0 540

Cited 54 times in

Regulation of bioenergetics through dual inhibition of aldehyde dehydrogenase and mitochondrial complex I suppresses glioblastoma tumorspheres

DC Field Value Language
dc.contributor.author강석구-
dc.contributor.author장종희-
dc.date.accessioned2018-08-28T17:21:18Z-
dc.date.available2018-08-28T17:21:18Z-
dc.date.issued2018-
dc.identifier.issn1522-8517-
dc.identifier.urihttps://ir.ymlib.yonsei.ac.kr/handle/22282913/162510-
dc.description.abstractBackground: Targeted approaches for treating glioblastoma (GBM) attempted to date have consistently failed, highlighting the imperative for treatment strategies that operate on different mechanistic principles. Bioenergetics deprivation has emerged as an effective therapeutic approach for various tumors. We have previously found that cancer cells preferentially utilize cytosolic NADH supplied by aldehyde dehydrogenase (ALDH) for ATP production through oxidative phosphorylation (OxPhos). This study is aimed at examining therapeutic responses and underlying mechanisms of dual inhibition of ALDH and OxPhos against GBM. Methods: For inhibition of ALDH and OxPhos, the corresponding inhibitors, gossypol and phenformin were used. Biological functions, including ATP levels, stemness, invasiveness, and viability, were evaluated in GBM tumorspheres (TSs). Gene expression profiles were analyzed using microarray data. In vivo anticancer efficacy was examined in a mouse orthotopic xenograft model. Results: Combined treatment of GBM TSs with gossypol and phenformin significantly reduced ATP levels, stemness, invasiveness, and cell viability. Consistently, this therapy substantially decreased expression of genes associated with stemness, mesenchymal transition, and invasion in GBM TSs. Supplementation of ATP using malate abrogated these effects, whereas knockdown of ALDH1L1 mimicked them, suggesting that disruption of ALDH-mediated ATP production is a key mechanism of this therapeutic combination. In vivo efficacy confirmed remarkable therapeutic responses to combined treatment with gossypol and phenformin. Conclusion: Our findings suggest that dual inhibition of tumor bioenergetics is a novel and effective strategy for the treatment of GBM.-
dc.description.statementOfResponsibilityrestriction-
dc.languageEnglish-
dc.publisherOxford University Press-
dc.relation.isPartOfNEURO-ONCOLOGY-
dc.rightsCC BY-NC-ND 2.0 KR-
dc.rightshttps://creativecommons.org/licenses/by-nc-nd/2.0/kr/-
dc.titleRegulation of bioenergetics through dual inhibition of aldehyde dehydrogenase and mitochondrial complex I suppresses glioblastoma tumorspheres-
dc.typeArticle-
dc.contributor.collegeCollege of Medicine-
dc.contributor.departmentDept. of Neurosurgery-
dc.contributor.googleauthorJunseong Park-
dc.contributor.googleauthorJin-Kyoung Shim-
dc.contributor.googleauthorJoon Hee Kang-
dc.contributor.googleauthorJunjeong Choi-
dc.contributor.googleauthorJong Hee Chang-
dc.contributor.googleauthorSoo-Youl Kim-
dc.contributor.googleauthorSeok-Gu Kang-
dc.identifier.doi10.1093/neuonc/nox243-
dc.contributor.localIdA00036-
dc.contributor.localIdA03470-
dc.relation.journalcodeJ02346-
dc.identifier.eissn1523-5866-
dc.identifier.pmid29294080-
dc.identifier.urlhttps://academic.oup.com/neuro-oncology/article/20/7/954/4773887-
dc.contributor.alternativeNameKang, Seok Gu-
dc.contributor.alternativeNameChang, Jong Hee-
dc.contributor.affiliatedAuthorKang, Seok Gu-
dc.contributor.affiliatedAuthorChang, Jong Hee-
dc.citation.volume20-
dc.citation.number7-
dc.citation.startPage954-
dc.citation.endPage965-
dc.identifier.bibliographicCitationNEURO-ONCOLOGY, Vol.20(7) : 954-965, 2018-
dc.identifier.rimsid60091-
dc.type.rimsART-
Appears in Collections:
1. College of Medicine (의과대학) > Dept. of Neurosurgery (신경외과학교실) > 1. Journal Papers

qrcode

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