[HTML][HTML] Exosome-mediated transfer of miR-10b promotes cell invasion in breast cancer

R Singh, R Pochampally, K Watabe, Z Lu, YY Mo - Molecular cancer, 2014 - Springer
R Singh, R Pochampally, K Watabe, Z Lu, YY Mo
Molecular cancer, 2014Springer
Introduction Exosomes are 30-100 nm membrane vesicles of endocytic origin, mediating
diverse biological functions including tumor cell invasion, cell-cell communication and
antigen presentation through transfer of proteins, mRNAs and microRNAs. Recent evidence
suggests that microRNAs can be released through ceramide-dependent secretory
machinery regulated by neutral sphingomyelinase 2 (nSMase2) enzyme encoded by the
smpd3 gene that triggers exosome secretion. However, whether exosome-mediated …
Introduction
Exosomes are 30-100 nm membrane vesicles of endocytic origin, mediating diverse biological functions including tumor cell invasion, cell-cell communication and antigen presentation through transfer of proteins, mRNAs and microRNAs. Recent evidence suggests that microRNAs can be released through ceramide-dependent secretory machinery regulated by neutral sphingomyelinase 2 (nSMase2) enzyme encoded by the smpd3 gene that triggers exosome secretion. However, whether exosome-mediated microRNA transfer plays any role in cell invasion remains poorly understood. Thus, the aim of this study was to identify the exosomal microRNAs involved in breast cancer invasion.
Methods
The expression level of endogenous and exosomal miRNAs were examined by real time PCR and the expression level of target proteins were detected by western blot. Scanning electron and confocal microscopy were used to characterize exosomes and to study its uptake and transfer. Luciferase reporter plasmids and its mutant were used to confirm direct targeting. Furthermore, the functional significance of exosomal miR-10b was estimated by invasion assay.
Results
In this study, we demonstrate that microRNA carrying exosomes can be transferred among different cell lines through direct uptake. miR-10b is highly expressed in metastatic breast cancer MDA-MB-231 cells as compared to non-metastatic breast cancer cells or non-malignant breast cells; it is actively secreted into medium via exosomes. In particular, nSMase2 or ceramide promotes the exosome-mediated miR-10b secretion whereas ceramide inhibitor suppresses this secretion. Moreover, upon uptake, miR-10b can suppress the protein level of its target genes such as HOXD10 and KLF4, indicating its functional significance. Finally, treatment with exosomes derived from MDA-MB-231 cells could induce the invasion ability of non-malignant HMLE cells.
Conclusion
Together, our results suggest that a set of specific microRNAs may play an important role in modulating tumor microenvironment through exosomes. Thus, a better understanding of this process may aid in the development of novel therapeutic agents.
Springer