Diverse Functions of Retinoic Acid in Brain Vascular Development

J Neurosci. 2016 Jul 20;36(29):7786-801. doi: 10.1523/JNEUROSCI.3952-15.2016.

Abstract

As neural structures grow in size and increase metabolic demand, the CNS vasculature undergoes extensive growth, remodeling, and maturation. Signals from neural tissue act on endothelial cells to stimulate blood vessel ingression, vessel patterning, and acquisition of mature brain vascular traits, most notably the blood-brain barrier. Using mouse genetic and in vitro approaches, we identified retinoic acid (RA) as an important regulator of brain vascular development via non-cell-autonomous and cell-autonomous regulation of endothelial WNT signaling. Our analysis of globally RA-deficient embryos (Rdh10 mutants) points to an important, non-cell-autonomous function for RA in the development of the vasculature in the neocortex. We demonstrate that Rdh10 mutants have severe defects in cerebrovascular development and that this phenotype correlates with near absence of endothelial WNT signaling, specifically in the cerebrovasculature, and substantially elevated expression of WNT inhibitors in the neocortex. We show that RA can suppress the expression of WNT inhibitors in neocortical progenitors. Analysis of vasculature in non-neocortical brain regions suggested that RA may have a separate, cell-autonomous function in brain endothelial cells to inhibit WNT signaling. Using both gain and loss of RA signaling approaches, we show that RA signaling in brain endothelial cells can inhibit WNT-β-catenin transcriptional activity and that this is required to moderate the expression of WNT target Sox17. From this, a model emerges in which RA acts upstream of the WNT pathway via non-cell-autonomous and cell-autonomous mechanisms to ensure the formation of an adequate and stable brain vascular plexus.

Significance statement: Work presented here provides novel insight into important yet little understood aspects of brain vascular development, implicating for the first time a factor upstream of endothelial WNT signaling. We show that RA is permissive for cerebrovascular growth via suppression of WNT inhibitor expression in the neocortex. RA also functions cell-autonomously in brain endothelial cells to modulate WNT signaling and its downstream target, Sox17. The significance of this is although endothelial WNT signaling is required for neurovascular development, too much endothelial WNT signaling, as well as overexpression of its target Sox17, are detrimental. Therefore, RA may act as a "brake" on endothelial WNT signaling and Sox17 to ensure normal brain vascular development.

Keywords: VEGF; WNT; brain vascular development; cerebrovasculature; endothelial cell; retinoic acid.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Age Factors
  • Alcohol Oxidoreductases / deficiency
  • Alcohol Oxidoreductases / genetics
  • Animals
  • Brain / cytology*
  • Brain / embryology
  • Cell Differentiation
  • Cells, Cultured
  • Cerebral Ventricles / cytology*
  • Cerebral Ventricles / embryology
  • Embryo, Mammalian
  • Endothelial Cells / metabolism
  • Ephrins / genetics
  • Ephrins / metabolism
  • Gene Expression Regulation, Developmental / drug effects
  • Gene Expression Regulation, Developmental / genetics*
  • Humans
  • Mice
  • Mice, Inbred C57BL
  • Mice, Transgenic
  • Nerve Tissue Proteins / genetics
  • Nerve Tissue Proteins / metabolism
  • Retinoic Acid Receptor alpha / genetics
  • Retinoic Acid Receptor alpha / metabolism*
  • Tretinoin / metabolism*
  • Tretinoin / pharmacology
  • Wnt Signaling Pathway / drug effects
  • Wnt Signaling Pathway / genetics
  • Wnt Signaling Pathway / physiology*
  • beta Catenin / genetics
  • beta Catenin / metabolism
  • beta-Galactosidase / genetics
  • beta-Galactosidase / metabolism

Substances

  • CTNNB1 protein, mouse
  • Ephrins
  • Nerve Tissue Proteins
  • Rara protein, mouse
  • Retinoic Acid Receptor alpha
  • beta Catenin
  • Tretinoin
  • Alcohol Oxidoreductases
  • trans-retinol dehydrogenase
  • beta-Galactosidase