Back to Search Start Over

Endothelial Progenitor Cells Promote Osteosarcoma Progression and Invasiveness via AKT/PI3K Signaling.

Authors :
Doppelt-Flikshtain, Ofri
Younis, Amin
Tamari, Tal
Ginesin, Ofir
Shentzer-Kutiel, Talia
Nikomarov, David
Bar-Sela, Gil
Coyac, Benjamin R.
Assaraf, Yehuda G.
Zigdon-Giladi, Hadar
Source :
Cancers; Mar2023, Vol. 15 Issue 6, p1818, 20p
Publication Year :
2023

Abstract

Simple Summary: Although progress has been made in the treatment and survival of patients with a range of malignancies, the prognosis for patients harboring metastatic osteosarcoma (bone cancer) remains dismal due to the limited therapeutic options available. Endothelial progenitor cells (EPCs) mediate the angiogenic (blood vessel formation) switch in several cancers. Spatial proximity between EPCs and osteosarcoma in the bone led to the hypothesis that EPCs-osteosarcoma interactions may possibly promote osteosarcoma (OS) progression and aggressiveness. In the current paper, we demonstrate the non-physical role that EPCs play in OS migration and invasion, along with deciphering a potential underlying molecular mechanism. Our findings may pave the way toward the development of new EPCs-targeted therapies to inhibit OS metastasis and hence enhance the therapeutic efficacy of this devastating bone cancer. Background: Osteosarcoma (OS) mortality is attributed to lung metastases. Endothelial progenitor cells (EPCs) mediate the angiogenic switch in several cancers. The spatial proximity between EPCs and OS in the bone led to the hypothesis that EPCs-osteosarcoma interactions may possibly promote OS progression and aggressiveness. Methods: A PI3K inhibitor, Bevacizumab (an anti-VEGF-A antibody), and an anti-FGF2 antibody were added to the EPCs' conditioned medium (EPC-CM), and their impacts on OS cell (U2-OS and 143B) proliferation, migration, invasion, MMP9 expression, and AKT phosphorylation were determined. The autocrine role of VEGF-A was assessed using Bevacizumab treatment and VEGF-A silencing in OS cells. Toward this end, an orthotopic mouse OS model was established. Mouse and human tumors were immunolabeled with antibodies to the abovementioned factors. Results: EPC-CM enhanced osteosarcoma MMP9 expression, invasiveness, and migration via the PI3K/AKT pathway. The addition of Bevacizumab and an anti-FGF2 antibody to the EPC-CM diminished OS cell migration. The autocrine role of VEGF-A was assessed using Bevacizumab and VEGF-A silencing in OS cells, resulting in decreased AKT phosphorylation and, consequently, diminished invasiveness and migration. Consistently, OS xenografts in mice displayed high VEGF-A and FGF2 levels. Remarkably, lung metastasis specimens derived from OS patients exhibited marked immunolabeling of CD31, VEGF-A, and FGF2. Conclusions: EPCs promote OS progression not only by physically incorporating into blood vessels, but also by secreting cytokines, which act via paracrine signaling. EPCs induced in vitro MMP9 overexpression, invasion, and migration. Additional animal studies are warranted to further expand these results. These findings may pave the way toward the development of novel EPCs-targeted therapeutics aimed at blocking OS metastasis. [ABSTRACT FROM AUTHOR]

Details

Language :
English
ISSN :
20726694
Volume :
15
Issue :
6
Database :
Complementary Index
Journal :
Cancers
Publication Type :
Academic Journal
Accession number :
162751217
Full Text :
https://doi.org/10.3390/cancers15061818