β-Amyloid accumulation in the human brain after one night of sleep deprivation

Ehsan Shokri-Kojori, Gene-Jack Wang, Corinde E Wiers, Sukru B Demiral, Min Guo, Sung Won Kim, Elsa Lindgren, Veronica Ramirez, Amna Zehra, Clara Freeman, Gregg Miller, Peter Manza, Tansha Srivastava, Susan De Santi, Dardo Tomasi, Helene Benveniste, Nora D Volkow, Ehsan Shokri-Kojori, Gene-Jack Wang, Corinde E Wiers, Sukru B Demiral, Min Guo, Sung Won Kim, Elsa Lindgren, Veronica Ramirez, Amna Zehra, Clara Freeman, Gregg Miller, Peter Manza, Tansha Srivastava, Susan De Santi, Dardo Tomasi, Helene Benveniste, Nora D Volkow

Abstract

The effects of acute sleep deprivation on β-amyloid (Aβ) clearance in the human brain have not been documented. Here we used PET and 18F-florbetaben to measure brain Aβ burden (ABB) in 20 healthy controls tested after a night of rested sleep (baseline) and after a night of sleep deprivation. We show that one night of sleep deprivation, relative to baseline, resulted in a significant increase in Aβ burden in the right hippocampus and thalamus. These increases were associated with mood worsening following sleep deprivation, but were not related to the genetic risk (APOE genotype) for Alzheimer's disease. Additionally, baseline ABB in a range of subcortical regions and the precuneus was inversely associated with reported night sleep hours. APOE genotyping was also linked to subcortical ABB, suggesting that different Alzheimer's disease risk factors might independently affect ABB in nearby brain regions. In summary, our findings show adverse effects of one-night sleep deprivation on brain ABB and expand on prior findings of higher Aβ accumulation with chronic less sleep.

Keywords: Alzheimer’s disease; beta amyloid; glymphatic system; hippocampus; sleep.

Conflict of interest statement

Conflict of interest statement: S.D.S. was an employee of Piramal Pharma Inc., which partly supported the radiotracer for this study.

Copyright © 2018 the Author(s). Published by PNAS.

Figures

Fig. 1.
Fig. 1.
Effects of one-night SD on ABB. (A) Voxelwise paired t test between RW and SD conditions highlighting the hippocampus as well as other subcortical structures (PFWE < 0.05, cluster-size corrected) (Table S1). (B) Subject-level changes in FBB SUVr (in the red cluster identified in A) from RW to SD. There was no significant effect of gender, or gender × sleep interaction (P > 0.15). (C) Association between changes in mood from RW to SD and changes in the FBB SUVr for the cluster identified in A. Mood change was quantified using the principal component of the changes in self-report measures from RW to SD, which accounted for 35.5% of the variance. Self-report measures of alert, friendly, happy, social, and energetic significantly decreased, and measures of tired and difficulty staying awake significantly increased from RW to SD (P < 0.001, two-tailed) (see also Fig. S1). (D) Average FBB SUVr in a priori hippocampus ROIs across subjects. Error bars show standard deviation (Methods).
Fig. 2.
Fig. 2.
Relationship between SH, APOE genotype, and ABB. (A) Regression of FBB SUVr (indexing ABB) for the red cluster shown in Fig. 1A at RW against SH. (B) Three-dimensional rendering of the areas showing an association between higher FBB SUVr at RW and lower SH (red clusters, PFWE < 0.05, cluster-size corrected) (Table S3), as well as areas showing an association between higher FBB SUVr and higher APOE-based genetic risk for AD (quantified as log of ORAD) (green clusters, PFWE < 0.05, cluster-size corrected, except the right-sided subcortical cluster, which was cluster size-corrected at qFDR < 0.05) (Table S4). The subcortical clusters related to SH and ORAD had minimal spatial overlap (18 voxels, <3%) (Tables S3 and S4). Notably, FBB SUVr within the SH-related subcortical clusters (red, B) was not associated with ORAD (P = 0.25) and FBB SUVr within the ORAD-related subcortical clusters (green, B) was not associated with SH (P = 0.2).

References

    1. Nedergaard M. Neuroscience. Garbage truck of the brain. Science. 2013;340:1529–1530.
    1. Mestre H, Kostrikov S, Mehta RI, Nedergaard M. Perivascular spaces, glymphatic dysfunction, and small vessel disease. Clin Sci (Lond) 2017;131:2257–2274.
    1. Xie L, et al. Sleep drives metabolite clearance from the adult brain. Science. 2013;342:373–377.
    1. Kang J-E, et al. Amyloid-β dynamics are regulated by orexin and the sleep-wake cycle. Science. 2009;326:1005–1007.
    1. Tabuchi M, et al. Sleep interacts with aβ to modulate intrinsic neuronal excitability. Curr Biol. 2015;25:702–712.
    1. Spira AP, et al. Self-reported sleep and β-amyloid deposition in community-dwelling older adults. JAMA Neurol. 2013;70:1537–1543.
    1. Brown BM, et al. AIBL Research Group The relationship between sleep quality and brain amyloid burden. Sleep (Basel) 2016;39:1063–1068.
    1. Sprecher KE, et al. Amyloid burden is associated with self-reported sleep in nondemented late middle-aged adults. Neurobiol Aging. 2015;36:2568–2576.
    1. Ju Y-ES, et al. Sleep quality and preclinical Alzheimer disease. JAMA Neurol. 2013;70:587–593.
    1. Ju Y-ES, Lucey BP, Holtzman DM. Sleep and Alzheimer disease pathology—A bidirectional relationship. Nat Rev Neurol. 2014;10:115–119.
    1. Mucke L, Selkoe DJ. Neurotoxicity of amyloid β-protein: Synaptic and network dysfunction. Cold Spring Harb Perspect Med. 2012;2:a006338.
    1. Jagust W. Is amyloid-β harmful to the brain? Insights from human imaging studies. Brain. 2016;139:23–30.
    1. Spira AP, Gottesman RF. Sleep disturbance: An emerging opportunity for Alzheimer’s disease prevention? Int Psychogeriatr. 2017;29:529–531.
    1. Lee H, et al. The effect of body posture on brain glymphatic transport. J Neurosci. 2015;35:11034–11044.
    1. Iliff JJ, et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid β. Sci Transl Med. 2012;4:147ra111.
    1. Ooms S, et al. Effect of 1 night of total sleep deprivation on cerebrospinal fluid β-amyloid 42 in healthy middle-aged men: A randomized clinical trial. JAMA Neurol. 2014;71:971–977.
    1. LaFerla FM, Green KN. Animal models of Alzheimer disease. Cold Spring Harb Perspect Med. 2012;2:a006320.
    1. Bu XL, et al. Blood-derived amyloid-β protein induces Alzheimer’s disease pathologies. Mol Psychiatry. October 31, 2017 doi: 10.1038/mp.2017.204.
    1. Deane R, Zlokovic BV. Role of the blood-brain barrier in the pathogenesis of Alzheimer’s disease. Curr Alzheimer Res. 2007;4:191–197.
    1. Wei M, et al. Sleep deprivation induced plasma amyloid-β transport disturbance in healthy young adults. J Alzheimers Dis. 2017;57:899–906.
    1. Lucey BP, et al. Effect of sleep on overnight CSF amyloid-β kinetics. Ann Neurol. December 8, 2017 doi: 10.1002/ana.25117.
    1. Klunk WE, et al. Imaging brain amyloid in Alzheimer’s disease with Pittsburgh compound-B. Ann Neurol. 2004;55:306–319.
    1. Rowe CC, et al. Imaging of amyloid β in Alzheimer’s disease with 18F-BAY94-9172, a novel PET tracer: Proof of mechanism. Lancet Neurol. 2008;7:129–135.
    1. Fodero-Tavoletti MT, et al. In vitro characterization of [18F]-florbetaben, an Aβ imaging radiotracer. Nucl Med Biol. 2012;39:1042–1048.
    1. Villemagne VL, et al. Amyloid imaging with (18)F-florbetaben in Alzheimer disease and other dementias. J Nucl Med. 2011;52:1210–1217.
    1. Sabri O, Seibyl J, Rowe C, Barthel H. Beta-amyloid imaging with florbetaben. Clin Transl Imaging. 2015;3:13–26.
    1. Ni R, Gillberg P-G, Bergfors A, Marutle A, Nordberg A. Amyloid tracers detect multiple binding sites in Alzheimer’s disease brain tissue. Brain. 2013;136:2217–2227.
    1. Yamin G, Teplow DB. Pittsburgh compound-B (PiB) binds amyloid β-protein protofibrils. J Neurochem. 2017;140:210–215.
    1. Schuff N, et al. Alzheimer’s Disease Neuroimaging Initiative MRI of hippocampal volume loss in early Alzheimer’s disease in relation to ApoE genotype and biomarkers. Brain. 2009;132:1067–1077.
    1. De Santi S, et al. Hippocampal formation glucose metabolism and volume losses in MCI and AD. Neurobiol Aging. 2001;22:529–539.
    1. Mander BA, Winer JR, Jagust WJ, Walker MP. Sleep: A novel mechanistic pathway, biomarker, and treatment target in the pathology of Alzheimer’s disease? Trends Neurosci. 2016;39:552–566.
    1. Ju YS, et al. Slow wave sleep disruption increases cerebrospinal fluid amyloid-β levels. Brain. 2017;140:2104–2111.
    1. Thal DR, Rüb U, Orantes M, Braak H. Phases of A β-deposition in the human brain and its relevance for the development of AD. Neurology. 2002;58:1791–1800.
    1. Aron L, Yankner BA. Neurodegenerative disorders: Neural synchronization in Alzheimer’s disease. Nature. 2016;540:207–208.
    1. Bero AW, et al. Neuronal activity regulates the regional vulnerability to amyloid-β deposition. Nat Neurosci. 2011;14:750–756.
    1. Cheng O, et al. Short-term sleep deprivation stimulates hippocampal neurogenesis in rats following global cerebral ischemia/reperfusion. PLoS One. 2015;10:e0125877.
    1. Fernandes C, et al. Detrimental role of prolonged sleep deprivation on adult neurogenesis. Front Cell Neurosci. 2015;9:140.
    1. Castellano JM, et al. Human apoE isoforms differentially regulate brain amyloid-β peptide clearance. Sci Transl Med. 2011;3:89ra57.
    1. Jack CR, Jr, et al. Alzheimer’s Disease Neuroimaging Initiative Brain beta-amyloid measures and magnetic resonance imaging atrophy both predict time-to-progression from mild cognitive impairment to Alzheimer’s disease. Brain. 2010;133:3336–3348.
    1. Vandenberghe R, et al. 18F-flutemetamol amyloid imaging in Alzheimer disease and mild cognitive impairment: A phase 2 trial. Ann Neurol. 2010;68:319–329.
    1. Meyer-Luehmann M, et al. Rapid appearance and local toxicity of amyloid-β plaques in a mouse model of Alzheimer’s disease. Nature. 2008;451:720–724.
    1. Condello C, Schain A, Grutzendler J. Multicolor time-stamp reveals the dynamics and toxicity of amyloid deposition. Sci Rep. 2011;1:19.
    1. Rothman SM, Herdener N, Frankola KA, Mughal MR, Mattson MP. Chronic mild sleep restriction accentuates contextual memory impairments, and accumulations of cortical Aβ and pTau in a mouse model of Alzheimer’s disease. Brain Res. 2013;1529:200–208.
    1. Zhao HY, et al. Chronic sleep restriction induces cognitive deficits and cortical beta-amyloid deposition in mice via BACE1-antisense activation. CNS Neurosci Ther. 2017;23:233–240.
    1. Camus V, et al. Using PET with 18F-AV-45 (florbetapir) to quantify brain amyloid load in a clinical environment. Eur J Nucl Med Mol Imaging. 2012;39:621–631.
    1. Montagne A, et al. Blood-brain barrier breakdown in the aging human hippocampus. Neuron. 2015;85:296–302.
    1. Cai Z, Hussain MD, Yan L-J. Microglia, neuroinflammation, and beta-amyloid protein in Alzheimer’s disease. Int J Neurosci. 2014;124:307–321.
    1. Zhu B, et al. Sleep disturbance induces neuroinflammation and impairment of learning and memory. Neurobiol Dis. 2012;48:348–355.
    1. Sehlin D, et al. Antibody-based PET imaging of amyloid beta in mouse models of Alzheimer’s disease. Nat Commun. 2016;7:10759.
    1. Wu JC, et al. Frontal lobe metabolic decreases with sleep deprivation not totally reversed by recovery sleep. Neuropsychopharmacology. 2006;31:2783–2792.
    1. Ma N, Dinges DF, Basner M, Rao H. How acute total sleep loss affects the attending brain: A meta-analysis of neuroimaging studies. Sleep (Basel) 2015;38:233–240.
    1. Bullich S, et al. Validation of non-invasive tracer kinetic analysis of 18F-florbetaben PET using a dual time-window acquisition protocol. J Nucl Med. 2017:jnumed.117.200964.
    1. Price JL, Drevets WC. Neurocircuitry of mood disorders. Neuropsychopharmacology. 2010;35:192–216.
    1. Mueller AD, Meerlo P, McGinty D, Mistlberger RE. Sleep and adult neurogenesis: Implications for cognition and mood. In: Meerlo P, Benca RM, Abel T, editors. Sleep, Neuronal Plasticity and Brain Function. Springer; Heidelberg, Germany: 2013. pp. 151–181.
    1. Dissel S, et al. Enhanced sleep reverses memory deficits and underlying pathology in Drosophila models of Alzheimer’s disease. Neurobiol Sleep Circadian Rhythms. 2017;2:15–26.
    1. Glasser MF, et al. WU-Minn HCP Consortium The minimal preprocessing pipelines for the Human Connectome Project. Neuroimage. 2013;80:105–124.
    1. Catafau AM, et al. Cerebellar amyloid-β plaques: How frequent are they, and do they influence 18F-florbetaben SUV ratios? J Nucl Med. 2016;57:1740–1745.
    1. Bullich S, et al. Optimal reference region to measure longitudinal amyloid-beta change with 18F-florbetaben PET. J Nucl Med. 2017:jnumed.116.187351.
    1. Heurling K, Buckley C, Van Laere K, Vandenberghe R, Lubberink M. Parametric imaging and quantitative analysis of the PET amyloid ligand [(18)F]flutemetamol. Neuroimage. 2015;121:184–192.
    1. Ossenkoppele R, Prins ND, van Berckel BN. Amyloid imaging in clinical trials. Alzheimers Res Ther. 2013;5:36.
    1. Gunn RN, Lammertsma AA, Hume SP, Cunningham VJ. Parametric imaging of ligand-receptor binding in PET using a simplified reference region model. Neuroimage. 1997;6:279–287.
    1. Lammertsma AA, Hume SP. Simplified reference tissue model for PET receptor studies. Neuroimage. 1996;4:153–158.
    1. Logan J, et al. Distribution volume ratios without blood sampling from graphical analysis of PET data. J Cereb Blood Flow Metab. 1996;16:834–840.
    1. Buysse DJ, Reynolds CF, 3rd, Monk TH, Berman SR, Kupfer DJ. The Pittsburgh Sleep Quality Index: A new instrument for psychiatric practice and research. Psychiatry Res. 1989;28:193–213.
    1. Choe YM, et al. Sleep quality in young and middle age-period is associated with cerebral amyloid burden in cognitively normal elderly people. Alzheimers Dement. 2016;12(Suppl):P171–P172.
    1. Westwood AJ, et al. Prolonged sleep duration as a marker of early neurodegeneration predicting incident dementia. Neurology. 2017;88:1172–1179.
    1. Liu C-C, Kanekiyo T, Xu H, Bu G. Apolipoprotein E and Alzheimer disease: Risk, mechanisms and therapy. Nat Rev Neurol. 2013;9:106–118, and erratum (2013), 10.1038/nmeurol.2013.32.
    1. Reiman EM, et al. Correlations between apolipoprotein E ε4 gene dose and brain-imaging measurements of regional hypometabolism. Proc Natl Acad Sci USA. 2005;102:8299–8302.
    1. Corneveaux JJ, et al. Association of CR1, CLU and PICALM with Alzheimer’s disease in a cohort of clinically characterized and neuropathologically verified individuals. Hum Mol Genet. 2010;19:3295–3301.
    1. Friston KJ, et al. Analysis of fMRI time-series revisited. Neuroimage. 1995;2:45–53.
    1. Poline J-B, Worsley KJ, Evans AC, Friston KJ. Combining spatial extent and peak intensity to test for activations in functional imaging. Neuroimage. 1997;5:83–96.

Source: PubMed

3
Subscribe