Abstract

The alterations of ribosome biogenesis and protein synthesis play a direct role in the development of tumors. The accessibility and transcription of ribosomal genes is controlled at several levels, with their epigenetic regulation being one of the most important. Here we explored the JmjC domain-containing histone demethylase 1B (JHDM1B) function in the epigenetic control of rDNA transcription. Since JHDM1B is a negative regulator of gene transcription, we focused on the effects induced by JHDM1B knock-down (KD). We studied the consequences of stable inducible JHDM1B silencing in cell lines derived from transformed and untransformed mammary epithelial cells. In these cellular models, prolonged JHDM1B downregulation triggered a surge of 45S pre-rRNA transcription and processing, associated with a re-modulation of the H3K36me2 levels at rDNA loci and with changes in DNA methylation of specific CpG sites in rDNA genes. We also found that after JHDM1B KD, cells showed a higher ribosome content: which were engaged in mRNA translation. JHDM1B KD and the consequent stimulation of ribosomes biogenesis conferred more aggressive features to the tested cellular models, which acquired a greater clonogenic, staminal and invasive potential. Taken together, these data indicate that the reduction of JHDM1B leads to a more aggressive cellular phenotype in mammary gland cells, by virtue of its negative regulatory activity on ribosome biogenesis.

Highlights

  • Ribosome biogenesis, the process of ribosome production, is frequently up-regulated in cancer in order to respond to the increased demand of protein synthesis in highly proliferating cells

  • MDA-MB231-derived cells, after JmjC domain-containing histone demethylase 1B (JHDM1B) KD, showed increased global levels of histone H3K4me3, while in the MCF 10A the H3K36me2 histone mark increased after JHDM1B KD, supporting a role played by this demethylase in the modification of these specific lysine residues

  • In this study we observed that the TRC-driven JHDM1B silencing in our cellular models caused a surge in 45S pre-rRNA transcription, the rate-limiting step of ribosome biogenesis

Read more

Summary

Introduction

The process of ribosome production, is frequently up-regulated in cancer in order to respond to the increased demand of protein synthesis in highly proliferating cells. Great importance has recently been attributed to the negative regulatory function of JHDM1B through lysine H3K36me demethylation over a group of genes involved in senescence control, mapping at the Ink4a/Arf/Ink4b locus [7, 8] These genes encode for a series of critical cell cycle regulators: p16Ink4a and p15Ink4b prevent G1/S phase transition through the inhibition of cyclin D binding to cyclin-dependent kinase 4 and 6 (Cdk4/6); p14Arf protein leads to cell cycle arrest or apoptosis, through p53 www.impactjournals.com/oncotarget pathway activation [9, 10]. To verify the stated hypothesis, we investigated the role of JHDM1B-mediated negative control in ribosome biogenesis and its relevance to cancer, by focusing on the effects of JHDM1B downregulation on ribosome biogenesis and cell behavior in transformed and untransformed cells derived from mammary gland epithelium

Objectives
Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call