Elsevier

Brain Research

Volume 1314, 16 February 2010, Pages 74-90
Brain Research

Research Report
Lateral hypothalamic orexin/hypocretin neurons: A role in reward-seeking and addiction

https://doi.org/10.1016/j.brainres.2009.09.106Get rights and content

Abstract

Orexins (synonymous with hypocretins) are recently discovered neuropeptides made exclusively in hypothalamus. Behavioral, anatomical, and neurophysiological studies show that a subset of these cells, specifically those in lateral hypothalamus (LH), are involved in reward processing and addictive behaviors. Fos expression in LH orexin neurons varied in proportion to conditioned place preference (CPP) for morphine, cocaine, or food. This relationship occurred both in drug-naïve rats and in animals during protracted morphine withdrawal, when drug preference was elevated but food preference was decreased. Inputs to the LH orexin cell field from lateral septum and bed nucleus of the stria terminalis were Fos-activated during cocaine CPP in proportion to the preference expressed in each animal. This implies that these inputs may be involved in driving the conditioned responses in LH orexin neurons. Related studies showed that LH orexin neurons that project to ventral tegmental area (VTA) had greater Fos induction in association with elevated morphine preference during protracted withdrawal than non-VTA-projecting orexin neurons, indicating that the VTA is an important site of action for orexin's role in reward processing. In addition, stimulation of LH orexin neurons, or microinjection of orexin into VTA, reinstated an extinguished morphine preference. In self-administration studies, the orexin 1 receptor antagonist SB-334867 (SB) blocked cocaine-seeking induced by discrete or contextual cues previously associated with cocaine, but not by a priming injection of cocaine. There was no effect of SB on cocaine self-administration itself, indicating that it did not interfere with the drug's reinforcing properties. Neurophysiological studies revealed that locally applied orexin often augmented responses of VTA dopamine (DA) neurons to activation of the medial prefrontal cortex (mPFC), consistent with the view that orexin facilitates activation of VTA DA neurons by stimulus–reward associations. This LH-to-VTA orexin pathway was found to be necessary for learning a morphine place preference. These findings are consistent with results showing that orexin facilitates glutamate-mediated responses, and is necessary for glutamate-dependent long-term potentiation in VTA DA neurons. We surmise from these studies that LH orexin neurons play an important role in reward processing and addiction and that LH orexin cells are an important input to VTA for behavioral effects associated with reward-paired stimuli.

Introduction

The neuropeptides orexin A and orexin B (synonymous with hypocretin 1 and hypocretin 2) are produced from a prepro-orexin molecule made solely in hypothalamic neurons. Since the discoveries by de Lecea et al. (1998) and Sakurai et al. (1998), considerable work has characterized this neurotransmitter system. Sakurai et al. (1998) characterized two receptors for the orexin system, termed OxR1 and OxR2 (also denoted HcrtR1 and HcrtR2). OxR1 binds orexin A with 30 nM affinity but has much lower affinity for orexin B, whereas OxR2 binds both orexin peptides with similar high affinity. Further, OxR1 is coupled exclusively to a Gq subclass of G proteins, and OxR2 is coupled to both Gq and Gi/o proteins (Zhu et al., 2003). The orexin neurons give rise to a highly divergent system of fiber projections that spans the entire neuraxis, including innervation in the cerebral cortex, hippocampus, thalamus, midbrain, and spinal cord (Peyron et al., 1998, Sutcliffe and de Lecea, 2002). Likewise, the two orexin receptors are widely distributed in the CNS but are regionally selective and largely non-overlapping (Kilduff and de Lecea, 2001, Lu et al., 2000, Marcus et al., 2001, Trivedi et al., 1998).

Great interest was focused on this system shortly after its discovery, when two groups virtually simultaneously reported that dysfunction in the orexin system is strongly associated with narcoleptic symptoms in animals (Chemelli et al., 1999, Lin et al., 1999). Subsequent work in humans verified that narcoleptic patients (particularly those with cataplexy) have little orexin in their CSF and lack most or all orexin neurons (Nishino et al., 2000, Nishino, 2007). With these compelling findings, the prevailing view of orexin function focused on arousal and maintenance of the waking state. Supporting this view were findings that major targets of orexin projections are classic brain arousal nuclei such as the locus coeruleus (Peyron et al., 1998, Sutcliffe and de Lecea, 2002) and that orexin application typically strongly activates these cells (Brown et al., 2001, Eriksson et al., 2001, Horvath et al., 1999, Ivanov and Aston-Jones, 2000, Korotkova et al., 2003).

However, a potential role for orexins in reward processing was evident from one of the first publications of their discovery. Sakurai et al. (1998) reported that administration of orexin A or orexin B into the lateral ventricle produced feeding in rats, which prompted them to name the new peptides “orexins,” meaning appetite. The first report of a possible role for orexins in effects of addictive drugs appeared in 2003 and showed that orexin neurons play a role in opiate withdrawal (Georgescu et al., 2003). Subsequent studies examined a possible role for this novel neuropeptide system in reward processing and drug abuse. As reviewed below, orexin appears to play a prominent role in conditioned responses to stimuli associated with food and drug rewards. This reward-associated function of the orexin system may be separate from its role in maintenance of the waking state and mediated by a separate population of (laterally located) orexin neurons.

Section snippets

Orexin neurons are activated by reward-associated stimuli

Orexin neurons in lateral hypothalamus (LH) play an active role in reward processing and drug abuse. Mice with a genetic deletion of orexin completely lack conditioned place preference (CPP) for morphine (Narita et al., 2006). Further, there is a strong association between Fos activation in orexin neurons and the expression of CPP for drug or natural rewards (Harris et al., 2005). Rats conditioned with morphine, cocaine, or food in a CPP paradigm exhibited substantially increased Fos staining

Functional differences for LH versus DMH/PeF orexin neurons

As reviewed above, orexin neurons appear to be involved both in arousal and in reward-seeking, and evidence indicates that orexin neurons are functionally dichotomous (Harris and Aston-Jones, 2006). Thus, reward-seeking functions are associated primarily with orexin cells in LH, whereas arousal- and stress-related processes are more associated with orexin neurons in the DMH and PeF. For example, Estabrooke et al. (2001) reported that PeF and DMH, but not LH, orexin neurons are Fos-activated

Orexin neurotransmission in VTA drives reinstatement

The orexin system projects widely throughout the CNS, so there are many possible targets where orexin might be acting during drug-seeking and reinstatement. One site that seemed likely was the VTA, where dopamine (DA) neurons that play a critical role in reward and reinforcement mechanisms are located. Microinjections of orexin directly into VTA robustly reinstated morphine preference in animals that had previously been extinguished (Harris et al., 2005), and intra-VTA orexin injections

Discussion and hypothesis

The recent data reviewed above indicate that orexin is involved in conditioned behavioral responding to drug-associated stimuli. Cocaine self-administration studies found that OxR1 antagonism significantly attenuated cocaine-seeking elicited by stimuli previously associated with cocaine. SB reduced cue- and context-induced reinstatement of extinguished cocaine-seeking, as well as relapse responding after 2 weeks of abstinence. On the other hand, SB had no effect on reinstatement of

Acknowledgments

This work was supported by PHS grants R37 DA06214, R01 DA017289, P50 DA015369, F31 DA019733, and T32 AA007474.

References (151)

  • ChangJ.Y. et al.

    Neuronal responses in prefrontal cortex and nucleus accumbens during heroin self-administration in freely moving rats

    Brain Res.

    (1997)
  • ChemelliR.M. et al.

    Narcolepsy in orexin knockout mice: molecular genetics of sleep regulation

    Cell

    (1999)
  • CrombagH.S. et al.

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

    Neuropsychopharmacology

    (2002)
  • DayasC.V. et al.

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

    Biol. Psychiatry

    (2008)
  • FadelJ. et al.

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

    Neuroscience

    (2002)
  • HamlinA.S. et al.

    Renewal of extinguished cocaine-seeking

    Neuroscience

    (2008)
  • HarrisG.C. et al.

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

    Neuropsychopharmacology

    (2001)
  • HarrisG. et al.

    Arousal and reward: a dichotomy in orexin function

    Trends Neurosci.

    (2006)
  • HarrisG.C. et al.

    Activation in extended amygdala corresponds to altered hedonic processing during protracted morphine withdrawal

    Behav. Brain Res.

    (2007)
  • HarrisG. et al.

    Glutamate-associated plasticity in the ventral tegmental area is necessary for conditioning environmental stimuli with morphine

    Neuroscience

    (2004)
  • HarrisG. et al.

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

    Behav. Brain Res.

    (2007)
  • HaynesA.C. et al.

    A selective orexin-1 receptor antagonist reduces food consumption in male and female rats

    Regul. Pept.

    (2000)
  • IkemotoS. et al.

    Regional differences within the rat ventral tegmental area for muscimol self-infusions

    Pharmacol. Biochem. Behav.

    (1998)
  • KolbB.

    Functions of the frontal cortex of the rat: a comparative review

    Brain Res.

    (1984)
  • LesseH. et al.

    Frequency-related, bidirectional limbic responses to cocaine: comparisons with amphetamine and lidocaine

    Brain Res.

    (1985)
  • LinL. et al.

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

    Cell

    (1999)
  • LuX.Y. et al.

    Differential distribution and regulation of OX1 and OX2 orexin/hypocretin receptor messenger RNA in the brain upon fasting

    Horm. Behav.

    (2000)
  • NakamuraT. et al.

    Orexin-induced hyperlocomotion and stereotypy are mediated by the dopaminergic system

    Brain Res.

    (2000)
  • NishinoS.

    The hypothalamic peptidergic system, hypocretin/orexin and vigilance control

    Neuropeptides

    (2007)
  • NishinoS. et al.

    Hypocretin (orexin) deficiency in human narcolepsy

    Lancet

    (2000)
  • PierceR.C. et al.

    The mesolimbic dopamine system: the final common pathway for the reinforcing effect of drugs of abuse?

    Neurosci. Biobehav. Rev.

    (2006)
  • Aston-JonesG. et al.

    The bed nucleus of the stria terminalis. A target site for noradrenergic actions in opiate withdrawal

    Ann. N. Y. Acad. Sci.

    (1999)
  • BorglandS.L. et al.

    Acute and chronic cocaine-induced potentiation of synaptic strength in the ventral tegmental area: electrophysiological and behavioral correlates in individual rats

    J. Neurosci.

    (2004)
  • BossertJ.M. et al.

    A role of ventral tegmental area glutamate in contextual cue-induced relapse to heroin seeking

    J. Neurosci.

    (2004)
  • BoutonM.E. et al.

    Role of conditioned contextual stimuli in reinstatement of extinguished fear

    J. Exp. Psychol. Anim. Behav. Process

    (1979)
  • BoutrelB. et al.

    Role for hypocretin in mediating stress-induced reinstatement of cocaine-seeking behavior

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

    (2005)
  • BrownE.E. et al.

    Evidence for conditional neuronal activation following exposure to a cocaine-paired environment: role of forebrain limbic structures

    J. Neurosci.

    (1992)
  • CamiJ. et al.

    Drug addiction

    N. Engl. J. Med.

    (2003)
  • CampbellR.E. et al.

    Orexin neurons express a functional pancreatic polypeptide Y4 receptor

    J. Neurosci.

    (2003)
  • CarlezonW.A. et al.

    Distinct sites of opiate reward and aversion within the midbrain identified using a herpes simplex virus vector expressing GluR1

    J. Neurosci.

    (2000)
  • CarrD.B. et al.

    Projections from the rat prefrontal cortex to the ventral tegmental area: target specificity in the synaptic associations with mesoaccumbens and mesocortical neurons

    J. Neurosci.

    (2000)
  • ChangJ.Y. et al.

    Single neuronal responses in medial prefrontal cortex during cocaine self-administration in freely moving rats

    Synapse

    (1997)
  • ChangJ.Y. et al.

    Comparison of mesocorticolimbic neuronal responses during cocaine and heroin self-administration in freely moving rats

    J. Neurosci.

    (1998)
  • ChildressA.R. et al.

    Limbic activation during cue-induced cocaine craving

    Am. J. Psychiatry

    (1999)
  • de LeceaL. et al.

    The hypocretins: hypothalamus-specific peptides with neuroexcitatory activity

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

    (1998)
  • DelfsJ.M. et al.

    Noradrenaline in the ventral forebrain is critical for opiate withdrawal-induced aversion

    Nature

    (2000)
  • DongH.W. et al.

    Organization of axonal projections from the anterolateral area of the bed nuclei of the stria terminalis

    J. Comp. Neurol.

    (2004)
  • DugovicC. et al.

    Blockade of orexin-1 receptors attenuates orexin-2 receptor antagonism-induced sleep promotion in the rat

    J. Pharmacol. Exp. Ther.

    (2009)
  • DumontE.C. et al.

    Self-administration enhances excitatory synaptic transmission in the bed nucleus of the stria terminalis

    Nat. Neurosci.

    (2005)
  • ErikssonK.S. et al.

    Orexin/hypocretin excites the histaminergic neurons of the tuberomammillary nucleus

    J. Neurosci.

    (2001)
  • Cited by (318)

    View all citing articles on Scopus
    View full text