Hostname: page-component-7c8c6479df-ws8qp Total loading time: 0 Render date: 2024-03-28T03:35:54.568Z Has data issue: false hasContentIssue false

Contribution of the mesolimbic dopamine system in mediating the effects of leptin and ghrelin on feeding

Published online by Cambridge University Press:  17 July 2012

R. van Zessen
Affiliation:
Department of Neuroscience and Pharmacology, Division of Neuroscience, Rudolf Magnus Institute, University Medical Center Utrecht, Utrecht, The Netherlands
G. van der Plasse
Affiliation:
Department of Neuroscience and Pharmacology, Division of Neuroscience, Rudolf Magnus Institute, University Medical Center Utrecht, Utrecht, The Netherlands
R. A. H. Adan*
Affiliation:
Department of Neuroscience and Pharmacology, Division of Neuroscience, Rudolf Magnus Institute, University Medical Center Utrecht, Utrecht, The Netherlands
*
*Corresponding author: Professor Roger Adan, fax +31 887569032, email r.a.h.adan@umcutrecht.
Rights & Permissions [Opens in a new window]

Abstract

Feeding behaviour is crucial for the survival of an organism and is regulated by different brain circuits. Among these circuits the mesolimbic dopamine (DA) system is implicated in the anticipation and motivation for food rewards. This system consists of the dopaminergic neurons in the ventral tegmental area (VTA), and their projections to different cortico-limbic structures such as the nucleus accumbens and medial prefrontal cortex. While the importance of this system in motivational drive for different rewards, including drugs of abuse, has been clearly established, its role in energy balance remains largely unexplored. Evidence suggests that peripheral hormones such as leptin and ghrelin are involved in the anticipation and motivation for food and this might be partially mediated through their effects on the VTA. Yet, it remains to be determined whether these effects are direct effects of ghrelin and leptin onto VTA DA neurons, and to what extent indirect effects through other brain areas contribute. Elucidation of the role of leptin and ghrelin signalling on VTA DA neurons in relation to disruptions of energy balance might provide important insights into the role of this neural circuit in obesity and anorexia nervosa.

Type
70th Anniversary Conference on ‘Body weight regulation – food, gut and brain signalling’
Copyright
Copyright © The Authors 2012

Abbreviations:
AgRP

agouti-related protein

D1R

dopamine 1 receptor

D2R

dopamine 2 receptor

DA

dopamine

FAA

food anticipatory activity

GABA

γ-aminobutyric acid

GHSR

growth hormone secretagogue receptor

HFHS

high fat high sucrose

LepR

leptin receptor

LH

lateral hypothalamus

mesDA

mesolimbic dopamine

NAc

nucleus accumbens

NPY

neuropeptide Y

Nts

neurotensin

POMC

pro-opiomelanocortin

PR

progressive ratio

VTA

ventral tegmental area

Feeding behaviour is regulated by several defined neural circuits that control different aspects of feeding behaviour such as motivational drive, satiety and the anticipation of food (e.g.( Reference Kelley, Baldo and Pratt 1 )). These neural circuits are under modulatory control of peripheral hormones such as leptin and ghrelin which signal information about the current metabolic state and that act on specific neurons within the circuit, such as hypothalamic neurons that regulate feeding and feeding-related behaviour. Apart from hormonal input into this feeding-circuitry, leptin and ghrelin also act on the mesolimbic dopamine (mesDA) system. This system signals specific reward-related information which can drive feeding behaviour. As such, the mesDA system may mediate how leptin and ghrelin are involved in the anticipation of and motivation for food reward.

The mesDA system consists of the ventral tegmental area (VTA) that contains dopamine (DA) neurons that project to cortico-limbic structures such as the nucleus accumbens (NAc), medial prefrontal cortex, hippocampus and amygdala( Reference Fields, Hjelmstad and Margolis 2 ). Due to this widespread dopaminergic innervation, changes in VTA DA activity, resulting in altered DA signalling, can modulate various reward circuits. These reward circuits are important for feeding behaviour( Reference Narayanan, Guarnieri and DiLeone 3 , Reference Palmiter 4 ). It is unclear to what extent the mesDA system specifically contributes to the effects of leptin and ghrelin signalling on feeding behaviour. To address this question, we will first describe the role of VTA DA neuronal activity in motivation and anticipation for rewards. We will then address this system's involvement in feeding, and discuss how the mesDA system adapts to positive and negative energy balance. Finally, we will address how this system is influenced by energy balance related signals such as leptin and ghrelin. These hormones have direct effects on the VTA, as well as on hypothalamic nuclei that are known to project to the VTA.

Ventral tegmental area dopaminergic involvement in motivation and reward

In order to understand how activity of VTA DA neurons affects the motivation for and anticipation of reward, we will first describe general characteristics of the DA system and the technologies used to determine its function in relation to behaviour.

The mesDA system originates from dopaminergic neurons in the VTA. The VTA itself is a heterogeneous brain area that contains dopaminergic (about 60%), γ-aminobutyric acid (GABA) ergic (about 30%) and glutamatergic (about 10%) neurons( Reference Margolis, Lock and Hjelmstad 5 Reference Yamaguchi, Wang and Li 7 ). VTA DA neurons typically show three modes of firing, but not all modes are always seen in individual neurons. Neurons can be silent or tonically active, and they can also fire in bursts. When they are tonically active they typically fire action potentials at low frequencies. During both conditions VTA neurons can fire in phasic burst patterns. Typically, bursts consist of an average of 3–4 action potential at a 20–50 Hz frequency( Reference Schultz 8 ).

For its actual function, the VTA relies on release and subsequent receptor binding of DA in target areas, such as the NAc. Compared with other neurotransmitters such as glutamate and GABA, DA levels stay elevated for a longer time, allowing for widespread trans-synaptic effects( Reference Fields, Hjelmstad and Margolis 2 ).

Neurons in DA-projection areas contain different DA receptors that have distinct affinities for DA. Dopamine 1 receptor (D1R) and dopamine 2 receptor (D2R) are the most abundant DA receptors in the striatum and have been best described in the effects of DA signalling. Due to tonic activity of VTA DA neurons, there is a basal level of DA in target areas, which is thought to preferentially activate the high affinity D2R. Burst firing induces a transient increase in DA levels, which also causes the activation of the lower affinity D1R( Reference Wightman and Robinson 9 , Reference Gonon 10 ).

While in vivo electrophysiological tools allow for direct measurement of VTA DA neuronal activity during behaviour, techniques such as fast scan cyclic voltammetry and microdialysis allow for the measurement of DA release in target areas. Between these latter two it has been suggested that microdialysis is only able to measure changes in tonic DA release, brought about by basal DA firing, while fast scan cyclic voltammetry is able to measure DA transients caused by VTA DA burst firing as well( Reference Wightman and Robinson 9 ).

Functionally, the mesDA system has been implicated in various behaviours, and different modes of VTA DA neuronal activity contribute to different behavioural aspects. While both tonic and phasic activity are important for motivation, tonic activity plays an important role in enabling locomotor activity, while phasic bursting has been linked to cue–reward seeking behaviour( Reference Schultz, Dayan and Montague 11 Reference Yun, Wakabayashi and Fields 13 ). The mesDA system has also been implicated in coding for salient or aversive events and stimuli, and cognitive functioning( Reference Schultz 8 ,Reference Schultz 12 , Reference Schultz 14 , Reference Bromberg-Martin, Matsumoto and Hikosaka 15 ). In this review, we only focus on the role of DA in motivation and cue–reward seeking.

The mesDA system is crucial for the development of cue–reward seeking in which neutral cues are associated with rewards they predict. In the context of feeding behaviour, it is important for animals to be able to assess nutritional value of foods when they encounter related cues, such as the smell or sight of particular food. Over time, specific cues illicit specific conditioned anticipatory responses that are coupled to the reward that the cue predicts. DA neurons show burst firing during unexpected primary rewards, such as food, and this burst firing is higher for larger rewards( Reference Tobler, Fiorillo and Schultz 16 ). When a cue is coupled to a reward over multiple sessions, animals learn that the cue predicts the reward and DA burst firing shifts to the cue onset( Reference Schultz, Dayan and Montague 11 , Reference Waelti, Dickinson and Schultz 17 , Reference Fiorillo, Tobler and Schultz 18 ). In this context, DA neurons have been shown to encode a reward prediction error that drives learning.

The reward prediction error encodes the difference between a predicted outcome and the actual outcome( Reference Schultz, Dayan and Montague 11 ). These characteristics are also seen in NAc DA release( Reference Day, Roitman and Wightman 19 Reference Roitman, Stuber and Phillips 21 ), as measured using fast-scan cyclic voltammetry. Importantly, when DA signalling in the NAc is disturbed( Reference Nicola, Taha and Kim 22 ) associative learning is impaired. Rather than only encoding the presence of a reward, this signal also correlates with the size of the reward( Reference Tobler, Fiorillo and Schultz 16 , Reference Roesch, Calu and Schoenbaum 23 , Reference Cohen, Haesler and Vong 24 ).

Overall, VTA DA neuronal firing encodes a reward prediction error that is important for cue–reward associative learning. This role of DA neurons is important in feeding behaviour. In the following section, we will discuss how the mesDA system has been implicated in feeding behaviour.

Dopamine and feeding behaviour

MesDA signalling is thought to be essential for the motivation to earn food rewards( Reference Salamone, Wisniecki and Carlson 25 Reference Nowend, Arizzi and Carlson 28 ), but not for the actual consumption of these rewards. The consumption of food is mediated by dopaminergic projections from substantia nigra neurons to the dorsolateral striatum( Reference Szczypka, Kwok and Brot 29 Reference Salamone, Mahan and Rogers 32 ). Furthermore, the anticipation of food rewards is thought to be mediated by a distributed neural circuit that besides the mesDA system includes hypothalamic nuclei( Reference Acosta-Galvan, Yi and van der Vliet 33 Reference King and Hentges 35 ). In this review, we focus only on the mesDA system.

In general terms, the mesDA system has been implicated in the rewarding effects of natural rewards such as food or sex, and drug rewards, as these have been shown to increase VTA DA firing activity and release of DA in target areas( Reference Schultz, Dayan and Montague 11 , Reference Schultz 36 Reference Berridge and Robinson 40 ). Moreover, VTA DA burst firing has been shown to be rewarding, as animals actively self-administer these bursts( Reference Witten, Steinberg and Lee 41 ) and direct activation of VTA DA neurons increases operant responses for food rewards( Reference Adamantidis, Tsai and Boutrel 42 ). In addition, induced burst firing of VTA DA neurons elicits conditioned place preference( Reference Tsai, Zhang and Adamantidis 43 ).

In the case of food rewards, it has been shown that food-restricted animals have lower basal DA levels in the NAc( Reference Pothos, Creese and Hoebel 44 ), although these changes are not always observed( Reference Verhagen, Luijendijk and Korte-Bouws 45 Reference Cadoni, Solinas and Valentini 47 ). However, most studies show an increased DA release when animals are given rewards such as food( Reference Pothos, Creese and Hoebel 44 , Reference Verhagen, Luijendijk and Korte-Bouws 45 , Reference Hernandez and Hoebel 48 ). Importantly, these animals also show increased motivation for food during food restriction( Reference Wise 37 , Reference Carr 49 ). Food-restricted animals exert more effort to obtain food with increasing energetic value, and also respond more in an operant task when they are able to gain more food( Reference Wise 37 , Reference Fulton 50 ). This increase in motivation can be modulated by changes in DA signalling. When synaptic DA levels are increased through local infusion of amphetamine into the NAc( Reference Sulzer, Chen and Lau 51 ), performance on a progressive ratio (PR) task for a sucrose pellet is increased( Reference Zhang, Balmadrid and Kelley 52 ). Similarly, when VTA DA neuronal firing is increased through optogenetic techniques, sucrose consumption is increased( Reference Domingos, Vaynshteyn and Voss 53 ).

By blocking DA signalling through systemic injection of a D1R or D2R antagonist, food-restricted rats show a decrease in performance on a PR for food( Reference Aberman, Ward and Salamone 54 ). Furthermore, there is a similar decrease in performance when the DA neurotoxin 6-hydroxy is introduced into the NAc. This neurotoxin destroys DA neurons, decreasing NAc DA signalling( Reference Aberman, Ward and Salamone 54 ). This indicates that the increased motivation for food in these food-restricted animals is mediated through mesDA signalling.

Overall these studies indicate that DA signalling is important for feeding behaviour. In food-restricted animals, there is a higher increase in DA signalling when animals are given food or food-related cues are presented. These animals also show more motivation for obtaining food, and this motivation might be mediated by dopaminergic signalling. However, it remains to be determined whether the changes in DA signalling seen in food-restricted animals are due to direct changes in VTA DA neuronal firing and to what extent this is mediated by modulation in neurotransmitter signalling in target areas such as the NAc.

Mesolimbic dopamine in obesity

Animals which are kept on a diet that contains a high-fat high-sucrose (HFHS) content typically gain body weight, display increased motivation to obtain sucrose( Reference Figlewicz, Bennett and Naleid 55 , Reference la Fleur, Vanderschuren and Luijendijk 56 ) and show changes in mesDA functioning. It is thought that some of these changes may be attributable to specific diets. Rats on a HFHS diet have decreased NAc D1R expression compared with controls( Reference Alsio, Olszewski and Norback 57 ) and when exposed to both regular chow and an energy-dense high-fat diet that resembles cafeteria diets, their striatal D2R levels are decreased( Reference Johnson and Kenny 58 ), similar to human obese patients( Reference Wang, Volkow and Logan 59 ). Furthermore VTA DA transporter levels are increased, while tyrosine hydroxylase levels are decreased in high-fat fed mice( Reference Alsio, Olszewski and Norback 57 , Reference Li, South and Han 60 ). In these animals, DA turnover in the NAc is decreased( Reference Davis, Tracy and Schurdak 61 ). Further supporting this, animals which are fed a high-fat diet are unable to illicit conditioned place preference for amphetamine( Reference Davis, Tracy and Schurdak 61 ), supporting a possible decrease in NAc DA levels. These molecular changes indicate that DA production and signalling might be decreased.

It has been hypothesised that initial excessive feeding triggers increased DA release, and as a compensatory mechanism DA receptors are then desensitised, resulting in a tolerance to the rewarding effects of excessive feeding. This then causes further excessive feeding in an attempt to increase DA signalling( Reference Wang, Volkow and Fowler 62 ). In accordance with this, rats that are chronically exposed to a HFHS diet are less susceptible to the rewarding effects of lateral hypothalamus (LH) self-stimulation, and this decrease is potentiated when the D2R is knocked down in the striatum( Reference Johnson and Kenny 58 ).

In summary, the mesDA system is important for feeding behaviour, and modulated during changes in energy balance. In the case of food restriction, mesDA signalling increases above free fed animals when subjected to food or tasks that are awarded by food. Opposite to this, obese animals seem to have lower DA-mediated signalling which animals try to compensate for by increasing their behaviour for obtaining food.

It should be taken into account that NAc DA levels do not necessarily reflect VTA neuronal activity, as they are also dependent on reuptake and clearance of synaptic DA. In addition, dopaminergic neurotransmission depends on the number and type of DA receptors. When animals are kept on a high-fat or HFHS diet, NAc DA-mediated signalling is decreased, indicated by lower D1R and D2R levels, lower DA turnover in the NAc and higher DA transporter expression in VTA neurons( Reference Alsio, Olszewski and Norback 57 , Reference Johnson and Kenny 58 , Reference Davis, Tracy and Schurdak 61 ). It remains to be investigated whether basal tonic and burst firing is changed in VTA DA neurons in obese animals. To fully understand the role of VTA DA neurons in this process, it is important to be able to record their activity directly in vivo.

Although our current understanding of the changes induced by increased or decreased food availability on VTA DA neurons is incomplete, it is apparent that this system changes when energy balance is disturbed. As leptin and ghrelin are strongly regulated during negative and positive energy balance, these peptides likely modulate the mesDA system, and might do so at the level of the VTA.

Inputs to the ventral tegmental area that are involved in feeding and energy balance

To understand how the mesDA system plays a role in energy balance, it is important to understand how different inputs important for feeding regulate this circuit. In this review, we only focus on the VTA, as the origin of the dopaminergic projections that make the mesDA system. The VTA receives inputs from peptide hormones, and several brain areas that play a role in the regulation of feeding behaviour and energy balance. Among these brain areas there are distinct hypothalamic nuclei that are important for feeding. This review is limited to the effects of the hormones leptin and ghrelin on the VTA, and on hypothalamic areas that are known to project to the VTA( Reference Fields, Hjelmstad and Margolis 2 , Reference King and Hentges 35 , Reference Philpot, Dallvechia-Adams and Smith 63 , Reference Greenwell, Zangen and Martin-Schild 64 ) (see Fig. 1).

Fig. 1. Schematic representation of ventral tegmental area (VTA) inputs from the hypothalamus which are modulated by leptin and ghrelin. VTA dopaminergic neuronal activity is decreased by leptin( Reference Hommel, Trinko and Sears 65 ) and increased by ghrelin( Reference Abizaid, Liu and Andrews 66 ). Orexin-producing neurons from the lateral hypothalamus (LH) are inhibited by leptin( Reference Yamanaka, Beuckmann and Willie 67 ) and excited by ghrelin( Reference Yamanaka, Beuckmann and Willie 67 , Reference Lawrence, Snape and Baudoin 68 ). Orexin A increases excitatory projections onto VTA dopamine (DA) neurons( Reference Borgland, Chang and Bowers 69 ). LH neurotensin (Nts)/γ-aminobutyric acid (GABA)-producing neurons are excited by leptin, and project onto LH orexin neurons, as well as VTA DA neurons( Reference Leinninger, Opland and Jo 70 ). VTA GABA neurons directly inhibit VTA DA neurons, and are also modulated by hypothalamic inputs. Endomorphin-producing neurons from the ventromedial hypothalamus (VMH) project to the VTA( Reference Thompson, Canteras and Swanson 71 ), and opioid release in the VTA inhibits VTA GABA neurons( Reference Johnson and North 72 ). It remains to be investigated whether these hypothalamic neurons are also leptin or ghrelin responsive. Pro-opiomelanocortin (POMC) neurons from the arcuate nucleus project to the VTA( Reference King and Hentges 35 ). They produce α-melanocyte stimulating hormone that excites VTA GABA neurons( Reference Korotkova, Sergeeva and Eriksson 73 ), and are themselves excited by leptin( Reference Cheung, Clifton and Steiner 74 , Reference Schwartz, Woods and Porte 75 ). These POMC neurons are inhibited by arcuate nucleus agouti-related protein (AgRP) neurons that in turn are inhibited by leptin and excited by ghrelin( Reference Schwartz, Seeley and Campfield 76 Reference Nakazato, Murakami and Date 78 ). Note that this schematic only includes hypothalamic inputs onto the VTA that have been described in the literature, but other hypothalamic inputs may exist. Also, projections shown here do not represent all the known projections of the different neuronal subtypes.

Effects of leptin on the ventral tegmental area and mesolimbic dopamine system

Leptin is an adipose-derived hormone that signals information on adiposity to the brain. It increases energy expenditure, and decreases food intake, resulting in a decrease in body mass( Reference Campfield, Smith and Guisez 79 Reference Pelleymounter, Cullen and Baker 81 ). Dysfunctional leptin signalling has been implicated in the development of obesity in the spontaneous obese ob/ob mice( Reference Zhang, Proenca and Maffei 82 , Reference Speakman, Hambly and Mitchell 83 ). Leptin binds to the leptin receptor (LepR), a single pass membrane receptor, leading to dimerisation, the activation of Janus kinase and phosphorylated signal transducer, and activator of transcription 3. Injection of leptin directly into the VTA increases phosphorylated signal transducer and activator of transcription 3 levels and activates Jak-2 signalling pathways( Reference Morton, Blevins and Kim 84 , Reference Fulton, Pissios and Manchon 85 ). Among its different sites of effects, it also acts on neurons in the arcuate nucleus of the hypothalamus and mediates signalling for a decrease in food intake. It does so by acting on neuropeptide Y (NPY)/Agouti-related protein (AgRP) neurons( Reference Schwartz, Seeley and Campfield 76 , Reference van den Top, Lee and Whyment 77 ) and pro-opiomelanocortin (POMC) neurons( Reference Cheung, Clifton and Steiner 74 , Reference Schwartz, Woods and Porte 75 ).

The LepR is also expressed in the VTA, where 75–90% of the LepR positive neurons are dopaminergic( Reference Hommel, Trinko and Sears 65 , Reference Figlewicz, Evans and Murphy 86 , Reference Leshan, Opland and Louis 87 ), although it was reported that this represents only a fraction of the total amount of VTA DA neurons( Reference Leshan, Opland and Louis 87 ). In VTA acute brain slices, leptin hyperpolarises putative DA neurons and decreases their neuronal firing frequency( Reference Hommel, Trinko and Sears 65 ). Furthermore leptin injections decrease concentrations of extracellular DA levels in the NAc( Reference Krugel, Schraft and Kittner 88 ). Some of the leptin responsive VTA DA neurons project to the NAc( Reference Fulton, Pissios and Manchon 85 ), while it has been argued that the majority of LepR-containing VTA neurons project to central amygdala neurons containing cocaine and amphetamine responsive transcript( Reference Leshan, Opland and Louis 87 ). Supporting a possible implication of amygdala-mediated signalling, mice show an anxiogenic effect when LepR is knocked down in VTA DA neurons specifically, and this phenotype can be saved by local microinjection of a D1R antagonist into the central amygdala( Reference Liu, Perez and Zhang 89 ). Overall LepR is expressed in VTA DA neurons that project to the central amygdala, and to a lesser extent, in neurons that project to the NAc. Activation of this receptor by leptin decreases DA neuronal firing and subsequent NAc DA release.

Mice that lack leptin (ob/ob mice) are obese, are hyperphagic and have decreased energy expenditure. They have reduced DA levels in the NAc and decreased tyrosine hydroxylase levels in the VTA( Reference Fulton, Pissios and Manchon 85 ). Moreover, similar to obese patients( Reference Wang, Volkow and Logan 59 ), they also have reduced D2R in the VTA( Reference Roseberry, Painter and Mark 90 ), and this can be restored by leptin treatment( Reference Pfaffly, Michaelides and Wang 91 ). Interestingly, rats that are unable to produce the LepR, termed fa/fa rats or Zucker rats also become obese, and, similar to ob/ob mice, their VTA D2R levels are reduced( Reference Thanos, Michaelides and Piyis 92 ). Overall, these studies indicate that DA signalling originating in the VTA is reduced in leptin-deficient animals. Curiously, intra-VTA administration of leptin also reduces DA release in the NAc( Reference Krugel, Schraft and Kittner 88 ), an effect that may be caused by a direct inhibitory effect of leptin on VTA DA neurons( Reference Hommel, Trinko and Sears 65 ). Apart from possible developmental changes that occur in ob/ob mice, the reduced DA release in these mice may be due to a lack of leptin signalling in neurons projecting to the VTA, such as those from the LH (see section ‘Indirect effects of peptide hormones from hypothalamic inputs’). These studies, using leptin-deficient mice clearly show that leptin signalling is important for mesDA signalling.

This requirement of leptin for mesDA signalling is supported by studies in which leptin was administered (rather than removed) and food-related behaviours were investigated. Leptin injections in the ventricle and in the VTA decrease food consumption, while overall locomotor activity is not impaired( Reference Morton, Blevins and Kim 84 , Reference Leshan, Opland and Louis 87 , Reference Krugel, Schraft and Kittner 88 ). Ventricular leptin injections decrease performance on a PR task for food( Reference Figlewicz, Bennett and Naleid 55 ), indicating that the motivation for a food reward is decreased. Taken together these data suggest an involvement of the mesDA system in leptin's effect on reduced food intake and food reward. In animals on a HFHS diet, ventricular injections have no effect on food consumption( Reference Figlewicz, Bennett and Naleid 55 ), suggesting a leptin resistance. Mice with leptin over-expression in the VTA gained less body weight over time when compared with controls, and interestingly, they preferred a high-fat diet more than controls. Phosphorylated signal transducer and activator of transcription 3 activation was lower in the VTA of these leptin over-expressing animals, while also being lower than chow controls in HFHS-fed animals( Reference Matheny, Shapiro and Tumer 93 ). This indicates that chronically increased VTA leptin levels induced by HFHS diets or by leptin overexpression in the VTA blunt VTA LepR-mediated signalling.

Viral knock down of the LepR in the VTA using RNA interference increases food intake under both balanced and high-fat diets( Reference Hommel, Trinko and Sears 65 ). These animals also show increased locomotor activity and preference for sucrose and high-fat food. However, in contrast to this, transgenic animals that have the LepR knocked out specifically in VTA DA neurons show no change in body weight or food intake on a chow or high-fat diet( Reference Liu, Perez and Zhang 89 ). It is possible that these discrepancies arise from the specificity of the knockdown. While Hommel and co-workers most likely knocked down the LepR in all VTA neurons, including GABA and glutamatergic neurons( Reference Hommel, Trinko and Sears 65 , Reference Figlewicz, Evans and Murphy 86 , Reference Leshan, Opland and Louis 87 , Reference Liu, Perez and Zhang 89 ), Liu et al.( Reference Liu, Perez and Zhang 89 ) knocked out the LepR specifically in DA neurons. Furthermore, the use of a transgenic knock out might facilitate compensatory changes. Further investigation into the role of leptin in the VTA remains necessary to understand its role on VTA-mediated feeding behaviour.

Overall, based on electrophysiological evidence in vitro and microdialysis studies in vivo leptin decreases the activity of VTA neurons, and subsequent DA release for instance in the NAc. By doing so it decreases motivation for food resulting in lower food consumption. However, when leptin levels are constantly elevated by diet or over-expression, leptin-mediated signalling is decreased and the effect of additional leptin is blunted (leptin resistance). Therefore it can be concluded that the effects of leptin-signalling are likely dependent on metabolic state and diet.

Effects of ghrelin on the ventral tegmental area and mesolimbic dopamine system

Ghrelin is a twenty-eight-amino acid peptide hormone that is primarily produced by the stomach. Its plasma levels rise during fasting and decreases after a meal, and it promotes feeding( Reference Nakazato, Murakami and Date 78 ). It acts on the growth hormone secretagogue receptor (GHSR), a G protein-coupled receptor that couples to Gαq signalling( Reference Wettschureck, Moers and Wallenwein 94 ). It is expressed in different brain areas closely involved in feeding behaviour, including hypothalamic areas (e.g. arcuate nucleus, dorsomedial hypothalamus and ventromedial hypothalamus and paraventricular nucleus), but, importantly, also in the VTA( Reference Zigman, Jones and Lee 95 , Reference Mondal, Date and Yamaguchi 96 ). Within the arcuate nucleus, the GHSR is expressed on NPY and AgRP neurons, through which ghrelin stimulates feeding behaviour by increasing meal frequency, but not meal size( Reference Nakazato, Murakami and Date 78 , Reference Tschop, Smiley and Heiman 97 , Reference Horvath, Diano and Schop 98 ). In addition, ghrelin is involved in the anticipation of meals. While its plasma levels are correlated with food anticipatory activity (FAA)( Reference Verhagen, Egecioglu and Luijendijk 99 ), FAA is decreased in animals in which ghrelin signalling is blocked, either in GHSR knock out mice or by injection of a GHSR antagonist( Reference Verhagen, Egecioglu and Luijendijk 99 Reference Blum, Patterson and Khazall 101 ). Chronic ghrelin injections furthermore increase c-Fos expression in different hypothalamic areas, in a pattern that is similar to that induced by food restriction( Reference Blum, Patterson and Khazall 101 ).

In the VTA, the GHSR is co-localised with tyrosine hydroxylase in 35–60% of the GHSR positive neurons( Reference Abizaid, Liu and Andrews 66 , Reference Zigman, Jones and Lee 95 , Reference Guan, Yu and Palyha 102 ). Opposite to leptin-induced inhibition of putative VTA DA neurons, ghrelin increases the firing rate of DA neurons in slices. Interestingly, this effect is abolished by blockade of glutamate, but not GABAergic neurotransmission( Reference Abizaid, Liu and Andrews 66 ), indicating that these effects are mediated by presynaptic glutamatergic inputs, and not by direct effects of GHSR on DA neurons. Furthermore, 90 min after intraperitoneal injection of ghrelin, electron microscopy shows that VTA DA neurons have increased incoming glutamatergic synapses and decreased GABAergic synapses. These results indicate that the effects of ghrelin might mediate an increased excitation and decreased inhibition. This is supported by the fact that ghrelin increases the frequency of miniature excitatory postsynaptic currents, but decreases miniature inhibitory postsynaptic currents onto VTA DA neurons( Reference Abizaid, Liu and Andrews 66 ).

Systemic ghrelin administration increases DA levels in the NAc shell( Reference Quarta, Di Francesco and Melotto 103 ), and DA turnover, an effect that is absent in GHSR knock out animals( Reference Abizaid, Liu and Andrews 66 ). More specifically, local infusion of ghrelin into the VTA increases the number of lever presses for a sucrose reward on a PR task and fixed ratio tasks, and also increases food intake in free-fed rats( Reference Skibicka, Hansson and Alvarez-Crespo 104 Reference Naleid, Grace and Cummings 106 ). Furthermore, intra-VTA ghrelin injections increase locomotor activity and DA release in the NAc( Reference Jerlhag, Egecioglu and Dickson 107 ). Chronic ghrelin infusion in the VTA also dose-dependently increases food intake( Reference King, Isaacs and O'Farrell 105 ). Importantly, increases in food intake by systemic injection of ghrelin are blocked by local ghrelin antagonist injection into the VTA and chronic blockade of VTA GHSR abolishes food intake increase on a high-fat diet( Reference Abizaid, Liu and Andrews 66 , Reference King, Isaacs and O'Farrell 105 ). These results indicate that the VTA at least partly mediates the increases in motivation and consumption of food by ghrelin.

In food-restricted animals, ghrelin receptor activation contributes to FAA( Reference Verhagen, Egecioglu and Luijendijk 99 ) and increases PR performance for food( Reference Skibicka, Hansson and Alvarez-Crespo 104 ). This increase in performance can be blocked by VTA injection of a GHSR antagonist( Reference Skibicka, Hansson and Alvarez-Crespo 104 ). Finally, ghrelin deficient and GHSR knock out animals do not increase their food intake over days when they are food restricted overnight( Reference Abizaid, Liu and Andrews 66 ).

Ghrelin signalling in the VTA also appears to play a role in food choice behaviour. When given the choice between standard chow and more palatable food, both transgenic GHSR knock out mice, as well as rats systemically treated with a ghrelin antagonist, show suppressed intake of palatable food, but not of chow intake( Reference Egecioglu, Jerlhag and Salome 108 ). Furthermore, ghrelin administration in the VTA increased intake of palatable food, but not chow, during an hour exposure to both types of food( Reference Egecioglu, Jerlhag and Salome 108 ).

Indirect effects of peptide hormones from hypothalamic inputs

Thus far we have discussed how the mesDA system is involved in motivated behaviour, feeding, and how VTA DA neurons might be affected directly through the actions of leptin and ghrelin. However, it is likely that these neurons are also affected by inputs from leptin- and ghrelin-responsive neurons in other brain areas. Among these brain areas, we focus on the effects on different hypothalamic nuclei, because of their importance in homoeostatic control of feeding behaviour.

Lateral hypothalamic control of the ventral tegmental area

The LH consists of multiple nuclei that receive inputs not only from the arcuate nucleus POMC/cocaine and amphetamine responsive transcript and NPY/AgRP neurons but also from NAc shell, cortical areas, amygdala, hippocampus and multiple brain stem nuclei. It projects to various brain areas, including the VTA, paraventricular and arcuate nuclei, amygdala, hippocampus and different cortical areas( Reference Berthoud and Munzberg 109 ). As such it is ideally situated to serve as a centre that integrates feeding-related information. LH neurons express different neuropeptides that are involved in feeding behaviour, including melanocortin-concentrating hormone, orexin/hypocretin, galanin, neurotensin (Nts), and cocaine and amphetamine responsive transcript( Reference Berthoud and Munzberg 109 ). Among these LH neurons projecting to the VTA, about 20% contain orexin( Reference Fadel and Deutch 110 ), a peptide stimulating food intake( Reference Cason, Smith and Tahsili-Fahadan 111 ). Some VTA DA neurons increase their firing rate to Orexin A and Orexin B, while others are unresponsive( Reference Korotkova, Sergeeva and Eriksson 73 ). However, it has also been reported that presumed VTA GABAergic neurons are similarly excited by orexins( Reference Korotkova, Sergeeva and Eriksson 73 ). Interestingly, LH orexin neurons play a role in the rewarding properties of food, as food-induced place preference increases c-Fos activation in LH orexin neurons and pharmacological blockade of the orexin receptor decreases PR performance for high-fat foods( Reference Borgland, Chang and Bowers 69 ). Their effects on the mesDA system can be modulated by changes in diet, as high-fat food self-administration increases orexins excitability of DA neurons( Reference Borgland, Chang and Bowers 69 ). Thus, LH orexin neurons that project to the VTA might contribute to dopaminergic responses for food rewards.

Indirect leptin effects on the ventral tegmental area via the lateral hypothalamus

Within the LH, the LepR is expressed on GABAergic neurons that project to the VTA and do not express orexin or melanocortin-concentrating hormone. In vitro, leptin induces a depolarisation in one-third of these neurons, while in vivo leptin increases expression of c-Fos in these neurons( Reference Leinninger, Jo and Leshan 112 ). With respect to feeding, leptin injections into the LH decrease body weight and food intake in rats and leptin-deficient ob/ob mice. In these mice, LH leptin injections also increase VTA tyrosine hydroxylase expression and NAc DA levels( Reference Leinninger, Jo and Leshan 112 ). In a follow-up study, it was shown that some of these LepR-positive LH neurons also express the neuropeptide Nts. These LepR- and Nts-expressing LH neurons are depolarised by leptin and they project both to orexin neurons in the LH, as well as to the VTA( Reference Leinninger, Opland and Jo 70 ). Direct application of Nts into the VTA dose-dependently decreases performance on an operant responding for food, mimicking the effects of systemic leptin( Reference Kelley, Cador and Stinus 113 ). Possibly, these LepR and Nts expressing GABA neurons inhibit the mesDA system by direct actions on the VTA, and by inhibiting LH orexin neurons. Supporting this, in slices a population of orexin-positive neurons in the LH are inhibited by leptin application( Reference Yamanaka, Beuckmann and Willie 67 ). A specific knock-out mouse line in which the LepR is deleted from Nts positive neurons in the LH shows lower NAc DA levels, and less amphetamine-induced locomotor activation, a process that is thought to be dependent on striatal DA levels( Reference Leinninger, Opland and Jo 70 ). Taken together these results suggest that leptin can reduce VTA DA activity, and subsequent DA release, through stimulation of inhibitory projections originating from the LH.

Indirect ghrelin effects on the ventral tegmental area via the lateral hypothalamus

Ghrelin might also have indirect effects on DA neurons through the LH, although this has not been studied extensively. Ghrelin activation increases food intake and activates orexin, but not melanocortin-concentrating hormone-positive neurons in the LH( Reference Lawrence, Snape and Baudoin 68 ). In acute brain slices, ghrelin also activates LH orexin neurons( Reference Yamanaka, Beuckmann and Willie 67 ). Ghrelin is found in terminals forming synapses with orexin-positive LH neurons. Ghrelin-stimulated food intake is reduced when orexin signalling is diminished and blocking NPY signalling completely abolishes ghrelin-mediated increase in food intake( Reference Toshinai, Date and Murakami 114 ). Overall, this indicates that interplay between direct ghrelin effects on LH orexin neurons and arcuate nucleus originating NPY and ghrelin might contribute to ghrelin effects on food intake.

Indirect effects via the ventromedial and dorsomedial hypothalamus

In food-restricted animals, cues that are associated with food can induce increased locomotor activity. Both the ventromedial hypothalamus and dorsomedial hypothalamus have been implicated in this FAA( Reference Verhagen, Luijendijk and de Groot 115 ), which is said to reflect both hunger and the motivation to eat( Reference Merkestein, Brans and Luijendijk 116 ). Interestingly, both the ventromedial hypothalamus and dorsomedial hypothalamus are sensitive to leptin and ghrelin, and have been implicated in the expression of FAA. Whereas decreased leptin levels increases FAA( Reference Ribeiro, Ceccarini and Dupre 117 Reference Persons, Stephan and Bays 119 ), reduced ghrelin signalling attenuates FAA( Reference Verhagen, Egecioglu and Luijendijk 99 Reference Blum, Patterson and Khazall 101 Reference Verhagen, Egecioglu and Luijendijk 99 Reference Blum, Patterson and Khazall 101 , Reference Davis, Choi and Clegg 120 ). Neurons in and near the dorsomedial hypothalamus and ventromedial hypothalamus project to the VTA( Reference Greenwell, Zangen and Martin-Schild 64 ), see also( Reference Thompson, Canteras and Swanson 71 ), and could, in principle, modulate VTA activity during FAA. These neurons projecting to the VTA produce the endogenous opioids endomorphin-1 and -2. These opioids act on μ opioid receptors that are expressed on VTA GABA neurons( Reference Garzon and Pickel 121 ), and upon activation inhibit VTA GABA neurons, thus causing a disinhibition of VTA DA neurons( Reference Johnson and North 72 ). Whether these neurons express receptors for leptin or ghrelin remains to be determined.

Indirect effects of arcuate nucleus inputs on the ventral tegmental area

The arcuate nucleus has been extensively implicated in the regulation of energy balance. Two prominent neuronal subtypes are NPY/AgRP-producing neurons and POMC-producing neurons. Activation of the POMC-positive neurons induces decreases in food intake. They primarily release α-melanocyte stimulating hormone and β-endorphin, although they can also release other neurotransmitters. A subpopulation of the arcuate nucleus POMC neurons projects to the VTA( Reference King and Hentges 35 ). In slice recordings of VTA neurons, α-melanocyte stimulating hormone was not seen to have an effect on DA neurons, but excited a subpopulation of VTA GABAergic neurons( Reference Korotkova, Brown and Sergeeva 122 ). Supporting this, it has also been found that local injection of α-melanocyte stimulating hormone into the VTA increases NAc DA levels, an effect that was found to be melanocortin receptor 4 receptor dependent( Reference Lindblom, Opmane and Mutulis 123 ). Leptin is known to activate POMC and doing so could alter VTA DA activity as well. Although ghrelin does not affect POMC neurons directly it can inhibit these neurons through inhibitory projections from AgRP cells onto POMC neurons( Reference Nakazato, Murakami and Date 78 ).

Concluding remarks

Among the circuits involved in feeding behaviour, the mesDA system is important for the motivational drive for food rewards and this circuit is affected by disturbances of energy balance. VTA DA neurons encode a reward prediction error that guides associative learning processes, but it remains unknown what the direct in vivo effects are of a disturbed energy balance on activity of VTA DA neurons. Ghrelin has been shown to directly activate VTA DA neurons and inhibition of ghrelin signalling in the VTA reduces motivation and intake of palatable food. However it remains unclear to what extent VTA DA neurons are indirectly affected by distal projections of ghrelin-responsive neurons onto these DA neurons. Potentially VTA GABAergic neurons or hypothalamic neurons from the arcuate nucleus and LH play an important role in mediating the effects of ghrelin on the mesDA system. Although there are LepR expressed on VTA neurons, it remains unclear to what extent these receptors contribute to the effects of leptin on feeding behaviour. Possibly leptin affects VTA DA neuronal activity predominantly via LepR on Nts positive neurons in the LH. In addition, leptin may affect VTA DA activity via LepR expressed on POMC and NPY/AgRP neurons projecting from the arcuate to the VTA. Further insights into the effects of a disturbance on energy balance, and the effects of leptin and ghrelin on the mesDA system will require in vivo recordings from VTA DA neurons to understand the role of this system in feeding behaviour.

Acknowledgement

The authors declare no conflict of interest. The current work was supported and funded by NeuroFAST and Full4Health. NeuroFAST and Full4Heatlh are funded by the European Union Seventh Framework Programme (FP7/2007–2013) under grant agreement nos. 245009 and 266408. R.v.Z., G.v.d.P. and R.A.H.A. wrote the manuscript.

References

1. Kelley, AE, Baldo, BA, Pratt, WE et al. (2005) Corticostriatal-hypothalamic circuitry and food motivation: integration of energy, action and reward. Physiol Behav 86, 773795.Google Scholar
2. Fields, HL, Hjelmstad, GO, Margolis, EB et al. (2007) Ventral tegmental area neurons in learned appetitive behavior and positive reinforcement. Annu Rev Neurosci 30, 289316.Google Scholar
3. Narayanan, NS, Guarnieri, DJ, DiLeone, RJ (2010) Metabolic hormones, dopamine circuits, and feeding. Front Neuroendocrinol 31, 104112.Google Scholar
4. Palmiter, RD (2007) Is dopamine a physiologically relevant mediator of feeding behavior? Trends Neurosci 30, 375381.Google Scholar
5. Margolis, EB, Lock, H, Hjelmstad, GO et al. (2006) The ventral tegmental area revisited: is there an electrophysiological marker for dopaminergic neurons? J Physiol 577, 907924.Google Scholar
6. Nair-Roberts, RG, Chatelain-Badie, SD, Benson, E et al. (2008) Stereological estimates of dopaminergic, GABAergic and glutamatergic neurons in the ventral tegmental area, substantia nigra and retrorubral field in the rat. Neuroscience 152, 10241031.Google Scholar
7. Yamaguchi, T, Wang, HL, Li, X et al. (2011) Mesocorticolimbic glutamatergic pathway. J Neurosci 31, 84768490.Google Scholar
8. Schultz, W (2007) Multiple dopamine functions at different time courses. Annu Rev Neurosci 30, 259288.Google Scholar
9. Wightman, RM, Robinson, DL (2002) Transient changes in mesolimbic dopamine and their association with ‘reward’. J Neurochem 82, 721735.Google Scholar
10. Gonon, F (1997) Prolonged and extrasynaptic excitatory action of dopamine mediated by D1 receptors in the rat striatum in vivo . J Neurosci 17, 59725978.Google Scholar
11. Schultz, W, Dayan, P, Montague, PR (1997) A neural substrate of prediction and reward. Science 275, 15931599.Google Scholar
12. Schultz, W (2007) Behavioral dopamine signals. Trends Neurosci 30, 203210.Google Scholar
13. Yun, IA, Wakabayashi, KT, Fields, HL et al. (2004) The ventral tegmental area is required for the behavioral and nucleus accumbens neuronal firing responses to incentive cues. J Neurosci 24, 29232933.Google Scholar
14. Schultz, W (2010) Multiple functions of dopamine neurons. F1000 Biol Rep 2, 2.Google Scholar
15. Bromberg-Martin, ES, Matsumoto, M, Hikosaka, O (2010) Dopamine in motivational control: rewarding, aversive, and alerting. Neuron 68, 815834.Google Scholar
16. Tobler, PN, Fiorillo, CD, Schultz, W (2005) Adaptive coding of reward value by dopamine neurons. Science 307, 16421645.Google Scholar
17. Waelti, P, Dickinson, A, Schultz, W (2001) Dopamine responses comply with basic assumptions of formal learning theory. Nature 412, 4348.Google Scholar
18. Fiorillo, CD, Tobler, PN, Schultz, W (2003) Discrete coding of reward probability and uncertainty by dopamine neurons. Science 299, 18981902.Google Scholar
19. Day, JJ, Roitman, MF, Wightman, RM et al. (2007) Associative learning mediates dynamic shifts in dopamine signaling in the nucleus accumbens. Nat Neurosci 10, 10201028.Google Scholar
20. Beyene, M, Carelli, RM, Wightman, RM (2010) Cue-evoked dopamine release in the nucleus accumbens shell tracks reinforcer magnitude during intracranial self-stimulation. Neuroscience 169, 16821688.Google Scholar
21. Roitman, MF, Stuber, GD, Phillips, PE et al. (2004) Dopamine operates as a subsecond modulator of food seeking. J Neurosci 24, 12651271.Google Scholar
22. Nicola, SM, Taha, SA, Kim, SW et al. (2005) Nucleus accumbens dopamine release is necessary and sufficient to promote the behavioral response to reward-predictive cues. Neuroscience 135, 10251033.Google Scholar
23. Roesch, MR, Calu, DJ, Schoenbaum, G (2007) Dopamine neurons encode the better option in rats deciding between differently delayed or sized rewards. Nat Neurosci 10, 16151624.Google Scholar
24. Cohen, JY, Haesler, S, Vong, L et al. (2012) Neuron-type-specific signals for reward and punishment in the ventral tegmental area. Nature 482, 8588.Google Scholar
25. Salamone, JD, Wisniecki, A, Carlson, BB et al. (2001) Nucleus accumbens dopamine depletions make animals highly sensitive to high fixed ratio requirements but do not impair primary food reinforcement. Neuroscience 105, 863870.Google Scholar
26. Cousins, MS, Sokolowski, JD, Salamone, JD (1993) Different effects of nucleus accumbens and ventrolateral striatal dopamine depletions on instrumental response selection in the rat. Pharmacol Biochem Behav 46, 943951.Google Scholar
27. Ikemoto, S, Panksepp, J (1996) Dissociations between appetitive and consummatory responses by pharmacological manipulations of reward-relevant brain regions. Behav Neurosci 110, 331345.Google Scholar
28. Nowend, KL, Arizzi, M, Carlson, BB et al. (2001) D1 or D2 antagonism in nucleus accumbens core or dorsomedial shell suppresses lever pressing for food but leads to compensatory increases in chow consumption. Pharmacol Biochem Behav 69, 373382.Google Scholar
29. Szczypka, MS, Kwok, K, Brot, MD et al. (2001) Dopamine production in the caudate putamen restores feeding in dopamine-deficient mice. Neuron 30, 819828.Google Scholar
30. Ungerstedt, U (1971) Adipsia and aphagia after 6-hydroxydopamine induced degeneration of the nigro-striatal dopamine system. Acta Physiol Scand Suppl 367, 95122.Google Scholar
31. Salamone, JD, Steinpreis, RE, McCullough, LD et al. (1991) Haloperidol and nucleus accumbens dopamine depletion suppress lever pressing for food but increase free food consumption in a novel food choice procedure. Psychopharmacology (Berlin) 104, 515521.Google Scholar
32. Salamone, JD, Mahan, K, Rogers, S (1993) Ventrolateral striatal dopamine depletions impair feeding and food handling in rats. Pharmacol Biochem Behav 44, 605610.Google Scholar
33. Acosta-Galvan, G, Yi, CX, van der Vliet, J et al. (2011) Interaction between hypothalamic dorsomedial nucleus and the suprachiasmatic nucleus determines intensity of food anticipatory behavior. Proc Natl Acad Sci USA 108, 58135818.Google Scholar
34. Ribeiro, AC, LeSauter, J, Dupre, C et al. (2009) Relationship of arousal to circadian anticipatory behavior: ventromedial hypothalamus: one node in a hunger-arousal network. Eur J Neurosci 30, 17301738.Google Scholar
35. King, CM, Hentges, ST (2011) Relative number and distribution of murine hypothalamic proopiomelanocortin neurons innervating distinct target sites. PLoS One 6, e25864.Google Scholar
36. Schultz, W (2010) Dopamine signals for reward value and risk: basic and recent data. Behav Brain Funct 6, 24.Google Scholar
37. Wise, RA (2004) Dopamine, learning and motivation. Nat Rev Neurosci 5, 483494.Google Scholar
38. Wise, RA (2006) Role of brain dopamine in food reward and reinforcement. Philos Trans R Soc Lond B Biol Sci 361, 11491158.Google Scholar
39. Berridge, KC (1996) Food reward: brain substrates of wanting and liking. Neurosci Biobehav Rev 20, 125.Google Scholar
40. Berridge, KC, Robinson, TE (1998) What is the role of dopamine in reward: hedonic impact, reward learning, or incentive salience? Brain Res Brain Res Rev 28, 309369.Google Scholar
41. Witten, IB, Steinberg, EE, Lee, SY et al. (2011) Recombinase-driver rat lines: tools, techniques, and optogenetic application to dopamine-mediated reinforcement. Neuron 72, 721733.Google Scholar
42. Adamantidis, AR, Tsai, HC, Boutrel, B et al. (2011) Optogenetic interrogation of dopaminergic modulation of the multiple phases of reward-seeking behavior. J Neurosci 31, 1082910835.Google Scholar
43. Tsai, HC, Zhang, F, Adamantidis, A et al. (2009) Phasic firing in dopaminergic neurons is sufficient for behavioral conditioning. Science 324, 10801084.Google Scholar
44. Pothos, EN, Creese, I, Hoebel, BG (1995) Restricted eating with weight loss selectively decreases extracellular dopamine in the nucleus accumbens and alters dopamine response to amphetamine, morphine, and food intake. J Neurosci 15, 66406650.Google Scholar
45. Verhagen, LA, Luijendijk, MC, Korte-Bouws, GA et al. (2009) Dopamine and serotonin release in the nucleus accumbens during starvation-induced hyperactivity. Eur Neuropsychopharmacol 19, 309316.Google Scholar
46. Stuber, GD, Evans, SB, Higgins, MS et al. (2002) Food restriction modulates amphetamine-conditioned place preference and nucleus accumbens dopamine release in the rat. Synapse 46, 8390.Google Scholar
47. Cadoni, C, Solinas, M, Valentini, V et al. (2003) Selective psychostimulant sensitization by food restriction: differential changes in accumbens shell and core dopamine. Eur J Neurosci 18, 23262334.Google Scholar
48. Hernandez, L, Hoebel, BG (1988) Food reward and cocaine increase extracellular dopamine in the nucleus accumbens as measured by microdialysis. Life Sci 42, 17051712.Google Scholar
49. Carr, KD (2007) Chronic food restriction: enhancing effects on drug reward and striatal cell signaling. Physiol Behav 91, 459472.Google Scholar
50. Fulton, S (2010) Appetite and reward. Front Neuroendocrinol 31, 85103.Google Scholar
51. Sulzer, D, Chen, TK, Lau, YY et al. (1995) Amphetamine redistributes dopamine from synaptic vesicles to the cytosol and promotes reverse transport. J Neurosci 15, 41024108.Google Scholar
52. Zhang, M, Balmadrid, C, Kelley, AE (2003) Nucleus accumbens opioid, GABaergic, and dopaminergic modulation of palatable food motivation: contrasting effects revealed by a progressive ratio study in the rat. Behav Neurosci 117, 202211.Google Scholar
53. Domingos, AI, Vaynshteyn, J, Voss, HU et al. (2011) Leptin regulates the reward value of nutrient. Nat Neurosci 14, 15621568.Google Scholar
54. Aberman, JE, Ward, SJ, Salamone, JD (1998) Effects of dopamine antagonists and accumbens dopamine depletions on time-constrained progressive-ratio performance. Pharmacol Biochem Behav 61, 341348.Google Scholar
55. Figlewicz, DP, Bennett, JL, Naleid, AM et al. (2006) Intraventricular insulin and leptin decrease sucrose self-administration in rats. Physiol Behav 89, 611616.Google Scholar
56. la Fleur, SE, Vanderschuren, LJ, Luijendijk, MC et al. (2007) A reciprocal interaction between food-motivated behavior and diet-induced obesity. Int J Obes (Lond) 31, 12861294.Google Scholar
57. Alsio, J, Olszewski, PK, Norback, AH et al. (2010) Dopamine D1 receptor gene expression decreases in the nucleus accumbens upon long-term exposure to palatable food and differs depending on diet-induced obesity phenotype in rats. Neuroscience 171, 779787.Google Scholar
58. Johnson, PM, Kenny, PJ (2010) Dopamine D2 receptors in addiction-like reward dysfunction and compulsive eating in obese rats. Nat Neurosci 13, 635641.Google Scholar
59. Wang, GJ, Volkow, ND, Logan, J et al. (2001) Brain dopamine and obesity. Lancet 357, 354357.Google Scholar
60. Li, Y, South, T, Han, M et al. (2009) High-fat diet decreases tyrosine hydroxylase mRNA expression irrespective of obesity susceptibility in mice. Brain Res 1268, 181189.Google Scholar
61. Davis, JF, Tracy, AL, Schurdak, JD et al. (2008) Exposure to elevated levels of dietary fat attenuates psychostimulant reward and mesolimbic dopamine turnover in the rat. Behav Neurosci 122, 12571263.Google Scholar
62. Wang, GJ, Volkow, ND, Fowler, JS (2002) The role of dopamine in motivation for food in humans: implications for obesity. Expert Opin Ther Targets 6, 601609.Google Scholar
63. Philpot, KB, Dallvechia-Adams, S, Smith, Y et al. (2005) A cocaine-and-amphetamine-regulated-transcript peptide projection from the lateral hypothalamus to the ventral tegmental area. Neuroscience 135, 915925.Google Scholar
64. Greenwell, TN, Zangen, A, Martin-Schild, S et al. (2002) Endomorphin-1 and -2 immunoreactive cells in the hypothalamus are labeled by fluoro-gold injections to the ventral tegmental area. J Comp Neurol 454, 320328.Google Scholar
65. Hommel, JD, Trinko, R, Sears, RM et al. (2006) Leptin receptor signaling in midbrain dopamine neurons regulates feeding. Neuron 51, 801810.Google Scholar
66. Abizaid, A, Liu, ZW, Andrews, ZB et al. (2006) Ghrelin modulates the activity and synaptic input organization of midbrain dopamine neurons while promoting appetite. J Clin Invest 116, 32293239.Google Scholar
67. Yamanaka, A, Beuckmann, CT, Willie, JT et al. (2003) Hypothalamic orexin neurons regulate arousal according to energy balance in mice. Neuron 38, 701713.Google Scholar
68. Lawrence, CB, Snape, AC, Baudoin, FM et al. (2002) Acute central ghrelin and GH secretagogues induce feeding and activate brain appetite centers. Endocrinology 143, 155162.Google Scholar
69. Borgland, SL, Chang, SJ, Bowers, MS et al. (2009) Orexin A/hypocretin-1 selectively promotes motivation for positive reinforcers. J Neurosci 29, 1121511225.Google Scholar
70. Leinninger, GM, Opland, DM, Jo, YH et al. (2011) Leptin action via neurotensin neurons controls orexin, the mesolimbic dopamine system and energy balance. Cell Metab 14, 313323.Google Scholar
71. Thompson, RH, Canteras, NS, Swanson, LW (1996) Organization of projections from the dorsomedial nucleus of the hypothalamus: a PHA-L study in the rat. J Comp Neurol 376, 143173.Google Scholar
72. Johnson, SW, North, RA (1992) Opioids excite dopamine neurons by hyperpolarization of local interneurons. J Neurosci 12, 483488.Google Scholar
73. Korotkova, TM, Sergeeva, OA, Eriksson, KS et al. (2003) Excitation of ventral tegmental area dopaminergic and nondopaminergic neurons by orexins/hypocretins. J Neurosci 23, 711.Google Scholar
74. Cheung, CC, Clifton, DK, Steiner, RA (1997) Proopiomelanocortin neurons are direct targets for leptin in the hypothalamus. Endocrinology 138, 44894492.Google Scholar
75. Schwartz, MW, Woods, SC, Porte, D Jr et al. (2000) Central nervous system control of food intake. Nature 404, 661671.Google Scholar
76. Schwartz, MW, Seeley, RJ, Campfield, LA et al. (1996) Identification of targets of leptin action in rat hypothalamus. J Clin Invest 98, 11011106.Google Scholar
77. van den Top, M, Lee, K, Whyment, AD et al. (2004) Orexigen-sensitive NPY/AgRP pacemaker neurons in the hypothalamic arcuate nucleus. Nat Neurosci 7, 493494.Google Scholar
78. Nakazato, M, Murakami, N, Date, Y et al. (2001) A role for ghrelin in the central regulation of feeding. Nature 409, 194198.Google Scholar
79. Campfield, LA, Smith, FJ, Guisez, Y et al. (1995) Recombinant mouse OB protein: evidence for a peripheral signal linking adiposity and central neural networks. Science 269, 546549.Google Scholar
80. Grill, HJ, Schwartz, MW, Kaplan, JM et al. (2002) Evidence that the caudal brainstem is a target for the inhibitory effect of leptin on food intake. Endocrinology 143, 239246.Google Scholar
81. Pelleymounter, MA, Cullen, MJ, Baker, MB et al. (1995) Effects of the obese gene product on body weight regulation in ob/ob mice. Science 269, 540543.Google Scholar
82. Zhang, Y, Proenca, R, Maffei, M et al. (1994) Positional cloning of the mouse obese gene and its human homologue. Nature 372, 425432.Google Scholar
83. Speakman, J, Hambly, C, Mitchell, S et al. (2007) Animal models of obesity. Obes Rev 8, Suppl. 1, 5561.Google Scholar
84. Morton, GJ, Blevins, JE, Kim, F et al. (2009) The action of leptin in the ventral tegmental area to decrease food intake is dependent on Jak-2 signaling. Am J Physiol Endocrinol Metab 297, E202E210.Google Scholar
85. Fulton, S, Pissios, P, Manchon, RP et al. (2006) Leptin regulation of the mesoaccumbens dopamine pathway. Neuron 51, 811822.Google Scholar
86. Figlewicz, DP, Evans, SB, Murphy, J et al. (2003) Expression of receptors for insulin and leptin in the ventral tegmental area/substantia nigra (VTA/SN) of the rat. Brain Res 964, 107115.Google Scholar
87. Leshan, RL, Opland, DM, Louis, GW et al. (2010) Ventral tegmental area leptin receptor neurons specifically project to and regulate cocaine- and amphetamine-regulated transcript neurons of the extended central amygdala. J Neurosci 30, 57135723.Google Scholar
88. Krugel, U, Schraft, T, Kittner, H et al. (2003) Basal and feeding-evoked dopamine release in the rat nucleus accumbens is depressed by leptin. Eur J Pharmacol 482, 185187.Google Scholar
89. Liu, J, Perez, SM, Zhang, W et al. (2011) Selective deletion of the leptin receptor in dopamine neurons produces anxiogenic-like behavior and increases dopaminergic activity in amygdala. Mol Psychiatry 16, 10241038.Google Scholar
90. Roseberry, AG, Painter, T, Mark, GP et al. (2007) Decreased vesicular somatodendritic dopamine stores in leptin-deficient mice. J Neurosci 27, 70217027.Google Scholar
91. Pfaffly, J, Michaelides, M, Wang, GJ et al. (2010) Leptin increases striatal dopamine D2 receptor binding in leptin-deficient obese (ob/ob) mice. Synapse 64, 503510.Google Scholar
92. Thanos, PK, Michaelides, M, Piyis, YK et al. (2008) Food restriction markedly increases dopamine D2 receptor (D2R) in a rat model of obesity as assessed with in-vivo muPET imaging ([11C] raclopride) and in-vitro ([3H] spiperone) autoradiography. Synapse 62, 5061.Google Scholar
93. Matheny, M, Shapiro, A, Tumer, N et al. (2011) Region-specific diet-induced and leptin-induced cellular leptin resistance includes the ventral tegmental area in rats. Neuropharmacology 60, 480487.Google Scholar
94. Wettschureck, N, Moers, A, Wallenwein, B et al. (2005) Loss of Gq/11 family G proteins in the nervous system causes pituitary somatotroph hypoplasia and dwarfism in mice. Mol Cell Biol 25, 19421948.Google Scholar
95. Zigman, JM, Jones, JE, Lee, CE et al. (2006) Expression of ghrelin receptor mRNA in the rat and the mouse brain. J Comp Neurol 494, 528548.Google Scholar
96. Mondal, MS, Date, Y, Yamaguchi, H et al. (2005) Identification of ghrelin and its receptor in neurons of the rat arcuate nucleus. Regul Pept 126, 5559.Google Scholar
97. Tschop, M, Smiley, DL, Heiman, ML (2000) Ghrelin induces adiposity in rodents. Nature 407, 908913.Google Scholar
98. Horvath, TL, Diano, ST, Schop, M (2003) Ghrelin in hypothalamic regulation of energy balance. Curr Top Med Chem 3, 921927.Google Scholar
99. Verhagen, LA, Egecioglu, E, Luijendijk, MC et al. (2011) Acute and chronic suppression of the central ghrelin signaling system reveals a role in food anticipatory activity. Eur Neuropsychopharmacol 21, 384392.Google Scholar
100. LeSauter, J, Hoque, N, Weintraub, M et al. (2009) Stomach ghrelin-secreting cells as food-entrainable circadian clocks. Proc Natl Acad Sci USA 106, 1358213587.Google Scholar
101. Blum, ID, Patterson, Z, Khazall, R et al. (2009) Reduced anticipatory locomotor responses to scheduled meals in ghrelin receptor deficient mice. Neuroscience 164, 351359.Google Scholar
102. Guan, XM, Yu, H, Palyha, OC et al. (1997) Distribution of mRNA encoding the growth hormone secretagogue receptor in brain and peripheral tissues. Brain Res Mol Brain Res 48, 2329.Google Scholar
103. Quarta, D, Di Francesco, C, Melotto, S et al. (2009) Systemic administration of ghrelin increases extracellular dopamine in the shell but not the core subdivision of the nucleus accumbens. Neurochem Int 54, 8994.Google Scholar
104. Skibicka, KP, Hansson, C, Alvarez-Crespo, M et al. (2011) Ghrelin directly targets the ventral tegmental area to increase food motivation. Neuroscience 180, 129137.Google Scholar
105. King, SJ, Isaacs, AM, O'Farrell, E et al. (2011) Motivation to obtain preferred foods is enhanced by ghrelin in the ventral tegmental area. Horm Behav 60, 572580.Google Scholar
106. Naleid, AM, Grace, MK, Cummings, DE et al. (2005) Ghrelin induces feeding in the mesolimbic reward pathway between the ventral tegmental area and the nucleus accumbens. Peptides 26, 22742279.Google Scholar
107. Jerlhag, E, Egecioglu, E, Dickson, SL et al. (2007) Ghrelin administration into tegmental areas stimulates locomotor activity and increases extracellular concentration of dopamine in the nucleus accumbens. Addict Biol 12, 616.Google Scholar
108. Egecioglu, E, Jerlhag, E, Salome, N et al. (2010) Ghrelin increases intake of rewarding food in rodents. Addict Biol 15, 304311.Google Scholar
109. Berthoud, HR, Munzberg, H (2011) The lateral hypothalamus as integrator of metabolic and environmental needs: from electrical self-stimulation to opto-genetics. Physiol Behav 104, 2939.Google Scholar
110. Fadel, J, Deutch, AY (2002) Anatomical substrates of orexin-dopamine interactions: lateral hypothalamic projections to the ventral tegmental area. Neuroscience 111, 379387.Google Scholar
111. Cason, AM, Smith, RJ, Tahsili-Fahadan, P et al. (2010) Role of orexin/hypocretin in reward-seeking and addiction: implications for obesity. Physiol Behav 100, 419428.Google Scholar
112. Leinninger, GM, Jo, YH, Leshan, RL et al. (2009) Leptin acts via leptin receptor-expressing lateral hypothalamic neurons to modulate the mesolimbic dopamine system and suppress feeding. Cell Metab 10, 8998.Google Scholar
113. Kelley, AE, Cador, M, Stinus, L et al. (1989) Neurotensin, substance P, neurokinin-alpha, and enkephalin: injection into ventral tegmental area in the rat produces differential effects on operant responding. Psychopharmacology (Berlin) 97, 243252.Google Scholar
114. Toshinai, K, Date, Y, Murakami, N et al. (2003) Ghrelin-induced food intake is mediated via the orexin pathway. Endocrinology 144, 15061512.Google Scholar
115. Verhagen, LA, Luijendijk, MC, de Groot, JW et al. (2011) Anticipation of meals during restricted feeding increases activity in the hypothalamus in rats. Eur J Neurosci 34, 14851491.Google Scholar
116. Merkestein, M, Brans, MA, Luijendijk, MC et al. (2012) Ghrelin mediates anticipation to a palatable meal in rats. Obesity (Silver Spring) 20, 963971.Google Scholar
117. Ribeiro, AC, Ceccarini, G, Dupre, C et al. (2011) Contrasting effects of leptin on food anticipatory and total locomotor activity. PLoS One 6, e23364.Google Scholar
118. Mistlberger, RE, Marchant, EG (1999) Enhanced food-anticipatory circadian rhythms in the genetically obese Zucker rat. Physiol Behav 66, 329335.Google Scholar
119. Persons, JE, Stephan, FK, Bays, ME (1993) Diet-induced obesity attenuates anticipation of food access in rats. Physiol Behav 54, 5564.Google Scholar
120. Davis, JF, Choi, DL, Clegg, DJ et al. (2011) Signaling through the ghrelin receptor modulates hippocampal function and meal anticipation in mice. Physiol Behav 103, 3943.Google Scholar
121. Garzon, M, Pickel, VM (2001) Plasmalemmal mu-opioid receptor distribution mainly in nondopaminergic neurons in the rat ventral tegmental area. Synapse 41, 311328.Google Scholar
122. Korotkova, TM, Brown, RE, Sergeeva, OA et al. (2006) Effects of arousal- and feeding-related neuropeptides on dopaminergic and GABAergic neurons in the ventral tegmental area of the rat. Eur J Neurosci 23, 26772685.Google Scholar
123. Lindblom, J, Opmane, B, Mutulis, F et al. (2001) The MC4 receptor mediates alpha-MSH induced release of nucleus accumbens dopamine. Neuroreport 12, 21552158.Google Scholar
Figure 0

Fig. 1. Schematic representation of ventral tegmental area (VTA) inputs from the hypothalamus which are modulated by leptin and ghrelin. VTA dopaminergic neuronal activity is decreased by leptin(65) and increased by ghrelin(66). Orexin-producing neurons from the lateral hypothalamus (LH) are inhibited by leptin(67) and excited by ghrelin(67,68). Orexin A increases excitatory projections onto VTA dopamine (DA) neurons(69). LH neurotensin (Nts)/γ-aminobutyric acid (GABA)-producing neurons are excited by leptin, and project onto LH orexin neurons, as well as VTA DA neurons(70). VTA GABA neurons directly inhibit VTA DA neurons, and are also modulated by hypothalamic inputs. Endomorphin-producing neurons from the ventromedial hypothalamus (VMH) project to the VTA(71), and opioid release in the VTA inhibits VTA GABA neurons(72). It remains to be investigated whether these hypothalamic neurons are also leptin or ghrelin responsive. Pro-opiomelanocortin (POMC) neurons from the arcuate nucleus project to the VTA(35). They produce α-melanocyte stimulating hormone that excites VTA GABA neurons(73), and are themselves excited by leptin(74,75). These POMC neurons are inhibited by arcuate nucleus agouti-related protein (AgRP) neurons that in turn are inhibited by leptin and excited by ghrelin(7678). Note that this schematic only includes hypothalamic inputs onto the VTA that have been described in the literature, but other hypothalamic inputs may exist. Also, projections shown here do not represent all the known projections of the different neuronal subtypes.