4.2 Article

Histone lysine demethylase 4B regulates general and unique gene expression signatures in hypoxic cancer cells

Journal

MEDCOMM
Volume 2, Issue 3, Pages 414-429

Publisher

WILEY
DOI: 10.1002/mco2.85

Keywords

histone lysine demethylase 4B; colorectal cancer; ovarian cancer; renal cell carcinoma; tumor hypoxia

Funding

  1. NCATS NIH HHS [UL1 TR002366] Funding Source: Medline
  2. NICHD NIH HHS [U54 HD090216] Funding Source: Medline
  3. NIGMS NIH HHS [P30 GM122731, P20 GM104936] Funding Source: Medline
  4. NIH HHS [S10 OD021743] Funding Source: Medline

Ask authors/readers for more resources

The study reveals that KDM4B is overexpressed in various cancer types and may have different impacts on cancer prognosis. KDM4B regulates different cellular pathways under hypoxic and normoxic conditions, playing complex roles in cancer processes.
The hypoxic tumor microenvironment promotes tumor survival by inducing the expression of genes involved in angiogenesis and metastasis. As a direct target of hypoxia-inducible factor, lysine demethylase 4B (KDM4B) is overexpressed in multiple cancers, suggesting that a general KDM4B regulatory mechanism may exist in these cancer types. In this study, we sought to further investigate the general and unique roles of KDM4B in ovarian, colon, and renal cancer cells. We first identified a set of potential KDM4B targets shared by SKOV3ip.1, HCT116, and RCC4 cell lines, as well as numerous genes specifically regulated in each cell line. Through Gene Ontology, KEGG, and Oncobox pathway analyses, we found that KDM4B primarily regulated biosynthetic and cell cycle pathways in normoxia, whereas in hypoxia, it regulated pathways associated with inflammatory response and migration. TCGA data analyses reveal high expression of KDM4B in multiple cancer types and differential expression across cancer stages. Kaplan-Meier plots suggest that elevated KDM4B expression may contribute to a better or worse prognosis in a manner specific to each cancer type. Overall, our findings suggest that KDM4B plays complex roles in regulating multiple cancer processes, providing a useful resource for the future development of cancer therapies that target KDM4B expression.

Authors

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

Reviews

Primary Rating

4.2
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available