Abstract

Tumor cells use broad spectrum proteolytic activity of plasmin to invade tissue and form metastatic foci. Cell surface-associated enolase-1 (ENO-1) enhances plasmin formation and thus participates in the regulation of pericellular proteolysis. Although increased levels of cell surface bound ENO-1 have been described in different types of cancer, the molecular mechanism responsible for ENO-1 exteriorization remains elusive. In the present study, increased ENO-1 protein levels were found in ductal breast carcinoma and on the cell surface of highly metastatic breast cancer cell line MDA-MB-231. Elevated cell surface-associated ENO-1 expression correlated with augmented MDA-MB-231 cell migratory and invasive properties. Exposure of MDA-MB-231 cells to LPS potentiated translocation of ENO-1 to the cell surface and its release into the extracellular space in the form of exosomes. These effects were independent of de novo protein synthesis and did not require the classical endoplasmic reticulum/Golgi pathway. LPS-triggered ENO-1 exteriorization was suppressed by pretreatment of MDA-MB-231 cells with the Ca(2+) chelator BAPTA or an inhibitor of endoplasmic reticulum Ca(2+)-ATPase pump, cyclopiazonic acid. In line with these observations, the stromal interaction molecule (STIM) 1 and the calcium release-activated calcium modulator (ORAI) 1-mediated store-operated Ca(2+) entry were found to regulate LPS-induced ENO-1 exteriorization. Pharmacological blockage or knockdown of STIM1 or ORAI1 reduced ENO-1-dependent migration of MDA-MB-231 cells. Collectively, our results demonstrate the pivotal role of store-operated Ca(2+) channel-mediated Ca(2+) influx in the regulation of ENO-1 exteriorization and thus in the modulation of cancer cell migratory and invasive properties.

Highlights

  • Cell surface-associated enolase-1 regulates plasmin formation and pericellular proteolysis

  • Because the highest ENO-1 expression was detected in ductal carcinoma of the breast, in further studies we focused on the role of ENO-1 in breast cancer cell motility

  • Enolase-1 is a glycolytic enzyme primarily localized in the cytoplasm of prokaryotic and eukaryotic cells [12]

Read more

Summary

Background

Cell surface-associated enolase-1 regulates plasmin formation and pericellular proteolysis. Under stimulatory conditions, it can be translocated to the cell surface where it interacts with PLG and participates in the regulation of pericellular proteolysis, allowing cells to invade tissue [13,14,15] This fact can explain the positive correlation between high levels of ENO-1 present on the cell surface of cancer cells (16 –19) and their increased invasive potential [20, 21]. Given the fact that LPS up-regulates cell surface expression of ENO-1 on monocytoic cells, thereby increasing their invasive potential [14], LPS-triggered regulation of ENO-1-dependent pericellular proteolytic activity could represent another mechanism that contributes to cancer cell spreading and to metastasis formation. We investigated the mechanism and the functional consequence of LPS-triggered ENO-1 transport to the cell surface and to the extracellular milieu

EXPERIMENTAL PROCEDURES
F: ATTGCCGACAGGATGCAGGAA
RESULTS
E GFP-EV
B Exosomes
A GFP-EV
F LPS BAPTA ENO-1
G SC peptide ENO-1 peptide SC peptide ENO-1 peptide
DISCUSSION
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call