Abstract

Glutamine, like glucose, is a major nutrient consumed by cancer cells, yet these cells undergo glutamine starvation in the cores of tumors, forcing them to evolve adaptive metabolic responses. Pharmacologically targeting glutamine metabolism or withdrawal has been exploited for therapeutic purposes, but does not always induce cancer cell death. The mechanism by which cancer cells adapt to resist glutamine starvation in cisplatin-resistant non-small-cell lung cancer (NSCLC) also remains uncertain. Here, we report the potential metabolic vulnerabilities of A549/DDP (drug-resistant human lung adenocarcinoma cell lines) cells, which were more easily killed by the iron chelator deferoxamine (DFO) during glutamine deprivation than their parental cisplatin-sensitive A549 cells. We demonstrate that phenotype resistance to cisplatin is accompanied by adaptive responses during glutamine deprivation partly via higher levels of autophagic activity and apoptosis resistance characteristics. Moreover, this adaptation could be explained by sustained glucose instead of glutamine-dominant complex II-dependent oxidative phosphorylation (OXPHOS). Further investigation revealed that cisplatin-resistant cells sustain OXPHOS partly via iron metabolism reprogramming during glutamine deprivation. This reprogramming might be responsible for mitochondrial iron-sulfur [Fe-S] cluster biogenesis, which has become an “Achilles’ heel,” rendering cancer cells vulnerable to DFO-induced autophagic cell death and apoptosis through c-Jun N-terminal kinase (JNK) signaling. Finally, in vivo studies using xenograft mouse models also confirmed the growth-slowing effect of DFO. In summary, we have elucidated the adaptive responses of cisplatin-resistant NSCLC cells, which balanced stability and plasticity to overcome metabolic reprogramming and permitted them to survive under stress induced by chemotherapy or glutamine starvation. In addition, for the first time, we show that suppressing the growth of cisplatin-resistant NSCLC cells via iron chelator-induced autophagic cell death and apoptosis was possible with DFO treatment. These findings provide a solid basis for targeting mitochondria iron metabolism in cisplatin-resistant NSCLC for therapeutic purposes, and it is plausible to consider that DFO facilitates in the improvement of treatment responses in cisplatin-resistant NSCLC patients.

Highlights

  • Lung cancer is the leading cause of cancer-related deaths worldwide

  • We observed the formation of more autophagosomes or initial autophagic vacuoles accompanied by fewer apoptotic bodies in A549/DDP cells than in A549 cells after glutamine deprivation for 48 h (Figure 2A)

  • To further characterize glutamine withdrawalinduced cell death between the two cell lines, we performed the Annexin V-FITC/PI apoptosis detection assay on the FCM, and the results showed that glutamine deprivation for 48 h notably elevated the apoptosis rate in A549 cells, but made no difference in A549/ DDP cells (Figure 2B)

Read more

Summary

Introduction

Lung cancer is the leading cause of cancer-related deaths worldwide. Non-small-cell lung cancer (NSCLC) represents 85% of lung cancer cases. Cisplatin is one of the most widely used chemotherapeutic agents; cisplatin resistance has become a major obstacle in clinical oncology [1,2,3]. Deoxyribonucleic acid (DNA) has been thought to be the primary target of cisplatin, but recent studies have shown that only a very tiny percentage of cisplatin (1%) interacts with nuclear DNA [4,5,6,7]. Most cisplatin interacts with mitochondria, which in turn has revealed the fundamental role of mitochondria in chemotherapy resistance [8, 9]

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