APOΕ4 lowers energy expenditure in females and impairs glucose oxidation by increasing flux through aerobic glycolysis

Mol Neurodegener. 2021 Sep 6;16(1):62. doi: 10.1186/s13024-021-00483-y.

Abstract

Background: Cerebral glucose hypometabolism is consistently observed in individuals with Alzheimer's disease (AD), as well as in young cognitively normal carriers of the Ε4 allele of Apolipoprotein E (APOE), the strongest genetic predictor of late-onset AD. While this clinical feature has been described for over two decades, the mechanism underlying these changes in cerebral glucose metabolism remains a critical knowledge gap in the field.

Methods: Here, we undertook a multi-omic approach by combining single-cell RNA sequencing (scRNAseq) and stable isotope resolved metabolomics (SIRM) to define a metabolic rewiring across astrocytes, brain tissue, mice, and human subjects expressing APOE4.

Results: Single-cell analysis of brain tissue from mice expressing human APOE revealed E4-associated decreases in genes related to oxidative phosphorylation, particularly in astrocytes. This shift was confirmed on a metabolic level with isotopic tracing of 13C-glucose in E4 mice and astrocytes, which showed decreased pyruvate entry into the TCA cycle and increased lactate synthesis. Metabolic phenotyping of E4 astrocytes showed elevated glycolytic activity, decreased oxygen consumption, blunted oxidative flexibility, and a lower rate of glucose oxidation in the presence of lactate. Together, these cellular findings suggest an E4-associated increase in aerobic glycolysis (i.e. the Warburg effect). To test whether this phenomenon translated to APOE4 humans, we analyzed the plasma metabolome of young and middle-aged human participants with and without the Ε4 allele, and used indirect calorimetry to measure whole body oxygen consumption and energy expenditure. In line with data from E4-expressing female mice, a subgroup analysis revealed that young female E4 carriers showed a striking decrease in energy expenditure compared to non-carriers. This decrease in energy expenditure was primarily driven by a lower rate of oxygen consumption, and was exaggerated following a dietary glucose challenge. Further, the stunted oxygen consumption was accompanied by markedly increased lactate in the plasma of E4 carriers, and a pathway analysis of the plasma metabolome suggested an increase in aerobic glycolysis.

Conclusions: Together, these results suggest astrocyte, brain and system-level metabolic reprogramming in the presence of APOE4, a 'Warburg like' endophenotype that is observable in young females decades prior to clinically manifest AD.

Keywords: APOE; Aerobic glycolysis; Alzheimer’s disease; Apolipoprotein E; Energy expenditure; Metabolism.

Publication types

  • Comparative Study
  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Adolescent
  • Adult
  • Aerobiosis*
  • Aged
  • Alzheimer Disease / diagnosis
  • Alzheimer Disease / genetics
  • Alzheimer Disease / metabolism
  • Animals
  • Apolipoprotein E4 / genetics
  • Apolipoprotein E4 / physiology*
  • Astrocytes / metabolism
  • Base Sequence
  • Brain Chemistry
  • Cells, Cultured
  • Early Diagnosis
  • Energy Metabolism
  • Female
  • Gas Chromatography-Mass Spectrometry
  • Gene Knock-In Techniques
  • Glucose / metabolism*
  • Glycolysis*
  • Humans
  • Metabolomics
  • Mice
  • Mice, Transgenic
  • Middle Aged
  • Nerve Tissue Proteins / genetics
  • Nerve Tissue Proteins / metabolism
  • Oxidation-Reduction
  • Oxidative Phosphorylation
  • Oxygen Consumption / genetics
  • Prodromal Symptoms*
  • Sex Characteristics
  • Single-Cell Analysis
  • Young Adult

Substances

  • Apolipoprotein E4
  • Nerve Tissue Proteins
  • Glucose

Associated data

  • ChiCTR/NCT03109661