Translational profiling identifies sex-specific metabolic and epigenetic reprogramming of cortical microglia/macrophages in APPPS1-21 mice with an antibiotic-perturbed-microbiome

Abstract Background Microglia, the brain-resident macrophages perform immune surveillance and engage with pathological processes resulting in phenotype changes necessary for maintaining homeostasis. In preceding studies, we showed that antibiotic-induced perturbations of the gut microbiome of APPPS1...

Full description

Bibliographic Details
Main Authors: Shabana M. Shaik, Yajun Cao, Joseph V. Gogola, Hemraj B. Dodiya, Xulun Zhang, Hejer Boutej, Weinong Han, Jasna Kriz, Sangram S. Sisodia
Format: Article
Language:English
Published: BMC 2023-12-01
Series:Molecular Neurodegeneration
Subjects:
Online Access:https://doi.org/10.1186/s13024-023-00668-7
_version_ 1797388070572523520
author Shabana M. Shaik
Yajun Cao
Joseph V. Gogola
Hemraj B. Dodiya
Xulun Zhang
Hejer Boutej
Weinong Han
Jasna Kriz
Sangram S. Sisodia
author_facet Shabana M. Shaik
Yajun Cao
Joseph V. Gogola
Hemraj B. Dodiya
Xulun Zhang
Hejer Boutej
Weinong Han
Jasna Kriz
Sangram S. Sisodia
author_sort Shabana M. Shaik
collection DOAJ
description Abstract Background Microglia, the brain-resident macrophages perform immune surveillance and engage with pathological processes resulting in phenotype changes necessary for maintaining homeostasis. In preceding studies, we showed that antibiotic-induced perturbations of the gut microbiome of APPPS1-21 mice resulted in significant attenuation in Aβ amyloidosis and altered microglial phenotypes that are specific to male mice. The molecular events underlying microglial phenotypic transitions remain unclear. Here, by generating ‘APPPS1-21-CD11br’ reporter mice, we investigated the translational state of microglial/macrophage ribosomes during their phenotypic transition and in a sex-specific manner. Methods Six groups of mice that included WT-CD11br, antibiotic (ABX) or vehicle-treated APPPS1-21-CD11br males and females were sacrificed at 7-weeks of age (n = 15/group) and used for immunoprecipitation of microglial/macrophage polysomes from cortical homogenates using anti-FLAG antibody. Liquid chromatography coupled to tandem mass spectrometry and label-free quantification was used to identify newly synthesized peptides isolated from polysomes. Results We show that ABX-treatment leads to decreased Aβ levels in male APPPS1-21-CD11br mice with no significant changes in females. We identified microglial/macrophage polypeptides involved in mitochondrial dysfunction and altered calcium signaling that are associated with Aβ-induced oxidative stress. Notably, female mice also showed downregulation of newly-synthesized ribosomal proteins. Furthermore, male mice showed an increase in newly-synthesized polypeptides involved in FcγR-mediated phagocytosis, while females showed an increase in newly-synthesized polypeptides responsible for actin organization associated with microglial activation. Next, we show that ABX-treatment resulted in substantial remodeling of the epigenetic landscape, leading to a metabolic shift that accommodates the increased bioenergetic and biosynthetic demands associated with microglial polarization in a sex-specific manner. While microglia in ABX-treated male mice exhibited a metabolic shift towards a neuroprotective phenotype that promotes Aβ clearance, microglia in ABX-treated female mice exhibited loss of energy homeostasis due to persistent mitochondrial dysfunction and impaired lysosomal clearance that was associated with inflammatory phenotypes. Conclusions Our studies provide the first snapshot of the translational state of microglial/macrophage cells in a mouse model of Aβ amyloidosis that was subject to ABX treatment. ABX-mediated changes resulted in metabolic reprogramming of microglial phenotypes to modulate immune responses and amyloid clearance in a sex-specific manner. This microglial plasticity to support neuro-energetic homeostasis for its function based on sex paves the path for therapeutic modulation of immunometabolism for neurodegeneration.
first_indexed 2024-03-08T22:35:26Z
format Article
id doaj.art-825646de8c4347d8908615a566a99239
institution Directory Open Access Journal
issn 1750-1326
language English
last_indexed 2024-03-08T22:35:26Z
publishDate 2023-12-01
publisher BMC
record_format Article
series Molecular Neurodegeneration
spelling doaj.art-825646de8c4347d8908615a566a992392023-12-17T12:29:51ZengBMCMolecular Neurodegeneration1750-13262023-12-0118113110.1186/s13024-023-00668-7Translational profiling identifies sex-specific metabolic and epigenetic reprogramming of cortical microglia/macrophages in APPPS1-21 mice with an antibiotic-perturbed-microbiomeShabana M. Shaik0Yajun Cao1Joseph V. Gogola2Hemraj B. Dodiya3Xulun Zhang4Hejer Boutej5Weinong Han6Jasna Kriz7Sangram S. Sisodia8Dept. of Neurobiology, The University of ChicagoDept. of Neurobiology, The University of ChicagoDept. of Neurobiology, The University of ChicagoDept. of Neurobiology, The University of ChicagoDept. of Neurobiology, The University of ChicagoCERVO Brain Research Centre and Department of Psychiatry and Neuroscience, Laval UniversityDept. of Neurobiology, The University of ChicagoCERVO Brain Research Centre and Department of Psychiatry and Neuroscience, Laval UniversityDept. of Neurobiology, The University of ChicagoAbstract Background Microglia, the brain-resident macrophages perform immune surveillance and engage with pathological processes resulting in phenotype changes necessary for maintaining homeostasis. In preceding studies, we showed that antibiotic-induced perturbations of the gut microbiome of APPPS1-21 mice resulted in significant attenuation in Aβ amyloidosis and altered microglial phenotypes that are specific to male mice. The molecular events underlying microglial phenotypic transitions remain unclear. Here, by generating ‘APPPS1-21-CD11br’ reporter mice, we investigated the translational state of microglial/macrophage ribosomes during their phenotypic transition and in a sex-specific manner. Methods Six groups of mice that included WT-CD11br, antibiotic (ABX) or vehicle-treated APPPS1-21-CD11br males and females were sacrificed at 7-weeks of age (n = 15/group) and used for immunoprecipitation of microglial/macrophage polysomes from cortical homogenates using anti-FLAG antibody. Liquid chromatography coupled to tandem mass spectrometry and label-free quantification was used to identify newly synthesized peptides isolated from polysomes. Results We show that ABX-treatment leads to decreased Aβ levels in male APPPS1-21-CD11br mice with no significant changes in females. We identified microglial/macrophage polypeptides involved in mitochondrial dysfunction and altered calcium signaling that are associated with Aβ-induced oxidative stress. Notably, female mice also showed downregulation of newly-synthesized ribosomal proteins. Furthermore, male mice showed an increase in newly-synthesized polypeptides involved in FcγR-mediated phagocytosis, while females showed an increase in newly-synthesized polypeptides responsible for actin organization associated with microglial activation. Next, we show that ABX-treatment resulted in substantial remodeling of the epigenetic landscape, leading to a metabolic shift that accommodates the increased bioenergetic and biosynthetic demands associated with microglial polarization in a sex-specific manner. While microglia in ABX-treated male mice exhibited a metabolic shift towards a neuroprotective phenotype that promotes Aβ clearance, microglia in ABX-treated female mice exhibited loss of energy homeostasis due to persistent mitochondrial dysfunction and impaired lysosomal clearance that was associated with inflammatory phenotypes. Conclusions Our studies provide the first snapshot of the translational state of microglial/macrophage cells in a mouse model of Aβ amyloidosis that was subject to ABX treatment. ABX-mediated changes resulted in metabolic reprogramming of microglial phenotypes to modulate immune responses and amyloid clearance in a sex-specific manner. This microglial plasticity to support neuro-energetic homeostasis for its function based on sex paves the path for therapeutic modulation of immunometabolism for neurodegeneration.https://doi.org/10.1186/s13024-023-00668-7Alzheimer’s diseaseMicrogliaMacrophageMicrobiomeProteomicsMetabolism
spellingShingle Shabana M. Shaik
Yajun Cao
Joseph V. Gogola
Hemraj B. Dodiya
Xulun Zhang
Hejer Boutej
Weinong Han
Jasna Kriz
Sangram S. Sisodia
Translational profiling identifies sex-specific metabolic and epigenetic reprogramming of cortical microglia/macrophages in APPPS1-21 mice with an antibiotic-perturbed-microbiome
Molecular Neurodegeneration
Alzheimer’s disease
Microglia
Macrophage
Microbiome
Proteomics
Metabolism
title Translational profiling identifies sex-specific metabolic and epigenetic reprogramming of cortical microglia/macrophages in APPPS1-21 mice with an antibiotic-perturbed-microbiome
title_full Translational profiling identifies sex-specific metabolic and epigenetic reprogramming of cortical microglia/macrophages in APPPS1-21 mice with an antibiotic-perturbed-microbiome
title_fullStr Translational profiling identifies sex-specific metabolic and epigenetic reprogramming of cortical microglia/macrophages in APPPS1-21 mice with an antibiotic-perturbed-microbiome
title_full_unstemmed Translational profiling identifies sex-specific metabolic and epigenetic reprogramming of cortical microglia/macrophages in APPPS1-21 mice with an antibiotic-perturbed-microbiome
title_short Translational profiling identifies sex-specific metabolic and epigenetic reprogramming of cortical microglia/macrophages in APPPS1-21 mice with an antibiotic-perturbed-microbiome
title_sort translational profiling identifies sex specific metabolic and epigenetic reprogramming of cortical microglia macrophages in appps1 21 mice with an antibiotic perturbed microbiome
topic Alzheimer’s disease
Microglia
Macrophage
Microbiome
Proteomics
Metabolism
url https://doi.org/10.1186/s13024-023-00668-7
work_keys_str_mv AT shabanamshaik translationalprofilingidentifiessexspecificmetabolicandepigeneticreprogrammingofcorticalmicrogliamacrophagesinappps121micewithanantibioticperturbedmicrobiome
AT yajuncao translationalprofilingidentifiessexspecificmetabolicandepigeneticreprogrammingofcorticalmicrogliamacrophagesinappps121micewithanantibioticperturbedmicrobiome
AT josephvgogola translationalprofilingidentifiessexspecificmetabolicandepigeneticreprogrammingofcorticalmicrogliamacrophagesinappps121micewithanantibioticperturbedmicrobiome
AT hemrajbdodiya translationalprofilingidentifiessexspecificmetabolicandepigeneticreprogrammingofcorticalmicrogliamacrophagesinappps121micewithanantibioticperturbedmicrobiome
AT xulunzhang translationalprofilingidentifiessexspecificmetabolicandepigeneticreprogrammingofcorticalmicrogliamacrophagesinappps121micewithanantibioticperturbedmicrobiome
AT hejerboutej translationalprofilingidentifiessexspecificmetabolicandepigeneticreprogrammingofcorticalmicrogliamacrophagesinappps121micewithanantibioticperturbedmicrobiome
AT weinonghan translationalprofilingidentifiessexspecificmetabolicandepigeneticreprogrammingofcorticalmicrogliamacrophagesinappps121micewithanantibioticperturbedmicrobiome
AT jasnakriz translationalprofilingidentifiessexspecificmetabolicandepigeneticreprogrammingofcorticalmicrogliamacrophagesinappps121micewithanantibioticperturbedmicrobiome
AT sangramssisodia translationalprofilingidentifiessexspecificmetabolicandepigeneticreprogrammingofcorticalmicrogliamacrophagesinappps121micewithanantibioticperturbedmicrobiome