Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Original Article
  • Published:

The absence of VGLUT3 predisposes to cocaine abuse by increasing dopamine and glutamate signaling in the nucleus accumbens

Abstract

Tonically active cholinergic interneurons (TANs) from the nucleus accumbens (NAc) are centrally involved in reward behavior. TANs express a vesicular glutamate transporter referred to as VGLUT3 and thus use both acetylcholine and glutamate as neurotransmitters. The respective roles of each transmitter in the regulation of reward and addiction are still unknown. In this study, we showed that disruption of the gene that encodes VGLUT3 (Slc17a8) markedly increased cocaine self-administration in mice. Concomitantly, the amount of dopamine (DA) release was strongly augmented in the NAc of VGLUT3−/− mice because of a lack of signaling by metabotropic glutamate receptors. Furthermore, dendritic spines and glutamatergic synaptic transmission on medium spiny neurons were increased in the NAc of VGLUT3−/− mice. Increased DA and glutamate signaling in the NAc are hallmarks of addiction. Our study shows that TANs use glutamate to reduce DA release and decrease reinforcing properties of cocaine in mice. Interestingly, we also observed an increased frequency of rare variations in SLC17A8 in a cohort of severe drug abusers compared with controls. Our findings identify VGLUT3 as an unexpected regulator of drug abuse.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1
Figure 2
Figure 3
Figure 4
Figure 5
Figure 6

Similar content being viewed by others

References

  1. Di Chiara G, Imperato A . Preferential stimulation of dopamine release in the nucleus accumbens by opiates, alcohol, and barbiturates: studies with transcerebral dialysis in freely moving rats. Ann N Y Acad Sci 1986; 473: 367–381.

    Article  CAS  Google Scholar 

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

    Article  Google Scholar 

  3. Luscher C, Ungless MA . The mechanistic classification of addictive drugs. PLoS Med 2006; 3: e437.

    Article  Google Scholar 

  4. Gras C, Amilhon B, Lepicard EM, Poirel O, Vinatier J, Herbin M et al. The vesicular glutamate transporter VGLUT3 synergizes striatal acetylcholine tone. Nat Neurosci 2008; 11: 292–300.

    Article  CAS  Google Scholar 

  5. Higley MJ, Gittis AH, Oldenburg IA, Balthasar N, Seal RP, Edwards RH et al. Cholinergic interneurons mediate fast VGluT3-dependent glutamatergic transmission in the striatum. PLoS One 2011; 6: e19155.

    Article  CAS  Google Scholar 

  6. Guzman MS, De Jaeger X, Raulic S, Souza IA, Li AX, Schmid S et al. Elimination of the vesicular acetylcholine transporter in the striatum reveals regulation of behaviour by cholinergic-glutamatergic co-transmission. PLoS Biol 2011; 9: e1001194.

    Article  CAS  Google Scholar 

  7. Crettol S, Deglon JJ, Besson J, Croquette-Krokar M, Hammig R, Gothuey I et al. ABCB1 and cytochrome P450 genotypes and phenotypes: influence on methadone plasma levels and response to treatment. Clin Pharmacol Ther 2006; 80: 668–681.

    Article  CAS  Google Scholar 

  8. Besnard A, Bouveyron N, Kappes V, Pascoli V, Pages C, Heck N et al. Alterations of molecular and behavioral responses to cocaine by selective inhibition of Elk-1 phosphorylation. J Neurosci 2011; 31: 14296–14307.

    Article  CAS  Google Scholar 

  9. Martin-Garcia E, Barbano MF, Galeote L, Maldonado R . New operant model of nicotine-seeking behaviour in mice. Int J Neuropsychopharmacol 2009; 12: 343–356.

    Article  CAS  Google Scholar 

  10. Soria G, Barbano MF, Maldonado R, Valverde O . A reliable method to study cue-, priming-, and stress-induced reinstatement of cocaine self-administration in mice. Psychopharmacology 2008; 199: 593–603.

    Article  CAS  Google Scholar 

  11. Soria G, Mendizabal V, Tourino C, Robledo P, Ledent C, Parmentier M et al. Lack of CB1 cannabinoid receptor impairs cocaine self-administration. Neuropsychopharmacology 2005; 30: 1670–1680.

    Article  CAS  Google Scholar 

  12. Richardson NR, Gratton A . Changes in nucleus accumbens dopamine transmission associated with fixed- and variable-time schedule-induced feeding. Eur J Neurosci 2008; 27: 2714–2723.

    Article  Google Scholar 

  13. Tolu S, Eddine R, Marti F, David V, Graupner M, Pons S et al. Co-activation of VTA DA and GABA neurons mediates nicotine reinforcement. Mol Psychiatry 2013; 18: 382–393.

    Article  CAS  Google Scholar 

  14. Grace AA, Bunney BS . Intracellular and extracellular electrophysiology of nigral dopaminergic neurons—1. Identification and characterization. Neuroscience 1983; 10: 301–315.

    Article  CAS  Google Scholar 

  15. Grace AA, Bunney BS . The control of firing pattern in nigral dopamine neurons: burst firing. J Neurosci 1984; 4: 2877–2890.

    Article  CAS  Google Scholar 

  16. Mameli M, Bellone C, Brown MT, Luscher C . Cocaine inverts rules for synaptic plasticity of glutamate transmission in the ventral tegmental area. Nat Neurosci 2011; 14: 414–416.

    Article  CAS  Google Scholar 

  17. Mameli M, Halbout B, Creton C, Engblom D, Parkitna JR, Spanagel R et al. Cocaine-evoked synaptic plasticity: persistence in the VTA triggers adaptations in the NAc. Nat Neurosci 2009; 12: 1036–1041.

    Article  CAS  Google Scholar 

  18. Amilhon B, Lepicard E, Renoir T, Mongeau R, Popa D, Poirel O et al. VGLUT3 (vesicular glutamate transporter type 3) contribution to the regulation of serotonergic transmission and anxiety. J Neurosci 2010; 30: 2198–2210.

    Article  CAS  Google Scholar 

  19. Bolte S, Cordelieres FP . A guided tour into subcellular colocalization analysis in light microscopy. J Microsc 2006; 224: 213–232.

    Article  CAS  Google Scholar 

  20. Jaskolski F, Mulle C, Manzoni OJ . An automated method to quantify and visualize colocalized fluorescent signals. J Neurosci Methods 2005; 146: 42–49.

    Article  CAS  Google Scholar 

  21. Heck N, Betuing S, Vanhoutte P, Caboche J . A deconvolution method to improve automated 3D-analysis of dendritic spines: application to a mouse model of Huntington's disease. Brain Structure Function 2012; 217: 421–434.

    Article  Google Scholar 

  22. Rodriguez A, Ehlenberger DB, Dickstein DL, Hof PR, Wearne SL . Automated three-dimensional detection and shape classification of dendritic spines from fluorescence microscopy images. PLoS One 2008; 3: e1997.

    Article  Google Scholar 

  23. Sanchis-Segura C, Spanagel R . Behavioural assessment of drug reinforcement and addictive features in rodents: an overview. Addict Biol 2006; 11: 2–38.

    Article  Google Scholar 

  24. Carboni E, Vacca C . Conditioned place preference. A simple method for investigating reinforcing properties in laboratory animals. Methods Mol Med 2003; 79: 481–498.

    CAS  PubMed  Google Scholar 

  25. Schmitz Y, Benoit-Marand M, Gonon F, Sulzer D . Presynaptic regulation of dopaminergic neurotransmission. J Neurochem 2003; 87: 273–289.

    Article  CAS  Google Scholar 

  26. Karasawa J, Yoshimizu T, Chaki S . A metabotropic glutamate 2/3 receptor antagonist, MGS0039, increases extracellular dopamine levels in the nucleus accumbens shell. Neurosci Lett 2006; 393: 127–130.

    Article  CAS  Google Scholar 

  27. Fitzjohn SM, Bortolotto ZA, Palmer MJ, Doherty AJ, Ornstein PL, Schoepp DD et al. The potent mGlu receptor antagonist LY341495 identifies roles for both cloned and novel mGlu receptors in hippocampal synaptic plasticity. Neuropharmacology 1998; 37: 1445–1458.

    Article  CAS  Google Scholar 

  28. Bertran-Gonzalez J, Bosch C, Maroteaux M, Matamales M, Herve D, Valjent E et al. Opposing patterns of signaling activation in dopamine D1 and D2 receptor-expressing striatal neurons in response to cocaine and haloperidol. J Neurosci 2008; 28: 5671–5685.

    Article  CAS  Google Scholar 

  29. Robinson TE, Kolb B . Alterations in the morphology of dendrites and dendritic spines in the nucleus accumbens and prefrontal cortex following repeated treatment with amphetamine or cocaine. Eur J Neurosci 1999; 11: 1598–1604.

    Article  CAS  Google Scholar 

  30. Russo SJ, Dietz DM, Dumitriu D, Morrison JH, Malenka RC, Nestler EJ . The addicted synapse: mechanisms of synaptic and structural plasticity in nucleus accumbens. Trends Neurosci 2010; 33: 267–276.

    Article  CAS  Google Scholar 

  31. Heck N, Dos Santos M, Amairi B, Salery M, Besnard A, Herzog E et al. A new automated 3D detection of synaptic contacts reveals the formation of cortico-striatal synapses upon cocaine treatment in vivo. Brain Structure Function 2014.

  32. Sesack SR, Grace AA . Cortico-basal ganglia reward network: microcircuitry. Neuropsychopharmacology 2010; 35: 27–47.

    Article  Google Scholar 

  33. Kourrich S, Rothwell PE, Klug JR, Thomas MJ . Cocaine experience controls bidirectional synaptic plasticity in the nucleus accumbens. J Neurosci 2007; 27: 7921–7928.

    Article  CAS  Google Scholar 

  34. Pascoli V, Turiault M, Luscher C . Reversal of cocaine-evoked synaptic potentiation resets drug-induced adaptive behaviour. Nature 2012; 481: 71–75.

    Article  CAS  Google Scholar 

  35. Genomes Project C, Abecasis GR, Auton A, Brooks LD, DePristo MA, Durbin RM et al. An integrated map of genetic variation from 1,092 human genomes. Nature 2012; 491: 56–65.

    Article  Google Scholar 

  36. Brown MT, Tan KR, O'Connor EC, Nikonenko I, Muller D, Luscher C . Ventral tegmental area GABA projections pause accumbal cholinergic interneurons to enhance associative learning. Nature 2012; 492: 452–456.

    Article  CAS  Google Scholar 

  37. Tepper JM, Bolam JP . Functional diversity and specificity of neostriatal interneurons. Curr Opin Neurobiol 2004; 14: 685–692.

    Article  CAS  Google Scholar 

  38. Hikida T, Kaneko S, Isobe T, Kitabatake Y, Watanabe D, Pastan I et al. Increased sensitivity to cocaine by cholinergic cell ablation in nucleus accumbens. Proc Natl Acad Sci USA 2001; 98: 13351–13354.

    Article  CAS  Google Scholar 

  39. Kitabatake Y, Hikida T, Watanabe D, Pastan I, Nakanishi S . Impairment of reward-related learning by cholinergic cell ablation in the striatum. Proc Natl Acad Sci USA 2003; 100: 7965–7970.

    CAS  Google Scholar 

  40. Surmeier DJ, Graybiel AM . A feud that wasn't: acetylcholine evokes dopamine release in the striatum. Neuron 2012; 75: 1–3.

    Article  CAS  Google Scholar 

  41. Exley R, Cragg SJ . Presynaptic nicotinic receptors: a dynamic and diverse cholinergic filter of striatal dopamine neurotransmission. Br J Pharmacol 2008; 153: S283–S297.

    Article  CAS  Google Scholar 

  42. Nelson AB, Bussert TG, Kreitzer AC, Seal RP . Striatal cholinergic neurotransmission requires VGLUT3. J Neurosci 2014; 34: 8772–8777.

    Article  CAS  Google Scholar 

  43. 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.

    Article  CAS  Google Scholar 

  44. Hikida T, Yawata S, Yamaguchi T, Danjo T, Sasaoka T, Wang Y et al. Pathway-specific modulation of nucleus accumbens in reward and aversive behavior via selective transmitter receptors. Proc Natl Acad Sci USA 2013; 110: 342–347.

    Article  CAS  Google Scholar 

  45. Lobo MK, Nestler EJ . The striatal balancing act in drug addiction: distinct roles of direct and indirect pathway medium spiny neurons. Front Neuroanat 2011; 5: 41.

    Article  Google Scholar 

  46. Kravitz AV, Tye LD, Kreitzer AC . Distinct roles for direct and indirect pathway striatal neurons in reinforcement. Nat Neurosci 2012; 15: 816–818.

    Article  CAS  Google Scholar 

  47. Berridge KC . The debate over dopamine's role in reward: the case for incentive salience. Psychopharmacology 2007; 191: 391–431.

    Article  CAS  Google Scholar 

  48. Berridge KC, Robinson TE, Aldridge JW . Dissecting components of reward: 'liking', 'wanting', and learning. Curr Opin Pharmacol 2009; 9: 65–73.

    Article  CAS  Google Scholar 

  49. Kalivas PW, Volkow N, Seamans J . Unmanageable motivation in addiction: a pathology in prefrontal-accumbens glutamate transmission. Neuron 2005; 45: 647–650.

    Article  CAS  Google Scholar 

  50. Wagner FA, Anthony JC . From first drug use to drug dependence; developmental periods of risk for dependence upon marijuana, cocaine, and alcohol. Neuropsychopharmacology 2002; 26: 479–488.

    Article  Google Scholar 

Download references

Acknowledgements

This research was supported by funds from ANR (ANR-09-MNPS-033, ANR-13-SAMA-0005-01), Équipe FRM DEQ20130326486, FRC, Brain Canada Multi-Investigator Research Initiative, Djavad Mowafaghian Foundation, ERANET-Neuron Joint Transnational Call for ‘European Research Projects on Mental Disorders’, INSERM, CNRS and UPMC. The research teams of s.e.m., SJ, MM, JC, PV, BG and FB are members of the Bio-Psy Laboratory of Excellence; this work was therefore supported by French state funds managed by the ANR within the Investissements d'Avenir program under reference ANR-11-IDEX-0004-02. DYS was funded by the École des Neurosciences de Paris. This work was also supported by the City of Paris and Inserm Atip-Avenir to MM. FV was supported by grants from the Mission Interministérielle de Lutte contre la Drogue et la Toxicomanie (MILDT, 2006); the Département de la Recherche Clinique et du Développement-Assistance Publique Hôpitaux de Paris (DRCD-APHP, OST07013); and from the Programme Hospitalier de Recherches Cliniques (PHRC program, AOM10165). The Cellular Imaging and Flow Cytometry Facility is supported by the Conseil Régional Ile-de-France. We thank Emily Nichols for recruiting patients and data analysis. We also thank Dr Ryad Tamouza for providing DNA sample controls from Africa.

Author contributions

DYS, LJM, EM-G and JG-C performed the behavioral experiments with the help of SD. FM performed the in vivo electrophysiology and voltammetry with the help of DYS, LM and AG. DYS and LJM performed the anatomical experiments with the help of SB and EH. DYS, LJM and AB performed the biochemical measurements with the help of NH, PV and JC. MM and SL performed the in vitro electrophysiology experiments. The genetic studies were designed by SJ and performed by SJ with the help of FV, AH and FB. CBE, SC, FV and FB recruited subjects with severe addiction and unaffected controls. VFP and MAMP generated the VAChT mutant mice and helped with the manuscript. SEM designed the study and wrote the manuscript with the help of DYS, FM, RM, BG, MM and SJ.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to S Jamain or S El Mestikawy.

Ethics declarations

Competing interests

The authors declare no conflict of interest.

Additional information

Supplementary Information accompanies the paper on the Molecular Psychiatry website

Supplementary information

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sakae, D., Marti, F., Lecca, S. et al. The absence of VGLUT3 predisposes to cocaine abuse by increasing dopamine and glutamate signaling in the nucleus accumbens. Mol Psychiatry 20, 1448–1459 (2015). https://doi.org/10.1038/mp.2015.104

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/mp.2015.104

This article is cited by

Search

Quick links