Abstract

Current treatments targeting amyloid beta in Alzheimer’s disease (AD) have minimal efficacy, which results in a huge unmet medical need worldwide. Accumulating data suggest that brain mitochondrial dysfunction play a critical role in AD pathogenesis. Targeting cellular mechanisms associated with mitochondrial dysfunction in AD create a novel approach for drug development. This study investigated the effects of nilotinib, as a selective tyrosine kinase inhibitor, in astroglia derived from 3xTg-AD mice versus their C57BL/6-controls. Parameters included oxygen consumption rates (OCR), ATP, cytochrome c oxidase (COX), citrate synthase (CS) activity, alterations in oxidative phosphorylation (OXPHOS), nuclear factor kappa B (NF-κB), key regulators of mitochondrial dynamics (mitofusin (Mfn1), dynamin-related protein 1 (Drp1)), and mitochondrial biogenesis (peroxisome proliferator-activated receptor gamma coactivator1-alpha (PGC-1α), calcium/calmodulin-dependent protein kinase II (CaMKII), and nuclear factor (erythroid-derived 2)-like 2 (Nrf2)). Nilotinib increased OCR, ATP, COX, Mfn1, and OXPHOS levels in 3xTg astroglia. No significant differences were detected in levels of Drp1 protein and CS activity. Nilotinib enhanced mitochondrial numbers, potentially through a CaMKII-PGC1α-Nrf2 pathway in 3xTg astroglia. Additionally, nilotinib-induced OCR increases were reduced in the presence of the NF-κB inhibitor, Bay11-7082. The data suggest that NF-κB signaling is intimately involved in nilotinib-induced changes in bioenergetics in 3xTg brain astroglia. Nilotinib increased translocation of the NF-κB p50 subunit into the nucleus of 3xTg astroglia that correlates with an increased expression and activation of NF-κB. The current findings support a role for nilotinib in improving mitochondrial function and suggest that astroglia may be a key therapeutic target in treating AD.

Highlights

  • Current treatments targeting amyloid beta in Alzheimer’s disease (AD) have minimal efficacy, which results in a huge unmet medical need worldwide

  • Approaches focusing on therapeutics that interfere with amyloid β plaques (Aβ), a classic hallmark of AD [2, 3], have failed for the most part to show efficacy for stopping or slowing cognitive decline

  • The mitochondrial oxygen consumption rate (OCR) was measured in astroglia derived from cortical brain regions from control (C57BL/6) and AD (3xTg) mice (Fig. 1A)

Read more

Summary

Introduction

Current treatments targeting amyloid beta in Alzheimer’s disease (AD) have minimal efficacy, which results in a huge unmet medical need worldwide. Accumulating data suggest that brain mitochondrial dysfunction play a critical role in AD pathogenesis. The data suggest that NF-κB signaling is intimately involved in nilotinib-induced changes in bioenergetics in 3xTg brain astroglia. The current findings support a role for nilotinib in improving mitochondrial function and suggest that astroglia may be a key therapeutic target in treating AD. As important central nervous system (CNS) cells, play key roles in the pathogenesis of neurodegenerative disorders such as AD; excessive oxidative stress, inflammation as well as mitochondrial impairment is observed in AD. Accumulating evidence highlights the critical role of astroglia in inflammatory, oxidative stress, modulation of synaptic activity, and bioenergetic changes present in AD [27,28,29,30,31]. The notion of testing nilotinib for AD may seem counterintuitive since AD involves cell loss and cancer involves unregulated cell proliferation, but the earliest deficits in the pathological progression of AD and cancer are associated with mitochondrial dysfunction; that is, before the robust appearance of neurotoxic proteins in AD and oncogene expression in cancer [4,5,6,7,8, 32,33,34,35,36,37,38,39]

Objectives
Methods
Results
Discussion
Conclusion
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