Abstract

BackgroundOncogenic KRAS mutations are prevalent in human cancers, but effective treatment of KRAS-mutant malignancies remains a major challenge in the clinic. Increasing evidence suggests that aberrant metabolism plays a central role in KRAS-driven oncogenic transformation. The aim of this study is to identify selective metabolic dependency induced by mutant KRAS and to exploit it for the treatment of the disease.MethodWe performed an integrated analysis of RNAi- and CRISPR-based functional genomic datasets (n = 5) to identify novel genes selectively required for KRAS-mutant cancer. We further screened a customized library of chemical inhibitors for candidates that are synthetic lethal with NOP56 depletion. Functional studies were carried out by genetic knockdown using siRNAs and shRNAs, knockout using CRISPR/Cas9, and/or pharmacological inhibition, followed by cell viability and apoptotic assays. Protein expression was determined by Western blot. Metabolic ROS was measured by flow cytometry-based quantification.ResultsWe demonstrated that nucleolar protein 5A (NOP56), a core component of small nucleolar ribonucleoprotein complexes (snoRNPs) with an essential role in ribosome biogenesis, confers a metabolic dependency by regulating ROS homeostasis in KRAS-mutant lung cancer cells and that NOP56 depletion causes synthetic lethal susceptibility to inhibition of mTOR. Mechanistically, cancer cells with reduced NOP56 are subjected to higher levels of ROS and rely on mTOR signaling to balance oxidative stress and survive. We also discovered that IRE1α-mediated unfolded protein response (UPR) regulates this process by activating mTOR through p38 MAPK. Consequently, co-targeting of NOP56 and mTOR profoundly enhances KRAS-mutant tumor cell death in vitro and in vivo.ConclusionsOur findings reveal a previously unrecognized mechanism in which NOP56 and mTOR cooperate to play a homeostatic role in the response to oxidative stress and suggest a new rationale for the treatment of KRAS-mutant cancers.

Highlights

  • Oncogenic Kirsten rat sarcoma viral oncogene homolog (KRAS) mutations are prevalent in human cancers, but effective treatment of KRAS-mutant malignancies remains a major challenge in the clinic

  • We demonstrated that nucleolar protein 5A (NOP56), a core component of small nucleolar ribonucleopro‐ tein complexes with an essential role in ribosome biogenesis, confers a metabolic dependency by regulat‐ ing reactive oxygen species (ROS) homeostasis in KRAS-mutant lung cancer cells and that Nucleolar protein 5A (NOP56) depletion causes synthetic lethal susceptibility to inhibition of Mechanistic target of rapamycin (mTOR)

  • We discovered that Inositol-requiring enzyme 1α (IRE1α)-mediated unfolded protein response (UPR) regulates this process by activating mTOR through p38 mitogenactivated protein kinase (MAPK)

Read more

Summary

Introduction

Oncogenic KRAS mutations are prevalent in human cancers, but effective treatment of KRAS-mutant malignancies remains a major challenge in the clinic. Oncogenic mutations in the RAS family (HRAS, KRAS, and NRAS) are the most common genetic alterations across human cancers and occur in approximately 25% of all tumors (COSMIC; http://cancer.sanger.ac.uk/cosmic). KRAS is the predominant isoform of the RAS family proteins activated by mutations (most frequently at codon 12, 13, and 61) in cancers and is responsible for 85% of all. Unlike NSCLC with less frequent oncogenic drivers (e.g., EGFR, ALK, MET1, and ROS1) that respond significantly to selective kinase inhibitors [2], effective therapies targeting KRAS-mutant cancers remains a challenge [2, 3]. Identification of new targets for innovative treatment strategies tailored to KRAS-mutant cancers still represents a pressing need [3]

Objectives
Methods
Results
Discussion
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