Molecular Psychiatry (2002) 7, 21–26  2002 Nature Publishing Group All rights reserved 1359-4184/02 $15.00 www.nature.com/mp REVIEW ARTICLE Cocaine, reward, movement and monoamine transporters GR Uhl1, FS Hall1 and I Sora1,2

1Molecular Neurobiology Branch, NIDA-IRP, NIH, Baltimore, MD 21224, USA

Recent evidence enriches our understanding of the molecular sites of action of cocaine reward and locomotor stimulation. Dopamine transporter blockade by cocaine appears a suf- ficient explanation for cocaine-induced locomotion. Variation in DAT appears to cause differ- ences in locomotion without stimulation. However, previously-held views that DAT block- ade was the sole site for cocaine reward have been replaced by a richer picture of multitransporter involvement with the rewarding and aversive actions of cocaine. These new insights, derived from studies of knockout mice with simultaneous deletions and/or blockade of multiple transporters, provide a novel model for the rewarding action of this heavily-abused substance and implicate multiple monoamine systems in cocaine’s hedonic activities. Molecular Psychiatry (2002) 7, 21–26. DOI: 10.1038/sj/mp/4000964 Keywords: reward; cocaine; aversion; knockout mouse; reinforcement; polygenic

Cocaine is a prototypical psychomotor stimulant that reward mechanisms. These studies in turn built on increases locomotor activity and elevates mood with prior pharmacologic blockade studies18–22 that revealed rewarding euphoria.1 It also produces fearful and jit- altered psychostimulant self-administration with dopa- tery aversive effects in many who take it.2,3 Cocaine mine depletions or with dopamine receptor blocking blocks monoamine uptake by neuronal plasma mem- . brane transporters for dopamine (DAT), serotonin This ‘DAT-is-it’ hypothesis was a major motivation (SERT) and norepinephrine (NET), and can also block for work that resulted in cloning DAT cDNAs, genes ligand- and voltage-gated channels with somewhat and gene variants in our laboratory and others.23–30 It lower potencies.4 Understanding the relationships drove characterization and study of DAT gene variants between cocaine’s molecular actions and its psycho- in humans29,31–53 and animal models.54,55 It led to the motor stimulant and aversive properties has remained production and characterization of DAT-over- and surprisingly incomplete. This review focuses on recent under-expressing and DAT-knockout mice strains.56–59 advances in our understanding of the complex These studies revealed an exquisite DAT-dependence relationships between cocaine’s molecular actions and of cocaine’s locomotor stimulation. By contrast, they its locomotor stimulant, rewarding and aversive also produced surprising results that refuted the strong properties. ‘DAT-is-it’ hypothesis of cocaine reward. The ‘DAT-is-it’ hypothesis drove thinking about cocaine reward for at least a decade, ideas widely Cocaine-induced locomotion: DAT is it linked to Kuhar, Spealman and their colleagues4–8 who DAT variation in humans and mice links robustly with gathered evidence from structure-activity relationships variation in both baseline locomotion and/or psycho- of transporter-blocking compounds with differential stimulant-induced locomotor activities. Three strains potencies at DAT, SERT and NET. They examined of DAT knockout or knockdown mice produced in sep- relationships between potencies of these compounds arate laboratories each have substantially elevated in tests of reward and binding affinities at each of the locomotor activities in novel environments.56,58,59 Sup- monoamine transporters. They each identified the best ranormal levels of locomotor activity remain elevated correlations for the rewarding effects of cocaine with even after several hours of habituation. When cocaine DAT blockade. These authors also cited prior work is administered, wild-type animals display substantial from lesion studies9–12 and electrical brain stimulation increases in their locomotion that are several times studies13–17 that implicated dopamine pathways in greater than their habituated baseline activity levels. However, DAT knockout mice display no further cocaine-induced increase in activity.56,58,60 Neither Correspondence: GR Uhl, MD, PhD, Molecular Neurobiology, Box SERT nor NET gene knockouts provide any comparable 5180, Baltimore, MD 21224, USA. E-mail: guhlȰintra.nida.nih. reduction in cocaine-induced locomotion.60,61 Knock- gov 2Current address: Psychopharmacology Laboratory, Tokyo Metro- out mice have a lifetime to adapt to loss of a single politan Institute of Psychiatry, Tokyo, Japan gene product, providing cautions in interpretation (see Received 20 June 2001; accepted 27 June 2001 below). Nevertheless, results from these mice clearly Cocaine, reward, movement and monoamine transporters GR Uhl et al 22 indicate that DAT is a necessary and principal site for mouse brain systems that normally expressed the cocaine-induced locomotor stimulation. absent transporters, maintaining cocaine reward. Genetic variation at the mouse and human DAT loci We tested whether DAT- and SERT-expressing sys- also contributes to interesting locomotor phenotypes. tems could each provide such redundancy in the long- Mouse quantitative trait locus (QTL) studies localize a term absence of the other transporter. We constructed portion of the B×D recombinant inbred strain differ- double knockout mice with deletions of both the DAT ence in baseline and psychomotor stimulant-induced and SERT genes, examined their baseline behavioral locomotion near the DAT locus, while additional vari- and biochemical features, and tested their preference ance maps to a second locus that appears to regulate for cocaine.60 Deletions of all of the DAT combined DAT expression.62 Human DAT gene markers have with removal of either half or all of the SERT com- reproducibly linked with attention deficit hyperactiv- pletely eliminated cocaine reward assessed in the CPP ity disorder (ADHD).29,63–68 DAT polymorphisms can paradigm. These results defined, for the first time, a be linked more strongly to the hyperactivity than to the minimal set of gene deletions necessary for elimination attention deficits in this disorder.65 Variation in DAT of cocaine reward. More recent evidence underscores activity is thus a significant contributor to ‘baseline’ the specificity of these observations. Double knockouts locomotor activity levels, while DAT is absolutely of both SERT and NET fail to reduce cocaine reward.70 necessary for cocaine-induced locomotor stimulation. In fact, SERT/NET double knockouts dramatically enhance cocaine place preference. While we have thus identified a specific group of Cocaine, reward and aversion: rich patterns of multi- sites whose elimination can totally block measures of transporter involvement cocaine reward, we have also identified potential sites This simple story for DAT and cocaine locomotion for some of the aversive effects reported by cocaine contrasts strikingly with results for rewarding proper- users. Data from reduced or enhanced cocaine reward ties of cocaine. We have tested DAT knockout mice in in the absence of specific transporters or transporter the conditioned place preference (CPP) paradigm58 that combinations can be tentatively assembled with prior measures preference for an environment paired with a data from other transporter blocking drugs as follows drug. Homozygous DAT knockout mice express per- (see also Figure 1): fectly intact cocaine conditioned place preference. DAT appears to be the transporter most associated Another DAT knockout strain was also rewarded by with rewarding properties of cocaine in conditioned cocaine in an operant self-administration paradigm.69 place preference tests. Compounds with substantial Two strains of DAT knockouts thus provided convinc- ing evidence that DAT was not necessary for cocaine reward, results fatal to any strong version of the ‘DAT- is-it’ hypothesis for cocaine reward. Studies of reward in mice with deletions of the other monoamine transporters at which cocaine acts did not provide any clear alternative single transporter site for cocaine’s rewarding actions. Cocaine was no less rewarding in SERT knockouts tested in the CPP para- digm58 or in NET knockout mice.61 The failure of any single monoamine transporter gene knockout strain to eliminate cocaine reinforcement and reward thus left open several possible roles for these transporters in cocaine reward/reinforcement in wild-type mice. These even included the possibility that non-trans- porter molecular sites of cocaine could be involved in cocaine reward/reinforcement. We chose to pursue a different hypothesis: that cocaine normally works as a ‘dirty drug’, producing rewarding effects through simultaneous actions at Figure 1 Model for the relative contributions of blockade of more than one transporter site. Multiple molecular the transporters for dopamine (DAT), serotonin (SERT) and sites for cocaine’s rewarding/reinforcing actions could norepinephrine (NET) on cocaine reward and aversive fea- provide redundancies so that no one site alone would tures in wild-type animals. DAT: Dopamine transporter block- be absolutely required for cocaine reward. Such redun- ade is drawn as exerting largely rewarding properties. SERT: dancies could even be enhanced by compensatory Serotonin transporter blockade is drawn as exerting a combi- nation of rewarding and aversive properties. Uneven distri- mechanisms active in mice that develop without one butions of these influences (lumps in the figure) are meant or more transporters. If cocaine normally altered activi- to suggest different properties of action at different serotonin ties in several parallel or interactive brain systems with receptor subtypes. NET: Norepinephrine transporter blockade substantial redundancies, the systems expressing the is drawn as exerting largely aversive properties. These contri- remaining transporter(s) might thus compensate for butions change significantly in knockout mice, as noted in loss of cocaine-modulated activities in knockout the text.

Molecular Psychiatry Cocaine, reward, movement and monoamine transporters GR Uhl et al 23 selectivity for DAT can produce place preferences and aversive features in some animal models.78–80 They fit are self-administered by animals, although often less with the failure of mazindol, a widely used combined avidly than cocaine (for review see Rothman and DAT/NET blocker, to exhibit any striking human clini- Glowa).71 However, as noted above, DAT is not ‘it’, cal abuse liability and with its not-infrequent discon- since DAT deletion alone does not eliminate cocaine tinuation due to jitteriness and sleep disturbances.81–83 reward.58 Data from DAT knockouts are also tempered by Caveats: brain changes in knockout mice and cocaine results examining the role of SERT and NET in the reward maintenance of cocaine reward in these mice. The The precise neuroadaptive changes that may contribute selective SERT blocker fluoxetine and the selective to the differences in the rewarding consequences of NET blocker nisoxetine both produce strong place pref- monoamine transporter blockade in single and mul- erences in DAT knockouts but not in wild-type mice tiple transporter knockout mice are unknown. The or in heterozygote mice with one knockout DAT allele, brains of mice of each of these knockout strains appear one wild-type DAT allele and half wild-type levels of grossly normal, although the smaller brains of DAT KO DAT expression.70 These data clearly indicate that mice are consistent with the smaller overall size of blockade of SERT or NET can acquire rewarding these mice.84 Each single transporter knockout exhibits properties in DAT’s lifelong absence. How much DAT a distinctive pattern of brain neurochemical rearrange- removal and how long this removal needs to be carried ments,58,60,61,85,86 many of which can be tentatively out to achieve these striking effects remain interesting linked to homeostatic mechanisms triggered by current questions. The adaptive mechanisms that make increased tonic levels of extracellular neurotransmit- fluoxetine and nisoxetine rewarding in DAT knockouts ters. Thus, the DAT knockouts that display enhanced could well contribute to the reward that cocaine con- extracellular dopamine levels56,87 and reduced DA fers in DAT knockout mice, since cocaine retains its clearance56,87,88 also display altered levels of ability to block both SERT and NET in these animals. expression of dopamine D1, D2 and D3 receptors levels Recent reports that the NET blocker roboxetine tyrosine hydroxylase, dopamine-modulated neuropep- enhances nucleus accumbens dopamine in DAT tides, and other features.89,90 Microarray studies of knockout but not in wild-type mice72 could contribute mRNA from the brains of DAT and of SERT knockouts one biochemical substrate for the nisoxetine reward also identify dozens of genes whose expression is con- found in DAT knockouts. Such mechanisms may not sistently altered more than two-fold.91 None of these be relevant for fluoxetine reward, however, since a characterizations provide evidence for large supra- selective SERT blocker did not increase nucleus additive knockout influences on any brain neurochem- accumbens dopamine levels in the same study. ical, microdialysis or gene expression profile when SERT blockade could well contribute to both mice with multiple knockouts have been examined to rewarding and aversive properties of cocaine. Such date, however. No current known neurochemical alter- explanations seem among the most plausible current ation follows the pattern of retained or absent cocaine explanations for otherwise apparently contradictory reward across these multiple knockout strains. data that demonstrate: (1) enhanced cocaine reward in In DAT knockouts, cocaine could also acquire novel SERT knockout mice;58 (2) enhanced fluoxetine reward properties by modulating dopamine acting as a ‘false in DAT and in NET knockout mice;70 and (3) ablated transmitter’ in cells in which it is not synthesized. cocaine reward in DAT/SERT combined knockout Since NET has good affinities for dopamine and SERT mice.60 can even accumulate dopamine with lower affinity, SERT blockade might normally produce a combi- some of the dopamine that is synthesized in dopami- nation of rewarding and aversive features by aug- nergic neurons of DAT knockout mice could become a menting serotonin actions at different serotonin recep- ‘false transmitter’ at adjacent norepinephrine or sero- tor subtypes expressed differentially in distinct tonin terminals. In a similar fashion norepinephrine serotonergic circuits. If these rewarding and aversive could be a false transmitter at adjacent dopaminergic features normally balanced each other to produce little or serotonergic terminals in NET knockouts, and/or net reward or aversion with SERT blockade, fluoxetine serotonin a false transmitter in SERT knockout dopa- or other selective serotonin reuptake inhibitors would mine or norepinephrine terminals.92,93 While some display minimal aversion and also little abuse liability false transmission thus seems likely in these knock- in humans, as has been observed.73–75 The fluoxetine outs, it seems an unlikely explanation for all of the cur- reward acquired by DAT and by NET knockout mice rent data derived from these mice. NET and SERT is consistent with the idea that each of these knockouts blockade by cocaine fails to provide the robust shifts the balance between rewarding and aversive ser- enhancements of striatal dopamine efflux in DAT otonergic components, although not necessarily in knockouts that would be expected if false transmission identical ways. were widespread in these mice.85 Although cocaine NET blockade may well contribute less to rewarding and the NET blocker reboxetine do enhance dopamine and more to aversive properties of cocaine in wildtype efflux in the nucleus accumbens of DAT knockouts, animals. These observations are consistent with fre- rewarding SERT blockade by fluoxetine fails to quent human clinical self-reports of ‘jitteriness’ with enhance this dopamine efflux.72 Further studies will be cocaine use.76,77 They accord with evidence for cocaine necessary to define how much of the cocaine, fluoxet-

Molecular Psychiatry Cocaine, reward, movement and monoamine transporters GR Uhl et al 24 ine and/or nisoxetine rewarding responses found in 10 Roberts DC, Koob GF, Klonoff P, Fibiger HC. Extinction and recov- DAT knockouts depend on false neurotransmission. ery of cocaine self-administration following 6-hydroxydopamine lesions of the nucleus accumbens. Pharmacol Biochem Behav Equally plausible explanations for current data include 1980; 12:781–787. adaptations in circuits such as those that regulate 11 Roberts DC, Koob GF. Disruption of cocaine self-administration dopamine cell firing rates and dopamine release after following 6-hydroxydopamine lesions of the ventral tegmental cocaine administration.94–103 area in rats. Pharmacol Biochem Behav 1982; 17: 901–904. 12 Lyness WH, Friedle NM, Moore KE. Destruction of dopaminergic nerve terminals in nucleus accumbens: effect on d-amphetamine self-administration. Pharmacol Biochem Behav 1979; 11:553– Conclusions 556. Lifelong deletions of DAT, SERT, NET or transporter 13 Edmonds DE, Gallistel CR. Reward versus performance in self- stimulation: electrode-specific effects of alpha-methyl-p-tyrosine combinations each distinctively alter the rewarding on reward in the rat. J Comp Physiol Psychol 1977; 91:962–974. effects of blocking the remaining transporters. These 14 Gallistel CR, Boytim M, Gomita Y, Klebanoff L. Does pimozide results are consistent with the idea that cocaine nor- block the reinforcing effect of brain stimulation? Pharmacol mally works as a dirty drug that provides both Biochem Behav 1982; 17: 769–781. 15 Wasserman EM, Gomita Y, Gallistel CR. Pimozide blocks rewarding and aversive properties by distinct actions reinforcement but not priming from MFB stimulation in the rat. at these three transporters. The combined observations Pharmacol Biochem Behav 1982; 17:783–787. document adaptations that occur in knockout mice. 16 Gomita Y, Gallistel CR. Effects of reinforcement-blocking doses of They also tentatively fit with the human abuse liability pimozide on neural systems driven by rewarding stimulation of of a number of widely used drug classes whose mem- the MFB: a 14C-2-deoxyglucose analysis. Pharmacol Biochem Behav 1982; 17:841–845. bers act differentially at these three monoamine trans- 17 Gallistel CR, Karras D. Pimozide and amphetamine have opposing porters. Current data from monoamine transporter effects on the reward summation function. Pharmacol Biochem knockouts thus provide several novel pathways to Behav 1984; 20:73–77. thinking about cocaine therapeutics. Drugs that block 18 Gunne LM, Anggard E, Jonsson LE. Clinical trials with ampheta- cocaine’s uptake-inhibiting actions at DAT and SERT mine-blocking drugs. Psychiatr Neurol Neurochir 1972; 75: 225– 226. but allow it to continue to block uptake by NET could 19 Wilson MC, Schuster CR. The effects of on psych- provide a possible means for cocaine antagonism at omotor stimulant self-administration in the rhesus monkey. Psy- ‘rewarding’ transporters and continued cocaine action chopharmacologia 1972; 26: 115–126. at its ‘aversive’ sites. Each of the observations summar- 20 Yokel RA, Wise RA. Attenuation of intravenous amphetamine reinforcement by central dopamine blockade in rats. Psychophar- ized above provides new pharmacological routes to macol (Berl) 1976; 48:311–318. dramatically alter the rewarding valences of cocaine. 21 Yokel RA, Wise RA. Increased lever pressing for amphetamine after pimozide in rats: implications for a dopamine theory of reward. Science 1975; 187:547–549. Acknowledgements 22 De Wit H, Wise RA. Blockade of cocaine reinforcement in rats with the dopamine receptor blocker pimozide, but not with the The authors are grateful to each of the co-investigators noradrenergic blockers phentolamine or phenoxybenzamine. Can who have contributed to this work, to helpful com- J Psychol 1977; 31:195–203. ments from Dr Steven Goldberg, and to financial sup- 23 Shimada S, Kitayama S, Lin CL, Patel A, Nanthakumar E, Gregor port from the NIDA-IRP. P et al. Cloning and expression of a cocaine-sensitive dopamine transporter complementary DNA. Science 1991; 254:576–578. 24 Kilty JE, Lorang D, Amara SG. Cloning and expression of a cocaine-sensitive rat dopamine transporter. Science 1991; 254: References 578–579. 1 Johanson CE, Fischman MW. The of cocaine 25 Giros B, el Mestikawy S, Godinot N, Zheng K, Han H, Yang-Feng T related to its abuse. Pharmacol Rev 1989; 41:3–52. et al. Cloning, pharmacological characterization, and chromosome 2 Gawin FH. Cocaine addiction: psychology and neurophysiology. assignment of the human dopamine transporter. Mol Pharmacol Science 1991; 251: 1580–1586. 1992; 42:383–390. 3 Satel SL, Southwick SM, Gawin FH. Clinical features of cocaine 26 Vandenbergh DJ, Persico AM, Hawkins AL, Griffin CA, Li X, Jabs induced paranoia. NIDA Res Monogr 1991; 105: 371. EW et al. Human dopamine transporter gene (DAT1) maps to chro- 4 Kuhar MJ, Ritz MC, Boja JW. The dopamine hypothesis of the mosome 5p15.3 and displays a VNTR. Genomics 1992; 14: reinforcing properties of cocaine. Trends Neurosci 1991; 14: 1104–1106. 299–302. 27 Vandenbergh DJ, Persico AM, Uhl GR. A human dopamine trans- 5 Spealman RD, Madras BK, Bergman J. Effects of cocaine and porter cDNA predicts reduced glycosylation, displays a novel related drugs in nonhuman primates. II. Stimulant effects on repetitive element and provides racially-dimorphic TaqI RFLPs. schedule-controlled behavior. J Pharmacol Exp Ther 1989; 251: Brain Res Mol Brain Res 1992; 15:161–166. 142–149. 28 Lossie AC, Vandenbergh DJ, Uhl GR, Camper SA. Localization of 6 Ritz MC, Lamb RJ, Goldberg SR, Kuhar MJ. Cocaine receptors on the dopamine transporter gene, Datl, on mouse chromosome 13. dopamine transporters are related to self-administration of Mamm Genome 1994; 5: 117–118. cocaine. Science 1987; 237: 1219–1223. 29 Vandenbergh DJ, Thompson MD, Cook EH, Bendahhou E, Nguyen 7 Kuhar MJ, Ritz MC, Sharkey J. Cocaine receptors on dopamine T, Krasowski MD et al. Human dopamine transporter gene: coding transporters mediate cocaine-reinforced behavior. NIDA Res Mon- region conservation among normal, Tourette’s disorder, alcohol ogr 1988; 88:14–22. dependence and attention-deficit hyperactivity disorder popu- 8 Ritz MC, Lamb RJ, Goldberg SR, Kuhar MJ. Cocaine self-adminis- lations. Mol Psychiatry 2000; 5:283–292. tration appears to be mediated by dopamine uptake inhibition. 30 Donovan DM, Vandenbergh DJ, Perry MP, Bird GS, Ingersoll R, Prog Neuropsychopharmacol Biol Psychiatry 1988; 12:233–239. Nanthakumar E et al. Human and mouse dopamine transporter 9 Roberts DC, Corcoran ME, Fibiger HC. On the role of ascending genes: conservation of 5Ј-flanking sequence elements and gene catecholaminergic systems in intravenous self-administration of structures. Brain Res Mol Brain Res 1995; 30:327–335. cocaine. Pharmacol Biochem Behav 1977; 6:615–620. 31 Persico AM, Macciardi F. Genotypic association between dopam-

Molecular Psychiatry Cocaine, reward, movement and monoamine transporters GR Uhl et al 25 ine transporter gene polymorphisms and schizophrenia. Am J Med porter gene polymorphisms are not associated with polysubstance Genet 1997; 74:53–57. abuse. Biol Psychiatry 1993; 34:265–267. 32 Persico AM, Catalano M. Lack of association between dopamine 51 Persico AM, Wang ZW, Black DW, Andreasen NC, Uhl GR, Crowe transporter gene polymorphisms and delusional disorder. Am J RR. Exclusion of close linkage of the dopamine transporter gene Med Genet 1998; 81:163–165. with schizophrenia spectrum disorders. Am J Psychiatry 1995; 33 Grunhage F, Schulze TG, Muller DJ, Lanczik M, Franzek E, Albus 152: 134–136. M et al. Systematic screening for DNA sequence variation in the 52 Sabol SZ, Nelson ML, Fisher C, Gunzerath L, Brody CL, Hu S et coding region of the human dopamine transporter gene (DAT1). al. A genetic association for cigarette smoking behavior. Health Mol Psychiatry 2000; 5:275–282. Psychol 1999; 18:7–13. 34 Hamilton SP, Haghighi F, Heiman GA, Klein DF, Hodge SE, Fyer 53 Lerman C, Caporaso NE, Audrain J, Main D, Bowman ED, Lock- AJ et al. Investigation of dopamine receptor (DRD4) and dopamine shin B et al. Evidence suggesting the role of specific genetic factors transporter (DAT) polymorphisms for genetic linkage or associ- in cigarette smoking. Health Psychol 1999; 18:14–20. ation to panic disorder. Am J Med Genet 2000; 96: 324–330. 54 Miller GM, Yatin SM, De La Garza R, 2nd, Goulet M, Madras BK. 35 Kim JW, Kim DH, Kim SH, Cha JK. Association of the dopamine Cloning of dopamine, norepinephrine and serotonin transporters transporter gene with Parkinson’s disease in Korean patients. J from monkey brain: relevance to cocaine sensitivity(1). Brain Res Korean Med Sci 2000; 15: 449–451. Mol Brain Res 2001; 87:124–143. 36 Martinez D, Gelernter J, Abi-Dargham A, van Dyck CH, Kegeles L, 55 Miller GM, De La Garza RD, 2nd, Novak MA, Madras BK. Single Innis RB et al. The variable number of tandem repeats polymor- nucleotide polymorphisms distinguish multiple dopamine trans- phism of the dopamine transporter gene is not associated with porter alleles in primates: implications for association with atten- significant change in dopamine transporter phenotype in humans. tion deficit hyperactivity disorder and other neuropsychiatric dis- 2001; 24: 553–560. orders. Mol Psychiatry 2001; 6:50–58. 37 Samochowiec J, Rybakowski F, Czerski P, Zakrzewska M, Stepie´ 56 Giros B, Jaber M, Jones SR, Wightman RM, Caron MG. Hyperloco- N et al. Polymorphisms in the dopamine, serotonin, and norepi- motion and indifference to cocaine and amphetamine in mice nephrine transporter genes and their relationship to temperamen- lacking the dopamine transporter. Nature 1996; 379:606–612. tal dimensions measured by the temperament and character 57 Donovan DM, Miner LL, Perry MP, Revay RS, Sharpe LG, Przed- inventory in healthy volunteers. Neuropsychobiology 2001; 43: borski S et al. Cocaine reward and MPTP toxicity: alteration by 248–253. regional variant dopamine transporter overexpression. Brain Res 38 Heinz A, Goldman D, Jones DW, Palmour R, Hommer D, Gorey JG Mol Brain Res 1999; 73:37–49. et al. Genotype influences in vivo dopamine transporter avail- 58 Sora I, Wichems C, Takahashi N, Li XF, Zeng Z, Revay R et al. ability in human striatum. Neuropsychopharmacology 2000; 22: Cocaine reward models: conditioned place preference can be 133–139. established in dopamine- and in serotonin-transporter knockout 39 Kang AM, Palmatier MA, Kidd KK. Global variation of a 40-bp mice. Proc Natl Acad Sci USA 1998; 95: 7699–7704. VNTR in the 3Ј-untranslated region of the dopamine transporter 59 Zhuang X, Oosting RS, Jones SR, Gainetdinov RR, Miller GW, gene (SLC6A3). Biol Psychiatry 1999; 46: 151–60. Caron MG et al. Hyperactivity and impaired response habituation 40 Jonsson EG, Nothen MM, Gustavsson JP, Neidt H, Bunzel R, in hyperdopaminergic mice. Proc Natl Acad Sci USA 2001; 98: Propping P et al. Polymorphisms in the dopamine, serotonin, and 1982–1987. norepinephrine transporter genes and their relationships to 60 Sora I, Hall FS, Andrews AM, Itokawa M, Li XF, Wei HB et al. monoamine metabolite concentrations in CSF of healthy volun- Molecular mechanisms of cocaine reward: combined dopamine teers. Psychiatry Res 1998; 79:1–9. and serotonin transporter knockouts eliminate cocaine place pref- 41 Leighton PW, Le Couteur DG, Pang CC, McCann SJ, Chan D, Law erence. Proc Natl Acad Sci USA 2001; 98: 5300–5305. LK et al. The dopamine transporter gene and Parkinson’s disease 61 Xu F, Gainetdinov RR, Wetsel WC, Jones SR, Bohn LM, Miller GW in a Chinese population. Neurology 1997; 49: 1577–1579. et al. Mice lacking the norepinephrine transporter are supersensi- 42 Dobashi I, Inada T, Hadano K. Alcoholism and gene polymor- tive to psychostimulants. Nat Neurosci 2000; 3: 465–471. phisms related to central dopaminergic transmission in the 62 Buck K, Lischka T, Dorow J, Crabbe J. Mapping quantitative trait Japanese population. Psychiatr Genet 1997; 7:87–91. loci that regulate sensitivity and tolerance to quinpirole, a dopa- 43 Plante-Bordeneuve V, Taussig D, Thomas F, Said G, Wood NW, mine mimetic selective for D(2)/D(3) receptors. Am J Med Genet Marsden CD et al. Evaluation of four candidate genes encoding 2000; 96:696–705. proteins of the dopamine pathway in familial and sporadic Park- 63 Barr CL, Xu C, Kroft J, Feng Y, Wigg K, Zai G et al. Haplotype inson’s disease: evidence for association of a DRD2 allele. Neurol- study of three polymorphisms at the dopamine transporter locus ogy 1997; 48:1589–1593. confirm linkage to attention-deficit/hyperactivity disorder. Biol 44 Fujiwara Y, Yamaguchi K, Tanaka Y, Tomita H, Shiro Y, Kashih- Psychiatry 2001; 49:333–339. ara K et al. Polymorphism of dopamine receptors and transporter 64 Daly G, Hawi Z, Fitzgerald M, Gill M. Mapping susceptibility loci genes in neuropsychiatric diseases. Eur Neurol 1997; 38 (Suppl in attention deficit hyperactivity disorder: preferential trans- 1): 6–10. mission of parental alleles at DAT1, DBH and DRD5 to affected 45 Inada T, Sugita T, Dobashi I, Inagaki A, Kitao Y, Matsuda G et al. children. Mol Psychiatry 1999; 4:192–196. Dopamine transporter gene polymorphism and psychiatric symp- 65 Waldman ID, Rowe DC, Abramowitz A, Kozel ST, Mohr JH, Sher- toms seen in schizophrenic patients at their first episode. Am J man SL et al. Association and linkage of the dopamine transporter Med Genet 1996; 67:406–408. gene and attention- deficit hyperactivity disorder in children: het- 46 Gomez-Casero E, Perez de Castro I, Saiz-Ruiz J, Llinares C, Fernan- erogeneity owing to diagnostic subtype and severity. Am J Hum dez-Piqueras J. No association between particular DRD3 and DAT Genet 1998; 63:1767–1776. gene polymorphisms and manic-depressive illness in a Spanish 66 Gill M, Daly G, Heron S, Hawi Z, Fitzgerald M. Confirmation of sample. Psychiatr Genet 1996; 6:209–212. association between attention deficit hyperactivity disorder and 47 Bodeau-Pean S, Laurent C, Campion D, Jay M, Thibaut F, Dollfus a dopamine transporter polymorphism. Mol Psychiatry 1997; 2: S et al. No evidence for linkage or association between the dopam- 311–313. ine transporter gene and schizophrenia in a French population. 67 Cook EH, Jr, Stein MA, Krasowski MD, Cox NJ, Olkon DM, Kieffer Psychiatry Res 1995; 59:1–6. JE et al. Association of attention-deficit disorder and the dopamine 48 Daniels J, Williams J, Asherson P, McGuffin P, Owen M. No transporter gene. Am J Hum Genet 1995; 56: 993–998. association between schizophrenia and polymorphisms within the 68 Swanson J, Posner M, Fusella J, Wasdell M, Sommer T, Fan J. genes for debrisoquine 4-hydroxylase (CYP2D6) and the dopamine Genes and attention deficit hyperactivity disorder. Curr Psychiatry transporter (DAT). Am J Med Genet 1995; 60:85–87. Rep 2001; 3:92–100. 49 Gelernter J, Kranzler HR, Satel SL, Rao PA. Genetic association 69 Rocha BA, Fumagalli F, Gainetdinov RR, Jones SR, Ator R, Giros between dopamine transporter protein alleles and cocaine- B et al. Cocaine self-administration in dopamine-transporter induced paranoia. Neuropsychopharmacology 1994; 11:195–200. knockout mice [published erratum appears in Nat Neurosci 1998; 50 Persico AM, Vandenbergh DJ, Smith SS, Uhl GR. Dopamine trans- 1: 330]. Nat Neurosci 1998; 1: 132–137.

Molecular Psychiatry Cocaine, reward, movement and monoamine transporters GR Uhl et al 26 70 Hall FS, Sora I, Li XF, Lesch KP, Murphy DL, Xu F et al. Cocaine the dopamine transporter. Proc Natl Acad Sci USA 1998; 95: mechanisms: enhanced cocaine, fluoxetine and nisoxetine place 4029–4034. preferences following monoamine transporter deletions. J Neuro- 88 Benoit-Marand M, Jaber M, Gonon F. Release and elimination of sci (submitted). dopamine in vivo in mice lacking the dopamine transporter: func- 71 Rothman RB, Glowa JR. A review of the effects of dopaminergic tional consequences. Eur J Neurosci 2000; 12: 2985–2992. agents on humans, animals, and drug-seeking behavior, and its 89 Jaber M, Dumartin B, Sagne C, Haycock JW, Roubert C, Giros B et implications for medication development. Focus on GBR 12909. al. Differential regulation of tyrosine hydroxylase in the basal gan- Mol Neurobiol 1995; 11:1–19. glia of mice lacking the dopamine transporter. Eur J Neurosci 72 Carboni E, Spielewoy C, Vacca C, Nosten-Bertrand M, Giros B, Di 1999; 11: 3499–3511. Chiara G. Cocaine and amphetamine increase extracellular dopa- 90 Fauchey V, Jaber M, Caron MG, Bloch B, Le Moine C. Differential mine in the nucleus accumbens of mice lacking the dopamine regulation of the dopamine D1, D2 and D3 receptor gene transporter gene. J Neurosci 2001; 21: RC141. expression and changes in the phenotype of the striatal neurons 73 Yen TT, Fuller RW. Preclinical pharmacology of fluoxetine, a sero- in mice lacking the dopamine transporter. Eur J Neurosci 2000; tonergic drug for weight loss. Am J Clin Nutr 1992; 55(1 Suppl): 12:19–26. 177S–180S. 91 Liu QR, Hall FS, Sora I, Uhl GR. Unpublished data. 74 Tella SR. Effects of monoamine reuptake inhibitors on cocaine 92 Stamford JA, Kruk ZL, Millar J. Striatal dopamine terminals self-administration in rats. Pharmacol Biochem Behav 1995; 51: release serotonin after 5-HTP pretreatment: in vivo voltammetric 687–692. data. Brain Res 1990; 515:173–180. 75 Zawertailo LA, Busto U, Kaplan HL, Sellers EM. Comparative 93 Vanhatalo S, Soinila S. Release of false transmitter serotonin from abuse liability of sertraline, alprazolam, and dextroamphetamine the dopaminergic nerve terminals of the rat pituitary intermediate in humans. J Clin Psychopharmacol 1995; 15:117–124. lobe. Neurosci Res 1995; 22: 367–374. 76 Grabowski J, Roache JD, Schmitz JM, Rhoades H, Creson D, 94 Yadid G, Pacak K, Kopin IJ, Goldstein DS. Endogenous serotonin Korszun A. Replacement medication for cocaine dependence: stimulates striatal dopamine release in conscious rats. J Pharma- methylphenidate. J Clin Psychopharmacol 1997; 17: 485–488. col Exp Ther 1994; 270: 1158–1165. 77 Lobl JK, Carbone LD. Emergency management of cocaine intoxi- 95 Bonhomme N, De Deurwaerdere P, Le Moal M, Spampinato U. cation. Counteracting the effects of today’s ‘favorite drug’. Post- Evidence for 5-HT4 receptor subtype involvement in the enhance- grad Med 1992; 91:161–162, 165–166. ment of striatal dopamine release induced by serotonin: a microdi- 78 Ettenberg A, Raven MA, Danluck DA, Necessary BD. Evidence for alysis study in the halothane-anesthetized rat. Neuropharmacol- opponent-process actions of intravenous cocaine. Pharmacol ogy 1995; 34: 269–279. Biochem Behav 1999; 64:507–512. 96 Boulenguez P, Rawlins JN, Chauveau J, Joseph MH, Mitchell SN, 79 DeVries AC, Pert A. Conditioned increases in anxiogenic-like Gray JA. Modulation of dopamine release in the nucleus accum- bens by 5-HT1B agonists: involvement of the hippocampo-accum- behavior following exposure to contextual stimuli associated with bens pathway. Neuropharmacology 1996; 35: 1521–1529. cocaine are mediated by corticotropin-releasing factor. Psycho- 97 Lejeune F, Millan MJ. Induction of burst firing in ventral tegmen- pharmacol (Berl) 1998; 137: 333–340. tal area dopaminergic neurons by activation of serotonin (5-HT)1A 80 DeVries AC, Taymans SE, Sundstrom JM, Pert A. Conditioned receptors: WAY 100,635- reversible actions of the highly selective release of corticosterone by contextual stimuli associated with ligands, flesinoxan and S 15535. Synapse 1998; 30:172–180. cocaine is mediated by corticotropin-releasing factor. Brain Res 98 West AR, Galloway MP. Regulation of serotonin-facilitated dopa- 1998; 786:39–46. mine release in vivo: the role of protein kinase A activating trans- 81 Pickworth WB, Klein SA, Bunker EB, Henningfield JE. Assessment duction mechanisms. Synapse 1996; 23:20–27. of mazindol for abuse liability. NIDA Res Monogr 1991; 105:443. 99 Berretta N, Bernardi G, Mercuri NB. Alpha(1)-adrenoceptor- 82 Chait LD, Uhlenhuth EH, Johanson CE. Reinforcing and subjective mediated excitation of substantia nigra pars reticulata neurons. effects of several anorectics in normal human volunteers. J Pharm- Neuroscience 2000; 98:599–604. acol Exp Ther 1987; 242: 777–783. 100 Bowers BJ, Henry MB, Thielen RJ, McBride WJ. Serotonin 5-HT(2) 83 Wilson MC, Schuster CR. Mazindol self-administration in the rhe- receptor stimulation of dopamine release in the posterior but not sus monkey. Pharmacol Biochem Behav 1976; 4: 207–210. anterior nucleus accumbens of the rat. J Neurochem 2000; 75: 84 Bosse R, Fumagalli F, Jaber M, Giros B, Gainetdinov RR, Wetsel 1625–1633. WC et al. Anterior pituitary hypoplasia and dwarfism in mice 101 Di Giovanni G, Di Matteo V, Di Mascio M, Esposito E. Preferential lacking the dopamine transporter. Neuron 1997; 19:127–138. modulation of mesolimbic vs nigrostriatal dopaminergic function 85 Gainetdinov RR, Jones SR, Caron MG. Functional hyperdopami- by serotonin(2C/2B) receptor agonists: a combined in vivo electro- nergia in dopamine transporter knock-out mice. Biol Psychiatry physiological and microdialysis study. Synapse 2000; 35:53–61. 1999; 46:303–311. 102 Di Matteo V, Di Mascio M, Di Giovanni G, Esposito E. Acute 86 Bengel D, Murphy DL, Andrews AM, Wichems CH, Feltner D, administration of amitriptyline and mianserin increases dopa- Heils A et al. Altered brain serotonin homeostasis and locomotor mine release in the rat nucleus accumbens: possible involvement insensitivity to 3,4-methylenedioxymethamphetamine (‘Ecstasy’) of serotonin2C receptors. Psychopharmacol (Berl) 2000; 150: in serotonin transporter-deficient mice. Mol Pharmacol 1998; 53: 45–51. 649–655. 103 Shi WX, Pun CL, Zhang XX, Jones MD, Bunney BS. Dual effects 87 Jones SR, Gainetdinov RR, Jaber M, Giros B, Wightman RM, Caron of D-amphetamine on dopamine neurons mediated by dopamine MG. Profound neuronal plasticity in response to inactivation of and nondopamine receptors. J Neurosci 2000; 20: 3504–3511.

This article is a ‘United States Government Work’ paper as defined by the US Copyright Act.

Molecular Psychiatry