Cargando…

Increased Glucose Transport into Neurons Rescues Aβ Toxicity in Drosophila

Glucose hypometabolism is a prominent feature of the brains of patients with Alzheimer’s disease (AD). Disease progression is associated with a reduction in glucose transporters in both neurons and endothelial cells of the blood-brain barrier. However, whether increasing glucose transport into eithe...

Descripción completa

Detalles Bibliográficos
Autores principales: Niccoli, Teresa, Cabecinha, Melissa, Tillmann, Anna, Kerr, Fiona, Wong, Chi T., Cardenes, Dalia, Vincent, Alec J., Bettedi, Lucia, Li, Li, Grönke, Sebastian, Dols, Jacqueline, Partridge, Linda
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Cell Press 2016
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5026704/
https://www.ncbi.nlm.nih.gov/pubmed/27524482
http://dx.doi.org/10.1016/j.cub.2016.07.017
_version_ 1782454145691680768
author Niccoli, Teresa
Cabecinha, Melissa
Tillmann, Anna
Kerr, Fiona
Wong, Chi T.
Cardenes, Dalia
Vincent, Alec J.
Bettedi, Lucia
Li, Li
Grönke, Sebastian
Dols, Jacqueline
Partridge, Linda
author_facet Niccoli, Teresa
Cabecinha, Melissa
Tillmann, Anna
Kerr, Fiona
Wong, Chi T.
Cardenes, Dalia
Vincent, Alec J.
Bettedi, Lucia
Li, Li
Grönke, Sebastian
Dols, Jacqueline
Partridge, Linda
author_sort Niccoli, Teresa
collection PubMed
description Glucose hypometabolism is a prominent feature of the brains of patients with Alzheimer’s disease (AD). Disease progression is associated with a reduction in glucose transporters in both neurons and endothelial cells of the blood-brain barrier. However, whether increasing glucose transport into either of these cell types offers therapeutic potential remains unknown. Using an adult-onset Drosophila model of Aβ (amyloid beta) toxicity, we show that genetic overexpression of a glucose transporter, specifically in neurons, rescues lifespan, behavioral phenotypes, and neuronal morphology. This amelioration of Aβ toxicity is associated with a reduction in the protein levels of the unfolded protein response (UPR) negative master regulator Grp78 and an increase in the UPR. We further demonstrate that genetic downregulation of Grp78 activity also protects against Aβ toxicity, confirming a causal effect of its alteration on AD-related pathology. Metformin, a drug that stimulates glucose uptake in cells, mimicked these effects, with a concomitant reduction in Grp78 levels and rescue of the shortened lifespan and climbing defects of Aβ-expressing flies. Our findings demonstrate a protective effect of increased neuronal uptake of glucose against Aβ toxicity and highlight Grp78 as a novel therapeutic target for the treatment of AD.
format Online
Article
Text
id pubmed-5026704
institution National Center for Biotechnology Information
language English
publishDate 2016
publisher Cell Press
record_format MEDLINE/PubMed
spelling pubmed-50267042016-09-23 Increased Glucose Transport into Neurons Rescues Aβ Toxicity in Drosophila Niccoli, Teresa Cabecinha, Melissa Tillmann, Anna Kerr, Fiona Wong, Chi T. Cardenes, Dalia Vincent, Alec J. Bettedi, Lucia Li, Li Grönke, Sebastian Dols, Jacqueline Partridge, Linda Curr Biol Article Glucose hypometabolism is a prominent feature of the brains of patients with Alzheimer’s disease (AD). Disease progression is associated with a reduction in glucose transporters in both neurons and endothelial cells of the blood-brain barrier. However, whether increasing glucose transport into either of these cell types offers therapeutic potential remains unknown. Using an adult-onset Drosophila model of Aβ (amyloid beta) toxicity, we show that genetic overexpression of a glucose transporter, specifically in neurons, rescues lifespan, behavioral phenotypes, and neuronal morphology. This amelioration of Aβ toxicity is associated with a reduction in the protein levels of the unfolded protein response (UPR) negative master regulator Grp78 and an increase in the UPR. We further demonstrate that genetic downregulation of Grp78 activity also protects against Aβ toxicity, confirming a causal effect of its alteration on AD-related pathology. Metformin, a drug that stimulates glucose uptake in cells, mimicked these effects, with a concomitant reduction in Grp78 levels and rescue of the shortened lifespan and climbing defects of Aβ-expressing flies. Our findings demonstrate a protective effect of increased neuronal uptake of glucose against Aβ toxicity and highlight Grp78 as a novel therapeutic target for the treatment of AD. Cell Press 2016-09-12 /pmc/articles/PMC5026704/ /pubmed/27524482 http://dx.doi.org/10.1016/j.cub.2016.07.017 Text en © 2016 The Author(s) http://creativecommons.org/licenses/by/4.0/ This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
spellingShingle Article
Niccoli, Teresa
Cabecinha, Melissa
Tillmann, Anna
Kerr, Fiona
Wong, Chi T.
Cardenes, Dalia
Vincent, Alec J.
Bettedi, Lucia
Li, Li
Grönke, Sebastian
Dols, Jacqueline
Partridge, Linda
Increased Glucose Transport into Neurons Rescues Aβ Toxicity in Drosophila
title Increased Glucose Transport into Neurons Rescues Aβ Toxicity in Drosophila
title_full Increased Glucose Transport into Neurons Rescues Aβ Toxicity in Drosophila
title_fullStr Increased Glucose Transport into Neurons Rescues Aβ Toxicity in Drosophila
title_full_unstemmed Increased Glucose Transport into Neurons Rescues Aβ Toxicity in Drosophila
title_short Increased Glucose Transport into Neurons Rescues Aβ Toxicity in Drosophila
title_sort increased glucose transport into neurons rescues aβ toxicity in drosophila
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5026704/
https://www.ncbi.nlm.nih.gov/pubmed/27524482
http://dx.doi.org/10.1016/j.cub.2016.07.017
work_keys_str_mv AT niccoliteresa increasedglucosetransportintoneuronsrescuesabtoxicityindrosophila
AT cabecinhamelissa increasedglucosetransportintoneuronsrescuesabtoxicityindrosophila
AT tillmannanna increasedglucosetransportintoneuronsrescuesabtoxicityindrosophila
AT kerrfiona increasedglucosetransportintoneuronsrescuesabtoxicityindrosophila
AT wongchit increasedglucosetransportintoneuronsrescuesabtoxicityindrosophila
AT cardenesdalia increasedglucosetransportintoneuronsrescuesabtoxicityindrosophila
AT vincentalecj increasedglucosetransportintoneuronsrescuesabtoxicityindrosophila
AT bettedilucia increasedglucosetransportintoneuronsrescuesabtoxicityindrosophila
AT lili increasedglucosetransportintoneuronsrescuesabtoxicityindrosophila
AT gronkesebastian increasedglucosetransportintoneuronsrescuesabtoxicityindrosophila
AT dolsjacqueline increasedglucosetransportintoneuronsrescuesabtoxicityindrosophila
AT partridgelinda increasedglucosetransportintoneuronsrescuesabtoxicityindrosophila