Skip to main content

Advertisement

Log in

Shifts in the neurobiological mechanisms motivating cocaine use with the development of an addiction-like phenotype in male rats

  • Original Investigation
  • Published:
Psychopharmacology Aims and scope Submit manuscript

Abstract

Rationale

The development of addiction is accompanied by a shift in the mechanisms motivating cocaine use from nucleus accumbens (NAc) dopamine D1 receptor (D1R) signaling to glutamate AMPA-kainate receptor (AMPA-R) signaling.

Objective

Here, we determined whether similar shifts occur for NAc-D2R signaling and following systemic manipulation of D1R, D2R, and AMPA-R signaling.

Methods

Male rats were given short-access (20 infusions/day) or extended-access to cocaine (24 h/day, 96 infusions/day, 10 days). Motivation for cocaine was assessed following 14 days of abstinence using a progressive-ratio schedule. Once responding stabilized, the effects of NAc-D2R antagonism (eticlopride; 0–10.0 μg/side) and systemic D1R (SCH-23390; 0–1.0 mg/kg), D2R (eticlopride; 0–0.1 mg/kg), and AMPA-R (CNQX; 0–1.5 mg/kg) antagonism, and NAc-dopamine-R gene expression (Drd1/2/3) were examined.

Results

Motivation for cocaine was markedly higher in the extended- versus short-access group confirming the development of an addiction-like phenotype in the extended-access group. NAc-infused eticlopride decreased motivation for cocaine in both the short- and extended-access groups although low doses (0.1–0.3 μg) were more effective in the short-access group and high doses (3–10 μg/side) tended to be more effective in the extended-access group. Systemic administration of eticlopride (0.1 mg/kg) was more effective in the extended-access group, and systemic administration of CNQX was effective in the extended- but not short-access group. NAc-Drd2 expression was decreased in both the short- and extended-access groups.

Conclusion

These findings indicate that in contrast to NAc-D1R, D2R remain critical for motivating cocaine use with the development of an addiction-like phenotype. These findings also indicate that shifts in the mechanisms motivating cocaine use impact the response to both site-specific and systemic pharmacological treatment.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

References

  • Abel JM, Nesil T, Bakhti-Suroosh A, Grant PA, Lynch WJ (2019) Mechanisms underlying the efficacy of exercise as an intervention for cocaine relapse: a focus on mGlu5 in the dorsal medial prefrontal cortex. Psychopharmacology 236:2155–2171

    CAS  PubMed  PubMed Central  Google Scholar 

  • Ahmed SH, Koob GF (2004) Changes in response to a dopamine receptor antagonist in rats with escalating cocaine intake. Psychopharmacology 172:450–454

    CAS  PubMed  Google Scholar 

  • Allain F, Samaha AN (2019) Revisiting long-access versus short-access cocaine self-administration in rats: intermittent intake promotes addiction symptoms independent of session length. Addict Biol 24:641–651

    CAS  PubMed  Google Scholar 

  • Amato L, Minozzi S, Pani PP, Davoli M (2007) Antipsychotic medications for cocaine dependence Cochrane Database Syst Rev: CD006306

  • American Psychiatric Association (2013) Diagnostic and statistical manual of mental disorders, 5th edn. American Psychiatric Publishing, Arlington VA

    Google Scholar 

  • Bachtell RK, Whisler K, Karanian D, Self DW (2005) Effects of intra-nucleus accumbens shell administration of dopamine agonists and antagonists on cocaine-taking and cocaine-seeking behaviors in the rat. Psychopharmacology 183:41–53

    CAS  PubMed  Google Scholar 

  • Backstrom P, Hyytia P (2003) Attenuation of cocaine-seeking behaviour by the AMPA/kainate receptor antagonist CNQX in rats. Psychopharmacology 166:69–76

    PubMed  Google Scholar 

  • Backstrom P, Hyytia P (2006) Ionotropic and metabotropic glutamate receptor antagonism attenuates cue-induced cocaine seeking. Neuropsychopharmacology 31:778–786

    PubMed  Google Scholar 

  • Baker DA, Fuchs RA, Specio SE, Khroyan TV, Neisewander JL (1998) Effects of intraaccumbens administration of SCH-23390 on cocaine-induced locomotion and conditioned place preference. Synapse 30:181–193

    CAS  PubMed  Google Scholar 

  • Bari AA, Pierce RC (2005) D1-like and D2 dopamine receptor antagonists administered into the shell subregion of the rat nucleus accumbens decrease cocaine, but not food, reinforcement. Neuroscience 135:959–968

    CAS  PubMed  Google Scholar 

  • Ben-Shahar O, Keeley P, Cook M, Brake W, Joyce M, Nyffeler M, Heston R, Ettenberg A (2007) Changes in levels of D1, D2, or NMDA receptors during withdrawal from brief or extended daily access to IV cocaine. Brain res 1131: 220-8.

  • Ben-Shahar O, Posthumus EJ, Waldroup SA, Ettenberg A (2008) Heightened drug-seeking motivation following extended daily access to self-administered cocaine. Prog Neuro-Psychopharmacol Biol Psychiatry 32:863–869

    CAS  Google Scholar 

  • Bibb JA, Snyder GL, Nishi A, Yan Z, Meijer L, Fienberg AA, Tsai LH, Kwon YT, Girault JA, Czernik AJ, Huganir RL, Hemmings HC Jr, Nairn AC, Greengard P (1999) Phosphorylation of DARPP-32 by Cdk5 modulates dopamine signalling in neurons. Nature 402:669–671

    CAS  PubMed  Google Scholar 

  • Binswanger IA, Stern MF, Deyo RA, Heagerty PJ, Cheadle A, Elmore JG, Koepsell TD (2007) Release from prison--a high risk of death for former inmates. N Engl J Med 356:157–165

    CAS  PubMed  PubMed Central  Google Scholar 

  • Binswanger IA, Nowels C, Corsi KF, Glanz J, Long J, Booth RE, Steiner JF (2012) Return to drug use and overdose after release from prison: a qualitative study of risk and protective factors. Addict Sci Clin Pract 7:3

    PubMed  PubMed Central  Google Scholar 

  • Bornebusch AB, Fink-Jensen A, Wortwein G, Seeley RJ, Thomsen M (2019) Glucagon-like peptide-1 receptor agonist treatment does not reduce abuse-related effects of opioid drugs. eNeuro 6

  • Boudreau AC, Wolf ME (2005) Behavioral sensitization to cocaine is associated with increased AMPA receptor surface expression in the nucleus accumbens. J Neurosci 25:9144–9151

    CAS  PubMed  PubMed Central  Google Scholar 

  • Briand LA, Flagel SB, Garcia-Fuster MJ, Watson SJ, Akil H, Sarter M, Robinson TE (2008) Persistent alterations in cognitive function and prefrontal dopamine D2 receptors following extended, but not limited, access to self-administered cocaine. Neuropsychopharmacology 33:2969–2980

    CAS  PubMed  PubMed Central  Google Scholar 

  • Conrad KL, Tseng KY, Uejima JL, Reimers JM, Heng LJ, Shaham Y, Marinelli M, Wolf ME (2008) Formation of accumbens GluR2-lacking AMPA receptors mediates incubation of cocaine craving. Nature 454:118–121

    CAS  PubMed  PubMed Central  Google Scholar 

  • Conrad KL, Ford K, Marinelli M, Wolf ME (2010) Dopamine receptor expression and distribution dynamically change in the rat nucleus accumbens after withdrawal from cocaine self-administration. Neuroscience 169:182–194

    CAS  PubMed  PubMed Central  Google Scholar 

  • de Lima MS, de Oliveira Soares BG, Reisser AA, Farrell M (2002) Pharmacological treatment of cocaine dependence: a systematic review. Addiction 97:931–949

    PubMed  Google Scholar 

  • Deroche-Gamonet V, Belin D, Piazza PV (2004) Evidence for addiction-like behavior in the rat. Science 305:1014–1017

    CAS  PubMed  Google Scholar 

  • Doyle SE, Ramôa C, Garber G, Newman J, Toor Z, Lynch WJ (2014) A shift in the role of glutamatergic signaling in the nucleus accumbens core with the development of an addicted phenotype. Biol Psychiatry 76:810–815

    CAS  PubMed  PubMed Central  Google Scholar 

  • Ducret E, Puaud M, Lacoste J, Belin-Rauscent A, Fouyssac M, Dugast E, Murray JE, Everitt BJ, Houeto JL, Belin D (2016) N-acetylcysteine facilitates self-imposed abstinence after escalation of cocaine intake. Biol Psychiatry 80:226–234

    CAS  PubMed  PubMed Central  Google Scholar 

  • Edwards S, Whisler KN, Fuller DC, Orsulak PJ, Self DW (2007) Addiction-related alterations in D1 and D2 dopamine receptor behavioral responses following chronic cocaine self-administration. Neuropsychopharmacology 32:354–366

    CAS  PubMed  Google Scholar 

  • Fischer KD, Houston AC, Rebec GV (2013) Role of the major glutamate transporter GLT1 in nucleus accumbens core versus shell in cue-induced cocaine-seeking behavior. J Neurosci 33:9319–9327

    CAS  PubMed  PubMed Central  Google Scholar 

  • Gentile TA, Simmons SJ, Barker DJ, Shaw JK, Espana RA, Muschamp JW (2018) Suvorexant, an orexin/hypocretin receptor antagonist, attenuates motivational and hedonic properties of cocaine. Addict Biol 23:247–255

    CAS  PubMed  Google Scholar 

  • Greenwell TN, Funk CK, Cottone P, Richardson HN, Chen SA, Rice KC, Zorrilla EP, Koob GF (2009) Corticotropin-releasing factor-1 receptor antagonists decrease heroin self-administration in long-but not short-access rats. Addict Biol 14:130–143

    CAS  PubMed  PubMed Central  Google Scholar 

  • Haile CN, Kosten TR (2013) Pharmacotherapy for stimulant-related disorders. Curr Psychiatry Rep 15:415

    PubMed  Google Scholar 

  • Hammad AM, Alasmari F, Althobaiti YS, Sari Y (2017) Modulatory effects of ampicillin/sulbactam on glial glutamate transporters and metabotropic glutamate receptor 1 as well as reinstatement to cocaine-seeking behavior. Behav Brain Res 332:288–298

    CAS  PubMed  PubMed Central  Google Scholar 

  • Hao Y, Martin-Fardon R, Weiss F (2010) Behavioral and functional evidence of metabotropic glutamate receptor 2/3 and metabotropic glutamate receptor 5 dysregulation in cocaine-escalated rats: factor in the transition to dependence. Biol Psychiatry 68:240–248

    CAS  PubMed  PubMed Central  Google Scholar 

  • Hemby SE, Tang W, Muly EC, Kuhar MJ, Howell L, Mash DC (2005) Cocaine-induced alterations in nucleus accumbens ionotropic glutamate receptor subunits in human and non-human primates. J Neurochem 95:1785–1793

    CAS  PubMed  Google Scholar 

  • James MH, Stopper CM, Zimmer BA, Koll NE, Bowrey HE, Aston-Jones G (2019) Increased number and activity of a lateral subpopulation of hypothalamic orexin/hypocretin neurons underlies the expression of an addicted state in rats. Biol Psychiatry 85:925–935

    CAS  PubMed  Google Scholar 

  • Kalivas PW, McFarland K (2003) Brain circuitry and the reinstatement of cocaine-seeking behavior. Psychopharmacology 168:44–56

    CAS  PubMed  Google Scholar 

  • Kalivas PW, Volkow ND (2005) The neural basis of addiction: a pathology of motivation and choice. Am J Psychiatry 162:1403–1413

    PubMed  Google Scholar 

  • Koob G, Kreek MJ (2007) Stress, dysregulation of drug reward pathways, and the transition to drug dependence. Am J Psychiatry 164:1149–1159

    PubMed  PubMed Central  Google Scholar 

  • Loweth JA, Tseng KY, Wolf ME (2014) Adaptations in AMPA receptor transmission in the nucleus accumbens contributing to incubation of cocaine craving. Neuropharmacology 76 Pt B: 287-300

  • Lynch WJ (2018) Modeling the development of drug addiction in male and female animals. Pharmacol Biochem Behav 164:50–61

    CAS  PubMed  Google Scholar 

  • Lynch WJ, Carroll ME (2001) Regulation of drug intake. Exp Clin Psychopharmacol 9:131–143

    CAS  PubMed  Google Scholar 

  • Martinez D, Greene K, Broft A, Kumar D, Liu F, Narendran R, Slifstein M, Van Heertum R, Kleber HD (2009a) Lower level of endogenous dopamine in patients with cocaine dependence: findings from PET imaging of D(2)/D(3) receptors following acute dopamine depletion. Am J Psychiatry 166:1170–1177

    PubMed  PubMed Central  Google Scholar 

  • Martinez D, Slifstein M, Narendran R, Foltin RW, Broft A, Hwang DR, Perez A, Abi-Dargham A, Fischman MW, Kleber HD, Laruelle M (2009b) Dopamine D1 receptors in cocaine dependence measured with PET and the choice to self-administer cocaine. Neuropsychopharmacology 34:1774–1782

    CAS  PubMed  PubMed Central  Google Scholar 

  • Mateo Y, Lack CM, Morgan D, Roberts DC, Jones SR (2005) Reduced dopamine terminal function and insensitivity to cocaine following cocaine binge self-administration and deprivation. Neuropsychopharmacology 30:1455–1463

    CAS  PubMed  Google Scholar 

  • McGregor A, Roberts DC (1993) Dopaminergic antagonism within the nucleus accumbens or the amygdala produces differential effects on intravenous cocaine self-administration under fixed and progressive ratio schedules of reinforcement. Brain Res 624:245–252

    CAS  PubMed  Google Scholar 

  • Meador-Woodruff JH, Little KY, Damask SP, Mansour A, Watson SJ (1993) Effects of cocaine on dopamine receptor gene expression: a study in the postmortem human brain. Biol Psychiatry 34:348–355

    CAS  PubMed  Google Scholar 

  • Morgan D, Roberts DC (2004) Sensitization to the reinforcing effects of cocaine following binge-abstinent self-administration. Neurosci Biobehav Rev 27:803–812

    CAS  PubMed  Google Scholar 

  • Morgan D, Brebner K, Lynch WJ, Roberts DC (2002) Increases in the reinforcing efficacy of cocaine after particular histories of reinforcement. Behav Pharmacol 13:389–396

    CAS  PubMed  Google Scholar 

  • Morgan D, Smith MA, Roberts DC (2005) Binge self-administration and deprivation produces sensitization to the reinforcing effects of cocaine in rats. Psychopharmacology 178:309–316

    CAS  PubMed  Google Scholar 

  • Murschall A, Hauber W (2005) Effects of a systemic AMPA/KA and NMDA receptor blockade on pavlovian-instrumental transfer. Psychopharmacology 182:290–296

    CAS  PubMed  Google Scholar 

  • Negus SS, Banks ML (2018) Modulation of drug choice by extended drug access and withdrawal in rhesus monkeys: implications for negative reinforcement as a driver of addiction and target for medications development. Pharmacol Biochem Behav 164:32–39

    CAS  PubMed  Google Scholar 

  • Neisewander JL, Cheung TH, Pentkowski NS (2014) Dopamine D3 and 5-HT1B receptor dysregulation as a result of psychostimulant intake and forced abstinence: implications for medications development. Neuropharmacology 76 Pt B: 301-19

  • Nicolas C, Tauber C, Lepelletier FX, Chalon S, Belujon P, Galineau L, Solinas M (2017) Longitudinal changes in brain metabolic activity after withdrawal from escalation of cocaine Self-administration. Neuropsychopharmacology 42:1981–1990

    CAS  PubMed  PubMed Central  Google Scholar 

  • O’Brien C (2005) Drug addiction and drug abuse. In: Brunton LL, Lazo JS, Parker K (eds). Goodman and Gilman’s: The pharmacological basis of therapeutics. 2005. 11th edition. McGraw-Hill: New York, USA. pp. 607–627

  • Oleson EB, Roberts DC (2008) Parsing the addiction phenomenon: self-administration procedures modeling enhanced motivation for drug and escalation of drug intake. Drug Discov Today Dis Models 5:217–226

    PubMed  PubMed Central  Google Scholar 

  • Oleson EB, Roberts DC (2009) Behavioral economic assessment of price and cocaine consumption following self-administration histories that produce escalation of either final ratios or intake. Neuropsychopharmacology 34:796–804

    PubMed  Google Scholar 

  • Palmatier MI, Kellicut MR, Brianna Sheppard A, Brown RW, Robinson DL (2014) The incentive amplifying effects of nicotine are reduced by selective and non-selective dopamine antagonists in rats. Pharmacol Biochem Behav 126:50–62

    CAS  PubMed  PubMed Central  Google Scholar 

  • Parvaz MA, Moeller SJ, Goldstein RZ (2016) Incubation of cue-induced craving in adults addicted to cocaine measured by electroencephalography. JAMA Psychiatry 73:1127–1134

    PubMed  PubMed Central  Google Scholar 

  • Pelloux Y, Degoulet M, Tiran-Cappello A, Cohen C, Lardeux S, George O, Koob GF, Ahmed SH, Baunez C (2018) Subthalamic nucleus high frequency stimulation prevents and reverses escalated cocaine use. Mol Psychiatry 23:2266–2276

    CAS  PubMed  Google Scholar 

  • Piazza PV, Deroche-Gamonent V, Rouge-Pont F, Le Moal M (2000) Vertical shifts in self-administration dose-response functions predict a drug-vulnerable phenotype predisposed to addiction. J Neurosci 20:4226–4232

    CAS  PubMed  PubMed Central  Google Scholar 

  • Pickens CL, Airavaara M, Theberge F, Fanous S, Hope BT, Shaham Y (2011) Neurobiology of the incubation of drug craving. Trends Neurosci 34:411–420

    CAS  PubMed  PubMed Central  Google Scholar 

  • Pierce RC, Kumaresan V (2006) The mesolimbic dopamine system: the final common pathway for the reinforcing effect of drugs of abuse? Neurosci Biobehav Rev 30:215–238

    CAS  PubMed  Google Scholar 

  • Ramôa CP, Doyle SE, Lycas MD, Chernau AK, Lynch WJ (2014) Diminished role of dopamine D1-receptor signaling with the development of an addicted phenotype in rats. Biol Psychiatry 76:8–14

    PubMed  Google Scholar 

  • Roberts DC, Morgan D, Liu Y (2007) How to make a rat addicted to cocaine. Prog Neuro-Psychopharmacol Biol Psychiatry 31:1614–1624

    CAS  Google Scholar 

  • Schenk S, Gittings D (2003) Effects of SCH 23390 and eticlopride on cocaine-seeking produced by cocaine and WIN 35,428 in rats. Psychopharmacology 168:118–123

    CAS  PubMed  Google Scholar 

  • Schmeichel BE, Herman MA, Roberto M, Koob GF (2017) Hypocretin neurotransmission within the central amygdala mediates escalated cocaine self-administration and stress-induced reinstatement in rats. Biol Psychiatry 81:606–615

    CAS  PubMed  Google Scholar 

  • Seth P, Scholl L, Rudd RA, Bacon S (2018) Overdose deaths involving opioids, cocaine, and psychostimulants-United States, 2015-2016. MMWR Morb Mortal Wkly Rep 67:349–358

    PubMed  PubMed Central  Google Scholar 

  • Seymour A, Black M, Simpson K, Oliver JS (2000) Drug related deaths amongst Glasgow city hostel dwellers. J Clin Forensic Med 7:183–187

    CAS  PubMed  Google Scholar 

  • Smith MA, Fronk GE, Abel JM, Lacy RT, Bills SE, Lynch WJ (2018) Resistance exercise decreases heroin self-administration and alters gene expression in the nucleus accumbens of heroin-exposed rats. Psychopharmacology 235:1245–1255

    CAS  PubMed  PubMed Central  Google Scholar 

  • Substance Abuse and Mental Health Services Administration (2019) Key substance use and mental health indicators in the United States: Results from the 2018 National Survey on Drug Use and Health (HHS Publication No. PEP19–5068, NSDUH Series H-54). Rockville, MD: Center for Behavioral Health Statistics and Quality, Substance Abuse and Mental Health Services Administration

  • Tang WX, Fasulo WH, Mash DC, Hemby SE (2003) Molecular profiling of midbrain dopamine regions in cocaine overdose victims. J Neurochem 85:911–924

    CAS  PubMed  PubMed Central  Google Scholar 

  • Valenza M, Butelman ER, Kreek MJ (2017) Effects of the novel relatively short-acting kappa opioid receptor antagonist LY2444296 in behaviors observed after chronic extended-access cocaine self-administration in rats. Psychopharmacology 234:2219–2231

    CAS  PubMed  PubMed Central  Google Scholar 

  • Venniro M, Caprioli D, Shaham Y (2016) Animal models of drug relapse and craving: from drug priming-induced reinstatement to incubation of craving after voluntary abstinence. Prog Brain Res 224:25–52

    PubMed  Google Scholar 

  • Volkow ND, Fowler JS, Wolf AP, Schlyer D, Shiue CY, Alpert R, Dewey SL, Logan J, Bendriem B, Christman D et al (1990) Effects of chronic cocaine abuse on postsynaptic dopamine receptors. Am J Psychiatry 147:719–724

    CAS  PubMed  Google Scholar 

  • Volkow ND, Fowler JS, Wang GJ, Hitzemann R, Logan J, Schlyer DJ, Dewey SL, Wolf AP (1993) Decreased dopamine D2 receptor availability is associated with reduced frontal metabolism in cocaine abusers. Synapse 14:169–177

    CAS  PubMed  Google Scholar 

  • Volkow ND, Wang GJ, Fowler JS, Logan J, Gatley SJ, Hitzemann R, Chen AD, Dewey SL, Pappas N (1997) Decreased striatal dopaminergic responsiveness in detoxified cocaine-dependent subjects. Nature 386:830–833

    CAS  PubMed  Google Scholar 

  • Vranjkovic O, Van Newenhizen EC, Nordness ME, Blacktop JM, Urbanik LA, Mathy JC, McReynolds JR, Miller AM, Doncheck EM, Kloehn TM, Stinnett GS, Gerndt CH, Ketchesin KD, Baker DA, Seasholtz AF, Mantsch JR (2018) Enhanced CRFR1-dependent regulation of a ventral tegmental area to prelimbic cortex projection establishes susceptibility to stress-induced cocaine seeking. J Neurosci 38:10657–10671

    CAS  PubMed  PubMed Central  Google Scholar 

  • Willuhn I, Wanat MJ, Clark JJ, Phillips PE (2010) Dopamine signaling in the nucleus accumbens of animals self-administering drugs of abuse. Curr Top Behav Neurosci 3:29–71

    PubMed  PubMed Central  Google Scholar 

  • Wolf ME, Tseng KY (2012) Calcium-permeable AMPA receptors in the VTA and nucleus accumbens after cocaine exposure: when, how, and why? Front Mol Neurosci 5:72

    CAS  PubMed  PubMed Central  Google Scholar 

  • Zhang Y, Svenningsson P, Picetti R, Schlussman SD, Nairn AC, Ho A, Greengard P, Kreek MJ (2006) Cocaine self-administration in mice is inversely related to phosphorylation at Thr34 (protein kinase a site) and Ser130 (kinase CK1 site) of DARPP-32. J Neurosci 26:2645–2651

    CAS  PubMed  PubMed Central  Google Scholar 

  • Zimmer BA, Oleson EB, Roberts DC (2012) The motivation to self-administer is increased after a history of spiking brain levels of cocaine. Neuropsychopharmacology 37:1901–1910

    CAS  PubMed  PubMed Central  Google Scholar 

  • Zimmer BA, Dobrin CV, Roberts DC (2013) Examination of behavioral strategies regulating cocaine intake in rats. Psychopharmacology 225:935–944

    CAS  PubMed  Google Scholar 

  • Zorrilla EP, Wee S, Zhao Y, Specio S, Boutrel B, Koob GF, Weiss F (2012) Extended access cocaine self-administration differentially activates dorsal raphe and amygdala corticotropin-releasing factor systems in rats. Addict Biol 17:300–308

    CAS  PubMed  Google Scholar 

Download references

Acknowledgments

We would like to acknowledge Lasya Pidaparthi for technical assistance.

Funding

This study was supported by the National Institute on Drug Abuse (2R01DA024716; WJL).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Wendy J. Lynch.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lynch, W.J., Bakhti-Suroosh, A., Abel, J.M. et al. Shifts in the neurobiological mechanisms motivating cocaine use with the development of an addiction-like phenotype in male rats. Psychopharmacology 238, 811–823 (2021). https://doi.org/10.1007/s00213-020-05732-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00213-020-05732-4

Keywords

Navigation