Abstract

The nucleolus is the site of ribosome biogenesis and has been recently described as important sensor for a variety of cellular stressors. In the last two decades, it has been largely demonstrated that many chemotherapeutics act by inhibiting early or late rRNA processing steps with consequent alteration of ribosome biogenesis and activation of nucleolar stress response. The overall result is cell cycle arrest and/or apoptotic cell death of cancer cells. Our previously data demonstrated that ribosomal protein uL3 is a key sensor of nucleolar stress activated by common chemotherapeutic agents in cancer cells lacking p53. We have also demonstrated that uL3 status is associated to chemoresistance; down-regulation of uL3 makes some chemotherapeutic drugs ineffective. Here, we demonstrate that in colon cancer cells, the uL3 status affects rRNA synthesis and processing with consequent activation of uL3-mediated nucleolar stress pathway. Transcriptome analysis of HCT 116p53−/− cells expressing uL3 and of a cell sub line stably depleted of uL3 treated with Actinomycin D suggests a new extra-ribosomal role of uL3 in the regulation of autophagic process. By using confocal microscopy and Western blotting experiments, we demonstrated that uL3 acts as inhibitory factor of autophagic process; the absence of uL3 is associated to increase of autophagic flux and to chemoresistance. Furthermore, experiments conducted in presence of chloroquine, a known inhibitor of autophagy, indicate a role of uL3 in chloroquine-mediated inhibition of autophagy. On the basis of these results and our previous findings, we hypothesize that the absence of uL3 in cancer cells might inhibit cancer cell response to drug treatment through the activation of cytoprotective autophagy. The restoration of uL3 could enhance the activity of many drugs thanks to its pro-apoptotic and anti-autophagic activity.

Highlights

  • Colon cancer is the most common cause of cancer-related death in the development countries

  • To investigate whether the observed reduction of 47S pre-rRNA in HCT 116p53−/− cells overexpressing uL3 was associated to the alteration in rRNA synthesis or to the increase of 47S pre-rRNA turnover, we firstly monitored the rate of 47S pre-rRNA degradation in presence or in absence of uL3

  • Increasing evidence suggest that tumor resistance to anticancer therapies can be correlated to induction of nucleolar stress and up-regulation of autophagy in different cancer cells [20,28]

Read more

Summary

Introduction

Colon cancer is the most common cause of cancer-related death in the development countries. Some anticancer treatments can induce autophagic cell death, many recent studies have concluded that in tumors, autophagy works as a prosurvival process that eliminates damaged organelles and recycles macromolecules in response to metabolic stresses induced by anticancer drugs [21]. In this way, autophagy has emerged as mechanism of chemoresistance. This study presents new insights into our understanding of the link between drug inducing nucleolar stress and autophagy, and provides a potential therapeutic strategy for the management of tumors lacking functional p53 and having decreased levels of uL3

Alteration of uL3 Intracellular Levels Causes Defects in rRNA Processing
Effect of uL3 Status on 47S pre-rRNA Synthesis and Stability
Effect of uL3 Restoration in uL3 Deleted Cells on Cell Cycle and Apoptosis
Discussion
Material and Methods
RNA Extraction and RT-qPCR
Library Preparation and Deep Sequencing
Computational Analysis of Deep Sequencing Data
Nuclear Run-On
Immunofluorescence
Western Blot Analysis
Cell Cycle Analysis
Cell Death Assay
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