4.7 Article

Essential role of the histone lysine demethylase KDM4A in the biology of malignant pleural mesothelioma (MPM)

Journal

BRITISH JOURNAL OF CANCER
Volume 125, Issue 4, Pages 582-592

Publisher

SPRINGERNATURE
DOI: 10.1038/s41416-021-01441-7

Keywords

-

Categories

Funding

  1. International Mesothelioma Program at Brigham and Women's Hospital
  2. Department of Defense
  3. National Institutes of Health [R01-CA120528, R01-CA160979]
  4. Brent Leahey Fund

Ask authors/readers for more resources

The study revealed that KDM4A is overexpressed in MPM patients and inhibiting its enzymatic activity efficiently reduces cell growth and tumor volume. KDM4A expression is associated with pathways involved in cell growth and DNA repair.
Background Malignant pleural mesothelioma (MPM) is a highly aggressive cancer with a dismal prognosis. There is increasing interest in targeting chromatin regulatory pathways in difficult-to-treat cancers. In preliminary studies, we found that KDM4A (lysine-specific histone demethylase 4) was overexpressed in MPM. Methods KDM4A protein expression was determined by immunohistochemistry or immunoblotting. Functional inhibition of KDM4A by targeted knockdown and small molecule drugs was correlated to cell growth using cell lines and a xenograft mouse model. Gene expression profiling was performed to identify KDM4A-dependent signature pathways. Results Levels of KDM4A were found to be significantly elevated in MPM patients compared to normal mesothelial tissue. Inhibiting the enzyme activity efficiently reduced cell growth in vitro and reduced tumour growth in vivo. KDM4A inhibitor-induced apoptosis was further enhanced by the BH3 mimetic navitoclax. KDM4A expression was associated with pathways involved in cell growth and DNA repair. Interestingly, inhibitors of the DNA damage and replication checkpoint regulators CHK1 (prexasertib) and WEE1 (adavosertib) within the DNA double-strand break repair pathway, cooperated in the inhibition of cell growth. Conclusions The results establish a novel and essential role for KDM4A in growth in preclinical models of MPM and identify potential therapeutic approaches to target KDM4A-dependent vulnerabilities.

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