File Download
  Links for fulltext
     (May Require Subscription)
Supplementary

Article: Distinct histone modifications denote early stress-induced drug tolerance in cancer

TitleDistinct histone modifications denote early stress-induced drug tolerance in cancer
Authors
KeywordsAcquired drug resistance
Dna methylation
Epigenetic reprogramming
Histone modification
Stress-induced resistance
Issue Date2018
Citation
Oncotarget, 2018, v. 9, n. 9, p. 8206-8222 How to Cite?
AbstractBesides somatic mutations or drug efflux, epigenetic reprogramming can lead to acquired drug resistance. We recently have identified early stress-induced multi-drug tolerant cancer cells termed induced drug-tolerant cells (IDTCs). Here, IDTCs were generated using different types of cancer cell lines; melanoma, lung, breast and colon cancer. A common loss of the H3K4me3 and H3K27me3 and gain of H3K9me3 mark was observed as a significant response to drug exposure or nutrient starvation in IDTCs. These epigenetic changes were reversible upon drug holidays. Microarray, qRT-PCR and protein expression data confirmed the up-regulation of histone methyltransferases (SETDB1 and SETDB2) which contribute to the accumulation of H3K9me3 concomitantly in the different cancer types. Genome-wide studies suggest that transcriptional repression of genes is due to concordant loss of H3K4me3 and regional increment of H3K9me3. Conversely, genome-wide CpG site-specific DNA methylation showed no common changes at the IDTC state. This suggests that distinct histone methylation patterns rather than DNA methylation are driving the transition from parental to IDTCs. In addition, silencing of SETDB1/2 reversed multi drug tolerance. Alterations of histone marks in early multi-drug tolerance with an increment in H3K9me3 and loss of H3K4me3/H3K27me3 is neither exclusive for any particular stress response nor cancer type specific but rather a generic response.
Persistent Identifierhttp://hdl.handle.net/10722/318702
PubMed Central ID

 

DC FieldValueLanguage
dc.contributor.authorEmran, Abdullah Al-
dc.contributor.authorMarzese, Diego M.-
dc.contributor.authorMenon, Dinoop Ravindran-
dc.contributor.authorStark, Mitchell S.-
dc.contributor.authorTorrano, Joachim-
dc.contributor.authorHammerlindl, Heinz-
dc.contributor.authorZhang, Gao-
dc.contributor.authorBrafford, Patricia-
dc.contributor.authorSalomon, Matthew P.-
dc.contributor.authorNelson, Nellie-
dc.contributor.authorHammerlindl, Sabrina-
dc.contributor.authorGupta, Deepesh-
dc.contributor.authorMills, Gordon B.-
dc.contributor.authorLu, Yiling-
dc.contributor.authorSturm, Richard A.-
dc.contributor.authorFlaherty, Keith-
dc.contributor.authorDave, S. B.Hoon-
dc.contributor.authorGabrielli, Brian-
dc.contributor.authorHerlyn, Meenhard-
dc.contributor.authorSchaider, Helmut-
dc.date.accessioned2022-10-11T12:24:21Z-
dc.date.available2022-10-11T12:24:21Z-
dc.date.issued2018-
dc.identifier.citationOncotarget, 2018, v. 9, n. 9, p. 8206-8222-
dc.identifier.urihttp://hdl.handle.net/10722/318702-
dc.description.abstractBesides somatic mutations or drug efflux, epigenetic reprogramming can lead to acquired drug resistance. We recently have identified early stress-induced multi-drug tolerant cancer cells termed induced drug-tolerant cells (IDTCs). Here, IDTCs were generated using different types of cancer cell lines; melanoma, lung, breast and colon cancer. A common loss of the H3K4me3 and H3K27me3 and gain of H3K9me3 mark was observed as a significant response to drug exposure or nutrient starvation in IDTCs. These epigenetic changes were reversible upon drug holidays. Microarray, qRT-PCR and protein expression data confirmed the up-regulation of histone methyltransferases (SETDB1 and SETDB2) which contribute to the accumulation of H3K9me3 concomitantly in the different cancer types. Genome-wide studies suggest that transcriptional repression of genes is due to concordant loss of H3K4me3 and regional increment of H3K9me3. Conversely, genome-wide CpG site-specific DNA methylation showed no common changes at the IDTC state. This suggests that distinct histone methylation patterns rather than DNA methylation are driving the transition from parental to IDTCs. In addition, silencing of SETDB1/2 reversed multi drug tolerance. Alterations of histone marks in early multi-drug tolerance with an increment in H3K9me3 and loss of H3K4me3/H3K27me3 is neither exclusive for any particular stress response nor cancer type specific but rather a generic response.-
dc.languageeng-
dc.relation.ispartofOncotarget-
dc.rightsThis work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.-
dc.subjectAcquired drug resistance-
dc.subjectDna methylation-
dc.subjectEpigenetic reprogramming-
dc.subjectHistone modification-
dc.subjectStress-induced resistance-
dc.titleDistinct histone modifications denote early stress-induced drug tolerance in cancer-
dc.typeArticle-
dc.description.naturepublished_or_final_version-
dc.identifier.doi10.18632/oncotarget.23654-
dc.identifier.pmid29492189-
dc.identifier.pmcidPMC5823586-
dc.identifier.scopuseid_2-s2.0-85041472961-
dc.identifier.volume9-
dc.identifier.issue9-
dc.identifier.spage8206-
dc.identifier.epage8222-
dc.identifier.eissn1949-2553-

Export via OAI-PMH Interface in XML Formats


OR


Export to Other Non-XML Formats