File Download

There are no files associated with this item.

  Links for fulltext
     (May Require Subscription)
Supplementary

Article: WDR5 supports an N-myc transcriptional complex that drives a protumorigenic gene expression signature in neuroblastoma

TitleWDR5 supports an N-myc transcriptional complex that drives a protumorigenic gene expression signature in neuroblastoma
Authors
Issue Date2015
Citation
Cancer Research, 2015, v. 75, n. 23, p. 5143-5154 How to Cite?
Abstract© 2015 American Association for Cancer Research. MYCN gene amplification in neuroblastoma drives a gene expression program that correlates strongly with aggressive disease. Mechanistically, trimethylation of histone H3 lysine 4 (H3K4) at target gene promoters is a strict prerequisite for this transcriptional program to be enacted. WDR5 is a histone H3K4 presenter that has been found to have an essential role in H3K4 trimethylation. For this reason, in this study, we investigated the relationship between WDR5-mediated H3K4 trimethylation and N-Myc transcriptional programs in neuroblastoma cells. N-Myc upregulated WDR5 expression in neuroblastoma cells. Gene expression analysis revealed that WDR5 target genes included those with MYC-binding elements at promoters such as MDM2. We showed that WDR5 could form a protein complex at the MDM2 promoter with N-Myc, but not p53, leading to histone H3K4 trimethylation and activation of MDM2 transcription. RNAi-mediated attenuation of WDR5 upregulated expression of wild-type but not mutant p53, an effect associated with growth inhibition and apoptosis. Similarly, a small-molecule antagonist of WDR5 reduced N-Myc/WDR5 complex formation, N-Myc target gene expression, and cell growth in neuroblastoma cells. In MYCN-transgenic mice, WDR5 was overexpressed in precancerous ganglion and neuroblastoma cells compared with normal ganglion cells. Clinically, elevated levels of WDR5 in neuroblastoma specimens were an independent predictor of poor overall survival. Overall, our results identify WDR5 as a key cofactor for N-Myc-regulated transcriptional activation and tumorigenesis and as a novel therapeutic target for MYCN-amplified neuroblastomas.
Persistent Identifierhttp://hdl.handle.net/10722/251138
ISSN
2023 Impact Factor: 12.5
2023 SCImago Journal Rankings: 3.468
ISI Accession Number ID

 

DC FieldValueLanguage
dc.contributor.authorSun, Yuting-
dc.contributor.authorBell, Jessica L.-
dc.contributor.authorCarter, Daniel-
dc.contributor.authorGherardi, Samuele-
dc.contributor.authorPoulos, Rebecca C.-
dc.contributor.authorMilazzo, Giorgio-
dc.contributor.authorWong, Jason W.H.-
dc.contributor.authorAl-Awar, Rima-
dc.contributor.authorTee, Andrew E.-
dc.contributor.authorLiu, Pei Y.-
dc.contributor.authorLiu, Bing-
dc.contributor.authorAtmadibrata, Bernard-
dc.contributor.authorWong, Matthew-
dc.contributor.authorTrahair, Toby-
dc.contributor.authorZhao, Quan-
dc.contributor.authorShohet, Jason M.-
dc.contributor.authorHaupt, Ygal-
dc.contributor.authorSchulte, Johannes H.-
dc.contributor.authorBrown, Peter J.-
dc.contributor.authorArrowsmith, Cheryl H.-
dc.contributor.authorVedadi, Masoud-
dc.contributor.authorMacKenzie, Karen L.-
dc.contributor.authorHüttelmaier, Stefan-
dc.contributor.authorPerini, Giovanni-
dc.contributor.authorMarshall, Glenn M.-
dc.contributor.authorBraithwaite, Antony-
dc.contributor.authorLiu, Tao-
dc.date.accessioned2018-02-01T01:54:43Z-
dc.date.available2018-02-01T01:54:43Z-
dc.date.issued2015-
dc.identifier.citationCancer Research, 2015, v. 75, n. 23, p. 5143-5154-
dc.identifier.issn0008-5472-
dc.identifier.urihttp://hdl.handle.net/10722/251138-
dc.description.abstract© 2015 American Association for Cancer Research. MYCN gene amplification in neuroblastoma drives a gene expression program that correlates strongly with aggressive disease. Mechanistically, trimethylation of histone H3 lysine 4 (H3K4) at target gene promoters is a strict prerequisite for this transcriptional program to be enacted. WDR5 is a histone H3K4 presenter that has been found to have an essential role in H3K4 trimethylation. For this reason, in this study, we investigated the relationship between WDR5-mediated H3K4 trimethylation and N-Myc transcriptional programs in neuroblastoma cells. N-Myc upregulated WDR5 expression in neuroblastoma cells. Gene expression analysis revealed that WDR5 target genes included those with MYC-binding elements at promoters such as MDM2. We showed that WDR5 could form a protein complex at the MDM2 promoter with N-Myc, but not p53, leading to histone H3K4 trimethylation and activation of MDM2 transcription. RNAi-mediated attenuation of WDR5 upregulated expression of wild-type but not mutant p53, an effect associated with growth inhibition and apoptosis. Similarly, a small-molecule antagonist of WDR5 reduced N-Myc/WDR5 complex formation, N-Myc target gene expression, and cell growth in neuroblastoma cells. In MYCN-transgenic mice, WDR5 was overexpressed in precancerous ganglion and neuroblastoma cells compared with normal ganglion cells. Clinically, elevated levels of WDR5 in neuroblastoma specimens were an independent predictor of poor overall survival. Overall, our results identify WDR5 as a key cofactor for N-Myc-regulated transcriptional activation and tumorigenesis and as a novel therapeutic target for MYCN-amplified neuroblastomas.-
dc.languageeng-
dc.relation.ispartofCancer Research-
dc.titleWDR5 supports an N-myc transcriptional complex that drives a protumorigenic gene expression signature in neuroblastoma-
dc.typeArticle-
dc.description.naturelink_to_OA_fulltext-
dc.identifier.doi10.1158/0008-5472.CAN-15-0423-
dc.identifier.pmid26471359-
dc.identifier.scopuseid_2-s2.0-84955493969-
dc.identifier.volume75-
dc.identifier.issue23-
dc.identifier.spage5143-
dc.identifier.epage5154-
dc.identifier.eissn1538-7445-
dc.identifier.isiWOS:000365603000014-
dc.identifier.issnl0008-5472-

Export via OAI-PMH Interface in XML Formats


OR


Export to Other Non-XML Formats