The Impact of Muscarinic Antagonism on Psychosis-Relevant Behaviors and Striatal [<sup>11</sup>C] Raclopride Binding in Tau Mouse Models of Alzheimer’s Disease

Psychosis that occurs over the course of Alzheimer’s disease (AD) is associated with increased caregiver burden and a more rapid cognitive and functional decline. To find new treatment targets, studies modeling psychotic conditions traditionally employ agents known to induce psychosis, utilizing out...

Full description

Bibliographic Details
Main Authors: Heidy Jimenez, Joseph Carrion, Leslie Adrien, Adam Wolin, John Eun, Ezra Cinamon, Eric H. Chang, Peter Davies, An Vo, Jeremy Koppel
Format: Article
Language:English
Published: MDPI AG 2023-07-01
Series:Biomedicines
Subjects:
Online Access:https://www.mdpi.com/2227-9059/11/8/2091
_version_ 1797585486759329792
author Heidy Jimenez
Joseph Carrion
Leslie Adrien
Adam Wolin
John Eun
Ezra Cinamon
Eric H. Chang
Peter Davies
An Vo
Jeremy Koppel
author_facet Heidy Jimenez
Joseph Carrion
Leslie Adrien
Adam Wolin
John Eun
Ezra Cinamon
Eric H. Chang
Peter Davies
An Vo
Jeremy Koppel
author_sort Heidy Jimenez
collection DOAJ
description Psychosis that occurs over the course of Alzheimer’s disease (AD) is associated with increased caregiver burden and a more rapid cognitive and functional decline. To find new treatment targets, studies modeling psychotic conditions traditionally employ agents known to induce psychosis, utilizing outcomes with cross-species relevance, such as locomotive activity and sensorimotor gating, in rodents. In AD, increased burdens of tau pathology (a diagnostic hallmark of the disease) and treatment with anticholinergic medications have, separately, been reported to increase the risk of psychosis. Recent evidence suggests that muscarinic antagonists may increase extracellular tau. Preclinical studies in AD models have not previously utilized muscarinic cholinergic antagonists as psychotomimetic agents. In this report, we utilize a human–mutant–tau model (P301L/COMTKO) and an over-expressed non-mutant human tau model (htau) in order to compare the impact of antimuscarinic (scopolamine 10 mg/kg/day) treatment with dopaminergic (reboxetine 20 mg/kg/day) treatment, for 7 days, on locomotion and sensorimotor gating. Scopolamine increased spontaneous locomotion, while reboxetine reduced it; neither treatment impacted sensorimotor gating. In the P301L/COMTKO, scopolamine treatment was associated with decreased muscarinic M4 receptor expression, as quantified with RNA-seq, as well as increased dopamine receptor D2 signaling, as estimated with Micro-PET [<sup>11</sup>C] raclopride binding. Scopolamine also increased soluble tau in the striatum, an effect that partially mediated the observed increases in locomotion. Studies of muscarinic agonists in preclinical tau models are warranted to determine the impact of treatment—on both tau and behavior—that may have relevance to AD and other tauopathies.
first_indexed 2024-03-11T00:06:48Z
format Article
id doaj.art-b50c03d26a1b4862ac7cfaf05a1423ef
institution Directory Open Access Journal
issn 2227-9059
language English
last_indexed 2024-03-11T00:06:48Z
publishDate 2023-07-01
publisher MDPI AG
record_format Article
series Biomedicines
spelling doaj.art-b50c03d26a1b4862ac7cfaf05a1423ef2023-11-19T00:19:19ZengMDPI AGBiomedicines2227-90592023-07-01118209110.3390/biomedicines11082091The Impact of Muscarinic Antagonism on Psychosis-Relevant Behaviors and Striatal [<sup>11</sup>C] Raclopride Binding in Tau Mouse Models of Alzheimer’s DiseaseHeidy Jimenez0Joseph Carrion1Leslie Adrien2Adam Wolin3John Eun4Ezra Cinamon5Eric H. Chang6Peter Davies7An Vo8Jeremy Koppel9The Litwin-Zucker Research Center for the Study of Alzheimer’s Disease, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USAThe Litwin-Zucker Research Center for the Study of Alzheimer’s Disease, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USAThe Litwin-Zucker Research Center for the Study of Alzheimer’s Disease, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USAThe Litwin-Zucker Research Center for the Study of Alzheimer’s Disease, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USAThe Litwin-Zucker Research Center for the Study of Alzheimer’s Disease, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USADepartment of Biochemistry, Queens College, Flushing, NY 11355, USAThe Litwin-Zucker Research Center for the Study of Alzheimer’s Disease, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USAThe Litwin-Zucker Research Center for the Study of Alzheimer’s Disease, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USAThe Litwin-Zucker Research Center for the Study of Alzheimer’s Disease, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USAThe Litwin-Zucker Research Center for the Study of Alzheimer’s Disease, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USAPsychosis that occurs over the course of Alzheimer’s disease (AD) is associated with increased caregiver burden and a more rapid cognitive and functional decline. To find new treatment targets, studies modeling psychotic conditions traditionally employ agents known to induce psychosis, utilizing outcomes with cross-species relevance, such as locomotive activity and sensorimotor gating, in rodents. In AD, increased burdens of tau pathology (a diagnostic hallmark of the disease) and treatment with anticholinergic medications have, separately, been reported to increase the risk of psychosis. Recent evidence suggests that muscarinic antagonists may increase extracellular tau. Preclinical studies in AD models have not previously utilized muscarinic cholinergic antagonists as psychotomimetic agents. In this report, we utilize a human–mutant–tau model (P301L/COMTKO) and an over-expressed non-mutant human tau model (htau) in order to compare the impact of antimuscarinic (scopolamine 10 mg/kg/day) treatment with dopaminergic (reboxetine 20 mg/kg/day) treatment, for 7 days, on locomotion and sensorimotor gating. Scopolamine increased spontaneous locomotion, while reboxetine reduced it; neither treatment impacted sensorimotor gating. In the P301L/COMTKO, scopolamine treatment was associated with decreased muscarinic M4 receptor expression, as quantified with RNA-seq, as well as increased dopamine receptor D2 signaling, as estimated with Micro-PET [<sup>11</sup>C] raclopride binding. Scopolamine also increased soluble tau in the striatum, an effect that partially mediated the observed increases in locomotion. Studies of muscarinic agonists in preclinical tau models are warranted to determine the impact of treatment—on both tau and behavior—that may have relevance to AD and other tauopathies.https://www.mdpi.com/2227-9059/11/8/2091taupsychosisscopolaminelocomotionAlzheimer’s diseasehopping
spellingShingle Heidy Jimenez
Joseph Carrion
Leslie Adrien
Adam Wolin
John Eun
Ezra Cinamon
Eric H. Chang
Peter Davies
An Vo
Jeremy Koppel
The Impact of Muscarinic Antagonism on Psychosis-Relevant Behaviors and Striatal [<sup>11</sup>C] Raclopride Binding in Tau Mouse Models of Alzheimer’s Disease
Biomedicines
tau
psychosis
scopolamine
locomotion
Alzheimer’s disease
hopping
title The Impact of Muscarinic Antagonism on Psychosis-Relevant Behaviors and Striatal [<sup>11</sup>C] Raclopride Binding in Tau Mouse Models of Alzheimer’s Disease
title_full The Impact of Muscarinic Antagonism on Psychosis-Relevant Behaviors and Striatal [<sup>11</sup>C] Raclopride Binding in Tau Mouse Models of Alzheimer’s Disease
title_fullStr The Impact of Muscarinic Antagonism on Psychosis-Relevant Behaviors and Striatal [<sup>11</sup>C] Raclopride Binding in Tau Mouse Models of Alzheimer’s Disease
title_full_unstemmed The Impact of Muscarinic Antagonism on Psychosis-Relevant Behaviors and Striatal [<sup>11</sup>C] Raclopride Binding in Tau Mouse Models of Alzheimer’s Disease
title_short The Impact of Muscarinic Antagonism on Psychosis-Relevant Behaviors and Striatal [<sup>11</sup>C] Raclopride Binding in Tau Mouse Models of Alzheimer’s Disease
title_sort impact of muscarinic antagonism on psychosis relevant behaviors and striatal sup 11 sup c raclopride binding in tau mouse models of alzheimer s disease
topic tau
psychosis
scopolamine
locomotion
Alzheimer’s disease
hopping
url https://www.mdpi.com/2227-9059/11/8/2091
work_keys_str_mv AT heidyjimenez theimpactofmuscarinicantagonismonpsychosisrelevantbehaviorsandstriatalsup11supcraclopridebindingintaumousemodelsofalzheimersdisease
AT josephcarrion theimpactofmuscarinicantagonismonpsychosisrelevantbehaviorsandstriatalsup11supcraclopridebindingintaumousemodelsofalzheimersdisease
AT leslieadrien theimpactofmuscarinicantagonismonpsychosisrelevantbehaviorsandstriatalsup11supcraclopridebindingintaumousemodelsofalzheimersdisease
AT adamwolin theimpactofmuscarinicantagonismonpsychosisrelevantbehaviorsandstriatalsup11supcraclopridebindingintaumousemodelsofalzheimersdisease
AT johneun theimpactofmuscarinicantagonismonpsychosisrelevantbehaviorsandstriatalsup11supcraclopridebindingintaumousemodelsofalzheimersdisease
AT ezracinamon theimpactofmuscarinicantagonismonpsychosisrelevantbehaviorsandstriatalsup11supcraclopridebindingintaumousemodelsofalzheimersdisease
AT erichchang theimpactofmuscarinicantagonismonpsychosisrelevantbehaviorsandstriatalsup11supcraclopridebindingintaumousemodelsofalzheimersdisease
AT peterdavies theimpactofmuscarinicantagonismonpsychosisrelevantbehaviorsandstriatalsup11supcraclopridebindingintaumousemodelsofalzheimersdisease
AT anvo theimpactofmuscarinicantagonismonpsychosisrelevantbehaviorsandstriatalsup11supcraclopridebindingintaumousemodelsofalzheimersdisease
AT jeremykoppel theimpactofmuscarinicantagonismonpsychosisrelevantbehaviorsandstriatalsup11supcraclopridebindingintaumousemodelsofalzheimersdisease
AT heidyjimenez impactofmuscarinicantagonismonpsychosisrelevantbehaviorsandstriatalsup11supcraclopridebindingintaumousemodelsofalzheimersdisease
AT josephcarrion impactofmuscarinicantagonismonpsychosisrelevantbehaviorsandstriatalsup11supcraclopridebindingintaumousemodelsofalzheimersdisease
AT leslieadrien impactofmuscarinicantagonismonpsychosisrelevantbehaviorsandstriatalsup11supcraclopridebindingintaumousemodelsofalzheimersdisease
AT adamwolin impactofmuscarinicantagonismonpsychosisrelevantbehaviorsandstriatalsup11supcraclopridebindingintaumousemodelsofalzheimersdisease
AT johneun impactofmuscarinicantagonismonpsychosisrelevantbehaviorsandstriatalsup11supcraclopridebindingintaumousemodelsofalzheimersdisease
AT ezracinamon impactofmuscarinicantagonismonpsychosisrelevantbehaviorsandstriatalsup11supcraclopridebindingintaumousemodelsofalzheimersdisease
AT erichchang impactofmuscarinicantagonismonpsychosisrelevantbehaviorsandstriatalsup11supcraclopridebindingintaumousemodelsofalzheimersdisease
AT peterdavies impactofmuscarinicantagonismonpsychosisrelevantbehaviorsandstriatalsup11supcraclopridebindingintaumousemodelsofalzheimersdisease
AT anvo impactofmuscarinicantagonismonpsychosisrelevantbehaviorsandstriatalsup11supcraclopridebindingintaumousemodelsofalzheimersdisease
AT jeremykoppel impactofmuscarinicantagonismonpsychosisrelevantbehaviorsandstriatalsup11supcraclopridebindingintaumousemodelsofalzheimersdisease