Abstract

In Alzheimer’s disease (AD), microglia are affected by disease processes, but may also drive pathogenesis. AD pathology-associated microglial populations have been identified with single-cell RNA-Seq, but have not been validated in human brain tissue with anatomical context. Here, we quantified myeloid cell markers to identify changes in AD pathology-associated microglial populations. We performed fluorescent immunohistochemistry on normal (n = 8) and AD (n = 8) middle temporal gyri, co-labelling the pan-myeloid cell marker, Iba1, with one of 11 markers of interest (MOIs): CD45, HLA-DR, CD14, CD74, CD33, CD206, CD32, CD163, P2RY12, TMEM119, L-Ferritin. Novel image analyses quantified the single-cell abundance of Iba1 and each MOI. Each cell was gated into one Iba1-MOI population (Iba1low MOIhigh, Iba1high MOIhigh, or Iba1high MOIlow) and the abundance of each population was compared between AD and control. Triple-labelling of L-Ferritin and Iba1 with a subset of MOIs was performed to investigate L-Ferritin-MOI co-expression on Iba1low cells. Iba1low MOIhigh myeloid cell populations delineated by MOIs CD45, HLA-DR, CD14, CD74, CD33, CD32, and L-Ferritin were increased in AD. Further investigation of the Iba1low MOIhigh populations revealed that their abundances correlated with tau, but not amyloid beta, load in AD. The Iba1low microglial population highly expressed L-Ferritin, reflecting microglial dysfunction. The L-Ferritinhigh CD74high HLA-DRhigh phenotype of the Iba1low population mirrors that of a human AD pathology-associated microglial subpopulation previously identified using single-cell RNA-Seq. Our high-throughput immunohistochemical data with anatomical context support the microglial dysfunction hypothesis of AD.

Highlights

  • Alzheimer’s disease (AD) pathology is characterised by the accumulation of amyloid beta plaques and hyperphosphorylated tau tangles [1]

  • An ­Iba1low myeloid cell population is more abundant in the AD temporal cortex We co-labelled Ionised calcium binding adaptor protein 1 (Iba1) with one of 11 myeloid cell markers using immunohistochemistry on normal and AD

  • We developed a custom MetaMorph image analysis pipeline which allowed for single-cell measurements of Iba1 and markers of interest (MOI) staining intensity in post-mortem human tissue (Fig. 2A)

Read more

Summary

Introduction

Alzheimer’s disease (AD) pathology is characterised by the accumulation of amyloid beta plaques and hyperphosphorylated tau tangles [1]. AD pathology-associated microglial subpopulations with unique transcriptomic signatures have been identified, including a population with enriched expression of FTL (L-Ferritin), HLA-DRA, HLA-DRB1, CD14, and CD74 [16, 17] (Additional file 1: Table S1). These studies require the dissociation of cells from whole tissue. As such, they are unable to resolve whether changes in microglial populations are driven by human AD pathology or quantify the anatomical location of microglia with unique expression profiles relative to amyloid beta and tau in the brain

Methods
Results
Discussion
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.