Show simple item record

dc.contributor.authorZagorac, Sen_US
dc.contributor.authorAlcala, Sen_US
dc.contributor.authorFernandez Bayon, Gen_US
dc.contributor.authorBou Kheir, Ten_US
dc.contributor.authorSchoenhals, Men_US
dc.contributor.authorGonzález-Neira, Aen_US
dc.contributor.authorFernandez Fraga, Men_US
dc.contributor.authorAicher, Aen_US
dc.contributor.authorHeeschen, Cen_US
dc.contributor.authorSainz, Ben_US
dc.date.accessioned2016-07-13T10:04:17Z
dc.date.available2016-04-27en_US
dc.date.issued2016-08-01en_US
dc.date.submitted2016-06-07T17:15:19.431Z
dc.identifier.urihttp://qmro.qmul.ac.uk/xmlui/handle/123456789/13464
dc.description.abstractPancreatic ductal adenocarcinoma (PDAC) and other carcinomas are hierarchically organized, with cancer stem cells (CSC) residing at the top of the hierarchy, where they drive tumor progression, metastasis, and chemoresistance. As CSC and non-CSC share an identical genetic background, we hypothesize that differences in epigenetics account for the striking functional differences between these two cell populations. Epigenetic mechanisms, such as DNA methylation, play an important role in maintaining pluripotency and regulating the differentiation of stem cells, but the role of DNA methylation in pancreatic CSC is obscure. In this study, we investigated the genome-wide DNA methylation profile of PDAC CSC, and we determined the importance of DNA methyltransferases for CSC maintenance and tumorigenicity. Using high-throughput methylation analysis, we discovered that sorted CSCs have a higher level of DNA methylation, regardless of the heterogeneity or polyclonality of the CSC populations present in the tumors analyzed. Mechanistically, CSC expressed higher DNMT1 levels than non-CSC. Pharmacologic or genetic targeting of DNMT1 in CSCs reduced their self-renewal and in vivo tumorigenic potential, defining DNMT1 as a candidate CSC therapeutic target. The inhibitory effect we observed was mediated in part through epigenetic reactivation of previously silenced miRNAs, in particular the miR-17-92 cluster. Together, our findings indicate that DNA methylation plays an important role in CSC biology and also provide a rationale to develop epigenetic modulators to target CSC plasticity and improve the poor outcome of PDAC patients. Cancer Res; 76(15); 4546-58. ©2016 AACR.en_US
dc.format.extent4546 - 4558en_US
dc.languageengen_US
dc.language.isoenen_US
dc.relation.ispartofCancer Resen_US
dc.subjectAdenocarcinomaen_US
dc.subjectCarcinoma, Pancreatic Ductalen_US
dc.subjectDNA (Cytosine-5-)-Methyltransferasesen_US
dc.subjectHumansen_US
dc.subjectMicroRNAsen_US
dc.subjectNeoplastic Stem Cellsen_US
dc.subjectUp-Regulationen_US
dc.titleDNMT1 Inhibition Reprograms Pancreatic Cancer Stem Cells via Upregulation of the miR-17-92 Cluster.en_US
dc.typeArticle
dc.rights.holder© 2016 American Association for Cancer Research.
dc.identifier.doi10.1158/0008-5472.CAN-15-3268en_US
pubs.author-urlhttps://www.ncbi.nlm.nih.gov/pubmed/27261509en_US
pubs.issue15en_US
pubs.notesNot knownen_US
pubs.publication-statusPublisheden_US
pubs.volume76en_US
dcterms.dateAccepted2016-04-27en_US


Files in this item

Thumbnail

This item appears in the following Collection(s)

Show simple item record