ROS production and mitochondrial dysfunction driven by PU.1-regulated NOX4-p22phox activation in Aβ-induced retinal pigment epithelial cell injury

Theranostics. 2020 Sep 19;10(25):11637-11655. doi: 10.7150/thno.48064. eCollection 2020.

Abstract

Rationale: Amyloid β (Aβ) deposition, an essential pathological process in age-related macular degeneration (AMD), causes retinal pigment epithelium (RPE) degeneration driven mostly by oxidative stress. However, despite intense investigations, the extent to which overoxidation contributes to Aβ-mediated RPE damage and its potential mechanism has not been fully elucidated. Methods: We performed tandem mass-tagged (TMT) mass spectrometry (MS) and bioinformatic analysis of the RPE-choroid complex in an Aβ1-40-induced mouse model of retinal degeneration to obtain a comprehensive proteomic profile. Key regulators in this model were confirmed by reactive oxygen species (ROS) detection, mitochondrial ROS assay, oxygen consumption rate (OCR) measurement, gene knockout experiment, chromatin immunoprecipitation (ChIP), and luciferase assay. Results: A total of 4243 proteins were identified, 1069 of which were significantly affected by Aβ1-40 and found to be enriched in oxidation-related pathways by bioinformatic analysis. Moreover, NADPH oxidases were identified as hub proteins in Aβ1-40-mediated oxidative stress, as evidenced by mitochondrial dysfunction and reactive oxygen species overproduction. By motif and binding site analyses, we found that the transcription factor PU.1/Spi1 acted as a master regulator of the activation of NADPH oxidases, especially the NOX4-p22phox complex. Also, PU.1 silencing impeded RPE oxidative stress and mitochondrial dysfunction and rescued the retinal structure and function. Conclusion: Our study suggests that PU.1 is a novel therapeutic target for AMD, and the regulation of PU.1 expression represents a potentially novel approach against excessive oxidative stress in Aβ-driven RPE injury.

Keywords: NADPH oxidases; PU.1; age-related macular degeneration; amyloid β; retinal pigment epithelial cells.

Publication types

  • Observational Study
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Amyloid beta-Peptides / administration & dosage
  • Amyloid beta-Peptides / metabolism*
  • Animals
  • Computational Biology
  • Cross-Sectional Studies
  • Cytochrome b Group / analysis
  • Cytochrome b Group / genetics*
  • Cytochrome b Group / metabolism
  • Disease Models, Animal
  • Epithelial Cells
  • Gene Knockdown Techniques
  • Humans
  • Intravitreal Injections
  • Macular Degeneration / diagnosis
  • Macular Degeneration / pathology*
  • Male
  • Mice
  • Mitochondria / pathology
  • NADPH Oxidase 4 / analysis
  • NADPH Oxidase 4 / genetics*
  • NADPH Oxidase 4 / metabolism
  • NADPH Oxidases / analysis
  • NADPH Oxidases / genetics*
  • NADPH Oxidases / metabolism
  • Oxidative Stress / genetics
  • Peptide Fragments / administration & dosage
  • Peptide Fragments / metabolism*
  • Primary Cell Culture
  • Proteomics / methods
  • Proto-Oncogene Proteins / genetics
  • Proto-Oncogene Proteins / metabolism*
  • Reactive Oxygen Species / metabolism
  • Retinal Pigment Epithelium / cytology
  • Retinal Pigment Epithelium / diagnostic imaging
  • Retinal Pigment Epithelium / pathology*
  • Signal Transduction / genetics
  • Tandem Mass Spectrometry
  • Tomography, Optical Coherence
  • Trans-Activators / genetics
  • Trans-Activators / metabolism*

Substances

  • Amyloid beta-Peptides
  • Cytochrome b Group
  • Peptide Fragments
  • Proto-Oncogene Proteins
  • Reactive Oxygen Species
  • Trans-Activators
  • amyloid beta-protein (1-40)
  • proto-oncogene protein Spi-1
  • NADPH Oxidase 4
  • NADPH Oxidases
  • Nox4 protein, mouse
  • Cyba protein, mouse