Bidirectional Relationship between Opioids and Disrupted Sleep: Putative Mechanisms

D Eacret, S C Veasey, J A Blendy, D Eacret, S C Veasey, J A Blendy

Abstract

Millions of Americans suffer from opiate use disorder, and over 100 die every day from opioid overdoses. Opioid use often progresses into a vicious cycle of abuse and withdrawal, resulting in very high rates of relapse. Although the physical and psychologic symptoms of opiate withdrawal are well-documented, sleep disturbances caused by chronic opioid exposure and withdrawal are less well-understood. These substances can significantly disrupt sleep acutely and in the long term. Yet poor sleep may influence opiate use, suggesting a bidirectional feed-forward interaction between poor sleep and opioid use. The neurobiology of how opioids affect sleep and how disrupted sleep affects opioid use is not well-understood. Here, we will summarize what is known about the effects of opioids on electroencephalographic sleep in humans and in animal models. We then discuss the neurobiology interface between reward-related brain regions that mediate arousal and wakefulness as well as the effect of opioids in sleep-related brain regions and neurotransmitter systems. Finally, we summarize what is known of the mechanisms underlying opioid exposure and sleep. A critical review of such studies, as well as recommendations of studies that evaluate the impact of manipulating sleep during withdrawal, will further our understanding of the cyclical feedback between sleep and opioid use. SIGNIFICANCE STATEMENT: We review recent studies on the mechanisms linking opioids and sleep. Opioids affect sleep, and sleep affects opioid use; however, the biology underlying this relationship is not understood. This review compiles recent studies in this area that fill this gap in knowledge.

Copyright © 2020 by The Author(s).

Figures

Fig. 1.
Fig. 1.
Opioid-related wake-promoting vs. opioid-related sleep-promoting systems. These regions and cell types have been shown in this review to be related to opioids and affect sleep. The wake-promoting systems are generally activated by opioids, and the sleep-promoting systems are generally inhibited by opioids.
Fig. 2.
Fig. 2.
Neural circuitry of reward/opioid regions involved in sleep/wake. A likely circuit mechanism regulating sleep/wake states that are engaged by opioid use. This figure infers the sleep/wake circuitry in response to opioids. Green indicates wake-promoting systems and red indicates sleep-promoting systems. Many brain regions were tested for neuronal activation or inhibition, but not all were tested for opioid effects on sleep. VLPO and RMTg neurons are sleep-promoting and are inhibited by opioids. This VLPO opioid exposure inhibits sleep-promoting neurons to produce wakefulness, whereas RMTg opioid exposure disinhibits VTA dopamine neurons. Connections between brain regions are drawn for studies in this review that have manipulated projections and examined sleep. For the VLPO, and for adenosine 2A receptor neurons in the nucleus accumbens, experiments examined manipulation of those neurons specifically, and projections to other reward-related brain regions were not established. 5HT, 5-hydroxytryptamine. DA, Dopamine. DRN, Dorsal Raphe Nucleus.

References

    1. Adamantidis AR, Zhang F, Aravanis AM, Deisseroth K, de Lecea L. (2007) Neural substrates of awakening probed with optogenetic control of hypocretin neurons. Nature 450:420–424.
    1. Alexandre C, Latremoliere A, Ferreira A, Miracca G, Yamamoto M, Scammell TE, Woolf CJ. (2017) Decreased alertness due to sleep loss increases pain sensitivity in mice. Nat Med 23:768–774.
    1. Baimel C, Borgland SL. (2015) Orexin signaling in the VTA gates morphine-induced synaptic plasticity. J Neurosci 35:7295–7303 DOI: 10.1523/JNEUROSCI.4385-14.2015.
    1. Berridge CW, España RA, Vittoz NM. (2010) Hypocretin/orexin in arousal and stress. Brain Res 1314:91–102.
    1. Beswick T, Best D, Rees S, Bearn J, Gossop M, Strang J. (2003) Major disruptions of sleep during treatment of the opiate withdrawal syndrome: differences between methadone and lofexidine detoxification treatments. Addict Biol 8:49–57.
    1. Bonafide CP, Aucutt-Walter N, Divittore N, King T, Bixler EO, Cronin AJ. (2008) Remifentanil inhibits rapid eye movement sleep but not the nocturnal melatonin surge in humans. Anesthesiology 108:627–633.
    1. Chemelli RM, Willie JT, Sinton CM, Elmquist JK, Scammell T, Lee C, Richardson JA, Williams SC, Xiong Y, Kisanuki Y, et al. (1999) Narcolepsy in orexin knockout mice: molecular genetics of sleep regulation. Cell 98:437–451.
    1. Cho JR, Treweek JB, Robinson JE, Xiao C, Bremner LR, Greenbaum A, Gradinaru V. (2017) Dorsal raphe dopamine neurons modulate arousal and promote wakefulness by salient stimuli. Neuron 94:1205–1219.e8 DOI: 10.1016/j.neuron.2017.05.020.
    1. Chowdhury S, Matsubara T, Miyazaki T, Ono D, Fukatsu N, Abe M, Sakimura K, Sudo Y, Yamanaka A. (2019) GABA neurons in the ventral tegmental area regulate non-rapid eye movement sleep in mice. eLife 8:e44928.
    1. Chung S, Weber F, Zhong P, Tan CL, Nguyen TN, Beier KT, Hörmann N, Chang W-C, Zhang Z, Do JP, et al. (2017) Identification of preoptic sleep neurons using retrograde labelling and gene profiling. Nature 545:477–481.
    1. Coffey AA, Guan Z, Grigson PS, Fang J. (2016) Reversal of the sleep-wake cycle by heroin self-administration in rats. Brain Res Bull 123:33–46.
    1. Coleman CG, Baghdoyan HA, Lydic R. (2006) Dialysis delivery of an adenosine A2A agonist into the pontine reticular formation of C57BL/6J mouse increases pontine acetylcholine release and sleep. J Neurochem 96:1750–1759.
    1. Cronin A, Keifer JC, Baghdoyan HA, Lydic R. (1995) Opioid inhibition of rapid eye movement sleep by a specific mu receptor agonist. Br J Anaesth 74:188–192.
    1. De Andrés I, Caballero A. (1989) Chronic morphine administration in cats: effects on sleep and EEG. Pharmacol Biochem Behav 32:519–526.
    1. de Lecea L, Kilduff TS, Peyron C, Gao X, Foye PE, Danielson PE, Fukuhara C, Battenberg EL, Gautvik VT, Bartlett FS, II, et al. (1998) The hypocretins: hypothalamus-specific peptides with neuroexcitatory activity. Proc Natl Acad Sci USA 95:322–327.
    1. Dimsdale JE, Norman D, DeJardin D, Wallace MS. (2007) The effect of opioids on sleep architecture. J Clin Sleep Med 3:33–36.
    1. Durmer JS, Dinges DF. (2005) Neurocognitive consequences of sleep deprivation. Semin Neurol 25:117–129.
    1. Eban-Rothschild A, Rothschild G, Giardino WJ, Jones JR, de Lecea L. (2016) VTA dopaminergic neurons regulate ethologically relevant sleep-wake behaviors. Nat Neurosci 19:1356–1366.
    1. Gauthier EA, Guzick SE, Brummett CM, Baghdoyan HA, Lydic R. (2011) Buprenorphine disrupts sleep and decreases adenosine concentrations in sleep-regulating brain regions of Sprague Dawley rat. Anesthesiology 115:743–753.
    1. Ge F, Mu P, Guo R, Cai L, Liu Z, Dong Y, Huang YH. (2019) Chronic sleep fragmentation enhances habenula cholinergic neural activity. Mol Psychiatry DOI: 10.1038/s41380-019-0419-z [published ahead of print].
    1. Georgescu D, Zachariou V, Barrot M, Mieda M, Willie JT, Eisch AJ, Yanagisawa M, Nestler EJ, Dileone RJ. (2003) Involvement of the lateral hypothalamic peptide orexin in morphine dependence and withdrawal. J Neurosci 23:3106–3111.
    1. Glick SD, Ramirez RL, Livi JM, Maisonneuve IM. (2006) 18-Methoxycoronaridine acts in the medial habenula and/or interpeduncular nucleus to decrease morphine self-administration in rats. Eur J Pharmacol 537:94–98 DOI: 10.1016/j.ejphar.2006.03.045.
    1. Goeldner C, Lutz P-E, Darcq E, Halter T, Clesse D, Ouagazzal A-M, Kieffer BL. (2011) Impaired emotional-like behavior and serotonergic function during protracted abstinence from chronic morphine. Biol Psychiatry 69:236–244.
    1. Greco MA, Fuller PM, Jhou TC, Martin-Schild S, Zadina JE, Hu Z, Shiromani P, Lu J. (2008) Opioidergic projections to sleep-active neurons in the ventrolateral preoptic nucleus. Brain Res 1245:96–107.
    1. Harris GC, Wimmer M, Randall-Thompson JF, Aston-Jones G. (2007) Lateral hypothalamic orexin neurons are critically involved in learning to associate an environment with morphine reward. Behav Brain Res 183:43–51.
    1. Hartwell EE, Pfeifer JG, McCauley JL, Moran-Santa Maria M, Back SE. (2014) Sleep disturbances and pain among individuals with prescription opioid dependence. Addict Behav 39:1537–1542.
    1. Hedegaard H, Miniño AM, Warner M, National Center for Health Statistics (2020) Drug overdose deaths in the United States, 1999–2018 no. 356, NCHS Data Brief, Hyattsville, MD.
    1. Howe RC, Phillips JL, Hegge FW. (1981) Acute heroin abstinence in man: IV. Sleep--waking state contingencies. Drug Alcohol Depend 7:163–176.
    1. Jouvet M. (1969) Biogenic amines and the states of sleep. Science 163:32–41.
    1. Jouvet M. (1972) The role of monoamines and acetylcholine-containing neurons in the regulation of the sleep-waking cycle. Ergeb Physiol 64:166–307.
    1. Keifer JC, Baghdoyan HA, Lydic R. (1992) Sleep disruption and increased apneas after pontine microinjection of morphine. Anesthesiology 77:973–982.
    1. Khazan N, Colasanti B. (1972) Protracted rebound in rapid movement sleep time and electroencephalogram voltage output in morphine-dependent rats upon withdrawal. J Pharmacol Exp Ther 183:23–30.
    1. Lewis SA, Oswald I, Evans JI, Akindele MO, Tompsett SL. (1970) Heroin and human sleep. Electroencephalogr Clin Neurophysiol 28:374–381.
    1. Li C, Sugam JA, Lowery-Gionta EG, McElligott ZA, McCall NM, Lopez AJ, McKlveen JM, Pleil KE, Kash TL. (2016) Mu opioid receptor modulation of dopamine neurons in the periaqueductal gray/dorsal raphe: a role in regulation of pain. Neuropsychopharmacology 41:2122–2132.
    1. Li Y, Li C-Y, Xi W, Jin S, Wu Z-H, Jiang P, Dong P, He X-B, Xu F-Q, Duan S, et al. (2019) Rostral and caudal ventral tegmental area GABAergic inputs to different dorsal raphe neurons participate in opioid dependence. Neuron 101:748–761.e5.
    1. Lim J, Dinges DF. (2008) Sleep deprivation and vigilant attention. Ann N Y Acad Sci 1129: 305–322.
    1. Lin L, Faraco J, Li R, Kadotani H, Rogers W, Lin X, Qiu X, de Jong PJ, Nishino S, Mignot E. (1999) The sleep disorder canine narcolepsy is caused by a mutation in the hypocretin (orexin) receptor 2 gene. Cell 98:365–376.
    1. Logan RW, Hasler BP, Forbes EE, Franzen PL, Torregrossa MM, Huang YH, Buysse DJ, Clark DB, McClung CA. (2018) Impact of sleep and circadian rhythms on addiction vulnerability in adolescents. Biol Psychiatry 83:987–996.
    1. Lopez-Rodriguez F, Wilson CL, Maidment NT, Poland RE, Engel J. (2003) Total sleep deprivation increases extracellular serotonin in the rat hippocampus. Neuroscience 121:523–530.
    1. Louter M, Aarden WCCA, Lion J, Bloem BR, Overeem S. (2012) Recognition and diagnosis of sleep disorders in Parkinson’s disease. J Neurol 259:2031–2040.
    1. Luo YJ, Li YD, Wang L, Yang SR, Yuan XS, Wang J, Cherasse Y, Lazarus M, Chen JF, Qu WM, et al. (2018) Nucleus accumbens controls wakefulness by a subpopulation of neurons expressing dopamine D1 receptors. Nat Commun 9:1576.
    1. Lydic R, Keifer JC, Baghdoyan HA, Becker L. (1993) Microdialysis of the pontine reticular formation reveals inhibition of acetylcholine release by morphine. Anesthesiology 79:1003–1012.
    1. Lydon-Staley DM, Cleveland HH, Huhn AS, Cleveland MJ, Harris J, Stankoski D, Deneke E, Meyer RE, Bunce SC. (2017) Daily sleep quality affects drug craving, partially through indirect associations with positive affect, in patients in treatment for nonmedical use of prescription drugs. Addict Behav 65:275–282.
    1. Marcus JN, Aschkenasi CJ, Lee CE, Chemelli RM, Saper CB, Yanagisawa M, Elmquist JK. (2001) Differential expression of orexin receptors 1 and 2 in the rat brain. J Comp Neurol 435:6–25.
    1. Margarit C, Ballester P, Inda MD, Roca R, Gomez L, Planelles B, Ajo R, Morales D, Peiró AM. (2019) OPRM1 gene interaction with sleep in chronic pain patients treated with opioids. Pain Physician 22:97–107.
    1. Martin WR, Jasinski DR, Haertzen CA, Kay DC, Jones BE, Mansky PA, Carpenter RW. (1973) Methadone--a reevaluation. Arch Gen Psychiatry 28:286–295.
    1. Matsui A, Jarvie BC, Robinson BG, Hentges ST, Williams JT. (2014) Separate GABA afferents to dopamine neurons mediate acute action of opioids, development of tolerance, and expression of withdrawal. Neuron 82:1346–1356.
    1. Maulik PK, Tripathi BM, Pal HR. (2002) Coping behaviors and relapse precipitants in opioid dependence: a study from North India. J Subst Abuse Treat 22:135–140.
    1. Mazei-Robison MS, Koo JW, Friedman AK, Lansink CS, Robison AJ, Vinish M, Krishnan V, Kim S, Siuta MA, Galli A, et al. (2011) Role for mTOR signaling and neuronal activity in morphine-induced adaptations in ventral tegmental area dopamine neurons. Neuron 72:977–990 DOI: 10.1016/j.neuron.2011.10.012.
    1. McDevitt RA, Tiran-Cappello A, Shen H, Balderas I, Britt JP, Marino RAM, Chung SL, Richie CT, Harvey BK, Bonci A. (2014) Serotonergic versus nonserotonergic dorsal raphe projection neurons: differential participation in reward circuitry. Cell Rep 8:1857–1869.
    1. Moore JT, Kelz MB. (2009) Opiates, sleep, and pain: the adenosinergic link. Anesthesiology 111:1175–1176.
    1. Mouret J, Bobillier P, Jouvet M. (1968) Insomnia following parachlorophenylalanine in the rat. Eur J Pharmacol 5:17–22.
    1. Nascimento DC, Andersen ML, Hipólide DC, Nobrega JN, Tufik S. (2007) Pain hypersensitivity induced by paradoxical sleep deprivation is not due to altered binding to brain μ-opioid receptors. Behav Brain Res 178:216–220.
    1. Nehlig A, Daval JL, Debry G. (1992) Caffeine and the central nervous system: mechanisms of action, biochemical, metabolic and psychostimulant effects. Brain Res Brain Res Rev 17:139–170.
    1. Nelson AM, Battersby AS, Baghdoyan HA, Lydic R. (2009) Opioid-induced decreases in rat brain adenosine levels are reversed by inhibiting adenosine deaminase. Anesthesiology 111:1327–1333.
    1. Nestler EJ. (2004) Molecular mechanisms of drug addiction. Neuropharmacology 47 (Suppl 1):24–32.
    1. Oikonomou G, Altermatt M, Zhang R-W, Coughlin GM, Montz C, Gradinaru V, Prober DA. (2019) The serotonergic raphe promote sleep in Zebrafish and mice. Neuron 103:686–701.e8.
    1. Oishi Y, Xu Q, Wang L, Zhang BJ, Takahashi K, Takata Y, Luo YJ, Cherasse Y, Schiffmann SN, de Kerchove d’Exaerde A, et al. (2017) Slow-wave sleep is controlled by a subset of nucleus accumbens core neurons in mice. Nat Commun 8:734.
    1. Onen SH, Alloui A, Gross A, Eschallier A, Dubray C. (2001) The effects of total sleep deprivation, selective sleep interruption and sleep recovery on pain tolerance thresholds in healthy subjects. J Sleep Res 10:35–42.
    1. Ren S, Wang Y, Yue F, Cheng X, Dang R, Qiao Q, Sun X, Li X, Jiang Q, Yao J, et al. (2018) The paraventricular thalamus is a critical thalamic area for wakefulness. Science 362:429–434.
    1. Roehrs T, Hyde M, Blaisdell B, Greenwald M, Roth T. (2006) Sleep loss and REM sleep loss are hyperalgesic. Sleep 29:145–151.
    1. Roehrs T, Roth T. (2005) Sleep and pain: interaction of two vital functions. Semin Neurol 25:106–116.
    1. Sagaspe P, Sanchez-Ortuno M, Charles A, Taillard J, Valtat C, Bioulac B, Philip P. (2006) Effects of sleep deprivation on Color-Word, Emotional, and Specific Stroop interference and on self-reported anxiety. Brain Cogn 60:76–87.
    1. Saito YC, Tsujino N, Abe M, Yamazaki M, Sakimura K, Sakurai T. (2018) Serotonergic input to orexin neurons plays a role in maintaining wakefulness and REM sleep architecture. Front Neurosci 12:892.
    1. Sakurai T, Amemiya A, Ishii M, Matsuzaki I, Chemelli RM, Tanaka H, Williams SC, Richardson JA, Kozlowski GP, Wilson S, et al. (1998) Orexins and orexin receptors: a family of hypothalamic neuropeptides and G protein-coupled receptors that regulate feeding behavior. Cell 92:573–585.
    1. Scammell TE, Gerashchenko DY, Mochizuki T, McCarthy MT, Estabrooke IV, Sears CA, Saper CB, Urade Y, Hayaishi O. (2001) An adenosine A2a agonist increases sleep and induces Fos in ventrolateral preoptic neurons. Neuroscience 107:653–663.
    1. Scholl L, Seth P, Kariisa M, Wilson N, Baldwin G. (2018) Drug and opioid-involved overdose deaths - United States, 2013-2017. MMWR Morb Mortal Wkly Rep 67:1419–1427.
    1. Sharkey KM, Kurth ME, Corso RP, Brower KJ, Millman RP, Stein MD. (2009) Home polysomnography in methadone maintenance patients with subjective sleep complaints. Am J Drug Alcohol Abuse 35:178–182.
    1. Shaw IR, Lavigne G, Mayer P, Choinière M. (2005) Acute intravenous administration of morphine perturbs sleep architecture in healthy pain-free young adults: a preliminary study [published correction appears in Sleep (2006) 29:136]. Sleep 28:677–682.
    1. Skinner GO, Damasceno F, Gomes A, de Almeida OMMS. (2011) Increased pain perception and attenuated opioid antinociception in paradoxical sleep-deprived rats are associated with reduced tyrosine hydroxylase staining in the periaqueductal gray matter and are reversed by L-dopa. Pharmacol Biochem Behav 99:94–99.
    1. Stein MD, Herman DS, Bishop S, Lassor JA, Weinstock M, Anthony J, Anderson BJ. (2004) Sleep disturbances among methadone maintained patients. J Subst Abuse Treat 26:175–180.
    1. Taylor NE, Van Dort CJ, Kenny JD, Pei J, Guidera JA, Vlasov KY, Lee JT, Boyden ES, Brown EN, Solt K. (2016) Optogenetic activation of dopamine neurons in the ventral tegmental area induces reanimation from general anesthesia. Proc Natl Acad Sci USA 113:12826–12831.
    1. Thannickal TC, John J, Shan L, Swaab DF, Wu MF, Ramanathan L, McGregor R, Chew KT, Cornford M, Yamanaka A, et al. (2018) Opiates increase the number of hypocretin-producing cells in human and mouse brain and reverse cataplexy in a mouse model of narcolepsy. Sci Transl Med 10:1–14.
    1. Tomim DH, Pontarolla FM, Bertolini JF, Arase M, Tobaldini G, Lima MMS, Fischer L. (2016) The pronociceptive effect of paradoxical sleep deprivation in rats: evidence for a role of descending pain modulation mechanisms. Mol Neurobiol 53:1706–1717.
    1. Tsujino N, Sakurai T. (2009) Orexin/hypocretin: a neuropeptide at the interface of sleep, energy homeostasis, and reward system. Pharmacol Rev 61:162–176 DOI: 10.1124/pr.109.001321.
    1. Urban DJ, Zhu H, Marcinkiewcz CA, Michaelides M, Oshibuchi H, Rhea D, Aryal DK, Farrell MS, Lowery-Gionta E, Olsen RH, et al. (2016) Elucidation of the behavioral program and neuronal network encoded by dorsal raphe serotonergic neurons. Neuropsychopharmacology 41:1404–1415.
    1. Ursin R. (2002) Serotonin and sleep. Sleep Med Rev 6:55–69.
    1. Valentinova K, Tchenio A, Trusel M, Clerke JA, Lalive AL, Tzanoulinou S, Matera A, Moutkine I, Maroteaux L, Paolicelli RC, et al. (2019) Morphine withdrawal recruits lateral habenula cytokine signaling to reduce synaptic excitation and sociability. Nat Neurosci 22:1053–1056 DOI: 10.1038/s41593-019-0421-4.
    1. Vowles KE, McEntee ML, Julnes PS, Frohe T, Ney JP, Van Der Goes DN. (2015) Rates of opioid misuse, abuse, and addiction in chronic pain: a systematic review and data synthesis. Pain 156:596–576.
    1. Wang HL, Zhang S, Qi J, Wang H, Cachope R, Mejias-Aponte CA, Gomez JA, Mateo-Semidey GE, Beaudoin GMJ, Paladini CA, et al. (2019) Dorsal raphe dual serotonin-glutamate neurons drive reward by establishing excitatory synapses on VTA mesoaccumbens dopamine neurons. Cell Rep 26:1128–1142.e7.
    1. Wang Q, Yue XF, Qu WM, Tan R, Zheng P, Urade Y, Huang ZL. (2013) Morphine inhibits sleep-promoting neurons in the ventrolateral preoptic area via mu receptors and induces wakefulness in rats. Neuropsychopharmacology 38:791–801.
    1. Weaver MD, Barger LK, Malone SK, Anderson LS, Klerman EB. (2018) Dose-dependent associations between sleep duration and unsafe behaviors among US high school students. JAMA Pediatr 172:1187–1189.
    1. Wells AM, Ridener E, Bourbonais CA, Kim W, Pantazopoulos H, Carroll FI, Kim K-S, Cohen BM, Carlezon WA., Jr (2017) Effects of chronic social defeat stress on sleep and circadian rhythms are mitigated by kappa-opioid receptor antagonism. J Neurosci 37:7656–7668.
    1. Xiao L, Tang YL, Smith AK, Xiang YT, Sheng LX, Chi Y, Du WJ, Guo S, Jiang ZN, Zhang GF, et al. (2010) Nocturnal sleep architecture disturbances in early methadone treatment patients. Psychiatry Res 179:91–95.
    1. Yang SR, Hu ZZ, Luo YJ, Zhao YN, Sun HX, Yin D, Wang CY, Yan YD, Wang DR, Yuan XS, et al. (2018) The rostromedial tegmental nucleus is essential for non-rapid eye movement sleep. PLoS Biol 16:e2002909.
    1. Yu X, Li W, Ma Y, Tossell K, Harris JJ, Harding EC, Ba W, Miracca G, Wang D, Li L, et al. (2019) GABA and glutamate neurons in the VTA regulate sleep and wakefulness. Nat Neurosci 22:106–119.
    1. Zant JC, Leenaars CHC, Kostin A, Van Someren EJ, Porkka-Heiskanen T. (2011) Increases in extracellular serotonin and dopamine metabolite levels in the basal forebrain during sleep deprivation. Brain Res 1399:40–48.
    1. Zhu Y, Wienecke CFR, Nachtrab G, Chen X. (2016) A thalamic input to the nucleus accumbens mediates opiate dependence. Nature 530:219–222.

Source: PubMed

3
Sottoscrivi