The E3 ubiquitin ligase RNF40 suppresses apoptosis in colorectal cancer cells

2019 | journal article. A publication with affiliation to the University of Göttingen.

Jump to: Cite & Linked | Documents & Media | Details | Version history

Cite this publication

​The E3 ubiquitin ligase RNF40 suppresses apoptosis in colorectal cancer cells​
Schneider, D.; Chua, R. L; Molitor, N.; Hamdan, F. H; Rettenmeier, E. M; Prokakis, E. & Mishra, V. K et al.​ (2019) 
Clinical Epigenetics11(1) art. 98​.​ DOI: https://doi.org/10.1186/s13148-019-0698-x 

Documents & Media

13148_2019_Article_698.pdf4.95 MBAdobe PDF

License

Published Version

Attribution 4.0 CC BY 4.0

Details

Authors
Schneider, Deborah; Chua, Robert L; Molitor, Nicole; Hamdan, Feda H; Rettenmeier, Eva M; Prokakis, Evangelos; Mishra, Vivek K; Kari, Vijayalakshmi; Wegwitz, Florian; Johnsen, Steven A; Kosinsky, Robyn L
Abstract
Abstract Background Colorectal cancer (CRC) is the fourth leading cause of cancer-related deaths worldwide, and deciphering underlying molecular mechanism is essential. The loss of monoubiquitinated histone H2B (H2Bub1) was correlated with poor prognosis of CRC patients and, accordingly, H2Bub1 was suggested as a tumor-suppressive mark. Surprisingly, our previous work revealed that the H2B ubiquitin ligase RING finger protein 40 (RNF40) might exert tumor-promoting functions. Here, we investigated the effect of RNF40 loss on tumorigenic features of CRC cells and their survival in vitro. Methods We evaluated the effects of RNF40 depletion in several human CRC cell lines in vitro. To evaluate cell cycle progression, cells were stained with propidium iodide and analyzed by flow cytometry. In addition, to assess apoptosis rates, caspase 3/7 activity was assessed in a Celigo® S-based measurement and, additionally, an Annexin V assay was performed. Genomic occupancy of H2Bub1, H3K79me3, and H3K27ac was determined by chromatin immunoprecipitation. Transcriptome-wide effects of RNF40 loss were evaluated based on mRNA-seq results, qRT-PCR, and Western blot. To rescue apoptosis-related effects, cells were treated with Z-VAD-FMK. Results Human CRC cell lines displayed decreased cell numbers in vitro after RNF40 depletion. While the differences in confluence were not mediated by changes in cell cycle progression, we discovered highly increased apoptosis rates after RNF40 knockdown due to elevated caspase 3/7 activity. This effect can be explained by reduced mRNA levels of anti-apoptotic and upregulation of pro-apoptotic BCL2 family members. Moreover, the direct occupancy of the RNF40-mediated H2B monoubiquitination was observed in the transcribed region of anti-apoptotic genes. Caspase inhibition by Z-VAD-FMK treatment rescued apoptosis in RNF40-depleted cells. However, knockdown cells still displayed decreased tumorigenic features despite the absence of apoptosis. Conclusions Our findings reveal that RNF40 is essential for maintaining tumorigenic features of CRC cells in vitro by controlling the expression of genes encoding central apoptotic regulators.
Issue Date
2019
Publisher
BioMed Central
Journal
Clinical Epigenetics 
Language
English

Reference

Citations


Social Media