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.

  • Review Article
  • Published:

Fractionating impulsivity: neuropsychiatric implications

Key Points

  • Although also being a characteristic of normal behaviour, excessive impulsivity is an important symptom of several neuropsychiatric and neurological disorders, including addiction, attention-deficit hyperactivity disorder and Parkinson disease.

  • However, impulsivity may comprise several apparently related forms that depend on distinct neuropsychological processes and neural systems. Of particular importance are interactions between frontostriatal systems and their neurochemical modulation, which are also providing new insights into the functions of these systems in behaviour.

  • The psychological and neural basis of impulsivity can be studied in a mutually profitable way in experimental animals and in humans. Striking parallels can be observed in the underlying neurobehavioural systems, allowing both macro- and micro-definition of functional circuits.

  • The dissection of impulsivity in this Review may represent a way in which complex behaviour relevant to psychiatric disorders can be broken down into its constituent parts, thus allowing for improved genetic understanding and more-precise treatments at the level of symptoms rather than according to categorical diagnoses of mental health disorders.

Abstract

The ability to make decisions and act quickly without hesitation can be advantageous in many settings. However, when persistently expressed, impulsive decisions and actions are considered risky, maladaptive and symptomatic of such diverse brain disorders as attention-deficit hyperactivity disorder, drug addiction and affective disorders. Over the past decade, rapid progress has been made in the identification of discrete neural networks that underlie different forms of impulsivity — from impaired response inhibition and risky decision making to a profound intolerance of delayed rewards. Herein, we review what is currently known about the neural and psychological mechanisms of impulsivity, and discuss the relevance and application of these new insights to various neuropsychiatric disorders.

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

Access options

Buy this article

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

Figure 1: Translatable experimental paradigms to assess impulsivity in rodents and humans.
Figure 2: Distinct loci of 'stopping' and 'waiting' impulsivity in the dorsal and ventral striatum.
Figure 3: Topographical organization of the corticostriatal circuitry and associated impulsivity constructs in humans.

Similar content being viewed by others

References

  1. Moeller, F. G. in The Oxford Handbook of Impulse Control Disorders (eds Grant, J. E. & Potenza, M. N.) 11–21 (Oxford Univ. Press, 2012).

    Google Scholar 

  2. Mischel, W., Shoda, Y. & Rodriguez, M. I. Delay of gratification in children. Science 244, 933–938 (1989).

    Article  CAS  PubMed  Google Scholar 

  3. Casey, B. J. et al. Behavioral and neural correlates of delay of gratification 40 years later. Proc. Natl Acad. Sci. USA 108, 14998–15003 (2011). This article provides an astonishing demonstration of the predictive capability of behaviour in early life for subsequent adult outcomes.

    Article  CAS  PubMed  Google Scholar 

  4. Cuthbert, B. N. & Insel, T. R. Toward the future of psychiatric diagnosis: the seven pillars of RDoC. BMC Med. 11, 126 (2013). This is a description of the controversial Research Domain Criteria alternative approach to psychiatric classification of mental illness that is based more firmly on advances in behavioural and cognitive neuroscience than on categorical diagnosis.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Patton, J. H., Stanford, M. S. & Barratt, E. S. Factor structure of the Barratt Impulsiveness Scale. J. Clin. Psychol. 51, 768–774 (1995).

    Article  CAS  PubMed  Google Scholar 

  6. Ainslie, G. Specious reward: a behavioral theory of impulsiveness and impulse control. Psychol. Bull. 82, 463–496 (1975).

    Article  CAS  PubMed  Google Scholar 

  7. Robbins, T. W. The 5-choice serial reaction time task: behavioural pharmacology and functional neurochemistry. Psychopharmacology (Berl.) 163, 362–380 (2002).

    Article  CAS  Google Scholar 

  8. Tecce, J. J. Contingent negative variation (CNV) and psychological processes in man. Psychol. Bull. 77, 73–108 (1972).

    Article  CAS  PubMed  Google Scholar 

  9. Kagan, J. Reflection-impulsivity: the generality and dynamics of conceptual tempo. J. Abnorm. Psychol. 71, 17–24 (1966).

    Article  CAS  PubMed  Google Scholar 

  10. Logan, G. D., Van Zandt, T., Verbruggen, F. & Wagenmakers, E. J. On the ability to inhibit thought and action: general and special theories of an act of control. Psychol. Rev. 121, 66–95 (2014). This is an example of the sophisticated psychological investigation of mechanisms underlying certain forms of impulse control.

    Article  PubMed  Google Scholar 

  11. Green, L. & Myerson, J. How many impulsivities? A discounting perspective. J. Exp. Anal. Behav. 99, 3–13 (2013).

    Article  PubMed  Google Scholar 

  12. Rangel, A., Camerer, C. & Montague, P. R. A framework for studying the neurobiology of value-based decision making. Nat. Rev. Neurosci. 9, 545–556 (2008). This review provides a brilliant theoretical synthesis of neuroeconomics and reinforcement learning theory in the service of understanding decision making and its neural correlates.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Dalley, J. W., Everitt, B. J. & Robbins, T. W. Impulsivity, compulsivity, and top-down cognitive control. Neuron 69, 680–694 (2011).

    Article  CAS  PubMed  Google Scholar 

  14. Heilbronner, S. R., Rodriguez-Romaguera, J., Quirk, G. J., Groenewegen, H. J. & Haber, S. N. Circuit-based corticostriatal homologies between rat and primate. Biol. Psychiatry 80, 509–521 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Cole, B. J. & Robbins, T. W. Effects of 6-hydroxydopamine lesions of the nucleus accumbens septi on performance of a 5-choice serial reaction time task in rats: implications for theories of selective attention and arousal. Behav. Brain Res. 33, 165–179 (1989).

    Article  CAS  PubMed  Google Scholar 

  16. Dalley, J. W. et al. Nucleus accumbens D2/3 receptors predict trait impulsivity and cocaine reinforcement. Science 315, 1267–1270 (2007). This article offers an experimental demonstration of how animal-based investigations of neurobehavioural syndromes may have relevance for understanding causal mechanisms in complex human disorders such as addiction.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Cardinal, R. N., Pennicott, D. R., Sugathapala, C. L., Robbins, T. W. & Everitt, B. J. Impulsive choice induced in rats by lesions of the nucleus accumbens core. Science 292, 2499–2501 (2001).

    Article  CAS  PubMed  Google Scholar 

  18. Basar, K. et al. Nucleus accumbens and impulsivity. Prog. Neurobiol. 92, 533–557 (2010).

    Article  PubMed  Google Scholar 

  19. Diergaarde, L. et al. Impulsive choice and impulsive action predict vulnerability to distinct stages of nicotine seeking in rats. Biol. Psychiatry 63, 301–308 (2008).

    Article  CAS  PubMed  Google Scholar 

  20. Jupp, B. et al. Dopaminergic and GABA-ergic markers of impulsivity in rats: evidence for anatomical localisation in ventral striatum and prefrontal cortex. Eur. J. Neurosci. 37, 1519–1528 (2013).

    Article  PubMed  Google Scholar 

  21. Besson, M. et al. Dissociable control of impulsivity in rats by dopamine D2/3 receptors in the core and shell subregions of the nucleus accumbens. Neuropsychopharmacology 35, 560–569 (2010).

    Article  CAS  PubMed  Google Scholar 

  22. Murphy, E. R., Robinson, E. S., Theobald, D. E., Dalley, J. W. & Robbins, T. W. Contrasting effects of selective lesions of nucleus accumbens core or shell on inhibitory control and amphetamine-induced impulsive behaviour. Eur. J. Neurosci. 28, 353–363 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Feja, M., Hayn, L. & Koch, M. Nucleus accumbens core and shell inactivation differentially affects impulsive behaviours in rats. Prog. Neuropsychopharmacol. Biol. Psychiatry 54, 31–42 (2014).

    Article  PubMed  Google Scholar 

  24. Sesia, T. et al. Deep brain stimulation of the nucleus accumbens core and shell: opposite effects on impulsive action. Exp. Neurol. 214, 135–139 (2008).

    Article  PubMed  Google Scholar 

  25. Sawiak, S. J. et al. In vivo γ-aminobutyric acid measurement in rats with spectral editing at 4.7T. J. Magn. Reson. Imaging 43, 1308–1312 (2016).

    Article  PubMed  Google Scholar 

  26. Caprioli, D. et al. Gamma aminobutyric acidergic and neuronal structural markers in the nucleus accumbens core underlie trait-like impulsive behavior. Biol. Psychiatry 75, 115–123 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Haber, S. N., Fudge, J. L. & McFarland, N. R. Striatonigrostriatal pathways in primates form an ascending spiral from the shell to the dorsolateral striatum. J. Neurosci. 20, 2369–2382 (2000).

    Article  CAS  PubMed  Google Scholar 

  28. Baunez, C. & Robbins, T. W. Bilateral lesions of the subthalamic nucleus induce multiple deficits in an attentional task in rats. Eur. J. Neurosci. 9, 2086–2099 (1997).

    Article  CAS  PubMed  Google Scholar 

  29. Buckholtz, J. W. et al. Dopaminergic network differences in human impulsivity. Science 329, 532 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Robinson, E. S. et al. Behavioural characterisation of high impulsivity on the 5-choice serial reaction time task: specific deficits in 'waiting' versus 'stopping'. Behav. Brain Res. 196, 310–316 (2009).

    Article  CAS  PubMed  Google Scholar 

  31. Cardinal, R. N., Robbins, T. W. & Everitt, B. J. The effects of d-amphetamine, chlordiazepoxide, α-flupenthixol and behavioural manipulations on choice of signalled and unsignalled delayed reinforcement in rats. Psychopharmacology (Berl.) 152, 362–375 (2000).

    Article  CAS  Google Scholar 

  32. Winstanley, C. A., Dalley, J. W., Theobald, D. E. & Robbins, T. W. Global 5-HT depletion attenuates the ability of amphetamine to decrease impulsive choice on a delay-discounting task in rats. Psychopharmacology (Berl.) 170, 320–331 (2003).

    Article  CAS  Google Scholar 

  33. Miyazaki, K. W. et al. Optogenetic activation of dorsal raphe serotonin neurons enhances patience for future rewards. Curr. Biol. 24, 2033–2040 (2014).

    Article  CAS  PubMed  Google Scholar 

  34. Tedford, S. E., Persons, A. L. & Napier, T. C. Dopaminergic lesions of the dorsolateral striatum in rats increase delay discounting in an impulsive choice task. PLoS ONE 10, e0122063 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Winstanley, C. A., Baunez, C., Theobald, D. E. & Robbins, T. W. Lesions to the subthalamic nucleus decrease impulsive choice but impair autoshaping in rats: the importance of the basal ganglia in Pavlovian conditioning and impulse control. Eur. J. Neurosci. 21, 3107–3116 (2005).

    Article  PubMed  Google Scholar 

  36. Eagle, D. M. et al. Stop-signal reaction-time task performance: role of prefrontal cortex and subthalamic nucleus. Cereb. Cortex 18, 178–188 (2008).

    Article  PubMed  Google Scholar 

  37. Cardinal, R. N. & Howes, N. J. Effects of lesions of the nucleus accumbens core on choice between small certain rewards and large uncertain rewards in rats. BMC Neurosci. 6, 37 (2005).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Stopper, C. M. & Floresco, S. B. Contributions of the nucleus accumbens and its subregions to different aspects of risk-based decision making. Cogn. Affect. Behav. Neurosci. 11, 97–112 (2011).

    Article  PubMed  Google Scholar 

  39. Stopper, C. M., Khayambashi, S. & Floresco, S. B. Receptor-specific modulation of risk-based decision making by nucleus accumbens dopamine. Neuropsychopharmacology 38, 715–728 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Cocker, P. J., Dinelle, K., Kornelson, R., Sossi, V. & Winstanley, C. A. Irrational choice under uncertainty correlates with lower striatal D2/3 receptor binding in rats. J. Neurosci. 32, 15450–15457 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Zalocusky, K. A. et al. Nucleus accumbens D2R cells signal prior outcomes and control risky decision-making. Nature 531, 642–646 (2016). This article offers a glimpse into the future of how new neuroscience tools such as optogenetics can be used to define specific functions of neural circuitry in experimental animals of relevance to human clinical disorders.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Eagle, D. M. & Robbins, T. W. Lesions of the medial prefrontal cortex or nucleus accumbens core do not impair inhibitory control in rats performing a stop-signal reaction time task. Behav. Brain Res. 146, 131–144 (2003).

    Article  CAS  PubMed  Google Scholar 

  43. Eagle, D. M. et al. Contrasting roles for dopamine D1 and D2 receptor subtypes in the dorsomedial striatum but not the nucleus accumbens core during behavioral inhibition in the stop-signal task in rats. J. Neurosci. 31, 7349–7356 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Robertson, C. L. et al. Striatal D1- and D2-type dopamine receptors are linked to motor response inhibition in human subjects. J. Neurosci. 35, 5990–5997 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Chudasama, Y. et al. Dissociable aspects of performance on the 5-choice serial reaction time task following lesions of the dorsal anterior cingulate, infralimbic and orbitofrontal cortex in the rat: differential effects on selectivity, impulsivity and compulsivity. Behav. Brain Res. 146, 105–119 (2003).

    Article  CAS  PubMed  Google Scholar 

  46. Murphy, E. R., Dalley, J. W. & Robbins, T. W. Local glutamate receptor antagonism in the rat prefrontal cortex disrupts response inhibition in a visuospatial attentional task. Psychopharmacology (Berl.) 179, 99–107 (2005).

    Article  CAS  Google Scholar 

  47. Abela, A. R., Dougherty, S. D., Fagen, E. D., Hill, C. J. & Chudasama, Y. Inhibitory control deficits in rats with ventral hippocampal lesions. Cereb. Cortex 23, 1396–1409 (2013).

    Article  PubMed  Google Scholar 

  48. Belin-Rauscent, A. et al. From impulses to maladaptive actions: the insula is a neurobiological gate for the development of compulsive behavior. Mol. Psychiatry 21, 491–499 (2016).

    Article  CAS  PubMed  Google Scholar 

  49. Donnelly, N. A., Paulsen, O., Robbins, T. W. & Dalley, J. W. Ramping single unit activity in the medial prefrontal cortex and ventral striatum reflects the onset of waiting but not imminent impulsive actions. Eur. J. Neurosci. 41, 1524–1537 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Muir, J. L., Everitt, B. J. & Robbins, T. W. The cerebral cortex of the rat and visual attentional function: dissociable effects of mediofrontal, cingulate, anterior dorsolateral, and parietal cortex lesions on a five-choice serial reaction time task. Cereb. Cortex 6, 470–481 (1996).

    Article  CAS  PubMed  Google Scholar 

  51. Dalley, J. W., Theobald, D. E., Eagle, D. M., Passetti, F. & Robbins, T. W. Deficits in impulse control associated with tonically-elevated serotonergic function in rat prefrontal cortex. Neuropsychopharmacology 26, 716–728 (2002).

    Article  CAS  PubMed  Google Scholar 

  52. Winstanley, C. A., Theobald, D. E., Cardinal, R. N. & Robbins, T. W. Contrasting roles of basolateral amygdala and orbitofrontal cortex in impulsive choice. J. Neurosci. 24, 4718–4722 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Cheung, T. H. & Cardinal, R. N. Hippocampal lesions facilitate instrumental learning with delayed reinforcement but induce impulsive choice in rats. BMC Neurosci. 6, 36 (2005).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Abela, A. R. & Chudasama, Y. Dissociable contributions of the ventral hippocampus and orbitofrontal cortex to decision-making with a delayed or uncertain outcome. Eur. J. Neurosci. 37, 640–647 (2013).

    Article  PubMed  Google Scholar 

  55. Mobini, S. et al. Effects of lesions of the orbitofrontal cortex on sensitivity to delayed and probabilistic reinforcement. Psychopharmacology (Berl.) 160, 290–298 (2002).

    Article  CAS  Google Scholar 

  56. Kheramin, S. et al. Effects of quinolinic acid-induced lesions of the orbital prefrontal cortex on inter-temporal choice: a quantitative analysis. Psychopharmacology (Berl.) 165, 9–17 (2002).

    Article  CAS  Google Scholar 

  57. Kheramin, S. et al. Effects of orbital prefrontal cortex dopamine depletion on inter-temporal choice: a quantitative analysis. Psychopharmacology (Berl.) 175, 206–214 (2004).

    Article  CAS  Google Scholar 

  58. Rudebeck, P. H., Walton, M. E., Smyth, A. N., Bannerman, D. M. & Rushworth, M. F. Separate neural pathways process different decision costs. Nat. Neurosci. 9, 1161–1168 (2006).

    Article  CAS  PubMed  Google Scholar 

  59. Zeeb, F. D., Floresco, S. B. & Winstanley, C. A. Contributions of the orbitofrontal cortex to impulsive choice: interactions with basal levels of impulsivity, dopamine signalling, and reward-related cues. Psychopharmacology (Berl.) 211, 87–98 (2010).

    Article  CAS  Google Scholar 

  60. Mar, A. C., Walker, A. L., Theobald, D. E., Eagle, D. M. & Robbins, T. W. Dissociable effects of lesions to orbitofrontal cortex subregions on impulsive choice in the rat. J. Neurosci. 31, 6398–6404 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Schoenbaum, G., Setlow, B. & Ramus, S. J. A systems approach to orbitofrontal cortex function: recordings in rat orbitofrontal cortex reveal interactions with different learning systems. Behav. Brain Res. 146, 19–29 (2003).

    Article  PubMed  Google Scholar 

  62. Stopper, C. M., Green, E. B. & Floresco, S. B. Selective involvement by the medial orbitofrontal cortex in biasing risky, but not impulsive, choice. Cereb. Cortex 24, 154–162 (2014).

    Article  PubMed  Google Scholar 

  63. St Onge, J. R. & Floresco, S. B. Prefrontal cortical contribution to risk-based decision making. Cereb. Cortex 20, 1816–1828 (2010).

    Article  PubMed  Google Scholar 

  64. St Onge, J. R., Stopper, C. M., Zahm, D. S. & Floresco, S. B. Separate prefrontal-subcortical circuits mediate different components of risk-based decision making. J. Neurosci. 32, 2886–2899 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Stopper, C. M. & Floresco, S. B. What's better for me? Fundamental role for lateral habenula in promoting subjective decision biases. Nat. Neurosci. 17, 33–35 (2014).

    Article  CAS  PubMed  Google Scholar 

  66. Luchicchi, A. et al. Sustained attentional states require distinct temporal involvement of the dorsal and ventral medial prefrontal cortex. Front. Neural Circuits 10, 70 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Passetti, F., Chudasama, Y. & Robbins, T. W. The frontal cortex of the rat and visual attentional performance: dissociable functions of distinct medial prefrontal subregions. Cereb. Cortex 12, 1254–1268 (2002).

    Article  PubMed  Google Scholar 

  68. Koike, H. et al. Chemogenetic inactivation of dorsal anterior cingulate cortex neurons disrupts attentional behavior in mouse. Neuropsychopharmacology 41, 1014–1023 (2016).

    Article  CAS  PubMed  Google Scholar 

  69. Boekhoudt, L. et al. Chemogenetic activation of midbrain dopamine neurons affects attention, but not impulsivity, in the five-choice serial reaction time task in rats. Neuropsychopharmacology http:dx.doi.org/10.1038/npp.2016.235 (2016).

  70. Voon, V. et al. Measuring “waiting” impulsivity in substance addictions and binge eating disorder in a novel analogue of rodent serial reaction time task. Biol. Psychiatry 75, 148–155 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  71. Morris, L. S. et al. Jumping the gun: mapping neural correlates of waiting impulsivity and relevance across alcohol misuse. Biol. Psychiatry 79, 499–507 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  72. Bari, A. & Robbins, T. W. Inhibition and impulsivity: behavioral and neural basis of response control. Prog. Neurobiol. 108, 44–79 (2013).

    Article  PubMed  Google Scholar 

  73. Aron, A. R., Fletcher, P. C., Bullmore, E. T., Sahakian, B. J. & Robbins, T. W. Stop-signal inhibition disrupted by damage to right inferior frontal gyrus in humans. Nat. Neurosci. 6, 115–116 (2003).

    Article  CAS  PubMed  Google Scholar 

  74. Aron, A. R. & Poldrack, R. A. Cortical and subcortical contributions to Stop signal response inhibition: role of the subthalamic nucleus. J. Neurosci. 26, 2424–2433 (2006).

    Article  CAS  PubMed  Google Scholar 

  75. Whelan, R. et al. Adolescent impulsivity phenotypes characterized by distinct brain networks. Nat. Neurosci. 15, 920–925 (2012). This is a multidisciplinary study of impulsivity using functional neuroimaging and genetics of 2,000 healthy adolescents screened for early drug use and ADHD characteristics.

    Article  CAS  PubMed  Google Scholar 

  76. Aron, A. R., Robbins, T. W. & Poldrack, R. A. Inhibition and the right inferior frontal cortex: one decade on. Trends Cogn. Sci. 18, 177–185 (2014).

    Article  PubMed  Google Scholar 

  77. Cai, W., Ryali, S., Chen, T., Li, C. S. & Menon, V. Dissociable roles of right inferior frontal cortex and anterior insula in inhibitory control: evidence from intrinsic and task-related functional parcellation, connectivity, and response profile analyses across multiple datasets. J. Neurosci. 34, 14652–14667 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Dodds, C. M., Morein-Zamir, S. & Robbins, T. W. Dissociating inhibition, attention, and response control in the frontoparietal network using functional magnetic resonance imaging. Cereb. Cortex 21, 1155–1165 (2011).

    Article  PubMed  Google Scholar 

  79. Rae, C. L., Hughes, L. E., Anderson, M. C. & Rowe, J. B. The prefrontal cortex achieves inhibitory control by facilitating subcortical motor pathway connectivity. J. Neurosci. 35, 786–794 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Aron, A. R. From reactive to proactive and selective control: developing a richer model for stopping inappropriate responses. Biol. Psychiatry 69, e55–e68 (2011).

    Article  PubMed  Google Scholar 

  81. Kable, J. W. & Glimcher, P. W. The neural correlates of subjective value during intertemporal choice. Nat. Neurosci. 10, 1625–1633 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. McClure, S. M., Laibson, D. I., Loewenstein, G. & Cohen, J. D. Separate neural systems value immediate and delayed monetary rewards. Science 306, 503–507 (2004).

    Article  CAS  PubMed  Google Scholar 

  83. Ballard, K. & Knutson, B. Dissociable neural representations of future reward magnitude and delay during temporal discounting. Neuroimage 45, 143–150 (2009).

    Article  PubMed  Google Scholar 

  84. Tanaka, S. C. et al. Prediction of immediate and future rewards differentially recruits cortico-basal ganglia loops. Nat. Neurosci. 7, 887–893 (2004).

    Article  CAS  PubMed  Google Scholar 

  85. Huettel, S. A., Stowe, C. J., Gordon, E. M., Warner, B. T. & Platt, M. L. Neural signatures of economic preferences for risk and ambiguity. Neuron 49, 765–775 (2006).

    Article  CAS  PubMed  Google Scholar 

  86. Pattij, T. & Vanderschuren, L. J. The neuropharmacology of impulsive behaviour. Trends Pharmacol. Sci. 29, 192–199 (2008).

    Article  CAS  PubMed  Google Scholar 

  87. Del Campo, N., Chamberlain, S. R., Sahakian, B. J. & Robbins, T. W. The roles of dopamine and noradrenaline in the pathophysiology and treatment of attention-deficit/hyperactivity disorder. Biol. Psychiatry 69, e145–e157 (2011).

    Article  CAS  PubMed  Google Scholar 

  88. Caprioli, D. et al. Dissociable rate-dependent effects of oral methylphenidate on impulsivity and D2/3 receptor availability in the striatum. J. Neurosci. 35, 3747–3755 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Robinson, E. S. et al. Similar effects of the selective noradrenaline reuptake inhibitor atomoxetine on three distinct forms of impulsivity in the rat. Neuropsychopharmacology 33, 1028–1037 (2008).

    Article  CAS  PubMed  Google Scholar 

  90. Economidou, D., Theobald, D. E., Robbins, T. W., Everitt, B. J. & Dalley, J. W. Norepinephrine and dopamine modulate impulsivity on the five-choice serial reaction time task through opponent actions in the shell and core sub-regions of the nucleus accumbens. Neuropsychopharmacology 37, 2057–2066 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Bari, A. et al. Prefrontal and monoaminergic contributions to stop-signal task performance in rats. J. Neurosci. 31, 9254–9263 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Chamberlain, S. R. et al. Atomoxetine modulates right inferior frontal activation during inhibitory control: a pharmacological functional magnetic resonance imaging study. Biol. Psychiatry 65, 550–555 (2009).

    Article  CAS  PubMed  Google Scholar 

  93. Rubia, K. et al. Effects of stimulants on brain function in attention-deficit/hyperactivity disorder: a systematic review and meta-analysis. Biol. Psychiatry 76, 616–628 (2014). This is a recent valuable synthesis of investigations of mechanisms underlying therapeutic effects of amphetamine-like drugs in ADHD.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Nagashima, M. et al. Acute neuropharmacological effects of atomoxetine on inhibitory control in ADHD children: a fNIRS study. Neuroimage Clin. 6, 192–201 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  95. Kehagia, A. A. et al. Targeting impulsivity in Parkinson's disease using atomoxetine. Brain 137, 1986–1997 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  96. Ye, Z. et al. Improving response inhibition in Parkinson's disease with atomoxetine. Biol. Psychiatry 77, 740–748 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Harrison, A. A., Everitt, B. J. & Robbins, T. W. Central 5-HT depletion enhances impulsive responding without affecting the accuracy of attentional performance: interactions with dopaminergic mechanisms. Psychopharmacology (Berl.) 133, 329–342 (1997).

    Article  CAS  Google Scholar 

  98. Worbe, Y., Savulich, G., Voon, V., Fernandez-Egea, E. & Robbins, T. W. Serotonin depletion induces 'waiting impulsivity' on the human four-choice serial reaction time task: cross-species translational significance. Neuropsychopharmacology 39, 1519–1526 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  99. Winstanley, C. A. et al. Intra-prefrontal 8-OH-DPAT and M100907 improve visuospatial attention and decrease impulsivity on the five-choice serial reaction time task in rats. Psychopharmacology (Berl.) 167, 304–314 (2003).

    Article  CAS  Google Scholar 

  100. Robinson, E. S. et al. Opposing roles for 5-HT2A and 5-HT2C receptors in the nucleus accumbens on inhibitory response control in the 5-choice serial reaction time task. Neuropsychopharmacology 33, 2398–2406 (2008).

    Article  CAS  PubMed  Google Scholar 

  101. Winstanley, C. A., Theobald, D. E., Dalley, J. W., Glennon, J. C. & Robbins, T. W. 5-HT2A and 5-HT2C receptor antagonists have opposing effects on a measure of impulsivity: interactions with global 5-HT depletion. Psychopharmacology (Berl.) 176, 376–385 (2004).

    Article  CAS  Google Scholar 

  102. Miyazaki, K., Miyazaki, K. W. & Doya, K. Activation of dorsal raphé serotonin neurons underlies waiting for delayed rewards. J. Neurosci. 31, 469–479 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Eagle, D. M. et al. Serotonin depletion impairs waiting but not stop-signal reaction time in rats: implications for theories of the role of 5-HT in behavioral inhibition. Neuropsychopharmacology 34, 1311–1321 (2009).

    Article  CAS  PubMed  Google Scholar 

  104. Chamberlain, S. R. et al. Neurochemical modulation of response inhibition and probabilistic learning in humans. Science 311, 861–863 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Bari, A., Eagle, D. M., Mar, A. C., Robinson, E. S. & Robbins, T. W. Dissociable effects of noradrenaline, dopamine, and serotonin uptake blockade on stop task performance in rats. Psychopharmacology (Berl.) 205, 273–283 (2009).

    Article  CAS  Google Scholar 

  106. Ye, Z. et al. Selective serotonin reuptake inhibition modulates response inhibition in Parkinson's disease. Brain 137, 1145–1155 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  107. Winstanley, C. A., Olausson, P., Taylor, J. R. & Jentsch, J. D. Insight into the relationship between impulsivity and substance abuse from studies using animal models. Alcohol Clin. Exp. Res. 34, 1306–1318 (2010).

    PubMed  PubMed Central  Google Scholar 

  108. Perry, J. L. & Carroll, M. E. The role of impulsive behavior in drug abuse. Psychopharmacology (Berl.) 200, 1–26 (2008).

    Article  CAS  Google Scholar 

  109. Morein-Zamir, S., Simon Jones, P., Bullmore, E. T., Robbins, T. W. & Ersche, K. D. Prefrontal hypoactivity associated with impaired inhibition in stimulant-dependent individuals but evidence for hyperactivation in their unaffected siblings. Neuropsychopharmacology 38, 1945–1953 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  110. Ersche, K. D. et al. Abnormal brain structure implicated in stimulant drug addiction. Science 335, 601–604 (2012).

    Article  CAS  PubMed  Google Scholar 

  111. Whelan, R. et al. Neuropsychosocial profiles of current and future adolescent alcohol misusers. Nature 512, 185–189 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Holmes, A. J., Hollinshead, M. O., Roffman, J. L., Smoller, J. W. & Buckner, R. L. Individual differences in cognitive control circuit anatomy link sensation seeking, impulsivity, and substance use. J. Neurosci. 36, 4038–4049 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Ersche, K. D., Williams, G. B., Robbins, T. W. & Bullmore, E. T. Meta-analysis of structural brain abnormalities associated with stimulant drug dependence and neuroimaging of addiction vulnerability and resilience. Curr. Opin. Neurobiol. 23, 615–624 (2013).

    Article  CAS  PubMed  Google Scholar 

  114. Everitt, B. J. & Robbins, T. W. Drug addiction: updating actions to habits to compulsions ten years on. Annu. Rev. Psychol. 67, 23–50 (2016).

    Article  PubMed  Google Scholar 

  115. Belin, D., Mar, A. C., Dalley, J. W., Robbins, T. W. & Everitt, B. J. High impulsivity predicts the switch to compulsive cocaine-taking. Science 320, 1352–1355 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Deserno, L. et al. Lateral prefrontal model-based signatures are reduced in healthy individuals with high trait impulsivity. Transl Psychiatry 5, e659 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Solanto, M. V. et al. The ecological validity of delay aversion and response inhibition as measures of impulsivity in AD/HD: a supplement to the NIMH multimodal treatment study of AD/HD. J. Abnorm. Child Psychol. 29, 215–228 (2001).

    Article  CAS  PubMed  Google Scholar 

  118. Dambacher, F. et al. Out of control: evidence for anterior insula involvement in motor impulsivity and reactive aggression. Soc. Cogn. Affect Neurosci. 10, 508–516 (2015).

    Article  PubMed  Google Scholar 

  119. Churchwell, J. C. & Yurgelun-Todd, D. A. Age-related changes in insula cortical thickness and impulsivity: significance for emotional development and decision-making. Dev. Cogn. Neurosci. 6, 80–86 (2013).

    Article  PubMed  Google Scholar 

  120. Damasio, A. Feelings of emotion and the self. Ann. NY Acad. Sci. 1001, 253–261 (2003).

    Article  PubMed  Google Scholar 

  121. Giovannoni, G., O'Sullivan, J. D., Turner, K., Manson, A. J. & Lees, A. J. Hedonistic homeostatic dysregulation in patients with Parkinson's disease on dopamine replacement therapies. J. Neurol. Neurosurg. Psychiatry 68, 423–428 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Frank, M. J., Seeberger, L. C. & O'Reilly, R. C. By carrot or by stick: cognitive reinforcement learning in parkinsonism. Science 306, 1940–1943 (2004).

    Article  CAS  PubMed  Google Scholar 

  123. Clark, L., Robbins, T. W., Ersche, K. D. & Sahakian, B. J. Reflection impulsivity in current and former substance users. Biol. Psychiatry 60, 515–522 (2006).

    Article  CAS  PubMed  Google Scholar 

  124. Evenden, J. L. Varieties of impulsivity. Psychopharmacology (Berl.) 146, 348–361 (1999). This early influential review helped to begin the analysis of different forms of impulsivity.

    Article  CAS  Google Scholar 

  125. Crockett, M. J. et al. Restricting temptations: neural mechanisms of precommitment. Neuron 79, 391–401 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Jupp, B. & Dalley, J. W. in Animal Models of Behavior Genetics (eds Gewirtz, J. & Kim, Y.-K.) (Springer, 2016).

    Google Scholar 

  127. Bevilacqua, L. & Goldman, D. Genes and addictions. Clin. Pharmacol. Ther. 85, 359–361 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Franke, B. et al. The genetics of attention deficit/hyperactivity disorder in adults, a review. Mol. Psychiatry 17, 960–987 (2012).

    Article  CAS  PubMed  Google Scholar 

  129. Bezdjian, S., Baker, L. A. & Tuvblad, C. Genetic and environmental influences on impulsivity: a meta-analysis of twin, family and adoption studies. Clin. Psychol. Rev. 31, 1209–1223 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  130. Le Foll, B., Gallo, A., Le Strat, Y., Lu, L. & Gorwood, P. Genetics of dopamine receptors and drug addiction: a comprehensive review. Behav. Pharmacol. 20, 1–17 (2009).

    Article  CAS  PubMed  Google Scholar 

  131. Cao, J. et al. Association of the HTR2A gene with alcohol and heroin abuse. Hum. Genet. 133, 357–365 (2014).

    Article  CAS  PubMed  Google Scholar 

  132. Comings, D. E. et al. Studies of the 48 bp repeat polymorphism of the DRD4 gene in impulsive, compulsive, addictive behaviors: Tourette syndrome, ADHD, pathological gambling, and substance abuse. Am. J. Med. Genet. 88, 358–368 (1999).

    Article  CAS  PubMed  Google Scholar 

  133. Wilson, D., da Silva Lobo, D. S., Tavares, H., Gentil, V. & Vallada, H. Family-based association analysis of serotonin genes in pathological gambling disorder: evidence of vulnerability risk in the 5HT-2A receptor gene. J. Mol. Neurosci. 49, 550–553 (2013).

    Article  CAS  PubMed  Google Scholar 

  134. Suda, A. et al. Dopamine D2 receptor gene polymorphisms are associated with suicide attempt in the Japanese population. Neuropsychobiology 59, 130–134 (2009).

    Article  CAS  PubMed  Google Scholar 

  135. Serretti, A. et al. HTR2C and HTR1A gene variants in German and Italian suicide attempters and completers. Am. J. Med. Genet. B Neuropsychiatr. Genet. 144B, 291–299 (2007).

    Article  CAS  PubMed  Google Scholar 

  136. Wu, J., Xiao, H., Sun, H., Zou, L. & Zhu, L. Q. Role of dopamine receptors in ADHD: a systematic meta-analysis. Mol. Neurobiol. 45, 605–620 (2012).

    Article  CAS  PubMed  Google Scholar 

  137. Zhao, A. L. et al. Association analysis of serotonin transporter promoter gene polymorphism with ADHD and related symptomatology. Int. J. Neurosci. 115, 1183–1191 (2005).

    Article  CAS  PubMed  Google Scholar 

  138. Bevilacqua, L. et al. A population-specific HTR2B stop codon predisposes to severe impulsivity. Nature 468, 1061–1066 (2010). This article describes a sophisticated genetic approach that now has to be adopted for the different forms of impulsivity (this one being especially relevant to aggression).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Colzato, L. S., van den Wildenberg, W. P., Van der Does, A. J. & Hommel, B. Genetic markers of striatal dopamine predict individual differences in dysfunctional, but not functional impulsivity. Neuroscience 170, 782–788 (2010).

    Article  CAS  PubMed  Google Scholar 

  140. Eisenberg, D. T. et al. Examining impulsivity as an endophenotype using a behavioral approach: a DRD2 TaqI A and DRD4 48-bp VNTR association study. Behav. Brain Funct. 3, 2 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Varga, G. et al. Additive effects of serotonergic and dopaminergic polymorphisms on trait impulsivity. Am. J. Med. Genet. B Neuropsychiatr. Genet. 159B, 281–288 (2012).

    Article  CAS  PubMed  Google Scholar 

  142. Limosin, F. et al. Association between dopamine receptor D3 gene BalI polymorphism and cognitive impulsiveness in alcohol-dependent men. Eur. Psychiatry 20, 304–306 (2005).

    Article  CAS  PubMed  Google Scholar 

  143. Congdon, E., Lesch, K. P. & Canli, T. Analysis of DRD4 and DAT polymorphisms and behavioral inhibition in healthy adults: implications for impulsivity. Am. J. Med. Genet. B Neuropsychiatr. Genet. 147B, 27–32 (2008).

    Article  CAS  PubMed  Google Scholar 

  144. Paloyelis, Y., Asherson, P., Mehta, M. A., Faraone, S. V. & Kuntsi, J. DAT1 and COMT effects on delay discounting and trait impulsivity in male adolescents with attention deficit/hyperactivity disorder and healthy controls. Neuropsychopharmacology 35, 2414–2426 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  145. Benko, A. et al. Significant association between the C(–1019)G functional polymorphism of the HTR1A gene and impulsivity. Am. J. Med. Genet. B Neuropsychiatr. Genet. 153B, 592–599 (2010).

    Article  CAS  PubMed  Google Scholar 

  146. Jakubczyk, A. et al. The CC genotype in HTR2A T102C polymorphism is associated with behavioral impulsivity in alcohol-dependent patients. J. Psychiatr. Res. 46, 44–49 (2012).

    Article  PubMed  Google Scholar 

  147. Bjork, J. M. et al. Serotonin 2a receptor T102C polymorphism and impaired impulse control. Am. J. Med. Genet. 114, 336–339 (2002).

    Article  PubMed  Google Scholar 

  148. Sonuga-Barke, E. J. et al. A functional variant of the serotonin transporter gene (SLC6A4) moderates impulsive choice in attention-deficit/hyperactivity disorder boys and siblings. Biol. Psychiatry 70, 230–236 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Soeiro-De-Souza, M. G., Stanford, M. S., Bio, D. S., Machado-Vieira, R. & Moreno, R. A. Association of the COMT Met158 allele with trait impulsivity in healthy young adults. Mol. Med. Rep. 7, 1067–1072 (2013).

    Article  CAS  PubMed  Google Scholar 

  150. Manuck, S. B., Flory, J. D., Ferrell, R. E., Mann, J. J. & Muldoon, M. F. A regulatory polymorphism of the monoamine oxidase-A gene may be associated with variability in aggression, impulsivity, and central nervous system serotonergic responsivity. Psychiatry Res. 95, 9–23 (2000).

    Article  CAS  PubMed  Google Scholar 

  151. Stoltenberg, S. F., Christ, C. C. & Highland, K. B. Serotonin system gene polymorphisms are associated with impulsivity in a context dependent manner. Prog. Neuropsychopharmacol. Biol. Psychiatry 39, 182–191 (2012).

    Article  CAS  PubMed  Google Scholar 

  152. Rogers, R. D. et al. Dissociable deficits in the decision-making cognition of chronic amphetamine abusers, opiate abusers, patients with focal damage to prefrontal cortex, and tryptophan-depleted normal volunteers: evidence for monoaminergic mechanisms. Neuropsychopharmacology 20, 322–339 (1999).

    Article  CAS  PubMed  Google Scholar 

  153. Logan, G. D. in Inhibitory Processes in Attention, Memory and Language (eds Dagenbach, D. & Carr, T. H.) (Academic Press, 1994).

    Google Scholar 

  154. Besson, M. et al. Cocaine modulation of frontostriatal expression of Zif268, D2, and 5-HT2c receptors in high and low impulsive rats. Neuropsychopharmacology 38, 1963–1973 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Luscher, C. & Bellone, C. Cocaine-evoked synaptic plasticity: a key to addiction? Nat. Neurosci. 11, 737–738 (2008).

    Article  CAS  PubMed  Google Scholar 

  156. Banca, P. et al. Reflection impulsivity in binge drinking: behavioural and volumetric correlates. Addict. Biol. 21, 504–515 (2016).

    Article  PubMed  Google Scholar 

  157. Voon, V. & Dalley, J. W. Translatable and back-translatable measurement of impulsivity and compulsivity: convergent and divergent processes. Curr. Top. Behav. Neurosci. 28, 53–91 (2016).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors acknowledge support from the Wellcome Trust (grant 104631/Z/14/Z), UK Medical Research Council (grants G0701500, G0802729 and G9536855) and the European Commission (IMAGEN LSHM-CT-2007-037286). The Cambridge University Behavioural and Clinical Neuroscience Institute is supported by a joint award from the Wellcome Trust (093875/Z/10/Z) and Medical Research Council (G1000183). The authors also thank L. Morris and V. Voon for the frontostriatal connectivity illustrations in figure 3.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Trevor W. Robbins.

Ethics declarations

Competing interests

T.W.R. discloses consultancy with Cambridge Cognition, Lundbeck, Otsuka and Mundipharma, as well as a research grant with Lundbeck, royalties for CANTAB and editorial honoraria with Springer-Verlag and Elsevier. J.W.D. discloses research grant support from Boehringer Ingelheim and editorial honoraria from Wiley and Sage.

PowerPoint slides

Glossary

Differential reinforcement of low rates of responding (DRL) schedules

Schedules of reinforcement of instrumental behaviour in which the animal must wait for a given time after the last reinforcer before making an instrumental response.

Sensation seeking

A type of behaviour in which individuals apparently seek certain types of experience (such as mountaineering) despite the associated risks.

5-choice serial reaction time task

(5CSRTT). A behavioural test of sustained attention; animals must detect brief signals that predict food rewards. Importantly, animals are punished for responding prematurely.

Nafadotride

A relatively selective dopamine D3 receptor antagonist.

Dopamine transporter

(DAT). A transmembrane protein that pumps dopamine from the synapse into the neuron. Some drugs (for example, cocaine, methylphenidate and amphetamine) increase synaptic dopamine levels by blocking DAT.

Amphetamine

A psychomotor stimulant drug (a catecholaminergic indirect agonist) that increases activity and arousal. It is used as an effective, although perhaps paradoxical, treatment for attention-deficit hyperactivity disorder.

Autoreceptors

Receptors found in the presynaptic neuronal membrane, at both the neuronal bodies and the terminals. Their activity negatively regulates neurotransmitter release.

Antidromic

Referring to conduction of an action potential in the opposite direction; that is, away from the axon terminal to the cell body.

Striatal indirect pathway

A striatal output pathway in which striatal medium spiny neurons project via inhibitory neurons, first to the globus pallidus externa and thence to the subthalamic nucleus, which disinhibits the substantia nigra pars reticulata–globus pallidus interna.

Striatal direct pathway

A striatal output pathway in which inhibitory neurons directly project onto the cells of the substantia nigra pars reticulata–globus pallidus interna.

Hyperdirect pathway

Direct excitatory projections from several cortical areas, including the motor cortex, premotor cortex, supplementary motor area, anterior cingulate and dorsolateral prefrontal cortex, to the subthalamic nucleus, by-passing the striatum.

Beta system

A set of brain regions, including the nucleus accumbens and medial prefrontal cortex, that are postulated to process immediate rewards and hypothetically interact functionally with the so-called delta system.

Delta system

A set of brain regions, including the dorsolateral and ventrolateral prefrontal cortex and parietal cortex, that are thought to discount rewards over longer time periods and to determine behaviour by interactions with the so-called beta system.

Endophenotypes

A term from genetic epidemiology, implying, in psychiatry, an intermediate phenotype with a possible heritable basis, present not only in patients but also in their clinically non-affected first-degree relatives.

Goal-directed

Instrumental or purposeful, conscious and volitional, and in pursuit of defined outcomes.

Habitual

Elicited automatically by stimuli in the environment without reference to a goal or outcome.

Model-free learning algorithms

Algorithms for learning that reflect immediate reinforcement learning contingencies and therefore are associated with habitual behaviour.

Conduct disorder

A mental disorder of childhood or adolescence in which violent or disruptive anti-social behaviour is the main characteristic.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dalley, J., Robbins, T. Fractionating impulsivity: neuropsychiatric implications. Nat Rev Neurosci 18, 158–171 (2017). https://doi.org/10.1038/nrn.2017.8

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrn.2017.8

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