Your browser doesn't support javascript.
loading
Staufen Impairs Autophagy in Neurodegeneration.
Paul, Sharan; Dansithong, Warunee; Gandelman, Mandi; Figueroa, Karla P; Zu, Tao; Ranum, Laura P W; Scoles, Daniel R; Pulst, Stefan M.
Afiliación
  • Paul S; Department of Neurology, University of Utah, Salt Lake City, UT.
  • Dansithong W; Department of Neurology, University of Utah, Salt Lake City, UT.
  • Gandelman M; Department of Neurology, University of Utah, Salt Lake City, UT.
  • Figueroa KP; Department of Neurology, University of Utah, Salt Lake City, UT.
  • Zu T; Center for NeuroGenetics and Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL.
  • Ranum LPW; Center for NeuroGenetics and Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL.
  • Scoles DR; Department of Neurology, University of Utah, Salt Lake City, UT.
  • Pulst SM; Department of Neurology, University of Utah, Salt Lake City, UT.
Ann Neurol ; 93(2): 398-416, 2023 02.
Article en En | MEDLINE | ID: mdl-36151701
ABSTRACT

OBJECTIVE:

The mechanistic target of rapamycin (mTOR) kinase is one of the master coordinators of cellular stress responses, regulating metabolism, autophagy, and apoptosis. We recently reported that staufen1 (STAU1), a stress granule (SG) protein, was overabundant in fibroblast cell lines from patients with spinocerebellar ataxia type 2 (SCA2), amyotrophic lateral sclerosis, frontotemporal degeneration, Huntington's, Alzheimer's, and Parkinson's diseases as well as animal models, and patient tissues. STAU1 overabundance is associated with mTOR hyperactivation and links SG formation with autophagy. Our objective was to determine the mechanism of mTOR regulation by STAU1.

METHODS:

We determined STAU1 abundance with disease- and chemical-induced cellular stressors in patient cells and animal models. We also used RNA-binding assays to contextualize STAU1 interaction with MTOR mRNA.

RESULTS:

STAU1 and mTOR were overabundant in bacterial artificial chromosome (BAC)-C9ORF72, ATXN2Q127 , and Thy1-TDP-43 transgenic mouse models. Reducing STAU1 levels in these mice normalized mTOR levels and activity and autophagy-related marker proteins. We also saw increased STAU1 levels in HEK293 cells transfected to express C9ORF72-relevant dipeptide repeats (DPRs). Conversely, DPR accumulations were not observed in cells treated by STAU1 RNA interference (RNAi). Overexpression of STAU1 in HEK293 cells increased mTOR levels through direct MTOR mRNA interaction, activating downstream targets and impairing autophagic flux. Targeting mTOR by rapamycin or RNAi normalized STAU1 abundance in an SCA2 cellular model.

INTERPRETATION:

STAU1 interaction with mTOR drives its hyperactivation and inhibits autophagic flux in multiple models of neurodegeneration. Staufen, therefore, constitutes a novel target to modulate mTOR activity and autophagy, and for the treatment of neurodegenerative diseases. ANN NEUROL 2023;93398-416.
Asunto(s)

Texto completo: 1 Base de datos: MEDLINE Asunto principal: Ataxias Espinocerebelosas / Serina-Treonina Quinasas TOR Idioma: En Revista: Ann Neurol Año: 2023 Tipo del documento: Article

Texto completo: 1 Base de datos: MEDLINE Asunto principal: Ataxias Espinocerebelosas / Serina-Treonina Quinasas TOR Idioma: En Revista: Ann Neurol Año: 2023 Tipo del documento: Article