Abstract

Reprogrammed glucose metabolism and increased glycolysis have been implicated in tumor chemoresistance. The aim was to investigate the distinct roles of the glucose metabolites pyruvate and ATP in chemoresistance mechanisms, including cell death and proliferation. Our data showed higher glucose transporters in colorectal cancer (CRC) from non-responsive patients than those responsive to chemotherapy. Human CRC cell lines exposed to 5-fluorouracil (5-FU) displayed elevated cell viability and larger tumors in xenograft mouse models if cultured in high-glucose medium. Glucose conferred resistance to 5-FU-induced necroptosis via pyruvate scavenging of mitochondrial free radicals, whereas ATP replenishment had no effect on cell death. Glucose attenuated the 5-FU-induced G0/G1 shift but not the S phase arrest. Opposing effects were observed by glucose metabolites; ATP increased while pyruvate decreased the G0/G1 shift. Lastly, 5-FU-induced tumor spheroid destruction was prevented by glucose and pyruvate, but not by ATP. Our finding argues against ATP as the main effector for glucose-mediated chemoresistance and supports a key role of glycolytic pyruvate as an antioxidant for dual modes of action: necroptosis reduction and a cell cycle shift to a quiescent state.

Highlights

  • Colorectal cancer (CRC) is the second leading cause of cancer-related mortality

  • Tumor chemoresistance represents a significant problem in clinical management, and refractory tumors became more aggressive after first-line and targeted therapies [1]

  • Our results showed for the first time the distinct modes of action through which glucose metabolites ATP and pyruvate fueled tumor chemoresistance by manipulating the balance between cell death and proliferation

Read more

Summary

Introduction

Colorectal cancer (CRC) is the second leading cause of cancer-related mortality. Resistance to chemotherapies such as first-line genotoxic agents represents a significant problem in the clinical management of CRC. Studies on tumor chemoresistance have focused on genetic changes in association with drug metabolism and efflux pathways [2, 3]. Accumulating evidence suggests that altered cellular bioenergetic status, which contributes to cancer growth, is involved in chemoresistance [4, 5]. Recurrence of CRC after preoperative chemotherapy was correlated with high GLUT1 expression and anaerobic glycolysis in tumors [11,12,13]. Recent evidence showed that hyperglycemia and high sugar content in the intestinal lumen

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