Abstract

Simple SummaryProteasome inhibitors are currently used in the treatment of certain blood cancers, and clinical trials to treat solid tumors, including liver cancer, have also been conducted. However, different malignancies are not equally susceptible to proteasome inhibitors, and resistance to the drug may develop during the therapy. Here, we characterize the molecular mechanisms underlying the resilience of liver cancer cells to the proteasome inhibitor bortezomib. The results demonstrate that the activity of the eIF2α–ATF4 stress response pathway affects the viability of cells treated with bortezomib. We found that the pseudokinase TRIB3, an endogenous regulator of ATF4 and a gene highly expressed in liver cancer, resides predominantly at the same chromatin sites as ATF4 and constrains ATF4 activity. The survival of bortezomib-exposed hepatoma cells proved sensitive to TRIB3 overexpression and inactivation. Thus, TRIB3 is a novel factor contributing to bortezomib resistance of liver cancer cells.The proteasome is an appealing target for anticancer therapy and the proteasome inhibitor bortezomib has been approved for the treatment of several types of malignancies. However, the molecular mechanisms underlying cancer cell resistance to bortezomib remain poorly understood. In the current article, we investigate how modulation of the eIF2α–ATF4 stress pathway affects hepatoma cell response to bortezomib. Transcriptome profiling revealed that many ATF4 transcriptional target genes are among the most upregulated genes in bortezomib-treated HepG2 human hepatoma cells. While pharmacological enhancement of the eIF2α–ATF4 pathway activity results in the elevation of the activities of all branches of the unfolded protein response (UPR) and sensitizes cells to bortezomib toxicity, the suppression of ATF4 induction delays bortezomib-induced cell death. The pseudokinase TRIB3, an inhibitor of ATF4, is expressed at a high basal level in hepatoma cells and is strongly upregulated in response to bortezomib. To map genome-wide chromatin binding loci of TRIB3 protein, we fused a Flag tag to endogenous TRIB3 in HepG2 cells and performed ChIP-Seq. The results demonstrate that TRIB3 predominantly colocalizes with ATF4 on chromatin and binds to genomic regions containing the C/EBP–ATF motif. Bortezomib treatment leads to a robust enrichment of TRIB3 binding near genes induced by bortezomib and involved in the ER stress response and cell death. Disruption of TRIB3 increases C/EBP–ATF-driven transcription, augments ER stress and cell death upon exposure to bortezomib, while TRIB3 overexpression enhances cell survival. Thus, TRIB3, colocalizing with ATF4 and limiting its transcriptional activity, functions as a factor increasing resistance to bortezomib, while pharmacological over-activation of eIF2α–ATF4 can overcome the endogenous restraint mechanisms and sensitize cells to bortezomib.

Highlights

  • Liver cancer is the seventh most frequent cancer and the third leading cause of cancer-associated mortality in the world, representing a considerable need for improved treatment [1]

  • Cancer cells exposed to proteasome inhibitors are known to activate the unfolded protein response, which can facilitate proteostasis, or, if cellular adaptations prove insufficient, give way to ER stress-induced cell death [3]

  • We study the transcriptional response programs and cell viability of hepatocellular carcinoma cells exposed to bortezomib in combination with pharmacological compounds that perturb intracellular signaling to increase or decrease (ISRIB) the level of ATF4, a key transcriptional regulator of cellular stress responses

Read more

Summary

Introduction

Liver cancer is the seventh most frequent cancer and the third leading cause of cancer-associated mortality in the world, representing a considerable need for improved treatment [1]. The proteasome inhibitor bortezomib has been approved by FDA for use in the treatment of multiple myeloma and mantle cell lymphoma, and a number of phase I or II clinical trials have been carried out to explore the potential of bortezomib in other types of cancer, including solid tumors such as hepatocellular carcinoma (reviewed in [3]). As different malignancies have not proven sensitive to bortezomib, and resistance to the drug may develop over the course of therapy, solid tumors have not proven amenable to proteasome inhibitor therapy [4]. Cells exposed to bortezomib suffer from ER stress and initiate the unfolded protein response (UPR), a cellular program consisting of three branches that begin with ER overload sensor proteins ATF6, IRE1α and PERK. The initiator of the third UPR branch, PERK, phosphorylates eIF2α, thereby reducing translation initiation and triggering the integrated stress response (ISR) transcriptional program controlled by ATF4. The genes activated by ATF4 include those that facilitate adaptation to stress as well as cell death-promoting genes, and the activity of the eIF2α–ATF4 pathway is regulated by several signaling mechanisms, including negative feedback regulation by the pseudokinase TRIB3

Methods
Results
Conclusion
Full Text
Paper version not known

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