4.6 Article

A potential target for liver cancer management, lysophosphatidic acid receptor 6 (LPAR6), is transcriptionally up-regulated by the NCOA3 coactivator

Journal

JOURNAL OF BIOLOGICAL CHEMISTRY
Volume 295, Issue 6, Pages 1474-1488

Publisher

ELSEVIER
DOI: 10.1074/jbc.RA119.009899

Keywords

epigenetics; transcription; hepatocellular carcinoma; cell signaling; hepatocyte growth factor; scatter factor (HGF; SF); G protein?coupled receptor (GPCR); histone acetyltransferase; lysophosphatidic acid receptor 6 (LPAR6); nuclear receptor coactivator 3 (NCOA3)

Funding

  1. National Key Research and Development Program of China [2018YFC1312300]
  2. National Natural Science Foundation of China [NSFC 81672782]
  3. Fundamental Research Funds for the Central Universities [20822041A4065]
  4. Sichuan Science and Technology Program [2018JY0018]
  5. 1.3.5 Project for Disciplines of Excellence, West China Hospital [ZYGD18003]
  6. Sichuan University

Ask authors/readers for more resources

Lysophosphatidic acid receptor 6 (LPAR6) is a G protein?coupled receptor that plays critical roles in cellular morphology and hair growth. Although LPAR6 overexpression is also critical for cancer cell proliferation, its role in liver cancer tumorigenesis and the underlying mechanism are poorly understood. Here, using liver cancer and matched paracancerous tissues, as well as functional assays including cell proliferation, quantitative real-time PCR, RNA-Seq, and ChIP assays, we report that LPAR6 expression is controlled by a mechanism whereby hepatocyte growth factor (HGF) suppresses liver cancer growth. We show that high LPAR6 expression promotes cell proliferation in liver cancer. More importantly, we find that LPAR6 is transcriptionally down-regulated by HGF treatment and that its transcriptional suppression depends on nuclear receptor coactivator 3 (NCOA3). We note that enrichment of NCOA3, which has histone acetyltransferase activity, is associated with histone 3 Lys-27 acetylation (H3K27ac) at the LPAR6 locus in response to HGF treatment, indicating that NCOA3 transcriptionally regulates LPAR6 through the HGF signaling cascade. Moreover, depletion of either LPAR6 or NCOA3 significantly inhibited tumor cell growth in vitro and in vivo (in mouse tumor xenograft assays), similar to the effect of the HGF treatment. Collectively, our findings indicate an epigenetic link between LPAR6 and HGF signaling in liver cancer cells, and suggest that LPAR6 can serve as a biomarker and new strategy for therapeutic interventions for managing liver cancer.

Authors

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

Reviews

Primary Rating

4.6
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available