Abstract

BackgroundImproved therapeutics aimed at ameliorating the devastating effects of neurodegenerative diseases, such as Alzheimer’s disease (AD), are pertinent to help attenuate their growing prevalence worldwide. One promising avenue for such therapeutics lies in botanical medicines that have been efficaciously employed in the likes of traditional medicine doctrines for millennium. Integral to this approach is the necessity of neuritogenesis and/or neuroprotection to counterbalance the deleterious effects of amyloid-β (Aβ) proteins. Senegenin, a principle saponin of Polygala tenuifolia Willd., which has empirically shown to improve cognition and intelligence, was chosen to evaluate its cytoprotective potential and possible neuritogenic and neuroprotective effects.MethodsThe purpose of the present study was then to analyze morphological changes in neurite development and altered protein expression of two proteins requisite to neuritogenesis, growth associated protein 43 (Gap-43) and microtubule-associated protein 2 (MAP2) in PC 12 cells. Neuritogenic analysis was conducted with immunofluorescence after incubation with Aβ (25–35) peptide, and to deduce information on cell viability and mitochondrial functionality MTT (3,(4,5-dimethylthiazol-2-yl)2,5-diphenyltetrazolium bromide) was employed.ResultsThis study found that cells pre-incubated with senegenin for 24 h (40 μg and 20 μg/ml) before introducing Aβ attenuated Aβ-cytotoxicity, and significantly increased cell viability by 23 % and 34 % (P < 0.001), respectively. In neurite outgrowth experiments, Aβ was compared to NGF positive control and senegenin treated groups which showed a drastic decrease in the quantity, average length and maximum length of neurites (P < 0.001). At concentrations of 1 μg/ml (P < 0.01) and 5 μg/ml (P < 0.05) senegenin triggered neuritogenesis with significant increases in total neurite number, average length and maximum length. This was additionally shown through the augmented expression of MAP2 and Gap-43.ConclusionsThese results suggest that senegenin possesses cytoprotective properties, can moderate neurite outgrowth and augment MAP2 and Gap-43, thus suggesting a potential therapeutic value for Polygala tenuifolia in neurodegenerative disorders.

Highlights

  • Improved therapeutics aimed at ameliorating the devastating effects of neurodegenerative diseases, such as Alzheimer’s disease (AD), are pertinent to help attenuate their growing prevalence worldwide

  • The progressive cognitive decline associated with AD is attributable to dystrophic neurites and neuronal atrophy, which are the corollaries of extracellular amyloid-β peptide (Aβ) deposition and the intracellular deposition of neurofibrillary tangles (NFT) [2]; the latter of which is comprised of hyperphosphorylated tau proteins, and posited to be the resultant effect of the toxic cascade initiated by Aβ

  • The effects of senegenin on Aβ-induced cytotoxicity of PC 12 cells MTT was used to determine the effects of senegenin on the cell viability of PC 12 cells after Aβ-induced cytotoxicity

Read more

Summary

Introduction

Improved therapeutics aimed at ameliorating the devastating effects of neurodegenerative diseases, such as Alzheimer’s disease (AD), are pertinent to help attenuate their growing prevalence worldwide. One promising avenue for such therapeutics lies in botanical medicines that have been efficaciously employed in the likes of traditional medicine doctrines for millennium. Integral to this approach is the necessity of neuritogenesis and/or neuroprotection to counterbalance the deleterious effects of amyloid-β (Aβ) proteins. The progressive cognitive decline associated with AD is attributable to dystrophic neurites and neuronal atrophy, which are the corollaries of extracellular amyloid-β peptide (Aβ) deposition and the intracellular deposition of neurofibrillary tangles (NFT) [2]; the latter of which is comprised of hyperphosphorylated tau proteins, and posited to be the resultant effect of the toxic cascade initiated by Aβ. Various research models have employed the use of the Aβ (25–35) peptide, which retains the toxic properties of the full length peptide (39–43), to test the pathogenic mechanisms of Aβ, making it a desirable model for testing AD, and chosen for the present study to examine its toxicity in a neuronal cell line

Objectives
Methods
Results
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