Your browser doesn't support javascript.
loading
Multiomics analysis of adaptation to repeated DNA damage in prostate cancer cells.
Challis, D; Lippis, T; Wilson, R; Wilkinson, E; Dickinson, J; Black, A; Azimi, I; Holloway, A; Taberlay, P; Brettingham-Moore, K.
Affiliation
  • Challis D; Tasmanian School of Medicine, University of Tasmania, Hobart, Tasmania, Australia.
  • Lippis T; Tasmanian School of Medicine, University of Tasmania, Hobart, Tasmania, Australia.
  • Wilson R; Central Science Laboratory, University of Tasmania, Hobart, Tasmania, Australia.
  • Wilkinson E; Menzies Institute of Medical Research, University of Tasmania, Hobart, Tasmania, Australia.
  • Dickinson J; Menzies Institute of Medical Research, University of Tasmania, Hobart, Tasmania, Australia.
  • Black A; Medical Oncology, Royal Hobart Hospital, Hobart, Tasmania, Australia.
  • Azimi I; School of Pharmacy and Pharmacology, University of Tasmania, Hobart, Tasmania, Australia.
  • Holloway A; Tasmanian School of Medicine, University of Tasmania, Hobart, Tasmania, Australia.
  • Taberlay P; Tasmanian School of Medicine, University of Tasmania, Hobart, Tasmania, Australia.
  • Brettingham-Moore K; Tasmanian School of Medicine, University of Tasmania, Hobart, Tasmania, Australia.
Epigenetics ; 18(1): 2214047, 2023 12.
Article de En | MEDLINE | ID: mdl-37196186
ABSTRACT
DNA damage is frequently utilized as the basis for cancer therapies; however, resistance to DNA damage remains one of the biggest challenges for successful treatment outcomes. Critically, the molecular drivers behind resistance are poorly understood. To address this question, we created an isogenic model of prostate cancer exhibiting more aggressive characteristics to better understand the molecular signatures associated with resistance and metastasis. 22Rv1 cells were repeatedly exposed to DNA damage daily for 6 weeks, similar to patient treatment regimes. Using Illumina Methylation EPIC arrays and RNA-seq, we compared DNA methylation and transcriptional profiles between the parental 22Rv1 cell line and the lineage exposed to prolonged DNA damage. Here we show that repeated DNA damage drives the molecular evolution of cancer cells to a more aggressive phenotype and identify molecular candidates behind this process. Total DNA methylation was increased while RNA-seq demonstrated these cells had dysregulated expression of genes involved in metabolism and the unfolded protein response (UPR) with Asparagine synthetase (ASNS) identified as central to this process. Despite the limited overlap between RNA-seq and DNA methylation, oxoglutarate dehydrogenase-like (OGDHL) was identified as altered in both data sets. Utilising a second approach we profiled the proteome in 22Rv1 cells following a single dose of radiotherapy. This analysis also highlighted the UPR in response to DNA damage. Together, these analyses identified dysregulation of metabolism and the UPR and identified ASNS and OGDHL as candidates for resistance to DNA damage. This work provides critical insight into molecular changes which underpin treatment resistance and metastasis.
Sujet(s)
Mots clés

Texte intégral: 1 Collection: 01-internacional Base de données: MEDLINE Sujet principal: Tumeurs de la prostate / Méthylation de l'ADN Type d'étude: Prognostic_studies Limites: Humans / Male Langue: En Journal: Epigenetics Sujet du journal: GENETICA Année: 2023 Type de document: Article Pays d'affiliation: Australie

Texte intégral: 1 Collection: 01-internacional Base de données: MEDLINE Sujet principal: Tumeurs de la prostate / Méthylation de l'ADN Type d'étude: Prognostic_studies Limites: Humans / Male Langue: En Journal: Epigenetics Sujet du journal: GENETICA Année: 2023 Type de document: Article Pays d'affiliation: Australie