Show simple item record

dc.contributor.authorMarhuenda, E
dc.contributor.authorFabre, C
dc.contributor.authorZhang, C
dc.contributor.authorMartin-Fernandez, M
dc.contributor.authorIskratsch, T
dc.contributor.authorSaleh, A
dc.contributor.authorBauchet, L
dc.contributor.authorCambedouzou, J
dc.contributor.authorHugnot, J-P
dc.contributor.authorDuffau, H
dc.contributor.authorDennis, JW
dc.contributor.authorCornu, D
dc.contributor.authorBakalara, N
dc.date.accessioned2021-06-02T15:53:32Z
dc.date.available2021-03-23
dc.date.available2021-06-02T15:53:32Z
dc.date.issued2021-04-24
dc.identifier.urihttps://qmro.qmul.ac.uk/xmlui/handle/123456789/72238
dc.description.abstractBACKGROUND: Glioblastomas stem-like cells (GSCs) by invading the brain parenchyma, remains after resection and radiotherapy and the tumoral microenvironment become stiffer. GSC invasion is reported as stiffness sensitive and associated with altered N-glycosylation pattern. Glycocalyx thickness modulates integrins mechanosensing, but details remain elusive and glycosylation enzymes involved are unknown. Here, we studied the association between matrix stiffness modulation, GSC migration and MGAT5 induced N-glycosylation in fibrillar 3D context. METHOD: To mimic the extracellular matrix fibrillar microenvironments, we designed 3D-ex-polyacrylonitrile nanofibers scaffolds (NFS) with adjustable stiffnesses by loading multiwall carbon nanotubes (MWCNT). GSCs neurosphere were plated on NFSs, allowing GSCs migration and MGAT5 was deleted using CRISPR-Cas9. RESULTS: We found that migration of GSCs was maximum at 166 kPa. Migration rate was correlated with cell shape, expression and maturation of focal adhesion (FA), Epithelial to Mesenchymal Transition (EMT) proteins and (β1,6) branched N-glycan binding, galectin-3. Mutation of MGAT5 in GSC inhibited N-glycans (β1-6) branching, suppressed the stiffness dependence of migration on 166 kPa NFS as well as the associated FA and EMT protein expression. CONCLUSION: MGAT5 catalysing multibranched N-glycans is a critical regulators of stiffness induced invasion and GSCs mechanotransduction, underpinning MGAT5 as a serious target to treat cancer.en_US
dc.format.extent139 - ?
dc.languageeng
dc.relation.ispartofJ Exp Clin Cancer Res
dc.rightsAttribution 3.0 United States*
dc.rights.urihttp://creativecommons.org/licenses/by/3.0/us/*
dc.subject3D-nanofibre scaffolden_US
dc.subjectBiomaterialen_US
dc.subjectEMTen_US
dc.subjectFocal adhesionen_US
dc.subjectGalectinen_US
dc.subjectGlioblastomaen_US
dc.subjectGlycosylationen_US
dc.subjectMechanotransductionen_US
dc.subjectMgat5en_US
dc.subjectMigrationen_US
dc.subjectStiffnessen_US
dc.titleGlioma stem cells invasive phenotype at optimal stiffness is driven by MGAT5 dependent mechanosensing.en_US
dc.typeArticleen_US
dc.identifier.doi10.1186/s13046-021-01925-7
pubs.author-urlhttps://www.ncbi.nlm.nih.gov/pubmed/33894774en_US
pubs.issue1en_US
pubs.notesNot knownen_US
pubs.publication-statusPublished onlineen_US
pubs.volume40en_US
dcterms.dateAccepted2021-03-23


Files in this item

Thumbnail
Thumbnail

This item appears in the following Collection(s)

Show simple item record

Attribution 3.0 United States
Except where otherwise noted, this item's license is described as Attribution 3.0 United States