Central to the pathophysiology of cognitive dysfunction in Alzheimer's disease (AD) is loss of synapses with an impairment of plasticity at surviving synapses. Therapeutic efforts to intervene in AD have focused on the A peptide as an upstream trigger for synaptic disease, but clinical trials have been disappointing so far. Additional validated targets for AD therapy are needed, in particular those focused more directly on synaptic deficits. By their very nature, genetic studies of Late Onset AD (LOAD) risk are of direct clinical relevance. The largest GWAS analysis of LOAD identified a short list of genes whose common variants alter risk, providing potential new targets for AD therapy. We considered whether any of these might be directly linked to synaptic dysfunction in AD. Nearly all of the LOAD risk genes are hypothesized to bind A, to alter A metabolism, to regulate cellular endocytosis, or to modulate immune function. Therefore, their action on synaptic dysfunction must be considered indirect, via A levels or via the immune reaction to pathology. From the list of AD genetic risk factors, Pyk2 (also PTK2B or FAK2) is the only gene recognized to encode a protein concentrated at post- synaptic densities with direct effects on synaptic plasticity. Here, we seek to assess the role of Pyk2 in AD genetically and mechanistically, and to evaluate the protein as a therapeutic target focused on synaptic dysfunction. Previously, we have studied the biochemical basis for A oligomer (Ao) toxicity in neurons and these studies have also implicated Pyk2. Using an unbiased genome-wide screening method we searched for A oligomer- specific binding sites expressed in brain, and identified PrPC. We defined an Ao?PrPC?mGluR5?Fyn cascade that damages synapses in AD models. Importantly, the Pyk2 protein physically associates with mGluR5 and Fyn, as well as being implicated in synaptic plasticity. We will evaluate the hypothesis that Pyk2 is essential for manifestations of human familial AD transgene phenotypes in mice, and we will assess Pyk2 dysregulation in human AD samples. We will also target the enzyme pharmacologically. These studies have the potential to couple a validated LOAD genetic risk to synaptic dysfunction in AD, and may establish a connection with the Ao?PrPC?mGluR5?Fyn cascade. Critically, these data may guide attempts to develop pharmacological tools to target the synaptic Pyk2 pathway for therapeutic intervention in AD.

Public Health Relevance

Disease-modifying therapy for Alzheimer's disease (AD) remains a massive unmet medical need. Therapies that preserve synaptic function in AD are crucial. Genetic risk studies of AD have identified one protein that may be directly linked to synapse loss in AD, namely Pyk2. Here, we will evaluate the genetic and mechanistic role of Pyk2 in synapse stability and plasticity, and its role in AD models. We will target the enzyme therapeutically in preclinical studies to evaluate its potential for AD modification.

Agency
National Institute of Health (NIH)
Institute
National Institute on Aging (NIA)
Type
Multi-Year Funded Research Project Grant (RF1)
Project #
1RF1AG053000-01A1
Application #
9196005
Study Section
Cell Death in Neurodegeneration Study Section (CDIN)
Program Officer
Yang, Austin Jyan-Yu
Project Start
2016-09-01
Project End
2021-06-30
Budget Start
2016-09-01
Budget End
2021-06-30
Support Year
1
Fiscal Year
2016
Total Cost
$2,090,105
Indirect Cost
$840,105
Name
Yale University
Department
Neurology
Type
Schools of Medicine
DUNS #
043207562
City
New Haven
State
CT
Country
United States
Zip Code
06520
Kostylev, Mikhail A; Tuttle, Marcus D; Lee, Suho et al. (2018) Liquid and Hydrogel Phases of PrPC Linked to Conformation Shifts and Triggered by Alzheimer's Amyloid-? Oligomers. Mol Cell 72:426-443.e12
Strittmatter, Stephen M (2018) Emerging Mechanisms in Alzheimer's Disease and Their Therapeutic Implications. Biol Psychiatry 83:298-299
Salazar, Santiago V; Cox, Timothy O; Lee, Suho et al. (2018) Alzheimer's Disease Risk Factor Pyk2 Mediates Amyloid-? Induced Synaptic Dysfunction and Loss. J Neurosci :
Brody, A Harrison; Strittmatter, Stephen M (2018) Synaptotoxic Signaling by Amyloid Beta Oligomers in Alzheimer's Disease Through Prion Protein and mGluR5. Adv Pharmacol 82:293-323
Smith, Levi M; Zhu, Rong; Strittmatter, Stephen M (2018) Disease-modifying benefit of Fyn blockade persists after washout in mouse Alzheimer's model. Neuropharmacology 130:54-61
Smith, Levi M; Strittmatter, Stephen M (2017) Binding Sites for Amyloid-? Oligomers and Synaptic Toxicity. Cold Spring Harb Perspect Med 7:
Salazar, Santiago V; Gallardo, Christopher; Kaufman, Adam C et al. (2017) Conditional Deletion of Prnp Rescues Behavioral and Synaptic Deficits after Disease Onset in Transgenic Alzheimer's Disease. J Neurosci 37:9207-9221
Haas, Laura T; Salazar, Santiago V; Smith, Levi M et al. (2017) Silent Allosteric Modulation of mGluR5 Maintains Glutamate Signaling while Rescuing Alzheimer's Mouse Phenotypes. Cell Rep 20:76-88
Takahashi, Hideyuki; Klein, Zoe A; Bhagat, Sarah M et al. (2017) Opposing effects of progranulin deficiency on amyloid and tau pathologies via microglial TYROBP network. Acta Neuropathol 133:785-807
Klein, Zoe A; Takahashi, Hideyuki; Ma, Mengxiao et al. (2017) Loss of TMEM106B Ameliorates Lysosomal and Frontotemporal Dementia-Related Phenotypes in Progranulin-Deficient Mice. Neuron 95:281-296.e6

Showing the most recent 10 out of 14 publications