Variation within the serotonin (5-HT) 5-HT₂C receptor system aligns with vulnerability to cocaine cue reactivity

N C Anastasio, S Liu, L Maili, S E Swinford, S D Lane, R G Fox, S C Hamon, D A Nielsen, K A Cunningham, F G Moeller, N C Anastasio, S Liu, L Maili, S E Swinford, S D Lane, R G Fox, S C Hamon, D A Nielsen, K A Cunningham, F G Moeller

Abstract

Cocaine dependence remains a challenging public health problem with relapse cited as a major determinant in its chronicity and severity. Environmental contexts and stimuli become reliably associated with its use leading to durable conditioned responses ('cue reactivity') that can predict relapse as well as treatment success. Individual variation in the magnitude and influence of cue reactivity over behavior in humans and animals suggest that cue-reactive individuals may be at greater risk for the progression to addiction and/or relapse. In the present translational study, we investigated the contribution of variation in the serotonin (5-HT) 5-HT2C receptor (5-HT2CR) system in individual differences in cocaine cue reactivity in humans and rodents. We found that cocaine-dependent subjects carrying a single nucleotide polymorphism (SNP) in the HTR2C gene that encodes for the conversion of cysteine to serine at codon 23 (Ser23 variant) exhibited significantly higher attentional bias to cocaine cues in the cocaine-word Stroop task than those carrying the Cys23 variant. In a model of individual differences in cocaine cue reactivity in rats, we identified that high cocaine cue reactivity measured as appetitive approach behavior (lever presses reinforced by the discrete cue complex) correlated with lower 5-HT2CR protein expression in the medial prefrontal cortex and blunted sensitivity to the suppressive effects of the selective 5-HT2CR agonist WAY163909. Our translational findings suggest that the functional status of the 5-HT2CR system is a mechanistic factor in the generation of vulnerability to cocaine-associated cues, an observation that opens new avenues for future development of biomarker and therapeutic approaches to suppress relapse in cocaine dependence.

Figures

Figure 1
Figure 1
Cocaine-dependent subjects exhibit attentional bias toward cocaine-related words on the cocaine-word Stroop task. The difference in mean reaction times between trials with cocaine-related words and those with neutral words was used as a measure of attentional bias toward cocaine-related words. (a) Cocaine-dependent subjects displayed longer reaction times (msec; mean±s.e.m.) to indicate the word color in trials with cocaine-related words vs trials with neutral words (P<0.01 vs neutral words). (b) Accuracy (ratio of correct responses to total trials on either neutral or cocaine-related word trials) did not differ between cocaine-related and neutral word trials in cocaine-dependent subjects.
Figure 2
Figure 2
The highest attentional bias is seen in African-American cocaine-dependent subjects with the Ser23 protein variant. Mean reaction times (msec±s.e.m.) for African-American subjects with the C/CC genotype which encodes for the Ser23 protein variant were significantly greater than that for African-American subjects with the G/GG genotype (*P<0.05 vs G/GG genotype).
Figure 3
Figure 3
Individual differences in appetitive approach behavior in rats are driven by discrete cocaine-associated stimuli. The levels of operant behavior within the cocaine-associated context (left) or reinforced by the discrete cue complex (right) at 24 h of withdrawal were assessed in rats stratified as high (HCR) vs low cue reactive (LCR). Previously-active lever presses did not differ between HCR (n=12) and LCR (n=12) rats upon exposure to the context in the absence of the discrete cue complex (first 10 min of session; NS). HCR rats displayed significantly higher cue-reinforced lever presses (second 60 min of session) vs LCR rats (*P<0.05 vs LCR rats).
Figure 4
Figure 4
High cue reactive (HCR) rats exhibit lower 5-HT2CR protein expression in medial prefrontal cortex (mPFC) relative to low cue reactive (LCR) rats. (a) Mean total lever presses (±s.e.m.) on the previously-active and inactive levers are presented for the cue reactivity test session. Each previously-active lever press resulted in the presentation of the discrete cue complex in the absence of cocaine delivery on an FR1. Rats identified as HCR (n=5) displayed significantly higher lever presses for cocaine-associated cues vs LCR rats (n=6; *P<0.01). Inactive lever presses did not differ between HCR and LCR rats. (b) Qualitative and (c) quantitative data demonstrate phenotypic differences in mPFC 5-HT2CR synaptosomal protein expression. HCR rats displayed lower cortical synaptosomal 5-HT2CR protein levels relative to LCR rats (*P<0.05). (d) An inverse correlation was observed between mPFC 5-HT2CR synaptosomal protein and responses on the previously-active lever for cocaine-associated cues in individual rats (r=0.815; P<0.01). The differential protein expression observed in HCR (0.051±0.002 arbitrary units) and LCR (0.076±0.008 arbitrary units) rats was not related to the cue reactivity test itself as comparable 5-HT2CR mPFC protein levels were observed in HCR (n=6; 0.049±0.01 arbitrary units) and LCR rats (n=6; 0.087±0.02 arbitrary units) that remained in their home cage until sacrifice.
Figure 5
Figure 5
High cue reactive (HCR) rats exhibit lower sensitivity to the suppressive effects of the selective 5-HT2CR agonist WAY163909 relative to low cue reactive (LCR) rats. Mean active lever presses (±s.e.m.) are presented for the cue reactivity test session. Each previously active lever press resulted in the presentation of the discrete cue complex in the absence of cocaine delivery on an FR1. Rats identified as HCR (n=16 rats per treatment) and LCR rats (n=16 rats per treatment) were injected with saline (15 min; 1 ml kg−1, intraperitoneal) or WAY163909 (15 min; 0.5 mg kg−1; intraperitoneal) prior to a cocaine cue reactivity test session on FA day 1. WAY163909 significantly attenuated lever presses for cocaine-associated cues in LCR, but not HCR, rats (*P<0.05 vs saline-treated LCR rats). FA, forced abstinence.

References

    1. Koob GF, Volkow ND. Neurocircuitry of addiction. Neuropsychopharmacology. 2010;35:217–238.
    1. Janes AC, Pizzagalli DA, Richardt S, deB FB, Chuzi S, Pachas G, et al. Brain reactivity to smoking cues prior to smoking cessation predicts ability to maintain tobacco abstinence. Biol Psychiatry. 2010;67:722–729.
    1. Back SE, Hartwell K, DeSantis SM, Saladin M, Rae-Clark AL, Price KL, et al. Reactivity to laboratory stress provocation predicts relapse to cocaine. Drug Alcohol Depend. 2010;106:21–27.
    1. Rohsenow DJ, Niaura RS, Childress AR, Abrams DB, Monti PM. Cue reactivity in addictive behaviors: theoretical and treatment implications. Int J Addict. 1990;25 ((7A-8A:957–993.
    1. Carter BL, Tiffany ST. Cue-reactivity and the future of addiction research. Addiction. 1999;94:349–351.
    1. Field M, Cox WM. Attentional bias in addictive behaviors: a review of its development, causes, and consequences. Drug Alcohol Depend. 2008;97 ((1-2:1–20.
    1. Modesto-Lowe V, Kranzler HR. Using cue reactivity to evaluate medications for treatment of cocaine dependence: a critical review. Addiction. 1999;94:1639–1651.
    1. Mahler SV, de Wit H. Cue-reactors: individual differences in cue-induced craving after food or smoking abstinence. PLoS One. 2010;5:e15475.
    1. Flagel SB, Robinson TE, Clark JJ, Clinton SM, Watson SJ, Seeman P, et al. An animal model of genetic vulnerability to behavioral disinhibition and responsiveness to reward-related cues: implications for addiction. Neuropsychopharmacology. 2010;35:388–400.
    1. Tomie A, Grimes KL, Pohorecky LA. Behavioral characteristics and neurobiological substrates shared by Pavlovian sign-tracking and drug abuse. Brain Res Rev. 2008;58:121–135.
    1. Hendershot CS, Witkiewitz K, George WH, Marlatt GA. Relapse prevention for addictive behaviors. Subst Abuse Treat Prev Policy. 2011;6:17.
    1. Prisciandaro JJ, Myrick H, Henderson S, Rae-Clark AL, Brady KT. Prospective associations between brain activation to cocaine and no-go cues and cocaine relapse. Drug Alcohol Depend. 2013;131 ((1-2:44–49.
    1. Goldstein RZ, Tomasi D, Alia-Klein N, Honorio CJ, Maloney T, Woicik PA, et al. Dopaminergic response to drug words in cocaine addiction. J Neurosci. 2009;29:6001–6006.
    1. Lidov HGW, Grzanna R, Molliver ME. The serotonin innervation of the cerebral cortex in the rat: An immunohistochemical analysis. Neuroscience. 1980;5:207–227.
    1. Vertes RP, Linley SB.Efferent and afferent connections of the dorsal and median raphe nuclei in the rat Monti JM, Pandi-Perumal SR, Jacobs BL, Nutt DJ.(eds).Serotonin and Sleep: Molecular, Functional and Clinical Aspects Birkhäuser Verlag: Basel, Switzerland; pp. 69–102., 2008.
    1. Cunningham KA, Anastasio NC. Serotonin at the nexus of impulsivity and cue ceactivity in cocaine addiction. Neuropharmacology. 2013;76 ((Pt B:460–478.
    1. Pompeiano M, Palacios JM, Mengod G. Distribution of the serotonin 5-HT2 receptor family mRNAs: Comparison between 5-HT2A and 5-HT2C receptors. Mol Brain Res. 1994;23:163–178.
    1. Uylings HB, Groenewegen HJ, Kolb B. Do rats have a prefrontal cortex. Behav Brain Res. 2003;146 ((1-2:3–17.
    1. Goldstein RZ, Tomasi D, Rajaram S, Cottone LA, Zhang L, Maloney T, et al. Role of the anterior cingulate and medial orbitofrontal cortex in processing drug cues in cocaine addiction. Neuroscience. 2007;144:1153–1159.
    1. Baeg EH, Jackson ME, Jedema HP, Bradberry CW. Orbitofrontal and anterior cingulate cortex neurons selectively process cocaine-associated environmental cues in the rhesus monkey. J Neurosci. 2009;29:11619–11627.
    1. Lu H, Chefer S, Kurup PK, Guillem K, Vaupel DB, Ross TJ, et al. fMRI response in the medial prefrontal cortex predicts cocaine but not sucrose self-administration history. Neuroimage. 2012;62:1857–1866.
    1. Pentkowski NS, Duke FD, Weber SM, Pockros LA, Teer AP, Hamilton EC, et al. Stimulation of medial prefrontal cortex serotonin 2C 5-HT2C receptors attenuates cocaine-seeking behavior. Neuropsychopharmacology. 2010;35:2037–2048.
    1. Cunningham KA, Anastasio NC, Fox RG, Stutz SJ, Bubar MJ, Swinford SE, et al. Synergism between a serotonin 5-HT2A receptor (5-HT2AR) antagonist and 5-HT2CR agonist suggests new pharmacotherapeutics for cocaine addiction. ACS Chem Neurosci. 2013;4:110–121.
    1. Neisewander JL, Acosta JI. Stimulation of 5-HT2C receptors attenuates cue and cocaine-primed reinstatement of cocaine-seeking behavior in rats. Behav Pharmacol. 2007;18:791–800.
    1. Fletcher PJ, Grottick AJ, Higgins GA. Differential effects of the 5-HT2A receptor antagonist M100,907 and the 5-HT2C receptor antagonist SB242,084 on cocaine-induced locomotor activity, cocaine self-administration and cocaine-induced reinstatement of responding. Neuropsychopharmacology. 2002;27:576–586.
    1. Grottick AJ, Fletcher PJ, Higgins GA. Studies to investigate the role of 5-HT(2C) receptors on cocaine- and food-maintained behavior. J Pharmacol Exp Ther. 2000;295:1183–1191.
    1. Cunningham KA, Fox RG, Anastasio NC, Bubar MJ, Stutz SJ, Moeller FG, et al. Selective serotonin 5-HT2C receptor activation suppresses the reinforcing efficacy of cocaine and sucrose but differentially affects the incentive-salience value of cocaine- vs sucrose-associated cues. Neuropharmacology. 2011;61:513–523.
    1. Manvich DF, Kimmel HL, Cooper DA, Howell LL. The serotonin 2C receptor antagonist SB 242084 exhibits abuse-related effects typical of stimulants in squirrel monkeys. J Pharmacol Exp Ther. 2012;342:761–769.
    1. Pelloux Y, Dilleen R, Economidou D, Theobald D, Everitt BJ. Reduced forebrain serotonin transmission is causally involved in the development of compulsive cocaine seeking in rats. Neuropsychopharmacology. 2012;37:2505–2514.
    1. Manvich DF, Kimmel HL, Howell LL. Effects of serotonin 2C receptor agonists on the behavioral and neurochemical effects of cocaine in squirrel monkeys. J Pharmacol Exp Ther. 2012;341:424–434.
    1. Anastasio NC, Stutz SJ, Fox RG, Sears RM, Emeson RB, DiLeone RJ, et al. Functional status of the serotonin 5-HT2C receptor (5-HT2CR) drives interlocked phenotypes that precipitate relapse-like behaviors in cocaine dependence. Neuropsychopharmacology. 2014;39:370–382.
    1. Lappalainen J, Zhang L, Dean M, Oz M, Ozaki N, Yu DH, et al. Identification, expression, and pharmacology of a Cys23-Ser23 substitution in the human 5-HT2c receptor gene (HTR2C) Genomics. 1995;27:274–279.
    1. Piva F, Giulietti M, Baldelli L, Nardi B, Bellantuono C, Armeni T, et al. Bioinformatic analyses to select phenotype affecting polymorphisms in HTR2C gene. Hum Psychopharmacol. 2011;26:365–372.
    1. Okada M, Northup JK, Ozaki N, Russell JT, Linnoila M, Goldman D. Modification of human 5-HT(2C) receptor function by Cys23Ser, an abundant, naturally occurring amino-acid substitution. Mol Psychiatry. 2004;9:55–64.
    1. Walstab J, Steinhagen F, BRUSS M, Gothert M, Bonisch H. Differences between human wild-type and C23S variant 5-HT2C receptors in inverse agonist-induced resensitization. Pharmacol Rep. 2011;63:45–53.
    1. Brasch-Andersen C, Moller MU, Christiansen L, Thinggaard M, Otto M, Brosen K, et al. A candidate gene study of serotonergic pathway genes and pain relief during treatment with escitalopram in patients with neuropathic pain shows significant association to serotonin receptor2C (HTR2C) Eur J Clin Pharmacol. 2011;67:1131–1137.
    1. Kuhn KU, Joe AY, Meyer K, Reichmann K, Maier W, Rao ML, et al. Neuroimaging and 5-HT2C receptor polymorphism: a HMPAO-SPECT study in healthy male probands using mCPP-challenge of the 5-HT2C receptor. Pharmacopsychiatry. 2004;37:286–291.
    1. Quested DJ, Whale R, Sharpley AL, McGavin CL, Crossland N, Harrison PJ, et al. Allelic variation in the 5-HT2C receptor (HTR2C) and functional responses to the 5-HT2C receptor agonist, m-chlorophenylpiperazine. Psychopharmacology (Berl) 1999;144:306–307.
    1. Saltzman AG, Morse B, Whitman MM, Ivanshchenko Y, Jaye M, Felder S. Cloning of the human serotonin 5-HT2 and 5-HT1C receptor subtypes. Biochem Biophys Res Commun. 1991;181:1469–1478.
    1. Xie EZ, Zhu LY, Zhao LY, Chang LS. The human serotonin 5-HT2c receptor: Complete cDNA, genomic structure, and alternatively spliced variant. Genomics. 1996;35:551–561.
    1. First MB, Spitzer RL, Gibbon M, Williams JB.Structured Clinical Interview for DSM-IV Axis I Disorders-Patient Edition Biometrics Research Department, New York State Psychiatric Institute: New York, NY, USA, 1996.
    1. Hester R, Dixon V, Garavan H. A consistent attentional bias for drug-related material in active cocaine users across word and picture versions of the emotional Stroop task. Drug Alcohol Depend. 2006;81:251–257.
    1. Liu S, Lane SD, Schmitz JM, Green CE, Cunningham KA, Moeller FG. Increased intra-individual reaction time variability in cocaine-dependent subjects: role of cocaine-related cues. Addict Behav. 2012;37:193–197.
    1. Liu S, Lane SD, Schmitz JM, Waters AJ, Cunningham KA, Moeller FG. Relationship between attentional bias to cocaine-related stimuli and impulsivity in cocaine-dependent subjects. Am J Drug Alcohol Abuse. 2011;37:117–122.
    1. Field M, Munafo MR, Franken IH. A meta-analytic investigation of the relationship between attentional bias and subjective craving in substance abuse. Psychol Bull. 2009;135:589–607.
    1. Cox WM, Fadardi JS, Pothos EM. The addiction-stroop test: Theoretical considerations and procedural recommendations. Psychol Bull. 2006;132:443–476.
    1. Kosten TR, Wu G, Huang W, Harding MJ, Hamon SC, Lappalainen J, et al. Pharmacogenetic randomized trial for cocaine abuse: disulfiram and dopamine beta-hydroxylase. Biol Psychiatry. 2013;73:219–224.
    1. Kosten TR, Domingo CB, Hamon SC, Nielsen DA. DBH gene as predictor of response in a cocaine vaccine clinical trial. Neurosci Lett. 2013;541:29–33.
    1. Nic Dhonnchadha BA, Fox RG, Stutz SJ, Rice KC, Cunningham KA. Blockade of the serotonin 5-HT2A receptor suppresses cue-evoked reinstatement of cocaine-seeking behavior in a rat self-administration model. Behav Neurosci. 2009;123:382–396.
    1. Anastasio NC, Lanfranco MF, Bubar MJ, Seitz PK, Stutz SJ, McGinnis AG, et al. Serotonin 5-HT(2C) receptor protein expression is enriched in synaptosomal and post-synaptic compartments of rat cortex. J Neurochem. 2010;113:1504–1515.
    1. Liu S, Bubar MJ, Lanfranco MF, Hillman GR, Cunningham KA. Serotonin(2C) receptor localization in GABA neurons of the rat medial prefrontal cortex: Implications for understanding the neurobiology of addiction. Neuroscience. 2007;146:1667–1688.
    1. Waters AJ, Marhe R, Franken IH. Attentional bias to drug cues is elevated before and during temptations to use heroin and cocaine. Psychopharmacology (Berl) 2012;219:909–921.
    1. Moeller SJ, Parvaz MA, Shumay E, Beebe-Wang N, Konova AB, Alia-Klein N, et al. Gene xabstinence effects on drug cue reactivity in addiction: multimodal evidence.J Neurosci 20133310027–10036.
    1. Smelson D, Yu L, Buyske S, Gonzalez G, Tischfield J, Deutsch CK, et al. Genetic association of GABA-A receptor alpha-2 and mu opioid receptor with cocaine cue-reactivity: evidence for inhibitory synaptic neurotransmission involvement in cocaine dependence. Am J Addict. 2012;21:411–415.
    1. Meyer PJ, Lovic V, Saunders BT, Yager LM, Flagel SB, Morrow JD, et al. Quantifying individual variation in the propensity to attribute incentive salience to reward cues. PLoS One. 2012;7:e38987.
    1. Brummett BH, Kuhn CM, Boyle SH, Babyak MA, Siegler IC, Williams RB. Cortisol responses to emotional stress in men: association with a functional polymorphism in the 5HTR2C gene. Biol Psychol. 2012;89:94–98.
    1. Mickey BJ, Sanford BJ, Love TM, Shen PH, Hodgkinson CA, Stohler CS, et al. Striatal dopamine release and genetic variation of the serotonin 2C receptor in humans. J Neurosci. 2012;32:9344–9350.
    1. Fentress HM, Grinde E, Mazurkiewicz JE, Backstrom JR, Herrick-Davis K, Sanders-Bush E. Pharmacological properties of the Cys23Ser single nucleotide polymorphism in human 5-HT2C receptor isoforms. Pharmacogenomics J. 2005;5:244–254.
    1. Drago A, Serretti A. Focus on HTR2C: A possible suggestion for genetic studies of complex disorders. Am J Med Genet B Neuropsychiatr Genet. 2009;150B:601–637.
    1. Sanders-Bush E, Fentress H, Hazelwood L. Serotonin 5-HT2 receptors: Molecular and genomic diversity. Mol Interv. 2003;3:319–330.
    1. Handelsman L, Kahn RS, Sturiano C, Rinaldi PJ, Gabriel S, Schmeidler JP, et al. Hostility is associated with a heightened prolactin response to meta-chlorophenylpiperazine in abstinent cocaine addicts. Psychiatry Res. 1998;80:1–12.
    1. Patkar AA, Mannelli P, Peindl K, Hill KP, Gopalakrishnan R, Berrettini WH. Relationship of disinhibition and aggression to blunted prolactin response to meta-chlorophenylpiperazine in cocaine-dependent patients. Psychopharmacology (Berl) 2006;185:123–132.
    1. Filip M, Bubar MJ, Cunningham KA. Contribution of serotonin (5-hydroxytryptamine; 5-HT) 5-HT2 receptor subtypes to the hyperlocomotor effects of cocaine: Acute and chronic pharmacological analyses. J Pharmacol Exp Ther. 2004;310:1246–1254.
    1. Robinson TE, Flagel SB. Dissociating the predictive and incentive motivational properties of reward-related cues through the study of individual differences. Biol Psychiatry. 2009;65:869–873.
    1. Parsons LH, Koob GF, Weiss F. Extracellular serotonin is decreased in the nucleus accumbens during withdrawal from cocaine self-administration. Behav Brain Res. 1995;73 ((1-2:225–228.
    1. Rajagopal S, Ahn S, Rominger DH, Gowen-MacDonald W, Lam CM, Dewire SM, et al. Quantifying ligand bias at seven-transmembrane receptors. Mol Pharmacol. 2011;80:367–377.
    1. Abe K, Fujimoto T, Akaishi T, Misawa M. Stimulation of basolateral amygdaloid serotonin 5-HT(2C) receptors promotes the induction of long-term potentiation in the dentate gyrus of anesthetized rats. Neurosci Lett. 2009;451:65–68.
    1. Potenza MN, Hong KI, Lacadie CM, Fulbright RK, Tuit KL, Sinha R. Neural correlates of stress-induced and cue-induced drug craving: influences of sex and cocaine dependence. Am J Psychiatry. 2012;169:406–414.
    1. Munafo MR, Johnstone EC, Mackintosh B. Association of serotonin transporter genotype with selective processing of smoking-related stimuli in current smokers and ex-smokers. Nicotine Tob Res. 2005;7:773–778.

Source: PubMed

3
Předplatit