English
 
Help Privacy Policy Disclaimer
  Advanced SearchBrowse

Item

ITEM ACTIONSEXPORT

Released

Journal Article

RTP801/REDD1 contributes to neuroinflammation severity and memory impairments in Alzheimer's disease

MPS-Authors

Anglada-Huguet,  Marta
Neuronal Cytoskeleton and Alzheimer's Disease, Cooperations, Center of Advanced European Studies and Research (caesar), Max Planck Society;
External Organizations;

/persons/resource/persons128150

Mandelkow,  Eva-Maria
Neuronal Cytoskeleton and Alzheimer's Disease, Cooperations, Center of Advanced European Studies and Research (caesar), Max Planck Society;
External Organizations;

External Resource
Fulltext (restricted access)
There are currently no full texts shared for your IP range.
Fulltext (public)
There are no public fulltexts stored in PuRe
Supplementary Material (public)
There is no public supplementary material available
Citation

Perez-Sisques, L., Sancho-Balsells, A., Solana-Balaguer, J., Campoy-Campos, G., Vives-Isern, M., Soler-Palazon, F., et al. (2021). RTP801/REDD1 contributes to neuroinflammation severity and memory impairments in Alzheimer's disease. Cell Death and Disease, 12: 616. doi:10.1038/s41419-021-03899-y.


Cite as: https://hdl.handle.net/21.11116/0000-0008-D5AC-6
Abstract
RTP801/REDD1 is a stress-regulated protein whose upregulation is necessary and sufficient to trigger neuronal death. Its
downregulation in Parkinson’s and Huntington’s disease models ameliorates the pathological phenotypes. In the context of
Alzheimer’s disease (AD), the coding gene for RTP801, DDIT4, is responsive to Aβ and modulates its cytotoxicity in vitro. Also,
RTP801 mRNA levels are increased in AD patients’ lymphocytes. However, the involvement of RTP801 in the pathophysiology of AD
has not been yet tested. Here, we demonstrate that RTP801 levels are increased in postmortem hippocampal samples from AD
patients. Interestingly, RTP801 protein levels correlated with both Braak and Thal stages of the disease and with GFAP expression.
RTP801 levels are also upregulated in hippocampal synaptosomal fractions obtained from murine 5xFAD and rTg4510 mice models
of the disease. A local RTP801 knockdown in the 5xFAD hippocampal neurons with shRNA-containing AAV particles ameliorates
cognitive deficits in 7-month-old animals. Upon RTP801 silencing in the 5xFAD mice, no major changes were detected in
hippocampal synaptic markers or spine density. Importantly, we found an unanticipated recovery of several gliosis hallmarks and
inflammasome key proteins upon neuronal RTP801 downregulation in the 5xFAD mice. Altogether our results suggest that RTP801
could be a potential future target for theranostic studies since it could be a biomarker of neuroinflammation and neurotoxicity
severity of the disease and, at the same time, a promising therapeutic target in the treatment of AD.