Cargando…

The Physiological Link between Metabolic Rate Depression and Tau Phosphorylation in Mammalian Hibernation

Abnormal phosphorylation and aggregation of tau protein are hallmarks of a variety of neurological disorders, including Alzheimer's disease (AD). Increased tau phosphorylation is assumed to represent an early event in pathogenesis and a pivotal aspect for aggregation and formation of neurofibri...

Descripción completa

Detalles Bibliográficos
Autores principales: Stieler, Jens T., Bullmann, Torsten, Kohl, Franziska, Tøien, Øivind, Brückner, Martina K., Härtig, Wolfgang, Barnes, Brian M., Arendt, Thomas
Formato: Texto
Lenguaje:English
Publicado: Public Library of Science 2011
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3022585/
https://www.ncbi.nlm.nih.gov/pubmed/21267079
http://dx.doi.org/10.1371/journal.pone.0014530
_version_ 1782196524226183168
author Stieler, Jens T.
Bullmann, Torsten
Kohl, Franziska
Tøien, Øivind
Brückner, Martina K.
Härtig, Wolfgang
Barnes, Brian M.
Arendt, Thomas
author_facet Stieler, Jens T.
Bullmann, Torsten
Kohl, Franziska
Tøien, Øivind
Brückner, Martina K.
Härtig, Wolfgang
Barnes, Brian M.
Arendt, Thomas
author_sort Stieler, Jens T.
collection PubMed
description Abnormal phosphorylation and aggregation of tau protein are hallmarks of a variety of neurological disorders, including Alzheimer's disease (AD). Increased tau phosphorylation is assumed to represent an early event in pathogenesis and a pivotal aspect for aggregation and formation of neurofibrillary tangles. However, the regulation of tau phosphorylation in vivo and the causes for its increased stage of phosphorylation in AD are still not well understood, a fact that is primarily based on the lack of adequate animal models. Recently we described the reversible formation of highly phosphorylated tau protein in hibernating European ground squirrels. Hence, mammalian hibernation represents a model system very well suited to study molecular mechanisms of both tau phosphorylation and dephosphorylation under in vivo physiological conditions. Here, we analysed the extent and kinetics of hibernation-state dependent tau phosphorylation in various brain regions of three species of hibernating mammals: arctic ground squirrels, Syrian hamsters and black bears. Overall, tau protein was highly phosphorylated in torpor states and phosphorylation levels decreased after arousal in all species. Differences between brain regions, hibernation-states and phosphosites were observed with respect to degree and kinetics of tau phosphorylation. Furthermore, we tested the phosphate net turnover of tau protein to analyse potential alterations in kinase and/or phosphatase activities during hibernation. Our results demonstrate that the hibernation-state dependent phosphorylation of tau protein is specifically regulated but involves, in addition, passive, temperature driven regulatory mechanisms. By determining the activity-state profile for key enzymes of tau phosphorylation we could identify kinases potentially involved in the differentially regulated, reversible tau phosphorylation that occurs during hibernation. We show that in black bears hibernation is associated with conformational changes of highly phosphorylated tau protein that are typically related to neuropathological alterations. The particular hibernation characteristics of black bears with a continuous torpor period and an only slightly decreased body temperature, therefore, potentially reflects the limitations of this adaptive reaction pattern and, thus, might indicate a transitional state of a physiological process.
format Text
id pubmed-3022585
institution National Center for Biotechnology Information
language English
publishDate 2011
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-30225852011-01-25 The Physiological Link between Metabolic Rate Depression and Tau Phosphorylation in Mammalian Hibernation Stieler, Jens T. Bullmann, Torsten Kohl, Franziska Tøien, Øivind Brückner, Martina K. Härtig, Wolfgang Barnes, Brian M. Arendt, Thomas PLoS One Research Article Abnormal phosphorylation and aggregation of tau protein are hallmarks of a variety of neurological disorders, including Alzheimer's disease (AD). Increased tau phosphorylation is assumed to represent an early event in pathogenesis and a pivotal aspect for aggregation and formation of neurofibrillary tangles. However, the regulation of tau phosphorylation in vivo and the causes for its increased stage of phosphorylation in AD are still not well understood, a fact that is primarily based on the lack of adequate animal models. Recently we described the reversible formation of highly phosphorylated tau protein in hibernating European ground squirrels. Hence, mammalian hibernation represents a model system very well suited to study molecular mechanisms of both tau phosphorylation and dephosphorylation under in vivo physiological conditions. Here, we analysed the extent and kinetics of hibernation-state dependent tau phosphorylation in various brain regions of three species of hibernating mammals: arctic ground squirrels, Syrian hamsters and black bears. Overall, tau protein was highly phosphorylated in torpor states and phosphorylation levels decreased after arousal in all species. Differences between brain regions, hibernation-states and phosphosites were observed with respect to degree and kinetics of tau phosphorylation. Furthermore, we tested the phosphate net turnover of tau protein to analyse potential alterations in kinase and/or phosphatase activities during hibernation. Our results demonstrate that the hibernation-state dependent phosphorylation of tau protein is specifically regulated but involves, in addition, passive, temperature driven regulatory mechanisms. By determining the activity-state profile for key enzymes of tau phosphorylation we could identify kinases potentially involved in the differentially regulated, reversible tau phosphorylation that occurs during hibernation. We show that in black bears hibernation is associated with conformational changes of highly phosphorylated tau protein that are typically related to neuropathological alterations. The particular hibernation characteristics of black bears with a continuous torpor period and an only slightly decreased body temperature, therefore, potentially reflects the limitations of this adaptive reaction pattern and, thus, might indicate a transitional state of a physiological process. Public Library of Science 2011-01-18 /pmc/articles/PMC3022585/ /pubmed/21267079 http://dx.doi.org/10.1371/journal.pone.0014530 Text en Stieler et al. http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are properly credited.
spellingShingle Research Article
Stieler, Jens T.
Bullmann, Torsten
Kohl, Franziska
Tøien, Øivind
Brückner, Martina K.
Härtig, Wolfgang
Barnes, Brian M.
Arendt, Thomas
The Physiological Link between Metabolic Rate Depression and Tau Phosphorylation in Mammalian Hibernation
title The Physiological Link between Metabolic Rate Depression and Tau Phosphorylation in Mammalian Hibernation
title_full The Physiological Link between Metabolic Rate Depression and Tau Phosphorylation in Mammalian Hibernation
title_fullStr The Physiological Link between Metabolic Rate Depression and Tau Phosphorylation in Mammalian Hibernation
title_full_unstemmed The Physiological Link between Metabolic Rate Depression and Tau Phosphorylation in Mammalian Hibernation
title_short The Physiological Link between Metabolic Rate Depression and Tau Phosphorylation in Mammalian Hibernation
title_sort physiological link between metabolic rate depression and tau phosphorylation in mammalian hibernation
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3022585/
https://www.ncbi.nlm.nih.gov/pubmed/21267079
http://dx.doi.org/10.1371/journal.pone.0014530
work_keys_str_mv AT stielerjenst thephysiologicallinkbetweenmetabolicratedepressionandtauphosphorylationinmammalianhibernation
AT bullmanntorsten thephysiologicallinkbetweenmetabolicratedepressionandtauphosphorylationinmammalianhibernation
AT kohlfranziska thephysiologicallinkbetweenmetabolicratedepressionandtauphosphorylationinmammalianhibernation
AT tøienøivind thephysiologicallinkbetweenmetabolicratedepressionandtauphosphorylationinmammalianhibernation
AT brucknermartinak thephysiologicallinkbetweenmetabolicratedepressionandtauphosphorylationinmammalianhibernation
AT hartigwolfgang thephysiologicallinkbetweenmetabolicratedepressionandtauphosphorylationinmammalianhibernation
AT barnesbrianm thephysiologicallinkbetweenmetabolicratedepressionandtauphosphorylationinmammalianhibernation
AT arendtthomas thephysiologicallinkbetweenmetabolicratedepressionandtauphosphorylationinmammalianhibernation
AT stielerjenst physiologicallinkbetweenmetabolicratedepressionandtauphosphorylationinmammalianhibernation
AT bullmanntorsten physiologicallinkbetweenmetabolicratedepressionandtauphosphorylationinmammalianhibernation
AT kohlfranziska physiologicallinkbetweenmetabolicratedepressionandtauphosphorylationinmammalianhibernation
AT tøienøivind physiologicallinkbetweenmetabolicratedepressionandtauphosphorylationinmammalianhibernation
AT brucknermartinak physiologicallinkbetweenmetabolicratedepressionandtauphosphorylationinmammalianhibernation
AT hartigwolfgang physiologicallinkbetweenmetabolicratedepressionandtauphosphorylationinmammalianhibernation
AT barnesbrianm physiologicallinkbetweenmetabolicratedepressionandtauphosphorylationinmammalianhibernation
AT arendtthomas physiologicallinkbetweenmetabolicratedepressionandtauphosphorylationinmammalianhibernation