Regulation of DNA damage and transcriptional output in the vasculature through a cytoglobin-HMGB2 axis

Redox Biol. 2023 Sep:65:102838. doi: 10.1016/j.redox.2023.102838. Epub 2023 Aug 9.

Abstract

Identifying novel regulators of vascular smooth muscle cell function is necessary to further understand cardiovascular diseases. We previously identified cytoglobin, a hemoglobin homolog, with myogenic and cytoprotective roles in the vasculature. The specific mechanism of action of cytoglobin is unclear but does not seem to be related to oxygen transport or storage like hemoglobin. Herein, transcriptomic profiling of injured carotid arteries in cytoglobin global knockout mice revealed that cytoglobin deletion accelerated the loss of contractile genes and increased DNA damage. Overall, we show that cytoglobin is actively translocated into the nucleus of vascular smooth muscle cells through a redox signal driven by NOX4. We demonstrate that nuclear cytoglobin heterodimerizes with the non-histone chromatin structural protein HMGB2. Our results are consistent with a previously unknown function by which a non-erythrocytic hemoglobin inhibits DNA damage and regulates gene programs in the vasculature by modulating the genome-wide binding of HMGB2.

Keywords: Carotid artery; Cytoglobin; DNA damage; DNA repair pathways; Hemoglobin; Hydrogen peroxide; Reactive oxygen species; Redox signal; Smooth muscle.

Publication types

  • Research Support, N.I.H., Extramural

MeSH terms

  • Animals
  • Cytoglobin / genetics
  • DNA Damage
  • Globins* / genetics
  • Globins* / metabolism
  • HMGB2 Protein* / genetics
  • HMGB2 Protein* / metabolism
  • Mice
  • Transcription Factors / genetics

Substances

  • Cytoglobin
  • Globins
  • HMGB2 Protein
  • Transcription Factors
  • Cygb protein, mouse