4.3 Article

Enhanced Activity of Exportin-1/CRM1 in Neurons Contributes to Autophagy Dysfunction and Senescent Features in Old Mouse Brain

Journal

OXIDATIVE MEDICINE AND CELLULAR LONGEVITY
Volume 2021, Issue -, Pages -

Publisher

HINDAWI LTD
DOI: 10.1155/2021/6682336

Keywords

-

Categories

Funding

  1. CONACyT [586932, 588372, FC921, CF2019514879]
  2. UNAMPAPIIT [IN206518-IN209221]
  3. Secretaria de Educacion, Ciencia, Tecnologia e Innovacion de la Ciudad de Mexico [SECTEI/008/2018, M-SECTEI/200/2020]
  4. FONDECYT [1190518, 3190608]
  5. Geroscience Center for Brain Health and Metabolism [FONDAP-15150012]

Ask authors/readers for more resources

Brain aging is linked to dysfunctional autophagy and cellular senescence, with CRM1 playing a crucial role in modulating these processes. Increased CRM1 activity during aging impairs autophagic flux in neurons, contributing to neuronal senescence. Inhibition of CRM1 has been shown to improve autophagy flux and reduce senescence in neuronal cells.
Brain aging is characterized by dysfunctional autophagy and cellular senescence, among other features. While autophagy can either promote or suppress cellular senescence in proliferating cells, in postmitotic cells, such as neurons, autophagy impairment promotes cellular senescence. CRM1 (exportin-1/XPO1) exports hundreds of nuclear proteins into the cytoplasm, including the transcription factors TFEB (the main inducer of autophagy and lysosomal biogenesis genes) and STAT3, another autophagy modulator. It appears that CRM1 is a modulator of aging-associated senescence and autophagy, because pharmacological inhibition of CRM1 improved autophagic degradation in flies, by increasing nuclear TFEB levels, and because enhanced CRM1 activity is mechanistically linked to senescence in fibroblasts from Hutchinson-Gilford progeria syndrome patients and old healthy individuals; furthermore, the exogenous overexpression of CRM1 induced senescence in normal fibroblasts. In this work, we tested the hypothesis that impaired autophagic flux during brain aging occurs due to CRM1 accumulation in the brain. We found that CRM1 levels and activity increased in the hippocampus and cortex during physiological aging, which resulted in a decrease of nuclear TFEB and STAT3. Consistent with an autophagic flux impairment, we observed accumulation of the autophagic receptor p62/SQSTM1 in neurons of old mice, which correlated with increased neuronal senescence. Using an in vitro model of neuronal senescence, we demonstrate that CRM1 inhibition improved autophagy flux and reduced SA-beta-gal activity by restoring TFEB nuclear localization. Collectively, our data suggest that enhanced CRM1-mediated export of proteins during brain aging perturbs neuronal homeostasis, contributing to autophagy impairment, and neuronal senescence.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.3
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available