Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Apr 1.
Published in final edited form as: Curr Opin Behav Sci. 2017 Nov 21;20:67–74. doi: 10.1016/j.cobeha.2017.11.005

Regulation of habit formation in the dorsal striatum

Melissa Malvaez 1, Kate M Wassum 1,2
PMCID: PMC5920535  NIHMSID: NIHMS921971  PMID: 29713658

Abstract

Habits are an essential and pervasive component of our daily lives that allow us to efficiently perform routine tasks. But their disruption contributes to the symptoms that underlie many psychiatric diseases. Emerging data are revealing the cellular and molecular mechanisms of habit formation in the dorsal striatum. New data suggest that in both the dorsolateral and dorsomedial striatum histone deacetylase (HDAC) activity acts as a critical negative regulator of the transcriptional processes underlying habit formation. In this review, we discuss this recent work and draw conclusions relevant to the treatment of diseases marked by maladaptive habits.


When making a decision one prospectively considers potential actions and their anticipated consequences [1,2]. This allows behavior to readily adapt when circumstances change, but is cognitively taxing. The brain has another, more resource efficient, strategy for routine behaviors: habits. Habits permit common behaviors to be executed more automatically on the basis of their past success, without thought of their consequences, freeing attention to be focused elsewhere [3,4]. The balance between the reflective, goal-directed, and reflexive, habit, systems promotes maximally adaptive and efficient behavior [5,6], but when it is disrupted can lead to the symptoms that underlie myriad psychiatric and neurodegenerative diseases [711]. Indeed, deficits in the acquisition and execution of behavioral habits are symptoms of both Huntington’s [12] and Parkinson’s disease [1315]. An overreliance on habit is associated with the various forms of compulsivity that manifest across a range of conditions [8,16], including obsessive-compulsive disorder [17,18], schizophrenia [19,20], addiction [11], alcoholism [21], and compulsive overeating [22]. Research over the past two decades has exposed the largely dissociable cortico-striatal-limbic brain circuits vital for goal-directed actions and habits (for review see: [2329]). New information is emerging on how habits are acquired and stored within these networks. This will be the focus of this review.

Diagnosing and understanding habits

Instrumental behaviors can be modeled by training subjects to perform an action (e.g., lever press, maze run, button push) to receive a reward (e.g., food). Behavioral strategy cannot, however, be determined from simple observation of performance. Outcome-specific devaluation provides one diagnostic tool. Following training, non-reinforced instrumental performance is assessed after devaluation of the earned reward, usually achieved by sensory-specific satiation or pairing with a nausea-inducing agent. If subjects are prospectively evaluating the outcome of their actions, they will reduce action performance following devaluation [1,5]. Insensitivity to devaluation is a mark of habits [4,5]. Habits are also resistant to degradation of the action-outcome contingency, e.g., reward omission [30]. Initially, instrumental behavior is under control of the cognitively-taxing, but less error-prone, goal-directed system [1,5]. With repeated practice (e.g., overtraining), habits will slowly form and will come to dominate behavioral control when enough successful repetition has proceeded to ensure sufficient accuracy of the habit [35]. For the purpose of this review, habits will be defined as behaviors that are demonstrably insensitive to outcome devaluation and/or action-outcome contingency manipulation. In some cases, we will refer to data on motor-skill, procedural, or action-sequence learning, performance of all of which have been argued to be reflective of behavioral habits.

Habits have long been proposed to rely on a stimulus-response (S-R) associative architecture [5,3134]. In this view, a reinforcing event functions to stamp in an association between the behavioral response that preceded it and the stimuli present when the response was executed, such that those stimuli become capable of automatically triggering the response. S-R associations are, however, inferred from a lack of evidence for action-outcome control. Although certainly a possibility, habits need not require S-R learning. Another, not necessarily mutually-exclusive, view is that habits result from the ‘chunking’ of commonly-performed action sequences into automatic, stereotyped routines [3,35,36]. One extension of this proposes that, rather than deliberating at each individual step, sequences of movements are concatenated into chunks that, once selected by the decision-maker, run off automatically [3739]. Insensitivity to devaluation results from mistakes in planning that will systematically arise when devaluation occurs offline and from slips in action [38]. Recording studies lend support for this view [36]. Data from our laboratory show that dopamine release in the nucleus accumbens (NAc) core relates to hierarchical motivational control over a large chunk of stereotyped action sequences [40]. Following overtraining, dopamine release will backpropagate away from individual sequence performance and, instead, be released in response to the earliest unexpected reward predictor, where its magnitude inversely correlates with the average speed of subsequent stereotyped action sequence performance. The hierarchical view, that deliberate choices are executed through habitual action sequences, implies that goal-directed and habit processes cooperate more than compete [6,41] to control behavior. This has important implications for the neural mechanisms of actions and habit, suggesting they might not be as discrete as originally thought.

Dorsal striatal encoding of habit

The goal-directed and habit systems have been demonstrated in mice [42], rats [4346], non-human primates [47,48], and humans [4951] to rely on separate cortico-striatal-limbic circuits centered on the dorsomedial (DMS) ‘associative’ striatum and dorsolateral (DLS) ‘sensorimotor’ striatum, respectively. DMS lesions or inactivation prevent subjects from acquiring or using action-outcome associations, forcing reliance on the habit strategy [4345]. Conversely, DLS lesions prevent the formation and execution of habits, preserving goal-directed control even after overtraining [46]. Neural recordings further support DMS v. DLS encoding of actions and habits. Whereas DMS medium spiny projection neuron (MSN) activity can relate to the deliberation required for goal-directed control [52,53], DLS activity is recruited with repeated training and inversely correlates with deliberation [54]. DLS, but not DMS, MSN activity has been shown to occur at the initiation and termination of instrumental action sequences [5557] and this ‘chunking’ activity pattern strengthens with training and persists with overtraining [52,54,55,58], suggesting a relationship to habit [54]. Continuous DLS activity has also been reported during execution of well-trained motor sequences and such activity has been shown to represent the contextual and kinematic information needed to consistently execute a behavioral habit [59].

Striatal MSNs either directly (dMSNs) or indirectly (iMSNs) project to basal ganglia output nuclei and these pathways have traditionally been thought to promote and oppose, respectively, both motor output and reinforcement [6063]. Evidence of opposing activity for instrumental behavioral control has been identified in the DMS, where augmented dMSN and attenuated iMSN post-synaptic strength has been associated with action-outcome learning [64]. In the DLS, depression of spontaneous excitatory events in iMSNs has been implicated in habit formation [65]. Interestingly however, there is also evidence of more coordinated dMSN/iMSN activity in the DLS [66,67]. Both DLS dMSNs and iMSNs are transiently active during execution of a well-trained instrumental behavior [66]. Correspondingly, strengthened ex vivo responses to cortical input in both DLS pathways has been associated with habit expression [68] and the synaptic strength of both projections is enhanced following extended training of a motor skill [69]. Activity in both DLS projections is also needed for well-trained behavior; optical inhibition of either DLS dMSNs or iMSNs will disrupt the initiation and execution of well-trained action sequences [67]. One interpretation of these coordinated activity data is that dMSN activity might facilitate output of desired habits, while iMSN activation inhibits competing motor programs [70]. In support of this, the disruption of well-trained sequence performance by DLS dMSN inactivation is caused by slowing of initiating movements, whereas DLS iMSN inactivation prompts a switch to other competing behaviors. The balance and timing of dMSN v. iMSN activation has also been suggested to be critical for habit execution, given evidence that faster dMSN relative to iMSN ex vivo activation correlates with habit [68]. More work is needed to clarify the unique and interacting functions of each subregion-specific striatal projection pathway in habit. It also remains to be well understood how habits are encoded in ensembles of DLS direct and indirect pathway projection neurons.

Molecular mechanisms of habit

Emerging evidence is indicating that striatal, especially DLS, MSN activity and neuroplasticity is associated with habit learning. Such long-lasting changes in neuronal excitability that support learning require changes in gene transcription and protein synthesis [71]. This has been borne out throughout the brain, including in the NAc, where considerable efforts have identified the intracellular signaling-transcriptional-translational pathways underlying the experience-dependent plasticity associated with reward learning, especially in the context of drug reward (for review see [7276]). Evidence of the dorsal striatal molecular mechanisms that might regulate the neuroplasticity underlying habit is, relatively, more limited, but developing.

The extracellular signal-relate kinase (ERK) pathway has been demonstrated to be central to synaptic plasticity and learning in several different brain regions [7779], including the dorsal striatum. In the striatum, ERK activation is regulated by glutamatergic and dopaminergic receptor signaling [8083], making this pathway ideally suited to initiate intracellular signaling based on coincident or patterned activity of the two major striatal inputs and, therefore, to mediate MSN corticostriatal plasticity [8487] and striatal-dependent learning. Indeed, dorsal striatal ERK signaling has been implicated in motor-skill learning and [8688] and DLS ERK has been suggested as a candidate molecular regulator of habit [85,89], though direct evidence of the latter is needed. Similarly, cyclin-dependent kinase 5 (Cdk5), which is both down- and upstream of ERK [90], has been demonstrated to regulate MSN excitability, and, in the DLS, to be necessary for motor-skill learning [91]. ERK signaling is thought to contribute to striatal plasticity via regulation of gene transcription. In support of this, deletion of ERK from striatal iMSNs suppresses activity-associated gene expression and attenuates functional and structural plasticity [87].

One way ERK signaling regulates gene transcription is through phosphorylation of transcription factors, such as cyclic AMP response element-binding protein (CREB) [92]. CREB activates the transcription of target genes in response to a diverse array of stimuli mediated via activation of several different signal transduction pathways, including PKA and Ca2+, in addition to MAPK/ERK [93]. CREB-mediated gene expression is required for synaptic plasticity and memory processes in many brain regions [93,94], including both the ventral [95] and dorsal striatum [96]. In the dorsal striatum, CREB is required for both long-term potentiation and depression [96]. CREB is activated in the DLS, in particular, as habits come to dominate behavioral control [97]. Correspondingly, disruption of striatal CREB function interferes with both procedural learning [96] and the development of a habit-like navigation strategies [98,99], supporting the possibility that CREB-mediated transcription is critical for habit formation. Which specific gene transcription CREB regulates to mediate habit is an avenue ripe for investigation.

Epigenetic mechanisms of habit

Gene transcription occurs in the context of chromatin, the DNA-histone protein complex that packages genomic DNA. Epigenetic mechanisms alter accessibility to DNA for the transcriptional machinery to coordinate gene expression and are, therefore, fundamental regulators of the transcriptional processes mediating neuronal plasticity and memory [100102]. Histone deacetylases (HDACs) are particularly interesting. Under basal conditions, HDACs, and their associated co-repressor complexes, are present at the promoter regions of actively transcribed genes. These HDACs remove acetyl groups from the histone tails, creating a repressive chromatin state that prevents active gene transcription [75,103105]. HDACs have, therefore, been proposed to act as a ‘molecular brake’ to maintain specific genes in a silent state [103,104,106]. This brake can, however, be released; strong activity-dependent signaling, such as that induced by a salient learning event, can trigger a cascade that temporarily removes HDACs from gene promotors. This allows histone acetylation, which neutralizes the positive charge of the lysine residues on the histone’s N-terminal tails, loosening the DNA-histone interaction to, generally, promote the active gene expression that can subserve synaptic plasticity and learning [75,103105]. Indeed, disrupting HDAC activity can produce hyperacetylation and has been shown to enhance hippocampal synaptic plasticity, transforming what would otherwise be transient, early-phase potentiation, into more persistent, transcription-dependent, long-term potentiation [107] and converting a subthreshold learning event into a long-term, transcription-dependent memory [107112]. HDACs have also been shown to regulate sensory cortical plasticity [113] and amygdala long-term potentiation [114]. In the NAc, considerable evidence has implicated HDACs in drug memory (for review see [76,115]). Until recent work from our laboratory, the function of such mechanisms for instrumental learning or in the dorsal striatum was not known.

We recently investigated this [116]. Systemic inhibition of class I HDACs following instrumental conditioning sessions was found to increase histone acetylation in the dorsal striatum and to accelerate habitual control of behavior, suggesting that HDACs might normally be engaged in the dorsal striatum to restrain the gene expression underlying habit formation. In support of this, early in training, when habits did not yet dominate behavioral control, occupancy of HDAC3, the most highly-expressed class I HDAC in the brain [110,117], was enriched at specific learning-related and CREB-regulated gene promoters in the DLS and then returned to baseline levels with the repeated training that promoted habit. Correspondingly, decreasing HDAC3 function in the DLS was found to accelerate habit formation, while DLS HDAC3 overexpression prevented habit. These data suggest that HDAC3 is normally initially engaged to constrain the CREB-regulated plasticity necessary for habits and under suitable conditions (e.g., repeated success), an instrumental learning opportunity can trigger activity-dependent signaling that removes HDAC3 to create a state permissive to CREB-mediated transcription in the DLS that may allow the neuroplasticity necessary for habits to strengthen and, eventually, come to control behavior.

Based on the canonical role of the DMS in goal-directed learning, it would be logical to predict that disrupting transcriptionally-repressive HDAC3 activity in the DMS would enhance the transcription necessary for action-outcome encoding, and thereby, enhance goal-directed behavioral control, and, congruently, that increasing DMS HDAC3 function would attenuate action-outcome learning and force reliance on habit. But, surprisingly, we found the opposite; HDAC3 activity was found to constrain habit learning in the DMS, similar to its function in the DLS [116]. Disrupting HDAC3 function specifically in the DMS accelerated habit formation, while DMS HDAC3 overexpression prevented the normal transition to habit that would otherwise occur with overtraining. This was supported by evidence that HDAC3 occupancy was engaged early in training and removed with overtraining in the DMS, though at different (but still CREB-regulated) learning-related gene promoters than in the DLS. Therefore, the repressive enzyme HDAC3 acts as a molecular ‘brake’ on the gene transcription underlying habit formation in both the DLS and the DMS.

The precise upstream mechanisms that regulate the HDAC3 brake during instrumental learning are unknown. Because its catalytic activity can be regulated by its phosphorylation state [118], HDAC3 might be directly regulated by presynaptic inputs that activate intracellular signaling cascades. To form an active enzyme complex, HDAC3 must be associated with a co-repressor, either nuclear receptor corepressor (NCoR) or silencing mediator of retinoic acid and thyroid hormone receptor (SMRT) [119]. The ERK pathway might also, therefore, modulate HDAC3 function by phosphorylating and facilitating the nuclear export of SMRT [120], which leads to its dissociation from HDAC3, thereby reducing HDAC3’s transcriptional repression [121]. Future investigation of HDAC3 regulation, its targeting to specific genes, and how this might differ depending on striatal subregion and projection pathway will be pivotal.

Update of medial and lateral dorsal striatal function in actions and habits

Bidirectional subregion-specific manipulations reveal that the epigenetic repressor HDAC3 activity normally functions to constrain habit formation in not only the DLS, but also in the DMS [116]. This implies that transcriptional and likely also structural and/or functional changes in the DMS must occur for habits to develop. These data, therefore, challenge the canonical strict dissociation between DMS and DLS function in goal-directed and habitual control of behavior. Previous data suggest that DLS circuits might store habit-related information [54,59,65,68], with memories vital for goal-directed control stored in the DMS [64,122,123]. One possibility to reconcile these results with the traditional DLS/DMS functional dichotomy is, therefore, that HDAC3 may regulate the formation and storage of habit memories in the DLS, while in the DMS it depotentiates the action-outcome memories underlying goal-directed control as deliberation becomes less required. This might occur specifically in DMS dMSNs, because enhanced plasticity in these cells has been associated with goal-directed learning [64]. In support of this possibility, recent data suggest attenuation of DMS activity is needed to permit the transition to habit [123]. A second possibility is that DMS HDAC3 activity promotes the concatenation of actions into stereotyped chunks. Third, it is possible that HDAC3 regulates activity changes in DMS indirect-pathway projections, activity in which might oppose goal-directed behavioral control and thereby promote habit [124]. These three non-mutually-exclusive hypotheses are ripe for future investigation.

Implications for disease

Dorsal striatal HDAC3 functions as a molecular brake over habit, remaining in place to slow the transition to habit and being removed when the conditions are ripe for habits to dominate [116]. HDAC3 dysfunction could, therefore, lead to the inability to form habits or the maladaptive compulsive habits that mark many neurodegenerative and psychiatric diseases. HDACs might, therefore, be a promising therapeutic target for a variety of conditions [104,125129]. Indeed, the aforementioned data support recent proposals of the utility of HDAC inhibitors for the treatment of Huntington’s and Parkinson’s disease [130], suggesting such treatment targeted to dorsal striatal HDAC3 might alleviate some of the deficits in habit that mark these diseases.

An overreliance on habit learning strategies is associated with a compulsive phenotype [8,16], including that found in patients diagnosed with addiction [8,131]. Similarly, chronic exposure to cocaine [132135], amphetamine [136], or alcohol [137,138], as well as binge-like consumption of highly palatable foods [139] can potentiate habit formation, resulting in reward-seeking behavior that is insensitive to its consequences, even when those consequences are negative. Chronic exposure to the most commonly abused substances also increases dorsal striatal histone acetylation [140142], creating a chromatin state ripe for potential maladaptive learning. Nicotine’s ability to do this has been proposed as a potential mechanism for it as a gateway drug [142,143]. Moreover, stress, a predisposing factor to mental illness, can lead to abnormal HDAC activity [144,145] and can potentiate habits [146148] via opposing structural changes in DMS and DLS circuits [146]. If dorsal striatal HDAC3 functions as a molecular brake on habit, then chronic stress and exposure to addictive substances might remove this brake, creating an epigenetic ‘tag’ that biases future behavioral strategy towards habit, even with this is not adaptive, producing the compulsivity that marks many mental illnesses. If this is true, then dorsal striatal HDAC3 might be a viable target to prevent or reverse maladaptive compulsive behavior.

Highlights.

  • Actions and habits rely on dorsal medial and lateral striatal activity, respectively.

  • The epigenetic repressor HDAC3 is a negative regulator of habit in both subregions.

  • Stress and addictive substances can alter striatal HDAC activity.

  • Disrupted dorsal striatal HDAC3 activity might underlie maladaptive habits.

Acknowledgments

This work was supported by National Institutes of Health grants DA035443 and MH106972 to KMW.

Footnotes

Conflicts of interest

The authors have no actual or potential conflicts of interests to declare.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Adams CD, Dickinson A. Instrumental responding following reinforcer devaluation. The Quarterly Journal of Experimental Psychology. 1981;33:109–121. [Google Scholar]
  • 2.Dickinson A, Balleine BW. Actions and responses: the dual psychology of behaviour. In: Eilan N, McCarthy R, Brewer MW, editors. Spatial representation. Basil Blackwell Ltd; 1993. pp. 277–293. [Google Scholar]
  • 3.Graybiel AM. Habits, rituals, and the evaluative brain. Annu Rev Neurosci. 2008;31:359–387. doi: 10.1146/annurev.neuro.29.051605.112851. [DOI] [PubMed] [Google Scholar]
  • 4.Adams CD. Variations in the sensitivity of instrumental responding to reinforcer devaluation. Quarterly Journal of Experimental Psychology. 1982;34:77–98. [Google Scholar]
  • 5.Dickinson A. Actions and Habits: the development of behavioural autonomy. Philosphical Transactions of the Royal Society of London. 1985;B308:67–78. [Google Scholar]
  • 6.Dolan RJ, Dayan P. Goals and habits in the brain. Neuron. 2013;80:312–325. doi: 10.1016/j.neuron.2013.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7•.Gillan CM, Kosinski M, Whelan R, Phelps EA, Daw ND. Characterizing a psychiatric symptom dimension related to deficits in goal-directed control. Elife. 2016:5. doi: 10.7554/eLife.11305. This study used a large-scale online assessment to find that deficits in goal-directed processing are associated with the compulsive phenotype of several psychiatric conditions. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Voon V, Derbyshire K, Rück C, Irvine MA, Worbe Y, Enander J, Schreiber LR, Gillan C, Fineberg NA, Sahakian BJ, et al. Disorders of compulsivity: a common bias towards learning habits. Mol Psychiatry. 2015;20:345–352. doi: 10.1038/mp.2014.44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Vandaele Y, Janak PH. Defining the place of habit in substance use disorders. Prog Neuropsychopharmacol Biol Psychiatry. 2017 doi: 10.1016/j.pnpbp.2017.06.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Hogarth L, Balleine BW, Corbit LH, Killcross S. Associative learning mechanisms underpinning the transition from recreational drug use to addiction. Ann N Y Acad Sci. 2012 doi: 10.1111/j.1749-6632.2012.06768.x. [DOI] [PubMed] [Google Scholar]
  • 11.Belin D, Belin-Rauscent A, Murray JE, Everitt BJ. Addiction: failure of control over maladaptive incentive habits. Curr Opin Neurobiol. 2013 doi: 10.1016/j.conb.2013.01.025. [DOI] [PubMed] [Google Scholar]
  • 12.Heindel WC, Butters N, Salmon DP. Impaired learning of a motor skill in patients with Huntington’s disease. Behav Neurosci. 1988;102:141–147. doi: 10.1037//0735-7044.102.1.141. [DOI] [PubMed] [Google Scholar]
  • 13.Cohen H, Pourcher E. Intact encoding, impaired consolidation in procedural learning in Parkinson’s disease. Exp Brain Res. 2007;179:703–708. doi: 10.1007/s00221-006-0827-6. [DOI] [PubMed] [Google Scholar]
  • 14.Agostino R, Sanes JN, Hallett M. Motor skill learning in Parkinson’s disease. J Neurol Sci. 1996;139:218–226. [PubMed] [Google Scholar]
  • 15.Knowlton BJ, Mangels JA, Squire LR. A neostriatal habit learning system in humans. Science. 1996;273:1399–1402. doi: 10.1126/science.273.5280.1399. [DOI] [PubMed] [Google Scholar]
  • 16.Gillan CM, Robbins TW, Sahakian BJ, van den Heuvel OA, van Wingen G. The role of habit in compulsivity. Eur Neuropsychopharmacol. 2016;26:828–840. doi: 10.1016/j.euroneuro.2015.12.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Gillan CM, Morein-Zamir S, Urcelay GP, Sule A, Voon V, Apergis-Schoute AM, Fineberg NA, Sahakian BJ, Robbins TW. Enhanced avoidance habits in obsessive-compulsive disorder. Biol Psychiatry. 2014;75:631–638. doi: 10.1016/j.biopsych.2013.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Gillan CM, Papmeyer M, Morein-Zamir S, Sahakian BJ, Fineberg NA, Robbins TW, de Wit S. Disruption in the balance between goal-directed behavior and habit learning in obsessive-compulsive disorder. Am J Psychiatry. 2011;168:718–726. doi: 10.1176/appi.ajp.2011.10071062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Morris RW, Quail S, Griffiths KR, Green MJ, Balleine BW. Corticostriatal control of goal-directed action is impaired in schizophrenia. Biol Psychiatry. 2015;77:187–195. doi: 10.1016/j.biopsych.2014.06.005. [DOI] [PubMed] [Google Scholar]
  • 20.Griffiths KR, Morris RW, Balleine BW. Translational studies of goal-directed action as a framework for classifying deficits across psychiatric disorders. Front Syst Neurosci. 2014;8:101. doi: 10.3389/fnsys.2014.00101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Corbit LH, Janak PH. Habitual Alcohol Seeking: Neural Bases and Possible Relations to Alcohol Use Disorders. Alcohol Clin Exp Res. 2016 doi: 10.1111/acer.13094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Corbit LH. Effects of obesogenic diets on learning and habitual responding. Current Opinion in Behavioral Sciences. 2016;9:84–90. doi: 10.1016/j.cobeha.2015.11.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Balleine BW, Dickinson A. Goal-directed instrumental action: contingency and incentive learning and their cortical substrates. Neuropharmacology. 1998;37:407–419. doi: 10.1016/s0028-3908(98)00033-1. [DOI] [PubMed] [Google Scholar]
  • 24.Balleine BW, O’Doherty JP. Human and Rodent Homologies in Action Control: Corticostriatal Determinants of Goal-Directed and Habitual Action. Neuropsychopharmacology. 2009 doi: 10.1038/npp.2009.131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Balleine BW, Delgado MR, Hikosaka O. The role of the dorsal striatum in reward and decision-making. J Neurosci. 2007;27:8161–8165. doi: 10.1523/JNEUROSCI.1554-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Yin HH, Knowlton BJ. The role of the basal ganglia in habit formation. Nat Rev Neurosci. 2006;7:464–476. doi: 10.1038/nrn1919. [DOI] [PubMed] [Google Scholar]
  • 27.Burton AC, Nakamura K, Roesch MR. From ventral-medial to dorsal-lateral striatum: neural correlates of reward-guided decision-making. Neurobiol Learn Mem. 2015;117:51–59. doi: 10.1016/j.nlm.2014.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Balleine BW, Liljeholm M, Ostlund SB. The integrative function of the basal ganglia in instrumental conditioning. Behav Brain Res. 2009;199:43–52. doi: 10.1016/j.bbr.2008.10.034. [DOI] [PubMed] [Google Scholar]
  • 29.Balleine BW. Neural bases of food-seeking: affect, arousal and reward in corticostriatolimbic circuits. Physiol Behav. 2005;86:717–730. doi: 10.1016/j.physbeh.2005.08.061. [DOI] [PubMed] [Google Scholar]
  • 30.Dickinson A, Squire S, Varga ZI, Smith JW. Omission learning after instrumental pretraining. Q J Exp Psychol. 1998;51B:297–304. [Google Scholar]
  • 31.Hull C. Principles of Behavior. New York: Appleton; 1943. [Google Scholar]
  • 32.Holland PC. Cognitive versus stimulus-response theories of learning. Learn Behav. 2008;36:227–241. doi: 10.3758/lb.36.3.227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.SPENCE KW. Cognitive versus stimulus-response theories of learning. Psychol Rev. 1950;57:159–172. doi: 10.1037/h0058250. [DOI] [PubMed] [Google Scholar]
  • 34.Guthrie ER. The psychology of learning. New York: Harper; 1935. [Google Scholar]
  • 35.Graybiel AM. The basal ganglia and chunking of action repertoires. Neurobiol Learn Mem. 1998;70:119–136. doi: 10.1006/nlme.1998.3843. [DOI] [PubMed] [Google Scholar]
  • 36.Smith KS, Graybiel AM. Habit formation. Dialogues Clin Neurosci. 2016;18:33–43. doi: 10.31887/DCNS.2016.18.1/ksmith. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37••.Dezfouli A, Balleine BW. Actions, action sequences and habits: evidence that goal-directed and habitual action control are hierarchically organized. PLoS Comput Biol. 2013;9:e1003364. doi: 10.1371/journal.pcbi.1003364. This study provides evidence for the hierarchical model that a goal-directed deliberator selects between actions and habitual action sequences. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Dezfouli A, Lingawi NW, Balleine BW. Habits as action sequences: hierarchical action control and changes in outcome value. Philos Trans R Soc Lond B Biol Sci. 2014:369. doi: 10.1098/rstb.2013.0482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Dezfouli A, Balleine BW. Habits, action sequences and reinforcement learning. Eur J Neurosci. 2012;35:1036–1051. doi: 10.1111/j.1460-9568.2012.08050.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40•.Collins AL, Greenfield VY, Bye JK, Linker KE, Wang AS, Wassum KM. Dynamic mesolimbic dopamine signaling during action sequence learning and expectation violation. Sci Rep. 2016;6:20231. doi: 10.1038/srep20231. In this study, we show that action sequences are associated with ramping mesolimbic dopamine concentration elevations that rescind as action sequences become stereotyped and backpropogates to the earliest reward-predicting cue where it relates to hierarchical motivation. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Daw ND, Niv Y, Dayan P. Uncertainty-based competition between prefrontal and dorsolateral striatal systems for behavioral control. Nat Neurosci. 2005;8:1704–1711. doi: 10.1038/nn1560. [DOI] [PubMed] [Google Scholar]
  • 42.Hilario M, Holloway T, Jin X, Costa RM. Different dorsal striatum circuits mediate action discrimination and action generalization. Eur J Neurosci. 2012;35:1105–1114. doi: 10.1111/j.1460-9568.2012.08073.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Yin HH, Knowlton BJ, Balleine BW. Blockade of NMDA receptors in the dorsomedial striatum prevents action-outcome learning in instrumental conditioning. Eur J Neurosci. 2005;22:505–512. doi: 10.1111/j.1460-9568.2005.04219.x. [DOI] [PubMed] [Google Scholar]
  • 44••.Yin HH, Ostlund SB, Knowlton BJ, Balleine BW. The role of the dorsomedial striatum in instrumental conditioning. Eur J Neurosci. 2005;22:513–523. doi: 10.1111/j.1460-9568.2005.04218.x. This study provides evidence that the development and execution of goal-directed actions requires the DMS. [DOI] [PubMed] [Google Scholar]
  • 45.Corbit LH, Janak PH. Posterior dorsomedial striatum is critical for both selective instrumental and Pavlovian reward learning. Eur J Neurosci. 2010;31:1312–1321. doi: 10.1111/j.1460-9568.2010.07153.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46••.Yin HH, Knowlton BJ, Balleine BW. Lesions of dorsolateral striatum preserve outcome expectancy but disrupt habit formation in instrumental learning. Eur J Neurosci. 2004;19:181–189. doi: 10.1111/j.1460-9568.2004.03095.x. This study provides evidence that the development and execution of habits requires the DLS. [DOI] [PubMed] [Google Scholar]
  • 47.Miyachi S, Hikosaka O, Lu X. Differential activation of monkey striatal neurons in the early and late stages of procedural learning. Exp Brain Res. 2002;146:122–126. doi: 10.1007/s00221-002-1213-7. [DOI] [PubMed] [Google Scholar]
  • 48.Miyachi S, Hikosaka O, Miyashita K, Kárádi Z, Rand MK. Differential roles of monkey striatum in learning of sequential hand movement. Exp Brain Res. 1997;115:1–5. doi: 10.1007/pl00005669. [DOI] [PubMed] [Google Scholar]
  • 49.Liljeholm M, Tricomi E, O’Doherty JP, Balleine BW. Neural correlates of instrumental contingency learning: differential effects of action-reward conjunction and disjunction. J Neurosci. 2011;31:2474–2480. doi: 10.1523/JNEUROSCI.3354-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Tricomi E, Balleine BW, O’Doherty JP. A specific role for posterior dorsolateral striatum in human habit learning. Eur J Neurosci. 2009;29:2225–2232. doi: 10.1111/j.1460-9568.2009.06796.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.McNamee D, Liljeholm M, Zika O, O’Doherty JP. Characterizing the associative content of brain structures involved in habitual and goal-directed actions in humans: a multivariate FMRI study. J Neurosci. 2015;35:3764–3771. doi: 10.1523/JNEUROSCI.4677-14.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52••.Thorn CA, Atallah H, Howe M, Graybiel AM. Differential dynamics of activity changes in dorsolateral and dorsomedial striatal loops during learning. Neuron. 2010;66:781–795. doi: 10.1016/j.neuron.2010.04.036. In this study, single-unit recordings were simultaneously made in the DMS and DLS and the data show simultaneous development of an action-bracketing pattern in the DLS and ensemble activity at choice points in the DMS. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Regier PS, Amemiya S, Redish AD. Hippocampus and subregions of the dorsal striatum respond differently to a behavioral strategy change on a spatial navigation task. J Neurophysiol. 2015;114:1399–1416. doi: 10.1152/jn.00189.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54••.Smith KS, Graybiel AM. A dual operator view of habitual behavior reflecting cortical and striatal dynamics. Neuron. 2013;79:361–374. doi: 10.1016/j.neuron.2013.05.038. This study provides evidence that an action-bracketing activity pattern that is insensitive to devaluation emerges in the DLS as habits form. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Jog MS, Kubota Y, Connolly CI, Hillegaart V, Graybiel AM. Building neural representations of habits. Science. 1999;286:1745–1749. doi: 10.1126/science.286.5445.1745. [DOI] [PubMed] [Google Scholar]
  • 56.Jin X, Costa RM. Start/stop signals emerge in nigrostriatal circuits during sequence learning. Nature. 2010;466:457–462. doi: 10.1038/nature09263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Jin X, Tecuapetla F, Costa RM. Basal ganglia subcircuits distinctively encode the parsing and concatenation of action sequences. Nat Neurosci. 2014;17:423–430. doi: 10.1038/nn.3632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Barnes TD, Kubota Y, Hu D, Jin DZ, Graybiel AM. Activity of striatal neurons reflects dynamic encoding and recoding of procedural memories. Nature. 2005;437:1158–1161. doi: 10.1038/nature04053. [DOI] [PubMed] [Google Scholar]
  • 59.Rueda-Orozco PE, Robbe D. The striatum multiplexes contextual and kinematic information to constrain motor habits execution. Nat Neurosci. 2015;18:453–460. doi: 10.1038/nn.3924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Albin RL, Young AB, Penney JB. The functional anatomy of basal ganglia disorders. Trends Neurosci. 1989;12:366–375. doi: 10.1016/0166-2236(89)90074-x. [DOI] [PubMed] [Google Scholar]
  • 61.Graybiel AM. The basal ganglia. Curr Biol. 2000;10:R509–511. doi: 10.1016/s0960-9822(00)00593-5. [DOI] [PubMed] [Google Scholar]
  • 62.Freeze BS, Kravitz AV, Hammack N, Berke JD, Kreitzer AC. Control of basal ganglia output by direct and indirect pathway projection neurons. J Neurosci. 2013;33:18531–18539. doi: 10.1523/JNEUROSCI.1278-13.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Kravitz AV, Tye LD, Kreitzer AC. Distinct roles for direct and indirect pathway striatal neurons in reinforcement. Nat Neurosci. 2012;15:816–818. doi: 10.1038/nn.3100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Shan Q, Ge M, Christie MJ, Balleine BW. The acquisition of goal-directed actions generates opposing plasticity in direct and indirect pathways in dorsomedial striatum. J Neurosci. 2014;34:9196–9201. doi: 10.1523/JNEUROSCI.0313-14.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Shan Q, Christie MJ, Balleine BW. Plasticity in striatopallidal projection neurons mediates the acquisition of habitual actions. Eur J Neurosci. 2015;42:2097–2104. doi: 10.1111/ejn.12971. [DOI] [PubMed] [Google Scholar]
  • 66.Cui G, Jun SB, Jin X, Pham MD, Vogel SS, Lovinger DM, Costa RM. Concurrent activation of striatal direct and indirect pathways during action initiation. Nature. 2013;494:238–242. doi: 10.1038/nature11846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Tecuapetla F, Jin X, Lima SQ, Costa RM. Complementary Contributions of Striatal Projection Pathways to Action Initiation and Execution. Cell. 2016 doi: 10.1016/j.cell.2016.06.032. [DOI] [PubMed] [Google Scholar]
  • 68••.O’Hare JK, Ade KK, Sukharnikova T, Van Hooser SD, Palmeri ML, Yin HH, Calakos N. Pathway-Specific Striatal Substrates for Habitual Behavior. Neuron. 2016;89:472–479. doi: 10.1016/j.neuron.2015.12.032. By imaging corticostriatal activity, these authors provide evidence that activity in both DLS direct and indirectly pathway projections correlates with habit formation and that direct pathway activity precedes indirect pathway activation. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Yin HH, Mulcare SP, Hilário MR, Clouse E, Holloway T, Davis MI, Hansson AC, Lovinger DM, Costa RM. Dynamic reorganization of striatal circuits during the acquisition and consolidation of a skill. Nat Neurosci. 2009;12:333–341. doi: 10.1038/nn.2261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Mink JW. The Basal Ganglia and involuntary movements: impaired inhibition of competing motor patterns. Arch Neurol. 2003;60:1365–1368. doi: 10.1001/archneur.60.10.1365. [DOI] [PubMed] [Google Scholar]
  • 71.Alberini CM. Transcription factors in long-term memory and synaptic plasticity. Physiol Rev. 2009;89:121–145. doi: 10.1152/physrev.00017.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Nestler EJ. Cellular basis of memory for addiction. Dialogues Clin Neurosci. 2013;15:431–443. doi: 10.31887/DCNS.2013.15.4/enestler. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Sorg BA. Reconsolidation of drug memories. Neurosci Biobehav Rev. 2012;36:1400–1417. doi: 10.1016/j.neubiorev.2012.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Hyman SE, Malenka RC, Nestler EJ. Neural mechanisms of addiction: the role of reward-related learning and memory. Annu Rev Neurosci. 2006;29:565–598. doi: 10.1146/annurev.neuro.29.051605.113009. [DOI] [PubMed] [Google Scholar]
  • 75.Robison AJ, Nestler EJ. Transcriptional and epigenetic mechanisms of addiction. Nat Rev Neurosci. 2011;12:623–637. doi: 10.1038/nrn3111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Walker DM, Cates HM, Heller EA, Nestler EJ. Regulation of chromatin states by drugs of abuse. Curr Opin Neurobiol. 2015;30:112–121. doi: 10.1016/j.conb.2014.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Impey S, Obrietan K, Wong ST, Poser S, Yano S, Wayman G, Deloulme JC, Chan G, Storm DR. Cross talk between ERK and PKA is required for Ca2+ stimulation of CREB-dependent transcription and ERK nuclear translocation. Neuron. 1998;21:869–883. doi: 10.1016/s0896-6273(00)80602-9. [DOI] [PubMed] [Google Scholar]
  • 78.Peng S, Zhang Y, Zhang J, Wang H, Ren B. ERK in learning and memory: a review of recent research. Int J Mol Sci. 2010;11:222–232. doi: 10.3390/ijms11010222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Sweatt JD. Mitogen-activated protein kinases in synaptic plasticity and memory. Curr Opin Neurobiol. 2004;14:311–317. doi: 10.1016/j.conb.2004.04.001. [DOI] [PubMed] [Google Scholar]
  • 80.Valjent E, Pascoli V, Svenningsson P, Paul S, Enslen H, Corvol JC, Stipanovich A, Caboche J, Lombroso PJ, Nairn AC, et al. Regulation of a protein phosphatase cascade allows convergent dopamine and glutamate signals to activate ERK in the striatum. Proc Natl Acad Sci U S A. 2005;102:491–496. doi: 10.1073/pnas.0408305102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Sgambato V, Pagès C, Rogard M, Besson MJ, Caboche J. Extracellular signal-regulated kinase (ERK) controls immediate early gene induction on corticostriatal stimulation. J Neurosci. 1998;18:8814–8825. doi: 10.1523/JNEUROSCI.18-21-08814.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Calabresi P, Picconi B, Tozzi A, Ghiglieri V, Di Filippo M. Direct and indirect pathways of basal ganglia: a critical reappraisal. Nat Neurosci. 2014;17:1022–1030. doi: 10.1038/nn.3743. [DOI] [PubMed] [Google Scholar]
  • 83.Krapivinsky G, Krapivinsky L, Manasian Y, Ivanov A, Tyzio R, Pellegrino C, Ben-Ari Y, Clapham DE, Medina I. The NMDA receptor is coupled to the ERK pathway by a direct interaction between NR2B and RasGRF1. Neuron. 2003;40:775–784. doi: 10.1016/s0896-6273(03)00645-7. [DOI] [PubMed] [Google Scholar]
  • 84.Birnbaum SG, Varga AW, Yuan LL, Anderson AE, Sweatt JD, Schrader LA. Structure and function of Kv4-family transient potassium channels. Physiol Rev. 2004;84:803–833. doi: 10.1152/physrev.00039.2003. [DOI] [PubMed] [Google Scholar]
  • 85.Shiflett MW, Balleine BW. Contributions of ERK signaling in the striatum to instrumental learning and performance. Behav Brain Res. 2011;218:240–247. doi: 10.1016/j.bbr.2010.12.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Mazzucchelli C, Vantaggiato C, Ciamei A, Fasano S, Pakhotin P, Krezel W, Welzl H, Wolfer DP, Pagès G, Valverde O, et al. Knockout of ERK1 MAP kinase enhances synaptic plasticity in the striatum and facilitates striatal-mediated learning and memory. Neuron. 2002;34:807–820. doi: 10.1016/s0896-6273(02)00716-x. [DOI] [PubMed] [Google Scholar]
  • 87.Hutton SR, Otis JM, Kim EM, Lamsal Y, Stuber GD, Snider WD. ERK/MAPK signaling is required for pathway-specific striatal motor functions. J Neurosci. 2017 doi: 10.1523/JNEUROSCI.0473-17.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Bureau G, Carrier M, Lebel M, Cyr M. Intrastriatal inhibition of extracellular signal-regulated kinases impaired the consolidation phase of motor skill learning. Neurobiol Learn Mem. 2010;94:107–115. doi: 10.1016/j.nlm.2010.04.008. [DOI] [PubMed] [Google Scholar]
  • 89.Shiflett MW, Brown RA, Balleine BW. Acquisition and performance of goal-directed instrumental actions depends on ERK signaling in distinct regions of dorsal striatum in rats. J Neurosci. 2010;30:2951–2959. doi: 10.1523/JNEUROSCI.1778-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Modi PK, Komaravelli N, Singh N, Sharma P. Interplay between MEK-ERK signaling, cyclin D1, and cyclin-dependent kinase 5 regulates cell cycle reentry and apoptosis of neurons. Mol Biol Cell. 2012;23:3722–3730. doi: 10.1091/mbc.E12-02-0125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91••.Hernandez A, Tan C, Mettlach G, Pozo K, Plattner F, Bibb JA. Cdk5 Modulates Long-Term Synaptic Plasticity and Motor Learning in Dorsolateral Striatum. Scientific Reports. 2016;6:29812. doi: 10.1038/srep29812. This study provides evidence that the protein kinase Cdk5 regulates corticostriatal synaptic plasticity and motor-skill learning. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Xing J, Ginty DD, Greenberg ME. Coupling of the RAS-MAPK pathway to gene activation by RSK2, a growth factor-regulated CREB kinase. Science. 1996;273:959–963. doi: 10.1126/science.273.5277.959. [DOI] [PubMed] [Google Scholar]
  • 93.Kida S, Serita T. Functional roles of CREB as a positive regulator in the formation and enhancement of memory. Brain Res Bull. 2014;105:17–24. doi: 10.1016/j.brainresbull.2014.04.011. [DOI] [PubMed] [Google Scholar]
  • 94.Benito E, Barco A. CREB’s control of intrinsic and synaptic plasticity: implications for CREB-dependent memory models. Trends Neurosci. 2010;33:230–240. doi: 10.1016/j.tins.2010.02.001. [DOI] [PubMed] [Google Scholar]
  • 95.Dong Y, Green T, Saal D, Marie H, Neve R, Nestler EJ, Malenka RC. CREB modulates excitability of nucleus accumbens neurons. Nat Neurosci. 2006;9:475–477. doi: 10.1038/nn1661. [DOI] [PubMed] [Google Scholar]
  • 96.Pittenger C, Fasano S, Mazzocchi-Jones D, Dunnett SB, Kandel ER, Brambilla R. Impaired bidirectional synaptic plasticity and procedural memory formation in striatum-specific cAMP response element-binding protein-deficient mice. J Neurosci. 2006;26:2808–2813. doi: 10.1523/JNEUROSCI.5406-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97•.Colombo PJ, Brightwell JJ, Countryman RA. Cognitive strategy-specific increases in phosphorylated cAMP response element-binding protein and c-Fos in the hippocampus and dorsal striatum. J Neurosci. 2003;23:3547–3554. doi: 10.1523/JNEUROSCI.23-08-03547.2003. This study provides evidence that formation of a habit-like response navigational strategy is associated with phosphorylation of CREB in both the DLS and DMS. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Brightwell JJ, Smith CA, Neve RL, Colombo PJ. Transfection of mutant CREB in the striatum, but not the hippocampus, impairs long-term memory for response learning. Neurobiol Learn Mem. 2008;89:27–35. doi: 10.1016/j.nlm.2007.09.004. [DOI] [PubMed] [Google Scholar]
  • 99.Lee AS, Duman RS, Pittenger C. A double dissociation revealing bidirectional competition between striatum and hippocampus during learning. Proc Natl Acad Sci U S A. 2008;105:17163–17168. doi: 10.1073/pnas.0807749105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Kouzarides T. Chromatin modifications and their function. Cell. 2007;128:693–705. doi: 10.1016/j.cell.2007.02.005. [DOI] [PubMed] [Google Scholar]
  • 101.Day JJ, Sweatt JD. Epigenetic mechanisms in cognition. Neuron. 2011;70:813–829. doi: 10.1016/j.neuron.2011.05.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Peixoto L, Abel T. The role of histone acetylation in memory formation and cognitive impairments. Neuropsychopharmacology. 2013;38:62–76. doi: 10.1038/npp.2012.86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Vogel-Ciernia A, Wood MA. Molecular brake pad hypothesis: pulling off the brakes for emotional memory. Rev Neurosci. 2012;23:607–626. doi: 10.1515/revneuro-2012-0050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.McQuown SC, Wood MA. HDAC3 and the molecular brake pad hypothesis. Neurobiol Learn Mem. 2011;96:27–34. doi: 10.1016/j.nlm.2011.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Renthal W, Nestler EJ. Histone acetylation in drug addiction. Semin Cell Dev Biol. 2009;20:387–394. doi: 10.1016/j.semcdb.2009.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Heinzel T, Lavinsky RM, Mullen TM, Söderstrom M, Laherty CD, Torchia J, Yang WM, Brard G, Ngo SD, Davie JR, et al. A complex containing N-CoR, mSin3 and histone deacetylase mediates transcriptional repression. Nature. 1997;387:43–48. doi: 10.1038/387043a0. [DOI] [PubMed] [Google Scholar]
  • 107.Vecsey CG, Hawk JD, Lattal KM, Stein JM, Fabian SA, Attner MA, Cabrera SM, McDonough CB, Brindle PK, Abel T, et al. Histone deacetylase inhibitors enhance memory and synaptic plasticity via CREB:CBP-dependent transcriptional activation. J Neurosci. 2007;27:6128–6140. doi: 10.1523/JNEUROSCI.0296-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Levenson JM, O’Riordan KJ, Brown KD, Trinh MA, Molfese DL, Sweatt JD. Regulation of histone acetylation during memory formation in the hippocampus. J Biol Chem. 2004;279:40545–40559. doi: 10.1074/jbc.M402229200. [DOI] [PubMed] [Google Scholar]
  • 109.Alarcón JM, Malleret G, Touzani K, Vronskaya S, Ishii S, Kandel ER, Barco A. Chromatin acetylation, memory, and LTP are impaired in CBP+/− mice: a model for the cognitive deficit in Rubinstein-Taybi syndrome and its amelioration. Neuron. 2004;42:947–959. doi: 10.1016/j.neuron.2004.05.021. [DOI] [PubMed] [Google Scholar]
  • 110.McQuown SC, Barrett RM, Matheos DP, Post RJ, Rogge GA, Alenghat T, Mullican SE, Jones S, Rusche JR, Lazar MA, et al. HDAC3 is a critical negative regulator of long-term memory formation. J Neurosci. 2011;31:764–774. doi: 10.1523/JNEUROSCI.5052-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Guan JS, Haggarty SJ, Giacometti E, Dannenberg JH, Joseph N, Gao J, Nieland TJ, Zhou Y, Wang X, Mazitschek R, et al. HDAC2 negatively regulates memory formation and synaptic plasticity. Nature. 2009;459:55–60. doi: 10.1038/nature07925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Malvaez M, McQuown SC, Rogge GA, Astarabadi M, Jacques V, Carreiro S, Rusche JR, Wood MA. HDAC3-selective inhibitor enhances extinction of cocaine-seeking behavior in a persistent manner. Proc Natl Acad Sci U S A. 2013;110:2647–2652. doi: 10.1073/pnas.1213364110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Bieszczad KM, Bechay K, Rusche JR, Jacques V, Kudugunti S, Miao W, Weinberger NM, McGaugh JL, Wood MA. Histone Deacetylase Inhibition via RGFP966 Releases the Brakes on Sensory Cortical Plasticity and the Specificity of Memory Formation. J Neurosci. 2015;35:13124–13132. doi: 10.1523/JNEUROSCI.0914-15.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Monsey MS, Ota KT, Akingbade IF, Hong ES, Schafe GE. Epigenetic alterations are critical for fear memory consolidation and synaptic plasticity in the lateral amygdala. PLoS One. 2011;6:e19958. doi: 10.1371/journal.pone.0019958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Peña CJ, Bagot RC, Labonté B, Nestler EJ. Epigenetic signaling in psychiatric disorders. J Mol Biol. 2014;426:3389–3412. doi: 10.1016/j.jmb.2014.03.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Malvaez M, Greenfield V, Matheos D, Angelillis N, Murphy M, Kennedy P, Wood M, Wassum K. Habits are negatively regulated by HDAC3 in the dorsal striatum. bioRxiv. 2017 doi: 10.1016/j.biopsych.2018.01.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Broide RS, Redwine JM, Aftahi N, Young W, Bloom FE, Winrow CJ. Distribution of histone deacetylases 1–11 in the rat brain. J Mol Neurosci. 2007;31:47–58. doi: 10.1007/BF02686117. [DOI] [PubMed] [Google Scholar]
  • 118.Zhang X, Ozawa Y, Lee H, Wen YD, Tan TH, Wadzinski BE, Seto E. Histone deacetylase 3 (HDAC3) activity is regulated by interaction with protein serine/threonine phosphatase 4. Genes Dev. 2005;19:827–839. doi: 10.1101/gad.1286005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Guenther MG, Barak O, Lazar MA. The SMRT and N-CoR corepressors are activating cofactors for histone deacetylase 3. Mol Cell Biol. 2001;21:6091–6101. doi: 10.1128/MCB.21.18.6091-6101.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Hong SH, Privalsky ML. The SMRT corepressor is regulated by a MEK-1 kinase pathway: inhibition of corepressor function is associated with SMRT phosphorylation and nuclear export. Mol Cell Biol. 2000;20:6612–6625. doi: 10.1128/mcb.20.17.6612-6625.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Stanya KJ, Kao HY. New insights into the functions and regulation of the transcriptional corepressors SMRT and N-CoR. Cell Div. 2009;4:7. doi: 10.1186/1747-1028-4-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Gremel CM, Costa RM. Orbitofrontal and striatal circuits dynamically encode the shift between goal-directed and habitual actions. Nat Commun. 2013;4:2264. doi: 10.1038/ncomms3264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Gremel CM, Chancey JH, Atwood BK, Luo G, Neve R, Ramakrishnan C, Deisseroth K, Lovinger DM, Costa RM. Endocannabinoid Modulation of Orbitostriatal Circuits Gates Habit Formation. Neuron. 2016;90:1312–1324. doi: 10.1016/j.neuron.2016.04.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Li Y, He Y, Chen M, Pu Z, Chen L, Li P, Li B, Li H, Huang ZL, Li Z, et al. Optogenetic Activation of Adenosine A2A Receptor Signaling in the Dorsomedial Striatopallidal Neurons Suppresses Goal-Directed Behavior. Neuropsychopharmacology. 2016;41:1003–1013. doi: 10.1038/npp.2015.227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Gräff J, Tsai LH. The potential of HDAC inhibitors as cognitive enhancers. Annu Rev Pharmacol Toxicol. 2013;53:311–330. doi: 10.1146/annurev-pharmtox-011112-140216. [DOI] [PubMed] [Google Scholar]
  • 126.Kelly TK, De Carvalho DD, Jones PA. Epigenetic modifications as therapeutic targets. Nat Biotechnol. 2010;28:1069–1078. doi: 10.1038/nbt.1678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Dietz KC, Casaccia P. HDAC inhibitors and neurodegeneration: at the edge between protection and damage. Pharmacol Res. 2010;62:11–17. doi: 10.1016/j.phrs.2010.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Hahnen E, Hauke J, Tränkle C, Eyüpoglu IY, Wirth B, Blümcke I. Histone deacetylase inhibitors: possible implications for neurodegenerative disorders. Expert Opin Investig Drugs. 2008;17:169–184. doi: 10.1517/13543784.17.2.169. [DOI] [PubMed] [Google Scholar]
  • 129.Chuang DM, Leng Y, Marinova Z, Kim HJ, Chiu CT. Multiple roles of HDAC inhibition in neurodegenerative conditions. Trends Neurosci. 2009;32:591–601. doi: 10.1016/j.tins.2009.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Abel T, Zukin RS. Epigenetic targets of HDAC inhibition in neurodegenerative and psychiatric disorders. Curr Opin Pharmacol. 2008;8:57–64. doi: 10.1016/j.coph.2007.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Sjoerds Z, de Wit S, van den Brink W, Robbins TW, Beekman AT, Penninx BW, Veltman DJ. Behavioral and neuroimaging evidence for overreliance on habit learning in alcohol-dependent patients. Transl Psychiatry. 2013;3:e337. doi: 10.1038/tp.2013.107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Leblanc KH, Maidment NT, Ostlund SB. Repeated cocaine exposure facilitates the expression of incentive motivation and induces habitual control in rats. PLoS One. 2013;8:e61355. doi: 10.1371/journal.pone.0061355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Corbit LH, Chieng BC, Balleine BW. Effects of repeated cocaine exposure on habit learning and reversal by N-acetylcysteine. Neuropsychopharmacology. 2014;39:1893–1901. doi: 10.1038/npp.2014.37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Miles FJ, Everitt BJ, Dickinson A. Oral cocaine seeking by rats: action or habit? Behav Neurosci. 2003;117:927–938. doi: 10.1037/0735-7044.117.5.927. [DOI] [PubMed] [Google Scholar]
  • 135.Zapata A, Minney VL, Shippenberg TS. Shift from goal-directed to habitual cocaine seeking after prolonged experience in rats. J Neurosci. 2010;30:15457–15463. doi: 10.1523/JNEUROSCI.4072-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Nelson A, Killcross S. Amphetamine exposure enhances habit formation. J Neurosci. 2006;26:3805–3812. doi: 10.1523/JNEUROSCI.4305-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Dickinson A, Wood N, Smith JW. Alcohol seeking by rats: action or habit? Q J Exp Psychol B. 2002;55:331–348. doi: 10.1080/0272499024400016. [DOI] [PubMed] [Google Scholar]
  • 138.Corbit LH, Nie H, Janak PH. Habitual alcohol seeking: time course and the contribution of subregions of the dorsal striatum. Biol Psychiatry. 2012;72:389–395. doi: 10.1016/j.biopsych.2012.02.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Furlong TM, Jayaweera HK, Balleine BW, Corbit LH. Binge-like consumption of a palatable food accelerates habitual control of behavior and is dependent on activation of the dorsolateral striatum. J Neurosci. 2014;34:5012–5022. doi: 10.1523/JNEUROSCI.3707-13.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Kumar A, Choi KH, Renthal W, Tsankova NM, Theobald DE, Truong HT, Russo SJ, Laplant Q, Sasaki TS, Whistler KN, et al. Chromatin remodeling is a key mechanism underlying cocaine-induced plasticity in striatum. Neuron. 2005;48:303–314. doi: 10.1016/j.neuron.2005.09.023. [DOI] [PubMed] [Google Scholar]
  • 141.Shen HY, Kalda A, Yu L, Ferrara J, Zhu J, Chen JF. Additive effects of histone deacetylase inhibitors and amphetamine on histone H4 acetylation, cAMP responsive element binding protein phosphorylation and DeltaFosB expression in the striatum and locomotor sensitization in mice. Neuroscience. 2008;157:644–655. doi: 10.1016/j.neuroscience.2008.09.019. [DOI] [PubMed] [Google Scholar]
  • 142.Kandel DB, Kandel ER. A molecular basis for nicotine as a gateway drug. N Engl J Med. 2014;371:2038–2039. doi: 10.1056/NEJMc1411785. [DOI] [PubMed] [Google Scholar]
  • 143.Levine A, Huang Y, Drisaldi B, Griffin EA, Pollak DD, Xu S, Yin D, Schaffran C, Kandel DB, Kandel ER. Molecular mechanism for a gateway drug: epigenetic changes initiated by nicotine prime gene expression by cocaine. Sci Transl Med. 2011;3:107ra109. doi: 10.1126/scitranslmed.3003062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144•.White AO, Wood MA. Does stress remove the HDAC brakes for the formation and persistence of long-term memory? Neurobiol Learn Mem. 2014;112:61–67. doi: 10.1016/j.nlm.2013.10.007. This study discusses the evidence that stress-related signaling drives long-term memory via an HDAC-dependent mechanism. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Bagot RC, Labonté B, Peña CJ, Nestler EJ. Epigenetic signaling in psychiatric disorders: stress and depression. Dialogues Clin Neurosci. 2014;16:281–295. doi: 10.31887/DCNS.2014.16.3/rbagot. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146••.Dias-Ferreira E, Sousa JC, Melo I, Morgado P, Mesquita AR, Cerqueira JJ, Costa RM, Sousa N. Chronic stress causes frontostriatal reorganization and affects decision-making. Science. 2009;325:621–625. doi: 10.1126/science.1171203. This study demonstrates that chronic unpredictable stress biases towards a habit strategy of behavioral control via opposing structural changes in associative and sensorimotor corticostriatal circuits. [DOI] [PubMed] [Google Scholar]
  • 147.Soares JM, Sampaio A, Ferreira LM, Santos NC, Marques F, Palha JA, Cerqueira JJ, Sousa N. Stress-induced changes in human decision-making are reversible. Transl Psychiatry. 2012;2:e131. doi: 10.1038/tp.2012.59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Schwabe L, Wolf OT. Stress-induced modulation of instrumental behavior: from goal-directed to habitual control of action. Behav Brain Res. 2011;219:321–328. doi: 10.1016/j.bbr.2010.12.038. [DOI] [PubMed] [Google Scholar]

RESOURCES