Show simple item record

dc.contributor.authorSimigdala, N
dc.contributor.authorGao, Q
dc.contributor.authorPancholi, S
dc.contributor.authorRoberg-Larsen, H
dc.contributor.authorZvelebil, M
dc.contributor.authorRibas, R
dc.contributor.authorFolkerd, E
dc.contributor.authorThompson, A
dc.contributor.authorBhamra, A
dc.contributor.authorDowsett, M
dc.contributor.authorMartin, L-A
dc.date.accessioned2017-03-01T11:16:13Z
dc.date.issued2016-06-01
dc.identifier.citationBreast cancer research : BCR, 2016, 18 (1), pp. 58 - ?
dc.identifier.issn1465-5411
dc.identifier.urihttps://repository.icr.ac.uk/handle/internal/414
dc.identifier.eissn1465-542X
dc.identifier.doi10.1186/s13058-016-0713-5
dc.description.abstractBACKGROUND: Therapies targeting estrogenic stimulation in estrogen receptor-positive (ER+) breast cancer (BC) reduce mortality, but resistance remains a major clinical problem. Molecular studies have shown few high-frequency mutations to be associated with endocrine resistance. In contrast, expression profiling of primary ER+ BC samples has identified several promising signatures/networks for targeting. METHODS: To identify common adaptive mechanisms associated with resistance to aromatase inhibitors (AIs), we assessed changes in global gene expression during adaptation to long-term estrogen deprivation (LTED) in a panel of ER+ BC cell lines cultured in 2D on plastic (MCF7, T47D, HCC1428, SUM44 and ZR75.1) or in 3D on collagen (MCF7) to model the stromal compartment. Furthermore, dimethyl labelling followed by LC-MS/MS was used to assess global changes in protein abundance. The role of target genes/proteins on proliferation, ER-mediated transcription and recruitment of ER to target gene promoters was analysed. RESULTS: The cholesterol biosynthesis pathway was the common upregulated pathway in the ER+ LTED but not the ER- LTED cell lines, suggesting a potential mechanism dependent on continued ER expression. Targeting the individual genes of the cholesterol biosynthesis pathway with siRNAs caused a 30-50 % drop in proliferation. Further analysis showed increased expression of 25-hydroxycholesterol (HC) in the MCF7 LTED cells. Exogenous 25-HC or 27-HC increased ER-mediated transcription and expression of the endogenous estrogen-regulated gene TFF1 in ER+ LTED cells but not in the ER- LTED cells. Additionally, recruitment of the ER and CREB-binding protein (CBP) to the TFF1 and GREB1 promoters was increased upon treatment with 25-HC and 27-HC. In-silico analysis of two independent studies of primary ER+ BC patients treated with neoadjuvant AIs showed that increased expression of MSMO1, EBP, LBR and SQLE enzymes, required for cholesterol synthesis and increased in our in-vitro models, was significantly associated with poor response to endocrine therapy. CONCLUSION: Taken together, these data provide support for the role of cholesterol biosynthesis enzymes and the cholesterol metabolites, 25-HC and 27-HC, in a novel mechanism of resistance to endocrine therapy in ER+ BC that has potential as a therapeutic target.
dc.formatElectronic
dc.format.extent58 - ?
dc.languageeng
dc.language.isoeng
dc.publisherBMC
dc.rights.urihttps://creativecommons.org/licenses/by/4.0
dc.subjectCell Line, Tumor
dc.subjectHumans
dc.subjectBreast Neoplasms
dc.subjectCholesterol
dc.subjectCholesterol Esters
dc.subjectProteome
dc.subjectReceptors, Estrogen
dc.subjectAntineoplastic Agents, Hormonal
dc.subjectEstrogens
dc.subjectPrognosis
dc.subjectTreatment Outcome
dc.subjectGene Expression Profiling
dc.subjectProteomics
dc.subjectCell Proliferation
dc.subjectGene Expression Regulation, Enzymologic
dc.subjectGene Expression Regulation, Neoplastic
dc.subjectRNA Interference
dc.subjectDrug Resistance, Neoplasm
dc.subjectPhenotype
dc.subjectFemale
dc.subjectBiosynthetic Pathways
dc.subjectTranscriptome
dc.subjectBiomarkers
dc.titleCholesterol biosynthesis pathway as a novel mechanism of resistance to estrogen deprivation in estrogen receptor-positive breast cancer.
dc.typeJournal Article
dcterms.dateAccepted2016-04-26
rioxxterms.versionofrecord10.1186/s13058-016-0713-5
rioxxterms.licenseref.urihttps://creativecommons.org/licenses/by/4.0
rioxxterms.licenseref.startdate2016-06
rioxxterms.typeJournal Article/Review
dc.relation.isPartOfBreast cancer research : BCR
pubs.issue1
pubs.notesNo embargo
pubs.organisational-group/ICR
pubs.organisational-group/ICR/Primary Group
pubs.organisational-group/ICR/Primary Group/ICR Divisions
pubs.organisational-group/ICR/Primary Group/ICR Divisions/Breast Cancer Research
pubs.organisational-group/ICR/Primary Group/ICR Divisions/Breast Cancer Research/Endocrinology
pubs.organisational-group/ICR/Primary Group/ICR Divisions/Molecular Pathology
pubs.organisational-group/ICR/Primary Group/ICR Divisions/Molecular Pathology/Endocrinology
pubs.organisational-group/ICR/Primary Group/ICR Divisions/Molecular Pathology/Endocrinology/Endocrinology (hon.)
pubs.organisational-group/ICR
pubs.organisational-group/ICR/Primary Group
pubs.organisational-group/ICR/Primary Group/ICR Divisions
pubs.organisational-group/ICR/Primary Group/ICR Divisions/Breast Cancer Research
pubs.organisational-group/ICR/Primary Group/ICR Divisions/Breast Cancer Research/Endocrinology
pubs.organisational-group/ICR/Primary Group/ICR Divisions/Molecular Pathology
pubs.organisational-group/ICR/Primary Group/ICR Divisions/Molecular Pathology/Endocrinology
pubs.organisational-group/ICR/Primary Group/ICR Divisions/Molecular Pathology/Endocrinology/Endocrinology (hon.)
pubs.publication-statusPublished
pubs.volume18
pubs.embargo.termsNo embargo
icr.researchteamEndocrinology
dc.contributor.icrauthorGao, Qiong
dc.contributor.icrauthorPancholi, Sunil
dc.contributor.icrauthorFolkerd, Elizabeth
dc.contributor.icrauthorMartin, Lesley-Ann


Files in this item

Thumbnail

This item appears in the following collection(s)

Show simple item record

https://creativecommons.org/licenses/by/4.0
Except where otherwise noted, this item's license is described as https://creativecommons.org/licenses/by/4.0