Your browser doesn't support javascript.
loading
Phenotypic screen for oxygen consumption rate identifies an anti-cancer naphthoquinone that induces mitochondrial oxidative stress.
Byrne, Frances L; Olzomer, Ellen M; Marriott, Gabriella R; Quek, Lake-Ee; Katen, Alice; Su, Jacky; Nelson, Marin E; Hart-Smith, Gene; Larance, Mark; Sebesfi, Veronica F; Cuff, Jeff; Martyn, Gabriella E; Childress, Elizabeth; Alexopoulos, Stephanie J; Poon, Ivan K; Faux, Maree C; Burgess, Antony W; Reid, Glen; McCarroll, Joshua A; Santos, Webster L; Quinlan, Kate Gr; Turner, Nigel; Fazakerley, Daniel J; Kumar, Naresh; Hoehn, Kyle L.
Affiliation
  • Byrne FL; School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia. Electronic address: frances.byrne@unsw.edu.au.
  • Olzomer EM; School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia.
  • Marriott GR; School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia.
  • Quek LE; School of Mathematics and Statistics, The University of Sydney, Sydney, Australia.
  • Katen A; School of Chemistry, University of New South Wales, Sydney, NSW, Australia.
  • Su J; School of Chemistry, University of New South Wales, Sydney, NSW, Australia.
  • Nelson ME; Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Australia.
  • Hart-Smith G; School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia.
  • Larance M; Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Australia.
  • Sebesfi VF; School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia.
  • Cuff J; School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia.
  • Martyn GE; School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia.
  • Childress E; Department of Chemistry and VT Center for Drug Discovery, Virginia Tech, Blacksburg, VA, USA.
  • Alexopoulos SJ; School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia.
  • Poon IK; Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia.
  • Faux MC; Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.
  • Burgess AW; Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.
  • Reid G; Concord Medical School, Asbestos Disease Research Institute, University of Sydney, Australia.
  • McCarroll JA; Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia.
  • Santos WL; Department of Chemistry and VT Center for Drug Discovery, Virginia Tech, Blacksburg, VA, USA.
  • Quinlan KG; School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia.
  • Turner N; School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia.
  • Fazakerley DJ; Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Australia.
  • Kumar N; School of Chemistry, University of New South Wales, Sydney, NSW, Australia.
  • Hoehn KL; School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia. Electronic address: k.hoehn@unsw.edu.au.
Redox Biol ; 28: 101374, 2020 01.
Article de En | MEDLINE | ID: mdl-31743887
ABSTRACT
A hallmark of cancer cells is their ability to reprogram nutrient metabolism. Thus, disruption to this phenotype is a potential avenue for anti-cancer therapy. Herein we used a phenotypic chemical library screening approach to identify molecules that disrupted nutrient metabolism (by increasing cellular oxygen consumption rate) and were toxic to cancer cells. From this screen we discovered a 1,4-Naphthoquinone (referred to as BH10) that is toxic to a broad range of cancer cell types. BH10 has improved cancer-selective toxicity compared to doxorubicin, 17-AAG, vitamin K3, and other known anti-cancer quinones. BH10 increases glucose oxidation via both mitochondrial and pentose phosphate pathways, decreases glycolysis, lowers GSHGSSG and NAPDH/NAPD+ ratios exclusively in cancer cells, and induces necrosis. BH10 targets mitochondrial redox defence as evidenced by increased mitochondrial peroxiredoxin 3 oxidation and decreased mitochondrial aconitase activity, without changes in markers of cytosolic or nuclear damage. Over-expression of mitochondria-targeted catalase protects cells from BH10-mediated toxicity, while the thioredoxin reductase inhibitor auranofin synergistically enhances BH10-induced peroxiredoxin 3 oxidation and cytotoxicity. Overall, BH10 represents a 1,4-Naphthoquinone with an improved cancer-selective cytotoxicity profile via its mitochondrial specificity.
Sujet(s)
Mots clés

Texte intégral: 1 Collection: 01-internacional Base de données: MEDLINE Sujet principal: Consommation d'oxygène / Naphtoquinones / Stress oxydatif / Mitochondries Limites: Humans Langue: En Journal: Redox Biol Année: 2020 Type de document: Article

Texte intégral: 1 Collection: 01-internacional Base de données: MEDLINE Sujet principal: Consommation d'oxygène / Naphtoquinones / Stress oxydatif / Mitochondries Limites: Humans Langue: En Journal: Redox Biol Année: 2020 Type de document: Article