Critical role for lysyl oxidase in mesenchymal stem cell-driven breast cancer malignancy

Publication Type:

Journal Article

Authors:

El-Haibi, Christelle P; Bell, George W; Zhang, Jiangwen; Collmann, Anthony Y; Wood, David; Scherber, Cally M; Csizmadia, Eva; Mariani, Odette; Zhu, Cuihua; Campagne, Antoine; Toner, Mehmet; Sangeeta N Bhatia; Irimia, Daniel; Vincent-Salomon, Anne; Karnoub, Antoine E

Source:

Proc Natl Acad Sci U S A, Volume 109, Issue 43, p.17460-5 (2012)

Abstract:

Mesenchymal stem cells (MSCs) are multipotent progenitor cells with the ability to differentiate into multiple mesoderm lineages in the course of normal tissue homeostasis or during injury. We have previously shown that MSCs migrate to sites of tumorigenesis, where they become activated by cancer cells to promote metastasis. However, the molecular and phenotypic attributes of the MSCinduced metastatic state of the cancer cells remained undetermined. Here, we show that bone marrow-derived human MSCs promote de novo production of lysyl oxidase (LOX) from human breast carcinoma cells, which is sufficient to enhance the metastasis of otherwise weakly metastatic cancer cells to the lungs and bones. We also show that LOX is an essential component of the CD44- Twist signaling axis, in which extracellular hyaluronan causes nuclear translocation of CD44 in the cancer cells, thus triggering LOX transcription by associating with its promoter. Processed and enzymatically active LOX, in turn, stimulates Twist transcription, which mediates the MSC-triggered epithelial-to-mesenchymal transition (EMT) of carcinoma cells. Surprisingly, although induction of EMT in breast cancer cells has been tightly associated with the generation of cancer stem cells, we find that LOX, despite being critical for EMT, does not contribute to the ability of MSCs to promote the formation of cancer stem cells in the carcinoma cell populations. Collectively, our studies highlight a critical role for LOX in cancer metastasis and indicate that the signaling pathways controlling stroma-induced EMT are distinct from pathways regulating the development of cancer stem cells.

Manuscript + Supporting information (PDF)

Previous
Previous

A combinatorial extracellular matrix platform identifies cell-extracellular matrix interactions that correlate with metastasis

Next
Next

Cancer Vulnerabilities Unveiled by Genomic Loss