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

Abstract 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...

Full description

Bibliographic Details
Main Authors: Christian Linke, Markus Wösle, Anja Harder
Format: Article
Language:English
Published: BMC 2020-09-01
Series:BMC Cancer
Subjects:
NF1
p53
Online Access:http://link.springer.com/article/10.1186/s12885-020-07397-w
id doaj-3771d58ab6bd4b4599c5d920fcc31f01
record_format Article
spelling doaj-3771d58ab6bd4b4599c5d920fcc31f012020-11-25T02:52:20ZengBMCBMC Cancer1471-24072020-09-0120111110.1186/s12885-020-07397-wAnti-cancer agent 3-bromopyruvate reduces growth of MPNST and inhibits metabolic pathways in a representative in-vitro modelChristian Linke0Markus Wösle1Anja Harder2Faculty of Health Sciences, joint Faculty of the Brandenburg University of Technology Cottbus – Senftenberg, the Brandenburg Medical School Theodor Fontane and the University of PotsdamClinic for Radiotherapy and Radiation Oncology, Dessau City HospitalFaculty of Health Sciences, joint Faculty of the Brandenburg University of Technology Cottbus – Senftenberg, the Brandenburg Medical School Theodor Fontane and the University of PotsdamAbstract 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.http://link.springer.com/article/10.1186/s12885-020-07397-wMPNSTNF13-BrPAGlycolysisMitochondrial respirationp53
collection DOAJ
language English
format Article
sources DOAJ
author Christian Linke
Markus Wösle
Anja Harder
spellingShingle Christian Linke
Markus Wösle
Anja Harder
Anti-cancer agent 3-bromopyruvate reduces growth of MPNST and inhibits metabolic pathways in a representative in-vitro model
BMC Cancer
MPNST
NF1
3-BrPA
Glycolysis
Mitochondrial respiration
p53
author_facet Christian Linke
Markus Wösle
Anja Harder
author_sort Christian Linke
title Anti-cancer agent 3-bromopyruvate reduces growth of MPNST and inhibits metabolic pathways in a representative in-vitro model
title_short Anti-cancer agent 3-bromopyruvate reduces growth of MPNST and inhibits metabolic pathways in a representative in-vitro model
title_full Anti-cancer agent 3-bromopyruvate reduces growth of MPNST and inhibits metabolic pathways in a representative in-vitro model
title_fullStr Anti-cancer agent 3-bromopyruvate reduces growth of MPNST and inhibits metabolic pathways in a representative in-vitro model
title_full_unstemmed Anti-cancer agent 3-bromopyruvate reduces growth of MPNST and inhibits metabolic pathways in a representative in-vitro model
title_sort anti-cancer agent 3-bromopyruvate reduces growth of mpnst and inhibits metabolic pathways in a representative in-vitro model
publisher BMC
series BMC Cancer
issn 1471-2407
publishDate 2020-09-01
description Abstract 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.
topic MPNST
NF1
3-BrPA
Glycolysis
Mitochondrial respiration
p53
url http://link.springer.com/article/10.1186/s12885-020-07397-w
work_keys_str_mv AT christianlinke anticanceragent3bromopyruvatereducesgrowthofmpnstandinhibitsmetabolicpathwaysinarepresentativeinvitromodel
AT markuswosle anticanceragent3bromopyruvatereducesgrowthofmpnstandinhibitsmetabolicpathwaysinarepresentativeinvitromodel
AT anjaharder anticanceragent3bromopyruvatereducesgrowthofmpnstandinhibitsmetabolicpathwaysinarepresentativeinvitromodel
_version_ 1724730704419356672