1. Academic Validation
  2. RPS23RG1 modulates tau phosphorylation and axon outgrowth through regulating p35 proteasomal degradation

RPS23RG1 modulates tau phosphorylation and axon outgrowth through regulating p35 proteasomal degradation

  • Cell Death Differ. 2021 Jan;28(1):337-348. doi: 10.1038/s41418-020-00620-y.
Dongdong Zhao 1 Yunqiang Zhou 1 Yuanhui Huo 1 Jian Meng 1 Xiaoxia Xiao 1 Linkun Han 1 Xian Zhang 1 Hong Luo 1 Dan Can 1 Hao Sun 1 Timothy Y Huang 2 Xin Wang 1 Jie Zhang 1 Fa-Rong Liu 3 Huaxi Xu 1 Yun-Wu Zhang 4 5
Affiliations

Affiliations

  • 1 Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China.
  • 2 Neuroscience Initiative, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA.
  • 3 Department of Psychology, Xiamen Xianyue Hospital, Xiamen, 361012, Fujian, China.
  • 4 Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China. yunzhang@xmu.edu.cn.
  • 5 Department of Neurology, The First Affiliated Hospital of Xiamen University, Xiamen, 361003, Fujian, China. yunzhang@xmu.edu.cn.
Abstract

Tauopathies are a group of neurodegenerative diseases characterized by hyperphosphorylation of the microtubule-binding protein, tau, and typically feature axon impairment and synaptic dysfunction. Cyclin-dependent kinase5 (CDK5) is a major tau kinase and its activity requires p35 or p25 regulatory subunits. P35 is subjected to rapid proteasomal degradation in its membrane-bound form and is cleaved by calpain under stress to a stable p25 form, leading to aberrant CDK5 activation and tau hyperphosphorylation. The type Ib transmembrane protein RPS23RG1 has been implicated in Alzheimer's disease (AD). However, physiological and pathological roles for RPS23RG1 in AD and other tauopathies are largely unclear. Herein, we observed retarded axon outgrowth, elevated p35 and p25 protein levels, and increased tau phosphorylation at major CDK5 phosphorylation sites in Rps23rg1 knockout (KO) mice. Both downregulation of p35 and the CDK5 Inhibitor roscovitine attenuated tau hyperphosphorylation and axon outgrowth impairment in Rps23rg1 KO neurons. Interestingly, interactions between the RPS23RG1 carboxyl-terminus and p35 amino-terminus promoted p35 membrane distribution and proteasomal degradation. Moreover, P301L tau transgenic (Tg) mice showed increased tau hyperphosphorylation with reduced RPS23RG1 levels and impaired axon outgrowth. Overexpression of RPS23RG1 markedly attenuated tau hyperphosphorylation and axon outgrowth defects in P301L tau Tg neurons. Our results demonstrate the involvement of RPS23RG1 in tauopathy disorders, and implicate a role for RPS23RG1 in inhibiting tau hyperphosphorylation through homeostatic p35 degradation and suppression of CDK5 activation. Reduced RPS23RG1 levels in tauopathy trigger aberrant Cdk5-p35 activation, consequent tau hyperphosphorylation, and axon outgrowth impairment, suggesting that RPS23RG1 may be a potential therapeutic target in tauopathy disorders.

Figures
Products