A novel target to reduce microglial inflammation and neuronal damage after deep hypothermic circulatory arrest

J Thorac Cardiovasc Surg. 2020 Jun;159(6):2431-2444.e7. doi: 10.1016/j.jtcvs.2019.06.115. Epub 2019 Aug 29.

Abstract

Background: Neuroinflammation acts as a contributor to neurologic deficits after deep hypothermic circulatory arrest. However, the molecular mechanism remains unclear. This study postulates that cold-inducible RNA-binding protein can promote deep hypothermic circulatory arrest-induced neuroinflammation.

Methods: Rats were randomly assigned into 3 groups (n = 5, each group): sham group, deep hypothermic circulatory arrest group, and deep hypothermic circulatory arrest + Cirp-/- group (Cirp-/- group). Murine microglial BV2 cells were administered by adeno-associated viral vectors containing cold-inducible RNA-binding protein small interference RNA or negative control small interference RNA at 2 days before 4-hour oxygen-glucose deprivation at 18°C. Microglial activation, cell death, neuroinflammation, and related protein expression were assessed in tissue samples and cell cultures.

Results: Cold-inducible RNA-binding protein was elevated along with evident neuroinflammation and neuronal damage in rats exposed to deep hypothermic circulatory arrest. In Cirp-/- rats, histologic injury (3.00 [interquartile range, 2.00-3.00] vs 1.00 [interquartile range, 1.00-1.50] neuropathological score, P < .001) and microglial activation (40 ± 4 vs 13 ± 7 CA1 area, P < .001) were alleviated after deep hypothermic circulatory arrest. With RNA-sequencing analysis, this associated with reduction of key proinflammatory cytokines induced by inhibiting Brd2-NF-κB signals. In BV2 cells treated with small interference RNA-cold-inducible RNA-binding protein, similar protective effects were observed, including decreased proinflammatory cytokines and cytotoxicity. Brd2-NF-κB signals were confirmed by the addition of Brd2 inhibitor JQ1. Notably, the conditioned medium from BV2 cells transfected with small interference RNA cold-inducible RNA-binding protein significantly reduced apoptosis in neural SH-SY5Y cells after oxygen-glucose deprivation, which was similar to that after JQ1 administration.

Conclusions: Enhanced cold-inducible RNA-binding protein in microglia aggravates neuronal injury by promoting the release of proinflammatory cytokines, which might be mediated through Brd2-NF-κB signals during deep hypothermic circulatory arrest.

Keywords: cold-inducible RNA-binding protein; deep hypothermic circulatory arrest; microglia; neuroinflammation.

Publication types

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

MeSH terms

  • Animals
  • Cell Death
  • Cell Line, Tumor
  • Circulatory Arrest, Deep Hypothermia Induced / adverse effects*
  • Cold Shock Proteins and Peptides / deficiency
  • Cold Shock Proteins and Peptides / genetics
  • Cold Shock Proteins and Peptides / metabolism*
  • Disease Models, Animal
  • Encephalitis / genetics
  • Encephalitis / metabolism
  • Encephalitis / pathology
  • Encephalitis / prevention & control*
  • Hippocampus / metabolism*
  • Hippocampus / pathology
  • Humans
  • Male
  • Mice
  • Microglia / metabolism*
  • Microglia / pathology
  • NF-kappa B / metabolism
  • Neurons / metabolism*
  • Neurons / pathology
  • RNA Interference*
  • RNA, Small Interfering / genetics
  • RNA, Small Interfering / metabolism*
  • RNA-Binding Proteins / genetics
  • RNA-Binding Proteins / metabolism*
  • Rats, Sprague-Dawley
  • Rats, Transgenic
  • Transcription Factors / metabolism

Substances

  • Brd2 protein, mouse
  • CIRBP protein, human
  • Cirbp protein, mouse
  • Cirbp protein, rat
  • Cold Shock Proteins and Peptides
  • NF-kappa B
  • RNA, Small Interfering
  • RNA-Binding Proteins
  • Transcription Factors