Abstract

BackgroundAlthough radiation therapy represents a core cancer treatment modality, its efficacy is hampered by radioresistance. The effect of ionizing radiations (IRs) is well known regarding their ability to induce genetic alterations; however, their impact on the epigenome landscape in cancer, notably at the CpG dinucleotide resolution, remains to be further deciphered. In addition, no evidence is available regarding the effect of IRs on the DNA methylome profile according to the methionine dependency phenotype, which represents a hallmark of metabolic adaptation in cancer.MethodsWe used a case–control study design with a fractionated irradiation regimen on four cancerous cell lines representative of HCC (HepG2), melanoma (MeWo and MeWo-LC1, which exhibit opposed methionine dependency phenotypes), and glioblastoma (U251). We performed high-resolution genome-wide DNA methylome profiling using the MethylationEPIC BeadChip on baseline conditions, irradiated cell lines (cumulative dose of 10 Gy), and non-irradiated counterparts. We performed epigenome-wide association studies to assess the effect of IRs and methionine-dependency-oriented analysis by carrying out epigenome-wide conditional logistic regression. We looked for epigenome signatures at the locus and single-probe (CpG dinucleotide) levels and through enrichment analyses of gene ontologies (GO). The EpiMet project was registered under the ID#AAP-BMS_003_211.ResultsEWASs revealed shared GO annotation pathways associated with increased methylation signatures for several biological processes in response to IRs, including blood circulation, plasma membrane-bounded cell projection organization, cell projection organization, multicellular organismal process, developmental process, and animal organ morphogenesis. Epigenome-wide conditional logistic regression analysis on the methionine dependency phenotype highlighted several epigenome signatures related to cell cycle and division and responses to IR and ultraviolet light.ConclusionsIRs generated a variation in the methylation level of a high number of CpG probes with shared biological pathways, including those associated with cell cycle and division, responses to IRs, sustained angiogenesis, tissue invasion, and metastasis. These results provide insight on shared adaptive mechanisms of the epigenome in cancerous cell lines in response to IR. Future experiments should focus on the tryptic association between IRs, the initiation of a radioresistance phenotype, and their interaction with methionine dependency as a hallmark of metabolic adaptation in cancer.Graphical abstract

Highlights

  • Epigenome alterations, including modifications of DNA methylation, represent a hallmark of cancer initiation, progression, metastasis, and recurrence [1,2,3,4,5]

  • Dependency to methionine, called methionine dependency, is a metabolic adaptation that occurs in association with cancerous transformation, and its interaction with epigenetic modifications is increasingly recognized as a driver of tumorigenesis [2, 4, 6,7,8]

  • Principal component analyses for DNA methylome profile assessment We performed DNA methylome profiling of the 36 samples corresponding to nine flasks for each of the four studied cell lines

Read more

Summary

Introduction

Epigenome alterations, including modifications of DNA methylation, represent a hallmark of cancer initiation, progression, metastasis, and recurrence [1,2,3,4,5]. Dependency to methionine, called methionine dependency, is a metabolic adaptation that occurs in association with cancerous transformation, and its interaction with epigenetic modifications is increasingly recognized as a driver of tumorigenesis [2, 4, 6,7,8]. In this context, epigenetic modifications have been hypothesized as key drivers for tumor aggressiveness, with resistant phenotype to conventional chemo- and radiotherapy and an ability to develop metastases [5]. No evidence is available regarding the effect of IRs on the DNA methylome profile according to the methionine dependency phenotype, which represents a hallmark of metabolic adaptation in cancer

Objectives
Methods
Results
Discussion
Conclusion
Full Text
Paper version not known

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.