Your browser doesn't support javascript.
loading
Regulation of bioenergetics through dual inhibition of aldehyde dehydrogenase and mitochondrial complex I suppresses glioblastoma tumorspheres.
Park, Junseong; Shim, Jin-Kyoung; Kang, Joon Hee; Choi, Junjeong; Chang, Jong Hee; Kim, Soo-Youl; Kang, Seok-Gu.
Affiliation
  • Park J; Department of Neurosurgery, Brain Tumor Center, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea.
  • Shim JK; Department of Neurosurgery, Brain Tumor Center, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea.
  • Kang JH; Cancer Cell and Molecular Biology Branch, Research Institute, National Cancer Center, Goyang, Republic of Korea.
  • Choi J; College of Pharmacy, Yonsei Institute of Pharmaceutical Science, Yonsei University, Incheon, Republic of Korea.
  • Chang JH; Department of Neurosurgery, Brain Tumor Center, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea.
  • Kim SY; Cancer Cell and Molecular Biology Branch, Research Institute, National Cancer Center, Goyang, Republic of Korea.
  • Kang SG; Department of Neurosurgery, Brain Tumor Center, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea.
Neuro Oncol ; 20(7): 954-965, 2018 06 18.
Article in En | MEDLINE | ID: mdl-29294080
ABSTRACT

Background:

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.
Subject(s)

Full text: 1 Database: MEDLINE Therapeutic Methods and Therapies TCIM: Terapias_energeticas / Bioenergetica Main subject: Oxidative Phosphorylation / Neoplastic Stem Cells / Brain Neoplasms / Glioblastoma / Electron Transport Complex I / Aldehyde Dehydrogenase / Energy Metabolism Type of study: Prognostic_studies Language: En Journal: Neuro Oncol Year: 2018 Type: Article

Full text: 1 Database: MEDLINE Therapeutic Methods and Therapies TCIM: Terapias_energeticas / Bioenergetica Main subject: Oxidative Phosphorylation / Neoplastic Stem Cells / Brain Neoplasms / Glioblastoma / Electron Transport Complex I / Aldehyde Dehydrogenase / Energy Metabolism Type of study: Prognostic_studies Language: En Journal: Neuro Oncol Year: 2018 Type: Article