Abstract

Simple SummaryCancer cells alter their metabolism to maintain their high need for energy, produce enough macromolecules for biosynthesis, and preserve their redox status. The investigation of cancer cell-specific metabolic alterations has vital importance to identify targets to be exploited for therapeutic development. The pentose phosphate pathway (PPP) is often highly activated in tumor cells to maintain redox level, as this pathway takes an important role in reactive oxygen species detoxification. PPP also yields ribose-5-phosphate, a five-carbon sugar essential for synthesizing nucleotides necessary for DNA replication and cell proliferation. In this study, we inhibited one of the key enzymes of this biochemical pathway and observed the main functions of this enzyme in breast cancer cells. We have demonstrated that inhibition of this enzyme reduces cell proliferation and leads to cell cycle arrest and apoptosis. Besides that, we showed that the inhibition of this enzyme causes an alteration in cellular metabolism. All these findings indicate that targeting this enzyme with specific pharmacological inhibitors is an effective strategy in fighting cancer.The pentose phosphate pathway (PPP) plays an essential role in the metabolism of breast cancer cells for the management of oxidative stress and the synthesis of nucleotides. 6-phosphogluconate dehydrogenase (6PGD) is one of the key enzymes of the oxidative branch of PPP and is involved in nucleotide biosynthesis and redox maintenance status. Here, we aimed to analyze the functional importance of 6PGD in a breast cancer cell model. Inhibition of 6PGD in MCF7 reduced cell proliferation and showed a significant decrease in glucose consumption and an increase in glutamine consumption, resulting in an important alteration in the metabolism of these cells. No difference in reactive oxygen species (ROS) production levels was observed after 6PGD inhibition, indicating that 6PGD, in contrast to glucose 6-phosphate dehydrogenase, is not involved in redox balance. We found that 6PGD inhibition also altered the stem cell characteristics and mammosphere formation capabilities of MCF7 cells, opening new avenues to prevent cancer recurrance after surgery or chemotherapy. Moreover, inhibition of 6PGD via chemical inhibitor S3 resulted in an induction of senescence, which, together with the cell cycle arrest and apoptosis induction, might be orchestrated by p53 activation. Therefore, we postulate 6PGD as a novel therapeutic target to treat breast cancer.

Highlights

  • Alteration in metabolism is one of the emerging hallmarks of cancer [1]

  • The pentose phosphate pathway (PPP) plays essential roles in nucleotide biosynthesis and the maintenance of redox status and in various aspects related to cancer cells viability, including proliferation, apoptosis, drug resistance, invasiveness, metastasis, and senescence [4,5,6,7,8]

  • We tested the functional role of 6phosphogluconate dehydrogenase (6PGD) in breast cancer using MCF7 cells as a cell model.Two siRNA sequences targeting different exonic regions of the 6PGD gene were tested and the knockdown of 6PGD was measured at mRNA level 72 h after transfection

Read more

Summary

Introduction

Alteration in metabolism is one of the emerging hallmarks of cancer [1]. Many observations made during the early period of cancer biology research revealed metabolic changes, such as the Warburg effect [2,3], to be a common feature of cancer cells. The pentose phosphate pathway (PPP) plays essential roles in nucleotide biosynthesis and the maintenance of redox status and in various aspects related to cancer cells viability, including proliferation, apoptosis, drug resistance, invasiveness, metastasis, and senescence [4,5,6,7,8]. Breast cancer cells are reported to be eight times more dependent on PPP compared to their non-cancerous counterparts to maintain their redox status [9]. Because of all these crucial aspects, cancer cells are significantly dependent on PPP to maintain their highly proliferative state [10,11]. The therapeutic potential of targeting PPP has attracted the interest of researchers in the field and both oxidative and non-oxidative branches of this metabolic pathway have been of keen interest to be used as a therapeutic drug target [5,10,12,13]

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