Cargando…

The molecular mechanism by which saturated lysophosphatidylcholine attenuates the metastatic capacity of melanoma cells

Lysophophatidylcholine (LysoPC) is an abundant constituent in human plasma. Patients with malignant cancer diseases have attenuated LysoPC plasma levels, and thus LysoPC has been examined as a metabolic biomarker for cancer prediction. Preclinical studies have shown that solid tumor cells drasticall...

Descripción completa

Detalles Bibliográficos
Autores principales: Ross, Thomas, Jakubzig, Bastian, Grundmann, Manuel, Massing, Ulrich, Kostenis, Evi, Schlesinger, Martin, Bendas, Gerd
Formato: Online Artículo Texto
Lenguaje:English
Publicado: John Wiley and Sons Inc. 2016
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5324772/
https://www.ncbi.nlm.nih.gov/pubmed/28255537
http://dx.doi.org/10.1002/2211-5463.12152
_version_ 1782510261666578432
author Ross, Thomas
Jakubzig, Bastian
Grundmann, Manuel
Massing, Ulrich
Kostenis, Evi
Schlesinger, Martin
Bendas, Gerd
author_facet Ross, Thomas
Jakubzig, Bastian
Grundmann, Manuel
Massing, Ulrich
Kostenis, Evi
Schlesinger, Martin
Bendas, Gerd
author_sort Ross, Thomas
collection PubMed
description Lysophophatidylcholine (LysoPC) is an abundant constituent in human plasma. Patients with malignant cancer diseases have attenuated LysoPC plasma levels, and thus LysoPC has been examined as a metabolic biomarker for cancer prediction. Preclinical studies have shown that solid tumor cells drastically degrade LysoPCs by incorporating their free fatty acids into cell membrane phospholipids. In this way, LysoPC C18:0 reduced the metastatic spread of murine melanoma B16.F10 cells in mice. Although membrane rigidification may have a key role in the attenuation of metastasis, evidence for this has yet to be shown. Therefore, the present study aimed to determine how LysoPC reduces the metastatic capacity of B16.F10 cells. Following cellular preincubation with LysoPC C18:0 at increasing concentrations and lengths of time, cell migration was most significantly attenuated with 450 μm LysoPC C18:0 at 72 h. Biosensor measurements suggest that, despite their abundance in B16.F10 cells, LysoPC‐sensitive G protein‐coupled receptors do not appear to contribute to this effect. Instead, the attenuated migration appears to result from changes in cell membrane properties and their effect on underlying signaling pathways, most likely the formation of focal adhesion complexes. Treatment with 450 μm LysoPC C18:0 activates protein kinase C (PKC)δ to phosphorylate syndecan‐4, accompanied by deactivation of PKCα. Subsequently, focal adhesion complex formation was attenuated, as confirmed by the reduced activity of focal adhesion kinase (FAK). Interestingly, 450 μm LysoPC C18:1 did not affect FAK activity, explaining its lower propensity to affect migration and metastasis. Therefore, membrane rigidification by LysoPC C18:0 appears to prevent the formation of focal adhesion complexes, thus affecting integrin activity as a key for metastatic melanoma spread.
format Online
Article
Text
id pubmed-5324772
institution National Center for Biotechnology Information
language English
publishDate 2016
publisher John Wiley and Sons Inc.
record_format MEDLINE/PubMed
spelling pubmed-53247722017-03-02 The molecular mechanism by which saturated lysophosphatidylcholine attenuates the metastatic capacity of melanoma cells Ross, Thomas Jakubzig, Bastian Grundmann, Manuel Massing, Ulrich Kostenis, Evi Schlesinger, Martin Bendas, Gerd FEBS Open Bio Research Articles Lysophophatidylcholine (LysoPC) is an abundant constituent in human plasma. Patients with malignant cancer diseases have attenuated LysoPC plasma levels, and thus LysoPC has been examined as a metabolic biomarker for cancer prediction. Preclinical studies have shown that solid tumor cells drastically degrade LysoPCs by incorporating their free fatty acids into cell membrane phospholipids. In this way, LysoPC C18:0 reduced the metastatic spread of murine melanoma B16.F10 cells in mice. Although membrane rigidification may have a key role in the attenuation of metastasis, evidence for this has yet to be shown. Therefore, the present study aimed to determine how LysoPC reduces the metastatic capacity of B16.F10 cells. Following cellular preincubation with LysoPC C18:0 at increasing concentrations and lengths of time, cell migration was most significantly attenuated with 450 μm LysoPC C18:0 at 72 h. Biosensor measurements suggest that, despite their abundance in B16.F10 cells, LysoPC‐sensitive G protein‐coupled receptors do not appear to contribute to this effect. Instead, the attenuated migration appears to result from changes in cell membrane properties and their effect on underlying signaling pathways, most likely the formation of focal adhesion complexes. Treatment with 450 μm LysoPC C18:0 activates protein kinase C (PKC)δ to phosphorylate syndecan‐4, accompanied by deactivation of PKCα. Subsequently, focal adhesion complex formation was attenuated, as confirmed by the reduced activity of focal adhesion kinase (FAK). Interestingly, 450 μm LysoPC C18:1 did not affect FAK activity, explaining its lower propensity to affect migration and metastasis. Therefore, membrane rigidification by LysoPC C18:0 appears to prevent the formation of focal adhesion complexes, thus affecting integrin activity as a key for metastatic melanoma spread. John Wiley and Sons Inc. 2016-11-24 /pmc/articles/PMC5324772/ /pubmed/28255537 http://dx.doi.org/10.1002/2211-5463.12152 Text en © 2016 The Authors. Published by FEBS Press and John Wiley & Sons Ltd. This is an open access article under the terms of the Creative Commons Attribution (http://creativecommons.org/licenses/by/4.0/) License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.
spellingShingle Research Articles
Ross, Thomas
Jakubzig, Bastian
Grundmann, Manuel
Massing, Ulrich
Kostenis, Evi
Schlesinger, Martin
Bendas, Gerd
The molecular mechanism by which saturated lysophosphatidylcholine attenuates the metastatic capacity of melanoma cells
title The molecular mechanism by which saturated lysophosphatidylcholine attenuates the metastatic capacity of melanoma cells
title_full The molecular mechanism by which saturated lysophosphatidylcholine attenuates the metastatic capacity of melanoma cells
title_fullStr The molecular mechanism by which saturated lysophosphatidylcholine attenuates the metastatic capacity of melanoma cells
title_full_unstemmed The molecular mechanism by which saturated lysophosphatidylcholine attenuates the metastatic capacity of melanoma cells
title_short The molecular mechanism by which saturated lysophosphatidylcholine attenuates the metastatic capacity of melanoma cells
title_sort molecular mechanism by which saturated lysophosphatidylcholine attenuates the metastatic capacity of melanoma cells
topic Research Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5324772/
https://www.ncbi.nlm.nih.gov/pubmed/28255537
http://dx.doi.org/10.1002/2211-5463.12152
work_keys_str_mv AT rossthomas themolecularmechanismbywhichsaturatedlysophosphatidylcholineattenuatesthemetastaticcapacityofmelanomacells
AT jakubzigbastian themolecularmechanismbywhichsaturatedlysophosphatidylcholineattenuatesthemetastaticcapacityofmelanomacells
AT grundmannmanuel themolecularmechanismbywhichsaturatedlysophosphatidylcholineattenuatesthemetastaticcapacityofmelanomacells
AT massingulrich themolecularmechanismbywhichsaturatedlysophosphatidylcholineattenuatesthemetastaticcapacityofmelanomacells
AT kostenisevi themolecularmechanismbywhichsaturatedlysophosphatidylcholineattenuatesthemetastaticcapacityofmelanomacells
AT schlesingermartin themolecularmechanismbywhichsaturatedlysophosphatidylcholineattenuatesthemetastaticcapacityofmelanomacells
AT bendasgerd themolecularmechanismbywhichsaturatedlysophosphatidylcholineattenuatesthemetastaticcapacityofmelanomacells
AT rossthomas molecularmechanismbywhichsaturatedlysophosphatidylcholineattenuatesthemetastaticcapacityofmelanomacells
AT jakubzigbastian molecularmechanismbywhichsaturatedlysophosphatidylcholineattenuatesthemetastaticcapacityofmelanomacells
AT grundmannmanuel molecularmechanismbywhichsaturatedlysophosphatidylcholineattenuatesthemetastaticcapacityofmelanomacells
AT massingulrich molecularmechanismbywhichsaturatedlysophosphatidylcholineattenuatesthemetastaticcapacityofmelanomacells
AT kostenisevi molecularmechanismbywhichsaturatedlysophosphatidylcholineattenuatesthemetastaticcapacityofmelanomacells
AT schlesingermartin molecularmechanismbywhichsaturatedlysophosphatidylcholineattenuatesthemetastaticcapacityofmelanomacells
AT bendasgerd molecularmechanismbywhichsaturatedlysophosphatidylcholineattenuatesthemetastaticcapacityofmelanomacells