Flow-cytometric microglial sorting coupled with quantitative proteomics identifies moesin as a highly-abundant microglial protein with relevance to Alzheimer’s disease

Abstract Background Proteomic characterization of microglia provides the most proximate assessment of functionally relevant molecular mechanisms of neuroinflammation. However, microglial proteomics studies have been limited by low cellular yield and contamination by non-microglial proteins using exi...

Full description

Bibliographic Details
Main Authors: Sruti Rayaprolu, Tianwen Gao, Hailian Xiao, Supriya Ramesha, Laura D. Weinstock, Jheel Shah, Duc M. Duong, Eric B. Dammer, James A. Webster, James J. Lah, Levi B. Wood, Ranjita Betarbet, Allan I. Levey, Nicholas T. Seyfried, Srikant Rangaraju
Format: Article
Language:English
Published: BMC 2020-05-01
Series:Molecular Neurodegeneration
Subjects:
Online Access:http://link.springer.com/article/10.1186/s13024-020-00377-5
id doaj-d7c72d1d44654f4abfd3eb413217f735
record_format Article
collection DOAJ
language English
format Article
sources DOAJ
author Sruti Rayaprolu
Tianwen Gao
Hailian Xiao
Supriya Ramesha
Laura D. Weinstock
Jheel Shah
Duc M. Duong
Eric B. Dammer
James A. Webster
James J. Lah
Levi B. Wood
Ranjita Betarbet
Allan I. Levey
Nicholas T. Seyfried
Srikant Rangaraju
spellingShingle Sruti Rayaprolu
Tianwen Gao
Hailian Xiao
Supriya Ramesha
Laura D. Weinstock
Jheel Shah
Duc M. Duong
Eric B. Dammer
James A. Webster
James J. Lah
Levi B. Wood
Ranjita Betarbet
Allan I. Levey
Nicholas T. Seyfried
Srikant Rangaraju
Flow-cytometric microglial sorting coupled with quantitative proteomics identifies moesin as a highly-abundant microglial protein with relevance to Alzheimer’s disease
Molecular Neurodegeneration
Microglia
Proteomics
Mass spectrometry
FACS
MACS
Alzheimer’s disease
author_facet Sruti Rayaprolu
Tianwen Gao
Hailian Xiao
Supriya Ramesha
Laura D. Weinstock
Jheel Shah
Duc M. Duong
Eric B. Dammer
James A. Webster
James J. Lah
Levi B. Wood
Ranjita Betarbet
Allan I. Levey
Nicholas T. Seyfried
Srikant Rangaraju
author_sort Sruti Rayaprolu
title Flow-cytometric microglial sorting coupled with quantitative proteomics identifies moesin as a highly-abundant microglial protein with relevance to Alzheimer’s disease
title_short Flow-cytometric microglial sorting coupled with quantitative proteomics identifies moesin as a highly-abundant microglial protein with relevance to Alzheimer’s disease
title_full Flow-cytometric microglial sorting coupled with quantitative proteomics identifies moesin as a highly-abundant microglial protein with relevance to Alzheimer’s disease
title_fullStr Flow-cytometric microglial sorting coupled with quantitative proteomics identifies moesin as a highly-abundant microglial protein with relevance to Alzheimer’s disease
title_full_unstemmed Flow-cytometric microglial sorting coupled with quantitative proteomics identifies moesin as a highly-abundant microglial protein with relevance to Alzheimer’s disease
title_sort flow-cytometric microglial sorting coupled with quantitative proteomics identifies moesin as a highly-abundant microglial protein with relevance to alzheimer’s disease
publisher BMC
series Molecular Neurodegeneration
issn 1750-1326
publishDate 2020-05-01
description Abstract Background Proteomic characterization of microglia provides the most proximate assessment of functionally relevant molecular mechanisms of neuroinflammation. However, microglial proteomics studies have been limited by low cellular yield and contamination by non-microglial proteins using existing enrichment strategies. Methods We coupled magnetic-activated cell sorting (MACS) and fluorescence activated cell sorting (FACS) of microglia with tandem mass tag-mass spectrometry (TMT-MS) to obtain a highly-pure microglial proteome and identified a core set of highly-abundant microglial proteins in adult mouse brain. We interrogated existing human proteomic data for Alzheimer’s disease (AD) relevance of highly-abundant microglial proteins and performed immuno-histochemical and in-vitro validation studies. Results Quantitative multiplexed proteomics by TMT-MS of CD11b + MACS-enriched (N = 5 mice) and FACS-isolated (N = 5 mice), from adult wild-type mice, identified 1791 proteins. A total of 203 proteins were highly abundant in both datasets, representing a core-set of highly abundant microglial proteins. In addition, we found 953 differentially enriched proteins comparing MACS and FACS-based approaches, indicating significant differences between both strategies. The FACS-isolated microglia proteome was enriched with cytosolic, endoplasmic reticulum, and ribosomal proteins involved in protein metabolism and immune system functions, as well as an abundance of canonical microglial proteins. Conversely, the MACS-enriched microglia proteome was enriched with mitochondrial and synaptic proteins and higher abundance of neuronal, oligodendrocytic and astrocytic proteins. From the 203 consensus microglial proteins with high abundance in both datasets, we confirmed microglial expression of moesin (Msn) in wild-type and 5xFAD mouse brains as well as in human AD brains. Msn expression is nearly exclusively found in microglia that surround Aβ plaques in 5xFAD brains. In in-vitro primary microglial studies, Msn silencing by siRNA decreased Aβ phagocytosis and increased lipopolysaccharide-induced production of the pro-inflammatory cytokine, tumor necrosis factor (TNF). In network analysis of human brain proteomic data, Msn was a hub protein of an inflammatory co-expression module positively associated with AD neuropathological features and cognitive dysfunction. Conclusions Using FACS coupled with TMT-MS as the method of choice for microglial proteomics, we define a core set of highly-abundant adult microglial proteins. Among these, we validate Msn as highly-abundant in plaque-associated microglia with relevance to human AD.
topic Microglia
Proteomics
Mass spectrometry
FACS
MACS
Alzheimer’s disease
url http://link.springer.com/article/10.1186/s13024-020-00377-5
work_keys_str_mv AT srutirayaprolu flowcytometricmicroglialsortingcoupledwithquantitativeproteomicsidentifiesmoesinasahighlyabundantmicroglialproteinwithrelevancetoalzheimersdisease
AT tianwengao flowcytometricmicroglialsortingcoupledwithquantitativeproteomicsidentifiesmoesinasahighlyabundantmicroglialproteinwithrelevancetoalzheimersdisease
AT hailianxiao flowcytometricmicroglialsortingcoupledwithquantitativeproteomicsidentifiesmoesinasahighlyabundantmicroglialproteinwithrelevancetoalzheimersdisease
AT supriyaramesha flowcytometricmicroglialsortingcoupledwithquantitativeproteomicsidentifiesmoesinasahighlyabundantmicroglialproteinwithrelevancetoalzheimersdisease
AT lauradweinstock flowcytometricmicroglialsortingcoupledwithquantitativeproteomicsidentifiesmoesinasahighlyabundantmicroglialproteinwithrelevancetoalzheimersdisease
AT jheelshah flowcytometricmicroglialsortingcoupledwithquantitativeproteomicsidentifiesmoesinasahighlyabundantmicroglialproteinwithrelevancetoalzheimersdisease
AT ducmduong flowcytometricmicroglialsortingcoupledwithquantitativeproteomicsidentifiesmoesinasahighlyabundantmicroglialproteinwithrelevancetoalzheimersdisease
AT ericbdammer flowcytometricmicroglialsortingcoupledwithquantitativeproteomicsidentifiesmoesinasahighlyabundantmicroglialproteinwithrelevancetoalzheimersdisease
AT jamesawebster flowcytometricmicroglialsortingcoupledwithquantitativeproteomicsidentifiesmoesinasahighlyabundantmicroglialproteinwithrelevancetoalzheimersdisease
AT jamesjlah flowcytometricmicroglialsortingcoupledwithquantitativeproteomicsidentifiesmoesinasahighlyabundantmicroglialproteinwithrelevancetoalzheimersdisease
AT levibwood flowcytometricmicroglialsortingcoupledwithquantitativeproteomicsidentifiesmoesinasahighlyabundantmicroglialproteinwithrelevancetoalzheimersdisease
AT ranjitabetarbet flowcytometricmicroglialsortingcoupledwithquantitativeproteomicsidentifiesmoesinasahighlyabundantmicroglialproteinwithrelevancetoalzheimersdisease
AT allanilevey flowcytometricmicroglialsortingcoupledwithquantitativeproteomicsidentifiesmoesinasahighlyabundantmicroglialproteinwithrelevancetoalzheimersdisease
AT nicholastseyfried flowcytometricmicroglialsortingcoupledwithquantitativeproteomicsidentifiesmoesinasahighlyabundantmicroglialproteinwithrelevancetoalzheimersdisease
AT srikantrangaraju flowcytometricmicroglialsortingcoupledwithquantitativeproteomicsidentifiesmoesinasahighlyabundantmicroglialproteinwithrelevancetoalzheimersdisease
_version_ 1724947121344348160
spelling doaj-d7c72d1d44654f4abfd3eb413217f7352020-11-25T02:03:35ZengBMCMolecular Neurodegeneration1750-13262020-05-0115112210.1186/s13024-020-00377-5Flow-cytometric microglial sorting coupled with quantitative proteomics identifies moesin as a highly-abundant microglial protein with relevance to Alzheimer’s diseaseSruti Rayaprolu0Tianwen Gao1Hailian Xiao2Supriya Ramesha3Laura D. Weinstock4Jheel Shah5Duc M. Duong6Eric B. Dammer7James A. Webster8James J. Lah9Levi B. Wood10Ranjita Betarbet11Allan I. Levey12Nicholas T. Seyfried13Srikant Rangaraju14Department of Neurology, Emory University School of MedicineDepartment of Neurology, Emory University School of MedicineDepartment of Neurology, Emory University School of MedicineDepartment of Neurology, Emory University School of MedicineParker H. Petit Institute for Bioengineering and Bioscience, Wallace H. Coulter Department of Biomedical Engineering, and Georgia W. Woodruff School of Mechanical Engineering, Georgia Institute of TechnologyDepartment of Neurology, Emory University School of MedicineDepartment of Neurology, Emory University School of MedicineSchool of Medicine, Emory UniversityDepartment of Neurology, Emory University School of MedicineDepartment of Neurology, Emory University School of MedicineParker H. Petit Institute for Bioengineering and Bioscience, Wallace H. Coulter Department of Biomedical Engineering, and Georgia W. Woodruff School of Mechanical Engineering, Georgia Institute of TechnologyDepartment of Neurology, Emory University School of MedicineDepartment of Neurology, Emory University School of MedicineDepartment of Neurology, Emory University School of MedicineDepartment of Neurology, Emory University School of MedicineAbstract Background Proteomic characterization of microglia provides the most proximate assessment of functionally relevant molecular mechanisms of neuroinflammation. However, microglial proteomics studies have been limited by low cellular yield and contamination by non-microglial proteins using existing enrichment strategies. Methods We coupled magnetic-activated cell sorting (MACS) and fluorescence activated cell sorting (FACS) of microglia with tandem mass tag-mass spectrometry (TMT-MS) to obtain a highly-pure microglial proteome and identified a core set of highly-abundant microglial proteins in adult mouse brain. We interrogated existing human proteomic data for Alzheimer’s disease (AD) relevance of highly-abundant microglial proteins and performed immuno-histochemical and in-vitro validation studies. Results Quantitative multiplexed proteomics by TMT-MS of CD11b + MACS-enriched (N = 5 mice) and FACS-isolated (N = 5 mice), from adult wild-type mice, identified 1791 proteins. A total of 203 proteins were highly abundant in both datasets, representing a core-set of highly abundant microglial proteins. In addition, we found 953 differentially enriched proteins comparing MACS and FACS-based approaches, indicating significant differences between both strategies. The FACS-isolated microglia proteome was enriched with cytosolic, endoplasmic reticulum, and ribosomal proteins involved in protein metabolism and immune system functions, as well as an abundance of canonical microglial proteins. Conversely, the MACS-enriched microglia proteome was enriched with mitochondrial and synaptic proteins and higher abundance of neuronal, oligodendrocytic and astrocytic proteins. From the 203 consensus microglial proteins with high abundance in both datasets, we confirmed microglial expression of moesin (Msn) in wild-type and 5xFAD mouse brains as well as in human AD brains. Msn expression is nearly exclusively found in microglia that surround Aβ plaques in 5xFAD brains. In in-vitro primary microglial studies, Msn silencing by siRNA decreased Aβ phagocytosis and increased lipopolysaccharide-induced production of the pro-inflammatory cytokine, tumor necrosis factor (TNF). In network analysis of human brain proteomic data, Msn was a hub protein of an inflammatory co-expression module positively associated with AD neuropathological features and cognitive dysfunction. Conclusions Using FACS coupled with TMT-MS as the method of choice for microglial proteomics, we define a core set of highly-abundant adult microglial proteins. Among these, we validate Msn as highly-abundant in plaque-associated microglia with relevance to human AD.http://link.springer.com/article/10.1186/s13024-020-00377-5MicrogliaProteomicsMass spectrometryFACSMACSAlzheimer’s disease