SNX27 Deletion Causes Hydrocephalus by Impairing Ependymal Cell Differentiation and Ciliogenesis

J Neurosci. 2016 Dec 14;36(50):12586-12597. doi: 10.1523/JNEUROSCI.1620-16.2016.

Abstract

Hydrocephalus is a brain disorder derived from CSF accumulation due to defects in CSF clearance. Although dysfunctional apical cilia in the ependymal cell layer are causal to the onset of hydrocephalus, mechanisms underlying proper ependymal cell differentiation are largely unclear. SNX27 is a trafficking component required for normal brain function and was shown previously to suppress γ-secretase-dependent amyloid precursor protein and Notch cleavage. However, it was unclear how SNX27-dependent γ-secretase inhibition could contribute to brain development and pathophysiology. Here, we describe and characterize an Snx27-deleted mouse model for the ependymal layer defects of deciliation and hydrocephalus. SNX27 deficiency results in reductions in ependymal cells and cilia density, as well as severe postnatal hydrocephalus. Inhibition of Notch intracellular domain signaling with γ-secretase inhibitors reversed ependymal cells/cilia loss and dilation of lateral ventricles in Snx27-deficient mice, giving strong indication that Snx27 deletion triggers defects in ependymal layer formation and ciliogenesis through Notch hyperactivation. Together, these results suggest that SNX27 is essential for ependymal cell differentiation and ciliogenesis, and its deletion can promote hydrocephalus pathogenesis.

Significance statement: Down's syndrome (DS) in humans and mouse models has been shown previously to confer a high risk for the development of pathological hydrocephalus. Because we have previously described SNX27 as a component that is consistently downregulated in DS, we present here a robust Snx27-deleted mouse model that produces hydrocephalus and associated ciliary defects with complete penetrance. In addition, we find that γ-secretase/Notch modulation may be a candidate drug target in SNX27-associated hydrocephalus such as that observed in DS. Based on these findings, we anticipate that future study will determine whether modulation of a SNX27/Notch/γ-secretase pathway can also be of therapeutic interest to congenital hydrocephalus.

Keywords: SNX27; cilia; ependymal cell; hydrocephalus; trafficking; γ-secretase.

Publication types

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

MeSH terms

  • Adherens Junctions / pathology
  • Amyloid Precursor Protein Secretases / antagonists & inhibitors
  • Animals
  • Cell Differentiation / physiology*
  • Cilia / pathology
  • Cilia / physiology*
  • Ependyma / cytology
  • Ependyma / pathology*
  • Fibroblasts / drug effects
  • Glutathione / metabolism
  • Humans
  • Hydrocephalus / genetics*
  • Hydrocephalus / pathology*
  • Mice
  • Mice, Inbred C57BL
  • Mice, Knockout
  • Primary Cell Culture
  • Receptors, Notch / metabolism
  • Sorting Nexins / genetics
  • Sorting Nexins / physiology*

Substances

  • Receptors, Notch
  • Snx27 protein, mouse
  • Sorting Nexins
  • Amyloid Precursor Protein Secretases
  • Glutathione