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.

The dopamine motive system: implications for drug and food addiction

Key Points

  • The motivation to eat, like the motivation to take addictive drugs, activates the forebrain dopamine systems.

  • Excessive activation of this system strengthens the specific habits that precede the activation, sensitizing the animal's responsiveness to the specific conditions that elicit those habits.

  • At the same time, overactivation of the dopamine system downregulates the dopamine receptors, leaving the subject less interested in other activities.

  • The repeated intake of high-impact foods or addictive drugs thus makes food consumption or drug taking more habitual and decreases the importance of stimuli calling for alternatives.

  • Repeated drug use erodes the function of brain networks necessary for self-regulation, thereby facilitating impulsive, inflexible and compulsive actions.

Abstract

Behaviours such as eating, copulating, defending oneself or taking addictive drugs begin with a motivation to initiate the behaviour. Both this motivational drive and the behaviours that follow are influenced by past and present experience with the reinforcing stimuli (such as drugs or energy-rich foods) that increase the likelihood and/or strength of the behavioural response (such as drug taking or overeating). At a cellular and circuit level, motivational drive is dependent on the concentration of extrasynaptic dopamine present in specific brain areas such as the striatum. Cues that predict a reinforcing stimulus also modulate extrasynaptic dopamine concentrations, energizing motivation. Repeated administration of the reinforcer (drugs, energy-rich foods) generates conditioned associations between the reinforcer and the predicting cues, which is accompanied by downregulated dopaminergic response to other incentives and downregulated capacity for top-down self-regulation, facilitating the emergence of impulsive and compulsive responses to food or drug cues. Thus, dopamine contributes to addiction and obesity through its differentiated roles in reinforcement, motivation and self-regulation, referred to here as the 'dopamine motive system', which, if compromised, can result in increased, habitual and inflexible responding. Thus, interventions to rebalance the dopamine motive system might have therapeutic potential for obesity and addiction.

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: Dopamine motive system.
Figure 2: Brain images of DA D2 receptor availability in individuals suffering from either cocaine use disorder or morbid obesity along with the images of matched controls.

References

  1. Schultz, W. Predictive reward signal of dopamine neurons. J. Neurophysiol. 80, 1–27 (1998).

    Article  CAS  PubMed  Google Scholar 

  2. Stuber, G. D., Wightman, R. M. & Carelli, R. M. Extinction of cocaine self-administration reveals functionally and temporally distinct dopaminergic signals in the nucleus accumbens. Neuron 46, 661–669 (2005).

    Article  CAS  PubMed  Google Scholar 

  3. Volkow, N. D. & Baler, R. D. NOW versus LATER brain circuits: implications for obesity and addiction. Trends Neurosci. 38, 345–352 (2015).

    Article  CAS  PubMed  Google Scholar 

  4. Tomasi, D. et al. Overlapping patterns of brain activation to food and cocaine cues in cocaine abusers: association to striatal D2/D3 receptors. Hum. Brain Mapp. 36, 120–136 (2015).

    Article  PubMed  Google Scholar 

  5. Volkow, N. D., Wang, G. J., Fowler, J. S. & Telang, F. Overlapping neuronal circuits in addiction and obesity: evidence of systems pathology. Philos. Trans. R. Soc. Lond. B Biol. Sci. 363, 3191–3200 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Appelhans, B. M. et al. Inhibiting food reward: delay discounting, food reward sensitivity, and palatable food intake in overweight and obese women. Obesity 19, 2175–2182 (2011).

    Article  PubMed  Google Scholar 

  7. Wise, R. A. Dopamine, learning and motivation. Nat. Rev. Neurosci. 5, 483–494 (2004).

    Article  CAS  PubMed  Google Scholar 

  8. Tang, Y. Y., Posner, M. I., Rothbart, M. K. & Volkow, N. D. Circuitry of self-control and its role in reducing addiction. Trends Cogn. Sci. 19, 439–444 (2015).

    Article  PubMed  Google Scholar 

  9. Corwin, R. L., Avena, N. M. & Boggiano, M. M. Feeding and reward: perspectives from three rat models of binge eating. Physiol. Behav. 104, 87–97 (2011).

    Article  CAS  PubMed  Google Scholar 

  10. Parsons, L. H. & Hurd, Y. L. Endocannabinoid signalling in reward and addiction. Nat. Rev. Neurosci. 16, 579–594 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Peciña, S., Cagniard, B., Berridge, K. C., Aldridge, J. W. & Zhuang, X. Hyperdopaminergic mutant mice have higher “wanting” but not “liking” for sweet rewards. J. Neurosci. 23, 9395–9402 (2003).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Sotak, B. N., Hnasko, T. S., Robinson, S., Kremer, E. J. & Palmiter, R. D. Dysregulation of dopamine signaling in the dorsal striatum inhibits feeding. Brain Res. 1061, 88–96 (2005).

    Article  CAS  PubMed  Google Scholar 

  13. Palmiter, R. D. Dopamine signaling in the dorsal striatum is essential for motivated behaviors: lessons from dopamine-deficient mice. Ann. NY Acad. Sci. 1129, 35–46 (2008).

    Article  CAS  PubMed  Google Scholar 

  14. Hnasko, T. S., Sotak, B. N. & Palmiter, R. D. Cocaine-conditioned place preference by dopamine-deficient mice is mediated by serotonin. J. Neurosci. 27, 12484–12488 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Yager, L. M., Garcia, A. F., Wunsch, A. M. & Ferguson, S. M. The ins and outs of the striatum: role in drug addiction. Neuroscience 301, 529–541 (2015).

    Article  CAS  PubMed  Google Scholar 

  16. Wise, R. A. Roles for nigrostriatal — not just mesocorticolimbic — dopamine in reward and addiction. Trends Neurosci. 32, 517–524 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Howard, C. D., Li, H., Geddes, C. E. & Jin, X. Dynamic nigrostriatal dopamine biases action selection. Neuron 93, 1436–1450.e8 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Parkes, S. L., Bradfield, L. A. & Balleine, B. W. Interaction of insular cortex and ventral striatum mediates the effect of incentive memory on choice between goal-directed actions. J. Neurosci. 35, 6464–6471 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Vo, K., Rutledge, R. B., Chatterjee, A. & Kable, J. W. Dorsal striatum is necessary for stimulus-value but not action-value learning in humans. Brain 137, 3129–3135 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Voorn, P., Vanderschuren, L. J., Groenewegen, H. J., Robbins, T. W. & Pennartz, C. M. Putting a spin on the dorsal-ventral divide of the striatum. Trends Neurosci. 27, 468–474 (2004).

    Article  CAS  PubMed  Google Scholar 

  21. Marcellino, D., Kehr, J., Agnati, L. F. & Fuxe, K. Increased affinity of dopamine for D2-like versus D1-like receptors. Relevance for volume transmission in interpreting PET findings. Synapse 66, 196–203 (2012).

    Article  CAS  PubMed  Google Scholar 

  22. Richfield, E. K., Penney, J. B. & Young, A. B. Anatomical and affinity state comparisons between dopamine D1 and D2 receptors in the rat central nervous system. Neuroscience 30, 767–777 (1989).

    Article  CAS  PubMed  Google Scholar 

  23. Bromberg-Martin, E. S., Matsumoto, M. & Hikosaka, O. Dopamine in motivational control: rewarding, aversive, and alerting. Neuron 68, 815–834 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Danjo, T., Yoshimi, K., Funabiki, K., Yawata, S. & Nakanishi, S. Aversive behavior induced by optogenetic inactivation of ventral tegmental area dopamine neurons is mediated by dopamine D2 receptors in the nucleus accumbens. Proc. Natl Acad. Sci. USA 111, 6455–6460 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Dreyer, J. K., Herrik, K. F., Berg, R. W. & Hounsgaard, J. D. Influence of phasic and tonic dopamine release on receptor activation. J. Neurosci. 30, 14273–14283 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Trifilieff, P. et al. Increasing dopamine D2 receptor expression in the adult nucleus accumbens enhances motivation. Mol. Psychiatry 18, 1025–1033 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Zweifel, L. S. et al. Disruption of NMDAR-dependent burst firing by dopamine neurons provides selective assessment of phasic dopamine-dependent behavior. Proc. Natl Acad. Sci. USA 106, 7281–7288 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Garris, P. A., Ciolkowski, E. L., Pastore, P. & Wightman, R. M. Efflux of dopamine from the synaptic cleft in the nucleus accumbens of the rat brain. J. Neurosci. 14, 6084–6093 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Lammel, S., Lim, B. K. & Malenka, R. C. Reward and aversion in a heterogeneous midbrain dopamine system. Neuropharmacology 76, 351–359 (2014).

    Article  CAS  PubMed  Google Scholar 

  30. Morales, M. & Margolis, E. B. Ventral tegmental area: cellular heterogeneity, connectivity and behaviour. Nat. Rev. Neurosci. 18, 73–85 (2017).

    Article  CAS  PubMed  Google Scholar 

  31. Marinelli, M. & McCutcheon, J. E. Heterogeneity of dopamine neuron activity across traits and states. Neuroscience 282, 176–197 (2014).

    Article  CAS  PubMed  Google Scholar 

  32. Saddoris, M. P., Cacciapaglia, F., Wightman, R. M. & Carelli, R. M. Differential dopamine release dynamics in the nucleus accumbens core and shell reveal complementary signals for error prediction and incentive motivation. J. Neurosci. 35, 11572–11582 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Robinson, D. L., Zitzman, D. L. & Williams, S. K. Mesolimbic dopamine transients in motivated behaviors: focus on maternal behavior. Front. Psychiatry 2, 23 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Schultz, W. Updating dopamine reward signals. Curr. Opin. Neurobiol. 23, 229–238 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Floresco, S. B., West, A. R., Ash, B., Moore, H. & Grace, A. A. Afferent modulation of dopamine neuron firing differentially regulates tonic and phasic dopamine transmission. Nat. Neurosci. 6, 968–973 (2003).

    Article  CAS  PubMed  Google Scholar 

  36. Wang, L. P. et al. NMDA receptors in dopaminergic neurons are crucial for habit learning. Neuron 72, 1055–1066 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Grace, A. A., Floresco, S. B., Goto, Y. & Lodge, D. J. Regulation of firing of dopaminergic neurons and control of goal-directed behaviors. Trends Neurosci. 30, 220–227 (2007).

    Article  CAS  PubMed  Google Scholar 

  38. Huerta-Ocampo, I., Mena-Segovia, J. & Bolam, J. P. Convergence of cortical and thalamic input to direct and indirect pathway medium spiny neurons in the striatum. Brain Struct. Function 219, 1787–1800 (2014).

    Article  Google Scholar 

  39. Gerfen, C. R. & Surmeier, D. J. Modulation of striatal projection systems by dopamine. Annu. Rev. Neurosci. 34, 441–466 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Namburi, P., Al-Hasani, R., Calhoon, G. G., Bruchas, M. R. & Tye, K. M. Architectural representation of valence in the limbic system. Neuropsychopharmacology 41, 1697–1715 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Luo, Z., Volkow, N. D., Heintz, N., Pan, Y. & Du, C. Acute cocaine induces fast activation of D1 receptor and progressive deactivation of D2 receptor striatal neurons: in vivo optical microprobe [Ca2+]i imaging. J. Neurosci. 31, 13180–13190 (2011). This study provides preliminary evidence that the rate dependency of the effect of acute cocaine may relate to its fast and short-lasting activation of D1R-expressing striatal neurons in contrast to the slower and longer-lasting deactivation of D2R-expressing neurons. It also corroborates that DA activates D1R-expressing neurons and inhibits D2R-expressing neurons in the striatum.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Ferre, S. The GPCR heterotetramer: challenging classical pharmacology. Trends Pharmacol Sci. 36, 145–152 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Fiorentini, C., Busi, C., Spano, P. & Missale, C. Dimerization of dopamine D1 and D3 receptors in the regulation of striatal function. Curr. Opin. Pharmacol. 10, 87–92 (2010).

    Article  CAS  PubMed  Google Scholar 

  44. Kravitz, A. V., Tye, L. D. & Kreitzer, A. C. Distinct roles for direct and indirect pathway striatal neurons in reinforcement. Nat. Neurosci. 15, 816–818 (2012). This study uses optogenetic stimulation to demonstrate that the activation of the striatal direct pathway is sufficient for persistent reinforcement, whereas activation of the indirect pathway is sufficient for transient punishment. It also shows that reinforcement is more effective than punishment at modifying long-term behaviour.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. 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 66, 896–907 (2010). This study shows that both direct and indirect pathways are engaged by psychostimulant rewards: the direct pathway is active in distinguishing associative rewarding stimuli from non-associative ones, and the indirect pathway is involved with rapid memory formation to avoid aversive stimuli.

    Article  CAS  PubMed  Google Scholar 

  46. Cui, G. et al. Concurrent activation of striatal direct and indirect pathways during action initiation. Nature 494, 238–242 (2013). This study uses optogenetics to retest the classic model of direct versus indirect pathway in animals performing an operant task. The investigators report that during an operant task, there were transient increases in neural activity in both direct-pathway and indirect-pathway spiny projection neurons when animals initiated actions but not when they were inactive, which challenges some classical views of basal ganglia function.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Nakanishi, S., Hikida, T. & Yawata, S. Distinct dopaminergic control of the direct and indirect pathways in reward-based and avoidance learning behaviors. Neuroscience 282, 49–59 (2014).

    Article  CAS  PubMed  Google Scholar 

  48. Vicente, A. M., Galvao-Ferreira, P., Tecuapetla, F. & Costa, R. M. Direct and indirect dorsolateral striatum pathways reinforce different action strategies. Curr. Biol. 26, R267–R269 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Kupchik, Y. M. et al. Coding the direct/indirect pathways by D1 and D2 receptors is not valid for accumbens projections. Nat. Neurosci. 18, 1230–1232 (2015). This study provides optogenetic and electrophysiological evidence to suggest that the model in which D1R -expressing MSNs convey information directly to the output nuclei of the basal ganglia, whereas D2R-expressing neurons do so indirectly via pallidal neurons, may not apply to the projections from the accumbens to the ventral pallidum.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Calabresi, P., Picconi, B., Tozzi, A., Ghiglieri, V. & Di Filippo, M. Direct and indirect pathways of basal ganglia: a critical reappraisal. Nat. Neurosci. 17, 1022–1030 (2014). This Review provides an updated model of the direct and indirect pathways presumed to have facilitating and inhibitory effects on movement, respectively that incorporates the role of intrastriatal connections during movement.

    Article  CAS  PubMed  Google Scholar 

  51. Koob, G. F., Everitt, E. J. & Robbins, T. W. in Fundamental Neuroscience 3rd edn (eds Squire, L. et al.) 987–1016 (Academic Press, 2008).

    Google Scholar 

  52. Volkow, N. D., Wang, G. J., Tomasi, D. & Baler, R. D. Unbalanced neuronal circuits in addiction. Curr. Opin. Neurobiol. 23, 639–648 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Volkow, N. D. & Morales, M. The brain on drugs: from reward to addiction. Cell 162, 712–725 (2015). This review provides an in-depth update of the circuit- and cell-level mechanisms underlying the addictive state and its co-option of pathways regulating reward, self-control and affect.

    Article  CAS  PubMed  Google Scholar 

  54. Chen, J., Papies, E. K. & Barsalou, L. W. A core eating network and its modulations underlie diverse eating phenomena. Brain Cogn. 110, 20–42 (2016).

    Article  PubMed  Google Scholar 

  55. Day, J. J., Jones, J. L., Wightman, R. M. & Carelli, R. M. Phasic nucleus accumbens dopamine release encodes effort- and delay-related costs. Biol. Psychiatry 68, 306–309 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Hamid, A. A. et al. Mesolimbic dopamine signals the value of work. Nat. Neurosci. 19, 117–126 (2016).

    Article  CAS  PubMed  Google Scholar 

  57. Parsons, L. H. & Justice, J. B. Jr. Extracellular concentration and in vivo recovery of dopamine in the nucleus accumbens using microdialysis. J. Neurochem. 58, 212–218 (1992).

    Article  CAS  PubMed  Google Scholar 

  58. Keithley, R. B. et al. Higher sensitivity dopamine measurements with faster-scan cyclic voltammetry. Anal. Chem. 83, 3563–3571 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Wightman, R. M. & Robinson, D. L. Transient changes in mesolimbic dopamine and their association with 'reward'. J. Neurochem. 82, 721–735 (2002).

    Article  CAS  PubMed  Google Scholar 

  60. Small, D. M., Zatorre, R. J., Dagher, A., Evans, A. C. & Jones-Gotman, M. Changes in brain activity related to eating chocolate: from pleasure to aversion. Brain 124, 1720–1733 (2001).

    Article  CAS  PubMed  Google Scholar 

  61. Sun, X. et al. The neural signature of satiation is associated with ghrelin response and triglyceride metabolism. Physiol. Behav. 136, 63–73 (2014).

    Article  CAS  PubMed  Google Scholar 

  62. Wise, R. A. et al. Fluctuations in nucleus accumbens dopamine concentration during intravenous cocaine self-administration in rats. Psychopharmacology 120, 10–20 (1995).

    Article  CAS  PubMed  Google Scholar 

  63. Hyman, S. E. Addiction: a disease of learning and memory. Am. J. Psychiatry 162, 1414–1422 (2005).

    Article  PubMed  Google Scholar 

  64. Nair, S. G., Adams-Deutsch, T., Epstein, D. H. & Shaham, Y. The neuropharmacology of relapse to food seeking: methodology, main findings, and comparison with relapse to drug seeking. Prog. Neurobiol. 89, 18–45 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Harnett, M. T., Bernier, B. E., Ahn, K. C. & Morikawa, H. Burst-timing-dependent plasticity of NMDA receptor-mediated transmission in midbrain dopamine neurons. Neuron 62, 826–838 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Madhavan, A., Argilli, E., Bonci, A. & Whistler, J. L. Loss of D2 dopamine receptor function modulates cocaine-induced glutamatergic synaptic potentiation in the ventral tegmental area. J. Neurosci. 33, 12329–12336 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Wang, B. et al. Cocaine experience establishes control of midbrain glutamate and dopamine by corticotropin-releasing factor: a role in stress-induced relapse to drug seeking. J. Neurosci. 25, 5389–5396 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. You, Z. B., Wang, B., Zitzman, D. & Wise, R. A. Acetylcholine release in the mesocorticolimbic dopamine system during cocaine seeking: conditioned and unconditioned contributions to reward and motivation. J. Neurosci. 28, 9021–9029 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Calabresi, P., Centonze, D., Gubellini, P., Marfia, G. A. & Bernardi, G. Glutamate-triggered events inducing corticostriatal long-term depression. J. Neurosci. 19, 6102–6110 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Graziane, N. M. et al. Opposing mechanisms mediate morphine- and cocaine-induced generation of silent synapses. Nat. Neurosci. 19, 915–925 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Vanderschuren, L. J., Di Ciano, P. & Everitt, B. J. Involvement of the dorsal striatum in cue-controlled cocaine seeking. J. Neurosci. 25, 8665–8670 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Kauer, J. A. & Malenka, R. C. Synaptic plasticity and addiction. Nat. Rev. Neurosci. 8, 844–858 (2007).

    Article  CAS  PubMed  Google Scholar 

  73. Conrad, K. L. et al. Formation of accumbens GluR2-lacking AMPA receptors mediates incubation of cocaine craving. Nature 454, 118–121 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Park, K., Volkow, N. D., Pan, Y. & Du, C. Chronic cocaine dampens dopamine signaling during cocaine intoxication and unbalances D1 over D2 receptor signaling. J. Neurosci. 33, 15827–15836 (2013). This study uses optical imaging to compare adaptation in the D1R-expressing and D2R-expressing MSNs to chronic cocaine administration. It documents an imbalance associated with repeated cocaine exposure that favours D1R over D2R signalling, which could help explain the compulsive patterns of cocaine intake in addiction.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Mancino, S., Mendonça-Netto, S., Martín- García, E. & Maldonado, R. Role of DOR in neuronal plasticity changes promoted by food-seeking behaviour. Addict. Biol. 22, 1179–1190 (2016).

    Article  CAS  PubMed  Google Scholar 

  76. Gutierrez-Cuesta, J. et al. Effects of genetic deletion of endogenous opioid system components on the reinstatement of cocaine-seeking behavior in mice. Neuropsychopharmacology 39, 2974–2988 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Guegan, T. et al. Operant behavior to obtain palatable food modifies ERK activity in the brain reward circuit. Eur. Neuropsychopharmacol. 23, 240–252 (2013).

    Article  CAS  PubMed  Google Scholar 

  78. Ren, Z. et al. Dopamine D1 and N-methyl-D-aspartate receptors and extracellular signal-regulated kinase mediate neuronal morphological changes induced by repeated cocaine administration. Neuroscience 168, 48–60 (2010).

    Article  CAS  PubMed  Google Scholar 

  79. Nogueiras, R. et al. The opioid system and food intake: homeostatic and hedonic mechanisms. Obes. Facts 5, 196–207 (2012).

    Article  PubMed  Google Scholar 

  80. Gosnell, B. A. & Levine, A. S. Reward systems and food intake: role of opioids. Int. J. Obes. 33 (Suppl. 2), S54–S58 (2009).

    Article  CAS  Google Scholar 

  81. Befort, K. Interactions of the opioid and cannabinoid systems in reward: Insights from knockout studies. Front. Pharmacol. 6, 6 (2015).

    PubMed  PubMed Central  Google Scholar 

  82. Wenzel, J. M. & Cheer, J. F. Endocannabinoid regulation of reward and reinforcement through interaction with dopamine and endogenous opioid signaling. Neuropsychopharmacology http://dx.doi.org/10.1038/npp.2017.126 (2017).

  83. Ball, K. T., Best, O., Luo, J. & Miller, L. R. Chronic restraint stress causes a delayed increase in responding for palatable food cues during forced abstinence via a dopamine D1-like receptor-mediated mechanism. Behav. Brain Res. 319, 1–8 (2017).

    Article  CAS  PubMed  Google Scholar 

  84. Ball, K. T., Combs, T. A. & Beyer, D. N. Opposing roles for dopamine D1- and D2-like receptors in discrete cue-induced reinstatement of food seeking. Behav. Brain Res. 222, 390–393 (2011). This study extends the applicability of a classical animal model of drug abuse relapse to the investigation of the reinstatement of food-seeking behaviours. It finds that pharmacological blockade of dopamine D1-like receptors abrogates discrete cue-induced reinstatement of food seeking, whereas D2-like receptor blockade increases responding during reinstatement tests.

    Article  CAS  PubMed  Google Scholar 

  85. Ball, K. T. et al. Effects of repeated yohimbine administration on reinstatement of palatable food seeking: involvement of dopamine D1-like receptors and food-associated cues. Addict. Biol. 21, 1140–1150 (2016).

    Article  CAS  PubMed  Google Scholar 

  86. Billes, S. K., Simonds, S. E. & Cowley, M. A. Leptin reduces food intake via a dopamine D2 receptor-dependent mechanism. Mol. Metab. 1, 86–93 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Han, W. et al. Striatal dopamine links gastrointestinal rerouting to altered sweet appetite. Cell Metab. 23, 103–112 (2016).

    Article  CAS  PubMed  Google Scholar 

  88. Adams, W. K. et al. Long-term, calorie-restricted intake of a high-fat diet in rats reduces impulse control and ventral striatal D2 receptor signalling — two markers of addiction vulnerability. Eur. J. Neurosci. 42, 3095–3104 (2015).

    Article  PubMed  Google Scholar 

  89. Johnson, P. M. & Kenny, P. J. Dopamine D2 receptors in addiction-like reward dysfunction and compulsive eating in obese rats. Nat. Neurosci. 13, 635–641 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. van de Giessen, E. et al. High fat/carbohydrate ratio but not total energy intake induces lower striatal dopamine D2/3 receptor availability in diet-induced obesity. Int. J. Obes. 37, 754–757 (2013).

    Article  CAS  Google Scholar 

  91. Tellez, L. A. et al. A gut lipid messenger links excess dietary fat to dopamine deficiency. Science 341, 800–802 (2013).

    Article  CAS  PubMed  Google Scholar 

  92. Zhu, X., Ottenheimer, D. & DiLeone, R. J. Activity of D1/2 receptor expressing neurons in the nucleus accumbens regulates running, locomotion, and food intake. Front. Behav. Neurosci. 10, 66 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Everitt, B. J. et al. Neural mechanisms underlying the vulnerability to develop compulsive drug-seeking habits and addiction. Philos. Trans. R. Soc. Lond. B Biol. Sci. 363, 3125–3135 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  94. Szczypka, M. S. et al. Dopamine production in the caudate putamen restores feeding in dopamine-deficient mice. Neuron 30, 819–828 (2001).

    Article  CAS  PubMed  Google Scholar 

  95. Volkow, N. D. et al. “Nonhedonic” food motivation in humans involves dopamine in the dorsal striatum and methylphenidate amplifies this effect. Synapse 44, 175–180 (2002).

    Article  CAS  PubMed  Google Scholar 

  96. Epel, E. S. et al. The reward-based eating drive scale: a self-report index of reward-based eating. PLoS ONE 9, e101350 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Volkow, N. D., Koob, G. F. & McLellan, A. T. Neurobiologic advances from the brain disease model of addiction. N. Engl. J. Med. 374, 363–371 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Moffitt, T. E. et al. A gradient of childhood self-control predicts health, wealth, and public safety. Proc. Natl Acad. Sci. USA 108, 2693–2698 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Tarter, R. E. et al. Neurobehavioral disinhibition in childhood predicts early age at onset of substance use disorder. Am. J. Psychiatry 160, 1078–1085 (2003).

    Article  PubMed  Google Scholar 

  100. Volkow, N. D. & Fowler, J. S. Addiction, a disease of compulsion and drive: involvement of the orbitofrontal cortex. Cereb. Cortex 10, 318–325 (2000).

    Article  CAS  PubMed  Google Scholar 

  101. Volkow, N. D. et al. High levels of dopamine D2 receptors in unaffected members of alcoholic families: possible protective factors. Arch. Gen. Psychiatry 63, 999–1008 (2006).

    Article  CAS  PubMed  Google Scholar 

  102. Wang, G. J. et al. Brain dopamine and obesity. Lancet 357, 354–357 (2001).

    Article  CAS  PubMed  Google Scholar 

  103. van de Giessen, E., Celik, F., Schweitzer, D. H., van den Brink, W. & Booij, J. Dopamine D2/3 receptor availability and amphetamine-induced dopamine release in obesity. J. Psychopharmacol. 28, 866–873 (2014).

    Article  CAS  PubMed  Google Scholar 

  104. de Weijer, B. A. et al. Lower striatal dopamine D2/3 receptor availability in obese compared with non-obese subjects. EJNMMI Res. 1, 37 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Steele, K. E. et al. Alterations of central dopamine receptors before and after gastric bypass surgery. Obes. Surg. 20, 369–374 (2010).

    Article  PubMed  Google Scholar 

  106. Volkow, N. D. et al. Low dopamine striatal D2 receptors are associated with prefrontal metabolism in obese subjects: possible contributing factors. Neuroimage 42, 1537–1543 (2008).

    Article  PubMed  Google Scholar 

  107. Volkow, N. D. et al. Decreased dopamine D2 receptor availability is associated with reduced frontal metabolism in cocaine abusers. Synapse 14, 169–177 (1993).

    Article  CAS  PubMed  Google Scholar 

  108. Volkow, N. D. et al. Low level of brain dopamine D2 receptors in methamphetamine abusers: association with metabolism in the orbitofrontal cortex. Am. J. Psychiatry 158, 2015–2021 (2001).

    Article  CAS  PubMed  Google Scholar 

  109. Dunn, J. P. et al. Relationship of dopamine type 2 receptor binding potential with fasting neuroendocrine hormones and insulin sensitivity in human obesity. Diabetes Care 35, 1105–1111 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Karlsson, H. K. et al. Obesity is associated with decreased μ-opioid but unaltered dopamine D2 receptor availability in the brain. J. Neurosci. 35, 3959–3965 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Martinez, D. et al. Dopamine D1 receptors in cocaine dependence measured with PET and the choice to self-administer cocaine. Neuropsychopharmacology 34, 1774–1782 (2009).

    Article  CAS  PubMed  Google Scholar 

  112. Sinha, R. & Li, C. S. Imaging stress- and cue-induced drug and alcohol craving: association with relapse and clinical implications. Drug Alcohol Rev. 26, 25–31 (2007).

    Article  PubMed  Google Scholar 

  113. Weygandt, M. et al. Impulse control in the dorsolateral prefrontal cortex counteracts post-diet weight regain in obesity. Neuroimage 109, 318–327 (2015).

    Article  PubMed  Google Scholar 

  114. Tomasi, D. & Volkow, N. D. Striatocortical pathway dysfunctionin addiction and obesity: differences and similarities. Crit. Rev. Biochem. Mol. Biol. 48, 1–19 (2013).

    Article  CAS  PubMed  Google Scholar 

  115. Wang, G. J. et al. BMI modulates calorie-dependent dopamine changes in accumbens from glucose intake. PLoS ONE 9, e101585 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  116. Cosgrove, K. P., Veldhuizen, M. G., Sandiego, C. M., Morris, E. D. & Small, D. M. Opposing relationships of BMI with BOLD and dopamine D2/3 receptor binding potential in the dorsal striatum. Synapse 69, 195–202 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Stice, E., Yokum, S., Blum, K. & Bohon, C. Weight gain is associated with reduced striatal response to palatable food. J. Neurosci. 30, 13105–13109 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Volkow, N. D. et al. Stimulant-induced dopamine increases are markedly blunted in active cocaine abusers. Mol. Psychiatry 19, 1037–1043 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Carelli, R. M. & West, E. A. When a good taste turns bad: neural mechanisms underlying the emergence of negative affect and associated natural reward devaluation by cocaine. Neuropharmacology 76, 360–369 (2014).

    Article  CAS  PubMed  Google Scholar 

  120. Parvaz, M. A., Alia-Klein, N., Woicik, P. A., Volkow, N. D. & Goldstein, R. Z. Neuroimaging for drug addiction and related behaviors. Rev. Neurosci. 22, 609–624 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Garcia-Garcia, I. et al. Functional connectivity in obesity during reward processing. Neuroimage 66, 232–239 (2013).

    Article  CAS  PubMed  Google Scholar 

  122. Volkow, N. D. et al. Motivation deficit in ADHD is associated with dysfunction of the dopamine reward pathway. Mol. Psychiatry 16, 1147–1154 (2011).

    Article  CAS  PubMed  Google Scholar 

  123. Volkow, N. D. et al. Methylphenidate-elicited dopamine increases in ventral striatum are associated with long-term symptom improvement in adults with attention deficit hyperactivity disorder. J. Neurosci. 32, 841–849 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Courtney, K. E., Schacht, J. P., Hutchison, K., Roche, D. J. & Ray, L. A. Neural substrates of cue reactivity: association with treatment outcomes and relapse. Addict. Biol. 21, 3–22 (2016).

    Article  PubMed  Google Scholar 

  125. Stice, E., Spoor, S., Bohon, C., Veldhuizen, M. G. & Small, D. M. Relation of reward from food intake and anticipated food intake to obesity: a functional magnetic resonance imaging study. J. Abnorm. Psychol. 117, 924–935 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  126. Volkow, N. D. Opioid-dopamine interactions: implications for substance use disorders and their treatment. Biol. Psychiatry 68, 685–686 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  127. Gilbert-Diamond, D. et al. Television food advertisement exposure and FTO rs9939609 genotype in relation to excess consumption in children. Int. J. Obes. 41, 23–29 (2017).

    Article  CAS  Google Scholar 

  128. Rapuano, K. M. et al. Genetic risk for obesity predicts nucleus accumbens size and responsivity to real-world food cues. Proc. Natl Acad. Sci. USA 114, 160–165 (2017).

    Article  CAS  PubMed  Google Scholar 

  129. Koob, G. F. & Le Moal, M. Plasticity of reward neurocircuitry and the 'dark side' of drug addiction. Nat. Neurosci. 8, 1442–1444 (2005).

    Article  CAS  PubMed  Google Scholar 

  130. Gramsch, C., Blasig, J. & Herz, A. Changes in striatal dopamine metabolism during precipitated morphine withdrawal. Eur. J. Pharmacol. 44, 231–240 (1977).

    Article  CAS  PubMed  Google Scholar 

  131. Wang, G. J. et al. Dopamine D2 receptor availability in opiate-dependent subjects before and after naloxone-precipitated withdrawal. Neuropsychopharmacology 16, 174–182 (1997).

    Article  CAS  PubMed  Google Scholar 

  132. Koob, G. F. et al. Addiction as a stress surfeit disorder. Neuropharmacology 76, 370–382 (2014).

    Article  CAS  PubMed  Google Scholar 

  133. Colantuoni, C. et al. Evidence that intermittent, excessive sugar intake causes endogenous opioid dependence. Obes. Res. 10, 478–488 (2002).

    Article  CAS  PubMed  Google Scholar 

  134. Parylak, S. L., Koob, G. F. & Zorrilla, E. P. The dark side of food addiction. Physiol. Behav. 104, 149–156 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. de Witt Huberts, J. C., Evers, C. & de Ridder, D. T. Double trouble: restrained eaters do not eat less and feel worse. Psychol. Health 28, 686–700 (2013).

    Article  PubMed  Google Scholar 

  136. Hill, A. J., Weaver, C. F. & Blundell, J. E. Food craving, dietary restraint and mood. Appetite 17, 187–197 (1991).

    Article  CAS  PubMed  Google Scholar 

  137. Sinha, R. & Jastreboff, A. M. Stress as a common risk factor for obesity and addiction. Biol. Psychiatry 73, 827–835 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Yau, Y. H. & Potenza, M. N. Stress and eating behaviors. Minerva Endocrinol. 38, 255–267 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  139. de Araujo, I. E. et al. Food reward in the absence of taste receptor signaling. Neuron 57, 930–941 (2008).

    Article  CAS  PubMed  Google Scholar 

  140. Narayanaswami, V. & Dwoskin, L. P. Obesity: current and potential pharmacotherapeutics and targets. Pharmacol. Ther. 170, 116–147 (2017).

    Article  CAS  PubMed  Google Scholar 

  141. Collins, G. T., Gerak, L. R., Javors, M. A. & France, C. P. Lorcaserin reduces the discriminative stimulus and reinforcing effects of cocaine in rhesus monkeys. J. Pharmacol. Exp. Ther. 356, 85–95 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Neelakantan, H. et al. Lorcaserin suppresses oxycodone self-administration and relapse vulnerability in rats. ACS Chem. Neurosci. 8, 1065–1073 (2017).

    Article  CAS  PubMed  Google Scholar 

  143. Higgins, G. A. et al. The 5-HT2C receptor agonist lorcaserin reduces nicotine self-administration, discrimination, and reinstatement: relationship to feeding behavior and impulse control. Neuropsychopharmacology 37, 1177–1191 (2012).

    Article  CAS  PubMed  Google Scholar 

  144. Howell, L. L. & Cunningham, K. A. Serotonin 5-HT2 receptor interactions with dopamine function: implications for therapeutics in cocaine use disorder. Pharmacol Rev. 67, 176–197 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Mooney, M. E. et al. Bupropion and naltrexone for smoking cessation: a double-blind randomized placebo-controlled clinical trial. Clin. Pharmacol. Ther. 100, 344–352 (2016).

    Article  CAS  PubMed  Google Scholar 

  146. Wilcox, C. S. et al. An open-label study of naltrexone and bupropion combination therapy for smoking cessation in overweight and obese subjects. Addict. Behav. 35, 229–234 (2010).

    Article  PubMed  Google Scholar 

  147. Vallof, D. et al. The glucagon-like peptide 1 receptor agonist liraglutide attenuates the reinforcing properties of alcohol in rodents. Addict. Biol. 21, 422–437 (2016).

    Article  CAS  PubMed  Google Scholar 

  148. Cahill, K. & Ussher, M. Cannabinoid type 1 receptor antagonists for smoking cessation. Cochrane Database Syst. Rev. 3, CD005353 (2011).

    Google Scholar 

  149. Justinova, Z., Panlilio, L. V. & Goldberg, S. R. Drug addiction. Curr. Top. Behav. Neurosci. 1, 309–346 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. King, W. C. et al. Prevalence of alcohol use disorders before and after bariatric surgery. JAMA 307, 2516–2525 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Svensson, P. A. et al. Alcohol consumption and alcohol problems after bariatric surgery in the Swedish obese subjects study. Obesity 21, 2444–2451 (2013).

    Article  PubMed  Google Scholar 

  152. Raebel, M. A. et al. Chronic use of opioid medications before and after bariatric surgery. JAMA 310, 1369–1376 (2013).

    Article  CAS  PubMed  Google Scholar 

  153. Polston, J. E. et al. Roux-en-Y gastric bypass increases intravenous ethanol self-administration in dietary obese rats. PLoS ONE 8, e83741 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Biegler, J. M., Freet, C. S., Horvath, N., Rogers, A. M. & Hajnal, A. Increased intravenous morphine self-administration following Roux-en-Y gastric bypass in dietary obese rats. Brain Res. Bull. 123, 47–52 (2016).

    Article  CAS  PubMed  Google Scholar 

  155. Steffen, K. J., Engel, S. G., Wonderlich, J. A., Pollert, G. A. & Sondag, C. Alcohol and other addictive disorders following bariatric surgery: prevalence, risk factors and possible etiologies. Eur. Eat Disord. Rev. 23, 442–450 (2015).

    Article  PubMed  Google Scholar 

  156. Bolloni, C. et al. Bilateral transcranial magnetic stimulation of the prefrontal cortex reduces cocaine intake: a pilot study. Front. Psychiatry 7, 133 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  157. Val-Laillet, D. et al. Neuroimaging and neuromodulation approaches to study eating behavior and prevent and treat eating disorders and obesity. Neuroimage Clin. 8, 1–31 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Jauch-Chara, K. et al. Repetitive electric brain stimulation reduces food intake in humans. Am. J. Clin. Nutr. 100, 1003–1009 (2014).

    Article  CAS  PubMed  Google Scholar 

  159. Childs, J. E., DeLeon, J., Nickel, E. & Kroener, S. Vagus nerve stimulation reduces cocaine seeking and alters plasticity in the extinction network. Learn. Mem. 24, 35–42 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Wing, V. C. et al. Brain stimulation methods to treat tobacco addiction. Brain Stimul. 6, 221–230 (2013).

    Article  PubMed  Google Scholar 

  161. Ceccanti, M. et al. Deep TMS on alcoholics: effects on cortisolemia and dopamine pathway modulation. A pilot study. Can. J. Physiol. Pharmacol. 93, 283–290 (2015).

    Article  CAS  PubMed  Google Scholar 

  162. Terraneo, A. et al. Transcranial magnetic stimulation of dorsolateral prefrontal cortex reduces cocaine use: a pilot study. Eur. Neuropsychopharmacol. 26, 37–44 (2016).

    Article  CAS  PubMed  Google Scholar 

  163. Franco, R. et al. DBS for obesity. Brain Sci. 6, 21 (2016).

    Article  PubMed Central  Google Scholar 

  164. McClelland, J., Bozhilova, N., Campbell, I. & Schmidt, U. A systematic review of the effects of neuromodulation on eating and body weight: evidence from human and animal studies. Eur. Eat Disord. Rev. 21, 436–455 (2013).

    Article  PubMed  Google Scholar 

  165. Guiraud, D. et al. Vagus nerve stimulation: state of the art of stimulation and recording strategies to address autonomic function neuromodulation. J. Neural Eng. 13, 041002 (2016).

    Article  PubMed  Google Scholar 

  166. Muller, U. J. et al. Nucleus accumbens deep brain stimulation for alcohol addiction — safety and clinical long-term results of a pilot trial. Pharmacopsychiatry 49, 170–173 (2016).

    Article  CAS  PubMed  Google Scholar 

  167. Burgess, E., Hassmen, P. & Pumpa, K. L. Determinants of adherence to lifestyle intervention in adults with obesity: a systematic review. Clin. Obes. 7, 123–135 (2017).

    Article  CAS  PubMed  Google Scholar 

  168. Zhou, Y., Zhao, M., Zhou, C. & Li, R. Sex differences in drug addiction and response to exercise intervention: from human to animal studies. Front. Neuroendocrinol. 40, 24–41 (2016).

    Article  PubMed  Google Scholar 

  169. Kravitz, A. V., O'Neal, T. J. & Friend, D. M. Do dopaminergic impairments underlie physical inactivity in people with obesity? Front. Hum. Neurosci. 10, 514 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Chen, W. et al. Moderate intensity treadmill exercise alters food preference via dopaminergic plasticity of ventral tegmental area-nucleus accumbens in obese mice. Neurosci. Lett. 641, 56–61 (2017).

    Article  CAS  PubMed  Google Scholar 

  171. Volkow, N. D. et al. Evidence that sleep deprivation downregulates dopamine D2R in ventral striatum in the human brain. J. Neurosci. 32, 6711–6717 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Verwey, M., Dhir, S. & Amir, S. Circadian influences on dopamine circuits of the brain: regulation of striatal rhythms of clock gene expression and implications for psychopathology and disease. F1000Res 5, 2062 (2016).

    Article  CAS  Google Scholar 

  173. Wiers, C. E. et al. Reduced sleep duration mediates decreases in striatal D2/D3 receptor availability in cocaine abusers. Transl Psychiatry 6, e752 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Carlucci, C., Petrof, E. O. & Allen-Vercoe, E. Fecal microbiota-based therapeutics for recurrent Clostridium difficile infection, ulcerative colitis and obesity. EBioMedicine 13, 37–45 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  175. Jayasinghe, T. N., Chiavaroli, V., Holland, D. J., Cutfield, W. S. & O'Sullivan, J. M. The new era of treatment for obesity and metabolic disorders: evidence and expectations for gut microbiome transplantation. Front. Cell. Infect. Microbiol. 6, 15 (2016). This study uses chemogenetics to show that GLP-1 released from nucleus of the tractus solitarius neurons reduces highly palatable food intake by suppressing mesolimbic DA signalling.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Engen, P. A., Green, S. J., Voigt, R. M., Forsyth, C. B. & Keshavarzian, A. The gastrointestinal microbiome: alcohol effects on the composition of intestinal microbiota. Alcohol Res. 37, 223–236 (2015). This review discusses the bidirectional interactions between energy homeostasis signals and neural circuits that control motivation and food intake, with a focus on the activity of specific cell types in these networks.

    PubMed  PubMed Central  Google Scholar 

  177. Kiraly, D. D. et al. Alterations of the host microbiome affect behavioral responses to cocaine. Sci. Rep. 6, 35455 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Wang, X. F. et al. Endogenous glucagon-like peptide-1 suppresses high-fat food intake by reducing synaptic drive onto mesolimbic dopamine neurons. Cell Rep. 12, 726–733 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Ferrario, C. R. et al. Homeostasis meets motivation in the battle to control food intake. J. Neurosci. 36, 11469–11481 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Hommel, J. D. et al. Leptin receptor signaling in midbrain dopamine neurons regulates feeding. Neuron 51, 801–810 (2006).

    Article  CAS  PubMed  Google Scholar 

  181. Figlewicz, D. P., Evans, S. B., Murphy, J., Hoen, M. & Baskin, D. G. Expression of receptors for insulin and leptin in the ventral tegmental area/substantia nigra (VTA/SN) of the rat. Brain Res. 964, 107–115 (2003).

    Article  CAS  PubMed  Google Scholar 

  182. Labouebe, G. et al. Insulin induces long-term depression of ventral tegmental area dopamine neurons via endocannabinoids. Nat. Neurosci. 16, 300–308 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Skibicka, K. P., Hansson, C., Alvarez-Crespo, M., Friberg, P. A. & Dickson, S. L. Ghrelin directly targets the ventral tegmental area to increase food motivation. Neuroscience 180, 129–137 (2011). This study uses optogenetics to investigate the role of GABAergic neurons in the lateral habenula VTA axis and documents that these neurons upregulate dopamine release in the nucleus accumbens by inhibiting local VTA GABAergic neurons; this finding is crucial for understanding how lateral habenula GABAergic cells increase the motivational salience of a stimulus.

    Article  CAS  PubMed  Google Scholar 

  184. Hahn, J. D. & Swanson, L. W. Distinct patterns of neuronal inputs and outputs of the juxtaparaventricular and suprafornical regions of the lateral hypothalamic area in the male rat. Brain Res. Rev. 64, 14–103 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  185. Bonnavion, P., Mickelsen, L. E., Fujita, A., de Lecea, L. & Jackson, A. C. Hubs and spokes of the lateral hypothalamus: cell types, circuits and behaviour. J. Physiol. 594, 6443–6462 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Nieh, E. H. et al. Inhibitory input from the lateral hypothalamus to the ventral tegmental area disinhibits dopamine neurons and promotes behavioral activation. Neuron 90, 1286–1298 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Borgland, S. L., Storm, E. & Bonci, A. Orexin B/hypocretin 2 increases glutamatergic transmission to ventral tegmental area neurons. Eur. J. Neurosci. 28, 1545–1556 (2008).

    Article  CAS  PubMed  Google Scholar 

  188. Leinninger, G. M. et al. Leptin action via neurotensin neurons controls orexin, the mesolimbic dopamine system and energy balance. Cell Metab. 14, 313–323 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Kempadoo, K. A. et al. Hypothalamic neurotensin projections promote reward by enhancing glutamate transmission in the VTA. J. Neurosci. 33, 7618–7626 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Harris, G. C. & Aston-Jones, G. Arousal and reward: a dichotomy in orexin function. Trends Neurosci. 29, 571–577 (2006).

    Article  CAS  PubMed  Google Scholar 

  191. Baimel, C. et al. Orexin/hypocretin role in reward: implications for opioid and other addictions. Br. J. Pharmacol. 172, 334–348 (2015).

    Article  CAS  PubMed  Google Scholar 

  192. Sakurai, T. et al. Orexins and orexin receptors: a family of hypothalamic neuropeptides and G protein-coupled receptors that regulate feeding behavior. Cell 92, 573–585 (1998).

    Article  CAS  PubMed  Google Scholar 

  193. Sheng, Z., Santiago, A. M., Thomas, M. P. & Routh, V. H. Metabolic regulation of lateral hypothalamic glucose-inhibited orexin neurons may influence midbrain reward neurocircuitry. Mol. Cell Neurosci. 62, 30–41 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Cone, R. D. Anatomy and regulation of the central melanocortin system. Nat. Neurosci. 8, 571–578 (2005).

    Article  CAS  PubMed  Google Scholar 

  195. Pandit, R. et al. Melanocortin 3 receptor signaling in midbrain dopamine neurons increases the motivation for food reward. Neuropsychopharmacology 41, 2241–2251 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Cansell, C., Denis, R. G., Joly-Amado, A., Castel, J. & Luquet, S. Arcuate AgRP neurons and the regulation of energy balance. Front. Endocrinol. 3, 169 (2012).

    Article  CAS  Google Scholar 

  197. Atasoy, D., Betley, J. N., Su, H. H. & Sternson, S. M. Deconstruction of a neural circuit for hunger. Nature 488, 172–177 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Fan, W., Boston, B. A., Kesterson, R. A., Hruby, V. J. & Cone, R. D. Role of melanocortinergic neurons in feeding and the agouti obesity syndrome. Nature 385, 165–168 (1997).

    Article  CAS  PubMed  Google Scholar 

  199. Baver, S. B. et al. Leptin modulates the intrinsic excitability of AgRP/NPY neurons in the arcuate nucleus of the hypothalamus. J. Neurosci. 34, 5486–5496 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Wang, Q. et al. Arcuate AgRP neurons mediate orexigenic and glucoregulatory actions of ghrelin. Mol. Metab. 3, 64–72 (2014).

    Article  CAS  PubMed  Google Scholar 

  201. Shen, M., Jiang, C., Liu, P., Wang, F. & Ma, L. Mesolimbic leptin signaling negatively regulates cocaine-conditioned reward. Transl Psychiatry 6, e972 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. You, Z. B. et al. Reciprocal inhibitory interactions between the reward-related effects of leptin and cocaine. Neuropsychopharmacology 41, 1024–1033 (2016).

    Article  CAS  PubMed  Google Scholar 

  203. Pfaffly, J. et al. Leptin increases striatal dopamine D2 receptor binding in leptin-deficient obese (ob/ob) mice. Synapse 64, 503–510 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Kiefer, F. et al. Leptin: a modulator of alcohol craving? Biol. Psychiatry 49, 782–787 (2001).

    Article  CAS  PubMed  Google Scholar 

  205. Aguiar-Nemer, A. S., Toffolo, M. C., da Silva, C. J., Laranjeira, R. & Silva-Fonseca, V. A. Leptin influence in craving and relapse of alcoholics and smokers. J. Clin. Med. Res. 5, 164–167 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  206. Reddy, I. A., Stanwood, G. D. & Galli, A. Moving beyond energy homeostasis: new roles for glucagon-like peptide-1 in food and drug reward. Neurochem. Int. 73, 49–55 (2014).

    Article  CAS  PubMed  Google Scholar 

  207. Sorensen, G. et al. The glucagon-like peptide 1 (GLP-1) receptor agonist exendin-4 reduces cocaine self-administration in mice. Physiol. Behav. 149, 262–268 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Engel, J. A. & Jerlhag, E. Role of appetite-regulating peptides in the pathophysiology of addiction: implications for pharmacotherapy. CNS Drugs 28, 875–886 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  209. al'Absi, M., Lemieux, A. & Nakajima, M. Peptide YY and ghrelin predict craving and risk for relapse in abstinent smokers. Psychoneuroendocrinology 49, 253–259 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Jiang, H., Betancourt, L. & Smith, R. G. Ghrelin amplifies dopamine signaling by cross talk involving formation of growth hormone secretagogue receptor/dopamine receptor subtype 1 heterodimers. Mol. Endocrinol. 20, 1772–1785 (2006).

    Article  CAS  PubMed  Google Scholar 

  211. Kern, A., Albarran-Zeckler, R., Walsh, H. E. & Smith, R. G. Apo-ghrelin receptor forms heteromers with DRD2 in hypothalamic neurons and is essential for anorexigenic effects of DRD2 agonism. Neuron 73, 317–332 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. Fetissov, S. O. Role of the gut microbiota in host appetite control: bacterial growth to animal feeding behaviour. Nat. Rev. Endocrinol. 13, 11–25 (2017).

    Article  CAS  PubMed  Google Scholar 

  213. Tremaroli, V. & Backhed, F. Functional interactions between the gut microbiota and host metabolism. Nature 489, 242–249 (2012).

    Article  CAS  PubMed  Google Scholar 

  214. Proctor, C., Thiennimitr, P., Chattipakorn, N. & Chattipakorn, S. C. Diet, gut microbiota and cognition. Metab. Brain Dis. 32, 1–17 (2017).

    Article  CAS  PubMed  Google Scholar 

  215. Zheng, P. et al. Gut microbiome remodeling induces depressive-like behaviors through a pathway mediated by the host's metabolism. Mol. Psychiatry 21, 786–796 (2016).

    Article  CAS  PubMed  Google Scholar 

  216. Lobo, M. K. et al. Cell type-specific loss of BDNF signaling mimics optogenetic control of cocaine reward. Science 330, 385–390 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  217. Wolf, M. E. Regulation of AMPA receptor trafficking in the nucleus accumbens by dopamine and cocaine. Neurotox. Res. 18, 393–409 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  218. al'Absi, M. et al. Circulating leptin levels are associated with increased craving to smoke in abstinent smokers. Pharmacol. Biochem. Behav. 97, 509–513 (2011).

    Article  CAS  PubMed  Google Scholar 

  219. Kiefer, F. et al. Increasing leptin precedes craving and relapse during pharmacological abstinence maintenance treatment of alcoholism. J. Psychiatr. Res. 39, 545–551 (2005).

    Article  PubMed  Google Scholar 

  220. von der Goltz, C. et al. Orexin and leptin are associated with nicotine craving: a link between smoking, appetite and reward. Psychoneuroendocrinology 35, 570–577 (2010).

    Article  CAS  PubMed  Google Scholar 

  221. Kraus, T. et al. Leptin is associated with craving in females with alcoholism. Addict. Biol. 9, 213–219 (2004).

    Article  CAS  PubMed  Google Scholar 

  222. Hillemacher, T. et al. Alteration of prolactin serum levels during alcohol withdrawal correlates with craving in female patients. Addict. Biol. 10, 337–343 (2005).

    Article  CAS  PubMed  Google Scholar 

  223. Wilhelm, J. et al. Prolactin serum levels during alcohol withdrawal are associated with the severity of alcohol dependence and withdrawal symptoms. Alcohol. Clin. Exp. Res. 35, 235–239 (2011).

    Article  PubMed  Google Scholar 

  224. Drazen, D. L., Vahl, T. P., D'Alessio, D. A., Seeley, R. J. & Woods, S. C. Effects of a fixed meal pattern on ghrelin secretion: evidence for a learned response independent of nutrient status. Endocrinology 147, 23–30 (2006).

    Article  CAS  PubMed  Google Scholar 

  225. Hong, S. I. et al. Phentermine induces conditioned rewarding effects via activation of the PI3K/Akt signaling pathway in the nucleus accumbens. Psychopharmacology 233, 1405–1413 (2016).

    Article  CAS  PubMed  Google Scholar 

  226. Rothman, R. B., Elmer, G. I., Shippenberg, T. S., Rea, W. & Baumann, M. H. Phentermine and fenfluramine. Preclinical studies in animal models of cocaine addiction. Ann. NY Acad. Sci. 844, 59–74 (1998).

    Article  CAS  PubMed  Google Scholar 

  227. Higgins, G. A., Sellers, E. M. & Fletcher, P. J. From obesity to substance abuse: therapeutic opportunities for 5-HT2C receptor agonists. Trends Pharmacol. Sci. 34, 560–570 (2013).

    Article  CAS  PubMed  Google Scholar 

  228. Schiffer, W. K. et al. Topiramate selectively attenuates nicotine-induced increases in monoamine release. Synapse 42, 196–198 (2001).

    Article  CAS  PubMed  Google Scholar 

  229. Arenas, M. C. et al. Topiramate increases the rewarding properties of cocaine in young-adult mice limiting its clinical usefulness. Psychopharmacology 233, 3849–3859 (2016).

    Article  CAS  PubMed  Google Scholar 

  230. Valenta, J. P. et al. μ-Opioid receptors in the stimulation of mesolimbic dopamine activity by ethanol and morphine in Long-Evans rats: a delayed effect of ethanol. Psychopharmacology 228, 389–400 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  231. Ascher, J. A. et al. Bupropion: a review of its mechanism of antidepressant activity. J. Clin. Psychiatry 56, 395–401 (1995).

    CAS  PubMed  Google Scholar 

  232. Fava, M. et al. 15 years of clinical experience with bupropion HCl: from bupropion to bupropion SR to bupropion XL. Prim. Care Companion J. Clin. Psychiatry 7, 106–113 (2005).

    Article  PubMed  PubMed Central  Google Scholar 

  233. Aboujaoude, E. & Salame, W. O. Naltrexone: a pan-addiction treatment? CNS Drugs 30, 719–733 (2016).

    Article  CAS  PubMed  Google Scholar 

  234. Cabrera, E. A. et al. Neuroimaging the effectiveness of substance use disorder treatments. J. Neuroimmune Pharmacol. 11, 408–433 (2016).

    Article  PubMed  Google Scholar 

  235. Fortin, S. M. & Roitman, M. F. Central GLP-1 receptor activation modulates cocaine-evoked phasic dopamine signaling in the nucleus accumbens core. Physiol. Behav. 176, 17–25 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  236. Reddy, I. A. et al. Glucagon-like peptide 1 receptor activation regulates cocaine actions and dopamine homeostasis in the lateral septum by decreasing arachidonic acid levels. Transl Psychiatry 6, e809 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  237. Sorensen, G., Caine, S. B. & Thomsen, M. Effects of the GLP-1 agonist exendin-4 on intravenous ethanol self-administration in mice. Alcohol. Clin. Exp. Res. 40, 2247–2252 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  238. Fink-Jensen, A. & Vilsboll, T. Glucagon-like peptide-1 (GLP-1) analogues: a potential new treatment for alcohol use disorder? Nord. J. Psychiatry 70, 561–562 (2016).

    Article  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Contributions

N.D.V.: researching data for article, substantial contribution to discussion of content, writing, review and editing of manuscript before submission. R.A.W.: researching data for article, substantial contribution to discussion of content, writing, review and editing of manuscript before submission. R.B.: substantial contribution to discussion of content, writing, review and editing of manuscript before submission.

Corresponding author

Correspondence to Nora D. Volkow.

Ethics declarations

Competing interests

The authors declare that they have no pertinent competing financial interests or any other conflict of interest in relation to the work described herein.

Supplementary information

Supplementary information S1 (table)

Influence of selected food-related signals on reward associated areas and behaviors (PDF 371 kb)

PowerPoint slides

Glossary

Obesity

Body weight that is above what is considered a healthy weight for a given height, normally ascertained through the screening tool referred to as the body mass index (BMI). Obesity (BMI >30) is associated with increased risk of illness, disability and death.

Relapse

Spontaneous recurrence or reinstatement of a learned behaviour after a given period of extinction, such as the reinstatement of compulsive drug use or the reinstatement of eating behaviours that lead to the reversal of diet-induced weight loss.

Addiction

A chronic brain disease associated with disruption of reward and motivation, memory and conditioning, executive and self-regulation, mood and stress neurocircuitry, the risk of which implicates environmental, genetic and social factors.

Reward

The subjective salience value of an object, stimulus or situation that has the potential to induce goal-oriented behaviour.

Motivation

A brain process triggered by intrinsic and/or extrinsic drivers that induce an animal or a person to move towards a goal.

Reinforcer

An event or stimulus that, once delivered, increases the probability of repeating the act that it follows; this term can apply to both food and drugs. Painful and aversive stimuli can also act as reinforcers but instead they increase the probability of avoiding the behaviours or circumstances that preceded the stimuli. Novel stimuli can also act as reinforcers by engaging attentional systems.

Tonic

Slow and gradual. Receptors activated by a tonic input typically adapt slowly throughout the stimulation period, conveying information about its duration.

Phasic

Sudden and transient, conveying information about sudden changes in stimulus intensity and rate and promoting rapid adaptation to the stimulus.

Striatum

A key region of the limbic system, dysfunctions of which have been associated with the pathophysiology of addiction and obesity.

Dorsal striatum

A region of the striatum associated with habits or stimulus–response learning.

Ventral tegmental area

(VTA). A cluster of midbrain neurons that sends dopaminergic projections to both limbic and cortical areas, thus playing a central role in reward-related and goal-directed behaviours. Note that while it has been traditionally believed that the VTA underlies reinforcement, recent optogenetic studies indicate that the SN also participates in this phenomenon.

Ventral striatum

A region of the striatum that contains the nucleus accumbens and is predominantly associated with reward and motivation.

Substantia nigra

(SN). A cluster of midbrain dopamine neurons that is predominantly associated with movement and involved in habit formation. More recent optogenetic studies also implicate it in reward functions.

Medium spiny neurons

A GABAergic striatal cell type of critical importance because of its pivotal roles not only in motor control, habituation and motivated behaviour but also in psychiatric disorders such as Parkinson disease, Huntington disease, schizophrenia and addiction.

Direct striatocortical pathway

Striatal pathway in which D1R-expressing (striatonigral projection) medium spiny neurons project from the striatum to the internal globus pallidum and the substantia nigra reticulata, which disinhibit thalamic excitatory neurons to the frontal cortex, facilitating movement.

Indirect striatocortical pathway

Striatal pathway in which D2R-expressing (striatopallidal projection) medium spiny neurons project from the striatum to the external globus pallidum and then to the subthalamic nucleus, which then projects into the internal pallidum and substantia nigra reticulata with a resultant inhibition of thalamic stimulation of the frontal cortex, inhibiting movement.

Heteromers

Receptors consisting of dimers and possibly higher-order entities with unique biochemical and functional characteristics, composed of different monomers from the same or different gene families.

Satiety

A feeling that follows food intake and that leads to meal termination. It is a complex psychological construct that can be linked to physicochemical measures related to stomach distention, blood levels of peptides such as cholecystokinin and glucagon-like peptide-1, peripheral biomarkers associated with meal termination, and neural activity related to sensory-stimulus-specific satiety.

Compulsive

Related to an uncontrollable, often unconscious urge to perform a specific act, often in a repetitive fashion.

Optogenetic stimulation and inhibition

A technique that allows the use of an external source of monochromatic light to stimulate or inhibit the activity of cells reversibly and with a high degree of spatiotemporal resolution (typically applied to selected neuronal populations that have been genetically modified to express light-sensitive ion channels).

Gut microbiome

The diverse collection of symbiotic microorganisms (flora) residing in the gastrointestinal tract that perform structural and histological functions and play significant roles in the regulation of host health maintenance and homeostasis.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Volkow, N., Wise, R. & Baler, R. The dopamine motive system: implications for drug and food addiction. Nat Rev Neurosci 18, 741–752 (2017). https://doi.org/10.1038/nrn.2017.130

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrn.2017.130

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing