Skip to main content
Top
Published in: Current Neurology and Neuroscience Reports 9/2018

01-09-2018 | Sleep (M Thorpy and M Billiard, Section Editors)

Cerebral Metabolic Changes During Sleep

Authors: Nadia Nielsen Aalling, Maiken Nedergaard, Mauro DiNuzzo

Published in: Current Neurology and Neuroscience Reports | Issue 9/2018

Login to get access

Abstract

Purpose of Review

The goal of the present paper is to review current literature supporting the occurrence of fundamental changes in brain energy metabolism during the transition from wakefulness to sleep.

Recent Findings

Latest research in the field indicates that glucose utilization and the concentrations of several brain metabolites consistently change across the sleep-wake cycle. Lactate, a product of glycolysis that is involved in synaptic plasticity, has emerged as a good biomarker of brain state. Sleep-induced changes in cerebral metabolite levels result from a shift in oxidative metabolism, which alters the reliance of brain metabolism upon carbohydrates.

Summary

We found wide support for the notion that brain energetics is state dependent. In particular, fatty acids and ketone bodies partly replace glucose as cerebral energy source during sleep. This mechanism plausibly accounts for increases in biosynthetic pathways and functional alterations in neuronal activity associated with sleep. A better account of brain energy metabolism during sleep might help elucidate the long mysterious restorative effects of sleep for the whole organism.
Literature
1.
go back to reference Chatelle C, Laureys S, Schnakers C. Disorders of consciousness: what do we know? In: Dehaene S, Christen Y, editors. Characterizing consciousness: from cognition to the clinic? Berlin: Springer; 2011. p. 85–98.CrossRef Chatelle C, Laureys S, Schnakers C. Disorders of consciousness: what do we know? In: Dehaene S, Christen Y, editors. Characterizing consciousness: from cognition to the clinic? Berlin: Springer; 2011. p. 85–98.CrossRef
13.
go back to reference Morland C, Lauritzen KH, Puchades M, Holm-Hansen S, Andersson K, Gjedde A, et al. The lactate receptor, G-protein-coupled receptor 81/hydroxycarboxylic acid receptor 1: expression and action in brain. J Neurosci Res. 2015;93(7):1045–55. https://doi.org/10.1002/jnr.23593. Morland C, Lauritzen KH, Puchades M, Holm-Hansen S, Andersson K, Gjedde A, et al. The lactate receptor, G-protein-coupled receptor 81/hydroxycarboxylic acid receptor 1: expression and action in brain. J Neurosci Res. 2015;93(7):1045–55. https://​doi.​org/​10.​1002/​jnr.​23593.
19.
go back to reference Hibi M, Kubota C, Mizuno T, Aritake S, Mitsui Y, Katashima M, et al. Effect of shortened sleep on energy expenditure, core body temperature, and appetite: a human randomised crossover trial. Sci Rep. 2017;7:39640. https://doi.org/10.1038/srep39640. Hibi M, Kubota C, Mizuno T, Aritake S, Mitsui Y, Katashima M, et al. Effect of shortened sleep on energy expenditure, core body temperature, and appetite: a human randomised crossover trial. Sci Rep. 2017;7:39640. https://​doi.​org/​10.​1038/​srep39640.
22.
go back to reference Wildenhoff KE, Johansen JP, Karstoft H, Yde H, Sorensen NS. Diurnal variations in the concentrations of blood acetoacetate and 3-hydroxybutyrate. The ketone body peak around midnight and its relationship to free fatty acids, glycerol, insulin, growth hormone and glucose in serum and plasma. Acta Med Scand. 1974;195:25–8.PubMedCrossRef Wildenhoff KE, Johansen JP, Karstoft H, Yde H, Sorensen NS. Diurnal variations in the concentrations of blood acetoacetate and 3-hydroxybutyrate. The ketone body peak around midnight and its relationship to free fatty acids, glycerol, insulin, growth hormone and glucose in serum and plasma. Acta Med Scand. 1974;195:25–8.PubMedCrossRef
24.
go back to reference Iwata S, Ozawa K, Shimahara Y, Mori K, Kobayashi N, Kumada K, et al. Diurnal fluctuations of arterial ketone body ratio in normal subjects and patients with liver dysfunction. Gastroenterology. 1991;100:1371–8. Iwata S, Ozawa K, Shimahara Y, Mori K, Kobayashi N, Kumada K, et al. Diurnal fluctuations of arterial ketone body ratio in normal subjects and patients with liver dysfunction. Gastroenterology. 1991;100:1371–8.
25.
go back to reference De Gasquet P, Griglio S, Pequignot-Planche E, Malewiak MI. Diurnal changes in plasma and liver lipids and lipoprotein lipase activity in heart and adipose tissue in rats fed a high and low fat diet. J Nutr. 1977;107:199–212.PubMedCrossRef De Gasquet P, Griglio S, Pequignot-Planche E, Malewiak MI. Diurnal changes in plasma and liver lipids and lipoprotein lipase activity in heart and adipose tissue in rats fed a high and low fat diet. J Nutr. 1977;107:199–212.PubMedCrossRef
30.
go back to reference • Bellesi M, Pfister-Genskow M, Maret S, Keles S, Tononi G, Cirelli C. Effects of sleep and wake on oligodendrocytes and their precursors. J Neurosci. 2013;33(36):14288–300. https://doi.org/10.1523/JNEUROSCI.5102-12.2013. Using translating ribosome affinity purification (TRAP) technology combined with microarray analysis, this study generated cell-type specific transcriptomic profiles of oligodendrocytes after longer periods of sleep, wake, or acute sleep deprivation. They show that genes involved in phospholipid synthesis and myelination or promoting OPC proliferation are transcribed preferentially during sleep, and experimentally validate some of their findings. PubMedPubMedCentralCrossRef • Bellesi M, Pfister-Genskow M, Maret S, Keles S, Tononi G, Cirelli C. Effects of sleep and wake on oligodendrocytes and their precursors. J Neurosci. 2013;33(36):14288–300. https://​doi.​org/​10.​1523/​JNEUROSCI.​5102-12.​2013. Using translating ribosome affinity purification (TRAP) technology combined with microarray analysis, this study generated cell-type specific transcriptomic profiles of oligodendrocytes after longer periods of sleep, wake, or acute sleep deprivation. They show that genes involved in phospholipid synthesis and myelination or promoting OPC proliferation are transcribed preferentially during sleep, and experimentally validate some of their findings. PubMedPubMedCentralCrossRef
31.
go back to reference Poduslo SE, Miller K. Ketone bodies as precursors for lipid synthesis in neurons, astrocytes, and oligodendroglia (myelin) in hyperthyroidism, hyperketonemia and hypoketonemia. Neurochem Int. 1991;18(1):85–8.PubMedCrossRef Poduslo SE, Miller K. Ketone bodies as precursors for lipid synthesis in neurons, astrocytes, and oligodendroglia (myelin) in hyperthyroidism, hyperketonemia and hypoketonemia. Neurochem Int. 1991;18(1):85–8.PubMedCrossRef
33.
go back to reference Netchiporouk L, Shram N, Salvert D, Cespuglio R. Brain extracellular glucose assessed by voltammetry throughout the rat sleep-wake cycle. Eur J Neurosci. 2001;13(7):1429–34.PubMedCrossRef Netchiporouk L, Shram N, Salvert D, Cespuglio R. Brain extracellular glucose assessed by voltammetry throughout the rat sleep-wake cycle. Eur J Neurosci. 2001;13(7):1429–34.PubMedCrossRef
34.
go back to reference Van den Noort S, Brine K. Effect of sleep on brain labile phosphates and metabolic rate. Am J Phys. 1970;218(5):1434–9. Van den Noort S, Brine K. Effect of sleep on brain labile phosphates and metabolic rate. Am J Phys. 1970;218(5):1434–9.
36.
go back to reference Cespuglio R, Netchiporouk L, Glucose SN. Lactate monitoring across the rat sleep–wake cycle. In: Marinesco S, Dale N, editors. Microelectrode Biosensors. Totowa, NJ: Humana Press; 2013. p. 241–56.CrossRef Cespuglio R, Netchiporouk L, Glucose SN. Lactate monitoring across the rat sleep–wake cycle. In: Marinesco S, Dale N, editors. Microelectrode Biosensors. Totowa, NJ: Humana Press; 2013. p. 241–56.CrossRef
38.
go back to reference McNay EC, Fries TM, Gold PE. Decreases in rat extracellular hippocampal glucose concentration associated with cognitive demand during a spatial task. Proc Natl Acad Sci U S A. 2000;97(6):2881–5.PubMedPubMedCentralCrossRef McNay EC, Fries TM, Gold PE. Decreases in rat extracellular hippocampal glucose concentration associated with cognitive demand during a spatial task. Proc Natl Acad Sci U S A. 2000;97(6):2881–5.PubMedPubMedCentralCrossRef
40.
go back to reference Merboldt KD, Bruhn H, Hanicke W, Michaelis T, Frahm J. Decrease of glucose in the human visual cortex during photic stimulation. Magn Reson Med. 1992;25(1):187–94.PubMedCrossRef Merboldt KD, Bruhn H, Hanicke W, Michaelis T, Frahm J. Decrease of glucose in the human visual cortex during photic stimulation. Magn Reson Med. 1992;25(1):187–94.PubMedCrossRef
41.
go back to reference Clegern WC, Moore ME, Schmidt MA, Wisor J. Simultaneous electroencephalography, real-time measurement of lactate concentration and optogenetic manipulation of neuronal activity in the rodent cerebral cortex. J Vis Exp. 2012;70:e4328. https://doi.org/10.3791/4328.CrossRef Clegern WC, Moore ME, Schmidt MA, Wisor J. Simultaneous electroencephalography, real-time measurement of lactate concentration and optogenetic manipulation of neuronal activity in the rodent cerebral cortex. J Vis Exp. 2012;70:e4328. https://​doi.​org/​10.​3791/​4328.CrossRef
42.
go back to reference Shram N, Netchiporouk L, Cespuglio R. Lactate in the brain of the freely moving rat: voltammetric monitoring of the changes related to the sleep-wake states. Eur J Neurosci. 2002;16(3):461–6.PubMedCrossRef Shram N, Netchiporouk L, Cespuglio R. Lactate in the brain of the freely moving rat: voltammetric monitoring of the changes related to the sleep-wake states. Eur J Neurosci. 2002;16(3):461–6.PubMedCrossRef
45.
go back to reference Richter D, Dawson RM. Brain metabolism in emotional excitement and in sleep. Am J Phys. 1948;154(1):73–9. Richter D, Dawson RM. Brain metabolism in emotional excitement and in sleep. Am J Phys. 1948;154(1):73–9.
48.
go back to reference Frahm J, Kruger G, Merboldt KD, Kleinschmidt A. Dynamic uncoupling and recoupling of perfusion and oxidative metabolism during focal brain activation in man. Magn Reson Med. 1996;35(2):143–8.PubMedCrossRef Frahm J, Kruger G, Merboldt KD, Kleinschmidt A. Dynamic uncoupling and recoupling of perfusion and oxidative metabolism during focal brain activation in man. Magn Reson Med. 1996;35(2):143–8.PubMedCrossRef
49.
go back to reference Frahm J, Krueger G, Merboldt KD, Kleinschmidt A. Dynamic NMR studies of perfusion and oxidative metabolism during focal brain activation. Advances in Experimental Medicine and Biology. 1997;413:195–203.PubMedCrossRef Frahm J, Krueger G, Merboldt KD, Kleinschmidt A. Dynamic NMR studies of perfusion and oxidative metabolism during focal brain activation. Advances in Experimental Medicine and Biology. 1997;413:195–203.PubMedCrossRef
50.
go back to reference Prichard J, Rothman D, Novotny E, Petroff O, Kuwabara T, Avison M, et al. Lactate rise detected by 1H NMR in human visual cortex during physiologic stimulation. Proc Natl Acad Sci U S A. 1991;88(13):5829–31. Prichard J, Rothman D, Novotny E, Petroff O, Kuwabara T, Avison M, et al. Lactate rise detected by 1H NMR in human visual cortex during physiologic stimulation. Proc Natl Acad Sci U S A. 1991;88(13):5829–31.
51.
go back to reference Sappey-Marinier D, Calabrese G, Fein G, Hugg JW, Biggins C, Weiner MW. Effect of photic stimulation on human visual cortex lactate and phosphates using 1H and 31P magnetic resonance spectroscopy. J Cereb Blood Flow Metab. 1992;12(4):584–92.PubMedCrossRef Sappey-Marinier D, Calabrese G, Fein G, Hugg JW, Biggins C, Weiner MW. Effect of photic stimulation on human visual cortex lactate and phosphates using 1H and 31P magnetic resonance spectroscopy. J Cereb Blood Flow Metab. 1992;12(4):584–92.PubMedCrossRef
52.
go back to reference •• Hinard V, Mikhail C, Pradervand S, Curie T, Houtkooper RH, Auwerx J, et al. Key electrophysiological, molecular, and metabolic signatures of sleep and wakefulness revealed in primary cortical cultures. J Neurosci. 2012;32(36):12506–17. https://doi.org/10.1523/JNEUROSCI.2306-12.2012. This is the only study that have investigated how sleep deprivation affects the mouse cortex metabolome. Based on results of this study and from stimulation of neuronal cell cultures, the authors suggest that sleep might play a major role in reestablishing the neuronal membrane homeostasis. •• Hinard V, Mikhail C, Pradervand S, Curie T, Houtkooper RH, Auwerx J, et al. Key electrophysiological, molecular, and metabolic signatures of sleep and wakefulness revealed in primary cortical cultures. J Neurosci. 2012;32(36):12506–17. https://​doi.​org/​10.​1523/​JNEUROSCI.​2306-12.​2012. This is the only study that have investigated how sleep deprivation affects the mouse cortex metabolome. Based on results of this study and from stimulation of neuronal cell cultures, the authors suggest that sleep might play a major role in reestablishing the neuronal membrane homeostasis.
54.
go back to reference Maquet P, Dive D, Salmon E, Sadzot B, Franco G, Poirrier R, et al. Cerebral glucose utilization during stage 2 sleep in man. Brain Res. 1992;571(1):149–53. Maquet P, Dive D, Salmon E, Sadzot B, Franco G, Poirrier R, et al. Cerebral glucose utilization during stage 2 sleep in man. Brain Res. 1992;571(1):149–53.
55.
go back to reference Buchsbaum MS, Gillin JC, Wu J, Hazlett E, Sicotte N, Dupont RM, et al. Regional cerebral glucose metabolic rate in human sleep assessed by positron emission tomography. Life Sci. 1989;45(15):1349–56. Buchsbaum MS, Gillin JC, Wu J, Hazlett E, Sicotte N, Dupont RM, et al. Regional cerebral glucose metabolic rate in human sleep assessed by positron emission tomography. Life Sci. 1989;45(15):1349–56.
56.
go back to reference Franck G, Salmon E, Poirrier R, Sadzot B, Franco G. Study of regional cerebral glucose metabolism, in man, while awake or asleep, by positron emission tomography. Rev Electroencephalogr Neurophysiol Clin. 1987;17(1):71–7.PubMedCrossRef Franck G, Salmon E, Poirrier R, Sadzot B, Franco G. Study of regional cerebral glucose metabolism, in man, while awake or asleep, by positron emission tomography. Rev Electroencephalogr Neurophysiol Clin. 1987;17(1):71–7.PubMedCrossRef
57.
go back to reference Maquet P, Dive D, Salmon E, Sadzot B, Franco G, Poirrier R, et al. Cerebral glucose utilization during sleep-wake cycle in man determined by positron emission tomography and [18F]2-fluoro-2-deoxy-D-glucose method. Brain Res. 1990;513(1):136–43. Maquet P, Dive D, Salmon E, Sadzot B, Franco G, Poirrier R, et al. Cerebral glucose utilization during sleep-wake cycle in man determined by positron emission tomography and [18F]2-fluoro-2-deoxy-D-glucose method. Brain Res. 1990;513(1):136–43.
58.
go back to reference Kennedy C, Gillin JC, Mendelson W, Suda S, Miyaoka M, Ito M, et al. Local cerebral glucose utilization in non-rapid eye movement sleep. Nature. 1982;297(5864):325–7. Kennedy C, Gillin JC, Mendelson W, Suda S, Miyaoka M, Ito M, et al. Local cerebral glucose utilization in non-rapid eye movement sleep. Nature. 1982;297(5864):325–7.
59.
go back to reference Ramm P, Frost BJ. Cerebral and local cerebral metabolism in the cat during slow wave and REM sleep. Brain Res. 1986;365(1):112–24.PubMedCrossRef Ramm P, Frost BJ. Cerebral and local cerebral metabolism in the cat during slow wave and REM sleep. Brain Res. 1986;365(1):112–24.PubMedCrossRef
60.
go back to reference Heiss WD, Pawlik G, Herholz K, Wagner R, Wienhard K. Regional cerebral glucose metabolism in man during wakefulness, sleep, and dreaming. Brain Res. 1985;327(1–2):362–6.PubMedCrossRef Heiss WD, Pawlik G, Herholz K, Wagner R, Wienhard K. Regional cerebral glucose metabolism in man during wakefulness, sleep, and dreaming. Brain Res. 1985;327(1–2):362–6.PubMedCrossRef
61.
go back to reference Fox PT, Raichle ME, Mintun MA, Dence C. Nonoxidative glucose consumption during focal physiologic neural activity. Science. 1988;241(4864):462–4.PubMedCrossRef Fox PT, Raichle ME, Mintun MA, Dence C. Nonoxidative glucose consumption during focal physiologic neural activity. Science. 1988;241(4864):462–4.PubMedCrossRef
62.
go back to reference Madsen PL, Schmidt JF, Wildschiodtz G, Friberg L, Holm S, Vorstrup S, et al. Cerebral O2 metabolism and cerebral blood flow in humans during deep and rapid-eye-movement sleep. J Appl Physiol (Bethesda, Md: 1985). 1991;70(6):2597–601.CrossRef Madsen PL, Schmidt JF, Wildschiodtz G, Friberg L, Holm S, Vorstrup S, et al. Cerebral O2 metabolism and cerebral blood flow in humans during deep and rapid-eye-movement sleep. J Appl Physiol (Bethesda, Md: 1985). 1991;70(6):2597–601.CrossRef
63.
go back to reference Madsen PL, Schmidt JF, Holm S, Vorstrup S, Lassen NA, Wildschiodtz G. Cerebral oxygen metabolism and cerebral blood flow in man during light sleep (stage 2). Brain Res. 1991;557(1–2):217–20.PubMedCrossRef Madsen PL, Schmidt JF, Holm S, Vorstrup S, Lassen NA, Wildschiodtz G. Cerebral oxygen metabolism and cerebral blood flow in man during light sleep (stage 2). Brain Res. 1991;557(1–2):217–20.PubMedCrossRef
65.
go back to reference Fox PT, Raichle ME. Focal physiological uncoupling of cerebral blood flow and oxidative metabolism during somatosensory stimulation in human subjects. Proc Natl Acad Sci U S A. 1986;83(4):1140–4.PubMedPubMedCentralCrossRef Fox PT, Raichle ME. Focal physiological uncoupling of cerebral blood flow and oxidative metabolism during somatosensory stimulation in human subjects. Proc Natl Acad Sci U S A. 1986;83(4):1140–4.PubMedPubMedCentralCrossRef
66.
go back to reference Roland PE, Eriksson L, Stone-Elander S, Widen L. Does mental activity change the oxidative metabolism of the brain? J Neurosci. 1987;7(8):2373–89.PubMed Roland PE, Eriksson L, Stone-Elander S, Widen L. Does mental activity change the oxidative metabolism of the brain? J Neurosci. 1987;7(8):2373–89.PubMed
67.
go back to reference •• Cirelli C, Gutierrez CM, Tononi G. Extensive and divergent effects of sleep and wakefulness on brain gene expression. Neuron. 2004;41(1):35–43. Using microarray technology, a neat experimental design and clever contextual integration, this study shows that ~10% of transcripts in the rat cerebral cortex are differentially expressed between day and night and that half of these are modulated by sleep and wakefulness independent of circadian rythms. PubMedCrossRef •• Cirelli C, Gutierrez CM, Tononi G. Extensive and divergent effects of sleep and wakefulness on brain gene expression. Neuron. 2004;41(1):35–43. Using microarray technology, a neat experimental design and clever contextual integration, this study shows that ~10% of transcripts in the rat cerebral cortex are differentially expressed between day and night and that half of these are modulated by sleep and wakefulness independent of circadian rythms. PubMedCrossRef
71.
go back to reference Neufeld-Cohen A, Robles MS, Aviram R, Manella G, Adamovich Y, Ladeuix B, et al. Circadian control of oscillations in mitochondrial rate-limiting enzymes and nutrient utilization by PERIOD proteins. Proc Natl Acad Sci U S A. 2016;113(12):E1673–82. https://doi.org/10.1073/pnas.1519650113. Neufeld-Cohen A, Robles MS, Aviram R, Manella G, Adamovich Y, Ladeuix B, et al. Circadian control of oscillations in mitochondrial rate-limiting enzymes and nutrient utilization by PERIOD proteins. Proc Natl Acad Sci U S A. 2016;113(12):E1673–82. https://​doi.​org/​10.​1073/​pnas.​1519650113.
72.
76.
go back to reference Clarke DD, Sokoloff L. Basic neurochemistry. In: Agranoff BW, Fisher RW, Uhler MD, editors. Siegel GJ. Philadelphia: Lippincott Williams & Wilkins; 1999. Clarke DD, Sokoloff L. Basic neurochemistry. In: Agranoff BW, Fisher RW, Uhler MD, editors. Siegel GJ. Philadelphia: Lippincott Williams & Wilkins; 1999.
78.
go back to reference Siesjo DP. Brain energy metabolism. New York: Wiley; 1978. Siesjo DP. Brain energy metabolism. New York: Wiley; 1978.
81.
go back to reference Kerpel-Fronius S. Discussion. In: Wolstenholme GEW, editor. Somatic stability in the newly born. London: J. & a. Churchill LTD; 1961. p. 73. Kerpel-Fronius S. Discussion. In: Wolstenholme GEW, editor. Somatic stability in the newly born. London: J. & a. Churchill LTD; 1961. p. 73.
82.
go back to reference Edmond J. Energy metabolism in developing brain cells. Can J Physiol Pharmacol. 1992;70(Suppl):S118–29.PubMedCrossRef Edmond J. Energy metabolism in developing brain cells. Can J Physiol Pharmacol. 1992;70(Suppl):S118–29.PubMedCrossRef
87.
go back to reference Kong J, Shepel PN, Holden CP, Mackiewicz M, Pack AI, Geiger JD. Brain glycogen decreases with increased periods of wakefulness: implications for homeostatic drive to sleep. J Neurosci. 2002;22(13):5581–7. doi:20026500.PubMedCrossRef Kong J, Shepel PN, Holden CP, Mackiewicz M, Pack AI, Geiger JD. Brain glycogen decreases with increased periods of wakefulness: implications for homeostatic drive to sleep. J Neurosci. 2002;22(13):5581–7. doi:20026500.PubMedCrossRef
94.
go back to reference Ebert D, Haller RG, Walton ME. Energy contribution of octanoate to intact rat brain metabolism measured by 13C nuclear magnetic resonance spectroscopy. J Neurosci. 2003;23:5928–35.PubMedCrossRef Ebert D, Haller RG, Walton ME. Energy contribution of octanoate to intact rat brain metabolism measured by 13C nuclear magnetic resonance spectroscopy. J Neurosci. 2003;23:5928–35.PubMedCrossRef
95.
go back to reference •• Miller JC, Gnaedinger JM, Rapoport SI. Utilization of plasma fatty acid in rat brain: distribution of [14C]palmitate between oxidative and synthetic pathways. J Neurochem. 1987;49:1507–14. Building on knowlegde from prior in vitro studies, this study shows that intravenously injected [14C]palmitate into awake rats are taken up by the brain in vivo, about 50% undergo beta-oxidation and are temporally incoorporated into aqueous metabolite pools. The rest is incoorporated into stable lipid and protein compartments. •• Miller JC, Gnaedinger JM, Rapoport SI. Utilization of plasma fatty acid in rat brain: distribution of [14C]palmitate between oxidative and synthetic pathways. J Neurochem. 1987;49:1507–14. Building on knowlegde from prior in vitro studies, this study shows that intravenously injected [14C]palmitate into awake rats are taken up by the brain in vivo, about 50% undergo beta-oxidation and are temporally incoorporated into aqueous metabolite pools. The rest is incoorporated into stable lipid and protein compartments.
96.
go back to reference Gnaedinger J, Miller J. Cerebral metabolism of plasma palmitate in awake, adult rat: subcellular localization. Neurochem Res. 1988;13:21–9.PubMedCrossRef Gnaedinger J, Miller J. Cerebral metabolism of plasma palmitate in awake, adult rat: subcellular localization. Neurochem Res. 1988;13:21–9.PubMedCrossRef
98.
go back to reference Arai T, Wakabayashi S-i, Channing MA, Dunn BB, Der MG, Bell JM, et al. Incorporation of [1-carbon-11]palmitate in monkey brain using PET. J Nucl Med. 1995;36:2261–7.PubMed Arai T, Wakabayashi S-i, Channing MA, Dunn BB, Der MG, Bell JM, et al. Incorporation of [1-carbon-11]palmitate in monkey brain using PET. J Nucl Med. 1995;36:2261–7.PubMed
99.
go back to reference • Karmi A, Iozzo P, Viljanen A, Hirvonen J, Fielding Ba, Virtanen K et al. Increased brain fftty acid uptake in etabolic syndrome. 2010;59. https://doi.org/10.2337/db09-0138. Using positron emission tomography (PE) this study shows that [ 11 C]-palmitate and [ 18 F]fluoro-6-thia-heptadecanoic acid are taken up by the human brain primarily in gray matter regions. • Karmi A, Iozzo P, Viljanen A, Hirvonen J, Fielding Ba, Virtanen K et al. Increased brain fftty acid uptake in etabolic syndrome. 2010;59. https://​doi.​org/​10.​2337/​db09-0138. Using positron emission tomography (PE) this study shows that [ 11 C]-palmitate and [ 18 F]fluoro-6-thia-heptadecanoic acid are taken up by the human brain primarily in gray matter regions.
101.
102.
go back to reference Seelig A. The role of size and charge for blood-brain barrier permeation of drugs and fatty acids. J Mol Neurosci. 2007;33(1):32–41.PubMedCrossRef Seelig A. The role of size and charge for blood-brain barrier permeation of drugs and fatty acids. J Mol Neurosci. 2007;33(1):32–41.PubMedCrossRef
108.
go back to reference Auestad N, Korsak RA, Morrow JW, Edmond J. Fatty acid oxidation and ketogenesis by astrocytes in primary culture. J Neurochem. 1991;56(4):1376–86.PubMedCrossRef Auestad N, Korsak RA, Morrow JW, Edmond J. Fatty acid oxidation and ketogenesis by astrocytes in primary culture. J Neurochem. 1991;56(4):1376–86.PubMedCrossRef
114.
go back to reference Ishii-Iwamoto EL, Ferrarese MLL, Constantin J, Salgueiro-Pagadigorria C, Bracht A. Effects of norepinephrine on the metabolism of fatty acid with different chain lengths in the perfused rat liver. Mol Cell Biochem. 2000;205:13–23.PubMedCrossRef Ishii-Iwamoto EL, Ferrarese MLL, Constantin J, Salgueiro-Pagadigorria C, Bracht A. Effects of norepinephrine on the metabolism of fatty acid with different chain lengths in the perfused rat liver. Mol Cell Biochem. 2000;205:13–23.PubMedCrossRef
115.
go back to reference Chang MC, Wakabayashi S, Bell JM. The effect of methyl palmoxirate on incorporation of [U-14C] palmitate into rat brain. Neurochem Res. 1994;19(9):1217–23.PubMedCrossRef Chang MC, Wakabayashi S, Bell JM. The effect of methyl palmoxirate on incorporation of [U-14C] palmitate into rat brain. Neurochem Res. 1994;19(9):1217–23.PubMedCrossRef
123.
go back to reference Nomura T, Iguchi A, Sakamoto N, Harris RA. Effects of octanoate and acetate upon hepatic glycolysis and lipogenesis. Biochim Biophys Acta. 1983;754(3):315–20.PubMedCrossRef Nomura T, Iguchi A, Sakamoto N, Harris RA. Effects of octanoate and acetate upon hepatic glycolysis and lipogenesis. Biochim Biophys Acta. 1983;754(3):315–20.PubMedCrossRef
125.
130.
go back to reference Guzman M, Blazquez C. Is there an astrocyte-neuron ketone body shuttle? Trends Endocrinol Metab. 2001;12(4):169–73.PubMedCrossRef Guzman M, Blazquez C. Is there an astrocyte-neuron ketone body shuttle? Trends Endocrinol Metab. 2001;12(4):169–73.PubMedCrossRef
131.
go back to reference Ruderman NB, Ross PS, Berger M, Goodman MN. Regulation of glucose and ketone-body metabolism in brain of anaesthetized rats. Biochem J. 1974;138(1):1–10.PubMedPubMedCentralCrossRef Ruderman NB, Ross PS, Berger M, Goodman MN. Regulation of glucose and ketone-body metabolism in brain of anaesthetized rats. Biochem J. 1974;138(1):1–10.PubMedPubMedCentralCrossRef
136.
go back to reference • Dash MB, Douglas CL, Vyazovskiy VV, Cirelli C, Tononi G. Long-term homeostasis of extracellular glutamate in the rat cerebral cortex across sleep and waking states. J Neurosci. 2009;29(3):620–9. https://doi.org/10.1523/jneurosci.5486-08.2009. This study reports the glutamate concentration in two cortical areas of freely behaving rats, recorded with high temporal resolution simultaneously with local field potentials (LFPs) and electroencephalograms (EEGs) from the contralateral cortex. They show that the concentration of glutamate varies as a function of behavioral state. PubMedPubMedCentralCrossRef • Dash MB, Douglas CL, Vyazovskiy VV, Cirelli C, Tononi G. Long-term homeostasis of extracellular glutamate in the rat cerebral cortex across sleep and waking states. J Neurosci. 2009;29(3):620–9. https://​doi.​org/​10.​1523/​jneurosci.​5486-08.​2009. This study reports the glutamate concentration in two cortical areas of freely behaving rats, recorded with high temporal resolution simultaneously with local field potentials (LFPs) and electroencephalograms (EEGs) from the contralateral cortex. They show that the concentration of glutamate varies as a function of behavioral state. PubMedPubMedCentralCrossRef
139.
141.
go back to reference Kimura I, Inoue D, Maeda T, Hara T, Ichimura A, Miyauchi S, et al. Short-chain fatty acids and ketones directly regulate sympathetic nervous system via G protein-coupled receptor 41 (GPR41). Proc Natl Acad Sci U S A. 2011;108(19):8030–5. https://doi.org/10.1073/pnas.1016088108. Kimura I, Inoue D, Maeda T, Hara T, Ichimura A, Miyauchi S, et al. Short-chain fatty acids and ketones directly regulate sympathetic nervous system via G protein-coupled receptor 41 (GPR41). Proc Natl Acad Sci U S A. 2011;108(19):8030–5. https://​doi.​org/​10.​1073/​pnas.​1016088108.
142.
go back to reference Trinder J, Kleiman J, Carrington M, Smith S, Breen S, Tan N, et al. Autonomic activity during human sleep as a function of time and sleep stage. J Sleep Res. 2001;10(4):253–64. Trinder J, Kleiman J, Carrington M, Smith S, Breen S, Tan N, et al. Autonomic activity during human sleep as a function of time and sleep stage. J Sleep Res. 2001;10(4):253–64.
146.
go back to reference • Schulz JG, Laranjeira A, Van Huffel L, Gartner A, Vilain S, Bastianen J, et al. Glial beta-oxidation regulates Drosophila energy metabolism. Sci Rep. 2015;5:7805. https://doi.org/10.1038/srep07805. Using Drosophila as a model organism, this is the first study showing that glial fatty acid beta-oxidation is important for energy-homeostasis and prevention of triacylglycerol build-up. • Schulz JG, Laranjeira A, Van Huffel L, Gartner A, Vilain S, Bastianen J, et al. Glial beta-oxidation regulates Drosophila energy metabolism. Sci Rep. 2015;5:7805. https://​doi.​org/​10.​1038/​srep07805. Using Drosophila as a model organism, this is the first study showing that glial fatty acid beta-oxidation is important for energy-homeostasis and prevention of triacylglycerol build-up.
150.
go back to reference Miyagawa T, Miyadera H, Tanaka S, Kawashima M, Shimada M, Honda Y, et al. Abnormally low serum acylcarnitine levels in narcolepsy patients. Sleep. 2011;34:349–53A. Miyagawa T, Miyadera H, Tanaka S, Kawashima M, Shimada M, Honda Y, et al. Abnormally low serum acylcarnitine levels in narcolepsy patients. Sleep. 2011;34:349–53A.
152.
154.
go back to reference •• Knobloch M, Pilz GA, Ghesquiere B, Kovacs WJ, Wegleiter T, Moore DL, et al. A fatty acid oxidation-dependent metabolic shift regulates adult neural stem cell activity. Cell Rep. 2017;20(9):2144–55. https://doi.org/10.1016/j.celrep.2017.08.029. The first study to show how fatty acid oxidation capacity changes the state and function of cell types in the mouse brain. Inhibition of fatty acid oxidation by increased malonyl-CoA levels shifts quiescents neural stem cells to become proliferative. PubMedPubMedCentralCrossRef •• Knobloch M, Pilz GA, Ghesquiere B, Kovacs WJ, Wegleiter T, Moore DL, et al. A fatty acid oxidation-dependent metabolic shift regulates adult neural stem cell activity. Cell Rep. 2017;20(9):2144–55. https://​doi.​org/​10.​1016/​j.​celrep.​2017.​08.​029. The first study to show how fatty acid oxidation capacity changes the state and function of cell types in the mouse brain. Inhibition of fatty acid oxidation by increased malonyl-CoA levels shifts quiescents neural stem cells to become proliferative. PubMedPubMedCentralCrossRef
155.
go back to reference Lamers JM, Stinis HT, Montfoort A, Hulsmann WC. The effect of lipid intermediates on Ca2+ and Na+ permeability and (Na+ / K+)-ATPase of cardiac sarcolemma. A possible role in myocardial ischemia. Biochim Biophys Acta. 1984;774(1):127–37.PubMedCrossRef Lamers JM, Stinis HT, Montfoort A, Hulsmann WC. The effect of lipid intermediates on Ca2+ and Na+ permeability and (Na+ / K+)-ATPase of cardiac sarcolemma. A possible role in myocardial ischemia. Biochim Biophys Acta. 1984;774(1):127–37.PubMedCrossRef
159.
go back to reference •• Díaz-Muñoz M, Hernández-Muñoz R, Suárez J, de Sánchez VC. Day-night cycle of lipid peroxidation in rat cerebral cortex and their relationship to the gperoxide dismutase activity. Neuroscience. 1985;16:859–63. https://doi.org/10.1016/0306-4522(85)90100-9. This study measures the state of lipoperoxidation, glutathione cycle components and superoxide dismutase activity every four hour in the the cerebral cortex of the rat, and shows that these parameters exhibit day-night rhythms. PubMedCrossRef •• Díaz-Muñoz M, Hernández-Muñoz R, Suárez J, de Sánchez VC. Day-night cycle of lipid peroxidation in rat cerebral cortex and their relationship to the gperoxide dismutase activity. Neuroscience. 1985;16:859–63. https://​doi.​org/​10.​1016/​0306-4522(85)90100-9. This study measures the state of lipoperoxidation, glutathione cycle components and superoxide dismutase activity every four hour in the the cerebral cortex of the rat, and shows that these parameters exhibit day-night rhythms. PubMedCrossRef
160.
Metadata
Title
Cerebral Metabolic Changes During Sleep
Authors
Nadia Nielsen Aalling
Maiken Nedergaard
Mauro DiNuzzo
Publication date
01-09-2018
Publisher
Springer US
Published in
Current Neurology and Neuroscience Reports / Issue 9/2018
Print ISSN: 1528-4042
Electronic ISSN: 1534-6293
DOI
https://doi.org/10.1007/s11910-018-0868-9

Other articles of this Issue 9/2018

Current Neurology and Neuroscience Reports 9/2018 Go to the issue

Headache (R B Halker, Section Editor)

Photophobia: When Light Hurts, a Review

Sleep (M Thorpy and M Billiard, Section Editors)

Neuroimaging in the Kleine-Levin Syndrome