HSulf-1 deficiency dictates a metabolic reprograming of glycolysis and TCA cycle in ovarian cancer

dc.citation.firstpage33705
dc.citation.issueNumber32
dc.citation.journalTitleOncotarget
dc.citation.lastpage33719
dc.citation.volumeNumber6
dc.contributor.authorMondal, Susmita
dc.contributor.authorRoy, Debarshi
dc.contributor.authorCamacho-Pereira, Juliana
dc.contributor.authorKhurana, Ashwani
dc.contributor.authorChini, Eduardo
dc.contributor.authorYang, Lifeng
dc.contributor.authorBaddour, Joelle
dc.contributor.authorStilles, Katherine
dc.contributor.authorPadmabandu, Seth
dc.contributor.authorLeung, Sam
dc.contributor.authorKalloger, Steve
dc.contributor.authorGilks, Blake
dc.contributor.authorLowe, Val
dc.contributor.authorDierks, Thomas
dc.contributor.authorHammond, Edward
dc.contributor.authorDredge, Keith
dc.contributor.authorNagrath, Deepak
dc.contributor.authorShridhar, Viji
dc.date.accessioned2016-03-28T20:49:40Z
dc.date.available2016-03-28T20:49:40Z
dc.date.issued2015
dc.description.abstractWarburg effect has emerged as a potential hallmark of many cancers. However, the molecular mechanisms that led to this metabolic state of aerobic glycolysis, particularly in ovarian cancer (OVCA) have not been completely elucidated. HSulf-1 predominantly functions by limiting the bioavailability of heparan binding growth factors and hence their downstream signaling. Here we report that HSulf-1, a known putative tumor suppressor, is a negative regulator of glycolysis. Silencing of HSulf-1 expression in OV202 cell line increased glucose uptake and lactate production by upregulating glycolytic genes such as Glut1, HKII, LDHA, as well as metabolites. Conversely, HSulf-1 overexpression in TOV21G cells resulted in the down regulation of glycolytic enzymes and reduced glycolytic phenotype, supporting the role of HSulf-1 loss in enhanced aerobic glycolysis. HSulf-1 deficiency mediated glycolytic enhancement also resulted in increased inhibitory phosphorylation of pyruvate dehydrogenase (PDH) thus blocking the entry of glucose flux into TCA cycle. Consistent with this, metabolomic and isotope tracer analysis showed reduced glucose flux into TCA cycle. Moreover, HSulf-1 loss is associated with lower oxygen consumption rate (OCR) and impaired mitochondrial function. Mechanistically, lack of HSulf-1 promotes c-Myc induction through HB-EGF-mediated p-ERK activation. Pharmacological inhibition of c-Myc reduced HB-EGF induced glycolytic enzymes implicating a major role of c-Myc in loss of HSulf-1 mediated altered glycolytic pathway in OVCA. Similarly, PG545 treatment, an agent that binds to heparan binding growth factors and sequesters growth factors away from their ligand also blocked HB-EGF signaling and reduced glucose uptake in vivo in HSulf-1 deficient cells.
dc.identifier.citationMondal, Susmita, Roy, Debarshi, Camacho-Pereira, Juliana, et al.. "HSulf-1 deficiency dictates a metabolic reprograming of glycolysis and TCA cycle in ovarian cancer." <i>Oncotarget,</i> 6, no. 32 (2015) Impact Journals, LLC.: 33705-33719. http://dx.doi.org/10.18632/oncotarget.5605.
dc.identifier.doihttp://dx.doi.org/10.18632/oncotarget.5605
dc.identifier.urihttps://hdl.handle.net/1911/88656
dc.language.isoeng
dc.publisherImpact Journals, LLC.
dc.rightsAll content, except where otherwise noted, is licensed under aᅠCreative Commons Attribution 3.0 License.ᅠ
dc.rights.urihttp://creativecommons.org/licenses/by/3.0/
dc.subject.keywordHB-EGF
dc.subject.keywordHSulf-1
dc.subject.keywordPG545
dc.subject.keywordWarburg effect
dc.subject.keywordc-Myc
dc.subject.keywordovarian cancer
dc.titleHSulf-1 deficiency dictates a metabolic reprograming of glycolysis and TCA cycle in ovarian cancer
dc.typeJournal article
dc.type.dcmiText
dc.type.publicationpublisher version
Files
Original bundle
Now showing 1 - 1 of 1
Loading...
Thumbnail Image
Name:
5605-76613-3-PB.pdf
Size:
4.86 MB
Format:
Adobe Portable Document Format
Description: