Cargando…

Amyloid β induces cardiac dysfunction and neuro-signaling impairment in the heart of an Alzheimer’s disease model

AIMS: Alzheimer’s disease (AD) is a complex neurodegenerative disorder characterized by cerebral amyloid β (Aβ) deposition and tau pathology. The AD-mediated degeneration of the brain neuro-signaling pathways, together with a potential peripheral amyloid accumulation, may also result in the derangem...

Descripción completa

Detalles Bibliográficos
Autores principales: Elia, Andrea, Parodi-Rullan, Rebecca, Vazquez-Torres, Rafael, Carey, Ashley, Javadov, Sabzali, Fossati, Silvia
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Cold Spring Harbor Laboratory 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10369880/
https://www.ncbi.nlm.nih.gov/pubmed/37502936
http://dx.doi.org/10.1101/2023.07.11.548558
_version_ 1785077854371840000
author Elia, Andrea
Parodi-Rullan, Rebecca
Vazquez-Torres, Rafael
Carey, Ashley
Javadov, Sabzali
Fossati, Silvia
author_facet Elia, Andrea
Parodi-Rullan, Rebecca
Vazquez-Torres, Rafael
Carey, Ashley
Javadov, Sabzali
Fossati, Silvia
author_sort Elia, Andrea
collection PubMed
description AIMS: Alzheimer’s disease (AD) is a complex neurodegenerative disorder characterized by cerebral amyloid β (Aβ) deposition and tau pathology. The AD-mediated degeneration of the brain neuro-signaling pathways, together with a potential peripheral amyloid accumulation, may also result in the derangement of the peripheral nervous system, culminating in detrimental effects on other organs, including the heart. However, whether and how AD pathology modulates cardiac function, neurotrophins, innervation, and amyloidosis is still unknown. Here, we report for the first time that cardiac remodeling, amyloid deposition, and neuro-signaling dysregulation occur in the heart of Tg2576 mice, a widely used model of AD and cerebral amyloidosis. METHODS AD RESULTS: Echocardiographic analysis showed significant deterioration of left ventricle function, evidenced by a decline of both ejection fraction and fraction shortening percentage in 12-month-old Tg2576 mice compared to age-matched WT littermates. Tg2576 mice hearts exhibited an accumulation of amyloid aggregates, including Aβ, an increase in interstitial fibrosis and severe cardiac nervous system dysfunction. The transgenic mice also showed a significant decrease in cardiac nerve fiber density, including both adrenergic and regenerating nerve endings. This myocardial denervation was accompanied by a robust reduction in NGF and BDNF protein expression as well as GAP-43 expression (regenerating fibers) in both the brain and heart of Tg2576 mice. Accordingly, cardiomyocytes and neuronal cells challenged with Aβ oligomers showed significant downregulation of BDNF and GAP-43, indicating a causal effect of Aβ on the loss of cardiac neurotrophic function. CONCLUSIONS: Overall, this study uncovers possible harmful effects of AD on the heart, revealing cardiac degeneration induced by Aβ through fibrosis and neuro-signaling pathway deregulation for the first time in Tg2576 mice. Our data suggest that AD pathology can cause deleterious effects on the heart, and the peripheral neurotrophic pathway may represent a potential therapeutic target to limit these effects.
format Online
Article
Text
id pubmed-10369880
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Cold Spring Harbor Laboratory
record_format MEDLINE/PubMed
spelling pubmed-103698802023-07-27 Amyloid β induces cardiac dysfunction and neuro-signaling impairment in the heart of an Alzheimer’s disease model Elia, Andrea Parodi-Rullan, Rebecca Vazquez-Torres, Rafael Carey, Ashley Javadov, Sabzali Fossati, Silvia bioRxiv Article AIMS: Alzheimer’s disease (AD) is a complex neurodegenerative disorder characterized by cerebral amyloid β (Aβ) deposition and tau pathology. The AD-mediated degeneration of the brain neuro-signaling pathways, together with a potential peripheral amyloid accumulation, may also result in the derangement of the peripheral nervous system, culminating in detrimental effects on other organs, including the heart. However, whether and how AD pathology modulates cardiac function, neurotrophins, innervation, and amyloidosis is still unknown. Here, we report for the first time that cardiac remodeling, amyloid deposition, and neuro-signaling dysregulation occur in the heart of Tg2576 mice, a widely used model of AD and cerebral amyloidosis. METHODS AD RESULTS: Echocardiographic analysis showed significant deterioration of left ventricle function, evidenced by a decline of both ejection fraction and fraction shortening percentage in 12-month-old Tg2576 mice compared to age-matched WT littermates. Tg2576 mice hearts exhibited an accumulation of amyloid aggregates, including Aβ, an increase in interstitial fibrosis and severe cardiac nervous system dysfunction. The transgenic mice also showed a significant decrease in cardiac nerve fiber density, including both adrenergic and regenerating nerve endings. This myocardial denervation was accompanied by a robust reduction in NGF and BDNF protein expression as well as GAP-43 expression (regenerating fibers) in both the brain and heart of Tg2576 mice. Accordingly, cardiomyocytes and neuronal cells challenged with Aβ oligomers showed significant downregulation of BDNF and GAP-43, indicating a causal effect of Aβ on the loss of cardiac neurotrophic function. CONCLUSIONS: Overall, this study uncovers possible harmful effects of AD on the heart, revealing cardiac degeneration induced by Aβ through fibrosis and neuro-signaling pathway deregulation for the first time in Tg2576 mice. Our data suggest that AD pathology can cause deleterious effects on the heart, and the peripheral neurotrophic pathway may represent a potential therapeutic target to limit these effects. Cold Spring Harbor Laboratory 2023-07-11 /pmc/articles/PMC10369880/ /pubmed/37502936 http://dx.doi.org/10.1101/2023.07.11.548558 Text en https://creativecommons.org/licenses/by-nc-nd/4.0/This work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License (https://creativecommons.org/licenses/by-nc-nd/4.0/) , which allows reusers to copy and distribute the material in any medium or format in unadapted form only, for noncommercial purposes only, and only so long as attribution is given to the creator.
spellingShingle Article
Elia, Andrea
Parodi-Rullan, Rebecca
Vazquez-Torres, Rafael
Carey, Ashley
Javadov, Sabzali
Fossati, Silvia
Amyloid β induces cardiac dysfunction and neuro-signaling impairment in the heart of an Alzheimer’s disease model
title Amyloid β induces cardiac dysfunction and neuro-signaling impairment in the heart of an Alzheimer’s disease model
title_full Amyloid β induces cardiac dysfunction and neuro-signaling impairment in the heart of an Alzheimer’s disease model
title_fullStr Amyloid β induces cardiac dysfunction and neuro-signaling impairment in the heart of an Alzheimer’s disease model
title_full_unstemmed Amyloid β induces cardiac dysfunction and neuro-signaling impairment in the heart of an Alzheimer’s disease model
title_short Amyloid β induces cardiac dysfunction and neuro-signaling impairment in the heart of an Alzheimer’s disease model
title_sort amyloid β induces cardiac dysfunction and neuro-signaling impairment in the heart of an alzheimer’s disease model
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10369880/
https://www.ncbi.nlm.nih.gov/pubmed/37502936
http://dx.doi.org/10.1101/2023.07.11.548558
work_keys_str_mv AT eliaandrea amyloidbinducescardiacdysfunctionandneurosignalingimpairmentintheheartofanalzheimersdiseasemodel
AT parodirullanrebecca amyloidbinducescardiacdysfunctionandneurosignalingimpairmentintheheartofanalzheimersdiseasemodel
AT vazqueztorresrafael amyloidbinducescardiacdysfunctionandneurosignalingimpairmentintheheartofanalzheimersdiseasemodel
AT careyashley amyloidbinducescardiacdysfunctionandneurosignalingimpairmentintheheartofanalzheimersdiseasemodel
AT javadovsabzali amyloidbinducescardiacdysfunctionandneurosignalingimpairmentintheheartofanalzheimersdiseasemodel
AT fossatisilvia amyloidbinducescardiacdysfunctionandneurosignalingimpairmentintheheartofanalzheimersdiseasemodel