Skip to main content
Top
Published in: Alzheimer's Research & Therapy 1/2023

Open Access 01-12-2023 | Alzheimer's Disease | Research

Altered dynamics of glymphatic flow in a mature-onset Tet-off APP mouse model of amyloidosis

Authors: Inès R. H. Ben-Nejma, Aneta J. Keliris, Verdi Vanreusel, Peter Ponsaerts, Annemie Van der Linden, Georgios A. Keliris

Published in: Alzheimer's Research & Therapy | Issue 1/2023

Login to get access

Abstract

Background

Alzheimer’s disease (AD) is an incurable neurodegenerative disorder characterised by the progressive buildup of toxic amyloid-beta (Aβ) and tau protein aggregates eventually leading to cognitive decline. Recent lines of evidence suggest that an impairment of the glymphatic system (GS), a brain waste clearance pathway, plays a key role in the pathology of AD. Moreover, a relationship between GS function and neuronal network integrity has been strongly implicated. Here, we sought to assess the efficacy of the GS in a transgenic Tet-Off APP mouse model of amyloidosis, in which the expression of mutant APP was delayed until maturity, mimicking features of late-onset AD—the most common form of dementia in humans.

Methods

To evaluate GS function, we used dynamic contrast-enhanced MRI (DCE-MRI) in 14-month-old Tet-Off APP (AD) mice and aged-matched littermate controls. Brain-wide transport of the Gd-DOTA contrast agent was monitored over time after cisterna magna injection. Region-of-interest analysis and computational modelling were used to assess GS dynamics while characterisation of brain tissue abnormalities at the microscale was performed ex vivo by immunohistochemistry.

Results

We observed reduced rostral glymphatic flow and higher accumulation of the contrast agent in areas proximal to the injection side in the AD group. Clustering and subsequent computational modelling of voxel time courses revealed significantly lower influx time constants in AD relative to the controls. Ex vivo evaluation showed abundant amyloid plaque burden in the AD group coinciding with extensive astrogliosis and microgliosis. The neuroinflammatory responses were also found in plaque-devoid regions, potentially impacting brain-fluid circulation.

Conclusions

In a context resembling late-onset AD in humans, we demonstrate the disruption of glymphatic function and particularly a reduction in brain-fluid influx in the AD group. We conjecture that the hindered circulation of cerebrospinal fluid is potentially caused by wide-spread astrogliosis and amyloid-related obstruction of the normal routes of glymphatic flow resulting in redirection towards caudal regions. In sum, our study highlights the translational potential of alternative approaches, such as targeting brain-fluid circulation as potential therapeutic strategies for AD.
Appendix
Available only for authorised users
Literature
1.
go back to reference Dubois B, et al. Preclinical Alzheimer’s disease: definition, natural history, and diagnostic criteria. Alzheimers Dement. 2016;12(3):292–323.CrossRef Dubois B, et al. Preclinical Alzheimer’s disease: definition, natural history, and diagnostic criteria. Alzheimers Dement. 2016;12(3):292–323.CrossRef
2.
go back to reference Long JM, Holtzman DM. Alzheimer disease: an update on pathobiology and treatment strategies. Cell. 2019;179(2):312–39.CrossRef Long JM, Holtzman DM. Alzheimer disease: an update on pathobiology and treatment strategies. Cell. 2019;179(2):312–39.CrossRef
3.
go back to reference Querfurth HW, LaFerla FM. Alzheimer’s disease. N Engl J Med. 2010;362(4):329–44.CrossRef Querfurth HW, LaFerla FM. Alzheimer’s disease. N Engl J Med. 2010;362(4):329–44.CrossRef
4.
go back to reference Zlokovic BV. Neurovascular pathways to neurodegeneration in Alzheimer’s disease and other disorders. Nat Rev Neurosci. 2011;12(12):723–38.CrossRef Zlokovic BV. Neurovascular pathways to neurodegeneration in Alzheimer’s disease and other disorders. Nat Rev Neurosci. 2011;12(12):723–38.CrossRef
5.
go back to reference Soto-Rojas LO, et al. The neurovascular unit dysfunction in Alzheimer’s disease. Int J Mol Sci. 2021;22(4):2022. Soto-Rojas LO, et al. The neurovascular unit dysfunction in Alzheimer’s disease. Int J Mol Sci. 2021;22(4):2022.
6.
go back to reference Zlokovic BV, et al. Clearance of amyloid beta-peptide from brain: transport or metabolism? Nat Med. 2000;6(7):718–9.CrossRef Zlokovic BV, et al. Clearance of amyloid beta-peptide from brain: transport or metabolism? Nat Med. 2000;6(7):718–9.CrossRef
7.
go back to reference Weller RO, et al. Perivascular drainage of amyloid-beta peptides from the brain and its failure in cerebral amyloid angiopathy and Alzheimer’s disease. Brain Pathol. 2008;18(2):253–66.CrossRef Weller RO, et al. Perivascular drainage of amyloid-beta peptides from the brain and its failure in cerebral amyloid angiopathy and Alzheimer’s disease. Brain Pathol. 2008;18(2):253–66.CrossRef
8.
go back to reference Iliff JJ, et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid beta. Sci Transl Med. 2012;4(147):147ra111. Iliff JJ, et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid beta. Sci Transl Med. 2012;4(147):147ra111.
9.
go back to reference Aspelund A, et al. A dural lymphatic vascular system that drains brain interstitial fluid and macromolecules. J Exp Med. 2015;212(7):991–9.CrossRef Aspelund A, et al. A dural lymphatic vascular system that drains brain interstitial fluid and macromolecules. J Exp Med. 2015;212(7):991–9.CrossRef
10.
go back to reference Tarasoff-Conway JM, et al. Clearance systems in the brain-implications for Alzheimer disease. Nat Rev Neurol. 2015;11(8):457–70.CrossRef Tarasoff-Conway JM, et al. Clearance systems in the brain-implications for Alzheimer disease. Nat Rev Neurol. 2015;11(8):457–70.CrossRef
11.
go back to reference Louveau A, et al. Understanding the functions and relationships of the glymphatic system and meningeal lymphatics. J Clin Invest. 2017;127(9):3210–9.CrossRef Louveau A, et al. Understanding the functions and relationships of the glymphatic system and meningeal lymphatics. J Clin Invest. 2017;127(9):3210–9.CrossRef
12.
go back to reference Hladky SB, Barrand MA. Elimination of substances from the brain parenchyma: efflux via perivascular pathways and via the blood-brain barrier. Fluids Barriers CNS. 2018;15(1):30.CrossRef Hladky SB, Barrand MA. Elimination of substances from the brain parenchyma: efflux via perivascular pathways and via the blood-brain barrier. Fluids Barriers CNS. 2018;15(1):30.CrossRef
13.
go back to reference Mestre H, Mori Y, Nedergaard M. The brain’s glymphatic system: current controversies. Trends Neurosci. 2020;43(7):458–66.CrossRef Mestre H, Mori Y, Nedergaard M. The brain’s glymphatic system: current controversies. Trends Neurosci. 2020;43(7):458–66.CrossRef
14.
go back to reference Xie L, et al. Sleep drives metabolite clearance from the adult brain. Science. 2013;342(6156):373–7.CrossRef Xie L, et al. Sleep drives metabolite clearance from the adult brain. Science. 2013;342(6156):373–7.CrossRef
15.
go back to reference Peng W, et al. Suppression of glymphatic fluid transport in a mouse model of Alzheimer’s disease. Neurobiol Dis. 2016;93:215–25.CrossRef Peng W, et al. Suppression of glymphatic fluid transport in a mouse model of Alzheimer’s disease. Neurobiol Dis. 2016;93:215–25.CrossRef
16.
go back to reference Iliff JJ, et al. Impairment of glymphatic pathway function promotes tau pathology after traumatic brain injury. J Neurosci. 2014;34(49):16180–93.CrossRef Iliff JJ, et al. Impairment of glymphatic pathway function promotes tau pathology after traumatic brain injury. J Neurosci. 2014;34(49):16180–93.CrossRef
17.
go back to reference Harrison IF, et al. Impaired glymphatic function and clearance of tau in an Alzheimer’s disease model. Brain. 2020;143(8):2576–93.CrossRef Harrison IF, et al. Impaired glymphatic function and clearance of tau in an Alzheimer’s disease model. Brain. 2020;143(8):2576–93.CrossRef
18.
go back to reference Iliff JJ, et al. Brain-wide pathway for waste clearance captured by contrast-enhanced MRI. J Clin Invest. 2013;123(3):1299–309.CrossRef Iliff JJ, et al. Brain-wide pathway for waste clearance captured by contrast-enhanced MRI. J Clin Invest. 2013;123(3):1299–309.CrossRef
19.
go back to reference Jessen NA, et al. The glymphatic system: a beginner’s guide. Neurochem Res. 2015;40(12):2583–99.CrossRef Jessen NA, et al. The glymphatic system: a beginner’s guide. Neurochem Res. 2015;40(12):2583–99.CrossRef
20.
go back to reference Iliff JJ, et al. Cerebral arterial pulsation drives paravascular CSF-interstitial fluid exchange in the murine brain. J Neurosci. 2013;33(46):18190–9.CrossRef Iliff JJ, et al. Cerebral arterial pulsation drives paravascular CSF-interstitial fluid exchange in the murine brain. J Neurosci. 2013;33(46):18190–9.CrossRef
21.
go back to reference Mestre H, et al. Flow of cerebrospinal fluid is driven by arterial pulsations and is reduced in hypertension. Nat Commun. 2018;9(1):4878.CrossRef Mestre H, et al. Flow of cerebrospinal fluid is driven by arterial pulsations and is reduced in hypertension. Nat Commun. 2018;9(1):4878.CrossRef
22.
go back to reference Kress BT, et al. Impairment of paravascular clearance pathways in the aging brain. Ann Neurol. 2014;76(6):845–61.CrossRef Kress BT, et al. Impairment of paravascular clearance pathways in the aging brain. Ann Neurol. 2014;76(6):845–61.CrossRef
23.
go back to reference Jankowsky JL, Zheng H. Practical considerations for choosing a mouse model of Alzheimer’s disease. Mol Neurodegener. 2017;12(1):89.CrossRef Jankowsky JL, Zheng H. Practical considerations for choosing a mouse model of Alzheimer’s disease. Mol Neurodegener. 2017;12(1):89.CrossRef
24.
go back to reference Radde R, et al. Abeta42-driven cerebral amyloidosis in transgenic mice reveals early and robust pathology. EMBO Rep. 2006;7(9):940–6.CrossRef Radde R, et al. Abeta42-driven cerebral amyloidosis in transgenic mice reveals early and robust pathology. EMBO Rep. 2006;7(9):940–6.CrossRef
25.
go back to reference Oblak AL, et al. Comprehensive evaluation of the 5XFAD mouse model for preclinical testing applications: a MODEL-AD study. Front Aging Neurosci. 2021;13: 713726.CrossRef Oblak AL, et al. Comprehensive evaluation of the 5XFAD mouse model for preclinical testing applications: a MODEL-AD study. Front Aging Neurosci. 2021;13: 713726.CrossRef
26.
go back to reference Xu Z, et al. Deletion of aquaporin-4 in APP/PS1 mice exacerbates brain Abeta accumulation and memory deficits. Mol Neurodegener. 2015;10:58.CrossRef Xu Z, et al. Deletion of aquaporin-4 in APP/PS1 mice exacerbates brain Abeta accumulation and memory deficits. Mol Neurodegener. 2015;10:58.CrossRef
27.
go back to reference Feng W, et al. Microglia prevent beta-amyloid plaque formation in the early stage of an Alzheimer’s disease mouse model with suppression of glymphatic clearance. Alzheimers Res Ther. 2020;12(1):125.CrossRef Feng W, et al. Microglia prevent beta-amyloid plaque formation in the early stage of an Alzheimer’s disease mouse model with suppression of glymphatic clearance. Alzheimers Res Ther. 2020;12(1):125.CrossRef
28.
go back to reference Zeppenfeld DM, et al. Association of perivascular localization of aquaporin-4 with cognition and Alzheimer disease in aging brains. JAMA Neurol. 2017;74(1):91–9.CrossRef Zeppenfeld DM, et al. Association of perivascular localization of aquaporin-4 with cognition and Alzheimer disease in aging brains. JAMA Neurol. 2017;74(1):91–9.CrossRef
29.
go back to reference Yang J, et al. Loss of astrocyte polarization in the tg-ArcSwe mouse model of Alzheimer’s disease. J Alzheimers Dis. 2011;27(4):711–22.CrossRef Yang J, et al. Loss of astrocyte polarization in the tg-ArcSwe mouse model of Alzheimer’s disease. J Alzheimers Dis. 2011;27(4):711–22.CrossRef
30.
go back to reference Kiviniemi V, et al. Ultra-fast magnetic resonance encephalography of physiological brain activity - glymphatic pulsation mechanisms? J Cereb Blood Flow Metab. 2016;36(6):1033–45.CrossRef Kiviniemi V, et al. Ultra-fast magnetic resonance encephalography of physiological brain activity - glymphatic pulsation mechanisms? J Cereb Blood Flow Metab. 2016;36(6):1033–45.CrossRef
31.
go back to reference Fultz NE, et al. Coupled electrophysiological, hemodynamic, and cerebrospinal fluid oscillations in human sleep. Science. 2019;366(6465):628–31.CrossRef Fultz NE, et al. Coupled electrophysiological, hemodynamic, and cerebrospinal fluid oscillations in human sleep. Science. 2019;366(6465):628–31.CrossRef
32.
go back to reference Han F, et al. Reduced coupling between cerebrospinal fluid flow and global brain activity is linked to Alzheimer disease-related pathology. PLoS Biol. 2021;19(6): e3001233.CrossRef Han F, et al. Reduced coupling between cerebrospinal fluid flow and global brain activity is linked to Alzheimer disease-related pathology. PLoS Biol. 2021;19(6): e3001233.CrossRef
33.
go back to reference Lloret MA, et al. Is sleep disruption a cause or consequence of Alzheimer’s disease? Reviewing its possible role as a biomarker. Int J Mol Sci. 2020;21(3):1168. Lloret MA, et al. Is sleep disruption a cause or consequence of Alzheimer’s disease? Reviewing its possible role as a biomarker. Int J Mol Sci. 2020;21(3):1168.
34.
go back to reference Mander BA, Winer JR, Walker MP. Sleep and human aging. Neuron. 2017;94(1):19–36.CrossRef Mander BA, Winer JR, Walker MP. Sleep and human aging. Neuron. 2017;94(1):19–36.CrossRef
35.
go back to reference Badhwar A, et al. Resting-state network dysfunction in Alzheimer’s disease: a systematic review and meta-analysis. Alzheimers Dement (Amst). 2017;8:73–85.CrossRef Badhwar A, et al. Resting-state network dysfunction in Alzheimer’s disease: a systematic review and meta-analysis. Alzheimers Dement (Amst). 2017;8:73–85.CrossRef
36.
go back to reference Greicius MD, et al. Default-mode network activity distinguishes Alzheimer’s disease from healthy aging: evidence from functional MRI. Proc Natl Acad Sci U S A. 2004;101(13):4637–42.CrossRef Greicius MD, et al. Default-mode network activity distinguishes Alzheimer’s disease from healthy aging: evidence from functional MRI. Proc Natl Acad Sci U S A. 2004;101(13):4637–42.CrossRef
37.
go back to reference Grandjean J, et al. Complex interplay between brain function and structure during cerebral amyloidosis in APP transgenic mouse strains revealed by multi-parametric MRI comparison. Neuroimage. 2016;134:1–11.CrossRef Grandjean J, et al. Complex interplay between brain function and structure during cerebral amyloidosis in APP transgenic mouse strains revealed by multi-parametric MRI comparison. Neuroimage. 2016;134:1–11.CrossRef
38.
go back to reference Shah D, et al. Resting state FMRI reveals diminished functional connectivity in a mouse model of amyloidosis. PLoS ONE. 2013;8(12): e84241.CrossRef Shah D, et al. Resting state FMRI reveals diminished functional connectivity in a mouse model of amyloidosis. PLoS ONE. 2013;8(12): e84241.CrossRef
39.
go back to reference Ben-Nejma IRH, et al. Increased soluble amyloid-beta causes early aberrant brain network hypersynchronisation in a mature-onset mouse model of amyloidosis. Acta Neuropathol Commun. 2019;7(1):180.CrossRef Ben-Nejma IRH, et al. Increased soluble amyloid-beta causes early aberrant brain network hypersynchronisation in a mature-onset mouse model of amyloidosis. Acta Neuropathol Commun. 2019;7(1):180.CrossRef
40.
go back to reference Rodgers SP, et al. Transgenic APP expression during postnatal development causes persistent locomotor hyperactivity in the adult. Mol Neurodegener. 2012;7:28.CrossRef Rodgers SP, et al. Transgenic APP expression during postnatal development causes persistent locomotor hyperactivity in the adult. Mol Neurodegener. 2012;7:28.CrossRef
41.
go back to reference Sri S, et al. Emergence of synaptic and cognitive impairment in a mature-onset APP mouse model of Alzheimer’s disease. Acta Neuropathol Commun. 2019;7(1):25.CrossRef Sri S, et al. Emergence of synaptic and cognitive impairment in a mature-onset APP mouse model of Alzheimer’s disease. Acta Neuropathol Commun. 2019;7(1):25.CrossRef
42.
go back to reference Jankowsky JL, et al. Persistent amyloidosis following suppression of Abeta production in a transgenic model of Alzheimer disease. PLoS Med. 2005;2(12): e355.CrossRef Jankowsky JL, et al. Persistent amyloidosis following suppression of Abeta production in a transgenic model of Alzheimer disease. PLoS Med. 2005;2(12): e355.CrossRef
43.
go back to reference Gaberel T, et al. Impaired glymphatic perfusion after strokes revealed by contrast-enhanced MRI: a new target for fibrinolysis? Stroke. 2014;45(10):3092–6.CrossRef Gaberel T, et al. Impaired glymphatic perfusion after strokes revealed by contrast-enhanced MRI: a new target for fibrinolysis? Stroke. 2014;45(10):3092–6.CrossRef
44.
go back to reference Xavier ALR, et al. Cannula implantation into the cisterna magna of rodents. J Vis Exp. 2018;(135):57378. Xavier ALR, et al. Cannula implantation into the cisterna magna of rodents. J Vis Exp. 2018;(135):57378.
45.
go back to reference Keith BJ, Franklin M, Paxinos G. The mouse brain in stereotaxic coordinates, compact: the coronal plates and diagrams. Academic Press; 2008. ISBN 10: 0123742447 ISBN 13: 9780123742445. Keith BJ, Franklin M, Paxinos G. The mouse brain in stereotaxic coordinates, compact: the coronal plates and diagrams. Academic Press; 2008. ISBN 10: 0123742447 ISBN 13: 9780123742445.
46.
go back to reference Hinz R, et al. An in-vivo digital atlas of the spatially reliable cerebral vasculature in mice. bioRxiv. 2021: p. 2021.10.21.465264. Hinz R, et al. An in-vivo digital atlas of the spatially reliable cerebral vasculature in mice. bioRxiv. 2021: p. 2021.10.21.465264.
47.
go back to reference Gakuba C, et al. General anesthesia inhibits the activity of the “glymphatic system.” Theranostics. 2018;8(3):710–22.CrossRef Gakuba C, et al. General anesthesia inhibits the activity of the “glymphatic system.” Theranostics. 2018;8(3):710–22.CrossRef
48.
go back to reference Bechet NB, et al. Light sheet fluorescence microscopy of optically cleared brains for studying the glymphatic system. J Cereb Blood Flow Metab. 2020;40(10):1975–86.CrossRef Bechet NB, et al. Light sheet fluorescence microscopy of optically cleared brains for studying the glymphatic system. J Cereb Blood Flow Metab. 2020;40(10):1975–86.CrossRef
49.
go back to reference Xue Y, et al. In vivo T1 mapping for quantifying glymphatic system transport and cervical lymph node drainage. Sci Rep. 2020;10(1):14592.CrossRef Xue Y, et al. In vivo T1 mapping for quantifying glymphatic system transport and cervical lymph node drainage. Sci Rep. 2020;10(1):14592.CrossRef
50.
go back to reference Busche MA, et al. Critical role of soluble amyloid-beta for early hippocampal hyperactivity in a mouse model of Alzheimer’s disease. Proc Natl Acad Sci U S A. 2012;109(22):8740–5.CrossRef Busche MA, et al. Critical role of soluble amyloid-beta for early hippocampal hyperactivity in a mouse model of Alzheimer’s disease. Proc Natl Acad Sci U S A. 2012;109(22):8740–5.CrossRef
51.
go back to reference Kazim SF, et al. Early-onset network hyperexcitability in presymptomatic Alzheimer’s disease transgenic mice is suppressed by passive immunization with anti-human APP/Abeta antibody and by mGluR5 blockade. Front Aging Neurosci. 2017;9:71.CrossRef Kazim SF, et al. Early-onset network hyperexcitability in presymptomatic Alzheimer’s disease transgenic mice is suppressed by passive immunization with anti-human APP/Abeta antibody and by mGluR5 blockade. Front Aging Neurosci. 2017;9:71.CrossRef
52.
go back to reference Shah D, et al. Early pathologic amyloid induces hypersynchrony of BOLD resting-state networks in transgenic mice and provides an early therapeutic window before amyloid plaque deposition. Alzheimers Dement. 2016;12(9):964–76.CrossRef Shah D, et al. Early pathologic amyloid induces hypersynchrony of BOLD resting-state networks in transgenic mice and provides an early therapeutic window before amyloid plaque deposition. Alzheimers Dement. 2016;12(9):964–76.CrossRef
53.
go back to reference Bero AW, et al. Neuronal activity regulates the regional vulnerability to amyloid-beta deposition. Nat Neurosci. 2011;14(6):750–6.CrossRef Bero AW, et al. Neuronal activity regulates the regional vulnerability to amyloid-beta deposition. Nat Neurosci. 2011;14(6):750–6.CrossRef
54.
go back to reference Li S, et al. Soluble oligomers of amyloid Beta protein facilitate hippocampal long-term depression by disrupting neuronal glutamate uptake. Neuron. 2009;62(6):788–801.CrossRef Li S, et al. Soluble oligomers of amyloid Beta protein facilitate hippocampal long-term depression by disrupting neuronal glutamate uptake. Neuron. 2009;62(6):788–801.CrossRef
55.
go back to reference Zott B, et al. A vicious cycle of beta amyloid-dependent neuronal hyperactivation. Science. 2019;365(6453):559–65.CrossRef Zott B, et al. A vicious cycle of beta amyloid-dependent neuronal hyperactivation. Science. 2019;365(6453):559–65.CrossRef
56.
go back to reference Wang M, et al. Focal solute trapping and global glymphatic pathway impairment in a murine model of multiple microinfarcts. J Neurosci. 2017;37(11):2870–7.CrossRef Wang M, et al. Focal solute trapping and global glymphatic pathway impairment in a murine model of multiple microinfarcts. J Neurosci. 2017;37(11):2870–7.CrossRef
57.
go back to reference Ma Q, et al. Lymphatic outflow of cerebrospinal fluid is reduced in glioma. Sci Rep. 2019;9(1):14815.CrossRef Ma Q, et al. Lymphatic outflow of cerebrospinal fluid is reduced in glioma. Sci Rep. 2019;9(1):14815.CrossRef
58.
go back to reference Hablitz LM, et al. Increased glymphatic influx is correlated with high EEG delta power and low heart rate in mice under anesthesia. Sci Adv. 2019;5(2):eaav5447. Hablitz LM, et al. Increased glymphatic influx is correlated with high EEG delta power and low heart rate in mice under anesthesia. Sci Adv. 2019;5(2):eaav5447.
59.
go back to reference Kim SH, et al. Cerebral amyloid angiopathy aggravates perivascular clearance impairment in an Alzheimer’s disease mouse model. Acta Neuropathol Commun. 2020;8(1):181.CrossRef Kim SH, et al. Cerebral amyloid angiopathy aggravates perivascular clearance impairment in an Alzheimer’s disease mouse model. Acta Neuropathol Commun. 2020;8(1):181.CrossRef
60.
go back to reference Ahn JH, et al. Meningeal lymphatic vessels at the skull base drain cerebrospinal fluid. Nature. 2019;572(7767):62–6.CrossRef Ahn JH, et al. Meningeal lymphatic vessels at the skull base drain cerebrospinal fluid. Nature. 2019;572(7767):62–6.CrossRef
61.
go back to reference de Leon MJ, et al. Cerebrospinal fluid clearance in Alzheimer disease measured with dynamic PET. J Nucl Med. 2017;58(9):1471–6.CrossRef de Leon MJ, et al. Cerebrospinal fluid clearance in Alzheimer disease measured with dynamic PET. J Nucl Med. 2017;58(9):1471–6.CrossRef
62.
go back to reference Ringstad G, et al. Brain-wide glymphatic enhancement and clearance in humans assessed with MRI. JCI Insight. 2018;3(13):e121537. Ringstad G, et al. Brain-wide glymphatic enhancement and clearance in humans assessed with MRI. JCI Insight. 2018;3(13):e121537.
63.
go back to reference Eide PK, Ringstad G. Delayed clearance of cerebrospinal fluid tracer from entorhinal cortex in idiopathic normal pressure hydrocephalus: a glymphatic magnetic resonance imaging study. J Cereb Blood Flow Metab. 2019;39(7):1355–68.CrossRef Eide PK, Ringstad G. Delayed clearance of cerebrospinal fluid tracer from entorhinal cortex in idiopathic normal pressure hydrocephalus: a glymphatic magnetic resonance imaging study. J Cereb Blood Flow Metab. 2019;39(7):1355–68.CrossRef
64.
go back to reference Stocklein SM, et al. Decreased craniocervical CSF flow in patients with normal pressure hydrocephalus: a pilot study. AJNR Am J Neuroradiol. 2022;43(2):230–7.CrossRef Stocklein SM, et al. Decreased craniocervical CSF flow in patients with normal pressure hydrocephalus: a pilot study. AJNR Am J Neuroradiol. 2022;43(2):230–7.CrossRef
65.
go back to reference Zhou Y, et al. Impairment of the glymphatic pathway and putative meningeal lymphatic vessels in the aging human. Ann Neurol. 2020;87(3):357–69.CrossRef Zhou Y, et al. Impairment of the glymphatic pathway and putative meningeal lymphatic vessels in the aging human. Ann Neurol. 2020;87(3):357–69.CrossRef
66.
go back to reference May C, et al. Cerebrospinal fluid production is reduced in healthy aging. Neurology. 1990;40(3 Pt 1):500–3.CrossRef May C, et al. Cerebrospinal fluid production is reduced in healthy aging. Neurology. 1990;40(3 Pt 1):500–3.CrossRef
67.
go back to reference Mestre H, et al. Perivascular spaces, glymphatic dysfunction, and small vessel disease. Clin Sci (Lond). 2017;131(17):2257–74.CrossRef Mestre H, et al. Perivascular spaces, glymphatic dysfunction, and small vessel disease. Clin Sci (Lond). 2017;131(17):2257–74.CrossRef
68.
go back to reference Mestre H, et al. Aquaporin-4-dependent glymphatic solute transport in the rodent brain. Elife. 2018;7:e40070. Mestre H, et al. Aquaporin-4-dependent glymphatic solute transport in the rodent brain. Elife. 2018;7:e40070.
69.
go back to reference Benveniste H, et al. The glymphatic system and waste clearance with brain aging: a review. Gerontology. 2019;65(2):106–19.CrossRef Benveniste H, et al. The glymphatic system and waste clearance with brain aging: a review. Gerontology. 2019;65(2):106–19.CrossRef
70.
go back to reference Varga AW, et al. Reduced slow-wave sleep is associated with high cerebrospinal fluid abeta42 levels in cognitively normal elderly. Sleep. 2016;39(11):2041–8.CrossRef Varga AW, et al. Reduced slow-wave sleep is associated with high cerebrospinal fluid abeta42 levels in cognitively normal elderly. Sleep. 2016;39(11):2041–8.CrossRef
71.
go back to reference Ju YE, et al. Sleep quality and preclinical Alzheimer disease. JAMA Neurol. 2013;70(5):587–93.CrossRef Ju YE, et al. Sleep quality and preclinical Alzheimer disease. JAMA Neurol. 2013;70(5):587–93.CrossRef
72.
go back to reference Varga AW, et al. Effects of aging on slow-wave sleep dynamics and human spatial navigational memory consolidation. Neurobiol Aging. 2016;42:142–9.CrossRef Varga AW, et al. Effects of aging on slow-wave sleep dynamics and human spatial navigational memory consolidation. Neurobiol Aging. 2016;42:142–9.CrossRef
73.
go back to reference Da Mesquita S, et al. Meningeal lymphatics affect microglia responses and anti-Abeta immunotherapy. Nature. 2021;593(7858):255–60.CrossRef Da Mesquita S, et al. Meningeal lymphatics affect microglia responses and anti-Abeta immunotherapy. Nature. 2021;593(7858):255–60.CrossRef
74.
go back to reference Ma Q, et al. Outflow of cerebrospinal fluid is predominantly through lymphatic vessels and is reduced in aged mice. Nat Commun. 2017;8(1):1434.CrossRef Ma Q, et al. Outflow of cerebrospinal fluid is predominantly through lymphatic vessels and is reduced in aged mice. Nat Commun. 2017;8(1):1434.CrossRef
75.
go back to reference Da Mesquita S, et al. Functional aspects of meningeal lymphatics in ageing and Alzheimer’s disease. Nature. 2018;560(7717):185–91.CrossRef Da Mesquita S, et al. Functional aspects of meningeal lymphatics in ageing and Alzheimer’s disease. Nature. 2018;560(7717):185–91.CrossRef
76.
go back to reference Mentis AA, Dardiotis E, Chrousos GP. Apolipoprotein E4 and meningeal lymphatics in Alzheimer disease: a conceptual framework. Mol Psychiatry. 2021;26(4):1075–97.CrossRef Mentis AA, Dardiotis E, Chrousos GP. Apolipoprotein E4 and meningeal lymphatics in Alzheimer disease: a conceptual framework. Mol Psychiatry. 2021;26(4):1075–97.CrossRef
77.
go back to reference Chen Z, Schwulst SJ, Mentis AA. APOE4-mediated Alzheimer disease and “vascular”-“meningeal lymphatic” components: towards a novel therapeutic era? Mol Psychiatry. 2021;26(10):5472–4.CrossRef Chen Z, Schwulst SJ, Mentis AA. APOE4-mediated Alzheimer disease and “vascular”-“meningeal lymphatic” components: towards a novel therapeutic era? Mol Psychiatry. 2021;26(10):5472–4.CrossRef
78.
go back to reference Kimbrough IF, et al. Vascular amyloidosis impairs the gliovascular unit in a mouse model of Alzheimer’s disease. Brain. 2015;138(Pt 12):3716–33.CrossRef Kimbrough IF, et al. Vascular amyloidosis impairs the gliovascular unit in a mouse model of Alzheimer’s disease. Brain. 2015;138(Pt 12):3716–33.CrossRef
79.
go back to reference Simon M, et al. Loss of perivascular aquaporin-4 localization impairs glymphatic exchange and promotes amyloid beta plaque formation in mice. Alzheimers Res Ther. 2022;14(1):59.CrossRef Simon M, et al. Loss of perivascular aquaporin-4 localization impairs glymphatic exchange and promotes amyloid beta plaque formation in mice. Alzheimers Res Ther. 2022;14(1):59.CrossRef
80.
go back to reference Iadecola C, Nedergaard M. Glial regulation of the cerebral microvasculature. Nat Neurosci. 2007;10(11):1369–76.CrossRef Iadecola C, Nedergaard M. Glial regulation of the cerebral microvasculature. Nat Neurosci. 2007;10(11):1369–76.CrossRef
81.
go back to reference Mahmoud S, et al. Astrocytes maintain glutamate homeostasis in the CNS by controlling the balance between glutamate uptake and release. Cells. 2019;8(2):184. Mahmoud S, et al. Astrocytes maintain glutamate homeostasis in the CNS by controlling the balance between glutamate uptake and release. Cells. 2019;8(2):184.
82.
go back to reference Atwell S, et al. Genome-wide association study of 107 phenotypes in Arabidopsis thaliana inbred lines. Nature. 2010;465(7298):627–31.CrossRef Atwell S, et al. Genome-wide association study of 107 phenotypes in Arabidopsis thaliana inbred lines. Nature. 2010;465(7298):627–31.CrossRef
83.
go back to reference Wu C, et al. Glutamate and γ-aminobutyric acid differentially modulate glymphatic clearance of amyloid β through pulsation- and aquaporin-4 dependent mechanisms. bioRxiv. 2020: p. 2020.01.31.928481. Wu C, et al. Glutamate and γ-aminobutyric acid differentially modulate glymphatic clearance of amyloid β through pulsation- and aquaporin-4 dependent mechanisms. bioRxiv. 2020: p. 2020.01.31.928481.
84.
go back to reference Chiang ACA, et al. Combination anti-Abeta treatment maximizes cognitive recovery and rebalances mTOR signaling in APP mice. J Exp Med. 2018;215(5):1349–64.CrossRef Chiang ACA, et al. Combination anti-Abeta treatment maximizes cognitive recovery and rebalances mTOR signaling in APP mice. J Exp Med. 2018;215(5):1349–64.CrossRef
85.
go back to reference Lee Y, et al. Improvement of glymphatic-lymphatic drainage of beta-amyloid by focused ultrasound in Alzheimer’s disease model. Sci Rep. 2020;10(1):16144.CrossRef Lee Y, et al. Improvement of glymphatic-lymphatic drainage of beta-amyloid by focused ultrasound in Alzheimer’s disease model. Sci Rep. 2020;10(1):16144.CrossRef
86.
go back to reference Lin Y, et al. Repetitive transcranial magnetic stimulation increases the brain’s drainage efficiency in a mouse model of Alzheimer’s disease. Acta Neuropathol Commun. 2021;9(1):102.CrossRef Lin Y, et al. Repetitive transcranial magnetic stimulation increases the brain’s drainage efficiency in a mouse model of Alzheimer’s disease. Acta Neuropathol Commun. 2021;9(1):102.CrossRef
87.
go back to reference Wang A, et al. Robust amyloid clearance in a mouse model of Alzheimer’s disease provides novel insights into the mechanism of amyloid-beta immunotherapy. J Neurosci. 2011;31(11):4124–36.CrossRef Wang A, et al. Robust amyloid clearance in a mouse model of Alzheimer’s disease provides novel insights into the mechanism of amyloid-beta immunotherapy. J Neurosci. 2011;31(11):4124–36.CrossRef
Metadata
Title
Altered dynamics of glymphatic flow in a mature-onset Tet-off APP mouse model of amyloidosis
Authors
Inès R. H. Ben-Nejma
Aneta J. Keliris
Verdi Vanreusel
Peter Ponsaerts
Annemie Van der Linden
Georgios A. Keliris
Publication date
01-12-2023
Publisher
BioMed Central
Published in
Alzheimer's Research & Therapy / Issue 1/2023
Electronic ISSN: 1758-9193
DOI
https://doi.org/10.1186/s13195-023-01175-z

Other articles of this Issue 1/2023

Alzheimer's Research & Therapy 1/2023 Go to the issue