Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Calcium ; 58(5): 457-66, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26162812

ABSTRACT

Amphetamine (AMPH) and its more potent enantiomer S(+)AMPH are psychostimulants used therapeutically to treat attention deficit hyperactivity disorder and have significant abuse liability. AMPH is a dopamine transporter (DAT) substrate that inhibits dopamine (DA) uptake and is implicated in DA release. Furthermore, AMPH activates ionic currents through DAT that modify cell excitability presumably by modulating voltage-gated channel activity. Indeed, several studies suggest that monoamine transporter-induced depolarization opens voltage-gated Ca(2+) channels (CaV), which would constitute an additional AMPH mechanism of action. In this study we co-express human DAT (hDAT) with Ca(2+) channels that have decreasing sensitivity to membrane depolarization (CaV1.3, CaV1.2 or CaV2.2). Although S(+)AMPH is more potent than DA in transport-competition assays and inward-current generation, at saturating concentrations both substrates indirectly activate voltage-gated L-type Ca(2+) channels (CaV1.3 and CaV1.2) but not the N-type Ca(2+) channel (CaV2.2). Furthermore, the potency to achieve hDAT-CaV electrical coupling is dominated by the substrate affinity on hDAT, with negligible influence of L-type channel voltage sensitivity. In contrast, the maximal coupling-strength (defined as Ca(2+) signal change per unit hDAT current) is influenced by CaV voltage sensitivity, which is greater in CaV1.3- than in CaV1.2-expressing cells. Moreover, relative to DA, S(+)AMPH showed greater coupling-strength at concentrations that induced relatively small hDAT-mediated currents. Therefore S(+)AMPH is not only more potent than DA at inducing hDAT-mediated L-type Ca(2+) channel currents but is a better depolarizing agent since it produces tighter electrical coupling between hDAT-mediated depolarization and L-type Ca(2+) channel activation.


Subject(s)
Amphetamine/pharmacology , Calcium Channel Agonists/pharmacology , Calcium Channels/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Calcium/metabolism , HEK293 Cells , Humans , Membrane Potentials
3.
Psychopharmacology (Berl) ; 227(3): 493-9, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23371489

ABSTRACT

RATIONALE: Psychoactive "bath salts" represent a relatively new drug of abuse combination that was placed in Schedule I in October 2011. Two common ingredients of bath salts include the cathinone analogs: mephedrone and methylenedioxypyrovalerone (MDPV). The mechanism of action of these synthetic cathinone analogs has not been well investigated. MATERIALS AND METHODS: Because cathinone and methcathinone are known to act as releasing agents at the human dopamine transporter (hDAT), mephedrone and MDPV were investigated at hDAT expressed in Xenopus oocytes. RESULTS: Whereas mephedrone was found to have the signature of a dopamine-releasing agent similar to methamphetamine or methcathinone, MDPV behaved as a cocaine-like reuptake inhibitor of dopamine. CONCLUSIONS: Mephedrone and MDPV produce opposite electrophysiological signatures through hDAT expressed in oocytes. Implications are that the combination (as found in bath salts) might produce effects similar to a combination of methamphetamine and cocaine.


Subject(s)
Benzodioxoles/pharmacology , Central Nervous System Stimulants/pharmacology , Designer Drugs/pharmacology , Dopamine Plasma Membrane Transport Proteins/metabolism , Methamphetamine/analogs & derivatives , Pyrrolidines/pharmacology , Animals , Benzodioxoles/chemistry , Central Nervous System Stimulants/chemistry , Designer Drugs/chemistry , Dopamine Plasma Membrane Transport Proteins/genetics , Dose-Response Relationship, Drug , Humans , In Vitro Techniques , Membrane Potentials/drug effects , Methamphetamine/chemistry , Methamphetamine/pharmacology , Molecular Structure , Oocytes/metabolism , Patch-Clamp Techniques , Pyrrolidines/chemistry , Structure-Activity Relationship , Synaptic Transmission/drug effects , Transfection , Xenopus laevis , Synthetic Cathinone
4.
Br J Pharmacol ; 168(7): 1750-7, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23170765

ABSTRACT

BACKGROUND AND PURPOSE: Bath salts is the street name for drug combinations that contain synthetic cathinone analogues, among them possibly mephedrone (MEPH) and certainly methylenedioxypyrovalerone (MDPV). In animal studies, cathinone and certain cathinone analogues release dopamine (DA), similar to the action of amphetamine (AMPH) and methamphetamine (METH). AMPH and METH act on the human DA transporter (hDAT); thus, we investigated MEPH and MDPV acting at hDAT. EXPERIMENTAL APPROACH: We recorded electrical currents mediated by hDAT expressed in Xenopus laevis oocytes and exposed to: DA, METH, a known hDAT stimulant and DA releaser, MEPH, MDPV, MEPH + MDPV, or cocaine, a known hDAT inhibitor. KEY RESULTS: DA, METH and MEPH induce an inward current (depolarizing) when the oocyte is held near the resting potential (-60 mV), therefore acting as excitatory hDAT substrates. Structurally analogous MDPV induces an outward (hyperpolarizing) current similar to cocaine, therefore acting as an inhibitory non-substrate blocker. CONCLUSIONS AND IMPLICATIONS: Two components of bath salts, MEPH and MDPV, produce opposite effects at hDAT that are comparable with METH and cocaine, respectively. In our assay, MEPH is nearly as potent as METH; however, MDPV is much more potent than cocaine and its effect is longer lasting. When applied in combination, MEPH exhibits faster kinetics than MDPV, viz., the MEPH depolarizing current occurs seconds before the slower MDPV hyperpolarizing current. Bath salts containing MEPH (or a similar drug) and MDPV might then be expected initially to release DA and subsequently prevent its reuptake via hDAT. Such combined action possibly underlies some of the reported effects of bath salts abuse.


Subject(s)
Benzodioxoles/pharmacology , Central Nervous System Stimulants/pharmacology , Dopamine Plasma Membrane Transport Proteins/metabolism , Methamphetamine/analogs & derivatives , Pyrrolidines/pharmacology , Amphetamine/pharmacology , Animals , Cocaine/pharmacology , Dopamine/pharmacology , Drug Synergism , Female , HEK293 Cells , Humans , Illicit Drugs , Male , Methamphetamine/pharmacology , Oocytes/metabolism , Xenopus laevis , Synthetic Cathinone
5.
Steroids ; 77(7): 774-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22504555

ABSTRACT

The endogenous neurosteroids, pregnenolone sulfate (PS) and 3α-hydroxy-5ß-pregnan-20-one sulfate (PREGAS), have been shown to differentially regulate the ionotropic glutamate receptor (iGluR) family of ligand-gated ion channels. Upon binding to these receptors, PREGAS decreases current flow through the channels. Upon binding to non-NMDA or NMDA receptors containing an GluN2C or GluN2D subunit, PS also decreases current flow through the channels, however, upon binding to NMDA receptors containing an GluN2A or GluN2B subunit, flow through the channels increases. To begin to understand this differential regulation, we have cloned the S1S2 and amino terminal domains (ATD) of the NMDA GluN2B and GluN2D and AMPA GluA2 subunits. Here we present results that show that PS and PREGAS bind to different sites in the ATD of the GluA2 subunit, which when combined with previous results from our lab, now identifies two binding domains for each neurosteroid. We also show both neurosteroids bind only to the ATD of the GluN2D subunit, suggesting that this binding is distinct from that of the AMPA GluA2 subunit, with both leading to iGluR inhibition. Finally, we provide evidence that both PS and PREGAS bind to the S1S2 domain of the NMDA GluN2B subunit. Neurosteroid binding to the S1S2 domain of NMDA subunits responsible for potentiation of iGluRs and to the ATD of NMDA subunits responsible for inhibition of iGluRs, provides an interesting option for therapeutic design.


Subject(s)
Neurotransmitter Agents/metabolism , Pregnanolone/analogs & derivatives , Pregnenolone/metabolism , Receptors, Ionotropic Glutamate/metabolism , Binding Sites , Cloning, Molecular , Models, Molecular , Pregnanolone/metabolism , Receptors, Ionotropic Glutamate/chemistry , Receptors, Ionotropic Glutamate/genetics , Spectrometry, Fluorescence
6.
Br J Pharmacol ; 165(8): 2749-57, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22014068

ABSTRACT

BACKGROUND AND PURPOSE: Wherever they are located, dopamine transporters (DATs) clear dopamine (DA) from the extracellular milieu to help regulate dopaminergic signalling. Exposure to amphetamine (AMPH) increases extracellular DA in the synaptic cleft, which has been ascribed to DAT reverse transport. Increased extracellular DA prolongs postsynaptic activity and reinforces abuse and hedonic behaviour. EXPERIMENTAL APPROACH: Xenopus laevis oocytes expressing human (h) DAT were voltage-clamped and exposed to DA, R(-)AMPH, or S(+)AMPH. KEY RESULTS: At -60mV, near neuronal resting potentials, S(+)AMPH induced a depolarizing current through hDAT, which after removing the drug, persisted for more than 30 min. This persistent leak in the absence of S(+)AMPH was in contrast to the currents induced by R(-)AMPH and DA, which returned to baseline immediately after their removal. Our data suggest that S(+)AMPH and Na(+) carry the initial S(+)AMPH-induced current, whereas Na+ and Cl(-) carry the persistent leak current. We propose that the persistent current results from the internal action of S(+)AMPH on hDAT because the temporal effect was consistent with S(+)AMPH influx, and intracellular S(+)AMPH activated the effect. The persistent current was dependent on Na(+) and was blocked by cocaine. Intracellular injection of S(+)AMPH also activated a DA-induced persistent leak current. CONCLUSIONS AND IMPLICATIONS: We report a hitherto unknown action of S(+)AMPH on hDAT that potentially affects AMPH-induced DA release. We propose that internal S(+)AMPH acts as a molecular stent that holds the transporter open even after external S(+)AMPH is removed. Amphetamine-induced persistent leak currents are likely to influence dopaminergic signalling, DA release mechanisms, and amphetamine abuse.


Subject(s)
Amphetamine/pharmacology , Dopamine Agents/pharmacology , Dopamine Plasma Membrane Transport Proteins/physiology , Dopamine/physiology , Amphetamine/chemistry , Animals , Dopamine Agents/chemistry , Humans , Oocytes , Stereoisomerism , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL
...