Cargando…

Differential LRRK2 signalling and gene expression in WT-LRRK2 and G2019S-LRRK2 mouse microglia treated with zymosan and MLi2

INTRODUCTION: Mutations in the Leucine Rich Repeat Kinase 2 (LRRK2) gene cause autosomal dominant Parkinson’s disease (PD) with the most common causative mutation being the LRRK2 p.G2019S within the kinase domain. LRRK2 protein is highly expressed in the human brain and also in the periphery, and hi...

Descripción completa

Detalles Bibliográficos
Autores principales: Nazish, Iqra, Mamais, Adamantios, Mallach, Anna, Bettencourt, Conceicao, Kaganovich, Alice, Warner, Tom, Hardy, John, Lewis, Patrick A., Pocock, Jennifer, Cookson, Mark R, Bandopadhyay, Rina
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Cold Spring Harbor Laboratory 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10515904/
https://www.ncbi.nlm.nih.gov/pubmed/37745519
http://dx.doi.org/10.1101/2023.09.14.557532
_version_ 1785109041503010816
author Nazish, Iqra
Mamais, Adamantios
Mallach, Anna
Bettencourt, Conceicao
Kaganovich, Alice
Warner, Tom
Hardy, John
Lewis, Patrick A.
Pocock, Jennifer
Cookson, Mark R
Bandopadhyay, Rina
author_facet Nazish, Iqra
Mamais, Adamantios
Mallach, Anna
Bettencourt, Conceicao
Kaganovich, Alice
Warner, Tom
Hardy, John
Lewis, Patrick A.
Pocock, Jennifer
Cookson, Mark R
Bandopadhyay, Rina
author_sort Nazish, Iqra
collection PubMed
description INTRODUCTION: Mutations in the Leucine Rich Repeat Kinase 2 (LRRK2) gene cause autosomal dominant Parkinson’s disease (PD) with the most common causative mutation being the LRRK2 p.G2019S within the kinase domain. LRRK2 protein is highly expressed in the human brain and also in the periphery, and high expression of dominant PD genes in immune cells suggest involvement of microglia and macrophages in inflammation related to PD. LRRK2 is known to respond to extracellular signalling including TLR4 resulting in alterations in gene expression, with the response to TLR2 signalling through zymosan being less known. METHODS: Here, we investigated the effects of zymosan, a TLR2 agonist and the potent and specific LRRK2 kinase inhibitor MLi-2 on gene expression in microglia from LRRK2-WT and LRRK2 p.G2019S knock-in mice by RNA-Sequencing analysis. RESULTS: We observed both overlapping and distinct zymosan and MLi-2 mediated gene expression profiles in microglia. At least two candidate Genome-Wide Association (GWAS) hits for PD, CathepsinB (Ctsb) and Glycoprotein-nmb (Gpnmb), were notably downregulated by zymosan treatment. Genes involved in inflammatory response and nervous system development were up and downregulated respectively with zymosan treatment while MLi-2 treatment particularly exhibited upregulated genes for ion transmembrane transport regulation. Furthermore, we observed the top twenty most significantly differentially expressed genes in LRRK2 p.G2019S microglia show enriched biological processes in iron transport and response to oxidative stress. DISCUSSION: Overall, these results suggest that microglial LRRK2 may contribute to PD pathogenesis through altered inflammatory pathways. Our findings should encourage future investigations of these putative avenues in the context of PD pathogenesis.
format Online
Article
Text
id pubmed-10515904
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Cold Spring Harbor Laboratory
record_format MEDLINE/PubMed
spelling pubmed-105159042023-09-23 Differential LRRK2 signalling and gene expression in WT-LRRK2 and G2019S-LRRK2 mouse microglia treated with zymosan and MLi2 Nazish, Iqra Mamais, Adamantios Mallach, Anna Bettencourt, Conceicao Kaganovich, Alice Warner, Tom Hardy, John Lewis, Patrick A. Pocock, Jennifer Cookson, Mark R Bandopadhyay, Rina bioRxiv Article INTRODUCTION: Mutations in the Leucine Rich Repeat Kinase 2 (LRRK2) gene cause autosomal dominant Parkinson’s disease (PD) with the most common causative mutation being the LRRK2 p.G2019S within the kinase domain. LRRK2 protein is highly expressed in the human brain and also in the periphery, and high expression of dominant PD genes in immune cells suggest involvement of microglia and macrophages in inflammation related to PD. LRRK2 is known to respond to extracellular signalling including TLR4 resulting in alterations in gene expression, with the response to TLR2 signalling through zymosan being less known. METHODS: Here, we investigated the effects of zymosan, a TLR2 agonist and the potent and specific LRRK2 kinase inhibitor MLi-2 on gene expression in microglia from LRRK2-WT and LRRK2 p.G2019S knock-in mice by RNA-Sequencing analysis. RESULTS: We observed both overlapping and distinct zymosan and MLi-2 mediated gene expression profiles in microglia. At least two candidate Genome-Wide Association (GWAS) hits for PD, CathepsinB (Ctsb) and Glycoprotein-nmb (Gpnmb), were notably downregulated by zymosan treatment. Genes involved in inflammatory response and nervous system development were up and downregulated respectively with zymosan treatment while MLi-2 treatment particularly exhibited upregulated genes for ion transmembrane transport regulation. Furthermore, we observed the top twenty most significantly differentially expressed genes in LRRK2 p.G2019S microglia show enriched biological processes in iron transport and response to oxidative stress. DISCUSSION: Overall, these results suggest that microglial LRRK2 may contribute to PD pathogenesis through altered inflammatory pathways. Our findings should encourage future investigations of these putative avenues in the context of PD pathogenesis. Cold Spring Harbor Laboratory 2023-09-14 /pmc/articles/PMC10515904/ /pubmed/37745519 http://dx.doi.org/10.1101/2023.09.14.557532 Text en https://creativecommons.org/licenses/by-nc-nd/4.0/This work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License (https://creativecommons.org/licenses/by-nc-nd/4.0/) , which allows reusers to copy and distribute the material in any medium or format in unadapted form only, for noncommercial purposes only, and only so long as attribution is given to the creator.
spellingShingle Article
Nazish, Iqra
Mamais, Adamantios
Mallach, Anna
Bettencourt, Conceicao
Kaganovich, Alice
Warner, Tom
Hardy, John
Lewis, Patrick A.
Pocock, Jennifer
Cookson, Mark R
Bandopadhyay, Rina
Differential LRRK2 signalling and gene expression in WT-LRRK2 and G2019S-LRRK2 mouse microglia treated with zymosan and MLi2
title Differential LRRK2 signalling and gene expression in WT-LRRK2 and G2019S-LRRK2 mouse microglia treated with zymosan and MLi2
title_full Differential LRRK2 signalling and gene expression in WT-LRRK2 and G2019S-LRRK2 mouse microglia treated with zymosan and MLi2
title_fullStr Differential LRRK2 signalling and gene expression in WT-LRRK2 and G2019S-LRRK2 mouse microglia treated with zymosan and MLi2
title_full_unstemmed Differential LRRK2 signalling and gene expression in WT-LRRK2 and G2019S-LRRK2 mouse microglia treated with zymosan and MLi2
title_short Differential LRRK2 signalling and gene expression in WT-LRRK2 and G2019S-LRRK2 mouse microglia treated with zymosan and MLi2
title_sort differential lrrk2 signalling and gene expression in wt-lrrk2 and g2019s-lrrk2 mouse microglia treated with zymosan and mli2
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10515904/
https://www.ncbi.nlm.nih.gov/pubmed/37745519
http://dx.doi.org/10.1101/2023.09.14.557532
work_keys_str_mv AT nazishiqra differentiallrrk2signallingandgeneexpressioninwtlrrk2andg2019slrrk2mousemicrogliatreatedwithzymosanandmli2
AT mamaisadamantios differentiallrrk2signallingandgeneexpressioninwtlrrk2andg2019slrrk2mousemicrogliatreatedwithzymosanandmli2
AT mallachanna differentiallrrk2signallingandgeneexpressioninwtlrrk2andg2019slrrk2mousemicrogliatreatedwithzymosanandmli2
AT bettencourtconceicao differentiallrrk2signallingandgeneexpressioninwtlrrk2andg2019slrrk2mousemicrogliatreatedwithzymosanandmli2
AT kaganovichalice differentiallrrk2signallingandgeneexpressioninwtlrrk2andg2019slrrk2mousemicrogliatreatedwithzymosanandmli2
AT warnertom differentiallrrk2signallingandgeneexpressioninwtlrrk2andg2019slrrk2mousemicrogliatreatedwithzymosanandmli2
AT hardyjohn differentiallrrk2signallingandgeneexpressioninwtlrrk2andg2019slrrk2mousemicrogliatreatedwithzymosanandmli2
AT lewispatricka differentiallrrk2signallingandgeneexpressioninwtlrrk2andg2019slrrk2mousemicrogliatreatedwithzymosanandmli2
AT pocockjennifer differentiallrrk2signallingandgeneexpressioninwtlrrk2andg2019slrrk2mousemicrogliatreatedwithzymosanandmli2
AT cooksonmarkr differentiallrrk2signallingandgeneexpressioninwtlrrk2andg2019slrrk2mousemicrogliatreatedwithzymosanandmli2
AT bandopadhyayrina differentiallrrk2signallingandgeneexpressioninwtlrrk2andg2019slrrk2mousemicrogliatreatedwithzymosanandmli2