4.7 Article

Keratins regulate colonic epithelial cell differentiation through the Notch1 signalling pathway

Journal

CELL DEATH AND DIFFERENTIATION
Volume 24, Issue 6, Pages 984-996

Publisher

NATURE PUBLISHING GROUP
DOI: 10.1038/cdd.2017.28

Keywords

-

Funding

  1. Academy of Finland [140759/126161, 218062]
  2. Sigrid Juselius Foundation
  3. Liv och Halsa foundation
  4. EU FP7 IRG
  5. Swedish Cultural Foundation
  6. Turku Doctoral Programme in Molecular Biosciences at AAU
  7. Turku Doctoral Programme of Biomedical Sciences
  8. Abo Akademi foundation
  9. Oskar Oflund Foundation
  10. K Albin Johansson foundation
  11. Magnus Ehrnrooth Foundation
  12. Waldemar von Frenckell Foundation
  13. Swedish Research Council
  14. EU
  15. AAU Center of Excellence of Cell Stress and Molecular Aging
  16. Academy of Finland (AKA) [218062, 218062] Funding Source: Academy of Finland (AKA)

Ask authors/readers for more resources

Keratins (K) are intermediate filament proteins important in stress protection and mechanical support of epithelial tissues. K8, K18 and K19 are the main colonic keratins, and K8-knockout (K8(-/-)) mice display a keratin dose-dependent hyperproliferation of colonic crypts and a colitis-phenotype. However, the impact of the loss of K8 on intestinal cell differentiation has so far been unknown. Here we show that K8 regulates Notch1 signalling activity and differentiation in the epithelium of the large intestine. Proximity ligation and immunoprecipitation assays demonstrate that K8 and Notch1 co-localize and interact in cell cultures, and in vivo in the colonic epithelial cells. K8 with its heteropolymeric partner K18 enhance Notch1 protein levels and activity in a dose dependent manner. The levels of the full-length Notch1 receptor (FLN), the Notch1 intracellular domain (NICD) and expression of Notch1 downstream target genes are reduced in the absence of K8, and the K8-dependent loss of Notch1 activity can be rescued with re-expression of K8/K18 in K8-knockout CRISPR/Cas9 Caco-2 cells protein levels. In vivo, K8 deletion with subsequent Notch1 downregulation leads to a shift in differentiation towards a goblet cell and enteroendocrine phenotype from an enterocyte cell fate. Furthermore, the K8(-/-) colonic hyperproliferation results from an increased number of transit amplifying progenitor cells in these mice. K8/K18 thus interact with Notch1 and regulate Notch1 signalling activity during differentiation of the colonic epithelium.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available