Please use this identifier to cite or link to this item: https://doi.org/10.1093/jnci/djv326
DC FieldValue
dc.titleST3GAL1-Associated Transcriptomic Program in Glioblastoma Tumor Growth, Invasion, and Prognosis
dc.contributor.authorChong Y.K.
dc.contributor.authorSandanaraj E.
dc.contributor.authorKoh L.W.H.
dc.contributor.authorThangaveloo M.
dc.contributor.authorTan M.S.Y.
dc.contributor.authorKoh G.R.H.
dc.contributor.authorToh T.B.
dc.contributor.authorLim G.G.Y.
dc.contributor.authorHolbrook J.D.
dc.contributor.authorKon O.L.
dc.contributor.authorNadarajah M.
dc.contributor.authorNg I.
dc.contributor.authorNg W.H.
dc.contributor.authorTan N.S.
dc.contributor.authorLim K.L.
dc.contributor.authorTang Soo Leng Carol
dc.contributor.authorAng B.T.
dc.date.accessioned2018-12-20T09:19:00Z
dc.date.available2018-12-20T09:19:00Z
dc.date.issued2016
dc.identifier.citationChong Y.K., Sandanaraj E., Koh L.W.H., Thangaveloo M., Tan M.S.Y., Koh G.R.H., Toh T.B., Lim G.G.Y., Holbrook J.D., Kon O.L., Nadarajah M., Ng I., Ng W.H., Tan N.S., Lim K.L., Tang Soo Leng Carol, Ang B.T. (2016). ST3GAL1-Associated Transcriptomic Program in Glioblastoma Tumor Growth, Invasion, and Prognosis. Journal of the National Cancer Institute 108 (2) : djv326. ScholarBank@NUS Repository. https://doi.org/10.1093/jnci/djv326
dc.identifier.issn278874
dc.identifier.urihttp://scholarbank.nus.edu.sg/handle/10635/150091
dc.description.abstractBackground: Cell surface sialylation is associated with tumor cell invasiveness in many cancers. Glioblastoma is the most malignant primary brain tumor and is highly infiltrative. ST3GAL1 sialyltransferase gene is amplified in a subclass of glioblastomas, and its role in tumor cell self-renewal remains unexplored. Methods: Self-renewal of patient glioma cells was evaluated using clonogenic, viability, and invasiveness assays. ST3GAL1 was identified from differentially expressed genes in Peanut Agglutinin-stained cells and validated in REMBRANDT (n = 390) and Gravendeel (n = 276) clinical databases. Gene set enrichment analysis revealed upstream processes. TGF? signaling on ST3GAL1 transcription was assessed using chromatin immunoprecipitation. Transcriptome analysis of ST3GAL1 knockdown cells was done to identify downstream pathways. A constitutively active FoxM1 mutant lacking critical anaphase-promoting complex/cyclosome ([APC/C]-Cdh1) binding sites was used to evaluate ST3Gal1-mediated regulation of FoxM1 protein. Finally, the prognostic role of ST3Gal1 was determined using an orthotopic xenograft model (3 mice groups comprising nontargeting and 2 clones of ST3GAL1 knockdown in NNI-11 [8 per group] and NNI-21 [6 per group]), and the correlation with patient clinical information. All statistical tests on patients' data were two-sided; other P values below are one-sided. Results: High ST3GAL1 expression defines an invasive subfraction with self-renewal capacity; its loss of function prolongs survival in a mouse model established from mesenchymal NNI-11 (P <. 001; groups of 8 in 3 arms: nontargeting, C1, and C2 clones of ST3GAL1 knockdown). ST3GAL1 transcriptomic program stratifies patient survival (hazard ratio [HR] = 2.47, 95% confidence interval [CI] = 1.72 to 3.55, REMBRANDT P = 1.92x10-8; HR = 2.89, 95% CI = 1.94 to 4.30, Gravendeel P = 1.05x10-11), independent of age and histology, and associates with higher tumor grade and T2 volume (P = 1.46x10-4). TGF? signaling, elevated in mesenchymal patients, correlates with high ST3GAL1 (REMBRANDT gliomacor = 0.31, P = 2.29x10-10; Gravendeel gliomacor = 0.50, P = 3.63x10-20). The transcriptomic program upon ST3GAL1 knockdown enriches for mitotic cell cycle processes. FoxM1 was identified as a statistically significantly modulated gene (P = 2.25x10-5) and mediates ST3Gal1 signaling via the (APC/C)-Cdh1 complex. Conclusions: The ST3GAL1-associated transcriptomic program portends poor prognosis in glioma patients and enriches for higher tumor grades of the mesenchymal molecular classification. We show that ST3Gal1-regulated self-renewal traits are crucial to the sustenance of glioblastoma multiforme growth. © 2015 The Author 2015.
dc.publisherOxford University Press
dc.sourceScopus
dc.typeArticle
dc.contributor.departmentCANCER SCIENCE INSTITUTE OF SINGAPORE
dc.contributor.departmentDEPT OF BIOCHEMISTRY
dc.contributor.departmentDEPT OF BIOLOGICAL SCIENCES
dc.contributor.departmentDEPT OF PHYSIOLOGY
dc.contributor.departmentDUKE-NUS MEDICAL SCHOOL
dc.description.doi10.1093/jnci/djv326
dc.description.sourcetitleJournal of the National Cancer Institute
dc.description.volume108
dc.description.issue2
dc.description.pagedjv326
dc.published.statePublished
dc.grant.idCIRG12nov024
dc.grant.idCIRGnov074
dc.grant.idNMRC/CSA/0058/2013
dc.grant.fundingagencyNMRC, National Medical Research Council
dc.grant.fundingagencyNMRC, National Medical Research Council
dc.grant.fundingagencyNMRC, National Medical Research Council
Appears in Collections:Staff Publications
Elements

Show simple item record
Files in This Item:
File Description SizeFormatAccess SettingsVersion 
djv326.pdf16.48 MBAdobe PDF

OPEN

PublishedView/Download

Google ScholarTM

Check

Altmetric


Items in DSpace are protected by copyright, with all rights reserved, unless otherwise indicated.