Elsevier

Physiology & Behavior

Volume 176, 1 July 2017, Pages 17-25
Physiology & Behavior

Central GLP-1 receptor activation modulates cocaine-evoked phasic dopamine signaling in the nucleus accumbens core

https://doi.org/10.1016/j.physbeh.2017.03.019Get rights and content

Highlights

  • Cocaine-evoked increases in dopamine signaling are greater in the NAc shell vs core.

  • Central GLP-1R activation suppresses phasic dopamine signaling in the NAc core.

  • GLP-1R effects are not due to alterations in dopamine reuptake.

Abstract

Drugs of abuse increase the frequency and magnitude of brief (1–3 s), high concentration (phasic) dopamine release events in terminal regions. These are thought to be a critical part of drug reinforcement and ultimately the development of addiction. Recently, metabolic regulatory peptides, including the satiety signal glucagon-like peptide-1 (GLP-1), have been shown to modulate cocaine reward-driven behavior and sustained dopamine levels after cocaine administration. Here, we use fast-scan cyclic voltammetry (FSCV) to explore GLP-1 receptor (GLP-1R) modulation of dynamic dopamine release in the nucleus accumbens (NAc) during cocaine administration. We analyzed dopamine release events in both the NAc shell and core, as these two subregions are differentially affected by cocaine and uniquely contribute to motivated behavior. We found that central delivery of the GLP-1R agonist Exendin-4 suppressed the induction of phasic dopamine release events by intravenous cocaine. This effect was selective for dopamine signaling in the NAc core. Suppression of phasic signaling in the core by Exendin-4 could not be attributed to interference with cocaine binding to one of its major substrates, the dopamine transporter, as cocaine-induced increases in reuptake were unaffected. The results suggest that GLP-1R activation, instead, exerts its suppressive effects by altering dopamine release – possibly by suppressing the excitability of dopamine neurons. Given the role of NAc core dopamine in the generation of conditioned responses based on associative learning, suppression of cocaine-induced dopamine signaling in this subregion by GLP-1R agonism may decrease the reinforcing properties of cocaine. Thus, GLP-1Rs remain viable targets for the treatment and prevention of cocaine seeking, taking and relapse.

Introduction

Drug addiction and dependence are major public health issues. There are currently few efficacious treatments for drug addiction due, in part, to an incomplete understanding of the neurobiology of the disease. While the mechanisms by which drugs of abuse drive behavior are complex and involve a diverse network of brain circuits, it is clear that increased release of the neurotransmitter dopamine plays a critical role in the reinforcing properties of abused drugs [1], [2]. Synchronous burst firing of ventral tegmental area (VTA) dopamine-containing neurons results in brief, high concentration (phasic) increases in nucleus accumbens (NAc) dopamine concentration [3], [4], [5]. These transient increases have been shown to be associated with reward based learning, motivation and reinforcement [6], [7], [8], [9] – all of which are central to the neurobiology of addiction. Cocaine, a highly abused drug with a well-investigated mechanism of action, augments NAc dopamine concentration by binding to and blocking dopamine reuptake by the dopamine transporter [10], [11]. However, cocaine also increases the frequency of dopamine release events [12], a key property for the development of addiction [13]. The increased frequency of dopamine release events would not be predicted by reuptake blockade alone but rather suggests increased burst firing of dopamine neurons which has gained empirical support [14]. Suppression of these cocaine-evoked transients represents a potential therapeutic approach to treating cocaine addiction.

Metabolic state and its proxies have been shown to bidirectionally modulate reward-driven behavior, whether directed at natural (e.g. food; see [15] for review) or artificial (e.g. cocaine; see [16] for review) rewards. The ability of metabolic state to potentiate or suppress drug seeking behavior has been demonstrated with multiple gut-derived peptides and across many classes of drugs of abuse [17], [18], [19], [20], [21], [22]. For example, exogenous administration of the orexigenic peptide ghrelin augments conditioned place preference for cocaine [23], while blockade of ghrelin signaling attenuates conditioned place preference for cocaine, amphetamine, and nicotine [21], [24]. Conversely, anorectic signals like the adiposity hormone leptin reduce food-deprivation induced heroin seeking [19]. Hormonal communicators of hunger and satiety that defend energy balance are therefore sufficient to alter drug reward-driven behavior.

A powerful modulator of food and drug reward is the incretin hormone glucagon-like peptide-1 (GLP-1), which suppresses food intake and body weight in rodents and humans by its actions on central and peripheral GLP-1Rs [25], [26], [27]. Much of the existing work on the anorectic effects of GLP-1 is heavily focused on GLP-1 signaling through metabolic control nuclei including nuclei of the hypothalamus and the nucleus of the solitary tract [28], [29], [30]. Recent work, however, has demonstrated that GLP-1 also decreases food intake by reducing the rewarding value of food [31], [32], [33], [34], implicating a physiological role for the peptide beyond energy balance regulation. Importantly, neurons in the NAc and VTA of the mesolimbic circuit express GLP-1Rs and receive direct projections from central GLP-1 producing neurons [32], [33]. Intra-VTA or NAc infusion of the GLP-1R agonist, Exendin-4, reduces palatable food intake [32], [33] and goal-directed behavior for food [31]. Interestingly GLP-1R agonists also suppress goal-directed behavior for cocaine reward [35], [36], [37], [38], [39], likely by attenuating dopamine signaling in the NAc [35], [39]. However, suppression of cocaine-evoked NAc dopamine release was demonstrated utilizing microdialysis, which lacks the spatiotemporal resolution to allow for investigation of phasic dopamine release patterns as well as potential regional differences. Importantly, the two subregions of the NAc (shell and core) respond differently to cocaine [40], [41] and are postulated to contribute in unique ways to the neurobiological basis of reward- directed behavior and drug addiction [42], [43], [44], [45], [46], [47], [48].

The studies here use fast-scan cyclic voltammetry (FSCV) to measure the pharmacological effects of Exendin-4 (a GLP-1R agonist) on intravenous cocaine-evoked phasic dopamine signaling in the NAc core and shell of rats. We replicate findings that non-contingent cocaine increases dopamine signaling in both the NAc core and shell [49], [50]. We show, for the first time, that central GLP-1R activation suppresses cocaine-evoked dopamine signaling but only within the NAc core. We also demonstrate that in cocaine-treated animals, Exendin-4 has no effect on the magnitude of electrically stimulated dopamine release or the rate of dopamine reuptake. This work suggests that GLP-1R agonism could curb cocaine-directed behavior, in part, by suppressing dopamine signaling in the NAc core. While future studies are needed to establish a causal relationship, GLP-1R targeted pharmacotherapies remain a promising approach for decreasing the reinforcing value of abused drugs like cocaine.

Section snippets

Subjects

Male Sprague-Dawley rats (n = 39; 350–425 g at the time of testing) were housed individually and maintained on a 12:12-h light-dark cycle (on at 7:00 AM). Rats were provided with ad libitum laboratory chow (LabDiet 5012) until post-operative recovery of body weight. Following recovery from the second surgery, animals were fed enough chow to maintain 85% of their ad libitum body weight. Experimental sessions were conducted after 4 days of restricted food access. Animals were treated in accordance

Intravenous cocaine evokes phasic dopamine release in the NAc

Fig. 1 illustrates representative dopamine transients evoked by intravenous cocaine infusion. The color plot (Fig. 1A) and associated concentration trace (Fig. 1B) were acquired from the NAc core of a rat with 15 previous cocaine infusions. Experiments utilizing CV Matches as an index of dopamine signaling used linear regression to compare cyclic voltammograms taken across the recording session to a cyclic voltammogram template for dopamine (Fig. 1C). Cyclic voltammogram templates were

Discussion

Cocaine is highly reinforcing, in part, because it induces phasic dopamine signaling in the NAc. We captured these phasic events using FSCV and examined the hypothesis that central GLP-1R agonism would suppress them. Indeed, the GLP1R agonist Exendin-4 suppressed dopamine signaling but only in the NAc core and not in the NAc shell. The central effects of Exendin-4 on cocaine-induced phasic dopamine signaling and its regional sensitivity is novel. Given the role of the NAc core dopamine in the

Conflict of interest

The authors have no known conflicts of interest associated with this publication and there has been no significant financial support for this work that could have influenced its outcome.

Acknowledgements

The authors gratefully acknowledge the support of The National Institutes of Health (DA025634 to MFR) and the University of Illinois at Chicago (University Fellowship to SMF). The authors would like to thank Dr. Robert Wheeler and Mykel Robbie for their assistance with jugular vein catheterization.

References (101)

  • G. Di Chiara et al.

    Reward system and addiction: what dopamine does and doesn't do

    Curr. Opin. Pharmacol.

    (2007)
  • G. Di Chiara

    Nucleus accumbens shell and core dopamine: differential role in behavior and addiction

    Behav. Brain Res.

    (2002)
  • S.E. Kanoski et al.

    The role of nausea in food intake and body weight suppression by peripheral GLP-1 receptor agonists, exendin-4 and liraglutide

    Neuropharmacology

    (2012)
  • J.T. Yorgason et al.

    Demon voltammetry and analysis software: analysis of cocaine-induced alterations in dopamine signaling using multiple kinetic measures

    J. Neurosci. Methods

    (2011)
  • S. Ikemoto

    Dopamine reward circuitry: two projection systems from the ventral midbrain to the nucleus accumbens-olfactory tubercle complex

    Brain Res. Rev.

    (2007)
  • L.H.L. Sellings et al.

    Characterization of dopamine-dependent rewarding and locomotor stimulant effects of intravenously-administered methylphenidate in rats

    Neuroscience

    (2006)
  • J.M. Uslaner et al.

    The attribution of incentive salience to a stimulus that signals an intravenous injection of cocaine

    Behav. Brain Res.

    (2006)
  • A.E. Kelley

    Ventral striatal control of appetitive motivation: role in ingestive behavior and reward-related learning

    Neurosci. Biobehav. Rev.

    (2004)
  • A.A. Bari et al.

    D1-like and D2 dopamine receptor antagonists administered into the shell subregion of the rat nucleus accumbens decrease cocaine, but not food, reinforcement

    Neuroscience

    (2005)
  • T.S. Dixit et al.

    Antipsychotic-like effect of GLP-1 agonist liraglutide but not DPP-IV inhibitor sitagliptin in mouse model for psychosis

    Physiol. Behav.

    (2013)
  • R.M. Wightman et al.

    Real-time characterization of dopamine overflow and uptake in the rat striatum

    Neuroscience

    (1988)
  • S. Liu et al.

    Regulation of the mesolimbic dopamine circuit by feeding peptides

    Neuroscience

    (2015)
  • J.D. Hommel et al.

    Leptin receptor signaling in midbrain dopamine neurons regulates feeding

    Neuron

    (2006)
  • K.P. Skibicka et al.

    Ghrelin directly targets the ventral tegmental area to increase food motivation

    Neuroscience

    (2011)
  • R.A. Wise

    Dopamine, learning and motivation

    Nat. Rev. Neurosci.

    (2004)
  • B.J. Everitt et al.

    Neural systems of reinforcement for drug addiction: from actions to habits to compulsion

    Nat. Neurosci.

    (2005)
  • P.A. Garris et al.

    Real-time measurement of electrically evoked extracellular dopamine in the striatum of freely moving rats

    J. Neurochem.

    (1997)
  • L.A. Sombers et al.

    Synaptic overflow of dopamine in the nucleus accumbens arises from neuronal activity in the ventral tegmental area

    J. Neurosci.

    (2009)
  • A.A. Hamid et al.

    Mesolimbic dopamine signals the value of work

    Nat. Neurosci.

    (2016)
  • E.E. Steinberg et al.

    A causal link between prediction errors, dopamine neurons and learning

    Nat. Neurosci.

    (2013)
  • N.F. Parker et al.

    Reward and choice encoding in terminals of midbrain dopamine neurons depends on striatal target

    Nat. Neurosci.

    (2016)
  • J. du Hoffmann et al.

    Dopamine invigorates reward seeking by promoting cue-evoked excitation in the nucleus accumbens

    J. Neurosci.

    (2014)
  • M. Ritz et al.

    Cocaine receptors on dopamine transporters are related to self-administration of cocaine

    Science (80-. )

    (1987)
  • R. Chen et al.

    Abolished cocaine reward in mice with a cocaine-insensitive dopamine transporter

    Proc. Natl. Acad. Sci. U. S. A.

    (2006)
  • S. Koulchitsky et al.

    Differential effects of cocaine on dopamine neuron firing in awake and anesthetized rats

    Neuropsychopharmacology

    (2012)
  • J.A. Engel et al.

    Role of appetite-regulating peptides in the pathophysiology of addiction: implications for pharmacotherapy

    CNS Drugs

    (2014)
  • M. Shen et al.

    Mesolimbic leptin signaling negatively regulates cocaine-conditioned reward

    Transl. Psychiatry

    (2016)
  • U. Shalev et al.

    Leptin attenuates acute food deprivation-induced relapse to heroin seeking

    J. Neurosci.

    (2001)
  • P.J. Wellman et al.

    Ghrelin and ghrelin receptor modulation of psychostimulant action

    Front. Neurosci.

    (2013)
  • E. Jerlhag et al.

    Ghrelin receptor antagonism attenuates cocaine- and amphetamine-induced locomotor stimulation, accumbal dopamine release, and conditioned place preference

    Psychopharmacology

    (2010)
  • S.E. Kanoski et al.

    Peripheral and central GLP-1 receptor populations mediate the anorectic effects of peripherally administered GLP-1 receptor agonists, liraglutide and exendin-4

    Endocrinology

    (2011)
  • M.R. Hayes et al.

    The common hepatic branch of the vagus is not required to mediate the glycemic and food intake suppressive effects of glucagon-like-peptide-1

    AJP Regul. Integr. Comp. Physiol.

    (2011)
  • E. Näslund et al.

    Energy intake and appetite are suppressed by glucagon-like peptide-1 (GLP-1) in obese men

    Int. J. Obes. Relat. Metab. Disord.

    (1999)
  • M.R. Hayes et al.

    Caudal brainstem processing is sufficient for behavioral, sympathetic, and parasympathetic responses driven by peripheral and hindbrain glucagon-like-peptide-1 receptor stimulation

    Endocrinology

    (2008)
  • R.R. Schick et al.

    Peptides that regulate food intake: glucagon-like peptide 1-(7-36) amide acts at lateral and medial hypothalamic sites to suppress feeding in rats

    Am. J. Physiol. Regul. Integr. Comp. Physiol.

    (2003)
  • A.L. Alhadeff et al.

    Endogenous glucagon-like peptide-1 receptor signaling in the nucleus tractus solitarius is required for food intake control

    Neuropsychopharmacology

    (2016)
  • S.L. Dickson et al.

    The glucagon-like peptide 1 (GLP-1) analogue, exendin-4, decreases the rewarding value of food: a new role for mesolimbic GLP-1 receptors

    J. Neurosci.

    (2012)
  • A.L. Alhadeff et al.

    GLP-1 neurons in the nucleus of the solitary tract project directly to the ventral tegmental area and nucleus accumbens to control for food intake

    Endocrinology

    (2012)
  • A.M. Dossat et al.

    Glucagon-like peptide 1 receptors in nucleus accumbens affect food intake

    J. Neurosci.

    (2011)
  • J.E. Richard et al.

    Activation of the GLP-1 receptors in the nucleus of the solitary tract reduces food reward behavior and targets the mesolimbic system

    PLoS One

    (2015)
  • Cited by (0)

    View full text