Cargando…

The role of reactive oxygen species and autophagy in safingol-induced cell death

Safingol is a sphingolipid with promising anticancer potential, which is currently in phase I clinical trial. Yet, the underlying mechanisms of its action remain largely unknown. We reported here that safingol-induced primarily accidental necrotic cell death in MDA-MB-231 and HT-29 cells, as shown b...

Descripción completa

Detalles Bibliográficos
Autores principales: Ling, L-U, Tan, K-B, Lin, H, Chiu, G N C
Formato: Texto
Lenguaje:English
Publicado: Nature Publishing Group 2011
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3101809/
https://www.ncbi.nlm.nih.gov/pubmed/21390063
http://dx.doi.org/10.1038/cddis.2011.12
_version_ 1782204310750232576
author Ling, L-U
Tan, K-B
Lin, H
Chiu, G N C
author_facet Ling, L-U
Tan, K-B
Lin, H
Chiu, G N C
author_sort Ling, L-U
collection PubMed
description Safingol is a sphingolipid with promising anticancer potential, which is currently in phase I clinical trial. Yet, the underlying mechanisms of its action remain largely unknown. We reported here that safingol-induced primarily accidental necrotic cell death in MDA-MB-231 and HT-29 cells, as shown by the increase in the percentage of cells stained positive for 7-aminoactinomycin , collapse of mitochondria membrane potential and depletion of intracellular ATP. Importantly, safingol treatment produced time- and concentration-dependent reactive oxygen species (ROS) generation. Autophagy was triggered following safingol treatment, as reflected by the formation of autophagosomes, acidic vacuoles, increased light chain 3-II and Atg biomarkers expression. Interestingly, scavenging ROS with N-acetyl--cysteine could prevent the autophagic features and reverse safingol-induced necrosis. Our data also suggested that autophagy was a cell repair mechanism, as suppression of autophagy by 3-methyladenine or bafilomycin A1 significantly augmented cell death on 2-5 μ safingol treatment. In addition, Bcl-xL and Bax might be involved in the regulation of safingol-induced autophagy. Finally, glucose uptake was shown to be inhibited by safingol treatment, which was associated with an increase in p-AMPK expression. Taken together, our data suggested that ROS was the mediator of safingol-induced cancer cell death, and autophagy is likely to be a mechanism triggered to repair damages from ROS generation on safingol treatment.
format Text
id pubmed-3101809
institution National Center for Biotechnology Information
language English
publishDate 2011
publisher Nature Publishing Group
record_format MEDLINE/PubMed
spelling pubmed-31018092011-07-05 The role of reactive oxygen species and autophagy in safingol-induced cell death Ling, L-U Tan, K-B Lin, H Chiu, G N C Cell Death Dis Original Article Safingol is a sphingolipid with promising anticancer potential, which is currently in phase I clinical trial. Yet, the underlying mechanisms of its action remain largely unknown. We reported here that safingol-induced primarily accidental necrotic cell death in MDA-MB-231 and HT-29 cells, as shown by the increase in the percentage of cells stained positive for 7-aminoactinomycin , collapse of mitochondria membrane potential and depletion of intracellular ATP. Importantly, safingol treatment produced time- and concentration-dependent reactive oxygen species (ROS) generation. Autophagy was triggered following safingol treatment, as reflected by the formation of autophagosomes, acidic vacuoles, increased light chain 3-II and Atg biomarkers expression. Interestingly, scavenging ROS with N-acetyl--cysteine could prevent the autophagic features and reverse safingol-induced necrosis. Our data also suggested that autophagy was a cell repair mechanism, as suppression of autophagy by 3-methyladenine or bafilomycin A1 significantly augmented cell death on 2-5 μ safingol treatment. In addition, Bcl-xL and Bax might be involved in the regulation of safingol-induced autophagy. Finally, glucose uptake was shown to be inhibited by safingol treatment, which was associated with an increase in p-AMPK expression. Taken together, our data suggested that ROS was the mediator of safingol-induced cancer cell death, and autophagy is likely to be a mechanism triggered to repair damages from ROS generation on safingol treatment. Nature Publishing Group 2011-03 2011-03-10 /pmc/articles/PMC3101809/ /pubmed/21390063 http://dx.doi.org/10.1038/cddis.2011.12 Text en Copyright © 2011 Macmillan Publishers Limited http://creativecommons.org/licenses/by-nc-nd/3.0/ This work is licensed under the Creative Commons Attribution-NonCommercial-No Derivative Works 3.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/3.0/
spellingShingle Original Article
Ling, L-U
Tan, K-B
Lin, H
Chiu, G N C
The role of reactive oxygen species and autophagy in safingol-induced cell death
title The role of reactive oxygen species and autophagy in safingol-induced cell death
title_full The role of reactive oxygen species and autophagy in safingol-induced cell death
title_fullStr The role of reactive oxygen species and autophagy in safingol-induced cell death
title_full_unstemmed The role of reactive oxygen species and autophagy in safingol-induced cell death
title_short The role of reactive oxygen species and autophagy in safingol-induced cell death
title_sort role of reactive oxygen species and autophagy in safingol-induced cell death
topic Original Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3101809/
https://www.ncbi.nlm.nih.gov/pubmed/21390063
http://dx.doi.org/10.1038/cddis.2011.12
work_keys_str_mv AT linglu theroleofreactiveoxygenspeciesandautophagyinsafingolinducedcelldeath
AT tankb theroleofreactiveoxygenspeciesandautophagyinsafingolinducedcelldeath
AT linh theroleofreactiveoxygenspeciesandautophagyinsafingolinducedcelldeath
AT chiugnc theroleofreactiveoxygenspeciesandautophagyinsafingolinducedcelldeath
AT linglu roleofreactiveoxygenspeciesandautophagyinsafingolinducedcelldeath
AT tankb roleofreactiveoxygenspeciesandautophagyinsafingolinducedcelldeath
AT linh roleofreactiveoxygenspeciesandautophagyinsafingolinducedcelldeath
AT chiugnc roleofreactiveoxygenspeciesandautophagyinsafingolinducedcelldeath