Journal Home > Volume 8 , Issue 4

Alzheimer’s disease (AD), the foremost variant of dementia, has been associated with a menagerie of risk factors, many of which are considered to be modifiable. Among these modifiable risk factors is circadian rhythm, the chronobiological system that regulates sleep-wake cycles, food consumption timing, hydration timing, and immune responses amongst many other necessary physiological processes. Circadian rhythm at the level of the suprachiasmatic nucleus (SCN), is tightly regulated in the human body by a host of biomolecular substances, principally the hormones melatonin, cortisol, and serotonin. In addition, photic information projected along afferent pathways to the SCN and peripheral oscillators regulates the synthesis of these hormones and mediates the manner in which they act on the SCN and its substructures. Dysregulation of this cycle, whether induced by environmental changes involving irregular exposure to light, or through endogenous pathology, will have a negative impact on immune system optimization and will heighten the deposition of Aβ and the hyperphosphorylation of the tau protein. Given these correlations, it appears that there is a physiologic association between circadian rhythm dysregulation and AD. This review will explore the physiology of circadian dysregulation in the AD brain, and will propose a basic model for its role in AD-typical pathology, derived from the literature compiled and referenced throughout.


menu
Abstract
Full text
Outline
About this article

Circadian dysregulation and Alzheimer’s disease: A comprehensive review

Show Author's information Peter Iacobelli( )
Department of Arts and Sciences, University of South Carolina, Columbia, USA

Abstract

Alzheimer’s disease (AD), the foremost variant of dementia, has been associated with a menagerie of risk factors, many of which are considered to be modifiable. Among these modifiable risk factors is circadian rhythm, the chronobiological system that regulates sleep-wake cycles, food consumption timing, hydration timing, and immune responses amongst many other necessary physiological processes. Circadian rhythm at the level of the suprachiasmatic nucleus (SCN), is tightly regulated in the human body by a host of biomolecular substances, principally the hormones melatonin, cortisol, and serotonin. In addition, photic information projected along afferent pathways to the SCN and peripheral oscillators regulates the synthesis of these hormones and mediates the manner in which they act on the SCN and its substructures. Dysregulation of this cycle, whether induced by environmental changes involving irregular exposure to light, or through endogenous pathology, will have a negative impact on immune system optimization and will heighten the deposition of Aβ and the hyperphosphorylation of the tau protein. Given these correlations, it appears that there is a physiologic association between circadian rhythm dysregulation and AD. This review will explore the physiology of circadian dysregulation in the AD brain, and will propose a basic model for its role in AD-typical pathology, derived from the literature compiled and referenced throughout.

Keywords: circadian, Alzheimer’s disease, tau, amyloid, pathology

References(222)

[1]
Homolak J, Mudrovčić M, Vukić B, et al. Circadian rhythm and Alzheimer’s disease. Med Sci: Basel 2018, 6(3): E52.
[2]
Rajmohan R, Reddy PH. Amyloid-beta and phosphorylated tau accumulations cause abnormalities at synapses of Alzheimer’s disease neurons. J Alzheimers Dis 2017, 57(4): 975–999.
[3]
Potter GDM, Skene DJ, Arendt J, et al. Circadian rhythm and sleep disruption: causes, metabolic consequences, and countermeasures. Endocr Rev 2016, 37(6): 584–608.
[4]
Blume C, Garbazza C, Spitschan M. Effects of light on human circadian rhythms, sleep and mood. Somnologie 2019, 23(3): 147–156.
[5]
James SM, Honn KA, Gaddameedhi S, et al. Shift work: disrupted circadian rhythms and sleep-implications for health and well-being. Curr Sleep Med Rep 2017, 3(2): 104–112.
[6]
Liang X, FitzGerald GA. Timing the microbes: the circadian rhythm of the gut microbiome. J Biol Rhythms 2017, 32(6): 505–515.
[7]
Hardeland R. Neurobiology, pathophysiology, and treatment of melatonin deficiency and dysfunction. Sci World J 2012, 2012: 640389.
[8]
Hadoush H, Alqudah A, Banihani SA, et al. Melatonin serum level, sleep functions, and depression level after bilateral anodal transcranial direct current stimulation in patients with Parkinson’s disease: a feasibility study. Sleep Sci 2021, 14(spec 1): 25–30.
[9]
Eugene AR, Masiak J. The neuroprotective aspects of sleep. MEDtube Sci 2015, 3(1): 35–40.
[10]
Zisapel N. New perspectives on the role of melatonin in human sleep, circadian rhythms and their regulation. Br J Pharmacol 2018, 175(16): 3190–3199.
[11]
Mohd Azmi NAS, Juliana N, Azmani S, et al. Cortisol on circadian rhythm and its effect on cardiovascular system. Int J Environ Res Public Health 2021, 18(2): E676.
[12]
Cagnacci A, Cannoletta M, Caretto S, et al. Increased cortisol level: a possible link between climacteric symptoms and cardiovascular risk factors. Menopause 2011, 18(3): 273–278.
[13]
Scher A, Hall WA, Zaidman-Zait A, et al. Sleep quality, cortisol levels, and behavioral regulation in toddlers. Dev Psychobiol 2010, 52(1): 44–53.
[14]
Reiter RJ, Tan DX, Korkmaz A, et al. Melatonin and stable circadian rhythms optimize maternal, placental and fetal physiology. Hum Reprod Update 2014, 20(2): 293–307.
[15]
Perez-Pozuelo I, Zhai B, Palotti J, et al. The future of sleep health: a data-driven revolution in sleep science and medicine. NPJ Digit Med 2020, 3: 42.
[16]
Khademi A, El-Manzalawy Y, Master L, et al. Personalized sleep parameters estimation from actigraphy: a machine learning approach. Nat Sci Sleep 2019, 11: 387–399.
[17]
Rasch B, Born J. About sleep’s role in memory. Physiol Rev 2013, 93(2): 681–766.
[18]
Shi M, Li C, Tian X, et al. Can control infections slow down the progression of Alzheimer’s disease? Talking about the role of infections in Alzheimer’s disease. Front Aging Neurosci 2021, 13: 685863.
[19]
Cross ZR, Kohler MJ, Schlesewsky M, et al. Sleep-dependent memory consolidation and incremental sentence comprehension: computational dependencies during language learning as revealed by neuronal oscillations. Front Hum Neurosci 2018, 12: 18.
[20]
Guo T, Zhang D, Zeng Y, et al. Molecular and cellular mechanisms underlying the pathogenesis of Alzheimer’s disease. Mol Neurodegener 2020, 15(1): 40.
[21]
Giggins OM, Doyle J, Hogan K, et al. The impact of a cycled lighting intervention on nursing home residents: A pilot study. Gerontol Geriatr Med 2019, 5: 233372141989745.
[22]
Targa ADS, Benítez ID, Dakterzada F, et al. The circadian rest-activity pattern predicts cognitive decline among mild-moderate Alzheimer’s disease patients. Alzheimers Res Ther 2021, 13(1): 161.
[23]
Haque RU, Levey AI. Alzheimer’s disease: a clinical perspective and future nonhuman primate research opportunities. PNAS 2019: 201912954.
[24]
O’Brien RJ, Wong PC. Amyloid precursor protein processing and Alzheimer’s disease. Annu Rev Neurosci 2011, 34: 185–204.
[25]
Chow VW, Mattson MP, Wong PC, et al. An overview of APP processing enzymes and products. Neuromolecular Med 2010, 12(1): 1–12.
[26]
Murphy MP, LeVine H. Alzheimer’s disease and the amyloid-beta peptide. J Alzheimers Dis 2010, 19(1): 311–323.
[27]
Šimić G, Babić Leko M, Wray S, et al. Tau protein hyperphosphorylation and aggregation in Alzheimer’s disease and other tauopathies, and possible neuroprotective strategies. Biomolecules 2016, 6(1): 6.
[28]
Nizynski B, Dzwolak W, Nieznanski K. Amyloidogenesis of tau protein. Protein Sci 2017, 26(11): 2126–2150.
[29]
Dehmelt L, Halpain S. The MAP2/Tau family of microtubule-associated proteins. Genome Biol 2005, 6(1): 204.
[30]
Morris M, Maeda S, Vossel K, et al. The many faces of tau. Neuron 2011, 70(3): 410–426.
[31]
Kinney JW, Bemiller SM, Murtishaw AS, et al. Inflammation as a central mechanism in Alzheimer’s disease. Alzheimers Dement: N Y 2018, 4: 575–590.
[32]
An Y, Varma VR, Varma S, et al. Evidence for brain glucose dysregulation in Alzheimer’s disease. Alzheimer’s Dement 2018, 14(3): 318–329.
[33]
Mietelska-Porowska A, Wasik U, Goras M, et al. Tau protein modifications and interactions: their role in function and dysfunction. Int J Mol Sci 2014, 15(3): 4671–4713.
[34]
Yu W, Jin H, Huang Y. Mitochondria-associated membranes (MAMs): a potential therapeutic target for treating Alzheimer’s disease. Clin Sci: Lond 2021, 135(1): 109–126.
[35]
Spires-Jones TL, Hyman BT. The intersection of amyloid beta and tau at synapses in Alzheimer’s disease. Neuron 2014, 82(4): 756–771.
[36]
Stefanis L. α-synuclein in Parkinson’s disease. Cold Spring Harb Perspect Med 2012, 2(2): a009399.
[37]
Lemere CA, Masliah E. Can Alzheimer disease be prevented by amyloid-beta immunotherapy? Nat Rev Neurol 2010, 6(2): 108–119.
[38]
Mucke L, Masliah E, Yu GQ, et al. High-level neuronal expression of abeta 1-42 in wild-type human amyloid protein precursor transgenic mice: synaptotoxicity without plaque formation. J Neurosci 2000, 20(11): 4050–4058.
[39]
Iqbal K, Grundke-Iqbal I. Alzheimer’s disease, a multifactorial disorder seeking multitherapies. Alzheimer’s Dement 2010, 6(5): 420–424.
[40]
Sienski G, Narayan P, Bonner JM, et al. APOE4 disrupts intracellular lipid homeostasis in human iPSC-derived Glia. Sci Transl Med 2021, 13(583): eaaz4564.
[41]
Liu CC, Kanekiyo T, Xu HX, et al. Apolipoprotein E and Alzheimer disease: risk, mechanisms and therapy. Nat Rev Neurol 2013, 9(2): 106–118.
[42]
Bagyinszky E, Youn YC, An SS, et al. The genetics of Alzheimer’s disease. Clin Interv Aging 2014, 9: 535–551.
[43]
Liu T, Zhang LY, Joo D, et al. NF-κB signaling in inflammation. Signal Transduct Target Ther 2017, 2: 17023.
[44]
Morgan MJ, Liu ZG. Crosstalk of reactive oxygen species and NF-κB signaling. Cell Res 2011, 21(1): 103–115.
[45]
Kany S, Vollrath JT, Relja B. Cytokines in inflammatory disease. Int J Mol Sci 2019, 20(23): E6008.
[46]
Huang WJ, Zhang X, Chen WW. Role of oxidative stress in Alzheimer’s disease. Biomed Rep 2016, 4(5): 519–522.
[47]
Sonnen JA, Breitner JC, Lovell MA, et al. Free radical-mediated damage to brain in Alzheimer’s disease and its transgenic mouse models. Free Radic Biol Med 2008, 45(3): 219–230.
[48]
Kurutas EB. The importance of antioxidants which play the role in cellular response against oxidative/nitrosative stress: current state. Nutr J 2016, 15(1): 71.
[49]
Sharifi-Rad M, Anil Kumar NV, Zucca P, et al. Lifestyle, oxidative stress, and antioxidants: back and forth in the pathophysiology of chronic diseases. Front Physiol 2020, 11: 694.
[50]
Traiffort E, Kassoussi A, Zahaf A, et al. Astrocytes and microglia as major players of myelin production in normal and pathological conditions. Front Cell Neurosci 2020, 14: 79.
[51]
Frost GR, Li YM. The role of astrocytes in amyloid production and Alzheimer’s disease. Open Biol 2017, 7(12): 170228.
[52]
Zilkha-Falb R, Rachutin-Zalogin T, Cleaver L, et al. RAM-589.555 favors neuroprotective and anti-inflammatory profile of CNS-resident glial cells in acute relapse EAE affected mice. J Neuroinflammation 2020, 17(1): 313.
[53]
Cheng AH, Cheng HM. Genesis of the master circadian pacemaker in mice. Front Neurosci 2021, 15: 659974.
[54]
Ma MA, Morrison EH. Neuroanatomy, Nucleus Suprachiasmatic. [Updated 2021 Jul 31]. Treasure Island (FL): StatPearls Publishing, 2022.
[55]
Dugué L, Merriam EP, Heeger DJ, et al. Differential impact of endogenous and exogenous attention on activity in human visual cortex. Sci Rep 2020, 10(1): 21274.
[56]
Paul JR, Davis JA, Goode LK, et al. Circadian regulation of membrane physiology in neural oscillators throughout the brain. Eur J Neurosci 2020, 51(1): 109–138.
[57]
Nomura M, Saito J, Ueta Y, et al. Enhanced up-regulation of corticotropin-releasing hormone gene expression in response to restraint stress in the hypothalamic paraventricular nucleus of oxytocin gene-deficient male mice. J Neuroendocrinol 2003, 15(11): 1054–1061.
[58]
Reghunandanan V, Reghunandanan R. Neurotransmitters of the suprachiasmatic nuclei. J Circadian Rhythms 2006, 4: 2.
[59]
Paul KN, Saafir TB, Tosini G. The role of retinal photoreceptors in the regulation of circadian rhythms. Rev Endocr Metab Disord 2009, 10(4): 271–278.
[60]
Bonmati-Carrion MA, Arguelles-Prieto R, Martinez-Madrid MJ, et al. Protecting the melatonin rhythm through circadian healthy light exposure. Int J Mol Sci 2014, 15(12): 23448–23500.
[61]
Morin LP. Neuroanatomy of the extended circadian rhythm system. Exp Neurol 2013, 243: 4–20.
[62]
Herzog ED, Hermanstyne T, Smyllie NJ, et al. Regulating the suprachiasmatic nucleus (SCN) circadian clockwork: interplay between cell-autonomous and circuit-level mechanisms. Cold Spring Harb Perspect Biol 2017, 9(1): a027706.
[63]
Ciranna L. Serotonin as a modulator of glutamate- and GABA-mediated neurotransmission: implications in physiological functions and in pathology. Curr Neuropharmacol 2006, 4(2): 101–114.
[64]
Brown RE, Basheer R, McKenna JT, et al. Control of sleep and wakefulness. Physiol Rev 2012, 92(3): 1087–1187.
[65]
Wang HL, Morales M. Pedunculopontine and laterodorsal tegmental nuclei contain distinct populations of cholinergic, glutamatergic and GABAergic neurons in the rat. Eur J Neurosci 2009, 29(2): 340–358.
[66]
Lanciego JL, Luquin N, Obeso JA. Functional neuroanatomy of the basal Ganglia. Cold Spring Harb Perspect Med 2012, 2(12): a009621.
[67]
Vigneswaran J, Muthukumar SA, Shafras M, et al. An insight into Alzheimer’s disease and its on-setting novel genes. Egypt J Neurol Psychiatry Neurosurg 2021, 57: 160.
[68]
Deurveilher S, Burns J, Semba K. Indirect projections from the suprachiasmatic nucleus to the ventrolateral preoptic nucleus: a dual tract-tracing study in rat. Eur J Neurosci 2002, 16(7): 1195–1213.
[69]
Pickel L, Sung HK. Feeding rhythms and the circadian regulation of metabolism. Front Nutr 2020, 7: 39.
[70]
Patke A, Young MW, Axelrod S. Molecular mechanisms and physiological importance of circadian rhythms. Nat Rev Mol Cell Biol 2020, 21(2): 67–84.
[71]
Hill NL, Kolanowski AM, Gill DJ. Plasticity in early Alzheimer’s disease: an opportunity for intervention. Top Geriatr Rehabil 2011, 27(4): 257–267.
[72]
Welsh DK, Takahashi JS, Kay SA. Suprachiasmatic nucleus: cell autonomy and network properties. Annu Rev Physiol 2010, 72: 551–577.
[73]
Hirotsu C, Tufik S, Andersen ML. Interactions between sleep, stress, and metabolism: from physiological to pathological conditions. Sleep Sci 2015, 8(3): 143–152.
[74]
Duffy JF, Czeisler CA. Effect of light on human circadian physiology. Sleep Med Clin 2009, 4(2): 165–177.
[75]
Brenna A, Albrecht U. Phosphorylation and circadian molecular timing. Front Physiol 2020, 11: 612510.
[76]
Kim YH, Lazar MA. Transcriptional control of circadian rhythms and metabolism: a matter of time and space. Endocr Rev 2020, 41(5): bnaa014.
[77]
Miyazaki K, Mezaki M, Ishida N. The role of phosphorylation and degradation of hPER protein oscillation in normal human fibroblasts. Novartis Found Symp 2003, 253: 238–248;discussion249.
[78]
Sulli G, Lam MTY, Panda S. Interplay between circadian clock and cancer: new frontiers for cancer treatment. Trends Cancer 2019, 5(8): 475–494.
[79]
Azmitia EC. Modern views on an ancient chemical: serotonin effects on cell proliferation, maturation, and apoptosis. Brain Res Bull 2001, 56(5): 413–424.
[80]
Gooley JJ, Chamberlain K, Smith KA, et al. Exposure to room light before bedtime suppresses melatonin onset and shortens melatonin duration in humans. J Clin Endocrinol Metab 2011, 96(3): E463–E472.
[81]
Grivas TB, Savvidou OD. Melatonin the “light of night” in human biology and adolescent idiopathic scoliosis. Scoliosis 2007, 2: 6.
[82]
Brennan R, Jan JE, Lyons CJ. Light, dark, and melatonin: emerging evidence for the importance of melatonin in ocular physiology. Eye 2007, 21(7): 901–908.
[83]
Todd WD. Potential pathways for circadian dysfunction and sundowning-related behavioral aggression in Alzheimer’s disease and related dementias. Front Neurosci 2020, 14: 910.
[84]
Tordjman S, Chokron S, Delorme R, et al. Melatonin: pharmacology, functions and therapeutic benefits. Curr Neuropharmacol 2017, 15(3): 434–443.
[85]
Betti L, Palego L, Demontis GC, et al. Hydroxyindole-O-methyltransferase (HIOMT) activity in the Retina of melatonin-proficient mice. Heliyon 2019, 5(9): e02417.
[86]
Pévet P. Melatonin. Dialogues Clin Neurosci 2002, 4(1): 57–72.
[87]
Aulinas A. Physiology of the pineal gland and melatonin. [Updated 2019 Dec 10]. In Endotext [Internet]. Feingold KR, Anawalt B, Boyce A, et al., Eds. South Dartmouth (MA): MDText.com, Inc., 2000.
[88]
Perez DM. α1-adrenergic receptors in neurotransmission, synaptic plasticity, and cognition. Front Pharmacol 2020, 11: 581098.
[89]
Jockers R, Maurice P, Boutin JA, et al. Melatonin receptors, heterodimerization, signal transduction and binding sites: what’s new? Br J Pharmacol 2008, 154(6): 1182–1195.
[90]
von Gall C, Stehle JH, Weaver DR. Mammalian melatonin receptors: molecular biology and signal transduction. Cell Tissue Res 2002, 309(1): 151–162.
[91]
Arendt J. Melatonin: countering chaotic time cues. Front Endocrinol (Lausanne) 2019, 10: 391.
[92]
Gupta R, Das S, Gujar K, et al. Clinical practice guidelines for sleep disorders. Indian J Psychiatry 2017, 59(suppl 1): S116–S138.
[93]
Thau L, Gandhi J, Sharma S. Physiology, Cortisol. [Updated 2021 Sep 6]. Treasure Island (FL): StatPearls Publishing, 2022.
[94]
Smith SM, Vale WW. The role of the hypothalamic-pituitary-adrenal axis in neuroendocrine responses to stress. Dialogues Clin Neurosci 2006, 8(4): 383–395.
[95]
Hu J, Zhang Z, Shen WJ, et al. Cellular cholesterol delivery, intracellular processing and utilization for biosynthesis of steroid hormones. Nutr Metab: Lond 2010, 7: 47.
[96]
Akerstedt T, Wright KP. Sleep loss and fatigue in shift work and shift work disorder. Sleep Med Clin 2009, 4(2): 257–271.
[97]
Chung S, Son GH, Kim K. Circadian rhythm of adrenal glucocorticoid: its regulation and clinical implications. Biochim Biophys Acta BBA Mol Basis Dis 2011, 1812(5): 581–591.
[98]
Allen MJ, Sharma S. Physiology, Adrenocorticotropic Hormone (ACTH) [Updated 2021 Aug 17]. Treasure Island (FL): StatPearls Publishing; 2022.
[99]
McIntyre CK, Roozendaal B. Adrenal Stress Hormones and Enhanced Memory for Emotionally Arousing Experiences. In Neural Plasticity and Memory: From Genes to Brain Imaging. Bermúdez-Rattoni F, Ed. Boca Raton (FL): CRC Press/Taylor & Francis, 2007.
DOI
[100]
Ranabir S, Reetu K. Stress and hormones. Indian J Endocrinol Metab 2011, 15(1): 18–22.
[100]
Hannibal KE, Bishop MD. Chronic stress, cortisol dysfunction, and pain: a psychoneuroendocrine rationale for stress management in pain rehabilitation. Phys Ther 2014, 94(12): 1816–1825.
[102]
Sharma A, Gerbarg PL, Brown RP. Non-pharmacological treatments for ADHD in youth. Adolesc Psychiatry: Hilversum 2015, 5(2): 84–95.
[103]
Singh A, Yeh CJ, Verma N, et al. Overview of attention deficit hyperactivity disorder in young children. Health Psychol Res 2015, 3(2): 2115.
[104]
McGrath LM, Stoodley CJ. Are there shared neural correlates between dyslexia and ADHD? A meta-analysis of voxel-based morphometry studies. J Neurodev Disord 2019, 11(1): 31.
[105]
Gan JH, Wang XD, Shi ZH, et al. The impact of rotating night shift work and daytime recharge on cognitive performance among retired nurses. Front Aging Neurosci 2022, 13: 827772.
[106]
Frazer A, Hensler JG. Serotonin. In Basic Neurochemistry: Molecular, Cellular and Medical Aspects. 6th ed. Siegel GJ, Agranoff BW, Albers RW, et al., Eds. Philadelphia: Lippincott-Raven, 1999.
[107]
Watts SW, Morrison SF, Davis RP, et al. Serotonin and blood pressure regulation. Pharmacol Rev 2012, 64(2): 359–388.
[108]
Sangkuhl K, Klein TE, Altman RB. Selective serotonin reuptake inhibitors pathway. Pharmacogenet Genomics 2009, 19(11): 907–909.
[109]
Best J, Nijhout HF, Reed M. Serotonin synthesis, release and reuptake in terminals: a mathematical model. Theor Biol Med Model 2010, 7: 34.
[110]
Waselus M, Valentino RJ, van Bockstaele EJ. Collateralized dorsal raphe nucleus projections: a mechanism for the integration of diverse functions during stress. J Chem Neuroanat 2011, 41(4): 266–280.
[111]
Alonso A, Merchán P, Sandoval JE, et al. Development of the serotonergic cells in murine raphe nuclei and their relations with rhombomeric domains. Brain Struct Funct 2013, 218(5): 1229–1277.
[112]
Morin LP. Serotonin and the regulation of mammalian circadian rhythmicity. Ann Med 1999, 31(1): 12–33.
[113]
Berg BA, Schoenbaum G, McDannald MA. The dorsal raphe nucleus is integral to negative prediction errors in Pavlovian fear. Eur J Neurosci 2014, 40(7): 3096–3101.
[114]
Vadnie CA, McClung CA. Circadian rhythm disturbances in mood disorders: insights into the role of the suprachiasmatic nucleus. Neural Plast 2017, 2017: 1504507.
[115]
Glass JD, DiNardo LA, Ehlen JC. Dorsal raphe nuclear stimulation of SCN serotonin release and circadian phase-resetting. Brain Res 2000, 859(2): 224–232.
[116]
Bedrosian TA, Nelson RJ. Timing of light exposure affects mood and brain circuits. Transl Psychiatry 2017, 7(1): e1017.
[117]
Prosser RA. Glutamate blocks serotonergic phase advances of the mammalian circadian pacemaker through AMPA and NMDA receptors. J Neurosci 2001, 21(19): 7815–7822.
[118]
Quintero JE, McMahon DG. Serotonin modulates glutamate responses in isolated suprachiasmatic nucleus neurons. J Neurophysiol 1999, 82(2): 533–539.
[119]
Jenkins TA, Nguyen JC, Polglaze KE, et al. Influence of tryptophan and serotonin on mood and cognition with a possible role of the gut-brain axis. Nutrients 2016, 8(1): E56.
[120]
Buguet A. Is sleeping sickness a circadian disorder? The serotonergic hypothesis. Chronobiol Int 1999, 16(4): 477–489.
[121]
Biello SM. Circadian clock resetting in the mouse changes with age. Age: Dordr 2009, 31(4): 293–303.
[122]
Thakkar MM. Histamine in the regulation of wakefulness. Sleep Med Rev 2011, 15(1): 65–74.
[123]
Wu HH, Williams CV, McLoon SC. Involvement of nitric oxide in the elimination of a transient retinotectal projection in development. Science 1994, 265(5178): 1593–1596.
[124]
Korshunov KS, Blakemore LJ, Trombley PQ. Dopamine: a modulator of circadian rhythms in the central nervous system. Front Cell Neurosci 2017, 11: 91.
[125]
Itri J, Michel S, Waschek JA, et al. Circadian rhythm in inhibitory synaptic transmission in the mouse suprachiasmatic nucleus. J Neurophysiol 2004, 92(1): 311–319.
[126]
McArthur AJ, Coogan AN, Ajpru S, et al. Gastrin-releasing peptide phase-shifts suprachiasmatic nuclei neuronal rhythms in vitro. J Neurosci 2000, 20(14): 5496–5502.
[127]
Yuan XS, Wei HH, Xu W, et al. Whole-brain monosynaptic afferent projections to the cholecystokinin neurons of the suprachiasmatic nucleus. Front Neurosci 2018, 12: 807.
[128]
Hirsch D, Zukowska Z. NPY and stress 30 years later: the peripheral view. Cell Mol Neurobiol 2012, 32(5): 645–659.
[129]
Chen G, van den Pol AN. Multiple NPY receptors coexist in pre- and postsynaptic sites: inhibition of GABA release in isolated self-innervating SCN neurons. J Neurosci 1996, 16(23): 7711–7724.
[130]
Gribkoff VK, Pieschl RL, Wisialowski TA, et al. Phase shifting of circadian rhythms and depression of neuronal activity in the rat suprachiasmatic nucleus by neuropeptide Y: mediation by different receptor subtypes. J Neurosci 1998, 18(8): 3014–3022.
[131]
Hasselmo ME. The role of acetylcholine in learning and memory. Curr Opin Neurobiol 2006, 16(6): 710–715.
[132]
Zatz M, Brownstein MJ. Central depressants rapidly reduce nocturnal serotonin N-acetyltransferase activity in the rat pineal gland. Brain Res 1979, 160(2): 381–385.
[133]
Choi HJ, Lee CJ, Schroeder A, et al. Excitatory actions of GABA in the suprachiasmatic nucleus. J Neurosci 2008, 28(21): 5450–5459.
[134]
Popova E. Ionotropic GABA receptors and distal retinal ON and OFF responses. Scientifica (Cairo) 2014, 2014: 149187.
[135]
Albers HE, Walton JC, Gamble KL, et al. The dynamics of GABA signaling: Revelations from the circadian pacemaker in the suprachiasmatic nucleus. Front Neuroendocrinol 2017, 44: 35–82.
[136]
Vosko A, van Diepen HC, Kuljis D, et al. Role of vasoactive intestinal peptide in the light input to the circadian system. Eur J Neurosci 2015, 42(2): 1839–1848.
[137]
Videnovic A, Lazar AS, Barker RA, et al. ‘The clocks that time us’—circadian rhythms in neurodegenerative disorders. Nat Rev Neurol 2014, 10(12): 683–693.
[138]
Ono D, Honma KI, Honma S. Roles of neuropeptides, VIP and AVP, in the mammalian central circadian clock. Front Neurosci 2021, 15: 650154.
[139]
Nielsen HS, Hannibal J, Fahrenkrug J. Vasoactive intestinal polypeptide induces per1 and per2 gene expression in the rat suprachiasmatic nucleus late at night. Eur J Neurosci 2002, 15(3): 570–574.
[140]
Francl JM, Kaur G, Glass JD. Regulation of vasoactive intestinal polypeptide release in the suprachiasmatic nucleus circadian clock. Neuroreport 2010, 21(16): 1055–1059.
[141]
Seggio JA, Logan RW, Rosenwasser AM. Chronic ethanol intake modulates photic and non-photic circadian phase responses in the Syrian hamster. Pharmacol Biochem Behav 2007, 87(3): 297–305.
[142]
Taub A, Carbajal Y, Rimu K, et al. Arginine vasopressin-containing neurons of the suprachiasmatic nucleus project to CSF. eNeuro 2021, 8(2): ENEURO.0363–20.2021.
[143]
Buijs RM, Hurtado-Alvarado G, Soto-Tinoco E. Vasopressin: An output signal from the suprachiasmatic nucleus to prepare physiology and behaviour for the resting phase. J Neuroendocrinol 2021, 33(7): e12998.
[144]
Antle MC, Kriegsfeld LJ, Silver R. Signaling within the master clock of the brain: localized activation of mitogen-activated protein kinase by gastrin-releasing peptide. J Neurosci 2005, 25(10): 2447–2454.
[145]
Decker MJ, Rye DB, Lee SY, et al. Paradoxical sleep suppresses immediate early gene expression in the rodent suprachiasmatic nuclei. Front Neurol 2010, 1: 122.
[146]
Buhr ED, Takahashi JS. Molecular components of the Mammalian circadian clock. Handb Exp Pharmacol 2013(217): 3–27.
[147]
Park J, Jung MS, Moon E, et al. Prediction of locomotor activity by infrared motion detector on sleep-wake state in mice. Clin Psychopharmacol Neurosci 2021, 19(2): 303–312.
[148]
Holth JK, Patel TK, Holtzman DM. Sleep in Alzheimer’s disease-beyond amyloid. Neurobiol Sleep Circadian Rhythms 2017, 2: 4–14.
[149]
Ishii M, Iadecola C. Metabolic and non-cognitive manifestations of Alzheimer’s disease: the hypothalamus as both culprit and target of pathology. Cell Metab 2015, 22(5): 761–776.
[150]
Leng Y, Musiek ES, Hu K, et al. Association between circadian rhythms and neurodegenerative diseases. Lancet Neurol 2019, 18(3): 307–318.
[151]
Carter B, Justin HS, Gulick D, et al. The molecular clock and neurodegenerative disease: a stressful time. Front Mol Biosci 2021, 8: 644747.
[152]
Palmer AL, Ousman SS. Astrocytes and aging. Front Aging Neurosci 2018, 10: 337.
[153]
Logan RW, McClung CA. Rhythms of life: circadian disruption and brain disorders across the lifespan. Nat Rev Neurosci 2019, 20(1): 49–65.
[154]
Harper DG, Stopa EG, McKee AC, et al. Dementia severity and Lewy bodies affect circadian rhythms in Alzheimer disease. Neurobiol Aging 2004, 25(6): 771–781.
[155]
Agostinelli LJ, Geerling JC, Scammell TE. Basal forebrain subcortical projections. Brain Struct Funct 2019, 224(3): 1097–1117.
[156]
Wisor JP, Edgar DM, Yesavage J, et al. Sleep and circadian abnormalities in a transgenic mouse model of Alzheimer’s disease: a role for cholinergic transmission. Neuroscience 2005, 131(2): 375–385.
[157]
Germain A, Kupfer DJ. Circadian rhythm disturbances in depression. Hum Psychopharmacol 2008, 23(7): 571–585.
[158]
Cordone S, Annarumma L, Rossini PM, et al. Sleep and β-amyloid deposition in alzheimer disease: insights on mechanisms and possible innovative treatments. Front Pharmacol 2019, 10: 695.
[159]
Kang JE, Lim MM, Bateman RJ, et al. Amyloid-beta dynamics are regulated by orexin and the sleep-wake cycle. Science 2009, 326(5955): 1005–1007.
[160]
Sadeghmousavi S, Eskian M, Rahmani F, et al. The effect of insomnia on development of Alzheimer’s disease. J Neuroinflammation 2020, 17(1): 289.
[161]
Benveniste H, Liu X, Koundal S, et al. The glymphatic system and waste clearance with brain aging: a review. Gerontology 2019, 65(2): 106–119.
[162]
Besedovsky L, Lange T, Haack M. The sleep-immune crosstalk in health and disease. Physiol Rev 2019, 99(3): 1325–1380.
[163]
Vladan B, Panfoli I. Melatonin and abeta, macular degeneration and alzheimers disease: same disease, different outcomes? Med Hypothesis Discov Innov Ophthalmol 2012, 1(2): 24–32.
[164]
Roy J, Tsui KC, Ng J, et al. Regulation of melatonin and neurotransmission in Alzheimer’s disease. Int J Mol Sci 2021, 22(13): 6841.
[165]
Mormino EC, Smiljic A, Hayenga AO, et al. Relationships between β-amyloid and functional connectivity in different components of the default mode network in aging. Cereb Cortex 2011, 21(10): 2399–2407.
[166]
Bastian L, Samanta A, Ribeiro de Paula D, et al. Spindle-slow oscillation coupling correlates with memory performance and connectivity changes in a hippocampal network after sleep. Hum Brain Mapp 2022, 43(13): 3923−3943.
[167]
Tan NYQ, Chew M, Tham YC, et al. Associations between sleep duration, sleep quality and diabetic retinopathy. PLoS One 2018, 13(5): e0196399.
[168]
Wang J, Liu J, Wang Z, et al. Dysfunctional interactions between the default mode network and the dorsal attention network in subtypes of amnestic mild cognitive impairment. Aging: Albany NY 2019, 11(20): 9147–9166.
[169]
Sperling RA, Dickerson BC, Pihlajamaki M, et al. Functional alterations in memory networks in early Alzheimer’s disease. Neuromolecular Med 2010, 12(1): 27–43.
[170]
Beason-Held LL. Dementia and the default mode. Curr Alzheimer Res 2011, 8(4): 361–365.
[171]
Jahn H. Memory loss in Alzheimer’s disease. Dialogues Clin Neurosci 2013, 15(4): 445–454.
[172]
Musiek ES. Circadian rhythms in AD pathogenesis: a critical appraisal. Curr Sleep Med Rep 2017, 3(2): 85–92.
[173]
Esposito E, Cuzzocrea S. Antiinflammatory activity of melatonin in central nervous system. Curr Neuropharmacol 2010, 8(3): 228–242.
[174]
Sulkava S, Muggalla P, Sulkava R, et al. Melatonin receptor type 1A gene linked to Alzheimer’s disease in old age. Sleep 2018, 41(7): zsy103.
[175]
Shukla M, Htoo HH, Wintachai P, et al. Melatonin stimulates the nonamyloidogenic processing of βAPP through the positive transcriptional regulation of ADAM10 and ADAM17. J Pineal Res 2015, 58(2): 151–165.
[176]
Phan TX, Malkani RG. Sleep and circadian rhythm disruption and stress intersect in Alzheimer’s disease. Neurobiol Stress 2019, 10: 100133.
[177]
Chen D, Zhang T, Lee TH. Cellular mechanisms of melatonin: insight from neurodegenerative diseases. Biomolecules 2020, 10(8): E1158.
[178]
Alkazemi D, Rahman A, Habra B. Alterations in glutathione redox homeostasis among adolescents with obesity and Anemia. Sci Rep 2021, 11(1): 3034.
[179]
Ansari MA, Scheff SW. Oxidative stress in the progression of Alzheimer disease in the frontal cortex. J Neuropathol Exp Neurol 2010, 69(2): 155–167.
[180]
Deng WG, Tang ST, Tseng HP, et al. Melatonin suppresses macrophage cyclooxygenase-2 and inducible nitric oxide synthase expression by inhibiting p52 acetylation and binding. Blood 2006, 108(2): 518–524.
[181]
Peter AE, Sandeep BV, Rao BG, et al. Calming the storm: natural immunosuppressants as adjuvants to target the cytokine storm in COVID-19. Front Pharmacol 2020, 11: 583777.
[182]
Cardinali DP, Vigo DE, Olivar N, et al. Melatonin therapy in patients with Alzheimer’s disease. Antioxidants: Basel 2014, 3(2): 245–277.
[183]
Ennis GE, An Y, Resnick SM, et al. Long-term cortisol measures predict Alzheimer disease risk. Neurology 2017, 88(4): 371–378.
[184]
Green KN, Billings LM, Roozendaal B, et al. Glucocorticoids increase amyloid-β and tau pathology in a mouse model of Alzheimer’s disease. J Neurosci 2006, 26(35): 9047–9056.
[185]
Abrahám I, Harkany T, Horvath KM, et al. Chronic corticosterone administration dose-dependently modulates Abeta(1-42)- and NMDA-induced neurodegeneration in rat magnocellular nucleus basalis. J Neuroendocrinol 2000, 12(6): 486–494.
[186]
Budas G, Coughlan CM, Seckl JR, et al. The effect of corticosteroids on amyloid beta precursor protein/amyloid precursor-like protein expression and processing in vivo. Neurosci Lett 1999, 276(1): 61–64.
[187]
McLean CA, Cherny RA, Fraser FW, et al. Soluble pool of Abeta amyloid as a determinant of severity of neurodegeneration in Alzheimer’s disease. Ann Neurol 1999, 46(6): 860–866.
DOI
[188]
Ouanes S, Popp J. High cortisol and the risk of dementia and Alzheimer’s disease: a review of the literature. Front Aging Neurosci 2019, 11: 43.
[189]
El-Farhan N, Rees DA, Evans C. Measuring cortisol in serum, urine and saliva - are our assays good enough? Ann Clin Biochem 2017, 54(3): 308–322.
[190]
Schrijvers EM, Direk N, Koudstaal PJ, et al. Associations of serum cortisol with cognitive function and dementia: the Rotterdam Study. J Alzheimers Dis 2011, 25(4): 671–677.
[191]
Edwards Iii GA, Gamez N, Escobedo G, et al. Modifiable risk factors for Alzheimer’s disease. Front Aging Neurosci 2019, 11: 146.
[192]
Mello AF, Mello MF, Carpenter L L, et al. Update on stress and depression: the role of the hypothalamic-pituitary-adrenal (HPA) axis. Braz J Psychiatry 2003, 25(4), 231–238.
[193]
Coplan JD, Andrews MW, Rosenblum LA, et al. Persistent elevations of cerebrospinal fluid concentrations of corticotropin-releasing factor in adult nonhuman Primates exposed to early-life stressors: implications for the pathophysiology of mood and anxiety disorders. Proc Natl Acad Sci USA 1996, 93(4): 1619–1623.
[194]
Byers AL, Yaffe K. Depression and risk of developing dementia. Nat Rev Neurol 2011, 7(6): 323–331.
[195]
Alloy LB, Ng TH, Titone MK, et al. Circadian rhythm dysregulation in bipolar spectrum disorders. Curr Psychiatry Rep 2017, 19(4): 21.
[196]
Alloy LB, Nusslock R, Boland EM. The development and course of bipolar spectrum disorders: an integrated reward and circadian rhythm dysregulation model. Annu Rev Clin Psychol 2015, 11: 213–250.
[197]
Hasler G. Pathophysiology of depression: do we have any solid evidence of interest to clinicians? World Psychiatry 2010, 9(3): 155–161.
[198]
Slavich GM, Irwin MR. From stress to inflammation and major depressive disorder: a social signal transduction theory of depression. Psychol Bull 2014, 140(3): 774–815.
[199]
van Galen KA, Ter Horst KW, Serlie MJ. Serotonin, food intake, and obesity. Obesity Rev 2021, 22(7): e13210.
[200]
Alexander R, Aragón OR, Bookwala J, et al. The neuroscience of positive emotions and affect: Implications for cultivating happiness and wellbeing. Neurosci Biobehav Rev 2021, 121: 220–249.
[201]
Cribbet MR, Logan RW, Edwards MD, et al. Circadian rhythms and metabolism: from the brain to the gut and back again. Ann N Y Acad Sci 2016, 1385(1): 21–40.
[202]
Besedovsky L, Lange T, Born J. Sleep and immune function. Pflugers Arch 2012, 463(1): 121–137.
[203]
Jessen NA, Munk AS, Lundgaard I, et al. The glymphatic system: a beginner’s guide. Neurochem Res 2015, 40(12): 2583–2599.
[204]
Serrano-Pozo A, Frosch MP, Masliah E, et al. Neuropathological alterations in Alzheimer disease. Cold Spring Harb Perspect Med 2011, 1(1): a006189.
[205]
Heneka MT, Carson MJ, El Khoury J, et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol 2015, 14(4): 388–405.
[206]
Sharkey KM, Eastman CI. Melatonin phase shifts human circadian rhythms in a placebo-controlled simulated night-work study. Am J Physiol Regul Integr Comp Physiol 2002, 282(2): R454–R463.
[207]
Cheikh M, Hammouda O, Gaamouri N, et al. Melatonin ingestion after exhaustive late-evening exercise improves sleep quality and quantity, and short-term performances in teenage Athletes. Chronobiol Int 2018, 35(9): 1281–1293.
[208]
Zakharov A, Khivintseva E. Clinical use of melatonin in the treatment of sleep disorders. In Melatonin – The Hormone of Darkness and it’s Therapeutic Potential and Perspectives. Vlachou M, Ed. London: IntechOpen, 2020.
[209]
Peck JS, LeGoff DB, Ahmed I, et al. Cognitive effects of exogenous melatonin administration in elderly persons. Am J Geriatr Psychiatry 2004, 12(4): 432–436.
[210]
Sharma A, Sethi G, Tambuwala MM, et al. Circadian rhythm disruption and Alzheimer’s disease: the dynamics of a vicious cycle. Curr Neuropharmacol 2021, 19(2): 248–264.
[211]
Petrie K, Dawson AG, Thompson L, et al. A double-blind trial of melatonin as a treatment for jet lag in international cabin crew. Biol Psychiatry 1993, 33(7): 526–530.
[212]
Cardinali DP, Vigo DE, Olivar N, et al. Therapeutic application of melatonin in mild cognitive impairment. Am J Neurodegener Dis 2012, 1(3): 280–291.
[213]
Morgan PT, Kehne JH, Sprenger KJ, et al. Retrograde effects of triazolam and zolpidem on sleep-dependent motor learning in humans. J Sleep Res 2010, 19(1 pt 2): 157–164.
[214]
Paul MA, Gray G, Lieberman H, et al. Management of circadian desynchrony (jetlag and shiftlag) in CF air operations. Technical Report 2010-002, Defence R&D Centre—Toronto, 2010.
[215]
Dodson ER, Zee PC. Therapeutics for circadian rhythm sleep disorders. Sleep Med Clin 2010, 5(4): 701–715.
[216]
Silva S, Bicker J, Falcão A, et al. Antidepressants and circadian rhythm: exploring their bidirectional interaction for the treatment of depression. Pharmaceutics 2021, 13(11): 1975.
[217]
Everitt H, Baldwin DS, Stuart B, et al. Antidepressants for insomnia in adults. Cochrane Database Syst Rev 2018, 5: CD010753.
[218]
Brendel M, Sauerbeck J, Greven S, et al. Serotonin selective reuptake inhibitor treatment improves cognition and grey matter atrophy but not amyloid burden during two-year follow-up in mild cognitive impairment and Alzheimer’s disease patients with depressive symptoms. J Alzheimers Dis 2018, 65(3): 793–806.
[219]
Kelley BJ, Petersen RC. Alzheimer’s disease and mild cognitive impairment. Neurol Clin 2007, 25(3): 577–609.
[220]
Yiannopoulou KG, Papageorgiou SG. Current and future treatments for Alzheimer’s disease. Ther Adv Neurol Disord 2013, 6(1): 19–33.
[221]
Ancoli-Israel S, Martin JL, Kripke DF, et al. Effect of light treatment on sleep and circadian rhythms in demented nursing home patients. J Am Geriatr Soc 2002, 50(2): 282–289.
[222]
Slama H, Deliens G, Schmitz R, et al. Afternoon nap and bright light exposure improve cognitive flexibility post lunch. PLoS One 2015, 10(5): e0125359.
Publication history
Copyright
Rights and permissions

Publication history

Received: 04 June 2022
Revised: 18 July 2022
Accepted: 31 August 2022
Published: 30 November 2022
Issue date: December 2022

Copyright

© The authors 2022.

Rights and permissions

This article is published with open access at journals.sagepub.com/home/BSA

Creative Commons Non Commercial CC BY-NC: This article is distributed under the terms of the Creative Commons Attribution-NonCommercial 4.0 License (http://www.creativecommons.org/licenses/by-nc/4.0/) which permits non-commercial use, reproduction and distribution of the work without further permission provided the original work is attributed as specified on the SAGE and Open Access pages (https://us.sagepub.com/en-us/nam/open-access-at-sage).

Return