Abstract

Simple SummaryGlioblastoma multiforme (GBM) is the most common and deadly primary brain tumor in adults and one of the most aggressive cancers. The use of Poly ADP-Ribose Polymerase (PARP) inhibitors is being expanded as therapeutic alternative in multiple types of cancer beyond BRCA1/2 mutant breast and ovarian cancer. Here we have analyzed glioma cells’ traits that limit the efficacy of PARPi as anti-glioma agents and we found that PARPi triggered the synthesis of lipid droplets (LDs) that fueled glioma cells by inducing pro-survival lipid consumption. Notably, blocking Fatty Acids utilization by inhibition of β-oxidation with etomoxir, increased PARPi-induced glioma cell death while treatment with oleic acid (OA) prevented the anti-glioma effect of PARPi. We uncover a novel mechanism by which glioblastoma escapes to anti-tumor agents through metabolic reprogramming, inducing the synthesis and utilization of LDs as a pro-survival strategy in response to PARP inhibition.Glioblastoma multiforme (GBM) is the most common and aggressive primary brain tumor in adults. Poly (ADP-ribose) polymerase inhibitors (PARPi) represent a new class of anti-neoplastic drugs. In the current study, we have characterized the mechanism by which glioblastoma cells evade the effect of PARPi as anti-tumor agents. We have found that suppression of PARP activity exerts an anti-stemness effect and has a dual impact on autophagy, inducing its activation in the first 24 h (together with down-regulation of the pro-survival mTOR pathway) and preventing autophagosomes fusion to lysosomes at later time-points, in primary glioma cells. In parallel, PARPi triggered the synthesis of lipid droplets (LDs) through ACC-dependent activation of de novo fatty acids (FA) synthesis. Notably, inhibiting β-oxidation and blocking FA utilization, increased PARPi-induced glioma cell death while treatment with oleic acid (OA) prevented the anti-glioma effect of PARPi. Moreover, LDs fuel glioma cells by inducing pro-survival lipid consumption as confirmed by quantitation of oxygen consumption rates using Seahorse respirometry in presence or absence of OA. In summary, we uncover a novel mechanism by which glioblastoma escapes to anti-tumor agents through metabolic reprogramming, inducing the synthesis and utilization of LDs as a pro-survival strategy in response to PARP inhibition.

Highlights

  • Glioblastoma multiforme (GBM) is the most common primary malignant brain tumor in adults

  • In a previous study we have reported that PTEN-deficient glioblastoma cells were sensitive to Poly (ADP-ribose) polymerase inhibitors (PARPi) because they aggravated homologous recombination repair deficiency and displayed genomic instability leading to mitotic catastrophe [12]

  • We have observed that PARP inhibition exerts a double effect: On the one hand, PARP inactivation blunts the AKT/mTOR pathway, resulting in autophagy activation; on the other, by reprogramming their metabolism GBM cells transform a cytotoxic response (PARPi-induced cell death) into a pro-survival signal leading to the accumulation of fatty acids in the form of lipid droplets.To elucidate the mechanism connecting PARP inactivation with lipid droplets formation we have focused on the de novo free fatty acids (FFA) synthesis pathway

Read more

Summary

Introduction

Glioblastoma multiforme (GBM) is the most common primary malignant brain tumor in adults. PARP inhibitors exhibit anti-tumor activity in part due to their ability to induce synthetic lethality in cells deficient for homologous recombination repair [3,4,5,6]. PARPi-induced LDs formation occurs through synthesis de novo, dependent on the activation of Acyl-CoA-Carboxylase after AMPK suppression as a consequence of PARPi-induced ATP recovery. This mechanism occurs concomitantly with the increase in the transcriptional activation of fatty acid synthase (FASN). Preventing lipid synthesis and utilization by glioma cells increased PARPi-induced glioma cell death suggesting that avoidance of metabolic reprogramming might result in potentiation of PARPi antitumor efficacy

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