Traumatic brain injury (TBI), a leading cause of death and disability worldwide, is an established risk factor for the development of dementia and Alzheimer's disease later in life. Neurofibrillary tangles, aggregates composed of the intracellula protein tau, are one of the hallmarks of the Alzheimer's disease brain and are thought to cause the death of neurons in this condition. Although these tau aggregates are a common feature of the brain after TBI, the changes that occur in the brain after traumatic injury that make it vulnerable to tau aggregation remain undefined and no treatments exist that can prevent the development of neurodegeneration after TBI. We have recently defined a series of paravascular channels throughout the brain that provide an avenue for the brain to clear wastes from between neural cells. Fluid movement along these pathways and through the brain interstitium is facilitated by the water channel aquaporin-4 (AQP4) which is expressed in a highly polarized manner in astrocyte membranes that directly face the brain vasculature. When this channel is knocked out, fluid movement along these paravascular pathways is lost and the clearance of wastes from the brain interstitium is markedly reduced. Tau, the protein that chiefly makes up neurofibrillary tangles yet is released into the interstitium under resting conditions, is cleared from the brain along these paravascular pathways, while interstitial solute clearance along these paravascular pathways is slowed after TBI. At the same time, AQP4, which is normally localized to vessel-facing astrocyte processes, loses its polarization. Based on these findings, we propose that the loss of perivascular AQP4 polarization after TBI impairs interstitial tau clearance, promoting tau aggregation and neurodegeneration after TBI. In this proposal, we will first utilize a transgenic mouse model to determine whether loss of perivascular AQP4 localization impairs interstitial tau clearance. We will then use an in vivo viral transfection approach to test whether upregulation of the AQP4-M1 variant after TBI causes the loss of perivascular AQP4 polarization after TBI. We will then use these approaches in a transgenic mouse line exhibiting spontaneous age-related neurofibrillary tangle formation to test whether loss of perivascular AQP4 polarization and impairment of interstitial tau clearance promote tau aggregation and neurodegeneration after TBI. If validated, then these studies may provide a mechanistic basis for the vulnerability of the post-traumatic brain to tau aggregation and neurodegeneration, and may identify a new therapeutic approach to preventing the development of neurodegeneration after TBI.

Public Health Relevance

Traumatic brain injury (TBI), one of the leading causes of death and disability in the United States, is also an established risk factor for the development o dementia and Alzheimer's disease later in life; however the changes that occur in the post-traumatic brain that render it vulnerable to neurodegeneration are not known. This proposal will investigate whether the brain's waste-clearance system fails after TBI, leading to the harmful buildup of protein aggregates that damage brain cells. These studies will help to define why the post-traumatic brain is susceptible to the development of dementia and Alzheimer's disease, and may lead to the development of new treatments to prevent these events in patients suffering TBI.

Agency
National Institute of Health (NIH)
Institute
National Institute of Neurological Disorders and Stroke (NINDS)
Type
Research Project (R01)
Project #
1R01NS089709-01A1
Application #
8961755
Study Section
Brain Injury and Neurovascular Pathologies Study Section (BINP)
Program Officer
Bellgowan, Patrick S F
Project Start
2015-09-01
Project End
2020-06-30
Budget Start
2015-09-01
Budget End
2016-06-30
Support Year
1
Fiscal Year
2015
Total Cost
$322,524
Indirect Cost
$103,774
Name
Oregon Health and Science University
Department
Anesthesiology
Type
Schools of Medicine
DUNS #
096997515
City
Portland
State
OR
Country
United States
Zip Code
97239
Simon, Matthew J; Murchison, Charles; Iliff, Jeffrey J (2018) A transcriptome-based assessment of the astrocytic dystrophin-associated complex in the developing human brain. J Neurosci Res 96:180-193
Boespflug, Erin L; Iliff, Jeffrey J (2018) The Emerging Relationship Between Interstitial Fluid-Cerebrospinal Fluid Exchange, Amyloid-?, and Sleep. Biol Psychiatry 83:328-336
Mestre, Humberto; Hablitz, Lauren M; Xavier, Anna Lr et al. (2018) Aquaporin-4-dependent glymphatic solute transport in the rodent brain. Elife 7:
Goodman, James R; Adham, Zachariah O; Woltjer, Randall L et al. (2018) Characterization of dural sinus-associated lymphatic vasculature in human Alzheimer's dementia subjects. Brain Behav Immun 73:34-40
Simon, Matthew J; Wang, Marie X; Murchison, Charles F et al. (2018) Transcriptional network analysis of human astrocytic endfoot genes reveals region-specific associations with dementia status and tau pathology. Sci Rep 8:12389
Wang, Minghuan; Ding, Fengfei; Deng, SaiYue et al. (2017) Focal Solute Trapping and Global Glymphatic Pathway Impairment in a Murine Model of Multiple Microinfarcts. J Neurosci 37:2870-2877
Simon, Matthew J; Iliff, Jeffrey J (2016) Regulation of cerebrospinal fluid (CSF) flow in neurodegenerative, neurovascular and neuroinflammatory disease. Biochim Biophys Acta 1862:442-51