Anesth Pain Med.  2022 Oct;17(4):343-351. 10.17085/apm.22227.

General anesthesia and sleep: like and unlike

Affiliations
  • 1Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea

Abstract

General anesthesia and sleep have long been discussed in the neurobiological context owing to their commonalities, such as unconsciousness, immobility, non-responsiveness to external stimuli, and lack of memory upon returning to consciousness. Sleep is regulated by complex interactions between wake-promoting and sleep-promoting neural circuits. Anesthetics exert their effects partly by inhibiting wake-promoting neurons or activating sleep-promoting neurons. Unconscious but arousable sedation is more related to sleep-wake circuitries, whereas unconscious and unarousable anesthesia is independent of them. General anesthesia is notable for its ability to decrease sleep propensity. Conversely, increased sleep propensity due to insufficient sleep potentiates anesthetic effects. Taken together, it is plausible that sleep and anesthesia are closely related phenomena but not the same ones. Further investigations on the relationship between sleep and anesthesia are warranted.

Keyword

General anesthesia; Neural pathways; Neurobiology; Sleep

Figure

  • Fig. 1. Similarities and differences in the mechanism of action of general anesthesia and sleep. General anesthesia and sleep have similar properties, as there is some degree of overlap in their neuronal circuitry. Sleep and anesthesia can be initiated by inhibiting the wake-promoting pathway or activating the sleep-promoting pathway. However, other mechanisms are thought to be used to reach the “Unconscious, Not arousable” state by general anesthesia, but the exact mechanism is not yet understood. LC: locus coeruleus, DRN: dorsal raphe nuclei, PPT: pedunculopontine tegmental nucleus, LDT: laterodorsal tegmental nucleus, VTA: ventral tegmental area, PB: parabrachial nucleus, TMN: tuberomammillary nuclei, LH: lateral hypothalamus, BF: basal forebrain, VLPO: ventrolateral preoptic area, PZ: parafacial zone, nNOS: neuronal nitric oxide synthase (nNOS)-containing neurons in the cortex, PPT: pedunculopontine tegmental nucleus, LDT: laterodorsal tegmental nucleus, SLD: sublaterodorsal nucleus. Figure were created using BioRender.com.


Reference

1. Fleming A. Note on the induction of sleep and anæsthesia by compression of the carotids. Br Foreign Med Chir Rev. 1855; 15:529–30.
2. Committee on Quality Management and Departmental Administration. Continuum of depth of sedation: definition of general anesthesia and levels of sedation/analgesia. American Society of Anesthesiologists [Internet]. 1999 Oct 13 [updated 2019 Oct 23; cited 2022 Oct 24]. Available from https://www.asahq.org/standards-and-guidelines/continuum-of-depth-of-sedation-definition-of-general-anesthesia-and-levels-of-sedationanalgesia.
3. Brown EN, Lydic R, Schiff ND. General anesthesia, sleep, and coma. N Engl J Med. 2010; 363:2638–50.
Article
4. Siegel JM. Clues to the functions of mammalian sleep. Nature. 2005; 437:1264–71.
Article
5. Kryger MH, Roth T, Goldstein CA, Dement WC. Principles and practice of sleep medicine. 7th ed. Philadelphia (PA), Elsevier. 2022, pp 13-26.
6. Swank RL, Watson CW. Effects of barbiturates and ether on spontaneous electrical activity of dog brain. J Neurophysiol. 1949; 12:137–60.
Article
7. Clark DL, Rosner BS. Neurophysiologic effects of general anesthetics. I. The electroencephalogram and sensory evoked responses in man. Anesthesiology. 1973; 38:564–82.
8. Ching S, Purdon PL, Vijayan S, Kopell NJ, Brown EN. A neurophysiological-metabolic model for burst suppression. Proc Natl Acad Sci U S A. 2012; 109:3095–100.
Article
9. Shanker A, Abel JH, Schamberg G, Brown EN. Etiology of burst suppression EEG patterns. Front Psychol. 2021; 12:673529.
Article
10. Marion DW, Penrod LE, Kelsey SF, Obrist WD, Kochanek PM, Palmer AM, et al. Treatment of traumatic brain injury with moderate hypothermia. N Engl J Med. 1997; 336:540–6.
Article
11. Stecker MM, Cheung AT, Pochettino A, Kent GP, Patterson T, Weiss SJ, et al. Deep hypothermic circulatory arrest: II. Changes in electroencephalogram and evoked potentials during rewarming. Ann Thorac Surg. 2001; 71:22–8.
Article
12. Westover MB, Ching S, Kumaraswamy VM, Akeju SO, Pierce E, Cash SS, et al. The human burst suppression electroencephalogram of deep hypothermia. Clin Neurophysiol. 2015; 126:1901–14.
Article
13. Purdon PL, Pierce ET, Mukamel EA, Prerau MJ, Walsh JL, Wong KF, et al. Electroencephalogram signatures of loss and recovery of consciousness from propofol. Proc Natl Acad Sci U S A. 2013; 110:E1142–51.
Article
14. Young GB. The EEG in coma. J Clin Neurophysiol. 2000; 17:473–85.
Article
15. Kales A, Rechtschaffen A. A manual of standardized terminology, techniques and scoring system for sleep stages of human subjects. Washington, D.C., US National Institute of Neurological Diseases and Blindness, Neurological Information Network. 1968.
16. Datta S. Cellular and chemical neuroscience of mammalian sleep. Sleep Med. 2010; 11:431–40.
Article
17. Moruzzi G. Reticular influences on the EEG. Electroencephalogr Clin Neurophysiol. 1964; 16:2–17.
Article
18. Moruzzi G. The sleep-waking cycle. Ergeb Physiol. 1972; 64:1–165.
Article
19. Moruzzi G, Magoun HW. Brain stem reticular formation and activation of the EEG. Electroencephalogr Clin Neurophysiol. 1949; 1:455–73.
Article
20. Datta S. Neuronal activity in the peribrachial area: relationship to behavioral state control. Neurosci Biobehav Rev. 1995; 19:67–84.
Article
21. Datta S, Maclean RR. Neurobiological mechanisms for the regulation of mammalian sleep-wake behavior: reinterpretation of historical evidence and inclusion of contemporary cellular and molecular evidence. Neurosci Biobehav Rev. 2007; 31:775–824.
Article
22. Deisseroth K. Optogenetics. Nat Methods. 2011; 8:26–9.
Article
23. Carter ME, Yizhar O, Chikahisa S, Nguyen H, Adamantidis A, Nishino S, et al. Tuning arousal with optogenetic modulation of locus coeruleus neurons. Nat Neurosci. 2010; 13:1526–33.
Article
24. Ito H, Yanase M, Yamashita A, Kitabatake C, Hamada A, Suhara Y, et al. Analysis of sleep disorders under pain using an optogenetic tool: possible involvement of the activation of dorsal raphe nucleus-serotonergic neurons. Mol Brain. 2013; 6:59.
Article
25. Boucetta S, Cissé Y, Mainville L, Morales M, Jones BE. Discharge profiles across the sleep-waking cycle of identified cholinergic, GABAergic, and glutamatergic neurons in the pontomesencephalic tegmentum of the rat. J Neurosci. 2014; 34:4708–27.
Article
26. Steriade M, Dossi RC, Paré D, Oakson G. Fast oscillations (20-40 Hz) in thalamocortical systems and their potentiation by mesopontine cholinergic nuclei in the cat. Proc Natl Acad Sci U S A. 1991; 88:4396–400.
Article
27. Kroeger D, Ferrari LL, Petit G, Mahoney CE, Fuller PM, Arrigoni E, et al. Cholinergic, glutamatergic, and GABAergic neurons of the pedunculopontine tegmental nucleus have distinct effects on sleep/wake behavior in mice. J Neurosci. 2017; 37:1352–66.
Article
28. Van Dort CJ, Zachs DP, Kenny JD, Zheng S, Goldblum RR, Gelwan NA, et al. Optogenetic activation of cholinergic neurons in the PPT or LDT induces REM sleep. Proc Natl Acad Sci U S A. 2015; 112:584–9.
Article
29. Cox J, Pinto L, Dan Y. Calcium imaging of sleep-wake related neuronal activity in the dorsal pons. Nat Commun. 2016; 7:10763.
Article
30. Eban-Rothschild A, Rothschild G, Giardino WJ, Jones JR, de Lecea L. VTA dopaminergic neurons regulate ethologically relevant sleep-wake behaviors. Nat Neurosci. 2016; 19:1356–66.
Article
31. Kaur S, Pedersen NP, Yokota S, Hur EE, Fuller PM, Lazarus M, et al. Glutamatergic signaling from the parabrachial nucleus plays a critical role in hypercapnic arousal. J Neurosci. 2013; 33:7627–40.
Article
32. Parmentier R, Zhao Y, Perier M, Akaoka H, Lintunen M, Hou Y, et al. Role of histamine H1-receptor on behavioral states and wake maintenance during deficiency of a brain activating system: a study using a knockout mouse model. Neuropharmacology. 2016; 106:20–34.
Article
33. Adamantidis AR, Zhang F, Aravanis AM, Deisseroth K, de Lecea L. Neural substrates of awakening probed with optogenetic control of hypocretin neurons. Nature. 2007; 450:420–4.
Article
34. Carter ME, Adamantidis A, Ohtsu H, Deisseroth K, de Lecea L. Sleep homeostasis modulates hypocretin-mediated sleep-to-wake transitions. J Neurosci. 2009; 29:10939–49.
Article
35. Carter ME, Brill J, Bonnavion P, Huguenard JR, Huerta R, de Lecea L. Mechanism for hypocretin-mediated sleep-to-wake transitions. Proc Natl Acad Sci U S A. 2012; 109:E2635–44.
Article
36. Tsunematsu T, Kilduff TS, Boyden ES, Takahashi S, Tominaga M, Yamanaka A. Acute optogenetic silencing of orexin/hypocretin neurons induces slow-wave sleep in mice. J Neurosci. 2011; 31:10529–39.
Article
37. Xu M, Chung S, Zhang S, Zhong P, Ma C, Chang WC, et al. Basal forebrain circuit for sleep-wake control. Nat Neurosci. 2015; 18:1641–7.
Article
38. Anaclet C, Ferrari L, Arrigoni E, Bass CE, Saper CB, Lu J, et al. The GABAergic parafacial zone is a medullary slow wave sleep-promoting center. Nat Neurosci 2014; 17: 1217-24. Erratum in: Nat Neurosci. 2014; 17:1841.
Article
39. Gerashchenko D, Wisor JP, Burns D, Reh RK, Shiromani PJ, Sakurai T, et al. Identification of a population of sleep-active cerebral cortex neurons. Proc Natl Acad Sci U S A. 2008; 105:10227–32.
Article
40. Hobson JA, McCarley RW, Wyzinski PW. Sleep cycle oscillation: reciprocal discharge by two brainstem neuronal groups. Science. 1975; 189:55–8.
Article
41. Sakai K, Crochet S, Onoe H. Pontine structures and mechanisms involved in the generation of paradoxical (REM) sleep. Arch Ital Biol. 2001; 139:93–107.
42. Saper CB, Scammell TE, Lu J. Hypothalamic regulation of sleep and circadian rhythms. Nature. 2005; 437:1257–63.
Article
43. Borbély AA. A two process model of sleep regulation. Hum Neurobiol. 1982; 1:195–204.
44. Porkka-Heiskanen T, Strecker RE, Thakkar M, Bjorkum AA, Greene RW, McCarley RW. Adenosine: a mediator of the sleep-inducing effects of prolonged wakefulness. Science. 1997; 276:1265–8.
Article
45. Campagna JA, Miller KW, Forman SA. Mechanisms of actions of inhaled anesthetics. N Engl J Med. 2003; 348:2110–24.
Article
46. Hemmings HC Jr, Akabas MH, Goldstein PA, Trudell JR, Orser BA, Harrison NL. Emerging molecular mechanisms of general anesthetic action. Trends Pharmacol Sci. 2005; 26:503–10.
Article
47. Orser BA, Canning KJ, Macdonald JF. Mechanisms of general anesthesia. Curr Opin Anaesthesiol. 2002; 15:427–33.
Article
48. Franks NP. Molecular targets underlying general anaesthesia. Br J Pharmacol. 2006; 147(Suppl 1):S72–81.
Article
49. Hemmings HC Jr. Sodium channels and the synaptic mechanisms of inhaled anaesthetics. Br J Anaesth. 2009; 103:61–9.
Article
50. Pearce RA. General anesthetic effects on GABAA receptors. In: Neural mechanisms of anesthesia. Edited by Antognini JF, Carstens E, Raines DE: Totowa (NJ), Humana Press. 2003, pp 265-82.
51. Chua HC, Chebib M. GABAA receptors and the diversity in their structure and pharmacology. Adv Pharmacol. 2017; 79:1–34.
52. Johnston GA. GABAA receptor pharmacology. Pharmacol Ther. 1996; 69:173–98.
Article
53. Anis NA, Berry SC, Burton NR, Lodge D. The dissociative anaesthetics, ketamine and phencyclidine, selectively reduce excitation of central mammalian neurones by N-methyl-aspartate. Br J Pharmacol. 1983; 79:565–75.
Article
54. Jevtović-Todorović V, Todorović SM, Mennerick S, Powell S, Dikranian K, Benshoff N, et al. Nitrous oxide (laughing gas) is an NMDA antagonist, neuroprotectant and neurotoxin. Nat Med. 1998; 4:460–3.
Article
55. Franks NP, Dickinson R, de Sousa SL, Hall AC, Lieb WR. How does xenon produce anaesthesia? Nature. 1998; 396:324.
Article
56. Lois F, De Kock M. Something new about ketamine for pediatric anesthesia? Curr Opin Anaesthesiol. 2008; 21:340–4.
Article
57. Chizh BA. Low dose ketamine: a therapeutic and research tool to explore N-methyl-D-aspartate (NMDA) receptor-mediated plasticity in pain pathways. J Psychopharmacol. 2007; 21:259–71.
Article
58. Nelson LE, Guo TZ, Lu J, Saper CB, Franks NP, Maze M. The sedative component of anesthesia is mediated by GABA(A) receptors in an endogenous sleep pathway. Nat Neurosci. 2002; 5:979–84.
Article
59. Nelson LE, Lu J, Guo T, Saper CB, Franks NP, Maze M. The alpha2-adrenoceptor agonist dexmedetomidine converges on an endogenous sleep-promoting pathway to exert its sedative effects. Anesthesiology. 2003; 98:428–36.
60. Saper CB. Organization of cerebral cortical afferent systems in the rat. II. Hypothalamocortical projections. J Comp Neurol. 1985; 237:21–46.
Article
61. Lin JS, Sakai K, Jouvet M. Evidence for histaminergic arousal mechanisms in the hypothalamus of cat. Neuropharmacology. 1988; 27:111–22.
Article
62. Du WJ, Zhang RW, Li J, Zhang BB, Peng XL, Cao S, et al. The locus coeruleus modulates intravenous general anesthesia of zebrafish via a cooperative mechanism. Cell Rep. 2018; 24:3146–55.e3.
Article
63. Vazey EM, Aston-Jones G. Designer receptor manipulations reveal a role of the locus coeruleus noradrenergic system in isoflurane general anesthesia. Proc Natl Acad Sci U S A. 2014; 111:3859–64.
Article
64. Kelz MB, Sun Y, Chen J, Cheng Meng Q, Moore JT, Veasey SC, et al. An essential role for orexins in emergence from general anesthesia. Proc Natl Acad Sci U S A. 2008; 105:1309–14.
Article
65. Zhao S, Wang S, Li H, Guo J, Li J, Wang D, et al. Activation of orexinergic neurons inhibits the anesthetic effect of desflurane on consciousness state via paraventricular thalamic nucleus in rats. Anesth Analg. 2021; 133:781–93.
Article
66. Lydic R, Baghdoyan HA. Sleep, anesthesiology, and the neurobiology of arousal state control. Anesthesiology. 2005; 103:1268–95.
Article
67. Franks NP. General anaesthesia: from molecular targets to neuronal pathways of sleep and arousal. Nat Rev Neurosci. 2008; 9:370–86.
Article
68. Moore JT, Chen J, Han B, Meng QC, Veasey SC, Beck SG, et al. Direct activation of sleep-promoting VLPO neurons by volatile anesthetics contributes to anesthetic hypnosis. Curr Biol. 2012; 22:2008–16.
Article
69. Nelson AB, Faraguna U, Tononi G, Cirelli C. Effects of anesthesia on the response to sleep deprivation. Sleep. 2010; 33:1659–67.
Article
70. Pal D, Lipinski WJ, Walker AJ, Turner AM, Mashour GA. State-specific effects of sevoflurane anesthesia on sleep homeostasis: selective recovery of slow wave but not rapid eye movement sleep. Anesthesiology. 2011; 114:302–10.
71. Hwang L, Ko IG, Jin JJ, Kim SH, Kim CJ, Chang B, et al. Dexmedetomidine ameliorates memory impairment in sleep-deprived mice. Anim Cells Syst (Seoul). 2019; 23:371–9.
Article
72. Tung A, Szafran MJ, Bluhm B, Mendelson WB. Sleep deprivation potentiates the onset and duration of loss of righting reflex induced by propofol and isoflurane. Anesthesiology. 2002; 97:906–11.
Article
73. Leung JM, Sands LP, Newman S, Meckler G, Xie Y, Gay C, et al. Preoperative sleep disruption and postoperative delirium. J Clin Sleep Med. 2015; 11:907–13.
Article
74. Eikermann M, Vetrivelan R, Grosse-Sundrup M, Henry ME, Hoffmann U, Yokota S, et al. The ventrolateral preoptic nucleus is not required for isoflurane general anesthesia. Brain Res. 2011; 1426:30–7.
Article
75. Eikermann M, Akeju O, Chamberlin NL. Sleep and anesthesia: the shared circuit hypothesis has been put to bed. Curr Biol. 2020; 30:R219–21.
Article
Full Text Links
  • APM
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr