Our funded U01 project in the AMP-AD consortium had the overarching aim of identifying therapeutic targets within innate immunity pathways. In the previous funding period, we made significant progress and met all milestones of our U01. We have nominated multiple targets in the immune system, and these targets are at various stages of validation. Nevertheless, key gaps in our understanding impede transformation of the collective AMP-AD knowledge to validated therapeutic targets. Despite identification of numerous perturbed transcriptional networks, some enriched for Alzheimer's disease (AD) risk genes, key tractable targets in these networks are not unequivocally identified and validated. Further, whether the observed transcriptional changes are a simple consequence of disease or actually, play a role in the pathologic cascade has, typically, not been determined. Finally, the direction of molecular changes that is beneficial vs. detrimental has, in most cases of nominated targets, not been established. To overcome these gaps in knowledge, we will i) leverage unique aspects of our existing data and tools along with data and analyses generated by the larger AMP-AD consortia, ii) generate complementary new data, and iii) apply innovative analytic approaches. Notably, our ability to perform comparative analysis of control (no pathology), PathAg, (Pathologic Aging, amyloid+), AD (tau+, amyloid+) and PSP (progressive supranuclear palsy, tau+) enables a framework to identify therapeutic targets that play a role in the transition to different disease stages of AD. Further, comparisons between two brain regions (TCX=affected and CER=largely spared in AD) and between AD and PSP can help distinguish whether the molecular changes identified are likely a cause or consequence of pathology. In this renewal application, we propose to: i) refine and genetically validate therapeutic targets ii) identify molecular mechanisms and targets that mediate disease transition from control to PathAg to AD iii) define the drug target mechanism(s) iv) evaluate select prioritized targets in relevant models, v) continue the collaboration with all AMP-AD partners to promote consortium wide- target validation and vi) share our data openly with the larger scientific community. These studies will include the final phase of modeling studies for more than 20 immune targets identified in the original grant cycle with a goal of making firm go, no-go, decisions on select targets. These studies will enable us to i) provide biologic insight into the mechanism of action of the proposed targets and ii) inform on the direction of change needed for therapeutic benefit. Identification of key targets that drive the transition from control to amyloid positivity, and then to tau pathology and neurodegeneration will enable us to propose alignments of future clinical testing of therapeutics that manipulate these targets in appropriate disease stages-increasing the likelihood to achieve clinical efficacy and avoiding costly testing of an agent in an intent to treat population that is unlikely to benefit.

Public Health Relevance

Given there are no approved disease modifying therapies, and the personal, societal, and economic toll of Alzheimer's disease (AD) continues to increase there is a huge unmet medical need. Generation, mining and sharing of information from system level data; with widespread collaborative validations in other human data and model systems provides novel insights to the disease processes. This data we will use to identify and validate novel therapeutic approaches for AD.

Agency
National Institute of Health (NIH)
Institute
National Institute on Aging (NIA)
Type
Research Project--Cooperative Agreements (U01)
Project #
3U01AG046139-08S1
Application #
10155949
Study Section
Special Emphasis Panel (ZAG1)
Program Officer
Petanceska, Suzana
Project Start
2013-09-20
Project End
2023-08-31
Budget Start
2020-09-08
Budget End
2021-08-31
Support Year
8
Fiscal Year
2020
Total Cost
Indirect Cost
Name
Mayo Clinic Jacksonville
Department
Type
DUNS #
153223151
City
Jacksonville
State
FL
Country
United States
Zip Code
32224
Lessard, Christian B; Malnik, Samuel L; Zhou, Yingyue et al. (2018) High-affinity interactions and signal transduction between A? oligomers and TREM2. EMBO Mol Med 10:
Conway, Olivia J; Carrasquillo, Minerva M; Wang, Xue et al. (2018) ABI3 and PLCG2 missense variants as risk factors for neurodegenerative diseases in Caucasians and African Americans. Mol Neurodegener 13:53
Deming, Yuetiva; Dumitrescu, Logan; Barnes, Lisa L et al. (2018) Sex-specific genetic predictors of Alzheimer's disease biomarkers. Acta Neuropathol 136:857-872
Li, Zeran; Del-Aguila, Jorge L; Dube, Umber et al. (2018) Genetic variants associated with Alzheimer's disease confer different cerebral cortex cell-type population structure. Genome Med 10:43
Dawson, Ted M; Golde, Todd E; Lagier-Tourenne, Clotilde (2018) Animal models of neurodegenerative diseases. Nat Neurosci 21:1370-1379
Ren, Yingxue; van Blitterswijk, Marka; Allen, Mariet et al. (2018) TMEM106B haplotypes have distinct gene expression patterns in aged brain. Mol Neurodegener 13:35
Pace, Michael C; Xu, Guilian; Fromholt, Susan et al. (2018) Differential induction of mutant SOD1 misfolding and aggregation by tau and ?-synuclein pathology. Mol Neurodegener 13:23
Ren, Yingxue; Reddy, Joseph S; Pottier, Cyril et al. (2018) Identification of missing variants by combining multiple analytic pipelines. BMC Bioinformatics 19:139
Ridge, Perry G; Karch, Celeste M; Hsu, Simon et al. (2018) Correction to: Linkage, whole genome sequence, and biological data implicate variants in RAB10 in Alzheimer's disease resilience. Genome Med 10:4
Sun, Wenyan; Samimi, Hanie; Gamez, Maria et al. (2018) Pathogenic tau-induced piRNA depletion promotes neuronal death through transposable element dysregulation in neurodegenerative tauopathies. Nat Neurosci 21:1038-1048

Showing the most recent 10 out of 61 publications