransmitter transporters evolved a reliance on extracellular Cl(-) to propel the transport cycle; what residue changes and helical rearrangements give rise to recognition of different substrates; and how drugs such as antidepressants, cocaine, and amphetamines halt (or reverse) the transport process.


Subject(s)
Bacterial Proteins/metabolism , Neurotransmitter Transport Proteins/metabolism , Antidepressive Agents/pharmacology , Bacterial Proteins/chemistry , Binding Sites , Humans , Models, Molecular , Multiprotein Complexes , Neurotransmitter Transport Proteins/antagonists & inhibitors , Neurotransmitter Transport Proteins/chemistry
16.
Curr Protoc Pharmacol ; Chapter 9: Unit9.9, 2006 Dec.
Article in English | MEDLINE | ID: mdl-22294181

ABSTRACT

This overview unit describes the core activities involved in the drug discovery and development process, from target identification - including preclinical biology, medicinal and process chemistry - to pharmacokinetics and metabolism (ADME), and also activities related to the to the drug approval process. The latter include the activities related to the filing of an IND (Investigational New Drug) application and also Phases I - III of clinical trials that form the basis of an NDA (New Drug Application) submission, as well as post-marketing Phase IV activities as required by the U.S. Food and Drug Administration (FDA) and the European and Japanese counterparts.


Subject(s)
Drug Discovery/methods , Pharmaceutical Preparations/chemical synthesis , Pharmacology, Clinical/methods , Biological Products/chemistry , Chemistry, Pharmaceutical/methods , Computers , Drug Approval , Drug-Related Side Effects and Adverse Reactions/prevention & control , Humans , Ligands , Neurotransmitter Transport Proteins/antagonists & inhibitors , Pharmaceutical Preparations/metabolism , Pharmacokinetics , RNA Interference/physiology , Receptors, Drug/drug effects , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...