University of Zurich
Institute for Regenerative Medicine
Christian Tackenberg is the Scientific Head of Stem Cell Research at the Institute for Regenerative Medicine (IREM), University of Zurich. Further, he is a group leader at the Neuroscience Center Zurich and a lecturer for the Faculty of Science. His research focus is the application of cellular (stem cell-derived) models to uncover pathomechanisms of Alzheimer’s disease and the establishment of stem cell-based regenerative therapies for brain diseases such as stroke.

Moderator of 1 Session

Session Type
SYMPOSIUM
Date
Thu, 17.03.2022
Session Time
09:10 AM - 11:10 AM
Room
ONSITE: 113

Presenter of 1 Presentation

APOE2, E3 AND E4 DIFFERENTIALLY MODULATE HOMEOSTATIC MECHANISMS IN ISOGENIC IPSC-DERIVED ASTROCYTES

Session Type
SYMPOSIUM
Date
Thu, 17.03.2022
Session Time
09:10 AM - 11:10 AM
Room
ONSITE: 113
Lecture Time
10:10 AM - 10:25 AM

Abstract

Aims

The APOE4 allele is the major genetic risk factor for AD while APOE3 is defined as average risk and APOE2 is protective. Despite recent advances, the fundamental role of different APOE alleles in brain homeostasis is still poorly understood. Here, we aim to uncover the functional role of APOE2, E3 and E4 in human astrocytes.

Methods

We differentiated human APOE-isogenic iPSCs (E4, E3, E2 and APOE-knockout (KO)) to functional astrocytes (hereafter “iAstrocytes”). iAstrocytes at baseline and after activation with Interleukin-1β were analyzed for proteomic profiles using unlabelled mass spectrometry. iAstrocyte functions and properties, such as uptake of glutamate and beta-amyloid, release of cytokine and lipid/cholesterol metabolism were characterized by various assays.

Results

APOE4 iAstrocytes showed lowest capacity for uptake of beta-amyloid and glutamate, while uptake was highest in APOE2 and APOE-KO cells. We observed a genotype-dependent reduction of cholesterol and lipid metabolic and biosynthetic proteomic pathways, and an increase in immunoregulatory pathways at baseline (E4>E3>E2). Cholesterol efflux and biosynthesis were reduced in E4 iAstrocytes, and subcellular localization of cholesterol in lysosomes was increased. APOE4 iAstrocytes showed an increase in inflammatory proteomic pathway, accompanied by highest release of cytokines at baseline, while APOE2 and APOE-KO iAstrocytes show lowest cytokine release (APOE4>E3>E2>KO).

Conclusions

We show that APOE plays a major role in several physiological and metabolic processes in human astrocytes with APOE4 pushing iAstrocytes to a disease-relevant phenotype, causing dysregulated cholesterol/lipid homeostasis, increased inflammatory signalling and reduced β-amyloid uptake while APOE2 iAstrocytes show opposing (protective) effects.

Hide