Reversal of β-Amyloid-Induced Microglial Toxicity In Vitro by Activation of Fpr2/3

Oxid Med Cell Longev. 2020 Jun 13:2020:2139192. doi: 10.1155/2020/2139192. eCollection 2020.

Abstract

Microglial inflammatory activity is thought to be a major contributor to the pathology of neurodegenerative conditions such as Alzheimer's disease (AD), and strategies to restrain their behaviour are under active investigation. Classically, anti-inflammatory approaches are aimed at suppressing proinflammatory mediator production, but exploitation of inflammatory resolution, the endogenous process whereby an inflammatory reaction is terminated, has not been fully investigated as a therapeutic approach in AD. In this study, we sought to provide proof-of-principle that the major proresolving actor, formyl peptide receptor 2, Fpr2, could be targeted to reverse microglial activation induced by the AD-associated proinflammatory stimulus, oligomeric β-amyloid (oAβ). The immortalised murine microglial cell line BV2 was employed as a model system to investigate the proresolving effects of the Fpr2 ligand QC1 upon oAβ-induced inflammatory, oxidative, and metabolic behaviour. Cytotoxic behaviour of BV2 cells was assessed through the use of cocultures with retinoic acid-differentiated human SH-SY5Y cells. Stimulation of BV2 cells with oAβ at 100 nM did not induce classical inflammatory marker production but did stimulate production of reactive oxygen species (ROS), an effect that could be reversed by subsequent treatment with the Fpr2 ligand QC1. Further investigation revealed that oAβ-induced ROS production was associated with NADPH oxidase activation and a shift in BV2 cell metabolic phenotype, activating the pentose phosphate pathway and NADPH production, changes that were again reversed by QC1 treatment. Microglial oAβ-stimulated ROS production was sufficient to induce apoptosis of bystander SH-SY5Y cells, an effect that could be prevented by QC1 treatment. In this study, we provide proof-of-concept data that indicate exploitation of the proresolving receptor Fpr2 can reverse damaging oAβ-induced microglial activation. Future strategies that are aimed at restraining neuroinflammation in conditions such as AD should examine proresolving actors as a mechanism to harness the brain's endogenous healing pathways and limit neuroinflammatory damage.

MeSH terms

  • Alzheimer Disease / metabolism
  • Alzheimer Disease / pathology
  • Amyloid beta-Peptides / toxicity*
  • Animals
  • Antioxidants / metabolism
  • Cell Line
  • Cell Respiration / drug effects
  • Enzyme Activation / drug effects
  • Humans
  • Inflammation / pathology
  • Mice
  • Microglia / drug effects
  • Microglia / metabolism
  • Microglia / pathology*
  • Mitochondria / drug effects
  • Mitochondria / metabolism
  • NADPH Oxidases / metabolism
  • Neurons / drug effects
  • Neurons / metabolism
  • Pentose Phosphate Pathway / drug effects
  • Reactive Oxygen Species / metabolism
  • Receptors, Formyl Peptide / agonists
  • Receptors, Formyl Peptide / metabolism*

Substances

  • Amyloid beta-Peptides
  • Antioxidants
  • Fpr3 protein, mouse
  • Reactive Oxygen Species
  • Receptors, Formyl Peptide
  • formyl peptide receptor 2, mouse
  • NADPH Oxidases