Targeting sphingolipid metabolism with the sphingosine kinase inhibitor SKI-II overcomes hypoxia-induced chemotherapy resistance in glioblastoma cells: effects on cell death, self-renewal, and invasion

Abstract Background Glioblastoma patients commonly develop resistance to temozolomide chemotherapy. Hypoxia, which supports chemotherapy resistance, favors the expansion of glioblastoma stem cells (GSC), contributing to tumor relapse. Because of a deregulated sphingolipid metabolism, glioblastoma ti...

Full description

Bibliographic Details
Main Authors: Nadia Sousa, Carsten Geiß, Laura Bindila, Ingo Lieberwirth, Ella Kim, Anne Régnier-Vigouroux
Format: Article
Language:English
Published: BMC 2023-08-01
Series:BMC Cancer
Subjects:
Online Access:https://doi.org/10.1186/s12885-023-11271-w
_version_ 1797452284391587840
author Nadia Sousa
Carsten Geiß
Laura Bindila
Ingo Lieberwirth
Ella Kim
Anne Régnier-Vigouroux
author_facet Nadia Sousa
Carsten Geiß
Laura Bindila
Ingo Lieberwirth
Ella Kim
Anne Régnier-Vigouroux
author_sort Nadia Sousa
collection DOAJ
description Abstract Background Glioblastoma patients commonly develop resistance to temozolomide chemotherapy. Hypoxia, which supports chemotherapy resistance, favors the expansion of glioblastoma stem cells (GSC), contributing to tumor relapse. Because of a deregulated sphingolipid metabolism, glioblastoma tissues contain high levels of the pro-survival sphingosine-1-phosphate and low levels of the pro-apoptotic ceramide. The latter can be metabolized to sphingosine-1-phosphate by sphingosine kinase (SK) 1 that is overexpressed in glioblastoma. The small molecule SKI-II inhibits SK and dihydroceramide desaturase 1, which converts dihydroceramide to ceramide. We previously reported that SKI-II combined with temozolomide induces caspase-dependent cell death, preceded by dihydrosphingolipids accumulation and autophagy in normoxia. In the present study, we investigated the effects of a low-dose combination of temozolomide and SKI-II under normoxia and hypoxia in glioblastoma cells and patient-derived GCSs. Methods Drug synergism was analyzed with the Chou-Talalay Combination Index method. Dose–effect curves of each drug were determined with the Sulforhodamine B colorimetric assay. Cell death mechanisms and autophagy were analyzed by immunofluorescence, flow cytometry and western blot; sphingolipid metabolism alterations by mass spectrometry and gene expression analysis. GSCs self-renewal capacity was determined using extreme limiting dilution assays and invasion of glioblastoma cells using a 3D spheroid model. Results Temozolomide resistance of glioblastoma cells was increased under hypoxia. However, combination of temozolomide (48 µM) with SKI-II (2.66 µM) synergistically inhibited glioblastoma cell growth and potentiated glioblastoma cell death relative to single treatments under hypoxia. This low-dose combination did not induce dihydrosphingolipids accumulation, but a decrease in ceramide and its metabolites. It induced oxidative and endoplasmic reticulum stress and triggered caspase-independent cell death. It impaired the self-renewal capacity of temozolomide-resistant GSCs, especially under hypoxia. Furthermore, it decreased invasion of glioblastoma cell spheroids. Conclusions This in vitro study provides novel insights on the links between sphingolipid metabolism and invasion, a hallmark of cancer, and cancer stem cells, key drivers of cancer. It demonstrates the therapeutic potential of approaches that combine modulation of sphingolipid metabolism with first-line agent temozolomide in overcoming tumor growth and relapse by reducing hypoxia-induced resistance to chemotherapy and by targeting both differentiated and stem glioblastoma cells.
first_indexed 2024-03-09T15:06:31Z
format Article
id doaj.art-0ba310161a6d4a26acf6da2093cbc059
institution Directory Open Access Journal
issn 1471-2407
language English
last_indexed 2024-03-09T15:06:31Z
publishDate 2023-08-01
publisher BMC
record_format Article
series BMC Cancer
spelling doaj.art-0ba310161a6d4a26acf6da2093cbc0592023-11-26T13:37:10ZengBMCBMC Cancer1471-24072023-08-0123112310.1186/s12885-023-11271-wTargeting sphingolipid metabolism with the sphingosine kinase inhibitor SKI-II overcomes hypoxia-induced chemotherapy resistance in glioblastoma cells: effects on cell death, self-renewal, and invasionNadia Sousa0Carsten Geiß1Laura Bindila2Ingo Lieberwirth3Ella Kim4Anne Régnier-Vigouroux5Institute of Developmental Biology & Neurobiology, Johannes Gutenberg University MainzInstitute of Developmental Biology & Neurobiology, Johannes Gutenberg University MainzClinical Lipidomics Unit, Institute of Physiological Chemistry, Medical University MainzMax Planck Institute for Polymer ResearchDepartment of Neurosurgery, Medical University of MainzInstitute of Developmental Biology & Neurobiology, Johannes Gutenberg University MainzAbstract Background Glioblastoma patients commonly develop resistance to temozolomide chemotherapy. Hypoxia, which supports chemotherapy resistance, favors the expansion of glioblastoma stem cells (GSC), contributing to tumor relapse. Because of a deregulated sphingolipid metabolism, glioblastoma tissues contain high levels of the pro-survival sphingosine-1-phosphate and low levels of the pro-apoptotic ceramide. The latter can be metabolized to sphingosine-1-phosphate by sphingosine kinase (SK) 1 that is overexpressed in glioblastoma. The small molecule SKI-II inhibits SK and dihydroceramide desaturase 1, which converts dihydroceramide to ceramide. We previously reported that SKI-II combined with temozolomide induces caspase-dependent cell death, preceded by dihydrosphingolipids accumulation and autophagy in normoxia. In the present study, we investigated the effects of a low-dose combination of temozolomide and SKI-II under normoxia and hypoxia in glioblastoma cells and patient-derived GCSs. Methods Drug synergism was analyzed with the Chou-Talalay Combination Index method. Dose–effect curves of each drug were determined with the Sulforhodamine B colorimetric assay. Cell death mechanisms and autophagy were analyzed by immunofluorescence, flow cytometry and western blot; sphingolipid metabolism alterations by mass spectrometry and gene expression analysis. GSCs self-renewal capacity was determined using extreme limiting dilution assays and invasion of glioblastoma cells using a 3D spheroid model. Results Temozolomide resistance of glioblastoma cells was increased under hypoxia. However, combination of temozolomide (48 µM) with SKI-II (2.66 µM) synergistically inhibited glioblastoma cell growth and potentiated glioblastoma cell death relative to single treatments under hypoxia. This low-dose combination did not induce dihydrosphingolipids accumulation, but a decrease in ceramide and its metabolites. It induced oxidative and endoplasmic reticulum stress and triggered caspase-independent cell death. It impaired the self-renewal capacity of temozolomide-resistant GSCs, especially under hypoxia. Furthermore, it decreased invasion of glioblastoma cell spheroids. Conclusions This in vitro study provides novel insights on the links between sphingolipid metabolism and invasion, a hallmark of cancer, and cancer stem cells, key drivers of cancer. It demonstrates the therapeutic potential of approaches that combine modulation of sphingolipid metabolism with first-line agent temozolomide in overcoming tumor growth and relapse by reducing hypoxia-induced resistance to chemotherapy and by targeting both differentiated and stem glioblastoma cells.https://doi.org/10.1186/s12885-023-11271-wHypoxiaGlioblastoma therapyDrug combinationTemozolomideSKI-IISphingolipids
spellingShingle Nadia Sousa
Carsten Geiß
Laura Bindila
Ingo Lieberwirth
Ella Kim
Anne Régnier-Vigouroux
Targeting sphingolipid metabolism with the sphingosine kinase inhibitor SKI-II overcomes hypoxia-induced chemotherapy resistance in glioblastoma cells: effects on cell death, self-renewal, and invasion
BMC Cancer
Hypoxia
Glioblastoma therapy
Drug combination
Temozolomide
SKI-II
Sphingolipids
title Targeting sphingolipid metabolism with the sphingosine kinase inhibitor SKI-II overcomes hypoxia-induced chemotherapy resistance in glioblastoma cells: effects on cell death, self-renewal, and invasion
title_full Targeting sphingolipid metabolism with the sphingosine kinase inhibitor SKI-II overcomes hypoxia-induced chemotherapy resistance in glioblastoma cells: effects on cell death, self-renewal, and invasion
title_fullStr Targeting sphingolipid metabolism with the sphingosine kinase inhibitor SKI-II overcomes hypoxia-induced chemotherapy resistance in glioblastoma cells: effects on cell death, self-renewal, and invasion
title_full_unstemmed Targeting sphingolipid metabolism with the sphingosine kinase inhibitor SKI-II overcomes hypoxia-induced chemotherapy resistance in glioblastoma cells: effects on cell death, self-renewal, and invasion
title_short Targeting sphingolipid metabolism with the sphingosine kinase inhibitor SKI-II overcomes hypoxia-induced chemotherapy resistance in glioblastoma cells: effects on cell death, self-renewal, and invasion
title_sort targeting sphingolipid metabolism with the sphingosine kinase inhibitor ski ii overcomes hypoxia induced chemotherapy resistance in glioblastoma cells effects on cell death self renewal and invasion
topic Hypoxia
Glioblastoma therapy
Drug combination
Temozolomide
SKI-II
Sphingolipids
url https://doi.org/10.1186/s12885-023-11271-w
work_keys_str_mv AT nadiasousa targetingsphingolipidmetabolismwiththesphingosinekinaseinhibitorskiiiovercomeshypoxiainducedchemotherapyresistanceinglioblastomacellseffectsoncelldeathselfrenewalandinvasion
AT carstengeiß targetingsphingolipidmetabolismwiththesphingosinekinaseinhibitorskiiiovercomeshypoxiainducedchemotherapyresistanceinglioblastomacellseffectsoncelldeathselfrenewalandinvasion
AT laurabindila targetingsphingolipidmetabolismwiththesphingosinekinaseinhibitorskiiiovercomeshypoxiainducedchemotherapyresistanceinglioblastomacellseffectsoncelldeathselfrenewalandinvasion
AT ingolieberwirth targetingsphingolipidmetabolismwiththesphingosinekinaseinhibitorskiiiovercomeshypoxiainducedchemotherapyresistanceinglioblastomacellseffectsoncelldeathselfrenewalandinvasion
AT ellakim targetingsphingolipidmetabolismwiththesphingosinekinaseinhibitorskiiiovercomeshypoxiainducedchemotherapyresistanceinglioblastomacellseffectsoncelldeathselfrenewalandinvasion
AT anneregniervigouroux targetingsphingolipidmetabolismwiththesphingosinekinaseinhibitorskiiiovercomeshypoxiainducedchemotherapyresistanceinglioblastomacellseffectsoncelldeathselfrenewalandinvasion