4.8 Article

The Histone-H3K4-Specific Demethylase KDM5B Binds to Its Substrate and Product through Distinct PHD Fingers

Journal

CELL REPORTS
Volume 6, Issue 2, Pages 325-335

Publisher

CELL PRESS
DOI: 10.1016/j.celrep.2013.12.021

Keywords

-

Categories

Funding

  1. NIH [GM096863, GM101664, R01HG007538, HL65440, GM068088, GM063873]
  2. CPRIT [RP110471]
  3. Welch Foundation [G1719]
  4. American Cancer Society [RSG-13-290-01-TBE, PF-13-085-01-DMC]
  5. NIH Postdoctoral Training Grant [T32AA007464]
  6. American Heart Association postdoctoral fellow
  7. Duncan Scholar Award
  8. Kimmel Scholar Award
  9. NATIONAL CANCER INSTITUTE [P30CA046934, T32CA009299] Funding Source: NIH RePORTER
  10. NATIONAL HEART, LUNG, AND BLOOD INSTITUTE [R01HL065440] Funding Source: NIH RePORTER
  11. NATIONAL HUMAN GENOME RESEARCH INSTITUTE [R01HG007538] Funding Source: NIH RePORTER
  12. NATIONAL INSTITUTE OF GENERAL MEDICAL SCIENCES [R01GM068088, R01GM101664, R01GM096863, R01GM063873] Funding Source: NIH RePORTER
  13. NATIONAL INSTITUTE ON ALCOHOL ABUSE AND ALCOHOLISM [T32AA007464] Funding Source: NIH RePORTER

Ask authors/readers for more resources

The histone lysine demethylase KDM5B regulates gene transcription and cell differentiation and is implicated in carcinogenesis. It contains multiple conserved chromatin-associated domains, including three PHD fingers of unknown function. Here, we show that the first and third, but not the second, PHD fingers of KDM5B possess histone binding activities. The PHD1 finger is highly specific for unmodified histone H3 (H3K4me0), whereas the PHD3 finger shows preference for the trimethylated histone mark H3K4me3. RNA-seq analysis indicates that KDM5B functions as a transcriptional repressor for genes involved in inflammatory responses, cell proliferation, adhesion, and migration. Biochemical analysis reveals that KDM5B associates with components of the nucleosome remodeling and deacetylase (NuRD) complex and may cooperate with the histone deacetylase 1 (HDAC1) in gene repression. KDM5B is downregulated in triple-negative breast cancer relative to estrogen-receptor-positive breast cancer. Overexpression of KDM5B in the MDA-MB 231 breast cancer cells suppresses cell migration and invasion, and the PHD1-H3K4me0 interaction is essential for inhibiting migration. These findings highlight tumor-suppressive functions of KDM5B in triple-negative breast cancer cells and suggest a multivalent mechanism for KDM5B-mediated transcriptional regulation.

Authors

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

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available