DOI: 10.1002/alz.073079 ISSN: 1552-5260

BAG3 regulates RAB35 mediates the Endosomal Sorting Complexes Required for Transport/Endolysosome pathway and Tau clearance

Heng Lin, Maoping Tang, Changyi Ji, Peter Girardi, Gregor Cvetojevic, Daniel Chen, Shon Koren, Gail VW Johnson
  • Psychiatry and Mental health
  • Cellular and Molecular Neuroscience
  • Geriatrics and Gerontology
  • Neurology (clinical)
  • Developmental Neuroscience
  • Health Policy
  • Epidemiology

Abstract

Background

Declining proteostasis with aging contributes to increased susceptibility to neurodegenerative diseases, including Alzheimer’s disease (AD). Emerging studies implicate impairment of the endosome‐lysosome pathway as a significant factor in the pathogenesis of these diseases. Our lab was the first to demonstrate that BAG3 regulates phosphorylated tau clearance. However, we did not fully define how BAG3 regulates endogenous tau proteostasis, especially in the early stages of disease progression.

Methods

Mass spectrometric analyses were performed to identify neuronal BAG3 interactors. Multiple biochemical assays were used to investigate the BAG3‐HSP70‐TBC1D10B‐Rab35‐Hrs regulatory networks. Live‐cell imaging was used to study the dynamic of endosomal pathway. Immunohistochemistry and immunoblotting were performed in human AD brains and BAG3 overexpressed P301S tau transgenic mice.

Results

The primary group of neuronal BAG3 interactors identified are involved in the endocytic pathway. Among them were key regulators of small GTPases, such as the Rab35 GTPase activating protein, TBC1D10B. We demonstrated that a BAG3‐HSP70‐TBC1D10B complex attenuates the ability of TBC1D10B to inactivate Rab35. Thus, BAG3 interacts with TBC1D10B to support the activation of Rab35 and recruitment of Hrs, initiating ESCRT‐mediated endosomal tau clearance. Further,TBC1D10B shows significantly less co‐localization with BAG3 in AD brains than in age‐matched controls. Overexpression of BAG3 in P301S tau transgenic mice increased the co‐localization of phospho‐tau with the ESCRT III protein CHMP2B and reduced the levels of the mutant human tau.

Conclusion

We identified a novel BAG3‐TBC1D10B‐Rab35 regulatory axis that modulates ESCRT‐dependent protein degradation machinery and tau clearance. Dysregulation of BAG3 could contribute to the pathogenesis of AD.

More from our Archive