Motor cortex transcriptome reveals microglial key events in amyotrophic lateral sclerosis

OBJECTIVETo identify transcriptomic changes, neuropathologic correlates, and cellular subpopulations in the motor cortex of sporadic amyotrophic lateral sclerosis (ALS). METHODSWe performed massive RNA sequencing of the motor cortex of patients with ALS (n = 11) and healthy controls (HCs; n = 8) and...

Full description

Saved in:
Bibliographic Details
Published inNeurology : neuroimmunology & neuroinflammation Vol. 7; no. 5; p. e829
Main Authors Dols-Icardo, Oriol, Montal, Víctor, Sirisi, Sònia, López-Pernas, Gema, Cervera-Carles, Laura, Querol-Vilaseca, Marta, Muñoz, Laia, Belbin, Olivia, Alcolea, Daniel, Molina-Porcel, Laura, Pegueroles, Jordi, Turón-Sans, Janina, Blesa, Rafael, Lleó, Alberto, Fortea, Juan, Rojas-García, Ricard, Clarimón, Jordi
Format Journal Article
LanguageEnglish
Published Hagerstown, MD American Academy of Neurology 01.09.2020
Lippincott Williams & Wilkins
Online AccessGet full text

Cover

Loading…
More Information
Summary:OBJECTIVETo identify transcriptomic changes, neuropathologic correlates, and cellular subpopulations in the motor cortex of sporadic amyotrophic lateral sclerosis (ALS). METHODSWe performed massive RNA sequencing of the motor cortex of patients with ALS (n = 11) and healthy controls (HCs; n = 8) and analyzed gene expression alterations, differential isoform usage, and gene coexpression networks. Furthermore, we used cell type deconvolution algorithms with human single-nucleus RNA sequencing data as reference to identify perturbations in cell type composition associated with ALS. We performed immunohistochemical techniques to evaluate neuropathologic changes in this brain region. RESULTSWe report extensive RNA expression alterations at gene and isoform levels, characterized by the enrichment of neuroinflammatory and synaptic-related pathways. The assembly of gene coexpression modules confirmed the involvement of these 2 major transcriptomic changes, which also showed opposite directions related to the disease. Cell type deconvolution revealed an overrepresentation of microglial cells in ALS compared with HC. Notably, microgliosis was driven by a subcellular population presenting a gene expression signature overlapping with the recently described disease-associated microglia (DAM). Using immunohistochemistry, we further evidenced that this microglial subpopulation is overrepresented in ALS and that the density of pTDP43 aggregates negatively correlates with the proportion of microglial cells. CONCLUSIONSDAM has a central role in microglia-related neuroinflammatory changes in the motor cortex of patients with ALS, and these alterations are coupled with a reduced expression of postsynaptic transcripts.
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
Go to Neurology.org/NN for full disclosures. Funding information is provided at the end of the article.
The Article Processing Charge was funded by the authors.
ISSN:2332-7812
2332-7812
DOI:10.1212/NXI.0000000000000829