Cargando…

The microglial activation state regulates migration and roles of matrix-dissolving enzymes for invasion

BACKGROUND: Microglial cells are highly mobile under many circumstances and, after central nervous system (CNS) damage, they must contend with the dense extracellular matrix (ECM) in order to reach their target sites. In response to damage or disease, microglia undergo complex activation processes t...

Descripción completa

Detalles Bibliográficos
Autores principales: Lively, Starlee, Schlichter, Lyanne C
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2013
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3693964/
https://www.ncbi.nlm.nih.gov/pubmed/23786632
http://dx.doi.org/10.1186/1742-2094-10-75
_version_ 1782274781509320704
author Lively, Starlee
Schlichter, Lyanne C
author_facet Lively, Starlee
Schlichter, Lyanne C
author_sort Lively, Starlee
collection PubMed
description BACKGROUND: Microglial cells are highly mobile under many circumstances and, after central nervous system (CNS) damage, they must contend with the dense extracellular matrix (ECM) in order to reach their target sites. In response to damage or disease, microglia undergo complex activation processes that can be modulated by environmental cues and culminate in either detrimental or beneficial outcomes. Thus, there is considerable interest in comparing their pro-inflammatory (‘classical’ activation) and resolving ‘alternative’ activation states. Almost nothing is known about how these activation states affect the ability of microglia to migrate and degrade ECM, or the enzymes used for substrate degradation. This is the subject of the present study. METHODS: Primary cultured rat microglial cells were exposed to lipopolysaccharide (LPS) to evoke classical activation or IL4 to evoke alternative activation. High-resolution microscopy was used to monitor changes in cell morphology and aspects of the cytoskeleton. We quantified migration in a scratch-wound assay and through open filter holes, and invasion through Matrigel™. A panel of inhibitors was used to analyze contributions of different matrix-degrading enzymes to migration and invasion, and quantitative real-time reverse transcriptase PCR (qRT-PCR) was used to assess changes in their expression. RESULTS: Vinculin- and F-actin-rich lamellae were prominent in untreated and IL4-treated microglia (but not after LPS). IL4 increased the migratory capacity of microglia but eliminated the preferential anterior nuclear-centrosomal axis polarity and location of the microtubule organizing center (MTOC). Microglia degraded fibronectin, regardless of treatment, but LPS-treated cells were relatively immobile and IL4-treated cells invaded much more effectively through Matrigel™. For invasion, untreated microglia primarily used cysteine proteases, but IL4-treated cells used a wider range of enzymes (cysteine proteases, cathepsin S and K, heparanase, and matrix metalloproteases). Untreated microglia expressed MMP2, MMP12, heparanase, and four cathepsins (B, K, L1, and S). Each activation stimulus upregulated a different subset of enzymes. IL4 increased MMP2 and cathepsins S and K; whereas LPS increased MMP9, MMP12, MMP14 (MT1-MMP), heparanase, and cathepsin L1. CONCLUSIONS: Microglial cells migrate during CNS development and after CNS damage or disease. Thus, there are broad implications of the finding that classically and alternatively activated microglia differ in morphology, cytoskeleton, migratory and invasive capacity, and in the usage of ECM-degrading enzymes.
format Online
Article
Text
id pubmed-3693964
institution National Center for Biotechnology Information
language English
publishDate 2013
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-36939642013-06-27 The microglial activation state regulates migration and roles of matrix-dissolving enzymes for invasion Lively, Starlee Schlichter, Lyanne C J Neuroinflammation Research BACKGROUND: Microglial cells are highly mobile under many circumstances and, after central nervous system (CNS) damage, they must contend with the dense extracellular matrix (ECM) in order to reach their target sites. In response to damage or disease, microglia undergo complex activation processes that can be modulated by environmental cues and culminate in either detrimental or beneficial outcomes. Thus, there is considerable interest in comparing their pro-inflammatory (‘classical’ activation) and resolving ‘alternative’ activation states. Almost nothing is known about how these activation states affect the ability of microglia to migrate and degrade ECM, or the enzymes used for substrate degradation. This is the subject of the present study. METHODS: Primary cultured rat microglial cells were exposed to lipopolysaccharide (LPS) to evoke classical activation or IL4 to evoke alternative activation. High-resolution microscopy was used to monitor changes in cell morphology and aspects of the cytoskeleton. We quantified migration in a scratch-wound assay and through open filter holes, and invasion through Matrigel™. A panel of inhibitors was used to analyze contributions of different matrix-degrading enzymes to migration and invasion, and quantitative real-time reverse transcriptase PCR (qRT-PCR) was used to assess changes in their expression. RESULTS: Vinculin- and F-actin-rich lamellae were prominent in untreated and IL4-treated microglia (but not after LPS). IL4 increased the migratory capacity of microglia but eliminated the preferential anterior nuclear-centrosomal axis polarity and location of the microtubule organizing center (MTOC). Microglia degraded fibronectin, regardless of treatment, but LPS-treated cells were relatively immobile and IL4-treated cells invaded much more effectively through Matrigel™. For invasion, untreated microglia primarily used cysteine proteases, but IL4-treated cells used a wider range of enzymes (cysteine proteases, cathepsin S and K, heparanase, and matrix metalloproteases). Untreated microglia expressed MMP2, MMP12, heparanase, and four cathepsins (B, K, L1, and S). Each activation stimulus upregulated a different subset of enzymes. IL4 increased MMP2 and cathepsins S and K; whereas LPS increased MMP9, MMP12, MMP14 (MT1-MMP), heparanase, and cathepsin L1. CONCLUSIONS: Microglial cells migrate during CNS development and after CNS damage or disease. Thus, there are broad implications of the finding that classically and alternatively activated microglia differ in morphology, cytoskeleton, migratory and invasive capacity, and in the usage of ECM-degrading enzymes. BioMed Central 2013-06-21 /pmc/articles/PMC3693964/ /pubmed/23786632 http://dx.doi.org/10.1186/1742-2094-10-75 Text en Copyright © 2013 Lively and Schlichter; licensee BioMed Central Ltd. http://creativecommons.org/licenses/by/2.0 This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
spellingShingle Research
Lively, Starlee
Schlichter, Lyanne C
The microglial activation state regulates migration and roles of matrix-dissolving enzymes for invasion
title The microglial activation state regulates migration and roles of matrix-dissolving enzymes for invasion
title_full The microglial activation state regulates migration and roles of matrix-dissolving enzymes for invasion
title_fullStr The microglial activation state regulates migration and roles of matrix-dissolving enzymes for invasion
title_full_unstemmed The microglial activation state regulates migration and roles of matrix-dissolving enzymes for invasion
title_short The microglial activation state regulates migration and roles of matrix-dissolving enzymes for invasion
title_sort microglial activation state regulates migration and roles of matrix-dissolving enzymes for invasion
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3693964/
https://www.ncbi.nlm.nih.gov/pubmed/23786632
http://dx.doi.org/10.1186/1742-2094-10-75
work_keys_str_mv AT livelystarlee themicroglialactivationstateregulatesmigrationandrolesofmatrixdissolvingenzymesforinvasion
AT schlichterlyannec themicroglialactivationstateregulatesmigrationandrolesofmatrixdissolvingenzymesforinvasion
AT livelystarlee microglialactivationstateregulatesmigrationandrolesofmatrixdissolvingenzymesforinvasion
AT schlichterlyannec microglialactivationstateregulatesmigrationandrolesofmatrixdissolvingenzymesforinvasion