Glymphatic clearance impaired in a mouse model of tauopathy: captured using contrast-enhanced MRI
Ian F Harrison1, Asif Machhada1, Niall Colgan1, Ozama Ismail1, James M O'Callaghan1, Holly E Holmes1, Jack A Wells1, Alexander V Gourine2, Tracey K Murray3, Zeshan Ahmed3, Ross A Johnson4, Emily C Collins4, Michael J O'Neill3, and Mark F Lythgoe1

1Centre for Advanced Biomedical Imaging, University College London, London, United Kingdom, 2Department of Neuroscience, Physiology & Pharmacology, University College London, London, United Kingdom, 3Eli Lilly and Company, Windlesham, United Kingdom, 4Eli Lilly and Company, Indianapolis, IN, United States

Synopsis

The ‘glymphatic’ clearance system is a brain-wide pathway for removal of waste solutes from the brain. It has recently been implicated in Alzheimer’s disease (AD), due to discovery that both amyloid and tau, accumulations of which lead to AD development, can be cleared from the brain via this pathway. We therefore hypothesise that an impairment of ‘glymphatic’ clearance occurs in the initial stages of disease development, leading to accumulation of amyloid and tau in the brain. Here, we determine whether this is the case, by using dynamic contrast-enhanced MRI to quantify glymphatic clearance in the brain of a mouse model of AD.

Purpose

Alzheimer’s disease (AD) is the most common form of dementia, with prevalence estimated currently to stand at around 24 million, a figure thought to quadruple by the year 2050 due to our ageing population1. Neuropathologically, AD is characterised by the formation of two species of toxic protein aggregates in the brain: extracellular accumulation of amyloid-β (Aβ) in the form of plaques, and intracellular accumulation of hyperphosphorylated tau in the form of neurofibrillary tangles (NFTs)2. Therefore failure of the clearance mechanisms of Aβ and tau from the brain parenchyma are becoming increasingly recognised in the pathogenesis of AD3.

A recently described mechanism of solute clearance from the brain is referred to as the ‘glymphatic’ clearance pathway, based on its appropriation of the lymphatic function of interstitial protein management, and its dependence upon glial water transport4. In this brain-wide pathway, cerebrospinal fluid (CSF) enters the brain along para-arterial routes via convective bulk flow, where it exchanges with interstitial fluid (ISF) and is cleared from the brain along para-venous routes, taking interstitial solutes with it4. Recently this pathway has been imaged using contrast-enhanced MRI and two photon microscopy, demonstrating that Aβ is indeed a substrate for ‘glymphatic’ clearance in mice5, and that tau follows the same para-venous route of clearance in the mouse brain6, hence the implication of impairment of this ‘glymphatic’ clearance pathway in AD pathogenesis.

Therefore in this study we sought to quantify the extent of ‘glymphatic’ clearance in the brain of an animal model of tauopathy using contrast-enhanced MRI and intracortical injection of tau to determine the effects of pathological tau accumulation on ‘glymphatic’ inflow and tau clearance. Furthermore, it is known that astrocytic expression of the water channel, aquaporin-4, and its polarisation to astrocytic endfeet surrounding blood vessels in the brain is crucially involved in facilitation of ‘glymphatic’ clearance. Hence molecular and cellular expression analysis of this channel in animal brains was additionally utilised to help elucidate the mechanism by which ‘glymphatic’ clearance is achieved and/or is impaired in this model.

Methods

Generation of homozygous rTg4510 transgenic mice has been reported previously7. Glymphatic inflow in the brains of 8.5 month old rTg4510 and litter-matched wildtype mice was captured using contrast-enhanced MRI. Briefly, an intrathecal cannula was surgically implanted into the cisterna magna of mice (see figure 1A). After baseline T1-weighted MR images were acquired, low molecular weight paramagnetic contrast agent Gadolinium (Magnevist®, 21mM Gd-DTPA), was infused intrathecally via the implanted cannula (0.6 µl/min, total infusion time, 50mins) and subsequent T1-weighted MR images acquired every 8mins for 120mins. 3D region-of-interest analysis of brain regions was then performed to determine the infiltration of Gadolinium-tagged into the brain parenchyma.

Additionally glymphatic clearance of tau from the cortex in these animals was quantified using intracortical injection of tau and its subsequent detection in extracted CSF. Briefly, tau was extracted from the brain of a late stage rTg4510 animal, and prepared for injection into either the rostral or caudal cortex. 60mins post injection, CSF was extracted from the cisterna magna and containing tau quantified using an ELISA.

Histological examination and laser capture microdissection of astrocytes surrounding blood vessels, and quantification of their aquaporin-4 expression was additionally performed to help understand the involvement of this water channel in mediating changes in glymphatic function in this animal model of tauopathy.

Results

Glymphatic inflow of MR contrast agent was significantly impaired in the caudal cortex of rTg4510 mice compared to wildtype animals (see figure 1), in line with reduced tau clearance and pathological accumulation of NFTs. Aquaporin-4 expression levels and the extent of astrocytic aquaporin-4 polarisation in this region suggest an involvement of this glial water channel in mediating glymphatic impairment in this animal model of tauopathy (see figure 2).

Conclusions

Aβ and tau are both known to be cleared from the brain via the recently described ‘glymphatic’ clearance pathway5,6. Based on these previous findings we therefore hypothesised that an impairment of ‘glymphatic’ clearance occurs during AD development leading to accumulation of these toxic proteins in the brain. Here we demonstrate that pathological accumulation of tau in the rTg4510 animal model of tauopathy is associated with impaired ‘glymphatic’ clearance from the brain. Expression levels and polarisation of astrocytic aquaporin-4 highlight a possible role for this protein in impairment of ‘glymphatic’ clearance in this model. This is the first investigation of glymphatic clearance in a tau model and warrants further investigation of the mechanisms involved. Furthermore, this study provides proof-of-concept that manipulation of the glymphatic clearance pathway may harbour new avenues of therapeutic intervention for AD.

Acknowledgements

This work was carried out in collaboration with, and funded by, Eli Lilly and Company.

References

[1] Reitz, C. and R. Mayeux, Alzheimer disease: Epidemiology, diagnostic criteria, risk factors and biomarkers. Biochemical Pharmacology, 2014. 88(4): p. 640-651.

[2] Huang, Y. and L. Mucke, Alzheimer Mechanisms and Therapeutic Strategies. Cell, 2012. 148(6): p. 1204-1222.

[3] Tarasoff-Conway, J.M., et al., Clearance systems in the brain - implications for Alzheimer disease. Nat Rev Neurol, 2015. 11(8): p. 457-470.

[4] Jessen, N.A., et al., The Glymphatic System: A Beginner’s Guide. Neurochemical Research, 2015: p. 1-17.

[5] Iliff, J.J., et al., A Paravascular Pathway Facilitates CSF Flow Through the Brain Parenchyma and the Clearance of Interstitial Solutes, Including Amyloid β. Science translational medicine, 2012. 4(147): p. 147ra111-147ra111.

[6] Iliff, J.J., et al., Impairment of Glymphatic Pathway Function Promotes Tau Pathology after Traumatic Brain Injury. The Journal of Neuroscience, 2014. 34(49): p. 16180-16193.

[7] Ramsden, M., et al., Age-Dependent Neurofibrillary Tangle Formation, Neuron Loss, and Memory Impairment in a Mouse Model of Human Tauopathy (P301L), The Journal of Neuroscience, 2005, 25: p. 10637-10647.

Figures

(A) Schematic showing location of intrathecal administration of Gadolinium (Magnevist®, 21mM Gd-DTPA) to the cisterna magna. Subset of representative serially acquired T1-weighted MR images following intrathecal infusion of contrast agent in (B) wildtype and (C) rTg4510 mice. (D) Intensity-time-plot showing 3D region of interest intensity analysis of the caudal cortex.

Immunofluorescent staining of blood vessel cross-sections in (A) wildtype, and (B) rTg4510 mouse brains, showing localisation of aquaporin-4 (red), CD31 (green), and DAPI (blue). Quantification of mean radial signal intensity in (C) wildtype, and (D) rTg4510 mouse blood vessels demonstrate reduced aquaporin polarisation surrounding blood vessels.



Proc. Intl. Soc. Mag. Reson. Med. 24 (2016)
4442