14.01.2013 Views

Regulation of the dopamine transporter - Addiction Research ...

Regulation of the dopamine transporter - Addiction Research ...

Regulation of the dopamine transporter - Addiction Research ...

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

DAT <strong>Regulation</strong> Schmitt & Reith<br />

by <strong>dopamine</strong>rgic neurons can affect <strong>the</strong> DAT. Of <strong>the</strong><br />

possible GPCR subtypes that can affect DAT function,<br />

<strong>dopamine</strong> D2-like autoreceptors are <strong>the</strong> most<br />

logical candidates, with <strong>the</strong>ir presynaptic localization<br />

and clearly defined role in regulatory inhibition<br />

<strong>of</strong> both tyrosine hydroxylase 92 and vesicular<br />

<strong>dopamine</strong> release. 93 However, although D2-like autoreceptors<br />

are <strong>the</strong> most thoroughly investigated,<br />

o<strong>the</strong>r GPCRs also appear to modulate <strong>dopamine</strong>rgic<br />

neurotransmission by altering DAT function.<br />

In particular, activation <strong>of</strong> both �-opioid receptors<br />

and TAAR1 receptors (a member <strong>of</strong> a recently discovered<br />

family <strong>of</strong> receptors for endogenous trace<br />

amines) has been shown to elicit changes in DAT<br />

function.<br />

Dopamine D2 and D3 autoreceptors<br />

The D2-like family <strong>of</strong> <strong>dopamine</strong> receptors comprises<br />

D2,D3,andD4 receptor subtypes. In addition<br />

to <strong>the</strong>ir role as classical postsynaptic receptors, <strong>the</strong><br />

D2 and D3 subtypes are expressed by <strong>dopamine</strong>rgic<br />

neurons <strong>the</strong>mselves, serving as presynaptic autoreceptors.<br />

Activation <strong>of</strong> D2-like autoreceptors attenuates<br />

<strong>dopamine</strong>rgic neurotransmission via several<br />

parallel mechanisms: in PC12 cells, for example,<br />

treatment with <strong>the</strong> D2/D3 agonist quinpirole causes<br />

feedback inhibition <strong>of</strong> tyrosine hydroxylase—<strong>the</strong><br />

rate-determining enzyme in <strong>dopamine</strong> syn<strong>the</strong>sis—<br />

and decreases K + -evoked <strong>dopamine</strong> release. 93 In<br />

addition to reducing stimulated <strong>dopamine</strong> release,<br />

recent evidence suggests that D2 and D3 receptor<br />

activation also reduces extracellular <strong>dopamine</strong> concentration<br />

via acute upregulation <strong>of</strong> DAT function.<br />

This effect was first demonstrated in <strong>the</strong> early 1990s<br />

by using rotating disk electrode voltammetry to<br />

measure <strong>the</strong> rate <strong>of</strong> extracellular <strong>dopamine</strong> clearance.<br />

That is, Meiergerd et al. demonstrated 94 that<br />

<strong>the</strong> D2/D3 agonist quinpirole increases <strong>the</strong> V max <strong>of</strong><br />

<strong>dopamine</strong> transport into rat striatal synaptosomes,<br />

an effect that is reversed by concomitant administration<br />

<strong>of</strong> <strong>the</strong> antagonist sulpiride. The authors also<br />

showed in vivo evidence <strong>of</strong> <strong>the</strong> inverse situation: a<br />

reductionin<strong>the</strong>V max <strong>of</strong> <strong>dopamine</strong> clearance in rats<br />

chronically treated with <strong>the</strong> potent D2 antagonist<br />

haloperidol. In support <strong>of</strong> this finding, localized intrastriatal<br />

application <strong>of</strong> <strong>the</strong> selective D2 antagonist<br />

raclopride has also been shown to decrease clearance<br />

<strong>of</strong> exogenous <strong>dopamine</strong> applied locally via a<br />

second micropipette. 95 Studies using selective D3<br />

receptor ligands paint a similar picture: <strong>the</strong> selective<br />

D3 agonist PD128907 increases electrochemically<br />

measured <strong>dopamine</strong> clearance in rat nucleus accumbens<br />

slices, producing an increase in transport V max<br />

with no effect on <strong>the</strong> K m <strong>of</strong> <strong>dopamine</strong> after a 10min<br />

preincubation. 96 In contrast, <strong>the</strong> D3 receptor<br />

antagonist GR103691 decreased <strong>dopamine</strong> significantly<br />

below vehicle levels. In Xenopus oocytes coexpressing<br />

hDAT and D2R transcripts, activation <strong>of</strong><br />

D2 receptors with apomorphine for as little as 5 min<br />

results in a 40% increase <strong>of</strong> whole-cell [ 3 H]CFT<br />

binding (Bmax) with no alteration in <strong>the</strong> Ki value<br />

for CFT, suggesting that activation <strong>of</strong> D2 receptors<br />

triggers rapid trafficking <strong>of</strong> intracellular DATs to <strong>the</strong><br />

plasma membrane. 97 Upregulation <strong>of</strong> surface DAT<br />

expression by D2 receptor agonists requires Gi/Go<br />

protein coupling, because pretreatment with pertussis<br />

toxin (PTX) blocked <strong>the</strong> increase in [ 3 H]CFT<br />

binding. In our collaborative study with <strong>the</strong> group<br />

<strong>of</strong> Garris, in vivo voltammetry was used to monitor<br />

<strong>dopamine</strong> uptake and release in rat striatum<br />

and nucleus accumbens. 98 We found that D2R antagonism<br />

reduced uptake at all frequencies used to<br />

stimulate <strong>the</strong> medial forebrain bundle, whereas release<br />

was enhanced only at lower frequencies. Be<br />

that as it may, both processes—D2R regulation <strong>of</strong><br />

<strong>dopamine</strong> uptake as well as release—serve as parallel<br />

feedback mechanisms aimed at reducing extracellular<br />

<strong>dopamine</strong> levels when high <strong>dopamine</strong> levels<br />

activate D2 receptors.<br />

Most investigations into <strong>the</strong> effects <strong>of</strong> D2-like receptor<br />

ligands on DAT function in vitro have used<br />

electrochemical methods to assess <strong>dopamine</strong> uptake<br />

in animal tissues, in lieu <strong>of</strong> more commonly<br />

used measurement <strong>of</strong> [ 3 H]<strong>dopamine</strong> uptake by heterologous<br />

cells. The reason for this methodological<br />

preference is purely technical: <strong>dopamine</strong> is a<br />

high-affinity agonist at D2 and D3 receptors, creating<br />

an obvious confound because <strong>the</strong> radioligand<br />

will activate <strong>the</strong> receptors as well. Although electrochemical<br />

detection has greater temporal resolution<br />

than is possible with cell models, 16 it does not allow<br />

direct observation <strong>of</strong> effects on DAT trafficking<br />

in concert with kinetic measures <strong>of</strong> DAT function.<br />

Recent studies have overcome this issue by using a<br />

novel syn<strong>the</strong>tic DAT substrate that is easily detected<br />

yet exhibits negligible affinity for <strong>dopamine</strong> receptors.<br />

The substrate 4-(4-(dimethylamino)styryl)-<br />

N-methylpyridinium (ASP + )—a styryl analogue<br />

<strong>of</strong> <strong>the</strong> neurotoxic substrate MPP + —has <strong>the</strong> added<br />

benefit <strong>of</strong> possessing a fluorescent �-conjugated<br />

326 Ann. N.Y. Acad. Sci. 1187 (2010) 316–340 c○ 2010 New York Academy <strong>of</strong> Sciences.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!