Abstract

Accumulating evidences supported that knock-down of DHCR24 is linked to the pathological risk factors of AD, suggesting a potential role of DHCR24 in AD pathogenesis. However, the molecular mechanism link between DHCR24 and tauopathy remains unknown. Here, in order to elucidate the relationship between DHCR24 and tauopathy, we will focus on the effect of DHCR24 on the tau hyperphosphorylation at some toxic sites. In present study, we found that DHCR24 knock-down significantly lead to the hyperphosphorylation of tau sites at Thr181, Ser199, Thr231, Ser262, Ser396. Moreover, DHCR24 knock-down also increase the accumulation of p62 protein, simultaneously decreased the ratio of LC3-II/LC3-I and the number of autophagosome compared to the control groups, suggesting the inhibition of autophagy activity. In contrast, DHCR24 knock-in obviously abolished the effect of DHCR24 knock-down on tau hyperphosphrylation and autophagy. In addition, to elucidate the association between DHCR24 and tauopathy, we further showed that the level of plasma membrane cholesterol, lipid raft-anchored protein caveolin-1, and concomitantly total I class PI3-K (p110α), phospho-Akt (Thr308 and Ser473) were significantly decreased, resulting in the disruption of lipid raft/caveola and inhibition of PI3-K/Akt signaling in silencing DHCR24 SH-SY5Y cells compared to control groups. At the same time, DHCR24 knock-down simultaneously decreased the level of phosphorylated GSK3β at Ser9 (inactive form) and increased the level of phosphorylated mTOR at Ser2448 (active form), leading to overactivation of GSK3β and mTOR signaling. On the contrary, DHCR24 knock-in largely increased the level of membrane cholesterol and caveolin-1, suggesting the enhancement of lipid raft/caveola. And synchronously DHCR24 knock-in also abolished the effect of DHCR24 knock-down on the inhibition of PI3-K/Akt signaling as well as the overactivation of GSK3β and mTOR signaling. Collectively, our data strongly supported DHCR24 knock-down lead to tau hyperphosphorylation and the inhibition of autophagy by a lipid raft-dependent PI3-K/Akt-mediated GSK3β and mTOR signaling. Taking together, our results firstly demonstrated that the decrease of plasma membrane cholesterol mediated by DHCR24 deficiency might contribute to the tauopathy in AD and other tauopathies.

Highlights

  • Previous studies found that there is a significant reduction in the expression of new gene in specific vulnerable brain regions in Alzheimer’s disease (AD) patients, which was named selective AD indicator 1 (Seladin-1), or 3β-hydroxysterol- 24 reductase (DHCR24) (Greeve et al, 2000; Waterham et al, 2001)

  • Primary antibodies against DHCR24 (#2033), AKt (#4691), Phospho-Akt (T308) (#13038), Phospho-Akt (Ser473) (#4060), PI3 Kinase p110α (#4249), GSK-3β (#12456), Phospho-glycogen synthase kinase-3β (GSK3β)(Ser9) (#5558), mammalian target of rapamycin (mTOR) (#2983), Phospho-mTOR (#2971),Tau (#46687), Phospho-Tau (Thr181) (#12885), Phospho-Tau (Ser199) (#29957), Phospho-Tau (Ser396) (#9632), LC3B (#3868), P62/sequestosome 1 (SQSTM1) (#23214), GAPDH (#2118), secondary antibodies anti-rabbit IgG, HRP-linked antibody (#7074), Anti-rabbit IgG (H+L) (#5366), Anti-mouse IgG, HRP-linked antibody (#7076) were purchased from Cell Signaling Tech (USA), Primary antibodies against Caveolin-1, Phospho-Tau (Ser262) were purchased from British abcam Company, Primary antibodies against Phospho-Tau (Thr231) (4137) was purchased from China ABclonal Company

  • In SH-SY5Y cells, we found that silencing DHCR24 facilitated abnormal hyperphosphorylation of the tau sites, including Thr181, Ser199, Thr231, Ser262, and Ser 396

Read more

Summary

Introduction

Previous studies found that there is a significant reduction in the expression of new gene in specific vulnerable brain regions in Alzheimer’s disease (AD) patients, which was named selective AD indicator 1 (Seladin-1), or 3β-hydroxysterol- 24 reductase (DHCR24) (Greeve et al, 2000; Waterham et al, 2001). Studies indicate that DHCR24 transcription is selectively down-regulated in brain regions vulnerable to Alzheimer’s disease in the transgenic mouse model of AD (Livonen et al, 2002; Vanmierlo et al, 2010; Orre et al, 2014). In AD patients, it has been reported that DHCR24 transcription and protein expression were selectively down-regulated in the brain areas affected in AD (Greeve et al, 2000; Livonen et al, 2002; Liang et al, 2008) It seems that DHCR24 is likely to establish a new direct link between the familial and sporadic AD. To elucidate a molecular mechanism link between DHCR24 and tauopathy will help to clarify the role of DHCR24 in the pathogenesis of AD and other tauopathies

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