Articles

  • the Korean Society for Brain and Neural Sciences

Article

Original Article

Exp Neurobiol 2013; 22(3): 224-231

Published online September 30, 2013

https://doi.org/10.5607/en.2013.22.3.224

© The Korean Society for Brain and Neural Sciences

Cocaine-Induced Behavioral Sensitization in Mice: Effects of Microinjection of Dopamine D2 Receptor Antagonist into the Nucleus Accumbens

Eun-Sol Jung, Hyo Jin Lee, Hye-Ri Sim and Ja-Hyun Baik*

Molecular Neurobiology Laboratory, College of Life Sciences and Biotechnology, Department of Life Sciences, Korea University, Seoul 136-701, Korea

Correspondence to: *To whom correspondence should be addressed.
TEL: 82-2-3290-3455, FAX: 82-2-927-9028
e-mail: jahyunb@korea.ac.kr

To determine the role of dopamine D2 receptor (D2R) in the nucleus accumbens (NAc) core in cocaine-induced behavioral sensitization, D2R antagonist, raclopride was bilaterally microinjected (2.5 or 5 nmol) into the NAc core of WT and D2R-/- mice and the initiation and expression phase of cocaine-mediated locomotor sensitization were analyzed. WT and D2R knockout (D2R-/-) mice received bilateral injections of either saline, or raclopride at the NAc core 30 min before each of five daily repeated injections of saline or cocaine (15 mg/kg i.p.). Following 2 weeks of withdrawal after repeated exposure to cocaine, the animals were pre-treated with an intra-accumbal injection of vehicle or raclopride before receiving a systemic cocaine challenge for the expression of sensitization. Animals which had been microinjected raclopride into NAc core displayed the enhancement of cocaine-induced behavioral response for the initiation but also for the expression of sensitization in WT as well as in D2R-/- mice, which was thus unaltered as compared to vehicle-injected control group. These results suggest that D2R in NAc core is not involved in cocaine-induced behavioral sensitization.

Keywords: dopamine receptor, nucleus accumbens, cocaine, addiction, behavioral sensitization

An initial exposure to psychostimulant such as cocaine induces enhanced locomotor stimulant effect to subsequent administration, a phenomenon known as sensitization [1, 2], producing enduring molecular, cellular and behavioral plasticity that resembles some addiction-related features in humans [1-5]. Process of behavioral sensitization includes two distinct phase; initiation and expression. The initiation phase refers to the period where the increased behavioral response following daily cocaine administration associated with an increase in extracellular dopamine concentration is observed. Behavioral sensitization continues to increase after cessation of cocaine administration and this procedure produces long-lasting sensitization, known as the expression of sensitization [1, 5, 6]. The expression phase shows a persistent drug hyper-responsiveness after cessation of drug, which is associated with a cascade of neuroadaptation [7-9].

Even though this phenomenon has been studied mostly in experimental animals, the neuronal plasticity underlying behavioral sensitization has been suggested to reflect the neuroadaptations that contribute to compulsive drug craving in human [9, 10].

Considerable evidences indicated that behavioral sensitization is associated with enhanced dopamine transmission in mesocorticolimbic system comprising thus, ventral tegmental area, prefrontal cortex and nucleus accumbens (NAc) involving also glutamatergic transmission. Animals behaviorally sensitized to cocaine, amphetamine, nicotine, or morphine [7,11] show enhanced dopamine release in the NAc in response to challenge drug exposure. In addition to changes in neurotransmitter release, dopamine binding to its receptors plays a key role in behavioral sensitization [5].

Dopamine is a neurotransmitter which plays an important role in diverse physiological functions such as locomotion, cognition, motivation and reward in central nervous system [12]. Dopamine receptors have been classified five types as D1-like receptor (D1 and D5) or D2-like receptor (D2, D3 and D4) on molecular structure and pharmacology [13]. Dopamine D2 receptor (D2R) is crucial for addictive-related behavior in mesolimbic dopaminergic pathway, consisting of the ventral tegmental area (VTA) and NAc received glutamatergic input from the corticolimbic structures [14-16].

While several studies have sought to elucidate the contribution of the dopamine receptors in NAc to cocaine-induced behavioral sensitization [17-19], relatively few systematic investigations are available for the role of dopamine receptors in specific period of behavioral sensitization, for example. Recently, we have observed that knockdown of D2R expression by infusing a lentiviral vector for encoding a short hairpin RNA (shRNA) specific for D2R mRNA (Lenti-shD2R) into the NAc core did not affect basal locomotor activity of saline-injected mice or cocaine-induced behavioral sensitization, compared with those observed in mice injected with the control virus [15]. As well, continuous infusion of the D2R antagonist in the NAc showed effects similar to those of D2R knockdown at NAc with Lenti-shD2R, displaying that absence of D2R does not affect the cocaine-induced behavioral sensitization [15].

In the present study, we investigated the effect of selective blocking of D2R in the NAc core on the initiation and the expression period of cocaine-induced behavioral sensitization, respectively, in WT and D2R-/- mice in order to dissect the role of D2R in each phase of cocaine-mediated behavioral sensitization.

Mice

All experiments were performed with wild-type and dopamine D2 receptor knockout (D2R-/-) mice, D2R-/- mice (B6;129S2-Drd2tmllow) were purchased from the Induced Mutant Resource at the Jackson Laboratory (Bar Harbor, ME) and produced from heterozygous D2R+/- mice. The mice were kept in a SPF barrier area, and the temperature (22±1℃) and humidity (50%) were carefully controlled with a 12:12 h light/dark cycle. Food (Purina Certified Rodent Diet, USA) and tap water (membrane filtered purified and autoclaved water) were provided ad libitum.

Drug

Cocaine hydrochloride (Macfarlan, U.K, 10 or 15 mg/kg, i.p.) and S(-)-Raclopride L-tartrate (Sigma-Aldrich, 2.5 or 5nmol/side) were dissolved in saline (0.9%).

Microinfusion in neucleus accumbens

Stereotaxic surgery was performed with male D2R-/- and WT control mice at 12 week -old mice. Animals were anesthetized with 1.6 µl/g Zoletil and 0.05 µl/g xylazine (Rompun, Bayer) intraperitoneallyAt least 5~7 day before experiments, all mice were placed in a Kopf stereotaxic frame and implanted with a 26-gauge bilateral guide cannula (plastics One, Inc., Roanoke, VA) at NAc core of the brain. Stereotaxic coordinates according to the atlas of Paxinos and Flanklin (2004): 1.7 mm anterior and 1.3 mm lateral relative to bregma and 4.5 mm depth. Guide cannula was implanted with the tip 0.3 mm above NAc core on each side. Internal cannula (33 gauge) extend 1mm below the tip of the guide cannula. Microinfusion of the raclopride (S(-)-Raclopride(+)-tartrate salt, Sigma-Aldrich, 5 nmol/side) was carried out using an automated syringe pump (KD Scientific Inc.) 30 min before locomotor activity measurement upon saline- or cocaine-challenge. A 22-gauge 25 µl Hamilton microsyringe connected to a 33-gauge internal cannula. 1µl of saline or raclopride was administered bilaterally into the NAc-core at a rate of 0.2 µl/min. The animals have inserted to stylet. Injection site was verified at the end of the experiments by 1% toluidine blue.

Cocaine induced behavioral sensitization

Apparatus and locomotor activity

Locomotor activity was evaluated using an Activity Monitor (MED Associates Inc., St. Albans, VT, USA), consisting of an open field chamber (43.2×43.2×30.5 cm) with 16×16 photocells for measuring horizontal movements. The locomotor activity was measured as the total distance traveled for 30 min at interval of 5 min. The experimental apparatus was cleaned with 70% ethanol between mice to remove odor cues.

Behavioral sensitization

For the experiments, age-matched WT and D2R-/- mice (12~20 weeks of age) were housed individually and allowed to acclimatize to the cage for 1 week. For each manipulation, mice were transferred to the experimental room 60 min before the onset of the experiment in order to allow for habituation and to reduce stress (brightness of the experimental room was 70 lux). All mice were implanted with a guide cannula at NAc core and allowed to recover 5~7 days in home cage. For initiation of sensitization, mice were habituated to saline injections (i.p.) for three consecutive days and were then injected with saline or cocaine (15 mg/kg, i.p) on five consecutive days. For microinjection of D2R antagonist into the NAc core, the mice received raclopride (2.5 nmol or 5 nmol/side) using an infusion pump. After 30 min, the mice have repeated intraperitoneal (i.p.) injection of saline or cocaine (15 mg/kg) and recorded 30 min for each consecutive five days. For the expression of sensitization, the mice received cocaine (15 mg/kg, i.p.) for consecutive five days and after 14 days of withdrawal, raclopride was infused into NAc core and mice were challenged cocaine (10 mg/kg, i.p.) to test the expression of sensitization.

Criteria for sensitization

Criteria for sensitization were based on the coefficient of variation (standard deviation/mean) for the day 5/day 1 locomotor count ratio in the saline group as described previously35. A cocaine-injected mouse was considered sensitized if its increase in activity over the course of cocaine treatment (day 5/day 1 locomotor count ratio) exceeded the coefficient of variation for the saline group. For this analysis, day5/day1 locomotor count ratios were calculated based on locomotor activity during the 30-min period after injection [20].

Assessment of cannula placement

At completion of the experiments, mice were decapitated, brain were removed and frozen at -70℃. Coronal sections (40 µm thick) were cut on a cryotome. Verifying the cannula placement of NAc core injected 1% toluidine blue. Approximate position of cannula tips for animals receiving microinjections in the core of the nucleus accumbens is presented in Fig. 1.

Statistical analysis

Data are presented as mean±s.e.m. and were analysed with the two-tailed student's t-test, or with two-way analysis of variance followed by Bonferroni's post hoc test. A p-value of <0.05 was considered statistically significant.

D2R antagonist did not affect the initiation of cocaine-induced behavioral sensitization

To determine the role of D2R in the initiation of cocaine-induced behavioral sensitization, we analyzed the development of cocaine-induced behavioral sensitization of WT and D2R knockout (D2R-/-) mice with following experimental schedule (Fig. 2A, B).

Mice were received D2R antagonist (Raclopride, 2.5 nmol) at NAc core by microinjection then mice were injected with cocaine (15 mg/kg) on five consecutive days, and locomotor responses were recorded for 30 min after each injection and cocaine-induced behavioral sensitization was analyzed (Fig. 2). Both WT and D2R-/- mice showed a marked increase in locomotor activity in response to the repeated cocaine injection (Fig. 2C). D2R-/- mice thus manifested cocaine-induced behavioral sensitization similar to that apparent in WT mice, despite the lower basal locomotor activity of the mutant animals as previously reported (WT, raclopride effect: F1.22=0.12 p=0.7361, D2R-/-, raclopride effect: F1.16=0.55 p=0.4692, genotype×cocaine interaction: F1.16=4.29 p=0.0548) (Fig. 2C, D) [15].

Microinjection of D2R antagonist, raclopride of 2.5 nmol at NAc core (Fig. 2) did not affect the cocaine-induced locomotor activity during the initiation phase of behavioral sensitization in WT mice but also in D2R-/- mice (Fig. 2C), showing no difference between vehicle- and antagonist-injected groups.

We then performed another series of experiment with higher concentration of antagonist, 5 nmol of raclopride. We have observed that higher concentration of D2R antagonist did neither alter the cocaine-induced locomotor activity during the initiation phase of behavioral sensitization in WT and D2R-/- mice (WT, raclopride effect: F1.22=0 p=0.99990, D2R-/- raclopride effect: F1.20=0.96 p=0.3381, genotype×cocaine interaction: F1.18=3.49 p=0.0781; Fig. 3B) (Fig. 3).

Therefore, Both WT and D2R-/- mice showed a marked increase in locomotor activity in response to the repeated cocaine injection (Fig. 2, 3), suggesting that the absence of D2R thus did not appear to affect the initiation of behavioral sensitization. As well, blocking the D2 receptor in NAc by the injection of D2R antagonist did not alter the initiation of cocaine-induced sensitization in both WT and D2R-/- mice.

Effect of D2R antagonist microinjection at NAc on the expression of cocaine-induced behavioral sensitization

We next assessed whether blocking of D2R in NAc can alter the expression phase of cocaine-induced behavioral sensitization. After induction of behavioral sensitization by repeated injection of cocaine for 5 days, expression of sensitization was evoked by challenge with a lower dose of cocaine after 14 days of withdrawal period (Fig. 4A). Both cocaine- and saline-treated WT or D2R-/- mice were divided into two groups: one group was injected with vehicle, whereas the other group, subjected to the microinjection of raclopride (Fig. 4A). Microinjection of raclopride (5 nmol) into the NAc core was performed before the cocaine challenge at the expression phase (Fig. 4A). A cocaine challenge after 14 days of withdrawal period produced a sensitized locomotor activity response, which corresponds to the expression of cocaine-induced behavioral sensitization in animals previously exposure to cocaine [cocaine (vehicle/cocaine)] as compared to animals which had not been subjected to the cocaine challenge before [saline (vehicle/cocaine)] in both WT and D2R-/- mice group. Animals which had been microinjected raclopride into NAc core did show also an enhancement of cocaine-induced behavioral response to cocaine challenge in WT but also in D2R-/- mice (for expression, WT, raclopride effect: F1.20=0.05 p=0.8323, raclopride x cocaine interaction: F1.20=0.71 p=0.4100, D2R-/-, raclopride effect: F1.25=0.35 p=0.5614, raclopride×cocaine interaction, F1.25=0.67 p=0.4191, vehicle group-genotype×cocaine interaction: F1.24=0 p=0.9638, Raclopride group-genotype×cocaine interaction: F1.27=0.7 p=0.4109) (Fig. 4B). Therefore the microinjection of D2R antagonist into NAc core did affect neither the initiation nor the expression of cocaine-induced behavioral sensitization in WT and D2R-/- mice.

In the present study, we determined the role of dopamine D2R in NAc core in the initiation and the expression of behavioral sensitization to cocaine respectively in WT and D2R-/- mice.

The nucleus accumbens is a heterogeneous structure [21], can be separated anatomically into core and shell subdivisions [22]. The distinct pattern of core and shell output targets, suggests that two regions may mediate different behavioral processes. It has been suggested that the nucleus accumbens core (NAcc) is important for the maintenance and induction of cocaine seeking behavior [23].

Recently, we have reported that the effects of knockdown of D2R expression at NAc core by infusing a lentiviral vector for a shRNA against D2R expression into the NAc of WT mice on the cocaine-induced sensitization [15]. Depletion of D2R in the NAc did not affect basal locomotor activity nor the cocaine-induced behavioral sensitization but conferred stress-induced inhibition of the expression of cocaine-induced behavioral sensitization [15]. In a similar set of experiments, the effects of infusion of the D2R antagonist raclopride into the NAc core of WT mice, during whole period of initiation but also for every other day during the withdrawal period of 2weeks before the cocaine challenge at the expression of behavioral sensitization, has been examined [15]. Such pharmacological blockade of D2R in the NAc did not affect the expression of sensitization in the control nonstressed group but only in stress-subjected animal group, the expression of sensitization was significantly attenuated by repeated stress during drug withdrawal [15]. Together with this study, our present investigation which separately examined the effect of D2R antagonist at the initiation and the expression respectively, now strongly suggest that the blockade of D2R in NAc core did not prevent the cocaine-mediated behavioral sensitization while certainly D2R at NAc core plays a key role in regulation of synaptic modification triggered by stress and drug addiction.

As to the locomotor sensitization induced by repetitive injection of cocaine, it has been reported that systemic administration of the dopamine D1-like receptor antagonist SCH-23390, or the dopamine D2R antagonists sulpiride, YM-09151-2, eticlopride or raclopride, has been shown to be contradictory, either not to affect or decrease the induction of cocaine sensitization [1, 24-29]. The direct intraaccumbal infusion of D2/D3R antagonist, sulpride in rats have shown that blockade of D2R reverses the acute cocaine-induced locomotion [17, 18], but these studies did not examine the effect on the cocaine-induced behavioral sensitization. Interestingly, it has been reported that the intra-medial prefrontal cortex injection of D2R agonist, quinpirole blocked the initiation and attenuated the expression of cocaine-induced behavioral sensitization [30].

Recent studies using genetically-engineered mice which can express Cre recombinase in cell-type specific manner, revealed some role of D2R-expressing cells in cocaine-addictive behaviors. For example, loss of DARPP-32 in D2R-expressing cells resulted in an enhanced acute locomotor response to cocaine [31]. Using DREADDs (designer receptors exclusively activated by designer drugs) strategies, with viral-mediated expression of an engineered GPCR (Gi/o-coupled human muscarinic M4 designer receptor exclusively activated by a designer drug, hM4D) that is activated by an otherwise pharmacologically inert ligand [32], showed that the activation of striatal D2R-expressing neurons facilitated the development of amphetamine-induced sensitization [32].

It would be possible that NAc D1R may have a more direct role in cocaine-mediated behavioral sensitization. Differently from previous pharmacological or D1R knockout mice studies [1, 24, 25, 33], recent reports using optogentics and other viral-mediated activation/inactivation of D1 and D2Rs revealed that D1R could be important in cocaine-induced behavioral sensitization [34, 35]. Hikida et al. demonstrated that with reversible inactivation of D1/D2 receptor-expressing MSNs with the tetanus toxin, they observed the predominant roles of the D1 receptor-expressing cells in reward learning and cocaine sensitization, but there was no change in sensitization caused by the inactivation of D2 receptor-expressing cells [34]. Moreover, optogenetically activated with conditional Channelrhodopsin2 viruses injected in NAc of D1-Cre mice showed an enhanced cocaine sensitization while the optogenetic activation of D2 receptor-expressing cells in the NAc induced no change in cocaine-induced behavioral sensitization [19]. As well, optogenetic inactivation of D1 receptor-expressing MSNs using the light activated chloride pump, halorhodopsin eNpHR3.0 during cocaine exposure resulted in an attenuation of cocaine-induced locomotor sensitization [35]. Therefore, on the whole, these evidences suggest that dopaminergic mechanisms critically mediate cocaine-induced addictive behaviors, although the precise contribution of D1 and D2Rs remains to be determined.

Since the common circuitry for behavioral sensitization includes dopamine projections from the VTA to the nucleus accumbens and glutamate projections from the mPFC to the nucleus accumbens [5, 36], it will be important to determine if D2R signaling in NAc also interacts with glutamate signaling in control of cocaine-induced behavioral sensitization, which play a significant role in drug relapse.

  1. Vanderschuren LJ, Kalivas PW. Alterations in dopaminergic and glutamatergic transmission in the induction and expression of behavioral sensitization: a critical review of preclinical studies. Psychopharmacology (Berl) 2000;151:99-120.
    Pubmed
  2. Robinson TE, Berridge KC. Addiction. Annu Rev Psychol 2003;54:25-53.
    Pubmed
  3. Kalivas PW, Volkow ND. The neural basis of addiction: a pathology of motivation and choice. Am J Psychiatry 2005;162:1403-1413.
    Pubmed
  4. Everitt BJ, Wolf ME. Psychomotor stimulant addiction: a neural systems perspective. J Neurosci 2002;22:3312-3320.
    Pubmed
  5. Steketee JD, Kalivas PW. Drug wanting: behavioral sensitization and relapse to drug-seeking behavior. Pharmacol Rev 2011;63:348-365.
    Pubmed
  6. Thomas MJ, Beurrier C, Bonci A, Malenka RC. Long-term depression in the nucleus accumbens: a neural correlate of behavioral sensitization to cocaine. Nat Neurosci 2001;4:1217-1223.
    Pubmed
  7. Kalivas PW, Duffy P. Effect of acute and daily cocaine treatment on extracellular dopamine in the nucleus accumbens. Synapse 1990;5:48-58.
    Pubmed
  8. Kalivas PW, Sorg BA, Hooks MS. The pharmacology and neural circuitry of sensitization to psychostimulants. Behav Pharmacol 1993;4:315-334.
    Pubmed
  9. Robinson TE, Berridge KC. The neural basis of drug craving: an incentive-sensitization theory of addiction. Brain Res Brain Res Rev 1993;18:247-291.
    Pubmed
  10. Kalivas PW, Pierce RC, Cornish J, Sorg BA. A role for sensitization in craving and relapse in cocaine addiction. J Psychopharmacol 1998;12:49-53.
    Pubmed
  11. Parsons LH, Justice JB. Serotonin and dopamine sensitization in the nucleus accumbens, ventral tegmental area, and dorsal raphe nucleus following repeated cocaine administration. J Neurochem 1993;61:1611-1619.
    Pubmed
  12. Salgado-Pineda P, Delaveau P, Blin O, Nieoullon A. Dopaminergic contribution to the regulation of emotional perception. Clin Neuropharmacol 2005;28:228-237.
    Pubmed
  13. Schwartz JC, Giros B, Martres MP, Sokoloff P. The dopamine receptor family: molecular biology and pharmacology. Semin Neurosci 1992;4:99-108.
  14. Kim SY, Choi KC, Chang MS, Kim MH, Kim SY, Na YS, Lee JE, Jin BK, Lee BH, Baik JH. The dopamine D2 receptor regulates the development of dopaminergic neurons via extracellular signal-regulated kinase and Nurr1 activation. J Neurosci 2006;26:4567-4576.
    Pubmed
  15. Sim HR, Choi TY, Lee HJ, Kang EY, Yoon S, Han PL, Choi SY, Baik JH. Role of dopamine D2 receptors in plasticity of stress-induced addictive behaviours. Nat Commun 2013;4:1579.
    Pubmed
  16. Bock R, Shin JH, Kaplan AR, Dobi A, Markey E, Kramer PF, Gremel CM, Christensen CH, Adrover MF, Alvarez VA. Strengthening the accumbal indirect pathway promotes resilience to compulsive cocaine use. Nat Neurosci 2013;16:632-638.
    Pubmed
  17. Neisewander JL, O'Dell LE, Redmond JC. Localization of dopamine receptor subtypes occupied by intra-accumbens antagonists that reverse cocaine-induced locomotion. Brain Res 1995;671:201-212.
    Pubmed
  18. Baker DA, Khroyan TV, O'Dell LE, Fuchs RA, Neisewander JL. Differential effects of intra-accumbens sulpiride on cocaine-induced locomotion and conditioned place preference. J Pharmacol Exp Ther 1996;279:392-401.
    Pubmed
  19. Lobo MK, Covington HE, Chaudhury D, Friedman AK, Sun H, Damez-Werno D, Dietz DM, Zaman S, Koo JW, Kennedy PJ, Mouzon E, Mogri M, Neve RL, Deisseroth K, Han MH, Nestler EJ. Cell type-specific loss of BDNF signaling mimics optogenetic control of cocaine reward. Science 2010;330:385-390.
    Pubmed
  20. Boudreau AC, Wolf ME. Behavioral sensitization to cocaine is associated with increased AMPA receptor surface expression in the nucleus accumbens. J Neurosci 2005;25:9144-9151.
    Pubmed
  21. Graybiel AM, Ragsdale CW. Histochemically distinct compartments in the striatum of human, monkeys, and cat demonstrated by acetylthiocholinesterase staining. Proc Natl Acad Sci U S A 1978;75:5723-5726.
    Pubmed
  22. Voorn P, Brady LS, Schotte A, Berendse HW, Richfield EK. Evidence for two neurochemical divisions in the human nucleus accumbens. Eur J Neurosci 1994;6:1913-1916.
    Pubmed
  23. Ito R, Robbins TW, Everitt BJ. Differential control over cocaine-seeking behavior by nucleus accumbens core and shell. Nat Neurosci 2004;7:389-397.
    Pubmed
  24. Mattingly BA, Hart TC, Lim K, Perkins C. Selective antagonism of dopamine D1 and D2 receptors does not block the development of behavioral sensitization to cocaine. Psychopharmacology (Berl) 1994;114:239-242.
    Pubmed
  25. Mattingly BA, Rowlett JK, Ellison T, Rase K. Cocaine-induced behavioral sensitization: effects of haloperidol and SCH 23390 treatments. Pharmacol Biochem Behav 1996;53:481-486.
    Pubmed
  26. Kuribara H. Dopamine D1 receptor antagonist SCH 23390 retards methamphetamine sensitization in both combined administration and early posttreatment schedules in mice. Pharmacol Biochem Behav 1995;52:759-763.
    Pubmed
  27. Steketee JD. Injection of SCH 23390 into the ventral tegmental area blocks the development of neurochemical but not behavioral sensitization to cocaine. Behav Pharmacol 1998;9:69-76.
    Pubmed
  28. White IM, Doubles L, Rebec GV. Cocaine-induced activation of striatal neurons during focused stereotypy in rats. Brain Res 1998;810:146-152.
    Pubmed
  29. Filip M, Frankowska M, Zaniewska M, Przegaliński E, Muller CE, Agnati L, Franco R, Roberts DC, Fuxe K. Involvement of adenosine A2A and dopamine receptors in the locomotor and sensitizing effects of cocaine. Brain Res 2006;1077:67-80.
    Pubmed
  30. Beyer CE, Steketee JD. Cocaine sensitization: modulation by dopamine D2 receptors. Cereb Cortex 2002;12:526-535.
    Pubmed
  31. Bateup HS, Santini E, Shen W, Birnbaum S, Valjent E, Surmeier DJ, Fisone G, Nestler EJ, Greengard P. Distinct subclasses of medium spiny neurons differentially regulate striatal motor behaviors. Proc Natl Acad Sci U S A 2010;107:14845-14850.
    Pubmed
  32. Ferguson SM, Eskenazi D, Ishikawa M, Wanat MJ, Phillips PE, Dong Y, Roth BL, Neumaier JF. Transient neuronal inhibition reveals opposing roles of indirect and direct pathways in sensitization. Nat Neurosci 2011;14:22-24.
    Pubmed
  33. Karlsson RM, Hefner KR, Sibley DR, Holmes A. Comparison of dopamine D1 and D5 receptor knockout mice for cocaine locomotor sensitization. Psychopharmacology (Berl) 2008;200:117-127.
    Pubmed
  34. Hikida T, Kimura K, Wada N, Funabiki K, Nakanishi S. Distinct roles of synaptic transmission in direct and indirect striatal pathways to reward and aversive behavior. Neuron 2010;66:896-907.
    Pubmed
  35. Chandra R, Lenz JD, Gancarz AM, Chaudhury D, Schroeder GL, Han MH, Cheer JF, Dietz DM, Lobo MK. Optogenetic inhibition of D1R containing nucleus accumbens neurons alters cocaine-mediated regulation of Tiam1. Front Mol Neurosci 2013;6:13.
    Pubmed
  36. Pierce RC, Kalivas PW. A circuitry model of the expression of behavioral sensitization to amphetamine-like psychostimulants. Brain Res Brain Res Rev 1997;25:192-216.
    Pubmed