APOE genotype is the strongest genetic risk factor for late-onset Alzheimer?s disease (AD). A complete understanding of its role in AD pathogenesis remains unclear. In addition to the effects of apoE on A? clearance and aggregation, it is also likely to also influence AD via other mechanisms. Over the last 4.5 years of this grant, we have utilized a mouse model that develops tauopathy and neurodegeneration (P301S Tau Tg mice) and found that all human isoforms of apoE significantly enhance tau-mediated brain volume/neuronal loss and disease associated microglial and astrocyte response with apoE4 having the strongest effects. Interestingly, tau-mediated neurodegeneration and the enhanced innate immune response was strongly attenuated in the absence of apoE. Using a cell culture model, we also found that glial-derived apoE, particularly apoE4, could exacerbate neuronal damage, an effect that appears to involve apoE?s ability to enhance inflammation. In preliminary data, we have now found that if we overexpress the low density lipoprotein receptor (LDLR) in the brain of P301S Tau Tg mice, we can markedly lower apoE and attenuate brain atrophy, decrease p-tau levels, and decrease the disease associated microglial-response. We also found that cultured astrocytes secrete apoE in cholesterol/phospholipid rich HDL-like particles whereas cultured microglia secrete apoE in small, poorly lipidated particles. While these findings suggest important roles for apoE in modulating tau-mediated neurodegeneration, there are a number of important unanswered questions to address to better understand these effects. They include 1) Are astrocytes or microglia the main source of apoE that mediates these effects? 2) In addition to effects on A?, does astrocyte vs. microglial apoE in an isoform-specific fashion affect A?-induced tau seeding and spreading? 3) How do astrocyte and microglial- derived apoE particles differ, what is their structure and how does it differ by isoform, and do these differences result in different biological effects? 4) How and what is the source of apoE that influences the brain?s innate immune response to influence tau-mediated neurodegeneration? These questions lead us to hypothesize that apoE, particularly apoE4, influences the brain?s innate immune response to exacerbate tau- mediated neurodegeneration and A?-induced tau seeding/spreading. We propose these Aims:
Aim 1 : To determine whether apoE produced by astrocytes or microglia influences tau-mediated neurodegeneration using newly generated conditional apoE KI mice.
Aim 2 : To determine whether apoE produced by astrocytes or microglia influences human AD tau seeding/spreading +/- A? as well as CSF and plasma levels of p-tau, tau, A?, and apoE using a well characterized mouse model that develops A? amyloidosis APPNL-F KI mice.
Aim 3 : To characterize apoE particles produced by astrocytes and microglia both in vitro and in vivo via lipidomics and cryoEM. We will utilize a variety of techniques including cryoEM and lipidomics to characterize microglial and astrocyte-produced apoE particles.

Public Health Relevance

Alzheimer?s disease is a major public health problem with no treatments that delay, slow, or prevent the disease. In this proposal, we are determining if we can better understand how the major genetic risk factor for Alzheimer?s disease, APOE, leads to increased risk. This should allow us to develop novel treatments.

Agency
National Institute of Health (NIH)
Institute
National Institute of Neurological Disorders and Stroke (NINDS)
Type
Research Project (R01)
Project #
2R01NS090934-24
Application #
9881454
Study Section
Cellular and Molecular Biology of Neurodegeneration Study Section (CMND)
Program Officer
Mcgavern, Linda
Project Start
1996-09-01
Project End
2025-01-31
Budget Start
2020-02-15
Budget End
2021-01-31
Support Year
24
Fiscal Year
2020
Total Cost
Indirect Cost
Name
Washington University
Department
Neurology
Type
Schools of Medicine
DUNS #
068552207
City
Saint Louis
State
MO
Country
United States
Zip Code
63130
Huynh, Tien-Phat V; Liao, Fan; Francis, Caroline M et al. (2017) Age-Dependent Effects of apoE Reduction Using Antisense Oligonucleotides in a Model of ?-amyloidosis. Neuron 96:1013-1023.e4
Shi, Yang; Yamada, Kaoru; Liddelow, Shane Antony et al. (2017) ApoE4 markedly exacerbates tau-mediated neurodegeneration in a mouse model of tauopathy. Nature 549:523-527
Huynh, Tien-Phat V; Davis, Albert A; Ulrich, Jason D et al. (2017) Apolipoprotein E and Alzheimer's disease: the influence of apolipoprotein E on amyloid-? and other amyloidogenic proteins. J Lipid Res 58:824-836
Leyns, Cheryl E G; Holtzman, David M (2017) Glial contributions to neurodegeneration in tauopathies. Mol Neurodegener 12:50
Chung, Won-Suk; Verghese, Philip B; Chakraborty, Chandrani et al. (2016) Novel allele-dependent role for APOE in controlling the rate of synapse pruning by astrocytes. Proc Natl Acad Sci U S A 113:10186-91
Tambini, Marc D; Pera, Marta; Kanter, Ellen et al. (2016) ApoE4 upregulates the activity of mitochondria-associated ER membranes. EMBO Rep 17:27-36
Achariyar, Thiyagaragan M; Li, Baoman; Peng, Weiguo et al. (2016) Glymphatic distribution of CSF-derived apoE into brain is isoform specific and suppressed during sleep deprivation. Mol Neurodegener 11:74
Han, Byung Hee; Zhou, Meng-Liang; Johnson, Andrew W et al. (2015) Contribution of reactive oxygen species to cerebral amyloid angiopathy, vasomotor dysfunction, and microhemorrhage in aged Tg2576 mice. Proc Natl Acad Sci U S A 112:E881-90
Musiek, Erik S; Holtzman, David M (2015) Three dimensions of the amyloid hypothesis: time, space and 'wingmen'. Nat Neurosci 18:800-6
Yan, Ping; Zhu, Alec; Liao, Fan et al. (2015) Minocycline reduces spontaneous hemorrhage in mouse models of cerebral amyloid angiopathy. Stroke 46:1633-1640

Showing the most recent 10 out of 12 publications