Abstract

The acetylcholinesterase inhibitors donepezil and rivastigmine have been used as therapeutic drugs for Alzheimer’s disease (AD), but their effects on LPS- and Aβ-induced neuroinflammatory responses and the underlying molecular pathways have not been studied in detail in vitro and in vivo. In the present study, we found that 10 or 50 μM donepezil significantly decreased the LPS-induced increases in the mRNA levels of a number of proinflammatory cytokines in BV2 microglial cells, whereas 50 μM rivastigmine significantly diminished only LPS-stimulated IL-6 mRNA levels. In subsequent experiments in primary astrocytes, donepezil suppressed only LPS-stimulated iNOS mRNA levels. To identify the molecular mechanisms by which donepezil regulates LPS-induced neuroinflammation, we examined whether donepezil alters LPS-stimulated proinflammatory responses by modulating LPS-induced downstream signaling and the NLRP3 inflammasome. Importantly, we found that donepezil suppressed LPS-induced AKT/MAPK signaling, the NLRP3 inflammasome, and transcription factor NF-kB/STAT3 phosphorylation to reduce neuroinflammatory responses. In LPS-treated wild-type mice, a model of neuroinflammatory disease, donepezil significantly attenuated LPS-induced microglial activation, microglial density/morphology, and proinflammatory cytokine COX-2 and IL-6 levels. In a mouse model of AD (5xFAD mice), donepezil significantly reduced Aβ-induced microglial and astrocytic activation, density, and morphology. Taken together, our findings indicate that donepezil significantly downregulates LPS- and Aβ-evoked neuroinflammatory responses in vitro and in vivo and may be a therapeutic agent for neuroinflammation-associated diseases such as AD.

Highlights

  • Before examining the effects of donepezil and rivastigmine on LPS-mediated proinflammatory cytokine levels, we first assessed the cytotoxicity of donepezil and rivastigmine at concentrations of 1–50 μM in BV2 microglial cells using the MTT assay

  • Vehicle (1% dimethyl sulfoxide (DMSO)) for 24 h (n = 6/group). (c,d) RT-PCR analysis of proinflammatory cytokine levels in BV2 microglial cells pretreated with LPS or PBS and treated with donepezil or vehicle as shown (n = 16/group). (e,f) RT-PCR analysis of proinflammatory cytokine levels in BV2 microglial cells pretreated with LPS or PBS and treated with rivastigmine or vehicle (1% DMSO) as shown (n = 8/group). (g) q-PCR analysis of proinflammatory cytokine levels in BV2 microglial cells pretreated with LPS or PBS and treated with 100 or 200 μM rivastigmine or vehicle (1% DMSO) as in (e,f) (n = 6/group). (h) q-PCR analysis of proinflammatory cytokine levels in primary astrocytes pretreated with LPS or PBS and treated with donepezil or vehicle (1% DMSO) as shown (n = 8/group)

  • Analysis of proinflammatory cytokines by RT-PCR showed that donepezil significantly reduced LPS-mediated COX-2, IL-1β, IL-6 and iNOS mRNA levels in BV2 microglial cells (Figure 1c,d), whereas 50 μM rivastigmine significantly decreased only LPS-mediated IL-6 mRNA levels (Figure 1e,f)

Read more

Summary

Introduction

Abnormal microglial and/or astrocyte activation accelerates the occurrence and development of neuroinflammatory responses [2]. Factor kappa-B (NF-κB)/p-signal transducer and activator of transcription–3 (STAT3) by binding to Toll-like receptor-4 (TLR-4) in in vitro and in vivo models [4,5]. Another inflammatory stimulus and a hallmark of AD, Aβ, leads to neurotoxicity by promoting microglial and astrocyte activation, which in turn triggers the release of proinflammatory cytokines through host cell-derived damage-associated molecular patterns secretion [1]. Regulating microglial and astrocyte activation as well as neuroinflammation-associated molecular targets may be an effective therapeutic strategy for neurodegenerative diseases

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