Oncotarget

Research Papers:

Divergent roles of lysyl oxidase family members in ornithine decarboxylase- and RAS-transformed mouse fibroblasts and human melanoma cells

Mari Kielosto, Johanna Eriksson, Pirjo Nummela, Miao Yin and Erkki Hölttä _

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2018; 9:37733-37752. https://doi.org/10.18632/oncotarget.26508

Metrics: PDF 1573 views  |   HTML 2027 views  |   ?  


Abstract

Mari Kielosto1, Johanna Eriksson1, Pirjo Nummela1,2, Miao Yin1 and Erkki Hölttä1

1Department of Pathology, University of Helsinki, Helsinki, Finland

2Current address: University of Helsinki, Genome-Scale Biology Research Program, Helsinki, Finland

Correspondence to:

Erkki Hölttä, email: [email protected]

Keywords: lysyl oxidase family; c-Jun; ODC; RAS; melanoma

Received: March 26, 2018     Accepted: December 13, 2018     Published: December 28, 2018

ABSTRACT

We have previously shown that proto-oncoprotein c-Jun is activated in ornithine decarboxylase (ODC)- and RAS-transformed mouse fibroblasts, and that the transformed morphology of these cells can be reversed by expressing the transactivation domain deletion mutant of c-Jun (TAM67). Here, we found that lysyl oxidase (Lox), encoding an extracellular matrix-modifying enzyme, is downregulated in a c-Jun-dependent manner in ODC-transformed fibroblasts (Odc cells). In addition to Lox, the Lox family members Lox-like 1 and 3 (Loxl1 and Loxl3) were found to be downregulated in Odc as well as in RAS-transformed fibroblasts (E4), whereas Lox-like 4 (Loxl4) was upregulated in Odc and downregulated in E4 cells compared to normal N1 fibroblasts. Tetracycline-regulatable LOX re-expression in Odc cells led to inhibition of cell growth and invasion in three-dimensional Matrigel in an activity-independent manner. On the contrary, LOX and especially LOXL2, LOXL3, and LOXL4 were found to be upregulated in several human melanoma cell lines, and LOX inhibitor B-aminopropionitrile inhibited the invasive growth of these cells particularly when co-cultured with fibroblasts in Matrigel. Knocking down the expression of LOX and especially LOXL2 in melanoma cells almost completely abrogated the invasive growth capability. Further, LOXL2 was significantly upregulated in clinical human primary melanomas compared to benign nevi, and high expression of LOXL2 in primary melanomas was associated with formation of metastases and shorter survival of patients. Thus, our studies reveal that inactive pro-LOX (together with Lox propeptide) functions as a tumor suppressor in ODC- and RAS-transformed murine fibroblasts by inhibiting cell growth and invasion, and active LOX and LOXL2 as tumor promoters in human melanoma cells by promoting their invasive growth.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 26508