Abstract

Simple SummaryDNA-damaging chemotherapeutics, such as platinum drugs, are cornerstones in cancer treatment. The efficacy of such treatment is intimately linked to the DNA repair capacity of the cancer cells, as DNA damage above a tolerable threshold culminates in cell death. Cancer cells often have deregulated DNA repair mechanisms, making them initially more sensitive to DNA-damaging chemotherapies. Unfortunately, over time, cancer cells often develop resistance to such treatments by rewiring their DNA damage response pathways. Here, we identify that targeting the recognized anti-cancer target 6-phosphofructo-2-kinase/fructose-2,6,-bisphophatase 3 (PFKFB3), commonly overexpressed in cancer, with the small molecule inhibitor KAN0438757, selectively sensitizes cancer cells to platinum drugs, including treatment-resistant cancer cells, while sparing normal cells. Mechanistically, PFKFB3 promotes tolerance to and the repair of platinum-induced DNA interstrand crosslinks (ICLs) through modulation of the Fanconi anemia (FA) DNA repair pathway. Thus targeting PFKFB3 opens up therapeutic possibilities to improve the efficacy of ICL-inducing cancer treatments.Replicative repair of interstrand crosslinks (ICL) generated by platinum chemotherapeutics is orchestrated by the Fanconi anemia (FA) repair pathway to ensure resolution of stalled replication forks and the maintenance of genomic integrity. Here, we identify novel regulation of FA repair by the cancer-associated glycolytic enzyme PFKFB3 that has functional consequences for replication-associated ICL repair and cancer cell survival. Inhibition of PFKFB3 displays a cancer-specific synergy with platinum compounds in blocking cell viability and restores sensitivity in treatment-resistant models. Notably, the synergies are associated with DNA-damage-induced chromatin association of PFKFB3 upon cancer transformation, which further increases upon platinum resistance. FA pathway activation triggers the PFKFB3 assembly into nuclear foci in an ATR- and FANCM-dependent manner. Blocking PFKFB3 activity disrupts the assembly of key FA repair factors and consequently prevents fork restart. This results in an incapacity to replicate cells to progress through S-phase, an accumulation of DNA damage in replicating cells, and fork collapse. We further validate PFKFB3-dependent regulation of FA repair in ex vivo cultures from cancer patients. Collectively, targeting PFKFB3 opens up therapeutic possibilities to improve the efficacy of ICL-inducing cancer treatments.

Highlights

  • The cellular pathways that promote the repair of DNA lesions and replication fork integrity are essential for genome stability [1]

  • We previously reported that PFKFB3 relocates to double strand break (DSB) upon ionizing radiation (IR), allowing for the assembly of key homologous recombination (HR) repair proteins into foci, required for functional DNA repair and cancer cell survival upon IR [25]

  • Inhibition of PFKFB3 leads to the defective recruitment of FANCM, BLM, FANCD2, and FANCI repair proteins, which are essential for successful interstrand crosslinks (ICL) repair during replication, recovery of DNA synthesis upon fork stalling, and cancer cell survival upon treatment with ICLinducing agents

Read more

Summary

Introduction

The cellular pathways that promote the repair of DNA lesions and replication fork integrity are essential for genome stability [1]. Upon FA pathway activation, ATR triggers the assembly of PFKFB3 into nuclear foci where PFKFB3 interacts with γH2AX and FANCD2 to enable the resolution of ICLs. Inhibition of PFKFB3 leads to the defective recruitment of FANCM, BLM, FANCD2, and FANCI repair proteins, which are essential for successful ICL repair during replication, recovery of DNA synthesis upon fork stalling, and cancer cell survival upon treatment with ICLinducing agents. Inhibition of PFKFB3 leads to the defective recruitment of FANCM, BLM, FANCD2, and FANCI repair proteins, which are essential for successful ICL repair during replication, recovery of DNA synthesis upon fork stalling, and cancer cell survival upon treatment with ICLinducing agents This modulatory mechanism of the FA response is associated with a PFKFB3 tumorigenic pattern of expression, as the synergistic anti-proliferative effect between PFKFB3 inhibition and crosslinking agents, carboplatin, and cisplatin, is specific to transformed cells

Cell Culture
Tumor Samples from High-Grade Ovarian Cancer Patients
Plasmid and siRNA Transfections
Establishment of shRNA Cell Lines
Quantitative Real-Time PCR (qRT-PCR) Analysis
Small-Molecule Inhibitors and Cytostatic
Cell Growth
End-Point Cell Viability Assays
Clonogenic Assays
2.10. Extracellular Flux Analysis
2.11. Flow Cytometry for Cell Cycle Progression of Replicating Cells
2.12. Immunoblotting and Whole Cell Protein Extraction
2.14. Immunoprecipitations
2.15. Fiber Assay
2.16. Immunofluorescence Microscopy
2.17. Statistics
Targeting PFKFB3 Enzymatic Activity Results in a Non-Reversible Cancer-Specific Synergy with Platinum Drugs
Platinum Drugs Do Not Induce a Preference for Glycolysis
Cancer-Specific PFKFB3 Chromatin
PFKFB3 Is Recruited into Foci upon Initiation of Replicative S-Phase FA Repair by FANCM and ATR
PFKFB3 Kinase Activity Promotes Assembly of FA Factors into Foci at the Chromatin
PFKFB3 Inhibition Impairs Replication Fork Restart upon ICL-Induction
Conclusions
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