Reduced forebrain serotonin transmission is causally involved in the development of compulsive cocaine seeking in rats

Yann Pelloux, Ruth Dilleen, Daina Economidou, David Theobald, Barry J Everitt, Yann Pelloux, Ruth Dilleen, Daina Economidou, David Theobald, Barry J Everitt

Abstract

Whereas the majority of cocaine users quit as they experience the negative consequences of drug use, some lose control over their drug taking and compulsively seek drugs. We report that 20% of rats compulsively seek cocaine despite intermittent negative outcomes after escalating their cocaine self-administration. This compulsive subgroup showed marked reductions in forebrain serotonin utilization; increasing serotonin transmission reduced their compulsive cocaine seeking. Depleting forebrain serotonin induced compulsive cocaine seeking in rats with a limited cocaine taking history; this was reversed by systemic treatment with a 5-hydroxytryptamine (5-HT2C) receptor agonist and mimicked by systemic treatment with a 5-HT2C receptor antagonist in intact animals. These results indicate the causal involvement of reduced serotoninergic transmission in the emergence of compulsive drug seeking after a long cocaine-taking history.

Figures

Figure 1
Figure 1
Emergence of a compulsive cocaine-seeking subpopulation of rats after an extended cocaine-taking history (a) Timeline of the experiment. (b) Early-loading phase during free access sessions, in animals having had 12 days of 1 h (ShA: gray dots) or 6 h (LgA: black dots) daily access to cocaine. Mean±SEM of 45 and 67 animals per group. Significant difference between ShA and LgA: *Fischer's LSD P<0.05. (c) Number of seeking cycles completed before (baseline) and during punishment of NA (white squares), ShA, and LgA sensitive (gray and black squares, respectively) or resistant (gray and black triangles). Mean±SEM of 3–53 animals per group. Significant differences between sensitive and resistant. &Fischer's LSD P<0.05.
Figure 2
Figure 2
Neurochemical correlates of compulsive cocaine seeking. Utilization of 5-HT (panel a), DA (b) and NE (c) in ShA (hashed columns) and ShA (filled columns), sensitive (solid-gray columns, respectively n=19 and 15) and resistant (solid-black columns, n=3 and 9) rats. The data are mean levels (±SEM.) of both left and right hemisphere. 5-HT and DA utilization indices were determined by the ratio of the respective metabolite of the parent transmitter. Significant differences between sensitive and resistant *Fischer's LSD P<0.05.
Figure 3
Figure 3
Reinstatement of punishment sensitivity by selective serotonin reuptake inhibition. (a) Effects of increasing doses of citalopram or (b) atomoxetine on the number of seeking cycles completed under punishment (upper panels) or the concomitant nose-poke responding for sucrose (lower panels) in resistant animals, mean±SEM of five animals. Significantly different from saline: #Fischer's LSD P<0.05.
Figure 4
Figure 4
Bidirectional modulation of compulsive cocaine seeking by serotoninergic manipulations (a) Timeline of the experiment; (b) number of seeking cycles completed before (baseline) and during punishment of cocaine-seeking responding; (c) the effects of increasing doses of mCPP on the number of seeking cycles completed under punishment in the ICV 5,7-DHT and sham-operated control animals: Mean±SEM of 20 and 28 animals. *: significant differences between sham and ICV 5,7-DHT, Fischer LSD P<0.03 #: significant differences from vehicle condition, Fischer's LSD P<0.001 (d) The effect of increasing doses of SB242084 and M100907 on the number of seeking cycles completed under punishment in animals after a short history of cocaine taking. Mean±SEM of 18 animals: significant differences from vehicle condition #: Fischer LSD P<0.04.

References

    1. Ahmed SH, Koob GF. Transition from moderate to excessive drug intake: change in hedonic set point. Science. 1998;282:298–300.
    1. American Psychiatric Association 2000Diagnostic and Statistical Manual of Mental Disorders4th edn.American Psychiatric Association: Washington, DC
    1. Anthony JC, Warner LA, Kessler RC. Comparative epidemiology of dependence on tobacco, alcohol, controlled substances, and inhalants. Exp Clin Psychopharmacol. 1994;2:244–268.
    1. Avanzi V, Silva RC, Macedo CE, Brandão ML. 5-HT mechanisms of median raphe nucleus in the conditioned freezing caused by light/foot-shock association. Physiol Behav. 2003;78:471–477.
    1. Batki SL, Washburn AM, Delucchi K, Jones RT. A controlled trial of fluoxetine in crack cocaine dependence. Drug Alcohol Depend. 1996;41:137–142.
    1. Belin D, Mar AC, Dalley JW, Robbins TW, Everitt BJ. High impulsivity predicts the switch to compulsive cocaine-taking. Science. 2008;320:1352–1355.
    1. Björklund A, Baumgarten HG, Lachenmayer L, Rosengren E. Recovery of brain noradrenaline after 5,7-dihydroxytryptamine-induced axonal lesions in the rat. Cell Tissue Res. 1975;161:145–155.
    1. Blacktop JM, Seubert C, Baker DA, Ferda N, Lee G, Graf EN, et al. Augmented cocaine seeking in response to stress or CRF delivered into the ventral tegmental area following long-access self-administration is mediated by CRF receptor type 1 but not CRF receptor type 2. J Neurosci. 2011;31:11396–11403.
    1. Calu DJ, Stalnaker TA, Franz TM, Singh T, Shaham Y, Schoenbaum G. Withdrawal from cocaine self-administration produces long-lasting deficits in orbitofrontal-dependent reversal learning in rats. Learn Mem. 2007;14:325–328.
    1. Castañé A, Theobald DE, Robbins TW. Selective lesions of the dorsomedial striatum impair serial spatial reversal learning in rats. Behav Brain Res. 2010;210:74–83.
    1. Clarke HF, Robbins TW, Roberts AC. Lesions of the medial striatum in monkeys produce perseverative impairments during reversal learning similar to those produced by lesions of the orbitofrontal cortex. J Neurosci. 2008;28:10972–10982.
    1. Cools R, Clark L, Owen AM, Robbins TW. Defining the neural mechanisms of probabilistic reversal learning using event-related functional magnetic resonance imaging. J Neurosci. 2002;22:4563–4567.
    1. Cooper A, Barnea-Ygael N, Levy D, Shaham Y, Zangen A. A conflict rat model of cue-induced relapse to cocaine seeking. Psychopharmacology. 2007;194:117–125.
    1. Covi L, Hess JM, Kreiter NA, Haertzen CA. Effects of combined fluoxetine and counseling in the outpatient treatment of cocaine abusers. Am J Drug Alcohol Abuse. 1995;21:327–344.
    1. Dalley JW, Theobald DE, Eagle DM, Passetti F, Robbins TW. Deficits in impulse control associated with tonically-elevated serotonergic function in rat prefrontal cortex. Neuropsychopharmacology. 2002;26:716–728.
    1. Dalley JW, Fryer TD, Brichard L, Robinson ES, Theobald DE, Lääne K, et al. Nucleus accumbens D2/3 receptors predict trait impulsivity and cocaine reinforcement. Science. 2007;315:1267–1270.
    1. Deroche-Gamonet V, Belin D, Piazza PV. Evidence for addiction-like behavior in the rat. Science. 2004;305:1014–1017.
    1. Economidou D, Pelloux Y, Robbins TW, Dalley JW, Everitt BJ. High impulsivity predicts relapse to cocaine-seeking after punishment-induced abstinence. Biol Psychiatry. 2009;65:851–856.
    1. Ersche KD, Barnes A, Jones PS, Morein-Zamir S, Robbins TW, Bullmore ET. Abnormal structure of frontostriatal brain systems is associated with aspects of impulsivity and compulsivity in cocaine dependence. Brain. 2011;134:2013–2024.
    1. Ersche KD, Roiser JP, Robbins TW, Sahakian BJ. Chronic cocaine but not chronic amphetamine use is associated with perseverative responding in humans. Psychopharmacology. 2008;197:421–431.
    1. Grabowski J, Rhoades H, Elk R, Schmitz J, Davis C, Creson D, et al. Fluoxetine is ineffective for treatment of cocaine dependence or concurrent opiate and cocaine dependence: two placebo-controlled double-blind trials. J Clin Psychopharmacol. 1995;15:163–174.
    1. Harrison AA, Everitt BJ, Robbins TW. Central 5-HT depletion enhances impulsive responding without affecting the accuracy of attentional performance: interactions with dopaminergic mechanisms. Psychopharmacology. 1997;133:329–342.
    1. Heyne A, Wolffgramm J. The development of addiction to d-amphetamine in an animal model: same principles as for alcohol and opiate. Psychopharmacology. 1998;140:510–518.
    1. Higgins GA, Fletcher PJ. Serotonin and drug reward: focus on 5-HT2C receptors. Eur J Pharmacol. 2003;480:151–162.
    1. Hodos W. Progressive ratio as a measure of reward strength. Science. 1961;134:943–944.
    1. Jacobsen LK, Staley JK, Malison RT, Zoghbi SS, Seibyl JP, Kosten TR, et al. Elevated central serotonin transporter binding availability in acutely abstinent cocaine-dependent patients. Am J Psychiatry. 2000;157:1134–1140.
    1. Letchworth SR, Nader MA, Smith HR, Friedman DP, Porrino LJ. Progression of changes in dopamine transporter binding site density as a result of cocaine self-administration in rhesus monkeys. J Neurosci. 2001;21:2799–2807.
    1. Little KY, McLaughlin DP, Zhang L, Livermore CS, Dalack GW, McFinton PR, et al. Cocaine, ethanol, and genotype effects on human midbrain serotonin transporter binding sites and mRNA levels. Am J Psychiatry. 1998;155:207–213.
    1. Loh EA, Roberts DC. Break-points on a progressive ratio schedule reinforced by intravenous cocaine increase following depletion of forebrain serotonin. Psychopharmacology. 1990;101:262–266.
    1. Melik E, Babar-Melik E, Ozgünen T, Binokay S. Median raphe nucleus mediates forming long-term but not short-term contextual fear conditioning in rats. Behav Brain Res. 2000;112:145–150.
    1. Moeller FG, Schmitz JM, Steinberg JL. Citalopram combined with behavioral therapy reduces cocaine use: A double-blind placebo-controlled trial. Am J Drug Alcohol Abuse. 2007;33:367–378.
    1. Panlilio LV, Thorndike EB, Schindler CW. Reinstatement of punishment-suppressed opioid self-administration in rats: an alternative model of relapse to drug abuse. Psychopharmacology. 2003;168:229–235.
    1. Paxinos G, Watson C. The Rat Brain in Stereotaxic Coordinate. Academic Press: New York; 1996.
    1. Pelloux Y, Everitt BJ, Dickinson A. Compulsive drug seeking by rats under punishment: effects of drug taking history. Psychopharmacology. 2007;194:127–137.
    1. Porrino LJ, Smith HR, Nader MA, Beveridge TJ. The effects of cocaine: a shifting target over the course of addiction. Prog Neuropsychopharmacol Biol Psychiatry. 2007;31:1593–1600.
    1. Porter RH, Benwell KR, Lamb H, Malcolm CS, Allen NH, Revell DF, et al. Functional characterization of agonists at recombinant human 5-HT2A, 5-HT2B and 5-HT2C receptors in CHO-K1 cells. Br J Pharmacol. 1999;128:13–20.
    1. 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.
    1. Rogers RD, Everitt BJ, Baldacchino A, Blackshaw AJ, Swainson R, Wynne K, et al. Dissociable deficits in the decision-making cognition of chronic amphetamine abusers, opiate abusers, patients with focal damage to prefrontal cortex, and tryptophan-depleted normal volunteers: evidence for monoaminergic mechanisms. Neuropsychopharmacology. 1999a;20:322–339.
    1. Rogers RD, Blackshaw AJ, Middleton HC, Matthews K, Hawtin K, Crowley C, et al. Tryptophan depletion impairs stimulus-reward learning while methylphenidate disrupts attentional control in healthy young adults: implications for the monoaminergic basis of impulsive behaviour. Psychopharmacology. 1999b;146:482–491.
    1. Sanders-Bush E, Breeding M, Knoth K, Tsutsumi M. Sertraline-induced desensitization of the serotonin 5HT-2 receptor transmembrane signaling system. Psychopharmacology. 1989;99:64–69.
    1. Schoenbaum G, Saddoris MP, Ramus SJ, Shaham Y, Setlow B. Cocaine-experienced rats exhibit learning deficits in a task sensitive to orbitofrontal cortex lesions. Eur J Neurosci. 2004;19:1997–2002.
    1. Seymour B, Daw ND, Roiser JP, Dayan P, Dolan R. Serotonin selectively modulates reward value in human decision-making. J Neurosci. 2012;32:5833–5842.
    1. Shaham Y, Shalev U, Lu L, De Wit H, Stewart J. The reinstatement model of drug relapse: history, methodology and major findings. Psychopharmacology. 2003;168:3–20.
    1. Spanagel R, Holter SM. Long-term alcohol self-administration with repeated alcohol deprivation phases: an animal model of alcoholism. Alcohol Alcohol. 1999;34:231–243.
    1. Thiébot MH, Hamon M, Soubrié P. The involvement of nigral serotonin innervation in the control of punishment-induced behavioral inhibition in rats. Pharmacol Biochem Behav. 1983;19:225–229.
    1. Vanderschuren LJ, Everitt BJ. Drug seeking becomes compulsive after prolonged cocaine self-administration. Science. 2004;305:1017–1019.
    1. Vayalapalli S, Vaughn M, Salles-Shahid K, Byrd-Sellers J, Drexler K. High-dose citalopram for cocaine dependence in veteran population-a pilot project. Am J Addict. 2011;20:485–486.
    1. Volkow ND, Chang L, Wang GJ, Fowler JS, Ding YS, Sedler M, et al. Low level of brain dopamine D-2 receptors in methamphetamine abusers: association with metabolism in the orbitofrontal cortex. Am J Psychiatry. 2001;158:2015–2021.
    1. Waldorf D, Reinarman C, Murphy S. Cocaine changes: the experience of using and quitting. Temple University Press: Philadelphia; 1991.
    1. Wilson JM, Kalasinsky KS, Levey A, Bergeron C, Reiber G, Anthony RM, et al. Striatal dopamine nerve terminal markers in human, chronic methamphetamine users. Nat Med. 1996;2:699–703.
    1. Winstanley CA, Theobald DE, Dalley JW, Glennon JC, Robbins TW. 5-HT2A and 5-HT2C receptor antagonists have opposing effects on a measure of impulsivity: interactions with global 5-HT depletion. Psychopharmacology. 2004;176:376–385.

Source: PubMed

3
Subskrybuj