Reduced HGF/MET Signaling May Contribute to the Synaptic Pathology in an Alzheimer's Disease Mouse Model

Alzheimer's disease (AD) is a neurodegenerative disorder strongly associates with aging. While amyloid plagues and neurofibrillary tangles are pathological hallmarks of AD, recent evidence suggests synaptic dysfunction and physical loss may be the key mechanisms that determine the clinical synd...

Full description

Bibliographic Details
Main Authors: Jing Wei, Xiaokuang Ma, Antoine Nehme, Yuehua Cui, Le Zhang, Shenfeng Qiu
Format: Article
Language:English
Published: Frontiers Media S.A. 2022-07-01
Series:Frontiers in Aging Neuroscience
Subjects:
Online Access:https://www.frontiersin.org/articles/10.3389/fnagi.2022.954266/full
_version_ 1811297155042770944
author Jing Wei
Xiaokuang Ma
Antoine Nehme
Yuehua Cui
Le Zhang
Shenfeng Qiu
author_facet Jing Wei
Xiaokuang Ma
Antoine Nehme
Yuehua Cui
Le Zhang
Shenfeng Qiu
author_sort Jing Wei
collection DOAJ
description Alzheimer's disease (AD) is a neurodegenerative disorder strongly associates with aging. While amyloid plagues and neurofibrillary tangles are pathological hallmarks of AD, recent evidence suggests synaptic dysfunction and physical loss may be the key mechanisms that determine the clinical syndrome and dementia onset. Currently, no effective therapy prevents neuropathological changes and cognitive decline. Neurotrophic factors and their receptors represent novel therapeutic targets to treat AD and dementia. Recent clinical literature revealed that MET receptor tyrosine kinase protein is reduced in AD patient's brain. Activation of MET by its ligand hepatocyte growth factor (HGF) initiates pleiotropic signaling in the developing brain that promotes neurogenesis, survival, synaptogenesis, and plasticity. We hypothesize that if reduced MET signaling plays a role in AD pathogenesis, this might be reflected in the AD mouse models and as such provides opportunities for mechanistic studies on the role of HGF/MET in AD. Examining the 5XFAD mouse model revealed that MET protein exhibits age-dependent progressive reduction prior to overt neuronal pathology, which cannot be explained by indiscriminate loss of total synaptic proteins. In addition, genetic ablation of MET protein in cortical excitatory neurons exacerbates amyloid-related neuropathology in 5XFAD mice. We further found that HGF enhances prefrontal layer 5 neuron synaptic plasticity measured by long-term potentiation (LTP). However, the degree of LTP enhancement is significantly reduced in 5XFAD mice brain slices. Taken together, our study revealed that early reduction of HGF/MET signaling may contribute to the synaptic pathology observed in AD.
first_indexed 2024-04-13T05:59:50Z
format Article
id doaj.art-bb3de869a8e3437aa7d158b8d208b2f9
institution Directory Open Access Journal
issn 1663-4365
language English
last_indexed 2024-04-13T05:59:50Z
publishDate 2022-07-01
publisher Frontiers Media S.A.
record_format Article
series Frontiers in Aging Neuroscience
spelling doaj.art-bb3de869a8e3437aa7d158b8d208b2f92022-12-22T02:59:29ZengFrontiers Media S.A.Frontiers in Aging Neuroscience1663-43652022-07-011410.3389/fnagi.2022.954266954266Reduced HGF/MET Signaling May Contribute to the Synaptic Pathology in an Alzheimer's Disease Mouse ModelJing WeiXiaokuang MaAntoine NehmeYuehua CuiLe ZhangShenfeng QiuAlzheimer's disease (AD) is a neurodegenerative disorder strongly associates with aging. While amyloid plagues and neurofibrillary tangles are pathological hallmarks of AD, recent evidence suggests synaptic dysfunction and physical loss may be the key mechanisms that determine the clinical syndrome and dementia onset. Currently, no effective therapy prevents neuropathological changes and cognitive decline. Neurotrophic factors and their receptors represent novel therapeutic targets to treat AD and dementia. Recent clinical literature revealed that MET receptor tyrosine kinase protein is reduced in AD patient's brain. Activation of MET by its ligand hepatocyte growth factor (HGF) initiates pleiotropic signaling in the developing brain that promotes neurogenesis, survival, synaptogenesis, and plasticity. We hypothesize that if reduced MET signaling plays a role in AD pathogenesis, this might be reflected in the AD mouse models and as such provides opportunities for mechanistic studies on the role of HGF/MET in AD. Examining the 5XFAD mouse model revealed that MET protein exhibits age-dependent progressive reduction prior to overt neuronal pathology, which cannot be explained by indiscriminate loss of total synaptic proteins. In addition, genetic ablation of MET protein in cortical excitatory neurons exacerbates amyloid-related neuropathology in 5XFAD mice. We further found that HGF enhances prefrontal layer 5 neuron synaptic plasticity measured by long-term potentiation (LTP). However, the degree of LTP enhancement is significantly reduced in 5XFAD mice brain slices. Taken together, our study revealed that early reduction of HGF/MET signaling may contribute to the synaptic pathology observed in AD.https://www.frontiersin.org/articles/10.3389/fnagi.2022.954266/fullAlzheimer's diseaseMET receptor tyrosine kinasehepatocyte growth factorsynaptic plasticityregeneration
spellingShingle Jing Wei
Xiaokuang Ma
Antoine Nehme
Yuehua Cui
Le Zhang
Shenfeng Qiu
Reduced HGF/MET Signaling May Contribute to the Synaptic Pathology in an Alzheimer's Disease Mouse Model
Frontiers in Aging Neuroscience
Alzheimer's disease
MET receptor tyrosine kinase
hepatocyte growth factor
synaptic plasticity
regeneration
title Reduced HGF/MET Signaling May Contribute to the Synaptic Pathology in an Alzheimer's Disease Mouse Model
title_full Reduced HGF/MET Signaling May Contribute to the Synaptic Pathology in an Alzheimer's Disease Mouse Model
title_fullStr Reduced HGF/MET Signaling May Contribute to the Synaptic Pathology in an Alzheimer's Disease Mouse Model
title_full_unstemmed Reduced HGF/MET Signaling May Contribute to the Synaptic Pathology in an Alzheimer's Disease Mouse Model
title_short Reduced HGF/MET Signaling May Contribute to the Synaptic Pathology in an Alzheimer's Disease Mouse Model
title_sort reduced hgf met signaling may contribute to the synaptic pathology in an alzheimer s disease mouse model
topic Alzheimer's disease
MET receptor tyrosine kinase
hepatocyte growth factor
synaptic plasticity
regeneration
url https://www.frontiersin.org/articles/10.3389/fnagi.2022.954266/full
work_keys_str_mv AT jingwei reducedhgfmetsignalingmaycontributetothesynapticpathologyinanalzheimersdiseasemousemodel
AT xiaokuangma reducedhgfmetsignalingmaycontributetothesynapticpathologyinanalzheimersdiseasemousemodel
AT antoinenehme reducedhgfmetsignalingmaycontributetothesynapticpathologyinanalzheimersdiseasemousemodel
AT yuehuacui reducedhgfmetsignalingmaycontributetothesynapticpathologyinanalzheimersdiseasemousemodel
AT lezhang reducedhgfmetsignalingmaycontributetothesynapticpathologyinanalzheimersdiseasemousemodel
AT shenfengqiu reducedhgfmetsignalingmaycontributetothesynapticpathologyinanalzheimersdiseasemousemodel