Abstract

AbstractBackgroundAlzheimer’s Disease (AD) etiology involves a complex interplay of genetics, age, and environmental factors that influence neuroepigenetic mediated neural gene control. Reduced histone acetylation in the AD brain is a widespread hallmark underlying neural gene repression and cognitive impairment in AD etiology. As histone acetylation is dynamically regulated by antagonistic histone deacetylases (HDACs) and histone acetyltransferases (HATs), pharmacological treatments aiming to restore histone acetylation via HDAC inhibition are a research hotspot for developing AD cognitive enhancing drugs. Although promising, HDAC inhibitor treatments in the AD animal models are rarely translated into clinical trials due to non‐specific global hyperacetylation. Alternatively, enhancing activity of specific HATs with non‐redundant functions serves as an exciting new therapeutic strategy that remains unexplored. To this end, our lab has identified and published a neuroprotective role for HAT Tip60 in multiple neural circuits impaired in AD pathology. Human and Drosophila AD brains exhibit reduced Tip60 and enhanced repressor HDAC2 that triggers histone deacetylation with concomitant repression of cognition genes. All these deficits are prevented by genetically increasing Tip60 in the Drosophila AD brain. Here, we test that pharmacologically enhancing Tip60’s HAT function replicates this AD neuroprotective function.MethodWe used in silico structural modeling and a novel pharmacophore based virtual screening approach to design and synthesize novel small molecule compounds strongly predicted to target and enhance Tip60’s HAT activity.ResultOur screen produced 19,000 hits that we individually docked on Tip60’s HAT domain using GOLD docking software. 13 compounds were identified that exhibited significantly high Tip60 docking scores, and optimal ease of synthesis, drug like properties and blood brain barrier peneteration. These compounds were custom synthesized. We show that several of these compounds fully rescue locomotor neural based deficits and cognition gene expression alterations in both Tip60 knockdown and AD Drosophila in vivo models with no observable side effects, underscoring their therapeutic effectiveness. These compounds will ultimately be assessed for further specificity and for their therapeutic effectiveness in AD animal models in vivo.ConclusionWe propose our first‐in‐class small molecule Tip60 HAT activators will serve as powerful novel chemical entities for development of specific acetylation‐based cognition enhancing drugs for AD.

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call