Abstract

BackgroundMicroglial dysfunction is implicated in frontotemporal lobar degeneration (FTLD). Although studies have reported excessive microglial activation or senescence (dystrophy) in Alzheimer’s disease (AD), few have explored this in FTLD. We examined regional patterns of microglial burden, activation and dystrophy in sporadic and genetic FTLD, sporadic AD and controls.MethodsImmunohistochemistry was performed in frontal and temporal grey and white matter from 50 pathologically confirmed FTLD cases (31 sporadic, 19 genetic: 20 FTLD-tau, 26 FTLD-TDP, four FTLD-FUS), five AD cases and five controls, using markers to detect phagocytic (CD68-positive) and antigen-presenting (CR3/43-positive) microglia, and microglia in general (Iba1-positive). Microglial burden and activation (morphology) were assessed quantitatively for each microglial phenotype. Iba1-positive microglia were assessed semi-quantitatively for dystrophy severity and qualitatively for rod-shaped and hypertrophic morphology. Microglia were compared in each region between FTLD, AD and controls, and between different pathological subtypes of FTLD, including its main subtypes (FTLD-tau, FTLD-TDP, FTLD-FUS), and subtypes of FTLD-tau, FTLD-TDP and genetic FTLD. Microglia were also compared between grey and white matter within each lobe for each group.ResultsThere was a higher burden of phagocytic and antigen-presenting microglia in FTLD and AD cases than controls, but activation was often not increased. Burden was generally higher in white matter than grey matter, but activation was greater in grey matter. However, microglia varied regionally according to FTLD subtype and disease mechanism. Dystrophy was more severe in FTLD and AD than controls, and more severe in white than grey matter, but this also varied regionally and was particularly extensive in FTLD due to progranulin (GRN) mutations. Presence of rod-shaped and hypertrophic microglia also varied by FTLD subtype.ConclusionsThis study demonstrates regionally variable microglial involvement in FTLD and links this to underlying disease mechanisms. This supports investigation of microglial dysfunction in disease models and consideration of anti-senescence therapies in clinical trials.

Highlights

  • Microglial dysfunction is implicated in frontotemporal lobar degeneration (FTLD)

  • At onset of symptoms (AAO) did not differ across FTLD-TDP subtypes or between sporadic and genetic FTLD-TDPA cases

  • FTLD-fused in sarcoma (FUS) cases had a younger Age at death (AAD) compared with controls (p = 0.006), FTLD-TDP (p = 0.008), FTLD-tau (p = 0.004) and Alzheimer’s disease (AD) (p = 0.012) cases (Fig. 4b)

Read more

Summary

Introduction

Microglial dysfunction is implicated in frontotemporal lobar degeneration (FTLD). Studies have reported excessive microglial activation or senescence (dystrophy) in Alzheimer’s disease (AD), few have explored this in FTLD. We examined regional patterns of microglial burden, activation and dystrophy in sporadic and genetic FTLD, sporadic AD and controls. Frontotemporal lobar degeneration (FTLD) encompasses a large heterogeneous group of neurodegenerative diseases. The main subtypes of FTLD (FTLD-TDP, FTLD-tau and FTLD-FUS) reflect inclusions of TDP-43, tau and fused in sarcoma (FUS) proteins. FTLD-TDP is further divided into five pathological subtypes, A to E [2], based on the nature of TDP-43 inclusions present. FTLD-tau encompasses genetic cases with tau inclusions due to mutations in microtubule-associated protein tau (MAPT) (FTLD-MAPT), and sporadic cases including those with Pick bodies (FTLD-Picks), progressive supranuclear palsy (FTLD-PSP) or corticobasal degeneration (FTLD-CBD). FTLD-FUS cases are rare and contain FUS inclusions [3,4,5]

Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

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