Elsevier

Physiology & Behavior

Volume 100, Issue 5, 14 July 2010, Pages 419-428
Physiology & Behavior

Role of orexin/hypocretin in reward-seeking and addiction: Implications for obesity

https://doi.org/10.1016/j.physbeh.2010.03.009Get rights and content

Abstract

Orexins (also named hypocretins) are recently discovered neuropeptides made exclusively in the hypothalamus. Recent studies have shown that orexin cells located specifically in lateral hypothalamus (LH) are involved in motivated behavior for drugs of abuse as well as natural rewards. Administration of orexin has been shown to stimulate food consumption, and orexin signaling in VTA has been implicated in intake of high-fat food. In self-administration studies, the orexin 1 receptor antagonist SB-334867 (SB) attenuated operant responding for high-fat pellets, sucrose pellets and ethanol, but not cocaine, demonstrating that signaling at orexin receptors is necessary for reinforcement of specific rewards. The orexin system is also implicated in associations between rewards and relevant stimuli. For example, Fos expression in LH orexin neurons varied in proportion to conditioned place preference (CPP) for food, morphine, or cocaine. This Fos expression was altered accordingly for CPP administered during protracted abstinence from morphine or cocaine, when preference for natural rewards was decreased and drug preference was increased. Additionally, orexin has been shown to be involved in reward–stimulus associations in the self-administration paradigm, where SB attenuated cue-induced reinstatement of extinguished sucrose- or cocaine-seeking. Although the specific circuitry mediating the effects of orexin on food reward remains unknown, VTA seems likely to be a critical target for at least some of these orexin actions. Thus, recent studies have established a role for orexin in reward-based feeding, and further investigation is warranted for determining whether function/dysfunction of the orexin system may contribute to the overeating associated with obesity.

The paper represents an invited review by a symposium, award winner or keynote speaker at the Society for the Study of Ingestive Behavior [SSIB] Annual Meeting in Portland, July 2009.

Introduction

The neuropeptides orexin A and orexin B (also denoted hypocretin 1 and hypocretin 2) were discovered in the late nineties and are synthesized solely in hypothalamic neurons [1], [2]. Sakurai et al. characterized two individual receptors for the orexin system, termed OxR1 and OxR2 (also denoted HcrtR1 and HcrtR2) [2]. OxR1 binds both orexin A and orexin B but has a much lower affinity for orexin B, whereas OxR2 binds both peptides with a similar high affinity. Both receptors couple with G proteins; OxR1 couples exclusively with Gq subunits while OxR2 couples to both Gq and Gi/o[3]. Orexin neurons send dense fiber projections throughout the brain including cerebral cortex, hippocampus, thalamus, midbrain and spinal cord [4], [5]. Furthermore, OxR1 and OxR2 are widely distributed throughout the CNS but are largely non-overlapping and are regionally selective [6], [7], [8], [9].

Orexin has been implicated in feeding behavior, in keeping with the location of these cells in the area of lateral hypothalamus (LH) specifically associated with feeding. The initial study by Sakurai et al. [2] demonstrated that central administration of orexin A or orexin B into the lateral ventricle stimulated food consumption, which prompted them to name the new peptides “orexin”, meaning appetite [2]. Furthermore, this study showed that prepro-orexin mRNA is upregulated following fasting [2]. Subsequent studies confirmed that orexin A stimulated feeding, whereas systemic administration of the OxR1 antagonist SB-334867 (SB) reduced feeding [10], [11], [12], [13], [14], [15]. Importantly, it was shown that SB reduced feeding by selectively enhancing the behavioral satiety sequence rather than through aversive mechanisms [11], [12], [13], [14]. Other studies showed that the orexin neuronal field has reciprocal connections with areas known to regulate food intake including neuropeptide Y (NPY) neurons in arcuate nucleus [16], [17], and that orexin A-induced feeding is partially attenuated by antagonism of the NPY-Y1 receptor [18]. Furthermore, obese mice (ob/ob and db/db) show decreased prepro-orexin gene expression [19], and antagonism of OxR1 produced a greater reduction of high-fat intake in Osborne–Mendel rats, a rat strain susceptible to dietary-induced obesity, as compared to obesity-resistant rats [20]. Taken together, these findings led to the view that the orexin neuropeptides modulate a homeostatic central feedback mechanism regulating feeding.

Further interest in the orexin system was generated when concurrent studies implicated it in another homeostatic function, sleep–wakefulness. Two groups simultaneously reported that dysfunction in the orexin system was linked with narcoleptic symptoms in mice and dogs [21], [22]. Later work in humans showed that narcoleptics (with cataplexy) lack orexin in their CSF and lack most orexin neurons in posterior hypothalamus [23], [24], [25]. Together these findings led to the view that the orexin system is involved in arousal and maintenance of the waking state. In support of this view, additional findings showed that orexin neurons send dense projections to brain arousal areas including the locus coeruleus, tuberomammillary nucleus, and basal forebrain cholinergic system [4], [5], [26], [27] and that orexin application typically strongly activates these cells [17], [28], [29], [30]. Together, the findings that the orexin system is involved in both feeding and arousal led to the hypothesis that the primary function of this system was in promoting arousal in response to food deprivation and in a fashion to promote food consumption [1], [2]. Recent studies, however, have investigated a possible role for orexins in reward-seeking for food and drugs of abuse independently of deprivation. As discussed below, this reward-associated function of the orexin system may be separated from its role in homeostatic feeding and maintenance of the arousal state, and may be mediated by a separate population of (laterally located) orexin neurons.

Much attention has been focused recently on the role of orexin in drug reward (reviewed in [[30], [31], [32], [33]]); however, with the current growing obesity epidemic, several studies have now linked the orexin system to reward-based eating. Dysfunction of the orexin system may be a contributing factor in the overeating associated with obesity. This seems a plausible hypothesis given the fact that hypothalamus is optimally located within the brain to communicate with lower brainstem nuclei controlling homeostatic processes and higher cortical and limbic areas associated with motivation. Furthermore, there is overlapping neural circuitry involved in food and drug reward [34]. We recently reviewed orexin's role in drug addiction [30], [31]. Therefore, this review will focus on orexin's role in reward-based feeding and will only briefly highlight studies involving drug reward.

Section snippets

Orexin and brain sites involved in reward-based eating

Endogenous opioids in nucleus accumbens (NAc) play an important role in control of appetite and are postulated to mediate the hedonic aspects of food intake [35]. Rats will consume highly palatable foods even when sated, and overriding of homeostatic control mechanisms by rewarding aspects of food has been postulated to contribute to obesity [34], [36]. A neural connection between NAc, a critical site for the regulation of reward-related behaviors, and hypothalamic orexin neurons may mediate

Orexin and food–stimulus associations

Orexin neurons in LH also play an active role in stimulus-driven reward-seeking. A strong association was found between Fos activation in orexin neurons and the expression of conditioned place preference (CPP) for food, cocaine, or morphine in rats [46]. Notably, this Fos induction was directly proportional to the degree of preference that animals exhibited on the CPP test day (r = 0.72 to 0.90, p < 0.01). Moreover, this correspondence between behavior and Fos induction was selective for orexin

Orexin and cocaine–stimulus associations

As noted above, our lab has shown that LH orexin neurons exhibit conditioned responses to cocaine, morphine and food-associated contexts in proportion to behavioral preference [46]. In addition, systemic administration of the orexin 1 receptor antagonist SB-334867 (SB) attenuated expression of morphine preference [46]. Preliminary studies from our laboratory indicate that SB has similar effects on cocaine preference (G. Sartor & G. Aston-Jones, unpublished data). In this study,

Orexin and ethanol intake

Several studies have implicated the orexin system in ethanol consumption and abuse. The area of LH expressing orexin mRNA is increased after ethanol drinking in alcohol-preferring rats [64]. As noted above, SB decreased operant responding for ethanol and attenuated cue-and yohimbine-induced reinstatement of ethanol-seeking [[41], [64]]. Furthermore, there was increased Fos expression in orexin neurons following context-induced reinstatement of ethanol-seeking [47].

Recent studies from our

Orexin and altered hedonic processing during protracted abstinence

Chronic exposure to cocaine or morphine followed by protracted forced abstinence results in dramatically altered preferences for natural and drug rewards. Specifically, preference for environments associated with natural rewards such as food or novelty is decreased, whereas preference for drug (morphine or cocaine) is increased in a CPP paradigm following 2 or 5 weeks of protracted forced abstinence [73], [74], [75], [76], [77]. This increased interest in drug and decreased interest in natural

Functional differences for LH versus DMH/PeF orexin neurons

As reviewed above, orexin neurons appear to be involved both in homeostatic processes that modulate arousal as well as feeding and reward-seeking; additional evidence indicates that orexin neurons are functionally dichotomous [79]. Thus, reward-seeking functions are associated primarily with orexin cells in LH, whereas arousal- and stress-related processes are linked with orexin neurons in the DMH and PeF [79]. Support for this hypothesis comes from several findings: PeF and DMH, but not LH,

Discussion

As described above, orexin is involved in reward-based feeding. Central orexin administration stimulated free-feeding in sated rats and antagonism of OxR1 blocked this effect. OxR1 antagonism in VTA attenuated high-fat feeding in sated rats induced by DAMGO in NAc. Additionally, self-administration studies found that central orexin administration increased fixed and progressive ratio responding for sweets, while recent studies from our lab have shown that OxR1 antagonism attenuates responding

References (95)

  • Y. Yamamoto et al.

    Down regulation of the prepro-orexin gene expression in genetically obese mice

    Brain Res Mol Brain Res

    (1999)
  • C.L. White et al.

    Effect of a selective OX1R antagonist on food intake and body weight in two strains of rats that differ in susceptibility to dietary-induced obesity

    Peptides

    (2005)
  • R.M. Chemelli et al.

    Narcolepsy in orexin knockout mice: molecular genetics of sleep regulation

    Cell

    (1999)
  • L. Lin et al.

    The sleep disorder canine narcolepsy is caused by a mutation in the hypocretin (orexin) receptor 2 gene

    Cell

    (1999)
  • S. Nishino et al.

    Hypocretin (orexin) deficiency in human narcolepsy

    Lancet

    (2000)
  • S. Nishino

    Clinical and neurobiological aspects of narcolepsy

    Sleep Med

    (2007)
  • E. Eggermann et al.

    Orexins/hypocretins excite basal forebrain cholinergic neurones

    Neuroscience

    (2001)
  • G. Aston-Jones et al.

    Role of lateral hypothalamic orexin neurons in reward processing and addiction

    Neuropharmacology

    (2009)
  • H. Zheng et al.

    Eating for pleasure or calories

    Curr Opin Pharmacol

    (2007)
  • C.V. Dayas et al.

    Stimuli linked to ethanol availability activate hypothalamic CART and orexin neurons in a reinstatement model of relapse

    Biol Psychiatry

    (2008)
  • H. Moore et al.

    Bidirectional modulation of stimulated cortical acetylcholine release by benzodiazephine receptor ligands

    Brain Res

    (1993)
  • F.M. Inglis et al.

    Enhanced acetylcholine release in hippocampus and cortex during anticipation and consumption of a palatable meal

    Neuroscience

    (1994)
  • D. Frederick-Duus et al.

    Food-elicited increases in cortical acetylcholine release require orexin transmission

    Neuroscience

    (2007)
  • R.J. McLaughlin et al.

    The role of different subregions of the basolateral amygdala in cue-induced reinstatement and extinction of food-seeking

    Neuroscience

    (2007)
  • S.B. Floresco et al.

    Opposing roles for the nucleus accumbens core and shell in cue-induced reinstatement of food-seeking behavior

    Behav Brain Res

    (2008)
  • G.J. Kirouac et al.

    Topographical organization of the nucleus accumbens afferents from the basolateral amygdala and efferents to the lateral hypothalamus

    Neuroscience

    (1995)
  • A.S. Hamlin et al.

    Renewal of an extinguished instrumental response: neural correlates and the role of D1 dopamine receptors

    Neuroscience

    (2006)
  • H.S. Crombag et al.

    Effect of dopamine receptor antagonists on renewal of cocaine seeking by reexposure to drug-associated contextual cues

    Neuropsychopharmacology

    (2002)
  • R.J. Smith et al.

    Orexin/hypocretin is necessary for context-driven cocaine-seeking

    Neuropharmacology

    (2010)
  • M.A. Akanmu et al.

    Selective stimulation of orexin receptor type 2 promotes wakefulness in freely behaving rats

    Brain Res

    (2005)
  • J.T. Willie et al.

    Distinct narcolepsy syndromes in orexin receptor-2 and orexin null mice: molecular genetic dissection of non-REM and REM sleep regulatory processes

    Neuron

    (2003)
  • D.E. Moorman et al.

    Orexin-1 receptor antagonism decreases ethanol consumption and preference selectively in high-ethanol-preferring Sprague Dawley rats

    Alcohol

    (2009)
  • G. Aston-Jones et al.

    Brain substrates for increased drug seeking during protracted withdrawal

    Neuropharmacology

    (2004)
  • G.C. Harris et al.

    Augmented accumbal serotonin levels decrease the preference for a morphine associated environment during withdrawal

    Neuropsychopharmacology

    (2001)
  • G.C. Harris et al.

    Lateral hypothalamic orexin neurons are critically involved in learning to associate an environment with morphine reward

    Behav Brain Res

    (2007)
  • G. Harris et al.

    Arousal and reward: a dichotomy in orexin function

    Trends Neurosci

    (2006)
  • R. Sharf et al.

    Orexin mediates the expression of precipitated morphine withdrawal and concurrent activation of the nucleus accumbens shell

    Biol Psychiatry

    (2008)
  • J. Fadel et al.

    Anatomical substrates of orexin–dopamine interactions: lateral hypothalamic projections to the ventral tegmental area

    Neuroscience

    (2002)
  • B.E. Schroeder et al.

    A common profile of prefrontal cortical activation following exposure to nicotine- or chocolate-associated contextual cues

    Neuroscience

    (2001)
  • A.E. Kelley et al.

    Neural systems recruited by drug- and food-related cues: studies of gene activation in corticolimbic regions

    Physiol Behav

    (2005)
  • L. de Lecea et al.

    The hypocretins: hypothalamus-specific peptides with neuroexcitatory activity

    Proc Natl Acad Sci U. S. A.

    (1998)
  • C. Peyron et al.

    Neurons containing hypocretin (orexin) project to multiple neuronal systems

    J Neurosci

    (1998)
  • J.G. Sutcliffe et al.

    The hypocretins: setting the arousal threshold

    Nat Rev Neurosci

    (2002)
  • T.S. Kilduff et al.

    Mapping of the mRNAs for the hypocretin/orexin and melanin-concentrating hormone receptors: networks of overlapping peptide systems

    J Comp Neurol

    (2001)
  • J.N. Marcus et al.

    Differential expression of orexin receptors 1 and 2 in the rat brain

    J Comp Neurol

    (2001)
  • R.J. Rodgers et al.

    SB-334867, a selective orexin-1 receptor antagonist, enhances behavioural satiety and blocks the hyperphagic effect of orexin-A in rats

    Eur J NeuroSci

    (2001)
  • C. Broberger et al.

    Hypocretin/orexin- and melanin-concentrating hormone-expressing cells form distinct populations in the rodent hypothalamus: relationship to neuropeptide Y and agouti gene-related protein systems

    J Comp Neurol

    (1998)
  • Cited by (197)

    View all citing articles on Scopus
    View full text