A microglial activity state biomarker panel differentiates FTD-granulin and Alzheimer's disease patients from controls

Mol Neurodegener. 2023 Sep 29;18(1):70. doi: 10.1186/s13024-023-00657-w.

Abstract

Background: With the emergence of microglia-modulating therapies there is an urgent need for reliable biomarkers to evaluate microglial activation states.

Methods: Using mouse models and human induced pluripotent stem cell-derived microglia (hiMGL), genetically modified to yield the most opposite homeostatic (TREM2-knockout) and disease-associated (GRN-knockout) states, we identified microglia activity-dependent markers. Non-targeted mass spectrometry was used to identify proteomic changes in microglia and cerebrospinal fluid (CSF) of Grn- and Trem2-knockout mice. Additionally, we analyzed the proteome of GRN- and TREM2-knockout hiMGL and their conditioned media. Candidate marker proteins were tested in two independent patient cohorts, the ALLFTD cohort (GRN mutation carriers versus non-carriers), as well as the proteomic data set available from the EMIF-AD MBD study.

Results: We identified proteomic changes between the opposite activation states in mouse microglia and CSF, as well as in hiMGL cell lysates and conditioned media. For further verification, we analyzed the CSF proteome of heterozygous GRN mutation carriers suffering from frontotemporal dementia (FTD). We identified a panel of six proteins (FABP3, MDH1, GDI1, CAPG, CD44, GPNMB) as potential indicators for microglial activation. Moreover, we confirmed three of these proteins (FABP3, GDI1, MDH1) to be significantly elevated in the CSF of Alzheimer's (AD) patients. Remarkably, each of these markers differentiated amyloid-positive cases with mild cognitive impairment (MCI) from amyloid-negative individuals.

Conclusions: The identified candidate proteins reflect microglia activity and may be relevant for monitoring the microglial response in clinical practice and clinical trials modulating microglial activity and amyloid deposition. Moreover, the finding that three of these markers differentiate amyloid-positive from amyloid-negative MCI cases in the AD cohort suggests that these proteins associate with a very early immune response to seeded amyloid. This is consistent with our previous findings in the Dominantly Inherited Alzheimer's Disease Network (DIAN) cohort, where soluble TREM2 increases as early as 21 years before symptom onset. Moreover, in mouse models for amyloidogenesis, seeding of amyloid is limited by physiologically active microglia further supporting their early protective role. The biological functions of some of our main candidates (FABP3, CD44, GPNMB) also further emphasize that lipid dysmetabolism may be a common feature of neurodegenerative disorders.

Keywords: Alzheimer; Biomarker; Frontotemporal Dementia; Microglial activity; Patient stratification; Therapeutic modulation.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Alzheimer Disease* / metabolism
  • Amyloid beta-Peptides / metabolism
  • Amyloidogenic Proteins / metabolism
  • Animals
  • Biomarkers / metabolism
  • Culture Media, Conditioned / pharmacology
  • Frontotemporal Dementia* / genetics
  • Frontotemporal Dementia* / metabolism
  • Granulins / metabolism
  • Humans
  • Induced Pluripotent Stem Cells* / metabolism
  • Membrane Glycoproteins / genetics
  • Mice
  • Mice, Knockout
  • Microglia / metabolism
  • Proteome
  • Proteomics

Substances

  • Amyloid beta-Peptides
  • Amyloidogenic Proteins
  • Biomarkers
  • Culture Media, Conditioned
  • GPNMB protein, human
  • Granulins
  • Membrane Glycoproteins
  • Proteome
  • Trem2 protein, mouse