Cargando…

Sex and APOE Genotype Alter the Basal and Induced Inflammatory States of Primary Microglia from APOE Targeted Replacement Mice

The sex and APOE4 genotype are significant risk factors for Alzheimer’s disease (AD); however, the mechanism(s) responsible for this interaction are still a matter of debate. Here, we assess the responses of mixed-sex and sex-specific APOE3 and APOE4 primary microglia (PMG) to lipopolysaccharide and...

Descripción completa

Detalles Bibliográficos
Autores principales: Mhatre-Winters, Isha, Eid, Aseel, Han, Yoonhee, Tieu, Kim, Richardson, Jason R.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: MDPI 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9456163/
https://www.ncbi.nlm.nih.gov/pubmed/36077227
http://dx.doi.org/10.3390/ijms23179829
_version_ 1784785745470291968
author Mhatre-Winters, Isha
Eid, Aseel
Han, Yoonhee
Tieu, Kim
Richardson, Jason R.
author_facet Mhatre-Winters, Isha
Eid, Aseel
Han, Yoonhee
Tieu, Kim
Richardson, Jason R.
author_sort Mhatre-Winters, Isha
collection PubMed
description The sex and APOE4 genotype are significant risk factors for Alzheimer’s disease (AD); however, the mechanism(s) responsible for this interaction are still a matter of debate. Here, we assess the responses of mixed-sex and sex-specific APOE3 and APOE4 primary microglia (PMG) to lipopolysaccharide and interferon-gamma. In our investigation, inflammatory cytokine profiles were assessed by qPCR and multiplex ELISA assays. Mixed-sex APOE4 PMG exhibited higher basal mRNA expression and secreted levels of TNFa and IL1b. In sex-specific cultures, basal expression and secreted levels of IL1b, TNFa, IL6, and NOS2 were 2–3 fold higher in APOE4 female PMG compared to APOE4 males, with both higher than APOE3 cells. Following an inflammatory stimulus, the expression of pro-inflammatory cytokines and the secreted cytokine level were upregulated in the order E4 female > E4 male > E3 female > E3 male in sex-specific cultures. These data indicate that the APOE4 genotype and female sex together contribute to a greater inflammatory response in PMG isolated from targeted replacement humanized APOE mice. These data are consistent with clinical data and indicate that sex-specific PMG may provide a platform for exploring mechanisms of genotype and sex differences in AD related to neuroinflammation and neurodegeneration.
format Online
Article
Text
id pubmed-9456163
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher MDPI
record_format MEDLINE/PubMed
spelling pubmed-94561632022-09-09 Sex and APOE Genotype Alter the Basal and Induced Inflammatory States of Primary Microglia from APOE Targeted Replacement Mice Mhatre-Winters, Isha Eid, Aseel Han, Yoonhee Tieu, Kim Richardson, Jason R. Int J Mol Sci Article The sex and APOE4 genotype are significant risk factors for Alzheimer’s disease (AD); however, the mechanism(s) responsible for this interaction are still a matter of debate. Here, we assess the responses of mixed-sex and sex-specific APOE3 and APOE4 primary microglia (PMG) to lipopolysaccharide and interferon-gamma. In our investigation, inflammatory cytokine profiles were assessed by qPCR and multiplex ELISA assays. Mixed-sex APOE4 PMG exhibited higher basal mRNA expression and secreted levels of TNFa and IL1b. In sex-specific cultures, basal expression and secreted levels of IL1b, TNFa, IL6, and NOS2 were 2–3 fold higher in APOE4 female PMG compared to APOE4 males, with both higher than APOE3 cells. Following an inflammatory stimulus, the expression of pro-inflammatory cytokines and the secreted cytokine level were upregulated in the order E4 female > E4 male > E3 female > E3 male in sex-specific cultures. These data indicate that the APOE4 genotype and female sex together contribute to a greater inflammatory response in PMG isolated from targeted replacement humanized APOE mice. These data are consistent with clinical data and indicate that sex-specific PMG may provide a platform for exploring mechanisms of genotype and sex differences in AD related to neuroinflammation and neurodegeneration. MDPI 2022-08-29 /pmc/articles/PMC9456163/ /pubmed/36077227 http://dx.doi.org/10.3390/ijms23179829 Text en © 2022 by the authors. https://creativecommons.org/licenses/by/4.0/Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
spellingShingle Article
Mhatre-Winters, Isha
Eid, Aseel
Han, Yoonhee
Tieu, Kim
Richardson, Jason R.
Sex and APOE Genotype Alter the Basal and Induced Inflammatory States of Primary Microglia from APOE Targeted Replacement Mice
title Sex and APOE Genotype Alter the Basal and Induced Inflammatory States of Primary Microglia from APOE Targeted Replacement Mice
title_full Sex and APOE Genotype Alter the Basal and Induced Inflammatory States of Primary Microglia from APOE Targeted Replacement Mice
title_fullStr Sex and APOE Genotype Alter the Basal and Induced Inflammatory States of Primary Microglia from APOE Targeted Replacement Mice
title_full_unstemmed Sex and APOE Genotype Alter the Basal and Induced Inflammatory States of Primary Microglia from APOE Targeted Replacement Mice
title_short Sex and APOE Genotype Alter the Basal and Induced Inflammatory States of Primary Microglia from APOE Targeted Replacement Mice
title_sort sex and apoe genotype alter the basal and induced inflammatory states of primary microglia from apoe targeted replacement mice
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9456163/
https://www.ncbi.nlm.nih.gov/pubmed/36077227
http://dx.doi.org/10.3390/ijms23179829
work_keys_str_mv AT mhatrewintersisha sexandapoegenotypealterthebasalandinducedinflammatorystatesofprimarymicrogliafromapoetargetedreplacementmice
AT eidaseel sexandapoegenotypealterthebasalandinducedinflammatorystatesofprimarymicrogliafromapoetargetedreplacementmice
AT hanyoonhee sexandapoegenotypealterthebasalandinducedinflammatorystatesofprimarymicrogliafromapoetargetedreplacementmice
AT tieukim sexandapoegenotypealterthebasalandinducedinflammatorystatesofprimarymicrogliafromapoetargetedreplacementmice
AT richardsonjasonr sexandapoegenotypealterthebasalandinducedinflammatorystatesofprimarymicrogliafromapoetargetedreplacementmice