A2A Adenosine Receptor Antagonism Reverts the Blood-Brain Barrier Dysfunction Induced by Sleep Restriction

PLoS One. 2016 Nov 28;11(11):e0167236. doi: 10.1371/journal.pone.0167236. eCollection 2016.

Abstract

Chronic sleep restriction induces blood-brain barrier disruption and increases pro-inflammatory mediators in rodents. Those inflammatory mediators may modulate the blood-brain barrier and constitute a link between sleep loss and blood-brain barrier physiology. We propose that adenosine action on its A2A receptor may be modulating the blood-brain barrier dynamics in sleep-restricted rats. We administrated a selective A2A adenosine receptor antagonist (SCH58261) in sleep-restricted rats at the 10th day of sleep restriction and evaluated the blood-brain barrier permeability to dextrans coupled to fluorescein (FITC-dextrans) and Evans blue. In addition, we evaluated by western blot the expression of tight junction proteins (claudin-5, occludin, ZO-1), adherens junction protein (E-cadherin), A2A adenosine receptor, adenosine-synthesizing enzyme (CD73), and neuroinflammatory markers (Iba-1 and GFAP) in the cerebral cortex, hippocampus, basal nuclei and cerebellar vermis. Sleep restriction increased blood-brain barrier permeability to FITC-dextrans and Evans blue, and the effect was reverted by the administration of SCH58261 in almost all brain regions, excluding the cerebellum. Sleep restriction increased the expression of A2A adenosine receptor only in the hippocampus and basal nuclei without changing the expression of CD73 in all brain regions. Sleep restriction reduced the expression of tight junction proteins in all brain regions, except in the cerebellum; and SCH58261 restored the levels of tight junction proteins in the cortex, hippocampus and basal nuclei. Finally, sleep restriction induced GFAP and Iba-1 overexpression that was attenuated with the administration of SCH58261. These data suggest that the action of adenosine on its A2A receptor may have a crucial role in blood-brain barrier dysfunction during sleep loss probably by direct modulation of brain endothelial cell permeability or through a mechanism that involves gliosis with subsequent inflammation and increased blood-brain barrier permeability.

MeSH terms

  • Adenosine A2 Receptor Antagonists / pharmacology*
  • Animals
  • Blood-Brain Barrier / drug effects*
  • Blood-Brain Barrier / metabolism
  • Blood-Brain Barrier / pathology
  • Cell Membrane Permeability / drug effects*
  • Male
  • Rats
  • Rats, Wistar
  • Receptor, Adenosine A2A / chemistry*
  • Sleep Deprivation / physiopathology*

Substances

  • Adenosine A2 Receptor Antagonists
  • Receptor, Adenosine A2A

Grants and funding

This work was supported by Grant CB-2012-180792 from Consejo Nacional de Ciencia y Tecnología (CONACyT-SEP) to BGG and Fellowship 371992 from Consejo Nacional de Ciencia y Tecnología (CONACyT-SEP) to GHA. URL: http://www.conacyt.mx. None of the funding sources had any role in the study design, analysis or interpretation of the data, nor in writing the report.