Anti-cancer agent 3-bromopyruvate reduces growth of MPNST and inhibits metabolic pathways in a representative invitro model

Background: Anticancer compound 3-bromopyruvate (3-BrPA) suppresses cancer cell growth via targeting glycolytic and mitochondrial metabolism. The malignant peripheral nerve sheath tumor (MPNST), a very aggressive, therapy resistant, and Neurofibromatosis type 1 associated neoplasia, shows a high met...

Verfasser: Linke, Christian
Wösle, Markus
Harder, Anja
Dokumenttypen:Artikel
Medientypen:Text
Erscheinungsdatum:2020
Publikation in MIAMI:06.03.2023
Datum der letzten Änderung:06.03.2023
Angaben zur Ausgabe:[Electronic ed.]
Quelle:BMC Cancer 20 (2020) 896, 1-11
Schlagwörter:MPNST; NF1; 3-BrPA; Glycolysis; Mitochondrial respiration; p53; Starvation; Cell cycle; PA28; B8 fibroblasts
Fachgebiet (DDC):610: Medizin und Gesundheit
Lizenz:CC BY 4.0
Sprache:English
Förderung:Finanziert über die DEAL-Vereinbarung mit Wiley 2019-2022.
Förderer: Ministerium für Wissenschaft, Forschung und Kultur / Projektnummer: Grant No. 06-GeCa: H228–05/002/008, Gesundheitscampus Brandenburg
Format:PDF-Dokument
URN:urn:nbn:de:hbz:6-51009474623
Weitere Identifikatoren:DOI: 10.17879/51009505729
Permalink:https://nbn-resolving.de/urn:nbn:de:hbz:6-51009474623
Verwandte Dokumente:
  • ist identisch zu:
  • Onlinezugriff:10.1186_s12885-020-07397-w.pdf

    Background: Anticancer compound 3-bromopyruvate (3-BrPA) suppresses cancer cell growth via targeting glycolytic and mitochondrial metabolism. The malignant peripheral nerve sheath tumor (MPNST), a very aggressive, therapy resistant, and Neurofibromatosis type 1 associated neoplasia, shows a high metabolic activity and affected patients may therefore benefit from 3-BrPA treatment. To elucidate the specific mode of action, we used a controlled cell model overexpressing proteasome activator (PA) 28, subsequently leading to p53 inactivation and oncogenic transformation and therefore reproducing an important pathway in MPNST and overall tumor pathogenesis. Methods: Viability of MPNST cell lines S462, NSF1, and T265 in response to increasing doses (0–120 μM) of 3-BrPA was analyzed by CellTiter-Blue® assay. Additionally, we investigated viability, reactive oxygen species (ROS) production (dihydroethidium assay), nicotinamide adenine dinucleotide dehydrogenase activity (NADH-TR assay) and lactate production (lactate assay) in mouse B8 fibroblasts overexpressing PA28 in response to 3-BrPA application. For all experiments normal and nutrient deficient conditions were tested. MPNST cell lines were furthermore characterized immunohistochemically for Ki67, p53, bcl2, bcl6, cyclin D1, and p21. Results: MPNST significantly responded dose dependent to 3-BrPA application, whereby S462 cells were most responsive. Human control cells showed a reduced sensitivity. In PA28 overexpressing cancer cell model 3-BrPA application harmed mitochondrial NADH dehydrogenase activity mildly and significantly failed to inhibit lactate production. PA28 overexpression was associated with a functional glycolysis as well as a partial resistance to stress provoked by nutrient deprivation. 3-BrPA treatment was not associated with an increase of ROS. Starvation sensitized MPNST to treatment. Conclusions: Aggressive MPNST cells are sensitive to 3-BrPA therapy in-vitro with and without starvation. In a PA28 overexpression cancer cell model leading to p53 inactivation, thereby reflecting a key molecular feature in human NF1 associated MPNST, known functions of 3-BrPA to block mitochondrial activity and glycolysis were reproduced, however oncogenic cells displayed a partial resistance. To conclude, 3-BrPA was sufficient to reduce NF1 associated MPNST viability potentially due inhibition of glycolysis which should lead to the initiation of further studies and promises a potential benefit for NF1 patients.