Elsevier

Physiology & Behavior

Volume 104, Issue 1, 25 July 2011, Pages 76-81
Physiology & Behavior

Cholinergic modulation of mesolimbic dopamine function and reward

https://doi.org/10.1016/j.physbeh.2011.04.052Get rights and content

Abstract

The substantial health risk posed by obesity and compulsive drug use has compelled a serious research effort to identify the neurobiological substrates that underlie the development these pathological conditions. Despite substantial progress, an understanding of the neurochemical systems that mediate the motivational aspects of drug-seeking and craving remains incomplete. Important work from the laboratory of Bart Hoebel has provided key information on neurochemical systems that interact with dopamine (DA) as potentially important components in both the development of addiction and the expression of compulsive behaviors such as binge eating. One such modulatory system appears to be cholinergic pathways that interact with DA systems at all levels of the reward circuit. Cholinergic cells in the pons project to DA-rich cell body regions in the ventral tegmental area (VTA) and substantial nigra (SN) where they modulate the activity of dopaminergic neurons and reward processing. The DA terminal region of the nucleus accumbens (NAc) contains a small but particularly important group of cholinergic interneurons, which have extensive dendritic arbors that make synapses with a vast majority of NAc neurons and afferents. Together with acetylcholine (ACh) input onto DA cell bodies, cholinergic systems could serve a vital role in gating information flow concerning the motivational value of stimuli through the mesolimbic system. In this report we highlight evidence that CNS cholinergic systems play a pivotal role in behaviors that are motivated by both natural and drug rewards. We argue that the search for underlying neurochemical substrates of compulsive behaviors, as well as attempts to identify potential pharmacotherapeutic targets to combat them, must include a consideration of central cholinergic systems.

Highlights

► Acetylcholine interacts with dopamine in the mesolimbic system. ► CNS cholinergic pathways influence motivation for both natural and drug rewards. ► Both ACh and DA are involved in modulating compulsive behaviors. ► Therapeutic targets for obesity and addiction should include cholinergic systems.

Introduction

More than three decades of research into the neurobiological substrates of reward have focused attention on the nucleus accumbens (NAc), a ventromedial component of the basal ganglia, as a key structure in the neural systems responsible for translating motivation to action [1], [2], [3], [4]. The preponderance of this research effort has centered on dopamine (DA) as the primary neurotransmitter in this regard. Many laboratories have contributed to our understanding of the role of DA in motivated behavior, and certainly the efforts of Dr. Bartley Hoebel in his laboratory at Princeton have been particularly noteworthy. Studies by Hoebel and colleagues have demonstrated increases in the release of DA in the NAc as a function of a variety of behaviors including feeding [5], [6], rehydration [7], models of binge eating [8], [9] and hypothalamic stimulation [10]. Moreover, his laboratory has been instrumental in demonstrating that many drugs with abuse liability (including cocaine, amphetamine, opiates, nicotine and alcohol) share a common mechanism in their ability to elevate DA levels in the NAc.

Of the many perspectives Hoebel brought to the study of neurochemical modulation of motivated behavior, perhaps the most salient for this review – and one that inspired much of the work presented herein – has been his view (along with that of Dr. Pedro Rada) that acetylcholine constitutes an integral component of the mesolimbic system [11]. Cholinergic pathways interact with key regions in the brain reward circuit, as illustrated in Fig. 1. ACh projections from the nucleus basalis provide input to cortical and subcortical regions of the hippocampus and amygdala. These projections may play a role in learning and memory processes that are implicated in drug-induced dysfunctions like craving and relapse [12], [13]. From the DA cell body region of the ventral tegmental area (VTA), DA terminals synapse on medium spiny gamma-aminobutyric acid (GABA)-containing cells and a smaller population of large, aspiny acetylcholine (ACh)-containing interneurons in the NAc [14]. Cholinergic interneurons have large dendritic arbors and an extensive network of axons that contact many cell bodies and terminals within both the core and shell subdivisions of the NAc [15], [16]. Thus, in conjunction with DA inputs from VTA, ACh interneurons can modulate the activity of the GABA projection neurons, the primary output neurons of the NAc [17], [18].

Cholinergic cells in the laterodorsal tegmental nucleus (LDTg) and the posterior component of the pedunculopontine tegmental nucleus (PPTg) project to the VTA [19], [20] where they modulate the activity of DA neurons and reward processing [21], [22], [23], [24], [25], [26]. The anterior portion of the PPTg sends cholinergic projections to the substantia nigra [20]. Together with glutamate fibers, the ACh connection from the LDTg to the VTA forms a loop between the midbrain DA cells, the pons and the prefrontal cortex. The ACh input onto DA cell bodies (in the VTA) is therefore a critical link for gating information flow through the mesocorticolimbic and mesostriatal systems.

Several lines of evidence suggest that stimulation of both nicotinic and muscarinic cholinergic receptors can affect mesolimbic DA levels and modify the reinforcing value of self-administered drugs. In this regard, the action of nicotine (the prototypical agonist at nicotinic ACh receptors) on the mesolimbic DA system has been studied extensively (for review see Ref. [27]) as has its interaction with other drugs of abuse such as alcohol [28], [29] and cocaine [30], [31]. These studies demonstrate that nicotinic activation increases extracellular DA in the NAc, stimulates locomotor activity with repeated exposure, and potentiates the reinforcing value of cocaine.

In comparison to the rather clear-cut situation for nicotinic activation, the effect of muscarinic drugs on stimulus reinforcement is less clear. There is evidence that muscarinic receptor activation can decrease amphetamine-induced hyperactivity [32] and inhibit amphetamine-induced DA release in the NAc [33]. Conversely, muscarinic antagonists enhance the locomotor stimulating effects of amphetamine and cocaine [32], [34], [35]. Systemic administration of muscarinic agonists and partial agonists has been reported to decrease cocaine self-administration rates in mice [36], whereas co-administration with the muscarinic antagonist, scopolamine decreases cocaine self-administration in rhesus monkeys [37]. In addition, mice lacking the muscarinic M5 type receptor self-administer less cocaine and show reduced cocaine conditioned place preference compared to their wild-type counterparts [38], suggesting that muscarinic receptors may mediate some component of cocaine reward.

Identifying the precise location of cholinergic effects on psychostimulant reward has been difficult because systemic administration of drugs and genetic deletion models affect receptors throughout the nervous system. However, recent work has focused attention on the NAc as a site of interaction between cholinergic mechanisms and cocaine reinforcement [39]. ACh interneurons in the NAc are known to be responsive to cocaine self-administration [40], [41]. Hikida et al. (2001) reported that selective ablation of cholinergic interneurons within the NAc using immunotoxin-mediated cell targeting resulted in increased cocaine-induced locomotor activity and reward value (as measured by conditioned place preference). In contrast, augmentation of ACh levels with acetylcholinesterase inhibitors in lesioned mice had the opposite effect [42], [43]. These studies provide strong evidence that cholinergic cells in the NAc play an important role in modulating the reinforcing value of psychostimulants, but the nature of the cholinergic receptors that are critical for this effect remains unclear.

Section snippets

Acetylcholine–dopamine interactions and ingestive behavior

Seminal studies by Hernandez and Hoebel (1988) demonstrated that food reward increased DA in the NAc as measured by microdialysis [5], and Radhakishun and colleagues reported a similar result in the same year [44]. A key question that remained to be addressed, however, was whether DA output in the NAc was part of a general stimulus-induced arousal process or, alternatively, did DA play a role in the transduction of ingestive reward valence. To address these issues, Hoebel and colleagues

Cholinergic interactions with mesencephalic dopamine systems and drug reward

Work done in the Hoebel laboratory over a decade ago was some of the first to demonstrate a relevant link between ACh in the VTA and feeding related behavior [61]. These findings suggested that an interaction between ACh from pontomesencephalic sources and VTA could also be particularly critical for determining the action of drugs such as cocaine, that increase DA signaling by reuptake blockade. The ability of cocaine to increase extracellular DA is impulse dependent, and the interaction of

Cholinergic antagonist prevents escalation of cocaine self-administration in rats

When rats are allowed increased access time to cocaine for self-administration (from 1-h/day to 6-h/day), they exhibit an escalation in their average daily intake of the drug [64], [65]. Rats maintained on 1-h/day access do not show an escalation. We have replicated this finding in our laboratory (see Fig. 7; red circles, solid line [69]). The escalation of drug intake phenomenon is a potential animal model with strong construct validity for the behavior of some human drug users who, as

Intra-VTA Injection of mecamylamine reduces high-level cocaine intake

A key goal was to identify whether inactivation of cholinergic input to VTA can reduce expression of escalated cocaine intake. In a recent study, we found that microinjection of the nAChR antagonist mecamylamine (60 μg/side) reduced escalated cocaine intake (Fig. 8) but did not reduce cocaine intake below pre-escalation levels. Rats self-administered cocaine for 1 h per day in the pre-escalation phase. The average level of cocaine intake in 1 h access sessions is represented by the dotted line.

Acknowledgments

I (GPM) would like to extend my sincerest gratitude to Dr. Bartley G. Hoebel for his guidance and support throughout the length of this work and all that preceded it. He has been an outstanding role model. The authors would like to thank Tammie Painter for her excellent technical assistance. This work was supported by NIH grants R01 DA14639, T32 DA07262, and P50 DA018165.

References (70)

  • G. Di Chiara et al.

    Modulatory functions of neurotransmitters in the striatum: ACh/dopamine/NMDA interactions

    Trends Neurosci

    (1994)
  • N.J. Woolf et al.

    Cholinergic systems in the rat brain: IV. Descending projections of the pontomesencephalic tegmentum

    Brain Res Bull

    (1989)
  • J.S. Yeomans

    Role of tegmental cholinergic neurons in dopamine activation, antimuscarinic psychosis and schizophrenia

    Neuropsychopharmacology

    (1995)
  • J. Yeomans et al.

    Both nicotinic and muscarinic receptors in ventral tegmental area contribute to brain-stimulation reward

    Pharmacol Biochem Behav

    (1997)
  • B. Soderpalm et al.

    Nicotinic mechanisms involved in the dopamine activating and reinforcing properties of ethanol

    Behav Brain Res

    (2000)
  • V. Zachariou et al.

    Nicotine receptor inactivation decreases sensitivity to cocaine

    Neuropsychopharmacology

    (2001)
  • J. Ichikawa et al.

    Cholinergic modulation of basal and amphetamine-induced dopamine release in rat medial prefrontal cortex and nucleus accumbens

    Brain Res

    (2002)
  • J.J. Hagan et al.

    Facilitation of amphetamine-induced rotation by muscarinic antagonists is correlated with M2 receptor affinity

    Brain Res

    (1987)
  • T. Rasmussen et al.

    Muscarinic receptor agonists decrease cocaine self-administration rates in drug-naive mice

    Eur J Pharmacol

    (2000)
  • M.L. Berlanga et al.

    Cholinergic interneurons of the nucleus accumbens and dorsal striatum are activated by the self-administration of cocaine

    Neuroscience

    (2003)
  • F.S. Radhakishun et al.

    Scheduled eating increases dopamine release in the nucleus accumbens of food-deprived rats as assessed with on-line brain dialysis

    Neuroscience Letters

    (1988)
  • G.P. Mark et al.

    A conditioned stimulus decreases extracellular dopamine in the nucleus accumbens after the development of a learned taste aversion

    Brain Research

    (1991)
  • G.P. Mark et al.

    An appetitively conditioned taste elicits a preferential increase in mesolimbic dopamine release

    Pharmacol Biochem Behav

    (1994)
  • L.L. Butcher

    Nature and mechanisms of cholinergic-monoaminergic interactions in the brain

    Life Sci

    (1977)
  • M.T. Martin-Iverson et al.

    Cholinergic-dopaminergic interactions and the mechanisms of action of antidepressants

    Eur J Pharmacol

    (1983)
  • S.G. Butcher et al.

    Origin and modulation of acetylcholine activity in the neostriatum

    Brain Res

    (1974)
  • D.B. Hoover et al.

    A mapping of the distribution of acetylcholine, choline acetyltransferase and acetylcholinesterase in discrete areas of rat brain

    Brain Res

    (1978)
  • J. Lehmann et al.

    The striatal cholinergic interneuron: synaptic target of dopaminergic terminals?

    Neuroscience

    (1983)
  • H. Stadler et al.

    Enhanced striatal acetylcholine release by chlorpromazine and its reversal by apomorphine

    Brain Res

    (1973)
  • J.C. Stoof et al.

    Stimulation of D2-dopamine receptors in rat neostriatum inhibits the release of acetylcholine and dopamine but does not affect the release of gamma-aminobutyric acid, glutamate or serotonin

    Eur J Pharmacol

    (1982)
  • C.E. Tedford et al.

    Effect of SCH 39166, a novel dopamine D1 receptor antagonist, on [3H]acetylcholine release in rat striatal slices

    Eur J Pharmacol

    (1992)
  • G.P. Mark et al.

    Extracellular acetylcholine is increased in the nucleus accumbens following the presentation of an aversively conditioned taste stimulus

    Brain Res

    (1995)
  • P.V. Rada et al.

    Acetylcholine release in ventral tegmental area by hypothalamic self-stimulation, eating, and drinking

    Pharmacol Biochem Behav

    (2000)
  • L.K. Dobbs et al.

    Comparison of systemic and local methamphetamine treatment on acetylcholine and dopamine levels in the ventral tegmental area in the mouse

    Neuroscience

    (2008)
  • S. Shabani et al.

    Muscarinic type 2 receptors in the lateral dorsal tegmental area modulate cocaine and food seeking behavior in rats

    Neuroscience

    (2010)
  • Cited by (0)

    1

    Current address: Dept of Neuroanatomy, University of Utah, Salt Lake City, UT.

    View full text