Oncotarget

Research Papers:

The anti-tumor drug 2-hydroxyoleic acid (Minerval) stimulates signaling and retrograde transport

Maria L. Torgersen _, Tove Irene Klokk, Simona Kavaliauskiene, Christian Klose, Kai Simons, Tore Skotland and Kirsten Sandvig

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2016; 7:86871-86888. https://doi.org/10.18632/oncotarget.13508

Metrics: PDF 2224 views  |   HTML 3637 views  |   ?  


Abstract

Maria L. Torgersen1, Tove Irene Klokk1, Simona Kavaliauskiene1,2, Christian Klose3, Kai Simons3, Tore Skotland1, Kirsten Sandvig1,2

1Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Norway, and Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Norway

2Department of Molecular Biosciences, University of Oslo, Norway

3Lipotype GmbH, Dresden, Germany

Correspondence to:

Maria L. Torgersen, email: [email protected]

Keywords: hydroxyoleic acid, minerval, membrane lipid therapy, EGFR, ricin

Received: June 20, 2016     Accepted: October 17, 2016     Published: November 22, 2016

ABSTRACT

2-hydroxyoleic acid (OHOA, Minerval®) is an example of a substance used for membrane lipid therapy, where the cellular membranes rather than specific proteins constitute the therapeutical target. OHOA is thought to mediate its anti-tumor effect by affecting the biophysical properties of membranes, which leads to altered recruitment and activation of amphitropic proteins, altered cellular signaling, and eventual cell death. Little is known about the initial signaling events upon treatment with OHOA, and whether the altered membrane properties would have any impact on the dynamic intracellular transport system. In the present study we demonstrate that treatment with OHOA led to a rapid release of intracellular calcium and activation of multiple signaling pathways in HeLa cells, including the PI3K-AKT1-MTOR pathway and several MAP kinases, in a process independent of the EGFR. By lipidomics we confirmed that OHOA was incorporated into several lipid classes. Concomitantly, OHOA potently increased retrograde transport of the plant toxin ricin from endosomes to the Golgi and further to the endoplasmic reticulum. The OHOA-stimulated ricin transport seemed to require several amphitropic proteins, including Src, phospholipase C, protein kinase C, and also Ca2+/calmodulin. Interestingly, OHOA induced a slight increase in endosomal localization of the retromer component VPS35. Thus, our data show that addition of a lipid known to alter membrane properties not only affects signaling, but also intracellular transport.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 13508