4.7 Article

Autophagy inhibition-mediated epithelial-mesenchymal transition augments local myofibroblast differentiation in pulmonary fibrosis

Journal

CELL DEATH & DISEASE
Volume 10, Issue -, Pages -

Publisher

NATURE PUBLISHING GROUP
DOI: 10.1038/s41419-019-1820-x

Keywords

-

Categories

Funding

  1. Academy of Medical Sciences/the Wellcome Trust Springboard Award [SBF002/1038]
  2. Wessex Medical Trust
  3. AAIR Charity
  4. Gerald Kerkut Charitable Trust
  5. University of Southampton Central VC Scholarship Scheme
  6. National Natural Science Foundation of China [81772827]
  7. Medical Research Foundation [MRF-091-0003-RG-CONFO]
  8. China Scholarship Council
  9. Institute for Life Sciences PhD Studentship
  10. Bloodwise grant [12044]
  11. Francis Crick Institute from Cancer Research UK [FC001070]
  12. UK Medical Research Council [FC001070]
  13. Wellcome Trust [FC001070, 100638/Z/12/Z]
  14. Ludwig Institute for Cancer Research
  15. NIHR Oxford Biomedical Research Centre
  16. Wessex Clinical Research Network
  17. National Institute of Health Research, UK
  18. Medical Research Foundation [MRF-091-0003-RG-CONFO] Funding Source: researchfish
  19. Wellcome Trust [100638/Z/12/Z] Funding Source: Wellcome Trust

Ask authors/readers for more resources

Idiopathic pulmonary fibrosis (IPF), the prototypic progressive fibrotic interstitial lung disease, is thought to be a consequence of repetitive micro-injuries to an ageing, susceptible alveolar epithelium. Ageing is a risk factor for IPF and incidence has been demonstrated to increase with age. Decreased (macro) autophagy with age has been reported extensively in a variety of systems and diseases, including IPF. However, it is undetermined whether the role of faulty autophagy is causal or coincidental in the context of IPF. Here, we report that in alveolar epithelial cells inhibition of autophagy promotes epithelial-mesenchymal transition (EMT), a process implicated in embryonic development, wound healing, cancer metastasis and fibrosis. We further demonstrate that this is attained, at least in part, by increased p62/SQSTM1 expression that promotes p65/RELA mediated-transactivation of an EMT transcription factor, Snail2 (SNAI2), which not only controls EMT but also regulates the production of locally acting profibrogenic mediators. Our data suggest that reduced autophagy induces EMT of alveolar epithelial cells and can contribute to fibrosis via aberrant epithelial-fibroblast crosstalk.

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