Cargando…

Alzheimer’s associated amyloid and tau deposition co-localizes with a homeostatic myelin repair pathway in two mouse models of post-stroke mixed dementia

The goal of this study was to determine the chronic impact of stroke on the manifestation of Alzheimer’s disease (AD) related pathology and behavioral impairments in mice. To accomplish this goal, we used two distinct models. First, we experimentally induced ischemic stroke in aged wildtype (wt) C57...

Descripción completa

Detalles Bibliográficos
Autores principales: Nguyen, Thuy-Vi V., Hayes, Megan, Zbesko, Jacob C., Frye, Jennifer B., Congrove, Nicole R., Belichenko, Nadia P., McKay, Brian S., Longo, Frank M., Doyle, Kristian P.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6154927/
https://www.ncbi.nlm.nih.gov/pubmed/30249297
http://dx.doi.org/10.1186/s40478-018-0603-4
_version_ 1783357789295345664
author Nguyen, Thuy-Vi V.
Hayes, Megan
Zbesko, Jacob C.
Frye, Jennifer B.
Congrove, Nicole R.
Belichenko, Nadia P.
McKay, Brian S.
Longo, Frank M.
Doyle, Kristian P.
author_facet Nguyen, Thuy-Vi V.
Hayes, Megan
Zbesko, Jacob C.
Frye, Jennifer B.
Congrove, Nicole R.
Belichenko, Nadia P.
McKay, Brian S.
Longo, Frank M.
Doyle, Kristian P.
author_sort Nguyen, Thuy-Vi V.
collection PubMed
description The goal of this study was to determine the chronic impact of stroke on the manifestation of Alzheimer’s disease (AD) related pathology and behavioral impairments in mice. To accomplish this goal, we used two distinct models. First, we experimentally induced ischemic stroke in aged wildtype (wt) C57BL/6 mice to determine if stroke leads to the manifestation of AD-associated pathological β-amyloid (Aβ) and tau in aged versus young adult wt mice. Second, we utilized a transgenic (Tg) mouse model of AD (hAPP-SL) to determine if stroke leads to the worsening of pre-existing AD pathology, as well as the development of pathology in brain regions not typically expressed in AD Tg mice. In the wt mice, there was delayed motor recovery and an accelerated development of cognitive deficits in aged mice compared to young adult mice following stroke. This corresponded with increased brain atrophy, increased cholinergic degeneration, and a focal increase of Aβ in areas of axonal degeneration in the ipsilateral hemisphere of the aged animals. By contrast, in the hAPP-SL mice, we found that ischemia induced aggravated behavioral deficits in conjunction with a global increase in Aβ(,) tau, and cholinergic pathology compared to hAPP-SL mice that underwent a sham stroke procedure. With regard to a potential mechanism, in both models, we found that the stroke-induced Aβ and tau deposits co-localized with increased levels of β-secretase 1 (BACE1), along with its substrate, neuregulin 1 (NGR1) type III, both of which are proteins integral for myelin repair. Based on these findings, we propose that the chronic sequelae of stroke may be ratcheting-up a myelin repair pathway, and that the consequent increase in BACE1 could be causing an inadvertent cleavage of its alternative substrate, AβPP, resulting in greater Aβ seeding and pathogenesis.
format Online
Article
Text
id pubmed-6154927
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-61549272018-09-26 Alzheimer’s associated amyloid and tau deposition co-localizes with a homeostatic myelin repair pathway in two mouse models of post-stroke mixed dementia Nguyen, Thuy-Vi V. Hayes, Megan Zbesko, Jacob C. Frye, Jennifer B. Congrove, Nicole R. Belichenko, Nadia P. McKay, Brian S. Longo, Frank M. Doyle, Kristian P. Acta Neuropathol Commun Research The goal of this study was to determine the chronic impact of stroke on the manifestation of Alzheimer’s disease (AD) related pathology and behavioral impairments in mice. To accomplish this goal, we used two distinct models. First, we experimentally induced ischemic stroke in aged wildtype (wt) C57BL/6 mice to determine if stroke leads to the manifestation of AD-associated pathological β-amyloid (Aβ) and tau in aged versus young adult wt mice. Second, we utilized a transgenic (Tg) mouse model of AD (hAPP-SL) to determine if stroke leads to the worsening of pre-existing AD pathology, as well as the development of pathology in brain regions not typically expressed in AD Tg mice. In the wt mice, there was delayed motor recovery and an accelerated development of cognitive deficits in aged mice compared to young adult mice following stroke. This corresponded with increased brain atrophy, increased cholinergic degeneration, and a focal increase of Aβ in areas of axonal degeneration in the ipsilateral hemisphere of the aged animals. By contrast, in the hAPP-SL mice, we found that ischemia induced aggravated behavioral deficits in conjunction with a global increase in Aβ(,) tau, and cholinergic pathology compared to hAPP-SL mice that underwent a sham stroke procedure. With regard to a potential mechanism, in both models, we found that the stroke-induced Aβ and tau deposits co-localized with increased levels of β-secretase 1 (BACE1), along with its substrate, neuregulin 1 (NGR1) type III, both of which are proteins integral for myelin repair. Based on these findings, we propose that the chronic sequelae of stroke may be ratcheting-up a myelin repair pathway, and that the consequent increase in BACE1 could be causing an inadvertent cleavage of its alternative substrate, AβPP, resulting in greater Aβ seeding and pathogenesis. BioMed Central 2018-09-24 /pmc/articles/PMC6154927/ /pubmed/30249297 http://dx.doi.org/10.1186/s40478-018-0603-4 Text en © The Author(s). 2018 Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
spellingShingle Research
Nguyen, Thuy-Vi V.
Hayes, Megan
Zbesko, Jacob C.
Frye, Jennifer B.
Congrove, Nicole R.
Belichenko, Nadia P.
McKay, Brian S.
Longo, Frank M.
Doyle, Kristian P.
Alzheimer’s associated amyloid and tau deposition co-localizes with a homeostatic myelin repair pathway in two mouse models of post-stroke mixed dementia
title Alzheimer’s associated amyloid and tau deposition co-localizes with a homeostatic myelin repair pathway in two mouse models of post-stroke mixed dementia
title_full Alzheimer’s associated amyloid and tau deposition co-localizes with a homeostatic myelin repair pathway in two mouse models of post-stroke mixed dementia
title_fullStr Alzheimer’s associated amyloid and tau deposition co-localizes with a homeostatic myelin repair pathway in two mouse models of post-stroke mixed dementia
title_full_unstemmed Alzheimer’s associated amyloid and tau deposition co-localizes with a homeostatic myelin repair pathway in two mouse models of post-stroke mixed dementia
title_short Alzheimer’s associated amyloid and tau deposition co-localizes with a homeostatic myelin repair pathway in two mouse models of post-stroke mixed dementia
title_sort alzheimer’s associated amyloid and tau deposition co-localizes with a homeostatic myelin repair pathway in two mouse models of post-stroke mixed dementia
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6154927/
https://www.ncbi.nlm.nih.gov/pubmed/30249297
http://dx.doi.org/10.1186/s40478-018-0603-4
work_keys_str_mv AT nguyenthuyviv alzheimersassociatedamyloidandtaudepositioncolocalizeswithahomeostaticmyelinrepairpathwayintwomousemodelsofpoststrokemixeddementia
AT hayesmegan alzheimersassociatedamyloidandtaudepositioncolocalizeswithahomeostaticmyelinrepairpathwayintwomousemodelsofpoststrokemixeddementia
AT zbeskojacobc alzheimersassociatedamyloidandtaudepositioncolocalizeswithahomeostaticmyelinrepairpathwayintwomousemodelsofpoststrokemixeddementia
AT fryejenniferb alzheimersassociatedamyloidandtaudepositioncolocalizeswithahomeostaticmyelinrepairpathwayintwomousemodelsofpoststrokemixeddementia
AT congrovenicoler alzheimersassociatedamyloidandtaudepositioncolocalizeswithahomeostaticmyelinrepairpathwayintwomousemodelsofpoststrokemixeddementia
AT belichenkonadiap alzheimersassociatedamyloidandtaudepositioncolocalizeswithahomeostaticmyelinrepairpathwayintwomousemodelsofpoststrokemixeddementia
AT mckaybrians alzheimersassociatedamyloidandtaudepositioncolocalizeswithahomeostaticmyelinrepairpathwayintwomousemodelsofpoststrokemixeddementia
AT longofrankm alzheimersassociatedamyloidandtaudepositioncolocalizeswithahomeostaticmyelinrepairpathwayintwomousemodelsofpoststrokemixeddementia
AT doylekristianp alzheimersassociatedamyloidandtaudepositioncolocalizeswithahomeostaticmyelinrepairpathwayintwomousemodelsofpoststrokemixeddementia