4.6 Article

HELLS, a chromatin remodeler is highly expressed in pancreatic cancer and downregulation of it impairs tumor growth and sensitizes to cisplatin by reexpressing the tumor suppressor TGFBR3

Journal

CANCER MEDICINE
Volume 10, Issue 1, Pages 350-364

Publisher

WILEY
DOI: 10.1002/cam4.3627

Keywords

chromatin remodeler; cisplatin; epigenetic; HELLS; pancreatic cancer

Categories

Funding

  1. National Natural Science Foundation of China [81472738]
  2. Hunan Provincial Natural Science Foundation [2020JJ4790]

Ask authors/readers for more resources

Pancreatic cancer is a highly malignant cancer with a poor prognosis, and chemotherapy with cisplatin is often the last option for patients with advanced disease. The chromatin remodeler HELLS has been identified as an oncogene in pancreatic cancer, with knockdown leading to increased sensitivity to cisplatin. This study found that HELLS silences the tumor suppressor TGFBR3 through epigenetic mechanisms, providing a potential biomarker for chemotherapy sensitivity in pancreatic cancer.
Pancreatic cancer (PC) is the most malignant cancer type in the digestive system with a poor prognosis. Chemotherapy such as cisplatin is the last chance for PC patients diagnosed with advanced or metastatic disease. Obtaining a deep understanding of the molecular mechanism underlying PC tumorigenesis and identifying optimal biomarkers to estimate chemotherapy sensitivity are essential for PC treatment. The chromatin remodeler HELLS was found to regulate various tumor suppressors through an epigenetic pathway in several cancers. We analyzed HELLS expression in clinical samples by Western blotting and immunohistochemical staining. Next, we identified the variation in tumor growth and cisplatin sensitivity after knockdown of HELLS and explored the downstream mediators of HELLS in PC via RNA-seq, chromatin immunoprecipitation, and gain- and loss-of-function assays. We found that HELLS is upregulated in PC tissues and correlates with advanced clinical stage and a poor prognosis, and the knockdown of HELLS leads to tumor growth arrest and increased sensitivity to cisplatin. Mechanistically, the tumor suppressor TGFBR3 is markedly reexpressed after HELLS knockdown; conversely, compromising TGFBR3 rescues HELLS knockdown-mediated effects in PC cells. Thus, our data provide evidence that HELLS can serve as a potential oncogene and suitable biomarker to evaluate chemotherapy sensitivity via epigenetically silencing the tumor suppressor TGFBR3 in PC.

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